201
|
Nomoto N. Inhibitory effect of free radical scavenger, MCI-186, in the increase of hydroxyl radical induced by iminodipropionitrile in rats. J Neurol Sci 2004; 219:41-4. [PMID: 15050436 DOI: 10.1016/j.jns.2003.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2003] [Revised: 11/15/2003] [Accepted: 12/01/2003] [Indexed: 11/30/2022]
Abstract
Beta,beta'-Iminodipropionitrile (IDPN) is known to produce permanent motor behavioral abnormalities in rats. This behavior syndrome is also termed as "ECC Syndrome", the animal model for Gilles de la Tourette syndrome in humans. Some reports showed that these behavioral abnormalities are caused by monoamine changes. However, there was little research on the relation between IDPN-induced behavioral abnormalities and free radical. 3-Methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), a newly synthesized free radical scavenger, exerts beneficial free radical scavenging and antioxidant characteristics. We investigated that MCI-186 inhibited the process of hydroxyl radical formation induced by IDPN administration in the rat brain. In the group of IDPN administration, hydroxyl radical levels exhibited predominant increase in most parts of the rat brain. In the group of IDPN and MCI-186 administration, hydroxyl radical levels marked significant decrease compared with those in the group of IDPN administration. Therefore, MCI-186 inhibited production of hydroxyl radical and might prove to be effective against ECC syndrome induced by IDPN.
Collapse
Affiliation(s)
- Nobuatsu Nomoto
- Fourth Department of Internal Medicine, Toho University School of Medicine, 2-17-6 Ohashi, Meguro, Tokyo 153-8515, Japan.
| |
Collapse
|
202
|
Dohi K, Ohtaki H, Inn R, Ikeda Y, Shioda HS, Aruga T. Peroxynitrite and caspase-3 expression after ischemia/reperfusion in mouse cardiac arrest model. ACTA NEUROCHIRURGICA. SUPPLEMENT 2004; 86:87-91. [PMID: 14753412 DOI: 10.1007/978-3-7091-0651-8_20] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NO is a putative neurotransmitter and neuromodulator in the brain. NO is not functioning as a direct neurotoxin. NO with the superoxide radical product peroxynitrite (ONOO-) is much more cytotoxic under tissue impairment conditions. Caspase-3, a potent effector of apoptosis that is triggered via several different signaling pathways, may play a very important role in neuronal cell death caused by various brain injuries. The relationship between mouse caspase-3 and peroxynitrite remains unclear. In the present study, we examined the in vivo expression of 3-nitrotyrosine (a metabolite of peroxinitrite) and caspase-3 after cerebral ischemia produced in a global ischemia model using mice (i.e., a cardiac arrest model). 3-nitrotyrosine immunoreactivity was detected in neuronal cells in the hippocampal dentate nucleus, and cortical regions starting at 12 hrs after ischemia. In particular, numerous neuronal cells were highly immunoreactive for 3-nitrotyrosine in the cortical regions. In hippocampal CA1 pyramidal neurons, 3-nitrotyrosine immunoreactivity was detected from 24 hrs. Caspase-3 immunopositive cells were observed in approximately the same area in which the positive reaction to the anti-nitrotyrosine antibody was observed. These results provide direct evidence for the induction of 3-nitrotyrosine and caspase-3 expression in vivo in an ischemia model using mice. The present findings suggest that peroxynitrite generated by cerebral ischemia/ reperfusion was strongly cytotoxic and induced neuronal cell death (apoptosis) mediated by caspase-3.
Collapse
Affiliation(s)
- K Dohi
- Department of Critical Care and Emergency Medicine, School of Medicine, Showa University, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
203
|
Nito C, Kamiya T, Amemiya S, Katoh K, Katayama Y. The neuroprotective effect of a free radical scavenger and mild hypothermia following transient focal ischemia in rats. ACTA NEUROCHIRURGICA. SUPPLEMENT 2004; 86:199-203. [PMID: 14753435 DOI: 10.1007/978-3-7091-0651-8_43] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Edaravone, a novel free radical scavenger, has been reported to reduce ischemic damage in rats subjected to transient focal ischemia. The aim of this study is, therefore, to investigate the effect of a combined therapy with edaravone and mild hypothermia of 35 degrees C. Sprague-Dawley rats were subjected to MCA occluding an intraluminal suture technique for 2 hrs. The rats were reperfused for 24 h and decapitated for infarct and edema analysis. Animals were randomly devided into four groups: (I) vehicle + normothermia (control) (II) vehicle + mild hypothermia (III) Edaravone + normothermia (IV) Edaravone + mild hypothermia. Mild hypothermia alone had no reduction of the brain damage. The edaravone alone significantly reduced edema volume. The combined treatment with edaravone and mild hypothermia reduced both infarct and edema volume. In addition, this treatment provided for the best functional outcome. These results demonstrate that free radical scavenger, edaravone attenuates brain edema and that the combined therapy with edaravone and mild hypothermia significantly reduces not only edema but also infarct on transient focal cerebral ischemia in rats. The neuroprotective effects seen in this study may be due to the combined interaction of antiedema activity between edaravone and mild hypothermia, suppressing free radical production.
Collapse
Affiliation(s)
- C Nito
- Division of Neurology, Second Department of Internal Medicine, Nippon Medical School, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
204
|
Watanabe K, Taniguchi M, Shinoda M. Synthesis of the metabolites of a free radical scavenger edaravone (MCI-186, Radicut). Redox Rep 2004; 8:157-61. [PMID: 12935313 DOI: 10.1179/135100003225001539] [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: 10/31/2022] Open
Abstract
Two metabolites of a free radical scavenger, edaravone, were synthesized. Edaravone glucuronate was synthesized by glycosylation of a glucuronic acid precursor using silver (I) trifluoromethane-sulfonate with edaravone. Edaravone sulfate was synthesized by sulfonylation of edaravone using a sulfur trioxide-pyridine complex. The two synthesized metabolites were identical to isolated metabolites. X-ray analysis identified edaravone glucuronate as beta-O-glucuronate, although there were three possible edaravone glucuronate tautomers.
Collapse
Affiliation(s)
- Kazutoshi Watanabe
- Research and Development Division, Mitsubishi Pharma Corporation, Yokohama, Japan.
| | | | | |
Collapse
|
205
|
Lee H, Bae JH, Lee SR. Protective effect of green tea polyphenol EGCG against neuronal damage and brain edema after unilateral cerebral ischemia in gerbils. J Neurosci Res 2004; 77:892-900. [PMID: 15334607 DOI: 10.1002/jnr.20193] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Previous studies have demonstrated that a green tea polyphenol, (-)-epigallocatechine gallate (EGCG), has a potent free radical scavenging and antioxidant effect. Glutamate leads to excitotoxicity and oxidative stress, which are important pathophysiologic responses to cerebral ischemia resulting in brain edema and neuronal damage. We investigated the effect of EGCG on excitotoxic neuronal damage in a culture system and the effect on brain edema formation and lesion after unilateral cerebral ischemia in gerbils. In vitro, excitotoxicity was induced by 24-hr incubation with N-methyl-D-aspartate (NMDA; 10 microM), AMPA (10 microM), or kainate (20 microM). EGCG (5 microM) was added to the culture media alone or with excitotoxins. We examined malondialdehyde (MDA) level and neuronal viability to evaluate the effect of EGCG. In vivo, unilateral cerebral ischemia was induced by occlusion of the right common carotid artery for 30, 60, or 90 min and followed by reperfusion of 24 hr. Brain edema, MDA, and infarction were examined to evaluate the protective effect of EGCG. EGCG (25 or 50 mg/kg, intraperitoneally) was administered twice, at 30 min before and immediately after ischemia. EGCG reduced excitotoxin-induced MDA production and neuronal damage in the culture system. In the in vivo study, treatment of gerbils with the lower EGCG dose failed to show neuroprotective effects; however, the higher EGCG dose attenuated the increase in MDA level caused by cerebral ischemia. EGCG also reduced the formation of postischemic brain edema and infarct volume. These results demonstrate EGCG may have future possibilities as a neuroprotective agent against excitotoxicity-related neurologic disorders such as brain ischemia.
Collapse
Affiliation(s)
- Hyung Lee
- Department of Neurology, School of Medicine, Keimyung University, Taegu, South Korea
| | | | | |
Collapse
|
206
|
Abe S, Kirima K, Tsuchiya K, Okamoto M, Hasegawa T, Houchi H, Yoshizumi M, Tamaki T. The Reaction Rate of Edaravone (3-Methyl-1-phenyl-2-pyrazolin-5-one (MCI-186)) with Hydroxyl Radical. Chem Pharm Bull (Tokyo) 2004; 52:186-91. [PMID: 14758002 DOI: 10.1248/cpb.52.186] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pyrazoline derivative edaravone is a potent hydroxyl radical scavenger that has been approved for attenuation of brain damage caused by ischemia-reperfusion. In the present work, we first determined the rate constant, k(r), at which edaravone scavenges radicals generated by a Fenton reaction in aqueous solution in the presence of the spin trap agent, 5,5-dimethyl-1-pyrroline-N-oxide (DMPO), which competed with edaravone. We detected the edaravone radicals in the process of hydroxyl radical scavenging and found that edaravone reacts with hydroxyl radical around the diffusion limit (k(r)=3.0 x 10(10) M(-1) s(-1)). The EPR (electron paramagnetic resonance) spectrum of the edaravone radical was observed by oxidation with a horseradish peroxidase-hydrogen peroxide system using the fast-flow method. This radical species is unstable and changed to another radical species with time. In addition, it was found that edaravone consumed molecular oxygen when it was oxidized by horseradish peroxidase (HRP)-H(2)O(2) system, and that edaravone was capable of providing two electrons to the electrophiles. The possible mechanisms for oxidation of edaravone were investigated from these findings.
Collapse
Affiliation(s)
- Shinji Abe
- Department of Pharmacology, The University of Tokushima School of Medicine, Tokushima, Japan
| | | | | | | | | | | | | | | |
Collapse
|
207
|
Takahashi G, Sakurai M, Abe K, Itoyama Y, Tabayashi K. MCI-186 prevents spinal cord damage and affects enzyme levels of nitric oxide synthase and Cu/Zn superoxide dismutase after transient ischemia in rabbits. J Thorac Cardiovasc Surg 2003; 126:1461-6. [PMID: 14666020 DOI: 10.1016/s0022-5223(03)00693-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE The mechanism of spinal cord injury is believed to be related to the vulnerability of spinal motor neuron cells against ischemia. We tested whether MCI-186, which is useful for treating ischemic damage in the brain, can protect against ischemic spinal cord damage. METHODS After induction of ischemia, MCI-186 or vehicle was injected intravenously. Cell damage was analyzed by observing the function of the lower limbs and by counting the number of motor neurons. To investigate the mechanism by which MCI-186 prevents ischemic spinal cord damage, we observed the immunoreactivity of Cu/Zn superoxide dismutase, neuronal nitric oxide synthase, and endothelial nitric oxide synthase. RESULTS MCI-186 eased the functional deficits and increased the number of motor neurons after ischemia. The induction of neuronal nitric oxide synthase was significantly reduced by the treatment with MCI-186. Furthermore, the increase in the induction of endothelial nitric oxide synthase and Cu/Zn superoxide dismutase was more pronounced. CONCLUSION These results indicate that MCI-186 may protect motor neurons from ischemic injury by reducing neuronal nitric oxide synthase and increasing endothelial nitric oxide synthase. MCI-186 may be a strong candidate for use as a therapeutic agent in the treatment of ischemic spinal cord injury.
Collapse
Affiliation(s)
- Goro Takahashi
- Department of Cardiovascular Surgery, Tohuko University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | |
Collapse
|
208
|
Kono H, Asakawa M, Fujii H, Maki A, Amemiya H, Yamamoto M, Matsuda M, Matsumoto Y. Edaravone, a novel free radical scavenger, prevents liver injury and mortality in rats administered endotoxin. J Pharmacol Exp Ther 2003; 307:74-82. [PMID: 12954792 DOI: 10.1124/jpet.103.053595] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We postulated that a novel free radical scavenger, 3-methyl-1-phenyl-2-pyrazolin-5-one (edaravone; EDA), would attenuate inflammatory cytokine and chemokine expression in the liver after lipopolysaccharide (LPS) challenge through its antioxidant effect. Rats were administered EDA (0.3, 1.5, 3.0, 6.0, and 12.0 mg/kg) or the same volume of saline intravenously just after LPS (10 mg/kg) injection and then was continued intermittently every 2 h (five administrations in total). Survival was assessed for the next 24 h. In separate experiments, rats were sacrificed at 60 min, 90 min, 6 h, and 9 h after LPS injection. Serum and liver sections were collected for further analysis. Survival was improved by EDA in a dose-dependent manner up to 3 mg/kg, and maximum effects were observed at a dose of 3 mg/kg. After LPS injection, alanine aminotransferase levels increased significantly to about 1,250 IU/l in the vehicle-treated group, whereas values were blunted by about 80% by EDA. Furthermore, increases in 4-hydroxynonenal-modified proteins were also blunted in the liver by EDA. Moreover, mRNA expressions of macrophage infiltrating protein-2, monocyte chemoattractant protein (MCP)-1 and MCP-5 were attenuated by EDA. As a result, increases in the number of infiltrating inflammatory cells and mRNA expression of inflammatory cytokines such as tumor necrosis factor-alpha and interleukin-6 were significantly blunted in the liver by EDA. This reduction was accompanied by a significant reduction of their serum levels. In conclusion, EDA prevented liver injury by both inhibition of recruitments of inflammatory cells and expression of inflammatory cytokine levels in the liver.
Collapse
Affiliation(s)
- Hiroshi Kono
- First Department of Surgery, University of Yamanashi, Tamaho, Nakakoma, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
209
|
Tada S, Nakamoto N, Kameyama K, Tsunematsu S, Kumagai N, Saito H, Ishii H. Clinical usefulness of edaravone for acute liver injury. J Gastroenterol Hepatol 2003; 18:851-7. [PMID: 12795759 DOI: 10.1046/j.1440-1746.2003.03064.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Edaravone, a newly synthesized radical scavenger, has shown an excellent effect on treating stroke patients. The effect of edaravone on carbon tetrachloride (CCl4)-induced acute liver injury was examined. METHODS Six rats were injected with CCl4 alone and six rats were intravenously injected with edaravone immediately after and 3 h after injection of CCl4. Another six rats were injected with olive oil alone. The animals were killed at 24 h after the CCl4 injection. RESULTS Injection of CCl4 was followed by a marked increase in serum alanine aminotranferase (ALT) level (CCl4, 1630.6 +/- 606.8 IU/L; olive oil, 21.0 +/- 2.6 IU/L; P < 0.001), lactate dehydrogenase (LDH) level (CCl4, 5068.0 +/- 2956.4 IU/L; olive oil, 203.6 +/- 30.5 IU/L; P < 0.005), and total bilirubin (TB) level (CCl4, 0.88 +/- 0.48 mg/dL; olive oil, 0.37 +/- 0.05 mg/dL; P < 0.01), whereas in the edaravone-treated rats, the ALT (119.4 +/- 113.5 IU/L, P < 0.001), LDH (369.7 +/- 288.2 IU/L, P < 0.005), and TB values (0.29 +/- 0.16 mg/dL, P < 0.01) were significantly decreased. Histological examination of the liver by hematoxylin and eosin and oil red O staining showed a marked reduction of steatosis in the CCl4 and edaravone-treated rats compared with the CCl4-injected rats. Significant inhibition of hepatocytic apoptosis was demonstrated by the terminal deoxynucleotidyl transferase-mediated UTP nick-end labeling (TUNEL) method in the edaravone-treated rats. CONCLUSIONS These results suggest that edaravone has a marked preventive effect on oxidative stress-induced acute liver injury.
Collapse
Affiliation(s)
- Shinchiro Tada
- Departments of Internal Medicine and Pathology, School of Medicine, Keio University and Center for Liver Diseases, Kitasato Institute Hospital, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
210
|
Saibara T, Toda K, Wakatsuki A, Ogawa Y, Ono M, Onishi S. Protective effect of 3-methyl-1-phenyl-2-pyrazolin-5-one, a free radical scavenger, on acute toxicity of paraquat in mice. Toxicol Lett 2003; 143:51-4. [PMID: 12697380 DOI: 10.1016/s0378-4274(03)00113-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Paraquat poisoning results in damages of multiple organs including liver, kidney and lung, and antioxidants have been proven to have protective effects. As a novel free radical scavenger, 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), was introduced to clinical use recently, its protective effect was studied on acute toxicity of paraquat in male ddY mice. When paraquat (175 mg/kg) were given orally, the survival rate was only 8% on the 6th day of paraquat ingestion in Control Group mice. Protective effect of MCI-186 was most evident and the survival rate was 42% on the 6th day and 38% on the 14th day of paraquat ingestion, respectively, when mice were treated immediately. A delay of 30 min in treatment resulted in an abrupt reduction of the survival rate. These results suggested that MCI-186 used in acute phase of paraquat intoxication might serve as a clinically available antidote for attenuating paraquat toxicity.
Collapse
Affiliation(s)
- Toshiji Saibara
- First Department of Internal Medicine, Kochi Medical School, Nankoku 783-8505, Japan.
| | | | | | | | | | | |
Collapse
|
211
|
The effects of a free radical scavenger, edaravone, combined with mild hypothermia on ischemic brain damage following transient middle cerebral artery occlusion in rats. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s0531-5131(03)00044-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
212
|
Satoh M, Kashihara N, Fujimoto S, Horike H, Tokura T, Namikoshi T, Sasaki T, Makino H. A novel free radical scavenger, edarabone, protects against cisplatin-induced acute renal damage in vitro and in vivo. J Pharmacol Exp Ther 2003; 305:1183-90. [PMID: 12649298 DOI: 10.1124/jpet.102.047522] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Accumulating evidence suggests that enhanced peroxidative damage caused by reactive oxygen species (ROS) may contribute to the pathogenesis of cisplatin-induced acute renal failure. Nevertheless, little is known about the involvement of oxygen radicals in cisplatin nephropathy. In this study, we investigated the effects of a novel free radical scavenger, 3-methyl-1-phenyl-pyrazolin-5-one (MCI-186; edarabone), on murine proximal tubular cell (PTC) damage induced by exposure to cisplatin in vitro and on renal function in an in vivo model of cisplatin-induced acute renal failure. Edarabone inhibited cisplatin-induced (40 microM, 24 h) cytotoxicity in a concentration-dependent manner (10-5 to 10-3 M). Edarabone also attenuated cisplatin-induced mitochondrial transmembrane potential loss and ROS production of PTCs. In the in vivo study, male Wistar rats were cotreated with cisplatin (5 mg/kg, i.p.) and edarabone (1 or 5 mg/kg, i.v.). Effects of edarabone on the kidney were examined 5 days after treatment. Cisplatin resulted in renal dysfunction, renal tubular damage, mitochondrial damage (assayed by histochemical staining for respiratory chain complex IV), renal protein oxidation (examined by Western blot analysis using a specific antibody for carbonyl group-containing proteins), and tubular apoptosis (determined by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling staining). The above changes were attenuated by edarabone treatment. Thus, edarabone exhibited cytoprotective effects in PTCs and renoprotective effects against cisplatin. Our findings suggest that ROS, in particular hydroxyl radicals, are involved in cisplatin nephropathy and that edarabone may be potentially useful in protecting the kidneys and prevention of acute renal failure.
Collapse
Affiliation(s)
- Minoru Satoh
- Division of Nephrology, Department of Internal Medicine, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama 701-0192, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
213
|
Toda N, Okamura T. The pharmacology of nitric oxide in the peripheral nervous system of blood vessels. Pharmacol Rev 2003; 55:271-324. [PMID: 12773630 DOI: 10.1124/pr.55.2.3] [Citation(s) in RCA: 221] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Unanticipated, novel hypothesis on nitric oxide (NO) radical, an inorganic, labile, gaseous molecule, as a neurotransmitter first appeared in late 1989 and into the early 1990s, and solid evidences supporting this idea have been accumulated during the last decade of the 20th century. The discovery of nitrergic innervation of vascular smooth muscle has led to a new understanding of the neurogenic control of vascular function. Physiological roles of the nitrergic nerve in vascular smooth muscle include the dominant vasodilator control of cerebral and ocular arteries, the reciprocal regulation with the adrenergic vasoconstrictor nerve in other arteries and veins, and in the initiation and maintenance of penile erection in association with smooth muscle relaxation of the corpus cavernosum. The discovery of autonomic efferent nerves in which NO plays key roles as a neurotransmitter in blood vessels, the physiological roles of this nerve in the control of smooth muscle tone of the artery, vein, and corpus cavernosum, and pharmacological and pathological implications of neurogenic NO have been reviewed. This nerve is a postganglionic parasympathetic nerve. Mechanical responses to stimulation of the nerve, mainly mediated by NO, clearly differ from those to cholinergic nerve stimulation. The naming "nitrergic or nitroxidergic" is therefore proposed to avoid confusion of the term "cholinergic nerve", from which acetylcholine is released as a major neurotransmitter. By establishing functional roles of nitrergic, cholinergic, adrenergic, and other autonomic efferent nerves in the regulation of vascular tone and the interactions of these nerves in vivo, especially in humans, progress in the understanding of cardiovascular dysfunctions and the development of pharmacotherapeutic strategies would be expected in the future.
Collapse
Affiliation(s)
- Noboru Toda
- Toyama Institute for Cardiovascular Pharmacology Research, Toyama Bldg., 7-13, 1-Chome, Azuchi-machi, Chuo-ku, Osaka 541-0052, Japan.
| | | |
Collapse
|
214
|
Hayashi T, Mori T, Sohmiya K, Okada Y, Inamoto S, Okuda N, Mori H, Kitaura Y. Efficacy of edaravone, a free radical scavenger, on left ventricular function and structure in diabetes mellitus. J Cardiovasc Pharmacol 2003; 41:923-9. [PMID: 12775971 DOI: 10.1097/00005344-200306000-00014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
This study was designed to assess the efficacy of edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one), a free radical scavenger that possesses anti-oxidant effects, on cardiac function and fine structure of the left ventricular myocardium in diabetes mellitus. Male Otsuka Long-Evans Tokushima Fatty (OLETF) rats, a model of spontaneous development of type II diabetes (30 weeks; n = 15) were divided into two groups and treated with edaravone 30 mg/kg/d or vehicle for 2 weeks. OLETF rats showed hyperglycemia (352 +/- 71 mg/dl vs normal control; 128 +/- 52 mg/dl), increased thiobarbituric acid-reactive substances (TBARS; 6.9 +/- 2.5 nM/ml vs 2.8 +/- 0.6 nM/ml), and decreased superoxide dismutase activity (21.5 +/- 0.9 U/ml vs 25.8 +/- 0.7 U/ml). Increased left ventricular end-diastolic pressure (12 +/- 3 mm Hg vs 6 +/- 2 mm Hg) and hypertrophied cardiocytes (23.1 +/- 1.4 vs 17.6 +/- 1.0 microm) were also observed (P < 0.05, respectively). Edaravone could not improve plasma glucose level and hemodynamic parameters but significantly decreased TBARS values (3.8 +/- 0.5) and increased superoxide dismutase activity (24.5 +/- 0.8) (vs OLETF, P < 0.05, respectively). Moreover, edaravone effectively preserved cardiocyte diameter (18.2 +/- 0.9 microm) and the fine structure of mitochondria. Thus, edaravone exhibits modest cardiac protection in diabetes mellitus independent of blood sugar level.
Collapse
Affiliation(s)
- Tetsuya Hayashi
- Third Department of Medicine, Osaka Medical College, Takatsuki, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
215
|
Okatani Y, Wakatsuki A, Enzan H, Miyahara Y. Edaravone protects against ischemia/reperfusion-induced oxidative damage to mitochondria in rat liver. Eur J Pharmacol 2003; 465:163-70. [PMID: 12650846 DOI: 10.1016/s0014-2999(03)01463-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This study investigated the effects of edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one, MCI-186), a potent free radical scavenger, on the prevention of mitochondrial injury induced by hepatic ischemia and reperfusion. Mature male rats were subjected to 70 min of hepatic ischemia and 2 h of reperfusion. The rats received vehicle or edaravone (10 mg/kg body weight) intravenously prior to ischemia, before reperfusion and 1 h after reperfusion. In the vehicle-treated animals, the respiratory control index, ADP/O, State 3 respiration and dinitrophenol-induced uncoupled respiration decreased markedly after ischemia/reperfusion and were restored by edaravone administration. Mitochondrial lipid peroxidation was elevated in the vehicle-treated group, which was attenuated by edaravone, while mitochondrial glutathione peroxidase activity decreased in the vehicle-treated group, which was similarly abrogated by edaravone treatment. Electron microscopic observation demonstrated that treatment with edaravone restored the ischemia/reperfusion-induced disorganization of mitochondrial structures. Edaravone protects against mitochondrial injury, which prevents mitochondrial oxidative stress and improves ischemia/reperfusion-induced hepatic energy metabolism.
Collapse
Affiliation(s)
- Yuji Okatani
- Department of Clinical Nursing Science, Kochi Medical School, Oko, Nankoku, Kochi 783-8505, Japan.
| | | | | | | |
Collapse
|
216
|
Dohi K, Satoh K, Ikeda Y, Ohtaki H, Shioda S, Aruga T. Neuroprotective effect from ischemia and direct free radical scavenging activity of Choto-san (kampo medicine). ACTA NEUROCHIRURGICA. SUPPLEMENT 2003; 86:123-7. [PMID: 14753419 DOI: 10.1007/978-3-7091-0651-8_27] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Choto-san is a kampo medicine that is widely used in patients with cerebral infarction, but the details of its mechanism of action remain unclear. We examined the neuroprotective effects of Choto-san using an experimental cerebral ischemia model (i.e., a rat cardiac arrest model). We also investigated the ability of Choto-san to eliminate or inhibit the activity of free radicals. It was found that Choto-san significantly prevents delayed neuronal cell death after ischemic reperfusion. Electron spin resonance demonstrated that the formation of hydroxyl- and superoxide-DMPO spin adducts were inhibited by Choto-san. The results of this study indicated that Choto-san prevents delayed neuronal cell death in the hippocampal CA1 region after ischemia. Direct free radical scavenging activity is among the pharmacological effects of Choto-san.
Collapse
Affiliation(s)
- K Dohi
- Department of Emergency and Critical Care Medicine, School of Medicine, Showa University, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
217
|
Nakagomi T, Yamakawa K, Sasaki T, Saito I, Takakura K. Effect of edaravone on cerebral vasospasm following experimental subarachnoid hemorrhage. J Stroke Cerebrovasc Dis 2003; 12:17-21. [PMID: 17903899 DOI: 10.1053/jscd.2003.2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2002] [Revised: 10/15/2002] [Indexed: 02/02/2023] Open
Abstract
The effect of the free radical scavenger edaravone on experimental cerebral vasospasm following subarachnoid hemorrhage (SAH) was investigated in a canine double hemorrhage model. Changes in the diameter of the basilar artery were assessed by serial angiography. The diameter ratio at day 7 was calculated as the percent of the basilar artery diameter of a given angiogram with respect to that of its control (day 0). The diameter ratios for the basilar artery following SAH in the control and vehicle-treated groups were 49.7% +/- 3.9% (mean +/- SEM) and 50.1% +/- 1.7%, respectively. Edaravone was administered either by continuous intravenous injection for 7 days or by bolus injection for 7 days. Continuous administration of edaravone (1 mg/kg/hr or 10 mg/kg/hr) significantly attenuated the narrowing of the basilar artery following SAH. The diameter ratios in these groups were 71.3% +/- 3.6% (1 mg/kg/hr) and 75.7% +/- 1.7% (10 mg/kg/hr). Bolus administration of edaravone (3 mg/kg, every 12 hours) reduced the arterial narrowing following SAH. The diameter ratio on day 7 was 60.1% +/- 3.3%, but the difference was not significant. These findings suggest that edaravone is effective in preventing cerebral vasospasm following SAH.
Collapse
Affiliation(s)
- Tadayoshi Nakagomi
- Department of Neurosurgery, Teikyo University School of Medicine, Tokyo, Japan
| | | | | | | | | |
Collapse
|
218
|
Abstract
Due to two developments that have taken place within the past few years, the treatment for stroke is about to change immensely. First, it has become possible to significantly inhibit the spread of hypoperfusive tissue damage by initiating antioxidant therapy within 24 hours of a stroke. As a result of this new therapy, many physicians now feel that the prognosis for stroke patients has improved considerably. Second, elucidation of the brain function of post-stroke rehabilitation has made headway as advanced functional brain imaging techniques have provided reliable data, thereby substantiating new rationales.
Collapse
Affiliation(s)
- Kyuya Kogure
- Foundation for Brain Function and Diseases, Institute of Neuropathology, 1216 Nakase, Fukaya, Saitama 366-0001, Japan.
| |
Collapse
|
219
|
Ikeda T, Xia YX, Kaneko M, Sameshima H, Ikenoue T. Effect of the free radical scavenger, 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), on hypoxia-ischemia-induced brain injury in neonatal rats. Neurosci Lett 2002; 329:33-6. [PMID: 12161256 DOI: 10.1016/s0304-3940(02)00573-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The free radical scavenger 3-methyl-1-phenyl-2-pyrazolin-5-one (MCI-186), which has been approved in Japan for use in patients with cerebral infarction, was used to treat ischemic-hypoxic brain damage in neonatal rats. Seven-day-old rat pups were subjected to a modified Levine procedure, then given either vehicle or MCI-186 (at one of three dosage levels: 3, 6, or 9 mg/kg), and the extent of brain damage was evaluated either 24 h or 7 days later. The administration of MCI-186 significantly attenuated damage, in a dose-dependent manner. These results indicate that MCI-186 is a promising candidate for the treatment of neonatal hypoxic-ischemic encephalopathy.
Collapse
Affiliation(s)
- Tomoaki Ikeda
- Department of Obstetrics and Gynecology, Miyazaki Medical College, 5200 Kihara, Kiyotake-Cho, Miyazaki 889-16, Japan.
| | | | | | | | | |
Collapse
|
220
|
Jin YJ, Mima T, Raicu V, Park KC, Shimizu K. Combined argatroban and edaravone caused additive neuroprotection against 15 min of forebrain ischemia in gerbils. Neurosci Res 2002; 43:75-9. [PMID: 12074843 DOI: 10.1016/s0168-0102(02)00019-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We investigated whether or not a combination of the selective thrombin inhibitor, argatroban, and the free radical scavenger, edaravone (MCI-186), ameliorates postischemic hypoperfusion and decreases mortality after 15 min of forebrain ischemia in the gerbil. Argatroban or edaravone alone significantly increased postischemic cerebral blood flow and attenuated brain edema after reperfusion. However, only the combination increased the survival ratio (P<0.05 by Mantel-Cox) and protected the damage of neuronal cells. The present study indicates that anticoagulants and free radical scavengers reciprocally function to inhibit the progression of ischemic cell damage and that a combination of these types of drugs will help to improve the outcomes after cerebral ischemia.
Collapse
Affiliation(s)
- Yong-Jian Jin
- Department of Neurosurgery, Kochi Medical School, Okohcho, Nankoku City, Kochi, Japan
| | | | | | | | | |
Collapse
|
221
|
Tanaka M. [Pharmacological and clinical profile of the free radical scavenger edaravone as a neuroprotective agent]. Nihon Yakurigaku Zasshi 2002; 119:301-8. [PMID: 12061142 DOI: 10.1254/fpj.119.301] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The involvement of oxygen radical species has been implicated in ischemic and post-ischemic brain cell damage. Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one; M.W. 174.20, MCI-186, Radicut Injection) has an inhibitory effect on lipid peroxidation by scavenging free radicals and prevents vascular endothelial cell injury. In rat brain ischemic models, post-ischemic treatment with edaravone reduces .OH production and infarction of the ischemic penumbral area and suppresses delayed neuronal death. It also improves neurological deficits and diminishes deterioration of brain edema observed after ischemia. We investigated the efficacy and safety of edaravone in acute ischemic stroke patients. Edaravone improved the core neurological deficits, impaired activities of daily living, and disability, without serious safety problems. Edaravone was approved in Japan for the treatment of acute brain infarction within 24 h after onset in April, 2001. We hope that edaravone represents a promising neuroprotective agent that can contribute to the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Masahiko Tanaka
- Pharmaceuticals Development Division, Mitsubishi Pharma Corporation, 2-2-6 Nihonbashi-Honcho, Chuo-ku, Tokyo 103-8405, Japan.
| |
Collapse
|
222
|
Abstract
Ischaemic brain oedema appears to involve two distinct processes, the relative contribution and time course of which depend on the duration and severity of ischaemia, and the presence of reperfusion. The first process involves an increase in tissue Na+ and water content accompanying increased pinocytosis and Na+, K+ ATPase activity across the endothelium. This is apparent during the early phase of infarction and before any structural damage is evident. This phenomenon is augmented by reperfusion. A second process results from a more indiscriminate and delayed BBB breakdown that is associated with infarction of both the parenchyma and the vasculature itself. Although, tissue Na+ level still seems to be the major osmotic force for oedema formation at this second stage, the extravasation of serum proteases is an additional potentially deleterious factor. The relative importance of protease action is not yet clear, however, degradation of the extracellular matrix conceivably leads to further BBB disruption and softening of the tissue, setting the stage for the most pronounced forms of brain swelling. A number of factors mediate or modulate ischaemic oedema formation, however, most current information comes from experimental models, and clinical data on this microcosmic level is lacking. Clinically significant brain oedema develops in a delayed fashion after large hemispheric strokes and is a cause of substantial mortality. Neurological signs appear to be at least as good as direct ICP measurement and neuroimaging in detecting and gauging the secondary damage produced by stroke oedema. The neuroimaging characteristics of the stroke, specifically the early involvement of greater than half of the MCA territory, are, however, highly predictive of the development of severe oedema over the subsequent hours and days. None of the available medical therapies provide substantial relief from the oedema and raised ICP, or at best, they are temporizing in most cases. Hemicraniectomy appears most promising as a method of avoiding death from brain compression, but the optimum timing and manner of patient selection are currently being investigated. All approaches to massive ischaemic brain swelling are clouded by the potential for survival with poor functional outcome. It is possible to manage blood pressure, serum osmolarity by way of selective fluid administration, and a number of other systemic factors that exaggerate brain oedema. Broad guidelines for treatment of stroke oedema can therefore be given at this time.
Collapse
Affiliation(s)
- Cenk Ayata
- Neurology Service, St. Elizabeth's Medical Center, Tufts University School of Medicine, Boston, MA 02135, USA
| | | |
Collapse
|
223
|
Zhang WR, Hayashi T, Sasaki C, Sato K, Nagano I, Manabe Y, Abe K. Attenuation of oxidative DNA damage with a novel antioxidant EPC-K1 in rat brain neuronal cells after transient middle cerebral artery occlusion. Neurol Res 2001; 23:676-80. [PMID: 11547942 DOI: 10.1179/016164101101199027] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
EPC-K1, L-ascorbic acid 2-[3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyltridecyl)-2H-1-benzopyran-6-yl-hydrogen phosphate] potassium salt, is a novel antioxidant. In this study, we investigated a reduction of oxidative neuronal cell damage with EPC-K1 by immunohistochemical analysis for 8-hydroxy-2'-deoxyguanosine (8-OHdG) in rat brain with 60 min transient middle cerebral artery occlusion, in association with terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ nick end labeling (TUNEL) and staining for total and active caspase-3. Treatment with EPC-K1 (20 mg kg(-1) i.v.) significantly reduced infarct size (p < 0.05) at 24 h of reperfusion. There were no positive cells for 8-OHdG and TUNEL in sham-operated brain, but numerous cells became positive for 8-OHdG, TUNEL and caspase-3 in the brains with ischemia. The number was markedly reduced in the EPC-K1 treated group. These reductions were particularly evident in the border zone of the infarct area, but the degree of reduction was less in caspase-3 staining than in 8-OHdG and TUNEL stainings. These results indicate EPC-K1 attenuates oxidative neuronal cell damage and prevents neuronal cell death.
Collapse
Affiliation(s)
- W R Zhang
- Department of Neurology, Okayama University School of Medicine and Dentistry, Japan.
| | | | | | | | | | | | | |
Collapse
|
224
|
Numagami Y, Ohnishi ST. S-allylcysteine inhibits free radical production, lipid peroxidation and neuronal damage in rat brain ischemia. J Nutr 2001; 131:1100S-5S. [PMID: 11238825 DOI: 10.1093/jn/131.3.1100s] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The efficacy of S-allylcysteine (SAC) as a free radical scavenger was studied using rat brain ischemia models. In a middle cerebral artery occlusion model, preischemic administration of SAC had the following effects: it improved motor performance and memory impairment and reduced water content and the infarct size. In a transient global ischemia model, the time course of free radical (alkoxyl radical) formation as studied by electron paramagnetic resonance (EPR) spectroscopy and alpha-phenyl-N-tert-butylnitrone (PBN) was biphasic; the first peak occurred at 5 min and the second at 20 min after reperfusion. Although SAC did not attenuate the first peak, it did affect the second peak, which is related to lipid peroxidation. The lipid peroxidation as estimated by thiobarbituric acid reactive substances (TBARS) increased significantly at 20 min after reperfusion. SAC decreased TBARS to the levels found without ischemia. These results suggest that SAC could have beneficial effects in brain ischemia and that the major protective mechanism may be the inhibition of free radical-mediated lipid peroxidation.
Collapse
Affiliation(s)
- Y Numagami
- Philadelphia Biomedical Research Institute, King of Prussia, PA 19406, USA
| | | |
Collapse
|
225
|
Rao AM, Hatcher JF, Kindy MS, Dempsey RJ. Arachidonic acid and leukotriene C4: role in transient cerebral ischemia of gerbils. Neurochem Res 1999; 24:1225-32. [PMID: 10492517 DOI: 10.1023/a:1020916905312] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Accumulation of arachidonic acid (AA) is greatest in brain regions most sensitive to transient ischemia. Free AA released after ischemia is either: 1) reincorporated into the membrane phospholipids, or 2) oxidized during reperfusion by lipoxygenases and cyclooxygenases, producing leukotrienes (LT), prostaglandins, thromboxanes and oxygen radicals. AA, its metabolite LTC4 and lipid peroxides (generated during AA metabolism) have been implicated in the blood-brain barrier (BBB) dysfunction, edema and neuronal death after ischemia/reperfusion. This report describes the time course of AA release, LTC4 accumulation and association with the physiological outcome during transient cerebral ischemia of gerbils. Significant amount of AA was detected immediately after 10 min ischemia (0 min reperfusion) which returned to sham levels within 30 min reperfusion. A later release of AA occurred after 1 d. LTC4 levels were elevated at 0-6 h and 1 d after ischemia. Increased lipid peroxidation due to AA metabolism was observed between 2-6 h. BBB dysfunction occurred at 6 h. Significant edema developed at 1 and 2 d after ischemia and reached maximum at 3 d. Ischemia resulted in approximately 80% neuronal death in the CA1 hippocampal region. Pretreatment with a 5-lipoxygenase inhibitor, AA861 resulted in significant attenuation of LTC4 levels (Baskaya et al. 1996. J. Neurosurg. 85: 112-116) and CA1 neuronal death. Accumulation of AA and LTC4, together with highly reactive oxygen radicals and lipid peroxides, may alter membrane permeability, resulting in BBB dysfunction, edema and ultimately to neuronal death.
Collapse
Affiliation(s)
- A M Rao
- Department of Neurological Surgery, University of Wisconsin, Madison 53792-3232, USA.
| | | | | | | |
Collapse
|
226
|
Igarashi T, Sakurai K, Oi T, Obara H, Ohya H, Kamada H. New sensitive agents for detecting singlet oxygen by electron spin resonance spectroscopy. Free Radic Biol Med 1999; 26:1339-45. [PMID: 10381208 DOI: 10.1016/s0891-5849(98)00291-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Free radicals are well-established transient intermediates in chemical and biological processes. Singlet oxygen, though not a free radical, is also a fairly common reactive chemical species. It is rare that singlet oxygen is studied with the electron spin resonance (ESR) technique in biological systems, because there are few suitable detecting agents. We have recently researched some semiquinone radicals. Specifically, our focus has been on bipyrazole derivatives, which slowly convert to semiquinone radicals in DMSO solution in the presence of potassium tert-butoxide and oxygen. These bipyrazole derivatives are dimers of 3-methyl-1-phenyl-2-pyrazolin-5-one and have anti-ischemic activities and free radical scavenging properties. In this work, we synthesized a new bipyrazole derivative, 4,4'-bis(1p-carboxyphenyl-3-methyl-5-hydroxyl)-pyrazole, DRD156. The resulting semiquinone radical, formed by reaction with singlet oxygen, was characterized by ESR spectroscopy. DRD156 gave no ESR signals from hydroxyl radical, superoxide, and hydrogen peroxide. DRD156, though, gives an ESR response with hypochlorite. This agent, nevertheless, has a much higher ability to detect singlet oxygen than traditional agents with the ESR technique.
Collapse
Affiliation(s)
- T Igarashi
- Research Center, Daiichi Radioisotope Laboratories, Ltd., Chiba, Japan.
| | | | | | | | | | | |
Collapse
|
227
|
Nakashima M, Niwa M, Iwai T, Uematsu T. Involvement of free radicals in cerebral vascular reperfusion injury evaluated in a transient focal cerebral ischemia model of rat. Free Radic Biol Med 1999; 26:722-9. [PMID: 10218662 DOI: 10.1016/s0891-5849(98)00257-3] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Free radicals have been suggested to be largely involved in the genesis of ischemic brain damage, as shown in the protective effects of alpha-phenyl-N-tert-butyl nitrone (PBN), a spin trapping agent, against ischemic cerebral injury. In the present study, the effects of PBN as well as MCI-186, a newly-developed free radical scavenger, and oxypurinol, an inhibitor of xanthine oxidase, were evaluated in a rat transient middle cerebral aretery (MCA) occlusion model to clarify the possible role of free radicals in the reperfusion injury of brain. The volume of cerebral infarction, induced by 2-h occlusion and subsequent 2-h reperfusion of MCA in Fisher-344 rats, was evaluated. The administration of PBN (100 mg/kg) and MCI-186 (100 mg/kg) just before reperfusion of MCA significantly reduced the infarction volume. In contrast, oxypurinol (100 mg/kg) failed to show any preventive effect on the infarction. These results suggest that free radical formation is involved in the cerebral damage induced by ischemia-reperfusion of MCA, and that hydroxyl radical is responsible for the reperfusion injury after transient focal brain ischemia. It is also suggested that xanthine oxidase is not a major source of free radicals.
Collapse
Affiliation(s)
- M Nakashima
- Department of Pharmacology, Gifu University School of Medicine, Japan
| | | | | | | |
Collapse
|
228
|
Takamatsu H, Kondo K, Ikeda Y, Umemura K. Neuroprotective effects depend on the model of focal ischemia following middle cerebral artery occlusion. Eur J Pharmacol 1998; 362:137-42. [PMID: 9874163 DOI: 10.1016/s0014-2999(98)00773-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The purpose of the present study was to compare the characteristics of the photochemical-induced thrombotic occlusion model and the thermocoagulated occlusion model of the middle cerebral artery in rats. We evaluated the neuroprotective effects of a NMDA receptor antagonist, (+)-MK-801 (dizocilpine, (+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cycloheptan-5,10-imine), an alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor antagonist, YM90K (6-(1H-imidazol-1-yl)-7-nitro-2,3(1H,4H)-quinoxalinedione monohydrochloride), a Ca2+ channel antagonist, S-312-d (S-(+)-methyl-4,7-dihydro-3-isobutyl-6-methyl-4-(3-nitrophenyl)-thieno[2 ,3-b]pyridine-5-carboxylate), the radical scavengers, MCI-186 (3-methyl-1-phenyl-2-pyrazolin-5-one) and EPC-K1 (L-ascorbic acid 2-[3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyl-tridecyl)-2H-1-be nzopyran-6yl-hydrogen phosphate] potassium salt), and a calcineurin inhibitor, FK506 (tacrolimus, Prograf). Although all tested agents in the present study attenuated the brain damage in the photochemical-induced thrombotic occlusion model, the radical scavengers did not attenuate the brain damage in the thermocoagulated occlusion model. The time course of brain damage and brain edema formation in the two models was examined. The time course of brain damage was not different in the two models, but the time course of brain edema was quite different. Brain edema formation in the photochemical-induced thrombotic occlusion model was significantly greater (P < 0.01) than that in the thermocoagulated occlusion model at all time point studied until 24 h after occlusion of the middle cerebral artery. The present study suggests that the photochemical-induced thrombotic occlusion model has characteristics of both permanent ischemia and ischemia-reperfusion.
Collapse
Affiliation(s)
- H Takamatsu
- Department of Pharmacology, Hamamatsu University School of Medicine, Japan.
| | | | | | | |
Collapse
|
229
|
Abstract
1. The original concept of the ischemic penumbra surrounding a focus of dense cerebral ischemia is based on electrophysiological observations. In the cortex of baboons following middle cerebral artery occlusion, complete failure of the cortical evoked potential was observed at a cerebral blood flow (CBF) threshold level of approx. 0.15 ml/g/min--a level at which extracellular potassium ion activity was only mildly elevated. With a greater CBF decrement to the range of 0.06-0.10 ml/g/min, massive increases in extracellular potassium occurred and were associated with complete tissue infarction. Thus, the ischemic penumbra has been conceptualized as a region in which CBF reduction has exceeded the threshold for failure of electrical function but not that for membrane failure. 2. Recent studies demonstrate that the penumbra as defined classically by the flow thresholds does not survive prolonged periods of ischemia. The correlation of CBF autoradiograms with diffusion-weighted MR images and the regional distribution of cerebral metabolites reveals that the ischemic core region enlarges when adjacent, formerly penumbral, areas undergo irreversible deterioration during the initial hours of vascular occlusion. At the same time, the residual penumbra becomes restricted to the periphery of the ischemic territory, and its fate may depend critically upon early therapeutic intervention. 3. In the border zone of brain infarcts, marked uncoupling of local CBF and glucose utilization is consistently observed. The correlation with electrophysiological measurements shows that metabolism-flow uncoupling is associated with sustained deflections of the direct current (DC) potential resembling transient depolarizations. Such penumbral cell depolarizations, which are associated with an increased metabolic workload, induce episodes of tissue hypoxia due to the constrained collateral flow, stimulate anaerobic glycolysis leading to lactacidosis, suppress protein synthesis, and, finally, compromise energy metabolism. The frequency of their occurrence correlates with the final volume of ischemic injury. Therefore, penumbral depolarizations are regarded as a key event in the pathogenesis of ischemic brain injury. Periinfarct DC deflections can be suppressed by NMDA and non-NMDA antagonists, resulting in a significant reduction of infarct size. 4. The histopathological sequelae within the penumbra consist of various degrees of scattered neuronal injury, also termed "incomplete infarction." The reduction of neuronal density at the infarct border is a flow- and time-dependent event which is accompanied by an early response of glial cells. As early as 3 hr after vascular occlusion a generalized microglial activation can be detected throughout the ipsilateral cortex. Astrocytic activation is observed in the intact parts of the ischemic hemisphere from 6 hr postocclusion onward. Thus, the penumbra is a spatially dynamic brain region of limited viability which is characterized by complex pathophysiological changes involving neuronal function as well as well as glial activation in response to local ischemic injury.
Collapse
Affiliation(s)
- T Back
- Department of Neurology, Ludwig Maximilians University, Munich, Germany.
| |
Collapse
|
230
|
McIntosh TK, Juhler M, Wieloch T. Novel pharmacologic strategies in the treatment of experimental traumatic brain injury: 1998. J Neurotrauma 1998; 15:731-69. [PMID: 9814632 DOI: 10.1089/neu.1998.15.731] [Citation(s) in RCA: 243] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The mechanisms underlying secondary or delayed cell death following traumatic brain injury are poorly understood. Recent evidence from experimental models suggests that widespread neuronal loss is progressive and continues in selectively vulnerable brain regions for months to years after the initial insult. The mechanisms underlying delayed cell death are believed to result, in part, from the release or activation of endogenous "autodestructive" pathways induced by the traumatic injury. The development of sophisticated neurochemical, histopathological and molecular techniques to study animal models of TBI have enabled researchers to begin to explore the cellular and genomic pathways that mediate cell damage and death. This new knowledge has stimulated the development of novel therapeutic agents designed to modify gene expression, synthesis, release, receptor or functional activity of these pathological factors with subsequent attenuation of cellular damage and improvement in behavioral function. This article represents a compendium of recent studies suggesting that modification of post-traumatic neurochemical and cellular events with targeted pharmacotherapy can promote functional recovery following traumatic injury to the central nervous system.
Collapse
Affiliation(s)
- T K McIntosh
- Department of Neurosurgery, University of Pennsylvania, Philadelphia 19104-6316, USA
| | | | | |
Collapse
|
231
|
Polidori MC, Frei B, Cherubini A, Nelles G, Rordorf G, Keaney JF, Schwamm L, Mecocci P, Koroshetz WJ, Beal MF. Increased plasma levels of lipid hydroperoxides in patients with ischemic stroke. Free Radic Biol Med 1998; 25:561-7. [PMID: 9741593 DOI: 10.1016/s0891-5849(98)00085-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A large body of experimental research indicates that the generation of free radicals leading to oxidative stress plays a role in the pathogenesis of ischemic brain injury, but evidence in humans is limited. We examined plasma levels of lipid hydroperoxides (measured as cholesteryl ester hydroperoxides, CEOOH) and ascorbic acid in 32 patients with cortical stroke, as compared with 13 patients with lacunar infarct. Patients with cortical stroke had significantly increased levels of CEOOH, which peaked on Day 5 after the ictus. Small decreases in ascorbic acid concentrations were not significant. There was a significant positive correlation of CEOOH with the NIH stroke scale, and a significant negative correlation with the Glasgow coma scale. Concentrations of CEOOH were significantly higher in patients with total anterior cerebral syndrome as compared with patients with partial anterior cerebral syndrome or posterior cerebral syndrome. Stroke volumes computed from CT or MRI scans were significantly correlated with plasma CEOOH levels. These findings implicate oxidative stress in ischemic brain injury in humans and suggest that measurements of CEOOH in plasma may be useful both prognostically as well as in monitoring therapeutic interventions.
Collapse
Affiliation(s)
- M C Polidori
- Neurology Service, Massachusetts General Hospital and Harvard Medical School, Boston 02114, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
232
|
Takamatsu H, Kondo K, Ikeda Y, Umemura K. Hydroxyl radical generation after the third hour following ischemia contributes to brain damage. Eur J Pharmacol 1998; 352:165-9. [PMID: 9716351 DOI: 10.1016/s0014-2999(98)00353-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The purpose of the present study was to determine after what time period hydroxyl radical formation contributes most to ischemic brain damage in focal ischemia, using a hydroxyl radical scavenger, EPC-K1, L-ascorbic acid 2-[3,4-dihydro-2,5,7,8-tetramethyl-2-(4,8,12-trimethyl-tridecyl)-2H-1-be nzopyran-6yl-hydrogen phosphate] potassium salt. Focal ischemia was produced by thrombotic occlusion of the left middle cerebral artery in rats. After evaluation of the pharmacokinetics of EPC-K in the brain tissue and plasma following 10 mg/kg intravenous bolus treatment of conscious rats, we investigated the neuroprotective effect of EPC-K1 in the middle cerebral artery occlusion model. A single intravenous bolus of EPC-K1 was given immediately, 3 or 6 h after ischemia, and cerebral brain damage was measured 24 h after ischemia. When EPC-K1 was injected 3 h after ischemia, a significant (P < 0.01) reduction of cerebral brain damage was observed. EPC-K1 delivered by intravenous infusion that started immediately after ischemia and lasted for 24 h, also significantly (P < 0.05) reduced brain damage, but the efficacy of the neuroprotective effect was the same as that of the 3 h after ischemia bolus treatment. These results may indicate that the period of hydroxyl radical formation most critical for ischemic brain damage is a few hours after the third hour following ischemia in this model.
Collapse
Affiliation(s)
- H Takamatsu
- Department of Pharmacology, Hamamatsu University School of Medicine, Japan
| | | | | | | |
Collapse
|
233
|
Mizuno A, Umemura K, Nakashima M. Inhibitory effect of MCI-186, a free radical scavenger, on cerebral ischemia following rat middle cerebral artery occlusion. GENERAL PHARMACOLOGY 1998; 30:575-8. [PMID: 9522178 DOI: 10.1016/s0306-3623(97)00311-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. In this study, we investigated the effect of a radical scavenger, MCI-186 (3-methyl-1-phenyl-2-pyrazolin-5-one), on cerebral damage induced by rat middle cerebral artery (MCA) occlusion, and further measured the hydroxyl radical level at the ischemic border zone using a microdialysis technique. 2. MCI-186, at a dose of 3 mg/kg per 30 min, was administered as a continuous infusion two times for 30 min, starting 20 min and then 80 min after Rose Bengal injection. 3. MCI-186 significantly (P < 0.05) reduced size of cerebral damage 24 hr after MCA occlusion and significantly (P < 0.05) reduced hydroxyl radical level.
Collapse
Affiliation(s)
- A Mizuno
- Department of Pharmacology, Hamamatsu University School of Medicine, Japan
| | | | | |
Collapse
|
234
|
Peters O, Back T, Lindauer U, Busch C, Megow D, Dreier J, Dirnagl U. Increased formation of reactive oxygen species after permanent and reversible middle cerebral artery occlusion in the rat. J Cereb Blood Flow Metab 1998; 18:196-205. [PMID: 9469163 DOI: 10.1097/00004647-199802000-00011] [Citation(s) in RCA: 235] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In barbiturate-anesthetized rats, we induced 3 hours of permanent middle cerebral artery occlusion (MCAO) by an intraluminal thread (n = 6), or 1 hour MCAO followed by 2 hours of reperfusion (n = 6). Through a closed cranial window over the parietal cortex, the production of reactive oxygen species (ROS) was measured in the infarct border using online in vivo chemiluminescence (CL) while monitoring the appearance of peri-infarct depolarizations (PID). The borderzone localization of the ROS and direct current (DC) potential measurements was confirmed in additional experiments using laser-Doppler scanning, mapping regional CBF changes through the cranial window after permanent (n = 5) or reversible (n = 5) MCAO. CL measurements revealed a short period (10 to 30 minutes) of reduced ROS formation after vessel occlusion, followed by a significant increase (to 162 +/- 51%; baseline = 100%; P < .05) from 100 minutes of permanent MCAO onward. Reperfusion after a 1-hour period of MCAO led to a burst-like pattern of ROS production (peak: 489 +/- 330%; P < .05). When the experiments were terminated 3 hours after induction of MCAO, CL was still significantly increased above baseline after permanent and reversible MCAO (to 190 +/- 67% and 211 +/- 64%, respectively; P < .05). Simultaneous DC potential recordings detected 6.4 +/- 2.7 PID in the first, 4.7 +/- 2.3 in the second, and 2.8 +/- 2.0 in the third hour after permanent MCAO. In animals with reversible MCAO, PID were abolished from 15-minutes recirculation onward. There was no temporal relationship between ROS production and peri-infarct DC potential shifts. In conclusion, using a high temporal resolution ROS detection technique (CL), we found that permanent MCAO (after an initial decrease) was accompanied by a steady increase of ROS production during the 3-hour observation period, while reperfusion after 1 hour of MCAO produced a burst in ROS formation. Both patterns of ROS production were not related to the occurrence of PID.
Collapse
Affiliation(s)
- O Peters
- Department of Neurology, Charité Hospital, Humboldt-University Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
235
|
Yuki S, Kogure K. The changes of LCGU and rCBF in the MCA occlusion-recirculation model in rats and the ameliorating effect of MCI-186, a novel free radical scavenger. MOLECULAR AND CHEMICAL NEUROPATHOLOGY 1997; 32:123-8. [PMID: 9437662 DOI: 10.1007/bf02815171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
We examined the change of regional cerebral blood flow (rCBF) and local cerebral glucose utilization (LCGU) in the middle cerebral artery (MCA) occlusion or recirculation model of rats, and tested anti-ischemic effects of a free radical scavenger, 3-methyl-1-phenyl-pyrazolon-5-one (MCI-186). A remarkable increase in LCGU was observed in the cortex supplied by the anterior cerebral artery after recirculation. This hypermetabolism of glucose was at least partly caused by the postischemic oxidative injury, since MCI-186 ameliorated the high LCGU in this area. These results suggested the usefulness of this type of free radical scavenger for inhibiting the postischemic injury.
Collapse
Affiliation(s)
- S Yuki
- Pharmaceuticals Laboratory 1, Mitsubishi Chemical Corporation, Yokohama, Japan.
| | | |
Collapse
|
236
|
Abstract
Traumatic injury to the central nervous system induces delayed neuronal death, which may be mediated by acute and chronic neurochemical changes. Experimental identification of these injury mechanisms and elucidation of the neurochemical cascade following trauma may provide enhanced opportunities for treatment with novel neuroprotective strategies.
Collapse
Affiliation(s)
- T K McIntosh
- Department of Neurosurgery, Bioengineering, and Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, USA
| | | | | | | |
Collapse
|
237
|
Chung HY, Baek BS, Song SH, Kim MS, Huh JI, Shim KH, Kim KW, Lee KH. Xanthine dehydrogenase/xanthine oxidase and oxidative stress. AGE 1997; 20:127-40. [PMID: 23604305 PMCID: PMC3455892 DOI: 10.1007/s11357-997-0012-2] [Citation(s) in RCA: 109] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Xanthine dehydrogenase (XDH) and xanthine oxidase (XOD) are single-gene products that exist in separate but interconvertible forms. XOD utilizes hypoxanthine or xanthine as a substrate and O2 as a cofactor to produce superoxide (·O2 (-)) and uric acid. XDH acts on these same substrates but utilizes NAD as a cofactor to produce NADH instead of ·O2 (-) and uric acid. XOD has been proposed as a source of oxygen radicals in polymorphonuclear, endothelial, epithelial, and connective tissue cells. However, several questions remain about the physiological significance and functions of XOD on aging and oxidative stress. XOD is reported to play an important role in cellular oxidative status, detoxification of aldehydes, oxidative injury in ischemia-reperfusion, and neutrophil mediation. For example, XOD may serve as a messenger or mediator in the activation of neutrophil, T cell, cytokines, or transcription in defense mechanisms rather than as a free radical generator of tissue damage. Emerging evidence on the synergistic interactions of ·O2 (-), a toxic product of XOD and nitric oxide, may be another illustration of XOD involvement in tissue injury and cytotoxicity in an emergent condition such as ischemia or inflammation.
Collapse
|
238
|
Ono S, Okazaki K, Sakurai M, Inoue Y. Density Functional Study of the Radical Reactions of 3-Methyl-1-phenyl-2-pyrazolin-5-one (MCI-186): Implication for the Biological Function of MCI-186 as a Highly Potent Antioxidative Radical Scavenger. J Phys Chem A 1997. [DOI: 10.1021/jp963267m] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Satoshi Ono
- Yokohama Research Center, Mitsubishi Chemical Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227, Japan, and Department of Biomolecular Engineering, Tokyo Institute of Technology, 4259, Nagatsuta-cho, Midori-ku, Yokohama 226, Japan
| | - Keiji Okazaki
- Yokohama Research Center, Mitsubishi Chemical Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227, Japan, and Department of Biomolecular Engineering, Tokyo Institute of Technology, 4259, Nagatsuta-cho, Midori-ku, Yokohama 226, Japan
| | - Minoru Sakurai
- Yokohama Research Center, Mitsubishi Chemical Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227, Japan, and Department of Biomolecular Engineering, Tokyo Institute of Technology, 4259, Nagatsuta-cho, Midori-ku, Yokohama 226, Japan
| | - Yoshio Inoue
- Yokohama Research Center, Mitsubishi Chemical Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama 227, Japan, and Department of Biomolecular Engineering, Tokyo Institute of Technology, 4259, Nagatsuta-cho, Midori-ku, Yokohama 226, Japan
| |
Collapse
|
239
|
|
240
|
|
241
|
Abstract
Reactive oxygen species are thought to be involved in a number of types of acute and chronic pathologic conditions in the brain and neural tissue. The metabolic antioxidant alpha-lipoate (thioctic acid, 1, 2-dithiolane-3-pentanoic acid; 1, 2-dithiolane-3 valeric acid; and 6, 8-dithiooctanoic acid) is a low molecular weight substance that is absorbed from the diet and crosses the blood-brain barrier. alpha-Lipoate is taken up and reduced in cells and tissues to dihydrolipoate, which is also exported to the extracellular medium; hence, protection is afforded to both intracellular and extracellular environments. Both alpha-lipoate and especially dihydrolipoate have been shown to be potent antioxidants, to regenerate through redox cycling other antioxidants like vitamin C and vitamin E, and to raise intracellular glutathione levels. Thus, it would seem an ideal substance in the treatment of oxidative brain and neural disorders involving free radical processes. Examination of current research reveals protective effects of these compounds in cerebral ischemia-reperfusion, excitotoxic amino acid brain injury, mitochondrial dysfunction, diabetes and diabetic neuropathy, inborn errors of metabolism, and other causes of acute or chronic damage to brain or neural tissue. Very few neuropharmacological intervention strategies are currently available for the treatment of stroke and numerous other brain disorders involving free radical injury. We propose that the various metabolic antioxidant properties of alpha-lipoate relate to its possible therapeutic roles in a variety of brain and neuronal tissue pathologies: thiols are central to antioxidant defense in brain and other tissues. The most important thiol antioxidant, glutathione, cannot be directly administered, whereas alpha-lipoic acid can. In vitro, animal, and preliminary human studies indicate that alpha-lipoate may be effective in numerous neurodegenerative disorders.
Collapse
Affiliation(s)
- L Packer
- Department of Molecular and Cell Biology, University of California, Berkeley 94720-3200, USA
| | | | | |
Collapse
|
242
|
Bacon PJ, Love SA, Gupta AK, Kirkpatrick PJ, Menon DK. Plasma antioxidant consumption associated with ischemia/reperfusion during carotid endarterectomy. Stroke 1996; 27:1808-11. [PMID: 8841335 DOI: 10.1161/01.str.27.10.1808] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE Thirteen patients undergoing carotid endarterectomy were studied to correlate changes in jugular bulb venous oxygen saturation (SjO2) with indices of free radical production during cerebral ischemia/reperfusion. Levels of oxidant species were also monitored in arterial samples to determine any change across the cerebral circulation. METHODS Blood was sampled from a venous catheter inserted in the ipsilateral jugular bulb and from an arterial catheter. Co-oximetry measurements were made to determine jugular bulb venous oxygenation saturation. To monitor changes in oxidant stress, a colorimetric assay was used to determine plasma antioxidant potential, and electron paramagnetic resonance spectroscopy was used to quantify free radical-spin trap adducts formed in a blood sample treated with the spin trap alpha-tert-butyl phenyl nitrone (PBN). RESULTS SjO2 decreased significantly from 68 +/- 11% to 61 +/- 10% (P < .05) during clamping of the internal carotid artery and returned to baseline (65 +/- 11%) when the carotid clamp was removed. Jugular venous plasma antioxidant potential decreased significantly from 32.76 +/- 5.42% inhibition to 28.02 +/- 6.77% inhibition (P < .05). There was no concomitant change in arterial plasma antioxidant potential values, indicating a decrease in antioxidant capacity across the cerebral circulation. However, analysis of spin trap-free radical adducts did not provide conclusive evidence for free radical production. CONCLUSIONS These results provide supportive evidence for oxidant production during cerebral ischemia/reperfusion in a clinical setting.
Collapse
Affiliation(s)
- P J Bacon
- University Department of Anesthesia, Addenbrooke's Hospital, Cambridge, UK
| | | | | | | | | |
Collapse
|
243
|
Inoue N, Yamamoto YL, Ito Y, Clemens JA, Panetta JK, Diksic M. The efficacy of retrograde infusion with LY231617 in a rat middle cerebral artery occlusion model. Neurol Sci 1996; 23:175-83. [PMID: 8862838 DOI: 10.1017/s0317167100038476] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND AND PURPOSE We examined the efficacy of the antioxidant LY231617 administered five hours following middle cerebral artery (MCA) occlusion in rats. METHODS The treatment was contrived for a two hour interval. Group A (n = 16) was left untreated. Group B (n = 16) received an intravenous infusion of LY231617. Group C (n = 16) received saline (86 microliters/min) by retrograde infusion of the cerebral vein (RICV). Group D (n = 22) was administered LY231617 (10mg/kg/2 hr) in saline (86 microliters/min) by RICV. Local cerebral blood flow with [14C]-iodoantipyrine and blood-brain transfer constant with 14C-alpha-amino-isobutyric acid were examined. Early ischemic damage was histologically examined with cresyl violet and Luxol fast blue and with triphenyl-tetrazolium chloride. RESULTS The results revealed a marked increase in local cerebral blood flow (over 600%, p < 0.01) after RICV with LY231617, with a significant improvement of BBB permeability in rats from group D. Ischemic brain damage measured with Luxol fast blue and triphenyl-tetrazolium chloride methods showed a significant improvement (50-91%) of ischemic damage in group D, as compared to groups B and C. CONCLUSION Retrograde infusion of the cerebral vein with LY231617 resulted in a significant amelioration at seven hours post MCA occlusion.
Collapse
Affiliation(s)
- N Inoue
- Neuroisotope Laboratory, Mc Gill University, Montreal, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
244
|
Minhaz U, Tanaka M, Tsukamoto H, Watanabe K, Koide S, Shohtsu A, Nakazawa H. Effect of MCI-186 on postischemic reperfusion injury in isolated rat heart. Free Radic Res 1996; 24:361-7. [PMID: 8733940 DOI: 10.3109/10715769609088034] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
MCI-186 (3-methyl-1-phenyl-2-pyrazolin-5-one) is a newly developed antioxidant which has been shown to reduce brain edema in cerebral ischemia through inhibition of the lipoxygenase pathway of arachidonic acid. However, its effect on myocardial reperfusion injury after prolonged ischemia has not yet been demonstrated. We compared the mode of the effect of MIC-186 and recombinant human CuZn superoxide dismutase (rh-SOD) in isolated perfused rat hearts subjected to 60-min ischemia followed by 60-min reperfusion. Left ventricular developed pressure (LVDP), necrotic area and the release of creatine phosphokinase (CPK) and endogenous CuZn superoxide dismutase (endoge-SOD) were measured to evaluate myocardial damage. The decrease in left coronary flow (CBF) was measured as an index of the damage of left coronary circulation. MCI-186 (14.5 mg/L) was perfused for 10 min in the MCI group and rh-SOD (70 mg/L) was perfused during the reperfusion period in the SOD group starting 5 min prior to reperfusion. The release patterns of CPK and endoge-SOD were analyzed to elucidate the difference in the mode of protection of MCI-186 and rh-SOD. The LVDP remained higher in both MCI and SOD groups than that of control (76 +/- 1, 77 +/- 2 and 69 +/- 1% of preischemic value, respectively). The necrotic area was significantly attenuated in both MCI and SOD groups compared with that in the control group (16 +/- 1, 14 +/- 1 and 32 +/- 1%, respectively, p < 0.05). Total CPK release was lower in both MCI and SOD groups than in the control (78 +/- 7, 100 +/- 2 and 116 +/- 4 x 10(3) units/g myocardium respectively). The decrease in CPK release was more marked in the MCI group than that in the SOD group (p < 0.05). The reduction in CBF was significantly attenuated by the treatment with rh-SOD or MCI-186, but the effect was much higher in the SOD group than in the MCI group (69 +/- 5, 58 +/- 2, and 48 +/- 2% in SOD, MCI and control groups, respectively). The release pattern of endoge-SOD was identical to that of CPK and thus this did not distinguish the mode of effect of MCI-186 from that of rh-SOD. These results indicate that MCI-186 reduces reperfusion injury in isolated perfused hearts with prolonged ischemia and the effect is more closely related to the reduction of myocyte damage than the preservation of the coronary circulation.
Collapse
Affiliation(s)
- U Minhaz
- First Department of Surgery, Tokai University School of Medicine, Kanagawa, Japan
| | | | | | | | | | | | | |
Collapse
|
245
|
|
246
|
Fischer S, Renz D, Schaper W, Karliczek GF. Effects of barbiturates on hypoxic cultures of brain derived microvascular endothelial cells. Brain Res 1996; 707:47-53. [PMID: 8866712 DOI: 10.1016/0006-8993(95)01219-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
An in vitro model of the blood-brain barrier (BBB) consisting of porcine brain derived microvascular endothelial cells (BMEC) seeded onto collagen-coated polycarbonate membranes was used to investigate the effects of the barbiturates, methohexital and thiopental, on permeability properties of the endothelial cell monolayer under hypoxia. The permeability of cultured BMEC to ions and sucrose increased significantly during 6 h of hypoxia in a reversible manner. Cells were resistant to hypoxia for up to 24 h, but 48 h resulted in marked damage as assessed by the release of lactate dehydrogenase activity into the culture medium. The hypoxia-induced increase of the permeability was unchanged in the presence of superoxide dismutase (SOD) and catalase. Methohexital and thiopental decreased the hypoxia-induced permeability increase in a concentration-dependent manner and permeability changes were abolished completely at the barbiturate concentration of 50 micrograms/ml. The barbiturates had no effect on the intracellular cAMP content which started to decline after 3 h of hypoxia. Results suggest that barbiturates at high concentrations might be able to prevent permeability changes of the BBB during cerebral ischemia.
Collapse
Affiliation(s)
- S Fischer
- Department of Anesthesiology and Intensive Care, Max-Planck Institute for Physiological and Clinical Research, Bad Nauheim, Germany
| | | | | | | |
Collapse
|
247
|
Morimoto T, Globus MY, Busto R, Martinez E, Ginsberg MD. Simultaneous measurement of salicylate hydroxylation and glutamate release in the penumbral cortex following transient middle cerebral artery occlusion in rats. J Cereb Blood Flow Metab 1996; 16:92-9. [PMID: 8530561 DOI: 10.1097/00004647-199601000-00011] [Citation(s) in RCA: 108] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Using the microdialysis technique and laser-Doppler flowmetry, we performed simultaneous measurement of salicylate hydroxylation and glutamate release along with local CBF in the ischemic penumbral cortex of rat brain subjected to normothermic transient middle cerebral artery (MCA) occlusion. Cortical CBF fell to 24 +/- 11% (mean +/- SD) during ischemia and recovered to 84 +/- 16% during reperfusion. Extracellular glutamate levels increased by 6.5-fold above baseline 10 min following MCA occlusion but subsequently returned to near baseline levels in spite of the persistent ischemia. Increase in 2,3- and 2,5-dihydroxybenzoic acid (DHBA) concentrations in the microdialysis perfusate was confirmed during both ischemia and reperfusion phase. Although the temporal profile and amount of salicylate hydroxylation were heterogeneous among individual animals, integrated 2,3-DHBA concentrations during reperfusion were correlated positively with integrated glutamate concentrations during ischemia and negatively with mean postischemic CBF. These relationships suggest a possible association of the enhanced production of 2,3-DHBA during reperfusion with larger amounts of intraischemic glutamate release and lower levels of post-ischemic CBF.
Collapse
Affiliation(s)
- T Morimoto
- Department of Neurology, University of Miami School of Medicine, Florida 33101, USA
| | | | | | | | | |
Collapse
|
248
|
Abstract
In this article we summarize a wide variety of properties of arachidonic acid (AA) in the mammalian nervous system especially in the brain. AA serves as a biologically-active signaling molecule as well as an important component of membrane lipids. Esterified AA is liberated from the membrane by phospholipase activity which is stimulated by various signals such as neurotransmitter-mediated rise in intracellular Ca2+. AA exerts many biological actions which include modulation of the activities of protein kinases and ion channels, inhibition of neurotransmitter uptake, and enhancement of synaptic transmission. AA serves also as a precursor of a variety of eicosanoids, which are formed by oxidative metabolism of AA. AA cascade is activated under several pathological conditions in the brain such as ischemia and seizures, and may be involved in irreversible tissue damage. On the other hand, AA can show beneficial influences on brain tissues and cells in several situations. In a recent study using cultured brain neurons, we have found that AA shows quite distinct actions at a narrow concentration range, such as induction of cell death, promotion of cell survival and enhancement of neurite extension. The neurotoxic action is mediated by free radicals generated by AA metabolism, whereas the neurotrophic actions are exerted by AA itself. The observed in vitro actions of AA might be related to important roles of AA in brain pathogenesis and neural development.
Collapse
Affiliation(s)
- H Katsuki
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, University of Tokyo, Japan
| | | |
Collapse
|
249
|
Abstract
During perinatal asphyxia several mechanisms aim to limit cerebral damage. However, when the degree of asphyxia passes beyond a certain threshold, brain damage is inevitable. This review focuses on the various factors determining the final cerebral outcome. Metabolic and biochemical events, such as the intracellular level of calcium, the formation of oxygen derived free radicals, the release of excitotoxic neurotransmitters and the interrelationship of these parameters are discussed. Furthermore, steps possibly useful to pharmacologic intervention aiming to reduce cerebral damage are presented.
Collapse
Affiliation(s)
- H H de Haan
- Department of Obstetrics and Gynecology, University Hospital, AZ Maastricht, The Netherlands
| | | |
Collapse
|
250
|
Safi HJ, Iliopoulos DC, Gopinath SP, Hess KR, Asimacopoulos PJ, Bartoli S, Raskin SA, Shaibani AT, Leveque CM, Yawn DH. Retrograde cerebral perfusion during profound hypothermia and circulatory arrest in pigs. Ann Thorac Surg 1995; 59:1107-12. [PMID: 7733705 DOI: 10.1016/0003-4975(95)00122-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The purpose of this study was to evaluate the use of retrograde cerebral perfusion via the superior vena cava during profound hypothermia and circulatory arrest (CA) in pigs. In three groups of 5 pigs each, group A (control) underwent cardiopulmonary bypass and normothermic CA for 1 hour, group B underwent cardiopulmonary bypass, profound hypothermia, and CA (15 degrees C nasopharyngeal) for 1 hour, and group C underwent the same procedure as group B plus retrograde cerebral perfusion. In group A none awoke. In group B, 2 of 5 did not awake and 3 of 5 awoke unable to stand, 2 with perceptive hind limb movement and 1 moving all extremities. In group C all awoke, 4 of 5 able to stand and 1 of 5 unable to stand but moving all limbs. In neurologic evaluation group B had significantly lower Tarlov scores than group C (p = 0.0090). Group B mean wake-up time, plus or minus standard error of the mean, was 124.6 +/- 4.6 minutes versus 29.2 +/- 5.1 in group C (p = 0.0090). In group B late phase CA cerebral blood flow dropped 30.9% +/- 4.8%, but in group C it rose 24.7% +/- 9.3% (p = 0.0007, pooled variance t test, two-tailed). In group B late phase CA brain oxygenation decreased 46.0% +/- 13.9% but it increased 26.1% +/- 5.4% in group C (p = 0.0013). This difference was reduced somewhat during rewarming (B, -21.2% +/- 14.9%; C, 16.4% +/- 4.7%; p = 0.043). Group B rewarming jugular venous O2 saturation was 30.8% +/- 2.5% versus 56.0% +/- 4.4% in group C (p = 0.0011). We conclude that in pigs retrograde cerebral perfusion combined with profound hypothermia during CA significantly reduces neurologic dysfunction, providing superior brain protection.
Collapse
Affiliation(s)
- H J Safi
- Baylor College of Medicine, Methodist Hospital, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|