51
|
Abstract
The past 25 years have seen a major expansion of knowledge concerning the cause of Parkinson's disease provided by an understanding of environmental and genetic factors that underlie the loss of nigral dopaminergic neurons. Based on the actions of toxins, postmortem investigations, and gene defects responsible for familial Parkinson's disease, there is now a general consensus about the mechanisms of cell death that contribute to neuronal loss in Parkinson's disease. Mitochondrial dysfunction, oxidative stress, altered protein handling, and inflammatory change are considered to lead to cell dysfunction and death by apoptosis or autophagy. Ageing is the single most important risk factor for Parkinson's disease, and the biochemical changes that are a consequence of aging amplify these abnormalities in Parkinson's disease brain. What remains to be determined is the combination and sequence of events leading to cell death and whether this is identical in all brain regions where pathology occurs and in all individuals with Parkinson's disease. Focusing on those events that characterize Parkinson's disease, namely, mitochondrial dysfunction and Lewy body formation, may be the key to further advancing the understanding of pathogenesis and to taking these mechanisms forward as a means of defining targets for neuroprotection.
Collapse
Affiliation(s)
- Anthony H Schapira
- Department of Clinical Neurosciences, Institute of Neurology, University College, London, United Kingdom.
| | | |
Collapse
|
52
|
Abstract
Parkinson's disease (PD) is a neurological movement disorder primarily resulting from damage to the nigrostriatal dopaminergic pathway. To elucidate the pathogenesis, mechanisms of cell death, and to evaluate therapeutic strategies for PD, numerous animal models have been developed. Understanding the strengths and limitations of these models can significantly impact the choice of model, experimental design, and data interpretation. The primary objectives of this article are twofold: First, to assist new investigators who are contemplating embarking on PD research to navigate through the available animal models. Emphasis will be placed on common neurotoxic murine models in which toxic molecules are used to lesion the nigrostriatal dopaminergic system. And second, to provide an overview of basic technical requirements for assessing the pathology, structure, and function of the nigrostriatal pathway.
Collapse
Affiliation(s)
- Kim Tieu
- Department of Neurology in the Center for Translational Neuromedicine, University of Rochester, Rochester, New York 14625, USA.
| |
Collapse
|
53
|
L'Episcopo F, Tirolo C, Testa N, Caniglia S, Morale MC, Impagnatiello F, Marchetti B. Switching the Microglial Harmful Phenotype Promotes Lifelong Restoration of Subtantia Nigra Dopaminergic Neurons from Inflammatory Neurodegeneration in Aged Mice. Rejuvenation Res 2011; 14:411-24. [DOI: 10.1089/rej.2010.1134] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- Francesca L'Episcopo
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
| | - Cataldo Tirolo
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
| | - Nunzio Testa
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
| | - Salvatore Caniglia
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
| | - Maria Concetta Morale
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
| | | | - Bianca Marchetti
- OASI Institute for Research and Care on Mental Retardation and Brain Aging, Neuropharmacology Section, Troina, Italy
- Department of Clinical and Molecular Biomedicine, Pharmacology Section, Faculty of Medicine, Catania, Italy
- Faculty of Pharmacy, University of Catania, Catania, Italy
| |
Collapse
|
54
|
Tufekci KU, Genc S, Genc K. The endotoxin-induced neuroinflammation model of Parkinson's disease. PARKINSON'S DISEASE 2011; 2011:487450. [PMID: 21331154 PMCID: PMC3034925 DOI: 10.4061/2011/487450] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Revised: 11/18/2010] [Accepted: 12/16/2010] [Indexed: 01/22/2023]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder characterized by the progressive loss of dopaminergic (DA) neurons in the substantia nigra. Although the exact cause of the dopaminergic neurodegeneration remains elusive, recent postmortem and experimental studies have revealed an essential role for neuroinflammation that is initiated and driven by activated microglial and infiltrated peripheral immune cells and their neurotoxic products (such as proinflammatory cytokines, reactive oxygen species, and nitric oxide) in the pathogenesis of PD. A bacterial endotoxin-based experimental model of PD has been established, representing a purely inflammation-driven animal model for the induction of nigrostriatal dopaminergic neurodegeneration. This model, by itself or together with genetic and toxin-based animal models, provides an important tool to delineate the precise mechanisms of neuroinflammation-mediated dopaminergic neuron loss. Here, we review the characteristics of this model and the contribution of neuroinflammatory processes, induced by the in vivo administration of bacterial endotoxin, to neurodegeneration. Furthermore, we summarize the recent experimental therapeutic strategies targeting endotoxin-induced neuroinflammation to elicit neuroprotection in the nigrostriatal dopaminergic system. The potential of the endotoxin-based PD model in the development of an early-stage specific diagnostic biomarker is also emphasized.
Collapse
Affiliation(s)
- Kemal Ugur Tufekci
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Sermin Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| | - Kursad Genc
- Department of Neuroscience, Health Science Institute, Dokuz Eylul University, Inciralti, 35340 Izmir, Turkey
| |
Collapse
|
55
|
Shih YT, Chen IJ, Wu YC, Lo YC. San-Huang-Xie-Xin-Tang Protects against Activated Microglia- and 6-OHDA-Induced Toxicity in Neuronal SH-SY5Y Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2011; 2011:429384. [PMID: 19339484 PMCID: PMC3135633 DOI: 10.1093/ecam/nep025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2008] [Accepted: 03/03/2009] [Indexed: 12/30/2022]
Abstract
San-Huang-Xie-Xin-Tang (SHXT), composed of Coptidis rhizoma, Scutellariae radix and Rhei rhizoma, is a traditional Chinese herbal medicine used to treat gastritis, gastric bleeding and peptic ulcers. This study investigated the neuroprotective effects of SHXT on microglia-mediated neurotoxicity using co-cultured lipopolysaccharide (LPS)-activated microglia-like BV-2 cells with neuroblastoma SH-SY5Y cells. Effects of SHXT on 6-hydroxydopamine (6-OHDA)-induced neurotoxicity were also examined in SH-SY5Y cells. Results indicated SHXT inhibited LPS-induced inflammation of BV-2 cells by downregulation of iNOS, NO, COX-2, PGE2, gp91phox, iROS, TNF-α, IL-1β, inhibition of IκBα degradation and upregulation of HO-1. In addition, SHXT increased cell viability and down regulated nNOS, COX-2 and gp91phox of SH-SY5Y cells co-cultured with LPS activated BV-2 cells. SHXT treatment increased cell viability and mitochondria membrane potential (MMP), decreased expression of nNOS, COX-2, gp91phox and iROS, and inhibited IκBα degradation in 6-OHDA-treated SH-SY5Y cells. SHXT also attenuated LPS activated BV-2 cells- and 6-OHDA-induced cell death in differentiated SH-SY5Y cells with db-cAMP. Furthermore, SHXT-inhibited nuclear translocation of p65 subunit of NF-κB in LPS treated BV-2 cells and 6-OHDA treated SH-SY5Y cells. In conclusion, SHXT showed protection from activated microglia- and 6-OHDA-induced neurotoxicity by attenuating inflammation and oxidative stress.
Collapse
Affiliation(s)
- Yu-Tzu Shih
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | | | | | | |
Collapse
|
56
|
Gonçalves J, Baptista S, Martins T, Milhazes N, Borges F, Ribeiro CF, Malva JO, Silva AP. Methamphetamine-induced neuroinflammation and neuronal dysfunction in the mice hippocampus: preventive effect of indomethacin. Eur J Neurosci 2010; 31:315-26. [PMID: 20074221 DOI: 10.1111/j.1460-9568.2009.07059.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Methamphetamine (METH) causes irreversible damage to brain cells leading to neurological and psychiatric abnormalities. However, the mechanisms underlying life-threatening effects of acute METH intoxication remain unclear. Indeed, most of the hypotheses focused on intra-neuronal events, such as dopamine oxidation, oxidative stress and excitotoxicity. Yet, recent reports suggested that glia may contribute to METH-induced neuropathology. In the present study, we investigated the hippocampal dysfunction induced by an acute high dose of METH (30 mg/kg; intraperitoneal injection), focusing on the inflammatory process and changes in several neuronal structural proteins. For that, 3-month-old male wild-type C57BL/6J mice were killed at different time-points post-METH. We observed that METH caused an inflammatory response characterized by astrocytic and microglia reactivity, and tumor necrosis factor (TNF) system alterations. Indeed, glial fibrillary acidic protein (GFAP) and CD11b immunoreactivity were upregulated, likewise TNF-alpha and TNF receptor 1 protein levels. Furthermore, the effect of METH on hippocampal neurons was also investigated, and we observed a downregulation in beta III tubulin expression. To clarify the possible neuronal dysfunction induced by METH, several neuronal proteins were analysed. Syntaxin-1, calbindin D28k and tau protein levels were downregulated, whereas synaptophysin was upregulated. We also evaluated whether an anti-inflammatory drug could prevent or diminish METH-induced neuroinflammation, and we concluded that indomethacin (10 mg/kg; i.p.) prevented METH-induced glia activation and both TNF system and beta III tubulin alterations. In conclusion, we demonstrated that METH triggers an inflammatory process and leads to neuronal dysfunction in the hippocampus, which can be prevented by an anti-inflammatory treatment.
Collapse
Affiliation(s)
- Joana Gonçalves
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Abstract
BACKGROUND The death of dopaminergic neurons in Parkinson's disease (PD) appears to have various causes, including oxidative stress, excitotoxicity, mitochondrial dysfunction (and associated apoptosis), ubiquitin/proteasomal dysfunction, and inflammation, any of which could in principle be the therapeutic target of a neuroprotective drug. The biology of dopaminergic neurons offers further potential targets, involving neurotrophic factors, dopamine-neuron genes, and even neurogenesis. OBJECTIVE To outline each hypothetical neuroprotective mechanism, the evidence suggesting its relevance to PD, and the research on pharmacologic intervention. METHODS A PubMed search was conducted to identify relevant preclinical and clinical literature published between 1989 and 2009. Additional articles were identified by reviewing the reference lists of papers selected in the original search. To circumscribe the survey and facilitate consideration of the conditions required for a neuroprotective effect, emphasis was placed on a single drug class, dopamine agonists, and in particular pramipexole. REVIEW OF THE FIELD: In a variety of in vitro and in vivo PD models, pramipexole exhibited preclinical evidence of neuroprotective actions of all hypothesized types, and in human neuroimaging studies it slowed the rate of loss of markers of dopaminergic function, consistent with drug-conferred neuroprotection in PD itself. Interpretation of the preclinical data was hampered by differences among models and by uncertainties concerning each model's mimicry of PD. Overall, the identified neuroprotection almost always required pretreatment (i.e., before insult) and high drug concentration. Interpretation of the clinical data was hampered by absence of placebo control and of a direct measure of neuroprotection. CONCLUSIONS Although the evidence is promising, neuroprotection in PD remains an elusive goal. In whatever form it emerges, neuroprotective therapy would be a strong argument against deferring PD treatment until symptoms are a significant life impediment, and thus would add urgency to early PD identification.
Collapse
|
58
|
Mastroeni D, Grover A, Leonard B, Joyce JN, Coleman PD, Kozik B, Bellinger DL, Rogers J. Microglial responses to dopamine in a cell culture model of Parkinson's disease. Neurobiol Aging 2009; 30:1805-17. [PMID: 18325635 PMCID: PMC2762863 DOI: 10.1016/j.neurobiolaging.2008.01.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 01/09/2008] [Accepted: 01/13/2008] [Indexed: 11/30/2022]
Abstract
Activated microglia appear to selectively attack dopamine (DA) neurons in the Parkinson's disease (PD) substantia nigra. We investigated potential mechanisms using culture models. As targets, human SH-SY5Y cells were left undifferentiated (UNDIFF) or were differentiated with retinoic acid (RA) or RA plus brain-derived neurotrophic factor (RA/BDNF). RA/BDNF-treated cells were immunoreactive for tyrosine hydroxylase and the DA transporter, took up exogenous DA, and released DA after K(+) stimulation. Undifferentiated and RA-treated cells lacked these characteristics of a DA phenotype. Co-culture of target cells with human elderly microglia resulted in elevated toxicity in DA phenotype (RA/BDNF) cells. Lipopolysaccharide (LPS) plus K(+)-stimulated DA release enhanced toxicity by 500-fold. DA induced microglial chemotaxis in Boyden chambers. Spiperone inhibited this effect. Cultured human elderly microglia expressed mRNAs for D1-D4 but not D5 DA receptors. The microglia, as well as PD microglia in situ, were also immunoreactive for D1-D4 but not D5 DA receptors. These findings demonstrate that activated microglia express DA receptors, and suggest that this mechanism may play a role in the selective vulnerability of DA neurons in PD.
Collapse
Affiliation(s)
- Diego Mastroeni
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| | - Andrew Grover
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| | - Brian Leonard
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| | - Jeffrey N. Joyce
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| | - Paul D. Coleman
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| | - Brooke Kozik
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Denise L. Bellinger
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92350
| | - Joseph Rogers
- Sun Health Research Institute, 10515 West Santa Fe Drive, Sun City, AZ 85372
| |
Collapse
|
59
|
Chao YX, He BP, Tay SSW. Mesenchymal stem cell transplantation attenuates blood brain barrier damage and neuroinflammation and protects dopaminergic neurons against MPTP toxicity in the substantia nigra in a model of Parkinson's disease. J Neuroimmunol 2009; 216:39-50. [PMID: 19819031 DOI: 10.1016/j.jneuroim.2009.09.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 01/14/2023]
Abstract
Immunomodulatory effects of transplanted mesenchymal stem cells (MSCs) in the treatment of Parkinson's disease were studied in the MPTP-induced mouse model. MPTP treatment induced a significant loss of dopaminergic neurons, decreased expressions of claudin 1, claudin 5 and occludin in the substantia nigra compacta (SNc), and functional damage of the blood brain barrier (BBB). Our study further discovered that infiltration of MBLs into the brain to bind with microglia was detected in the SNc of MPTP-treated mice, suggesting that the BBB compromise and MBL infiltration might be involved in the pathogenesis of MPTP-induced PD. In addition, MPTP treatment also increased the expression of mannose-binding lectins (MBLs) in the liver tissue. Intravenous transplantation of MSCs into MPTP-treated mice led to recovery of BBB integrity, suppression of MBL infiltration at SNc and MBL expression in the liver, suppression of microglial activation and prevention of dopaminergic neuron death. No transplanted MSCs were observed to differentiate into dopaminergic neurons, while the MSCs migrated into the SNc and released TGF-beta1 there. Therefore, intravenous transplantation of MSCs which protect dopaminergic neurons from MPTP toxicity may be engaged in anyone or a combination of these mechanisms: repair of the BBB, reduction of MBL in the brain, inhibition of microglial cytotoxicity, and direct protection of dopaminergic neurons.
Collapse
Affiliation(s)
- Yin Xia Chao
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
60
|
Klintworth H, Garden G, Xia Z. Rotenone and paraquat do not directly activate microglia or induce inflammatory cytokine release. Neurosci Lett 2009; 462:1-5. [PMID: 19559752 DOI: 10.1016/j.neulet.2009.06.065] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/29/2009] [Accepted: 06/19/2009] [Indexed: 11/28/2022]
Abstract
Both epidemiological and pathological data suggest an inflammatory response including microglia activation and neuro-inflammation in the Parkinsonian brain. Treatments with lipopolysaccharide (LPS), rotenone and paraquat have been used as models for Parkinson's disease, as they cause dopaminergic neuron degeneration in culture and in animals. Recent studies have suggested that rotenone and paraquat induce neuro-inflammation, however, it is not known if they can directly activate microglia. Here, we use primary cultured microglia to address this question. Microglia activation was analyzed by morphological changes and release of nitric oxide and inflammatory cytokines. Treatment with LPS was used as a positive control. While LPS induced morphological changes characteristic of microglial activation and release of nitric oxide and inflammatory cytokines, rotenone and paraquat did not. Our results suggest that paraquat and rotenone do not act directly on microglia and that neuro-inflammation and microglial activation in animals treated with these agents are likely non-cell autonomous, and may occur as a result of dopaminergic neuron damage or factors released by neurons and other cells.
Collapse
Affiliation(s)
- Heather Klintworth
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195-7234, USA
| | | | | |
Collapse
|
61
|
Zhang D, Hu X, Qian L, Wilson B, Lee C, Flood P, Langenbach R, Hong JS. Prostaglandin E2 released from activated microglia enhances astrocyte proliferation in vitro. Toxicol Appl Pharmacol 2009; 238:64-70. [PMID: 19397918 DOI: 10.1016/j.taap.2009.04.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2009] [Accepted: 04/21/2009] [Indexed: 12/16/2022]
Abstract
Microglial activation has been implicated in many astrogliosis-related pathological conditions including astroglioma; however, the detailed mechanism is not clear. In this study, we used primary enriched microglia and astrocyte cultures to determine the role of microglial prostaglandin E(2) (PGE(2)) in the proliferation of astrocytes. The proliferation of astrocytes was measured by BrdU incorporation. The level of PGE(2) was measured by ELISA method. Pharmacological inhibition or genetic ablation of COX-2 in microglia were also applied in this study. We found that proliferation of astrocytes increased following lipopolysaccharide (LPS) treatment in the presence of microglia. Furthermore, increased proliferation of astrocytes was observed in the presence of conditioned media from LPS-treated microglia. The potential involvement of microglial PGE(2) in enhanced astrocyte proliferation was suggested by the findings that PGE(2) production and COX-2 expression in microglia were increased by LPS treatment. In addition, activated microglia-induced increases in astrocyte proliferation were blocked by the PGE(2) antagonist AH6809, COX-2 selective inhibitor DuP-697 or by genetic knockout of microglial COX-2. These findings were further supported by the finding that addition of PGE(2) to the media significantly induced astrocyte proliferation. These results indicate that microglial PGE(2) plays an important role in astrocyte proliferation, identifying PGE(2) as a key neuroinflammatory molecule that triggers the pathological response related to uncontrollable astrocyte proliferation. These findings are important in elucidating the role of activated microglia and PGE(2) in astrocyte proliferation and in suggesting a potential avenue in the use of anti-inflammatory agents for the therapy of astroglioma.
Collapse
Affiliation(s)
- Dan Zhang
- Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Jenner P. Functional models of Parkinson's disease: a valuable tool in the development of novel therapies. Ann Neurol 2009; 64 Suppl 2:S16-29. [PMID: 19127585 DOI: 10.1002/ana.21489] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Functional models of Parkinson's disease (PD) have led to effective treatment for the motor symptoms. Toxin-based models, such as the 6-hydroxydopamine-lesioned rat and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated primate, have resulted in novel dopaminergic therapies and new therapeutic strategies. They have also been used to study processes underlying motor complications, particularly dyskinesia, and for developing pharmacological approaches to dyskinesia avoidance and suppression. Symptomatic models of PD based on nigrostriatal degeneration have a high degree of predictability of clinical effect of dopaminergic drugs on motor symptoms in humans. However, the effects of nondopaminergic drugs in these models do not translate effectively into clinical efficacy. Newer experimental models of PD have attempted to reproduce the pathogenic process and to involve all areas of the brain pathologically affected in humans. In addition, models showing progressive neuronal death have been sought but so far unsuccessfully. Pathogenic modeling has been attempted using a range of toxins, as well as through the use of transgenic models of gene defects in familial PD and mutant rodent strains. However, there are still no accepted progressive models of PD that mimic the processes known to occur during cell death and that result in the motor deficits, pathology, biochemistry, and drug responsiveness as seen in humans. Nevertheless, functional models of PD have led to many advances in treating the motor symptoms of the disorder, and we have been fortunate to have them available. They are an important reason the treatment of PD is so much better compared with treatments for related illnesses.
Collapse
Affiliation(s)
- Peter Jenner
- Neurodegenerative Disease Research Centre, School of Health and Biomedical Sciences, King's College, London, United Kingdom.
| |
Collapse
|
63
|
Ahmad I, Kumar A, Shukla S, Prasad Pandey H, Singh C. The involvement of nitric oxide in maneb- and paraquat-induced oxidative stress in rat polymorphonuclear leukocytes. Free Radic Res 2008; 42:849-62. [PMID: 18985485 DOI: 10.1080/10715760802513733] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oxidative stress plays a crucial role in the manifestations of maneb (MB) and paraquat (PQ)-induced toxicity including MB+PQ-induced Parkinson's disease (PD). Polymorphonuclear leukocytes (PMNs) actively participate in the oxidative stress-mediated inflammation and organ toxicity. The present study was undertaken to investigate the MB- and/or PQ-induced alterations in the indices of oxidative stress in rat PMNs. Animals were treated with or without MB and/or PQ in an exposure time dependent manner. In some sets of experiments, the animals were pre-treated with NOS inhibitors N(G)-nitro-L-arginine methyl ester (L-NAME) and aminoguanidine (AG) along with respective controls. A significant increase in myeloperoxidase (MPO), superoxide dismutase (SOD), nitric oxide, iNOS expression and lipid peroxidation (LPO) was observed in PMNs of MB- and/or PQ-treated animals, while catalase and glutathione S-transferase (GST) activities were attenuated. L-NAME and AG significantly reduced the augmented nitrite content, iNOS expression and MPO activity to control level in MB and PQ exposed animals. Although the augmented LPO was also reduced significantly in L-NAME and AG treated rat PMNs, the level was still higher as compared with controls. Alterations induced in SOD and GST activities were not affected by NOS inhibitors. The results thus suggest that MB and/or PQ induce iNOS-mediated nitric oxide production, which in turn increases MPO activity and lipid peroxidation, thereby oxidative stress.
Collapse
Affiliation(s)
- Israr Ahmad
- Indian Institute of Toxicology Research, Lucknow, India
| | | | | | | | | |
Collapse
|
64
|
Byler SL, Boehm GW, Karp JD, Kohman RA, Tarr AJ, Schallert T, Barth TM. Systemic lipopolysaccharide plus MPTP as a model of dopamine loss and gait instability in C57Bl/6J mice. Behav Brain Res 2008; 198:434-9. [PMID: 19070633 DOI: 10.1016/j.bbr.2008.11.027] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Revised: 11/08/2008] [Accepted: 11/19/2008] [Indexed: 12/20/2022]
Abstract
In most environmental models of Parkinson's disease (PD), a single neurodegenerative agent is introduced to cause nigrostriatal dopamine depletion. However, cell loss in human PD often might derive, at least in part, from multiple toxins or vulnerabilities, any one of which alone does not inevitably lead to chronic dopamine depletion. In the present research, male C57BL/6J mice were systemically administered the inflammatory bacterial endotoxin, lipopolysaccharide (LPS) and the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) alone or in combination and the behavior as well as striatal dopamine levels were compared to saline-treated mice. Mice in the combination (LPS+MPTP) group, but not in the single-factor groups, showed both dopamine depletion and parkinsonian symptoms, i.e., reduced stride length, at 4 months post-injection. MPTP alone acutely reduced striatal dopamine levels but this effect was transient as striatal dopamine recovered to normal levels after time (4 months). The LPS-only group showed no dopamine depletion or reduced stride length. These data are consistent with the view that nigrostriatal dopamine neurons might succumb after time to multiple toxic agents that independently may have only a transient, adverse effect.
Collapse
Affiliation(s)
- Stefanie L Byler
- Texas Christian University, Department of Psychology, Fort Worth, TX, USA.
| | | | | | | | | | | | | |
Collapse
|
65
|
Holden LJ, Coleman MD. Further preliminary assessment of three human glioma cell lines as models of human astrocytic toxicity in vitro. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2008; 26:290-296. [PMID: 21791377 DOI: 10.1016/j.etap.2008.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 05/22/2008] [Accepted: 05/29/2008] [Indexed: 05/31/2023]
Abstract
Three human astroglioma lines U251-MG, U373-MG and CCF-STTG1 have been evaluated further as possible models for astrocytotoxicity (GFAP and IL-6 release). The effects of bacterial lipopolysaccharide, chloroquine diphosphate and acrylamide were studied on GFAP expression and LPS, chloroquine diphosphate, ethanol, trimethyltin chloride (TMTC) and acrylamide were examined on interleukin-6 (IL-6) release in the U373-MG line only. At 4-h LPS elevated GFAP (17.0±5.0% P<0.05) above control in the U251-MG cell line only. Chloroquine diphosphate over 4h in the U251-MG line resulted in an increase in GFAP-IR to 20.3±4.2% and 21.1±4.1% above control levels 0.1μM (P<0.05) and 1μM (P<0.05) respectively. CQD was associated with decreases in MTT turnover, particularly after 24h incubation. With the U373-MG line, LPS (0.5μg/ml) increased IL-6 expression 640% above control (P<0.001), whilst chloroquine diphosphate (100μM), ethanol (10mM) and TMTC chloride (1μM) also increased IL-6. It is possible that batteries of astrocytic human glioma cell lines may be applicable to the sensitive evaluation of toxicants on astrogliotic expression markers such as GFAP and IL-6.
Collapse
Affiliation(s)
- Lindsay J Holden
- School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
| | | |
Collapse
|
66
|
Zhao F, Cai T, Liu M, Zheng G, Luo W, Chen J. Manganese induces dopaminergic neurodegeneration via microglial activation in a rat model of manganism. Toxicol Sci 2008; 107:156-64. [PMID: 18836210 DOI: 10.1093/toxsci/kfn213] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Manganese is an essential trace element required for normal development and bodily functions. However, exposure of the brain to excessive amounts of manganese results in neurotoxicity. Although previous studies examining manganese neurotoxicity have focused on neuronal injury, especially direct injury to dopaminergic neurons, the effects of manganese-induced neurotoxicity on glial cells have not been reported. The current study was designed to examine the effect of manganese on microglial activation, and the underlying mechanism of manganese-induced dopaminergic neuronal injury in vivo. We established an animal model of manganism by intrastriatal injection of MnCl(2).4H(2)O into male Sprague-Dawley rats. One day after administration of manganese, a few microglial cells in the substantia nigra (SN) were activated, although the number of tyrosine hydroxylase (TH)-immunoreactive neurons in the SN was unaffected. Seven days after administration of manganese, a marked reduction in the number of TH-immunoreactive neurons was observed in the SN, and the majority of microglial cells were activated. We found that manganese upregulated inducible nitric oxide synthase (iNOS) and tumor necrosis factor alpha (TNF-alpha) gene expression, as well as iNOS, TNF-alpha, and interleukin-1beta (IL-1beta) protein levels in the SN. Furthermore, treatment with minocycline, an inhibitor of microglial activation, attenuated microglial activation and mitigated IL-1beta, TNF-alpha, and iNOS production as well as dopaminergic neurotoxicity induced by manganese. These results suggested that dopaminergic neurons could be damaged by manganese neurotoxicity, and that the activated microglial cells and their associated activation products played an important role in this neurodegenerative process.
Collapse
Affiliation(s)
- Fang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | |
Collapse
|
67
|
Roy A, Jana A, Yatish K, Freidt MB, Fung YK, Martinson JA, Pahan K. Reactive oxygen species up-regulate CD11b in microglia via nitric oxide: Implications for neurodegenerative diseases. Free Radic Biol Med 2008; 45:686-99. [PMID: 18590811 PMCID: PMC2701551 DOI: 10.1016/j.freeradbiomed.2008.05.026] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 04/18/2008] [Accepted: 05/21/2008] [Indexed: 12/29/2022]
Abstract
Microglial activation is considered as a hallmark of several neurodegenerative disorders. During microglial activation, the expression of CD11b, the beta-integrin marker of microglia, is increased. However, the molecular mechanism behind increased microglial CD11b expression is poorly understood. The present study was undertaken to explore the role of reactive oxygen species (ROS) in the expression of CD11b in microglial cells. Bacterial lipopolysaccharide (LPS) stimulated the expression of CD11b in mouse BV-2 microglial cells and primary microglia, the effect that was blocked by antioxidants such as N-acetylcysteine (NAC) and pyrrolidine dithiocarbamate (PDTC). Furthermore, comicroinjection of either NAC or PDTC with LPS was also able to suppress LPS-stimulated expression of CD11b in striatum in vivo. Similarly, other neurotoxic molecules, such as interleukin-1beta (IL-1beta), IL-12 p40(2), fibrillar amyloid-beta (Abeta) peptides, HIV-1 gp120, and double-stranded RNA (poly(IC)), also stimulated the expression of CD11b in microglia through the involvement of ROS. Complete inhibition of LPS-stimulated expression of CD11b by catalase, induction of CD11b expression by H2O2 alone, and inhibition of superoxide-stimulated CD11b expression by catalase suggest that H2O2, but not superoxide, is in fact involved in the expression of CD11b. Interestingly, we also demonstrate that ROS stimulated the expression of CD11b after the induction of nitric oxide (NO) production and failed to stimulate CD11b when NO production was inhibited by either 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (carboxy-PTIO) or L-N6-(1-iminoethyl)-L-lysine (L-NIL). Taken together, these studies suggest that the up-regulation of CD11b in microglia is redox sensitive and that ROS up-regulates CD11b via NO.
Collapse
Affiliation(s)
- Avik Roy
- Department of Neurological Science, Rush University Medical Center, Chicago, IL 60612, USA
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
| | - Arundhati Jana
- Department of Neurological Science, Rush University Medical Center, Chicago, IL 60612, USA
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
| | - Kavitha Yatish
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
| | - Matthew B. Freidt
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
| | - Yiu K. Fung
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
| | - Jeffrey A. Martinson
- Department of Immunology and Microbiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Kalipada Pahan
- Department of Neurological Science, Rush University Medical Center, Chicago, IL 60612, USA
- Section of Neuroscience, Department of Oral Biology, University of Nebraska Medical Center College of Dentistry, Lincoln, NE 68583, USA
- Corresponding author. Department of Neurological Science, Rush University Medical Center, 1735 W Harrison Street, Chicago, IL 60612, USA. Fax: +1 312 563 3571. E-mail address: (K. Pahan)
| |
Collapse
|
68
|
Dutta G, Zhang P, Liu B. The lipopolysaccharide Parkinson's disease animal model: mechanistic studies and drug discovery. Fundam Clin Pharmacol 2008; 22:453-64. [PMID: 18710400 DOI: 10.1111/j.1472-8206.2008.00616.x] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Research in the last two decades has unveiled an important role for neuroinflammation in the degeneration of the nigrostriatal dopaminergic (DA) pathway that constitutes the pathological basis of the prevailing movement disorder, Parkinson's disease (PD). Neuroinflammation is characterized by the activation of brain glial cells, primarily microglia and astrocytes that release various soluble factors that include free radicals (reactive oxygen and nitrogen species), cytokines, and lipid metabolites. The majority of these glia-derived factors are proinflammatory and neurotoxic and are particularly deleterious to oxidative damage-vulnerable nigral DA neurons. As a proof of concept, various immunologic stimuli have been employed to directly induce glial activation to model DA neurodegeneration in PD. The bacterial endotoxin, lipopolysaccharide (LPS), has been the most extensively utilized glial activator for the induction of inflammatory DA neurodegeneration. In this review, we will summarize the various in vitro and in vivo LPS PD models. Furthermore, we will highlight the contribution of the LPS PD models to the mechanistic studies of PD pathogenesis and the search for neuroprotective agents for the treatment of PD.
Collapse
Affiliation(s)
- Garima Dutta
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | | | | |
Collapse
|
69
|
Armbrust E, Röhl C. Time- and activation-dependency of the protective effect of microglia on astrocytes exposed to peroxide-induced oxidative stress. Toxicol In Vitro 2008; 22:1399-404. [DOI: 10.1016/j.tiv.2008.02.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2007] [Revised: 02/08/2008] [Accepted: 02/11/2008] [Indexed: 10/22/2022]
|
70
|
Johnston LC, Su X, Maguire-Zeiss K, Horovitz K, Ankoudinova I, Guschin D, Hadaczek P, Federoff HJ, Bankiewicz K, Forsayeth J. Human interleukin-10 gene transfer is protective in a rat model of Parkinson's disease. Mol Ther 2008; 16:1392-9. [PMID: 18545225 PMCID: PMC2725180 DOI: 10.1038/mt.2008.113] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In Parkinson's disease (PD) chronic inflammation occurs in the substantia nigra (SNc) concurrently with dopaminergic neurodegeneration. In models of PD, microglial activation precedes neurodegeneration in the SNc, suggesting that the underlying pathogenesis involves a complex response in the nigrostriatal pathway, and that the innate immune system plays a significant role. We have investigated the neuroprotective effect of an adeno-associated viral type-2 (AAV2) vector containing the complementary DNA (cDNA) for human interleukin-10 (hIL-10) in the unilateral 6-hydroxydopamine (6-OHDA) rat model of PD. AAV2-hIL-10 reduced the 6-OHDA-induced loss of tyrosine hydroxylase (TH)-positive neurons in the SNc, and also reduced loss of striatal dopamine (DA). Pretreatment with AAV2-hIL-10 reduced glial activation in the SNc but did not attenuate striatal release of the inflammatory cytokine IL-1beta. Assessment of rotational behavior in response to apomorphine challenge showed absence of asymmetry, confirming protection of dopaminergic innervation of the lesioned striatum. At baseline, 6-OHDA-lesioned animals displayed a deficit in contralateral forelimb use, but pretreatment with AAV2-hIL-10 reduced this forelimb akinesia. Transcriptional analyses revealed alteration of a few genes by AAV2-hIL-10; these alterations may contribute to neuroprotection. This study supports the need for further investigations relating to gene therapies aimed at reducing neuroinflammation in early PD.
Collapse
Affiliation(s)
- Louisa C Johnston
- Department of Neurosurgery, University of California San Francisco, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Iravani MM, Sadeghian M, Leung CCM, Tel BC, Rose S, Schapira AH, Jenner P. Continuous subcutaneous infusion of pramipexole protects against lipopolysaccharide-induced dopaminergic cell death without affecting the inflammatory response. Exp Neurol 2008; 212:522-31. [PMID: 18571649 DOI: 10.1016/j.expneurol.2008.04.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2008] [Revised: 04/02/2008] [Accepted: 04/30/2008] [Indexed: 12/27/2022]
Abstract
The D2/D3 dopamine receptor agonist pramipexole, protects against toxin-induced dopaminergic neuronal destruction but its mechanism of action is unknown. Inflammation following glial cell activation contributes to cell death in Parkinson's disease and we now report on the effects of acute or chronic administration of pramipexole on lipopolysaccharide (LPS) induced inflammation and nigral dopaminergic cell death in the rat. At 48 h and 30 days following supranigral administration of LPS, approximately 70% of tyrosine hydroxylase (TH) immunoreactive (-ir) cells in substantia nigra had degenerated with a corresponding loss of TH-ir terminals in the striatum. In rats acutely treated with pramipexole (2x1 mg/kg; s.c.) 48 h following LPS application, there was no difference in the number of TH-ir cells or terminals compared to LPS-treated rats receiving vehicle. However, the continuous subcutaneous infusion of pramipexole for 7 days prior to LPS and 21 days subsequently, produced a marked preservation of both TH-ir cells and terminals. At 48 h or 30 days, LPS induced an up-regulation of ubiquitin-ir within the nigral TH-ir neurones, which was reduced by pramipexole treatment. Thirty days following supranigral LPS administration (9 days after the end of infusion), (+)-amphetamine (5 mg/kg, i.p.) caused robust ipsiversive rotation. In rats treated with LPS but receiving continuous subcutaneous administration of pramipexole, (+)-amphetamine-induced rotation was markedly reduced. LPS-induced increase in the levels of inflammatory markers, were not affected by either acute administration or continuous infusion of pramipexole. Continuous infusion of pramipexole protected dopaminergic neurones against inflammation induced degeneration but without modification of the inflammatory response.
Collapse
Affiliation(s)
- Mahmoud M Iravani
- Neurodegenerative Disease Research Centre, School of Health and Biomedical Sciences, King's College, London SE1 1UL, UK
| | | | | | | | | | | | | |
Collapse
|
72
|
Meredith GE, Sonsalla PK, Chesselet MF. Animal models of Parkinson's disease progression. Acta Neuropathol 2008; 115:385-98. [PMID: 18273623 DOI: 10.1007/s00401-008-0350-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Revised: 01/30/2008] [Accepted: 01/31/2008] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder whose etiology is not understood. This disease occurs both sporadically and through inheritance of single genes, although the familial types are rare. Over the past decade or so, experimental and clinical data suggest that PD could be a multifactorial, neurodegenerative disease that involves strong interactions between the environment and genetic predisposition. Our understanding of the pathophysiology and motor deficits of the disease relies heavily on fundamental research on animal models and the last few years have seen an explosion of toxin-, inflammation-induced and genetically manipulated models. The insight gained from the use of such models has strongly advanced our understanding of the progression and stages of the disease. The models have also aided the development of novel therapies to improve symptomatic management, and they are critical for the development of neuroprotective strategies. This review critically evaluates these in vivo models and the roles they play in mimicking the progression of PD.
Collapse
Affiliation(s)
- Gloria E Meredith
- Department of Cellular and Molecular Pharmacology, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road, North Chicago, IL 60064, USA.
| | | | | |
Collapse
|
73
|
Miller RL, James-Kracke M, Sun GY, Sun AY. Oxidative and Inflammatory Pathways in Parkinson’s Disease. Neurochem Res 2008; 34:55-65. [DOI: 10.1007/s11064-008-9656-2] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2007] [Accepted: 03/07/2008] [Indexed: 10/22/2022]
|
74
|
Sawada H, Hishida R, Hirata Y, Ono K, Suzuki H, Muramatsu SI, Nakano I, Nagatsu T, Sawada M. Activated microglia affect the nigro-striatal dopamine neurons differently in neonatal and aged mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. J Neurosci Res 2007; 85:1752-61. [PMID: 17469135 DOI: 10.1002/jnr.21241] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Microglia play an important role in the inflammatory process that occurs in Parkinson's disease (PD). Activated microglia produce cytokines and neurotrophins and may have neurotoxic or neurotrophic effects. Because microglia are most proliferative and easily activated during the neonatal period, we examined the effects of neonatal microglia activated with lipopolysaccharide (LPS) on the nigro-striatal dopamine neurons in mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), in comparison with activated microglia from the aged mice. By MPTP administration to neonatal mice, the number of dopamine neurons in the substantia nigra (SN) was decreased significantly, whereas that in the mice treated with LPS and MPTP was recovered to normal, along with significant microglial activation. Tyrosine hydroxylase (TH) activity, the levels of dopamine and 3,4-dihydroxyphenylacetic acid (DOPAC), and the levels of pro-inflammatory cytokines IL-1beta and IL-6 in the midbrain were elevated in the neonates treated with LPS and MPTP. On the contrary, although the number of dopamine neurons in the 60-week-old mice treated with MPTP was also decreased significantly, the microglial activation by LPS treatment caused a further decrease in their number. These results suggest that the activated microglia in neonatal mice are different from those in aged mice, with the former having neurotrophic potential toward the dopamine neurons in the SN, in contrast to the neurotoxic effect of the latter.
Collapse
Affiliation(s)
- Hirohide Sawada
- School of Medicine, Fujita Health University, Toyoake, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Hamby ME, Gragnolati AR, Hewett SJ, Hewett JA. TGF beta 1 and TNF alpha potentiate nitric oxide production in astrocyte cultures by recruiting distinct subpopulations of cells to express NOS-2. Neurochem Int 2007; 52:962-71. [PMID: 18035449 DOI: 10.1016/j.neuint.2007.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 09/26/2007] [Accepted: 10/09/2007] [Indexed: 11/25/2022]
Abstract
Nitric oxide (NO) synthase-2 (NOS-2), a key source of NO at sites of neuroinflammation, is induced in astrocyte cultures treated with lipopolysaccharide (LPS) plus interferon-gamma (IFN gamma). A recent study examining the regulation of astrocytic NOS-2 expression demonstrated that transforming growth factor-beta1 (TGF beta 1) potentiated LPS plus IFN gamma-induced NOS-2 expression via expansion of the pool of astrocytes that express NOS-2. Results in the current report indicate that this population-based mechanism of increasing NOS-2 expression is not restricted to TGF beta 1, since it also accounts for the potentiation of NO production in astrocyte cultures by tumor necrosis factor-alpha (TNFalpha). In contrast to TGF beta 1, which required 24h preincubation for optimal potentiation of NO production, TNF alpha was maximally effective when added concurrently with LPS plus IFN gamma. Nevertheless, under conditions that optimally potentiated NO production, both cytokines recruited similar numbers of astrocytes to express NOS-2 (% NOS-2-positive cells after LPS plus IFN gamma alone or with TNFalpha or TGF beta 1 was 9.5+/-1.2, 25.3+/-2.9, and 32.4+/-3.0, respectively). Interestingly, stimulation of astrocytes in the presence of both TGF beta 1 and TNFalpha additively increased the number of astrocytes that expressed NOS-2 protein (% NOS-2-positive cells was 61.0+/-4.2) relative to each cytokine alone. Potentiation of NO production by either TNF alpha or TGF beta 1 was not ablated by neutralizing antibodies to TGF beta 1 or TNFalpha, respectively. Thus, the two cytokines act independently to recruit separate pools of astrocytes to express NOS-2. These results are consistent with the notion that astrocytes possess an innate heterogeneity with respect to responsiveness to these cytokines.
Collapse
Affiliation(s)
- Mary E Hamby
- Department of Neuroscience MC 3401, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030-3401, USA
| | | | | | | |
Collapse
|
76
|
Tripanichkul W, Sripanichkulchai K, Duce JA, Finkelstein DI. 17β-Estradiol reduces nitrotyrosine immunoreactivity and increases SOD1 and SOD2 immunoreactivity in nigral neurons in male mice following MPTP insult. Brain Res 2007; 1164:24-31. [PMID: 17640623 DOI: 10.1016/j.brainres.2007.05.076] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 05/15/2007] [Accepted: 05/21/2007] [Indexed: 10/23/2022]
Abstract
Emerging evidence suggests the beneficial effects of estrogen on Parkinson's disease (PD), yet the mechanisms of action implicated remain elusive. While experimental evidence suggests that estrogen possesses potent antioxidative properties, it is still unknown whether the hormone exhibits a neuroprotection in a PD animal model through its antioxidant activities. This study therefore investigated the effects of 17beta-estradiol (E2) on the immunoreactivity of nigral neurons and glia for nitrotyrosine (NT, a stable marker for oxidative stress), Cu/Zn superoxide dismutase (SOD1) and Mn superoxide dismutase (SOD2) in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model. Adult male mice were treated with E2 or vehicle for 11 days during which they were injected with MPTP or saline on the sixth day. The brains were collected on day 11 and quantitative immunohistochemistry was used to assess the number of NT-, SOD1- and SOD2-immunoreactive (IR) cells in the substantia nigra pars compacta (SNpc). In saline-treated group, E2 decreased NT-IR neuronal number and raised SOD1 and SOD2 expression in neurons and glia in the SNpc. MPTP induced a significant increase in the number of NT- and SOD2-IR neurons, but decreased the number of SOD1-IR neurons. MPTP also triggered a significant increase of SOD2- and SOD1-IR glial number. E2 pretreatment in MPTP mice reduced the number of NT-IR neurons, increased the number of SOD1- and SOD2-IR neurons, but did not alter the MPTP effect on glia immunoreactive to either SOD. Stimulation of SOD1 and SOD2 expression in nigral neurons suggests that E2 provides neuroprotection against MPTP-induced oxidative stress, partly through its ability to act as an antioxidant.
Collapse
Affiliation(s)
- Wanida Tripanichkul
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Bangkok, 10110, Thailand
| | | | | | | |
Collapse
|
77
|
Lambeth JD. Nox enzymes, ROS, and chronic disease: an example of antagonistic pleiotropy. Free Radic Biol Med 2007; 43:332-47. [PMID: 17602948 PMCID: PMC2013737 DOI: 10.1016/j.freeradbiomed.2007.03.027] [Citation(s) in RCA: 478] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2007] [Revised: 03/27/2007] [Accepted: 03/28/2007] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are considered to be chemically reactive with and damaging to biomolecules including DNA, protein, and lipid, and excessive exposure to ROS induces oxidative stress and causes genetic mutations. However, the recently described family of Nox and Duox enzymes generates ROS in a variety of tissues as part of normal physiological functions, which include innate immunity, signal transduction, and biochemical reactions, e.g., to produce thyroid hormone. Nature's "choice" of ROS to carry out these biological functions seems odd indeed, given its predisposition to cause molecular damage. This review describes normal biological roles of Nox enzymes as well as pathological conditions that are associated with ROS production by Nox enzymes. By far the most common conditions associated with Nox-derived ROS are chronic diseases that tend to appear late in life, including atherosclerosis, hypertension, diabetic nephropathy, lung fibrosis, cancer, Alzheimer's disease, and others. In almost all cases, with the exception of a few rare inherited conditions (e.g., related to innate immunity, gravity perception, and hypothyroidism), diseases are associated with overproduction of ROS by Nox enzymes; this results in oxidative stress that damages tissues over time. I propose that these pathological roles of Nox enzymes can be understood in terms of antagonistic pleiotropy: genes that confer a reproductive advantage early in life can have harmful effects late in life. Such genes are retained during evolution despite their harmful effects, because the force of natural selection declines with advanced age. This review discusses some of the proposed physiologic roles of Nox enzymes, and emphasizes the role of Nox enzymes in disease and the likely beneficial effects of drugs that target Nox enzymes, particularly in chronic diseases associated with an aging population.
Collapse
Affiliation(s)
- J David Lambeth
- 148 Whitehead Biomedical Research Building, Department of Pathology and Laboratory Medicine, 615 Michael Street, Atlanta, GA 30322, USA.
| |
Collapse
|
78
|
Helkamaa T, Reenilä I, Tuominen RK, Soinila S, Väänänen A, Tilgmann C, Rauhala P. Increased catechol-O-methyltransferase activity and protein expression in OX-42-positive cells in the substantia nigra after lipopolysaccharide microinfusion. Neurochem Int 2007; 51:412-23. [PMID: 17573159 DOI: 10.1016/j.neuint.2007.04.020] [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: 03/23/2006] [Revised: 04/20/2007] [Accepted: 04/20/2007] [Indexed: 11/22/2022]
Abstract
Activated microglial cells are found in the substantia nigra and the striatum of Parkinson's disease patients. These cells have been shown to express catechol-O-methyltransferase activity which may increase during pathological conditions. Lipopolysaccharides are potent activators of microglial cells. After paranigral lipopolysaccharide infusion to rats we observed intense microglial activation around the lesion area followed by a delayed injury in nigrostriatal pathway in 2 weeks. Simultaneously, catechol-O-methyltransferase activity in the substantia nigra was gradually increased up to 213%. In the Western blot the amount of soluble COMT and membrane bound COMT proteins were increased by 255% and 86%, respectively. Increased catechol-O-methyltransferase immunoreactivity was located primarily into the activated microglial cells in the lesion area. Interestingly, catechol-O-methyltransferase and OX-42 stained also intensively microglia/macrophage-like cells which surrounded the adjacent blood vessels. Inhibition of catechol-O-methyltransferase activity by tolcapone or entacapone did not increase lipopolysaccharide-induced neurotoxicity. We conclude that catechol-O-methyltransferase activity and protein expression were increased in the substantia nigra after inflammation induced by lipopolysaccharides. These changes in glial and perivascular catechol-O-methyltransferase activity may have clinical relevance for Parkinson's disease drug treatment due to increased metabolism of levodopa in the brain.
Collapse
Affiliation(s)
- Teemu Helkamaa
- Institute of Biomedicine, Pharmacology, University of Helsinki, Finland.
| | | | | | | | | | | | | |
Collapse
|
79
|
Choi HB, Ryu JK, Kim SU, McLarnon JG. Modulation of the purinergic P2X7 receptor attenuates lipopolysaccharide-mediated microglial activation and neuronal damage in inflamed brain. J Neurosci 2007; 27:4957-68. [PMID: 17475804 PMCID: PMC6672082 DOI: 10.1523/jneurosci.5417-06.2007] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 03/13/2007] [Accepted: 03/28/2007] [Indexed: 12/30/2022] Open
Abstract
We investigated the involvement and roles of the ionotropic purinergic receptor P2X(7)R in microglia in mediating lipopolysaccharide (LPS)-induced inflammatory responses and neuronal damage in rat striatum. A detailed in vivo study showed that LPS injection into striatum markedly increased the expression of P2X(7)R in microglia compared with control (saline)-injected animals. Additionally, LPS injection upregulated a broad spectrum of proinflammatory mediators, including inducible nitric oxide synthase (nitric oxide production marker), 3-nitrotyrosine (peroxynitrite-mediated nitration marker), 4-hydroxynonenal (lipid peroxidation marker), and 8-hydroxy-2'-deoxyguanosine (oxidative DNA damage marker), and reduced neuronal viability. The P2X(7)R antagonist oxidized ATP (oxATP) was effective in attenuating expressions of all inflammatory mediators and in addition inhibited LPS-induced activation of the cellular signaling factors p38 mitogen-activated protein kinase and transcriptional factor nuclear factor kappaB. Most importantly, in vivo, oxATP blockade of P2X(7)R also reduced numbers of caspase-3-positive neurons and increased neuronal survival in LPS-injected brain. In vitro, LPS stimulation of cultured human microglia enhanced cellular expressions of a host of proinflammatory factors, including cyclooxygenase-2, interleukin-1beta (IL-1beta), IL-6, IL-12, and tumor necrosis factor-alpha; all factors were inhibited by oxATP. A novel finding was that LPS potentiated intracellular [Ca(2+)](i) mobilization induced by the P2X(7)R ligand 2',3'-O-(4-benzoyl-benzoyl) ATP, which could serve as a mechanistic link for P2X(7)R amplification of inflammatory responses. Our results suggest critical roles for P2X(7)R in mediating inflammation and inhibition of this subtype purinergic receptor as a novel therapeutic approach to reduce microglial activation and confer neuroprotection in inflamed and diseased brain.
Collapse
Affiliation(s)
- Hyun B. Choi
- Department of Anesthesiology, Pharmacology, and Therapeutics and
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3, and
| | - Jae K. Ryu
- Department of Anesthesiology, Pharmacology, and Therapeutics and
| | - Seung U. Kim
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3, and
- Brain Disease Research Center, Ajou University, Suwon, Korea 443-749
| | | |
Collapse
|
80
|
Tian DS, Dong Q, Pan DJ, He Y, Yu ZY, Xie MJ, Wang W. Attenuation of astrogliosis by suppressing of microglial proliferation with the cell cycle inhibitor olomoucine in rat spinal cord injury model. Brain Res 2007; 1154:206-14. [PMID: 17482149 DOI: 10.1016/j.brainres.2007.04.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2007] [Revised: 04/02/2007] [Accepted: 04/02/2007] [Indexed: 11/29/2022]
Abstract
Microglial activation/proliferation and reactive astrogliosis are commonly observed and have been considered to be closely relevant pathological processes during spinal cord injury (SCI). However, the molecular mechanisms underlying this microglial-astroglial interaction are still poorly understood. We showed recently that the continuous injection of the cell cycle inhibitor olomoucine not only markedly suppressed microglial proliferation and associated release of pro-inflammatory cytokines, but also attenuated astroglial scar formation and the lesion cavity and mitigated the functional deficits in rat SCI animal model. In this study, we asked whether microglial activation/proliferation plays an initial role and also necessary in maintaining astrogliosis in SCI model. Our results showed that traumatic induced microglial activation/proliferation precedes astrogliosis, and the up-regulated GFAP expression at both mRNA and protein levels was temporally posterior to the microglial activation. Furthermore, when the cell cycle inhibitor olomoucine was administered only once 1 h post-SCI that should selectively suppress microglial proliferation, the subsequent SCI induced increase in GFAP expression at 1, 2 and 4 weeks was significantly attenuated, suggesting that microglial activation/proliferation played an important role for the later onset astrogliosis after SCI. Consistent with the results that microglial proliferation always precedes astroglial proliferation and there is at present no evidence of other astroglial precursors, which as always does not mean that they will not be uncovered by further searching, and in view of the fact that microglial-derived pro-inflammatory cytokines promote astrogliosis as we reported recently, these findings together suggest that by release of cytokines and other soluble products, the early onset microglial activation/proliferation can significantly influence the subsequent development of reactive astrogliosis and glial scar formation in SCI animal model.
Collapse
Affiliation(s)
- Dai-shi Tian
- Department of Neurology, Affiliated Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, PR China
| | | | | | | | | | | | | |
Collapse
|
81
|
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: 471] [Impact Index Per Article: 27.7] [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
|
82
|
Ohtaki N, Kamitani W, Watanabe Y, Hayashi Y, Yanai H, Ikuta K, Tomonaga K. Downregulation of an astrocyte-derived inflammatory protein, S100B, reduces vascular inflammatory responses in brains persistently infected with Borna disease virus. J Virol 2007; 81:5940-8. [PMID: 17376896 PMCID: PMC1900267 DOI: 10.1128/jvi.02137-06] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Borna disease virus (BDV) is a neurotropic virus that causes a persistent infection in the central nervous system (CNS) of many vertebrate species. Although a severe reactive gliosis is observed in experimentally BDV-infected rat brains, little is known about the glial reactions contributing to the viral persistence and immune modulation in the CNS. In this regard, we examined the expression of an astrocyte-derived factor, S100B, in the brains of Lewis rats persistently infected with BDV. S100B is a Ca(2+)-binding protein produced mainly by astrocytes. A prominent role of this protein appears to be the promotion of vascular inflammatory responses through interaction with the receptor for advanced glycation end products (RAGE). Here we show that the expression of S100B is significantly reduced in BDV-infected brains despite severe astrocytosis with increased glial fibrillary acidic protein immunoreactivity. Interestingly, no upregulation of the expression of S100B, or RAGE, was observed in the persistently infected brains even when incited with several inflammatory stimuli, including lipopolysaccharide. In addition, expression of the vascular cell adhesion molecule 1 (VCAM-1), as well as the infiltration of encephalitogenic T cells, was significantly reduced in persistently infected brains in which an experimental autoimmune encephalomyelitis was induced by immunization with myelin-basic protein. Furthermore, we demonstrated that the continuous activation of S100B in the brain may be necessary for the progression of vascular immune responses in neonatally infected rat brains. Our results suggested that BDV infection may impair astrocyte functions via a downregulation of S100B expression, leading to the maintenance of a persistent infection.
Collapse
Affiliation(s)
- Naohiro Ohtaki
- Department of Virology, Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
83
|
Burgos M, Calvo S, Molina F, Vaquero CF, Samarel A, Llopis J, Tranque P. PKCε induces astrocyte stellation by modulating multiple cytoskeletal proteins and interacting with Rho A signalling pathways: implications for neuroinflammation. Eur J Neurosci 2007; 25:1069-78. [PMID: 17331203 DOI: 10.1111/j.1460-9568.2007.05364.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the importance of stellation to maintain astrocyte functionality, the intracellular signals controlling morphology in these cells are poorly characterized. Our goal was to examine the implication of protein kinase C epsilon (PKCepsilon) in astrocyte stellation. We found that the morphological transformation of astrocytes induced by exposure to the pro-inflammatory agent lipopolysaccharide is enhanced by adenoviral expression of wild-type PKCepsilon, and that activation of PKCepsilon is sufficient to trigger a dramatic stellation. Such an effect is mediated by the rearrangement of microtubules and filaments of glial fibrillary acidic protein, disorganization of stress fibres, and formation of new actin filaments within growing cellular processes. Furthermore, PKCepsilon regulates actin-interacting elements such as non-muscle myosin and proteins of the ezrin/radixin/moesin family. We also observed that at least part of the actions of PKCepsilon depend on its catalytic activity. Finally, stellation by PKCepsilon could be blocked by the expression of a constitutively active form of Rho A implicated in the stability of the flat astrocytic morphology. In summary, PKCepsilon stands out as a key intracellular regulator of morphological plasticity in astrocytes, affecting a large range of cytoskeletal elements and inactivating Rho A-dependent pathways. These morphological effects of PKCepsilon may play essential roles during the course of neuroinflammation.
Collapse
Affiliation(s)
- Miguel Burgos
- Medical School and Regional Center for Biomedical Research (CRIB), Castilla-La Mancha University, C/Almansa 14, Albacete 02006, Spain
| | | | | | | | | | | | | |
Collapse
|
84
|
Zhu Y, Carvey PM, Ling Z. Altered glutathione homeostasis in animals prenatally exposed to lipopolysaccharide. Neurochem Int 2007; 50:671-80. [PMID: 17291629 PMCID: PMC1868495 DOI: 10.1016/j.neuint.2006.12.013] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 12/07/2006] [Accepted: 12/20/2006] [Indexed: 11/23/2022]
Abstract
We previously reported that injection of bacterial lipopolysaccharide (LPS) into gravid female rats at embryonic day 10.5 resulted in a birth of offspring with fewer than normal dopamine (DA) neurons along with innate immunity dysfunction and many characteristics seen in Parkinson's disease (PD) patients. The LPS-exposed animals were also more susceptible to secondary toxin exposure as indicated by an accelerated DA neuron loss. Glutathione (GSH) is an important antioxidant in the brain. A disturbance in glutathione homeostasis has been proposed for the pathogenesis of PD. In this study, animals prenatally exposed to LPS were studied along with an acute intranigral LPS injection model for the status of glutathione homeostasis, lipid peroxidation, and related enzyme activities. Both prenatal LPS exposure and acute LPS injection produced a significant GSH reduction and increase in oxidized GSH (GSSG) and lipid peroxide (LPO) production. Activity of gamma-glutamylcysteine synthetase (GCS), the rate-limiting enzyme in de novo GSH synthesis, was up-regulated in acute supranigral LPS model but was reduced in the prenatal LPS model. The GCS light subunit protein expression was also down-regulated in prenatal LPS model. GSH redox recycling enzyme activities (glutathione peroxidase, GPx and glutathione reducdase, GR) and glutathione-S-transferase (GST), gamma-glutamyl transpeptidase (gamma-GT) activities were all increased in prenatal LPS model. Prenatal LPS exposure and aging synergized in GSH level and GSH-related enzyme activities except for those (GR, GST, and gamma-GT) with significant regional variations. Additionally, prenatal LPS exposure produced a reduction of DA neuron count in the substantia nigra (SN). These results suggest that prenatal LPS exposure may cause glutathione homeostasis disturbance in offspring brain and render DA neurons susceptible to the secondary neurotoxin insult.
Collapse
Affiliation(s)
- Yuangui Zhu
- Department of Pharmacology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
- Fujian Institute of Geriatrics, Union Hospital, Fujian Medical University, Fuzhou, Fujian 350001, China
| | - Paul M. Carvey
- Department of Pharmacology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
| | - Zaodung Ling
- Department of Pharmacology, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL 60612, USA
- Division of Mental Health and Substance Abuse Research, National Health Research Institutes, Taiwan
- *Corresponding author: Zaodung Ling, M.D. Division of Mental Health and Substance Abuse Research, National Health Research Institutes, Taiwan, 35 Keyan Road, Zhunan, Miaoli County, Taiwan 350, Tel: 011-886-37-246-166 ext 36717, Fax: 011-886-37-586-453, E-mail:
| |
Collapse
|
85
|
Röhl C, Lucius R, Sievers J. The effect of activated microglia on astrogliosis parameters in astrocyte cultures. Brain Res 2006; 1129:43-52. [PMID: 17169340 DOI: 10.1016/j.brainres.2006.10.057] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 09/13/2006] [Accepted: 10/27/2006] [Indexed: 10/23/2022]
Abstract
In the diseased central nervous system, astrogliosis is accompanied by microglial activation. Depending on the context of their activation, reactive astrocytes are involved in neuronal survival and regeneration in an either protective or impedimental way. Major reactive changes of astrocytes in vivo are the upregulation of the intermediate filaments GFAP (glial fibrillary acidic protein) and vimentin with accompanying cellular hypertrophy and/or hyperplasia. To examine the involvement of activated microglia in the onset and maintenance of astrogliosis, we used an in vitro model of purified cultures of astrocytes and assessed as parameters for astrogliosis GFAP, vimentin, astroglial hypertrophy and cell growth after treatment with medium conditioned by LPS (lipopolysaccarides)-stimulated microglia. Furthermore, IL-6 as a typically upregulated cytokine in proinflammatory processes in the brain was determined in treated astrocytes. GFAP, the classical marker for astrogliosis, was downregulated on its protein and in parallel with vimentin on its mRNA level. The expression of actin, another cytoskeleton protein used as control, remained unchanged. Ultrastructural studies of astroglial intermediate filaments supported these findings. No hypertrophy was found. Nevertheless, LPS-activated microglia stimulated astrocytes as demonstrated by an increased cell number and an enhanced mRNA expression of IL-6. Resting microglia did not change any of the determined parameters. Our results suggest that the role of activated microglia in astrogliotic processes following injury of the brain has to be reevaluated, as microglia in their activated state might support the onset of astrogliosis on the one hand, but might delay or reduce subsequent glial scar formation on the other hand.
Collapse
Affiliation(s)
- Claudia Röhl
- Department of Anatomy, University of Kiel, Olshausenstr. 40, D-24098 Kiel, Germany.
| | | | | |
Collapse
|
86
|
Croisier E, Graeber MB. Glial degeneration and reactive gliosis in alpha-synucleinopathies: the emerging concept of primary gliodegeneration. Acta Neuropathol 2006; 112:517-30. [PMID: 16896905 DOI: 10.1007/s00401-006-0119-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2006] [Revised: 07/04/2006] [Accepted: 07/04/2006] [Indexed: 01/06/2023]
Abstract
The concept of gliodegenerative diseases has not been widely established although there is accumulating evidence that glial cells may represent a primary target of degenerative disease processes. In the central nervous system (CNS), examples that provide a "proof of concept" include at least one alpha-synucleinopathy, multiple system atrophy (MSA), but this disease is conventionally discussed under the heading of "neurodegeneration". Additional evidence in support of primary glial affection has been reported in neurodegenerative disorders such as Parkinson's disease, Alzheimer's disease and transmissible spongiform encephalopathies. Based on biochemical, genetic and transcriptomic studies it is also becoming increasingly clear that the molecular changes measured in whole tissue extracts, e.g. obtained from Parkinson's disease brain, are not based on a purely neuronal contribution. This important evidence has been missed in cell culture or laser capture work focusing on the neuronal cell population. Studies of animal and in vitro models of disease pathogenesis additionally suggest glial accountability for some CNS degenerative processes. This review provides a critical analysis of the evidence available to date in support of the concept of gliodegeneration, which we propose to represent an essential although largely disregarded component of the spectrum of classical "neurodegeneration". Examples from the spectrum of alpha-synucleinopathies are presented.
Collapse
Affiliation(s)
- Emilie Croisier
- University Department of Neuropathology, Imperial College London and Hammersmith Hospitals Trust, Charing Cross Campus, Fulham Palace Road, London, UK
| | | |
Collapse
|
87
|
Armentero MT, Levandis G, Nappi G, Bazzini E, Blandini F. Peripheral inflammation and neuroprotection: systemic pretreatment with complete Freund's adjuvant reduces 6-hydroxydopamine toxicity in a rodent model of Parkinson's disease. Neurobiol Dis 2006; 24:492-505. [PMID: 17023164 DOI: 10.1016/j.nbd.2006.08.016] [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] [Received: 01/20/2006] [Revised: 07/29/2006] [Accepted: 08/14/2006] [Indexed: 12/22/2022] Open
Abstract
Complete Freund's adjuvant (CFA), a pro-inflammatory agent, was inoculated, subcutaneously, to Sprague-Dawley rats prior to the intrastriatal injection of 6-hydroxydopamine (6-OHDA). Animals were sacrificed 7 and 28 days following 6-OHDA injection; neuronal damage, glial activation and cytokine levels, within the nigrostriatal system, were then investigated. Nigrostriatal degeneration induced by 6-OHDA was accompanied by early microglial and astroglial activation, which preceded the onset of dopaminergic cell loss, in the SNc, without significant changes in cytokine levels. CFA pretreatment markedly reduced the SNc neuronal loss and associated microglial activation, as well as the rotational response to apomorphine. These changes were associated with moderate, transient increases in the nigrostriatal levels of glial-cell-derived neurotrophic factor (GDNF) and pro-inflammatory cytokines, including interleukin (IL)-1alpha, IL-1beta and IL-6. Our results show that prior delivery of a peripheral, pro-inflammatory stimulus induces neuroprotection, in a rodent model of Parkinson's disease, possibly through the modulation of cytokine production at the nigrostriatal level.
Collapse
Affiliation(s)
- Marie-Thérèse Armentero
- Laboratory of Functional Neurochemistry, Neurological Institute C. Mondino, Via Mondino, 2 27100 Pavia, Italy
| | | | | | | | | |
Collapse
|
88
|
Liu B. Modulation of microglial pro-inflammatory and neurotoxic activity for the treatment of Parkinson's disease. AAPS JOURNAL 2006; 8:E606-21. [PMID: 17025278 PMCID: PMC2668934 DOI: 10.1208/aapsj080369] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a debilitating movement disorder resulting from a progressive degeneration of the nigrostriatal dopaminergic pathway and depletion of neurotransmitter dopamine in the striatum. Molecular cloning studies have identified nearly a dozen genes or loci that are associated with small clusters of mostly early onset and genetic forms of PD. The etiology of the vast majority of PD cases remains unknown, and the precise molecular and biochemical processes governing the selective and progressive degeneration of the nigrostriatal dopaminergic pathway are poorly understood. Current drug therapies for PD are symptomatic and appear to bear little effect on the progressive neurodegenerative process. Studies of postmortem PD brains and various cellular and animal models of PD in the last 2 decades strongly suggest that the generation of pro-inflammatory and neurotoxic factors by the resident brain immune cells, microglia, plays a prominent role in mediating the progressive neurodegenerative process. This review discusses literature supporting the possibility of modulating the activity of microglia as a neuroprotective strategy for the treatment of PD.
Collapse
Affiliation(s)
- Bin Liu
- Department of Pharmacodynamics, College of Pharmacy, the McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
89
|
Ruano D, Revilla E, Gavilán MP, Vizuete ML, Pintado C, Vitorica J, Castaño A. Role of p38 and inducible nitric oxide synthase in the in vivo dopaminergic cells' degeneration induced by inflammatory processes after lipopolysaccharide injection. Neuroscience 2006; 140:1157-68. [PMID: 16713109 DOI: 10.1016/j.neuroscience.2006.02.073] [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: 10/13/2005] [Revised: 02/24/2006] [Accepted: 02/25/2006] [Indexed: 01/28/2023]
Abstract
Accumulating evidences suggest that neuroinflammation is involved in the progressive death of dopaminergic neurons in Parkinson's disease. Several studies have shown that intranigral injection of lipopolysaccharide induces inflammation in the substantia nigra leading to death of tyrosine hydroxylase-positive cells. To better understand how the inflammatory response gives rise to neurotoxicity we induced inflammation in substantia nigra by injecting lipopolysaccharide. The damage of substantia nigra dopaminergic neurons was evaluated by immunohistochemistry, reverse transcription-PCR and Western blot analysis of tyrosine hydroxylase. In parallel, activation of microglial cells, a hallmark of inflammation in CNS, was revealed by immunohistochemistry. Similarly the expression of molecules involved in the inflammatory response and apoptotic pathway was also tested, such as cytokines (tumor necrosis factor-alpha, interleukin-1beta, interleukin-6), inducible nitric oxide synthase and caspase-11. Tyrosine hydroxylase expression (both mRNA and protein) started to decrease around 3 days post-injection. At the mRNA level, our results showed that the cytokines expression peaked shortly (3-6 h) after lipopolysaccharide injection, followed by the induction of inducible nitric oxide synthase and caspase-11 (14 h). However, inducible nitric oxide synthase protein peaked at 24 h and lasted for 14 days. The lipopolysaccharide-induced loss of substantia nigra dopaminergic neurons was partially inhibited by co-injection of lipopolysaccharide with S-methylisothiourea, an inducible nitric oxide synthase inhibitor. Co-injections of lipopolysaccharide with SB203580, a p38 MAP kinase inhibitor, reduced inducible nitric oxide synthase and caspase-11 mRNA expression, and also rescued dopaminergic neurons in substantia nigra. In summary, this is the first report to describe in vivo the temporal profile of the expression of these inflammatory mediators and proteins involved in dopaminergic neuronal death after intranigral injection of lipopolysaccharide. Moreover data strongly support that lipopolysaccharide-induced dopaminergic cellular death in substantia nigra could be mediated, at least in part, by the p38 signal pathway leading to activation of inducible nitric oxide synthase and caspase-11.
Collapse
Affiliation(s)
- D Ruano
- Departamento de Bioquímica, Bromatología, Toxicología y Medicina Legal, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | | | | | | | | | | | | |
Collapse
|
90
|
Iravani MM, Haddon CO, Rose S, Jenner P. 3-Nitrotyrosine-dependent dopaminergic neurotoxicity following direct nigral administration of a peroxynitrite but not a nitric oxide donor. Brain Res 2006; 1067:256-62. [PMID: 16388785 DOI: 10.1016/j.brainres.2005.10.086] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Revised: 10/15/2005] [Accepted: 10/21/2005] [Indexed: 10/25/2022]
Abstract
The presence of 3-nitrotyrosine (3-NT) adducts in Lewy bodies in Parkinson's disease suggests a role for nitrative stress in dopaminergic cell death. Whether this is a direct effect of increased nitric oxide (NO) formation or requires its reaction with superoxide to form peroxynitrite is not clear. In the present study, we show that direct nigral administration of a NO donor, SNOG, in the rat produced only local toxicity to dopaminergic neurones pre-labeled with fluorogold with no 3-NT formation. However, administration of a peroxynitrite donor, SIN-1, caused widespread damage to dopaminergic neurones and marked expression of 3-NT immunoreactivity. Importantly, dopaminergic cell loss and the expression of 3-NT were completely prevented when SIN-1 was co-administered with the NO/peroxynitrite scavenger, carboxy-PTIO. The results suggest that increased NO formation is not inherently toxic to dopaminergic neurons, but when both oxidative and nitrative stress combine to cause peroxynitrite formation, neurotoxicity occurs.
Collapse
Affiliation(s)
- M M Iravani
- Neurodegenerative Disease Research Centre, Guy's, King's and St Thomas' School of Biomedical Sciences, King's College, London SE1 1UL, UK
| | | | | | | |
Collapse
|