701
|
Neuroinflammatory responses after experimental diffuse traumatic brain injury. J Neuropathol Exp Neurol 2007; 66:989-1001. [PMID: 17984681 DOI: 10.1097/nen.0b013e3181588245] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Little is known about microglial activation and macrophage localization after diffuse brain injury (DBI). DBI-mediated perisomatic traumatic axonal injury (TAI) was recently identified within the neocortex, hippocampus, and thalamus, providing an opportunity to characterize immune cell responses within diffusely injured brain loci uncomplicated by contusion. By using moderate midline/central fluid percussion injury, microglial/macrophage responses were examined with antibodies targeting immune cell phenotypes and amyloid precursor protein, a marker of TAI. Parallel assessments of blood-brain barrier alterations were also performed. Within 6 to 48 hours postinjury, microglial activation within injured loci was observed, whereas microglia within non-TAI-containing regions maintained a resting phenotype. Microglial activation shared a spatiotemporal relationship with TAI though no clear interactions were observed. By 7 to 28 days postinjury, activated microglia contained myelin debris, yet revealed limited aggregation. Immunophenotypic macrophages were also localized to injured loci. Select macrophages approximated somatic membranes of perisomatically axotomized neurons with evidence of bouton disruption. No causality was established between blood-brain barrier alterations and these inflammatory responses. These findings indicate rapid, yet initially nonspecific, and persistent microglial/macrophage responses to DBI. DBI-mediated inflammatory responses suggest further expansion of traumatic brain injury histopathologic evaluations to identify neuroinflammation indicative of diffuse pathology.
Collapse
|
702
|
Syed MM, Phulwani NK, Kielian T. Tumor necrosis factor-alpha (TNF-alpha) regulates Toll-like receptor 2 (TLR2) expression in microglia. J Neurochem 2007; 103:1461-71. [PMID: 17961202 DOI: 10.1111/j.1471-4159.2007.04838.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Microglia represent one effector arm of CNS innate immunity as evident by their role in pathogen recognition. We previously reported that exposure of microglia to Staphylococcus aureus (S. aureus), a prevalent CNS pathogen, led to elevated Toll-like receptor 2 (TLR2) expression, a pattern recognition receptor capable of recognizing conserved structural motifs associated with gram-positive bacteria such as S. aureus. In this study, we demonstrate that the proinflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) enhances TLR2 expression in microglia, whereas interleukin-1beta has no significant effect. To determine the downstream signaling events responsible for elevated microglial TLR2 expression in response to TNF-alpha, a series of signal transduction inhibitors were employed. Treatment with caffeic acid phenethyl ester, an inhibitor of redox-mediated nuclear factor-kappa B activation, significantly attenuated TNF-alpha-induced TLR2 expression. Similar results were observed with the IKK-2 and IkappaB-alpha inhibitors SC-514 and BAY 11-7082, respectively. In contrast, no significant alterations in TLR2 expression were observed with protein kinase C or p38 mitogen-activated protein kinase inhibitors. A definitive role for TNF-alpha was demonstrated by the inability of S. aureus to augment TLR2 expression in microglia isolated from TNF-alpha knockout mice. In addition, TLR2 expression was significantly attenuated in brain abscesses of TNF-alpha knockout mice. Collectively, these results indicate that in response to S. aureus, TNF-alpha acts in an autocrine/paracrine manner to enhance TLR2 expression in microglia and that this effect is mediated, in part, by activation of the nuclear factor-kappa B pathway.
Collapse
Affiliation(s)
- Mohsin Md Syed
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | | | | |
Collapse
|
703
|
Inhibition of toll-like receptor signaling in primary murine microglia. J Neuroimmune Pharmacol 2007; 3:5-11. [PMID: 18066668 DOI: 10.1007/s11481-007-9097-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2007] [Accepted: 11/01/2007] [Indexed: 01/17/2023]
Abstract
Microglial cells respond to the herpes simplex virus (HSV)-1 by producing proinflammatory cytokines and chemokines. After this inflammatory burst, these cells undergo apoptotic cell death. We have recently demonstrated that both virus-induced immune mediator production and apoptosis were induced through Toll-like receptor 2 (TLR2) signaling. Based upon these findings, we hypothesized that the inhibition of TLR2 signaling may serve as a means to alleviate excessive neuroinflammation. In the present study, we cloned four vaccinia virus (VV) proteins, which have been reported to disrupt either TLR signaling or NF-kappaB activation, and overexpressed them in HEK293T cells stably expressing murine TLR2 and in primary murine microglia. Using an NF-kappaB-driven luciferase reporter gene assay, we show that upon stimulation with HSV and Listeria monocytogenes, all four vaccinia proteins inhibited TLR2 signaling with different levels of inhibition in the TLR2-expressing cell line and primary microglia. We found similar results when microglial cells were stimulated with the TLR4 ligand LPS and the TLR9 ligand CpG ODN. Taken together, these data provide evidence that these VV proteins can function as inhibitors of TLR signaling in primary microglial cells.
Collapse
|
704
|
Lin YC, Uang HW, Lin RJ, Chen IJ, Lo YC. Neuroprotective effects of glyceryl nonivamide against microglia-like cells and 6-hydroxydopamine-induced neurotoxicity in SH-SY5Y human dopaminergic neuroblastoma cells. J Pharmacol Exp Ther 2007; 323:877-87. [PMID: 17855475 DOI: 10.1124/jpet.107.125955] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glyceryl nonivamide (GLNVA), a vanilloid receptor (VR) agonist, has been reported to have calcitonin gene-related peptide-associated vasodilatation and to prevent subarachnoid hemorrhage-induced cerebral vasospasm. In this study, we investigated the neuroprotective effects of GLNVA on activated microglia-like cell mediated- and proparkinsonian neurotoxin 6-hydroxydopamine (6-OHDA)-induced neurotoxicity in human dopaminergic neuroblastoma SH-SY5Y cells. In coculture conditions, we used lipopolysaccharide (LPS)-stimulated BV-2 cells as a model of activated microglia. LPS-induced neuronal death was significantly inhibited by diphenylene iodonium (DPI), an inhibitor of NADPH oxidase. However, capsazepine, the selective VR1 antagonist, did not block the neuroprotective effects of GLNVA. GLNVA reduced LPS-activated microglia-mediated neuronal death, but it lacked protection in DPI-pretreated cultures. GLNVA also decreased LPS activated microglia induced overexpression of neuronal nitric-oxide synthase (nNOS) and glycoprotein 91 phagocyte oxidase (gp91(phox)) on SH-SY5Y cells. Pretreatment of BV-2 cells with GLNVA diminished LPS-induced nitric oxide production, overexpression of inducible nitric-oxide synthase (iNOS), and gp91(phox) and intracellular reactive oxygen species (iROS). GLNVA also reduced cyclooxygenase (COX)-2 expression, inhibitor of nuclear factor (NF)-kappaB (IkappaB)alpha/IkappaBbeta degradation, NF-kappaB activation, and the overproduction of tumor necrosis factor-alpha, interleukin (IL)-1beta, and prostaglandin E2 in BV-2 cells. However, GLNVA augmented anti-inflammatory cytokine IL-10 production on LPS-stimulated BV-2 cells. Furthermore, in 6-OHDA-treated SH-SY5Y cells, GLNVA rescued the changes in condensed nuclear and apoptotic bodies, prevented the decrease in mitochondrial membrane potential, and reduced cells death. GLNVA also suppressed accumulation of iROS and up-regulated heme oxygenase-1 expression. 6-OHDA-induced overexpression of nNOS, iNOS, COX-2, and gp91(phox) was also reduced by GLNVA. In summary, the neuroprotective effects of GLNVA are mediated, at least in part, by decreasing the inflammation- and oxidative stress-associated factors induced by microglia and 6-OHDA.
Collapse
Affiliation(s)
- Yi-Chin Lin
- Department of Pharmacology and Graduate Institute of Pharmacology, College of Medicine, Kaohsiung Medical University, 100 Shih-Chuan 1st Rd., Kaohsiung 807, Taiwan
| | | | | | | | | |
Collapse
|
705
|
Aravalli RN, Peterson PK, Lokensgard JR. Toll-like receptors in defense and damage of the central nervous system. J Neuroimmune Pharmacol 2007; 2:297-312. [PMID: 18040848 DOI: 10.1007/s11481-007-9071-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 03/01/2007] [Indexed: 12/13/2022]
Abstract
Members of the Toll-like receptor (TLR) family play critical roles as regulators of innate and adaptive immune responses. TLRs function by recognizing diverse molecular patterns on the surface of invading pathogens. In the brain, microglial cells generate neuroimmune responses through production of proinflammatory mediators. The upregulation of cytokines and chemokines in response to microbial products and other stimuli has both beneficial and deleterious effects. Emerging evidence demonstrates a central role for TLRs expressed on microglia as a pivotal factor in generating these neuroimmune responses. Therefore, understanding the basis of TLR signaling in producing these responses may provide insights into how activated microglia attempt to strike a balance between defense against invading pathogens and inflicting irreparable brain damage. These insights may lead to innovative therapies for CNS infections and neuroinflammatory diseases based on the modulation of microglial cell activation through TLR signaling.
Collapse
Affiliation(s)
- Rajagopal N Aravalli
- Neuroimmunology Laboratory, Center for Infectious Diseases and Microbiology Translational Research, University of Minnesota Medical School, 2001 6th Street SE, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
706
|
Orsini H, Bondan EF, Sanchez M, Lallo MA, Maiorka PC, Dagli MLZ, Graça DL. Marcação imunoistoquímica da expressão astrocitária de proteína glial fibrilar ácida e de vimentina no sistema nervoso central de cães com cinomose. ARQUIVOS DE NEURO-PSIQUIATRIA 2007; 65:1070-7. [DOI: 10.1590/s0004-282x2007000600032] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 07/30/2007] [Indexed: 11/21/2022]
Abstract
Uma vez que muitos dos aspectos envolvidos na patogenia dos processos desmielinizantes do sistema nervoso central (SNC) são ainda pouco esclarecidos e que os astrócitos parecem estar envolvidos na mediação de tais processos, este estudo analisou morfologicamente a participação astrocitária na desmielinização do SNC por meio da marcação imunoistoquímica de duas proteínas dos filamentos intermediários astrocitários - a proteína glial fibrilar ácida (GFAP) e a vimentina (VIM) -, comparando amostras de cerebelo e de tronco encefálico de oito cães com cinomose e de dois cães normais, de diferentes raças e com idades entre um e quatro anos. Cortes histológicos dos tecidos foram submetidos à marcação pelo método indireto da avidina-biotina-peroxidase (ABC) e a reatividade astrocitária, observada em microscopia de luz, foi quantificada em um sistema computacional de análise de imagens. Observou-se, na maioria dos cortes de animais doentes, a presença de lesões degenerativas compatíveis com desmielinização. A marcação para a GFAP e para a VIM foi mais intensa nos animais com cinomose do que nos animais normais, especialmente nas regiões circunventriculares e nas adjacentes às áreas de degeneração tecidual. Não houve diferença significativa entre a imunomarcação (GFAP e VIM) dos animais com cinomose com e sem infiltração inflamatória da substância branca do cerebelo. O aumento da imunorreatividade dos astrócitos para a GFAP e a reexpressão de VIM nas áreas lesionais indicam o envolvimento astrocitário na resposta do tecido nervoso às lesões desmielinizantes induzidas pelo vírus da cinomose (CDV) no SNC.
Collapse
Affiliation(s)
- Heloísa Orsini
- Universidade Paulista; Universidade Cruzeiro do Sul, Brasil
| | | | | | | | | | | | | |
Collapse
|
707
|
Lee S, Lee J, Kim S, Park JY, Lee WH, Mori K, Kim SH, Kim IK, Suk K. A dual role of lipocalin 2 in the apoptosis and deramification of activated microglia. THE JOURNAL OF IMMUNOLOGY 2007; 179:3231-41. [PMID: 17709539 DOI: 10.4049/jimmunol.179.5.3231] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Activated microglia are thought to undergo apoptosis as a self-regulatory mechanism. To better understand molecular mechanisms of the microglial apoptosis, apoptosis-resistant variants of microglial cells were selected and characterized. The expression of lipocalin 2 (lcn2) was significantly down-regulated in the microglial cells that were resistant to NO-induced apoptosis. lcn2 expression was increased by inflammatory stimuli in microglia. The stable expression of lcn2 as well as the addition of rLCN2 protein augmented the sensitivity of microglia to the NO-induced apoptosis, while knockdown of lcn2 expression using short hairpin RNA attenuated the cell death. Microglial cells with increased lcn2 expression were more sensitive to other cytotoxic agents as well. Thus, inflammatory activation of microglia may lead to up-regulation of lcn2 expression, which sensitizes microglia to the self-regulatory apoptosis. Additionally, the stable expression of lcn2 in BV-2 microglia cells induced a morphological change of the cells into the round shape with a loss of processes. Treatment of primary microglia cultures with the rLCN2 protein also induced the deramification of microglia. The deramification of microglia was closely related with the apoptosis-prone phenotype, because other deramification-inducing agents such as cAMP-elevating agent forskolin, ATP, and calcium ionophore also rendered microglia more sensitive to cell death. Taken together, our results suggest that activated microglia may secrete LCN2 protein, which act in an autocrine manner to sensitize microglia to the self-regulatory apoptosis and to endow microglia with an amoeboid form, a canonical morphology of activated microglia in vivo.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Kyungpook National University School of Medicine, 101 Dong-in, Joong-gu, Daegu 700-422, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
708
|
Jinno S, Fleischer F, Eckel S, Schmidt V, Kosaka T. Spatial arrangement of microglia in the mouse hippocampus: a stereological study in comparison with astrocytes. Glia 2007; 55:1334-47. [PMID: 17647290 DOI: 10.1002/glia.20552] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Microglia are classically considered to be immune cells in the brain, but have now been proven to be involved in neuronal activity as well. Here we stereologically analyzed the spatial arrangement of microglia in the mouse hippocampus. First, we estimated the numerical densities (NDs) of microglia identified by ionized calcium-binding adaptor molecule 1 (Iba1). Despite that microglia appeared to be evenly distributed throughout the hippocampal area, the NDs demonstrated significant dorsoventral, interregional, and interlaminar differences. Briefly, the NDs in the ventral hippocampus were significantly lower in the CA3 region than in the CA1 region and dentate gyrus, although no interregional differences were detectable in the dorsal hippocampus. Both in the CA1 and CA3 regions, the NDs were significantly higher in the stratum lacunosum-moleculare than in the remaining layers. Next, we investigated the spatial patterns of distribution of Iba1-labeled microglia and S100beta-labeled astrocytes. So far as we examined, the somato-somatic contacts were not seen among microglia or among astrocytes, whereas the close apposition between microglia and astrocytes were occasionally detected. The 3D point process analysis showed that the spatial distribution of microglia was significantly repulsive. Because the statistical territory of single microglia was larger than that estimated from process tracing, they are not likely to touch each other with their processes. Astrocytes were distributed slightly repulsively with overlapping areas. The 3D point process analysis also revealed a significant spatial attraction between microglia and astrocytes. The present findings provide a novel anatomical basis for glial research.
Collapse
Affiliation(s)
- Shozo Jinno
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Kyushu University, Higashi-ku, Fukuoka, Japan.
| | | | | | | | | |
Collapse
|
709
|
Tansey MG, McCoy MK, Frank-Cannon TC. Neuroinflammatory mechanisms in Parkinson's disease: potential environmental triggers, pathways, and targets for early therapeutic intervention. Exp Neurol 2007; 208:1-25. [PMID: 17720159 PMCID: PMC3707134 DOI: 10.1016/j.expneurol.2007.07.004] [Citation(s) in RCA: 437] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 07/02/2007] [Accepted: 07/05/2007] [Indexed: 12/11/2022]
Abstract
Most acute and chronic neurodegenerative conditions are accompanied by neuroinflammation; yet the exact nature of the inflammatory processes and whether they modify disease progression is not well understood. In this review, we discuss the key epidemiological, clinical, and experimental evidence implicating inflammatory processes in the progressive degeneration of the dopaminergic (DA) nigrostriatal pathway and their potential contribution to the pathophysiology of Parkinson's disease (PD). Given that interplay between genetics and environment are likely to contribute to risk for development of idiopathic PD, recent data showing interactions between products of genes linked to heritable PD that function to protect DA neurons against oxidative or proteolytic stress and inflammation pathways will be discussed. Cellular mechanisms activated or enhanced by inflammatory processes that may contribute to mitochondrial dysfunction, oxidative stress, or apoptosis of dopaminergic (DA) neurons will be reviewed, with special emphasis on tumor necrosis factor (TNF) and interleukin-1-beta (IL-1beta) signaling pathways. Epigenetic factors which have the potential to trigger neuroinflammation, including environmental exposures and age-associated chronic inflammatory conditions, will be discussed as possible 'second-hit' triggers that may affect disease onset or progression of idiopathic PD. If inflammatory processes have an active role in nigrostriatal pathway degeneration, then evidence should exist to indicate that such processes begin in the early stages of disease and that they contribute to neuronal dysfunction and/or hasten neurodegeneration of the nigrostriatal pathway. Therapeutically, if anti-inflammatory interventions can be shown to rescue nigral DA neurons from degeneration and lower PD risk, then timely use of anti-inflammatory therapies should be investigated further in well-designed clinical trials for their ability to prevent or delay the progressive loss of nigral DA neurons in genetically susceptible populations.
Collapse
Affiliation(s)
- Malú G Tansey
- Department of Physiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.
| | | | | |
Collapse
|
710
|
Cardona AE, Huang D, Sasse ME, Ransohoff RM. Isolation of murine microglial cells for RNA analysis or flow cytometry. Nat Protoc 2007; 1:1947-51. [PMID: 17487181 DOI: 10.1038/nprot.2006.327] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is increasing interest in the isolation of adult microglia to study their functions at a morphological and molecular level during normal and neuroinflammatory conditions. Microglia have important roles in brain homeostasis, and in disease states they exert neuroprotective or neurodegenerative functions. To assay expression profiles or functions of microglia, we have developed a method to isolate microglial cells and infiltrating leukocytes from adult mouse brain. This protocol uses a digestion cocktail containing collagenase and dispase, and it involves separation over discontinuous percoll gradients. Isolated cells can be used for RNA analysis, including RNase protection analysis (RPA), quantitative RT-PCR, high-density microarray, proteomic or flow cytometric characterization of cell surface markers or adoptive transfer. Cell isolation can be completed in less than 4 h.
Collapse
Affiliation(s)
- Astrid E Cardona
- Neuroinflammation Research Center, Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | | | |
Collapse
|
711
|
Lee KH, Yun SJ, Nam KN, Gho YS, Lee EH. Activation of microglial cells by ceruloplasmin. Brain Res 2007; 1171:1-8. [PMID: 17727827 DOI: 10.1016/j.brainres.2007.07.053] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2007] [Revised: 07/26/2007] [Accepted: 07/31/2007] [Indexed: 12/20/2022]
Abstract
Ceruloplasmin (Cp) is the major copper transport protein in plasma and catalyzes the conversion of toxic ferrous iron to the safer ferric iron. As an acute-phase protein, Cp is induced during inflammation. It is synthesized primarily in the liver and is expressed in several other tissues, including the brain. Elevated Cp levels have been observed in the brain of patients with neurodegenerative conditions, including Alzheimer's, Parkinson's, and Huntington's diseases. However, the exact role(s) of Cp in inflammatory and neuropathological conditions remains unclear. Microglia are the prime effector cells involved in immune and inflammatory responses in the central nervous system (CNS). They are activated during pathological conditions to restore CNS homeostasis, but chronic microglial activation endangers neuronal survival. Consequently, it is important to identify the regulators of microglial activation and the underlying mechanisms. We sought to examine whether Cp might modulate microglial activation. We observed that Cp induced nitric oxide (NO) release and inducible NO synthase mRNA expression in BV2 microglial cells and rat brain microglia. Cp also increased levels of mRNAs encoding tumor necrosis factor-alpha, interleukin-1beta, cyclooxygenase-2, and NADPH oxidase. Treatment of BV2 cells and primary microglia with Cp induced phosphorylation of p38 MAP kinase. Moreover, Cp induced nuclear factor (NF)-kappaB activation, showing a more sustained pattern than seen with bacterial lipopolysaccharide. Cp-stimulated NO induction was significantly attenuated by a p38 inhibitor, SB203580, and the NF-kappaB inhibitor SN50. Cp induced secretion of TNF-alpha and prostaglandin E(2) in primary microglial cultures. These results suggest that Cp may play an important role in neuropathological conditions by stimulating various proinflammatory and neurotoxic molecules in microglia.
Collapse
Affiliation(s)
- Kyung-Hee Lee
- Department of Medical Science, Graduate School of East-West Medical Science, Kyung Hee University, Yongin-Si, Republic of Korea
| | | | | | | | | |
Collapse
|
712
|
Kielian T, Esen N, Liu S, Phulwani NK, Syed MM, Phillips N, Nishina K, Cheung AL, Schwartzman JD, Ruhe JJ. Minocycline modulates neuroinflammation independently of its antimicrobial activity in staphylococcus aureus-induced brain abscess. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:1199-214. [PMID: 17717149 PMCID: PMC1988870 DOI: 10.2353/ajpath.2007.070231] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Minocycline exerts beneficial immune modulatory effects in several noninfectious neurodegenerative disease models; however, its potential to influence the host immune response during central nervous system bacterial infections, such as brain abscess, has not yet been investigated. Using a minocycline-resistant strain of Staphylococcus aureus to dissect the antibiotic's bacteriostatic versus immune modulatory effects in a mouse experimental brain abscess model, we found that minocycline significantly reduced mortality rates within the first 24 hours following bacterial exposure. This protection was associated with a transient decrease in the expression of several proinflammatory mediators, including interleukin-1beta and CCL2 (MCP-1). Minocycline was also capable of protecting the brain parenchyma from necrotic damage as evident by significantly smaller abscesses in minocycline-treated mice. In addition, minocycline exerted anti-inflammatory effects when administered as late as 3 days following S. aureus infection, which correlated with a significant decrease in brain abscess size. Finally, minocycline was capable of partially attenuating S. aureus-dependent microglial and astrocyte activation. Therefore, minocycline may afford additional therapeutic benefits extending beyond its antimicrobial activity for the treatment of central nervous system infectious diseases typified by a pathogenic inflammatory component through its ability to balance beneficial versus detrimental inflammation.
Collapse
Affiliation(s)
- Tammy Kielian
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, 4301 W. Markham St., Slot 846, Little Rock, AR 72205, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
713
|
Bassotti G, Villanacci V, Fisogni S, Rossi E, Baronio P, Clerici C, Maurer CA, Cathomas G, Antonelli E. Enteric glial cells and their role in gastrointestinal motor abnormalities: introducing the neuro-gliopathies. World J Gastroenterol 2007; 13:4035-4041. [PMID: 17696219 PMCID: PMC4205302 DOI: 10.3748/wjg.v13.i30.4035] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2007] [Revised: 05/03/2007] [Accepted: 05/12/2007] [Indexed: 02/06/2023] Open
Abstract
The role of enteric glial cells has somewhat changed from that of mere mechanical support elements, gluing together the various components of the enteric nervous system, to that of active participants in the complex interrelationships of the gut motor and inflammatory events. Due to their multiple functions, spanning from supporting elements in the myenteric plexuses to neurotransmitters, to neuronal homeostasis, to antigen presenting cells, this cell population has probably more intriguing abilities than previously thought. Recently, some evidence has been accumulating that shows how these cells may be involved in the pathophysiological aspects of some diseases. This review will deal with the properties of the enteric glial cells more strictly related to gastrointestinal motor function and the human pathological conditions in which these cells may play a role, suggesting the possibility of enteric neuro-gliopathies.
Collapse
|
714
|
Nair A, Hunzeker J, Bonneau RH. Modulation of microglia and CD8(+) T cell activation during the development of stress-induced herpes simplex virus type-1 encephalitis. Brain Behav Immun 2007; 21:791-806. [PMID: 17349776 DOI: 10.1016/j.bbi.2007.01.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2006] [Revised: 12/20/2006] [Accepted: 01/08/2007] [Indexed: 01/07/2023] Open
Abstract
The central nervous system (CNS) has been shown to be vulnerable to a variety of insults in animals exposed to glucocorticoids. For example, psychological stress, a known inducer of glucocorticoid production, enhances the susceptibility of mice to herpes simplex virus type-1 (HSV-1) infection and results in the development of HSV-1 encephalitis (HSE). To determine the immune mechanisms by which stress promotes the development of HSE, we examined the role of the glucocorticoid receptor (GR) and the N-methyl-d-aspartate (NMDA) receptor in the development of HSE. Our findings demonstrate that blockade of either the GR or the NMDA receptor enhances survival following HSV-1 infection in stressed mice to levels similar to non-stressed mice. Subsequent studies determined the effect of GR and NMDA receptor blockade on immune function by specifically examining both microglia and CD8(+) T cell activation. Stress inhibited the expression of MHC class I by microglia and other brain-derived antigen presenting cells (CD45(hi)) independent of either the glucocorticoid receptor or the NMDA receptor, suggesting that stress-induced suppression of MHC class I expression in the brain does not affect survival during HSE. Blockade of the NMDA receptor, however, diminished HSV-1-induced increases in class I expression by CD45(hi) cells, suggesting that blockade of the NMDA receptor may limit CNS inflammation. Also, while CD8(+) T cell activation and function in the brain were not affected by stress, the number of CD8(+) T cells in the superficial cervical lymph nodes (SCLN) was decreased in stressed mice via GR-mediated mechanisms. These findings indicate that stress-induced hypocellularity is mediated by the GR while NMDA receptor activation is responsible for enhancing CNS inflammation. The combined effects of GR-mediated hypocellularity of the SCLN and NMDA receptor-mediated CNS inflammation during stress promote the development of HSE.
Collapse
MESH Headings
- Animals
- Brain/cytology
- Brain/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Cytokines/metabolism
- Disease Models, Animal
- Encephalitis, Herpes Simplex/immunology
- Encephalitis, Herpes Simplex/psychology
- Encephalitis, Herpes Simplex/virology
- Herpesvirus 1, Human/immunology
- Major Histocompatibility Complex/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Microglia/immunology
- Microglia/metabolism
- Microglia/virology
- Receptors, Glucocorticoid/antagonists & inhibitors
- Receptors, Glucocorticoid/physiology
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Receptors, N-Methyl-D-Aspartate/physiology
- Stress, Psychological/complications
- Stress, Psychological/immunology
- Stress, Psychological/virology
Collapse
Affiliation(s)
- Aji Nair
- Graduate Program in Neuroscience, The Pennsylvania State University College of Medicine, Milton S Hershey Medical Center, Hershey, PA 17033, USA
| | | | | |
Collapse
|
715
|
de Haas AH, Boddeke HWGM, Brouwer N, Biber K. Optimized isolation enables ex vivo analysis of microglia from various central nervous system regions. Glia 2007; 55:1374-84. [PMID: 17661344 DOI: 10.1002/glia.20554] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ex vivo analysis is an accurate and convenient way to study in vivo microglia phenotype and function. However, current microglia isolation protocols for ex vivo analysis show many differences in isolation steps (perfusion, removal of meninges and blood vessels, mechanical dissociation, enzymatic dissociation, density separation, immunomagnetic separation, and fluorescence-activated cell sorting), often without addressing their effects on microglia purity, number, phenotype, and function. Therefore, the aim of this study was to provide an optimized isolation protocol with emphasis on microglia purity and number to enable ex vivo analysis of adult mouse microglia. The application of this protocol for ex vivo phenotype and functional analysis is corroborated by results from flow cytometry, gene expression analysis, chemotaxis, and phagocytosis assays. In addition, this study shows the possibility to analyze microglia isolated from various central nervous system regions such as optic nerve, striatum, hippocampus, spinal cord, cerebellum, and cerebral cortex. Furthermore, this is the first study presenting DRAQ5 as a superior alternative to propidium iodide for the discrimination between living and dead cells. DRAQ5 staining facilitated the identification of microglia upon flow cytometry without the need of additional fluorescent markers. Along with a favorable emission spectrum, DRAQ5 proved a valuable tool for flow cytometry of microglia. The presented optimized microglia isolation protocol for ex vivo analysis offers the opportunity to obtain more insight into both general and region-specific microglia behavior.
Collapse
Affiliation(s)
- Alexander H de Haas
- Department of Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
716
|
Gemma C, Bickford PC. Interleukin-1beta and caspase-1: players in the regulation of age-related cognitive dysfunction. Rev Neurosci 2007; 18:137-48. [PMID: 17593876 DOI: 10.1515/revneuro.2007.18.2.137] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Scientific research on the unprecedented and growing number of older adults in the United States and other industrialized countries has focused much attention on the health consequences of aging. Over the last few decades, inflammation in the brain and its implication in the progression of aging and age-related cognitive dysfunction has been an area of increasing importance to neuroscientists and is now considered as one of the most interesting and promising topics for aging research. One of the critical aspects of inflammatory processes is that the activation of one upstream inflammatory molecule initiates a cascade of self-sustaining inflammatory events which leads to the activation of a number of different downstream functions. Recently, a great deal of attention has been given to the interplay between inflammatory and apoptotic processes and the regulation of these processes by the caspases. The caspase family of proteases can be divided into proapoptotic and pro-inflammatory members. The present review summarizes recent observations of the interactions between the inflammatory cytokine interleuldn-1 (IL-1) beta and the inflammatory/apoptotic caspase-1 and their involvement in age-related impairments in cognition. A comprehensive understanding of these mechanisms could potentially lead to the development of preventive or protective therapies that reduce or inhibit the cognitive decline associated with aging and age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Carmelina Gemma
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery, University of South Florida College of Medicine, Tampa, FL 33612, USA
| | | |
Collapse
|
717
|
Lee TS, Mane S, Eid T, Zhao H, Lin A, Guan Z, Kim JH, Schweitzer J, King-Stevens D, Weber P, Spencer SS, Spencer DD, de Lanerolle NC. Gene expression in temporal lobe epilepsy is consistent with increased release of glutamate by astrocytes. Mol Med 2007; 13:1-13. [PMID: 17515952 PMCID: PMC1869627 DOI: 10.2119/2006-00079.lee] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 12/07/2006] [Indexed: 11/06/2022] Open
Abstract
Patients with temporal lobe epilepsy (TLE) often have a shrunken hippocampus that is known to be the location in which seizures originate. The role of the sclerotic hippocampus in the causation and maintenance of seizures in temporal lobe epilepsy (TLE) has remained incompletely understood despite extensive neuropathological investigations of this substrate. To gain new insights and develop new testable hypotheses on the role of sclerosis in the pathophysiology of TLE, the differential gene expression profile was studied. To this end, DNA microarray analysis was used to compare gene expression profiles in sclerotic and non-sclerotic hippocampi surgically removed from TLE patients. Sclerotic hippocampi had transcriptional signatures that were different from non-sclerotic hippocampi. The differentially expressed gene set in sclerotic hippocampi revealed changes in several molecular signaling pathways, which included the increased expression of genes associated with astrocyte structure (glial fibrillary acidic protein, ezrin-moesin-radixin, palladin), calcium regulation (S100 calcium binding protein beta, chemokine (C-X-C motif) receptor 4) and blood-brain barrier function (Aquaaporin 4, Chemokine (C-C- motif) ligand 2, Chemokine (C-C- motif) ligand 3, Plectin 1, intermediate filament binding protein 55kDa) and inflammatory responses. Immunohistochemical localization studies show that there is altered distribution of the gene-associated proteins in astrocytes from sclerotic foci compared with non-sclerotic foci. It is hypothesized that the astrocytes in sclerotic tissue have activated molecular pathways that could lead to enhanced release of glutamate by these cells. Such glutamate release may excite surrounding neurons and elicit seizure activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhong Guan
- Epidemiology and Public Health
- Department of Mathematical Sciences, Indiana University, South Bend, Indiana
| | | | - Jeffrey Schweitzer
- Department of Neurosurgery, Kaiser Permanente Medical Center, Los Angeles, California
| | | | | | | | | | - Nihal C de Lanerolle
- Neurosurgery
- Address correspondence and reprint requests to Nihal C. de Lanerolle, Department of Neurosurgery FMB414, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520-8082. Phone: 203-785-3258; Fax: 203-737-2159; E-mail:
| |
Collapse
|
718
|
Svoboda N, Zierler S, Kerschbaum HH. cAMP mediates ammonia-induced programmed cell death in the microglial cell line BV-2. Eur J Neurosci 2007; 25:2285-95. [PMID: 17445227 DOI: 10.1111/j.1460-9568.2007.05452.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Although ammonia is a well-known neuropathogenic factor, the cellular mechanisms of ammonia toxicity are less characterized. Up to now, the main focus of ammonia toxicity has been on astrocytes and neurons. Despite the significance of microglia in neurodegenerative diseases, little is known about their responsiveness to ammonia. In the present study, we found that ammonia triggered mitosis at concentrations between 30 microm and 3.0 mm but apoptosis at concentrations >or= 1.0 mm in the murine microglial cell line BV-2. Most apoptotic cells showed an accumulation of condensed chromatin at the nuclear envelope, blebbing of the plasma membrane, formation of apoptotic bodies and an increase in caspase 3/7 activity. Blockade of caspase 3/7 activity by Ac-DEVD-CHO suppressed ammonia-induced apoptosis. Surprisingly, some BV-2 cells exposed to ammonia displayed clear signs of mitotic catastrophe, a type of cell death occurring during mitosis. In a further series of experiments, we found that cyclic adenosine 3',5'-monophosphate (cAMP) mediated the apoptogenic effects of ammonia, because (i) ammonia dose-dependently elevated the intracellular cAMP level, (ii) blockade of the adenylyl cyclase by SQ-22536 suppressed ammonia-induced apoptosis, (iii) inhibition of phosphodiesterases (PDEs) by the nonselective PDE inhibitor IBMX, or by the PDE4-selective inhibitor rolipram, increased the relative number of apoptotic cells, and (iv) the cAMP analogues 8-bromoadenosine cAMP and Sp-cAMP mimicked the effect of ammonia and triggered apoptosis. Taken together, our results indicate that distinct concentrations of ammonia trigger opposite signalling pathways in microglial cells.
Collapse
Affiliation(s)
- Nina Svoboda
- Department of Cell Biology, University of Salzburg, Hellbrunnerstr. 34, 5020 Salzburg, Austria
| | | | | |
Collapse
|
719
|
Piccio L, Buonsanti C, Mariani M, Cella M, Gilfillan S, Cross AH, Colonna M, Panina-Bordignon P. Blockade of TREM-2 exacerbates experimental autoimmune encephalomyelitis. Eur J Immunol 2007; 37:1290-301. [PMID: 17407101 DOI: 10.1002/eji.200636837] [Citation(s) in RCA: 204] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Triggering receptor expressed on myeloid cells (TREM-2) is a membrane receptor associated with DAP12 that is expressed primarily in myeloid cells, including dendritic cells and microglia, and promotes fusion of osteoclast precursors into multinucleated cells. A rare autosomal recessive condition, Nasu-Hakola disease (NHD) is associated with loss-of-function mutations in DAP12 and TREM-2. The brain pathology observed in NHD patients suggests that disruption of the TREM-2/DAP12 pathway leads to neurodegeneration with demyelination and axonal loss. In this study, we have characterized TREM-2 protein expression on microglia using a newly produced monoclonal antibody directed against the mouse TREM-2 receptor. We report that TREM-2 expression is up-regulated in the spinal cord during both the early inflammatory and chronic phases of myelin oligodendrocyte glycoprotein (MOG)(35-55)peptide-induced experimental autoimmune encaphalomyelitis (EAE). We also demonstrate that TREM-2 is highly expressed on microglial cells in the central nervous system (CNS) during EAE and that blockade of TREM-2 during the effector phase of EAE results in disease exacerbation with more diffuse CNS inflammatory infiltrates and demyelination in the brain parenchyma. These results demonstrate a critical role for TREM-2 during inflammatory responses in the CNS.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Brain/immunology
- Brain/metabolism
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Fluorescent Antibody Technique
- Macrophages/immunology
- Macrophages/metabolism
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice
- Microglia/metabolism
- RNA, Messenger/analysis
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Spinal Cord/immunology
- Spinal Cord/metabolism
- Spinal Cord/pathology
- Up-Regulation
Collapse
|
720
|
Butovsky O, Bukshpan S, Kunis G, Jung S, Schwartz M. Microglia can be induced by IFN-γ or IL-4 to express neural or dendritic-like markers. Mol Cell Neurosci 2007; 35:490-500. [PMID: 17560122 DOI: 10.1016/j.mcn.2007.04.009] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2006] [Revised: 04/18/2007] [Accepted: 04/24/2007] [Indexed: 01/30/2023] Open
Abstract
Microglia are resident cells in the central nervous system (CNS), of hematopoietic origin with a high plasticity. In this study, we examined whether adaptive immune system, involving in CNS maintenance and repair, can induce microglia to express markers of neural cells. We show that long exposure (above 10 days) of microglia to low doses (10 ng/ml) of the 'proinflammatory' T-cell derived cytokine, IFN-gamma, induced them to express neuronal markers including gamma-aminobutyric acid (GABA) and glutamic acid decarboxylase (GAD-67). In contrast, exposure of microglia to low doses (10 ng/ml) of the 'anti-inflammatory' T-cell derived cytokine, IL-4, induced the expression of oligodendrocyte markers and dendritic cell (DC) marker, CD11c. The microglial origin of the neural-like cells was confirmed using microglia from transgenic mice expressing GFP under promoter of the chemokine fractalkine receptor CX(3)CR1, and diphtheria toxin receptor, under CD11c promoter. This study emphasizes that microglial plasticity includes their ability to give rise to neural-like cells and shows that cytokines produced by the adaptive immune system are involved in these processes.
Collapse
Affiliation(s)
- Oleg Butovsky
- Department of Neurobiology, The Weizmann Institute of Science, 76100 Rehovot, Israel
| | | | | | | | | |
Collapse
|
721
|
Wirenfeldt M, Dissing-Olesen L, Anne Babcock A, Nielsen M, Meldgaard M, Zimmer J, Azcoitia I, Leslie RGQ, Dagnaes-Hansen F, Finsen B. Population control of resident and immigrant microglia by mitosis and apoptosis. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:617-31. [PMID: 17600121 PMCID: PMC1934543 DOI: 10.2353/ajpath.2007.061044] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microglial population expansion occurs in response to neural damage via processes that involve mitosis and immigration of bone marrow-derived cells. However, little is known of the mechanisms that regulate clearance of reactive microglia, when microgliosis diminishes days to weeks later. We have investigated the mechanisms of microglial population control in a well-defined model of reactive microgliosis in the mouse dentate gyrus after perforant pathway axonal lesion. Unbiased stereological methods and flow cytometry demonstrate significant lesion-induced increases in microglial numbers. Reactive microglia often occurred in clusters, some having recently incorporated bromodeoxyuridine, showing that proliferation had occurred. Annexin V labeling and staining for activated caspase-3 and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling showed that apoptotic mechanisms participate in dissolution of the microglial response. Using bone marrow chimeric mice, we found that the lesion-induced proliferative capacity of resident microglia superseded that of immigrant microglia, whereas lesion-induced kinetics of apoptosis were comparable. Microglial numbers and responses were severely reduced in bone marrow chimeric mice. These results broaden our understanding of the microglial response to neural damage by demonstrating that simultaneously occurring mitosis and apoptosis regulate expansion and reduction of both resident and immigrant microglial cell populations.
Collapse
Affiliation(s)
- Martin Wirenfeldt
- Medical Biotechnology Center, Institute of Medical Biology, University of Southern Denmark, Odense, Denmark.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
722
|
Silva AR, Pinheiro AM, Souza CS, Freitas SRVB, Vasconcellos V, Freire SM, Velozo ES, Tardy M, El-Bachá RS, Costa MFD, Costa SL. The flavonoid rutin induces astrocyte and microglia activation and regulates TNF-alpha and NO release in primary glial cell cultures. Cell Biol Toxicol 2007; 24:75-86. [PMID: 17549591 DOI: 10.1007/s10565-007-9017-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2006] [Accepted: 04/18/2007] [Indexed: 10/23/2022]
Abstract
Astrocyte and microglia cells play an important role in the central nervous system (CNS). They react to various external aggressions by becoming reactive and releasing neurotrophic and/or neurotoxic factors. Rutin is a flavonoid found in many plants and has been shown to have some biological activities, but its direct effects on cells of the CNS have not been well studied. To investigate its potential effects on CNS glial cells, we used both astrocyte primary cultures and astrocyte/microglia mixed primary cell cultures derived from newborn rat cortical brain. The cultures were treated for 24 h with rutin (50 or 100 micromol/L) or vehicle (0.5% dimethyl sulfoxide). Mitochondrial function on glial cells was not evidenced by the MTT test. However, an increased lactate dehydrogenase activity was detected in the culture medium of both culture systems when treated with 100 micromol/L rutin, suggesting loss of cell membrane integrity. Astrocytes exposed to 50 micromol/L rutin became reactive as revealed by glial fibrillary acidic protein (GFAP) overexpression and showed a star-like phenotype revealed by Rosenfeld's staining. The number of activated microglia expressing OX-42 increased in the presence of rutin. A significant increase of nitric oxide (NO) was observed only in mixed cultures exposed to 100 micromol/L rutin. Enhanced TNFalpha release was observed in astrocyte primary cultures treated with 100 micromol/L rutin and in mixed primary cultures treated with 50 and 100 micromol/L, suggesting different sensitivity of both activated cell types. These results demonstrated that rutin affects astrocytes and microglial cells in culture and has the capacity to induce NO and TNFalpha production in these cells. Hence, the impact of these effects on neurons in vitro and in vivo needs to be studied.
Collapse
Affiliation(s)
- A R Silva
- Departamento de Biofunção, Laboratório de Neuroquímica e Biologia Celular, Instituto de Ciências da Saúde, Universidade Federal da Bahia, Salvador, Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
723
|
Bédard A, Tremblay P, Chernomoretz A, Vallières L. Identification of genes preferentially expressed by microglia and upregulated during cuprizone-induced inflammation. Glia 2007; 55:777-89. [PMID: 17285589 DOI: 10.1002/glia.20477] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Microglia, monocytes, and peripheral macrophages share a common origin and many characteristics, but what distinguishes them from each other at the level of gene expression remains largely unknown. In this study, we compared the transcriptional profiles of freshly purified microglia, monocytes, and spleen macrophages using Affymetrix Mouse Genome arrays to identify genes predominantly expressed by microglia. Among tens of thousands of genes assayed, 127 potential candidates were found, including nine newly discovered genes encoding plasma membrane and extracellular proteins. In the brain, the latter were selectively expressed by microglia, as revealed by in situ hybridization. Three of them were confirmed to be exclusively (MSR2) or predominantly (GPR12, GPR34) expressed in the brain compared to the other tissues examined. Furthermore, all of these genes were upregulated in activated microglia after treatment with the demyelinating toxin cuprizone, suggesting that they play roles in neuroinflammation. In conclusion, this study reports the identification of new selective markers for microglia, which should prove useful not only to identify and isolate these cells, but also to better understand their distinctive properties.
Collapse
Affiliation(s)
- Andréanne Bédard
- Department of Oncology and Molecular Endocrinology, Laval University Hospital Research Center, Québec City, Québec G1V 4G2, Canada
| | | | | | | |
Collapse
|
724
|
Nikodemova M, Watters JJ, Jackson SJ, Yang SK, Duncan ID. Minocycline Down-regulates MHC II Expression in Microglia and Macrophages through Inhibition of IRF-1 and Protein Kinase C (PKC)α/βII. J Biol Chem 2007; 282:15208-16. [PMID: 17395590 DOI: 10.1074/jbc.m611907200] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Experimental allergic encephalomyelitis, an autoimmune disorder mediated by T cells, results in demyelination, inflammation, and axonal loss in the central nervous system (CNS). Microglia play a critical role in major histocompatibility complex class II (MHC II)-dependent antigen presentation and in reactivation of CNS-infiltrated encephalitogenic T cells. Minocycline, a tetracycline anti-biotic, has profound anti-inflammatory properties and is experimentally used for treatment of many CNS disorders; however, the mechanisms involved in minocycline effects remain unknown. We show that administration of minocycline for 2 weeks ameliorated clinical severity of experimental allergic encephalomyelitis, an effect that partially involves the down-regulation of MHC II proteins in the spinal cord. Therefore, we sought to elucidate the molecular mechanisms of minocycline inhibitory effects on MHC II expression in microglia. Although complex, the co-activator class II transactivator (CIITA) is a key regulator of MHC II expression. Here we show that minocycline inhibited interferongamma (IFNgamma)-induced CIITA and MHC II mRNA. Interestingly, however, it was without effect on STAT1 phosphorylation or IRF-1 expression, transcription factors that are activated by IFNgamma and necessary for CIITA expression. Further experiments revealed that MHC II expression is down-regulated in the presence of the PKC(alpha) inhibitor Gö6976. Minocycline inhibited IFNgamma-induced PKC(alpha/betaII) phosphorylation and the nuclear translocation of both PKC(alpha/betaII) and IRF-1 that subsequently inhibits CIITA expression. Our present data delineate a molecular pathway of minocycline action that includes inhibitory effects on PKC(alpha/betaII) and transcription factors that regulate the expression of critical inflammatory genes such as MHC II. Such a fundamental mechanism may underlie the pleiotropic effects of minocycline in CNS inflammatory disorders.
Collapse
Affiliation(s)
- Maria Nikodemova
- Department of Medical Sciences, School of Veterinary Medicine, University of Wisconsin, Madison, Wisconsin 53706, USA.
| | | | | | | | | |
Collapse
|
725
|
Toll-like receptor 2 signaling is a mediator of apoptosis in herpes simplex virus-infected microglia. J Neuroinflammation 2007; 4:11. [PMID: 17470292 PMCID: PMC1866225 DOI: 10.1186/1742-2094-4-11] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Accepted: 04/30/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Information regarding the response of brain cells to infection with herpes simplex virus (HSV)-1 is needed for a complete understanding of viral neuropathogenesis. We have recently demonstrated that microglial cells respond to HSV infection by producing a number of proinflammatory cytokines and chemokines through a mechanism involving Toll-like receptor 2 (TLR2). Following this cytokine burst, microglial cells rapidly undergo cell death by apoptosis. We hypothesized that TLR2 signaling might mediate the cell death process as well. METHODS To test this hypothesis, we investigated HSV-induced cell death of microglia obtained from both wild-type and TLR2-/- mice. Cell death was studied by oligonucleosomal ELISA and TUNEL staining, and the mechanisms of apoptosis were further analyzed using murine apoptosis-specific microarrays. The data obtained from microarray analysis were then validated using quantitative real-time PCR assays. RESULTS HSV infection induced apoptotic cell death in microglial cells from wild-type as well as TLR2 cells. However, the cell death at 24 h p.i. was markedly lower in knockout cells. Furthermore, microarray analyses clearly showed that the expression of pro-apoptotic genes was down-regulated at the time when wild-type cells were actively undergoing apoptosis, indicating a differential response to HSV in cells with or without TLR2. CONCLUSION We demonstrate here that HSV induces an apoptotic response in microglial cells which is mediated through TLR2 signaling.
Collapse
|
726
|
Hasegawa S, Yamaguchi M, Nagao H, Mishina M, Mori K. Enhanced cell-to-cell contacts between activated microglia and pyramidal cell dendrites following kainic acid-induced neurotoxicity in the hippocampus. J Neuroimmunol 2007; 186:75-85. [PMID: 17428546 DOI: 10.1016/j.jneuroim.2007.03.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2006] [Revised: 02/08/2007] [Accepted: 03/02/2007] [Indexed: 12/18/2022]
Abstract
Microglia participate in immune responses in the brain. However, little is known about the contact-mediated interaction between microglia and neurons. We report here that the cell-to-cell contacts between microglial processes and dendrites of hippocampal CA1 neurons were dramatically increased in density and area following local injection of kainic acid (KA). A similar KA-induced increase in the degree of intercellular contacts was observed in mice lacking telencephalin (TLCN), a neuronal dendritic adhesion molecule of ICAM family. The results suggest that adhesive contacts independent of TLCN and contact-mediated interactions between microglia and dendrites were promoted by excitotoxic brain injury.
Collapse
Affiliation(s)
- Sanae Hasegawa
- Department of Physiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | | | | | | | | |
Collapse
|
727
|
Li M, Wang Y, Guo R, Bai Y, Yu Z. Glucocorticoids impair microglia ability to induce T cell proliferation and Th1 polarization. Immunol Lett 2007; 109:129-37. [PMID: 17360046 DOI: 10.1016/j.imlet.2007.02.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2006] [Revised: 01/22/2007] [Accepted: 02/04/2007] [Indexed: 10/23/2022]
Abstract
Glucocorticoids (GC) are essential neuroendocrine regulators of the immune system during stress, and prolonged psychological stress has been shown to be immunosuppressive. However, little is known about how GC influence the role of microglia, the most potent antigen presenting cell (APC) residing in the central nervous system (CNS), in the T cell immune response during stress. Therefore, we investigated whether GC could modulate the function of microglia and thus affect T cell response in vitro. In interferon (IFN)-gamma-stimulated microglia, GC reduced secretion of the pro-inflammatory cytokines interleukin (IL)-12, IL-6 and tumor necrosis factor (TNF)-alpha, inhibited expression of major histocompatibility complex (MHC) class II, and costimulators CD40 and CD80 on microglia, but up-regulated the expression of co-inhibitors B7-H1 and B7-DC. In addition, GC induced the apoptosis of microglia directly. As a result, treatment of microglia with GC reduced their ability to stimulate CD4(+) Th cell proliferation primed by anti-CD3 monoclonal antibody (mAb), and induced a shift to the Th2 response with the imbalance between Th1 and Th2 cytokines. Our data suggest that the inhibitory effects of GC on the APC function of microglia may contribute to the stress-induced suppression of T cell response in the CNS.
Collapse
Affiliation(s)
- Maoquan Li
- Department of Public Health, Chengdu Medical College, Chengdu, Sichuan, PR China
| | | | | | | | | |
Collapse
|
728
|
Shie FS, Ling Z. Therapeutic strategy at the crossroad of neuroinflammation and oxidative stress in age-related neurodegenerative diseases. Expert Opin Ther Pat 2007. [DOI: 10.1517/13543776.17.4.419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
729
|
Whitton PS. Inflammation as a causative factor in the aetiology of Parkinson's disease. Br J Pharmacol 2007; 150:963-76. [PMID: 17339843 PMCID: PMC2013918 DOI: 10.1038/sj.bjp.0707167] [Citation(s) in RCA: 474] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2006] [Revised: 12/12/2006] [Accepted: 01/11/2007] [Indexed: 12/21/2022] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder affecting mainly the elderly, although a small proportion of PD patients develop the illness at a much younger age. In the former group, idiopathic PD patients, the causes of the illness have been the subject of longstanding debate with environmental toxins, mitochondrial dysfunction, abnormal protein handling and oxidative stress being suggested. One problem has been that the epidemiology of PD has offered few clues to provide evidence for a single major causative factor. Comparatively recently it has been found that in both patients and experimental models of PD in animals neuroinflammation appears to be a ubiquitous finding. These cases present with all of the classical features of inflammation including phagocyte activation, increased synthesis and release of proinflammatory cytokines and complement activation. Although this process is vital for normal function and protection in both the CNS, as in the periphery, it is postulated that in the aetiology of PD this process may spiral out of control with over activation of microglia, over production of cytokines and other proinflammatory mediators as well as the release of destructive molecules such as reactive oxygen species. Given that dopaminergic neurons in the substantia nigra are relatively vulnerable to 'stress' and the region has a large population of microglia in comparison to other CNS structures, these events may easily trigger neurodegeneration. These factors are examined in this review along with a consideration of the possible use of anti-inflammatory drugs in PD.
Collapse
Affiliation(s)
- P S Whitton
- 1Department of Pharmacology, The School of Pharmacy, London, UK.
| |
Collapse
|
730
|
Sriram K, O'Callaghan JP. Divergent roles for tumor necrosis factor-alpha in the brain. J Neuroimmune Pharmacol 2007; 2:140-53. [PMID: 18040839 DOI: 10.1007/s11481-007-9070-6] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 03/01/2007] [Indexed: 01/05/2023]
Abstract
Proinflammatory cytokines and chemokines have been implicated in the pathogenesis of several neurological and neurodegenerative disorders. Prominent among such factors is the pleiotropic cytokine, tumor necrosis factor (TNF)-alpha. Under normal physiological conditions, TNF-alpha orchestrates a diverse array of functions involved in immune surveillance and defense, cellular homeostasis, and protection against certain neurological insults. However, paradoxical effects of this cytokine have been observed. TNF-alpha is elicited in the brain following injury (ischemia, trauma), infection (HIV, meningitis), neurodegeneration (Alzheimer's, Parkinson's), and chemically induced neurotoxicity. The multifarious identity for this cytokine appears to be influenced by several mechanisms. Among the most prominent are the regulation of TNFalpha-induced NF-kappaB activation by adapter proteins such as TRADD and TRAF, and second, the heterogeneity of microglia and their distribution pattern across brain regions. Here, we review the differential role of TNF-alpha in response to brain injury, with emphasis on neurodegeneration, and discuss the possible mechanisms for such diverse and region-specific effects.
Collapse
Affiliation(s)
- Krishnan Sriram
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, CDC-NIOSH, 1095 Willowdale Road, Morgantown, WV 26505, USA
| | | |
Collapse
|
731
|
Sanders P, De Keyser J. Janus faces of microglia in multiple sclerosis. ACTA ACUST UNITED AC 2007; 54:274-85. [PMID: 17383006 DOI: 10.1016/j.brainresrev.2007.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/02/2007] [Accepted: 03/02/2007] [Indexed: 12/25/2022]
Abstract
Multiple sclerosis (MS) is the most common cause of neurological disability in young adults. The disease is characterized by inflammatory reactions, demyelination and axonal loss in the brain, spinal cord and optic nerves. Microglia seem to play an important role in the inflammatory processes in MS, since they are found in actively demyelinating lesions. Their role in the differentiation of T cells could led to the expansion of inflammation and tissue destruction. However, microglia are also involved in the termination of an inflammatory response and produce protective factors. To be able to therapeutically manipulate microglia, their exact function in the onset and development of MS needs to be clarified. This review provides an overview of the functions of the most important microglia-associated molecules in MS, being CD40, B7-1 and B7-2, interferon-gamma, tumor necrosis factor-alpha, chemokines, prostanoids, and nitric oxide.
Collapse
Affiliation(s)
- Patricia Sanders
- Department of Neurology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
732
|
Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol 2007; 28:138-45. [PMID: 17276138 DOI: 10.1016/j.it.2007.01.005] [Citation(s) in RCA: 965] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2006] [Revised: 01/02/2007] [Accepted: 01/19/2007] [Indexed: 02/06/2023]
Abstract
Innate immunity is a constitutive component of the central nervous system (CNS) and relies strongly on resident myeloid cells, the microglia. However, evidence is emerging that the most abundant glial cell population of the CNS, the astrocyte, participates in the local innate immune response triggered by a variety of insults. Astrocytes display an array of receptors involved in innate immunity, including Toll-like receptors, nucleotide-binding oligomerization domains, double-stranded RNA-dependent protein kinase, scavenger receptors, mannose receptor and components of the complement system. Following activation, astrocytes are endowed with the ability to secrete soluble mediators, such as CXCL10, CCL2, interleukin-6 and BAFF, which have an impact on both innate and adaptive immune responses. The role of astrocytes in inflammation and tissue repair is elaborated by recent in vivo studies employing cell-type specific gene targeting.
Collapse
Affiliation(s)
- Cinthia Farina
- Neuroimmunology and Neuromuscular Disorders Unit, National Neurological Institute Carlo Besta, 20133 Milan, Italy.
| | | | | |
Collapse
|
733
|
Burster T, Beck A, Poeschel S, Øren A, Baechle D, Reich M, Roetzschke O, Falk K, Boehm BO, Youssef S, Kalbacher H, Overkleeft H, Tolosa E, Driessen C. Interferon-gamma regulates cathepsin G activity in microglia-derived lysosomes and controls the proteolytic processing of myelin basic protein in vitro. Immunology 2007; 121:82-93. [PMID: 17302735 PMCID: PMC2265925 DOI: 10.1111/j.1365-2567.2007.02540.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The serine protease cathepsin (Cat) G dominates the proteolytic processing of the multiple sclerosis (MS)-associated autoantigen myelin basic protein (MBP) in lysosomes from primary human B cells and dendritic cells. This is in contrast to B-lymphoblastoid cell lines, where the asparagine endopeptidase (AEP) is responsible for this task. We have analysed microglia-derived lysosomal proteases for their ability to process MBP in vitro. In lysosomes derived from primary murine microglia, CatD, CatS, AEP and CatG were involved in the processing of MBP. Interestingly, when microglia were treated with interferon-gamma to mimic a T helper type 1-biased cytokine milieu in MS, CatG was drastically down-regulated, in contrast to CatS, CatB, CatL, CatD or AEP. This resulted in significantly increased stability of MBP and a selective lack of CatG-derived proteolytic fragments; however, it did not affect the gross pattern of MBP processing. Inhibition of serine proteases eliminated the processing differences between lysosomal extracts from resting microglia compared to interferon-stimulated microglia. Thus, the cytokine environment modulates lysosomal proteases in microglia by a selective down-regulation of CatG, leading to decreased MBP-processing by microglia-derived lysosomal proteases in vitro.
Collapse
Affiliation(s)
- Timo Burster
- Department of Medicine II, University of Tübingen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
734
|
Correa SG, Maccioni M, Rivero VE, Iribarren P, Sotomayor CE, Riera CM. Cytokines and the immune–neuroendocrine network: What did we learn from infection and autoimmunity? Cytokine Growth Factor Rev 2007; 18:125-34. [PMID: 17347025 DOI: 10.1016/j.cytogfr.2007.01.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The initial view of the neuroendocrine-immune communication as the brake of immune activation is changing. Recent evidence suggests that the optimization of the body's overall response to infection could be actually the role of the immune-endocrine network. In gradually more complex organisms, the multiplicity of host-pathogen interfaces forced the development of efficient and protective responses. Molecules such as cytokines and Toll-like receptors (TLRs) are distributed both in the periphery and in the brain to participate in a coordinated adaptive function. When sustained release of inflammatory mediators occurs, as in autoimmune diseases, undesirable pathological consequences become evident with different manifestations and outcomes. Clearly, organisms are not well adapted to that disregulated condition yet, suggesting that additional partners within neuroendocrine-immune interactions might emerge from the evolutionary road.
Collapse
Affiliation(s)
- Silvia G Correa
- Immunology, Department of Biochemical Chemistry, CIBICI (CONICET), Faculty of Chemical Sciences, National University of Cordoba, Haya de la Torre y Medina Allende, 5000 Cordoba, Argentina.
| | | | | | | | | | | |
Collapse
|
735
|
Tezel G, Yang X, Luo C, Peng Y, Sun SL, Sun D. Mechanisms of immune system activation in glaucoma: oxidative stress-stimulated antigen presentation by the retina and optic nerve head glia. Invest Ophthalmol Vis Sci 2007; 48:705-714. [PMID: 17251469 PMCID: PMC2494942 DOI: 10.1167/iovs.06-0810] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Evidence supports the immune system activity accompanying glaucomatous neurodegeneration. This study aimed to determine the in vitro effects of reactive oxygen species (ROS) on the phenotype and antigen-presenting function of the retina and optic nerve head glia. METHODS Cultures of rat retina and optic nerve head glia were treated with a mixture of ROS-generating compounds for 24 and 48 hours. Pretreated glial cells were then coincubated with syngeneic CD4(+) T cells for 48 hours. ROS generation and cell viability were assessed with the use of dihydroethidium and calcein assays, respectively. Flow cytometry and immunocytochemistry were used to determine major histocompatibility complex (MHC) class II molecules. In addition, functional experiments were performed to determine the proliferation and cytokine secretion of T cells using [(3)H]-thymidine incorporation and TNF-alpha assays, respectively. RESULTS MHC class II molecules were upregulated on glial cells exposed to ROS. Compared with the control glia, glial cells in ROS-generating systems were found to be more potent inducers of T-cell activation in a cell density- and time-dependent manner, as assessed by increased T-cell proliferation (approximately threefold) and TNF-alpha secretion (approximately sixfold; P < 0.01). When an ROS scavenging treatment was applied, MHC class II upregulation on glial cells persisted, but antigen-mediated T-cell activation was significantly decreased (P < 0.01), indicating an additional costimulatory function of ROS during antigen presentation. CONCLUSIONS These in vitro findings support that ROS regulate the immune response by stimulating the antigen-presenting ability of glial cells and functioning as costimulatory molecules for antigen presentation.
Collapse
Affiliation(s)
- Gülgün Tezel
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Kentucky Lions Eye Center, 301 E. Muhammad Ali Boulevard, Louisville, KY 40202, USA.
| | | | | | | | | | | |
Collapse
|
736
|
Ebert S, Zeretzke M, Nau R, Michel U. Microglial cells and peritoneal macrophages release activin A upon stimulation with Toll-like receptor agonists. Neurosci Lett 2007; 413:241-4. [PMID: 17194540 DOI: 10.1016/j.neulet.2006.11.065] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2006] [Revised: 11/28/2006] [Accepted: 11/29/2006] [Indexed: 01/30/2023]
Abstract
Activin A levels are elevated in the cerebrospinal fluid (CSF) of patients with meningitis and in the sera of patients with sepsis. The source(s) of the elevated concentrations of activin A in CSF and serum have not yet been discovered. Here we demonstrate that primary mouse microglial cells and peritoneal macrophages release activin A after treatment with agonists of Toll-like receptor (TLR) 2, 4, and 9. These findings provide further evidence for a role of activin in the innate immune response and suggest that microglial cells and macrophages are a source of elevated activin A concentrations observed in the CSF during bacterial meningitis and in the systemic circulation during sepsis.
Collapse
Affiliation(s)
- Sandra Ebert
- Department of Neurology, University of Göttingen, Robert-Koch-Street 40, 37075 Göttingen, Germany.
| | | | | | | |
Collapse
|
737
|
Nakamichi K, Saiki M, Kitani H, Kuboyama Y, Morimoto K, Takayama-Ito M, Kurane I. Roles of NF-kappaB and MAPK signaling pathways in morphological and cytoskeletal responses of microglia to double-stranded RNA. Neurosci Lett 2007; 414:222-7. [PMID: 17284350 DOI: 10.1016/j.neulet.2006.12.058] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 12/19/2006] [Accepted: 12/20/2006] [Indexed: 11/26/2022]
Abstract
Following virus infection of the central nervous system, microglia become activated and undergo morphological as well as functional transformations, thereby initiating effective antiviral actions. Herein, we have examined the contribution of nuclear factor kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK) signaling pathways to cell shape determination and cytoskeletal organization in microglia upon stimulation with double-stranded RNA (dsRNA), a conserved molecular pattern of virus infection. Under non-proliferative condition, microglial MG6-1 cells displayed a distinctive morphology with spinescent processes and small somata. Following dsRNA stimulation, the process-bearing microglial cells exhibited swift and drastic changes in cell morphology, filamentous actin (F-actin) structure, and intracellular signaling. In the dsRNA-stimulated microglial cells, the activation of c-Jun N-terminal kinase (JNK) pathway was involved in morphological alteration into an ameboid state. We also found that p38 signaling pathway negatively regulates the formation of cytoplasmic vacuoles in microglial cells. Furthermore, the dsRNA-induced accumulation of F-actin was partly mediated by NF-kappaB, JNK, and p38 pathways. These results indicate that NF-kappaB and MAPK signaling pathways mediate morphological and cytoskeletal changes during dsRNA-induced microglial activation.
Collapse
Affiliation(s)
- Kazuo Nakamichi
- Laboratory of Neurovirology, Department of Virology 1, National Institute of Infectious Diseases, Shinjuku, Tokyo 162-8640, Japan.
| | | | | | | | | | | | | |
Collapse
|
738
|
Perreau M, Mennechet F, Serratrice N, Glasgow JN, Curiel DT, Wodrich H, Kremer EJ. Contrasting effects of human, canine, and hybrid adenovirus vectors on the phenotypical and functional maturation of human dendritic cells: implications for clinical efficacy. J Virol 2007; 81:3272-84. [PMID: 17229706 PMCID: PMC1866049 DOI: 10.1128/jvi.01530-06] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Antipathogen immune responses create a balance between immunity, tolerance, and immune evasion. However, during gene therapy most viral vectors are delivered in substantial doses and are incapable of expressing gene products that reduce the host's ability to detect transduced cells. Gene transfer efficacy is also modified by the in vivo transduction of dendritic cells (DC), which notably increases the immunogenicity of virions and vector-encoded genes. In this study, we evaluated parameters that are relevant to the use of canine adenovirus serotype 2 (CAV-2) vectors in the clinical setting by assaying their effect on human monocyte-derived DC (hMoDC). We compared CAV-2 to human adenovirus (HAd) vectors containing the wild-type virion, functional deletions in the penton base RGD motif, and the CAV-2 fiber knob. In contrast to the HAd type 5 (HAd5)-based vectors, CAV-2 poorly transduced hMoDC, provoked minimal upregulation of major histocompatibility complex class I/II and costimulatory molecules (CD40, CD80, and CD86), and induced negligible morphological changes indicative of DC maturation. Functional maturation assay results (e.g., reduced antigen uptake; tumor necrosis factor alpha, interleukin-1beta [IL-1beta], gamma interferon [IFN-gamma], IL-10, IL-12, and IFN-alpha/beta secretion; and stimulation of heterologous T-cell proliferation) were also significantly lower for CAV-2. Our data suggested that this was due, in part, to the use of an alternative receptor and a block in vesicular escape. Additionally, HAd5 vector-induced hMoDC maturation was independent of the aforementioned cytokines. Paradoxically, an HAd5/CAV-2 hybrid vector induced the greatest phenotypical and functional maturation of hMoDC. Our data suggest that CAV-2 and the HAd5/CAV-2 vector may be the antithesis of Adenoviridae immunogenicity and that each may have specific clinical advantages.
Collapse
Affiliation(s)
- Matthieu Perreau
- Institut de Génétique Moléculaire de Montpellier, CNRS 5535, 1919 Route de Mende, 34293 Montpellier, France
| | | | | | | | | | | | | |
Collapse
|
739
|
Dijkstra IM, de Haas AH, Brouwer N, Boddeke HWGM, Biber K. Challenge with innate and protein antigens induces CCR7 expression by microglia in vitro and in vivo. Glia 2007; 54:861-72. [PMID: 16977602 DOI: 10.1002/glia.20426] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Since activated microglia are able to phagocytose damaged cells and subsequently express major histocompatibility complex class II (MHC-II) and co-stimulatory proteins, they are considered to function as antigen presenting cells (APCs) in the central nervous system. The maturation and migratory potential of professional APCs is associated with the expression of chemokine receptor CCR7. We therefore investigated whether the immunological activation of microglia induces CCR7 expression. We here present that activation of cultured microglia by both the innate antigen lipopolysaccharide and protein antigen ovalbumin rapidly induces CCR7 expression, accompanied by increased MHC-II expression. Moreover, it is shown that CCR7 expression in IBA-1 positive cells is induced during the symptom onset and progression of experimental autoimmune encephalomyelitis, a rodent model for multiple sclerosis. These results suggest that microglia express CCR7 under specific inflammatory conditions, corroborating the idea that microglia develop into APCs with migratory potential toward lymphoid chemokines.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigen Presentation/immunology
- Antigen-Presenting Cells/immunology
- Antigens/immunology
- Chemotaxis/immunology
- Disease Models, Animal
- Encephalitis/immunology
- Encephalitis/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Female
- Gliosis/immunology
- Histocompatibility Antigens Class II/immunology
- Lipopolysaccharides/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/immunology
- Ovalbumin/immunology
- Phagocytosis/immunology
- Receptors, CCR7
- Receptors, Chemokine/genetics
- Receptors, Chemokine/immunology
Collapse
Affiliation(s)
- I M Dijkstra
- Department of Medical Physiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
740
|
Dimayuga FO, Wang C, Clark JM, Dimayuga ER, Dimayuga VM, Bruce-Keller AJ. SOD1 overexpression alters ROS production and reduces neurotoxic inflammatory signaling in microglial cells. J Neuroimmunol 2007; 182:89-99. [PMID: 17097745 PMCID: PMC1797892 DOI: 10.1016/j.jneuroim.2006.10.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 09/20/2006] [Accepted: 10/04/2006] [Indexed: 02/04/2023]
Abstract
Activation of the oxidative burst is one of the earliest biochemical events in microglial activation, but it is not understood yet how free radicals participate in inflammatory signaling. To determine the role that specific reactive oxygen species play in microglial activation, the levels of SOD1 were manipulated in N9 murine microglia. Stable overexpression of SOD1 caused significant decreases in superoxide and nitric oxide production, with concurrent increases in hydrogen peroxide following LPS. However, LPS-induced activation of NFkappaB, and release of TNFalpha and IL-6 were significantly attenuated in SOD1 overexpressing cells, as was the ability of microglia to induce toxicity in cultured neurons. Conversely, acute inhibition of SOD1 with disulfiram was associated with increased nitric oxide and cytokine release, and increased neurotoxicity. Together, these data suggest that superoxide radicals in microglia play important roles in directing redox-sensitive inflammatory signaling and initiating neurotoxic inflammation.
Collapse
Affiliation(s)
- Filomena O Dimayuga
- Mn 222 Chandler Medical Center, Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY 40536, United States
| | | | | | | | | | | |
Collapse
|
741
|
Frank MG, Baratta MV, Sprunger DB, Watkins LR, Maier SF. Microglia serve as a neuroimmune substrate for stress-induced potentiation of CNS pro-inflammatory cytokine responses. Brain Behav Immun 2007; 21:47-59. [PMID: 16647243 DOI: 10.1016/j.bbi.2006.03.005] [Citation(s) in RCA: 442] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2006] [Revised: 03/06/2006] [Accepted: 03/09/2006] [Indexed: 02/07/2023] Open
Abstract
Prior exposure to a stressor can potentiate CNS pro-inflammatory immune responses to a peripheral immune challenge. However, the neuroimmune substrate(s) mediating this effect has not been determined. The present investigation examined whether microglia serve as this neuroimmune substrate given that microglia are the primary immune effector cell in the CNS. The effect of inescapable shock (IS) on glial activation (MHC II, CD11b, Iba-1, and GFAP) and regulatory markers (CD200) in vivo, and microglia pro-inflammatory responses (interleukin-1beta; IL-1beta) to lipopolysaccharide (LPS) ex vivo, were assessed in rat hippocampus. IS upregulated the microglia activation marker MHC II 24h post-IS, while the astroglia marker GFAP was unaffected. IS also downregulated the neuronal glycoprotein CD200, which functions to hold microglia in a quiescent state. Moreover, IS potentiated the pro-inflammatory response to LPS ex vivo 24h post-IS in isolated hippocampal microglia. Finally, the behavioral controllability of shock was manipulated and the effect of escapable (controllable) shock was comparable to the effect of IS on hippocampal microglia responses to LPS ex vivo. The present results suggest that stress can activate microglia, thereby sensitizing the pro-inflammatory reactivity of microglia to immunogenic stimuli.
Collapse
Affiliation(s)
- Matthew G Frank
- Department of Psychology and Center for Neuroscience, Campus Box 345, University of Colorado, Boulder, CO 80309-0345, USA.
| | | | | | | | | |
Collapse
|
742
|
Quintana A, Giralt M, Molinero A, Campbell IL, Penkowa M, Hidalgo J. Analysis of the cerebral transcriptome in mice subjected to traumatic brain injury: importance of IL-6. Neuroimmunomodulation 2007; 14:139-43. [PMID: 18073505 DOI: 10.1159/000110637] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Traumatic brain injury is one of the leading causes of incapacity and death among young people. Injury to the brain elicits a potent inflammatory response, comprising recruitment of inflammatory cells, reactive astrogliosis and activation of brain macrophages. Under the influence of presumably several cytokines and growth factors, a cascade of events is activated that result ultimately in increased oxidative stress and tissue damage, but also in activation of counterregulatory factors and tissue regeneration. The complexity of this response is being unraveled by high-throughput methodologies such as microarrays. The combination of these modern techniques with the comparison of normal and genetically modified mice boosts the significance of the results obtained. With this approach, we have demonstrated that a cytokine such as interleukin-6 is one of the key players in the response of the brain to injury.
Collapse
Affiliation(s)
- Albert Quintana
- Institute of Neurosciences and Department of Cellular Biology, Physiology and Immunology, Animal Physiology Unit, Faculty of Sciences, Autonomous University of Barcelona, Bellaterra, Spain
| | | | | | | | | | | |
Collapse
|
743
|
Santos AM, Calvente R, Tassi M, Carrasco MC, Martín-Oliva D, Marín-Teva JL, Navascués J, Cuadros MA. Embryonic and postnatal development of microglial cells in the mouse retina. J Comp Neurol 2007; 506:224-39. [DOI: 10.1002/cne.21538] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
744
|
Bruce-Keller AJ, Dimayuga FO, Reed JL, Wang C, Angers R, Wilson ME, Dimayuga VM, Scheff SW. Gender and Estrogen Manipulation Do Not Affect Traumatic Brain Injury in Mice. J Neurotrauma 2007; 24:203-15. [PMID: 17263684 DOI: 10.1089/neu.2006.0163] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
As epidemiological data have suggested that female patients may have improved clinical prognoses following traumatic brain injury (TBI) compared to males, we designed experiments to determine the role of gender and estrogen in TBI-induced brain injury and inflammation in rodents. To this end, male and female C57Bl/6 mice were separated into the following four groups: intact males, intact females with vehicle supplementation, ovariectomized females with vehicle supplementation, and ovariectomized females with estrogen supplementation. All mice were subjected to a controlled cortical impact model of TBI, and cortical injury, hippocampal degeneration, microglial activation, and brain cytokine expression were analyzed after injury. Additionally, the spleens were harvested and cytokine release from cultured splenic cells was measured in response to specific stimuli. Data indicate that TBI-induced cortical and hippocampal injury, as well as injury-related microglial activation were not significantly affected by gender or estrogen manipulation. Conversely, brain levels of MCP-1 and IL-6 were significantly increased in males and intact females following TBI, but not in female mice that had been ovariectomized and supplemented with either estrogen or vehicle. Evaluation of splenic responses showed that the spleen was only moderately affected by TBI, and furthermore that spleens isolated from mice that had been given estrogen supplementation showed significantly higher release of the anti-inflammatory cytokine IL-4, regardless of the presence of absence of TBI. Overall, these data indicate that while estrogen can modulate immune responses, and indeed can predispose splenic responses towards and anti-inflammatory phenotype, these effects do not translate to decreased brain injury or inflammation following TBI in mice.
Collapse
|
745
|
Wang XJ, Yan ZQ, Lu GQ, Stuart S, Chen SD. Parkinson disease IgG and C5a-induced synergistic dopaminergic neurotoxicity: Role of microglia. Neurochem Int 2007; 50:39-50. [PMID: 16971022 DOI: 10.1016/j.neuint.2006.07.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2006] [Revised: 07/04/2006] [Accepted: 07/07/2006] [Indexed: 11/18/2022]
Abstract
Increasing evidence suggests the involvement of immune/inflammatory system in Parkinson's disease (PD). Many immune/inflammatory factors may synergistically participate in PD. In this study, we demonstrated that immunoglobulin G from the serum of 4/11 PD patients (PDIgG, 60microg/ml) and recombinant human C5a (0.1nM) synergistically induced selective dopaminergic neurodegeneration in rat mesencephalic neuron-glia cultures, while that PDIgG alone or C5a alone was minimally toxic or nontoxic. IgG from 17 disease controls and from 7 normal controls did not significantly induce dopaminergic neurotoxicity in the cultures even in the presence of C5a. Using mesencephalic neuron-enriched cultures, we found that the synergistic dopaminergic neurotoxicity was mediated by glia. The results from microglia-supplemented neuronal cultures, astroglia-supplemented neuronal cultures and neuron-astroglia cocultures indicated that microglia, not astroglia, played a pivotal role in the neurotoxicity. Through immunocytochemistry analysis and assay of proinflammatory factors, we observed that each of the four PDIgGs (60microg/ml) and C5a (0.1nM) synergistically induced microglia activation and production of superoxide and nitric oxide (NO) in neuron-glia cultures. Further investigations indicated that superoxide and NO were both responsible for the synergistic neurotoxicity. Finally, using F(ab')(2) fragments of PDIgG, we demonstrated that microglial Fc receptors may play an important role in the neurotoxicity. Our work provides new evidence for the involvement of the immune/inflammatory system in PD and helpful clues for studying the combined effect of antibody and complement on microglia.
Collapse
Affiliation(s)
- Xi-Jin Wang
- Department of Neurology & Institute of Neurology, Ruijin Hospital affiliated to Shanghai Jiao-Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | |
Collapse
|
746
|
Mrass P, Weninger W. Immune cell migration as a means to control immune privilege: lessons from the CNS and tumors. Immunol Rev 2006; 213:195-212. [PMID: 16972905 DOI: 10.1111/j.1600-065x.2006.00433.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Certain organs, such as the brain, eye, and gonads, are particularly sensitive to damage by inflammation. Therefore, these tissues have developed unique immunological properties that curtail inflammatory responses, a phenomenon termed immune privilege. In addition, by co-opting some of the regulatory cues operant in immune privilege in normal organs, tumors can evade immunosurveillance. While many different mechanisms contribute to immune privilege, there is evidence that leukocyte migration is an important checkpoint in its control. This hypothesis is based on the fact that leukocyte entry into these organs is restricted by physical barriers and that the collapse of these obstacles marks a critical step in the development of inflammatory/autoimmune disease at these sites. Numerous studies in a variety of experimental systems have characterized the molecular and cellular mechanisms involved in leukocyte homing to immune-privileged organs. Recently, two-photon microscopy has revealed critical insights into the events occurring in the extravascular space of immune-privileged organs, including locomotion patterns and interactive behavior of leukocytes in the interstitial space. Here, we review our current understanding of immune cell migration to and within immune-privileged organs and highlight how this knowledge may be exploited for immunotherapeutic purposes.
Collapse
Affiliation(s)
- Paulus Mrass
- Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | |
Collapse
|
747
|
Narita M, Miyoshi K, Narita M, Suzuki T. Involvement of microglia in the ethanol-induced neuropathic pain-like state in the rat. Neurosci Lett 2006; 414:21-5. [PMID: 17284346 DOI: 10.1016/j.neulet.2006.10.062] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 10/17/2006] [Accepted: 10/18/2006] [Indexed: 12/31/2022]
Abstract
Central mechanisms of neuropathy induced by chronic ethanol treatment are almost unknown. In this study, rats were treated with ethanol-diet for 72 days. Mechanical hyperalgesia was observed during ethanol consumption, even after ethanol withdrawal. Under these conditions, a microglial marker ionized calcium-binding adaptor molecule 1-, but not a neuron marker microtuble associated protein-2-, like immunoreactivies were increased in the rat spinal cord. Furthermore, hypertrophy of microglia was clearly observed following chronic ethanol consumption. These findings support the idea that the activation and hypertrophy of microglia in the spinal cord may be, at least in part, associated with in the induction of ethanol-dependent neuropathic pain-like state.
Collapse
Affiliation(s)
- Minoru Narita
- Department of Toxicology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, 2-4-41 Ebara, Shinagawa-ku, Tokyo 142-8501, Japan.
| | | | | | | |
Collapse
|
748
|
Abstract
This review summarizes the current knowledge on the significance of human leukocyte antigen G (HLA-G) in the nervous system under physiologic and pathologic conditions. The central nervous system (CNS) has classically been viewed as an immune-privileged organ. Immune reactions triggered by, or directed against, CNS structures proceed along specific pathways. The expression and the functioning of the immune-tolerogenic major histocompatibility complex (MHC) molecule HLA-G have revealed novel insights into the endogenous immune-regulatory mechanisms exerted by resident cells within the nervous system, as well as how migrating immune cells contribute to this under pathologic conditions. HLA-G has been evidenced in certain neurologic disorders, including those of autoimmune, infectious, and neoplastic origin. This review compiles the current state of knowledge: how HLA-G is considered to be operative at different levels of the respective pathogenetic cascades of neurologic disorders. These findings are relevant both from a pathogenetic as well as from a therapeutic viewpoint.
Collapse
Affiliation(s)
- Heinz Wiendl
- Department of Neurology, Bayerische Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
749
|
Selak S, Paternain AV, Fritzler MJ, Lerma J. Human autoantibodies against early endosome antigen-1 enhance excitatory synaptic transmission. Neuroscience 2006; 143:953-64. [PMID: 17113235 DOI: 10.1016/j.neuroscience.2006.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 10/05/2006] [Accepted: 10/07/2006] [Indexed: 11/17/2022]
Abstract
Early endosome antigen 1 (EEA1), a peripheral membrane protein associated with the cytoplasmic face of early endosomes, controls vesicle fusion during endocytosis, as extensively studied in non-neuronal cells. In neurons, early endosomes are involved in recycling of synaptic vesicles and neurotransmitter receptors. Since certain patients bearing autoantibodies that target EEA1 develop neurological disease, we studied the subcellular distribution of EEA1 in neurons and the effect on neurotransmission of purified immunoglobulins from the serum of a patient bearing EEA1 autoantibodies. EEA1 was localized in the soma and in the postsynaptic nerve terminals. Electrophysiological recordings in hippocampal slices including purified EEA1 antibodies in the patch pipette solution, revealed a run-up of AMPA, N-methyl-D-aspartate and kainate receptor-mediated excitatory post-synaptic currents recorded from CA3 pyramidal neurons, which was absent in the recordings obtained in the presence of control human immunoglobulin G. Inclusion of human EEA1 antibodies had no effect on inhibitory post-synaptic responses. Recordings in the presence of a dominant-negative C-terminal EEA1 deletion mutant produced a similar effect as observed with human anti-EEA1 antibodies. This specific effect on the excitatory synaptic transmission may be due to the impairment of internalization of specific glutamate receptors and their subsequent accumulation in the synapse. These results may account for the neurological deficits observed in some patients developing EEA1 autoantibodies.
Collapse
Affiliation(s)
- S Selak
- Instituto de Neurociencias de Alicante, Consejo Superior de Investigaciones Científicas and Universidad Miguel Hernández, Aptdo 18, 03550 Sant Joan d'Alacant, Spain
| | | | | | | |
Collapse
|
750
|
Takefuji S, Murase T, Sugimura Y, Takagishi Y, Hayasaka S, Oiso Y, Murata Y. Role of microglia in the pathogenesis of osmotic-induced demyelination. Exp Neurol 2006; 204:88-94. [PMID: 17126835 DOI: 10.1016/j.expneurol.2006.09.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 09/28/2006] [Accepted: 09/30/2006] [Indexed: 11/30/2022]
Abstract
Osmotic demyelination is a serious disease caused by rapid correction of hyponatremia. In humans, demyelinative lesions occur preferentially in the central pons, and thus are termed central pontine myelinolysis. Although accumulation of microglia has been reported in such demyelinative lesions, their role in the pathogenesis of osmotic demyelination remains unclear. We examined the expression of cytokines in microglia that accumulated in the demyelinative lesions in a rat model of osmotic demyelination. Hyponatremia was induced in rats by a combination of dDAVP infusion and liquid diet feeding. After 7 days, serum sodium levels were rapidly corrected by hypertonic saline injection. The rats developed severe motor deficits, and marked demyelinative lesions were found in the midbrain and cerebral cortex. In the area of the demyelinative lesions, massive accumulations of microglia were observed that expressed the proinflammatory cytokines TNF-alpha and IFN-gamma as well as iNOS. In contrast, in hyponatremia corrected rats treated with lovastatin, which is known to inhibit microglial infiltration in various animal models of CNS disease, neurological impairments and the degree of demyelination were significantly ameliorated. Lovastatin also reduced the accumulation of microglia and decreased the expression of TNF-alpha in the demyelinative lesions. These results indicate that microglia play a detrimental role in the pathogenesis of osmotic demyelination by producing proinflammatory cytokines, and further suggest that lovastatin may be useful in repressing the demyelination.
Collapse
Affiliation(s)
- Seiko Takefuji
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya 464-8601, Japan
| | | | | | | | | | | | | |
Collapse
|