51
|
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
|
52
|
Hauck EF, Hoffmann JF, Heimann A, Kempski O. EndothelinA receptor antagonist BSF-208075 causes immune modulation and neuroprotection after stroke in gerbils. Brain Res 2007; 1157:138-45. [PMID: 17506996 DOI: 10.1016/j.brainres.2007.04.047] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2005] [Revised: 03/12/2007] [Accepted: 04/19/2007] [Indexed: 11/21/2022]
Abstract
UNLABELLED Leukocytes contribute to the ischemia-reperfusion injury. Recent studies suggested endothelins could be important mediators for leukocyte activation in stroke. We tested if the endothelinA receptor antagonist BSF-208075 (ambrisentan) could reduce an ischemic lesion by modulation of leukocyte-endothelium interactions. Twenty-four gerbils underwent either a sham operation (n=6) or 15 min of bilateral carotid artery occlusion resulting in global cerebral ischemia. Ischemic animals received normal saline (n=6), 5 mg/kg BSF-208075 (n=6) or 30 mg/kg (n=6) administered intravenously at 10 min of reperfusion. Leukocytes rolling or adhering to endothelium were counted by intravital microscopy in parietal subsurface venules through a closed cranial window. BSF-208075 dose-dependently reduced postischemic leukocytes rolling (7.3+/-2.3 vs. 3.3+/-1.4 vs. 0.7+/-0.7 [n/100 microm/min]; p<0.05) and adhering (5.3+/-1.4 vs. 2.7+/-1.6 vs. 1.3+/-0.5 [n/100 microm/min]; p<0.05). Cerebral blood flow was not significantly changed by BSF-208075. Cortical neurons [n/mm2] in an area corresponding to the in vivo microscopy were dose-dependently preserved 7 days after ischemia (2456+/-687 vs. 3254+/-245 vs. 3780+/-168; p<0.05). CONCLUSION Endothelins mediate leukocyte activation in ischemic stroke. The endothelinA receptor antagonist BSF-208075 administered during reperfusion reduces the postischemic leukocyte activation and causes neuroprotection.
Collapse
Affiliation(s)
- Erik F Hauck
- Division of Neurosurgery, University of Texas Medical Branch, Galveston, USA
| | | | | | | |
Collapse
|
53
|
Villa P, Triulzi S, Cavalieri B, Di Bitondo R, Bertini R, Barbera S, Bigini P, Mennini T, Gelosa P, Tremoli E, Sironi L, Ghezzi P. The interleukin-8 (IL-8/CXCL8) receptor inhibitor reparixin improves neurological deficits and reduces long-term inflammation in permanent and transient cerebral ischemia in rats. MOLECULAR MEDICINE (CAMBRIDGE, MASS.) 2007; 13:125-33. [PMID: 17592546 PMCID: PMC1892761 DOI: 10.2119/2007–00008.villa] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 02/05/2007] [Indexed: 01/30/2023]
Abstract
Leukocyte infiltration is viewed as a pharmacological target in cerebral ischemia. We previously reported that reparixin, a CXCL8 receptor blocker that inhibits neutrophil infiltration, and related molecules can reduce infarct size in a rat model of transient middle cerebral artery occlusion (MCAO). The study aims were to compare the effects of reparixin in transient and permanent MCAO using varied treatment schedules and therapeutic windows to evaluate effects on long-term neurological deficits and late inflammatory response. Reparixin, administered for 1 to 3 days, 3.5 to 6 h after MCAO, ameliorates neurological function recovery and inhibits long-term inflammation. The infarct size reduction at 24 h, evaluated by TTC staining, is more pronounced in transient MCAO. MRI analysis identified a decrease in the progression of infarct size by reparixin that was more evident at 48 h in permanent MCAO, and was associated with a significantly improved recovery from long-term neurological deficits.
Collapse
Affiliation(s)
- Pia Villa
- Mario Negri Institute, 20157 Milan, Italy
- CNR, Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | | | | | | | | | - Paolo Gelosa
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Elena Tremoli
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
- Centro Cardiologico Monzino, IRCCS, 20138 Milan, Italy
| | - Luigi Sironi
- Department of Pharmacological Sciences, University of Milan, 20133 Milan, Italy
| | - Pietro Ghezzi
- Mario Negri Institute, 20157 Milan, Italy
- Address correspondence and reprint requests to Pietro Ghezzi, Laboratory of Neuroim-munology, Mario Negri Institute, via Eritrea 62, 20157 Milan, Italy. Phone: + 39-02-39014486; Fax: + 39-02-3546277; E-mail:
| |
Collapse
|
54
|
Abstract
PURPOSE OF REVIEW Inflammation is implicated in ischaemic stroke as a general cardiovascular risk factor, a possible immediate trigger, a component (and possible exacerbating factor) of the response to tissue injury, a marker of future risk, and as a therapeutic target. Each aspect is reviewed. RECENT FINDINGS Evidence of epidemiological association of inflammatory markers, particularly C-reactive protein, has accrued, but the independence of inflammation from more conventional risk indicators is under question. Other inflammatory markers are associated with intermediate phenotypes such as hypertension. Tissue inflammation in atherosclerotic plaque is of probable relevance in identifying recently symptomatic carotid disease. Both humoral and cellular inflammation are evident following stroke, with evidence that these responses may exacerbate tissue injury. Blockade of interleukin-1, or of neutrophil chemotaxis, has reduced infarct volume in models of stroke but has yet to show benefit in clinical trials. Other anti-inflammatory strategies are promising. SUMMARY Inflammation is implicated in several aspects of acute ischaemic stroke. It remains to be established whether the inflammatory response is a truly independent risk factor in general, or whether specific anti-inflammatory interventions are beneficial either in prevention or acute treatment.
Collapse
Affiliation(s)
- Keith W Muir
- Division of Medicine and Neurosciences, University of Manchester, Hope Hospital, Salford, UK.
| | | | | | | |
Collapse
|
55
|
Yi JH, Park SW, Kapadia R, Vemuganti R. Role of transcription factors in mediating post-ischemic cerebral inflammation and brain damage. Neurochem Int 2007; 50:1014-27. [PMID: 17532542 PMCID: PMC2040388 DOI: 10.1016/j.neuint.2007.04.019] [Citation(s) in RCA: 169] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Revised: 04/16/2007] [Accepted: 04/18/2007] [Indexed: 12/20/2022]
Abstract
Inflammation is a known precipitator of neuronal death after cerebral ischemia. The mechanisms that promote or curtail the start and spread of inflammation in brain are still being debated. By virtue of their capability to modulate gene expression, several transcription factors induced in the ischemic brain can modulate the post-ischemic inflammation. While the induction of transcription factors such as IRF1, NF-kappaB, ATF-2, STAT3, Egr1 and C/EBPbeta is thought to promote post-ischemic inflammation, activation of transcription factors such as HIF-1, CREB, c-fos, PPARalpha, PPARgamma and p53 is thought to prevent post-ischemic inflammation and neuronal damage. Of these, PPARgamma which is a ligand-activated transcription factor was recently shown to prevent inflammatory gene expression in several animal models CNS disorders. This review article discusses some of the molecular mechanisms of PPARgamma induction by its agonists following focal cerebral ischemia.
Collapse
Affiliation(s)
- Jae-Hyuk Yi
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Seung-Won Park
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Department of Neurological Surgery (SP), Chung-Ang University, Seoul, Korea
| | - Ramya Kapadia
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
- Cardiovascular Research Center, University of Wisconsin, Madison, WI, USA
- Regenerative Medicine Program, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
56
|
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
|
57
|
Hayakawa K, Mishima K, Nozako M, Hazekawa M, Irie K, Fujioka M, Orito K, Abe K, Hasebe N, Egashira N, Iwasaki K, Fujiwara M. Delayed treatment with cannabidiol has a cerebroprotective action via a cannabinoid receptor-independent myeloperoxidase-inhibiting mechanism. J Neurochem 2007; 102:1488-1496. [PMID: 17437545 DOI: 10.1111/j.1471-4159.2007.04565.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We examined the neuroprotective mechanism of cannabidiol, non-psychoactive component of marijuana, on the infarction in a 4 h mouse middle cerebral artery (MCA) occlusion model in comparison with Delta(9)-tetrahydrocannabinol (Delta(9)-THC). Release of glutamate in the cortex was measured at 2 h after MCA occlusion. Myeloperoxidase (MPO) and cerebral blood flow were measured at 1 h after reperfusion. In addition, infarct size and MPO were determined at 24 and 72 h after MCA occlusion. The neuroprotective effect of cannabidiol was not inhibited by either SR141716 or AM630. Both pre- and post-ischemic treatment with cannabidiol resulted in potent and long-lasting neuroprotection, whereas only pre-ischemic treatment with Delta(9)-THC reduced the infarction. Unlike Delta(9)-THC, cannabidiol did not affect the excess release of glutamate in the cortex after occlusion. Cannabidiol suppressed the decrease in cerebral blood flow by the failure of cerebral microcirculation after reperfusion and inhibited MPO activity in neutrophils. Furthermore, the number of MPO-immunopositive cells was reduced in the ipsilateral hemisphere in cannabidiol-treated group. Cannabidiol provides potent and long-lasting neuroprotection through an anti-inflammatory CB(1) receptor-independent mechanism, suggesting that cannabidiol will have a palliative action and open new therapeutic possibilities for treating cerebrovascular disorders.
Collapse
Affiliation(s)
- Kazuhide Hayakawa
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Kenichi Mishima
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Masanori Nozako
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Mai Hazekawa
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Keiichi Irie
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Masayuki Fujioka
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Kensuke Orito
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Kohji Abe
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Nobuyoshi Hasebe
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Nobuaki Egashira
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| | - Michihiro Fujiwara
- Department of Neuropharmacology, Faculty of Pharmaceutical SciencesAdvanced Materials Institute, Fukuoka University, Fukuoka, JapanDepartment of Drug Safety Evaluation, Developmental Research Laboratories, Toyonaka, Osaka, JapanDepartment of Veterinary Pharmacology, School of Veterinary Medicine, Fuchinobe Sagamihara Kanagawa, Japan
| |
Collapse
|
58
|
Weston RM, Jones NM, Jarrott B, Callaway JK. Inflammatory cell infiltration after endothelin-1-induced cerebral ischemia: histochemical and myeloperoxidase correlation with temporal changes in brain injury. J Cereb Blood Flow Metab 2007; 27:100-14. [PMID: 16736051 DOI: 10.1038/sj.jcbfm.9600324] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Accumulation of neutrophils in brain after transient focal stroke remains controversial with some studies showing neutrophils to be deleterious, whereas others suggest neutrophils do not contribute to ischemic injury. Myeloperoxidase (MPO) has been used extensively as a marker for quantifying neutrophil accumulation, but is an indirect method and does not detect neutrophils alone. To elucidate the interaction of macrophages in the neutrophil inflammatory response, we conducted double-label immunofluorescence in brain sections at 0, 1, 2, 3, 7, and 15 days after ischemia. Each of these results was obtained from the same animal to determine correlations between neutrophil infiltration and ischemic damage. It was found that MPO activity increased up to 3 days after cerebral ischemia. Dual-staining revealed that macrophages engulf neutrophils in the brain and that this engulfment of neutrophils increased with time, with 50% of neutrophils in the brain engulfed at 3 days and approximately 85% at 15 days (N=5, P<0.05). Interestingly, at 7 days the amount of dual-staining was decreased to 20% (N=5, P<0.05). Neutrophil infiltration was positively correlated with ischemic damage in both the cortex and striatum (r(2)=0.86 and 0.80, respectively, P<0.01). The results of this study indicate that the MPO from neutrophils phagocytized by macrophages may continue to contribute to the overall MPO activity, and that previous assessments that have utilized this marker to measure neutrophil accumulation may have mis-calculated the number of neutrophils within the ischemic territory and hence their contribution to the evolution of the infarct at later time points. Thus any biphasic infiltration of neutrophils may have been masked by the accumulation of macrophages.
Collapse
Affiliation(s)
- Robert M Weston
- Department of Pharmacology, Monash University, Clayton, Victoria, Australia
| | | | | | | |
Collapse
|
59
|
Matsumoto H, Kumon Y, Watanabe H, Ohnishi T, Shudou M, Ii C, Takahashi H, Imai Y, Tanaka J. Antibodies to CD11b, CD68, and lectin label neutrophils rather than microglia in traumatic and ischemic brain lesions. J Neurosci Res 2007; 85:994-1009. [PMID: 17265469 DOI: 10.1002/jnr.21198] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Resident quiescent microglia have been thought to respond rapidly to various pathologic events in the brain by proliferating and producing many bioactive substances, including proinflammatory cytokines and nitric oxide (NO). In this study, we investigated the reaction of microglia in traumatic and ischemic lesions caused by stab wounds and the transient 90-min occlusion of middle cerebral artery in a mature rat brain. Although many Iba1(+) resident microglia underwent apoptotic degeneration in the lesion core within 24 hr after the onset of the brain insult as revealed by TUNEL staining, numerous small, round, isolectin B4(+)/CD11b(+)/CD68(+) cells were localized in the lesion core. These small, round cells with diameters of 7-9 mum and polymorph nuclei expressed neutrophil-specific elastase, alkaline phosphatase, and platelet-activating factor receptor. Accordingly, they were not activated microglia but neutrophils. Immunohistochemical staining with antibodies to inducible NO synthase (iNOS) showed that most iNOS(+) cells were neutrophils. The results from spatial and kinetic analyses using RT-PCR and immunoblotting were consistent with the immunohistochemical observations. These results suggest the necessity of reevaluating the traditional view on the roles of activated microglia in severe neuropathologic events. Note that the traditional microglial markers isolectin B4, CD11b, and CD68 are not specific for microglia, particularly in a pathologic brain.
Collapse
MESH Headings
- Alkaline Phosphatase/metabolism
- Animals
- Antibodies
- Antigens, CD/analysis
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Surface/immunology
- Biomarkers/analysis
- Biomarkers/metabolism
- Brain Infarction/immunology
- Brain Infarction/pathology
- Brain Infarction/physiopathology
- Brain Injuries/immunology
- Brain Injuries/pathology
- Brain Injuries/physiopathology
- Brain Ischemia/immunology
- Brain Ischemia/pathology
- Brain Ischemia/physiopathology
- CD11 Antigens/analysis
- CD11 Antigens/immunology
- CD11 Antigens/metabolism
- Chemotaxis, Leukocyte/immunology
- Female
- Infarction, Middle Cerebral Artery/immunology
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Leukocyte Elastase/metabolism
- Male
- Microglia/immunology
- Neutrophils/immunology
- Nitric Oxide Synthase Type II/metabolism
- Plant Lectins/immunology
- Platelet Membrane Glycoproteins/metabolism
- Rats
- Rats, Wistar
- Receptors, G-Protein-Coupled/metabolism
Collapse
Affiliation(s)
- Hiroaki Matsumoto
- Department of Neurosurgery, Graduate School of Medicine, Ehime University, Ehime, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
60
|
Oh JK, Hyun SY, Oh HR, Jung JW, Park C, Lee SY, Park JH, Kim SY, Kim KH, Kim YK, Ryu JH. Effects of Anemarrhena asphodeloides on Focal Ischemic Brain Injury Induced by Middle Cerebral Artery Occlusion in Rats. Biol Pharm Bull 2007; 30:38-43. [PMID: 17202656 DOI: 10.1248/bpb.30.38] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The preventive effect of Anemarrhena asphodeloides Bunge (Liliaceae), a traditional Chinese medicine, on ischemia-reperfusion-induced brain injury was evaluated in the rat brain. Ischemia was induced by intraluminal occlusion of the right middle cerebral artery for 2 h and reperfusion was continued for 22 h. Water extract of Anemarrhena asphodeloides (WEAA) was orally administered promptly prior to and 2 h after reperfusion. Total infarct volume and edema in the ipsilateral hemispheres of ischemia-reperfusion rats were significantly reduced by treatment with WEAA in a dose-dependent manner (p<0.05). The therapeutic time window of WEAA was 3 h in this ischemia-reperfusion rat model. WEAA also significantly inhibited increased neutrophil infiltration of ischemic brain tissue as estimated by myeloperoxidase (MPO) activity and immunohistochemical analysis. MPO-positive cells were markedly reduced by WEAA administration in striatal and cortical areas. These findings suggest that WEAA plays a crucial protective role in ischemia-induced brain injury, and suggest that WEAA could serve as a lead medicinal herb for the development of neuroprotective agents following transient focal ischemic brain injury.
Collapse
MESH Headings
- Anemarrhena
- Animals
- Biphenyl Compounds/chemistry
- Brain/drug effects
- Brain/metabolism
- Brain/pathology
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Free Radical Scavengers/chemistry
- Free Radical Scavengers/pharmacology
- Free Radical Scavengers/therapeutic use
- Hydrazines/chemistry
- Immunohistochemistry
- Infarction, Middle Cerebral Artery/complications
- Ischemic Attack, Transient/drug therapy
- Ischemic Attack, Transient/etiology
- Ischemic Attack, Transient/metabolism
- Ischemic Attack, Transient/pathology
- Male
- Neuroprotective Agents/chemistry
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Neutrophil Infiltration/drug effects
- Peroxidase/metabolism
- Picrates
- Rats
- Rats, Sprague-Dawley
- Reperfusion Injury/etiology
- Reperfusion Injury/metabolism
- Reperfusion Injury/pathology
- Reperfusion Injury/prevention & control
- Rhizome
- Saponins/analysis
- Time Factors
- Triterpenes/analysis
Collapse
Affiliation(s)
- Jin Kyung Oh
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Bu Y, Rho S, Kim J, Kim MY, Lee DH, Kim SY, Choi H, Kim H. Neuroprotective effect of tyrosol on transient focal cerebral ischemia in rats. Neurosci Lett 2006; 414:218-21. [PMID: 17316989 DOI: 10.1016/j.neulet.2006.08.094] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 08/24/2006] [Accepted: 08/24/2006] [Indexed: 10/23/2022]
Abstract
Tyrosol (2-(4-hydroxyphenyl)ethanol) is a well-known phenolic compound with antioxidant properties that is present in wine, olive oil, and other plant-derived products. The purpose of this study was to determine the neuroprotective effect of tyrosol in a stroke animal model. By using the transient middle cerebral artery occlusion rat model (2 h of occlusion, 22 h of reperfusion), we investigated the effects of tyrosol on infarct volume and sensory motor function deficit by performing 2,3,5-triphenyltetrazolium chloride staining and behavior tests after ischemia. Tyrosol showed a dose-dependent neuroprotective effect that peaked at 64.9% in rats treated with 30 mg/kg of tyrosol. In rotarod, beam balance, and foot fault tests, tyrosol exhibited protective effects against the sensory motor dysfunction. In conclusion, our results suggest that tyrosol is an appropriate candidate to be used in stroke therapy.
Collapse
Affiliation(s)
- Youngmin Bu
- Department of Herbal Pharmacology, College of Pharmacy, Woosuk University, Chonbuk 565-701, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
62
|
Role of inflammation and cellular stress in brain injury and central nervous system diseases. ACTA ACUST UNITED AC 2006. [DOI: 10.1016/j.cnr.2006.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
63
|
Huang J, Upadhyay UM, Tamargo RJ. Inflammation in stroke and focal cerebral ischemia. ACTA ACUST UNITED AC 2006; 66:232-45. [PMID: 16935624 DOI: 10.1016/j.surneu.2005.12.028] [Citation(s) in RCA: 496] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 12/26/2005] [Indexed: 10/24/2022]
Abstract
BACKGROUND A growing number of recent investigations have established a critical role for leukocytes in propagating tissue damage after ischemia and reperfusion in stroke. Experimental data obtained from animal models of middle cerebral artery occlusion implicate inflammatory cell adhesion molecules, chemokines, and cytokines in the pathogenesis of this ischemic damage. METHODS Data from recent animal and human studies were reviewed to demonstrate that inflammatory events occurring at the blood-endothelium interface of the cerebral capillaries underlie the resultant ischemic tissue damage. RESULTS After arterial occlusion, the up-regulated expression of cytokines including IL-1, and IL-6 act upon the vascular endothelium to increase the expression of intercellular adhesion molecule-1, P-selectin, and E-selectin, which promote leukocyte adherence and accumulation. Integrins then serve to structurally modify the basal lamina and extracellular matrix. These inflammatory signals then promote leukocyte transmigration across the endothelium and mediate inflammatory cascades leading to further cerebral infarction. CONCLUSIONS Inflammatory interactions that occur at the blood-endothelium interface, involving cytokines, adhesion molecules, chemokines and leukocytes, are critical to the pathogenesis of tissue damage in cerebral infarction. Exploring these pathophysiological mechanisms underlying ischemic tissue damage may direct rational drug design in the therapeutic treatment of stroke.
Collapse
Affiliation(s)
- Judy Huang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | |
Collapse
|
64
|
He W, Xu XJ. Attenuation of brain inflammatory response after focal cerebral ischemia/reperfusion with Xuesaitong Injection in rats. Chin J Integr Med 2006; 12:203-6. [PMID: 17005082 DOI: 10.1007/bf02836523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the neuro-protective effect of Xuesaitong Injection (XST) on brain inflammatory response after transient focal cerebral ischemia/reperfusion in rats. METHODS Focal cerebral ischemia/reperfusion models of male rats were induced by transient occlusion for 2 h of middle cerebral artery (MCA) which was followed by 24 h reperfusion. XST was administered through intraperitoneal injection of 25 mg/kg or 50 mg/kg at 4 h after the onset of ischemia. After reperfusion for 24 h, the neurological function score was evaluated, the brain edema was detected with dry-wet weight method, the myeloperoxidase (MPO) activity and the expression of intercellular adhesion molecule-1 (ICAM-1) of ischemic cerebral cortex and caudate putamen was determined by spectrophotometry and immunohistochemistry respectively. RESULTS XST not only lowered neurological function score at the dose of 50 mg/kg, but reduced brain edema and inhibited MPO activity and ICAM-1 expression as compared with the ischemia/reperfusion model group (P < 0.01). CONCLUSION XST has a definite effect on inhibiting the expression of ICAM-1 and neutrophil infiltration in rats with cerebral ischemia/reperfusion when treatment started at 4 h after ischemia onset, and also attenuates inflammation in the infarcted cerebral area.
Collapse
Affiliation(s)
- Wei He
- Department of Pharmacology, Gannan Medical College, Jiangxi, China.
| | | |
Collapse
|
65
|
Furlan JC, Fehlings MG. A Web-based systematic review on traumatic spinal cord injury comparing the "citation classics" with the consumers' perspectives. J Neurotrauma 2006; 23:156-69. [PMID: 16503800 DOI: 10.1089/neu.2006.23.156] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Although the citation index of an article is not a direct measure of its quality or importance, it is a measure of recognition that may suggest its impact on the scientific community. This study was undertaken to examine the characteristics of the top 100 most frequently cited articles (so-called "citation classics") on traumatic spinal cord injury (SCI) that were published between 1986 and 2003, and to compare this selected professional literature with the consumers' perspective on the key issues in SCI research. The 100 top-cited articles on traumatic SCI were identified using the Internet database of the Science Citation Index Expanded and the Web of Science with the terms "spinal cord injury" and "spinal cord injuries." Meeting abstracts, letters, and editorials were excluded. No language restriction was applied. From a consumers' perspective, the areas of greatest interest for people with SCI as reported in two previous large-scale surveys include motor function, bowel and bladder control, sexual function, and pain. The final list of citation classics on traumatic SCI included 82 original articles and 18 article reviews, which were cited 146 times on average. Topics on basic science (63%) were more frequent than clinical studies (37%). The years of publication were distributed in a bell-shape curve with a peak between 1992 and 1994. North American and European centers (99%) led the list of the citation classics. Most of the top 100 most frequently cited articles on traumatic SCI (63%) explicitly focused on at least one of the topics of greatest interest to individuals with SCI. Motor function was the leading topic in the matching list between professional literature and consumers' perspective. This bibliometric analysis, for the first time, identifies the key features of the citation classics on traumatic SCI between 1986 and 2003, a period that represents one of an unprecedented increase in knowledge in this field. The 100 top-cited peer-reviewed articles have been predominantly focused on basic science SCI research indicating a need for greater bench-to-bedside translational studies in SCI research. Although the body of this top-cited professional literature mostly matches with the consumers' perspective, most of this research has been focused on motor function assessment and recovery following SCI.
Collapse
Affiliation(s)
- Julio C Furlan
- Department of Surgery, Division of Neurosurgery, University of Toronto, Spinal Program, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
66
|
Chiba Y, Shimada A, Satoh M, Saitoh Y, Kawamura N, Hanai A, Keino H, Ide Y, Shimizu T, Hosokawa M. Sensory system-predominant distribution of leukotriene A4 hydrolase and its colocalization with calretinin in the mouse nervous system. Neuroscience 2006; 141:917-927. [PMID: 16716527 DOI: 10.1016/j.neuroscience.2006.04.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2005] [Revised: 04/05/2006] [Accepted: 04/06/2006] [Indexed: 10/24/2022]
Abstract
Leukotriene B4 is a potent lipid mediator, which has been identified as a potent proinflammatory and immunomodulatory compound. Although there has been robust evidence indicating that leukotriene B4 is synthesized in the normal brain, detailed distribution and its functions in the nervous system have been unclear. To obtain insight into the possible neural function of leukotriene B4, we examined the immunohistochemical distribution of leukotriene A4 hydrolase, an enzyme catalyzing the final and committed step in leukotriene B4 biosynthesis, in the mouse nervous system. Immunoreactivity for leukotriene A4 hydrolase showed widespread distribution with preference to the sensory-associated structures; i.e. neurons in the olfactory epithelium and vomeronasal organ, olfactory glomeruli, possibly amacrine cells, neurons in the ganglion cell layer and three bands in the inner plexiform layer of the retina, axons in the optic nerve and tract up to the superior colliculus, inner and outer hair cells and the spiral ganglion cells in the cochlea, vestibulocochlear nerve bundle, spinal trigeminal tract, and lamina II of the spinal cord. Double immunofluorescence staining demonstrated that most of the leukotriene A4-hydrolase-immunopositive neurons coexpressed calretinin, a calcium-binding protein in neurons. The ubiquitous distribution of leukotriene A4 hydrolase was in sharp contrast with the distribution of leukotriene C4 synthase [Shimada A, Satoh M, Chiba Y, Saitoh Y, Kawamura N, Keino H, Hosokawa M, Shimizu T (2005) Highly selective localization of leukotriene C4 synthase in hypothalamic and extrahypothalamic vasopressin systems of mouse brain. Neuroscience 131:683-689] which was confined to the hypothalamic and extrahypothalamic vasopressinergic neurons. These results suggest that leukotriene B4 may exert some neuromodulatory function mainly in the sensory nervous system, in concert with calretinin.
Collapse
Affiliation(s)
- Y Chiba
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| | - A Shimada
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan; Core Research for Evolutional Science and Technology (CREST) of Japan Science and Technology Corporation, Japan.
| | - M Satoh
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan; Core Research for Evolutional Science and Technology (CREST) of Japan Science and Technology Corporation, Japan
| | - Y Saitoh
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| | - N Kawamura
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| | - A Hanai
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| | - H Keino
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| | - Y Ide
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
| | - T Shimizu
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, Tokyo 113-0033, Japan; Core Research for Evolutional Science and Technology (CREST) of Japan Science and Technology Corporation, Japan
| | - M Hosokawa
- Department of Pathology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya-cho, Kasugai, Aichi 480-0392, Japan
| |
Collapse
|
67
|
Strbian D, Karjalainen-Lindsberg ML, Tatlisumak T, Lindsberg PJ. Cerebral mast cells regulate early ischemic brain swelling and neutrophil accumulation. J Cereb Blood Flow Metab 2006; 26:605-12. [PMID: 16163296 DOI: 10.1038/sj.jcbfm.9600228] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We previously observed degranulated mast cells (MC) in association with perivascular brain edema formation during focal cerebral ischemia. Brain MC are typically located perivascularly and contain potent fast-acting vasoactive and proteolytic substances. We examined in a rat model of transient middle cerebral artery occlusion (MCAO) whether, in the early phase of ischemia, MC regulate microcirculation, the blood-brain barrier (BBB) permeability, and edema formation. First, animals received MC inhibitor (cromoglycate), MC-degranulating drug (compound 48/80), or saline. Thereafter, we performed transient MCAO in gene-manipulated MC-deficient rats and their wild-type (WT) littermates, calculating brain swelling, visualizing BBB leakage by intravenously administered Evans blue albumin, and determining neutrophil infiltration with light microscopy. Cerebral blood flow, monitored by laser-Doppler flowmetry in separate experiments, was similar among pharmacological treatments. Ischemic swelling resulted in increased hemispheric volume of 13.4%+/-1.0% in controls, 8.1%+/-0.4% (39% reduction) after cromoglycate, and 25.2%+/-2.0% (89% increase) after compound 48/80 (P<0.05). Early ischemic BBB leakage was reduced by 51% after cromoglycate, and 50% enhanced by compound 48/80 (P<0.05). The cromoglycate group showed 37% less postischemic neutrophil infiltration than did controls (P<0.05). Furthermore, MC-deficient rats responded to focal ischemia with 58% less brain swelling (6.7%+/-1.2%) than did their WT littermates (15.8%+/-1.4%, P<0.05). Blood-brain barrier damage was 47% lower in MC-deficient rats than in the WT (P<0.05). Neutrophil infiltration after MCAO was decreased 47% in MC-deficient rats in comparison to WT (P<0.05). Pharmacological MC inhibition thus appears to deserve further investigation regarding reduction of brain swelling and inflammation early after stroke.
Collapse
Affiliation(s)
- Daniel Strbian
- Department of Neurology, Helsinki University Central Hospital and Neuroscience Program, University of Helsinki, Helsinki, Finland
| | | | | | | |
Collapse
|
68
|
Koh SH, Chang DI, Kim HT, Kim J, Kim MH, Kim KS, Bae I, Kim H, Kim DW, Kim SH. Effect of 3-aminobenzamide, PARP inhibitor, on matrix metalloproteinase-9 level in plasma and brain of ischemic stroke model. Toxicology 2005; 214:131-9. [PMID: 16085349 DOI: 10.1016/j.tox.2005.06.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Revised: 06/19/2005] [Accepted: 06/20/2005] [Indexed: 11/17/2022]
Abstract
We investigated the effect of poly(ADP-ribose) polymerase (PARP) inhibitor on the levels of plasma and brain matrix metalloproteinase-9 (MMP-9) and the expression of nuclear factor kappa B (NF-kappaB) during experimental focal cerebral ischemia. The 3-aminobenzamide (3-AB), a PARP inhibitor, and saline were administered to 80 Sprague-Dawley rats [3-AB group; 5 rats for plasma sampling, 35 for brain sampling, and 40 for TTC staining] and to 85 rats (10, 35, and 40, respectively), respectively, 10 min before the occlusion of the left middle cerebral artery (MCAo) for 2 h. Infarct volume was measured by TTC staining, the serial levels of plasma and brain MMP-9 were measured by zymography just before and 2, 4, 8, 24, 48, and 72 h after MCAo, brain NF-kappaB activity was determined by Western blotting, and neutrophil infiltration was evaluated by assessing myeloperoxidase activity. Compared with control group, the levels of plasma and brain MMP-9, brain NF-kappaB, and MPO activities were significantly reduced in 3-AB group at each time point (p<0.05). Plasma MMP-9 increased maximally at 4h and then decreased rapidly, brain MMP-9 increased maximally at 24 h and persisted until 72 h, and NF-kappaB increased maximally at 24h and then decreased slowly in both groups. Therefore, the PARP inhibitor reduces the expression of MMP-9 and NF-kappaB and the infiltration of neutrophils in ischemic stroke.
Collapse
Affiliation(s)
- Seong-Ho Koh
- Department of Neurology, Institute of Biomedical Science, College of Medicine, Hanyang University, #17 Haengdang-Dong, Seongdong-gu, Seoul 133-791, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Harmon D, Ghori K, Lan W, Shorten G. Effect of aprotinin on in vitro cerebral endothelial ICAM-1 expression induced by astrocyte-conditioned medium. Eur J Anaesthesiol 2005; 22:277-82. [PMID: 15892405 DOI: 10.1017/s0265021505000463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND OBJECTIVE Aprotinin administration may decrease the incidence of stroke associated with coronary artery bypass surgery by an unknown mechanism. Astrocytes exposed to hypoxia produce proinflammatory cytokines and upregulate intercellular adhesion molecule (ICAM)-1 on cerebral endothelium. This study investigated the effects of aprotinin on cerebral endothelial activation by hypoxic astrocytes in vitro. METHODS Mouse astrocytes were exposed to hypoxia in an anaerobic chamber for 4 h followed by reoxygenation for 24 h. Astrocyte-conditioned medium (ACM) collected from mouse astrocytes subjected to hypoxia/reoxygenation (HR) or normoxia were applied to mouse cerebral endothelial cell (MCEC) cultures for 4 and 24 h in normoxia. Endothelial cells were preincubated for 1 h with aprotinin (1600 KIU mL(-1)) prior to exposure to ACM. Flow cytometry was used to estimate endothelial ICAM-1 expression. Interleukin (IL)-1beta space concentrations in ACM were estimated with enzyme-linked immunosorbent assay (ELISA). Repeated comparisons were made using analysis of variance (ANOVA) and post hoc Tukey test as appropriate. P < 0.05 was considered significant. Data is presented as mean (standard deviation, SD). RESULTS MCEC ICAM-1 expression was greater after 24 h exposure to HR-ACM compared to normoxic-ACM (mean channel flouresence (MCF) 107.5 (4.5) vs. 74.3 (4.5), respectively, P < 0.001). ICAM-1 expression was decreased by aprotinin preincubation compared to control (MCF 91.0 (1.1) vs. 107.5 (2.1), P = 0.006). Supernatant IL-1beta concentrations in astrocytes exposed to HR were greater than those exposed to normoxia (7.1 (0.2) vs. 4.1 (0.2), P = 0.01). CONCLUSIONS This may be a neuroprotective mechanism associated with aprotinin administration.
Collapse
Affiliation(s)
- D Harmon
- Walton Centre for Neurology and Neurosurgery, Liverpool, UK.
| | | | | | | |
Collapse
|
70
|
Jeon SB, Ji KA, You HJ, Kim JH, Jou I, Joe EH. Nordihydroguaiaretic acid inhibits IFN-γ-induced STAT tyrosine phosphorylation in rat brain astrocytes. Biochem Biophys Res Commun 2005; 328:595-600. [PMID: 15694390 DOI: 10.1016/j.bbrc.2005.01.025] [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] [Received: 01/06/2005] [Indexed: 01/05/2023]
Abstract
The Janus kinase (JAK) and signal transducers and activators of transcription (STAT) signal cascades are major pathways that mediate the inflammatory functions of interferon-gamma (IFN-gamma), an important pro-inflammatory cytokine. Therefore, regulation of JAK/STAT signaling should modulate IFN-gamma-mediated inflammation. In this study, we found that nordihydroguaiaretic acid (NDGA), a well-known lipoxygenase (LO) inhibitor, suppressed IFN-gamma-induced inflammatory responses in brain astrocytes. In the presence of NDGA, interferon regulatory factor-1 expression was significantly reduced. Expression of monocyte chemotactic protein-1 and interferon-gamma inducible protein-10 mRNA in response to IFN-gamma was significantly suppressed in the presence of NDGA, as was tyrosine-phosphorylation of JAK and STAT. However, the 5-LO products, leukotriene B(4) (LTB(4)) and leukotriene C(4), were not detected in cells treated with IFN-gamma, indicating that the effect of NDGA seemed to be independent of 5-LO inhibition. In addition, two other 5-LO inhibitors (Rev5901 and AA861) did not mimic the effect of NDGA, and the 5-LO metabolites, 5-hydroxyeicosatetraenoic acid and LTB(4), were unable to reverse NDGA-driven suppression of STAT activation or affect basal STAT phosphorylation. Taken together, these results suggest that NDGA regulates IFN-gamma-mediated inflammation through mechanisms unrelated to the inhibition of 5-LO.
Collapse
Affiliation(s)
- Sae-Bom Jeon
- Interdisciplinary Course for Neuroscience and Technology, Ajou University School of Medicine, Suwon 442-721, South Korea
| | | | | | | | | | | |
Collapse
|
71
|
Barone FC. Emerging therapeutic targets in focal stroke and brain trauma: cytokines and the brain inflammatory response to injury. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.2.2.17] [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]
|
72
|
Nakamura K, Ueno T, Yamamoto H, Iguro Y, Yamada K, Sakata R. Relationship between cerebral injury and inflammatory responses in patients undergoing cardiac surgery with cardiopulmonary bypass. Cytokine 2005; 29:95-104. [PMID: 15613277 DOI: 10.1016/j.cyto.2004.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2004] [Revised: 10/04/2004] [Accepted: 10/05/2004] [Indexed: 10/26/2022]
Abstract
This study was performed to evaluate whether cytokines, adhesion molecules, ghrelin and S-100B are useful markers in predicting the cerebral infarction after cardiac surgery with cardioplumomary bypass (CPB). The patients (n=20) were classified into two groups; group A (n=4) showed postoperative organized cerebral damage, while group B (n=16) consisted of patients without occurrence of postoperative strokes. Before CPB, serum levels of S-100B in both groups A and B were low (<0.5 ng/mL), while ghrelin concentrations in group A (all patients had history of strokes) were much higher than those in group B. After CPB, when serum levels of S-100B in group A at 24h were higher than those in group B, ghrelin in group A at same time point showed high levels in comparison to group B. At 12 and 24h after CPB, levels of tumor necrosis factor (TNF)-alpha, interleukin-10 and soluble TNF-receptor I in group A were significantly higher than those in group B. In conclusion, it is considered that ghrelin as well as S-100B can be a useful marker for the prediction of stoke after CPB. Increase of TNF-alpha, interleukin-10 and soluble TNF-receptor I after CPB may be involved in the pathogenesis of stroke after CPB.
Collapse
Affiliation(s)
- Kazuo Nakamura
- Department of Clinical Pharmacy and Pharmacology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| | | | | | | | | | | |
Collapse
|
73
|
Arumugam TV, Salter JW, Chidlow JH, Ballantyne CM, Kevil CG, Granger DN. Contributions of LFA-1 and Mac-1 to brain injury and microvascular dysfunction induced by transient middle cerebral artery occlusion. Am J Physiol Heart Circ Physiol 2004; 287:H2555-60. [PMID: 15308480 DOI: 10.1152/ajpheart.00588.2004] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Although the beta2-integrins have been implicated in the pathogenesis of cerebral ischemia-reperfusion (I/R) injury, the relative contributions of the alpha-subunits to the pathogenesis of ischemic stroke remains unclear. The objective of this study was to determine whether and how genetic deficiency of either lymphocyte function-associated antigen-1 (LFA-1) or macrophage-1 (Mac-1) alters the blood cell-endothelial cell interactions, tissue injury, and organ dysfunction in the mouse brain exposed to focal I/R. Middle cerebral artery occlusion was induced for 1 h (followed by either 4 or 24 h of reperfusion) in wild-type mice and in mice with null mutations for either LFA-1 or Mac-1. Neurological deficit and infarct volume were monitored for 24 h after reperfusion. Platelet- and leukocyte-vessel wall adhesive interactions were monitored in cortical venules by intravital microscopy. Mice with null mutations for LFA-1 or Mac-1 exhibited significant reductions in infarct volume. This was associated with a significant improvement in the I/R-induced neurological deficit. Leukocyte adhesion in cerebral venules did not differ between wild-type and mutant mice at 4 h after reperfusion. However, after 24 h of reperfusion, leukocyte adhesion was reduced in both LFA-1- and Mac-1-deficient mice compared with their wild-type counterparts. Platelet adhesion was also reduced at both 4 and 24 h after reperfusion in the LFA-1- and Mac-1-deficient mice. These findings indicate that both alpha-subunits of the beta2-integrins contribute to the brain injury and blood cell-vessel wall interactions that are associated with transient focal cerebral ischemia.
Collapse
Affiliation(s)
- Thiruma V Arumugam
- Dept. of Molecular and Cellular Physiology, Louisiana State Univ. Health Sciences Center, 1500 Kings Highway, Shreveport, LA 71130, USA
| | | | | | | | | | | |
Collapse
|
74
|
Furuichi Y, Noto T, Li JY, Oku T, Ishiye M, Moriguchi A, Aramori I, Matsuoka N, Mutoh S, Yanagihara T. Multiple modes of action of tacrolimus (FK506) for neuroprotective action on ischemic damage after transient focal cerebral ischemia in rats. Brain Res 2004; 1014:120-30. [PMID: 15212998 DOI: 10.1016/j.brainres.2004.04.031] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2004] [Indexed: 01/21/2023]
Abstract
While the immunosuppressant tacrolimus (FK506) is known to be neuroprotective following cerebral ischemia, the mechanisms underlying its neuroprotective properties are not fully understood. To determine the mode of action by which tacrolimus ameliorates neurodegeneration after transient focal ischemia, we therefore evaluated the effect of tacrolimus on DNA damage, release of cytochrome c, activation of microglia and infiltration of neutrophils following a 60-min occlusion of the middle cerebral artery (MCA) in rats. In this model, cortical brain damage gradually expanded until 24 h after reperfusion, whereas brain damage in the caudate putamen was fully developed within 5 h. Tacrolimus (1 mg/kg) administered immediately after MCA occlusion significantly reduced ischemic damage in the cerebral cortex, but not in the caudate putamen. Tacrolimus decreased both apoptotic and necrotic cell death at 24 h and reduced the number of cytochrome c immunoreactive cells at 8 h after reperfusion in the ischemic penumbra in the cerebral cortex. In contrast, tacrolimus did not show significant neuroprotection for necrotic cell death and reduction of cytochrome c immunoreactive cells in the caudate putamen. Tacrolimus also significantly decreased microglial activation at 8 h and inflammatory markers (cytokine-induced neutrophil chemoattractant and myeloperoxidase [MPO] activity) at 24 h after reperfusion in the ischemic cortex but not in the caudate putamen. These results collectively suggest that tacrolimus ameliorates the gradually expanded brain damage by inhibiting both apoptotic and necrotic cell death, as well as suppressing inflammatory reactions.
Collapse
Affiliation(s)
- Yasuhisa Furuichi
- Medicinal Biology Research Laboratories, Fujisawa Pharmaceutical Co. Ltd, 2-1-6, Kashima, Osaka 532-8514, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Kataoka H, Kim SW, Plesnila N. Leukocyte-endothelium interactions during permanent focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2004; 24:668-76. [PMID: 15181374 DOI: 10.1097/01.wcb.0000117812.35136.5b] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The contribution of leukocyte infiltration to brain damage after permanent focal cerebral ischemia and the underlying molecular mechanisms are still unclear. Therefore, the aim of this study was to establish a mouse model for the visualization of leukocytes in the cerebral microcirculation in vivo and to investigate leukocyte-endothelial interaction (LEI) after permanent middle cerebral artery occlusion (MCAO). Sham-operated 129/Sv mice showed physiologic LEI in pial venules as observed by intravital fluorescent microscopy. Permanent focal cerebral ischemia induced a significant increase of LEI predominantly in pial venules. The number of rolling and adherent leukocytes reached 36.5 +/- 13.2/100 microm x min and 22.5 +/- 7.9/100 microm x min, respectively at 120 minutes after MCAO (P = 0.016 vs. control). Of note, rolling and adherent leukocytes were also observed in arterioles of ischemic animals (7.3 +/- 3.0/100 microm x min rolling and 3.0 +/- 3.6/100 microm x min adherent). Capillary density was not different between groups. These results demonstrate that leukocytes accumulate in the brain not only after transient but also after permanent focal cerebral ischemia and may therefore contribute to brain damage after stroke without reperfusion.
Collapse
Affiliation(s)
- Hiroharu Kataoka
- Institute for Surgical Research, Ludwig-Maximilians-University, Munich, Germany
| | | | | |
Collapse
|
76
|
Schuhmann MU, Mokhtarzadeh M, Stichtenoth DO, Skardelly M, Klinge PM, Gutzki FM, Samii M, Brinker T. Temporal profiles of cerebrospinal fluid leukotrienes, brain edema and inflammatory response following experimental brain injury. Neurol Res 2003; 25:481-91. [PMID: 12866196 DOI: 10.1179/016164103101201896] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The post-traumatic changes of leukotrienes LTC4, LTD4, LTE4, and LTB4 in cerebrospinal fluid of rats from 10 min to 7 days were investigated after controlled cortical impact in relation to brain edema and cellular inflammatory response. LTC4 increased five-fold at 4 h, normalized at 24 h, and showed another four-fold increase at 7 days. The same pattern was observed for LTD4 and LTE4. LTB4 however, behaved differently: concentrations were lower and levels peaked two-fold at 24 h. Edema in the injured hemisphere increased continuously up to 24 h without change contralaterally. Leukocyte infiltration, macrophage presence and microglia activation were most prominent at 24 h, 7 days and 24 h respectively. Leukotriene changes in CSF seem to reflect those in the affected tissue, with a time delay and in lower concentrations, and were not linearly correlated to brain edema. The initially high leukotriene levels are rather likely to contribute to the cytotoxic edema than to enhance a vasogenic edema component. The profile of LTB4 was parallel to the time course of leukocyte infiltration, indicating initiation of infiltration as well as prolonged production by leukocytes themselves. The second leukotriene peak at 7 days is likely to follow a different pathway and might be related to a production in macrophages or activated glia.
Collapse
|
77
|
Miljkovic-Lolic M, Silbergleit R, Fiskum G, Rosenthal RE. Neuroprotective effects of hyperbaric oxygen treatment in experimental focal cerebral ischemia are associated with reduced brain leukocyte myeloperoxidase activity. Brain Res 2003; 971:90-4. [PMID: 12691841 DOI: 10.1016/s0006-8993(03)02364-3] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Hyperbaric oxygen (HBO) reduces cerebral infarct size after middle cerebral artery occlusion (MCAO) in rats through an unknown mechanism. In other forms of injury, cellular protection with HBO is associated with diminished infiltration of polymorphonuclear neutrophils (PMN). We hypothesized that HBO given prior to or after MCAO reduces PMN infiltration into the brain, and that decreased PMN infiltration is associated with improved functional and anatomic outcome. METHODS Forty rats underwent MCAO and were randomized to pretreatment with HBO (3 ATA) immediately prior to (n=13), or posttreatment immediately after surgery (n=12), or to control (air 1 ATA) (n=15). Five rats underwent sham surgery. Neurologic outcome was measured at 24 h in all animals. Brain myeloperoxidase (MPO) activity (n=22) and infarct volume (n=23) were determined. RESULTS MPO activity was significantly higher in controls (mean 0.28, 95% C.I. 0.17-0.38) than in the HBO pretreatment group (0.12, 0.08-0.16), HBO posttreatment group (0.16, 0.13-0.19), and the sham group (0.02, -0.02 to 0.05). HBO treated animals also had better neurologic outcomes (pretreatment 1.5, 0.9-2.1, posttreatment 2.6, 2.0-3.2) and smaller infarcts (pretreatment 27%, 18-37%, posttreatment 28%, 19-37%) than controls (neurologic outcome 3.7, 3.1-4.4, infarct volume 39%, 30-48%). Neurologic outcomes correlate better with MPO activity (R(2)=0.75) than with infarct volume (R(2)=0.25). CONCLUSION These data confirm the neuroprotective effects of HBO in cerebral ischemia and suggest that the mechanism of this action may involve inhibition of PMN infiltration in the injured brain.
Collapse
|
78
|
Antezana DF, Clatterbuck RE, Alkayed NJ, Murphy SJ, Anderson LG, Frazier J, Hurn PD, Traystman RJ, Tamargo RJ. High-dose ibuprofen for reduction of striatal infarcts during middle cerebral artery occlusion in rats. J Neurosurg 2003; 98:860-6. [PMID: 12691413 DOI: 10.3171/jns.2003.98.4.0860] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Ibuprofen is an antiinflammatory drug that disrupts leukocyte-endothelial cell interactions by limiting expression of endothelial adhesion molecules such as intercellular adhesion molecule-1 (ICAM-1), also known as CD54. The authors hypothesized that ibuprofen could reduce the size of the infarct associated with transient focal ischemia by inhibition of ICAM-1 expression, and they evaluated its effects in rats treated with middle cerebral artery (MCA) occlusion. Ibuprofen treatment was compared with mild systemic hypothermia, which is known to be neuroprotective and is commonly used during neurosurgical procedures. METHODS The maximum ibuprofen dose (240 mg/kg/day) that could be tolerated with no systemic toxicity was established in the initial experiments. In the efficacy experiment, rats were pretreated with vehicle, ibuprofen, or hypothermia (33 degrees C) prior to 2 hours of MCA occlusion; then their brains were harvested at 24 hours of reperfusion for histological studies. End-ischemic cerebral blood flow (CBF) was evaluated using [14C]iodoantipyrine autoradiography in additional cohorts. Expression of ICAM-1 within ischemic compared with nonischemic caudate nucleus and putamen (striatum) or cortex was evaluated using immunohistochemical studies. Compared with vehicle treatment, ibuprofen produced a 46.2% reduction (p = 0.01) in striatal infarcts, which was comparable to hypothermia (48.7% reduction, p = 0.02). Ibuprofen did not alter end-ischemic CBF in any region studied, and the ibuprofen treatment group had the lowest proportion of animals with marked ICAM-1 staining. CONCLUSIONS Ibuprofen given in maximum tolerated doses reduces the striatal infarct size after focal cerebral ischemia. The neuroprotective mechanism does not work through preservation of intraischemic CBF and is consistent with inhibition of ICAM-1 expression; however, at the doses used in this study, other effects of ibuprofen on platelet and endothelial function are possible.
Collapse
Affiliation(s)
- David F Antezana
- Department of Neurosurgery, The Johns Hopkins Hospital, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Berti R, Williams AJ, Velarde LC, Moffett JR, Elliott PJ, Adams J, Yao C, Dave JR, Tortella FC. Effect of the proteasome inhibitor MLN519 on the expression of inflammatory molecules following middle cerebral artery occlusion and reperfusion in the rat. Neurotox Res 2003; 5:505-14. [PMID: 14715434 DOI: 10.1007/bf03033160] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anti-inflammatory treatment with the proteasome inhibitor MLN519 has been previously reported to be neuroprotective against ischemic brain injury in rats. These effects have been related to inhibition of the transcription factor NF-kappaB, which is activated through ubiquitin-proteasomal degradation. The aim of this study was to evaluate the effects of MLN519 to alter the expression of several inflammatory genes under the control of NF-kappaB. Male Sprague-Dawley rats underwent middle cerebral artery occlusion (MCAo) followed by vehicle or MLN519 (1.0 g/kg, i.v.) treatment immediately after reperfusion of blood to the brain at 2h. Gene expression was evaluated 3-72 h post-MCAo. The most striking effects of intravenous treatment with MLN519 were associated with reductions in ICAM-1 expression at 3 h followed by reductions in E-selectin (12-72 h). Less dramatic reductions were observed in IL-1Beta (3-24 h) and TNF-Alpha (24 h) with no apparent effects on IL-6 and VCAM-1 mRNA levels. Immunohistochemical analysis revealed that the genes most dramatically affected by MLN519 had highest expression in endothelial cells and leukocytes (E-selectin, ICAM-1),indicating that these cell types may be the primary targets of intravenously delivered MLN519 treatment.
Collapse
Affiliation(s)
- R Berti
- Division of Neurosciences, Walter Reed Army Institute of Research, Silver Spring, MD 20910, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Akisu M, Huseyinov A, Baka M, Yalaz M, Kultursay N. The effect of dietary supplementation with n-3 polyunsaturated fatty acids on the generation of platelet-activating factor and leukotriene B4 in hypoxic-ischemic brain in young mice. Prostaglandins Leukot Essent Fatty Acids 2002; 67:429-33. [PMID: 12468264 DOI: 10.1054/plef.2002.0453] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Platelet-activating factor (PAF), leukotriene B(4) (LTB(4)) and other cytokines have been indicated to be responsible for the neuronal damage in hypoxic-ischemic brain. Diets in omega-3 (n-3) fatty acids appear to have an antiinflammatory effect, which is thought to be due to decrease in active prostaglandins and leukotrienes production after incorporation of these fatty acids into cell membrane phospholipids. We investigated the effect of dietary supplementation with n-3 fatty acids on endogenous PAF and LTB(4) biosynthesis in hypoxic-ischemic brain of young mice. Young mice were randomly divided into four groups: Group 1 mice were fed standard chow (n-3 polyunsaturated fatty acids free); Group 2 and Group 3 mice were given standard diet supplemented with 10% by weight of fish oil, as source of n-3 polyunsaturated fatty acids, for 3 and 6 weeks, respectively. Group 4 mice served as control. We injured the right cerebral hemisphere of young mice by ligating the right common carotid artery and exposing the mice to 8% oxygen for 60 min. Approximately 10-fold increase in PAF concentration was determined in hypoxic-ischemic brain tissue of Group 1 mice. Tissue concentration of PAF showed a profound decline in Group 3 mice compared to Groups 1 and 2 (P<0.01, P<0.05, respectively). LTB(4) was also significantly elevated in the brain of Group 1 mice when compared to the brain of control mice (P<0.001). A striking decline was observed in the concentration of LTB(4) in both Group 2 and Group 3 mice compared to Group 1 mice (P<0.05, P<0.01, respectively). The present study shows that n-3 fatty-acid-enriched diet inhibits endogenous PAF and LTB(4) generation in hypoxic-ischemic brain tissue; however it demonstrates that 6 weeks of dietary supplementation with n-3 fatty acids results in a significant decrease in tissue level of PAF in the brain.
Collapse
Affiliation(s)
- M Akisu
- Department of Pediatrics, Medical Faculty, Ege University, Izmir, Turkey.
| | | | | | | | | |
Collapse
|
81
|
Sheng H, Enghild JJ, Bowler R, Patel M, Batinić-Haberle I, Calvi CL, Day BJ, Pearlstein RD, Crapo JD, Warner DS. Effects of metalloporphyrin catalytic antioxidants in experimental brain ischemia. Free Radic Biol Med 2002; 33:947-61. [PMID: 12361805 DOI: 10.1016/s0891-5849(02)00979-6] [Citation(s) in RCA: 83] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Reactive oxygen species play a role in the response of brain to ischemia. The effects of metalloporphyrin catalytic antioxidants (AEOL 10113 and AEOL 10150) were examined after murine middle cerebral artery occlusion (MCAO). Ninety minutes after reperfusion from 90 min MCAO in the rat, AEOL 10113, AEOL 10150, or vehicle were given intracerebroventricularly. AEOL 10113 and AEOL 10150 similarly reduced infarct size (35%) and neurologic deficit. AEOL 10113 caused behavioral side effects at twice the neuroprotective dose while AEOL 10150 required a 15-fold increase from the neuroprotective dose to cause behavioral changes. AEOL 10150, given 6 h after 90 min MCAO, reduced total infarct size by 43% without temperature effects. Brain AEOL 10150 elimination t(1/2) was 10 h. In the mouse, intravenous AEOL 10150 infusion post-MCAO reduced both infarct size (25%) and neurologic deficit. Brain AEOL 10150 uptake, greater in the ischemic hemisphere, was dose- and time-dependent. AEOL 10150 had direct effects on proteomic events and ameliorated changes caused by ischemia. In primary mixed neuronal/glial cultures exposed to 2 h of O(2)/glucose deprivation, AEOL 10150 reduced lactate dehydrogenase release dose-dependently and selectively preserved aconitase activity in concentrations consistent with neuroprotection in vivo. AEOL 10150 is an effective neuroprotective compound offering a wide therapeutic window with a large margin of safety against adverse behavioral side effects.
Collapse
Affiliation(s)
- Huaxin Sheng
- Department of Anesthesiology, Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Berti R, Williams AJ, Moffett JR, Hale SL, Velarde LC, Elliott PJ, Yao C, Dave JR, Tortella FC. Quantitative real-time RT-PCR analysis of inflammatory gene expression associated with ischemia-reperfusion brain injury. J Cereb Blood Flow Metab 2002; 22:1068-79. [PMID: 12218412 DOI: 10.1097/00004647-200209000-00004] [Citation(s) in RCA: 180] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ischemia-reperfusion brain injury initiates an inflammatory response involving the expression of adhesion molecules and cytokines, some of which are regulated by the nuclear transcription factor NF-kappaB. In this study the authors examined mRNA expression levels for several important genes associated with inflammation at five time points (3, 6, 12, 24, and 72 hours) after transient middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats. A sensitive and quantitative technique (TaqMan real-time QRT-PCR) was used to simultaneously measure mRNA levels for key cell adhesion molecules and inflammatory cytokines. Gene expression increased significantly in the injured hemisphere for interleukin (IL)-1beta (12-fold increase at 24 hours), IL-6 (25-fold increase at 6 hours) and ICAM-1 (4-fold increase at 24 hours), and the interhemispheric differences for these genes were significant for every time point examined (P < 0.05 for all values). Tumor necrosis factor-alpha mRNA was upregulated in the injured versus uninjured hemisphere from 3 to 24 hours (5-fold increase at 6 hours), while E-selectin showed a significant increase in mRNA levels from 6 to 24 hours after MCAO (10-fold increase at 6 hours) (P < 0.05 for all values). VCAM-1 mRNA levels did not respond differentially to injury at any time point between the two brain hemispheres. At all time points examined, activated NF-kappaB immunoreactivity was observed in cells throughout the infarct-damaged tissue. These results are consistent with the proinflammatory properties of the induced molecules, which are involved in the initiation of the inflammatory cascade, and may thus contribute to secondary cellular responses that lead to further brain damage.
Collapse
Affiliation(s)
- Rossana Berti
- Neuropharmacology and Molecular Biology Department, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Suzuki Y, Matsumoto Y, Ikeda Y, Kondo K, Ohashi N, Umemura K. SM-20220, a Na(+)/H(+) exchanger inhibitor: effects on ischemic brain damage through edema and neutrophil accumulation in a rat middle cerebral artery occlusion model. Brain Res 2002; 945:242-8. [PMID: 12126886 DOI: 10.1016/s0006-8993(02)02806-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The Na(+)/H(+) exchanger (NHE) is activated during ischemia-reperfusion in an effort to restore intracellular pH to normal levels. The NHE is recognized to exist as a distinct protein in the plasma membranes of a variety of cells. We investigated the pharmacological effects of a Na(+)/H(+) exchanger inhibitor, SM-20220 (N-(aminoiminomethyl)-1-methyl-1-H-indole-2-carboxamide methanesulfonate), on ischemic brain damage, edema and neutrophil accumulation at 72 h after middle cerebral artery (MCA) occlusion in a rat MCA occlusion model. SM-20220 was intravenously administered as a bolus injection immediately after occlusion, followed by a continuous infusion over 2.5 h. At 72 h after occlusion, the infract area was measured using hematoxylin-eosin staining and, using the same slices, neutrophils in the brain were immuno-stained with anti-myeloperoxidase (n=11). In a separate study, rat behavior was scored and scaled, and brains removed for the determination of water content by the dry-weight method. SM-20220 significantly (P<0.05) attenuated cerebral infarct volume, water content, and the neutrophil accumulation at 72 h after the MCA occlusion, and ameliorated neurological deficits. SM-20220, an NHE inhibitor prevented the progress of cerebral ischemic damage and edema following MCA occlusion in rats though a possible mechanism that may be due to the inhibition of neutrophil accumulation. The NHE in neutrophils may enhance the progress of cerebral damage following cerebral ischemia-reperfusion.
Collapse
Affiliation(s)
- Yasuhiro Suzuki
- Department of Pharmacology, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu 431-3192, Japan.
| | | | | | | | | | | |
Collapse
|
84
|
Altura BM, Gebrewold A, Zhang A, Altura BT. Role of leukocytes in ethanol-induced microvascular injury in the rat brain in situ: potential role in alcohol brain pathology and stroke. Eur J Pharmacol 2002; 448:89-94. [PMID: 12126976 DOI: 10.1016/s0014-2999(02)01899-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Effects of acute and chronic alcohol ethanol administration on in vivo microvascular-leukocyte dynamics was studied in brains of naive and leukocyte-depleted rats by direct, quantitative intravital high-resolution TV microscopy, fluorescence microscopy and myeloperoxidase staining. Administration of alcohol produced dose-dependent venular vasospasm, and rolling and adherence of leukocytes to venular walls; leukocyte velocity concomitantly decreased. Intermediate to high doses of ethanol resulted in infiltration of leukocytes and macrophages across venular walls, and concentration-dependent increases in myeloperoxidase staining in parenchyma, and rupture of postcapillary venules with focal hemorrhages. Use of phosphorus 31-nuclear magnetic resonance spectroscopy on intact animals revealed that the latter were associated with whole brain losses in intracellular levels of ATP and phosphocreatine with concomitant rises in intracellular inorganic phosphate and hydrogen ion concentration. Vinblastine-depletion of circulating leukocytes prevented or ameliorated greatly the alcohol-induced microvascular damage and proinflammatory-like reactions. These new results, when viewed in light of other recent findings, suggest that alcohol-induced cerebral vascular and brain damage is dependent, to a large extent, on recruitment of leukocytes.
Collapse
Affiliation(s)
- Burton M Altura
- Department of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA.
| | | | | | | |
Collapse
|
85
|
Emerich DF, Dean RL, Bartus RT. The role of leukocytes following cerebral ischemia: pathogenic variable or bystander reaction to emerging infarct? Exp Neurol 2002; 173:168-81. [PMID: 11771949 DOI: 10.1006/exnr.2001.7835] [Citation(s) in RCA: 129] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Data accumulated over the last 10 years have led to the popular hypothesis that neutrophils and other inflammatory cells play a prominent role in the neuropathology of cerebral ischemia. This hypothesis was derived from a large number of studies involving three general observations: (1) leukocytes, particularly neutrophils, are present in ischemic tissue at the approximate time that substantial neuronal death occurs; (2) neutropenia is sometimes associated with reduced ischemic damage; and (3) treatments that prevent leukocyte vascular adhesion and extravasation into the brain parenchyma can be neuroprotective. This review reexamines the literature to ascertain its support for a pathogenic role for neutrophils in ischemia-induced neuronal loss. To accomplish this goal, we employed several logical theorems of "cause-effect" relationships, as they pertain to leukocytes and ischemic brain damage. Since the majority of studies focused on neutrophils as the most likely pathogenic inflammatory cell, this review necessarily does so here. We reasoned that if neutrophils play an important pathogenic (i.e., cause-effect) role in the neuronal damage that follows a stroke, then one should expect to find clear evidence that: (1) neutrophils invade the ischemic area prior to terminal stage infarction, (2) greater numbers of early appearing neutrophils are accompanied by evidence of greater neuronal loss, and (3) dose-related inhibition of neutrophil trafficking or activity produces a corresponding decrease in the degree of brain damage following ischemia. This review of the literature reveals that the existing evidence does not readily support any of these predictions and that, therefore, it consistently falls short of establishing a clear cause-effect relationship between leukocyte recruitment and the pathogenesis of ischemia. While the available evidence does not necessarily rule out a potential pathogenic role of neutrophils and other leukocytes, it nevertheless does expose serious weaknesses in the existing studies intended to support that hypothesis. For this reason we also offer suggestions for additional experiments and the inclusion of control groups that, in the future, might provide more effective or conclusive tests of the hypothesis.
Collapse
Affiliation(s)
- Dwaine F Emerich
- Life Sciences Research and Development, Alkermes, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
86
|
Drew KL, Rice ME, Kuhn TB, Smith MA. Neuroprotective adaptations in hibernation: therapeutic implications for ischemia-reperfusion, traumatic brain injury and neurodegenerative diseases. Free Radic Biol Med 2001; 31:563-73. [PMID: 11522441 DOI: 10.1016/s0891-5849(01)00628-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Brains of hibernating mammals are protected against a variety of insults that are detrimental to humans and other nonhibernating species. Such protection is associated with a number of physiological adaptations including hypothermia, increased antioxidant defense, metabolic arrest, leukocytopenia, immunosuppression, and hypocoagulation. It is intriguing that similar manipulations provide considerable protection as experimental treatments for central nervous system injury. This review focuses on neuroprotective mechanisms employed during hibernation that may offer novel approaches in the treatment of stroke, traumatic brain injury, and neurodegenerative diseases in humans.
Collapse
Affiliation(s)
- K L Drew
- Institute of Arctic Biology, University of Alaska Fairbanks, Fairbanks, AK 99775-7000, USA.
| | | | | | | |
Collapse
|
87
|
Rossi P, Kuukasjärvi P, Salenius JP, Riutta A. The effect of revascularisation of an ischaemic lower limb on leukotriene E4 production. Prostaglandins Leukot Essent Fatty Acids 2001; 64:289-90. [PMID: 11427036 DOI: 10.1054/plef.2001.0274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
It has previously been shown that leukotriene E4 production is increased both in acute and chronic lower limb ischaemia. The aim of this study was to measure the effect of revascularisation on leuktriene E4 excretion in chronic lower limb ischaemia. Revascularisation did not affect significantly on leukotriene E4 excretion (preop. 34.9+/-7.1 pg/mg creatinine, postop. 24.5+/-4.7 pg/mg creatinine, n=10, P<0.238). We suggest that the enhanced leukotriene E4 production continues after revascularisation which may have a therapeutical implication.
Collapse
Affiliation(s)
- P Rossi
- Department of Surgery, Central Hospital of Central Finland, FIN-40600 Jyväskylä, Finland.
| | | | | | | |
Collapse
|
88
|
Barone FC, Parsons AA. Therapeutic potential of anti-inflammatory drugs in focal stroke. Expert Opin Investig Drugs 2000; 9:2281-306. [PMID: 11060807 DOI: 10.1517/13543784.9.10.2281] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The importance of cytokines, especially TNF-alpha and IL-1beta, are emphasised in the propagation and maintenance of the brain inflammatory response to injury. Much data supports the case that ischaemia and trauma elicit an inflammatory response in the injured brain. This inflammatory response consists of mediators (cytokines, chemokines and adhesion molecules) followed by cells (neutrophils early after the onset of brain injury and then a later monocyte infiltration). De novo upregulation of pro-inflammatory cytokines, chemokines and endothelial-leukocyte adhesion molecules occurs soon after focal ischaemia and trauma, as well as at the time when the tissue injury is evolving. The significance of this brain inflammatory response and its contribution to brain injury is now becoming more understood. In this review, we discuss the role of TNF-alpha and IL-1beta in traumatic and ischaemic brain injury and associated inflammation and the co-operative actions of chemokines and adhesion molecules in this process. We also address novel approaches to target cytokines and reduce the brain inflammatory response and thus brain injury, in stroke and neurotrauma. The mitogen-activated protein kinase (MAPK), p38, has been linked to inflammatory cytokine production and cell death following cellular stress. Stroke-induced p38 enzyme activation in the brain has been demonstrated and treatment with a second generation p38 MAPK inhibitor, SB-239063, provides a significant reduction in infarct size, neurological deficits and inflammatory cytokine expression produced by focal stroke. SB-239063 can also provide direct protection of cultured brain tissue to in vitro ischaemia. This robust SB-239063-induced neuroprotection emphasises a significant opportunity for targeting MAPK pathways in ischaemic stroke injury and also suggests that p38 inhibition should be evaluated for protective effects in other experimental models of nervous system injury and neurodegeneration.
Collapse
Affiliation(s)
- F C Barone
- Department of Cardiovascular Pharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA.
| | | |
Collapse
|
89
|
Aspey BS, Taylor FL, Terruli M, Harrison MJ. Temporary middle cerebral artery occlusion in the rat: consistent protocol for a model of stroke and reperfusion. Neuropathol Appl Neurobiol 2000; 26:232-42. [PMID: 10886681 DOI: 10.1046/j.1365-2990.2000.00221.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The intraluminal suture method of middle cerebral artery occlusion (MCAO) in the rat (the suture model) is a model of stroke which readily lends itself to studying the pathophysiology of post-ischaemic reperfusion. Unfortunately, variability of outcome has compromised the potential of the model, but systematic studies might characterize a consistent protocol. Therefore, the clinical and neuropathological outcome of temporary MCAO and reperfusion in the suture model were systematically investigated. Two hours or 4 h of MCAO were employed, measuring the extent of infarction at 24 h with triphenyltetrazolium chloride or at 72 h with histopathological techniques. Outcome was compared in three rat strains. Following 2 h of MCAO, motor function improved during reperfusion in Sprague-Dawley, but not in Wistar or Fischer-344 rats. All Sprague-Dawley and Wistar rats survived the protocol to 72 h, but 33% of Fischer-344 rats died. The extents of infarction and oedema were greater and less variable in Wistar and Fischer-344 than Sprague-Dawley rats, and in all three strains, the extent of infarction increased with reperfusion time. Following 4 h of MCAO, there was no improvement in motor function during reperfusion in Sprague-Dawley rats, and mortality was high at 24 h in Wistar (33%) and Fischer-344 rats (83%). Outcome was only pursued in Sprague-Dawley rats to 72 h, where the extent of infarction was quite variable. It was concluded that the extent and variability of outcome following temporary MCAO in the suture model is strain-dependent, and a consistent protocol with zero mortality was found in Wistar rats using 2 h of MCAO and 70 h of reperfusion.
Collapse
MESH Headings
- Animals
- Body Temperature
- Brain/blood supply
- Brain/pathology
- Brain/physiopathology
- Brain Edema/physiopathology
- Disease Models, Animal
- Infarction, Middle Cerebral Artery/mortality
- Infarction, Middle Cerebral Artery/pathology
- Infarction, Middle Cerebral Artery/physiopathology
- Ischemic Attack, Transient/mortality
- Ischemic Attack, Transient/pathology
- Ischemic Attack, Transient/physiopathology
- Male
- Motor Activity
- Rats
- Rats, Inbred F344
- Rats, Sprague-Dawley
- Rats, Wistar
- Reperfusion Injury/mortality
- Reperfusion Injury/pathology
- Reperfusion Injury/physiopathology
- Species Specificity
- Stroke/mortality
- Stroke/pathology
- Stroke/physiopathology
- Survival Analysis
Collapse
Affiliation(s)
- B S Aspey
- Reta Lila Weston Institute of Neurological Studies, UCL Medical School, London, UK.
| | | | | | | |
Collapse
|
90
|
Ellison JA, Barone FC, Feuerstein GZ. Matrix remodeling after stroke. De novo expression of matrix proteins and integrin receptors. Ann N Y Acad Sci 2000; 890:204-22. [PMID: 10668427 DOI: 10.1111/j.1749-6632.1999.tb07996.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Following an ischemic insult to the central nervous system a reorganization of cells and tissue takes place as the surrounding cells attempt to limit the injury, repair the damage, and restore normal architecture of the brain. This tissue remodeling requires de novo synthesis of genes and proteins which enables cells to actively change their relationship with the existing extracellular matrix and with other cells to reorganize the damaged tissue. We have identified two key molecular components of the matrix remodeling process after focal ischemia: osteopontin (OPN) and its integrin receptor alpha v beta 3 (alpha v beta 3). OPN is initially expressed by activated macrophages and microglia in the periinfarct region (24-48 hr) and at later times (5-15 days) in the core infarct. After focal stroke the alpha v beta 3 was upregulated by astrocytes in the periinfarct region. Spatial and temporal analyses demonstrated that at 5 days after injury the alpha v beta 3-positive astrocytes were at a distance from the osteopontin-expressing macrophages; by 15 days the alpha v beta 3-expressing astrocytes were localized within an osteopontin-rich matrix. In vitro OPN was shown to induce migration of astrocytes in a Boyden chamber system. These data suggest that OPN derived from microglia at the infarct border zone (and possible macrophages in the infarct core) may serve as an "astrokine" (suggested term for astrocyte chemoattractant) to organize the astrocyte scar after focal stroke. Our data demonstrate profound changes in brain matrix remodeling after focal ischemic stroke, including the synthesis and release of matrix proteins alien to the normal brain, the expression of integrin receptors that ligate these proteins, and possibly a novel function for microglial-derived OPN in astrocyte migration after focal ischemia that may drive glial activation, organization, and repair functions.
Collapse
Affiliation(s)
- J A Ellison
- Department of Cardiovascular Pharmacology, SmithKline Beecham Pharmaceuticals, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
91
|
Toyoda T, Kassell NF, Lee KS. Induction of tolerance against ischemia/reperfusion injury in the rat brain by preconditioning with the endotoxin analog diphosphoryl lipid A. J Neurosurg 2000; 92:435-41. [PMID: 10701530 DOI: 10.3171/jns.2000.92.3.0435] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Inflammatory responses and oxygen free radicals have increasingly been implicated in the development of ischemic brain injury. In some cases, an attenuation of inflammation or free-radical injury can provide tissue protection. Diphosphoryl lipid A (DPL) is a detoxified derivative of a lipopolysaccharide (endotoxin) of Salmonella minnesota strain R595, which is capable of stimulating the immune system without eliciting direct toxic effects. In this study the authors examined the influence of preconditioning with DPL on ischemia/reperfusion injury in rats. METHODS Sprague-Dawley rats were injected intravenously with either DPL or vehicle. Twenty-four hours later, some animals were tested for superoxide dismutase (SOD) activity. Others were subjected to a 3-hour period of focal cerebral ischemia and, after a reperfusion period of 24 hours, were killed. Infarction volume, SOD activity, and myeloperoxidase (MPO) activity were assayed in the postischemic animals. Pretreatment with DPL produced significant reductions in cerebral infarction and MPO activity in the ischemic penumbra. A significant enhancement of basal SOD activity was observed 24 hours after DPL treatment (that is, before ischemia), and a further enhancement of SOD activity was seen in the ischemic penumbra 24 hours after reperfusion. CONCLUSIONS These data provide the first evidence of a neuroprotective effect of preconditioning with DPL in an in vivo model of cerebral ischemia. Although the precise mechanisms through which DPL exerts its neuroprotective influence remain to be established, an inhibition of the complex inflammatory response to ischemia and an enhancement of endogenous antioxidant activity are leading candidates.
Collapse
Affiliation(s)
- T Toyoda
- Department of Neuroscience, University of Virginia Health Sciences Center, Charlottesville 22908, USA
| | | | | |
Collapse
|
92
|
Neutrophil and Platelet Activity and Quantification Following Delayed tPA Therapy in a Rabbit Model of Thromboembolic Stroke. J Thromb Thrombolysis 1999; 1:179-185. [PMID: 10603528 DOI: 10.1007/bf01062576] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Although there is considerable interest in the role of neutrophils and platelets in acute cerebral ischemia-reperfusion, there are very little data related to the effect of systemic thrombolytic therapy on these blood elements. In the present study a rabbit model was used to examine the effects of cerebral ischemia, tissue-plasminogen activator therapy, or both on neutrophil and platelet peripheral counts and activity, the latter studied by stimulated neutrophil and platelet impedance aggregation and neutrophil oxygen-free radical chemiluminescence. New Zealand white rabbits (n = 25) were randomized to receive either tissue-plasminogen activator (6.3 mg/kg IV; 20% bolus, remainder as a 2-hour infusion) or vehicle (0.9% saline) 3 hours following either autologous clot embolization or sham carotid artery isolation. Thus, four groups were examined: sham (n = 4), tPA only (n = 4), stroke only (n = 8), and stroke plus tPA (n = 9). Two hours after completion of thrombolytic therapy or vehicle infusion, the experiments were terminated, that is, 7 hours following autologous clot embolization or sham instrumentation. Blood was sampled from the thoracic aorta, and neutrophil and platelet peripheral counts and activity were determined prior to embolization and 0.5, 2.0, 4.0, and 7.0 hours following autologous clot embolization. No significant difference in platelet counts, either over time or between groups, was noted. In contrast to the platelet counts, the neutruphil count significantly increased over time, rising approximately 2.5-fold from baseline in all four groups (p < 0.001). No significant increase in neutrophil accumulation (myeloperoxidase assay; 10 (7) PMNs/g tissue; mean +/- SEM) was noted within infarcted regions of either the stroke (1.26 +/- 0.07; n = 5) or stroke plus tissue-plasminogen activator (1.26 +/- 0.09; n = 5) groups when compared to either viable brain regions within the ischemic hemisphere (1.29 +/- 0.03; n = 4) or in sham controls (1.36 +/- 0.35; n = 4). Neutrophil activity (aggregation, oxygen-free radical release) in both groups undergoing autologous clot embolization demonstrated a trend toward higher values when compared to the two sham-operated groups. Tissue-plasrninogen activator administration did not significantly affect ex vivo neutrophil activity. In contrast, platelet aggregation was significantly reduced by the administration of tPA with (p = 0.001) or without (p < 0.01) autologous clot embolization. Thus, in the present rabbit model platelet but not neutrophil activity is modulated by the administration of tissue-plasminogen activator, while autologous clot embolization results in a trend toward acute neutrophil activation.
Collapse
|
93
|
He K, Nukada H, McMorran PD, Murphy MP. Protein carbonyl formation and tyrosine nitration as markers of oxidative damage during ischaemia-reperfusion injury to rat sciatic nerve. Neuroscience 1999; 94:909-16. [PMID: 10579583 DOI: 10.1016/s0306-4522(99)00350-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We have investigated the role of oxidative damage in peripheral nerve ischaemia-reperfusion injury using a rat sciatic nerve model. After 5 h ischaemia blood flow to the sciatic nerve was restarted and markers of oxidative damage measured after various times of reperfusion. As a marker of protein oxidative damage, protein carbonyl formation was measured using a sensitive enzyme-linked immunosorbent assay. Protein carbonyl content was unaffected by ischaemia alone, but increased by 55% after 12-18 h reperfusion, correlating with the onset of nerve pathology. Pretreatment with the xanthine oxidase inhibitor allopurinol prevented these abnormalities, suggesting that xanthine oxidase activity is proximal to oxidative damage during reperfusion injury. To determine whether formation of the potent oxidant peroxynitrite from nitric oxide and superoxide contributed to ischaemia-reperfusion injury, we measured the accumulation of 3-nitrotyrosine residues in proteins. Only one protein of 49,000 mol. wt contained significant amounts of 3-nitrotyrosine residues which was shown to be glial fibrillary acidic protein, an abundant cytoskeletal protein in Schwann cells. However glial fibrillary acidic protein contained 3-nitrotyrosine residues prior to ischaemia-reperfusion, and the amount of nitrated tyrosine residues in total glial fibrillary acidic protein did not increase significantly during reperfusion, therefore it was not possible to draw conclusions about the role of peroxynitrite in nerve reperfusion injury.
Collapse
Affiliation(s)
- K He
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | | | | | | |
Collapse
|
94
|
Abstract
This review is directed at understanding how neuronal death occurs in two distinct insults, global ischemia and focal ischemia. These are the two principal rodent models for human disease. Cell death occurs by a necrotic pathway characterized by either ischemic/homogenizing cell change or edematous cell change. Death also occurs via an apoptotic-like pathway that is characterized, minimally, by DNA laddering and a dependence on caspase activity and, optimally, by those properties, additional characteristic protein and phospholipid changes, and morphological attributes of apoptosis. Death may also occur by autophagocytosis. The cell death process has four major stages. The first, the induction stage, includes several changes initiated by ischemia and reperfusion that are very likely to play major roles in cell death. These include inhibition (and subsequent reactivation) of electron transport, decreased ATP, decreased pH, increased cell Ca(2+), release of glutamate, increased arachidonic acid, and also gene activation leading to cytokine synthesis, synthesis of enzymes involved in free radical production, and accumulation of leukocytes. These changes lead to the activation of five damaging events, termed perpetrators. These are the damaging actions of free radicals and their product peroxynitrite, the actions of the Ca(2+)-dependent protease calpain, the activity of phospholipases, the activity of poly-ADPribose polymerase (PARP), and the activation of the apoptotic pathway. The second stage of cell death involves the long-term changes in macromolecules or key metabolites that are caused by the perpetrators. The third stage of cell death involves long-term damaging effects of these macromolecular and metabolite changes, and of some of the induction processes, on critical cell functions and structures that lead to the defined end stages of cell damage. These targeted functions and structures include the plasmalemma, the mitochondria, the cytoskeleton, protein synthesis, and kinase activities. The fourth stage is the progression to the morphological and biochemical end stages of cell death. Of these four stages, the last two are the least well understood. Quite little is known of how the perpetrators affect the structures and functions and whether and how each of these changes contribute to cell death. According to this description, the key step in ischemic cell death is adequate activation of the perpetrators, and thus a major unifying thread of the review is a consideration of how the changes occurring during and after ischemia, including gene activation and synthesis of new proteins, conspire to produce damaging levels of free radicals and peroxynitrite, to activate calpain and other Ca(2+)-driven processes that are damaging, and to initiate the apoptotic process. Although it is not fully established for all cases, the major driving force for the necrotic cell death process, and very possibly the other processes, appears to be the generation of free radicals and peroxynitrite. Effects of a large number of damaging changes can be explained on the basis of their ability to generate free radicals in early or late stages of damage. Several important issues are defined for future study. These include determining the triggers for apoptosis and autophagocytosis and establishing greater confidence in most of the cellular changes that are hypothesized to be involved in cell death. A very important outstanding issue is identifying the critical functional and structural changes caused by the perpetrators of cell death. These changes are responsible for cell death, and their identity and mechanisms of action are almost completely unknown.
Collapse
Affiliation(s)
- P Lipton
- Department of Physiology, University of Wisconsin School of Medicine, Madison, Wisconsin, USA
| |
Collapse
|
95
|
Abstract
Contrary to previous dogmas, it is now well established that brain cells can produce cytokines and chemokines, and can express adhesion molecules that enable an in situ inflammatory reaction. The accumulation of neutrophils early after brain injury is believed to contribute to the degree of brain tissue loss. Support for this hypothesis has been drawn from many studies where neutrophil-depletion blockade of endothelial-leukocyte interactions has been achieved by various techniques. The inflammation reaction is an attractive pharmacologic opportunity, considering its rapid initiation and progression over many hours after stroke and its contribution to evolution of tissue injury. While the expression of inflammatory cytokines that may contribute to ischemic injury has been repeatedly demonstrated, cytokines may also provide "neuroprotection" in certain conditions by promoting growth, repair, and ultimately, enhanced functional recovery. Significant additional basic work is required to understand the dynamic, complex, and time-dependent destructive and protective processes associated with inflammation mediators produced after brain injury. The realization that brain ischemia and trauma elicit robust inflammation in the brain provides fertile ground for discovery of novel therapeutic agents for stroke and neurotrauma. Inhibition of the mitogen-activated protein kinase (MAPK) cascade via cytokine suppressive anti-inflammatory drugs, which block p38 MAPK and hence the production of interleukin-1 and tumor necrosis factor-alpha, are most promising new opportunities. However, spatial and temporal considerations need to be exercised to elucidate the best opportunities for selective inhibitors for specific inflammatory mediators.
Collapse
Affiliation(s)
- F C Barone
- Department of Cardiovascular Pharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, Pennsylvania, USA
| | | |
Collapse
|
96
|
Petullo D, Masonic K, Lincoln C, Wibberley L, Teliska M, Yao DL. Model development and behavioral assessment of focal cerebral ischemia in rats. Life Sci 1999; 64:1099-108. [PMID: 10210272 DOI: 10.1016/s0024-3205(99)00038-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Stroke in humans is usually focal and occurs in the Middle Cerebral Artery (MCA) distribution. There are several rat models that mimic strokes clinically seen in human. Severity of ischemia can be determined by occlusion time, arteries occluded i.e. MCA alone or combined with one or both Common Carotid Arteries (CCA), and/or location of the occlusion. In this study three focal cerebral infarctions (stroke) were induced for 90 and 120 minutes due to the occlusion of: the MCA alone (MCAo); MCA plus unilateral CCA (MCAo+1CCAo); and MCA plus bilateral CCA (MCAo+2CCAo). Histological parameters included infarct lesion size and hemispheric swelling. Since functional recovery of clinical deficits in stroke often correlates with the efficacy of anti-ischemic therapy, we focused on the behavioral recovery. We combined many sources to obtain comprehensive guidelines for clinical behavior evaluation. Tests included limb flexion, torso twisting, circling, lateral push resistance, beam balancing and walking, hindlimb placing, and inverted angle-board gripping. Occlusion lasting 90 minutes was found to have consistent and repeatable deficits. Results from our study demonstrate 120 minutes of occlusion produced a 60% morality rate and was therefore dropped. Body weight changes between groups showed that increased occlusion time produced more weight loss. Behavior changes indicated that MCAo+2CCAo for 90 minutes demonstrated assessable and consistent clinical deficits for the screening of potential therapeutics.
Collapse
Affiliation(s)
- D Petullo
- Human Genome Sciences, Inc., Rockville, Maryland 20850, USA
| | | | | | | | | | | |
Collapse
|
97
|
Fabian RH, Kent TA. Superoxide anion production during reperfusion is reduced by an antineutrophil antibody after prolonged cerebral ischemia. Free Radic Biol Med 1999; 26:355-61. [PMID: 9895227 DOI: 10.1016/s0891-5849(98)00215-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neutrophils may be involved in the pathophysiology of reperfusion injury following cerebral ischemia. One potential mechanism of reperfusion injury by neutrophils is through production of the superoxide anion. We hypothesized that, due to progressive endothelial damage during ischemia, neutrophil activation would be more prominent after longer periods of ischemia prior to reperfusion. Thus, neutrophils would contribute more to pathological processes such as superoxide anion formation after longer than after shorter periods of ischemia. A reversible middle cerebral artery occlusion model in rats was employed and superoxide anion concentration was measured with a cytochrome c coated electrode placed on the cortical penumbral region. Occlusion times were varied from 60 min to 2 h, and neutrophils were inhibited with an antiCD18 antibody administered prior to occlusion. Neutrophil accumulation and reduction with antibody treatment was confirmed immunohistochemically. Superoxide anion (O2*-) concentration was detected during the hours following 60 min of occlusion, and increased further with 2 h of occlusion. Treatment with the antiCD18 antibody had no effect on O2*- concentration during reperfusion in the 60-90 min occlusion groups, but O2*- concentration was significantly lower in the antiCD18 antibody treated group than in the control group during reperfusion after 120 min of ischemia. The antibody also reduced cortical neutrophil accumulation in the 120 min ischemia group. These results indicate for the first time that superoxide production by neutrophils becomes more important with longer periods of ischemia, and other quantitatively less important sources of superoxide predominate with shorter periods of ischemia. This phenomenon may explain some of the variation seen between different models of ischemia with different durations of ischemia when targeting reactive oxygen species, and supports an approach to combination therapy to extend the therapeutic window and reduce the deleterious effects of reperfusion.
Collapse
Affiliation(s)
- R H Fabian
- Department of Neurology, and Marine Biomedical Institute of the University of Texas Medical Branch at Galveston, TX, USA.
| | | |
Collapse
|
98
|
Jean WC, Spellman SR, Nussbaum ES, Low WC. Reperfusion Injury after Focal Cerebral Ischemia: The Role of Inflammation and the Therapeutic Horizon. Neurosurgery 1998. [DOI: 10.1227/00006123-199812000-00076] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
99
|
Jean WC, Spellman SR, Nussbaum ES, Low WC. Reperfusion injury after focal cerebral ischemia: the role of inflammation and the therapeutic horizon. Neurosurgery 1998; 43:1382-96; discussion 1396-7. [PMID: 9848853 DOI: 10.1097/00006123-199812000-00076] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recent evidence indicates that thrombolysis may be an effective therapy for the treatment of acute ischemic stroke. However, the reperfusion of ischemic brain comes with a price. In clinical trials, patients treated with thrombolytic therapy have shown a 6% rate of intracerebral hemorrhage, which was balanced against a 30% improvement in functional outcome over controls. Destruction of the microvasculature and extension of the infarct area occur after cerebral reperfusion. We have reviewed the existing data indicating that an inflammatory response occurring after the reestablishment of circulation has a causative role in this reperfusion injury. The recruitment of neutrophils to the area of ischemia, the first step to inflammation, involves the coordinated appearance of multiple proteins. Intercellular adhesion molecule-1 and integrins are adhesion molecules that are up-regulated in endothelial cells and leukocytes. Tumor necrosis factor-alpha, interleukin-1, and platelet-activating factor also participate in leukocyte accumulation and subsequent activation. Therapies that interfere with the functions of these factors have shown promise in reducing reperfusion injury and infarct extension in the experimental setting. They may prove to be useful adjuncts to thrombolytic therapy in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- W C Jean
- Department of Neurosurgery, University of Minnesota, Minneapolis 55455, USA
| | | | | | | |
Collapse
|
100
|
Eppihimer MJ. The role of leukocyte-endothelial cell adhesion in cardiovascular disease. PATHOPHYSIOLOGY 1998. [DOI: 10.1016/s0928-4680(98)00023-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|