TY - JOUR T1 - Endothelium-protective, histone-neutralizing properties of the polyanionic agent defibrotide JF - medRxiv DO - 10.1101/2021.02.21.21252160 SP - 2021.02.21.21252160 AU - Hui Shi AU - Alex A. Gandhi AU - Stephanie A. Smith AU - Diane Chiang AU - Srilakshmi Yalavarthi AU - Ramadan A. Ali AU - Chao Liu AU - Gautam Sule AU - Pei-Suen Tsou AU - Yu Zuo AU - Yogendra Kanthi AU - James H. Morrissey AU - Jason S. Knight Y1 - 2021/01/01 UR - http://medrxiv.org/content/early/2021/02/28/2021.02.21.21252160.abstract N2 - Neutrophil-mediated activation and injury of the endothelium play a role in the pathogenesis of diverse disease states ranging from autoimmunity to cancer to COVID-19. Neutralization of cationic proteins (such as neutrophil extracellular trap/NET-derived histones) with polyanionic compounds has been suggested as a potential strategy for protecting the endothelium from such insults. Here, we report that the FDA-approved polyanionic agent defibrotide (a pleotropic mixture of oligonucleotides) directly engages histones and thereby blocks their pathological effects on endothelium. In vitro, defibrotide counteracted endothelial cell activation and cell death, whether triggered by purified NETs, COVID-19 serum containing high levels of NETs, or recombinant histone H4. In vivo, defibrotide stabilized the endothelium and protected against histone-accelerated inferior vena cava thrombosis in mice. Mechanistically, defibrotide demonstrated direct and tight binding to histone H4 as detected by both electrophoretic mobility shift assay and surface plasmon resonance. Taken together, these data provide insights into the potential role of polyanionic compounds in protecting the endothelium from thromboinflammation with potential implications for myriad NET- and histone-accelerated disease states.Competing Interest StatementThe authors have declared no competing interest.Funding StatementYZ was supported by a career development grant from the Rheumatology Research Foundation. YK was supported by the Intramural Research Program of the NIH and NHLBI, Lasker Foundation, NIH (K08HL131993, R01HL150392), Falk Medical Research Trust Catalyst Award, and the JOBST-American Venous Forum Award. JHM and SAS were supported by the NIH (R35 HL135823). JSK was additionally supported by grants from the NIH (R01HL115138), Burroughs Wellcome Fund, Rheumatology Research Foundation, and Lupus Research Alliance. The work was also partially supported by a grant from Jazz Pharmaceuticals, which did not participate in study design or data analysis.   Author DeclarationsI confirm all relevant ethical guidelines have been followed, and any necessary IRB and/or ethics committee approvals have been obtained.YesThe details of the IRB/oversight body that provided approval or exemption for the research described are given below:This study complied with all relevant ethical regulations and was approved by the University of Michigan Institutional Review Board.All necessary patient/participant consent has been obtained and the appropriate institutional forms have been archived.YesI understand that all clinical trials and any other prospective interventional studies must be registered with an ICMJE-approved registry, such as ClinicalTrials.gov. I confirm that any such study reported in the manuscript has been registered and the trial registration ID is provided (note: if posting a prospective study registered retrospectively, please provide a statement in the trial ID field explaining why the study was not registered in advance).YesI have followed all appropriate research reporting guidelines and uploaded the relevant EQUATOR Network research reporting checklist(s) and other pertinent material as supplementary files, if applicable.YesUpon publication, RNA sequencing data will be uploaded to a public server. Other data will be made available by email to the corresponding author. ER -