Abstract

The release of extracellular vesicles (EVs) is increased under cellular stress and cardiomyocyte damaging conditions. However, whether the cardiomyocyte-derived EVs eventually reach the systemic circulation and whether their number in the bloodstream reflects cardiac injury, remains unknown. Wild type C57B/6 and conditional transgenic mice expressing green fluorescent protein (GFP) by cardiomyocytes were studied in lipopolysaccharide (LPS)-induced systemic inflammatory response syndrome (SIRS). EVs were separated both from platelet-free plasma and from the conditioned medium of isolated cardiomyocytes of the left ventricular wall. Size distribution and concentration of the released particles were determined by Nanoparticle Tracking Analysis. The presence of GFP + cardiomyocyte-derived circulating EVs was monitored by flow cytometry and cardiac function was assessed by echocardiography. In LPS-treated mice, systemic inflammation and the consequent cardiomyopathy were verified by elevated plasma levels of TNFα, GDF-15, and cardiac troponin I, and by a decrease in the ejection fraction. Furthermore, we demonstrated elevated levels of circulating small- and medium-sized EVs in the LPS-injected mice. Importantly, we detected GFP+ cardiomyocyte-derived EVs in the circulation of control mice, and the number of these circulating GFP+ vesicles increased significantly upon intraperitoneal LPS administration (P = 0.029). The cardiomyocyte-derived GFP+ EVs were also positive for intravesicular troponin I (cTnI) and muscle-associated glycogen phosphorylase (PYGM). This is the first direct demonstration that cardiomyocyte-derived EVs are present in the circulation and that the increased number of cardiac-derived EVs in the blood reflects cardiac injury in LPS-induced systemic inflammation (SIRS).

Highlights

  • MethodsCardiovascular diseases represent the number one cause of death worldwide

  • We used mTmG (B6;129-Gt(ROSA)26Sortm11(CAGtdTomato*,GFP*)Nat/J) indicator mice (Jackson Laboratories), engineered to constitutively express a conditional tdTomato transgene that converts to green fluorescent protein (GFP) expression following exposure to Cre recombinase

  • We isolated mEVs from the conditioned media of cultured cardiomyocytes and characterized them by flow cytometry (FACSCalibur, Becton Dickinson). We found that both clusterin and PYGM were present in cardiomyocyte EVs (CMEVs), and the fluorescence intensity of mEVs immunostained for PYGM increased significantly (P = 0.0024, T-test) upon in vivo LPS injection of mice (Fig. 5)

Read more

Summary

Introduction

Cardiovascular diseases represent the number one cause of death worldwide. Despite major research efforts focusing on cardiovascular diseases, there is still a demand for early and sensitive diagnostic markers for myocardial injury. Extracellular vesicles (EVs) are subcellular, phospholipidbilayer enclosed particles present both in tissues and biological fluids [1]. Outstanding interest has been attracted by EVs as a potential next-generation biomarker. EVs have been shown to cross biological barriers and serve as promising complex diagnostic and prognostic tools [2]. The guidelines of the International Society for Extracellular Vesicles (MISEV2018) recommended referring to EVs either by their physical or molecular characteristics given that in most studies, the precise biogenesis route of EVs is not investigated and confirmed [3]

Objectives
Results
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.