Abstract
Background
Blood transfusion is a life-saving intervention for millions of recipients worldwide. Over the last 15 years, the advent of high-throughput, affordable omics technologies – including genomics, proteomics, lipidomics, and metabolomics – has allowed transfusion medicine to revisit the biology of blood donors, stored blood products, and transfusion recipients.
Summary
Omics approaches have shed light on the genetic and non-genetic factors (environmental or other exposures) impacting the quality of stored blood products and efficacy of transfusion events, based on the current Food and Drug Administration guidelines (e.g., hemolysis and post-transfusion recovery for stored red blood cells). As a treasure trove of data accumulates, the implementation of machine learning approaches promises to revolutionize the field of transfusion medicine, not only by advancing basic science. Indeed, computational strategies have already been used to perform high-content screenings of red blood cell morphology in microfluidic devices, generate in silico models of erythrocyte membrane to predict deformability and bending rigidity, or design systems biology maps of the red blood cell metabolome to drive the development of novel storage additives.
Key Message
In the near future, high-throughput testing of donor genomes via precision transfusion medicine arrays and metabolomics of all donated products will be able to inform the development and implementation of machine learning strategies that match, from vein to vein, donors, optimal processing strategies (additives, shelf life), and recipients, realizing the promise of personalized transfusion medicine.
Key Words: Machine learning, Omics, Red blood cells, Storage, Transfusion
Overview
In this review, we will summarize the most recent advances in the field of omics technologies and machine learning in transfusion. Owing to the author's own limited expertise, this short review will not focus on the implementation of artificial intelligence in blood inventory management [1], recipient profiling [2], the prediction of recipient comorbidities, and outcomes in the emergency and acute care setting [3, 4]. Rather, we will focus on molecular aspects of blood storage, with an emphasis on packed red blood cell (RBC) products and the crossroads of omics characterization and deep learning strategies for the identification of markers of storage quality and transfusion outcomes [5]. The focus on RBCs is justified both by the disproportionately higher numbers of omics papers in the field of RBC storage biology compared to other products but also and foremost because of the disproportionately higher number of packed RBC units transfused daily in the USA (∼29,000 units, as opposed to the nearly 5,000 units of platelets and 6,500 units of plasma). While recent comprehensive reviews on omics and RBC storage are available in the literature [6, 7], the rapid evolution of the field has generated a novel, solid body of knowledge that complements and expands on the literature reviewed in those recent reports. As such, the main goal of this short review is to bridge that gap.
RBCs: More Than a Circulating Bag of Hemoglobin
RBCs are by far the most abundant cell in the human body: 25 out of 30 × 1012 human cells are RBCs [8, 9]. The mature erythrocyte contains ∼250–270 million molecules of hemoglobin (Hb)/cell, which facilitate the transport of up to 1 billion molecules of oxygen/cell [10]. RBC function, specifically Hb's capacity to bind and off-load oxygen, is tightly regulated by metabolic mechanisms. These mechanisms involve direct allosteric modulators such as 2,3-diphosphoglycerate (DPG) and adenosine triphosphate (ATP) [11, 12]. Using a combination of omics technologies and single cell measurements of oxygen kinetics via microfluidic devices and biophysics approaches, we recently identified a whole new series of correlates to the Hb capacity to bind and off-load oxygen, including the indirect modulators adenosine, sphingosine-1-phosphate (S1P), recently validated in mechanistic studies on high-altitude hypoxia [13, 14, 15, 16, 17, 18] and RBC storage [19].
While Hb facilitates oxygen delivery, Hb autoxidation promotes oxidant stress in the RBC, with Hb iron (66% of bodily iron is in RBCs) fueling radical-generating Fenton and Haber-Weiss reactions. Specifically, RBC Hb is an oxygen-dependent buffer for glutathione – the main small molecule antioxidant in the cell [20] – through beta chain cysteine 93, which regulates recycling of key antioxidant enzymes like peroxiredoxin 2 [21]. Since no new proteins can be synthesized by mature RBCs, owing to the lack of nuclei and organelles, during their ∼120 day lifespan, RBCs have to cope with oxidant stress through mechanisms independent of gene regulation or protein expression [22]. Therefore, RBCs represent an excellent cell model to investigate metabolic responses to oxidant damage [10], which is relevant not just for storage biology but also for related iatrogenic interventions (e.g., RBC responses to hypoxia and oxidant stress upon transfusion in unhealthy, heterologous recipients).
Despite the lack of nuclei and organelles and the overwhelming abundance of a single class of proteins (Hb represents 98% of the cytosolic proteome), RBCs are not as simple as previously thought. Recent studies [23, 24, 25], including some from our group [26], have elucidated an unanticipated complexity of the RBC protein machinery. Despite Hb making up ∼90% of the total RBC proteome, more than 3,000 proteins are identified in RBCs [23, 26], including 77 transporters for 267 small molecules [27]. By combining tracing experiments with 13C- and 2H-labeled substrates and functional proteomics assays, we – among other groups – are showing one by one (e.g., carboxylic acid metabolism [28, 29, 30, 31], fatty acid desaturases [32], nitric oxide synthase [33]), that these enzymes are not inert remnants of maturation processes but rather they are active in mature RBCs and catalyze reactions that were once thought to be exclusive to specific organs. Thus, we provocatively introduced the concept of RBCs as an organ with functions beyond O2 transport [10]. All the considerations above are relevant examples of how omics approaches can be used for basic science that unravels biology relevant to transfusion medicine, in that all the pathways mentioned above are critical regulators of stored RBC metabolism, as we will summarize below.
RBC Omics in Health and Disease Paves the Way for Mechanistic Understanding of the Storage Lesion
Altered RBC antioxidant capacity triggers intra- [34] or extra-vascular hemolysis [35, 36], resulting in excess circulating heme and iron [37], a phenomenon that underlies the etiology of many diseases in which oxidant stress plays a central role [37] and co-morbidities in transfusion recipients, such as acute lung injury, kidney dysfunction [38, 39, 40], microbiome dysbiosis [41], or septic complications in patients infected by siderophilic bacteria [42]. As such, despite intrinsic limitations of animal models [43], comparative investigations of RBC metabolism in humans and animals – where alternative strategies to cope with such stress may have evolved to regulate hemolytic propensity – can thus further our understanding of the role of (transfused) RBCs in system physiology and its pathological alterations – other than having critical direct implications for veterinary transfusion medicine purposes [44, 45, 46, 47].
The identification of strategies to mitigate oxidant stress on RBCs holds immediate and critical biomedical implications. Indeed, RBC storage in the blood bank is a logistic necessity to make ∼110 million units/year available for blood transfusion, the most common in hospital procedure worldwide after vaccination. However, as storage progresses, RBCs undergo a series of biochemical and morphological modifications that are mostly triggered by oxidant stress [6, 7]. An overview of how omics technologies helped elucidate the targets of oxidant damage elicited by oxidant stress is provided below.
Omics in Transfusion Medicine
In the last 6 years, we and others have developed high-throughput proteomics and metabolomics methods – as rapid as 1 min per sample – to ensure feasibility of clinical omics studies [48, 49, 50, 51], which allowed rapid responses to rapid threats, such as the case of the COVID-19 pandemics and the impact of SARS-CoV-2 infection on RBC biology [52, 53, 54, 55, 56, 57, 58, 59]. In parallel, a burgeoning literature has emerged focusing on omics applications to transfusion medicine [6, 7]. Early omics studies in transfusion medicine have investigated the impact of RBC storage duration in all currently licensed storage additive solutions (AS), including SAGM, AS-1, 3, 5, PAGGSM, and novel alkaline additives, such as AS7, ESOL5, and PAGGGSM [29, 60, 61, 62, 63, 64, 65]. These studies revealed a significant impact of storage additives on the kinetics of the storage lesion, with alkaline additives better preserving energy metabolism by means of enhancement of the activity of pH-sensitive rate-limiting enzymes of glycolysis (phosphofructokinase) and the Rapoport-Luebering shunt (bisphosphoglycerate mutase) [66]. Based on these studies, we and others identified metabolic markers of the storage lesion [67, 68], and correlated storage-induced metabolic changes to the gold standard markers of storage quality as per the Food and Drug Administration guidelines, i.e., storage hemolysis and post-transfusion recovery (PTR) [34, 69, 70, 71, 72, 73, 74].
Through murine models of blood storage developed by Dr. Zimring [75], we identified the ferrireductase STEAP3 as a genetic and mechanistic factor contributing to the poor post-transfusion recovery of stored RBCs from mouse strains like FVB but not C57BL/6 [76]. Specifically, lipidomics studies have shown that iron metabolism in stored RBCs is tied to an elevation in oxidant stress, ultimately triggering lipid peroxidation and initiating a process that closely resembles that of iron-induced non-apoptotic cell death, or ferroptosis [77]. Of note, STEAP3, also known as tumor suppressor p53-activated pathway-6 (TSAP6), is a target of p53 transcriptional activity [78]. Both p53 and STEAP3 regulate erythropoiesis and limit maturing erythroid cell iron content [79, 80, 81], thus constraining substrate availability for one key reactant in Fenton chemistry. Even more interestingly, the rate of detoxification of oxidized lipids (and ferroptosis) is regulated by glutathione peroxidase 4 (GPX4) – a glutathione-dependent enzyme that is expressed in mature RBCs and correlates with hemolytic propensity [82, 83].
One of the main targets of oxidant stress in stored RBCs is the most abundant membrane protein – anion exchanger 1 (AE1) – with 1 million copies/cell [24]. AE1, also known as band 3, regulates the chloride/bicarbonate exchange (chloride shift), critical to CO2 and pH homeostasis [84] and thus pH-dependent activity of metabolic enzymes. AE1 has pleiotropic functions mediated by structural interactions [85, 86, 87]. The N-terminal AE1 residues 1–23 serve as a docking site for deoxygenated Hb under hypoxia and glycolytic enzymes at high oxygen saturation [85, 86, 87]. Under pro-oxidant (high SO2) conditions, AE1 binding inhibits glycolytic enzymes and promotes fluxes via the pentose phosphate pathway (PPP), which generates NADPH, a reducing cofactor necessary for recycling of oxidized glutathione and NADPH-dependent antioxidant enzymes [22]. Under hypoxia, deoxyHb binding displaces glycolytic enzymes from AE1, favoring glycolysis and the generation of allosteric modulators ATP and DPG, and thus oxygen off-loading [19, 88]. This O2-dependent metabolic “switch” (hereon, “AE1-Hb switch”) favors anti-oxidant metabolism when oxidant stress is high (e.g., during storage in the blood bank), energy metabolism, and O2 release when oxidant stress is low (e.g., in peripheral tissues or in response to high altitude). Through omics technologies, it has been shown that oxidant stress [60, 89] and protease activity [90] promote the fragmentation of the AE-1 N-terminus, impairing the capacity to cope with storage-induced oxidant stress – to the extent that RBCs from mice lacking the AE1 N-terminus store poorly, while treatment with a membrane-permeable AE11-57 peptide partially rescues the phenotype [74]. Indeed, we proved that oxidation and fragmentation of the cytosolic N-terminus of AE1 impair the capacity to bind glyceraldehyde 3-phosphate dehydrogenase and inhibit glycolysis [74], thereby limiting the capacity to activate the PPP [74, 91], a critical pathway for the generation of the cofactor NADPH, which is essential in many reducing reactions (for example, recycling of oxidized glutathione) [22]. This process is in part counteracted by the activation of protein L-isoaspartyl o-methyltransferase [92, 93] and by the compensatory activity of the rate-limiting enzyme of the PPP, glucose 6-phosphate dehydrogenase (G6PD). We then showed that G6PD polymorphisms are common in the donor population – as they are across all humans (∼6% of mankind, 500 million people around the world [94]) – and impact storage quality and transfusion efficacy [73, 95, 96, 97].
Blood Donors as a Window on Population Health: Toward Personalized (Transfusion) Medicine
While most of the studies described above focused on relatively small-scale studies in humans and animal models, the advent of high-throughput technologies affords much broader investigations to characterize the blood donor population with sample sets in the tens to hundreds of thousands [98]. For example, the US National Heart, Lung, and Blood Institute-sponsored Recipient Epidemiology and Donor Evaluation Study (REDS) has recently leveraged genomics approaches to characterize 879,000 polymorphisms from >13,000 donor volunteers[99]. Of note, through genomics studies, almost all of the enzymes mentioned in the previous paragraph were found to be polymorphic in the REDS RBC Omics blood donor population, and functional single nucleotide polymorphisms were associated with the hemolytic propensity of stored blood [82] and Hb increments in transfusion recipients [100].
A subset of the donors from the screening phase of the REDS study (n = 643) were identified as extreme hemolyzers (either spontaneous hemolysis, or upon oxidative and osmotic insults) and asked to donate a second unit of blood for multi-omics testing at storage day 10, 23, and 42 – a dataset that was in part disseminated through pilot metabolomics studies on a subset of these samples (∼600 out of a total of ∼2,000 – with the whole cohort of index and recalled donor just recently tested by our group). As proof of principle for cross-omics analyses from large transfusion medicine cohorts, we performed a pilot metabolite quantitative trait loci – mQTL analysis on just 250 of the 13K donors. We identified 2,831 high-confidence SNP-metabolite linkages (p < 5.0 × 10−8) [101]. We thus generated novel murine models for one of the most significant polymorphisms – G6PD African (V68M/N126D) and Mediterranean (S188F) variants – for future functional studies relevant to transfusion medicine and hematology [101].
Through genomics and metabolomics data in the REDS RBC Omics cohort, it has been noted that biological factors such as donor sex, sex hormones, age, body mass index contribute to storage quality [32, 73, 95, 96, 97, 102], as gleaned by hemolytic propensity [103], omics phenotypes [34], and Hb increments in recipients of units donated by the same donor volunteers [100, 104]. We then showed that female donors have younger circulating RBCs at the time of donation, which are more resistant to oxidant challenge in storage and have more resilient energy and antioxidant metabolism during storage [105]. Later, we introduced the concept of the blood donor exposome [51], showing that dietary (e.g., alcohol, caffeine consumption [106, 107]) or other exposures (e.g., smoking [108], exercise [109, 110], diet [111], prior infection by flaviviruses [112] or coronaviruses [54, 113], drugs that are not grounds for donor deferral [51]) all contribute to modulating stored RBC energy and redox metabolism, ultimately impacting Hb increments upon transfusion [100, 104]. We concluded that the chronological age of blood – in terms of days elapsed since the time of donation – is qualitatively distinct from the metabolic age of the unit – the “molecular storage lesion” [114]. We then tested alternative storage strategies (cryopreservation [115], rejuvenation [116], hypoxic storage [16, 30, 117, 118]), as well as leveraged a novel combination of high-throughput omics and 96-well plate-based scaled storage systems to develop novel additives [19, 119].
While currently transfusion of RBC relies on the altruistic gift of an estimated 6.8 million volunteers in the USA alone every year, omics studies have been preliminary used to inform or validate the development of novel bio-inspired synthetic blood products [120] or the ex vivo expansion and differentiation of hematopoietic stem cells [23, 121], as well as the testing of RBCs after hypotonic dialysis-based drug encapsulation process of enzymes for therapeutics intervention in the oncological setting [122].
From Blood Donors to Recipients
On top of the focus on the blood donors and products, omics technologies in transfusion medicine have been used to investigate transfusion recipients, either healthy autologous volunteers [123] or non-healthy heterologous recipients – with a special focus on trauma patients (massively transfused [124]) and sickle cell patients, either receiving standard red cell exchanges [125] or rejuvenated red cell exchanges [126]. With a combination of biotinylation studies and single cell oxygenation strategies, we determined that the storage lesion impacts RBC oxygen kinetics (transport and off-loading [116, 127]), which is only partially reversible upon transfusion of RBCs in healthy autologous recipients [128]. Indeed, at the net of the survival bias impacting the readout, omics data generated on stored biotinylated RBCs – in time course studies where the transfused RBCs were flow-sorted out of the bloodstream of the recipient – showed that while some of the metabolic storage lesion (e.g., low ATP and DPG levels) can be partially restored over time in circulation (at kinetics that may be too slow for example in the hypoxic critically ill patient), while other pathways – such as the PPP – are not restored at all [128].
Through (in vivo) metabolic tracing [129, 130] in tractable animal models (e.g., rodents, including mice and rats; porcine or non-human primates) [130, 131, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142], we are now using omics technologies to investigate the role of genetic background across multiple species as a driver of the storage lesion (the Zoomics project) [44, 45, 46, 47], as well as the impact of transfusion in pre-clinical models [134, 138, 140, 142, 143]. In parallel, we are exploring recipient outcomes in massively (e.g., trauma patients) [131, 140, 144] or chronically transfused patients (e.g., sickle cell patients) [17, 125, 145, 146, 147, 148, 149, 150].
In parallel, we are exploring the role of genetic abnormalities that can result in the need for transfusion, such as those linked to hemoglobinopathies (e.g., sickle cell trait, sickle cell disease; beta-thalassemia) [147, 149, 151, 152, 153] or inborn errors of metabolism beyond G6PD deficiency (e.g., pyruvate kinase deficiency, propionic acidemia) [154, 155], especially in under-represented populations in science (e.g., the Amish Mennonites and other populations in rural America [154, 155]; individuals with Down syndrome [156, 157], transgender individuals during gender reassignment therapy with sex hormones [158], or hypogonadic individuals who develop erythrocytosis and donate blood as a strategy to counteract such effects of hormonal therapies [159]).
Machine Learning: From Biomarkers to Systems Biology to In Silico Models of RBC Membranes
We mentioned above the use of machine learning approaches (including random forest and lasso regression) to identify markers of RBC storage quality and transfusion performance in healthy autologous volunteers and trauma patients [67, 68, 117, 124]. More recently, machine learning approaches have been coupled to high-content screening that leverages microfluidic devices and microscopy in the presence or absence of flow to determine the impact of a given treatment (storage or even SARS-CoV-2 infection) on RBC morphology in unperturbed or perturbed systems [5, 113]. Indeed, RBC morphology is altered as a function of damage to energy and redox metabolism, which ultimately results in vesiculation of oxidized components [64], loss of the discocytic phenotype and acquisition of progressively irreversibly altered morphologies (spheroechinocyte, spherocyte [160]), increased surface-to-volume ratios, increased mechanical fragility, and ultimately, sequestration in the spleen that triggers extravascular hemolysis [36, 161], and erythrophagocytosis [162]. Of note, a combination of RBC membrane protein and lipid composition as a function of storage can be fed into novel in silico models of the erythrocyte membrane, which can be used to predict membrane rigidity and deformability in fresh and stored RBCs, and predict actual direct biophysical measurements of RBC mechanics [163, 164]. Feeding of quantitative and functional metabolomics (e.g., tracing experiments) and proteomics data to computational models of RBC metabolic pathways has fueled recent progress in the field of systems biology of the RBC [27, 28, 165, 166], which can be used to drive the development of novel storage solutions [167] or simply investigate RBC aging in the bloodstream in vivo [168].
Application of machine learning and artificial intelligence approaches in healthcare will face key challenges, as reviewed extensively elsewhere [169]. One limitation of most existing studies in this space to date has been that benchmarking of machine learning algorithms has mostly been performed on retrospective data from large, already available databases. Performance of these models is likely to suffer when tested against real-world data from prospective studies as opposed to historically labeled data used for algorithm training. Another challenge that this field will encounter is based on the appreciation that machine learning algorithms will use input signals to achieve the best possible (prediction) performance in the dataset used; algorithms may thus end up exploiting signals from unknown confounders that may not be reliable, impairing the algorithm's ability to generalize to new datasets [169]. Depending on training sets, algorithms may develop implicit bias, which would limit their fitness for generalization and the accuracy of clinical predictions. The development of robust regulation and a rigorous quality control strategy will be a challenge facing this new, yet promising field.
Despite early success in the above-mentioned applications, the machine learning area that holds the strongest promise is that of personalized transfusion medicine approaches. The opportunity is there to match donors and recipients based on a treasure trove of data that can now be generated, in a cost-effective fashion, with ultra-high-throughput omics approaches. It is possible to imagine a not-so-distant future when artificial intelligence-based approaches could quickly identify optimal storage additives for blood based on donor biology and omics signatures at donation, shorten or extend the shelf-life of the product accordingly, or make indications for a specific recipient (matching the right product for acutely bleeding, massively transfused trauma patient vs. chronically transfused patient with hemoglobinopathies). These features could be made possible by the implementation in central lab testing practices of blood donor genomics characterization with precision transfusion medicine arrays [99] (including blood group antigen, rare polymorphisms associated with stored blood quality), along with metabolic characterization of non-genetic factors, e.g., metabolites of dietary or other exposures, from habits like smoking all the way to prescription drugs that are not grounds for blood donor deferral – in combination with donor and recipient demographics and clinical records.
Conflict of Interest Statement
A.D. is a founder of Omix Technologies Inc. and Altis Biosciences LLC, a consultant for Hemanext Inc., Macopharma Inc., and Forma Therapeutics.
Funding Sources
A.D. was supported by funds from the National Institute of General and Medical Sciences (RM1GM131968) and the National Heart, Lung, and Blood Institute (R01HL146442, R01HL149714, R01HL148151, R01HL161004, R21HL150032). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Author Contributions
A.D. wrote the manuscript.
Acknowledgments
The title is loosely inspired by Ray Kurzweil's book “Singularity is near,” a 2005 non-fiction book (ISBN: 978-0-670-03384-3) about artificial intelligence and the future of humanity.
Funding Statement
A.D. was supported by funds from the National Institute of General and Medical Sciences (RM1GM131968) and the National Heart, Lung, and Blood Institute (R01HL146442, R01HL149714, R01HL148151, R01HL161004, R21HL150032). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
References
- 1.Li N, Arnold DM, Down DG, Barty R, Blake J, Chiang F, et al. From demand forecasting to inventory ordering decisions for red blood cells through integrating machine learning statistical modeling and inventory optimization. Transfusion. 2022 Jan;62((1)):87–99. doi: 10.1111/trf.16739. [DOI] [PubMed] [Google Scholar]
- 2.Choorapoikayil S, Hof L, Old O, Steinbicker A, Meybohm P, Zacharowski K. How do I/we forecast tomorrow's transfusion? A focus on recipients' profiles. Transfus Clin Biol. 2023;30((1)):27–30. doi: 10.1016/j.tracli.2022.09.063. [DOI] [PubMed] [Google Scholar]
- 3.Abujaber A, Fadlalla A, Gammoh D, Abdelrahman H, Mollazehi M, El-Menyar A. Prediction of in-hospital mortality in patients on mechanical ventilation post traumatic brain injury machine learning approach. BMC Med Inform Decis Mak. 2020 Dec 14;20((1)):336. doi: 10.1186/s12911-020-01363-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Zhang Z, Navarese EP, Zheng B, Meng Q, Liu N, Ge H, et al. Analytics with artificial intelligence to advance the treatment of acute respiratory distress syndrome. J Evid Based Med. 2020 Nov;13((4)):301–312. doi: 10.1111/jebm.12418. [DOI] [PubMed] [Google Scholar]
- 5.Doan M, Sebastian JA, Caicedo JC, Siegert S, Roch A, Turner TR, et al. Objective assessment of stored blood quality by deep learning. Proc Natl Acad Sci U S A. 2020;117((35)):21381–90. doi: 10.1073/pnas.2001227117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.D'Alessandro A, Kriebardis AG, Rinalducci S, Antonelou MH, Hansen KC, Papassideri IS, et al. An update on red blood cell storage lesions as gleaned through biochemistry and omics technologies. Transfusion. 2015 Jan;55((1)):205–219. doi: 10.1111/trf.12804. [DOI] [PubMed] [Google Scholar]
- 7.Yoshida T, Prudent M, D'Alessandro A. Red blood cell storage lesion causes and potential clinical consequences. Blood Transfus. 2019 Jan;17((1)):27–52. doi: 10.2450/2019.0217-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, et al. An estimation of the number of cells in the human body. Ann Hum Biol. 2013 Nov–Dec;40((6)):463–471. doi: 10.3109/03014460.2013.807878. [DOI] [PubMed] [Google Scholar]
- 9.Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016 Aug;14((8)):e1002533. doi: 10.1371/journal.pbio.1002533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Nemkov T, Reisz JA, Xia Y, Zimring JC, D'Alessandro A. Red blood cells as an organ? How deep omics characterization of the most abundant cell in the human body highlights other systemic metabolic functions beyond oxygen transport. Expert Rev Proteomics. 2018 Nov;15((11)):855–864. doi: 10.1080/14789450.2018.1531710. [DOI] [PubMed] [Google Scholar]
- 11.Duhm J, Gerlach E. On the mechanisms of the hypoxia-induced increase of 2 3-diphosphoglycerate in erythrocytes. Studies on rat erythrocytes in vivo and on human erythrocytes in vitro. Pflugers Arch. 1971 Sep 1;326((3)):254–269. doi: 10.1007/BF00592506. [DOI] [PubMed] [Google Scholar]
- 12.Changeux JP. 50 years of allosteric interactions the twists and turns of the models. Nat Rev Mol Cell Biol. 2013 Dec;14((12)):819–829. doi: 10.1038/nrm3695. [DOI] [PubMed] [Google Scholar]
- 13.D'Alessandro A, Nemkov T, Sun K, Liu H, Song A, Monte AA, et al. AltitudeOmics red blood cell metabolic adaptation to high altitude hypoxia. J Proteome Res. 2016 Oct 7;15((10)):3883–3895. doi: 10.1021/acs.jproteome.6b00733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Liu H, Zhang Y, Wu H, D'Alessandro A, Yegutkin GG, Song A, et al. Beneficial role of erythrocyte adenosine A2B receptor-mediated AMP-activated protein kinase activation in high-altitude hypoxia. Circulation. 2016 Aug 2;134((5)):405–421. doi: 10.1161/CIRCULATIONAHA.116.021311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Song A, Zhang Y, Han L, Yegutkin GG, Liu H, Sun K, et al. Erythrocytes retain hypoxic adenosine response for faster acclimatization upon re-ascent. Nat Commun. 2017 Feb 7;8:14108. doi: 10.1038/ncomms14108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Nemkov T, Sun K, Reisz JA, Song A, Yoshida T, Dunham A, et al. Hypoxia modulates the purine salvage pathway and decreases red blood cell and supernatant levels of hypoxanthine during refrigerated storage. Haematologica. 2018 Feb;103((2)):361–372. doi: 10.3324/haematol.2017.178608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.D'Alessandro A, Xia Y. Erythrocyte adaptive metabolic reprogramming under physiological and pathological hypoxia. Curr Opin Hematol. 2020 May;27((3)):155–162. doi: 10.1097/MOH.0000000000000574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Xu P, Chen C, Zhang Y, Dzieciatkowska M, Brown BC, Zhang W, et al. Erythrocyte transglutaminase-2 combats hypoxia and chronic kidney disease by promoting oxygen delivery and carnitine homeostasis. Cell Metab. 2022 Feb 1;34((2)):299.e6–316.e6. doi: 10.1016/j.cmet.2021.12.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Hay AM, Nemkov T, Gamboni F, Dzieciatkowska M, Key A, Galbraith M, et al. S1P has a negative effect on RBC storage quality. Blood Adv. 2022 Dec 5; doi: 10.1182/bloodadvances.2022008936. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Fenk S, Melnikova EV, Anashkina AA, Poluektov YM, Zaripov PI, Mitkevich VA, et al. Hemoglobin is an oxygen-dependent glutathione buffer adapting the intracellular reduced glutathione levels to oxygen availability. Redox Biol. 2022 Nov 16;58:102535. doi: 10.1016/j.redox.2022.102535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Harper VM, Oh JY, Stapley R, Marques MB, Wilson L, Barnes S, et al. Peroxiredoxin-2 recycling is inhibited during erythrocyte storage. Antioxid Redox Signal. 2015 Feb 1;22((4)):294–307. doi: 10.1089/ars.2014.5950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.D'Alessandro A, Hansen KC, Eisenmesser EZ, Zimring JC. Protect destroy or sacrifice a role of oxidative stress biology in inter-donor variability of blood storage? Blood Transfus. 2019 Jul;17((4)):281–288. doi: 10.2450/2019.0072-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Wilson MC, Trakarnsanga K, Heesom KJ, Cogan N, Green C, Toye AM, et al. Comparison of the proteome of adult and cord erythroid cells and changes in the proteome following reticulocyte maturation. Mol Cell Proteomics. 2016 Jun;15((6)):1938–1946. doi: 10.1074/mcp.M115.057315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bryk AH, Wiśniewski JR. Quantitative analysis of human red blood cell proteome. J Proteome Res. 2017 Aug 4;16((8)):2752–2761. doi: 10.1021/acs.jproteome.7b00025. [DOI] [PubMed] [Google Scholar]
- 25.Gautier EF, Leduc M, Cochet S, Bailly K, Lacombe C, Mohandas N, et al. Absolute proteome quantification of highly purified populations of circulating reticulocytes and mature erythrocytes. Blood Adv. 2018 Oct 23;2((20)):2646–2657. doi: 10.1182/bloodadvances.2018023515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.D'Alessandro A, Dzieciatkowska M, Nemkov T, Hansen KC. Red blood cell proteomics update is there more to discover? Blood Transfus. 2017 Mar;15((2)):182–187. doi: 10.2450/2017.0293-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Bordbar A, Jamshidi N, Palsson BO. iAB-RBC-283 a proteomically derived knowledge-base of erythrocyte metabolism that can be used to simulate its physiological and patho-physiological states. BMC Syst Biol. 2011;5((1)):110. doi: 10.1186/1752-0509-5-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bordbar A, Yurkovich JT, Paglia G, Rolfsson O, Sigurjónsson ÓE, Palsson BO. Elucidating dynamic metabolic physiology through network integration of quantitative time-course metabolomics. Sci Rep. 2017 Apr 7;7:46249. doi: 10.1038/srep46249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.D'Alessandro A, Nemkov T, Yoshida T, Bordbar A, Palsson BO, Hansen KC. Citrate metabolism in red blood cells stored in additive solution-3. Transfusion. 2017 Feb;57((2)):325–336. doi: 10.1111/trf.13892. [DOI] [PubMed] [Google Scholar]
- 30.Nemkov T, Sun K, Reisz JA, Yoshida T, Dunham A, Wen EY, et al. Metabolism of citrate and other carboxylic acids in erythrocytes as a function of oxygen saturation and refrigerated storage. Front Med. 2017;4:175. doi: 10.3389/fmed.2017.00175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Rolfsson Ó, Sigurjonsson ÓE, Magnusdottir M, Johannsson F, Paglia G, Guðmundsson S, et al. Metabolomics comparison of red cells stored in four additive solutions reveals differences in citrate anticoagulant permeability and metabolism. Vox Sang. 2017 May;112((4)):326–335. doi: 10.1111/vox.12506. [DOI] [PubMed] [Google Scholar]
- 32.Thomas T, Cendali F, Fu X, Gamboni F, Morrison EJ, Beirne J, et al. Fatty acid desaturase activity in mature red blood cells and implications for blood storage quality. Transfusion. 2021 Apr 26;61((6)):1867–1883. doi: 10.1111/trf.16402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Leo F, Suvorava T, Heuser SK, Li J, LoBue A, Barbarino F, et al. Red blood cell and endothelial eNOS independently regulate circulating nitric oxide metabolites and blood pressure. Circulation. 2021 Sep 14;144((11)):870–889. doi: 10.1161/CIRCULATIONAHA.120.049606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.D'Alessandro A, Fu X, Kanias T, Reisz JA, Culp-Hill R, Guo Y, et al. Donor sex age and ethnicity impact stored red blood cell antioxidant metabolism through mechanisms in part explained by glucose 6-phosphate dehydrogenase levels and activity. Haematologica. 2021 May 1;106((5)):1290–1302. doi: 10.3324/haematol.2020.246603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.D'Alessandro A. In vivo clearance of stored red blood cells. Blood. 2021 Apr 29;137((17)):2275–2276. doi: 10.1182/blood.2021010946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Roussel C, Morel A, Dussiot M, Marin M, Colard M, Fricot-Monsinjon A, et al. Rapid clearance of storage-induced microerythrocytes alters transfusion recovery. Blood. 2021 Apr 29;137((17)):2285–2298. doi: 10.1182/blood.2020008563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Vallelian F, Buehler PW, Schaer DJ. Hemolysis free hemoglobin toxicity and scavenger protein therapeutics. Blood. 2022 Oct 27;140((17)):1837–1844. doi: 10.1182/blood.2022015596. [DOI] [PubMed] [Google Scholar]
- 38.Nath KA, Hebbel RP. Sickle cell disease renal manifestations and mechanisms. Nat Rev Nephrol. 2015 Mar;11((3)):161–171. doi: 10.1038/nrneph.2015.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bissinger R, Nemkov T, D'Alessandro A, Grau M, Dietz T, Bohnert BN, et al. Proteinuric chronic kidney disease is associated with altered red blood cell lifespan deformability and metabolism. Kidney Int. 2021 Dec;100((6)):1227–1239. doi: 10.1016/j.kint.2021.08.024. [DOI] [PubMed] [Google Scholar]
- 40.van Wonderen SF, Klanderman RB, Vlaar APJ. Understanding transfusion-related acute lung injury (TRALI) and its complex pathophysiology. Blood Transfus. 2022 Nov;20((6)):443–445. doi: 10.2450/2022.0232-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.La Carpia F, Wojczyk BS, Annavajhala MK, Rebbaa A, Culp-Hill R, D'Alessandro A, et al. Transfusional iron overload and intravenous iron infusions modify the mouse gut microbiota similarly to dietary iron. NPJ Biofilms Microbiomes. 2019;5((1)):26. doi: 10.1038/s41522-019-0097-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.La Carpia F, Slate A, Bandyopadhyay S, Wojczyk BS, Godbey EA, Francis KP, et al. Red blood cell transfusion-induced non-transferrin-bound iron promotes Pseudomonas aeruginosa biofilms in human sera and mortality in catheterized mice. Br J Haematol. 2022;196((4)):1105–1110. doi: 10.1111/bjh.17934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hay AM, Howie HL, Gorham JD, D'Alessandro A, Spitalnik SL, Hudson KE, et al. Mouse background genetics in biomedical research the devil's in the details. Transfusion. 2021 Oct;61((10)):3017–3025. doi: 10.1111/trf.16628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bertolone L, Shin HK, Stefanoni D, Baek JH, Gao Y, Morrison EJ, et al. ZOOMICS comparative metabolomics of red blood cells from old world monkeys and humans. Front Physiol. 2020;11:593841. doi: 10.3389/fphys.2020.593841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Stefanoni D, Shin HKH, Baek JH, Champagne DP, Nemkov T, Thomas T, et al. Red blood cell metabolism in Rhesus macaques and humans comparative biology of blood storage. Haematologica. 2020 Aug;105((8)):2174–2186. doi: 10.3324/haematol.2019.229930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Bertolone L, Shin HKH, Baek JH, Gao Y, Spitalnik SL, Buehler PW, et al. ZOOMICS comparative metabolomics of red blood cells from Guinea pigs, humans, and non-human primates during refrigerated storage for up to 42 days. Front Physiol. 2022;13:845347. doi: 10.3389/fphys.2022.845347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Miglio A, Maslanka M, Di Tommaso M, Rocconi F, Nemkov T, Buehler PW, et al. ZOOMICS comparative metabolomics of red blood cells from dogs, cows, horses and donkeys during refrigerated storage for up to 42 days. Blood Transfus. 2022 Jul 25; doi: 10.2450/2022.0118-22. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Nemkov T, Hansen KC, D'Alessandro A. A three-minute method for high-throughput quantitative metabolomics and quantitative tracing experiments of central carbon and nitrogen pathways. Rapid Commun Mass Spectrom. 2017 Apr 30;31((8)):663–673. doi: 10.1002/rcm.7834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Nemkov T, Reisz JA, Gehrke S, Hansen KC, D'Alessandro A. High-throughput metabolomics isocratic and gradient mass spectrometry-based methods. Methods Mol Biol. 2019;1978:13–26. doi: 10.1007/978-1-4939-9236-2_2. [DOI] [PubMed] [Google Scholar]
- 50.Reisz JA, Zheng C, D'Alessandro A, Nemkov T. Untargeted and semi-targeted lipid analysis of biological samples using mass spectrometry-based metabolomics. Methods Mol Biol. 2019;1978:121–135. doi: 10.1007/978-1-4939-9236-2_8. [DOI] [PubMed] [Google Scholar]
- 51.Nemkov T, Stefanoni D, Bordbar A, Issaian A, Palsson BO, Dumont LJ, et al. Blood donor exposome and impact of common drugs on red blood cell metabolism. JCI Insight. 2021;6((3)):e146175. doi: 10.1172/jci.insight.146175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.D'Alessandro A, Thomas T, Dzieciatkowska M, Hill RC, Francis RO, Hudson KE, et al. Serum proteomics in COVID-19 patients altered coagulation and complement status as a function of IL-6 level. J Proteome Res. 2020 Aug 14;19:4417–4427. doi: 10.1021/acs.jproteome.0c00365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Freedman R, Hunter SK, Law AJ, D'Alessandro A, Noonan K, Wyrwa A, et al. Maternal choline and respiratory coronavirus effects on fetal brain development. J Psychiatr Res. 2020 May 25;128:1–4. doi: 10.1016/j.jpsychires.2020.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Thomas T, Stefanoni D, Dzieciatkowska M, Issaian A, Nemkov T, Hill RC, et al. Evidence for structural protein damage and membrane lipid remodeling in red blood cells from COVID-19 patients. medRxiv. 2020 Jun 30; doi: 10.1021/acs.jproteome.0c00606. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Thomas T, Stefanoni D, Reisz JA, Nemkov T, Bertolone L, Francis RO, et al. COVID-19 infection alters kynurenine and fatty acid metabolism correlating with IL-6 levels and renal status. JCI Insight. 2020 Jul 23;5((14)):e140327. doi: 10.1172/jci.insight.140327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.D'Alessandro A, Thomas T, Akpan IJ, Reisz JA, Cendali FI, Gamboni F, et al. Biological and clinical factors contributing to the metabolic heterogeneity of hospitalized patients with and without COVID-19. Cells. 2021 Sep 2;10((9)):2293. doi: 10.3390/cells10092293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Galbraith MD, Kinning KT, Sullivan KD, Baxter R, Araya P, Jordan KR, et al. Seroconversion stages COVID19 into distinct pathophysiological states. Elife. 2021 Mar 16;10:e65508. doi: 10.7554/eLife.65508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Sullivan KD, Galbraith MD, Kinning KT, Bartsch KW, Levinsky NC, Araya P, et al. The COVIDome Explorer researcher portal. Cell Rep. 2021 Aug 17;36((7)):109527. doi: 10.1016/j.celrep.2021.109527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Guntur VP, Nemkov T, de Boer E, Mohning MP, Baraghoshi D, Cendali FI, et al. Signatures of mitochondrial dysfunction and impaired fatty acid metabolism in plasma of patients with post-acute sequelae of COVID-19 (PASC) Metabolites. 2022;12((11)):1026. doi: 10.3390/metabo12111026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.D'Alessandro A, D'Amici GM, Vaglio S, Zolla L. Time-course investigation of SAGM-stored leukocyte-filtered red bood cell concentrates from metabolism to proteomics. Haematologica. 2012 Jan;97((1)):107–115. doi: 10.3324/haematol.2011.051789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.D'Alessandro A, Hansen KC, Silliman CC, Moore EE, Kelher M, Banerjee A. Metabolomics of AS-5 RBC supernatants following routine storage. Vox Sang. 2015 Feb;108((2)):131–140. doi: 10.1111/vox.12193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.D'Alessandro A, Nemkov T, Hansen KC, Szczepiorkowski ZM, Dumont LJ. Red blood cell storage in additive solution-7 preserves energy and redox metabolism a metabolomics approach. Transfusion. 2015 Dec;55((12)):2955–2966. doi: 10.1111/trf.13253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.D'Alessandro A, Nemkov T, Kelher M, West FB, Schwindt RK, Banerjee A, et al. Routine storage of red blood cell (RBC) units in additive solution-3 a comprehensive investigation of the RBC metabolome. Transfusion. 2015 Jun;55((6)):1155–1168. doi: 10.1111/trf.12975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.D'Alessandro A, Dzieciatkowska M, Hill RC, Hansen KC. Supernatant protein biomarkers of red blood cell storage hemolysis as determined through an absolute quantification proteomics technology. Transfusion. 2016 Jun;56((6)):1329–1339. doi: 10.1111/trf.13483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.D'Alessandro A, Reisz JA, Culp-Hill R, Korsten H, van Bruggen R, de Korte D. Metabolic effect of alkaline additives and guanosine/gluconate in storage solutions for red blood cells. Transfusion. 2018 Apr 6;58((8)):1992–2002. doi: 10.1111/trf.14620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Hess, D'Alessandro A. In Rossi's Principles of Transfusion Medicine. Hoboken (NJ): John Wiley & Sons, Incorporated; 2022. Red blood cell metabolism and preservation; pp. 143–157. [Google Scholar]
- 67.Paglia G, D'Alessandro A, Rolfsson O, Sigurjonsson OE, Bordbar A, Palsson S, et al. Biomarkers defining the metabolic age of red blood cells during cold storage. Blood. 2016 Sep 29;128((13)):e43–e50. doi: 10.1182/blood-2016-06-721688. [DOI] [PubMed] [Google Scholar]
- 68.D'Alessandro A, Nemkov T, Reisz J, Dzieciatkowska M, Wither MJ, Hansen KC. Omics markers of the red cell storage lesion and metabolic linkage. Blood Transfus. 2017 Mar;15((2)):137–144. doi: 10.2450/2017.0341-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.van 't Erve TJ, Wagner BA, Martin SM, Knudson CM, Blendowski R, Keaton M, et al. The heritability of metabolite concentrations in stored human red blood cells. Transfusion. 2014;54((8)):2055–2063. doi: 10.1111/trf.12605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Van 't Erve TJ, Wagner BA, Martin SM, Knudson CM, Blendowski R, Keaton M, et al. The heritability of hemolysis in stored human red blood cells. Transfusion. 2015 Jun;55((6)):1178–1185. doi: 10.1111/trf.12992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.de Wolski K, Fu X, Dumont LJ, Roback JD, Waterman H, Odem-Davis K, et al. Metabolic pathways that correlate with post-transfusion circulation of stored murine red blood cells. Haematologica. 2016 May;101((5)):578–586. doi: 10.3324/haematol.2015.139139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Francis RO, Mahajan S, Rapido F, La Carpia F, Soffing M, Divgi C, et al. Reexamination of the chromium-51-labeled posttransfusion red blood cell recovery method. Transfusion. 2019 Jul;59((7)):2264–2275. doi: 10.1111/trf.15310. [DOI] [PubMed] [Google Scholar]
- 73.Francis RO, D'Alessandro A, Eisenberger A, Soffing M, Yeh R, Coronel E, et al. Donor glucose-6-phosphate dehydrogenase deficiency decreases blood quality for transfusion. J Clin Invest. 2020 May 1;130((5)):2270–2285. doi: 10.1172/JCI133530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Issaian A, Hay A, Dzieciatkowska M, Roberti D, Perrotta S, Darula Z, et al. The interactome of the N-terminus of band 3 regulates red blood cell metabolism and storage quality. haematol. 2021 May 13;106((11)):2971–2985. doi: 10.3324/haematol.2020.278252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Zimring JC, Smith N, Stowell, Johnsen JM, Bell LN, Francis RO, et al. Strain-specific red blood cell storage and eicosanoid generation in a mouse model. Transfusion. 2014 Jan;54((1)):137–148. doi: 10.1111/trf.12264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Howie HL, Hay AM, de Wolski K, Waterman H, Lebedev J, Fu X, et al. Differences in Steap3 expression are a mechanism of genetic variation of RBC storage and oxidative damage in mice. Blood Adv. 2019 Aug 13;3((15)):2272–2285. doi: 10.1182/bloodadvances.2019000605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Stockwell BR. Ferroptosis turns 10 emerging mechanisms, physiological functions, and therapeutic applications. Cell. 2022;185((14)):2401–2421. doi: 10.1016/j.cell.2022.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Passer BJ, Nancy-Portebois V, Amzallag N, Prieur S, Cans C, Roborel de Climens A, et al. The p53-inducible TSAP6 gene product regulates apoptosis and the cell cycle and interacts with Nix and the Myt1 kinase. Proc Natl Acad Sci U S A. 2003 Mar 4;100((5)):2284–2289. doi: 10.1073/pnas.0530298100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Lambe T, Simpson RJ, Dawson S, Bouriez-Jones T, Crockford TL, Lepherd M, et al. Identification of a Steap3 endosomal targeting motif essential for normal iron metabolism. Blood. 2009 Feb 19;113((8)):1805–1808. doi: 10.1182/blood-2007-11-120402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Blanc L, Papoin J, Debnath G, Vidal M, Amson R, Telerman A, et al. Abnormal erythroid maturation leads to microcytic anemia in the TSAP6/Steap3 null mouse model. Am J Hematol. 2015 Mar;90((3)):235–241. doi: 10.1002/ajh.23920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Le Goff S, Boussaid I, Floquet C, Raimbault A, Hatin I, Andrieu-Soler C, et al. p53 activation during ribosome biogenesis regulates normal erythroid differentiation. Blood. 2021 Jan 7;137((1)):89–102. doi: 10.1182/blood.2019003439. [DOI] [PubMed] [Google Scholar]
- 82.Page GP, Kanias T, Guo YJ, Lanteri MC, Zhang X, Mast AE, et al. Multiple-ancestry genome-wide association study identifies 27 loci associated with measures of hemolysis following blood storage. J Clin Invest. 2021 Jul 1;131((13)):e146077. doi: 10.1172/JCI146077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Stolwijk JM, Stefely JA, Veling MT, van 't Erve TJ, Wagner BA, Raife TJ, et al. Red blood cells contain enzymatically active GPx4 whose abundance anticorrelates with hemolysis during blood bank storage. Redox Biol. 2021 Oct;46:102073. doi: 10.1016/j.redox.2021.102073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Nemkov T, Hansen KC, Dumont LJ, D'Alessandro A. Metabolomics in transfusion medicine. Transfusion. 2016 Apr;56((4)):980–993. doi: 10.1111/trf.13442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Rettig MP, Orendorff CJ, Campanella E, Low PS. Effect of pH on the self-association of erythrocyte band 3 in situ. Biochim Biophys Acta. 2001 Nov 1;1515((1)):72–81. doi: 10.1016/s0005-2736(01)00397-2. [DOI] [PubMed] [Google Scholar]
- 86.Campanella ME, Chu H, Low PS. Assembly and regulation of a glycolytic enzyme complex on the human erythrocyte membrane. Proc Natl Acad Sci U S A. 2005 Feb 15;102((7)):2402–2407. doi: 10.1073/pnas.0409741102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Lewis IA, Campanella ME, Markley JL, Low PS. Role of band 3 in regulating metabolic flux of red blood cells. Proc Natl Acad Sci U S A. 2009 Nov 3;106((44)):18515–20. doi: 10.1073/pnas.0905999106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Sun K, Zhang Y, D'Alessandro A, Nemkov T, Song A, Wu H, et al. Sphingosine-1-phosphate promotes erythrocyte glycolysis and oxygen release for adaptation to high-altitude hypoxia. Nat Commun. 2016 Jul 15;7:12086. doi: 10.1038/ncomms12086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Lange PF, Huesgen PF, Nguyen K, Overall CM. Annotating N termini for the human proteome project N termini and Nα-acetylation status differentiate stable cleaved protein species from degradation remnants in the human erythrocyte proteome. J Proteome Res. 2014 Apr 4;13((4)):2028–2044. doi: 10.1021/pr401191w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rinalducci S, Ferru E, Blasi B, Turrini F, Zolla L. Oxidative stress and caspase-mediated fragmentation of cytoplasmic domain of erythrocyte band 3 during blood storage. Blood Transfus. 2012 May;10((Suppl 2)):s55–62. doi: 10.2450/2012.009S. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Rogers SC, Ge X, Brummet M, Lin X, Timm DD, d'Avignon A, et al. Quantifying dynamic range in red blood cell energetics evidence of progressive energy failure during storage. Transfusion. 2021 May;61((5)):1586–1599. doi: 10.1111/trf.16395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Reisz JA, Nemkov T, Dzieciatkowska M, Culp-Hill R, Stefanoni D, Hill RC, et al. Methylation of protein aspartates and deamidated asparagines as a function of blood bank storage and oxidative stress in human red blood cells. Transfusion. 2018 Dec;58((12)):2978–2991. doi: 10.1111/trf.14936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.D'Alessandro A, Hay A, Dzieciatkowska M, Brown BC, Morrison EJ, Hansen KC, et al. Protein-L-isoaspartate O-methyltransferase is required for in vivo control of oxidative damage in red blood cells. Haematologica. 2021 Oct 1;106((10)):2726–2739. doi: 10.3324/haematol.2020.266676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Luzzatto L, Ally M, Notaro R. Glucose-6-phosphate dehydrogenase deficiency. Blood. 2020 Sep 10;136((11)):1225–1240. doi: 10.1182/blood.2019000944. [DOI] [PubMed] [Google Scholar]
- 95.Tzounakas VL, Kriebardis AG, Georgatzakou HT, Foudoulaki-Paparizos LE, Dzieciatkowska M, Wither MJ, et al. Glucose 6-phosphate dehydrogenase deficient subjects may be better “storers” than donors of red blood cells. Free Radic Biol Med. 2016 Jul;96:152–165. doi: 10.1016/j.freeradbiomed.2016.04.005. [DOI] [PubMed] [Google Scholar]
- 96.Tzounakas VL, Kriebardis AG, Georgatzakou HT, Foudoulaki-Paparizos LE, Dzieciatkowska M, Wither MJ, et al. Data on how several physiological parameters of stored red blood cells are similar in glucose 6-phosphate dehydrogenase deficient and sufficient donors. Data Brief. 2016 Sep;8:618–627. doi: 10.1016/j.dib.2016.06.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.D'Alessandro A, Howie HL, Hay AM, Dziewulska KH, Brown BC, Wither MJ, et al. Hematologic and systemic metabolic alterations due to Mediterranean class II G6PD deficiency in mice. JCI Insight. 2021 Jul 22;6((14)):e147056. doi: 10.1172/jci.insight.147056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Surendran P, Stewart ID, Au Yeung VPW, Pietzner M, Raffler J, Wörheide MA, et al. Rare and common genetic determinants of metabolic individuality and their effects on human health. Nat Med. 2022 Nov;28((11)):2321–2332. doi: 10.1038/s41591-022-02046-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Guo Y, Busch MP, Seielstad M, Endres-Dighe S, Westhoff CM, Keating B, et al. Development and evaluation of a transfusion medicine genome wide genotyping array. Transfusion. 2019 Jan;59((1)):101–111. doi: 10.1111/trf.15012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Roubinian NH, Reese SE, Qiao H, Plimier C, Fang F, Page GP, et al. Donor genetic and nongenetic factors affecting red blood cell transfusion effectiveness. JCI Insight. 2022 Jan 11;7((1)):e152598. doi: 10.1172/jci.insight.152598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Moore A, Busch MP, Dziewulska K, Francis RO, Hod EA, Zimring JC, et al. Genome-wide metabolite quantitative trait loci analysis (mQTL) in red blood cells from volunteer blood donors. J Biol Chem. 2022;298((12)):102706. doi: 10.1016/j.jbc.2022.102706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.D'Alessandro A, Fu X, Kanias T, Reisz JA, Culp-Hill R, Guo Y, et al. Donor sex, age and ethnicity impact stored red blood cell antioxidant metabolism through mechanisms in part explained by glucose 6-phosphate dehydrogenase levels and activity. Haematologica. 2020 Apr 2;106((5)) doi: 10.3324/haematol.2020.246603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kanias T, Lanteri MC, Page GP, Guo Y, Endres SM, Stone M, et al. Ethnicity and age are determinants of red blood cell storage and stress hemolysis results of the REDS-III RBC-Omics study. Blood Adv. 2017 Jun 27;1((15)):1132–1141. doi: 10.1182/bloodadvances.2017004820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Roubinian NH, Plimier C, Woo JP, Lee C, Bruhn R, Liu VX, et al. Effect of donor and recipient characteristics on hemoglobin increments following red blood cell transfusion. Blood. 2019 Sep 26;134((13)):1003–1013. doi: 10.1182/blood.2019000773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Mykhailova O, Olafson C, Turner TR, D'Alessandro A, Acker JP. Donor-dependent aging of young and old red blood cell subpopulations metabolic and functional heterogeneity. Transfusion. 2020 Aug 19;60((11)):2633–2646. doi: 10.1111/trf.16017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.D'Alessandro A, Fu X, Reisz JA, Kanias T, Page GP, Stone M, et al. Stored RBC metabolism as a function of caffeine levels. Transfusion. 2020 Jun;60((6)):1197–1211. doi: 10.1111/trf.15813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.D'Alessandro A, Fu X, Reisz JA, Stone M, Kleinman S, Zimring JC, et al. Ethyl glucuronide a marker of alcohol consumption correlates with metabolic markers of oxidant stress but not with hemolysis in stored red blood cells from healthy blood donors. Transfusion. 2020 Jun;60((6)):1183–1196. doi: 10.1111/trf.15811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Stefanoni D, Fu X, Reisz JA, Kanias T, Nemkov T, Page GP, et al. Nicotine exposure increases markers of oxidant stress in stored red blood cells from healthy donor volunteers. Transfusion. 2020 Jun;60((6)):1160–1174. doi: 10.1111/trf.15812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.San-Millan I, Stefanoni D, Martinez JL, Hansen KC, D'Alessandro A, Nemkov T. Metabolomics of endurance capacity in world tour professional cyclists. Front Physiol. 2020;11:578. doi: 10.3389/fphys.2020.00578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Nemkov T, Skinner SC, Nader E, Stefanoni D, Robert M, Cendali F, et al. Acute cycling exercise induces changes in red blood cell deformability and membrane lipid remodeling. Int J Mol Sci. 2021 Jan 18;22((2)):896. doi: 10.3390/ijms22020896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Kim CY, Johnson H, Peltier S, Spitalnik SL, Hod EA, Francis RO, et al. Deuterated linoleic acid attenuates the RBC storage lesion in a mouse model of poor RBC storage. Front Physiol. 2022;13:868578. doi: 10.3389/fphys.2022.868578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Catala A, Stone M, Busch MP, D'Alessandro A. Reprogramming of red blood cell metabolism in Zika virus–infected donors. Transfusion. 2022;62((5)):1045–1064. doi: 10.1111/trf.16851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Recktenwald SM, Simionato G, Lopez MGM, Gamboni F, Dzieciatkowska M, Meybohm P, et al. Cross-talk between red blood cells and plasma influences blood flow and omics phenotypes in severe COVID-19. medRxiv. 2022 doi: 10.7554/eLife.81316. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.D'Alessandro A, Zimring JC, Busch M. Chronological storage age and metabolic age of stored red blood cells are they the same? Transfusion. 2019 May;59((5)):1620–1623. doi: 10.1111/trf.15248. [DOI] [PubMed] [Google Scholar]
- 115.D'Alessandro A, Gray AD, Szczepiorkowski ZM, Hansen K, Herschel LH, Dumont LJ. Red blood cell metabolic responses to refrigerated storage and frozen storage. Transfusion. 2017 Apr;57((4)):1019–1030. doi: 10.1111/trf.14034. [DOI] [PubMed] [Google Scholar]
- 116.Donovan K, Meli A, Cendali F, Park KC, Cardigan R, Stanworth S, et al. Stored blood has compromised oxygen unloading kinetics that can be normalized with rejuvenation and predicted from corpuscular side-scatter. Haematologica. 2022;107((1)):298–302. doi: 10.3324/haematol.2021.279296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.D'Alessandro A, Yoshida T, Nestheide S, Nemkov T, Stocker S, Stefanoni D, et al. Hypoxic storage of red blood cells improves metabolism and post-transfusion recovery. Transfusion. 2020 Apr;60((4)):786–798. doi: 10.1111/trf.15730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Hay A, Dziewulska K, Gamboni F, Nerguizian D, Dzieciatkowska M, Zimring JC, et al. Hypoxic storage of murine red blood cells improves energy metabolism and post-transfusion recoveries. Blood Transfus. 2023;21((1)):50–61. doi: 10.2450/2022.0172-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Nemkov T, Yoshida T, Nikulina M, D'Alessandro A. High-throughput metabolomics platform for the rapid data-driven development of novel additive solutions for blood storage. Front Physiol. 2022;13:833242. doi: 10.3389/fphys.2022.833242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Doctor A. Bio-inspired artificial red blood cell design, pre-clinical results and novel indications. Blood. 2019;134((Suppl. 1)):SCI-4. [Google Scholar]
- 121.Trakarnsanga K, Griffiths RE, Wilson MC, Blair A, Satchwell TJ, Meinders M, et al. An immortalized adult human erythroid line facilitates sustainable and scalable generation of functional red cells. Nat Commun. 2017 Mar 14;8:14750. doi: 10.1038/ncomms14750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Robert M, Laperrousaz B, Piedrahita D, Gautier EF, Nemkov T, Dupuy F, et al. Multiparametric characterization of red blood cell physiology after hypotonic dialysis based drug encapsulation process. Acta Pharm Sin B. 2022 Apr;12((4)):2089–2102. doi: 10.1016/j.apsb.2021.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.D'Alessandro A, Reisz JA, Zhang Y, Gehrke S, Alexander K, Kanias T, et al. Effects of aged stored autologous red blood cells on human plasma metabolome. Blood Adv. 2019 Mar 26;3((6)):884–896. doi: 10.1182/bloodadvances.2018029629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.LaCroix IS, Cohen M, Moore EE, Dzieciatkowska M, Nemkov T, Schaid TR Jr., et al. Omics markers of red blood cell transfusion in trauma. Int J Mol Sci. 2022 Nov 10;23((22)):13815. doi: 10.3390/ijms232213815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Culp-Hill R, Srinivasan AJ, Gehrke S, Kamyszek R, Ansari A, Shah N, et al. Effects of red blood cell (RBC) transfusion on sickle cell disease recipient plasma and RBC metabolism. Transfusion. 2018 Dec;58((12)):2797–2806. doi: 10.1111/trf.14931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Gehrke S, Srinivasan AJ, Culp-Hill R, Reisz JA, Ansari A, Gray A, et al. Metabolomics evaluation of early-storage red blood cell rejuvenation at 4°C and 37°C. Transfusion. 2018;58((8)):1980–1991. doi: 10.1111/trf.14623. [DOI] [PubMed] [Google Scholar]
- 127.Rabcuka J, Blonski S, Meli A, Sowemimo-Coker S, Zaremba D, Stephenson D, et al. Metabolic reprogramming under hypoxic storage preserves faster oxygen unloading from stored red blood cells. Blood Adv. 2022;6((18)):5415–5428. doi: 10.1182/bloodadvances.2022007774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.de Bruin S, Peters AL, Wijnberge M, van Baarle FEHP, AbdelRahman AHA, Vermeulen C, et al. Storage of red blood cells in alkaline PAGGGM improves metabolism but has no effect on recovery after transfusion. Blood Adv. 2022 Jul 12;6((13)):3899–3910. doi: 10.1182/bloodadvances.2022006987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.D'Alessandro A, Slaughter AL, Peltz ED, Moore EE, Silliman CC, Wither M, et al. Trauma/hemorrhagic shock instigates aberrant metabolic flux through glycolytic pathways as revealed by preliminary (13)C-glucose labeling metabolomics. J Transl Med. 2015 Aug 5;13((1)):253. doi: 10.1186/s12967-015-0612-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Slaughter AL, D'Alessandro A, Moore EE, Banerjee A, Silliman CC, Hansen KC, et al. Glutamine metabolism drives succinate accumulation in plasma and the lung during hemorrhagic shock. J Trauma Acute Care Surg. 2016 Dec;81((6)):1012–1019. doi: 10.1097/TA.0000000000001256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Peltz ED, D'Alessandro A, Moore EE, Chin T, Silliman CC, Sauaia A, et al. Pathologic metabolism an exploratory study of the plasma metabolome of critical injury. J Trauma Acute Care Surg. 2015 Apr;78((4)):742–751. doi: 10.1097/TA.0000000000000589. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Wiener G, Moore HB, Moore EE, Gonzalez E, Diamond S, Zhu S, et al. Shock releases bile acid inducing platelet inhibition and fibrinolysis. J Surg Res. 2015 May 15;195((2)):390–395. doi: 10.1016/j.jss.2015.01.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.D'Alessandro A, Moore HB, Moore EE, Wither MJ, Nemkov T, Morton AP, et al. Plasma first resuscitation reduces lactate acidosis enhances redox homeostasis amino acid and purine catabolism in a rat model of profound hemorrhagic shock. Shock. 2016 Aug;46((2)):173–182. doi: 10.1097/SHK.0000000000000588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Clendenen N, Nunns GR, Moore EE, Reisz JA, Gonzalez E, Peltz E, et al. Hemorrhagic shock and tissue injury drive distinct plasma metabolome derangements in swine. J Trauma Acute Care Surg. 2017;83((4)):635–642. doi: 10.1097/TA.0000000000001504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Reisz JA, Slaughter AL, Culp-Hill R, Moore EE, Silliman CC, Fragoso M, et al. Red blood cells in hemorrhagic shock a critical role for glutaminolysis in fueling alanine transamination in rats. Blood Adv. 2017 Jul 25;1((17)):1296–305. doi: 10.1182/bloodadvances.2017007187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Banerjee A, Silliman CC, Moore EE, Dzieciatkowska M, Kelher M, Sauaia A, et al. Systemic hyperfibrinolysis after trauma a pilot study of targeted proteomic analysis of superposed mechanisms in patient plasma. J Trauma Acute Care Surg. 2018 Jun;84((6)):929–938. doi: 10.1097/TA.0000000000001878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Slaughter AL, Nunns GR, D'Alessandro A, Banerjee A, Hansen KC, Moore EE, et al. The metabolopathy of tissue injury hemorrhagic shock and resuscitation in a rat model. Shock. 2018 May;49((5)):580–590. doi: 10.1097/SHK.0000000000000948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Coleman, Moore EE, Kelher MR, Samuels JM, Cohen MJ, Sauaia A, et al. Female platelets have distinct functional activity compared with male platelets implications in transfusion practice and treatment of trauma-induced coagulopathy. J Trauma Acute Care Surg. 2019 Nov;87((5)):1052–1060. doi: 10.1097/TA.0000000000002398. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Henriksen HH, McGarrity S, SigurÐardóttir RS, Nemkov T, D'Alessandro A, Palsson BO, et al. Metabolic systems analysis of shock-induced endotheliopathy (SHINE) in trauma a new research paradigm. Ann Surg. 2020 Dec;272((6)):1140–8. doi: 10.1097/SLA.0000000000003307. [DOI] [PubMed] [Google Scholar]
- 140.Nunns GR, Vigneshwar N, Kelher MR, Stettler GR, Gera L, Reisz JA, et al. Succinate activation of SUCNR1 predisposes severely injured patients to neutrophil-mediated ARDS. Ann Surg. 2022;276((6)):e944–e954. doi: 10.1097/SLA.0000000000004644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Williams AT, Jani VP, Nemkov T, Lucas A, Yoshida T, Dunham A, et al. Transfusion of anaerobically or conventionally stored blood after hemorrhagic shock. Shock. 2020 Mar;53((3)):352–362. doi: 10.1097/SHK.0000000000001386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Cralley AL, Moore EE, Fox CJ, Kissau D, DeBot M, Schaid TR, et al. Zone 1 REBOA in a combat DCBI swine model does not worsen brain injury. Surgery. 2022 Aug;172((2)):751–758. doi: 10.1016/j.surg.2022.04.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Hadley JB, Kelher MR, Coleman, DeBot M, Eitel AP, Moore EE, et al. Testosterone and sex affect platelet responsiveness in vitro. J Am Coll Surgeons. 2021;233((5)):e208–e209. [Google Scholar]
- 144.D'Alessandro A, Nemkov T, Moore HB, Moore EE, Wither M, Nydam T, et al. Metabolomics of trauma-associated death shared and fluid-specific features of human plasma vs lymph. Blood Transfus. 2016 May;14((2)):185–194. doi: 10.2450/2016.0208-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sun K, D'Alessandro A, Ahmed MH, Zhang Y, Song A, Ko TP, et al. Structural and functional insight of sphingosine 1-phosphate-mediated pathogenic metabolic reprogramming in sickle cell disease. Sci Rep. 2017 Nov 10;7((1)):15281. doi: 10.1038/s41598-017-13667-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Gehrke S, Shah N, Gamboni F, Kamyszek R, Srinivasan AJ, Gray A, et al. Metabolic impact of red blood cell exchange with rejuvenated red blood cells in sickle cell patients. Transfusion. 2019 Oct;59((10)):3102–3112. doi: 10.1111/trf.15467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Buehler PW, Swindle D, Pak DI, Fini MA, Hassell K, Nuss R, et al. Murine models of sickle cell disease and beta-thalassemia demonstrate pulmonary hypertension with distinctive features. Pulm Circ. 2021 Oct-Dec;11((4)):20458940211055996. doi: 10.1177/20458940211055996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Moriconi C, Dzieciatkowska M, Roy M, D'Alessandro A, Roingeard P, Lee JY, et al. Retention of functional mitochondria in mature red blood cells from patients with sickle cell disease. Br J Haematol. 2022 Aug;198((3)):574–586. doi: 10.1111/bjh.18287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Nemkov T, Skinner S, Diaw M, Diop S, Samb A, Connes P, et al. Plasma levels of acyl-carnitines and carboxylic acids correlate with cardiovascular and kidney function in subjects with sickle cell trait. Front Physiol. 2022;13:916197. doi: 10.3389/fphys.2022.916197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Song A, Wen AQ, Wen YE, Dzieciatkowska M, Kellems RE, Juneja HS, et al. p97 dysfunction underlies a loss of quality control of damaged membrane proteins and promotes oxidative stress and sickling in sickle cell disease. FASEB J. 2022 May;36((5)):e22246. doi: 10.1096/fj.202101500RR. [DOI] [PubMed] [Google Scholar]
- 151.Tzounakas VL, Anastasiadi AT, Dzieciatkowska M, Karadimas DG, Stamoulis K, Papassideri IS, et al. Proteome of stored RBC membrane and vesicles from heterozygous beta thalassemia donors. Int J Mol Sci. 2021 Mar 25;22((7)):3369. doi: 10.3390/ijms22073369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Tzounakas VL, Anastasiadi AT, Stefanoni D, Cendali F, Bertolone L, Gamboni F, et al. Beta thalassemia minor is a beneficial determinant of red blood cell storage lesion. Haematologica. 2022;107((1)):112–125. doi: 10.3324/haematol.2020.273946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Anastasiadi AT, Tzounakas VL, Dzieciatkowska M, Arvaniti V-Z, Papageorgiou EG, Papassideri IS, et al. Innate variability in physiological and omics aspects of the beta thalassemia trait-specific donor variation effects. Front Physiol. 2022;13:907444. doi: 10.3389/fphys.2022.907444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Roy MK, Cendali F, Ooyama G, Gamboni F, Morton H, D'Alessandro A. Red blood cell metabolism in pyruvate kinase deficient patients. Front Physiol. 2021;12:735543. doi: 10.3389/fphys.2021.735543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Roy MK, Cendali FI, Ooyama G, Gamboni F, Morton H, D'Alessandro A. Red blood cell metabolism in patients with propionic acidemia. Separations. 2021;8((9)):142. doi: 10.3389/fphys.2021.735543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Culp-Hill R, Zheng C, Reisz JA, Smith K, Rachubinski A, Nemkov T, et al. Red blood cell metabolism in Down syndrome hints on metabolic derangements in aging. Blood Adv. 2017 Dec 26;1((27)):2776–2780. doi: 10.1182/bloodadvances.2017011957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Powers RK, Culp-Hill R, Ludwig MP, Smith KP, Waugh KA, Minter R, et al. Trisomy 21 activates the kynurenine pathway via increased dosage of interferon receptors. Nat Commun. 2019 Oct 18;10((1)):4766. doi: 10.1038/s41467-019-12739-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Kalani Roy M, Wilkerson RB, Alexander K, Nokoff NJ, Cree-Green M, D'Alessandro A. Longitudinal metabolic study of red blood cells from patients undergoing gender-affirming testosterone therapy. Blood Adv. 2022 doi: 10.1182/bloodadvances.2022008061. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Alexander K, Hazegh K, Fang F, Sinchar D, Kiss JE, Page GP, et al. Testosterone replacement therapy in blood donors modulates erythrocyte metabolism and susceptibility to hemolysis in cold storage. Transfusion. 2021 Jan;61((1)):108–123. doi: 10.1111/trf.16141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Blasi B, D'Alessandro A, Ramundo N, Zolla L. Red blood cell storage and cell morphology. Transfus Med. 2012;22((2)):90–96. doi: 10.1111/j.1365-3148.2012.01139.x. [DOI] [PubMed] [Google Scholar]
- 161.Rapido F, Brittenham GM, Bandyopadhyay S, La Carpia F, L'Acqua C, McMahon DJ, et al. Prolonged red cell storage before transfusion increases extravascular hemolysis. J Clin Invest. 2017 Jan 3;127((1)):375–382. doi: 10.1172/JCI90837. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Youssef LA, Rebbaa A, Pampou S, Weisberg SP, Stockwell BR, Hod EA, et al. Increased erythrophagocytosis induces ferroptosis in red pulp macrophages in a mouse model of transfusion. Blood. 2018;131((23)):2581–2593. doi: 10.1182/blood-2017-12-822619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Himbert S, Qadri SM, Sheffield WP, Schubert P, D'Alessandro A, Rheinstädter MC. Blood bank storage of red blood cells increases RBC cytoplasmic membrane order and bending rigidity. PLoS One. 2021;16((11)):e0259267. doi: 10.1371/journal.pone.0259267. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Himbert S, D'Alessandro A, Qadri SM, Majcher MJ, Hoare T, Sheffield WP, et al. The bending rigidity of the red blood cell cytoplasmic membrane. PLoS One. 2022;17((8)):e0269619. doi: 10.1371/journal.pone.0269619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Bordbar A, Monk JM, King ZA, Palsson BO. Constraint-based models predict metabolic and associated cellular functions. Nat Rev Genet. 2014 Feb;15((2)):107–120. doi: 10.1038/nrg3643. [DOI] [PubMed] [Google Scholar]
- 166.Yurkovich JT, Palsson BO. Quantitative -omic data empowers bottom-up systems biology. Curr Opin Biotechnol. 2018 Jun;51:130–136. doi: 10.1016/j.copbio.2018.01.009. [DOI] [PubMed] [Google Scholar]
- 167.Yurkovich JT, Bordbar A, Sigurjónsson ÓE, Palsson BO. Systems biology as an emerging paradigm in transfusion medicine. BMC Syst Biol. 2018 Mar 7;12((1)):31. doi: 10.1186/s12918-018-0558-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Jamshidi N, Xu X, von Löhneysen K, Soldau K, Mohney RP, Karoly ED, et al. Metabolome changes during in vivo red cell aging reveal disruption of key metabolic pathways. iScience. 2020;23((10)):101630. doi: 10.1016/j.isci.2020.101630. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Kelly CJ, Karthikesalingam A, Suleyman M, Corrado G, King D. Key challenges for delivering clinical impact with artificial intelligence. BMC Med. 2019;17((1)):195. doi: 10.1186/s12916-019-1426-2. [DOI] [PMC free article] [PubMed] [Google Scholar]