Reactive oxygen species mediate modification of glycocalyx during ischemia-reperfusion injury

2006 ◽  
Vol 290 (6) ◽  
pp. H2247-H2256 ◽  
Author(s):  
Ivan Rubio-Gayosso ◽  
Steven H. Platts ◽  
Brian R. Duling

The glycocalyx (Gcx) is a complex and poorly understood structure covering the luminal surface of endothelial cells. It is known to be a determinant of vascular rheology and permeability and may be a key control site for the vascular injuries caused by ischemia-reperfusion (I/R). We used intravital-microscopy to evaluate the effects of I/R injury on two properties of Gcx in mouse cremasteric microvessels: exclusion of macromolecules (anionic-dextrans) and intracapillary distribution of red blood cells (RBC). In this model, the Gcx is rapidly modified by I/R injury with an increase in 70-kDa anionic-dextran penetration without measurable effect on the penetration of 580-kDa anionic-dextran or on RBC exclusion. The effects of I/R injury appear to be mediated by the rapid production of reactive oxygen species (ROS) because they are ameliorated by the addition of exogenous superoxide dismutase-catalase. Intravenous application of allopurinol or heparin also inhibited the effects of I/R injury, and we interpret efficacy of allopurinol as evidence for a role for xanthine-oxidoreductase (XOR) in the response to I/R injury. Heparin, which is hypothesized to displace XOR from a heparin-binding domain in the Gcx, reduced the effects of I/R. The effects of I/R injury were also partially prevented or fully reversed by the intravascular infusion of exogenous hyaluronan. These data demonstrate: 1) the liability of Gcx during I/R injury; 2) the importance of locally produced ROS in the injury to Gcx; and 3) the potential importance of heparin-binding sites in modulating the ROS production. Our findings further highlight the relations between glycosaminoglycans and the pathophysiology of Gcx in vivo.

2021 ◽  
Vol 2021 ◽  
pp. 1-11
Author(s):  
Yaqian Weng ◽  
Hui Li ◽  
Lin Gao ◽  
Wenjing Guo ◽  
Shiyuan Xu ◽  
...  

The main objective of this study was to investigate the diurnal differences in Period 2 (PER2) expression in myocardial ischemia-reperfusion (I/R) injury. We investigated diurnal variations in oxidative stress and energy metabolism after myocardial I/R in vitro and in vivo. In addition, we also analyzed the effects of H2O2 treatment and serum shock in H9c2 cells transfected with silencing RNA against Per2 (siRNA-Per2) in vitro. We used C57BL/6 male mice to construct a model of I/R injury at zeitgeber time (ZT) 2 and ZT14 by synchronizing the circadian rhythms. Our in vivo analysis demonstrated that there were diurnal differences in the severity of injury caused by myocardial infarctions, with more injury occurring in the daytime. PER2 was significantly reduced in heart tissue in the daytime and was higher at night. Our results also showed that more severe injury of mitochondrial function in daytime produced more reactive oxygen species (ROS) and less ATP, which increased myocardial injury. In vitro, our findings presented a similar trend showing that apoptosis of H9c2 cells was increased when PER2 expression was lower. Meanwhile, downregulation of PER2 disrupted the oxidative balance by increasing ROS and mitochondrial injury. The result was a reduction in ATP and failure to provide sufficient energy protection for cardiomyocytes.


2018 ◽  
Vol 49 (6) ◽  
pp. 2320-2332 ◽  
Author(s):  
Guo Zu ◽  
Tingting Zhou ◽  
Ningwei Che ◽  
Xiangwen Zhang

Background/Aims: Ischemia-reperfusion (I/R) adversely affects the intestinal mucosa. The major mechanisms of I/R are the generation of reactive oxygen species (ROS) and apoptosis. Salvianolic acid A (SalA) is suggested to be an effective antioxidative and antiapoptotic agent in numerous pathological injuries. The present study investigated the protective role of SalA in I/R of the intestine. Methods: Adult male Sprague-Dawley rats were subjected to intestinal I/R injury in vivo. In vitro experiments were performed in IEC-6 cells subjected to hypoxia/ reoxygenation (H/R) stimulation to simulate intestinal I/R. TNF-α, IL-1β, and IL-6 levels were measured using enzyme-linked immunosorbent assay. Malondialdehyde and myeloperoxidase and glutathione peroxidase levels were measured using biochemical analysis. Apoptosis was measured by terminal deoxynucleotidyl transferase mediated dUTP nick-end labeling staining or flow cytometry in vivo and in vitro. The level of reactive oxygen species (ROS) was measured by dichlorodihydrofluorescin diacetate (DCFH-DA) staining. Western blotting was performed to determine the expression of heme oxygenase-1 (HO-1), Nrf2 and proteins associated with apoptosis. The mRNA expressions of Nrf2 and HO-1 were detected by quantitative real-time polymerase chain reaction in vivo and in vitro. Results: Malondialdehyde level and myeloperoxidase and glutathione peroxidase, TNF-α, IL-1β, and IL-6 levels group in intestinal tissue decreased significantly in the SalA pretreatment groups compared to the I/R group. SalA markedly abolished intestinal injury compared to the I/R group. SalA significantly attenuated apoptosis and increased Nrf2/HO-1 expression in vivo and in vitro. However, Nrf2 siRNA treatment partially abrogated the above mentioned effects of SalA in H/R-induced ROS and apoptosis in IEC-6 cells. Conclusion: The present study demonstrated that SalA ameliorated oxidation, inhibited the release of pro-inflammatory cytokines and alleviated apoptosis in I/R-induced injury and that these protective effects may partially occur via regulation of the Nrf2/ HO-1 pathways.


2019 ◽  
Vol 11 (3) ◽  
pp. 292-297 ◽  
Author(s):  
Jonathan E. Palmer ◽  
Breanna M. Brietske ◽  
Tyler C. Bate ◽  
Erik A. Blackwood ◽  
Manasa Garg ◽  
...  

2019 ◽  
Vol 317 (1) ◽  
pp. H156-H163 ◽  
Author(s):  
Aleksandra Stamenkovic ◽  
Grant N. Pierce ◽  
Amir Ravandi

Cell death is an important component of the pathophysiology of any disease. Myocardial disease is no exception. Understanding how and why cells die, particularly in the heart where cardiomyocyte regeneration is limited at best, becomes a critical area of study. Ferroptosis is a recently described form of nonapoptotic cell death. It is an iron-mediated form of cell death that occurs because of accumulation of lipid peroxidation products. Reactive oxygen species and iron-mediated phospholipid peroxidation is a hallmark of ferroptosis. To date, ferroptosis has been shown to be involved in cell death associated with Alzheimer’s disease, Huntington’s disease, cancer, Parkinson’s disease, and kidney degradation. Myocardial reperfusion injury is characterized by iron deposition as well as reactive oxygen species production. These conditions, therefore, favor the induction of ferroptosis. Currently there is no available treatment for reperfusion injury, which accounts for up to 50% of the final infarct size. This review will summarize the evidence that ferroptosis can induce cardiomyocyte death following reperfusion injury and the potential for this knowledge to open new therapeutic approaches for myocardial ischemia-reperfusion injury.


2017 ◽  
Vol 2017 ◽  
pp. 1-7 ◽  
Author(s):  
Si-Ming Wei ◽  
Yu-Min Huang ◽  
Jian Zhou

This study investigated the effect of probucol, a potent antioxidant, on testicular torsion/detorsion-induced ischemia/reperfusion injury attributable to excess reactive oxygen species released by neutrophils. Sixty male Sprague-Dawley rats were randomly divided into sham-operated control, ischemia-reperfusion, and probucol-treated groups. In the ischemia-reperfusion group, testicular detorsion was performed after 2 hours of left testicular torsion. In the probucol-treated group, after performing the same surgical procedures as in the ischemia-reperfusion group, probucol was given intraperitoneally at testicular detorsion. Orchiectomy was performed to evaluate protein expression of E-selectin which is an endothelial cell adhesion molecule and mediates neutrophil adhesion to vascular endothelium, myeloperoxidase activity (a mark of neutrophil accumulation in the testis), malondialdehyde level (an indicator of reactive oxygen species), and spermatogenesis. E-selectin protein expression, myeloperoxidase activity, and malondialdehyde level were significantly increased, and testicular spermatogenesis was significantly decreased in the ipsilateral testes in the ischemia-reperfusion group, compared with the control group. The probucol-treated group showed significant decreases in E-selectin protein expression, myeloperoxidase activity, and malondialdehyde level and significant increase in testicular spermatogenesis in the ipsilateral testes, compared with the ischemia-reperfusion group. These findings indicate that probucol can protect testicular spermatogenesis by reducing overgeneration of reactive oxygen species by inhibiting E-selectin protein expression and neutrophil accumulation in the testis.


1997 ◽  
Vol 272 (5) ◽  
pp. L897-L902 ◽  
Author(s):  
J. J. Zulueta ◽  
R. Sawhney ◽  
F. S. Yu ◽  
C. C. Cote ◽  
P. M. Hassoun

Reactive oxygen species (ROS) play an important role in the pathogenesis of ischemia-reperfusion injury. Extracellular H2O2 generation from bovine pulmonary artery endothelial cells (EC) is known to increase in response to anoxia-reoxygenation (A-R). To determine potential sources of intracellular ROS formation in EC in response to A-R, a fluorometric assay based on the oxidation of 2',7'-dichlorofluorescin was used. Intracellular ROS production declined 40% during 6 h of anoxia (P < 0.05). After A-R, the rates of intracellular ROS formation increased to 148 +/- 9% (P < 0.001) that of normoxic EC (100 +/- 3%). In EC exposed to A-R, allopurinol and NG-methyl-L-arginine (L-NMMA), inhibitors of xanthine oxidase (XO) and nitric oxide synthase (NOS), respectively, reduced intracellular ROS formation by 25 +/- 1% (P < 0.001) and 36 +/- 4% (P < 0.01). Furthermore, at low doses (i.e., 20 microM), deferoxamine and diethylenetriaminepentaacetic acid (DTPA) significantly inhibited intracellular ROS formation. However, at 100 microM, only deferoxamine caused further reduction in DCF fluorescence. In summary, EC respond to A-R by generating increased amounts of XO- and NOS-derived intracellular ROS. The inhibition, to a similar extent, caused by allopurinol and L-NMMA, as well as the effect of deferoxamine and DTPA suggest that the ROS detected is peroxynitrite. Based on these findings and previous work, we conclude that EC generate ROS in response to A-R from at least two different sources: a plasma membrane-bound NADPH oxidase-like enzyme that releases H2O2 extracellularly and XO, which generates intracellular O2-, which in turn may react with nitric oxide to form peroxynitrite.


Sign in / Sign up

Export Citation Format

Share Document