The collection and storage of red blood cells (RBCs) is a logistical necessity to provide sufficient blood products. However, RBC storage is an unnatural state, resulting in complicated biological changes, referred to collectively as the “storage lesion.” Specifics of the storage lesion have been studied for decades, including alterations to cellular properties, morphology, molecular biology of carbohydrates, proteins and lipids, and basic metabolism. Recently, mass spectrometry–based “omics” technology has been applied to the RBC storage lesion, resulting in many new observations, the initial effects of which are more information than understanding. Meanwhile, clinical research on RBC transfusion is considering both the efficacy and also the potential untoward effects of transfusing stored RBCs of different ages and storage conditions. The myriad biological changes that have now been observed during the storage lesion have been extensively reviewed elsewhere. This article focuses rather on an analysis of our current understanding of the biological effects of different elements of the storage lesion, in the context of evolving new clinical understanding. A synopsis is presented of both established and theoretical considerations of the RBC storage lesion and ongoing efforts to create a safer and more efficacious product.

The practice of storing red blood cells (RBCs) prior to transfusion has been in place for close to a century.1,2  Indeed, the storage and banking of RBCs is a necessity to provide a sufficient volume of RBCs for patient needs. However, banking blood is an unnatural state for RBCs to exist: sitting in the static environment of a refrigerator shelf, in a solution of glucose, citrate, and other additives, in the absence of a kidney or liver to detoxify products of ongoing metabolism and likewise with no reticuloendothelial system to remove senescent or damaged RBCs. Moreover, additional factors are introduced, such as plasticizers from the bag, which both inadvertently not only improve RBC integrity3,4  but also introduce concerns regarding toxicity.5,6  In this artificial environment, it is thus not surprising that stored RBCs undergo myriad changes, which are in aggregate referred to as the “storage lesion.”

In recent years, the scope of clinical concern regarding transfusion of stored RBCs has widened from traditional issues of replacing lost RBCs with stored RBCs that could deliver oxygen to peripheral tissues, to concerns regarding the accumulation of toxicological entities in stored RBCs that could lead to medical sequelae upon transfusion. Much like the difficulties in studying tissue oxygenation by RBCs, analyzing sequelae of transfusing stored RBCs (if such sequelae even exist) is a very challenging process.7  Retrospective studies have been useful, but give a wide variety of results, with a large number of studies showing worse medical outcomes as a function of RBC storage time, others showing no effect, and some reporting worse outcomes with fresh RBCs. Although of great potential utility from the standpoint of using existing medical data to generate observational hypotheses, retrospective studies inevitably suffer a series of intrinsic biases. For this reason, several prospective randomized clinical trials (RCTs) were launched,8  3 of which have now reported no difference between groups receiving fresher vs older blood. The Age of Red Blood Cells in Premature Infants (ARIPI) study detected no difference in fresher vs older RBC units in very low-birth-weight infants,9  whereas the Red Cell Storage Duration Study (RECESS)10  recently reported no difference in cardiac surgery patients. Likewise, the Age of Blood Evaluation (ABLE) trial recently reported no difference in intensive care unit patients.11  Formal publication of the results of RECESS and ABLE (outside of abstract form) have not yet occurred and additional trials remain under way.8,12 

RCTs are of tremendous use in helping to resolve this important issue. Although it is inevitable that some challenges to methodologies and analysis will be raised, these trials nevertheless provide very reassuring data indicating that large effects are not present in the particular patient populations that have been studied, with the RBC storage methodologies being used, and with the definitions of old vs fresh blood that were used. However, an additional concern remains, due to the tremendous number of transfused patients. In the United States alone, ∼5 000 000 patients receive an RBC transfusion each year; thus, a difference as little as 1% would equate to 50 000 patients annually. Regrettably, the cost and logistical difficulties of carrying out RCTs is such that trials powered to detect such small outcomes are unlikely to be forthcoming.13  Conversely, this means that large and clinically relevant improvements would need to be made to RBC storage conditions before any new RBC products could be demonstrated to be superior to current products based upon RCTs.

There are substantial data to indicate that among the substances that accumulate during RBC storage, a number of entities are generated with both theoretical and actual toxicology in the context of in vitro human and whole animal models. Such substances include:

  • scavengers of nitric oxide (NO) and inhibitors of NO generation that may result in lack of vascular relaxation (eg, hemoglobin, both free and in microparticles,14  alterations in RBCs themselves,15  and asymmetric dimethylarginine that may inhibit NO synthase16,17 );

  • microparticles and RBCs themselves that may have procoagulant effects18-21  and/or other effects on innate and/or adaptive immunity and also physiology;22,23 

  • free and nontransferrin bound iron that may promote growth of siderophilic bacteria;24-30 

  • clearance of damaged RBCs that may result in activation of innate immunity;27,31 

  • the generation of bioactive lipids that prime neutrophils and may contribute to transfusion-related acute lung injury,32-34  and

  • cytokines and chemokines, especially in units that are not filter leukoreduced.35,36 

In some cases, the biological effects of stored/aged blood in whole animal models has resulted in profound pathology and pathophysiology, with substantial morbidity and mortality of recipients of the stored (but not fresh) units of blood.24,27,29,30,37,38  Such animal studies must be interpreted with a number of caveats, as animal biology may not reflect human biology and such studies are generated to maximize potential effects (ie, recipients of stored RBCs get all units at outdate and receive large volumes of blood). Like any system, the toxicology of animal studies has titratable dose response curves, and toxicological effects are confined to dose ranges that likewise may not translate into most humans.

The ARIPI, RECESS, and ABLE trials described above bring substantial intellectual comfort that within the context of our current practice and standard of care, there are not large effects between groups receiving “fresher” vs “older” stored RBCs. Nevertheless, these RCTs must likewise be interpreted with their own caveats, including:

  1. the limited power to detect small effects (even below 20% depending upon the nature of the storage lesion),13 

  2. the fact that few patients in these trials received multiple units of blood at outdate but, at some frequency, that such patients likely exist in real practice, and

  3. that just as animal biology may not reflect human biology, the clinical susceptibilities of the patients in the existing RCTs may not reflect susceptibilities in other patient populations.

Thus, both the animal studies and the human RCTs are compelling and essential parts of an evolving landscape of data, and must be given the intellectual influence and weight they are due, without overinferring or generalizing to unobserved situations to which they may not apply.39 

Primary metrics that have historically guided development and refinement of RBC storage conditions have focused on RBC cellular integrity. RBCs that hemolyze in the storage bag are clearly no longer viable as a therapy for anemia; thus, the US Food and Drug Administration (FDA) requires that on average, hemolysis in the bag be <1%. However, just because an RBC is intact does not mean it will circulate upon transfusion. Accordingly, the number of RBCs that remain in circulation 24 hours posttransfusion has been a primary criteria for the conditions and length of RBC storage that the FDA will approve (there must be a mean “24-hour recovery” of at least 75% with a standard deviation of <9% and with a 1-sided 95% lower confidence limit for the population proportion of successes of >70%). The cutoff of 75% is based upon historical expert opinion, but is nevertheless an arbitrary metric.40-43  The rational basis for the 24-hour recovery is that the removal of irrevocably damaged RBCs appears to occur within this 24-hour window, and RBCs which are still circulating 24 hours posttransfusion have a normal RBC circulatory lifespan.42 

It seems a fair statement that RBCs which do not circulate cannot deliver oxygen; thus, using 24-hour recoveries is a meaningful metric. However, even the word “circulate” requires certain biological scrutiny in this context. It might seem a reasonable conclusion that if a labeled RBC is injected IV and 24 hours later is recovered from a peripheral blood draw, it has been circulating. It might also seem a reasonable conclusion that if 75% of transfused RBCs are recovered 24 hours after infusion, then 25% of the transfused RBCs are no longer circulating. However, both of these assumptions have undergone a critical evaluation. With regards to the first notion, it has been rightly pointed out that the flow dynamics of large vessels are profoundly different than issues of the microvasculature of capillary beds. As the vast majority of oxygen exchange occurs in the microvasculature embedded in organ parenchyma, it is the actual behavior of RBCs in microcirculation that likely affect oxygen delivery capacity. It is for this reason that a great deal of study has gone into the rheological properties of RBCs, using a variety of instruments that measure RBC flexibility, the ability to bend and contort, and the ability to navigate artificial microvasculature with a variety of properties meant to model capillaries. Indeed, stored RBCs show progressively diminished flexibility and ability to perform the contortions necessary for microcirculation44-46  and even show negative effects upon circulation itself46 ; although studies to the contrary have also been reported.47  Moreover, transfusion of RBCs stored for longer periods has a statistically significant inverse correlation with both thenar muscle tissue oxygen saturation and perfused capillary vascular density in trauma patients.48  With regard to the assumption that RBCs that are no longer detected in venous blood are no longer circulating, it is important to note that some transfused RBCs may not be detected in peripheral circulation until over 24 hours posttransfusion, presumably due to some manner of adhesion or sequestration.49 

Regrettably, just because an RBC can circulate (even through the microvasculature), does not mean that it is in fact capable of delivering oxygen with normal efficiency. Indeed, there are data to indicate that stored RBCs may not optimally deliver oxygen until they have “recovered” after a period of time circulating in the recipient.50  This issue remains a matter of some dispute, with evidence on both sides; however, what the experimental data do demonstrate is that RBCs are capable of a phenotype in which they circulate but have decreased efficiency of oxygen delivery. Thus, this must remain a concern in guiding development of RBC storage conditions and has led to the view by some that stored RBCs do not function well. An argument that is often forwarded to the contrary is that many trauma patients who have had their entire blood volume replaced with stored RBCs survive, and patients requiring exchange transfusion do better clinically, indicating that stored RBCs can function to a reasonable extent. This seems undeniably true, and is a credit to the efficacy of transfusing stored RBCs. However, the fact that patients are routinely transfused with stored RBCs and many have good outcomes cannot logically justify the claim that stored RBCs function optimally nor does it demonstrate that stored RBCs are not damaging to patients in some ways.

Early characterization of the storage lesion focused on a number of parameters for which there was a rational basis to assume that they would reflect RBC function, including: (1) metabolic changes (eg, adenosine triphosphate [ATP], 2,3-diphosphoglycerate, etc), (2) enzymatic changes, (3) changes in rheological properties, and (4) physiologic changes (eg, shape change, membrane remodeling), which have been recently reviewed.51,52  Oxidative changes have also been thoroughly characterized, have been shown to correlate in many ways with listed items 1 through 4, and have been posited as playing a causal role in the RBC storage lesion.51,52  However, although it seems a reasonable prediction that some of the above metrics would predict how stored RBCs would perform after transfusion, such is regrettably not the case.

A major challenge to the field has been that although the above measures do reflect observable alterations in RBCs during storage, they have only an “asymmetric correlation” to the ability of RBCs to survive storage (as measured by 24-hour recoveries). In other words, RBCs with extreme changes can be shown to have poor 24-hour recoveries; however, none or a small amount of change does not necessarily predict good 24-hour recoveries. Such is even the case for ATP levels, which have been a mainstay measure of RBC storage quality and have guided the addition of adenine to RBC storage solutions. Indeed, ATP at the end of storage has been the best predictor of 24-hour RBC recoveries; however, even ATP has only a 40% correlation with 24-hour RBC recoveries (with a mean 75% to 80% recovery).53  This poor correlation may be in part due to intrinsic difficulties in measurements of high accuracy,54  but may likewise reflect a lack of biological correlation. The lack of correlation between in vitro measures and in vivo circulation raises the serious concern that current metrics may be untethered to relevant biology.

In more recent decades, the concept of in vivo RBC function has evolved well beyond the issue of oxygen delivery. The current paradigms now include a role of RBCs as not only a vehicle of oxygen delivery, but also as both an agent of CO2 removal, a sensor of tissue oxygenation, and a regulator of biological processes extrinsic to the RBC (eg, vascular tone through NO biology).55  Thus, additional metrics of RBC storage have been added to the above-mentioned details, including distinguishing hemoglobin outside of intact RBCs (eg, either free or in the form of microparticles that may scavenge NO), the amount of S-nitrosylation of hemoglobin itself (SNO-Hg),55  adhesion of RBCs to endothelial beds,56  effects of stored RBCs on models of vascular tone (eg, isolated aortic rings),57  and in vivo measures of how stored RBCs affect vascular tone, both in animals and directly in humans.58  However, as with the long list of more traditional measures, the clinical meaning of these indications remains largely undetermined (and in some cases controversial) and the ability to predict RBC function is unclear.

The era of “omics” biology has now made its way to the study of the RBC storage lesion.59  As in many fields, the application of “big data” platforms (eg, proteomics, lipomics, glycomics, and metabolomics) has led to a progression toward a seemingly encyclopedic list of the changes that take place to RBCs over 42 days of storage.34,60-71  Genomics of RBC storage is a bit more abstract, as mature RBCs do not have nuclei and do not express gene products; however, it is well known that there is substantial variation in RBC storage across donors and it has also been reported that some characteristics of RBC storage are heritable traits in humans.68,69  The generation of such large quantities of data are of great potential, and have led to the ability to rapidly characterize storage biology to greater depths. These studies have detected an entire panoply of new molecular changes that correlate with RBC age, as well as confirming historical observations. Importantly, such data have also allowed systems biology approaches to RBC storage biology, which will likely be of great scientific utility moving forward.72  However, like the older measures, it is as yet unclear which, if any, of the new measurements correlate with the in vivo behavior of the RBCs. Thus, for the time being (at least), the generation of large volumes of new data has only exacerbated the central problem of which variables to study. Thus, a downside to the new data are that given the thousands of changes RBCs undergo during storage, and the experimental difficulty (or impossibility) of changing and testing a given variable in isolation from other variables, the actual significance of any of the observed changes remains unassessed by experimental methodologies and thus remains somewhat obscure.

So, the question is raised, how will the field move forward in sorting out the proverbial wheat from the chaff? One pathway is through experimental modification of storage solutions that give a better RBC product, as assessed by traditional metrics, if not better medical outcomes. Analysis of what variables correlate with a better RBC storage solution, and which do not, will not isolate individual changes, but will nevertheless help to establish the functional significance of at least different groups of changes. Controlled animal experimentation will also be of use in helping to isolate pathways, and will certainly provide a much greater degree of experimental control than can be ethically or practically achieved in humans, with the decidedly negative bedfellow that animal RBC biology will differ from human RBC biology, to an as yet unknown extent.39  Thus, animal studies will likely find their greatest utility in sorting through the multiplicity of known changes in discovery-phase searches that will then allow more focused hypothesis testing in humans. Of central importance will be the combination of omics analysis with units of RBCs that are subsequently transfused into humans, and at the very least subjected to traditional measures of in the bag hemolysis and 24-hour recoveries. First steps to this approach have been taken in platelet storage biology, and have isolated initial candidates for changes that predict posttransfusion performance.73-76  Similar approaches to RBC storage are likely to bear similar fruits, and seem a logical next step.

It is clear that certain storage lesion–based changes correlate with poor RBC performance (eg, hemolysis, extreme changes in morphology [echinospherocytosis], high levels of protein and lipid oxidation, loss of sialic acid, cross-linking of hemoglobin to itself and other proteins, clustering of band 3, etc). The significance of such changes may be somewhat exaggerated in the context of confusing the distinction between that which can occur and that which clearly does occur. Just as one example, the observation that transfused RBCs clear rapidly if first treated with neuraminidase (which removes terminal sialic acids from glycans) does indicate that if sialic acids are lost then RBCs will clear; however, this does not equate to establishing that the degree of spontaneous sialic acid loss during storage is sufficient to result in RBC clearance. This is an issue that afflicts essentially all of the proposed mechanisms of poststorage clearance of damaged/aged RBCs. Unless and until a variable can be reversed in isolation from other variables, and the effects of such reversal can be assessed in vivo, then the requirement of a given pathway cannot be firmly tested. Such maneuvers are technically challenging, if not impossible, given the massive number of changes that are known to take place during RBC storage, and present a substantial challenge even in tractable animal models, let alone the technical and ethical challenges of human studies. Moreover, even if such experimental sophistication were achievable, it can only answer questions of what is “required” but cannot exclude what is “involved” due to issues of biological redundancy. Indeed, the lack of clear understanding that has been perpetuated by this obstacle is not limited to clearance of RBCs poststorage; rather, it seems a fair statement that discrete answers to the question of how RBCs are cleared in vivo as a part of normal RBC senescence remain undetermined. As with RBC storage, multiple pathways have been proposed and demonstrated as being possible, but identification of those that are clearly involved and/or essential has been elusive.

An additional concern to the issue of sorting out the storage lesion and its effects on RBC efficacy and/or toxicity is the large number of approved RBC products and the myriad medical conditions for which stored RBCs are transfused. RBCs may be anticoagulated in CPD and CP2D (and for apheresis in ACD); units may also be supplemented with a variety of additive solutions (eg, SAGM, ADSOL-1, ADSOL-3, ADSOL-5, ADSOL-7, [SOLX], etc); the different components of these solutions have been recently reviewed.59  However, in general, the solutions differ in concentrations of NaCl, NaHCO3, Na2HPO4, citric acid, sodium citrate, adenine, guanosine, glucose, and mannitol.59  Moreover, RBC units may be leukoreduced or not, depending upon country and region. Finally, there are additional prestorage variables (eg, time at room temperature after collection, irradiation, apheresis vs whole blood, etc) that have not been included in many studies, but for which there is a good faith basis to suggest a role in biology.77  Indeed, differences in storage solutions in Europe and Canada vs the United States have been posited as a potential cause for different results of analysis of patient outcomes as a function of transfusing stored RBCs.77  Perhaps ever more confounding is the wide variety of medical indications for which RBCs are given, and how the different, and at times opposing, pathophysiologies may confound observations of RBC efficacy. For instance, it has been observed that stored RBCs develop a “procoagulant activity.”18-21  If such activity correlates to the promotion of clotting in vivo, then such procoagulant activity may simultaneously be therapeutic to a patient suffering hemorrhage and lethal to a patient suffering thrombosis. This is both a concern as far as adjusting medical practice to individual patient needs, but also for the validity of clinical trials in which patients with diseases of different pathologies are included together in groups assessing medical outcomes. In the above context, the general overarching question as stated, “Is older blood bad for you?”, appears to be oversimplified to the point of being neither testable nor applicable, and more focused questions must be asked.

In the field of blood product collection and transfusion, the notion of “process control” is well established and in place to variable degrees in different settings. That is to say that there is an attempt to ensure that all products are collected and distributed within the context of certain controlled parameters. In other words, blood products are all collected and treated the same, and attempts are made to sample a small number of units, from which one extrapolates a general understanding of the quality of a large number of products, which are not themselves directly tested. However, neither the scientific nor technical notion of “product control” has yet been developed or implemented. That is to say, release criteria from the blood bank (for individual units) is limited to testing negative for select pathogens, the recipient not having a detectable alloantibody against the donor unit, the unit meeting process control, and passing a visual inspection.

Of the panoply of changes that RBCs are known to undergo during storage, none is used as a quality control measure at the time of RBC release, on a unit-per-unit basis. As explained in this article, this is in large part because the field has yet to identify storage lesion measures known to predict efficacy or toxicity. However, should such factors be described, then the application to individual units is not necessarily farfetched; in the recent past (prior to electronic cross-match), essentially every unit that was transfused was cross-matched to the individual recipient.

Thus, how are we to answer the question of “What are the established and theoretical factors to consider in assessing the red cell storage lesion?” As detailed in this article and as reviewed extensively elsewhere, there is a well-defined list of factors that are traditional measures of the storage lesion, and a panoply of new and evolving measures, as the omics era makes its way to RBC storage. As of yet, no components of the storage lesion have been identified that adequately or accurately predict 24-hour recoveries. Moreover, although RBC circulation remains a necessary property, it is unclear that 24-hour recoveries predict efficacy beyond eliminating lack thereof due to nonviable RBCs. Because some metrics can predict blood that has lost its integrity, in vitro measures remain a guiding factor in development and refinement of new storage solutions. Nevertheless, the lack of an in vitro metric known to correlate strongly with how well RBCs will circulate posttransfusion, and the lack of knowledge as to whether 24-hour recoveries correlate to medical outcomes, remains a major obstacle. The field has access to the ability to carry out human trials and studies, use animal models, and harness systems biology of RBC storage. Future refinement of methodologies and metrics in the field will necessitate combining these approaches with a distinct focus on identifying the changes that correlate and/or are causally associated with increased efficacy and decrease in untoward effects, with a mindful consideration of different patient characteristics and clinical situations. The identification of distinct components of the storage lesion with such predictive qualities will not only guide clinical care, but also provide a rational basis for rapid storage system development and refinement. In the meantime, the lack of such metrics remains a major challenge to blood storage research.

Contribution: J.C.Z. wrote the manuscript.

Conflict-of-interest disclosure: J.C.Z. has a sponsored research agreement with Immucor Inc unrelated to the topic of the current manuscript.

Correspondence: James C. Zimring, Puget Sound Blood Center Research Institute, 1551 Eastlake Ave E, Seattle, WA 98102; e-mail: jzimring@psbc.org.

1
Weil
 
R
Sodium citrate in the transfusion of blood.
JAMA
1915
, vol. 
LXIV
 
5
(pg. 
425
-
426
)
2
Rous
 
P
Turner
 
JR
The preservation of living red blood cells in vitro. II. The transfusion of kept cells.
J Exp Med
1916
, vol. 
23
 
2
(pg. 
239
-
248
)
3
Draper
 
CJ
Greenwalt
 
TJ
Dumaswala
 
UJ
Biochemical and structural changes in RBCs stored with different plasticizers: the role of hexanol.
Transfusion
2002
, vol. 
42
 
7
(pg. 
830
-
835
)
4
Hill
 
HR
Oliver
 
CK
Lippert
 
LE
Greenwalt
 
TJ
Hess
 
JR
The effects of polyvinyl chloride and polyolefin blood bags on red blood cells stored in a new additive solution.
Vox Sang
2001
, vol. 
81
 
3
(pg. 
161
-
166
)
5
Dumont
 
LJ
Baker
 
S
Dumont
 
DF
, et al. 
Exploratory in vitro study of red blood cell storage containers formulated with an alternative plasticizer.
Transfusion
2012
, vol. 
52
 
7
(pg. 
1439
-
1445
)
6
Prowse
 
CV
de Korte
 
D
Hess
 
JR
van der Meer
 
PF
Biomedical Excellence for Safer Transfusion (BEST) Collaborative
Commercially available blood storage containers.
Vox Sang
2014
, vol. 
106
 
1
(pg. 
1
-
13
)
7
Glynn
 
SA
The red blood cell storage lesion: a method to the madness.
Transfusion
2010
, vol. 
50
 
6
(pg. 
1164
-
1169
)
8
Zimring
 
JC
Fresh versus old blood: are there differences and do they matter?
Hematology Am Soc Hematol Educ Program
2013
, vol. 
2013
 (pg. 
651
-
655
)
9
Fergusson
 
DA
Hébert
 
P
Hogan
 
DL
, et al. 
Effect of fresh red blood cell transfusions on clinical outcomes in premature, very low-birth-weight infants: the ARIPI randomized trial.
JAMA
2012
, vol. 
308
 
14
(pg. 
1443
-
1451
)
10
Steiner
 
ME
Assmann
 
SF
Levy
 
JH
, et al. 
Addressing the question of the effect of RBC storage on clinical outcomes: the Red Cell Storage Duration Study (RECESS) (Section 7).
Transfus Apheresis Sci
2010
, vol. 
43
 
1
(pg. 
107
-
116
)
11
Lacroix
 
J
 
Three decades of transfusion. Presented at the Scientific Presentations of the Critical Care Canada Forum 2014, October 30, 2014, Toronto, Canada
12
Lacroix
 
J
Hébert
 
P
Fergusson
 
D
, et al. 
ABLE Study Group
The Age of Blood Evaluation (ABLE) randomized controlled trial: study design.
Transfus Med Rev
2011
, vol. 
25
 
3
(pg. 
197
-
205
)
13
Pereira
 
A
Will clinical studies elucidate the connection between the length of storage of transfused red blood cells and clinical outcomes? An analysis based on the simulation of randomized controlled trials.
Transfusion
2013
, vol. 
53
 
1
(pg. 
34
-
40
)
14
Liu
 
C
Zhao
 
W
Christ
 
GJ
Gladwin
 
MT
Kim-Shapiro
 
DB
Nitric oxide scavenging by red cell microparticles.
Free Radic Biol Med
2013
, vol. 
65
 (pg. 
1164
-
1173
)
15
Liu
 
C
Liu
 
X
Janes
 
J
, et al. 
Mechanism of faster NO scavenging by older stored red blood cells.
Redox Biol
2014
, vol. 
2
 (pg. 
211
-
219
)
16
Billecke
 
SS
D’Alecy
 
LG
Platel
 
R
, et al. 
Blood content of asymmetric dimethylarginine: new insights into its dysregulation in renal disease.
Nephrol Dial Transplant
2009
, vol. 
24
 
2
(pg. 
489
-
496
)
17
Billecke
 
SS
Kitzmiller
 
LA
Northrup
 
JJ
, et al. 
Contribution of whole blood to the control of plasma asymmetrical dimethylarginine.
Am J Physiol Heart Circ Physiol
2006
, vol. 
291
 
4
(pg. 
H1788
-
H1796
)
18
Cardo
 
LJ
Hmel
 
P
Wilder
 
D
Stored packed red blood cells contain a procoagulant phospholipid reducible by leukodepletion filters and washing.
Transfus Apheresis Sci
2008
, vol. 
38
 
2
(pg. 
141
-
147
)
19
Gao
 
Y
Lv
 
L
Liu
 
S
Ma
 
G
Su
 
Y
Elevated levels of thrombin-generating microparticles in stored red blood cells.
Vox Sang
2013
, vol. 
105
 
1
(pg. 
11
-
17
)
20
Keating
 
FK
Butenas
 
S
Fung
 
MK
Schneider
 
DJ
Platelet-white blood cell (WBC) interaction, WBC apoptosis, and procoagulant activity in stored red blood cells.
Transfusion
2011
, vol. 
51
 
5
(pg. 
1086
-
1095
)
21
Lu
 
C
Shi
 
J
Yu
 
H
Hou
 
J
Zhou
 
J
Procoagulant activity of long-term stored red blood cells due to phosphatidylserine exposure.
Transfus Med
2011
, vol. 
21
 
3
(pg. 
150
-
157
)
22
Cata
 
JP
Wang
 
H
Gottumukkala
 
V
Reuben
 
J
Sessler
 
DI
Inflammatory response, immunosuppression, and cancer recurrence after perioperative blood transfusions.
Br J Anaesth
2013
, vol. 
110
 
5
(pg. 
690
-
701
)
23
Vamvakas
 
EC
Blajchman
 
MA
Transfusion-related immunomodulation (TRIM): an update.
Blood Rev
2007
, vol. 
21
 
6
(pg. 
327
-
348
)
24
Cortés-Puch
 
I
Wang
 
D
Sun
 
J
, et al. 
Washing older blood units before transfusion reduces plasma iron and improves outcomes in experimental canine pneumonia.
Blood
2014
, vol. 
123
 
9
(pg. 
1403
-
1411
)
25
Hod
 
EA
Spitalnik
 
SL
Harmful effects of transfusion of older stored red blood cells: iron and inflammation.
Transfusion
2011
, vol. 
51
 
4
(pg. 
881
-
885
)
26
Hod
 
EA
Spitalnik
 
SL
Stored red blood cell transfusions: iron, inflammation, immunity, and infection.
Transfus Clin Biol
2012
, vol. 
19
 
3
(pg. 
84
-
89
)
27
Hod
 
EA
Zhang
 
N
Sokol
 
SA
, et al. 
Transfusion of red blood cells after prolonged storage produces harmful effects that are mediated by iron and inflammation.
Blood
2010
, vol. 
115
 
21
(pg. 
4284
-
4292
)
28
Hu
 
H
Xenocostas
 
A
Chin-Yee
 
N
Lu
 
X
Chin-Yee
 
I
Feng
 
Q
Transfusion of fresh but not old stored blood reduces infarct size and improves cardiac function after acute myocardial infarction in anemic rats*.
Crit Care Med
2012
, vol. 
40
 
3
(pg. 
740
-
746
)
29
Solomon
 
SB
Wang
 
D
Sun
 
J
, et al. 
Mortality increases after massive exchange transfusion with older stored blood in canines with experimental pneumonia.
Blood
2013
, vol. 
121
 
9
(pg. 
1663
-
1672
)
30
Wang
 
D
Cortés-Puch
 
I
Sun
 
J
, et al. 
Transfusion of older stored blood worsens outcomes in canines depending on the presence and severity of pneumonia.
Transfusion
2014
, vol. 
54
 
7
(pg. 
1712
-
1724
)
31
Callan
 
MB
Patel
 
RT
Rux
 
AH
, et al. 
Transfusion of 28-day-old leucoreduced or non-leucoreduced stored red blood cells induces an inflammatory response in healthy dogs.
Vox Sang
2013
, vol. 
105
 
4
(pg. 
319
-
327
)
32
Eun
 
JC
Moore
 
EE
Banerjee
 
A
, et al. 
Leukotriene b4 and its metabolites prime the neutrophil oxidase and induce proinflammatory activation of human pulmonary microvascular endothelial cells.
Shock
2011
, vol. 
35
 
3
(pg. 
240
-
244
)
33
Silliman
 
CC
Moore
 
EE
Kelher
 
MR
Khan
 
SY
Gellar
 
L
Elzi
 
DJ
Identification of lipids that accumulate during the routine storage of prestorage leukoreduced red blood cells and cause acute lung injury.
Transfusion
2011
, vol. 
51
 
12
(pg. 
2549
-
2554
)
34
Silliman
 
CC
Voelkel
 
NF
Allard
 
JD
, et al. 
Plasma and lipids from stored packed red blood cells cause acute lung injury in an animal model.
J Clin Invest
1998
, vol. 
101
 
7
(pg. 
1458
-
1467
)
35
Sparrow
 
RL
Patton
 
KA
Supernatant from stored red blood cell primes inflammatory cells: influence of prestorage white cell reduction.
Transfusion
2004
, vol. 
44
 
5
(pg. 
722
-
730
)
36
Weisbach
 
V
Wanke
 
C
Zingsem
 
J
Zimmermann
 
R
Eckstein
 
R
Cytokine generation in whole blood, leukocyte-depleted and temporarily warmed red blood cell concentrates.
Vox Sang
1999
, vol. 
76
 
2
(pg. 
100
-
106
)
37
Baek
 
JH
D’Agnillo
 
F
Vallelian
 
F
, et al. 
Hemoglobin-driven pathophysiology is an in vivo consequence of the red blood cell storage lesion that can be attenuated in guinea pigs by haptoglobin therapy.
J Clin Invest
2012
, vol. 
122
 
4
(pg. 
1444
-
1458
)
38
Prestia
 
K
Bandyopadhyay
 
S
Slate
 
A
, et al. 
Transfusion of stored blood impairs host defenses against Gram-negative pathogens in mice.
Transfusion
2014
, vol. 
54
 
11
(pg. 
2842
-
2851
)
39
Zimring
 
JC
Spitalnik
 
SL
On the appropriate use and interpretation of animal models in transfusion medicine research.
Transfusion
2013
, vol. 
53
 
10
(pg. 
2334
-
2339
)
40
Food and Drug Administration
 
In: Proceedings from the 91st Meeting of the Blood Products Advisory Committee; May 1-2, 2008; Rockville, MD. Pages 1-5
41
Grindon
 
AJ
McClatchey
 
KD
Blood collection.
Clinical Laboratory Medicine
2001
Baltimore, MD
Lippincott Williams & Wilkins
(pg. 
1526
-
1528
)
42
Klein
 
HG
Anstee
 
DJ
Mollison’s Blood Transfusion in Clinical Medicine,
2014
12th ed
Malden, MA
Blackwell Publishing Inc
43
Ross
 
JF
Finch
 
CA
Peacock
 
WC
Sammons
 
ME
The in vitro preservation and post-transfusion survival of stored blood.
J Clin Invest
1947
, vol. 
26
 
4
(pg. 
687
-
703
)
44
Berezina
 
TL
Zaets
 
SB
Morgan
 
C
, et al. 
Influence of storage on red blood cell rheological properties.
J Surg Res
2002
, vol. 
102
 
1
(pg. 
6
-
12
)
45
Daly
 
A
Raval
 
JS
Waters
 
JH
Yazer
 
MH
Kameneva
 
MV
Effect of blood bank storage on the rheological properties of male and female donor red blood cells.
Clin Hemorheol Microcirc
2014
, vol. 
56
 
4
(pg. 
337
-
345
)
46
Yalcin
 
O
Ortiz
 
D
Tsai
 
AG
Johnson
 
PC
Cabrales
 
P
Microhemodynamic aberrations created by transfusion of stored blood.
Transfusion
2014
, vol. 
54
 
4
(pg. 
1015
-
1027
)
47
Henkelman
 
S
Dijkstra-Tiekstra
 
MJ
de Wildt-Eggen
 
J
Graaff
 
R
Rakhorst
 
G
van Oeveren
 
W
Is red blood cell rheology preserved during routine blood bank storage?
Transfusion
2010
, vol. 
50
 
4
(pg. 
941
-
948
)
48
Weinberg
 
JA
MacLennan
 
PA
Vandromme-Cusick
 
MJ
, et al. 
The deleterious effect of red blood cell storage on microvascular response to transfusion.
J Trauma Acute Care Surg
2013
, vol. 
75
 
5
(pg. 
807
-
812
)
49
Hod
 
EA
Brittenham
 
GM
Billote
 
GB
, et al. 
Transfusion of human volunteers with older, stored red blood cells produces extravascular hemolysis and circulating non-transferrin-bound iron.
Blood
2011
, vol. 
118
 
25
(pg. 
6675
-
6682
)
50
Raat
 
NJ
Ince
 
C
Oxygenating the microcirculation: the perspective from blood transfusion and blood storage.
Vox Sang
2007
, vol. 
93
 
1
(pg. 
12
-
18
)
51
Hess
 
JR
Red cell changes during storage.
Transfus Apheresis Sci
2010
, vol. 
43
 
1
(pg. 
51
-
59
)
52
Hess
 
JR
Red cell storage.
J Proteomics
2010
, vol. 
73
 
3
(pg. 
368
-
373
)
53
Hess
 
JR
Measures of stored red blood cell quality.
Vox Sang
2014
, vol. 
107
 
1
(pg. 
1
-
9
)
54
Hess
 
JR
Improving the predictive value of red blood cell storage trials: lessons from the Biomedical Excellence for Safer Transfusion (BEST) Collaborative Trial 41.
Transfusion
2011
, vol. 
51
 
suppl 1
(pg. 
34S
-
37S
)
55
Singel
 
DJ
Stamler
 
JS
Chemical physiology of blood flow regulation by red blood cells: the role of nitric oxide and S-nitrosohemoglobin.
Annu Rev Physiol
2005
, vol. 
67
 (pg. 
99
-
145
)
56
Anniss
 
AM
Sparrow
 
RL
Variable adhesion of different red blood cell products to activated vascular endothelium under flow conditions.
Am J Hematol
2007
, vol. 
82
 
6
(pg. 
439
-
445
)
57
Alexander
 
JT
El-Ali
 
AM
Newman
 
JL
, et al. 
Red blood cells stored for increasing periods produce progressive impairments in nitric oxide-mediated vasodilation.
Transfusion
2013
, vol. 
53
 
11
(pg. 
2619
-
2628
)
58
Neuman
 
RB
Hayek
 
S
Rahman
 
A
, et al. 
Effects of storage-aged red blood cell transfusions on endothelial function in hospitalized patients [published online ahead of print November 13, 2014].
Transfusion
59
Sparrow
 
RL
Time to revisit red blood cell additive solutions and storage conditions: a role for “omics” analyses.
Blood Transfus
2012
, vol. 
10
 
suppl 2
(pg. 
s7
-
s11
)
60
Anniss
 
AM
Glenister
 
KM
Killian
 
JJ
Sparrow
 
RL
Proteomic analysis of supernatants of stored red blood cell products.
Transfusion
2005
, vol. 
45
 
9
(pg. 
1426
-
1433
)
61
Bosman
 
GJ
Lasonder
 
E
Luten
 
M
, et al. 
The proteome of red cell membranes and vesicles during storage in blood bank conditions.
Transfusion
2008
, vol. 
48
 
5
(pg. 
827
-
835
)
62
Dzieciatkowska
 
M
Silliman
 
CC
Moore
 
EE
, et al. 
Proteomic analysis of the supernatant of red blood cell units: the effects of storage and leucoreduction.
Vox Sang
2013
, vol. 
105
 
3
(pg. 
210
-
218
)
63
Lion
 
N
Crettaz
 
D
Rubin
 
O
Tissot
 
JD
Stored red blood cells: a changing universe waiting for its map(s).
J Proteomics
2010
, vol. 
73
 
3
(pg. 
374
-
385
)
64
Nikolovski
 
Z
De La Torre
 
C
Chiva
 
C
, et al. 
Alterations of the erythrocyte membrane proteome and cytoskeleton network during storage—a possible tool to identify autologous blood transfusion.
Drug Test Anal
2012
, vol. 
4
 
11
(pg. 
882
-
890
)
65
Pallotta
 
V
Gevi
 
F
D’alessandro
 
A
Zolla
 
L
Storing red blood cells with vitamin C and N-acetylcysteine prevents oxidative stress-related lesions: a metabolomics overview.
Blood Transfus
2014
, vol. 
12
 
3
(pg. 
376
-
387
)
66
Patel
 
RM
Roback
 
JD
Uppal
 
K
Yu
 
T
Jones
 
DP
Josephson
 
CD
Metabolomics profile comparisons of irradiated and nonirradiated stored donor red blood cells [published online ahead of print October 21, 2014].
Transfusion
67
Roback
 
JD
Josephson
 
CD
Waller
 
EK
, et al. 
Metabolomics of ADSOL (AS-1) red blood cell storage.
Transfus Med Rev
2014
, vol. 
28
 
2
(pg. 
41
-
55
)
68
van ’t Erve
 
TJ
Doskey
 
CM
Wagner
 
BA
, et al. 
Heritability of glutathione and related metabolites in stored red blood cells.
Free Radic Biol Med
2014
, vol. 
76
 (pg. 
107
-
113
)
69
van ’t Erve
 
TJ
Wagner
 
BA
Martin
 
SM
, et al. 
The heritability of metabolite concentrations in stored human red blood cells.
Transfusion
2014
, vol. 
54
 
8
(pg. 
2055
-
2063
)
70
Walpurgis
 
K
Kohler
 
M
Thomas
 
A
, et al. 
Storage-induced changes of the cytosolic red blood cell proteome analyzed by 2D DIGE and high-resolution/high-accuracy MS.
Proteomics
2012
, vol. 
12
 
21
(pg. 
3263
-
3272
)
71
Walpurgis
 
K
Kohler
 
M
Thomas
 
A
, et al. 
Effects of gamma irradiation and 15 days of subsequent ex vivo storage on the cytosolic red blood cell proteome analyzed by 2D-DIGE and Orbitrap MS.
Proteomics Clin Appl
2013
, vol. 
7
 
7-8
(pg. 
561
-
570
)
72
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
, vol. 
5
 pg. 
110
 
73
Schubert
 
P
Culibrk
 
B
Karwal
 
S
Slichter
 
SJ
Devine
 
DV
Optimization of platelet concentrate quality: application of proteomic technologies to donor management.
J Proteomics
2012
, vol. 
76
 
spec no
(pg. 
329
-
336
)
74
Devine
 
DV
Schubert
 
P
Proteomic applications in blood transfusion: working the jigsaw puzzle.
Vox Sang
2011
, vol. 
100
 
1
(pg. 
84
-
91
)
75
Thon
 
JN
Schubert
 
P
Devine
 
DV
Platelet storage lesion: a new understanding from a proteomic perspective.
Transfus Med Rev
2008
, vol. 
22
 
4
(pg. 
268
-
279
)
76
Thon
 
JN
Schubert
 
P
Duguay
 
M
, et al. 
Comprehensive proteomic analysis of protein changes during platelet storage requires complementary proteomic approaches.
Transfusion
2008
, vol. 
48
 
3
(pg. 
425
-
435
)
77
van de Watering
 
LM
Age of blood: does older blood yield poorer outcomes?
Curr Opin Hematol
2013
, vol. 
20
 
6
(pg. 
526
-
532
)
Sign in via your Institution