51
|
NADPH oxidase mediates TNF-α-evoked in vitro brain barrier dysfunction: roles of apoptosis and time. Mol Cell Neurosci 2014; 61:72-84. [PMID: 24907586 DOI: 10.1016/j.mcn.2014.06.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/20/2014] [Accepted: 06/02/2014] [Indexed: 11/20/2022] Open
Abstract
The pro-inflammatory cytokine TNF-α severely perturbs the integrity of the blood-brain barrier (BBB). This study explored the specific roles of NADPH oxidase and associated downstream effectors by using human brain microvascular endothelial cells (HBMECs) and human astrocytes (HAs), the key components of BBB, alone or in co-cultures to mimic human BBB. Exposure to TNF-α (6h) impaired BBB integrity as evidenced by marked decreases in transendothelial electrical resistance and concurrent increases in paracellular flux which appeared to subside with time (24h). Increased barrier dysfunction concurred with increases in endothelial NADPH oxidase activity, O2(-) production, actin stress fibre formation, MMP-2/9 activities and concomitant decreases in antioxidant (CuZn-SOD and catalase) and tight junction (claudin-5 and occludin) protein expressions. Conversely, TNF-α did not affect astrocytic MMP activities and antioxidant enzyme expressions. Unlike BBB damage, rates of HBMEC and HA apoptosis increased by time. Suppression of NADPH oxidase by apocynin or diphenyleneiodonium prevented TNF-α-evoked morphological changes and apoptosis, attenuated endothelial MMP activity and helped retain usual tight junction protein expression and barrier function. In conclusion, HBMECs constitute the main source of oxidative stress and basement-membrane degrading endopeptidases in inflammatory conditions associated with excessive release of TNF-α where targeting NADPH oxidase may prove extremely beneficial in maintaining proper barrier activity through prevention of cytoskeletal and tight junction reorganisations.
Collapse
|
52
|
Janata A, Magnet IAM, Uray T, Stezoski JP, Janesko-Feldman K, Tisherman SA, Kochanek PM, Drabek T. Regional TNFα mapping in the brain reveals the striatum as a neuroinflammatory target after ventricular fibrillation cardiac arrest in rats. Resuscitation 2014; 85:694-701. [PMID: 24530249 PMCID: PMC4034695 DOI: 10.1016/j.resuscitation.2014.01.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Revised: 01/22/2014] [Accepted: 01/31/2014] [Indexed: 12/14/2022]
Abstract
Cardiac arrest (CA) triggers neuroinflammation that could play a role in a delayed neuronal death. In our previously established rat model of ventricular fibrillation (VF) CA characterized by extensive neuronal death, we tested the hypothesis that individual brain regions have specific neuroinflammatory responses, as reflected by regional brain tissue levels of tumor necrosis factor (TNF)α and other cytokines. In a prospective study, rats were randomized to 6min (CA6), 8min (CA8) or 10min (CA10) of VF CA, or sham group. Cortex, striatum, hippocampus and cerebellum were evaluated for TNFα and interleukin (IL)-1α, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12 and interferon gamma at 3h, 6h or 14 d after CA by ELISA and Luminex. Immunohistochemistry was used to determine the cell source of TNFα. CA resulted in a selective TNFα response with significant regional and temporal differences. At 3h after CA, TNFα-levels increased in all regions depending on the duration of the insult. The most pronounced increase was observed in striatum that showed 20-fold increase in CA10 vs. sham, and 3-fold increase vs. CA6 or CA8 group, respectively (p<0.01). TNFα levels in striatum decreased between 3h and 6h, but increased in other regions between 3h and 14 d. TNFα levels remained twofold higher in CA6 vs. shams across brain regions at 14 d (p<0.01). In contrast to pronounced TNFα response, other cytokines showed only a minimal increase in CA6 and CA8 groups vs. sham in all brain regions with the exception that IL-1β increased twofold in cerebellum and striatum (p<0.01). TNFα colocalized with neurons. In conclusion, CA produced a duration-dependent acute TNFα response, with dramatic increase in the striatum where TNFα colocalized with neurons. Increased TNFα levels persist for at least two weeks. This TNFα surge contrasts the lack of an acute increase in other cytokines in brain after CA. Given that striatum is a selectively vulnerable brain region, our data suggest possible role of neuronal TNFα in striatum after CA and identify therapeutic targets for future experiments. This study was approved by the University of Pittsburgh IACUC 1002340A-3.
Collapse
Affiliation(s)
- Andreas Janata
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ingrid A M Magnet
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Thomas Uray
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jason P Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Samuel A Tisherman
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patrick M Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Department of Anesthesiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
53
|
Drabek T, Janata A, Wilson CD, Stezoski J, Janesko-Feldman K, Tisherman SA, Foley LM, Verrier J, Kochanek PM. Minocycline attenuates brain tissue levels of TNF-α produced by neurons after prolonged hypothermic cardiac arrest in rats. Resuscitation 2014; 85:284-291. [PMID: 24513126 PMCID: PMC3952024 DOI: 10.1016/j.resuscitation.2013.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 09/24/2013] [Accepted: 10/15/2013] [Indexed: 12/14/2022]
Abstract
Neuro-cognitive disabilities are a well-recognized complication of hypothermic circulatory arrest. We and others have reported that prolonged cardiac arrest (CA) produces neuronal death and microglial proliferation and activation that are only partially mitigated by hypothermia. Microglia, and possibly other cells, are suggested to elaborate tumor necrosis factor alpha (TNF-α), which can trigger neuronal death cascades and exacerbate edema after CNS insults. Minocycline is neuroprotective in some brain ischemia models in part by blunting the microglial response. We tested the hypothesis that minocycline would attenuate neuroinflammation as reflected by brain tissue levels of TNF-α after hypothermic CA in rats. Rats were subjected to rapid exsanguination, followed by a 6 min normothermic CA. Hypothermia (30 °C) was then induced by an aortic saline flush. After a total of 20 min CA, resuscitation was achieved via cardiopulmonary bypass (CPB). After 5 min reperfusion, minocycline (90 mg kg−1; n = 6) or vehicle (PBS; n = 6) was given. Hypothermia (34 °C) was maintained for 6 h. Rats were sacrificed at 6 or 24 h. TNF-α was quantified (ELISA) in four brain regions (cerebellum, CEREB; cortex, CTX; hippocampus, HIP; striatum, STRI). Naïve rats (n = 6) and rats subjected to the same anesthesia and CPB but no CA served as controls (n = 6). Immunocytochemistry was used to localize TNF-α. Naïve rats and CPB controls had no detectable TNF-α in any brain region. CA markedly increased brain TNF-α. Regional differences were seen, with the highest TNF-α levels in striatum in CA groups (10-fold higher, P < 0.05 vs. all other brain regions). TNF-α was undetectable at 24 h. Minocycline attenuated TNF-α levels in CTX, HIP and STRI (P < 0.05). TNF-α showed unique co-localization with neurons. In conclusion, we report region-dependent early increases in brain TNF-α levels after prolonged hypothermic CA, with maximal increases in striatum. Surprisingly, TNF-α co-localized in neurons and not microglia. Minocycline attenuated TNF-α by approximately 50% but did not totally ablate its production. That minocycline decreased brain TNF-α levels suggests that it may represent a therapeutic adjunct to hypothermia in CA neuroprotection. University of Pittsburgh IACUC 0809278B-3.
Collapse
Affiliation(s)
- Tomas Drabek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Andreas Janata
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Caleb D. Wilson
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jason Stezoski
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Anesthesiology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Keri Janesko-Feldman
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samuel A. Tisherman
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lesley M. Foley
- Pittsburgh NMR Center for Biomedical Research, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Jonathan Verrier
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
54
|
Chen Y, Won SJ, Xu Y, Swanson RA. Targeting microglial activation in stroke therapy: pharmacological tools and gender effects. Curr Med Chem 2014; 21:2146-55. [PMID: 24372213 PMCID: PMC4076056 DOI: 10.2174/0929867321666131228203906] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 06/02/2013] [Accepted: 06/03/2013] [Indexed: 12/20/2022]
Abstract
Ischemic stroke is caused by critical reductions in blood flow to brain or spinal cord. Microglia are the resident immune cells of the central nervous system, and they respond to stroke by assuming an activated phenotype that releases cytotoxic cytokines, reactive oxygen species, proteases, and other factors. This acute, innate immune response may be teleologically adapted to limit infection, but in stroke this response can exacerbate injury by further damaging or killing nearby neurons and other cell types, and by recruiting infiltration of circulating cytotoxic immune cells. The microglial response requires hours to days to fully develop, and this time interval presents a clinically accessible time window for initiating therapy. Because of redundancy in cytotoxic microglial responses, the most effective therapeutic approach may be to target the global gene expression changes involved in microglial activation. Several classes of drugs can do this, including histone deacetylase inhibitors, minocycline and other PARP inhibitors, corticosteroids, and inhibitors of TNFα and scavenger receptor signaling. Here we review the pre-clinical studies in which these drugs have been used to suppress microglial activation after stroke. We also review recent advances in the understanding of sex differences in the CNS inflammatory response, as these differences are likely to influence the efficacy of drugs targeting post-stroke brain inflammation.
Collapse
Affiliation(s)
| | | | | | - R A Swanson
- Dept. of Neurology, University of California San Francisco; and Neurology Service, San Francisco Veterans Affairs Medical Center, 4150 Clement St, San Francisco, CA 94121, USA.
| |
Collapse
|
55
|
Pelekanou V, Notas G, Kampa M, Tsentelierou E, Stathopoulos EN, Tsapis A, Castanas E. BAFF, APRIL, TWEAK, BCMA, TACI and Fn14 proteins are related to human glioma tumor grade: immunohistochemistry and public microarray data meta-analysis. PLoS One 2013; 8:e83250. [PMID: 24376672 PMCID: PMC3869762 DOI: 10.1371/journal.pone.0083250] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 11/01/2013] [Indexed: 11/30/2022] Open
Abstract
Gliomas are common and lethal tumors of the central nervous system (CNS). Genetic alterations, inflammatory and angiogenic processes have been identified throughout tumor progression; however, treatment still remains palliative for most cases. Biological research on parameters influencing cell survival, invasion and tumor heterogeneity identified several cytokines interfering in CNS inflammation, oxidative stress and malignant transformation, including TNF-superfamily (TNFSF) members. In this report we performed a meta-analysis of public gene-array data on the expression of a group of TNFSF ligands (BAFF, APRIL, TWEAK) and their receptors (BAFF-R, TACI, BCMA, Fn14) in gliomas. In addition, we investigated by immunohistochemistry (IHC) the tumor cells' expression of these ligands and receptors in a series of 56 gliomas of different grade. We show that in IHC, BAFF and APRIL as well as their cognate receptors (BCMA, TACI) and Fn14 expression correlate with tumor grade. This result was not evidenced in micro-arrays meta-analysis. Finally, we detected for the first time Fn14, BAFF, BCMA and TACI in glioma-related vascular endothelium. Our data, combined with our previous report in glioma cell lines, suggest a role for these receptors and ligands in glioma biology and advance these molecules as potential markers for the classification of these tumors to the proliferative, angiogenic or stem-like molecular subtype.
Collapse
Affiliation(s)
- Vassiliki Pelekanou
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
- Laboratories of Pathology, University of Crete, School of Medicine, Heraklion, Greece
| | - George Notas
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
| | - Marilena Kampa
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
| | | | | | - Andreas Tsapis
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
- INSERM U976, Hôpital Saint Louis, Paris, France; (4) Université Paris Diderot, Paris, France
| | - Elias Castanas
- Laboratories of Experimental Endocrinology, University of Crete, School of Medicine, Heraklion, Greece
| |
Collapse
|
56
|
Kalay S, Oztekin O, Tezel G, Aldemir H, Sahin E, Koksoy S, Akcakus M, Oygur N. The effects of intraperitoneal pentoxifylline treatment in rat pups with hypoxic-ischemic encephalopathy. Pediatr Neurol 2013; 49:319-23. [PMID: 23993832 DOI: 10.1016/j.pediatrneurol.2013.05.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/22/2013] [Accepted: 05/24/2013] [Indexed: 11/15/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the effects of postischemic treatment with pentoxifylline on the cytokine gene expressions and neuronal apoptosis in neonatal rat model of hypoxic-ischemic encephalopathy. METHODS Seven-day-old Wistar rat pups (n = 40) of either sex, delivered spontaneously, were used in this experimental study. Control group (n = 8): after median neck incision was made, neither ligation nor hypoxia was performed, ischemia group (n = 16): 0.5 mL of saline was injected intraperitoneally immediately after hypoxia. Pentoxifylline and ischemia groups (n = 16): the rat pups were administered intraperitoneally 60 mg/kg of pentoxifylline immediately after hypoxia. Eight rats from ischemia and pentoxifylline + ischemia groups were sacrificed 4 and 24 hours after drug administration. Control group mice were decapitated 4 hours after hypoxia. Caspase-3 activity, interleukin-1β, and tumor necrosis factor-α messenger RNA expression levels were studied in the left half of the brain. RESULTS Induction of cerebral ischemia increased tumor necrosis factor-α and interleukin-1β messenger RNA expression levels significantly at 4 hours and 24 hours following ischemia in the left ischemic hemispheres in the ischemia group as compared with the control group. Systemic administration of pentoxifylline immediately after hypoxic-ischemic encephalopathy significantly reduced the tumor necrosis factor-α and interleukin-1β messenger RNA expression levels in ischemic tissue as compared with the ischemia group. Caspase-3 activities in the left half of the brains of ischemia group were found to be increased significantly as compared with control group. Caspase-3 activities in the brains of pentoxifylline + ischemia groups were significantly lower than in that of ischemia group. CONCLUSIONS Based on the significantly lower interleukin-1β and tumor necrosis factor-α gene expression measured after 4 and 24 hours and significantly reduced caspase-3 activity measured colorimetrically in the animals treated with pentoxifylline, our findings suggest that pentoxifylline may reduce brain damage due to hypoxic-ischemic injury.
Collapse
Affiliation(s)
- Salih Kalay
- Division of Neonatology, Department of Pediatrics, School of Medicine, Akdeniz University, Antalya, Turkey
| | | | | | | | | | | | | | | |
Collapse
|
57
|
Zhang L, Dong LY, Li YJ, Hong Z, Wei WS. The microRNA miR-181c controls microglia-mediated neuronal apoptosis by suppressing tumor necrosis factor. J Neuroinflammation 2012; 9:211. [PMID: 22950459 PMCID: PMC3488569 DOI: 10.1186/1742-2094-9-211] [Citation(s) in RCA: 123] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 08/18/2012] [Indexed: 12/01/2022] Open
Abstract
Background Post-ischemic microglial activation may contribute to neuronal damage through the release of large amounts of pro-inflammatory cytokines and neurotoxic factors. The involvement of microRNAs (miRNAs) in the pathogenesis of disorders related to the brain and central nervous system has been previously studied, but it remains unknown whether the production of pro-inflammatory cytokines is regulated by miRNAs. Methods BV-2 and primary rat microglial cells were activated by exposure to oxygen-glucose deprivation (OGD). Global cerebral ischemia was induced using the four-vessel occlusion (4-VO) model in rats. Induction of pro-inflammatory and neurotoxic factors, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric oxide (NO), were assessed by ELISA, immunofluorescence, and the Griess assay, respectively. The miRNA expression profiles of OGD-activated BV-2 cells were subsequently compared with the profiles of resting cells in a miRNA microarray. BV-2 and primary rat microglial cells were transfected with miR-181c to evaluate its effects on TNF-α production after OGD. In addition, a luciferase reporter assay was conducted to confirm whether TNF-α is a direct target of miR-181c. Results OGD induced BV-2 microglial activation in vitro, as indicated by the overproduction of TNF-α, IL-1β, and NO. Global cerebral ischemia/reperfusion injury induced microglial activation and the release of pro-inflammatory cytokines in the hippocampus. OGD also downregulated miR-181c expression and upregulated TNF-α expression. Overproduction of TNF-α after OGD-induced microglial activation provoked neuronal apoptosis, whereas the ectopic expression of miR-181c partially protected neurons from cell death caused by OGD-activated microglia. RNAinterference-mediated knockdown of TNF-α phenocopied the effect of miR-181c-mediated neuronal protection, whereas overexpression of TNF-α blocked the miR-181c-dependent suppression of apoptosis. Further studies showed that miR-181c could directly target the 3′-untranslated region of TNF-α mRNA, suppressing its mRNA and protein expression. Conclusions Our data suggest a potential role for miR-181c in the regulation of TNF-α expression after ischemia/hypoxia and microglia-mediated neuronal injury.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurology, Huadong Hospital, Fudan University, 221 West Yan An Road, Shanghai, 200040, China
| | | | | | | | | |
Collapse
|
58
|
Therapeutic effects of postischemic treatment with hypotensive doses of an angiotensin II receptor blocker on transient focal cerebral ischemia. J Hypertens 2012; 29:2210-9. [PMID: 21934531 DOI: 10.1097/hjh.0b013e32834bbb30] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Neurovascular protection against cerebral ischemia is not consistently observed with a postischemia hypotensive dose of candesartan. The aim of this study was to determine the levels of brain angiotensin II after reperfusion and the efficacy and therapeutic time window of postischemic treatments with hypotensive doses of candesartan for the treatment of cerebral ischemia. METHOD Occlusions of the right middle cerebral artery (60 min) followed by reperfusion were performed using the thread method under halothane anesthesia in Sprague-Dawley (SD) rats. Protein levels of brain angiotensin II and mRNA levels of renin-angiotensin system components were evaluated following reperfusion (n=184 in total). Low-dose or high-dose treatments with candesartan cilexetil (1 or 10 mg/kg per day, respectively) were administered orally immediately following reperfusion once daily for 4 or 7 days (n = 119 in total). An additional group was treated with low-dose candesartan cilexetil after a 12-h delay based on the brain angiotensin II levels (n = 14). RESULTS Levels of brain angiotensin II transiently increased 4-12 h after reperfusion, which followed an increase in angiotensinogen mRNA. Candesartan cilexetil treatments significantly reduced blood pressure (BP) in rats administered the high dose and moderately in rats receiving the low dose. A low dose of candesartan cilexetil reduced the infarct size, cerebral edema, and neurological deficits, whereas the high-dose treatments showed limited reductions. Furthermore, oxidative stress following reperfusion was reduced with the low-dose treatments. The therapeutic time window was open for at least 12 h after reperfusion when brain angiotensin II levels had peaked. CONCLUSION Postischemic treatments using low hypotensive doses of candesartan cilexetil provided protection against cerebral ischemic injury and may have a clinically relevant therapeutic time window.
Collapse
|
59
|
Leung PY, Stevens SL, Packard AEB, Lessov NS, Yang T, Conrad VK, van den Dungen NNAM, Simon RP, Stenzel-Poore MP. Toll-like receptor 7 preconditioning induces robust neuroprotection against stroke by a novel type I interferon-mediated mechanism. Stroke 2012; 43:1383-9. [PMID: 22403050 DOI: 10.1161/strokeaha.111.641522] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Systemic administration of Toll-like receptor (TLR) 4 and TLR9 agonists before cerebral ischemia have been shown to reduce ischemic injury by reprogramming the response of the brain to stroke. Our goal was to explore the mechanism of TLR-induced neuroprotection by determining whether a TLR7 agonist also protects against stroke injury. METHODS C57Bl/6, TNF(-/-), interferon (IFN) regulatory factor 7(-/-), or type I IFN receptor (IFNAR)(-/-) mice were subcutaneously administered the TLR7 agonist Gardiquimod (GDQ) 72 hours before middle cerebral artery occlusion. Infarct volume and functional outcome were determined after reperfusion. Plasma cytokine responses and induction of mRNA for IFN-related genes in the brain were measured. IFNAR(-/-) mice also were treated with the TLR4 agonist (lipopolysaccharide) or the TLR9 agonist before middle cerebral artery occlusion and infarct volumes measured. RESULTS The results show that GDQ reduces infarct volume as well as functional deficits in mice. GDQ pretreatment provided robust neuroprotection in TNF(-/-) mice, indicating that TNF was not essential. GDQ induced a significant increase in plasma IFNα levels and both IRF7(-/-) and IFNAR(-/-) mice failed to be protected, implicating a role for IFN signaling in TLR7-mediated protection. CONCLUSIONS Our studies provide the first evidence that TLR7 preconditioning can mediate neuroprotection against ischemic injury. Moreover, we show that the mechanism of protection is unique from other TLR preconditioning ligands in that it is independent of TNF and dependent on IFNAR.
Collapse
Affiliation(s)
- Philberta Y Leung
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Zhu J, Jiang Y, Wu L, Lu T, Xu G, Liu X. Suppression of local inflammation contributes to the neuroprotective effect of ginsenoside Rb1 in rats with cerebral ischemia. Neuroscience 2011; 202:342-51. [PMID: 22173011 DOI: 10.1016/j.neuroscience.2011.11.070] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 11/08/2011] [Accepted: 11/30/2011] [Indexed: 11/15/2022]
Abstract
Local inflammation accounts for the progression of cerebral ischemic insult. Ginsenoside Rb1 (GRb1) is a natural product extracted from Panax ginseng C.A. Meyer. It has been reported to have beneficial effects in cerebral ischemia and to inhibit the inflammatory cascade in sepsis. In this study, to determine whether modulating local inflammation contributed to the neuroprotection of GRb1, male Sprague-Dawley rats were treated with GRb1 or vehicle intranasally for 1 week before being subjected to temporary occlusion of the right middle cerebral artery and reperfusion. Neuroprotection of GRb1 was evaluated with a focus on the key elements of central nervous system (CNS) inflammation, such as inflammatory cells, proinflammatory cytokines, and transcriptional factor. GRb1 reduced infarction volume by 57% (n=6, P<0.01) and significantly alleviated the neurological deficit (n=12, modified neurological severity scores [mNSS]: 6.6±1.1 vs. 8.6±1.1, P<0.05). GRb1 depressed the activation of microglia in the penumbra by 15%-27% from 24 h to 72 h after reperfusion and its further convention into phagocytic microglia/macrophages. In GRb1 group, the peak mRNA level of tumor necrosis factor α (TNF-α) mRNA was decreased by 35% 12 h after reperfusion, whereas the protein level was significantly reduced by 43%-57%. Downregulation by GRb1 of both interleukin (IL)-6 gene and protein after GRb1 administration was also observed. GRb1 partially inhibited the activation of nuclear factor-κB (NF-κB) pathway from 6 h to 72 h after ischemia and reperfusion onset, as determined by the expression of total and phosphorylated NF-κB/p65, inhibitor protein of κB (IκB)-α, and IκB-kinase complex (IKK)-α. All these results indicate that suppression of local inflammation after cerebral ischemia might be one mechanism that contributes to the neuroprotection of GRb1.
Collapse
Affiliation(s)
- J Zhu
- Department of Neurology, Jinling Hospital, Nanjing University School of Medicine, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, China
| | | | | | | | | | | |
Collapse
|
61
|
Combination treatment with low-dose Niaspan and tissue plasminogen activator provides neuroprotection after embolic stroke in rats. J Neurol Sci 2011; 309:96-101. [PMID: 21802695 DOI: 10.1016/j.jns.2011.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 06/29/2011] [Accepted: 07/06/2011] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Niaspan, an extended-release formulation of niacin (vitamin B3), has been widely used to increase high density lipoprotein (HDL) cholesterol and to prevent cardiovascular diseases and stroke. We have previously demonstrated that Niaspan (40 mg/kg) administered at 2h after stroke induces neuroprotection, while low dose Niaspan (20mg/kg) does not reduce infarct volume. Tissue plasminogen activator (tPA) is an effective therapy for acute stroke, but its use remains limited by a narrow therapeutic window. We have previously demonstrated that intravenous administration of tPA 4h after stroke in rats does not reduce infarct volume. In this study, we tested whether combination treatment with low-dose Niaspan (20mg/kg) and tPA administered 4h after embolic stroke in a rat model reduces infarct volume and provides neuroprotection. METHODS Adult male Wistar rats were subjected to embolic middle cerebral artery occlusion (MCAo) and treated with low-dose Niaspan (20mg/kg) alone (n = 7), tPA (10mg/kg) alone (n = 7), combination of low-dose Niaspan and tPA (n = 7), or saline control (n = 9), 4h after stroke. A battery of functional outcome tests was performed. Rats were sacrificed at 7 days after MCAo and lesion volumes were measured. To investigate the underlying mechanism of combination treatment neuroprotective effect, deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), cleaved caspase-3, tumor necrosis factor alpha (TNF-alpha), and toll-like receptor 4 (TLR-4) immunostaining were performed. RESULTS Combination treatment with low-dose Niaspan and tPA significantly improved functional outcome compared to the saline control group (p<0.05), while treatment with Niaspan or tPA alone did not significantly improve functional outcome compared to saline control group. Additionally, combination treatment significantly reduced infarct volume compared to saline control group (p = 0.006) and infarct volume was significantly correlated with functional outcome (p = 0.0008; r = 0.63). Monotherapy with Niaspan or tPA did not significantly decrease infarct volume compared to saline control group. Combination treatment reduced apoptosis as measured by significant reduction in the number of TUNEL-positive cells and cleaved caspase-3 expression in the ischemic brain compared to saline control group (p<0.05). Combination treatment also significantly reduced the expression of TNF-alpha and TLR-4 in the ischemic brain compared to Niaspan, tPA and saline treatment groups (p<0.05). A significant interaction between Niaspan and tPA on the TNF-alpha expression was detected (p<0.05), indicating a synergy effect in the combination treatment group. CONCLUSION Treatment of stroke with combination of low-dose Niaspan and tPA at 4h after embolic stroke reduces infarct volume, improves neurological outcome and provides neuroprotection. The neuroprotective effects of combination treatment were associated with reduction of apoptosis and attenuation of TNF-alpha and TLR-4 expression.
Collapse
|
62
|
Frentzou GA, Bradford C, Harkness KA, Haddock G, Woodroofe MN, Cross AK. IL-1β down-regulates ADAMTS-13 mRNA expression in cells of the central nervous system. J Mol Neurosci 2011; 46:343-51. [PMID: 21732076 DOI: 10.1007/s12031-011-9591-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/22/2011] [Indexed: 01/06/2023]
Abstract
ADAMTS-13 is the Von Willebrand factor (vWF) cleaving protease, responsible for the cleavage and down-regulation of the pro-thrombotic properties of ultra large VWF multimers. It is expressed predominantly by the hepatic stellate cells of the liver, but is also found to be expressed in other tissues, including brain. Reduced ADAMTS-13 is associated with a variety of thrombotic microangiopathies. Since the cellular origin and regulation of ADAMTS-13 expression in the brain is unknown, we aimed to investigate this in four different central nervous system (CNS)-derived cell lines, SHSY-5Y (human neuroblastoma), U373 (human astroglioma), CHME-3 (human foetal microglia) and hCMEC/D3 (adult human brain endothelial cells). All cell lines expressed ADAMTS-13 mRNA constitutively with neuroblastoma cells showing the highest expression. Interleukin (IL)-1β down-regulated ADAMTS-13 mRNA expression in astroglioma cells and microglial cells whereas TNF and IL-6 treatment showed no significant differences in ADAMTS-13 mRNA expression in any cell line tested. ADAMTS-13 protein expression was reduced in a dose-dependent manner only in astroglioma cells following stimulation by IL-1β. The ability of IL-1β to significantly reduce ADAMTS-13 mRNA expression in human microglia and astroglioma cells suggests a role in the haemostasis of the local microenvironment under inflammatory conditions. This is the first report of ADAMTS-13 expression in cells of the CNS; however, its function remains to be determined.
Collapse
Affiliation(s)
- G Alkistis Frentzou
- Biomedical Research Centre, Faculty of Health and Wellbeing, Sheffield Hallam University, Howard Street, Sheffield, S1 1WB, UK.
| | | | | | | | | | | |
Collapse
|
63
|
Wang T, Jiang N, Han B, Liu W, Liu T, Fu F, Zhao D. Escin attenuates cerebral edema induced by acute omethoate poisoning. Toxicol Mech Methods 2011; 21:400-5. [DOI: 10.3109/15376516.2010.550337] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
64
|
Tanskanen M, Myllykangas L, Saarialho-Kere U, Paetau A. Matrix metalloproteinase-β19 expressed in cerebral amyloid angiopathy. Amyloid 2011; 18:3-9. [PMID: 21261556 DOI: 10.3109/13506129.2010.541960] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy (CAA) is a frequent finding in the brains of patients with Alzheimer's disease (AD). CAA may be complicated with CAA-associated intracerebral haemorrhage (CAAH). Previous studies have revealed matrix metalloproteinase (MMP) expression in a mouse model of CAA and in human intracerebral haemorrhage. Here we studied the involvement of MMPs in human CAA and CAAH. MATERIAL AND METHODS To investigate the putative expression of MMPs in human CAA and CAAH (Step 1), immunohistochemistry (IHC) against MMPs-1, -2, -7, -9, -19 and -26 was applied on tissue microarray (TMA) constructed of cerebral samples from 29 individuals with AD, 15 with CAAH and 2 controls. The findings in TMA were confirmed (Step 2) in tissue samples from 64 individuals, 45 presenting with CAA (including 36 with CAAH) and 19 without CAA (including 11 with hypertensive cerebral haemorrhage). RESULTS In Step 1, immunoreactivity against MMPs-19 and -26 was detected in cerebral blood vessels in CAA. The results were confirmed in Step 2, where CAA (p<0.001) and intracerebral haemorrhage (p=0.045) were associated with vascular immunoreactivity against MMP-19. Multivariate analysis showed that the association between vascular MMP-19 and intracerebral haemorrhage was dependent from CAA. MMP-26 associated with CAA (p=0.021) but not with intracerebral haemorrhage. CONCLUSION This is the first human study showing local MMP-19 immunoreactivity in the Aβ-amyloid-laden blood vessels in CAA, suggesting that MMPs may be involved in CAA.
Collapse
Affiliation(s)
- Maarit Tanskanen
- Department of Pathology, University of Helsinki and Helsinki University Central Hospital, Helsinki, Finland.
| | | | | | | |
Collapse
|
65
|
Vakili A, Mojarrad S, Akhavan MM, Rashidy-Pour A. Pentoxifylline attenuates TNF-α protein levels and brain edema following temporary focal cerebral ischemia in rats. Brain Res 2011; 1377:119-25. [PMID: 21219888 DOI: 10.1016/j.brainres.2011.01.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 12/04/2010] [Accepted: 01/03/2011] [Indexed: 10/18/2022]
Abstract
Cerebral edema is the most common cause of neurological deterioration and mortality during acute ischemic stroke. Despite the clinical importance of cerebral ischemia, the underlying mechanisms remain poorly understood. Recent studies suggest a role for TNF-α in the brain edema formation. To further investigate whether TNF-α would play a role in brain edema formation, we examined the effects of pentoxifylline (PTX, an inhibitor of TNF-α synthesis) on the brain edema and TNF-α levels in a model of transient focal cerebral ischemia. The right middle cerebral artery (MCA) of rats was occluded for 60 min using the intraluminal filament method. The animals received PTX (60 mg/kg) immediately, 1, 3, or 6h post-ischemic induction. Twenty-four hours after induction of ischemic injury, permeability of the blood-brain barrier (BBB) and brain edema were determined by in situ brain perfusion of Evans Blue (EB) and wet-to-dry weight ratio, respectively. TNF-α protein levels in ischemic cortex were also measured at 1, 4, and 24h after the beginning of an ischemic stroke by using an enzyme-linked immunosorbent assay method. The administration of PTX up to 6h after occlusion of the MCA significantly reduced the brain edema. Moreover, PTX significantly reduced the concentration of TNF-α in ischemic brain cortex up to 4h post-transient focal stroke (P<0.002). Finally, treatment by PTX led to a significant decrease in EB extravasations (P<0.001). Our data demonstrate that PTX administration up to 6h after ischemia can reduce brain edema in a model of transient focal cerebral ischemia. The beneficial effects of PTX may be mediated, at least in part, through a decline in TNF-α production and BBB breakdown.
Collapse
Affiliation(s)
- Abedin Vakili
- Department and Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| | | | | | | |
Collapse
|
66
|
Ceulemans AG, Zgavc T, Kooijman R, Hachimi-Idrissi S, Sarre S, Michotte Y. The dual role of the neuroinflammatory response after ischemic stroke: modulatory effects of hypothermia. J Neuroinflammation 2010; 7:74. [PMID: 21040547 PMCID: PMC2988764 DOI: 10.1186/1742-2094-7-74] [Citation(s) in RCA: 230] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 11/01/2010] [Indexed: 12/15/2022] Open
Abstract
Neuroinflammation is a key element in the ischemic cascade after cerebral ischemia that results in cell damage and death in the subacute phase. However, anti-inflammatory drugs do not improve outcome in clinical settings suggesting that the neuroinflammatory response after an ischemic stroke is not entirely detrimental. This review describes the different key players in neuroinflammation and their possible detrimental and protective effects in stroke. Because of its inhibitory influence on several pathways of the ischemic cascade, hypothermia has been introduced as a promising neuroprotective strategy. This review also discusses the influence of hypothermia on the neuroinflammatory response. We conclude that hypothermia exerts both stimulating and inhibiting effects on different aspects of neuroinflammation and hypothesize that these effects are key to neuroprotection.
Collapse
Affiliation(s)
- An-Gaëlle Ceulemans
- Department of Pharmaceutical Chemistry and Drug Analysis, Research Group Experimental Neuropharmacology, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
67
|
Adén U, Favrais G, Plaisant F, Winerdal M, Felderhoff-Mueser U, Lampa J, Lelièvre V, Gressens P. Systemic inflammation sensitizes the neonatal brain to excitotoxicity through a pro-/anti-inflammatory imbalance: key role of TNFalpha pathway and protection by etanercept. Brain Behav Immun 2010; 24:747-58. [PMID: 19861157 DOI: 10.1016/j.bbi.2009.10.010] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2009] [Revised: 10/21/2009] [Accepted: 10/22/2009] [Indexed: 10/20/2022] Open
Abstract
Systemic inflammation sensitizes the perinatal brain to an ischemic/excitotoxic insult but the mechanisms are poorly understood. We hypothesized that the mechanisms involve an imbalance between pro- and anti-inflammatory factors. A well characterized mouse model where a systemic injection of IL-1beta during the first five postnatal days (inflammatory insult) is combined with an intracerebral injection of the glutamatergic analogue ibotenate (excitotoxic insult) at postnatal day 5 was used. Following the inflammatory insult alone, there was a transient induction of IL-1beta and TNFalpha, compared with controls measured by quantitative PCR, ELISA, and Western blot. Following the combined inflammatory and excitotoxic insult, there was an induction of IL-1beta, TNFalpha, and IL-6 but not of IL-10 and TNFR1, indicating an altered pro-/anti-inflammatory balance after IL-1beta sensitized lesion. We then tested the hypothesis that the TNFalpha pathway plays a key role in the sensitization and insult using TNFalpha blockade (etanercept) and TNFalpha(-/-) mice. Etanercept given before the insult did not affect brain damage, but genetic deletion of TNFalpha or TNFalpha blockade by etanercept given after the combined inflammatory and excitotoxic insult reduced brain damage by 50%. We suggest this protective effect was centrally mediated, since systemic TNFalpha administration in the presence of an intact blood-brain barrier did not aggravate the damage and etanercept almost abolished cerebral TNFalpha production. In summary, sensitization was, at least partly, mediated by an imbalance between pro- and anti-inflammatory cytokines. Cerebral TNFalpha played a key role in mediating brain damage after the combined inflammatory and excitatory insult.
Collapse
|
68
|
Williams-Karnesky RL, Stenzel-Poore MP. Adenosine and stroke: maximizing the therapeutic potential of adenosine as a prophylactic and acute neuroprotectant. Curr Neuropharmacol 2010; 7:217-27. [PMID: 20190963 PMCID: PMC2769005 DOI: 10.2174/157015909789152209] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 05/15/2009] [Accepted: 05/18/2009] [Indexed: 12/12/2022] Open
Abstract
Stroke is a leading cause of morbidity and mortality in the United States. Despite intensive research into the development of treatments that lessen the severity of cerebrovascular injury, no major therapies exist. Though the potential use of adenosine as a neuroprotective agent in the context of stroke has long been realized, there are currently no adenosine-based therapies for the treatment of cerebral ischemia and reperfusion. One of the major obstacles to developing adenosine-based therapies for the treatment of stroke is the prevalence of functional adenosine receptors outside the central nervous system. The activities of peripheral immune and vascular endothelial cells are particularly vulnerable to modulation via adenosine receptors. Many of the pathophysiological processes in stroke are a direct result of peripheral immune infiltration into the brain. Ischemic preconditioning, which can be induced by a number of stimuli, has emerged as a promising area of focus in the development of stroke therapeutics. Reprogramming of the brain and immune responses to adenosine signaling may be an underlying principle of tolerance to cerebral ischemia. Insight into the role of adenosine in various preconditioning paradigms may lead to new uses for adenosine as both an acute and prophylactic neuroprotectant.
Collapse
Affiliation(s)
- Rebecca L Williams-Karnesky
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, 3181 Sam Jackson Park Road, Portland, OR 97239, USA.
| | | |
Collapse
|
69
|
Niaspan treatment induces neuroprotection after stroke. Neurobiol Dis 2010; 40:277-83. [PMID: 20554037 DOI: 10.1016/j.nbd.2010.05.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 05/26/2010] [Accepted: 05/30/2010] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Niaspan, an extended-release formulation of Niacin (vitamin B3), has been widely used to increase high density lipoprotein (HDL) cholesterol and to prevent cardiovascular diseases and stroke. In this study, we tested whether Niaspan administered acutely after stroke is neuroprotective. METHODS Adult male rats (n=8/group) were subjected to 2h of middle cerebral artery occlusion (MCAo) and treated with or without different doses of Niaspan (20, 40 or 80 mg/kg) at 2 and 24h after MCAo. A battery of functional outcome tests was performed, and serum HDL and triglycerides were measured. Rats were sacrificed at 7 days after MCAo and lesion volumes were measured. The optimal dose of Niaspan treatment of stroke was chosen for immunostaining: deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), cleaved caspase-3, tumor necrosis factor alpha (TNF-alpha), vascular endothelial growth factor (VEGF) and phosphorylated phosphatidylinositol 3-kinase (p-PI3K). Another set of rats (n=4/group) were killed at 7 days after MCAo for Western blot assay. RESULTS Niaspan dose-dependently reduced infarct volume and improved functional outcome after stroke. No significant difference in HDL and triglyceride levels was detected between Niaspan treatments and MCAo control groups. Niaspan treatment significantly decreased the number of TUNEL-positive cells (105+/-17) and cleaved caspase-3 expression (381+/-33) in the ischemic brain compared to MCAo control (165+/-18; 650+/-61, respectively; p<or=0.05). Niaspan treatment significantly reduced the expression of TNF-alpha (9.7+/-1.1% vs. 16+/-2.2%; p<or=0.05) and negative correlations were observed between the functional tests and the expression of TNF-alpha (r=-0.71, p<or=0.05). Niaspan treatment also significantly increased the expression of VEGF (5.2+/-0.9%) and PI3K/Akt (0.381+/-0.04%) in the ischemic brain compared with non-treated MCAo control (2.6+/-0.4%; 0.24+/-0.03, respectively; p<or=0.05). The functional outcome was positively correlated with p-PI3K (r=0.7, p<or=0.05). CONCLUSIONS Treatment of stroke with Niaspan at 2h after MCAo reduces infarct volume and improves neurological outcome and provides neuroprotection. The neuroprotective effects of Niaspan were associated with reduction of apoptosis and attenuation of TNF-alpha expression. VEGF and the PI3K/Akt pathway may contribute to the Niaspan-induced neuroprotection after stroke.
Collapse
|
70
|
Lescot T, Fulla-Oller L, Palmier B, Po C, Beziaud T, Puybasset L, Plotkine M, Gillet B, Meric P, Marchand-Leroux C. Effect of Acute Poly(ADP-Ribose) Polymerase Inhibition by 3-AB on Blood–Brain Barrier Permeability and Edema Formation after Focal Traumatic Brain Injury in Rats. J Neurotrauma 2010; 27:1069-79. [DOI: 10.1089/neu.2009.1188] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Thomas Lescot
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
| | - Laurence Fulla-Oller
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Bruno Palmier
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Christelle Po
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Tiphaine Beziaud
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Louis Puybasset
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
| | - Michel Plotkine
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Brigitte Gillet
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Philippe Meric
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Catherine Marchand-Leroux
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| |
Collapse
|
71
|
Liu J, Feng ZC. Increased umbilical cord plasma interleukin-1 beta levels was correlated with adverse outcomes of neonatal hypoxic-ischemic encephalopathy. J Trop Pediatr 2010; 56:178-182. [PMID: 19822562 DOI: 10.1093/tropej/fmp098] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
To explore whether or not the umbilical blood levels of cytokines can be used to indicate the adverse outcomes of hypoxic-ischemic encephalopathy (HIE) patients. Umbilical artery blood and peripheral venous blood samples were collected on the 1st, 3rd and 7th days after birth to detect the levels of IL-1 beta, IL-8 and TNF-alpha. Neurological examination and Denver developmental screening test (DDST-II) were performed at the 6 and 12 months evaluations to detect any neurodevelopmental abnormalities. The results showed: (i) the serum concentrations of IL-1 beta, IL-8 and TNF-alpha in umbilical and peripheral blood were significantly higher in HIE patients than control groups; (ii) the umbilical blood concentrations of IL-1 beta exhibited the best positive correlation with HIE grades, when compared with IL-8 and TNF-alpha; and (iii) abnormal neurological outcomes at 6 and 12 months of age were best predicted by umbilical levels of IL-1 beta. Thus, umbilical concentrations of IL-1 beta were associated with the grades and adverse outcomes of HIE.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neonatology and NICU Feng, Bayi Children's Hospital Affiliated with Beijing Military Region General Hospital, Beijing 100700, China.
| | | |
Collapse
|
72
|
Vinukonda G, Csiszar A, Hu F, Dummula K, Pandey NK, Zia MT, Ferreri NR, Ungvari Z, LaGamma EF, Ballabh P. Neuroprotection in a rabbit model of intraventricular haemorrhage by cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-alpha inhibition. ACTA ACUST UNITED AC 2010; 133:2264-80. [PMID: 20488889 DOI: 10.1093/brain/awq107] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intraventricular haemorrhage is a major complication of prematurity that results in neurological dysfunctions, including cerebral palsy and cognitive deficits. No therapeutic options are currently available to limit the catastrophic brain damage initiated by the development of intraventricular haemorrhage. As intraventricular haemorrhage leads to an inflammatory response, we asked whether cyclooxygenase-2, its derivative prostaglandin E2, prostanoid receptors and pro-inflammatory cytokines were elevated in intraventricular haemorrhage; whether their suppression would confer neuroprotection; and determined how cyclooxygenase-2 and cytokines were mechanistically-linked. To this end, we used our rabbit model of intraventricular haemorrhage where premature pups, delivered by Caesarian section, were treated with intraperitoneal glycerol at 2 h of age to induce haemorrhage. Intraventricular haemorrhage was diagnosed by head ultrasound at 6 h of age. The pups with intraventricular haemorrhage were treated with inhibitors of cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-α; and cell-infiltration, cell-death and gliosis were compared between treated-pups and vehicle-treated controls during the first 3 days of life. Neurobehavioural performance, myelination and gliosis were assessed in pups treated with cyclooxygenase-2 inhibitor compared to controls at Day 14. We found that both protein and messenger RNA expression of cyclooxygenase-2, prostaglandin E2, prostanoid receptor-1, tumour necrosis factor-α and interleukin-1β were consistently higher in the forebrain of pups with intraventricular haemorrhage relative to pups without intraventricular haemorrhage. However, cyclooxygenase-1 and prostanoid receptor 2-4 levels were comparable in pups with and without intraventricular haemorrhage. Cyclooxygenase-2, prostanoid receptor-1 or tumour necrosis factor-α inhibition reduced inflammatory cell infiltration, apoptosis, neuronal degeneration and gliosis around the ventricles of pups with intraventricular haemorrhage. Importantly, cyclooxygenase-2 inhibition alleviated neurological impairment, improved myelination and reduced gliosis at 2 weeks of age. Cyclooxygenase-2 or prostanoid receptor-1 inhibition reduced tumour necrosis factor-α level, but not interleukin-1β. Conversely, tumour necrosis factor-α antagonism did not affect cyclooxygenase-2 expression. Hence, prostanoid receptor-1 and tumour necrosis factor-α are downstream to cyclooxygenase-2 in the inflammatory cascade induced by intraventricular haemorrhage, and cyclooxygenase-2-inhibition or suppression of downstream molecules--prostanoid receptor-1 or tumour necrosis factor-α--might be a viable neuroprotective strategy for minimizing brain damage in premature infants with intraventricular haemorrhage.
Collapse
Affiliation(s)
- Govindaiah Vinukonda
- Department of Paediatrics, New York Medical College-Westchester Medical Centre, Valhalla, NY 10595, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Lee ST, Chu K, Jung KH, Kang KM, Kim JH, Bahn JJ, Jeon D, Kim M, Lee SK, Roh JK. Cholinergic anti-inflammatory pathway in intracerebral hemorrhage. Brain Res 2009; 1309:164-71. [PMID: 19900419 DOI: 10.1016/j.brainres.2009.10.076] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Revised: 10/30/2009] [Accepted: 10/30/2009] [Indexed: 01/25/2023]
Abstract
Stimulated vagus nerve excretes acetylcholine into the peripheral immune organs such as the spleen, reducing innate inflammation. Here, we investigated whether activation of this "cholinergic anti-inflammatory pathway" can be used to reduce cerebral inflammation in a model of hemorrhagic stroke. Experimental intracerebral hemorrhage (ICH) was induced by stereotaxic collagenase injection in rats. Muscarine, an activator of the vagus nerve, or phosphate-buffered saline (control) was injected into the lateral ventricle after induction of ICH. Intraventricular muscarine injection increased heart rate variability in the ICH model, suggesting increased vagus nerve output. Muscarine-injected ICH rats showed improved neurologic outcomes, reduced brain water content, and decreased levels of inflammatory mediators in both brain and spleen. Central muscarine injection was ineffective at reducing cerebral edema without spleen, suggesting that the effect of muscarine is mediated through the vagus nerve-spleen pathway rather than through a direct interaction with the brain. Our results suggest that the brain possesses a cholinergic anti-inflammatory pathway that counteracts the inflammatory responses after ICH, thereby limiting damage to the brain itself.
Collapse
Affiliation(s)
- Soon-Tae Lee
- Stroke and Stem cell Laboratory in Clinical Research Institute, Stem Cell Research Center, Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Reyes R, Guo M, Swann K, Shetgeri SU, Sprague SM, Jimenez DF, Barone CM, Ding Y. Role of tumor necrosis factor-alpha and matrix metalloproteinase-9 in blood-brain barrier disruption after peripheral thermal injury in rats. J Neurosurg 2009; 110:1218-26. [PMID: 19199470 DOI: 10.3171/2008.8.jns08382] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECT A relationship has been found between peripheral thermal injury and cerebral complications leading to injury and death. In the present study, the authors examined whether tumor necrosis factor-alpha (TNF-alpha) and matrix metalloproteinase-9 (MMP-9) play a causative role in blood-brain barrier (BBB) disruption after peripheral thermal injury. METHODS Thirty-two male Sprague-Dawley rats were subjected to thermal injury. One hour later, 8 rats were injected with TNF-alpha neutralizing antibody, and 8 were injected with doxycycline, an inhibitor of the MMP family proteins; 16 rats did not receive any treatment. Brain tissue samples obtained 7 hours after injury in the treated animals were examined for BBB function by using fluorescein isothiocyanate-dextran and by assessing parenchymal water content. Protein expression of basement membrane components (collagen IV, laminin, and fibronectin) was quantified on Western blot analysis, and MMP-9 protein expression and enzyme activity were determined using Western blot and gelatin zymography. Thermally injured rats that did not receive treatment were killed at 3, 7, or 24 hours after injury and tested for BBB functioning at each time point. Histological analysis for basement membrane proteins was also conducted in untreated rats killed at 7 hours after injury. Results of testing in injured rats were compared with those obtained in a control group of rats that did not undergo thermal injury. RESULTS At 7 hours after thermal injury, a significant increase in the fluorescein isothiocyanate-dextran and water content of the brain was found (p < 0.05), but BBB dysfunction was significantly decreased in the rats that received TNF-alpha antibody or doxycycline (p < 0.05). In addition, the components of the basal lamina were significantly decreased at 7 hours after thermal injury (p < 0.01), and there were significant increases in MMP-9 protein expression and enzyme activity (p < 0.05). The basal lamina damage was reversed by inhibition of TNF-alpha and MMP-9, and the increase in MMP-9 protein was reduced in the presence of doxycycline (p < 0.05). The authors found that MMP-9 enzyme activity was significantly increased after thermal injury (p < 0.01) but decreased in the presence of either TNF-alpha antibody or doxycycline (p < 0.01). CONCLUSIONS The dual, inhibitory activity of both TNF-alpha and MMP-9 in brain injury suggests that a TNF-alpha and MMP-9 cascade may play a key role in BBB disruption. These results offer a better understanding of the pathophysiology of burn injuries, which may open new avenues for burn treatment beyond the level of current therapies.
Collapse
Affiliation(s)
- Raul Reyes
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, Texas, USA
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Abstract
Stroke is the major cause of disability in the Western world and is the third greatest cause of death, but there are no widely effective treatments to prevent the devastating effects of stroke. Extensive and growing evidence implicates inflammatory and immune processes in the occurrence of stroke and particularly in the subsequent injury. Several inflammatory mediators have been identified in the pathogenesis of stroke including specific cytokines, adhesion molecules, matrix metalloproteinases, and eicosanoids. An early clinical trial suggests that inhibiting interleukin-1 may be of benefit in the treatment of acute stroke.
Collapse
|
76
|
Badiola N, Malagelada C, Llecha N, Hidalgo J, Comella JX, Sabriá J, Rodríguez-Alvarez J. Activation of caspase-8 by tumour necrosis factor receptor 1 is necessary for caspase-3 activation and apoptosis in oxygen-glucose deprived cultured cortical cells. Neurobiol Dis 2009; 35:438-47. [PMID: 19555759 DOI: 10.1016/j.nbd.2009.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Revised: 05/22/2009] [Accepted: 06/16/2009] [Indexed: 01/08/2023] Open
Abstract
TNF-alpha has been reported to be relevant in stroke-induced neuronal death. However the precise function of TNF-alpha in brain ischemia remains controversial since there are data supporting either a detrimental or a protective effect. Here we show that TNF-alpha is released after oxygen-glucose deprivation (OGD) of cortical cultures and is a major contributor to the apoptotic death observed without affecting the OGD-mediated necrotic cell death. In this paradigm, apoptosis depends on TNF-alpha-induced activation of caspase-8 and -3 without affecting the activation of caspase-9. By using knock-out mice for TNF-alpha receptor 1, we show that the activation of both caspase-3 and -8 by TNF-alpha is mediated by TNF-alpha receptor 1. The pro-apoptotic role of TNF-alpha in OGD is restricted to neurons and microglia, since astrocytes do not express either TNF-alpha or TNF-alpha receptor 1. Altogether, these results show that apoptosis of cortical neurons after OGD is mediated by TNF-alpha/TNF-alpha receptor 1.
Collapse
Affiliation(s)
- Nahuai Badiola
- Institut de Neurociencies, Departament de Bioquímica i Biología Molecular, Universitat Autònoma de Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
77
|
Bruno RDB, Marques TF, Batista TMT, Lima JCSS, de Arruda KG, Lima PFSF, Santos NDS, Cunha GM, Vitor HVN, Viana GSDB. Pentoxifylline treatment improves neurological and neurochemical deficits in rats subjected to transient brain ischemia. Brain Res 2009; 1260:55-64. [DOI: 10.1016/j.brainres.2008.12.064] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 12/16/2008] [Accepted: 12/17/2008] [Indexed: 11/29/2022]
|
78
|
Yagi K, Kitazato KT, Uno M, Tada Y, Kinouchi T, Shimada K, Nagahiro S. Edaravone, a Free Radical Scavenger, Inhibits MMP-9–Related Brain Hemorrhage in Rats Treated With Tissue Plasminogen Activator. Stroke 2009; 40:626-31. [DOI: 10.1161/strokeaha.108.520262] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Kenji Yagi
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Keiko T. Kitazato
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Masaaki Uno
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yoshiteru Tada
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Tomoya Kinouchi
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Kenji Shimada
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Shinji Nagahiro
- From the Department of Neurosurgery, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| |
Collapse
|
79
|
Pettigrew LC, Kindy MS, Scheff S, Springer JE, Kryscio RJ, Li Y, Grass DS. Focal cerebral ischemia in the TNFalpha-transgenic rat. J Neuroinflammation 2008; 5:47. [PMID: 18947406 PMCID: PMC2583993 DOI: 10.1186/1742-2094-5-47] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Accepted: 10/23/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND To determine if chronic elevation of the inflammatory cytokine, tumor necrosis factor-alpha (TNFalpha), will affect infarct volume or cortical perfusion after focal cerebral ischemia. METHODS Transgenic (TNFalpha-Tg) rats overexpressing the murine TNFalpha gene in brain were prepared by injection of mouse DNA into rat oocytes. Brain levels of TNFalpha mRNA and protein were measured and compared between TNFalpha-Tg and non-transgenic (non-Tg) littermates. Mean infarct volume was calculated 24 hours or 7 days after one hour of reversible middle cerebral artery occlusion (MCAO). Cortical perfusion was monitored by laser-Doppler flowmetry (LDF) during MCAO. Cortical vascular density was quantified by stereology. Post-ischemic cell death was assessed by immunohistochemistry and regional measurement of caspase-3 activity or DNA fragmentation. Unpaired t tests or analysis of variance with post hoc tests were used for comparison of group means. RESULTS In TNFalpha-Tg rat brain, the aggregate mouse and rat TNFalpha mRNA level was fourfold higher than in non-Tg littermates and the corresponding TNFalpha protein level was increased fivefold (p CONCLUSION Chronic elevation of TNFalpha protein in brain increases susceptibility to ischemic injury but has no effect on vascular density. TNFalpha-Tg animals are more susceptible to apoptotic cell death after MCAO than are non-Tg animals. We conclude that the TNFalpha-Tg rat is a valuable new tool for the study of cytokine-mediated ischemic brain injury.
Collapse
Affiliation(s)
- L Creed Pettigrew
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
- Department of Neurology, University of Kentucky, Lexington, Kentucky, USA
- Veterans Administration (VA) Medical Center, Lexington, Kentucky, USA
| | - Mark S Kindy
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina, USA
- Ralph H. Johnson VA Medical Center, Charleston, South Carolina, USA
| | - Stephen Scheff
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Joe E Springer
- Department of Physical Medicine & Rehabilitation, University of Kentucky, Lexington, Kentucky, USA
| | - Richard J Kryscio
- Department of Statistics and School of Public Health, University of Kentucky, Lexington, Kentucky, USA
| | - Yizhao Li
- Jinan Great Wall Hospital, Jinan, Shandong, PR China
| | | |
Collapse
|
80
|
Kim Y, So HS, Moon BS, Youn MJ, Kim HJ, Shin YI, Moon SK, Song MS, Choi KY, Song J, Park R. Sasim attenuates LPS-induced TNF-alpha production through the induction of HO-1 in THP-1 differentiated macrophage-like cells. JOURNAL OF ETHNOPHARMACOLOGY 2008; 119:122-128. [PMID: 18602978 DOI: 10.1016/j.jep.2008.06.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 05/26/2008] [Accepted: 06/11/2008] [Indexed: 05/26/2023]
Abstract
AIM OF THE STUDY Sasim, a traditional prescription composed of seven herbal mixtures, has been widely used as an oriental medicine for the treatment of cerebral infarction in Korea. However, the regulatory mechanisms by which the formula affects immune processing in cerebral infarction patients remain unknown. MATERIALS AND METHODS The levels of secretory protein of tumor necrosis factor (TNF)-alpha were determined in both THP-1 differentiated macrophage-like (THP-1/M) cells and Peripheral blood mononuclear cells (PBMCs) from cerebral infarction patients. Also, the levels of protein and mRNA of TNF-alpha and heme oxygenase-1 (HO-1) were detected in THP-1/M cells under our experimental condition. RESULTS Sasim markedly suppressed lipopolysaccharide (LPS)-induced TNF-alpha at the levels of secretory protein and mRNA in both PBMCs from cerebral infarction patients and THP-1/M cells. Interestingly, Sasim strongly induced HO-1, the rate-limiting enzyme of heme catabolism, at both the protein and mRNA levels in THP-1/M cells. Treatment with tin protoporphyrin IX (SnPP), an inhibitor of the catalytic activity of HO, significantly abolished the suppressive effect of Sasim on LPS-induced TNF-a production in THP-1/M cells. CONCLUSIONS These data indicate that Sasim may be beneficial in the cessation of inflammatory processes associated with cerebral infarction through the induction of HO-1 expression.
Collapse
Affiliation(s)
- Yunha Kim
- Vestibulocochlear System Research Center and Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk 570-749, South Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Iwanami J, Mogi M, Li JM, Tsukuda K, Min LJ, Sakata A, Fujita T, Iwai M, Horiuchi M. Deletion of Angiotensin II Type 2 Receptor Attenuates Protective Effects of Bone Marrow Stromal Cell Treatment on Ischemia–Reperfusion Brain Injury in Mice. Stroke 2008; 39:2554-9. [DOI: 10.1161/strokeaha.107.513275] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Jun Iwanami
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaki Mogi
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Jian-Mei Li
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Kana Tsukuda
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Li-Juan Min
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Akiko Sakata
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Teppei Fujita
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masaru Iwai
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| | - Masatsugu Horiuchi
- From the Department of Molecular Cardiovascular Biology and Pharmacology, Ehime University, Graduate School of Medicine, Tohon, Ehime, Japan
| |
Collapse
|
82
|
Sandoval KE, Witt KA. Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol Dis 2008; 32:200-19. [PMID: 18790057 DOI: 10.1016/j.nbd.2008.08.005] [Citation(s) in RCA: 757] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2008] [Revised: 07/29/2008] [Accepted: 08/10/2008] [Indexed: 01/01/2023] Open
Abstract
The blood-brain barrier (BBB) is formed by the endothelial cells of cerebral microvessels, providing a dynamic interface between the peripheral circulation and the central nervous system. The tight junctions (TJs) between the endothelial cells serve to restrict blood-borne substances from entering the brain. Under ischemic stroke conditions decreased BBB TJ integrity results in increased paracellular permeability, directly contributing to cerebral vasogenic edema, hemorrhagic transformation, and increased mortality. This loss of TJ integrity occurs in a phasic manner, which is contingent on several interdependent mechanisms (ionic dysregulation, inflammation, oxidative and nitrosative stress, enzymatic activity, and angiogenesis). Understanding the inter-relation of these mechanisms is critical for the development of new therapies. This review focuses on those aspects of ischemic stroke impacting BBB TJ integrity and the principle regulatory pathways, respective to the phases of paracellular permeability.
Collapse
Affiliation(s)
- Karin E Sandoval
- Department of Pharmaceutical Sciences, School of Pharmacy, Southern Illinois University Edwardsville, Edwardsville, IL 62026, USA
| | | |
Collapse
|
83
|
Abstract
Central nervous system (CNS) infections presenting to the emergency room include meningitis, encephalitis, brain and spinal epidural abscess, subdural empyema, and ventriculitis. These conditions often require admission to an intensive care unit (ICU) and are complications of ICU patients with neurologic injury, contributing significantly to morbidity and mortality. Reducing morbidity and mortality is critically dependent on rapid diagnosis and, perhaps more importantly, on the timely initiation of appropriate antimicrobial therapy. New insights into the role of inflammation and the immune response in CNS infections have contributed to development of new diagnostic strategies using markers of inflammation, and to the study of agents with focused immunomodulatory activity, which may lead to further adjunctive therapy in human disease.
Collapse
|
84
|
Guo M, Lin V, Davis W, Huang T, Carranza A, Sprague S, Reyes R, Jimenez D, Ding Y. Preischemic induction of TNF-alpha by physical exercise reduces blood-brain barrier dysfunction in stroke. J Cereb Blood Flow Metab 2008; 28:1422-30. [PMID: 18414498 DOI: 10.1038/jcbfm.2008.29] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
This study explores the neuroprotective action of tumor necrosis factor-alpha (TNF-alpha) induced during physical exercise, which, consequently, reduces matrix metalloproteinase-9 (MMP-9) activity and ameliorates blood-brain barrier (BBB) dysfunction in association with extracellular signal-regulated kinase 1 and 2 (ERK1/2) phosphorylation. Adult male Sprague-Dawley rats were subjected to exercise on a treadmill for 3 weeks. A 2-h middle cerebral artery occlusion and reperfusion was administered to exercised and nonexercised animals to induce stroke. Exercised ischemic rats were subjected to TNF-alpha inhibition and ERK1/2 by TNF-alpha antibody or UO126. Nissl staining of coronal sections revealed the infarct volume. Evans blue extravasation and water content evaluated BBB function. Western blot was performed to analyze protein expression of TNF-alpha, ERK1/2, phosphorylated ERK1/2, the basal laminar protein collagen IV, and MMP-9. The activity of MMP-9 was determined by gelatin zymography. Tumor necrosis factor-alpha expression and ERK1/2 phosphorylation were upregulated during exercise. Infarct volume, brain edema, and Evans blue extravasation all significantly decreased in exercised ischemic rats. Collagen IV production increased in exercised rats and remained high after stroke, whereas MMP-9 protein level and activity decreased. These results were negated and returned toward nonexercised values once TNF-alpha or ERK1/2 was blocked. We concluded that preischemic, exercise-induced TNF-alpha markedly decreases BBB dysfunction by using the ERK1/2 pathway.
Collapse
Affiliation(s)
- Miao Guo
- Department of Neurosurgery, The University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Adibhatla RM, Hatcher JF. Tissue plasminogen activator (tPA) and matrix metalloproteinases in the pathogenesis of stroke: therapeutic strategies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2008; 7:243-53. [PMID: 18673209 PMCID: PMC2562687 DOI: 10.2174/187152708784936608] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Today there exists only one FDA-approved treatment for ischemic stroke; i.e., the serine protease tissue-type plasminogen activator (tPA). In the aftermath of the failed stroke clinical trials with the nitrone spin trap/radical scavenger, NXY-059, a number of articles raised the question: are we doing the right thing? Is the animal research truly translational in identifying new agents for stroke treatment? This review summarizes the current state of affairs with plasminogen activators in thrombolytic therapy. In addition to therapeutic value, potential side effects of tPA also exist that aggravate stroke injury and offset the benefits provided by reperfusion of the occluded artery. Thus, combinational options (ultrasound alone or with microspheres/nanobubbles, mechanical dissociation of clot, activated protein C (APC), plasminogen activator inhibitor-1 (PAI-1), neuroserpin and CDP-choline) that could offset tPA toxic side effects and improve efficacy are also discussed here. Desmoteplase, a plasminogen activator derived from the saliva of Desmodus rotundus vampire bat, antagonizes vascular tPA-induced neurotoxicity by competitively binding to low-density lipoprotein related-receptors (LPR) at the blood-brain barrier (BBB) interface, minimizing the tPA uptake into brain parenchyma. tPA can also activate matrix metalloproteinases (MMPs), a family of endopeptidases comprised of 24 mammalian enzymes that primarily catalyze the turnover and degradation of the extracellular matrix (ECM). MMPs have been implicated in BBB breakdown and neuronal injury in the early times after stroke, but also contribute to vascular remodeling, angiogenesis, neurogenesis and axonal regeneration during the later repair phase after stroke. tPA, directly or by activation of MMP-9, could have beneficial effects on recovery after stroke by promoting neurovascular repair through vascular endothelial growth factor (VEGF). However, any treatment regimen directed at MMPs must consider their pleiotropic nature and the likelihood of either beneficial or detrimental effects that might depend on the timing of the treatment in relation to the stage of brain injury.
Collapse
Affiliation(s)
- Rao Muralikrishna Adibhatla
- Department of Neurological Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | | |
Collapse
|
86
|
Obrenovitch TP. Molecular physiology of preconditioning-induced brain tolerance to ischemia. Physiol Rev 2008; 88:211-47. [PMID: 18195087 DOI: 10.1152/physrev.00039.2006] [Citation(s) in RCA: 176] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Ischemic tolerance describes the adaptive biological response of cells and organs that is initiated by preconditioning (i.e., exposure to stressor of mild severity) and the associated period during which their resistance to ischemia is markedly increased. This topic is attracting much attention because preconditioning-induced ischemic tolerance is an effective experimental probe to understand how the brain protects itself. This review is focused on the molecular and related functional changes that are associated with, and may contribute to, brain ischemic tolerance. When the tolerant brain is subjected to ischemia, the resulting insult severity (i.e., residual blood flow, disruption of cellular transmembrane gradients) appears to be the same as in the naive brain, but the ensuing lesion is substantially reduced. This suggests that the adaptive changes in the tolerant brain may be primarily directed against postischemic and delayed processes that contribute to ischemic damage, but adaptive changes that are beneficial during the subsequent test insult cannot be ruled out. It has become clear that multiple effectors contribute to ischemic tolerance, including: 1) activation of fundamental cellular defense mechanisms such as antioxidant systems, heat shock proteins, and cell death/survival determinants; 2) responses at tissue level, especially reduced inflammatory responsiveness; and 3) a shift of the neuronal excitatory/inhibitory balance toward inhibition. Accordingly, an improved knowledge of preconditioning/ischemic tolerance should help us to identify neuroprotective strategies that are similar in nature to combination therapy, hence potentially capable of suppressing the multiple, parallel pathophysiological events that cause ischemic brain damage.
Collapse
Affiliation(s)
- Tihomir Paul Obrenovitch
- Division of Pharmacology, School of Life Sciences, University of Bradford, Bradford, United Kingdom.
| |
Collapse
|
87
|
Kim Y, So HS, Kim SJ, Youn MJ, Lee JH, Kim NS, Lee JH, Woo WH, Lee DW, Cho KH, Moon BS, Park R. Antiinflammatory effect of Daesiho, a Korean traditional prescription for cerebral infarct patients. Phytother Res 2008; 22:829-35. [DOI: 10.1002/ptr.2389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
88
|
Lee ST, Chu K, Jung KH, Kim SJ, Kim DH, Kang KM, Hong NH, Kim JH, Ban JJ, Park HK, Kim SU, Park CG, Lee SK, Kim M, Roh JK. Anti-inflammatory mechanism of intravascular neural stem cell transplantation in haemorrhagic stroke. ACTA ACUST UNITED AC 2007; 131:616-29. [PMID: 18156155 DOI: 10.1093/brain/awm306] [Citation(s) in RCA: 314] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neural stem cell (NSC) transplantation has been investigated as a means to reconstitute the damaged brain after stroke. In this study, however, we investigated the effect on acute cerebral and peripheral inflammation after intracerebral haemorrhage (ICH). NSCs (H1 clone) from fetal human brain were injected intravenously (NSCs-iv, 5 million cells) or intracerebrally (NSCs-ic, 1 million cells) at 2 or 24 h after collagenase-induced ICH in a rat model. Only NSCs-iv-2 h resulted in fewer initial neurologic deteriorations and reduced brain oedema formation, inflammatory infiltrations (OX-42, myeloperoxidase) and apoptosis (activated caspase-3, TUNEL) compared to the vehicle-injected control animals. Rat neurosphere-iv-2 h, but not human fibroblast-iv-2 h, also reduced the brain oedema and the initial neurologic deficits. Human NSCs-iv-2 h also attenuated both cerebral and splenic activations of tumour necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and nuclear factor-kappa B (NF-kappaB). However, we observed only a few stem cells in brain sections of the NSCs-iv-2 h group; in the main, they were detected in marginal zone of spleens. To investigate whether NSCs interact with spleen to reduce cerebral inflammation, we performed a splenectomy prior to ICH induction, which eliminated the effect of NSCs-iv-2 h transplantation on brain water content and inflammatory infiltrations. NSCs also inhibited in vitro macrophage activations after lipopolysaccharide stimulation in a cell-to-cell contact dependent manner. In summary, early intravenous NSC injection displayed anti-inflammatory functionality that promoted neuroprotection, mainly by interrupting splenic inflammatory responses after ICH.
Collapse
Affiliation(s)
- Soon-Tae Lee
- Stroke & Stem Cell Laboratory, Clinical Research Institute, Stem Cell Research Center, Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Kim Y, So HS, Youn MJ, Kim ES, Song MS, Chai KY, Woo WH, Cho KH, Moon BS, Park R. Anti-inflammatory effect of So-Pung-Tang, a Korean traditional prescription for cerebral infarction patients. JOURNAL OF ETHNOPHARMACOLOGY 2007; 114:425-431. [PMID: 17931810 DOI: 10.1016/j.jep.2007.08.041] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Revised: 07/18/2007] [Accepted: 08/16/2007] [Indexed: 05/25/2023]
Abstract
So-Pung-Tang (Sopung), a prescription composed of 14 herbal mixtures, has been widely used in the treatment of cerebral infarction in Oriental Medicine. However, the mechanisms by which the formula affects on the production of pro-inflammatory cytokines in cerebral infarction patients remain unknown yet. The levels of secretory protein of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-alpha, interlukin (IL)-1beta, and IL-6, were significantly increased in both THP-1 differentiated macrophage-like cells (THP-1/M) and peripheral blood mononuclear cells (PBMCs) from cerebral infarction patients after stimulation. However, pretreatment with Sopung markedly inhibited the secretion of TNF-alpha and IL-6, but not IL-1beta, in lipopolysaccharide (LPS)-stimulated THP-1/M cells and PBMCs treated with LPS and phytohemagglutinin (PHA). Furthermore, Sopung significantly inhibited LPS-induced phosphorylation of extracellular signal-regulated kinase (ERK1/2) and c-jun N-terminal kinase (JNK), but not p38 in THP-1/M cells. These data indicate that Sopung may be beneficial in the cessation of inflammatory processes of cerebral infarction through suppression of ERK1/2 and JNK activation.
Collapse
Affiliation(s)
- Yunha Kim
- Vestibulocochlear System Research Center and Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk 570-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Campbell SJ, Jiang Y, Davis AEM, Farrands R, Holbrook J, Leppert D, Anthony DC. Immunomodulatory effects of etanercept in a model of brain injury act through attenuation of the acute-phase response. J Neurochem 2007; 103:2245-55. [PMID: 17883399 DOI: 10.1111/j.1471-4159.2007.04928.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
TNF-alpha has proved to be a successful target in the treatment of many peripheral inflammatory diseases, but the same interventions worsen immune-mediated CNS disease. However, anti-TNF-alpha strategies may offer promise as therapy for non-immune CNS injury. In this study, we have microinjected IL-1beta or lipopolysaccharide (LPS) into the rat brain as a simple model of brain injury and have systemically administered the TNF-alpha antagonist etanercept to discover whether hepatic TNF-alpha, produced as part of the acute-phase response to CNS injury, modulates the inflammatory response in the brain. We report a significant reduction in neutrophil numbers recruited to the IL-1beta- or LPS-challenged brain as a result of TNF-alpha inhibition. We also show an attenuation in the levels of hepatic mRNA including TNF-alpha mRNA and of TNF-alpha-induced genes, such as the chemokines CCL-2, CXCL-5, and CXCL-10, although other chemokines elevated by the injury were not significantly changed. The reduction in hepatic chemokine synthesis results in reduced numbers of circulating neutrophils, and also a reduction in the numbers recruited to the liver as a consequence of brain injury. These findings suggest that TNF-alpha inhibitors may reduce CNS inflammatory responses by targeting the hepatic acute-phase response, and thus therapies for brain injury need not cross the blood-brain barrier to be effective.
Collapse
Affiliation(s)
- Sandra J Campbell
- Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford, UK
| | | | | | | | | | | | | |
Collapse
|
91
|
Lapchak PA. Tumor necrosis factor-α is involved in thrombolytic-induced hemorrhage following embolic strokes in rabbits. Brain Res 2007; 1167:123-8. [PMID: 17673188 DOI: 10.1016/j.brainres.2007.06.072] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 06/15/2007] [Accepted: 06/15/2007] [Indexed: 12/14/2022]
Abstract
The present study assessed whether tumor necrosis factor-alpha (TNFalpha) is involved in hemorrhage following large clot embolism-induced ischemia in New Zealand white rabbits by intracisternally administering either TNFalpha or a goat-anti-rabbit-TNFalpha antibody following a stroke. The first aim of the study showed that TNFalpha administration increased stroke-induced hemorrhage incidence to 53.3% from 18.5% (an increase of 188%) in the control group and also increased hemorrhage volume by 87% (p<0.05). The second aim showed that administration of tissue plasminogen activator (tPA) using a standard dose of 3.3 mg/kg increased hemorrhage incidence in rabbits to 76.5% from 18.5% (an increase of 314%) and this effect was reversed by administration of an anti-TNFalpha antibody. In the tPA-anti-TNFalpha antibody group, the absolute hemorrhage rate was 38.8% and the hemorrhage volume was 98% of control. In conclusion, following an embolic stroke, TNFalpha administration increased the incidence and volume of hemorrhage and an anti-TNFalpha antibody counteracted tPA-induced hemorrhage. The results suggest that TNFalpha may either be directly or indirectly involved in vascular damage following an embolic stroke. Moreover, TNFalpha may mediate some of the detrimental effects of tPA on the vascular compartment. Based upon our studies, TNFalpha receptor antagonists or TNFalpha processing inhibitors should be further pursued as targets for the treatment of hemorrhagic stroke as adjuvant treatment for stroke patients receiving thrombolytic treatment.
Collapse
Affiliation(s)
- Paul A Lapchak
- Department of Neuroscience, University of California San Diego, La Jolla, CA 92093-0624, USA.
| |
Collapse
|
92
|
Kim Y, So HS, Kim JK, Park C, Lee JH, Woo WH, Cho KH, Moon BS, Park R. Anti-inflammatory effect of oyaksungisan in peripheral blood mononuclear cells from cerebral infarction patients. Biol Pharm Bull 2007; 30:1037-41. [PMID: 17541150 DOI: 10.1248/bpb.30.1037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oyaksungisan, the herbal prescription composed of eleven herbs, has been widely used in treatment of cerebral infarct in Oriental Medicine. However, the mechanisms by which the herbal formula affects on the production of pro- and anti-inflammatory cytokines in cerebral infarction patients remain unknown yet. The secretory levels of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta, and IL-6, and IL-10 were significantly increased in both LPS and PHA-stimulated peripheral blood mononuclear cells (PBMCs) from cerebral infarction patients. However, pretreatment with oyaksungisan significantly inhibited the secretion of pro- and anti-inflammatory in PBMCs. Also, oyaksungisan induced a significant increase of transforming growth factor (TGF)-beta1 in PBMCs. Thus, these data indicate that oyaksungisan may be beneficial in the cessation of inflammatory processes of cerebral infarct through suppression of TNF-alpha, IL-1beta, IL-6, and IL-10 and induction of TGF-beta1.
Collapse
Affiliation(s)
- Yunha Kim
- Department of Microbiology, Wonkwang University School of Medicine, Jeonbuk, Korea (South)
| | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Kim Y, So HS, Youn MJ, Kim HJ, Woo WH, Shin SH, Lee I, Moon BS, Cho KH, Park R. Anti-inflammatory effect of Sasim extracts in PHA-stimulated THP-1 and peripheral blood mononuclear cells from cerebral infarction patients. JOURNAL OF ETHNOPHARMACOLOGY 2007; 112:32-9. [PMID: 17349756 DOI: 10.1016/j.jep.2007.01.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 12/26/2006] [Accepted: 01/25/2007] [Indexed: 05/14/2023]
Abstract
Sasim, a prescription composed of seven herbal mixtures, has been widely used for the treatment of cerebral infarction as an oriental medicine in Korea. However, the mechanisms by which the formula affects on the production of pro-inflammatory cytokines in cerebral infarct patients remain unknown yet. The levels of secretory protein and mRNA of pro-inflammatory cytokines, including tumor necrosis factor (TNF)-alpha, interlukin (IL)-1beta, and IL-6, were significantly increased in both THP-1 differentiated macrophage-like cells (T/M) and peripheral blood mononuclear cells (PBMCs) from cerebral infarct patients at 24h after stimulation with phytohemagglutinin (PHA) (p<0.05). However, pretreatment of Sasim strongly suppressed the secretion of pro-inflammatory cytokines in PHA-stimulated T/M cells and PBMCs. Moreover, Sasim significantly suppressed the transcriptional levels of pro-inflammatory cytokines in PHA-stimulated THP-1/M cells. These data indicate that Sasim may be beneficial in the cessation of inflammatory processes of cerebral infarction through suppression on the production of pro-inflammatory cytokines via inhibition of mRNA expression.
Collapse
Affiliation(s)
- Yunha Kim
- Vestibulocochlear System Research Center and Department of Microbiology, Wonkwang University School of Medicine, Iksan, Jeonbuk 570-749, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Ziai WC, Lewin JJ. Advances in the management of central nervous system infections in the ICU. Crit Care Clin 2007; 22:661-94; abstract viii-ix. [PMID: 17239749 DOI: 10.1016/j.ccc.2006.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This chapter focuses on early aggressive management of common infections of the central nervous system that require monitoring in an ICU setting. These include meningitis, encephalitis, brain and epidural abscess, subdural empyema and ventriculitis. It emphasizes priorities in evaluation and management due to increasing morbidity and mortality as a result of failure to appreciate non-specific symptoms or administer timely therapy. The emergence of organisms resistant to penicillin and cephalosporins has also further complicated the early management of bacterial meningitis. Current antimicrobial guidelines are provided along with discussion of new diagnostic and therapeutic strategies and controversial aspects of management.
Collapse
Affiliation(s)
- Wendy C Ziai
- Division of Neurosciences Critical Care, Department of Neurology, The Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA.
| | | |
Collapse
|
95
|
Gosselin D, Rivest S. Role of IL-1 and TNF in the brain: twenty years of progress on a Dr. Jekyll/Mr. Hyde duality of the innate immune system. Brain Behav Immun 2007; 21:281-9. [PMID: 17275255 DOI: 10.1016/j.bbi.2006.12.004] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Revised: 12/05/2006] [Accepted: 12/06/2006] [Indexed: 11/26/2022] Open
Abstract
The immune-privileged status of the central nervous system (CNS) has changed quite dramatically during the past two decades. Leukocytes have the ability to infiltrate the CNS and cytokines are produced by resident cells, especially during injuries and diseases. Although the cellular source and role of these immune ligands are better known, their exact contribution to brain protection, repair or diseases still remains highly debated today. The ultimate fate of the immune reaction depends on the cytokines involved and the experimental models. It is now generally accepted that microglia play a central role in this response, at least for the production of cytokines participating in the innate immune system. As macrophages, resident microglia produce numerous cytokines and two of them have been largely studied since the beginning of this field of research. Twenty years ago, interleukin 1 (IL-1) and tumor-necrosis factor (TNF) were cloned and recombinant forms were used to investigate their functions ranging from normal neurophysiological responses to pathological conditions. This review presents the history of these two cytokines during immune responses in the brain and where we are now two decades later.
Collapse
Affiliation(s)
- David Gosselin
- Laboratory of Molecular Endocrinology, CHUL Research Center and Department of Anatomy and Physiology, Laval University 2705, Boulevard Laurier, Que., Canada G1V 4G2
| | | |
Collapse
|
96
|
Dimitrijevic OB, Stamatovic SM, Keep RF, Andjelkovic AV. Absence of the chemokine receptor CCR2 protects against cerebral ischemia/reperfusion injury in mice. Stroke 2007; 38:1345-53. [PMID: 17332467 DOI: 10.1161/01.str.0000259709.16654.8f] [Citation(s) in RCA: 278] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND AND PURPOSE The chemokine, monocyte chemoattractant protein-1 (CCL2), is a major factor driving leukocyte infiltration into the brain parenchyma in a variety of neuropathologic conditions associated with inflammation, including stroke. In addition, recent studies indicate that CCL2 and its receptor (CCR2) could have an important role in regulating blood-brain barrier (BBB) permeability. This study evaluated the role of the CCL2/CCR2 axis in regulating postischemic inflammation, BBB breakdown, and vasogenic edema formation. METHODS CCR2(-/-) and CCR2(+/+) mice were subjected to focal transient cerebral ischemia. BBB permeability and brain edema formation were observed at days 1 and 5 of reperfusion by evaluating the product surface area for fluorescein isothiocyanate-albumin and measuring water and electrolyte contents. Immunohistochemistry was used to assess leukocyte infiltration. cDNA gene and protein arrays for inflammatory cytokines were used to assess inflammatory profiles in CCR2(+/+) and CCR2(-/-) mice. RESULTS CCR2(-/-) mice had reduced infarct sizes and significantly reduced BBB permeability and brain edema formation in the affected ischemic hemisphere compared with CCR2(+/+) mice. This reduction in injury was closely associated with reduced infiltration of not only monocytes but also neutrophils (7- and 4-fold decreases, respectively). In addition, CCR2(-/-) mice had reduced expression/production of inflammatory cytokines during reperfusion. CONCLUSIONS These data suggest that inhibiting the CCL2/CCR2 axis affects brain reperfusion outcome by reducing brain edema, leukocyte infiltration, and inflammatory mediator expression.
Collapse
Affiliation(s)
- Oliver B Dimitrijevic
- Department of Pathology, University of Michigan, Medical School, Ann Arbor, MI 48109-0532, USA
| | | | | | | |
Collapse
|
97
|
Planas AM, Gorina R, Chamorro A. Signalling pathways mediating inflammatory responses in brain ischaemia. Biochem Soc Trans 2007; 34:1267-70. [PMID: 17073799 DOI: 10.1042/bst0341267] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Stroke causes neuronal necrosis and generates inflammation. Pro-inflammatory molecules intervene in this process by triggering glial cell activation and leucocyte infiltration to the injured tissue. Cytokines are major mediators of the inflammatory response. Pro-inflammatory and anti-inflammatory cytokines are released in the ischaemic brain. Anti-inflammatory cytokines, such as interleukin-10, promote cell survival, whereas pro-inflammatory cytokines, such as TNFalpha (tumour necrosis factor alpha), can induce cell death. However, deleterious effects of certain cytokines can turn to beneficial actions, depending on particular features such as the concentration, time point and the very intricate network of intracellular signals that become activated and interact. A key player in the intracellular response to cytokines is the JAK (Janus kinase)/STAT (signal transducer and activator of transcription) pathway that induces alterations in the pattern of gene transcription. These changes are associated either with cell death or survival depending, among other things, on the specific proteins involved. STAT1 activation is related to cell death, whereas STAT3 activation is often associated with survival. Yet, it is clear that STAT activation must be tightly controlled, and for this reason the function of JAK/STAT modulators, such as SOCS (suppressors of cytokine signalling) and PIAS (protein inhibitor of activated STAT), and phosphatases is most relevant. Besides local effects in the ischaemic brain, cytokines are released to the circulation and affect the immune system. Unbalanced pro-inflammatory and anti-inflammatory plasma cytokine concentrations favouring an 'anti-inflammatory' state can decrease the immune response. Robust evidence now supports that stroke can induce an immunodepression syndrome, increasing the risk of infection. The contribution of individual cytokines and their intracellular signalling pathways to this response needs to be further investigated.
Collapse
Affiliation(s)
- A M Planas
- IIBB (Institute for Biomedical Research)--CSIC (Spanish Research Council), IDIBAPS (Institute of Biomedical Investigation 'August Pi i Sunyer'), Rosselló 161, planta 6, E-08036 Barcelona, Spain.
| | | | | |
Collapse
|
98
|
Abstract
There is now considerable evidence from both experimental and clinical studies that immune and inflammatory processes can contribute to the onset of stroke and the neurologic and psychologic outcomes. Several specific therapeutic targets have been identified that may significantly improve the devastating impact of stroke.
Collapse
Affiliation(s)
- Robert Skinner
- Faculty of Life Sciences, Michael Smith Building (C2210), University of Manchester, Acker Street, Manchester M13 9PT, UK.
| | | | | | | |
Collapse
|
99
|
Adibhatla RM, Hatcher JF. Secretory phospholipase A2 IIA is up-regulated by TNF-alpha and IL-1alpha/beta after transient focal cerebral ischemia in rat. Brain Res 2007; 1134:199-205. [PMID: 17204250 PMCID: PMC1855193 DOI: 10.1016/j.brainres.2006.11.080] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 11/22/2006] [Accepted: 11/27/2006] [Indexed: 10/23/2022]
Abstract
Cerebral ischemia initiates an inflammatory response in the brain that is associated with the induction of a variety of cytokines, including tumor necrosis factor-alpha (TNF-alpha) and interleukin-1alpha/beta (IL-1alpha/beta) that contributes to stroke injury. Transient middle cerebral artery occlusion (tMCAO) in spontaneously hypertensive rat (SHR) resulted in significant increases in TNF-alpha and IL-1beta levels. We have previously demonstrated up-regulation of secretory phospholipase A2 IIA (sPLA2 IIA) mRNA and protein expression, increased PLA2 activity, and loss of phosphatidylcholine after 1-h tMCAO and 24-h reperfusion in SHR. Treatment with TNF-alpha antibody or IL-1 receptor antagonist significantly attenuated infarction volume, sPLA2 IIA protein expression, PLA2 activity and significantly restored phosphatidylcholine levels after tMCAO. This suggests that cytokine induction up-regulates sPLA2 IIA protein expression, resulting in altered lipid metabolism that contributes to stroke injury.
Collapse
Affiliation(s)
- Rao Muralikrishna Adibhatla
- Department of Neurological Surgery, University of Wisconsin, and Veterans Administration Hospital, Madison, WI 53705, USA.
| | | |
Collapse
|
100
|
Konsman JP, Drukarch B, Van Dam AM. (Peri)vascular production and action of pro-inflammatory cytokines in brain pathology. Clin Sci (Lond) 2006; 112:1-25. [PMID: 17132137 DOI: 10.1042/cs20060043] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In response to tissue injury or infection, the peripheral tissue macrophage induces an inflammatory response through the release of IL-1β (interleukin-1β) and TNFα (tumour necrosis factor α). These cytokines stimulate macrophages and endothelial cells to express chemokines and adhesion molecules that attract leucocytes into the peripheral site of injury or infection. The aims of the present review are to (i) discuss the relevance of brain (peri)vascular cells and compartments to bacterial meningitis, HIV-1-associated dementia, multiple sclerosis, ischaemic and traumatic brain injury, and Alzheimer's disease, and (ii) to provide an overview of the production and action of pro-inflammatory cytokines by (peri)vascular cells in these pathologies of the CNS (central nervous system). The brain (peri)vascular compartments are highly relevant to pathologies affecting the CNS, as infections are almost exclusively blood-borne. Insults disrupt blood and energy flow to neurons, and active brain-to-blood transport mechanisms, which are the bottleneck in the clearance of unwanted molecules from the brain. Perivascular macrophages are the most reactive cell type and produce IL-1β and TNFα after infection or injury to the CNS. The main cellular target for IL-1β and TNFα produced in the brain (peri)vascular compartment is the endothelium, where these cytokines induce the expression of adhesion molecules and promote leucocyte infiltration. Whether this and other effects of IL-1 and TNF in the brain (peri)vascular compartments are detrimental or beneficial in neuropathology remains to be shown and requires a clear understanding of the role of these cytokines in both damaging and repair processes in the CNS.
Collapse
Affiliation(s)
- Jan P Konsman
- Laboratory of Integrative Neurobiology, CNRS FRE 2723/INRA UR 1244/University Bordeaux2, Institut François Magendie, Bordeaux, France
| | | | | |
Collapse
|