scholarly journals Protection of renal tissue from ischemia-reperfusion injury by inhibition of Fas-induced apoptosis

2007 ◽  
Vol 27 (2) ◽  
pp. 124-129
Author(s):  
Manae S. Kurokawa ◽  
Zhiyun Ozaki-Chen ◽  
Hideshi Yoshikawa ◽  
Noboru Suzuki
2014 ◽  
Vol 115 (suppl_1) ◽  
Author(s):  
Chi K Lam ◽  
Wen Zhao ◽  
Wenfeng Cai ◽  
Guansheng Liu ◽  
Phil Bidwell ◽  
...  

Sarcoplasmic reticulum (SR) calcium handling is central not only in the control of heart function during excitation-contraction coupling but also in mitochondrial energetics and apoptosis. Recent studies have identified the anti-apoptotic protein, HS-1 associated protein X-1 (HAX-1) as a novel regulator of SR calcium cycling. Although HAX-1 has been shown to localize to mitochondria in various tissues, we found out that it also localizes to SR through its interaction with phospholamban (PLN) in cardiac muscle. Acute or chronic overexpression of HAX-1 in cardiomyocytes promoted PLN inhibition on the calcium ATPase (SERCA) and decreased cardiomyocyte calcium kinetics and contractile parameters. Accordingly, ablation of HAX-1 significantly enhanced SERCA activity and calcium kinetics. Furthermore, the HAX-1/PLN interaction appeared to also regulate cardiomyocyte survival. Indeed, overexpression of HAX-1 and the associated depressed SR Ca-load attenuated endoplasmic reticulum stress induced apoptosis, as evidenced by reduction of both caspase-12 activation and pro-apoptotic transcription factor C/EBP homologous protein induction during ischemia/reperfusion injury. In addition, the depressed SR Ca-cycling by HAX-1 overexpression was associated with reduced mitochondrial Ca-load as reflected by: a) hyper-phosphorylation of pyruvate dehydrogenase (PDH) and decreases in its activity, to diminish ATP production consistent with the attenuated energetic demand in these hearts; and b) reduced levels of reactive oxygen species, indicating protection from oxidative damage and preserved mitochondrial integrity. These findings suggest that HAX-1 is a key regulator of Ca-cycling, apoptosis and energetics in the heart. Thus, decreases in HAX-1 levels, observed during ischemia/reperfusion injury, may contribute to the deteriorated function and progression to heart failure development.


2020 ◽  
Author(s):  
Chengyu Mao ◽  
Dongjiu Li ◽  
En Zhou ◽  
Erhe Gao ◽  
Tiantian Zhang ◽  
...  

Abstract BackgroundExosomes derived from adipose-derived mesenchymal stem cells can potentially protect cardiomyocytes from myocardial ischemia reperfusion injury. It's notable that exosomes derived from adipose-derived mesenchymal stem cells underwent anoxia preconditioning showed a better cardioprotective effect than that without anoxia. Here, in vitro and in vivo studies were used to investigate the cardioprotective effects against myocardial ischemia reperfusion injury of exosomes derived from adipose-derived mesenchymal stem cells with (Int-EXO) or without anoxia (NC-EXO), respectively. Methods: Adipose-derived mesenchymal stem cells and exosomes were identified by western blot, flow cytometry, transmission electron microscopy, and nanosight. An exosome tracer assay identified exosomes absorbed by cells. An in vitro model using mice cardiomyocytes for studying anoxia-reoxygenation and an in vivo mice model of MIRI were used to investigate the cardioprotective effects of NC-Exo and Int-Exo, respectively.ResultsWe discovered that treatment with NC-EXO and Int-EXO significantly reduced the infarct size and attenuated cardiomyocyte apoptosis, In addition, Int-EXO group had a less infarct size and apoptosis degree. The mechanism revealed by RNA sequencing showed that 40 miRNAs were up-regulated in Int-EXO compared to NC-EXO. 10 of these miRNAs could bind thioredoxin-interacting protein as a downstream target gene; among these, the top-discrepant miRNA224-5p was selected for further study. Dual luciferase reporter assay and rescue study verified TXNIP as a target gene for miR-224-5p. Furthermore, the cellular death signaling pathway which Int-EXO involved in mediating was in a special form of apoptosis, not pyroptosis, induced by activated thioredoxin-interacting protein-pyroptosis-caspase1 pathway in gasdermin D-deficient cells. ConclusionThe research demonstrated adipose-derived mesenchymal stem cells exosomes attenuated MIRI by inhibiting pyroptosis-induced apoptosis in cardiomyocytes which are lack of gasdermin D. The cardioprotective effect of Int-EXO was more significant than that of NC-EXO, possibly due to treated with anoxia preconditioning, adipose-derived mesenchymal stem cells product more miRNAs targeting thioredoxin-interacting protein in exosomes to alleviate pyroptosis-induced apoptosis. These findings provide new insights into the pathogenesis and methods for intervention of myocardial ischemia reperfusion injury.


2015 ◽  
Vol 117 (suppl_1) ◽  
Author(s):  
Dongchao Lv ◽  
Shengguang Ding ◽  
Ping Chen ◽  
Yihua Bei ◽  
Chongjun Zhong ◽  
...  

Ischemia-reperfusion injury (IRI) following acute myocardial infarction (AMI) has no effective treatment and a poor prognosis. microRNA (miRNA)-19b is a key functional member of miRNA-19-72 cluster family, regulating cellular proliferation, apoptosis, differentiation, and metabolism. Dysregulation of the miR-19b cluster is critically involved in a spectrum of cardiovascular diseases. However, the role of miR-19b in myocardial IRI is unknown. In this study, we found that miR-19b was downregulated in a mouse model of IRI. Meanwhile, about 50% downregulation of miR-19b was detected in H2O2-treated H9C2 cells mimicking myocardial IRI. We also found that overexpression of miR-19b decreased H2O2-induced apoptosis (36.02%±3.92% vs 29.34%±0.79% in nc-mimics vs miR-19b-mimics, respectively) and necrosis (23.11%±1.64% vs 18.76%±0.71% in nc-mimics vs miR-19b-mimics, respectively), and increased proliferation of H9C2 cells in vitro, while downregulation of miR-19b had reverse effects. Furthermore, PTEN, a previously validated target gene of miR-19b, has been found to be negatively regulated by miR-19b at protein levels in H9C2 cells. These data reveal the potential of miR-19b as a therapeutic target for myocardial IRI.


2019 ◽  
Vol 24 (44) ◽  
pp. 5334-5341 ◽  
Author(s):  
Xiaofei Jiang ◽  
Kar-Sheng Lew ◽  
Qiying Chen ◽  
Arthur M. Richards ◽  
Peipei Wang

Background: Human mesenchymal stem cell-derived exosomes (hMSC-Exo) have been shown to reduce ischemia/reperfusion injury (I/R) in multiple models. I/R-induced apoptosis or autophagy play important roles in cell death. However, little or no reports demonstrate any roles of hMSC-Exo in this regards. Objective: To test the hypothesis that the inhibition of I/R-induced apoptosis and autophagy play a pivotal role in the cardioprotection of hMSC-Exo. Methods: Myoblast H9c2 cells and isolated rat hearts underwent hypoxia/re-oxygenate (H/R) or ischemia/ reperfusion (I/R) respectively. H9c2 were treated with 1.0 μg/ml Exo, in comparison with 3-MA or rapamycin (Rapa), a known anti- or pro-autophagic agent respectively. Hearts were treated with 0.5, 1.0 and 2.0 μg/ml Exo for 20 min in the beginning of reperfusion. Cell viability, WST assay, LDH release, Annexin-V staining apoptosis assay and GFP-LC3 labeled autophagosomes formation, cardiac function and Western blot were measured. Results: Exo significantly reduced H/R injury as indicated by increased cell viability and reduced LDH and apoptosis. 3-MA, while Rapa, showed increased or decreased protective effects. Rapa-induced injury was partially blocked by Exo. Exo decreased LC3-II/I ratio and increased p62, inhibited autophagosome formation, an indication of autophagy inhibition. In isolated heart, Exo increased cardiac functional recovery and reduced LDH release in I/R. Bcl-2 was significantly upregulated by Exo but not 3-MA. Exo downregulated Traf6 and upregulated mTORC1/p-4eBP1. Conclusion: Exo reduce I/R-induced apoptosis and autophagy. Up-regulation of Bcl-2 is the cross-talk between these two processes. The down-regulation of Traf6 and activation of mTORC1 are additional mechanisms in the inhibition of apoptosis and autophagy.


Sign in / Sign up

Export Citation Format

Share Document