Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Brain Behav Immun ; 72: 22-33, 2018 08.
Article in English | MEDLINE | ID: mdl-29175543

ABSTRACT

Inflammation is a ubiquitous but poorly understood consequence of spinal cord injury (SCI). The mechanisms controlling this response are unclear but culminate in the sequential activation of resident and recruited immune cells. Collectively, these cells can exert divergent effects on cell survival and tissue repair. HMGB1 is a ubiquitously expressed DNA binding protein and also a potent inflammatory stimulus. Necrotic cells release HGMB1, but HMGB1 also is actively secreted by inflammatory macrophages. A goal of this study was to quantify spatio-temporal patterns of cellular HMGB1 expression in a controlled mouse model of experimental SCI then determine the effects of HMGB1 on post-SCI neuroinflammation and recovery of function. We documented SCI-induced changes in nuclear and cytoplasmic distribution of HMGB1 in various cell types after SCI. The data reveal a time-dependent increase in HMGB1 mRNA and protein with protein reaching maximal levels 24-72 h post-injury then declining toward baseline 14-28 days post-SCI. Although most cells expressed nuclear HMGB1, reduced nuclear labeling with increased cytoplasmic expression was found in a subset of CNS macrophages suggesting that those cells begin to secrete HMGB1 at the injury site. In vitro data indicate that extracelluar HMGB1 helps promote the development of macrophages with a neurotoxic phenotype. The ability of HMGB1 to elicit neurotoxic macrophage functions was confirmed in vivo; 72 h after injecting 500 ng of recombinant HMGB1 into intact spinal cord ventral horn, inflammatory CNS macrophages co-localized with focal areas of neuronal killing. However, attempts to confer neuroprotection after SCI by blocking HMGB1 with a neutralizing antibody were unsuccessful. Collectively, these data implicate HMGB1 as a novel regulator of post-SCI inflammation and suggest that inhibition of HMGB1 could be a novel therapeutic target after SCI. Future studies will need to identify better methods to deliver optimal concentrations of HMGB1 antagonists to the injured spinal cord.


Subject(s)
HMGB1 Protein/metabolism , Spinal Cord Injuries/immunology , Spinal Cord Injuries/metabolism , Alarmins/metabolism , Alarmins/physiology , Animals , Biomarkers/blood , Brain/metabolism , Disease Models, Animal , Female , HMGB1 Protein/physiology , Inflammation/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurotoxicity Syndromes/metabolism , Seizures/etiology , Signal Transduction/physiology , Spinal Cord/metabolism , Spinal Cord Injuries/physiopathology , Toll-Like Receptor 4/metabolism
2.
Exp Neurol ; 233(1): 333-41, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22079587

ABSTRACT

It is widely believed that microglia and monocyte-derived macrophages (collectively referred to as central nervous system (CNS) macrophages) cause excitotoxicity in the diseased or injured CNS. This view has evolved mostly from in vitro studies showing that neurotoxic concentrations of glutamate are released from CNS macrophages stimulated with lipopolysaccharide (LPS), a potent inflammogen. We hypothesized that excitotoxic killing by CNS macrophages is more rigorously controlled in vivo, requiring both the activation of the glutamate/cystine antiporter (system x(c)(-)) and an increase in extracellular cystine, the substrate that drives glutamate release. Here, we show that non-traumatic microinjection of low-dose LPS into spinal cord gray matter activates CNS macrophages but without causing overt neuropathology. In contrast, neurotoxic inflammation occurs when LPS and cystine are co-injected. Simultaneous injection of NBQX, an antagonist of AMPA glutamate receptors, reduces the neurotoxic effects of LPS+cystine, implicating glutamate as a mediator of neuronal cell death in this model. Surprisingly, neither LPS nor LPS+cystine adversely affects survival of oligodendrocytes or oligodendrocyte progenitor cells. Ex vivo analyses show that redox balance in microglia and macrophages is controlled by induction of system x(c)(-) and that high GSH:GSSG ratios predict the neurotoxic potential of these cells. Together, these data indicate that modulation of redox balance in CNS macrophages, perhaps through regulating system x(c)(-), could be a novel approach for attenuating injurious neuroinflammatory cascades.


Subject(s)
Excitatory Amino Acids/toxicity , Glutamic Acid/metabolism , Macrophages/metabolism , Microglia/metabolism , Spinal Cord Diseases/chemically induced , Spinal Cord Diseases/pathology , Animals , Cystine/metabolism , Disease Models, Animal , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/toxicity , Glutathione/metabolism , Laser Capture Microdissection/methods , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Oxidation-Reduction/drug effects , Quinoxalines/pharmacology
3.
J Neurosci ; 31(27): 9910-22, 2011 Jul 06.
Article in English | MEDLINE | ID: mdl-21734283

ABSTRACT

Macrophages exert divergent effects in the injured CNS, causing either neurotoxicity or regeneration. The mechanisms regulating these divergent functions are not understood but can be attributed to the recruitment of distinct macrophage subsets and the activation of specific intracellular signaling pathways. Here, we show that impaired signaling via the chemokine receptor CX3CR1 promotes recovery after traumatic spinal cord injury (SCI) in mice. Deficient CX3CR1 signaling in intraspinal microglia and monocyte-derived macrophages (MDMs) attenuates their ability to synthesize and release inflammatory cytokines and oxidative metabolites. Also, impaired CX3CR1 signaling abrogates the recruitment or maturation of MDMs with presumed neurotoxic effects after SCI. Indeed, in wild-type mice, Ly6C(lo)/iNOS(+)/MHCII(+)/CD11c(-) MDMs dominate the lesion site, whereas CCR2(+)/Ly6C(hi)/MHCII(-)/CD11c(+) monocytes predominate in the injured spinal cord of CX3CR1-deficient mice. Replacement of wild-type MDMs with those unable to signal via CX3CR1 resulted in anatomical and functional improvements after SCI. Thus, blockade of CX3CR1 signaling represents a selective anti-inflammatory therapy that is able to promote neuroprotection, in part by reducing inflammatory signaling in microglia and MDMs and recruitment of a novel monocyte subset.


Subject(s)
Antigens, Ly/metabolism , Macrophages/metabolism , Nitric Oxide Synthase Type II/metabolism , Receptors, Chemokine/deficiency , Recovery of Function/genetics , Signal Transduction/physiology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology , Analysis of Variance , Animals , CD11 Antigens/metabolism , CX3C Chemokine Receptor 1 , Cells, Cultured , Chemokine CXCL1/genetics , Chemokine CXCL1/metabolism , Disease Models, Animal , Flow Cytometry , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/genetics , Motor Activity/physiology , Myelin Basic Protein/metabolism , Nitric Oxide/metabolism , Signal Transduction/genetics , Spinal Cord Injuries/genetics
4.
J Biol Chem ; 284(13): 8703-13, 2009 Mar 27.
Article in English | MEDLINE | ID: mdl-19196714

ABSTRACT

The type 1 interleukin-1 receptor (IL-1R1) mediates diverse functions of interleukin-1 (IL-1) in the nervous, immune, and neuroendocrine systems. It has been suggested previously that the versatile functions of IL-1 may in part be conferred by the multiple promoters of IL-1R1 that have been identified for the human IL-1R1 gene. Promoters for murine IL-1R1 (mIL-1R1) gene have not been studied in detail. We performed 5'-rapid amplification of cDNA ends to determine the transcription start sites (TSS) in mIL-1R1, using mRNAs derived from 24 different tissues. The results revealed three putative TSSs of mIL-1R1. Three full-length cDNAs containing these distinct TSSs were recovered in screens of cloned cDNA libraries. Translation of these cDNAs produced IL-1R1 proteins that were verified by Western blot analysis. IL-1 stimulation of the individual IL-1R1 proteins resulted in the activation of NF-kappaB. Promoter-reporter assay for genomic DNA sequences immediately upstream of the three TSSs validated that the sequences possess promoter activity in a cell type-specific manner. These promoters are termed P1, P2, and P3 of the mIL-1R1, in 5' to 3' order. Quantitative PCR analysis of P1-, P2-, and P3-specific mIL-1R1 mRNAs showed that there is tissue-specific distribution of these mRNAs in vivo, and there are distinct patterns of P1, P2, and P3 mRNA expression in different cell lines. In the brain, P3 mRNA is expressed preferentially in the dentate gyrus. Further, glucocorticoids differentially regulate these promoters in a cell type-specific manner. Together, these results suggest that the different IL-1R1 promoters contribute to the discrete and diverse actions of IL-1.


Subject(s)
Gene Expression Regulation/physiology , Interleukin-1/metabolism , Promoter Regions, Genetic/physiology , Receptors, Interleukin-1/biosynthesis , Animals , DNA, Complementary/genetics , Gene Library , Glucocorticoids/genetics , Glucocorticoids/metabolism , Interleukin-1/genetics , Mice , Organ Specificity/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Interleukin-1/genetics
5.
J Neurosci ; 27(39): 10476-86, 2007 Sep 26.
Article in English | MEDLINE | ID: mdl-17898219

ABSTRACT

Interleukin-1 (IL-1) has been implicated as a critical mediator of neuroimmune communication. In the brain, the functional receptor for IL-1, type 1 IL-1 receptor (IL-1R1), is localized primarily to the endothelial cells. In this study, we created an endothelial-specific IL-1R1 knockdown model to test the role of endothelial IL-1R1 in mediating the effects of IL-1. Neuronal activation in the hypothalamus was measured by c-fos expression in the paraventricular nucleus and the ventromedial preoptic area. In addition, two specific sickness symptoms, febrile response and reduction of locomotor activity, were studied. Intracerebroventricular injection of IL-1 induced leukocyte infiltration into the CNS, activation of hypothalamic neurons, fever, and reduced locomotor activity in normal mice. Endothelial-specific knockdown of IL-1R1 abrogated all these responses. Intraperitoneal injection of IL-1 also induced neuronal activation in the hypothalamus, fever, and reduced locomotor activity, without inducing leukocyte infiltration into the brain. Endothelial-specific knockdown of IL-1R1 suppressed intraperitoneal IL-1-induced fever, but not the induction of c-fos in hypothalamus. When IL-1 was given intravenously, endothelial knockdown of IL-1R1 abolished intravenous IL-1-induced CNS activation and the two monitored sickness symptoms. In addition, endothelial-specific knockdown of IL-1R1 blocked the induction of cyclooxygenase-2 expression induced by all three routes of IL-1 administration. These results show that the effects of intravenous and intracerebroventricular IL-1 are mediated by endothelial IL-1R1, whereas the effects of intraperitoneal IL-1 are partially dependent on endothelial IL-1R1.


Subject(s)
Central Nervous System/drug effects , Endothelial Cells/metabolism , Immunologic Factors/administration & dosage , Interleukin-1/administration & dosage , Motor Activity/physiology , Receptors, Interleukin-1 Type I/physiology , Animals , Cyclooxygenase 2/biosynthesis , Fever/physiopathology , Genes, fos/physiology , Hypothalamus/drug effects , Mice , Mice, Transgenic , Motor Activity/drug effects , Neuroimmunomodulation/drug effects , Receptors, Interleukin-1 Type I/drug effects
6.
J Neurochem ; 102(4): 1083-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17663750

ABSTRACT

Spinal cord injury (SCI) elicits a neuroinflammatory reaction dominated by microglia and monocyte-derived macrophages (MDM). Because MDM do not infiltrate the spinal cord until days after injury, it may be possible to control whether they differentiate into neuroprotective or neurotoxic effector cells. However, doing so will require better understanding of the factors controlling MDM differentiation and activation. Our goal was to develop an in vitro model of MDM that is relevant in the context of SCI. This tool would allow future studies to define mechanisms and intracellular signaling pathways that are associated with MDM-mediated neuroprotection or neurotoxicity. We first characterized SCI-induced cytokine expression in MDM using laser capture microdissection and real-time PCR. Based on this data, we assessed which easily procurable primary macrophage subset would mimic this phenotype in vitro. We established the baseline and inductive potential of resident peritoneal, thioglycollate-elicited peritoneal and bone marrow-derived macrophages (BMDM) at the molecular, cellular and functional level. Of these cells, only BMDM retained the phenotypic, molecular and functional characteristics of MDM that infiltrate the injured spinal cord. Thus, peripheral macrophages should not be used interchangeably in vitro to model the functional consequences of the MDM response elicited by SCI.


Subject(s)
Macrophages/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Animals , B7-1 Antigen/metabolism , CD11b Antigen/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry/methods , Gene Expression Regulation/physiology , Histocompatibility Antigens Class II/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Phagocytosis/physiology , RNA, Messenger/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction/methods , Time Factors
7.
J Neurochem ; 102(1): 37-50, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17403033

ABSTRACT

Activation of macrophages via toll-like receptors (TLRs) is important for inflammation and host defense against pathogens. Recent data suggest that non-pathogenic molecules released by trauma also can trigger inflammation via TLR2 and TLR4. Here, we tested whether TLRs are regulated after sterile spinal cord injury (SCI) and examined their effects on functional and anatomical recovery. We show that mRNA for TLR1, 2, 4, 5, and 7 are increased after SCI as are molecules associated with TLR signaling (e.g. MyD88, NFkappaB). The significance of in vivo TLR2 and TLR4 signaling was evident in SCI TLR4 mutant (C3H/HeJ) and TLR2 knockout (TLR2-/-) mice. In C3H/HeJ mice, sustained locomotor deficits were observed relative to SCI wild-type control mice and were associated with increased demyelination, astrogliosis, and macrophage activation. These changes were preceded by reduced intraspinal expression of interleukin-1beta mRNA. In TLR2-/- mice, locomotor recovery also was impaired relative to SCI wild-type controls and novel patterns of myelin pathology existed within ventromedial white matter--an area important for overground locomotion. Together, these data suggest that in the absence of pathogens, TLR2 and TLR4 are important for coordinating post-injury sequelae and perhaps in regulating inflammation and gliosis after SCI.


Subject(s)
Gliosis/pathology , Inflammation/pathology , Myelin Sheath/pathology , Spinal Cord Injuries/pathology , Toll-Like Receptor 2/physiology , Toll-Like Receptor 4/physiology , Animals , Axons/pathology , Chemokines/biosynthesis , Cytokines/biosynthesis , Female , Immunohistochemistry , In Situ Hybridization , Locomotion/physiology , Macrophages/pathology , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microdissection , Nitric Oxide Synthase Type II/biosynthesis , Nitric Oxide Synthase Type II/genetics , Oligonucleotide Array Sequence Analysis , RNA/biosynthesis , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spinal Cord Injuries/physiopathology
8.
Eur J Neurosci ; 22(8): 1977-86, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16262636

ABSTRACT

Proinflammatory cytokines, such as interleukin-1beta and tumour necrosis factor-alpha, are released by activated glial cells in the spinal cord and play a major role in pain facilitation. These cytokines exert their actions, at least partially, through the activation of the transcription factor, nuclear factor kappaB (NF-kappaB). In turn, NF-kappaB regulates the transcription of many inflammatory mediators, including cytokines. We have previously shown that intrathecal injection of the human immunodeficiency virus-1 (HIV-1) envelope glycoprotein, gp120, induces mechanical allodynia via the release of proinflammatory cytokines. Here, we investigated whether NF-kappaB is involved in gp120-induced pain behaviour in Sprague-Dawley rats. Intrathecal administration of NF-kappaB inhibitors, pyrrolidinedithiocarbamate (PDTC) and SN50, prior to gp120 partially attenuated gp120-induced allodynia. In addition, PDTC delayed and reversed allodynia in a model of neuropathic pain induced by sciatic nerve inflammation. These observations suggest that intrathecal gp120 may lead to activation of NF-kappaB within the spinal cord. To reveal NF-kappaB activation, we assessed inhibitory factor kappaBalpha (IkappaBalpha) mRNA expression by in situ hybridization, as NF-kappaB activation up-regulates IkappaBalpha gene expression as part of an autoregulatory feedback loop. No or low levels of IkappaBalpha mRNA were detected in the lumbar spinal cord of vehicle-injected rats, whereas IkappaBalpha mRNA expression was markedly induced in the spinal cord following intrathecal gp120 in predominantly astrocytes and endothelial cells. Moreover, IkappaBalpha mRNA expression positively correlated with proinflammatory cytokine protein levels in lumbosacral cerebrospinal fluid. Together, these results demonstrate that spinal cord NF-kappaB activation is involved, at least in part, in exaggerated pain states.


Subject(s)
Cytokines/cerebrospinal fluid , Gene Expression Regulation/physiology , NF-kappa B/metabolism , Pain/metabolism , Spinal Cord/metabolism , Animals , Antioxidants , Calcium-Binding Proteins/metabolism , Cell Count/methods , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Activation , Enzyme Inhibitors/administration & dosage , Enzyme-Linked Immunosorbent Assay/methods , Functional Laterality , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/metabolism , HIV Envelope Protein gp120/pharmacology , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Microfilament Proteins , Pain/chemically induced , Pain Measurement/methods , Peptides/administration & dosage , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Proline/administration & dosage , Proline/analogs & derivatives , Rats , Rats, Sprague-Dawley , Thiocarbamates/administration & dosage , Time Factors
9.
Brain Behav Immun ; 19(2): 127-37, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15664785

ABSTRACT

This study investigates the role of type I IL-1 receptor (IL-1R1) in mediating the recruitment of leukocytes into the brain parenchyma in mice. Intracerebroventricular (icv) injection of interleukin IL-1beta induced infiltration of leukocytes between 8 and 72 h after the injection. Leukocytes were rarely found in the brain tissue of saline-injected animals. At 8h after IL-1beta injection, leukocytes were seen lining the blood vessels of the brain and sparsely scattered infiltration of leukocytes was found in the cortex. Peak infiltration of leukocytes, which distributed evenly throughout the brain, was seen at 16 h post-injection. The number of leukocytes in the brain declined thereafter and no leukocytes were found 72 h post-injection. This phenomenon was replicated in mice deficient in lymphotoxin-alpha (LT(alpha)), IL-6, interferon (IFN)-gamma receptor, or the tumor necrosis factor (TNF)-alpha receptor, but abrogated in animals deficient in IL-1R1. ICV injection of IFN-gamma or TNF-alpha, but not IL-6 or IL-12, also induced leukocyte infiltration into the brain. Injection of IL-1beta, IFN-gamma, TNF-alpha, IL-6, and IL-12 induced IL-1beta expression in the brain, with IL-6 and IL-12 being the least effective. Leukocyte infiltration induced by icv IFN-gamma and TNF-alpha was also abrogated in IL-1R1-knockout animals. The induced infiltrating leukocytes were identified as neutrophils. Chronic infection with Trypanosoma brucei resulted in the recruitment of T cells, but no other cell types, into the brain. This did not occur in IL-1R1-knockout mice. Thus, IL-1R1 appears to be important for the recruitment of leukocytes across the blood-brain barrier.


Subject(s)
Brain/immunology , Cell Movement/immunology , Leukocytes/cytology , Receptors, Interleukin-1/immunology , Animals , Blood-Brain Barrier/immunology , Brain/blood supply , Brain/parasitology , Interferon-gamma/pharmacology , Interleukin-1/genetics , Interleukin-1/pharmacology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/pharmacology , Leukocytes/immunology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/immunology , Male , Mice , Mice, Knockout , Neuroimmunomodulation/immunology , RNA, Messenger/analysis , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/immunology , Trypanosoma brucei brucei , Trypanosomiasis, African/immunology , Tumor Necrosis Factor-alpha/pharmacology , Interferon gamma Receptor
10.
J Neuroimmunol ; 138(1-2): 92-8, 2003 May.
Article in English | MEDLINE | ID: mdl-12742658

ABSTRACT

Infection of Trypanosoma brucei causes specific patterns of neurodegeneration in association with chronic expression of proinflammatory cytokines in the brain. To investigate whether the induction of proinflammatory cytokines contributed to the observe pathology in this disease, we infected rats with T. brucei and treated them with intracerebral infusion of the cytokine antagonists interleukin-1 receptor antagonist (IL-1ra) and/or soluble type-I receptor of the tumor necrosis factor (sTNFr1). Infusion of IL-1ra, not sTNFr1, restored the reduction of body weight gain induced by the infection. Infusion of IL-1ra+sTNFr1 reduced the expression of IL-1beta and the cytokine response gene IkappaBalpha, but not TNFalpha. Infusion of sTNFr1 reduced trypanosome-induced neurodegeneration. Further reduction of neurodegeneration was seen after IL-1ra+sTNFr1 infusion. Infusion of IL-1ra alone, however, did not significantly affect the patterns of neurodegeneration. These results suggest that TNFalpha is a major mediator for trypanosome-induced neurodegeneration although its neurotoxic effects can be augmented by IL-1.


Subject(s)
Interleukin-1/antagonists & inhibitors , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/parasitology , Trypanosoma brucei brucei , Trypanosomiasis, African/immunology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Antigens, CD/administration & dosage , Body Weight/immunology , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Cytokines/genetics , Injections, Intraventricular , Interleukin 1 Receptor Antagonist Protein , Interleukin-1/biosynthesis , Interleukin-1/genetics , Male , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis , Rats , Rats, Sprague-Dawley , Receptors, Tumor Necrosis Factor/administration & dosage , Receptors, Tumor Necrosis Factor, Type I , Sialoglycoproteins/administration & dosage , Solubility , Trypanosomiasis, African/pathology , Trypanosomiasis, African/physiopathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
11.
J Neuroimmunol ; 137(1-2): 51-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12667647

ABSTRACT

Splenocytes from socially stressed male mice display functional glucocorticoid (GC) resistance, viz., the antiproliferative effects of GC on lipopolysaccharide (LPS)-stimulated splenocytes is absent. In this study, we investigated changes in the structure and function of the glucocorticoid receptor (GR) in socially stressed animals. Changes of GR at both DNA and RNA levels were excluded. Reduced GR function was restricted to macrophages (CD11b(+)) in association with impaired nuclear translocation of GR after GC stimulation. Consequently, GC failed to block the activation of NF-kappa B in these cells. Thus, impaired nuclear translocation of GR and the lack of transcriptional suppression of NF-kappa B by GC were identified as the molecular mechanisms responsible for the observed GC resistance in spleens of socially stressed mice.


Subject(s)
Corticosterone/pharmacology , Spleen/immunology , Stress, Psychological/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Drug Resistance/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , Stress, Psychological/metabolism , Stress, Psychological/psychology
12.
Brain Res Bull ; 59(6): 447-52, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12576141

ABSTRACT

Peripheral injection of bacterial endotoxin lipopolysaccharide (LPS) activates the paraventricular nuclei of the hypothalamus (PVN), and consequently the hypothalamus-pituitary adrenal axis. Inflammatory cytokine interleukin-1 (IL-1) has been considered as a key mediator that translates the peripheral LPS stimulation into neuronal activation in the PVN. Several studies attempting to localize the expression of receptors for IL-1 (IL-1R), however, have failed to detect IL-1R on PVN neurons. It remains unclear, therefore, how IL-1 might stimulate the neurons of the PVN. In this study, we traced the cellular responsiveness to IL-1 by measuring the mRNA production of the cytokine responsive gene IkappaBalpha in the PVN. After either peripheral injection LPS or intracerebroventricular (i.c.v.) injection of IL-1beta, IkappaBalpha mRNA was found mostly in endothelial cells of the brain with the highest level of expression in PVN blood vessels. In addition, both injections also induced the expression of cyclooxygenase-2 in brain endothelial cells. Pretreatment with indomethacin, a cyclooxygenase inhibitor, blocked LPS and IL-1 induced neuronal activation in the PVN, but did not reduce the induction of IkappaBalpha in PVN endothelium. These results show that IL-1 acting on the endothelial cells of the brain, particularly in the PVN, may be an intermediate step relating peripheral immune signals to the brain.


Subject(s)
Blood-Brain Barrier/immunology , Endothelium, Vascular/metabolism , Lipopolysaccharides/pharmacology , Paraventricular Hypothalamic Nucleus/blood supply , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Blood-Brain Barrier/drug effects , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Gene Expression/drug effects , I-kappa B Proteins/genetics , Indomethacin/pharmacology , Interleukin-1/pharmacology , Isoenzymes/analysis , Isoenzymes/genetics , Male , NF-KappaB Inhibitor alpha , Paraventricular Hypothalamic Nucleus/immunology , Prostaglandin-Endoperoxide Synthases/analysis , Prostaglandin-Endoperoxide Synthases/genetics , Proto-Oncogene Proteins c-fos/genetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL