• Title/Summary/Keyword: ischemic injury

검색결과 424건 처리시간 0.029초

Matrix Metalloproteinase Inhibitors Attenuate Neuroinflammation Following Focal Cerebral Ischemia in Mice

  • Park, Cheol-Hong;Shin, Tae-Kyeong;Lee, Ho-Youn;Kim, So-Jung;Lee, Won-Suk
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제15권2호
    • /
    • pp.115-122
    • /
    • 2011
  • The aim of this study was to investigate whether matrix metalloproteinase (MMP) inhibitors attenuate neuroinflammation in an ischemic brain following photothrombotic cortical ischemia in mice. Male C57BL/6 mice were anesthetized, and Rose Bengal was systemically administered. Permanent focal ischemia was induced in the medial frontal and somatosensory cortices by irradiating the skull with cold white light. MMP inhibitors, such as doxycycline, minocycline, and batimastat, significantly reduced the cerebral infarct size, and the expressions of monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-${\alpha}$ (TNF-${\alpha}$), and indoleamine 2,3-dioxygenase (IDO). However, they had no effect on the expressions of heme oxygenase-1 and neuroglobin in the ischemic cortex. These results suggest that MMP inhibitors attenuate ischemic brain injury by decreasing the expression levels of MCP-1, TNF-${\alpha}$, and IDO, thereby providing a therapeutic benefit against cerebral ischemia.

NO 억제제가 허혈전처치의 심장 보호효과에 미치는 영향 (Effect of Inhibitor of Nitric Oxide Synthesis on the Ischemic Reconditioning in Isolated Heart of Rat.)

  • 유호진;조은용
    • Journal of Chest Surgery
    • /
    • 제29권8호
    • /
    • pp.807-815
    • /
    • 1996
  • 허혈전처치(ischemic preconditioniiIE)의 허혈심장 보호효과와 그 기전을 규명하기 위한 일환으로 citric oxide(HO)가 허혈전처치의 심보호 효과에 미치는 영향을 검토하였다. 흰쥐 적출심장의 Langendorrr관류표본에서 실험적인 허할(30분)-재관류(30분) 손상을 유도하였고, 허혈전처치는 재관류손상 유도 전에 5분 허혈 - 5분 재관류를 3회 반복하여 시행하였다. 허혈심근 손상의 지표로 심수축기능 세질효소 유출 및 미세형태학적 변화를, 그리고 HO 합성 억제제인 L-HAME 를 투여하여 허혈전처치와 비전처치 허혈-재관류 심장들에서 손상의 정도를 비교하였다. 그 결과 허혈- 재관류 심장에서 심기능의 저하및 세포질 유출이 현저하게 증가하였고 전자현미경상의 미세구조에서도 세포내 소기관 및 myofibril의 파괴가 관찰되 어 심근손상이 심함을 알 수 있었다. 허 혈-재관류에 의한 심 장손상은 허혈전처치를 시행한 허혈-재관류 심장에서는 현격하게 감소돼 심회복률이 77%로 증가하였 고 세포질유출도 현저하게 감소되었으며 미세소견에서도 세포구조가 비교적 잘 보존되었다. 허혈전처 치에 의한 심보호 효과에 NO가 관여하는지를 관찰하기 위하여 NO합성 억제제인 L-NAME를 투여하 여 허혈전처치를 시행하였다. 결과 L-UAME투여로 허혈전처치에 의하여 회복된 심기능 및 LDH유출 감소에 아무런 영향을 주지 않았고 허혈전처치에 의하여 비교적 잘 보존된 미세구조 역시 영향을 받지 않았다. 이상의 결과들로부터 허혈전처치는 세포수준에서 허혈심근의 재관류손상을 방지하며, NO합성의 증가가 횐쥐 적출 심장에서 허혈전처치에 의한 허혈심장 보호효과에 크게 기여하지 않을 것으로 사료되었다.

  • PDF

Apple pectin, a dietary fiber, ameliorates myocardial injury by inhibiting apoptosis in a rat model of ischemia/reperfusion

  • Lim, Sun Ha;Kim, Mi Young;Lee, Jongwon
    • Nutrition Research and Practice
    • /
    • 제8권4호
    • /
    • pp.391-397
    • /
    • 2014
  • BACKGROUND/OBJECTIVE: Myocardial cell death due to occlusion of the coronary arteries leads to myocardial infarction, a subset of coronary heart disease (CHD). Dietary fiber is known to be associated with a reduced risk of CHD, the underlying mechanisms of which were suggested to delay the onset of occlusion by ameliorating risk factors. In this study, we tested a hypothesis that a beneficial role of dietary fiber could arise from protection of myocardial cells against ischemic injury, manifested after occlusion of the arteries. MATERIALS/METHODS: Three days after rats were fed apple pectin (AP) (with 10, 40, 100, and 400 mg/kg/day), myocardial ischemic injury was induced by 30 min-ligation of the left anterior descending coronary artery, followed by 3 hr-reperfusion. The area at risk and infarct area were evaluated using Evans blue dye and 2,3,5-triphenyltetrazolium chloride (TTC) staining, respectively. DNA nicks reflecting the extent of myocardial apoptosis were assessed by TUNEL assay. Levels of cleaved caspase-3, Bcl-2, and Bax were assessed by immunohistochemistry. RESULTS: Supplementation of AP (with 100 and 400 mg/kg/day) resulted in significantly attenuated infarct size (IS) (ratio of infarct area to area at risk) by 21.9 and 22.4%, respectively, in the AP-treated group, compared with that in the control group. This attenuation in IS showed correlation with improvement in biomarkers involved in the apoptotic cascades: reduction of apoptotic cells, inhibition of conversion of procaspase-3 to caspase-3, and increase of Bcl-2/Bax ratio, a determinant of cell fate. CONCLUSIONS: The findings indicate that supplementation of AP results in amelioration of myocardial infarction by inhibition of apoptosis. Thus, the current study suggests that intake of dietary fiber reduces the risk of CHD, not only by blocking steps leading to occlusion, but also by protecting against ischemic injury caused by occlusion of the arteries.

흰쥐를 이용한 심근경색모델에서 진피(秦皮)의 심장손상 보호효과 (Protective Effect of Cortex Fraxini on Heart Injury in a Rat Model of Myocardial Infarction)

  • 임선하;이종원
    • 대한본초학회지
    • /
    • 제26권4호
    • /
    • pp.149-154
    • /
    • 2011
  • Objectives : Myocardial infarction is caused by heart cell death in a region where coronary arteries supplying blood to the region are occluded. In the present study, we determined whether ethanol extract of Cortex fraxini (HY5053) could attenuate heart injury by inhibiting apoptosis. Methods : Improvement of survival of HepG2 cells, a human hepatocellular carcinoma cell line, and reduction of apoptosis under hypoxic conditions (3% $O_2$) were assessed by trypan blue staining and DNA fragmentation assay, respectively. To assess the impact of HY5053 on the heart injury, Sprague-Dawley rats underwent 1 day of the left anterior descending coronary artery occlusion. HY5053 was given by intraperitoneal injection (200 mg/kg) 1 hr prior to the occlusion. Subsequently, the heart were harvested, excised into 4 slices, and the slices were stained with 2,3,5-triphenyl tetrazolium chloride. Finally, the extent of heart injury represented as ischemic index (%) was assessed. Results : Addition of HY5053 (400 ${\mu}g$/mL) into the culture medium for 1 day under ischemic conditions improved the cell survival by 50%, compared with control (0 ${\mu}g$/mL), consequently delayed apoptosis in 6 hr difference. Also, HY5053 (200 mg/kg) reduced the ischemic index by 44%, compared with control (0 mg/kg). Conclusions : These findings suggested that HY5053 attenuated myocardial infarction by inhibiting apoptosis. Thus, Cortex fraxini could be developed as a novel cardioprotectant to complement a currently available treatment, coronary angioplasty.

Curcumin targets vascular endothelial growth factor via activating the PI3K/Akt signaling pathway and improves brain hypoxic-ischemic injury in neonatal rats

  • Li, Jia;An, Yan;Wang, Jia-Ning;Yin, Xiao-Ping;Zhou, Huan;Wang, Yong-Sheng
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제24권5호
    • /
    • pp.423-431
    • /
    • 2020
  • This study aimed to evaluate the effect of curcumin on brain hypoxic-ischemic (HI) damage in neonatal rats and whether the phosphoinositide 3-kinase (PI3K)/Akt/vascular endothelial growth factor (VEGF) signaling pathway is involved. Brain HI damage models were established in neonatal rats, which received the following treatments: curcumin by intraperitoneal injection before injury, insulin-like growth factor 1 (IGF-1) by subcutaneous injection after injury, and VEGF by intracerebroventricular injection after injury. This was followed by neurological evaluation, hemodynamic measurements, histopathological assessment, TUNEL assay, flow cytometry, and western blotting to assess the expression of p-PI3K, PI3K, p-Akt, Akt, and VEGF. Compared with rats that underwent sham operation, rats with brain HI damage showed remarkably increased neurological deficits, reduced right blood flow volume, elevated blood viscosity and haematocrit, and aggravated cell damage and apoptosis; these injuries were significantly improved by curcumin pretreatment. Meanwhile, brain HI damage induced the overexpression of p-PI3K, p-Akt, and VEGF, while curcumin pretreatment inhibited the expression of these proteins. In addition, IGF-1 treatment rescued the curcumin-induced down-regulated expression of p-PI3K, p-Akt, and VEGF, and VEGF overexpression counteracted the inhibitory effect of curcumin on brain HI damage. Overall, pretreatment with curcumin protected against brain HI damage by targeting VEGF via the PI3K/Akt signaling pathway in neonatal rats.

Ex Vivo Lung Perfusion of Cardiac-death Donor Lung in Pigs

  • Paik, Hyo Chae;Haam, Seok Jin;Park, Moo Suk;Song, Joo Han
    • 대한이식학회지
    • /
    • 제28권3호
    • /
    • pp.154-159
    • /
    • 2014
  • Background: Lung transplantation (LTx) is a life-saving treatment for patients with end-stage lung disease; however, the shortage of donor lungs has been a major limiting factor to increasing the number of LTx. Growing experience following LTx using donor lungs after cardiac death (DCD) has been promising, although concerns remain. The purpose of this study was to develop a DCD lung harvest model using an ex vivo lung perfusion (EVLP) system and to assess the function of presumably damaged lungs harvested from the DCD donor in pigs. Methods: The 40 kg pigs were randomly divided into the control group with no ischemic lung injury (n=5) and the study group (n=5), which had 1 hour of warm ischemic lung injury after cardiac arrest. Harvested lungs were placed in the EVLP circuit and oxygen capacities (OC), pulmonary vascular resistance (PVR), and peak airway pressure (PAP) were evaluated every hour for 4 hours. At the end of EVLP, specimens were excised for pathologic review and wet/dry ratio. Results: No statistically significant difference in OC (P=0.353), PVR (P=0.951), and PAP (P=0.651) was observed in both groups. Lung injury severity score (control group vs. study group: 0.700±0.303 vs. 0.870±0.130; P=0.230) and wet/dry ratio (control group vs. study group: 5.89±0.97 vs. 6.20±0.57; P=0.560) also showed no statistically significant difference between the groups. Conclusions: The function of DCD lungs assessed using EVLP showed no difference from that of control lungs without ischemic injury; therefore, utilization of DCD lungs can be a new option to decrease the number of deaths on the waiting list.

Dexmedetomidine alleviates blood-brain barrier disruption in rats after cerebral ischemia-reperfusion by suppressing JNK and p38 MAPK signaling

  • Canmin Zhu;Dili Wang;Chang Chang;Aofei Liu;Ji Zhou;Ting Yang;Yuanfeng Jiang;Xia Li;Weijian Jiang
    • The Korean Journal of Physiology and Pharmacology
    • /
    • 제28권3호
    • /
    • pp.239-252
    • /
    • 2024
  • Dexmedetomidine displays multiple mechanisms of neuroprotection in ameliorating ischemic brain injury. In this study, we explored the beneficial effects of dexmedetomidine on blood-brain barrier (BBB) integrity and neuroinflammation in cerebral ischemia/reperfusion injury. Sprague-Dawley rats were subjected to middle cerebral artery occlusion (MCAO) for 1.5 h and reperfusion for 24 h to establish a rat model of cerebral ischemia/reperfusion injury. Dexmedetomidine (9 ㎍/kg) was administered to rats 30 min after MCAO through intravenous injection, and SB203580 (a p38 MAPK inhibitor, 200 ㎍/kg) was injected intraperitoneally 30 min before MCAO. Brain damages were evaluated by 2,3,5-triphenyltetrazolium chloride staining, hematoxylin-eosin staining, Nissl staining, and brain water content assessment. BBB permeability was examined by Evans blue staining. Expression levels of claudin-5, zonula occludens-1, occludin, and matrix metalloproteinase-9 (MMP-9) as well as M1/M2 phenotypes-associated markers were assessed using immunofluorescence, RT-qPCR, Western blotting, and gelatin zymography. Enzyme-linked immunosorbent assay was used to examine inflammatory cytokine levels. We found that dexmedetomidine or SB203580 attenuated infarct volume, brain edema, BBB permeability, and neuroinflammation, and promoted M2 microglial polarization after cerebral ischemia/reperfusion injury. Increased MMP-9 activity by ischemia/reperfusion injury was inhibited by dexmedetomidine or SB203580. Dexmedetomidine inhibited the activation of the ERK, JNK, and p38 MAPK pathways. Moreover, activation of JNK or p38 MAPK reversed the protective effects of dexmedetomidine against ischemic brain injury. Overall, dexmedetomidine ameliorated brain injury by alleviating BBB permeability and promoting M2 polarization in experimental cerebral ischemia/reperfusion injury model by inhibiting the activation of JNK and p38 MAPK pathways.