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 187 毫秒
1.
近年来,血红素氧合酶-1/一氧化碳(HO-1/CO)体系对各脏器保护作用的研究较为广泛.HO-1/CO体系几乎分布于所有的组织和器官,除血红素外,在缺血、缺氧、炎症、应激、感染性休克、机械性损伤、重金属和细胞因子等因素诱导下其活性可显著增强,对多种脏器起保护作用.本文就HO-1/CO体系在多个系统、器官的脏器保护作用做一综述. HO-1/CO的生物学特性 血红素氧合酶(HO)是血红素代谢过程中的限速酶,在血红素降解代谢的第一个限速步骤起催化作用,它降解血红素产生一氧化碳(CO)、胆绿素(biliverdin)和Fe2+.这个反应的三种产物全部具有抗氧化、抗炎、抗凋亡和抗增殖的作用.  相似文献   

2.
血红素加氧酶-1诱导对鼠肝缺血再灌注损伤的保护作用   总被引:1,自引:0,他引:1  
目的研究血红素加氧酶-1(heme oxygenase-1,HO-1)在鼠肝缺血再灌注损伤肝组织中的表达及其作用。方法建立小鼠部分肝脏热缺血再灌注损伤模型,36只清洁级Balb/C小鼠随机分为3组: 假手术组(S组)、缺血/再灌注损伤组(I/R组)、HO-1诱导剂氯化高铁血红素(hemin)预处理组(HM组)。免疫组化半定量分析肝组织HO-1蛋白的表达,检测血清AST和ALT,肝组织丙二醛(MDA)含量及超氧化物歧化酶(SOD)活性,并观察肝组织的病理变化。结果与S组比较,I/R组HO-1蛋白表达显著增强,hemin预处理后,HO-1蛋白表达较I/R组增高(P<0.01)。I/R组AST,ALT活性和MDA的含量显著高于S组,而HM组均显著低于I/R组(P<0.01);I/R组SOD活性下降,而HM组显著高于I/R组(P<0.01)。HM组病理损伤程度明显轻于I/R组。结论 HO-1在鼠肝缺血再灌注损伤肝组织中表达上调,对肝脏具有保护效应。  相似文献   

3.
感染性休克引起的急性肺损伤( ALI)是临床常见的急危重症之一,发病率和病死率较高,其发病机制及防治措施已成为国内外学者研究的重点.血红素氧合酶(heme oxygenase,HO)是催化血红素生成一氧化碳(CO)、胆绿素和铁离子的一种限速酶[1].已有研究表明,HO-1/CO对感染性休克肺损伤有保护作用[2,3].本文就HO-1/CO系统在感染性休克肺损伤中的保护机制进行综述.  相似文献   

4.
器官缺血/再灌注(ischemia/reperfusion,I/R)损伤是一种复杂的、多因素参与、非抗原依赖性炎症反应,对器官功能的早期和远期影响以及患者的生命安全有重要的影响,也是目前阻碍器官移植发展的一道难题.传统的理论认为器官I/R损伤主要是固有性免疫系统参与的,如巨噬细胞、中性粒细胞等.但是在近几年的研究中,人们发现适应性免疫系统在器官I/R损伤中也发挥着重要的作用,尤其是T淋巴细胞.T淋巴细胞主要在再灌注后1 h内浸润,通过分泌因子和调节其他炎症细胞浸润引起器官I/R损伤.  相似文献   

5.
目的 探讨人参皂甙Rb1对肠缺血/再灌注致急性肺损伤的保护效应及核因子NF-E2相关因子2(Nrf2)/血红素氧合酶-1(HO-1)通路参与该效应的分子机制.方法 成年雄性C57BL/6J小鼠随机分为5组:假手术组(S组);肠缺血/再灌注组(I/R组);再灌注+Rb1组(I/R +Rb1组);全反式维甲酸(ATRA)+再灌注组(ATRA+ I/R组);ATRA+再灌注+Rb1组(ATRA+ I/R+Rb1组).采用肠缺血/再灌注模型,Western blot检测肺组织Nrf2、HO-1表达变化;酶联免疫吸附试验(ELISA)法检测肺组织肿瘤坏死因子(TNF)-α、白细胞介素(IL)-6、IL-10水平;检测超氧化物歧化酶(SOD)活性及丙二醛(MDA)含量;检测肺湿/干比及肺组织病理损伤评分.结果 与S组比较,其他4组Nrf2、HO-1蛋白表达,TNF-α、IL-6、MDA含量,肺组织湿/干重比及肺组织病理评分增高(P<0.05);与1/R组比较,1/R+ Rb1组Nrf2,HO-1蛋白表达,TNF-α,IL-6,MDA含量,肺组织湿/干重比及肺组织病理评分降低(P<0.05);与I/R+ Rb1组比较,ATRA+ I/R组,ATRA+ I/R+ Rb1组Nrf2、HO-1蛋白表达,NF-α、IL-6、MDA含量,肺组织湿/干重比及肺组织病理评分增高(P<0.05).SOD活性、IL-10水平与上述变化相反.结论 肠缺血/再灌注可引起急性肺损伤,人参皂甙Rb1后处理能通过激活Nrf2/HO-1通路减轻肠缺血/再灌注所致肺损伤.  相似文献   

6.
血红素氧合酶(hemeoxygenase,HO)广泛存在于人类及哺乳动物体内,近年来发现它不仅可降解血红素,而且能调节体内许多生理和病理过程,进而保护机体组织器官.体内HO有3种存在形式,即HO-1、HO-2、HO-3.在氧化应激过程中,HO-1在肾小管损伤和肾血管损伤中起重要的保护作用.本文就近年来HO-1与肾脏疾病的研究进展作一综述.  相似文献   

7.
缺血再灌注损伤(I/R)是恢复某些缺血组织器官的血液灌流以及氧供后, 组织损伤不但没有减轻、反而进一步加重的病理过程。心、脑、肺、肾、肝等重要器官I/R发生率高, 致死率高, 严重影响人类健康。三结构域蛋白(TRIM)具有共同的氨基末端三方结构域排列-RING-Bbox1/2-卷曲螺旋(RBCC), 调节各种生物过程。本文系统地总结了TRIM家族蛋白的分类与功能以及其在器官缺血再灌注损伤中的作用机制, 为I/R的诊治提供理论依据。  相似文献   

8.
目的 探讨内源性一氧化碳 (CO)在大鼠肢体缺血再灌注 (I/ R)致远隔多器官氧化性损伤中的作用机制。方法 将 6 4只大鼠随机分为 4组 :假手术 (Sham )组 ;Sham 特异性血红素氧化酶阻断剂—锌原卟啉 (Zn PP)组 ;肢体缺血 2小时和再灌注 4小时 (I/ R)组 ;I/ R Zn PP组。测定各组心、肺、肝和肾组织匀浆中丙二醛 (MDA )含量、超氧化物歧化酶 (SOD)活性及血液内碳氧血红蛋白 (COHb)的变化 ,观察动物 2 4小时存活率。结果 与 Sham组相比 ,I/ R组各脏器 MDA含量及血液内 COHb水平均显著增高 ,组织中 SOD活性和动物 2 4小时存活率显著降低 ,有统计学意义(P<0 .0 5 ) ;I/ R Zn PP组与 I/ R组相比各脏器 MDA含量进一步增高 ,血液内 COHb水平、组织中 SOD活性和动物的2 4小时存活率显著降低 ,也有统计学意义 (P<0 .0 5 )。结论 肢体缺血再灌注可导致多器官的氧化性损伤 ,并使 CO产生增多 ,后者在大鼠抗缺血再灌注所致的远隔多器官损伤中具有重要作用。  相似文献   

9.
目的 研究胱硫醚β-合酶(CBS)/硫化氢(H2S)体系和血红素氧合酶-1(HO-1)/一氧化碳(CO)体系在大鼠脑缺血再灌注损伤中的作用。方法 30只Wistar大鼠随机分为5组(n=6):对照组(C组)、脑缺血再灌注组(I/R组)、脑缺血再灌注+锌原卟啉(HO-1抑制剂)组(I/R+Z组)、脑缺血再灌注+羟氨(CBS抑制剂)组(I/R+H组)、脑缺血再灌注+锌原卟啉+羟氨组(I/R+Z+H组)。采用四血管阻断法建立全脑缺血再灌注损伤模型,I/R+Z组、I/R+H组和I/R+Z+H组夹闭两侧颈总动脉前30min分别腹腔注射锌原卟啉45/zmol/kg、羟氨5mmol/L、羟氨5mmol/L+锌原卟啉45/maol/kg1ml,C、I/R组给予等量生理盐水。再灌注6h时处死大鼠,取海马,测定海马组织H2S、CO、还原型谷胱甘肽(GSH)、丙二醛(MDA)、超氧化物歧化酶(SOD)水平及CBSmRNA和HO-1mRNA的表达;电镜下观察海马线粒体变性情况。结果 与C组比较,I/R组海马组织CO、H2S、CBSmRNA和HO-1mRNA、MDA水平及线粒体变性率均升高,海马组织SOD、GSH水平降低(P〈0.01);与I/R组比较,I/R+Z组海马组织CO、HO-1mRNA水平降低,海马组织H2S、CBSmRNA、GSH、MDA水平升高,I/R+H组海马组织CO、HO-1mRNA、MDA水平升高,H2S、CBSmRNA、GSH水平降低;I/R+Z+H组海马组织CO、RS、HO-1mRNA和CBSmRNA、SOD、GSH水平降低,线粒体变性率升高(P〈0.05)。结论 CBS/H1S体系和HO-1/CO体系可拮抗大鼠脑缺血再灌注损伤,其作用可相互代偿。  相似文献   

10.
背景 沉默信息调节蛋白1 (silent information regulator protein 1,Sirt1)是尼克酰胺腺嘌呤二核苷酸(nicotinamide adenine dinucleotide,NAD)依赖的第3类组蛋白/非组蛋白去乙酰酶,参与细胞内多种生物功能的调节.Sirt1不仅在抗衰老中发挥重要作用,同样在心肌缺血/再灌注( ischemia/reperfusion,I/R)时保护受损细胞.目的 对Sirt1在心肌I/R中的保护机制以及几种激活Sirt1活性的方法进行综述. 内容 Sirt1能使活性氧清除剂产生增加、抗心肌细胞凋亡、促进细胞内自噬、减轻炎症反应、促进胞内正常线粒体合成等而减轻I/R的心肌细胞损伤.通过多种方法提高I/R时Sirt1的活性,是减轻损伤的重要措施. 趋向 随着Sin1在心肌I/R中的作用机制不断被揭示,其将成为缓解心肌I/R损伤的重要靶点.  相似文献   

11.
BACKGROUND: Heme oxygenase (HO)-1 is induced as a unique stress response and leads to a transient resistance against oxidative damage, including ischemia and reperfusion (I/R) injury. In the present study, we examined whether HO-1 induction may confer a protection against I/R injury in the rat kidney. METHODS: Lewis rats were divided into four groups as follows: (1) vehicle group; (2) group treated with ferri-protoporphyrin IX (hemin), an inducer of HO; (3) group treated with low-dose tin-protoporphyrin IX (SnPP), an inhibitor of HO; and (4) group treated with high-dose SnPP. Renal warm ischemia for 60 minutes was performed 24 hours after each treatment. RESULTS: At 24 hours after treatment, hemin induced a significant increase in renal HO activity, but failed to induce HO-1 protein synthesis. Although both low- and high-dose SnPP reduced HO activity, a marked HO-1 expression was observed only in the high-dose SnPP-treated kidney. Hemin exacerbated the renal function after reperfusion, while high-dose SnPP significantly suppressed the intercellular adhesion molecule (ICAM)-1 expression, the infiltration of ED-1-positive macrophages and the expression of activated caspase-3, which resulted in attenuation of apoptotic cell death and ameliorated I/R injury. CONCLUSION: These results suggest that prior induction of HO-1 protein by high-dose SnPP may lead to anti-inflammatory and antiapoptotic effects on warm renal I/R injury independently of its enzyme activity, and that HO enzyme activation may not always act as an antioxidant, especially under I/R-induced oxidative stress.  相似文献   

12.
目的 评价细胞穿透肽PEP-1介导血红素加氧酶-1(HO-1)对大鼠肠缺血再灌注损伤的影响.方法 雄性SD大鼠18只,周龄7~9周,体重210~260 g,采用随机数字表法,将大鼠随机分为3组(n=6):假手术组(S组)、肠缺血再灌注组(IR组)和融合蛋白PEP-1/HO-1+肠缺血再灌注组(HO组).采用夹闭肠系膜上动脉45 min,恢复灌注120 min的方法制备大鼠肠缺血再灌注损伤模型.HO组夹闭肠系膜上动脉前30 min,左侧髂静脉注射融合蛋白PEP-1/HO-1 0.5 mg,S组不夹闭肠系膜上动脉,余操作同IR组.于再灌注120 min时处死大鼠取小肠组织,称重后计算肠湿/干重比,测定丙二醛(MDA)含量、超氧化物歧化酶(SOD)活性和HO-1活性,免疫组化法检测肠组织HO-1蛋白的表达,光镜下观察肠组织结构并进行损伤评分.结果 与S组比较,IR组和HO组肠湿/干重比和MDA含量升高,SOD活性降低,HO-1活性和蛋白表达水平升高,损伤评分升高(P<0.05);与IR组比较,HO组肠湿/干重比、MDA含量降低,SOD活性升高,HO-1活性和蛋白表达水平升高,损伤评分降低(P<0.05).HO组大鼠肠组织病理学损伤较IR组减轻.结论 细胞穿透肽PEP-1可将HO-1成功导人大鼠肠组织中的细胞并减轻肠缺血再灌注损伤.
Abstract:
Objective To investigate the effects of heme oxygenase-1 (HO-1) mediated by cell penetrating peptide PEP-1 on intestinal ischemia/reperfusion (I/R) injuiy in tats. Methods Eighteen male SD rats aged 7-9 weeks weighing 210-260 g were randomly divided into 3 groups (re = 6 each): sham operation group (group S) , I/R group and PEP-1/HO-1 + I/R group (group HO) . To establish a model of intestinal I/R injury, intestines were exteriorized and the superior mesenteric artery was exposed and occluded for 45 min ischemia, and then the clamp was removed for 120 min reperfusion. The PEP-1/HO-1 fusion protein 0.5 mg was injected via the left iliac vein 30 min prior to ischemia in group HO. The superior mesenteric artery was exposed but not occluded in group S. At the end of reperfusion, the rats were sacrificed and intestinal tissues obtained to determine the intestinal wet/ dry ratio, malondialdehyde (MDA) level, activities of superoxide dismutase (SOD) and HO-1, and HO-1 protein expression. The histological changes in the intestinal mucosa were examined and the injuiy was scored. Results Compared with group S, the intestinal wet/dry ratio, MDA level, HO-1 activity, HO-1 protein expression and injury score were significantly increased, while the SOD activity was significantly decreased in groups I/R and HO ( P < 0.05) . Compared with group I/R, the intestinal wet/dry ratio, MDA level and injury score were significantly decreased, while the SOD activity, HO-1 activity and HO-1 protein expression increased in group HO ( P < 0.05) . The pathologic changes were significantly attenuated in group HO compared with group I/R.Conclusion HO-1 protein can be successfully delivered into intestinal tissues by PEP-1 and has protective effects against intestinal I/R injury.  相似文献   

13.
This study investigated the role of heme oxygenase (HO)-1 in the cardiac tissue injury of acute ischemia/reperfusion (I/R) in diabetic streptozotocin (STZ)-induced hyperglycemic rats. The effects of 1) hemin, an inducer of HO expression and activity, and 2) zinc protoporphyrin IX (ZnPP-IX), an inhibitor of HO activity, have also been investigated on the tissue injury by I/R and some mediators released in these circumstances. STZ hyperglycemic rats had impaired levels of HO-1 within the cardiac tissue and increased myocardial infarct size (IS) following I/R, as compared with the nondiabetic rats. In these rats, administration of hemin 4 mg/kg 18 h before I/R increases the levels of HO-1 within the tissue. However, the values of HO-1 assayed in these circumstances were significantly lower (P < 0.01) than those assayed in nondiabetic animals subjected to the same procedures; IS was much more extended (P < 0.01) than in the parent nondiabetic group. STZ hyperglycemic rats also predisposed the heart to produce high levels of the cytokines interleukin (IL)-1beta and CXCL8. Subsequent I/R further increased (P < 0.01) the cytokine production, an effect partly prevented by hemin treatment. This recovered the huge number of infiltrated polymorphonuclear (PMN) leukocytes within the cardiac tissue associated with the STZ hyperglycemic state and I/R damage.  相似文献   

14.
BACKGROUND: Expression of inducible heme-oxygenase (HO-1) has been shown to be increased in various inflammatory disorders, which may confer a protective role. The aim of our study was to assess pulmonary expression of HO-1 after ischemia/reperfusion (I/R) of the lower limbs in rats. MATERIALS AND METHODS: We compared three groups of rats (n = 5/group): one Sham group, and two I/R groups (aorta cross-clamped for 2 h followed by 2 h of reperfusion), one of which pre-treated with Zn-protoporphyrin (Zn-PP), a competitive inhibitor of HO (50 micromol/kg, i.p.). At the end of experiment, lungs were harvested for determination of HO activity and HO-1 expression by Western blot and immunohistochemistry. Lung injury was assessed by bronchoalveolar lavage, histological study, and determination of the lung Evans Blue dye content, an index of microvascular permeability. RESULTS: I/R of the lower limbs was responsible for acute lung injury (ALI), characterized by neutrophilic infiltration (87 +/- 20 x 10(3) neutrophils/mm(3), Sham group versus 191 +/- 38 x 10(3) neutrophils/mm(3), I/R group; P < 0.002) and an increase in lung Evans blue dye content: (74 +/- 6 microg/g, Sham group versus 122 +/- 48 microg/g, I/R group; P < 0.05). Pre-treatment with Zn-PP further increases the Evans Blue content (122 +/- 48 microg/g, I/R group versus 179 +/- 23 microg/g Zn-PP group P < 0.05) and the neutrophilic infiltration. Pulmonary heme-oxygenase activity, and HO-1 content were increased after I/R. (10.5 +/- 12 pmol bilirubin/mg protein/h, Sham group versus 101.2 +/- 66 pmol bilirubin/mg protein/h, I/R group; P < 0.02). Immunohistochemistry revealed that the expression of HO-1 was mainly localized to inflammatory cells. CONCLUSIONS: ALI following I/R of the lower limbs in rats is associated with an increase of pulmonary expression of HO-1, inhibition of this expression increase the severity of ALI.  相似文献   

15.
Lai IR  Ma MC  Chen CF  Chang KJ 《Transplantation》2004,77(7):1004-1008
BACKGROUND: Hypoxic preconditioning (HP) confers cytoprotection against ischemia/reperfusion (I/R) injury. This effect is in part attributable to the induction of heme oxygenase (HO)-1. This experiment evaluates liver cell damage after I/R injury in HP rats. METHODS: HP rats were prepared by exposure (15 hr/day) to an altitude chamber (5500 m) for 2 weeks. Partial hepatic ischemia was produced in the left lobes for 45 min followed by 180 min of reperfusion. Zinc (Zn) protoporphyrin (PP), a specific inhibitor of HO enzymatic activity, was subcutaneously injected 1 hr before the I/R injury into separate groups of sea-level (SL) control and HP rats. Serum alanine aminotransferase (ALT) levels, liver HO-1 mRNA and protein, and HO enzymatic activity were measured. RESULTS: HO-1 was induced in the livers of rats exposed to HP. The levels of HO-1 mRNA and protein were obviously overexpressed after 2 weeks of HP. HP diminished the elevation of serum ALT levels after I/R injury (83.7+/- 4.9 U/L) when compared with SL controls (280.8+/-19.4 U/L) and HP+ZnPP-pretreated groups (151.3+/-4.4 U/L). The HO activity in treated rats also was correlated with these results (237.9+/-19.8 pmol/mg of protein per hour for the HP group, 164.3+/-12.7 pmol/mg of protein per hour for the HP+ZnPP group, and 182.6+/-8 pmol/mg of protein per hour for the SL controls). CONCLUSIONS: The authors' results indicated that the induction of HO-1 in hypoxic preconditioning played a protective role against hepatic I/R injury.  相似文献   

16.
Upregulation of heme oxygenase (HO)-1, a heat shock protein 32, protects against hepatic ischemia/reperfusion (I/R) injury. Activation of "innate" toll-like receptor (TLR) 4 system triggers the I/R injury cascade. This study explores cytoprotective functions of HO-1 overexpression following exogenous administration of cobalt protoporphyrin (CoPP), and its relationship with the TLR4 pathway in a model of mouse partial hepatic warm I/R injury. CoPP treatment markedly improved hepatic function and histology, and suppressed pro-inflammatory cytokine elaboration profile, as compared with untreated controls. Although administration of CoPP did not affect intrahepatic TLR4, it downregulated IFN-inducible protein 10 (IP-10) expression. As IP-10 is the major product of type-1 IFN pathway downstream of TLR4, we then infused recombinant IFN-beta (rIFN-beta) directly into mouse livers. Interestingly, infusion of rIFN-beta upregulated hepatic IP-10 expression. In contrast, adjunctive CoPP treatment decreased IP-10 levels in mouse livers infused with rIFN-beta. Thus, CoPP-induced HO-1 upregulation suppresses type-1 IFN pathway downstream of TLR4 system in hepatic warm I/R injury model.  相似文献   

17.
Lai IR  Chang KJ  Chen CF  Tsai HW 《Transplantation》2006,81(9):1311-1317
BACKGROUND: We have reported the protective role of heme oxygenase-1 (HO-1) in the mechanism of hypoxic preconditioning. We wish to investigate the role of HO-1 in remote preconditioning (RP) against hepatic ischemia/reperfusion (I/R) injury in rats. METHODS: The remote preconditioning was produced by four cycles of 10-min ischemia-reperfusion of the hind limb of rats. Partial hepatic ischemia was produced in the left lobes for 45 min followed by 240 min of reperfusion. Zinc-protoporphyrin IX (ZnPP), a specific inhibitor of HO enzymatic activity, was intra-peritoneally injected 1 hr before the ischemia-reperfusion injury in separate groups of RP rats. Serum alanine transaminase (ALT) levels, expression of hepatic HO-1 protein and mRNA, immunohistochemical staining and HO enzymatic activity were measured. RESULTS: HO-1 was induced in the livers of rats 4 hr after the RP stimuli, and the overexpression persisted for 24 hr. Immunohistochemical staining demonstrated induction of HO-1 in the hepatocytes. The peripheral lymphocytes did not express HO-1 after RP. RP diminished the elevation of serum ALT levels 4 hr after I/R injury (283.7+/-167.4 U L) when compared with controls (1297.7+/-729.3 U L) and RP+ ZnPP pretreated groups (1429.9+/-750.9 U L). The heme oxygenase activity in treated rats also correlated these results (286.8+/-34.3 pmol mg protein hr for the RP group, 156.3+/-27.5 pmol mg protein hr for the RP+ ZnPP pretreated group, and 170.6+/-19.4 pmol mg protein hr for the control group, 144.8+/-7.8 pmol mg protein hr for the control+ ZnPP pretreated group). CONCLUSION: Our results indicated that the induction of HO-1 in remote preconditioning played a protective role against hepatic I/R injury.  相似文献   

18.
BACKGROUND: We have shown that both intraischemic hypothermia and hypertonic saline resuscitation provide dramatic protection against gut ischemia/reperfusion (I/R) injury that is in part mediated by heme oxygenase-1 (HO-1). We therefore hypothesized that induction of HO-1 by hemin would lessen damage and improve function after gut I/R. MATERIALS AND METHODS: Male Sprague-Dawley rats were treated with 50 micromol/kg hemin (HO-1 inducer ferric protoporphyrin IX chloride) sq or vehicle 2 h before superior mesenteric artery occlusion for 60 min or sham laparotomy. After 6 h of reperfusion, transit was determined by quantitation of percentage of tracer in 10 equal segments of small intestine 30 min following injection into the duodenum (expressed as mean geometric center). Ileum was harvested for assessment of mucosal histologic injury (Chiu score 0-5 by blinded observer), myeloperoxidase activity (MPO, index of inflammation), and HO-1 protein expression. RESULTS: Hemin treatment was associated with increased HO-1 protein expression, lessened mucosal injury, decreased MPO activity, and improved intestinal transit following gut I/R. CONCLUSION: These data corroborate that HO-1 plays an important role in protecting the gut against I/R-induced injury.  相似文献   

19.
BACKGROUND: Heme oxygenase (HO)-1 system has been shown to provide protection against oxidative stress through the degradation of heme to biliverdin, free iron, and carbon monoxide (CO). This study investigated cytoprotective efficacy of CO at a low concentration on cold ischemia/reperfusion (I/R) injury of transplanted intestine. METHODS: Lewis rat recipients of syngenic orthotopic small intestinal transplantation with 6 hours UW cold preservation were either kept in room air (air-treated control) or exposed to CO (250 ppm) for 1 hour before and 24 hours after surgery. RESULTS: In air-treated grafts, mRNA levels for interleukin-6, intracellular adhesion molecule-1, cyclooxygenase-2, and inducible nitric oxide synthase promptly increased. Sequential histopathologic analysis of untreated grafts revealed initial rapid epithelial loss, subsequent recruitment of inflammatory infiltrates, and local hemorrhage in the lamina propria, which extended downward to the epithelial crypt and muscle layer with time. CO effectively blocked proinflammatory cascade during I/R injury, inhibited upregulation of inflammatory molecules and ameliorated intestinal tissue injuries. Beneficial effects of CO were associated with improved graft blood flow without inhibiting endogenous HO-1 activity. Recipient animal survival was significantly improved with CO to 100% versus 58% in air-treated controls. CONCLUSIONS: These results indicate a significant role for CO in protecting the intestine from cold I/R injury associating with small intestinal transplantation.  相似文献   

20.
PURPOSE: Gut ischemia/reperfusion (I/R) elicits an inflammatory response that impairs intestinal transit. We have previously shown that regional intraischemic hypothermia (IH) protects against moderate gut I/R-induced mucosal injury, is associated with decreased NF-kappaB activity and inducible nitric oxide synthase induction and preserves heme oxygenase-1 (HO-1) expression. HO-1 provides cytoprotection in various models of oxidant stress. We, therefore, tested the hypothesis that IH protects against gut I/R-induced impaired intestinal transit via HO-1 induction. MATERIALS AND METHODS: At laparotomy (lap), Sprague-Dawley rats had duodenal catheters placed followed by sham or gut I/R (superior mesenteric artery occlusion for 75 min) with or without regional IH (15 degrees C). Each animal was placed on a heating blanket maintaining systemic normothermia (37 degrees C). At 12 or 24 h of reperfusion, small intestinal transit was determined by quantitating the distribution of a tracer (FITC dextran) in the intestine 30 min after instillation (expressed as geometric center of distribution). Ileal samples were obtained for histology and HO-1 expression, assessed by Western immunoblot at 12 and 24 h of reperfusion. In separate experiments, rats were pretreated with an HO-1 inhibitor Sn protoporphyrin IX (25 mumol/kg, ip), 1 h before superior mesenteric artery occlusion and transit measured as above. RESULTS: Rats treated with I/R had increased histological injury and impaired intestinal transit at both 12 and 24 h compared with sham. Rats treated with I/R+IH exhibited histological injury and transit comparable with sham controls. I/R induced HO-1 expression at 12 and 24 h of reperfusion and IH augmented this I/R-induced HO-1 expression. Sn protoporphyrin IX abrogated IH protection against histological injury and impaired transit. CONCLUSION: We conclude that intraischemic regional hypothermia protects against histological injury and impaired intestinal transit caused by severe gut I/R injury. Hypothermic protection under these conditions is in part due to HO-1 expression.  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