1
|
Neuroprotective Effect of 1,3-dipalmitoyl-2-oleoylglycerol Derived from Rice Bran Oil against Cerebral Ischemia-Reperfusion Injury in Rats. Nutrients 2022; 14:nu14071380. [PMID: 35405992 PMCID: PMC9003438 DOI: 10.3390/nu14071380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/17/2022] [Accepted: 03/24/2022] [Indexed: 02/05/2023] Open
Abstract
1,3-Dipalmitoyl-2-oleoylglycerol (POP) is a triacylglyceride found in oils from various natural sources, including palm kernels, sunflower seeds, and rice bran. In the current study, the neuroprotective effects and the specific mechanism of POP derived from rice bran oil were investigated for the first time using the middle cerebral artery occlusion/reperfusion (MCAO/R) model in rats. Orally administered POP at 1, 3, or 5 mg/kg (three times: 0.5 h before MCAO, after 1 h of MCAO, and after 1 h of reperfusion) markedly reduced the MCAO/R-induced infarct/edema volume and neurobehavioral deficits. Glutathione depletion and the oxidative degradation of lipids in the rat brain induced by MCAO/R were prevented by POP administration. The upregulation of phosphorylated p38 MAPKs, inflammatory factors (inducible nitric oxide synthase (i-NOS) and cyclooxygenase-2 (COX-2)), and pro-apoptotic proteins (B-cell lymphoma-2 (Bcl-2) associated X protein (Bax) and cleaved caspase-3) and the downregulation of the anti-apoptotic protein (Bcl-2) in the ischemic brain were significantly inhibited by POP administration. In addition, downregulation of phosphatidylinositol 3′-kinase (PI3K), phosphorylated protein kinase B (Akt), and phosphorylated cyclic (adenosine monophosphate) AMP responsive element-binding protein (CREB) expression in the ischemic brain was inhibited by POP administration. These results suggest that POP might exert neuroprotective effects by inhibition of p38 MAPK and activation of PI3K/Akt/CREB pathway, which is associated with anti-oxidant, anti-apoptotic, and anti-inflammatory action. From the above results, the present study provides evidence that POP might be effectively applied for the management of cerebral ischemia-related diseases.
Collapse
|
2
|
Wierońska JM, Cieślik P, Kalinowski L. Nitric Oxide-Dependent Pathways as Critical Factors in the Consequences and Recovery after Brain Ischemic Hypoxia. Biomolecules 2021; 11:biom11081097. [PMID: 34439764 PMCID: PMC8392725 DOI: 10.3390/biom11081097] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/17/2021] [Accepted: 07/20/2021] [Indexed: 12/14/2022] Open
Abstract
Brain ischemia is one of the leading causes of disability and mortality worldwide. Nitric oxide (NO•), a molecule that is involved in the regulation of proper blood flow, vasodilation, neuronal and glial activity constitutes the crucial factor that contributes to the development of pathological changes after stroke. One of the early consequences of a sudden interruption in the cerebral blood flow is the massive production of reactive oxygen and nitrogen species (ROS/RNS) in neurons due to NO• synthase uncoupling, which leads to neurotoxicity. Progression of apoptotic or necrotic neuronal damage activates reactive astrocytes and attracts microglia or lymphocytes to migrate to place of inflammation. Those inflammatory cells start to produce large amounts of inflammatory proteins, including pathological, inducible form of NOS (iNOS), which generates nitrosative stress that further contributes to brain tissue damage, forming vicious circle of detrimental processes in the late stage of ischemia. S-nitrosylation, hypoxia-inducible factor 1α (HIF-1α) and HIF-1α-dependent genes activated in reactive astrocytes play essential roles in this process. The review summarizes the roles of NO•-dependent pathways in the early and late aftermath of stroke and treatments based on the stimulation or inhibition of particular NO• synthases and the stabilization of HIF-1α activity.
Collapse
Affiliation(s)
- Joanna M Wierońska
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Paulina Cieślik
- Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna Street 12, 31-343 Kraków, Poland; (J.M.W.); (P.C.)
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Biobank Fahrenheit BBMRI.pl, Medical University of Gdansk, Debinki Street 7, 80-211 Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Debinki Street 7, 80-211 Gdansk, Poland
- BioTechMed Center/Department of Mechanics of Materials and Structures, Gdansk University of Technology, Narutowicza 11/12, 80-223 Gdansk, Poland
- Correspondence: ; Tel.: +48-58-349-1182
| |
Collapse
|
3
|
Chen HS, Chen X, Li WT, Shen JG. Targeting RNS/caveolin-1/MMP signaling cascades to protect against cerebral ischemia-reperfusion injuries: potential application for drug discovery. Acta Pharmacol Sin 2018; 39:669-682. [PMID: 29595191 PMCID: PMC5943912 DOI: 10.1038/aps.2018.27] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/26/2018] [Indexed: 02/07/2023]
Abstract
Reactive nitrogen species (RNS) play important roles in mediating cerebral ischemia-reperfusion injury. RNS activate multiple signaling pathways and participate in different cellular events in cerebral ischemia-reperfusion injury. Recent studies have indicated that caveolin-1 and matrix metalloproteinase (MMP) are important signaling molecules in the pathological process of ischemic brain injury. During cerebral ischemia-reperfusion, the production of nitric oxide (NO) and peroxynitrite (ONOO−), two representative RNS, down-regulates the expression of caveolin-1 (Cav-1) and, in turn, further activates nitric oxide synthase (NOS) to promote RNS generation. The increased RNS further induce MMP activation and mediate disruption of the blood-brain barrier (BBB), aggravating the brain damage in cerebral ischemia-reperfusion injury. Therefore, the feedback interaction among RNS/Cav-1/MMPs provides an amplified mechanism for aggravating ischemic brain damage during cerebral ischemia-reperfusion injury. Targeting the RNS/Cav-1/MMP pathway could be a promising therapeutic strategy for protecting against cerebral ischemia-reperfusion injury. In this mini-review article, we highlight the important role of the RNS/Cav-1/MMP signaling cascades in ischemic stroke injury and review the current progress of studies seeking therapeutic compounds targeting the RNS/Cav-1/MMP signaling cascades to attenuate cerebral ischemia-reperfusion injury. Several representative natural compounds, including calycosin-7-O-β-D-glucoside, baicalin, Momordica charantia polysaccharide (MCP), chlorogenic acid, lutein and lycopene, have shown potential for targeting the RNS/Cav-1/MMP signaling pathway to protect the brain in ischemic stroke. Therefore, the RNS/Cav-1/MMP pathway is an important therapeutic target in ischemic stroke treatment.
Collapse
|
4
|
Ding R, Lin C, Wei S, Zhang N, Tang L, Lin Y, Chen Z, Xie T, Chen X, Feng Y, Wu L. Therapeutic Benefits of Mesenchymal Stromal Cells in a Rat Model of Hemoglobin-Induced Hypertensive Intracerebral Hemorrhage. Mol Cells 2017; 40:133-142. [PMID: 28190323 PMCID: PMC5339504 DOI: 10.14348/molcells.2017.2251] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/29/2016] [Accepted: 01/09/2017] [Indexed: 12/31/2022] Open
Abstract
Previous studies have shown that bone marrow mesenchymal stromal cell (MSC) transplantation significantly improves the recovery of neurological function in a rat model of intracerebral hemorrhage. Potential repair mechanisms involve anti-inflammation, anti-apoptosis and angiogenesis. However, few studies have focused on the effects of MSCs on inducible nitric oxide synthase (iNOS) expression and subsequent peroxynitrite formation after hypertensive intracerebral hemorrhage (HICH). In this study, MSCs were transplanted intracerebrally into rats 6 hours after HICH. The modified neurological severity score and the modified limb placing test were used to measure behavioral outcomes. Blood-brain barrier disruption and neuronal loss were measured by zonula occludens-1 (ZO-1) and neuronal nucleus (NeuN) expression, respectively. Concomitant edema formation was evaluated by H&E staining and brain water content. The effect of MSCs treatment on neuroinflammation was analyzed by immunohistochemical analysis or polymerase chain reaction of CD68, Iba1, iNOS expression and subsequent peroxynitrite formation, and by an enzyme-linked immunosorbent assay of pro-inflammatory factors (IL-1β and TNF-α). The MSCs-treated HICH group showed better performance on behavioral scores and lower brain water content compared to controls. Moreover, the MSC injection increased NeuN and ZO-1 expression measured by immunochemistry/immunofluorescence. Furthermore, MSCs reduced not only levels of CD68, Iba1 and pro-inflammatory factors, but it also inhibited iNOS expression and peroxynitrite formation in perihematomal regions. The results suggest that intracerebral administration of MSCs accelerates neurological function recovery in HICH rats. This may result from the ability of MSCs to suppress inflammation, at least in part, by inhibiting iNOS expression and subsequent peroxynitrite formation.
Collapse
Affiliation(s)
- Rui Ding
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - Chunnan Lin
- Department of Neurosurgery, Maoming People’s Hospital, Maoming 525000, Guangdong,
China
| | - ShanShan Wei
- Department of Hematology, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - Naichong Zhang
- Department of Neurosurgery, Maoming People’s Hospital, Maoming 525000, Guangdong,
China
| | - Liangang Tang
- Department of Neurosurgery, Maoming People’s Hospital, Maoming 525000, Guangdong,
China
| | - Yumao Lin
- Department of Neurosurgery, Maoming People’s Hospital, Maoming 525000, Guangdong,
China
| | - Zhijun Chen
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - Teng Xie
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - XiaoWei Chen
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - Yu Feng
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| | - LiHua Wu
- Department of Neurosurgery, Jingmen No. 1 People’s Hospital, Jingmen 448000, Hubei,
China
| |
Collapse
|
5
|
Yu G, Liang Y, Huang Z, Jones DW, Pritchard KA, Zhang H. Inhibition of myeloperoxidase oxidant production by N-acetyl lysyltyrosylcysteine amide reduces brain damage in a murine model of stroke. J Neuroinflammation 2016; 13:119. [PMID: 27220420 PMCID: PMC4879722 DOI: 10.1186/s12974-016-0583-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 05/13/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Oxidative stress plays an important and causal role in the mechanisms by which ischemia/reperfusion (I/R) injury increases brain damage after stroke. Accordingly, reducing oxidative stress has been proposed as a therapeutic strategy for limiting damage in the brain after stroke. Myeloperoxidase (MPO) is a highly potent oxidative enzyme that is capable of inducing both oxidative and nitrosative stress in vivo. METHODS To determine if and the extent to which MPO-generated oxidants contribute to brain I/R injury, we treated mice subjected to middle cerebral artery occlusion (MCAO) with N-acetyl lysyltyrosylcysteine amide (KYC), a novel, specific and non-toxic inhibitor of MPO. Behavioral testing, ischemic damage, blood-brain-barrier disruption, apoptosis, neutrophils infiltration, microglia/macrophage activation, and MPO oxidation were analyzed within a 7-day period after MCAO. RESULTS Our studies show that KYC treatment significantly reduces neurological severity scores, infarct size, IgG extravasation, neutrophil infiltration, loss of neurons, apoptosis, and microglia/macrophage activation in the brains of MCAO mice. Immunofluorescence studies show that KYC treatment reduces the formation of chlorotyrosine (ClTyr), a fingerprint biomarker of MPO oxidation, nitrotyrosine (NO2Tyr), and 4-hydroxynonenal (4HNE) in MCAO mice. All oxidative products colocalized with MPO in the infarcted brains, suggesting that MPO-generated oxidants are involved in forming the oxidative products. CONCLUSIONS MPO-generated oxidants play detrimental roles in causing brain damage after stroke which is effectively reduced by KYC.
Collapse
Affiliation(s)
- Guoliang Yu
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Ye Liang
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Ziming Huang
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA.,Department of Breast Surgery, Maternal and Child Health Hospital of Hubei Province, 745 WuLuo Road, Hongshan District, Wuhan City, Hubei Province, 430070, China
| | - Deron W Jones
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Kirkwood A Pritchard
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Hao Zhang
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA.
| |
Collapse
|
6
|
Chen X, Bakillah A, Zhou L, Pan X, Hoepfner F, Jacob M, Jiang XC, Lazar J, Schlitt A, Hussain MM. Nitrated apolipoprotein AI/apolipoprotein AI ratio is increased in diabetic patients with coronary artery disease. Atherosclerosis 2016; 245:12-21. [PMID: 26687998 PMCID: PMC4738057 DOI: 10.1016/j.atherosclerosis.2015.11.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Recent studies have suggested that determination of HDL function may be more informative than its concentration in predicting its protective role in coronary artery disease (CAD). Apolipoprotein AI (apoAI), the major protein of HDL, is nitrosylated in vivo to nitrated apoAI (NT-apoAI) that might cause dysfunction. We hypothesized that NT-apoAI/apoAI ratio might be associated with diabetes mellitus (DM) in CAD patients. METHODS We measured plasma NT-apoAI and apoAI levels in 777 patients with coronary artery disease (CAD) by ELISA. Further, we measured plasma cholesterol efflux potential in subjects with similar apoAI but different NT-apoAI levels. RESULTS We found that median NT-apoAI/apoAI ratio was significantly higher in diabetes mellitus (DM) (n = 327) versus non-diabetic patients (n = 450). Further analysis indicated that DM, thiobarbituric acid-reactive substances and C-reactive protein levels were independent predictors of higher NT-apoAI/apoAI ratio. There was negative correlation between NT-apoAI/apoAI and use of anti-platelet and lipid lowering drugs. The cholesterol efflux capacity of plasma from 67 individuals with differing NT-apoAI but similar apoAI levels from macrophages in vitro was negatively correlated with NT-apoAI/apoAI ratio. CONCLUSIONS Higher NT-apoAI/apoAI ratio is significantly associated with DM in this relatively large German cohort with CAD and may contribute to associated complications by reducing cholesterol efflux capacity.
Collapse
Affiliation(s)
- Xueying Chen
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ahmed Bakillah
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Xiaoyue Pan
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | | | - Marrit Jacob
- Department of Medicine III, University Clinic Halle, Germany
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY 11209, USA
| | - Jason Lazar
- Division of Cardiovascular Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Axel Schlitt
- Department of Medicine III, University Clinic Halle, Germany; Paracelsus-Harz-Clinic Bad Suderode, Quedlinburg, Germany
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY 11209, USA; Department of Pediatrics, SUNY Downstate Medical Center, Brooklyn, NY, USA.
| |
Collapse
|
7
|
Iqbal S, Hayman EG, Hong C, Stokum JA, Kurland DB, Gerzanich V, Simard JM. Inducible nitric oxide synthase (NOS-2) in subarachnoid hemorrhage: Regulatory mechanisms and therapeutic implications. Brain Circ 2016; 2:8-19. [PMID: 27774520 PMCID: PMC5074544 DOI: 10.4103/2394-8108.178541] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) typically carries a poor prognosis. Growing evidence indicates that overabundant production of nitric oxide (NO) may be responsible for a large part of the secondary injury that follows SAH. Although SAH modulates the activity of all three isoforms of nitric oxide synthase (NOS), the inducible isoform, NOS-2, accounts for a majority of NO-mediated secondary injuries after SAH. Here, we review the indispensable physiological roles of NO that must be preserved, even while attempting to downmodulate the pathophysiologic effects of NO that are induced by SAH. We examine the effects of SAH on the function of the various NOS isoforms, with a particular focus on the pathological effects of NOS-2 and on the mechanisms responsible for its transcriptional upregulation. Finally, we review interventions to block NOS-2 upregulation or to counteract its effects, with an emphasis on the potential therapeutic strategies to improve outcomes in patients afflicted with SAH. There is still much to be learned regarding the apparently maladaptive response of NOS-2 and its harmful product NO in SAH. However, the available evidence points to crucial effects that, on balance, are adverse, making the NOS-2/NO/peroxynitrite axis an attractive therapeutic target in SAH.
Collapse
Affiliation(s)
- Sana Iqbal
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Erik G Hayman
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Caron Hong
- Department of Anesthesiology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Jesse A Stokum
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - David B Kurland
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - J Marc Simard
- Department of Neurosurgery, School of Medicine, University of Maryland, Baltimore, Maryland, USA; Department of Pathology, School of Medicine, University of Maryland, Baltimore, Maryland, USA; Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland, USA
| |
Collapse
|
8
|
Wei D, Xiong X, Zhao H. Tim-3 cell signaling and iNOS are involved in the protective effects of ischemic postconditioning against focal ischemia in rats. Metab Brain Dis 2015; 30:483-90. [PMID: 24771108 PMCID: PMC4213319 DOI: 10.1007/s11011-014-9543-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/04/2014] [Indexed: 11/28/2022]
Abstract
The protective effect of ischemic postconditioning (IPostC) against stroke has been well-established, and the underlying mechanisms are known to involve inhibited-inflammation and free radical production. Nevertheless, how IPostC affects protein expression of iNOS, nitrotyrosine, and COX-2 has not been characterized. In addition, the role of the galectin-9/Tim-3 cell signaling pathway--a novel inflammatory pathway--in IPostC has not been studied. We examined whether iNOS, nitrotyrosine, and COX-2, as well as galectin-9/Tim-3 are involved in the protective effects of IpostC in a rat focal ischemia model. Western blot and confocal immunofluoresent staining results indicate that IPostC significantly inhibited Tim-3 expression, and that galectin-9 expression was also inhibited. In addition, IPostC attenuated production of iNOS and nitrotyrosine, but not COX-2, suggesting that IPostC has distinct effects on these inflammatory factors. Furthermore, the inflammation inhibitor minocycline blocked Tim-3 and iNOS expression induced by stroke. Taken together, we show that the galectin-9/Tim-3 cell signaling pathway is involved in inflammation induced by stroke, and IPostC may reduce infarction by attenuating this novel pathway as well as the inflammatory factors iNOS and nitrotyrosine, but not COX-2.
Collapse
Affiliation(s)
- Dingtai Wei
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Fujian Medical University Ningde Hospital, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| | - Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
- Stroke Center, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
9
|
Wu JX, Zhu HW, Chen X, Wei JL, Zhang XF, Xu MY. Inducible nitric oxide synthase inhibition reverses pulmonary arterial dysfunction in lung transplantation. Inflamm Res 2014; 63:609-18. [PMID: 24760104 DOI: 10.1007/s00011-014-0733-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 03/30/2014] [Accepted: 03/31/2014] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) after lung transplantation remains a significant cause of morbidity and mortality. Lung IRI induces nitric oxide synthesis (iNOS) and reactive nitrogen species, decreasing nitric oxide bioavailability. We hypothesized that ischemia-induced iNOS intensifies with reperfusion and contributes to IRI-induced pulmonary arterial regulatory dysfunction, which may lead to early graft failure and cause pulmonary edema. The aim of this study was to determine whether ischemia-reperfusion alters inducible and endothelial nitric oxide synthase expression, potentially affecting pulmonary perfusion. We further evaluated the role of iNOS in post-transplantation pulmonary arterial disorder. METHODS We randomized 32 Sprague-Dawley rats into two groups. The control group was given a sham operation whilst the experimental group received orthotropic lung transplants with a modified three-cuff technique. Changes in lung iNOS, and endothelial nitric oxide synthase expression were measured after lung transplantation by enzyme-linked immunosorbent assay (ELISA). Vasoconstriction in response to exogenous phenylephrine and vasodilation in response to exogenous acetylcholine of pulmonary arterial rings were measured in vitro as a measure of vascular dysfunction. To elucidate the roles of iNOS in regulating vascular function, an iNOS activity inhibitor (N6-(1-iminoethyl)-L-lysine, L-NIL) was used to treat isolated arterial rings. In order to test whether iNOS inhibition has a therapeutic effect, we further used L-NIL to pre-treat transplanted lungs and then measured post-transplantation arterial responses. RESULTS Lung transplantation caused upregulation of iNOS expression. This was also accompanied by suppression of both vasoconstriction and vasodilation of arterial rings from transplanted lungs. Removal of endothelium did not interfere with the contraction of pulmonary arterial rings from transplanted lungs. In contrast, iNOS inhibition rescued the vasoconstriction response to exogenous phenylephrine of pulmonary arterial rings from transplanted lungs. In addition, lung transplantation led to suppression of PaO2/FiO2 ratio, increased intrapulmonary shunt (Q s/Q t), and increase of lung wet to dry ratio (W/D), malondialdehyde and myeloperoxidase levels, all of which were reversed upon iNOS inhibition. Furthermore, inhibition of iNOS significantly rescued vascular function and alleviated edema and inflammatory cell infiltration in the transplanted lung. CONCLUSIONS Our data suggest that lung transplantation causes upregulation of iNOS expression, and pulmonary vascular dysfunction. iNOS inhibition reverses the post-transplantational pulmonary vascular dysfunction.
Collapse
Affiliation(s)
- Jing-Xiang Wu
- Department of Anesthesiology, Shanghai Chest Hospital, Shanghai Jiaotong University, 241 West Huaihai Road, Shanghai, 200030, People's Republic of China
| | | | | | | | | | | |
Collapse
|
10
|
Onufriev MV, Stepanichev MY, Tishkina AO, Sidorova SV, Gulyaeva NV. Neuroimmune aspects of brain damage after focal ischemia. NEUROCHEM J+ 2014. [DOI: 10.1134/s1819712414010127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
11
|
Capacity of HSYA to inhibit nitrotyrosine formation induced by focal ischemic brain injury. Nitric Oxide 2013; 35:144-51. [PMID: 24126016 DOI: 10.1016/j.niox.2013.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2013] [Revised: 09/25/2013] [Accepted: 10/01/2013] [Indexed: 12/21/2022]
Abstract
Peroxynitrite-mediated protein tyrosine nitration represents a crucial pathogenic mechanism of stroke. Hydroxysafflor yellow A (HSYA) is the most important active component of the safflower plant. Here we assess the neuroprotective efficacy of HSYA and investigate the mechanism through anti-nitrative pathway. Rats were subjected to 60-min ischemia followed by reperfusion. HSYA (2.5-10mg/kg) was injected at 1h after ischemia onset. Other groups received HSYA (10mg/kg) treatment at 3-9h after onset. Infarct volume, brain edema, and neurological score were evaluated at 24h after ischemia. Nitrotyrosine and inducible NO synthase (iNOS) expression, as well as NO level (nitrate/nitrite) in ischemic cortex was examined within 24h after ischemia. The ability of HSYA to scavenge peroxynitrite was evaluated in vitro. Infarct volume was significantly decreased by HSYA (P<0.05), with a therapeutic window of 3h after ischemia at dose of 10mg/kg. HSYA treatment also reduced brain edema and improved neurological score (P<0.05). Nitrotyrosine formation was dose- and time-dependently inhibited by HSYA. The time window of HSYA in decreasing protein tyrosine nitration paralleled its action in infarct volume. HSYA also greatly reduced iNOS expression and NO content at 24h after ischemia, suggesting prevention of peroxynitrite generation from iNOS. In vitro, HSYA blocked authentic peroxynitrite-induced tyrosine nitration in bovine serum albumin and primary cortical neurons. Collectively, our results indicated that post-ischemic HSYA treatment attenuates brain ischemic injury which is at least partially due to reducing nitrotyrosine formation, possibly by the combined mechanism of its peroxynitrite scavenging ability and its reduction in iNOS production.
Collapse
|
12
|
Woitzik J, Weinzierl N, Schilling L. Early administration of a second-generation perfluorochemical decreases ischemic brain damage in a model of permanent middle cerebral artery occlusion in the rat. Neurol Res 2013; 27:509-15. [PMID: 15978177 DOI: 10.1179/016164105x15677] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Perfluorochemicals (PFCs) may exert a neuroprotective function in the early phase of ischemia by improving the oxygen supply to the endangered tissue. We have, therefore, investigated the effect of Oxycyte, a second-generation perfluorocarbon solution, on the extent of early ischemic brain damage in a model of permanent focal cerebral ischemia. METHODS Eight hours of permanent focal cerebral ischemia was induced in isoflurane anesthetized male Sprague-Dawley rats by unilateral middle cerebral artery (MCA) thread occlusion under the control of laser Doppler flowmetry (LDF). Animals were assigned to one of the following treatment groups: nO2-NaCl and hO2-NaCl-NaCl (0.9%, 1 ml/100 g i.v.) and nO2-Oxycyte and hO2-Oxycyte-Oxycyte (1 ml/100 g i.v.). The injection of NaCl or Oxycyte was performed immediately after MCA occlusion. After injection, breathing was changed to pure oxygen in groups hO2-NaCl and hO2-Oxycyte while animals in groups nO2-NaCl and nO2-Oxycyte were allowed to breathe air. The necrotic volume was calculated from serial coronal sections stained with silver-nitrate. In addition, nitrotyrosine production was studied by immunohistochemistry. RESULTS Upon MCA occlusion, animals showed a reduction of cerebral blood flow of approximately 80% of the LDF signal in all groups. Hemodynamic and metabolic parameters were not affected by the infusion of Oxycyte. The total infarct volume was reduced in hO2-Oxycyte animals [group nO2-NaCl: 341+/-31 mm3 (mean+/-SD), group hO2-NaCl: 351+/-33 mm3, group nO2-Oxycyte: 354+/-24 mm3, and group hO2-Oxycyte: 300+/-29 mm3, p < 0.05 versus all other groups]. Moreover, hO2-Oxycyte animals showed lesser intensity of nitrotyrosine staining when compared with hO2-NaCl animals. DISCUSSION These results suggest that Oxycyte administered immediately after the onset of vascular occlusion may exert neuroprotective effects in the early phase of brain ischemia.
Collapse
Affiliation(s)
- Johannes Woitzik
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany.
| | | | | |
Collapse
|
13
|
Goenka S, Peelukhana SV, Kim J, Stringer KF, Banerjee RK. Dependence of vascular damage on higher frequency components in the rat-tail model. INDUSTRIAL HEALTH 2013; 51:373-385. [PMID: 23518603 DOI: 10.2486/indhealth.2012-0060] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Hand-Arm Vibration Syndrome (HAVS) is caused by hand-transmitted vibration in industrial workers. Current ISO guidelines (ISO 5349) might underestimate vascular injury associated with range of vibration frequencies near resonance. A rat-tail model was used to investigate the effects of higher frequencies >100 Hz on early vascular damage. 13 Male Sprague-Dawley rats (250 ± 15 gm) were used. Rat-tails were vibrated at 125 Hz and 250 Hz (49 m/s(2)) for 1D, 5D and 10D; D=days (4 h/day). Structural damage of the ventral artery was quantified by vacuole count using Toluidine blue staining whereas biochemical changes were assessed by nitrotyrosine (NT) staining. The results were analyzed using one-way repeated measures mixed-model ANOVA at p<0.05 level of significance. The structural damage increased at 125 Hz causing significant number of vacuoles (40.62 ± 9.8) compared to control group (8.36 ± 2.49) and reduced at 250 Hz (12.33 ± 2.98) compared to control group (8.36 ± 2.49). However, the biochemical alterations (NT-signal) increased significantly for 125 Hz (143.35 ± 5.8 gray scale value, GSV) and for 250 Hz (155.8 ± 7.35 GSV) compared to the control group (101.7 ± 4.18 GSV). Our results demonstrate that vascular damage in the form of structural and bio chemical disruption is significant at 125 Hz and 250 Hz. Hence the current ISO guidelines might underestimate vascular damage at frequencies>100 Hz.
Collapse
Affiliation(s)
- Shilpi Goenka
- School of Energy, Environmental, Biological and Medical Engineering, Materials Engineering Program, University of Cincinnati, USA
| | | | | | | | | |
Collapse
|
14
|
Awooda HA. Down-regulation of Rho-kinases induce tolerance in Ischemic preconditioning model after transient cerebral ischemia/reperfusion in rats. Health (London) 2013. [DOI: 10.4236/health.2013.57a5002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Wei D, Ren C, Chen X, Zhao H. The chronic protective effects of limb remote preconditioning and the underlying mechanisms involved in inflammatory factors in rat stroke. PLoS One 2012; 7:e30892. [PMID: 22347410 PMCID: PMC3275571 DOI: 10.1371/journal.pone.0030892] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Accepted: 12/23/2011] [Indexed: 11/19/2022] Open
Abstract
We recently demonstrated that limb remote preconditioning (LRP) protects against focal ischemia measured 2 days post-stroke. Here, we studied whether LRP provides long-term protection and improves neurological function. We also investigated whether LRP transmits its protective signaling via the afferent nerve pathways from the preconditioned limb to the ischemic brain and whether inflammatory factors are involved in LRP, including the novel galectin-9/Tim-3 inflammatory cell signaling pathway, which induces cell death in lymphocytes. LRP in the left hind femoral artery was performed immediately before stroke. LRP reduced brain injury size both at 2 days and 60 days post-stroke and improved behavioral outcomes for up to 2 months. The sensory nerve inhibitors capsaicin and hexamethonium, a ganglion blocker, abolished the protective effects of LRP. In addition, LRP inhibited edema formation and blood-brain barrier (BBB) permeability measured 2 days post-stroke. Western blot and immunostaining analysis showed that LRP inhibited protein expression of both galectin-9 and T-cell immunoglobulin domain and mucin domain 3 (Tim-3), which were increased after stroke. In addition, LRP decreased iNOS and nitrotyrosine protein expression after stroke. In conclusion, LRP executes long-term protective effects against stroke and may block brain injury by inhibiting activities of the galectin-9/Tim-3 pathway, iNOS, and nitrotyrosine.
Collapse
Affiliation(s)
- Dingtai Wei
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- Department of Radiology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Radiology, Fujian Medical University Ningde Hospital, Fujian, China
| | - Chuancheng Ren
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- Shanghai No.5 Hospital, Fudan University, Shanghai, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Heng Zhao
- Department of Neurosurgery, Stanford University, Stanford, California, United States of America
- Stroke Center, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
16
|
Onufriev MV, Savos’kina IV, Kaimovskii IL, Lebedeva AV, Gusev EI, Guekht AB, Gulyaeva NV. Indices of nitrosative stress and immune response in the liquor of patients at the early stage of stroke. NEUROCHEM J+ 2011. [DOI: 10.1134/s1819712411010077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
|
18
|
Kunz A, Dirnagl U, Mergenthaler P. Acute pathophysiological processes after ischaemic and traumatic brain injury. Best Pract Res Clin Anaesthesiol 2010; 24:495-509. [DOI: 10.1016/j.bpa.2010.10.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Accepted: 10/11/2010] [Indexed: 12/23/2022]
|
19
|
Onufriev MV. Nitrosative stress in the brain: Autoantibodies to nitrotyrosine in the liquor as a potential marker. NEUROCHEM J+ 2010. [DOI: 10.1134/s1819712410030116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Inhibition of gp91(phox) contributes towards normobaric hyperoxia afforded neuroprotection in focal cerebral ischemia. Brain Res 2010; 1348:174-80. [PMID: 20547141 DOI: 10.1016/j.brainres.2010.05.082] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2010] [Revised: 05/25/2010] [Accepted: 05/26/2010] [Indexed: 02/02/2023]
Abstract
Oxygen therapy is a promising treatment strategy for ischemic stroke. One potential safety concern with oxygen therapy, however, is the possibility of increased generation of reactive oxygen species (ROS), which could exacerbate ischemic brain injury. Our previous study indicated that normobaric hyperoxia (NBO, 95% O(2) with 5% CO(2)) treatment during ischemia salvaged ischemic brain tissue and significantly reduced ROS generation in transient experimental stroke. In this follow-up study, we tested the hypothesis that suppression of NADPH oxidase is an important mechanism for NBO-induced reduction of ROS generation in focal cerebral ischemia. Male Sprague-Dawley rats were given NBO (95% O(2)) or normoxia (21% O(2)) during 90-min filament occlusion of the middle cerebral artery, followed by 22.5-hour reperfusion. NBO treatment increased the tissue oxygen partial pressure (pO(2)) level in the ischemic penumbra close to the pre-ischemic value, as measured by electronic paramagnetic resonance (EPR), and led to a 30.2% reduction in magnetic resonance imaging (MRI) apparent diffusion coefficients (ADC) lesion volume. Real time PCR and western blot analyses showed that the mRNA and protein expression of NADPH oxidase catalytic subunit gp91(phox) were upregulated in the ischemic brain, which was significantly inhibited by NBO. As a consequence of gp91(phox) inhibition, NBO treatment reduced NADPH oxidase activity in the ischemic brain. Our results suggest that NBO treatment given during ischemia reduces ROS generation via inhibiting NADPH oxidase, which may serve as an important mechanism underlying NBO's neuroprotection in acute ischemic stroke.
Collapse
|
21
|
Honoré JC, Kooli A, Hou X, Hamel D, Rivera JC, Picard É, Hardy P, Tremblay S, Varma DR, Jankov RP, Mancini JA, Balazy M, Chemtob S. Sustained hypercapnia induces cerebral microvascular degeneration in the immature brain through induction of nitrative stress. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1522-30. [DOI: 10.1152/ajpregu.00807.2009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Hypercapnia is regularly observed in chronic lung disease, such as bronchopulmonary dysplasia in preterm infants. Hypercapnia results in increased nitric oxide synthase activity and in vitro formation of nitrates. Neural vasculature of the immature subject is particularly sensitive to nitrative stress. We investigated whether exposure to clinically relevant sustained high CO2 causes microvascular degeneration in the newborn brain by inducing nitrative stress, and whether this microvascular degeneration has an impact on brain growth. Newborn rat pups were exposed to 10% CO2 as inspired gas (PaCO2 = 60–70 mmHg) starting within 24 h of birth until postnatal day 7 (P7). Brains were notably collected at different time points to measure vascular density, determine brain cortical nitrite/nitrate, and trans-arachidonic acids (TAAs; products of nitration) levels as effectors of vessel damage. Chronic exposure of rat pups to high CO2 (PaCO2 ≈ 65 mmHg) induced a 20% loss in cerebrovascular density at P3 and a 15% decrease in brain mass at P7; at P30, brain mass remained lower in CO2-exposed animals. Within 24 h of exposure to CO2, brain eNOS expression and production of nitrite/nitrate doubled, lipid nitration products (TAAs) increased, and protein nitration (3-nitrotyrosine immunoreactivity) was also coincidently augmented on brain microvessels (lectin positive). Intracerebroventricular injection of TAAs (10 μM) replicated cerebrovascular degeneration. Treatment of rat pups with NOS inhibitor (l-Nω-nitroarginine methyl ester) or a peroxynitrite decomposition catalyst (FeTPPS) prevented hypercapnia-induced microvascular degeneration and preserved brain mass. Cytotoxic effects of high CO2 were reproduced in vitro /ex vivo on cultured endothelial cells and sprouting microvessels. In summary, hypercapnia at values frequently observed in preterm infants with chronic lung disease results in increased nitrative stress, which leads to cerebral cortical microvascular degeneration and curtails brain growth.
Collapse
Affiliation(s)
- Jean-Claude Honoré
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
| | - Amna Kooli
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Quebec, Canada
| | - Xin Hou
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
| | - David Hamel
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
| | - José Carlos Rivera
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
| | - Émilie Picard
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
| | - Pierre Hardy
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
| | - Sophie Tremblay
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
| | - Daya R. Varma
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Quebec, Canada
| | - Robert P. Jankov
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada; and
| | - Joseph A. Mancini
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
| | - Michael Balazy
- Department of Pathology, New York Medical College, New York, New York
| | - Sylvain Chemtob
- Department of Pediatrics, Research Center-Centre Hospitalier Universitaire Ste-Justine, Montréal, Quebec, Canada
- Department of Pharmacology, Université de Montréal, Quebec, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
22
|
Toda N, Ayajiki K, Okamura T. Cerebral blood flow regulation by nitric oxide: recent advances. Pharmacol Rev 2009; 61:62-97. [PMID: 19293146 DOI: 10.1124/pr.108.000547] [Citation(s) in RCA: 283] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
Nitric oxide (NO) is undoubtedly quite an important intercellular messenger in cerebral and peripheral hemodynamics. This molecule, formed by constitutive isomers of NO synthase, endothelial nitric-oxide synthase, and neuronal nitric-oxide synthase, plays pivotal roles in the regulation of cerebral blood flow and cell viability and in the protection of nerve cells or fibers against pathogenic factors associated with cerebral ischemia, trauma, and hemorrhage. Cerebral blood flow is increased and cerebral vascular resistance is decreased by NO derived from endothelial cells, autonomic nitrergic nerves, or brain neurons under resting and stimulated conditions. Somatosensory stimulation also evokes cerebral vasodilatation mediated by neurogenic NO. Oxygen and carbon dioxide alter cerebral blood flow and vascular tone mainly via constitutively formed NO. Endothelial dysfunction impairs cerebral hemodynamics by reducing the bioavailability of NO and increasing the production of reactive oxygen species (ROS). The NO-ROS interaction is an important issue in discussing blood flow and cell viability in the brain. Recent studies on brain circulation provide quite useful information concerning the physiological roles of NO produced by constitutive isoforms of nitric-oxide synthase and how NO may promote cerebral pathogenesis under certain conditions, including cerebral ischemia/stroke, cerebral vasospasm after subarachnoid hemorrhage, and brain injury. This information would contribute to better understanding of cerebral hemodynamic regulation and its dysfunction and to development of novel therapeutic measures to treat diseases of the central nervous system.
Collapse
Affiliation(s)
- Noboru Toda
- Shiga University of Medical Science, Toyama Institute for Cardiovascular Pharmacology Research, 7-13, 1-Chome, Azuchi-machi, Chuo-ku, Osaka 541-0052, Japan.
| | | | | |
Collapse
|
23
|
Takizawa S, Fukuyama N, Hirabayashi H, Nakazawa H, Shinohara Y. Reperfusion enhances nitrotyrosine formation in rat focal cerebral ischemia. J Stroke Cerebrovasc Dis 2007; 12:196-200. [PMID: 17903926 DOI: 10.1016/s1052-3057(03)00074-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2003] [Accepted: 05/02/2003] [Indexed: 11/22/2022] Open
Abstract
The widespread benefit of thrombolysis has been emphasized, but relatively little is known about reperfusion injury. The purpose of this study is to evaluate the difference in nitrotyrosine formation and infarct volume between permanent and transient focal ischemia in rats. Permanent (n = 14) or transient (n = 12) focal ischemia was induced by permanent or 2-hour occlusion of the middle cerebral artery, respectively, with the permanent ligation of the bilateral common carotid arteries in Sprague-Dawley rats. In both models all animals were killed 24 hours after the start of occlusion. The ratio of nitrotyrosine in the peri-infarct and core-of-infarct regions in transient focal ischemia was significantly higher than in permanent focal ischemia (P < .01). Infarct volume in the cortex, but not caudoputamen or whole brain, was significantly larger in transient ischemia than in permanent ischemia (P < .05), with a significant expansion of brain swelling. These results may reflect the higher production of superoxide and nitric oxide owing to reperfusion, and suggest the need to administer neuroprotective drugs such as anti-oxidants as well as thrombolytic agents in the treatment of acute ischemic cerebral damage.
Collapse
Affiliation(s)
- Shunya Takizawa
- Department of Neurology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
24
|
Hsiao G, Lee JJ, Chen YC, Lin JH, Shen MY, Lin KH, Chou DS, Sheu JR. Neuroprotective effects of PMC, a potent α-tocopherol derivative, in brain ischemia-reperfusion: Reduced neutrophil activation and anti-oxidant actions. Biochem Pharmacol 2007; 73:682-93. [PMID: 17157267 DOI: 10.1016/j.bcp.2006.11.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2006] [Revised: 10/18/2006] [Accepted: 11/13/2006] [Indexed: 10/23/2022]
Abstract
2,2,5,7,8-Pentamethyl-6-hydroxychromane (PMC) is the most potent analogue of alpha-tocopherol for anti-oxidation. It is more hydrophilic than other alpha-tocopherol derivatives and has potent free radical-scavenging activity. In the present study, PMC significantly attenuated middle cerebral artery occlusion (MCAO)-induced focal cerebral ischemia in rats. Administration of PMC at 20mg/kg, showed marked reductions in infarct size compared with that of control rats. MCAO-induced focal cerebral ischemia was associated with increases in HIF-1alpha, active caspase-3, iNOS, and nitrotyrosine expressions in ischemic regions. These expressions were markedly inhibited by treatment with PMC (20mg/kg). In addition, PMC (4-12 microM) inhibited respiratory bursts in human neutrophils stimulated by fMLP (800 nM) and PMA (320 nM). Furthermore, PMC (6, 12, and 60 microM) also significantly inhibited neutrophil migration stimulated by leukotriene B(4) (160 nM). An electron spin resonance (ESR) method was conducted on the scavenging activity of PMC on the free radicals formed. PMC (12 microM) greatly reduced the ESR signal intensities of superoxide anion, hydroxyl radical, and methyl radical formation. In conclusion, we demonstrate a potent neuroprotective effect of PMC on MCAO-induced focal cerebral ischemia in vivo. This effect may be mediated, at least in part, by inhibition of free radical formation, followed by inhibition of HIF-1alpha activation, apoptosis formation (active caspase-3), neutrophil activation, and inflammatory responses (i.e., iNOS and nitrotyrosine expressions), resulting in a reduction in the infarct volume in ischemia-reperfusion brain injury. Thus, PMC treatment may represent a novel approach to lowering the risk or improving function in ischemia-reperfusion brain injury-related disorders.
Collapse
Affiliation(s)
- George Hsiao
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The discovery that mammalian cells have the ability to synthesize the free radical nitric oxide (NO) has stimulated an extraordinary impetus for scientific research in all the fields of biology and medicine. Since its early description as an endothelial-derived relaxing factor, NO has emerged as a fundamental signaling device regulating virtually every critical cellular function, as well as a potent mediator of cellular damage in a wide range of conditions. Recent evidence indicates that most of the cytotoxicity attributed to NO is rather due to peroxynitrite, produced from the diffusion-controlled reaction between NO and another free radical, the superoxide anion. Peroxynitrite interacts with lipids, DNA, and proteins via direct oxidative reactions or via indirect, radical-mediated mechanisms. These reactions trigger cellular responses ranging from subtle modulations of cell signaling to overwhelming oxidative injury, committing cells to necrosis or apoptosis. In vivo, peroxynitrite generation represents a crucial pathogenic mechanism in conditions such as stroke, myocardial infarction, chronic heart failure, diabetes, circulatory shock, chronic inflammatory diseases, cancer, and neurodegenerative disorders. Hence, novel pharmacological strategies aimed at removing peroxynitrite might represent powerful therapeutic tools in the future. Evidence supporting these novel roles of NO and peroxynitrite is presented in detail in this review.
Collapse
Affiliation(s)
- Pál Pacher
- Section on Oxidative Stress Tissue Injury, Laboratory of Physiologic Studies, National Institutes of Health, National Institute of Alcohol Abuse and Alcoholism, Bethesda, Maryland, USA.
| | | | | |
Collapse
|
26
|
Hong H, Zeng JS, Kreulen DL, Kaufman DI, Chen AF. Atorvastatin protects against cerebral infarction via inhibition of NADPH oxidase-derived superoxide in ischemic stroke. Am J Physiol Heart Circ Physiol 2006; 291:H2210-5. [PMID: 16766636 DOI: 10.1152/ajpheart.01270.2005] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Statins have recently been shown to exert neuronal protection in ischemic stroke. Reactive oxygen species, specifically superoxide formed during the early phase of reperfusion, augment neuronal injury. NADPH oxidase is a key enzyme for superoxide production. The present study tested the hypothesis that atorvastatin protects against cerebral infarction via inhibition of NADPH oxidase-derived superoxide in transient focal ischemia. Transient focal ischemia was created in halothane-anesthetized adult male Sprague-Dawley rats (250-300 g) by middle cerebral artery occlusion (MCAO). Atorvastatin (Lipitor, 10 mg/kg sc) was administered three times before MCAO. Infarct volume was measured by triphenyltetrazolium chloride staining. NADPH oxidase enzymatic activity and superoxide levels were quantified in the ischemic core and penumbral regions by lucigenin (5 microM)-enhanced chemiluminescence. Expression of NADPH oxidase membrane subunit gp91(phox) and membrane-translocated subunit p47(phox) and small GTPase Rac-1 was analyzed by Western blot. NADPH oxidase activity and superoxide levels increased after reperfusion and peaked within 2 h of reperfusion in the penumbra, but not in the ischemic core, in MCAO rats. Atorvastatin pretreatment prevented these increases, blunted expression of membrane subunit gp91(phox), and prevented translocation of cytoplasmic subunit p47(phox) to the membrane in the penumbra 2 h after reperfusion. Consequently, cerebral infarct volume was significantly reduced in atorvastatin-treated compared with nontreated MCAO rats 24 h after reperfusion. These results indicate that atorvastatin protects against cerebral infarction via inhibition of NADPH oxidase-derived superoxide in transient focal ischemia.
Collapse
Affiliation(s)
- Hua Hong
- Depts. of Pharmacology and Neurology, B403 Life Sciences Bldg., Michigan State Univ., East Lansing, MI 48824-1317, USA
| | | | | | | | | |
Collapse
|
27
|
Leduc M, Kermorvant-Duchemin E, Checchin D, Sennlaub F, Sirinyan M, Kooli A, Lachapelle P, Chemtob S. Hypercapnia- and trans-arachidonic acid-induced retinal microvascular degeneration: implications in the genesis of retinopathy of prematurity. Semin Perinatol 2006; 30:129-38. [PMID: 16813971 DOI: 10.1053/j.semperi.2006.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
High oxygen tension is a major factor in the genesis of retinopathy of prematurity (ROP). However, clinical and experimental evidence suggests a significant role for high carbon dioxide (CO(2)) tension as well. Along these lines, although ischemia is often considered to be synonymous with an oxygen deficit, it is also associated with a concomitant local elevation of CO(2) that can lead to impaired developmental and ischemic neovascularization. The mechanisms by which hypercapnia induces retinal microvascular degeneration, a critical step which precedes the subsequent proliferative preretinal neovascularization, are not known. Nitrative stress has an important role in microvascular degeneration leading to ischemia in conditions such as ROP. Hypercapnia is a facilitator of nitration in vitro. We hereby present evidence that prolonged exposure to CO(2) impairs developmental retinal neovascularization through a mechanism involving increased endothelial nitric oxide synthase and induction of a nitrative stress; effects of hypercapnia are independent of its hyperaemic effects. Moreover, we demonstrate that an in vivo nitrative stress associated with retinal vasoobliteration results in nitration of arachidonic acids into trans-arachidonic acids (TAAs), which can act as mediators of nitrative stress by causing microvascular degeneration by inducing expression of the antiangiogenic factor thrombospondin-1. These recent findings establish a previously unexplored means by which hypercapnia hinders efficient neovascularization and provide new insight into the molecular mechanisms of nitrative stress on microvascular injury involving TAA, and suggest new therapeutic avenues in the management of nitrative stress disorders such as in ischemic retinopathies (of prematurity and of diabetes) and encephalopathies.
Collapse
Affiliation(s)
- Martin Leduc
- Department of Pediatrics, Ophthalmology and Pharmacology, Research Center, Hôpital Ste-Justine, 3175 Ch. Côte-Sainte-Catherine, Montréal, Québec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Park EM, Cho S, Frys KA, Glickstein SB, Zhou P, Anrather J, Ross ME, Iadecola C. Inducible nitric oxide synthase contributes to gender differences in ischemic brain injury. J Cereb Blood Flow Metab 2006; 26:392-401. [PMID: 16049426 DOI: 10.1038/sj.jcbfm.9600194] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Estrogens have antiinflammatory actions and protect the brain from ischemic injury. Cerebral ischemia is accompanied by an inflammatory reaction that contributes to the tissue damage, an effect mediated in part by toxic amounts of nitric oxide (NO) produced by the inducible isoform of NO synthase (iNOS). Therefore, estrogens may protect the female brain by modulating postischemic iNOS expression. To test this hypothesis, we studied whether iNOS plays a role in the mechanisms of the reduced susceptibility to ischemic injury observed in female mice. The middle cerebral artery was occluded for 20 mins using an intraluminal filament in C57Bl/6 mice, and infarct volume was assessed 3 days later in cresyl violet-stained sections. Infarcts were 53% smaller in female mice than in males (P < 0.05), a reduction abolished by ovariectomy (OVX) and reinstated by estrogen replacement. In normal female mice, postischemic iNOS mRNA was lower than in males (P < 0.05). Ovariectomy increased iNOS mRNA after ischemia and estrogen replacement blocked this effect. Furthermore, the iNOS inhibitor aminoguanidine reduced infarct volume in male, but not in female, mice. Similarly, male iNOS-null mice had smaller infarcts than wild-type mice, but female iNOS nulls were not protected. Ovariectomy and OVX with estrogen replacement did not affect infarct volume in iNOS-null female mice. The findings suggest that the neuroprotection conferred by estrogens is, in part, related to attenuation of iNOS expression. Such attenuation could result from the potent antiinflammatory effects of estrogens that downregulate iNOS expression via transcriptional or posttranscriptional mechanisms.
Collapse
Affiliation(s)
- Eun-Mi Park
- Division of Neurobiology, Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Lau D, Baldus S. Myeloperoxidase and its contributory role in inflammatory vascular disease. Pharmacol Ther 2006; 111:16-26. [PMID: 16476484 DOI: 10.1016/j.pharmthera.2005.06.023] [Citation(s) in RCA: 200] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2005] [Accepted: 06/28/2005] [Indexed: 11/21/2022]
Abstract
Myeloperoxidase (MPO), a heme protein abundantly expressed in polymorphonuclear neutrophils (PMN), has long been viewed to function primarily as a bactericidal enzyme centrally linked to innate host defense. Recent observations now extend this perspective and suggest that MPO is profoundly involved in the regulation of cellular homeostasis and may play a central role in initiation and propagation of acute and chronic vascular inflammatory disease. For example, low levels of MPO-derived hypochlorous acid (HOCl) interfere with intracellular signaling events, MPO-dependent oxidation of lipoproteins modulates their affinity to macrophages and the vessel wall, MPO-mediated depletion of endothelial-derived nitric oxide (NO) impairs endothelium-dependent vasodilatation, and nitrotyrosine (NO(2)Tyr) formation by MPO sequestered into the vessel wall may affect matrix protein structure and function. Future studies are needed to further elucidate the significance of MPO in the development of acute and chronic vascular disease and to evaluate MPO as a potential target for treatment.
Collapse
Affiliation(s)
- Denise Lau
- Department of Cardiology, University Hospital Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | | |
Collapse
|
30
|
Checchin D, Sennlaub F, Sirinyan M, Brault S, Zhu T, Kermorvant-Duchemin E, Hardy P, Balazy M, Chemtob S. Hypercapnia prevents neovascularization via nitrative stress. Free Radic Biol Med 2006; 40:543-53. [PMID: 16443170 DOI: 10.1016/j.freeradbiomed.2005.09.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 09/01/2005] [Accepted: 09/06/2005] [Indexed: 02/02/2023]
Abstract
Neovascularization after an ischemic insult is a beneficial attempt to salvage the injured tissue. Yet, despite the production of angiogenic factors within ischemic tissues, compensatory growth of new vessels fails to provide adequate vascularization. Thus, we hypothesized that local factors counter efficient revascularization. Whereas ischemia is often considered to be synonymous with an oxygen deficit, it is also associated with a concomitant local elevation of carbon dioxide (CO2). Although studies suggest that hypercapnia impacts tissue neovascularization, its significance relative to the abundantly described effects of hypoxia and its underlying mechanisms have yet to be elucidated. Therefore, we investigated the effects of hypercapnia on blood vessel growth in models of developmental and ischemic neovascularization. Acute and prolonged CO2 exposure inhibited developmental neovascularization of the rodent retina, as well as revascularization of the ischemic retina. Hypercapnia induced early increases in endothelial nitric oxide synthase and nitrative stress, associated with astrocyte impairment and endothelial cell death, as well as downregulation of the proangiogenic prostaglandin E2 receptor EP3. These results establish a previously unexplored means by which hypercapnia hinders efficient neovascularization, a mechanism that may contribute to ischemic tissue injury.
Collapse
Affiliation(s)
- Daniella Checchin
- Department of Pediatrics, Department of Ophthalmology, and Department of Pharmacology, Research Center, Hôpital Ste. Justine, Montreal, QC, Canada H3T 1C5
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wang YH, Wang WY, Chang CC, Liou KT, Sung YJ, Liao JF, Chen CF, Chang S, Hou YC, Chou YC, Shen YC. Taxifolin ameliorates cerebral ischemia-reperfusion injury in rats through its anti-oxidative effect and modulation of NF-kappa B activation. J Biomed Sci 2005; 13:127-41. [PMID: 16283433 DOI: 10.1007/s11373-005-9031-0] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2005] [Accepted: 09/08/2005] [Indexed: 12/17/2022] Open
Abstract
Infarction in adult rat brain was induced by middle cerebral arterial occlusion (MCAO) followed by reperfusion to examine whether taxifolin could reduce cerebral ischemic reperfusion (CI/R) injury. Taxifolin administration (0.1 and 1.0 microg/kg, i.v.) 60 min after MCAO ameliorated infarction (by 42%+/-7% and 62%+/-6%, respectively), which was accompanied by a dramatic reduction in malondialdehyde and nitrotyrosine adduct formation, two markers for oxidative tissue damage. Overproduction of reactive oxygen species (ROS) and nitric oxide (NO) via oxidative enzymes (e.g., COX-2 and iNOS) was responsible for this oxidative damage. Taxifolin inhibited leukocyte infiltration, and COX-2 and iNOS expressions in CI/R-injured brain. Taxifolin also prevented Mac-1 and ICAM-1 expression, two key counter-receptors involved in firm adhesion/transmigration of leukocytes to the endothelium, which partially accounted for the limited leukocyte infiltration. ROS, generated by leukocytes and microglial cells, activated nuclear factor-kappa B (NF-kappaB) that in turn signaled up-regulation of inflammatory proteins. NF-kappaB activity in CI/R was enhanced 2.5-fold over that of sham group and was inhibited by taxifolin. Production of both ROS and NO by leukocytes and microglial cells was significantly antagonized by taxifolin. These data suggest that amelioration of CI/R injury by taxifolin may be attributed to its anti-oxidative effect, which in turn modulates NF-kappaB activation that mediates CI/R injury.
Collapse
Affiliation(s)
- Yea-Hwey Wang
- Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ozben T, Balkan E, Balkan S, Serteser M, Gümüslü S. Effects of MK-801 on nitrite and cGMP levels during focal cerebral ischemia in rats. Nitric Oxide 2005; 13:210-5. [PMID: 16122952 DOI: 10.1016/j.niox.2005.07.005] [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] [Received: 04/14/2005] [Revised: 06/08/2005] [Accepted: 07/08/2005] [Indexed: 11/28/2022]
Abstract
Glutamate is a major excitatory neurotransmitter in the mammalian central nervous system and initiates the events leading to ischemic brain damage. Glutamate receptor antagonists are being used to reduce neuronal damage observed after hypoxia and ischemia. The glutamate receptor antagonist, (+)-5-methyl-10,11-dihydro-5H-dibenzo-(a,d)-cyclohepten-5,10-imine maleate (MK-801) crosses the blood-brain barrier readily and produces a non-competitive use-dependent blockade of the N-methyl-D-aspartate subtype of glutamate receptor. The aim of this study was to investigate effects of MK-801 administered before and just after the onset of ischemia in rats on nitrite and cyclic guanosine monophosphate (cGMP) levels. Focal cerebral ischemia in rats was produced by permanent occlusion of right middle cerebral artery (MCAO). Nitrite and cGMP levels were measured in both cortex and cerebellum at 0, 10, and 60 min following MCAO. The same parameters were measured in rats treated with MK-801 (0.5 mg/kg, i.p.) 30 min before or just after MCAO. Ipsilateral cortical nitrite levels were increased relative to contralateral cortex after MCAO. No significant changes were observed in cerebellum. The cGMP concentrations in both sides of the cortex and cerebellum were increased at 10 and 60 min compared with 0 min values. cGMP level in the ipsilateral cortex was higher than contralateral cortex, whereas the opposite was found for the cerebellum. MK-801 treatment before or just after MCAO decreased significantly nitrite and cGMP production. Our data indicate that MK-801 treatment before or just after focal ischemia prevents the increase in NO and cGMP production.
Collapse
Affiliation(s)
- Tomris Ozben
- Department of Biochemistry, Akdeniz University, Medical Faculty, 07070 Antalya, Turkey.
| | | | | | | | | |
Collapse
|
33
|
Margaill I, Plotkine M, Lerouet D. Antioxidant strategies in the treatment of stroke. Free Radic Biol Med 2005; 39:429-43. [PMID: 16043015 DOI: 10.1016/j.freeradbiomed.2005.05.003] [Citation(s) in RCA: 305] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Revised: 04/19/2005] [Accepted: 05/04/2005] [Indexed: 11/22/2022]
Abstract
Excessive production of free radicals is known to lead to cell injury in a variety of diseases, such as cerebral ischemia. In this review, we describe some of the numerous studies that have examined this oxidative stress and the efficiency of antioxidant strategies in focal cerebral ischemia. Besides using genetically modified mice, these strategies can be divided into three groups: (1) inhibition of free radical production, (2) scavenging of free radicals, and (3) increase of free radical degradation by using agents mimicking the enzymatic activity of endogenous antioxidants. Finally, the clinical trials that have tested or are currently testing the efficiency of antioxidants in patients suffering from stroke are reviewed. The results presented here lead us to consider that antioxidants are very promising drugs for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Isabelle Margaill
- Faculté des Sciences Pharmaceutiques et Biologiques, Laboratoire de Pharmacologie (UPRES EA 2510), Université René Descartes, 4 avenue de l'Observatoire, 75006 Paris, France.
| | | | | |
Collapse
|
34
|
Acarin L, Peluffo H, Barbeito L, Castellano B, González B. Astroglial nitration after postnatal excitotoxic damage: correlation with nitric oxide sources, cytoskeletal, apoptotic and antioxidant proteins. J Neurotrauma 2005; 22:189-200. [PMID: 15665612 DOI: 10.1089/neu.2005.22.189] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Oxygen free radicals and nitric oxide (NO) participate in the pathogenesis of acute central nervous system (CNS) injury by forming peroxynitrite, which promotes oxidative damage and tyrosine nitration. Neuronal nitration is associated with cell death, but little is known of the characteristics and cell fate of nitrated astrocytes. In this study, we have used a postnatal excitotoxic lesion model (intracortical NMDA injection) and our aims were (i) to evaluate the temporal and spatial pattern of astroglial nitration in correlation with the neuropathological process and the sources of NO; and (ii) to establish, if any, the correlation among astrocyte nitration and other events such as expression of cytoskeletal proteins, antioxidant enzymes, and cell death markers to cope with nitration and/or undergo cell death. Our results show that after postnatal excitotoxic damage two distinct waves of nitration were observed in relation to astrocytes. At 24 h post-lesion, early-nitrated astrocytes were found within the neurodegenerating area, coinciding with the time of maximal cell death. These early-nitrated astrocytes are highly ramified protoplasmic cells, showing diffuse glial fibrillary acidic protein (GFAP) content and expressing inducible NOS. At later time-points, when astrogliosis is morphologically evident, nitrated hypertrophied reactive astrocytes are observed in the penumbra and the neurodegenerated area, displaying increased expression of GFAP and vimentin cytoskeletal proteins and of metallothionein I-II and Cu/Zn superoxide dismutase antioxidant proteins. Moreover, despite revealing activated caspase-3, they do not show TUNEL labeling. In summary, we show that nitrated astrocytes in vivo constitute a subpopulation of highly reactive astrocytes which display high resistance towards oxidative stress induced cell death.
Collapse
Affiliation(s)
- Laia Acarin
- Unit of Histology, School of Medicine, Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autonoma Barcelona, Bellaterra 08193, Spain.
| | | | | | | | | |
Collapse
|
35
|
Luthra A, Gupta N, Kaufman PL, Weinreb RN, Yücel YH. Oxidative injury by peroxynitrite in neural and vascular tissue of the lateral geniculate nucleus in experimental glaucoma. Exp Eye Res 2005; 80:43-9. [PMID: 15652525 DOI: 10.1016/j.exer.2004.08.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2004] [Accepted: 08/14/2004] [Indexed: 11/18/2022]
Abstract
In glaucoma, recent studies show that neural degeneration extends beyond the retinal ganglion cells to include target neurons in the lateral geniculate nucleus of the brain. The pathobiology of LGN degeneration in glaucoma is as yet unknown. We investigated whether peroxynitrite-mediated oxidative stress plays a role in glaucomatous degeneration of the LGN. Nitrotyrosine (NT), a marker for peroxynitrite-mediated oxidative injury, was studied in right LGN sections from monkeys with experimental unilateral glaucoma in the right eye and from normal controls. Immunoreactivity for NT was analyzed using bright-field microscopy. The density of NT profiles localized in neural tissue was determined for LGN layers (2,3,5) connected to the glaucoma eye and LGN layers (1,4,6) connected to the non-glaucoma eye. Density was calculated for each LGN layer by dividing the number of NT profiles by the cross-sectional area of each LGN layer. Blood vessels in each LGN were examined for NT formation. NT formation was detected in LGN layers of all monkeys with glaucoma. Quantitative analysis revealed that compared to controls, the density of NT profiles was increased in monkeys with glaucoma in LGN layers connected to glaucoma and non-glaucoma eyes. The mean density of NT profiles (+/-SEM) in neural tissue was significantly increased in glaucoma LGN layers compared to those of controls (2.30+/-0.56 vs. 0.29+/-0.12; P=0.016). Nitrotyrosine was readily apparent in LGN blood vessel endothelium in glaucoma, and not detected in blood vessels of control LGNs. The presence of NT in neural and vascular tissue of the glaucomatous LGN implicates peroxynitrite-mediated oxidative cell injury in the pathobiology of central neural degeneration in glaucoma.
Collapse
Affiliation(s)
- Anchla Luthra
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, St. Michael's Hospital, University of Toronto, Toronto, ON M5B 1W8, Canada
| | | | | | | | | |
Collapse
|
36
|
Lee JC, Cho GS, Kim HJ, Lim JH, Oh YK, Nam W, Chung JH, Kim WK. Accelerated cerebral ischemic injury by activated macrophages/microglia after lipopolysaccharide microinjection into rat corpus callosum. Glia 2005; 50:168-81. [PMID: 15702482 DOI: 10.1002/glia.20164] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In cerebral ischemic insults, activated inflammatory cells such as microglia and macrophages may be implicated in the pattern and degree of ischemic injury by producing various bioactive mediators. In the present study, we provide the evidence that activated microglia/macrophages accelerate cerebral ischemic injury by overexpression of inducible nitric oxide synthase (iNOS). To activate microglia/macrophages, a potent inflammation inducer lipopolysaccharide (LPS, 5 microg/5 microl) was microinjected into rat corpus callosum. Isolectin B4-positive microglia/macrophages were abundantly observed in ipsilateral hemisphere at 1 day after LPS injection. RT-PCR showed that LPS injection induced iNOS mRNA expression mostly in microglia/macrophages, peaking in intensity at 15 h after LPS injection. While ischemic injury was little evoked in control rats by 2-h middle cerebral artery occlusion (MCAO) followed by 3-h reperfusion, it was markedly increased in rats pre-injected with LPS 1 day before MCAO. However, no significant difference between control and LPS-pretreated groups was observed after 24-h reperfusion. The increased ischemic injury in LPS-treated rats was well correlated with iNOS level expressed over 3 orders of magnitude than in LPS-untreated rats. Immunohistochemical studies showed that iNOS- and nitrotyrosine (a peroxynitrite marker)-positive cells were prominent throughout the infarct area. NOS inhibitors aminoguanidine or N(G)-nitro-L-arginine, simultaneously injected with LPS, reduced the iNOS immunoreactivity and infarct volume, especially in penumbra regions. Total glutathione levels in ischemic regions were decreased more in LPS pre-injected rats than in control ones. Further defining the role of NO in cerebral ischemic insults would provide the rationale for new therapeutic strategies based on modulation of microglial and macrophageal NO production in the brain.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Pharmacology, College of Medicine, Laboratory of Neurodegenerative Diseases, Ewha Institute of Neuroscience, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Patel P, Qi WN, Allen DM, Chen LE, Seaber AV, Stamler JS, Urbaniak JR. Inhibition of iNOS with 1400W improves contractile function and alters nos gene and protein expression in reperfused skeletal muscle. Microsurgery 2004; 24:324-31. [PMID: 15274192 DOI: 10.1002/micr.20029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study examined the effects of 1400W, an inhibitor of inducible nitric oxide (iNOS), on contractile function and iNOS expression in reperfused skeletal muscle. The right extensor digitorum longus (EDL) muscle of 104 rats underwent a sham operation or 3-h ischemia followed by 3-h or 24-h reperfusion (I/R). Rats received 3 mg/kg 1400W, 10 mg/kg 1400W, or water subcutaneously. Results showed that EDL contractile function in both 1400W-treated groups significantly outperformed the controls at 24-h but not at 3-h reperfusion. Although iNOS expression increased in all three I/R groups during reperfusion, a significantly smaller increase was found in 1400W-treated muscles after 3-h reperfusion, and more dramatically so after 24-h reperfusion. Our results indicate that inhibition of iNOS preserved the contractile function in reperfused skeletal muscle, perhaps via downregulating iNOS expression. Protection by 1400W at 24-h reperfusion suggests that the role of iNOS in exaggerating reperfusion injury is more prominent in the later stages of injury.
Collapse
Affiliation(s)
- Prerana Patel
- Orthopaedic Microsurgery Laboratory, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Qi WN, Chen LE, Zhang L, Eu JP, Seaber AV, Urbaniak JR. Reperfusion injury in skeletal muscle is reduced in inducible nitric oxide synthase knockout mice. J Appl Physiol (1985) 2004; 97:1323-8. [PMID: 15180976 DOI: 10.1152/japplphysiol.00380.2004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Inducible nitric oxide synthase (iNOS) participates in many pathological events, and selective inhibition of iNOS has been shown to reduce ischemia-reperfusion (I/R) injury in different tissues. To further confirm its role in this injury process, I/R injury was observed in denervated cremaster muscles of iNOS-deficient (iNOS−/−) and wild-type mice. After 3-h ischemia and 90-min reperfusion, blood flow in reperfused muscle was 80 ± 8.5% (mean ± SE) of baseline at 10-min reperfusion and completely returned to the preischemia baseline after 20 min in iNOS−/− mice. In contrast, blood flow was 32 ± 7.4% at 10 min and increased to 60 ± 20% of the baseline level at 90 min in wild-type mice ( P < 0.001 vs. iNOS−/− mice at all time points). The increased muscle blood flow in iNOS−/− mice was associated with significantly less vasospasm in all three sizes of arterial vessel size categories. The weight ratio to the contralateral muscle not subjected to I/R was greater in wild-type mice (173 ± 11%) than in iNOS−/− mice (117 ± 3%; P < 0.01). Inflammation and neutrophil extravasation were also more severe in wild-type mice. Western blot analysis demonstrated an absence of iNOS protein band in iNOS−/− mice and upregulation of iNOS protein expression in wild-type mice. Our results confirm the importance of iNOS in I/R injury. Upregulated iNOS exacerbates I/R injury and appears to be a therapeutic target in protection of tissues against this type of injury.
Collapse
Affiliation(s)
- Wen-Ning Qi
- Orthopaedic Research Laboratory, Duke Univ. Medical Center, Box 3093, Durham, NC 27710, USA
| | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Abstract
Reactive oxygen species have been implicated in brain injury after cerebral ischemia. These oxidants can damage proteins, lipids, and DNA, and lead to cell injury and necrosis. Oxidants are also initiators in intracellular cell death signaling pathways that may lead to apoptosis. The possible targets of this redox signaling include mitochondria, death membrane receptors, and DNA repair enzymes. Genetic manipulation of intrinsic antioxidants and the factors in the signaling pathways has provided substantial progress in understanding the mechanisms in cell death signaling pathways and involvement of oxygen radicals in ischemic brain injury. Future studies of these pathways may provide novel therapeutic strategies in clinical stroke.
Collapse
Affiliation(s)
- Taku Sugawara
- Department of Neurosurgery, Department of Neurology and Neurological Sciences, and Program in Neurosciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | |
Collapse
|
41
|
Schneider R, Raff U, Vornberger N, Schmidt M, Freund R, Reber M, Schramm L, Gambaryan S, Wanner C, Schmidt HHHW, Galle J. L-Arginine counteracts nitric oxide deficiency and improves the recovery phase of ischemic acute renal failure in rats. Kidney Int 2003; 64:216-25. [PMID: 12787412 DOI: 10.1046/j.1523-1755.2003.00063.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND In ischemic acute renal failure (ARF), nitric oxide-dependent regulation of renal hemodynamics and glomerular function is disturbed. Previous studies indicate that the nitric oxide precursor l-arginine (l-Arg) has beneficial effects on renal function. Here we further analyzed the impact of l-Arg on functional and biochemical parameters of nitric oxide signaling during the course of ischemic ARF. METHODS Ischemic ARF was induced in rats by bilateral clamping of renal arteries for 45 minutes. l-Arg was applied intraperitoneally during clamping, and orally during 14 days of follow-up. Glomerular filtration rate (GFR) and renal plasma flow (RPF) were measured, and biochemical parameters analyzed by protein immunoblots. RESULTS Clamping resulted in 70% to 90% reduction of GFR and RPF, with a gradual recovery by day 14. Using an in situ assay with the oxidative fluorescent dye hydroethidine, increased tubular generation of O2- was detected in the early course of ischemic ARF, indicating enhanced oxidative stress. These findings were accompanied by up-regulation of the nitric oxide receptor, soluble guanylate cyclase, and by significant regulatory changes of inducible nitric oxide synthase (iNOS) and endothelial NOS expression. l-Arg had a beneficial effect on GFR and RPF, decreased O2- production, diminished up-regulation of soluble guanylate cyclase, and prevented up-regulation of iNOS. CONCLUSION Ischemic ARF is accompanied by marked alterations in the expression of key enzymes of the nitric oxide pathway, indicative for deficiency of constitutive NOS activity. l-Arg supplementation reduces O2- generation and significantly improves the expression of nitric oxide signaling proteins as well as the recovery phase of ischemic ARF.
Collapse
Affiliation(s)
- Reinhard Schneider
- Division of Nephrology, Department of Medicine, Julius-Maximilians-University, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Muhs A, Heublein B, Schletter J, Herrmann A, Rüdiger M, Sturm M, Grust A, Malms J, Schrader J, von der Leyen HE. Preclinical evaluation of inducible nitric oxide synthase lipoplex gene therapy for inhibition of stent-induced vascular neointimal lesion formation. Hum Gene Ther 2003; 14:375-83. [PMID: 12659678 DOI: 10.1089/104303403321208970] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Several reports have established the concept of nitric oxide synthase (NOS) gene transfer for inhibiting smooth muscle cell (SMC) proliferation after vascular injury. To minimize potential risks associated with viral gene transfer, we developed a liposome-based gene transfer approach employing inducible NOS (iNOS) overexpression for inhibition of stent-induced neointimal lesion formation. Therapeutic lipoplexes were transferred to femoral or coronary arteries of Goettingen minipigs, using the Infiltrator local drug delivery device. Efficiency of local iNOS lipoplex transfer was analyzed by iNOS-specific immunohistochemistry. NO-mediated inhibition of stent-induced neointimal lesion formation was analyzed by intravascular ultrasound (IVUS) and computerized morphometry. Gene transfer efficiency increased dose dependently to a maximum of 44.3 +/- 4.2% iNOS-positive vessel area (dose, 2 microg of iNOS lipoplex). Proliferating cell nuclear antigen (PCNA) expression of medial SMCs (immunohistochemistry) was inhibited significantly by transfer of 2 microg of iNOS lipoplexes (111 +/- 27 cells [iNOS] versus 481 +/- 67 cells [control; PCNA-positive medial cells]). IVUS analysis demonstrated that local transfer of iNOS lipoplexes resulted in a significant reduction of femoral in-stent plaque area (control, 40.85 +/- 6.37 mm(2); iNOS, 24.69 +/- 1.8 mm(2); p = 0.03). Coronary in-stent lesion formation was reduced by about 45% as determined by histologic morphometry (control, 4.0 +/- 0.29; iNOS, 2.2 +/- 0.30; p < 0.01). In conclusion, this study demonstrates that local intramural delivery of iNOS lipoplexes can exert therapeutic effects in inhibiting stent-induced neointimal lesion formation. Together with the nonviral character of this gene therapy approach, these findings may have important impact on the transition of NOS-based gene therapy to clinical practice.
Collapse
Affiliation(s)
- Andreas Muhs
- Cardion AG, Max Planck Strasse 15A, 40699 Erkrath, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ding-Zhou L, Marchand-Verrecchia C, Croci N, Plotkine M, Margaill I. L-NAME reduces infarction, neurological deficit and blood-brain barrier disruption following cerebral ischemia in mice. Eur J Pharmacol 2002; 457:137-46. [PMID: 12464359 DOI: 10.1016/s0014-2999(02)02686-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of nitric oxide (NO) in the development of post-ischemic cerebral infarction has been extensively examined, but fewer studies have investigated its role in other outcomes. In the present study, we first determined the temporal evolution of infarct volume, NO production, neurological deficit and blood-brain barrier disruption in a model of transient focal cerebral ischemia in mice. We then examined the effect of the nonselective NO-synthase inhibitor N(omega)-nitro-L-arginine-methylester (L-NAME). L-NAME given at 3 mg/kg 3 h after ischemia reduced by 20% the infarct volume and abolished the increase in brain NO production evaluated by its metabolites (nitrites/nitrates) 48 h after ischemia. L-NAME with this protocol also reduced the neurological deficit evaluated by the grip test and decreased by 65% the extravasation of Evans blue, an index of blood-brain barrier breakdown. These protective activities of L-NAME suggest that NO has multiple deleterious effects in cerebral ischemia.
Collapse
Affiliation(s)
- Li Ding-Zhou
- Laboratoire de Pharmacologie, Université René Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | | | | | | | | |
Collapse
|
44
|
Baldus S, Eiserich JP, Brennan ML, Jackson RM, Alexander CB, Freeman BA. Spatial mapping of pulmonary and vascular nitrotyrosine reveals the pivotal role of myeloperoxidase as a catalyst for tyrosine nitration in inflammatory diseases. Free Radic Biol Med 2002; 33:1010. [PMID: 12361810 DOI: 10.1016/s0891-5849(02)00993-0] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Nitrotyrosine (NO(2)Tyr) formation is a hallmark of acute and chronic inflammation and has been detected in a wide variety of human pathologies. However, the mechanisms responsible for this posttranslational protein modification remain elusive. While NO(2)Tyr has been considered a marker of peroxynitrite (ONOO(-)) formation previously, there is growing evidence that heme-protein peroxidase activity, in particular neutrophil-derived myeloperoxidase (MPO), significantly contributes to NO(2)Tyr formation in vivo via the oxidation of nitrite (NO(2)(-)) to nitrogen dioxide (.NO(2)). Coronary arteries from a patient with coronary artery disease, liver and lung tissues from a sickle cell disease patient, and an open lung biopsy from a lung transplant patient undergoing rejection were analyzed immunohistochemically to map relative tissue distributions of MPO and NO(2)Tyr. MPO immunodistribution was concentrated along the subendothelium in coronary tissue and hepatic veins as well as in the alveolar epithelial compartment of lung tissue from patients with sickle cell disease or acute rejection. MPO immunoreactivity strongly colocalized with NO(2)Tyr formation, which was similarly distributed in the subendothelial and epithelial regions of these tissues. The extracellular matrix protein fibronectin (FN), previously identified as a primary site of MPO association in vascular inflammatory reactions, proved to be a major target protein for tyrosine nitration, with a strong colocalization of MPO, NO(2)Tyr, and tissue FN occurring. Finally, lung tissue from MPO(-/-) mice, having tissue inflammatory responses stimulated by intraperitoneal zymosan administration, revealed less subendothelial NO(2)Tyr immunoreactivity than tissue from wild-type mice, confirming the significant role that MPO plays in catalyzing tissue nitration reactions. These observations reveal that (i) sequestration of neutrophil-derived MPO in vascular endothelial and alveolar epithelial compartments is an important aspect of MPO distribution and action in vivo, (ii) MPO-catalyzed NO(2)Tyr formation occurs in diverse vascular and pulmonary inflammatory pathologies, and (iii) extracellular matrix FN is an important target of tyrosine nitration in these inflammatory processes.
Collapse
Affiliation(s)
- Stephan Baldus
- Department of Anesthesiology, University of Alabama, Birmingham, AL 35233, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Thomas DD, Espey MG, Vitek MP, Miranda KM, Wink DA. Protein nitration is mediated by heme and free metals through Fenton-type chemistry: an alternative to the NO/O2- reaction. Proc Natl Acad Sci U S A 2002; 99:12691-6. [PMID: 12226478 PMCID: PMC130522 DOI: 10.1073/pnas.202312699] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The chemical origins of nitrated tyrosine residues (NT) formed in proteins during a variety of pathophysiological conditions remain controversial. Although numerous studies have concluded that NT is a signature for peroxynitrite (ONOO(-)) formation, other works suggest the primary involvement of peroxidases. Because metal homeostasis is often disrupted in conditions bearing NT, the role of metals as catalysts for protein nitration was examined. Cogeneration of nitric oxide (NO) and superoxide (O(2)(-)), from spermine/NO (2.7 microM/min) and xanthine oxidase (1-28 microM O(2)(-)/min), respectively, resulted in protein nitration only when these species were produced at approximately equivalent rates. Addition of ferriprotoporphyrin IX (hemin) to this system increased nitration over a broad range of O(2)(-) concentrations with respect to NO. Nitration in the presence of superoxide dismutase but not catalase suggested that ONOO(-) might not be obligatory to this process. Hemin-mediated NT formation required only the presence of NO(2)(-) and H(2)O(2), which are stable end-products of NO and O(2)(-) degradation. Ferrous, ferric, and cupric ions were also effective catalysts, indicating that nitration is mediated by species capable of Fenton-type chemistry. Although ONOO(-) can nitrate proteins, there are severe spatial and temporal constraints on this reaction. In contrast, accumulation of metals and NO(2)(-) subsequent to NO synthase activity can result in far less discriminate nitration in the presence of an H(2)O(2) source. Metal catalyzed nitration may account for the observed specificity of protein nitration seen under pathological conditions, suggesting a major role for translocated metals and the labilization of heme in NT formation.
Collapse
Affiliation(s)
- Douglas D Thomas
- Tumor Biology Section, Radiation Biology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
46
|
Suzuki M, Tabuchi M, Ikeda M, Tomita T. Concurrent formation of peroxynitrite with the expression of inducible nitric oxide synthase in the brain during middle cerebral artery occlusion and reperfusion in rats. Brain Res 2002; 951:113-20. [PMID: 12231464 DOI: 10.1016/s0006-8993(02)03145-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peroxynitrite is assumed to play a crucial role in brain damage associated with the overproduction of nitric oxide (NO). The purpose of this study is to examine time-dependent changes of nitrite and nitrate (NOx) concentration in the circulation, and peroxynitrite formation as well as the expression of inducible nitric oxide synthase (iNOS) in the penumbra of rat brains during transient middle cerebral artery occlusion (MCAO) of Wistar rat for 2 h and reperfusion for 4-70 h. NOx concentration in the circulation was continuously monitored at the right jugular vein by microdialysis. The expression of iNOS was detected at 22-70 h after reperfusion in vascular walls and the cortex. Nitrotyrosine, a marker of peroxynitrite, appeared 4 h after reperfusion in the cortex, increasing substantially at 22-46 h in vascular walls. NOx level in dialysate increased immediately after MCAO. After a gradual decrease, the level increased again 4 h after reperfusion, reaching a maximum at 46 h. Brain myeloperoxidase activity, a marker of neutrophil infiltration, was not detected 4 h after reperfusion, but greatly increased at 22 h and then decreased. These results suggest that a marked increase of NOx level in the circulation might reflect the expression of iNOS, while neuronal NOS may contribute to peroxynitrite formation in the cortex observed at an earlier phase of reperfusion. This study indicates that monitoring NOx level in the circulation serves to assess the progress of stroke, and to determine appropriate therapeutic measures.
Collapse
Affiliation(s)
- Motohisa Suzuki
- Graduate School of Health Sciences, University of Shizuoka, 52-1, Yada, Shizuoka 422-8526, Japan
| | | | | | | |
Collapse
|
47
|
Zhu DY, Deng Q, Yao HH, Wang DC, Deng Y, Liu GQ. Inducible nitric oxide synthase expression in the ischemic core and penumbra after transient focal cerebral ischemia in mice. Life Sci 2002; 71:1985-96. [PMID: 12175893 DOI: 10.1016/s0024-3205(02)01970-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The present observations examined the hypothesis that the iNOS expression in the ischemic penumbra after a transient focal ischemic insult is involved in the recruitment of penumbra into infarction. The middle cerebral artery in mice was occluded for 2 h by an intraluminal filament and then recirculated. The measurement of iNOS activity, iNOS protein formation and NO concentration in the ischemic core and penumbra, and the determination of infarct volume were performed at 6, 12, 24 and 48 h after reperfusion. iNOS protein and iNOS enzymatic activity appeared at 6 h, peaked at 24 h, and declined at 48 h in the penumbra after reperfusion. iNOS protein was not detectable in contralateral area and in sham-operated brains. The time course of iNOS protein, enzymatic activity and NO concentration in the penumbra but not in the core matched the process of infarct maturation. Treatment with iNOS inhibitor aminoguanidine (100 mg.kg(-1), i.p.) at 6 and 12 h after reperfusion inhibited iNOS activity by 88.0 +/- 10.4% and reduced NO concentration by 48.5 +/- 8.3% in the penumbra, and lessened infarct size by 48.8 +/- 7.2%. The iNOS activity and NO level in the core were not affected by the administration of aminoguanidine. These results suggest that iNOS expression in the ischemic penumbra is involved in the recruitment of penumbra into infarction and thereby contributing to the enlargement of infarct.
Collapse
Affiliation(s)
- Dong-Ya Zhu
- Pharmacology Department, New Drug Research Center, China Pharmaceutical University, Tong Jia Xiang 24#, Nanjing 210009, China.
| | | | | | | | | | | |
Collapse
|
48
|
Borkowski A, Younge BR, Szweda L, Mock B, Björnsson J, Moeller K, Goronzy JJ, Weyand CM. Reactive nitrogen intermediates in giant cell arteritis: selective nitration of neocapillaries. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 161:115-23. [PMID: 12107096 PMCID: PMC1850706 DOI: 10.1016/s0002-9440(10)64163-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Arterial wall damage in giant cell arteritis (GCA) is mediated by several different macrophage effector functions, including the production of metalloproteinases and lipid peroxidation. Tissue-invading macrophages also express nitric oxide synthase (NOS)-2, but it is not known whether nitric oxide-related mechanisms contribute to the disease process. Nitric oxide can form nitrating agents, including peroxynitrite, a nitric oxide congener formed in the presence of reactive oxygen intermediates. Protein nitration selectively targets tyrosine residues and can result in a gain, as well as a loss, of protein function. Nitrated tyrosine residues in GCA arteries were detected almost exclusively on endothelial cells of newly formed microcapillaries in the media, whereas microvessels in the adventitia and the intima were spared. Nitration correlated with endothelial NOS-3 expression and not with NOS-2-producing macrophages, which preferentially homed to the hyperplastic intima. The restriction of nitration to the media coincided with the production of reactive oxygen intermediates as demonstrated by the presence of the toxic aldehyde, 4-hydroxynonenal. Depletion of tissue-infiltrating macrophages in human temporal artery-SCID mouse chimeras disrupted nitrotyrosine generation, demonstrating a critical role of macrophages in the nitration process that targeted medial microvessels. Thus, protein nitration in GCA is highly compartmentalized, reflecting the production of reactive oxygen and reactive nitrogen intermediates in the inflamed arterial wall. Heterogeneity of microvessels in NOS-3 regulation may be an additional determinant contributing to this compartmentalization and could explain the preferential targeting of newly generated capillary beds.
Collapse
Affiliation(s)
- Astrid Borkowski
- Department of Medicine and Immunology, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Influence of mild hypothermia on inducible nitric oxide synthase expression and reactive nitrogen production in experimental stroke and inflammation. J Neurosci 2002. [PMID: 12019311 DOI: 10.1523/jneurosci.22-10-03921.2002] [Citation(s) in RCA: 135] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Mild hypothermia is neuroprotective, but the reasons are not well known. Inflammation contributes to ischemic damage; therefore, we examined whether the protection by hypothermia may be attributable to alterations in the inflammation. We examined whether hypothermia might alter the inflammatory cell-associated inducible nitric oxide synthase (iNOS) and subsequent nitric oxide (NO) and peroxynitrite generation in experimental stroke and inflammation. Rats underwent 2 hr of middle cerebral artery occlusion (MCAO). Brain inflammation was modeled by intravenous lipopolysaccharide (LPS) (2 mg/kg) injection. Temperature was maintained at 33 degrees C for 2 hr immediately after MCAO and LPS injection, delayed 2 hr after MCAO or maintained at 38 degrees C. Cultured microglia were activated with LPS and then incubated at 33 or 37 degrees C. Both intraischemic and delayed mild hypothermia attenuated infarct size by 40% (p < 0.05). Immunohistochemistry was performed to identify cell type, iNOS, and peroxynitrite. The majority of iNOS- and peroxynitrite-positive cells were activated microglia-macrophages, and mild hypothermia significantly decreased the numbers of immunoreactive cells at 72 hr by >50% (p < 0.05). After ischemia, mild hypothermia decreased NO production by 40%. Similarly, hypothermia attenuated NO and iNOS in LPS-injected rats, as well as in cultured microglia. Aminoguanidine, an iNOS inhibitor, also attenuated infarct size and NO in ischemic and inflammation models. We conclude that mild hypothermia significantly inhibits the inflammatory response by affecting microglial iNOS-NO generation. Therapies directed against microglia or their activation may be useful in treating stroke.
Collapse
|
50
|
Chatzipanteli K, Garcia R, Marcillo AE, Loor KE, Kraydieh S, Dietrich WD. Temporal and segmental distribution of constitutive and inducible nitric oxide synthases after traumatic spinal cord injury: effect of aminoguanidine treatment. J Neurotrauma 2002; 19:639-51. [PMID: 12042098 DOI: 10.1089/089771502753754109] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nitric oxide (NO) has been shown to play an important role in the pathophysiology of traumatic brain injury (TBI) and cerebral ischemia. However, its contribution to the pathogenesis of traumatic spinal cord injury (SCI) remains to be clarified. This study determined the time course of constitutive and inducible nitric oxide synthases (cNOS and iNOS, respectively) after SCI. Rats underwent moderate SCI at T10 using the NYU impactor device and were allowed to survive for 3, 6, or 24 h and 3 days after SCI (n = 5 in each group). For the determination of enzymatic activities, spinal cords were dissected into five segments, including levels rostral and caudal (remote) to the injury site. Other rats were perfusion fixed for the immunohistochemical localization of iNOS protein levels. cNOS activity was significantly decreased at 3 and 6 h within the traumatized T10 segment and at 3, 6, and 24 h at the rostral (T9) level (p < 0.05). Rostral (T8) and caudal (T11, T12) to the injury site cNOS activity was also decreased at 3 h after injury (p < 0.05). However, cNOS activity returned to control levels within 6 h at T8, T11 and T12 and at one day at T10 and T9 segments. iNOS enzymatic activity was elevated at all time points tested (p < 0.05), with the most robust increase observed at 24 h. Immunostaining for iNOS at 24 h revealed that a significant cellular source of iNOS protein appeared to be invading polymorphonuclear leukocytes (PMNLs). To assess the functional consequences of iNOS inhibition, aminoguanidine treatment was initiated 5 min after SCI and rats tested using the BBB open field locomotor score. Treated rats demonstrated significantly improved hindlimb function up to 7 weeks after SCI. Histopathological analysis of contusion volume showed that aminoguanidine treatment decreased lesion volume by 37% (p < 0.05). In conclusion, these results indicate that (1) cNOS and iNOS activities are regionally and temporally affected after moderate SCI, (2) the early accumulation of PMNLs are a potentially significant source of NO-induced cytotoxic products, and (3) acute aminoguanidine treatment significantly improves functional and histopathological outcome after SCI.
Collapse
Affiliation(s)
- Katina Chatzipanteli
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami School of Medicine, Miami, Florida 33101, USA.
| | | | | | | | | | | |
Collapse
|