首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
韩笑  刘建勋 《中国药理学通报》2006,22(10):1211-1214
目的探讨双参通冠方药物血清对培养心肌细胞缺氧/复氧损伤的Ca2+超载、NOS-NO系统的影响。方法培养心肌细胞,建立缺氧/复氧损伤模型。运用血清药理学方法研究双参通冠方药物血清对缺氧/复氧心肌Ca2+超载、NOS-NO系统的影响。荧光分光光度法测定心肌细胞胞质[Ca2+]i,比色法检测心肌细胞培养上清NOS活性、NO含量。结果缺氧/复氧后[Ca2+]i增高,总NOS(T-NOS)及诱导型NOS(iNOS)活性增高,双参通冠方药物血清能降低缺氧/复氧心肌细胞[Ca2+]i,并能降低T-NOS、iNOS活性和NO含量(P<0.05)。结论双参通冠方药物血清可抑制缺氧/复氧损伤时心肌细胞Ca2+超载及NOS-NO系统的激活。  相似文献   

2.
目的:研究羟丁酸钠(SO)在大鼠海马脑片缺氧/复氧(H/R)损伤中的保护作用,揭示SO在缺血性脑损伤中的保护作用机制。方法:采用大鼠海马脑片H/R损伤模型。设正常对照组,H/R组,SO1、10、100μmol/L组,NCS-382100μmol/L+SO100μmol/L(NCS-382+SO)组、NCS-356100μmol/L(NCS-356)组。测定脑片孵育液中乳酸脱氢酶(LDH)释放率,γ-氨基丁酸(GABA)、谷氨酸(Glu)含量;脑片进行TTC染色计算组织损伤百分率;HE染色观察组织病理形态学变化,流式细胞仪测定细胞内钙;酶组织化学法检测一氧化氮合酶(NOS)的表达。结果:SO明显降低H/R海马脑片LDH释放率(P〈0.01),提高TTC染色的A490值,降低组织损伤百分率(P〈0.01),升高GABA/Glu比值(P〈0.01),减轻H/R所致的组织病理损伤。H/R组脑片细胞内钙荧光强度和NOS阳性神经元明显高于正常对照组(P〈0.01);SO100μmol/L组、NCS-356组细胞内钙荧光强度较H/R组显著减弱(P〈0.01),NOS阳性神经元的数目明显减少(P〈0.01)。用γ-羟基丁酸(GHB)受体选择性阻断剂NCS-382后再使用SO,细胞内钙荧光强度、NOS阳性神经元的数目均接近H/R组。结论:SO对大鼠海马脑片H/R损伤有明显的保护作用,其机制可能与升高GABA/Glu比值,激动GHB受体抑制细胞内钙的增高及NOS的表达有关。  相似文献   

3.
NITRIC OXIDE: ROLE IN NEUROTOXICITY   总被引:1,自引:0,他引:1  
1. Nitric oxide (NO) is a novel neuronal messenger molecule which interacts with surrounding neurones, not by synaptic transmission but by diffusion between cells. 2. No is produced following stimulation of the enzyme, NO synthase (NOS). After synthesis, NO exerts its biological actions by diffusion to the site of action. Therefore, the way to regulate the physiological actions of NO is to regulate NOS. 3. NOS is activated by the influx of calcium from glutamate-activated N-methyl-D-aspartate receptors. Overactivation of these receptors leads to overproduction of NO and neuronal cell death. 4. NOS can be regulated at the catalytic site, at the flavoproteins, at the calmodulin site and by phosphorylation. 5. In excess, NO is toxic to neurones. This toxicity is mediated largely by an interaction with the superoxide anion, presumably through the generation of the oxidant, peroxynitrite. 6. NO or peroxynitrite-mediated neuronal injury involves the activation of the nuclear protein, poly(ADP-ribose)synthetase.  相似文献   

4.
Oxidative stress results from an oxidant/antioxidant imbalance, an excess of oxidants and/or a depletion of antioxidants. A vast amount of circumstantial evidence implicates oxygen-derived free radicals (especially, superoxide and hydroxyl radical) and high energy oxidants (such as peroxynitrite) as mediators of secondary damage associated with spinal cord injury. Reactive oxygen species (ROS) (e.g., superoxide, peroxynitrite, hydroxyl radical and hydrogen peroxide) are all potential reactants capable of initiating DNA single strand breakage, with subsequent activation of the nuclear enzyme poly (ADP ribose) synthetase (PARS), leading to eventual severe energy depletion of the cells, and necrotic-type cell death. Moreover, Poly(ADP-ribosyl)ation is regulated by the synthesizing enzyme poly(ADP-ribose) polymerase-1 (PARP-1) and the degrading enzyme poly(ADP-ribose) glycohydrolase (PARG). Here, we review the roles of ROS, PARP-1 and PARG in spinal cord injury as well as the beneficial effect of the in vivo treatment with novel pharmacological tools (e.g. peroxynitrite decomposition catalysts, selective superoxide dismutase mimetics (SODm), PARP-1 and PARG inhibitors.  相似文献   

5.
目的研究从玉郎伞(Yulangsan,YLS)中首次分离的两种黄酮单体对体外培养大鼠乳鼠心肌细胞缺氧/复氧损伤的保护作用,并初步探讨其作用机制。方法建立体外培养大鼠乳鼠心肌细胞缺氧/复氧损伤模型,倒置显微镜下观察加入YLS两种单体的含药血清(10%、5%、2.5%)后,各组缺氧/复氧心肌细胞形态学和搏动频率的变化;以MTT法检测各组细胞的存活率;用ELISA法测定心肌细胞Na+,K+-ATP酶、Ca2+,Mg2+-ATP酶活性及细胞培养上清液中总超氧化物歧化酶(T-SOD)、乳酸脱氢酶(LDH)、一氧化氮合酶(NOS)活性和丙二醛(MDA)含量。结果与模型组相比,YLS两种单体含药血清的高、中剂量均能明显增加心肌细胞的存活率,增强Na+,K+-ATP酶、Ca2+,Mg2+-ATP酶活性,降低细胞培养上清液LDH、NOS活性、MDA含量,提高T-SOD活性并呈剂量依赖性(P<0.05)。结论两种YLS单体对体外培养大鼠乳鼠心肌细胞缺氧/复氧损伤具有保护作用,其机制可能与清除自由基、抑制心肌细胞Ca2+超载有关。  相似文献   

6.
Recent studies evidence that peroxynitrite is spontaneously formed when nitric oxide (NO) and superoxide coexist and suggest that it is likely to be involved in the destruction of the pancreatic beta cells. We examined whether drugs that inhibit nitric oxide synthase (NOS) or scavenge peroxynitrite could abrogate STZ-induced hyperglycemia in rats. Blood glucose levels were measured before (0 h) and 24, 48, and 72 h following intraperitoneal administration of 60 mg/kg streptozotocin (STZ). The levels of blood sugar in STZ-treated control animals were significantly elevated at all time points of observation with a peak increase at 48 h. The hyperglycemic response of STZ was found to be significantly reduced in animals pretreated with aminoguanidine (50 mg/kg i.p.), an inducible isoform-selective NOS (iNOS) inhibitor with antioxidant property, and by melatonin (6 mg/kg i.p.), an antioxidant that also prevents peroxynitrite formation but not by Nw-nitro-L-arginine methyl ester (L-NAME, 10 mg/kg i.p.), and 7-nitroindazole (7-NI, 50 mg/kg i.p.), the constitutive inhibitors of endothelial and neuronal NOS, respectively. These findings indicate the possible participation of iNOS-derived NO as well as oxygen free radicals in STZ-induced pancreatic beta cell destruction and compounds that act as scavengers of peroxynitrite may offer protection against such a damage.  相似文献   

7.
The substance 4-(4-fluorophenyl)-2-methyl-6-(5-piperidinopentyloxy) pyrimidine hydrochloride (NS-7) has been developed recently as a cerebroprotective compound with Na+ and Ca2+ channel blocking action. In the present study, the effect of NS-7 in an in vitro model of hypoxic injury was examined and the possible involvement of Na+ and Ca2+ channels in the hypoxic injury subsequently determined. When slices of rat cerebral cortex were exposed to hypoxia/glucose deprivation followed by reoxygenation and restoration of the glucose supply, marked leakage of lactate dehydrogenase (LDH) occurred 3–6 h after reoxygenation. This hypoxia/reoxygenation-induced injury was blocked almost completely by the removal of extracellular Ca2+ or by chelating intracellular Ca2+ with 1,2-bis(o-aminophenoxy)ethane-N,N,N’,N’-tetraacetic acid tetra(acetoxymethyl)ester (BAPTA/AM). In addition, combined treatment with the N-type Ca2+ channel blocker ω-conotoxin GVIA and the P/Q-type Ca2+ channel blocker ω-agatoxin IVA significantly reduced LDH leakage, although neither of these Ca2+ channel blockers alone, nor nimodipine, an L-type Ca2+ channel blocker, was effective. On the other hand, several Na+ channel blockers, including tetrodotoxin, local anaesthetics and antiepileptics, significantly reduced the hypoxic injury. NS-7 (3–30 μM) concentration-dependently inhibited LDH leakage caused by hypoxia/reoxygenation, but had no influence on the reduction of tissue ATP content and energy charge during hypoxia and glucose deprivation. It is suggested that blockade of Na+ and Ca2+ channels is implicated in the cerebroprotective action of NS-7. Received: 10 March 1998 / Accepted: 19 April 1998  相似文献   

8.
1-Methyl-3-nitro-1-nitrosoguanidine (MNNG) is a DNA alkylating agent. DNA alkylation by MNNG is known to trigger accelerated poly(ADP-ribose) metabolism. Various nitroso compounds release nitric oxide (NO). Therefore, we set out to investigate whether MNNG functions as NO donor and whether MNNG-derived NO or secondary NO metabolites such as peroxynitrite contribute to MNNG-induced cytotoxicity. MNNG in aqueous solutions resulted in time- and concentration-dependent NO release and nitrite/nitrate formation. Moreover, various proteins in MNNG-treated thymocytes were found to be nitrated, indicating that MNNG-derived NO may combine with cellular superoxide to form peroxynitrite, a nitrating agent. MNNG also caused DNA breakage and increased poly(ADP-ribose) polymerase activity and cytotoxicity in thymocytes. MNNG-induced DNA damage (measured by the comet assay) and thymocyte death (measured by propidium iodide uptake) was prevented by the PARP inhibitor PJ-34 and by glutathione (GSH) or N-acetylcysteine (NAC). The cytoprotection provided by PJ-34 against necrotic parameters was paralleled by increased outputs in apoptotic parameters (caspase activity, DNA laddering) indicating that PARP activation diverts apoptotic death toward necrosis. As MNNG-induced cytotoxicity showed many similarities to peroxynitrite-induced cell death, we tested whether peroxynitrite was responsible for at least part of the cytotoxicity induced by MNNG. Cell-permeable enzymic antioxidants (superoxide dismutase and catalase), the NO scavenger cPTIO or the peroxynitrite decomposition catalyst FP15 failed to inhibit MNNG-induced DNA breakage and cytotoxicity. In conclusion, MNNG induces tyrosine nitration in thymocytes. Furthermore, MNNG damages DNA by a radical mechanism that does not involve NO or peroxynitrite.  相似文献   

9.
一氧化氮参与预处理心肌细胞早期保护作用   总被引:2,自引:2,他引:0  
目的 明确一氧化氮 (NO)是否诱导预处理心肌细胞早期保护作用。方法 取体外培养的新生大鼠心肌细胞 ,分为如下各组 :①阴性对照组 (Normal组 ) ;②SNAP(5 0 0 μmol·L-1)预处理组 (NO组 ) ;③缺氧预处理组 (HP组 ) ;④NO合成抑制剂L NAME作用细胞后再行缺氧预处理组 (L NAME +HP组 ) ;⑤L 精氨酸 (L Arg)预处理心肌细胞组(L Arg组 ) ;⑥L NAME与L Arg共同作用于心肌细胞组(L NAME +L Arg组 ) ;⑦缺氧复氧损伤组 (H/R组 )。②~⑥组细胞在给予干预因素后使细胞经历 6h缺氧及 3h复氧 ,阳性对照组不予任何处理直接使其缺氧 6h复氧 3h。分别检测心肌细胞存活率及乳酸脱氢酶 (LDH)活性 ,以判定心肌细胞损伤程度。结果 NO预处理心肌细胞后使其缺氧复氧损伤减轻 ,表现为与单纯缺氧复氧组细胞比较其培养上清LDH活性降低 ,细胞存活率提高 (P <0 0 1) ;缺氧预处理和L Arg预处理细胞均可减轻心肌细胞缺氧复氧损伤 (P<0 0 1) ,但此作用可被NOS抑制剂L NAME所拮抗。结论 内源性及外源性NO均可诱导心肌细胞预处理早期保护作用  相似文献   

10.
The toxic effects of nitrogen dioxide (NO2) and ozone (O3) are mediated through the formation of free radicals, which can cause DNA strand breaks. The present study demonstrates that exposure to NO2 and O3 causes a stimulation of poly(ADP-ribose) (polyADPR) synthetase in alveolar macrophages of rats. Three-month-old male Sprague-Dawley rats, specific pathogen free, were exposed to either 1.2 ppm NO2 or 0.3 ppm O3 alone or a combination of these 2 oxidants continuously for 3 days. The control group was exposed to filtered room air. To evaluate whether exposure to these two oxidants (NO2 and O3) caused DNA damage to lung cells, the activity of polyADPR synthetase was measured. Cellular DNA repair is dependent upon the formation of poly(ADP-ribose) polymerase, which is catalyzed by polyADPR synthetase. PolyADPR synthetase is known to be activated in response of DNA damage. The results showed that the enzyme activity was stimulated after exposure to O3 or exposure to NO2 + O3. Ozone exposure caused a 25% increase in the enzyme activity as compared to the control. Combined exposure to NO2 + O3 showed a 53% increase in the enzyme activity. These results were statistically significant as compared to the control and NO(2) exposure groups. Other parameters such as total cell count, cell viability, and differential cell count were also determined. The stimulation of polyADPR synthetase activity after O3 exposure or NO2 + O3 exposure reflects a response to lung cellular DNA repair, which may be used as an indicator for assessing DNA damage caused by oxidant injury.  相似文献   

11.
We have previously reported that free radical-mediated injury induced by cephaloridine (CER) is enhanced by phorbol 12-myristate 13-acetate (PMA), a protein kinase C (PKC) activator, in rat renal cortical slices. We have also shown that PKC activation in mitochondria is involved in CER-induced nephrotoxicity in rats. We investigated the role of a downstream PKC pathway, a MEK/ERK pathway, in free radical-induced injury in rat renal cortical slices exposed to CER. Immediately after preparing slices from rat renal cortex, the slices were incubated in the medium containing MEK inhibitors. ERK1/2 activation was determined by Western blot analysis for phosphorylated ERK (pERK) 1/2 protein in nucleus fraction prepared from the slices exposed to CER. Prominently, CER caused not only increases in lipid peroxidation as an index of free radical generation and in LDH leakage as that of cell injury in the slices, but also marked activation of ERK1/2 in nucleus fraction. PD98059 and U0126, MEK1/2 inhibitors, significantly attenuated CER-induced increases in lipid peroxidation and LDH leakage in the slices. PD98059 also suppressed ERK1/2 activation in nucleus fraction prepared from the slices treated with CER. Inhibition of other MAP kinase pathways, p38 MAP kinase and c-Jun N-terminal kinase (JNK) had no effect on CER-induced increases in lipid peroxidation level and LDH leakage in the slices. The present results suggest that a MEK/ERK pathway down stream of a PKC pathway is probably involved in free radical-induced injury in rat renal cortical slices exposed to CER.  相似文献   

12.
A vast amount of circumstantial evidence implicates oxygen-derived free radicals (especially superoxide and hydroxyl radical) and high-energy oxidants (such as peroxynitrite) as mediators of inflammation, shock, and ischemia/reperfusion injury. The aim of this review is to describe recent developments in the field of oxidative stress research. The first part of the review focuses on the roles of reactive oxygen species (ROS) in shock, inflammation, and ischemia/reperfusion injury. The second part of the review deals with the novel findings using recently identified pharmacological tools (e.g., peroxynitrite decomposition catalysts and selective superoxide dismutase mimetics (SODm) in shock, ischemia/reperfusion, and inflammation. 1) The role of ROS consists of immunohistochemical and biochemical evidence that demonstrates the production of ROS in shock, inflammation, and ischemia/reperfusion injury. ROS can initiate a wide range of toxic oxidative reactions. These include initiation of lipid peroxidation, direct inhibition of mitochondrial respiratory chain enzymes, inactivation of glyceraldehyde-3-phosphate dehydrogenase, inhibition of membrane sodium/potassium ATPase activity, inactivation of membrane sodium channels, and other oxidative modifications of proteins. All these toxicities are likely to play a role in the pathophysiology of shock, inflammation, and ischemia/reperfusion. 2) Treatment with either peroxynitrite decomposition catalysts, which selectively inhibit peroxynitrite, or with SODm, which selectively mimic the catalytic activity of the human superoxide dismutase enzymes, have been shown to prevent in vivo the delayed vascular decompensation and the cellular energetic failure associated with shock, inflammation, and ischemia/reperfusion injury. ROS (e.g., superoxide, peroxynitrite, hydroxyl radical, and hydrogen peroxide) are all potential reactants capable of initiating DNA single-strand breakage, with subsequent activation of the nuclear enzyme poly(ADP-ribose) synthetase, leading to eventual severe energy depletion of the cells and necrotic-type cell death. Antioxidant treatment inhibits the activation of poly(ADP-ribose) synthetase and prevents the organ injury associated with shock, inflammation, and ischemia/reperfusion.  相似文献   

13.
Heart failure is the major cause of hospitalization, morbidity and mortality worldwide. Previous experimental and clinical studies have suggested that there is an increased production of reactive oxygen species (ROS: superoxide, hydrogen peroxide, hydroxyl radical) both in animals and in patients with acute and chronic heart failure. The possible source of increased ROS in the failing myocardium include xanthine and NAD(P)H oxidoreductases, cyclooxygenase, the mitochondrial electron transport chain and activated neutrophils among many others. The excessively produced nitric oxide (NO) derived from NO synthases (NOS) has also been implicated in the pathogenesis of chronic heart failure (CHF). The combination of NO and superoxide yields peroxynitrite, a reactive oxidant, which has been shown to impair cardiac function via multiple mechanisms. Increased oxidative and nitrosative stress also activates the nuclear enzyme poly(ADP-ribose) polymerase (PARP), which importantly contributes to the pathogenesis of cardiac and endothelial dysfunction associated with myocardial infarction, chronic heart failure, diabetes, atherosclerosis, hypertension, aging and various forms of shock. Recent studies have demonstrated that pharmacological inhibition of xanthine oxidase derived superoxide formation, neutralization of peroxynitrite or inhibition of PARP provide significant benefit in various forms of cardiovascular injury. This review discusses the role of oxidative/nitrosative stress and downstream pathways in various forms of cardiomyopathy and heart failure.  相似文献   

14.
Urantide对心肌缺血性损伤的保护作用   总被引:1,自引:0,他引:1  
姚华  陈志武 《药学学报》2008,43(2):150-156
研究UT受体拮抗剂——urantide对心肌缺血性损伤的保护作用及其机制。小鼠心肌缺血采用皮下注射(sc)异丙肾上腺素(Iso)法,观察心电图ST段的变化,测定小鼠血清中乳酸脱氢酶(LDH)、一氧化氮合酶(NOS)的活性以及丙二醛(MDA)、一氧化氮(NO)的含量, HE染色观察心肌组织病理损伤情况。建立乳鼠原代心肌细胞缺氧再给氧模型,采用ELISA法观察urantide对细胞培养上清液中肌钙蛋白I(cTnI)的影响,比色法测LDH活性;MTT法观察细胞存活率及激光共聚焦检测细胞内钙离子浓度的变化。实验揭示urantide (3~30 μg·kg-1)可明显抑制小鼠心电图ST段的抬高; urantide (10及30 μg·kg-1)可显著降低小鼠血清中LDH活性和MDA含量,明显升高NOS活性和NO含量,同时减轻异丙肾上腺素诱导的心肌病理损伤。在乳鼠原代心肌细胞缺氧再给氧模型中, urantide (1×10-6和1×10-7 mol·L-1)能明显增加细胞存活率,降低培养上清液中LDH的活性; urantide (1×10-6~1×10-9 mol·L-1)能显著降低细胞培养上清液中cTnI的增高和细胞内钙离子浓度的上升。上述结果表明, urantide对心肌缺血及缺氧再给氧所致心肌细胞的损伤具有一定的保护作用,其作用机制可能与增加NOS活性、促进NO合成及抑制钙超载有关。  相似文献   

15.
In reconstructive surgery, skeletal muscle may endure protracted ischemia before reperfusion, which can lead to significant ischemia/reperfusion injury. Ischemic postconditioning induced by brief cycles of reperfusion/reocclusion at the end of ischemia has been shown to salvage skeletal muscle from ischemia/reperfusion injury in several animal models. However, ischemic postconditioning has not been confirmed in human skeletal muscle. Using an established in vitro human skeletal muscle hypoxic conditioning model, we tested our hypothesis that hypoxic postconditioning salvages ex vivo human skeletal muscle from hypoxia/reoxygenation injury and the mechanism involves inhibition of opening of the mitochondrial permeability transition pore (mPTP) and preservation of ATP synthesis. Muscle strips (~0.5×0.5×15mm) from human rectus abdominis muscle biopsies were cultured in Krebs-Henseleit-HEPES buffer, bubbled with 95%N(2)/5%CO(2) (hypoxia) or 95%O(2)/5%CO(2) (reoxygenation). Samples were subjected to 3h hypoxia/2h reoxygenation. Hypoxic postconditioning was induced by one or two cycles of 5min reoxygenation/5min hypoxia after 3h hypoxia. Muscle injury, viability and ATP synthesis after 2h of reoxygenation were assessed by measuring lactate dehydrogenase (LDH) release, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) reduction and ATP content, respectively. Hypoxic postconditioning or treatment with the mPTP-opening inhibitors Cyclosporine A (CsA, 5×10(-6)M) or N-Methyl-4-isoleucine Cyclosporine (NIM811, 5×10(-6)M) 10min before reoxygenation decreased LDH release, increased MTT reduction and increased muscle ATP content (n=7 patients; P<0.05). Conversely, treatment with the mPTP opener Atractyloside (5×10(-6)M) 10min before hypoxic postconditioning abolished its protective effect (n=7 patients; P<0.05). We conclude that hypoxic postconditioning effectively salvages human skeletal muscle from hypoxia/reoxygenation injury by inhibition of mPTP opening and preservation of ATP synthesis during reoxygenation.  相似文献   

16.
Beneficial effects of n-acetylcysteine on ischaemic brain injury   总被引:4,自引:0,他引:4  
1. Nitric oxide (NO), peroxynitrite, formed from NO and superoxide anion, poly (ADP-ribole) synthetase have been implicated as mediators of neuronal damage following focal ischaemia. Here we have investigated the effects of n-acetylcysteine (NAC) treatment in Mongolian gerbils subjected to cerebral ischaemia. 2. Treatment of gerbils with NAC (20 mg kg(-1) 30 min before reperfusion and 1, 2 and 6 h after reperfusion) reduced the formation of post-ischaemic brain oedema, evaluated by water content. 3. NAC also attenuated the increase in the brain levels of malondialdehyde (MDA) and the increase in the hippocampus of myeloperoxidase (MPO) caused by cerebral ischaemia. 4. Positive staining for nitrotyrosine was found in the hippocampus in Mongolian gerbils subjected to cerebral ischaemia. Hippocampus tissue sections from Mongolian gerbils subjected to cerebral ischaemia also showed positive staining for poly (ADP-ribose) synthetase (PARS). The degree of staining for nitrotyrosine and for PARS were markedly reduced in tissue sections obtained from animals that received NAC. 5. NAC treatment increased survival and reduced hyperactivity linked to neurodegeneration induced by cerebral ischaemia and reperfusion. 6. Histological observations of the pyramidal layer of CA1 showed a reduction of neuronal loss in animals that received NAC. 7. These results show that NAC improves brain injury induced by transient cerebral ischaemia.  相似文献   

17.
The organophosphate insecticide mevinphos (Mev) acts on the rostral ventrolateral medulla (RVLM), where sympathetic vasomotor tone originates, to elicit phasic cardiovascular responses via nitric oxide (NO) generated by NO synthase (NOS) I and II. We evaluated the contribution of soluble guanylyl cyclase (sGC)/cyclic guanosine monophosphate (cGMP)/protein kinase G (PKG) cascade and peroxynitrite in this process. PKG expression in ventrolateral medulla of Sprague-Dawley rats manifested an increase during the sympathoexcitatory phase (Phase I) of cardiovascular responses induced by microinjection of Mev bilaterally into the RVLM that was antagonized by co-administration of 7-nitroindazole or Nomega-propyl-L-arginine, two selective NOS I inhibitors or 1-H-[1,2,4]oxadiaolo[4,3-a]quinoxalin-1-one (ODQ), a selective sGC antagonist. Co-microinjection of ODQ or two PKG inhibitors, KT5823 or Rp-8-Br-cGMPS, also blunted the Mev-elicited sympathoexcitatory effects. However, the increase in nitrotyrosine, a marker for peroxynitrite, and the sympathoinhibitory circulatory actions during Phase II Mev intoxication were antagonized by co-administration of S-methylisothiourea, a selective NOS II inhibitor, Mn(III)-tetrakis-(4-benzoic acid) porphyrin, a superoxide dismutase mimetic, 5,10,15,20-tetrakis-N-methyl-4'-pyridyl)-porphyrinato iron (III), a peroxynitrite decomposition catalyst, or L-cysteine, a peroxynitrite scavenger. We conclude that sGC/cGMP/PKG cascade and peroxynitrite formation may participate in Mev-induced phasic cardiovascular responses as signals downstream to NO generated respectively by NOS I and II in the RVLM.  相似文献   

18.
钴原卟啉对H9c2心肌细胞缺氧/复氧损伤的保护作用   总被引:3,自引:2,他引:1  
目的研究钴原卟啉(COPP)预处理在H9c2心肌细胞缺氧/复氧损伤中的作用及分子机制。方法建立H9C2心肌细胞缺氧/复氧模型。在H9c2心肌细胞缺氧/复氧前用COPP预处理,并用锌原卟啉(Znpp),全反视黄酸(ATAR)分别抑制HO-1及Nrf2-ARE。检测细胞上清液中LDH、CK的水平变化;用RT-PCR法分析HO-1mRNA表达水平;Westernblot分析HO-1、Nrf2的蛋白表达水平。结果与缺氧/复氧组比,COPP预处理组中LDH和CK水平均明显降低,而HO-1mRNA水平,蛋白水平及细胞核的Nrf2蛋白水平均明显增加;Znpp的加入阻断了COPP预处理对心肌细胞的保护作用;ATAR的加入抑制了Nrf2在细胞核的聚集,进而抑制了COPP对HO-1的诱导。结论COPP预处理诱导H9c2心肌细胞HO-1过表达,具有抗心肌细胞缺氧/复氧损伤的保护作用;其机制与Nrf2-ARE信号通路相关。  相似文献   

19.
目的研究吡那地尔(pinacidil,Pin)、美托洛尔(metoprolol,Met)、谷氨酰胺(glutamine,Glu)、胰岛素(insu-lin,Ins)4药联用对缺氧/复氧(hypoxia/reoxygenation,H/R)所致H9c2心肌细胞损伤的抗凋亡保护作用并探讨其作用机制。方法将培养的H9c2心肌细胞随机分为7组,即①正常对照(Con)组;②缺氧/复氧(H/R)组;③吡那地尔(Pin)组;④美托洛尔(Met)组;⑤谷氨酰胺(Glu)组;⑥胰岛素(Ins)组;⑦吡那地尔、美托洛尔、谷氨酰胺和胰岛素4药联用(PMGI)组。细胞经药物处理建立H/R损伤模型,并测定各组H9c2心肌细胞的存活率;收集培养液测定乳酸脱氢酶(lactate dehydrogenase,LDH)活性;流式细胞仪检测心肌细胞凋亡百分率;罗丹明123(Rhodamine123,Rh123)荧光探针标记线粒体,检测荧光强度以反映心肌细胞线粒体膜电位(mitochondrial membrane potential,△Ψm)变化。结果对于H/R损伤的H9c2心肌细胞,吡那地尔、美托洛尔、谷氨酰胺与胰岛素4药联用组与药物单用组或H/R组相比能明显提高细胞存活率,保护其细胞膜,减少LDH渗漏;减少细胞凋亡率;提高△Ψm。结论吡那地尔、美托洛尔、谷氨酰胺和胰岛素4药联用后,通过不同途径保护心肌细胞,达到协同抗凋亡作用。  相似文献   

20.
Livers isolated from 18 hours fasted rats were subjected to N(2) hypoxia (for 45 min) followed by reoxygenation (for 45 min). The perfusion medium used was Krebs-Henseleit bicarbonate buffer (KHBB, pH 7.4). Lactate and alanine were added as gluconeogenic and ureagenic substrates and Trolox C was also added to perfusate. Oxygen consumption, lactate dehydrogenase (LDH), alanine transaminase (ALT), total glutathione, oxidized glutathione, bile flow, glucose and urea were measured. After hypoxia oxygen consumption significantly dropped but Trolox C had no influence on this decrease. ALT and LDH were significantly increased by hypoxia/reoxygenation. This increase was markedly attenuated in the presence of Trolox C. The total glutathione and oxidized glutathione efflux increased following hypoxia, which were prevented by the treatment of Trolox C. Bile flow rate decreased following hypoxia/reoxygenation but did not continue to decrease in the reoxygenation phase by Trolox C. Following hypoxia/reoxygenation glucose and urea releases decreased. Trolox C had no influence on inhibition of glucose and urea production. These results suggest that Trolox C protected the liver cells against hypoxia/reoxygenation injury, yielding further evidence for a causative role of oxidative stress in this model.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号