1
|
García-Revilla J, Herrera AJ, de Pablos RM, Venero JL. Inflammatory Animal Models of Parkinson’s Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:S165-S182. [PMID: 35662128 PMCID: PMC9535574 DOI: 10.3233/jpd-213138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Accumulating evidence suggests that microglia and peripheral immune cells may play determinant roles in the pathogenesis of Parkinson’s disease (PD). Consequently, there is a need to take advantage of immune-related models of PD to study the potential contribution of microglia and peripheral immune cells to the degeneration of the nigrostriatal system and help develop potential therapies for PD. In this review, we have summarised the main PD immune models. From a historical perspective, we highlight first the main features of intranigral injections of different pro-inflammogens, including lipopolysaccharide (LPS), thrombin, neuromelanin, etc. The use of adenoviral vectors to promote microglia-specific overexpression of different molecules in the ventral mesencephalon, including α-synuclein, IL-1β, and TNF, are also presented and briefly discussed. Finally, we summarise different models associated with peripheral inflammation whose contribution to the pathogenesis of neurodegenerative diseases is now an outstanding question. Illustrative examples included systemic LPS administration and dextran sulfate sodium-induced colitis in rodents.
Collapse
Affiliation(s)
- Juan García-Revilla
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Antonio J. Herrera
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - Rocío M. de Pablos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | - José Luis Venero
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Diffusion kurtosis imaging to evaluate the effect and mechanism of tetramethylpyrazine on cognitive impairment induced by lipopolysaccharide in rats. Brain Imaging Behav 2021; 15:2492-2501. [PMID: 33570727 DOI: 10.1007/s11682-021-00449-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/11/2020] [Accepted: 01/03/2021] [Indexed: 12/11/2022]
Abstract
Using diffusion kurtosis imaging (DKI) to evaluate the brain changes, the therapeutic effect and mechanism of tetramethylpyrazine in rats with dementia induced by lipopolysaccharide. Thirty-six male Sprague-Dawley rats were randomly divided into control group and five groups pretreated with sham operation, lipopolysaccharide(150ug) and three doses of tetramethylpyrazine(5, 10, and 20 mg/mL respectively). The Morris water maze test was used to evaluate cognitive ability. DKI and histology were performed. Low-dose of tetramethylpyrazine pretreated rats showed lower escape latency(6th day: 15.92seconds(s) vs. 5.11 s, P = 0.001), spent more time in the target quadrant(15.67 s vs. 29.83 s, P = 0.009) and crossed the platform area more frequently(3.50 vs. 9.17, P = 0.001) than rats in the LPS-treated group. Compared to sham group, the fractional anisotropy (FA), axial diffusion (Da), mean kurtosis (MK), and axial kurtosis (Ka) values in the cortex of lipopolysaccharide group were lower (P = 0.021,0.003,0.003,0.001,respectively).The MK, Ka, Kr, and FA values in the hippocampus of the lipopolysaccharide group were higher (P = 0.01, 0.026,0.007,0.003,respectively),while MD and Da values were lower (P = 0.045,0.044, respectively). Tetramethylpyrazine-pretreated rats showed higher values of FA, MD, Da, MK, and Ka in the cortex, lower MK, Ka, Kr, and FA values and higher MD,Da values in the hippocampus than the lipopolysaccharide group. Histologically, prominent inflammatory cells infiltration in the brain parenchyma of lipopolysaccharide group were observed, while groups pretreated using tetramethylpyrazine were alleviated. Tetramethylpyrazine can improve cognitive dysfunction induced by lipopolysaccharide. DKI can sensitively detect microstructure integrity of brain parenchyma in a non-invasive manner.
Collapse
|
3
|
Crisafulli U, Xavier AM, Dos Santos FB, Cambiaghi TD, Chang SY, Porcionatto M, Castilho BA, Malnic B, Glezer I. Topical Dexamethasone Administration Impairs Protein Synthesis and Neuronal Regeneration in the Olfactory Epithelium. Front Mol Neurosci 2018; 11:50. [PMID: 29559887 PMCID: PMC5845685 DOI: 10.3389/fnmol.2018.00050] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/06/2018] [Indexed: 11/13/2022] Open
Abstract
Chronic inflammatory process in the nasal mucosa is correlated with poor smell perception. Over-activation of immune cells in the olfactory epithelium (OE) is generally associated with loss of olfactory function, and topical steroidal anti-inflammatory drugs have been largely used for treating such condition. Whether this therapeutic strategy could directly affect the regenerative process in the OE remains unclear. In this study, we show that nasal topical application of dexamethasone (DEX; 200 or 800 ng/nostril), a potent synthetic anti-inflammatory steroid, attenuates OE lesion caused by Gram-negative bacteria lipopolysaccharide (LPS) intranasal infusion. In contrast, repeated DEX (400 ng/nostril) local application after lesion establishment limited the regeneration of olfactory sensory neurons after injury promoted by LPS or methimazole. Remarkably, DEX effects were observed when the drug was infused as 3 consecutive days regimen. The anti-inflammatory drug does not induce OE progenitor cell death, however, disturbance in mammalian target of rapamycin downstream signaling pathway and impairment of protein synthesis were observed during the course of DEX treatment. In addition, in vitro studies conducted with OE neurospheres in the absence of an inflammatory environment showed that glucocorticoid receptor engagement directly reduces OE progenitor cells proliferation. Our results suggest that DEX can interfere with the intrinsic regenerative cellular mechanisms of the OE, raising concerns on the use of topical anti-inflammatory steroids as a risk factor for progressive olfactory function impairment.
Collapse
Affiliation(s)
- Umberto Crisafulli
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Biochemistry, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - André M Xavier
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fabiana B Dos Santos
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Tavane D Cambiaghi
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Seo Y Chang
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Marimélia Porcionatto
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Beatriz A Castilho
- Department of Microbiology, Immunology and Parasitology, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Bettina Malnic
- Department of Biochemistry, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Isaias Glezer
- Department of Biochemistry, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
4
|
Stucky EC, Erndt-Marino J, Schloss RS, Yarmush ML, Shreiber DI. Prostaglandin E 2 Produced by Alginate-Encapsulated Mesenchymal Stromal Cells Modulates the Astrocyte Inflammatory Response. NANO LIFE 2017; 7:1750005. [PMID: 29682085 PMCID: PMC5903452 DOI: 10.1142/s1793984417500052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Astroglia are well known for their role in propagating secondary injury following brain trauma. Modulation of this injury cascade, including inflammation, is essential to repair and recovery. Mesenchymal stromal cells (MSCs) have been demonstrated as trophic mediators in several models of secondary CNS injury, however, there has been varied success with the use of direct implantation due to a failure to persist at the injury site. To achieve sustained therapeutic benefit, we have encapsulated MSCs in alginate microspheres and evaluated the ability of these encapsulated MSCs to attenuate neuro-inflammation. In this study, astroglial cultures were administered lipopolysaccharide (LPS) to induce inflammation and immediately co-cultured with encapsulated or monolayer human MSCs. Cultures were assayed for the pro-inflammatory cytokine tumor necrosis factor alpha (TNF-α) produced by astroglia, MSC-produced prostaglandin E2, and expression of neurotrophin-associated genes. We found that encapsulated MSCs significantly reduced TNF-α produced by LPS-stimulated astrocytes, more effectively than monolayer MSCs, and this enhanced benefit commences earlier than that of monolayer MSCs. Furthermore, in support of previous findings, encapsulated MSCs constitutively produced high levels of PGE2, while monolayer MSCs required the presence of inflammatory stimuli to induce PGE2 production. The early, constitutive presence of PGE2 significantly reduced astrocyte-produced TNF-α, while delayed administration had no effect. Finally, MSC-produced PGE2 was not only capable of modulating inflammation, but appears to have an additional role in stimulating astrocyte neurotrophin production. Overall, these results support the enhanced benefit of encapsulated MSC treatment, both in modulating the inflammatory response and providing neuroprotection.
Collapse
Affiliation(s)
- Elizabeth C Stucky
- Department of Chemical and Biochemical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - Joshua Erndt-Marino
- Department of Biomedical Engineering, The College of New Jersey, 2000 Pennington Road, Ewing, New Jersey 08628, USA
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers University, 599 Taylor Road, Piscataway, New Jersey 08854, USA
| |
Collapse
|
5
|
Abstract
The innate immune response is a coordinated set of reactions involving cells of myeloid lineage and a network of signaling molecules. Such a response takes place in the CNS during trauma, stroke, spinal cord injury, and neurodegenerative diseases, suggesting that macrophages/microglia are the cells that perpetuate the progressive neuronal damage. However, there is accumulating evidence that these cells and their secreted proinflammatory molecules have more beneficial effects than detrimental consequences for the neuronal elements. Indeed, a timely controlled innate immune response may limit toxicity in swiftly eliminating foreign materials and debris that are known to interfere with recovery and regeneration. Each step of the immune cascade is under the tight control of stimulatory and inhibitory signals. Glucocorticoids (GCs) act as the critical negative feedback on all myeloid cells, including those present within the brain parenchyma. Because too little is like too much, both an inappropriate feedback of GCs on microglia and high circulating GC levels in stressed individuals have been associated with deleterious consequences for the brain. In this review, the authors discuss both sides of the story with a particular emphasis on the neuro-protective role of endogenous GCs during immune challenges and the problems in determining whether GCs can be a good therapy for the treatment of neuropathological conditions.
Collapse
Affiliation(s)
- Isaias Glezer
- Laboratory of Molecular Endocrinology, CHUL Research Center, Department of Anatomy and Physiology, Laval University, Québec, Canada
| | | |
Collapse
|
6
|
Stucky EC, Schloss RS, Yarmush ML, Shreiber DI. Alginate micro-encapsulation of mesenchymal stromal cells enhances modulation of the neuro-inflammatory response. Cytotherapy 2015; 17:1353-64. [PMID: 26210574 PMCID: PMC5928499 DOI: 10.1016/j.jcyt.2015.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 04/29/2015] [Accepted: 05/11/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND AIMS Modulation of inflammation after brain trauma is a key therapeutic goal aimed at limiting the consequences of the subsequent injury cascade. Mesenchymal stromal cells (MSCs) have been demonstrated to dynamically regulate the inflammatory environment in several tissue systems, including the central nervous system. There has been limited success, however, with the use of direct implantation of cells in the brain caused by low viability and engraftment at the injury site. To circumvent this, we encapsulated MSCs in alginate microspheres and evaluated the ability of these encapsulated MSCs to attenuate inflammation in rat organotypic hippocampal slice cultures (OHSC). METHODS OHSC were administered lipopolysaccharide to induce inflammation and immediately co-cultured with encapsulated or monolayer human MSCs. After 24 h, culture media was assayed for the pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-α) produced by OHSC, as well as MSC-produced trophic mediators. RESULTS Encapsulated MSCs reduced TNF-α more effectively than did monolayer MSCs. Additionally, there was a strong correlation between increased prostaglandin E2 (PGE2) and reduction of TNF-α. In contrast to monolayer MSCs, inflammatory signals were not required to stimulate PGE2 production by encapsulated MSCs. Further encapsulation-stimulated changes were revealed in a multiplex panel analyzing 27 MSC-produced cytokines and growth factors, from which additional mediators with strong correlations to TNF-α levels were identified. CONCLUSIONS These results suggest that alginate encapsulation of MSCs may not only provide an improved delivery vehicle for transplantation but may also enhance MSC therapeutic benefit for treating neuro-inflammation.
Collapse
Affiliation(s)
- Elizabeth C Stucky
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Rene S Schloss
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Martin L Yarmush
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA; Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA; Center for Engineering in Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA.
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA.
| |
Collapse
|
7
|
Houdek HM, Larson J, Watt JA, Rosenberger TA. Bacterial lipopolysaccharide induces a dose-dependent activation of neuroglia and loss of basal forebrain cholinergic cells in the rat brain. INFLAMMATION AND CELL SIGNALING 2014; 1. [PMID: 26052539 DOI: 10.14800/ics.47] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In a rat model of neuroinflammation induced with a low-dose infusion lipopolysaccharide (5.0 ng/hr, LPS), we reported that brain arachidonic acid (ARA, 20:4 n-6), but not docosahexaenoic acid (DHA, 22:6n-3), metabolism is increased compared to control rats. To further characterize the impact LPS has on the induction of injury in this model, we quantified the dose-dependent activation of neuroglia and the loss of cholinergic cells in rats subjected to increasing doses of LPS. In this study, we found that LPS produced a statistically significant and linear dose-dependent increase in the percentage of activated CD11b-positive microglia ranging from 26% to 82% following exposure to doses ranging between 0.05 and 500 ng/hr, respectively. The percentage of activated GFAP-positive astrocytes also increased linearly and significantly from 35% to 91%. Significant astroglial scaring was evident at the lateral ventricular boarder of rats treated with 50 and 500 ng/hr LPS, but not evident in control treated rats or rats treated with lower doses of LPS. A dose-dependent decrease in the numbers of ChAT-positive cells in the basal forebrain of LPS-treated rats was found at higher doses of LPS (5, 50, and 500 ng/hr) but not at lower doses. The numbers of ChAT-positive cells within individual regions of the basal forebrain (medial septum and diagonal bands) and the composite basal forebrain were similar in their response. These data demonstrate that extremely low doses of LPS are sufficient to induce significant neuroglia activation while moderate doses above 5.0 ng/hr are required to induce cholinergic cell loss.
Collapse
Affiliation(s)
- Heidi M Houdek
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Jordan Larson
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - John A Watt
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| | - Thad A Rosenberger
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58203, USA
| |
Collapse
|
8
|
Abstract
Animal models of neuroinflammatory processes are needed to study the involvement of inflammation in neurodegenerative disorders such as Parkinson's and Alzheimer's diseases. One of the models used is based on lipopolysaccharide (LPS) as brain inflammation-inducing agent. This toxin is a potent inducer of inflammation and has different effects on cells of the immune system, as microglial cells. This chapter describes a protocol for the model of brain inflammation in rats based on the unilateral stereotaxic injection of LPS, which mimics the inflammatory milieu produced in some brain diseases.
Collapse
|
9
|
Hoban DB, Connaughton E, Connaughton C, Hogan G, Thornton C, Mulcahy P, Moloney TC, Dowd E. Further characterisation of the LPS model of Parkinson's disease: a comparison of intra-nigral and intra-striatal lipopolysaccharide administration on motor function, microgliosis and nigrostriatal neurodegeneration in the rat. Brain Behav Immun 2013; 27:91-100. [PMID: 23044176 DOI: 10.1016/j.bbi.2012.10.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 09/20/2012] [Accepted: 10/01/2012] [Indexed: 01/06/2023] Open
Abstract
Chronic neuroinflammation has been established as one of the many processes involved in the pathogenesis of Parkinson's disease (PD). Because of this, researchers have attempted to replicate this pathogenic feature in animal models using the potent inflammagen, lipopolysaccharide (LPS), in order to gain better understanding of immune-mediated events in PD. However, although the effect of intra-cerebral LPS on neuroinflammation and neurodegeneration has been relatively well characterised, its impact on motor function has been less well studied. Therefore, the aim of this study was to further characterise the neuropathological and behavioural impact of intra-nigral and intra-striatal administration of LPS. To do, LPS (10 μg) or vehicle (sterile saline) were stereotaxically injected into the adult rat substantia nigra or striatum on one side only. The effect of LPS administration on lateralised motor function was assessed using the Corridor, Stepping and Whisker tests for two weeks post-injection, after which, amphetamine-induced rotational asymmetry was completed. Post-mortem, the impact of LPS on nigrostriatal degeneration and microgliosis was assessed using quantitative tyrosine hydroxylase and OX-42 immunohistochemistry respectively. We found that intra-nigral administration of LPS led to localised microgliosis in the substantia nigra and this was accompanied by nigrostriatal neurodegeneration and stable spontaneous motor deficits. In contrast, intra-striatal administration of LPS led to localised microgliosis in the striatum but this did not lead to any nigrostriatal neurodegeneration and only induced transient motor dysfunction. In conclusion, this study reveals the impact of intra-cerebral LPS administration on PD-related neuropathology and motor function, and it indicates that the intra-nigral model may be a highly relevant model as it is associated with stable motor decline underpinned by nigral microgliosis and nigrostriatal neurodegeneration.
Collapse
Affiliation(s)
- Deirdre B Hoban
- The Department of Pharmacology & Therapeutics, School of Medicine, National University of Ireland, Galway, Ireland
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Norazit A, Nguyen M, Dickson C, Tuxworth G, Goss B, Mackay-Sim A, Meedeniya A. Vascular endothelial growth factor and platelet derived growth factor modulates the glial response to a cortical stab injury. Neuroscience 2011; 192:652-60. [DOI: 10.1016/j.neuroscience.2011.06.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 05/20/2011] [Accepted: 06/11/2011] [Indexed: 12/31/2022]
|
11
|
Machado A, Herrera AJ, Venero JL, Santiago M, de Pablos RM, Villarán RF, Espinosa-Oliva AM, Argüelles S, Sarmiento M, Delgado-Cortés MJ, Mauriño R, Cano J. Inflammatory Animal Model for Parkinson's Disease: The Intranigral Injection of LPS Induced the Inflammatory Process along with the Selective Degeneration of Nigrostriatal Dopaminergic Neurons. ISRN NEUROLOGY 2011; 2011:476158. [PMID: 22389821 PMCID: PMC3263561 DOI: 10.5402/2011/476158] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 03/17/2011] [Indexed: 12/15/2022]
Abstract
We have developed an animal model of degeneration of the nigrostriatal dopaminergic neurons, the neuronal system involved in Parkinson's disease (PD). The implication of neuroinflammation on this disease was originally established in 1988, when the presence of activated microglia in the substantia nigra (SN) of parkinsonians was reported by McGeer et al. Neuroinflammation could be involved in the progression of the disease or even has more direct implications. We injected 2 μg of the potent proinflammatory compound lipopolysaccharide (LPS) in different areas of the CNS, finding that SN displayed the highest inflammatory response and that dopaminergic (body) neurons showed a special and specific sensitivity to this process with the induction of selective dopaminergic degeneration. Neurodegeneration is induced by inflammation since it is prevented by anti-inflammatory compounds. The special sensitivity of dopaminergic neurons seems to be related to the endogenous dopaminergic content, since it is overcome by dopamine depletion. Compounds that activate microglia or induce inflammation have similar effects to LPS. This model suggest that inflammation is an important component of the degeneration of the nigrostriatal dopaminergic system, probably also in PD. Anti-inflammatory treatments could be useful to prevent or slow down the rate of dopaminergic degeneration in this disease.
Collapse
Affiliation(s)
- A Machado
- - Departmento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Yu JT, Lee CH, Yoo KY, Choi JH, Li H, Park OK, Yan B, Hwang IK, Kwon YG, Kim YM, Won MH. Maintenance of anti-inflammatory cytokines and reduction of glial activation in the ischemic hippocampal CA1 region preconditioned with lipopolysaccharide. J Neurol Sci 2010; 296:69-78. [PMID: 20580380 DOI: 10.1016/j.jns.2010.06.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2010] [Revised: 05/27/2010] [Accepted: 06/02/2010] [Indexed: 12/31/2022]
Abstract
Lipopolysaccharide (LPS) induces a strong immune response, and pretreatment with low dose of LPS suppresses the production of proinflammatory mediators. In the present study, we investigated the effect of LPS preconditioning on the delayed neuronal death in the gerbil hippocampal CA1 region after 5 min of transient cerebral ischemia. LPS preconditioning showed neuroprotective effects against ischemic damage in the hippocampal CA1 region after ischemic insult: about 92% of neurons in the CA1 region survived in the LPS-treated ischemia group. LPS preconditioning maintained anti-inflammatory cytokines, such as interleukin (IL)-4 and IL-13, in pyramidal neurons in the CA1 region after ischemia/reperfusion. In addition, IL-4 and IL-13 protein levels in the CA1 region of the LPS-treated ischemia group were similar to the vehicle-treated sham group. We found that reactive gliosis was markedly attenuated in the CA1 region of the LPS-treated ischemia group compared to the vehicle-treated ischemia group using immunohistochemistry of glial fibrillary acidic protein for astrocytes, and ionized calcium-binding adapter molecule 1 and isolectin B4 for microglia. These results indicate that LPS preconditioning may provide neuroprotection in the ischemic hippocampal CA1 region via maintenance of anti-inflammatory cytokines and suppression of glial activation.
Collapse
Affiliation(s)
- Jia Tian Yu
- Department of Anatomy and Neurobiology, and Institute of Neurodegeneration and Neuroregeneration, College of Medicine, Hallym University, Chuncheon 200-702, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Batchelor PE, Tan S, Wills TE, Porritt MJ, Howells DW. Comparison of inflammation in the brain and spinal cord following mechanical injury. J Neurotrauma 2009; 25:1217-25. [PMID: 18986223 DOI: 10.1089/neu.2007.0308] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inflammation in the CNS predominantly involves microglia and macrophages, and is believed to be a significant cause of secondary injury following trauma. This study compares the microglial and macrophage response in the rat brain and spinal cord following discrete mechanical injury to better appreciate the degree to which these cells could contribute to secondary damage in these areas. We find that, 1 week after injury, the microglial and macrophage response is significantly greater in the spinal cord compared to the brain. This is the case for injuries to both gray and white matter. In addition, we observed a greater inflammatory response in white matter compared to gray matter within both the brain and spinal cord. Because activated microglia and macrophages appear to be effectors of secondary damage, a greater degree of inflammation in the spinal cord is likely to result in more extensive secondary damage. Tissue saving strategies utilizing anti-inflammatory treatments may therefore be more useful in traumatic spinal cord than brain injury.
Collapse
Affiliation(s)
- Peter E Batchelor
- Department of Medicine, University of Melbourne, Austin Health, Victoria, Australia
| | | | | | | | | |
Collapse
|
14
|
Seki T, Fehlings MG. Mechanistic insights into posttraumatic syringomyelia based on a novel in vivo animal model. Laboratory investigation. J Neurosurg Spine 2008; 8:365-75. [PMID: 18377322 DOI: 10.3171/spi/2008/8/4/365] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECT Although posttraumatic syringomyelia (PTS) develops in up to 30% of patients after spinal cord injury (SCI), the pathophysiology of this debilitating complication is incompletely understood. To provide greater insight into the mechanisms of this degenerative sequela of SCI, the authors developed and characterized a novel model of PTS. METHODS The spinal cords of 64 female Wistar rats were injured by 35-g modified aneurysm clip compression at the level of T6-7. Kaolin (5 microl of 500 mg/ml solution) was then injected into the subarachnoid space rostral to the site of the injury to induce inflammatory arachnoiditis in 22 rats. Control groups received SCI alone (in 21 rats), kaolin injection alone (in 15 rats), or laminectomy and durotomy alone without injury (sham surgery in 6 rats). RESULTS The combination of SCI and subarachnoid kaolin injection resulted in a significantly greater syrinx formation and perilesional myelomalacia than SCI alone; SCI and kaolin injection significantly attenuated locomotor recovery and exacerbated neuropathic pain (mechanical allodynia) compared with SCI alone. We observed that combined SCI and kaolin injection significantly increased the number of terminal deoxytransferase-mediated deoxyuridine triphosphate nick-end labeled-positive cells at 7 days after injury (p<0.05 compared with SCI alone) and resulted in a significantly greater extent of astrogliosis and macrophage/microglial-associated inflammation at the lesion (p<0.05). CONCLUSIONS The combination of compressive/contusive SCI with induced arachnoiditis results in severe PTS and perilesional myelomalacia, which is associated with enhanced inflammation, astrogliosis, and apoptotic cell death. The development of delayed neurobehavioral deficits and neuropathic pain in this model accurately reflects the key pathological and clinical conditions of PTS in humans.
Collapse
Affiliation(s)
- Toshitaka Seki
- Division of Cell and Molecular Biology, Toronto Western Research Institute, Krembil Neuroscience Center, University Health Network, University of Toronto, Ontario, Canada
| | | |
Collapse
|
15
|
Weinstein JR, Swarts S, Bishop C, Hanisch UK, Möller T. Lipopolysaccharide is a frequent and significant contaminant in microglia-activating factors. Glia 2008; 56:16-26. [PMID: 17910052 PMCID: PMC2926344 DOI: 10.1002/glia.20585] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Lipopolysaccharide (LPS/endotoxin) is a potent immunologic stimulant. Many commercial-grade reagents used in research are not screened for LPS contamination. LPS induces a wide spectrum of proinflammatory responses in microglia, the immune cells of the brain. Recent studies have demonstrated that a broad range of endogenous factors including plasma-derived proteins and bioactive phospholipids can also activate microglia. However, few of these studies have reported either the LPS levels found in the preparations used or the effect of LPS inhibitors such as polymyxin B (PMX) on factor-induced responses. Here, we used the Limulus amoebocyte lysate assay to screen a broad range of commercial- and pharmaceutical-grade proteins, peptides, lipids, and inhibitors commonly used in microglia research for contamination with LPS. We then characterized the ability of PMX to alter a representative set of factor-induced microglial activation parameters including surface antigen expression, metabolic activity/proliferation, and NO/cytokine/chemokine release in both the N9 microglial cell line and primary microglia. Significant levels of LPS contamination were detected in a number of commercial-grade plasma/serum- and nonplasma/serum-derived proteins, phospholipids, and synthetic peptide preparations, but not in pharmaceutical-grade recombinant proteins or pharmacological inhibitors. PMX had a significant inhibitory effect on the microglia-activating potential of a number of commercial-, but not pharmaceutical-grade, protein preparations. Novel PMX-resistant responses to alpha(2)-macroglobulin and albumin were incidentally observed. Our results indicate that LPS is a frequent and significant contaminant in commercial-grade preparations of previously reported microglia-activating factors. Careful attention to LPS levels and appropriate controls are necessary for future studies in the neuroinflammation field.
Collapse
Affiliation(s)
- Jonathan R Weinstein
- Department of Neurology, School of Medicine, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | |
Collapse
|
16
|
Babcock AA, Wirenfeldt M, Holm T, Nielsen HH, Dissing-Olesen L, Toft-Hansen H, Millward JM, Landmann R, Rivest S, Finsen B, Owens T. Toll-like receptor 2 signaling in response to brain injury: an innate bridge to neuroinflammation. J Neurosci 2006; 26:12826-37. [PMID: 17151286 PMCID: PMC6674840 DOI: 10.1523/jneurosci.4937-05.2006] [Citation(s) in RCA: 150] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Reactive gliosis is a prominent feature of neurodegenerative and neuroinflammatory disease in the CNS, yet the stimuli that drive this response are not known. There is growing appreciation that signaling through Toll-like receptors (TLRs), which is key to generating innate responses to infection, may have pathogen-independent roles. We show that TLR2 was selectively upregulated by microglia in the denervated zones of the hippocampus in response to stereotactic transection of axons in the entorhinal cortex. In mice lacking TLR2, there were transient, selective reductions in lesion-induced expression of cytokines and chemokines. Recruitment of T cells, but not macrophages, was delayed in TLR2-deficient mice, as well as in mice lacking TNFR1 (tumor necrosis factor receptor 1). TLR2 deficiency also affected microglial proliferative expansion, whereas all of these events were unaffected in TLR4-mutant mice. Consistent with the fact that responses in knock-out mice had all returned to wild-type levels by 8 d, there was no evidence for effects on neuronal plasticity at 20 d. These results identify a role for TLR2 signaling in the early glial response to brain injury, acting as an innate bridge to neuroinflammation.
Collapse
Affiliation(s)
- Alicia A. Babcock
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
- Neuroimmunology Unit, Montreal Neurological Institute, Montreal, Quebec, Canada H3A 2B4
| | - Martin Wirenfeldt
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
| | - Thomas Holm
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
| | - Helle H. Nielsen
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
| | - Lasse Dissing-Olesen
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
| | - Henrik Toft-Hansen
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
- Neuroimmunology Unit, Montreal Neurological Institute, Montreal, Quebec, Canada H3A 2B4
| | - Jason M. Millward
- Neuroimmunology Unit, Montreal Neurological Institute, Montreal, Quebec, Canada H3A 2B4
| | - Regine Landmann
- Division of Infectious Diseases, Department of Research, University Hospital, 4031 Basel, Switzerland, and
| | - Serge Rivest
- Laboratory of Molecular Endocrinology, Centre Hospitalier Université Laval Research Center, and Department of Anatomy and Physiology, Laval University, Quebec, Quebec, Canada G1V 4G2
| | - Bente Finsen
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
| | - Trevor Owens
- Medical Biotechnology Center, University of Southern Denmark, 5000 Odense C, Denmark
- Neuroimmunology Unit, Montreal Neurological Institute, Montreal, Quebec, Canada H3A 2B4
| |
Collapse
|
17
|
Hossain-Ibrahim MK, Rezajooi K, MacNally JK, Mason MRJ, Lieberman AR, Anderson PN. Effects of lipopolysaccharide-induced inflammation on expression of growth-associated genes by corticospinal neurons. BMC Neurosci 2006; 7:8. [PMID: 16433912 PMCID: PMC1403789 DOI: 10.1186/1471-2202-7-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2005] [Accepted: 01/24/2006] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Inflammation around cell bodies of primary sensory neurons and retinal ganglion cells enhances expression of neuronal growth-associated genes and stimulates axonal regeneration. We have asked if inflammation would have similar effects on corticospinal neurons, which normally show little response to spinal cord injury. Lipopolysaccharide (LPS) was applied onto the pial surface of the motor cortex of adult rats with or without concomitant injury of the corticospinal tract at C4. Inflammation around corticospinal tract cell bodies in the motor cortex was assessed by immunohistochemistry for OX42 (a microglia and macrophage marker). Expression of growth-associated genes c-jun, ATF3, SCG10 and GAP-43 was investigated by immunohistochemistry or in situ hybridisation. RESULTS Application of LPS induced a gradient of inflammation through the full depth of the motor cortex and promoted c-Jun and SCG10 expression for up to 2 weeks, and GAP-43 upregulation for 3 days by many corticospinal neurons, but had very limited effects on neuronal ATF3 expression. However, many glial cells in the subcortical white matter upregulated ATF3. LPS did not promote sprouting of anterogradely labelled corticospinal axons, which did not grow into or beyond a cervical lesion site. CONCLUSION Inflammation produced by topical application of LPS promoted increased expression of some growth-associated genes in the cell bodies of corticospinal neurons, but was insufficient to promote regeneration of the corticospinal tract.
Collapse
Affiliation(s)
- MK Hossain-Ibrahim
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - K Rezajooi
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - JK MacNally
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - MRJ Mason
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - AR Lieberman
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - PN Anderson
- Department of Anatomy and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
18
|
Richardson RL, Kim EM, Gardiner T, O'Hare E. Chronic intracerebroventricular infusion of lipopolysaccharide: effects of ibuprofen treatment and behavioural and histopathological correlates. Behav Pharmacol 2005; 16:531-41. [PMID: 16170230 DOI: 10.1097/01.fbp.0000179278.03868.96] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Twenty male Wistar rats were trained under an alternating-lever cyclic-ratio (ALCR) schedule of food reinforcement. When responding showed no trends, each subject was subcutaneously implanted with an Alzet osmotic mini-pump, connected to a chronic indwelling cannula extending into the lateral ventricle of the brain. The mini-pumps were primed to infuse 0.25 microl lipopolysaccharide (LPS) (1.0 microg/0.25 ml) or 0.25 microl artificial cerebrospinal fluid (aCSF) per hour and were implanted for 28 days. LPS infusion produced behavioural deficits which chronic ibuprofen treatment (40 mg/kg every 12 h) alleviated. Infusion of LPS induced R 1282-positive amyloid deposits, and activation of microglia and astrocytes. Ibuprofen treatment reduced the numbers of activated microglia, and withdrawal of ibuprofen resulted in an increase in activated microglia; however, ibuprofen treatment had no effect on numbers of activated astrocytes in the LPS-infused subjects.
Collapse
Affiliation(s)
- R L Richardson
- School of Psychology, University of Ulster, Shore Road, Newtownabbey, Co. Antrim, N. Ireland, UK
| | | | | | | |
Collapse
|
19
|
Vallières N, Berard JL, David S, Lacroix S. Systemic injections of lipopolysaccharide accelerates myelin phagocytosis during Wallerian degeneration in the injured mouse spinal cord. Glia 2005; 53:103-13. [PMID: 16206158 DOI: 10.1002/glia.20266] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The phagocytic cell response within the injured spinal cord is inefficient, allowing myelin debris to remain for prolonged periods of time within white matter tracts distal to the injury. Several proteins associated with this degenerating myelin are inhibitory to axon growth and therefore prevent severed axons from regenerating. Inflammatory agents such as lipopolysaccharide (LPS) can stimulate both the migration and phagocytic activity of macrophages. Using in situ hybridization, we found that the expression of the LPS membrane receptor, CD14, was enhanced in the mouse dorsal column following a dorsal hemisection. Double labeling studies showed that microglia and macrophages are the two major cell types expressing CD14 mRNA following spinal cord injury (SCI). We therefore tested whether systemic injections of LPS would increase the number and phagocytic activity of macrophages/microglia in the ascending sensory tract (AST) of the mouse dorsal column following a dorsal hemisection. Mice were treated daily via intraperitoneal injections of either LPS or phosphate-buffered saline (PBS). At 7 days post-SCI, greater numbers of activated mononuclear phagocytes were present in the AST undergoing Wallerian degeneration (WD) in LPS-treated animals compared with controls. Animals treated with LPS also exhibited greater Oil Red O staining, which is specific for degenerating myelin and macrophages phagocytosing myelin debris. Myelin clearance was confirmed at 7 days using Luxol Fast Blue staining and on toluidine blue-stained semi-thin sections. These results indicate that it is possible to manipulate the innate immune response to accelerate myelin clearance during WD in the injured mouse spinal cord.
Collapse
Affiliation(s)
- Nicolas Vallières
- Department of Anatomy and Physiology, Laval University, Ste-Foy, Québec, Canada
| | | | | | | |
Collapse
|
20
|
Abstract
The aim of this study was to determine whether glutamate receptors modulate the innate immune response in the brain of C3H/HeN and C3H/HeJ mice; the latter bear a loss of function in the toll-like receptor (TLR) 4 gene. Mice received an intrastriatal (IS) infusion of lipopolysaccharide (LPS), the exogenous ligand for TLR4, and were killed at several times thereafter. This treatment activated the transcription of a wide variety of genes involved in the control of the innate immune response. MK-801, an antagonist of NMDA glutamate receptor subtype, exacerbated the effects of the endotoxin in the brain of C3H/HeN mice but not in TLR4-deficient animals. The ipsilateral side of C3H/HeN mice exhibited stronger hybridization signals for the mRNA encoding TLR2, CD14, tumor necrosis factor-alpha, and inhibitory factor-kappaBalpha at various times after the treatment combining MK-801 and LPS. This robust inflammatory response in the brain of C3H/HeN mice was not associated with any convincing signs of neurodegeneration or demyelination that was verified via numerous approaches and at time up to 2 weeks after injection. However, animals that received long-term IS infusion of LPS, together with MK-801, exhibited a significant increase in demyelination levels within the ipsilateral side. Our results demonstrate that binding of glutamate to its cognate NMDA receptor modulates LPS-induced innate immune reaction in a TLR4-dependent manner. This acute response may be crucial to eliminate bacterial cell wall components and minimizing tissue injury. However, sustained deregulation of proinflammatory signaling involving NMDA receptors leads to demyelination and is likely to be a mechanism participating in such pathological conditions.
Collapse
|
21
|
Logan A, Berry M. Cellular and molecular determinants of glial scar formation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003; 513:115-58. [PMID: 12575819 DOI: 10.1007/978-1-4615-0123-7_4] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Ann Logan
- Molecular Neuroscience, Department of Medicine, Wolfson Research Laboratories, Queen Elizabeth Hospital, Edgbaston, Birmingham, B15 2TH, UK
| | | |
Collapse
|
22
|
Liu B, Gao HM, Wang JY, Jeohn GH, Cooper CL, Hong JS. Role of nitric oxide in inflammation-mediated neurodegeneration. Ann N Y Acad Sci 2002; 962:318-31. [PMID: 12076984 DOI: 10.1111/j.1749-6632.2002.tb04077.x] [Citation(s) in RCA: 330] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Increasing evidence has suggested that inflammation in the brain is closely associated with the pathogenesis of several degenerative neurologic disorders, including Parkinson's disease, Alzheimer's diseases, multiple sclerosis, amyotrophic lateral sclerosis, and AIDS dementia. The hallmark of brain inflammation is the activation of glial cells, especially that of microglia that produce a variety of proinflammatory and neurotoxic factors, including cytokines, fatty acid metabolites, free radicals--such as nitric oxide (NO) and superoxide. Excessive production of NO, as a consequence of nitric oxide synthase induction in activated glia, has been attributed to participate in neurodegeneration. Using primary mixed neuron-glia cultures and glia-enriched cultures prepared from embryonic rodent brain tissues, we have systemically studied the relationship between the production of NO and neurodegeneration in response to stimulation by the inflammagen lipopolysaccharide. This review summarizes our recent findings on the kinetics of NO generation, the relative contribution of microglia and astrocytes to NO accumulation, the relationship between NO production and neurodegeneration, and points of intervention along the pathways associated with NO generation to achieve neuroprotection. We also describe our results relating to the effect of several opioid-related agents on microglial activation and neuroprotection. Among these agents, the opioid receptor antagonist naloxone, especially its non-opioid enantiomer (+)-naloxone, promises to be of potential therapeutic value for the treatment of inflammation-related diseases.
Collapse
Affiliation(s)
- Bin Liu
- Neuropharmacology Section, Laboratory of Pharmacology and Chemistry, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, North Carolina 27710, USA.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
In order to fully evaluate the effects of colchicine treatment on learning ability in rats, colchicine was administered, and both Morris water maze (MWM) and step-through type passive avoidance (PA) learning tests were conducted. In both learning tests, infusion of colchicine into the rat dentate gyrus, at two distinct bilateral rostrocaudal locations, potently impaired memory function in a dose-dependent manner (0.01-2.0 microg/site), whereas systemic injection of colchicine (50-300 microg/kg) did not. In the MWM test, memory impairment was observed even at doses where there was no evidence of any histological changes in the dentate granule cells. This suggests that functional deterioration, that is, learning impairment was induced by the dysfunction of microtubules and/or axons, was caused by colchicine. Moreover, ameliorated learning behavior was observed with chronic treatment of beta-estradiol 3-benzoate, which has been suggested to have an important role as an adjuvant treatment for younger Alzheimer's disease (AD), immediately after colchicine infusion (0.3 microg). These results indicate that the animal model accompanying the colchicine-induced functional defect showing early tau pathology, but not neuronal cell degeneration, may well mimic comparatively early stage of AD.
Collapse
Affiliation(s)
- Takahiro Nakayama
- BF Research Institute, Inc., c/o National Cardiovascular Center, 7-1, 5-Chome, Fujishiro-dai, Suita, Osaka 565-0873, Japan.
| | | |
Collapse
|
24
|
Berger R, Garnier Y, Pfeiffer D, Jensen A. Lipopolysaccharides do not alter metabolic disturbances in hippocampal slices of fetal guinea pigs after oxygen-glucose deprivation. Pediatr Res 2000; 48:531-5. [PMID: 11004246 DOI: 10.1203/00006450-200010000-00018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The aim of the present study was to clarify whether endotoxins [lipopolysaccharides (LPS)] have a toxic effect on fetal brain tissue after cerebral ischemia, while excluding their effect on the cardiovascular system. Experiments were therefore performed on hippocampal slices prepared from mature fetal guinea pigs. In particular, we studied the influence of LPS on nitric oxide production, energy metabolism, and protein synthesis after oxygen-glucose deprivation (OGD). Incubating hippocampal slices in LPS (4 mg/L) for as long as 12 h did not alter cGMP tissue concentrations significantly. However, 10 min after OGD of 40-min duration, cGMP tissue concentrations were substantially increased in relation to controls, and this increase was almost completely blocked by the application of 100 microM N:(omega)-nitro-L-arginine, indicating that nitric oxide synthase was activated after OGD in fetal brain tissue. Again, LPS did not have any effect on cGMP tissue concentrations after OGD. Furthermore, addition of LPS altered neither protein synthesis nor energy metabolism measured 12 h after OGD. We therefore conclude that, apart from their well-known influence on the cardiovascular system, LPS do not alter metabolic disturbances in hippocampal slices of fetal guinea pigs 12 h after OGD. A direct toxic effect of LPS on immature brain tissue within this interval does not therefore seem to be very likely. However, delayed activation of LPS-sensitive pathways that may be involved in cell death, or damage limited to a small subgroup of cells such as oligodendrocyte progenitors, cannot be fully excluded.
Collapse
Affiliation(s)
- R Berger
- Department of Obstetrics and Gynecology, Ruhr-Universität Bochum, In der Schornau 23-25, D-44892 Bochum, Germany
| | | | | | | |
Collapse
|
25
|
Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma. J Neurosci 1999. [PMID: 10493720 DOI: 10.1523/jneurosci.19-19-08182.1999] [Citation(s) in RCA: 396] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Post-traumatic cystic cavitation, in which the size and severity of a CNS injury progress from a small area of direct trauma to a greatly enlarged secondary injury surrounded by glial scar tissue, is a poorly understood complication of damage to the brain and spinal cord. Using minimally invasive techniques to avoid primary physical injury, this study demonstrates in vivo that inflammatory processes alone initiate a cascade of secondary tissue damage, progressive cavitation, and glial scarring in the CNS. An in vitro model allowed us to test the hypothesis that specific molecules that stimulate macrophage inflammatory activation are an important step in initiating secondary neuropathology. Time-lapse video analyses of inflammation-induced cavitation in our in vitro model revealed that this process occurs primarily via a previously undescribed cellular mechanism involving dramatic astrocyte morphological changes and rapid migration. The physical process of cavitation leads to astrocyte abandonment of neuronal processes, neurite stretching, and secondary injury. The macrophage mannose receptor and the complement receptor type 3 beta2-integrin are implicated in the cascade that induces cavity and scar formation. We also demonstrate that anti-inflammatory agents modulating transcription via the nuclear hormone receptor peroxisome proliferator-activated receptor-gamma may be therapeutic in preventing progressive cavitation by limiting inflammation and subsequent secondary damage after CNS injury.
Collapse
|
26
|
Cahill CM, Dray A, Coderre TJ. Priming enhances endotoxin-induced thermal hyperalgesia and mechanical allodynia in rats. Brain Res 1998; 808:13-22. [PMID: 9795108 DOI: 10.1016/s0006-8993(98)00786-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Central inflammation is an integral component and contributor of the pathology of many debilitating diseases and has been shown to produce spontaneous pain and hyperalgesia. Recently, administration of lipopolysaccharide (LPS) into the lateral ventricle of rats was shown to elicit both thermal hyperalgesia and tactile allodynia [K. Walker, A. Dray, M. Perkins, Hyperalgesia in rats following intracerebroventricular administration of endotoxin: effect of bradykinin B1 and B2 receptor antagonist treatment, Pain 65 (1996) 211-219]. In this study, we have replicated the LPS model with some adaptations and correlated the nociceptive behaviors with an increased expression of activated macrophages in the central nervous system. We also examined the effects of priming on LPS-induced decreases in thermal nociceptive thresholds and mechanical response thresholds following either central or peripheral administration. Intracerebroventricular (i.c.v.) administration of LPS (0.2 microgram/rat) did not alter either thermal (hot plate) or mechanical (von Frey filaments) thresholds compared to baseline values in the first few hours after injection. However, priming rats by pretreating with i.c.v. LPS (0.2 microgram) 24 h prior to testing with i.c.v. LPS (0.2 microgram) produced significant mechanical allodynia and thermal hyperalgesia. The mechanical allodynia had an onset of 80 min after injection and a duration of 5 h. A similar time course was observed for thermal hyperalgesia, although its expression was less pronounced. Immunohistochemical studies indicated an increased expression of activated macrophages in the brain parenchyma of primed rats but not in unprimed rats. Intraperitoneal (i.p., 2 mg/kg) administration of LPS had no significant effect on either thermal or mechanical thresholds in the first few hours after injection; however, priming rats via i.p. (0.2 mg/kg) or i.c.v. (0.2 microgram) LPS produced a reduction in both thermal nociceptive thresholds and mechanical response thresholds in rats given a subsequent i.p. injection of LPS. This study demonstrates that priming is an effective protocol for the induction of central inflammation and increases the duration of these behaviors after i.c. v. administration.
Collapse
Affiliation(s)
- C M Cahill
- Pain Mechanisms Laboratory, Clinical Research Institute of Montreal, 110 Pine Ave. West, Montreal, Canada.
| | | | | |
Collapse
|
27
|
Hauss-Wegrzyniak B, Lukovic L, Bigaud M, Stoeckel ME. Brain inflammatory response induced by intracerebroventricular infusion of lipopolysaccharide: an immunohistochemical study. Brain Res 1998; 794:211-24. [PMID: 9622633 DOI: 10.1016/s0006-8993(98)00227-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory processes may play a critical role in the pathogenesis of the degenerative changes associated with Alzheimer's disease (AD). In the present study, we used an animal model of brain inflammation in order to study a possible mechanism involved in AD. Lipopolysaccharide (LPS) was used to produce global microglial reactivity within the brain of young rats. Time-dependent changes in the inflammatory reaction and the participation of glial cells after acute injection of LPS (50 or 100 microg) into the lateral ventricle or the fourth ventricle were compared with the chronic infusion of LPS (0.15, 0.5, 1.5 or 5.0 microg/h) into the fourth ventricle (14 days). Several immunohistochemical markers were used to characterize the microglial response. Acute and chronic exposure to LPS induced major histocompatibility complex class II (MHC II) antigen expression, detected with OX-6 antibody, in a sub-population of microglial cells in defined brain areas. The morphological features and distribution of OX-6 positive cells observed in the proximity of the cannula track after LPS injection into the lateral ventricle suggested the recruitment of monocytes/macrophages from the periphery. The activation of the resident microglial cells was delayed and mainly concentrated within the temporal lobe regions and the limbic system. Chronic infusion to LPS into the fourth ventricle induced a comparable activation of microglial cells. Quantitative analysis of OX-6 positive cells showed a dose-dependent response to LPS exposure.
Collapse
Affiliation(s)
- B Hauss-Wegrzyniak
- Arizona research Laboratories, Division of Neural Systems, Memory and Aging, University of Arizona, Tucson, AZ, USA.
| | | | | | | |
Collapse
|
28
|
Fitch MT, Silver J. Activated macrophages and the blood-brain barrier: inflammation after CNS injury leads to increases in putative inhibitory molecules. Exp Neurol 1997; 148:587-603. [PMID: 9417835 DOI: 10.1006/exnr.1997.6701] [Citation(s) in RCA: 198] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The cellular responses to spinal cord or brain injury include the production of molecules that modulate wound healing. This study examined the upregulation of chondroitin sulfate proteoglycans, a family of molecules present in the wound healing matrix that may inhibit axon regeneration in the central nervous system (CNS) after trauma. We have demonstrated increases in these putative inhibitory molecules in brain and spinal cord injury models, and we observed a close correlation between the tissue distribution of their upregulation and the presence of inflammation and a compromised blood-brain barrier. We determined that the presence of degenerating and dying axons injured by direct trauma does not provide a sufficient signal to induce the increases in proteoglycans observed after injury. Activated macrophages, their products, or other serum components that cross a compromised blood-brain barrier may provide a stimulus for changes in extracellular matrix molecules after CNS injury. While gliosis is associated with increased levels of proteoglycans, not all reactive astrocytes are associated with augmented amounts of these extracellular matrix molecules, which suggests a heterogeneity among glial cells that exhibit a reactive phenotype. Chondroitin sulfate also demarcates developing cavities of secondary necrosis, implicating these types of boundary molecules in the protective response of the CNS to trauma.
Collapse
Affiliation(s)
- M T Fitch
- Department of Neurosciences, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, USA
| | | |
Collapse
|