PT - JOURNAL ARTICLE AU - Travis Nemkov AU - Davide Stefanoni AU - Aarash Bordbar AU - Aaron Issaian AU - Bernhard O. Palsson AU - Larry J Dumont AU - Ariel Hay AU - Anren Song AU - Yang Xia AU - Jasmina S. Redzic AU - Elan Z. Eisenmesser AU - James C Zimring AU - Steve Kleinman AU - Kirk C. Hansen AU - Michael P. Busch AU - Angelo D’Alessandro AU - for the Recipient Epidemiology and Donor Evaluation Study-III (REDS-III) RBC-Omics Study TI - Blood donor exposome and impact of common drugs on red blood cell metabolism AID - 10.1101/2020.08.17.20176891 DP - 2020 Jan 01 TA - medRxiv PG - 2020.08.17.20176891 4099 - http://medrxiv.org/content/early/2020/08/21/2020.08.17.20176891.short 4100 - http://medrxiv.org/content/early/2020/08/21/2020.08.17.20176891.full AB - Computational models based on recent maps of the red blood cell proteome suggest that mature erythrocytes may harbor targets for common drugs. This prediction is relevant to red blood cell storage in the blood bank, in which the impact of small molecule drugs or other xenometabolites deriving from dietary, iatrogenic or environmental exposures (“exposome”) may alter erythrocyte energy and redox metabolism and, in so doing, affect red cell storage quality and post-transfusion efficacy. To test this prediction, here we provide a comprehensive characterization of the blood donor exposome, including the detection of common prescription and off-the-counter drugs in 250 units donated by healthy volunteers from the REDS-III RBC Omics study. Based on high-throughput drug screenings of 1,366 FDA-approved drugs, we report a significant impact of ∼65% of the tested drugs on erythrocyte metabolism. Machine learning models built using metabolites as predictors were able to accurately predict drugs for several drug classes/targets (bisphosphonates, anticholinergics, calcium channel blockers, adrenergics, proton-pump inhibitors, antimetabolites, selective serotonin reuptake inhibitors, and mTOR) suggesting that these drugs have a direct, conserved, and significant impact on erythrocyte metabolism. We then focused on ranitidine – a common antiacid – as a representative drug with the potential to improve human erythrocyte storage quality and post-transfusion performances in mice. By combining tracing experiments with 1,2,3-13C3-glucose, proteome integral solubility alteration assays, genetic ablation of S1P synthesis capacity, in silico docking and 1D NMR, we show that ranitidine triggers metabolic mechanisms involving sphingosine 1-phosphate (S1P)-dependent modulation of erythrocyte glycolysis and/or direct binding to hemoglobin.RBC exposome from the REDS III study revealed that blood from a subset of donors contains traces of the most common drugs in the United States. RBCs can uptake these drugs, in some cases can metabolize them to their bioactive metabolites and in others the drug can directly impact RBC metabolism during storage.Key pointsBlood donor exposomes include metabolites of environmental exposure, traces of common prescription or off-the-counter drugs;65% of 1366 FDA- approved drug significantly affect RBC metabolism. Ranitidine significantly impacts glycolysis and S1P metabolism.Competing Interest StatementThe authors declare that AD, TN and KCH are founders of Omix Technologies Inc and Altis Biosciencens LLC. AB and BOP are founders of Sinopia Biosciences Inc. JCZ and AD are consultants for Rubius Therapeutics. AD is an advisory board member for Hemanext Inc. and Forma Therapeutics Inc. All the other authors disclose no conflicts of interest relevant to this study.Clinical TrialBioLINCC Study: HLB02071919aFunding StatementResearch reported in this publication was funded by the NHLBI Recipient Epidemiology and Donor Evaluation Study-III (REDS-III), which was supported by NHLBI contracts NHLBI HHSN2682011-00001I, -00002I, -00003I, -00004I, -00005I, -00006I, -00007I, -00008I, and -00009I, as well as funds from the the National Institute of General and Medical Sciences (RM1GM131968 to ADA and KCH), the Boettcher Webb-Waring Investigator Award (ADA), and R01HL146442 (ADA), R01HL149714 (ADA), R01HL148151 (ADA, JCZ), R21HL150032 (ADA), and T32 HL007171 (TN) from the National Heart, Lung, and Blood Institute. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. . The authors would like to express their deep gratitude Dr. Simone Glynn of NHLBI for her support throughout this study, the RBC-Omics research staff at all participating blood centers and testing labs for their contribution to this project, and to all blood donors who agreed to participate in this study..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:Donor selection and recruitment for the RBC-Omics study under approved protocols (BioLINCC Study: HLB02071919a) were previously detailed in references 27-29 of this manuscript.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.YesAll the raw data are provided in the Supplementary Table of this manuscript.