scholarly journals The pulmonary microvasculature entraps induced vascular progenitor cells (iVPCs) systemically delivered after cardiac ischemia-reperfusion injury: Indication for preservation of heart function via paracrine effects beyond engraftment

2018 ◽  
Vol 26 (2) ◽  
pp. e12493 ◽  
Author(s):  
Melanie Ziegler ◽  
Katharina Haigh ◽  
Thao Nguyen ◽  
Xiaowei Wang ◽  
Bock Lim ◽  
...  
2020 ◽  
Author(s):  
Emily Dookun ◽  
Anna Walaszczyk ◽  
Rachael Redgrave ◽  
Pawel Palmowski ◽  
Simon Tual-Chalot ◽  
...  

AbstractA key component of cardiac ischemia-reperfusion injury (IRI) is the increased generation of reactive oxygen species, leading to enhanced inflammation and tissue dysfunction in patients following intervention for myocardial infarction. In this study we hypothesized that oxidative stress, due to ischemia-reperfusion, induces senescence which contributes to the pathophysiology of cardiac IRI. We demonstrate that IRI induces cellular senescence in both cardiomyocytes and interstitial cell populations and treatment with the senolytic drug navitoclax after ischemia-reperfusion improves left ventricular function, increases myocardial vascularization, and decreases scar size. SWATH-MS based proteomics reveal that biological processes associated with fibrosis and inflammation, that were increased following ischemia-reperfusion, were attenuated upon senescent cell clearance. Furthermore, navitoclax treatment reduced the expression of proinflammatory, profibrotic and anti-angiogenic cytokines, including interferon gamma-induced protein-10, TGF-β3, interleukin-11, interleukin-16 and fractalkine. Our study provides proof-of-concept evidence that cellular senescence contributes to impaired heart function and adverse remodeling following cardiac ischemia-reperfusion. We also establish that post-IRI senescent cells play a considerable role in the inflammatory response. Subsequently, senolytic treatment, at a clinically feasible time point, attenuates multiple components of this response and improves clinically important parameters. Thus, cellular senescence represents a potential novel therapeutic avenue to improve patient outcomes following cardiac ischemia-reperfusion.


2004 ◽  
Vol 43 (5) ◽  
pp. A533
Author(s):  
Alok S Pachori ◽  
Luis G Melo ◽  
Lunan Zhang ◽  
Richard E Pratt ◽  
Victor J Dzau

ACS Nano ◽  
2014 ◽  
Vol 9 (1) ◽  
pp. 279-289 ◽  
Author(s):  
Naviin Hardy ◽  
Helena M. Viola ◽  
Victoria P. A. Johnstone ◽  
Tristan D. Clemons ◽  
Henrietta Cserne Szappanos ◽  
...  

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.


2008 ◽  
Vol 49 (5) ◽  
pp. 735 ◽  
Author(s):  
Hakan Ceyran ◽  
Figen Narin ◽  
Nazmi Narin ◽  
Hülya Akgün ◽  
A. Bahar Ceyran ◽  
...  

PLoS ONE ◽  
2017 ◽  
Vol 12 (3) ◽  
pp. e0173657 ◽  
Author(s):  
Janine C. Deddens ◽  
Dries A. Feyen ◽  
Peter-Paul Zwetsloot ◽  
Maike A. Brans ◽  
Sailay Siddiqi ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document