Hundreds of billions of platelets are cleared daily from circulation via efficient and highly regulated mechanisms. These mechanisms may be stimulated by exogenous reagents or environmental changes to accelerate platelet clearance, leading to thrombocytopenia. The interplay between antiapoptotic Bcl-xL and proapoptotic molecules Bax and Bak sets an internal clock for the platelet lifespan, and BH3-only proteins, mitochondrial permeabilization, and phosphatidylserine (PS) exposure may also contribute to apoptosis-induced platelet clearance. Binding of plasma von Willebrand factor or antibodies to the ligand-binding domain of glycoprotein Ibα (GPIbα) on platelets can activate GPIb-IX in a shear-dependent manner by inducing unfolding of the mechanosensory domain therein, and trigger downstream signaling in the platelet including desialylation and PS exposure. Deglycosylated platelets are recognized by the Ashwell-Morell receptor and potentially other scavenger receptors, and are rapidly cleared by hepatocytes and/or macrophages. Inhibitors of platelet clearance pathways, including inhibitors of GPIbα shedding, neuraminidases, and platelet signaling, are efficacious at preserving the viability of platelets during storage and improving their recovery and survival in vivo. Overall, common mechanisms of platelet clearance have begun to emerge, suggesting potential strategies to extend the shelf-life of platelets stored at room temperature or to enable refrigerated storage.

In addition to their vital role in hemostasis and thrombosis, platelets are involved in many diverse biological processes including inflammation, tissue repair, and antimicrobial host defense. To maintain a steady count of 150 000 to 400 000 platelets per microliter of whole blood, the body produces and clears platelets at a rate of 1011 platelets per day. Platelet genesis or thrombopoiesis has been extensively characterized, and new elements in the process are still being discovered.1  In recent years, many critical advances in the studies of platelet clearance have been made. This review focuses on the current understanding of the molecular mechanisms underlying platelet clearance, and how this knowledge is used to improve platelet storage.

Three methods are typically used to monitor platelet clearance. The first method is to measure the ability of a compound or molecule to induce platelet clearance. The compound is administered into the body and blood counting is performed periodically thereafter, producing a plot of relative platelet count over time, expressed typically as a percentage of that prior to administration (Figure 1A).2,3  An acute drop in platelet count illustrates the compound’s clearing effect. Once the compound is metabolized or removed from the body, the platelet count rises to normal due to continuous thrombopoiesis. The second method is to measure the lifespan of endogenous platelets. A radioisotopic or fluorescent compound is administered into humans or mice to pulse label the circulating platelets.4-6  Thereafter, blood is collected periodically, and platelets are isolated from the whole blood. The percentage or the radioactivity of labeled platelets in the whole platelet population is measured and plotted over time (Figure 1B). These plots demonstrated that the lifespans of human and murine platelets are 7 to 10 and 4 to 5 days, respectively.4,5  The third method is to measure the clearance of transfused platelets. Platelets obtained from humans or animals are processed in vitro, labeled with radioisotopes7  (eg, 51Cr or 111In) or chromophores8,9  (eg, carboxyfluorescein succinimidyl ester, 5-chloromethylfluorescein diacetate, or N-hydroxysuccinimido biotin), and transfused into a different host. The resulting plot of transfused platelets over time typically consists of 2 parameters (Figure 1C). The first parameter, known as platelet recovery, denotes the appearance of transfused platelets in peripheral circulation. The second, known as platelet survival, denotes the clearance of transfused platelets from circulation. Compared with the first 2, the third method enables the assessment of effects of in vitro treatment (eg, storage) of platelets. Also, protocols have been developed to monitor function and survival of human platelets in animals.10 

Figure 1.

Measurement of platelet clearance kinetics. (A) Endogenous platelet count is monitored over time following the injection of a reagent to assess its effect on platelet clearance. (B) A radioisotopic or fluorescent compound is administered into human or mice. Thereafter, the percentage or radioactivity of labeled platelets in the whole platelet population is measured over time. (C) Exogenous platelets are labeled with radioisotopes or chromophores, and transfused into a host. The percentage of these exogenous labeled platelets is measured over time. The recovery indicates the initial appearance of transfused platelet in the circulation, and the survival means the time that the transfused platelets stay in the circulation.

Figure 1.

Measurement of platelet clearance kinetics. (A) Endogenous platelet count is monitored over time following the injection of a reagent to assess its effect on platelet clearance. (B) A radioisotopic or fluorescent compound is administered into human or mice. Thereafter, the percentage or radioactivity of labeled platelets in the whole platelet population is measured over time. (C) Exogenous platelets are labeled with radioisotopes or chromophores, and transfused into a host. The percentage of these exogenous labeled platelets is measured over time. The recovery indicates the initial appearance of transfused platelet in the circulation, and the survival means the time that the transfused platelets stay in the circulation.

Close modal

All 3 methods have been applied to humans and mice. Overall, the results suggest that despite some differences such as lifespan, human and mouse platelet clearance mechanisms share many common features.

Similar to many nucleated cells, platelet apoptosis depends on the balance between proapoptotic and antiapoptotic machinery (Figure 2). Antiapoptotic Bcl-2 family proteins restrain the proapoptotic molecules Bak and Bax. Several Bcl-2 family proteins, including Bcl-2, Bcl-w, and Bcl-xL are expressed in both human and murine platelets.11,12  Platelet-specific knockout of Bcl-2 and systemic knockout of Bcl-w does not alter platelet lifespan.13,14  Treatment with ABT-199, which specifically inhibits Bcl-2, causes cell death in Bcl-2–dependent tumors but not thrombocytopenia.15  Alternatively, specific pharmacological inhibition16  or Cre-mediated deletion of Bcl-xL,17  or broad inhibition of Bcl-2-family proteins such as by ABT-737,18  led to platelet apoptosis and thrombocytopenia. Furthermore, double deletion of Bak and Bax prolongs platelet lifespan, and can rescue thrombocytopenia caused by loss of Bcl-xL.12,19  Single deletions have revealed that Bak is likely the major regulator of lifespan whereas Bax plays a smaller role.6,19 

Figure 2.

Apoptotic machinery in platelet clearance and lifespan. The anti-apoptotic Bcl-xL restrains the proapoptotic Bax/Bak in platelets. Mitochondrial damage induced by CCCP, an ionophore, leads to robust ectodomain shedding of GPIbα. If inhibition by Bcl-xL is blocked pharmacologically, Bax/Bak will induce mitochondrial damage, leading to the apoptotic cascade. The BH3-only initiator of apoptosis Bad may also affect platelet lifespan, though further study would help to elucidate its role. Apoptotic cells redistribute PS from the inner to the outer leaflet of their plasma membranes. One calcium-independent pathway may involve Xkr8. Another pathway present in platelets is facilitated by TMEM16F, a calcium-activated phospholipid scramblase.

Figure 2.

Apoptotic machinery in platelet clearance and lifespan. The anti-apoptotic Bcl-xL restrains the proapoptotic Bax/Bak in platelets. Mitochondrial damage induced by CCCP, an ionophore, leads to robust ectodomain shedding of GPIbα. If inhibition by Bcl-xL is blocked pharmacologically, Bax/Bak will induce mitochondrial damage, leading to the apoptotic cascade. The BH3-only initiator of apoptosis Bad may also affect platelet lifespan, though further study would help to elucidate its role. Apoptotic cells redistribute PS from the inner to the outer leaflet of their plasma membranes. One calcium-independent pathway may involve Xkr8. Another pathway present in platelets is facilitated by TMEM16F, a calcium-activated phospholipid scramblase.

Close modal

In many apoptotic cells, Bcl-2 family proteins are inhibited by BH3-only initiators of apoptosis, ultimately leaving Bax/Bak free to initiate mitochondrial membrane damage and trigger the apoptotic cascade. Of the 4 BH3-only proteins expressed in platelets (Bid, Bim, Bad, and Bik), genetic deletions of Bid or Bim did not alter the platelet count in mice.12  Loss of Bad leads to only a small increase in platelet count and lifespan.20  The expression of BH3-only proteins in platelets may imply their involvement in regulating intrinsic apoptosis (Figure 2), but future studies are needed to fully elucidate their roles. Similarly, platelets express certain components of the extrinsic apoptosis pathway including caspase 8, but the limited data so far do not support their critical role in regulating platelet lifespan.21,22 

In many cells undergoing apoptosis, the redistribution of phosphatidylserine (PS) from the inner to the outer leaflet of the plasma membrane serves as a molecular cue for engulfment and clearance by phagocytes. Although lactadherin and the scavenger machinery can mediate clearance of platelet-derived PS-expressing microvesicles,23  whether they mediate clearance of apoptotic platelets, as well as the identity of the “clear-me” sign on apoptotic platelets, remains to be fully elucidated. Earlier studies have ruled out several markers of platelet activation, such as P-selectin, as “clear-me” signs for platelet clearance.24  Platelets possess 2 distinct pathways through which they expose PS on their surface25,26  (Figure 2). One is dependent on intracellular Ca2+ and TMEM16F, a Ca2+-activated phospholipid scramblase and ion channel.27,28  The other is associated with apoptosis, and may involve Xk-related protein 8 (Xkr8), a 10-transmembrane domain scramblase, instead of TMEM16F.25  Earlier studies suggest that apoptosis-associated morphological changes in platelets, such as PS exposure and recognition by phagocyte scavenger receptors, are not inhibited by broad-spectrum caspase inhibitor zVAD-fmk.29  Whether or how Xkr8 and/or TMEM16F are involved in regulating the platelet lifespan and mediating its clearance in a caspase-independent manner remains to be characterized.

In most apoptosis pathways, mitochondrial outer membrane permeabilization is a critical step, resulting in decrease of the mitochondrial electrochemical gradient and release of cytochrome C. Carbonyl cyanide 3-chlorophenylhydrazone (CCCP) is a lipid-soluble protonophore and oxidative phosphorylation uncoupler that induces mitochondrial permeabilization and loss of membrane potential.30  When platelets are pretreated with CCCP, posttransfusion recovery of these platelets is greatly reduced, indicating that the bulk of CCCP-treated platelets are cleared rapidly in vivo.31  Those that are not cleared rapidly do not have reduced lifespan. It is also noteworthy that CCCP treatment induces modest PS exposure but significant ectodomain shedding of glycoprotein Ibα (GPIbα; CD42b) in platelets.31  These results link the mitochondrial damage to accelerated platelet clearance and, as described in “GPIb-IX signaling: a trigger for platelet clearance,” implicate the shedding of GPIbα as a key step (Figure 2).

Another common mechanism of platelet clearance involves opsonization by antiplatelet antibodies, Fc-receptor–mediated recognition, and subsequent clearance. In patients with immune thrombocytopenia (ITP), autoantibodies targeting platelet surface glycoproteins, primarily GPIIb-IIIa and GPIb-IX, lead to Fc-dependent clearance via macrophages.32  Antiplatelet autoantibodies may also target the precursors to platelets, megakaryocytes.33  Additionally, infusion of monoclonal antibodies (MAbs) targeting the N-terminal ligand-binding domain (LBD) of GPIbα causes fast depletion of nearly all platelets from animals.34-37  Common treatments for ITP include immunosuppressive steroids and IV immunoglobulin G (IVIG).38  However, some patients are refractory to these treatments,39  implying at least 1 parallel Fc-independent clearance mechanism (discussed in “GPIb-IX signaling: a trigger for platelet clearance”).

Recent studies have highlighted the role of glycan modifications on platelets in mediating their clearance. In circulation, loss of terminal sialic acid (a derivative of neuraminic acid) from the platelet surface has been linked with senescent platelet removal.40  Neuraminidases (sialidases) are glycoside hydrolase enzymes that remove the terminal sialic acid residues on glycans. Injection of neuraminidase in animal models leads to rapid platelet clearance and transient thrombocytopenia.41  Also, certain bacterial infections are marked by a release of pathogen-derived neuraminidase resulting in thrombocytopenia.42  Furthermore, there is evidence that endogenous, platelet-derived neuraminidase plays a role in fast clearance of refrigerated platelets.43,44  Relatedly, many antibodies targeting the N-terminal ligand-binding domain (LBD) of GPIbα induce platelet signaling and surface presentation of lysosomal neuraminidase (Neu1), leading to increased desialylation of platelets and acute thrombocytopenia in mice.45  Treatment with 2,3-dehydro-2-deoxy-N-acetylneuraminic acid (DANA), a neuraminidase inhibitor, reduces desialylation and leads to amelioration of thrombocytopenia.45  Similarly, binding of plasma von Willebrand factor (VWF) to GPIbα on platelets under shear produces similar signaling events including desialylation.46 

In general, the terminal residues in both N- and O-glycans are sialic acid, linked to a penultimate β-galactose (β-gal). Desialylation of platelets therefore leads to the increased exposure of β-gal (Figure 3). The exposed β-gal on the platelet surface can be recognized by the Ashwell-Morell receptor (AMR), a multimeric endocytic receptor complex also known as the asialoglycoprotein receptor,47  on the surface of hepatocytes and/or liver macrophages (Kupffer cells), inducing the clearance of the platelet from circulation.43,48,49  The AMR exhibits higher affinity and ligand preference for tetra- or triantennary galactoses than di- or monoantennary ones.47,50  Mice lacking the AMR have elevated platelet count (mild thrombocytosis), and fast clearance of platelets in response to neuraminidase injection is abolished in them.3,51  On the other hand, St3gal4−/− mice, which have deficiencies in terminal sialic acid residues on platelet surface glycoproteins due to genetic loss of an important sialyltransferase, suffer from thrombocytopenia as a result of accelerated platelet clearance via the hepatic AMR.48  In addition to mediating clearance and removal of senescent platelets, the AMR also leads to stimulation of platelet production, forming a clearance/thrombopoiesis feedback loop for platelet homeostasis.51 

Figure 3.

Protein desialylation as a clear-me sign in platelets. Over the platelet lifespan, surface glycoproteins lose the terminal sialic acid residues in their glycans, a process associated with clearance. Neuraminidases are glycoside hydrolases that can remove terminal sialic acid from glycans. Neuraminidases are found in platelets, which present neuraminidase on their surface downstream of GPIb-IX complex signaling. In many glycans, desialylation leads to exposure of the penultimate galactose residues on glycans. These can in turn be recognized by the AMR. Further deglycosylation leads to exposed GlcNAc residues, which may be recognized by other carbohydrate receptors and potentially mediate their uptake by macrophages.

Figure 3.

Protein desialylation as a clear-me sign in platelets. Over the platelet lifespan, surface glycoproteins lose the terminal sialic acid residues in their glycans, a process associated with clearance. Neuraminidases are glycoside hydrolases that can remove terminal sialic acid from glycans. Neuraminidases are found in platelets, which present neuraminidase on their surface downstream of GPIb-IX complex signaling. In many glycans, desialylation leads to exposure of the penultimate galactose residues on glycans. These can in turn be recognized by the AMR. Further deglycosylation leads to exposed GlcNAc residues, which may be recognized by other carbohydrate receptors and potentially mediate their uptake by macrophages.

Close modal

In addition to the interaction between galactose and the AMR, other carbohydrates and their receptors may also play a role in platelet clearance (Figure 3). It was reported that integrin αMβ2 recognizes refrigerated platelets via binding to exposed GlcNAc on the platelet surface and mice lacking the αM subunit show a small increase in platelet count.52,53  Clodronate depletion of macrophages alleviates thrombocytopenia in a mouse model of von Willebrand disease (VWD) type 2B.54  Furthermore, preinjection of GlcNAc into guinea pigs prior to induction of antibody-induced thrombocytopenia partly protects against depletion of platelets.55  However, although galactosylation of GlcNAc residues via treatment of uridine 5′-diphosphogalactose (UDP-galactose) results in the normal survival of short-term refrigerated platelets, it does not ameliorate the survival of long-term (48 hours) refrigerated human and murine platelets.56  The degrees of contribution of various glycans to platelet clearance remain to be clarified.

Platelet GPIbα is heavily decorated with sialic acid residues, accounting for as much as 70% to 80% of the total sialic acid on the platelet surface. Unlike human GPIbα, the murine GPIbα amino acid sequence lacks any N-glycosylation consensus sequences. Grewal et al showed recently that even in mice lacking GPIbα, neuraminidase treatment leads to platelet clearance, albeit at a slower rate than in wild-type (WT) mice.3  This suggests that the glycans on GPIbα are necessary to set off a rapid rate of AMR-dependent clearance, but the exposed galactoses on other platelet glycoproteins may also be counterreceptors for the AMR. Considering these intriguing observations regarding glycosylation of GPIbα, further work is likely required in order to understand the full contributions of these phenomena to platelet clearance.

The GPIb-IX-V (CD42) complex has been implicated in platelet clearance under a number of scenarios. Among the scenarios are VWF-platelet agglutinated complexes,54,57,58  Fc-independent anti-GPIb-IX antibody-induced clearance,2,45  platelet surface desialylation,3,48  and ectodomain shedding of GPIbα during platelet storage8,31  (discussed in “Platelet storage at room temperature”).

GPIb-IX is a multimeric platelet receptor complex composed of the GPIbα, GPIbβ, and GPIX subunits. GPIbα is the major subunit of the complex and is responsible for binding to all known ligands of GPIb-IX including VWF. When immobilized under flow at sites of injury in the endothelium, VWF undergoes a conformational change that enables it to bind GPIbα and recruit platelets to the injury. Alternatively, circulating VWF does not spontaneously associate with the LBD of GPIbα. In patients with type 2B VWD, mutant VWF exhibits increased spontaneous association to GPIbα. Type 2B VWD patients present with accelerated platelet clearance and thrombocytopenia of variable severity, depending on the underlying causative mutation.46,59,60  Furthermore, transgenic mice expressing type 2B VWF exhibit thrombocytopenia due to clearance of large VWF-platelet complexes in the liver and/or spleen.54  In addition to type 2B VWD, several other situations that facilitate binding of soluble VWF to GPIbα also result in accelerated platelet clearance. For example, ristocetin, which induces spontaneous association of VWF to GPIbα, was pulled from clinical use because it caused thrombocytopenia and clotting.57  Injection of botrocetin, a snake venom that induces VWF binding to GPIbα via a different mechanism, causes acute thrombocytopenia in animals.58,61  Thrombocytopenia is also observed in many patients who have received implantations of left-ventricular assist devices,62  which generate abnormal shear flow conditions and may potentially induce VWF association with GPIbα. It was recently reported that binding of plasma VWF to GPIbα on platelets under shear induces GPIb-IX signaling including platelet desialylation, thereby leading to platelet clearance.46 

Platelet clearance by anti-LBD MAbs in mice can occur in an Fc-independent manner and is largely unaffected by IVIG pretreatment.2,37,63  This is because anti-LBD MAbs can directly activate GPIb-IX and induce platelet intracellular signaling, particularly desialylation, and subsequent platelet clearance by hepatocytes and/or macrophages.45,55  Analyses of plasma from ITP patients in multicenter cohort studies revealed that the presence of autoantibodies targeting GPIb-IX is an effective predictor for refractoriness to steroid or IVIG therapy.64,65  Regarding the mechanistic requirements of anti-LBD MAb-induced platelet signaling, 4 key observations have emerged from the literature. First, the F(ab′)2 but not the Fab fragment of an anti-LBD MAb induces platelet clearance,2,37  indicating that the bivalent structure of an antibody is required for activating GPIb-IX. Second, most anti-LBD antibodies clear platelets rapidly, regardless of their epitope in the LBD.34-37,45  Third, a small subset of anti-LBD MAbs is ineffective at inducing Fc-independent clearance.55,66  Fourth, most MAbs targeting regions other than the LBD in GPIb-IX do not induce Fc-independent platelet clearance.36,67,68 

A GPIbα clustering model has been proposed as the mechanism of GPIb-IX activation69,70  and applied to explain the observed effects of anti-LBD MAbs. In this model, an anti-LBD MAb binds 1 copy of GPIbα with each Fab, inducing lateral dimerization or “clustering,” and thereby transmitting a signal into the platelet that subsequently leads to fast clearance.55  VWF, being a multimeric ligand, is also capable of clustering GPIb-IX.69,70  The clustering model can explain the aforementioned first and second observations about anti-LBD MAbs. However, it is difficult to conceive how the clustering model accounts for the third and fourth observations. Particularly, a MAb targeting the mechanosensory domains (MSDs) of GPIbα binds to 2 copies of GPIbα on the platelet, but induces neither platelet activation in vitro nor thrombocytopenia in mice.68  Moreover, the requirement of shear in VWF-mediated GPIb-IX signaling is well documented71  but remains to be addressed by the clustering model.

An alternative model for GPIb-IX activation, the trigger model, has recently been proposed (Figure 4).46  The model is built on a membrane-proximal MSD that was recently identified between the macroglycopeptide region and the transmembrane domain of GPIbα.72  Under physiological shear, binding of soluble VWF to the LBD generates a pulling force on GPIbα, and induces MSD unfolding on the platelet surface, exposure of the membrane-proximal trigger sequence therein and subsequent platelet signaling including desialylation. This model of GPIb-IX activation accounts for the requirement of shear force, as well as all 4 aforementioned observations regarding antibody-induced signaling. The dimeric structure of activating ligands is used to crosslink platelets via GPIb-IX and induce MSD unfolding.66  In the trigger model, the defining characteristic of an activating ligand to GPIb-IX is its ability to bind the LBD and sustain sufficient tensile force to induce MSD unfolding.46,66  Thus, it is conceivable that ligands with similar binding affinities and binding sites but disparate mechanical properties, such as different anti-LBD MAbs or VWF bearing different type 2B mutations,60,66  may differentially activate GPIb-IX and induce platelet clearance.

Figure 4.

The trigger model of GPIb-IX-mediated signaling that leads to platelet clearance. A soluble multimeric ligand, such as plasma VWF or anti-LBD antibodies, can bind to the LBD of GPIbα and crosslink platelets. Under physiological shear, the crosslinking can generate a pulling force on GPIbα and induce unfolding of the MSD therein. Consequently, it induces platelet signaling as illustrated, including desialylation (the exposure of β-gal), leading to rapid clearance of platelets. Adapted from Deng et al with permission.46 

Figure 4.

The trigger model of GPIb-IX-mediated signaling that leads to platelet clearance. A soluble multimeric ligand, such as plasma VWF or anti-LBD antibodies, can bind to the LBD of GPIbα and crosslink platelets. Under physiological shear, the crosslinking can generate a pulling force on GPIbα and induce unfolding of the MSD therein. Consequently, it induces platelet signaling as illustrated, including desialylation (the exposure of β-gal), leading to rapid clearance of platelets. Adapted from Deng et al with permission.46 

Close modal

In circulating platelets, the ectodomain of GPIbα is continuously cleaved or shed by ADAM17.73  The ADAM17 cleavage site of GPIbα is located in the MSD, preceding the trigger sequence.46,74  It appears to be on the MSD surface and is accessible when MSD is folded,75  consistent with the observation that shedding of GPIbα occurs continuously on resting platelets. However, MSD unfolding induced by ligand binding and pulling could further expose the ADAM17 shedding cleavage site, thereby boosting shedding of GPIbα.46,66  On the other hand, upon shedding of GPIbα, and subsequent separation of glycocalicin from the platelet, the structure of the MSD is disrupted and the membrane-proximal trigger sequence therein unprotected (Figure 4). Thus, it is conceivable that shedding of GPIbα may achieve MSD unfolding and induce GPIb-IX signaling. This is consistent with the observation that mutations in the MSD that cause MSD unfolding and trigger sequence exposure can induce ligand-free signaling from GPIb-IX.46 

Intracellular signaling pathways that connect activated GPIb-IX to the surface expression of Neu1 and other clearance-related cellular changes remain to be determined. Soluble VWF binding to GPIbα under shear was reported to induce apoptotic signaling events in human platelets and Chinese hamster ovary cells expressing human GPIb-IX.76  This effect is dependent on 14-3-3 protein ζ isoform,76  which binds the cytoplasmic domains of both GPIbα and GPIbβ. GPIbα binding to VWF immobilized at the injury site is critical to platelet adhesion and activation, in which GPIb-IX-mediated signaling helps to mediate activation of GPIIb-IIIa.77  However, the difference and similarity between GPIb-IX–signaling pathways leading to platelet clearance and platelet activation remains to be clarified.

Platelet transfusion is a widely used therapy to treat patients with thrombocytopenia. Prior to transfusion, platelets derived from healthy donors, mixed in gas-permeable plastic bags with donor plasma (at ∼3 × 1011 platelets in 300 mL), are stored under constant agitation at room temperature for up to 5 days. The 5-day shelf-life is adopted primarily to reduce the risk of bacterial growth and secondarily to curtail the platelet storage lesion (PSL). PSL develops with the storage time, and its severity correlates with the reduced recovery and survival of infused platelets.78,79  Recent application of pathogen inactivation and detection technologies raise the possibility that PSL will become a limiting factor for platelet storage and efforts to reduce PSL may help to extend the platelet shelf-life beyond 5 days. Several factors have been identified to influence the development of PSL. For example, centrifugation can damage platelets and cause platelets to release both lactate dehydrogenase and granules.80  The storage conditions, including storage temperature and duration, composition of storage media, and storage containers, are also known to affect the quality of stored platelets.81  Here, we focus on recent developments targeting the molecular machineries in the platelet that mediates its clearance and function.

The onset of apoptosis may mediate PSL because caspase 3 is activated and gelsolin subsequently cleaved during storage.82  Addition of caspase 3 inhibitor Z-DEVD-FMK significantly increases platelet viability in the methyl-thiazolyl tetrazolium assay.83  Furthermore, utility of anandamide, which can inhibit platelet apoptosis through the phosphatidylinositol 3-kinase/Akt pathway,84  is able to reduce PS exposure and soluble P-selectin content in platelets after 7 days of storage although it has no effects during 5 days.85  On the other hand, partial inhibition of caspase 3 activation by complement inhibitor compstatin during storage does not reduce PSL.86  These studies suggest that apoptosis is ongoing during storage, but whether its inhibition is sufficient to preserve the viability of stored platelets requires additional investigation.

The mitochondrial transmembrane potential in stored platelets was reported to remain unchanged compared with fresh platelets, even though apoptotic signals such as caspase activation and PS exposure were enhanced.87  However, it was significantly higher than fresh platelets in another report.88  A recent study found that increasing the storage time was associated with mitochondrial dysfunction.89  Additionally, platelet mitochondria injury induced by CCCP treatment led to a significantly reduced posttransfusion recovery in mice.31  Acetyl-l-carnitine or ascorbic acid, which preserves mitochondrial function during platelet storage, helps but is not fully sufficient to maintain platelet viability.90 

Significant ectodomain shedding of GPIbα and accumulation of its product glycocalicin during storage is consistently observed in various studies.91,92  A tight correlation between GPIbα shedding and the extent of PSL has been noted in laboratory studies,93  although whether glycocalicin can serve as a biomarker for the quality of stored platelets requires testing in clinical settings. Furthermore, the utility of a broad-spectrum metalloproteinase inhibitor GM6001 significantly improved the posttransfusion recovery and survival of in vitro aged or CCCP-treated murine platelets (Figure 2, 5).31  Genetic ablation of ADAM17 or addition of inhibitors of p38 MAPK, which is an activator of ADAM17, during platelet storage achieved similar effects.94  More definitively, addition of MAb 5G6, which binds specifically to human GPIbα and block its shedding, during prolonged storage at room temperature improved the recovery and survival of stored platelets (Figure 5).8,95  In mice, platelets stored with the aforementioned shedding inhibitors exhibited significantly better in vivo hemostatic function than those stored without, likely because GPIbα is critically involved in primary hemostasis.8,31,94  These studies suggest that GPIbα shedding could accelerate platelet clearance, and that inhibition of GPIbα shedding could improve recovery and survival of stored platelets.

Figure 5.

Platelet storage at room temperature. At room temperature, platelets can only be stored for up to 5 days, which is mainly due to the risk of bacteria growth. In addition, GPIbα shedding is also tightly correlated to platelet storage lesion. Inhibiting GPIbα shedding by using GM6001 or 5G6 significantly improves the posttransfusion recovery and survival of room temperature–stored platelets.

Figure 5.

Platelet storage at room temperature. At room temperature, platelets can only be stored for up to 5 days, which is mainly due to the risk of bacteria growth. In addition, GPIbα shedding is also tightly correlated to platelet storage lesion. Inhibiting GPIbα shedding by using GM6001 or 5G6 significantly improves the posttransfusion recovery and survival of room temperature–stored platelets.

Close modal

The risk of microbial contamination during platelet storage at room temperature limits the shelf-life of stored platelets. Checking for the pathogens during storage adds significantly to the cost of blood banking. Refrigeration of platelets at 1°C to 6°C offers an alternative storage option because the cold temperature could effectively minimize microbial proliferation and slow down metabolism in the platelet. However, refrigerated platelets are rapidly cleared after transfusion.96  In the past few years, several studies have been carried out to critically advance our understanding of the underlying molecular mechanism.

It was noticed that platelets became desialylated following 48 hours of refrigeration.43  This is because refrigeration and subsequent rewarming of the platelets induces surface expression of Neu1, which removes sialic acid from platelet glycoproteins, particularly GPIbα.44  The exposed β-gal is recognized by the AMR and the platelets are quickly cleared by hepatocytes.43  Adding DANA, a neuraminidase inhibitor, to murine platelets during refrigeration improves the posttransfusion recovery and survival of refrigerated platelets.44  Likewise, AMR inhibitor asialofetuin significantly blocks the fast clearance of refrigerated platelets (Figure 6A).43  These inhibitory effects are similar to those on platelet desialylation and thrombocytopenia induced by anti-LBD antibodies.45 

Figure 6.

Platelet storage by refrigeration. (A) Desialylation-mediated clearance. Sialic acid is removed by Neu1 from platelet glycoproteins following refrigeration. The exposed β-gal is recognized by the AMR, and the platelets are cleared by hepatocytes. The utility of neuraminidase inhibitors such as DANA or the AMR inhibitor asialofetuin can impede the clearance of desialylated platelets. (B) GPIbα clustering–mediated clearance. GPIbα clusters on platelet surface, and14-3-3ζ dissociates from Bad and associates with GPIbα after refrigeration. This induces the platelet apoptosis process. A broad caspase inhibitor Q-VD-Oph or arachidonic acid depletion can inhibit the apoptosis process of refrigerated platelets and improve the posttransfusion recovery and survival. (C) VWF binding–mediated clearance. Refrigeration leads to binding of plasma VWF to GPIbα. Upon transfusion and thus exposure to the shear flow, VWF binding may generate a pulling force and induces MSD unfolding, leading to rapid platelet clearance. OGE cleaves off the LBD of GPIbα, therefore precludes the VWF-GPIbα interaction and subsequently platelet clearance.

Figure 6.

Platelet storage by refrigeration. (A) Desialylation-mediated clearance. Sialic acid is removed by Neu1 from platelet glycoproteins following refrigeration. The exposed β-gal is recognized by the AMR, and the platelets are cleared by hepatocytes. The utility of neuraminidase inhibitors such as DANA or the AMR inhibitor asialofetuin can impede the clearance of desialylated platelets. (B) GPIbα clustering–mediated clearance. GPIbα clusters on platelet surface, and14-3-3ζ dissociates from Bad and associates with GPIbα after refrigeration. This induces the platelet apoptosis process. A broad caspase inhibitor Q-VD-Oph or arachidonic acid depletion can inhibit the apoptosis process of refrigerated platelets and improve the posttransfusion recovery and survival. (C) VWF binding–mediated clearance. Refrigeration leads to binding of plasma VWF to GPIbα. Upon transfusion and thus exposure to the shear flow, VWF binding may generate a pulling force and induces MSD unfolding, leading to rapid platelet clearance. OGE cleaves off the LBD of GPIbα, therefore precludes the VWF-GPIbα interaction and subsequently platelet clearance.

Close modal

Clustering of GPIbα on the platelet surface was noticed following refrigeration of platelets.53  The clustered GPIbα may contribute to the recognition of refrigerated platelets by integrin αMβ2 on hepatic macrophages, in which glycans may play a role.52  In addition, refrigeration-induced GPIbα clustering was thought to induce platelet apoptosis, as 14-3-3ζ dissociates from Bad and associates with GPIbα following refrigeration, leading to Bad activation, cytochrome C release, caspase 9 activation, PS exposure, and increased platelet phagocytosis in vitro.97  The process is inhibited by Q-VD-Oph, which is a broad caspase inhibitor, and N-acetyl-d-glucosamine, which blocks the platelet-macrophage interaction and potentially GPIbα clustering (Figure 6B).53,97,98  Furthermore, refrigeration-mediated 14-3-3ζ–GPIbα association is dependent on arachidonic acid, as the depletion of arachidonic acid during refrigeration inhibited apoptotic signals and improved the posttransfusion recovery and survival of refrigerated platelets (Figure 6B).99 

Treatment of murine platelets with O-sialoglycoprotein endopeptidase (OGE), which cleaves off the LBD of GPIbα, prior to refrigeration significantly improves the recovery and survival of these platelets in mice.43,97  Because murine GPIbα does not have N-glycosylation sequence motifs, it could not be involved in direct binding with the AMR.50  Instead, because refrigeration induces binding of plasma VWF to GPIbα on the platelet,43,100  treatment of OGE could conceivably preclude the VWF-GPIbα interaction and subsequent clustering of GPIbα in refrigerated platelets (Figure 6C). Consistent with the aforementioned trigger model, shear treatment of refrigerated WT platelets, but not VWF−/− ones, results in MSD unfolding, platelet desialylation, and PS exposure.100  Furthermore, refrigerated VWF−/− platelets, or refrigerated WT platelets incubated with a peptide that inhibits GPIbα interaction with VWF, exhibit markedly higher posttransfusion recovery than WT.100  Thus, it appears that VWF binding, GPIbα clustering, platelet desialylation, and PS exposure are key steps to the fast clearance of refrigerated platelets.

In summary, although several questions remain to be addressed, common mechanisms of platelet clearance have begun to emerge. Studies of platelet storage at room temperature and under refrigerating conditions have provided critical insights. Reciprocally, several inhibitors have shown promising efficacies in preserving the viability and improving the recovery and survival of stored platelets in animals. It is time to translate the newly gained knowledge of the platelet clearance mechanisms into viable strategies to treat thrombocytopenia and to improve platelet storage for transfusion.

The authors are very grateful to their many colleagues and collaborators for their critical insights and helpful comments on this review.

This work was supported in part by National Institutes of Health National Heart, Lung, and Blood Institute grants HL082808, HL123984 (R.L.), and F31HL134241 (M.E.Q.).

Contribution: M.E.Q., W.C., and R.L. reviewed literature and wrote the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Renhao Li, Department of Pediatrics, Emory University, 2015 Uppergate Dr NE, Room 440, Atlanta, GA 30322; e-mail: renhao.li@emory.edu.

1.
Lefrançais
E
,
Ortiz-Muñoz
G
,
Caudrillier
A
, et al
.
The lung is a site of platelet biogenesis and a reservoir for haematopoietic progenitors
.
Nature
.
2017
;
544
(
7648
):
105
-
109
.
2.
Nieswandt
B
,
Bergmeier
W
,
Rackebrandt
K
,
Gessner
JE
,
Zirngibl
H
.
Identification of critical antigen-specific mechanisms in the development of immune thrombocytopenic purpura in mice
.
Blood
.
2000
;
96
(
7
):
2520
-
2527
.
3.
Grewal
PK
,
Aziz
PV
,
Uchiyama
S
, et al
.
Inducing host protection in pneumococcal sepsis by preactivation of the Ashwell-Morell receptor
.
Proc Natl Acad Sci USA
.
2013
;
110
(
50
):
20218
-
20223
.
4.
Cohen
JA
,
Leeksma
CH
.
Determination of the life span of human blood platelets using labelled diisopropylfluorophosphonate
.
J Clin Invest
.
1956
;
35
(
9
):
964
-
969
.
5.
Odell
TT
Jr
,
McDonald
TP
.
Life span of mouse blood platelets
.
Proc Soc Exp Biol Med
.
1961
;
106
(
1
):
107
-
108
.
6.
Mason
KD
,
Carpinelli
MR
,
Fletcher
JI
, et al
.
Programmed anuclear cell death delimits platelet life span
.
Cell
.
2007
;
128
(
6
):
1173
-
1186
.
7.
van der Meer
PF
,
Tomson
B
,
Brand
A
.
In vivo tracking of transfused platelets for recovery and survival studies: an appraisal of labeling methods
.
Transfus Apheresis Sci
.
2010
;
42
(
1
):
53
-
61
.
8.
Chen
W
,
Liang
X
,
Syed
AK
, et al
.
Inhibiting GPIbα shedding preserves post-transfusion recovery and hemostatic function of platelets after prolonged storage
.
Arterioscler Thromb Vasc Biol
.
2016
;
36
(
9
):
1821
-
1828
.
9.
Baker
GR
,
Sullam
PM
,
Levin
J
.
A simple, fluorescent method to internally label platelets suitable for physiological measurements
.
Am J Hematol
.
1997
;
56
(
1
):
17
-
25
.
10.
Newman
PJ
,
Aster
R
,
Boylan
B
.
Human platelets circulating in mice: applications for interrogating platelet function and survival, the efficacy of antiplatelet therapeutics, and the molecular basis of platelet immunological disorders
.
J Thromb Haemost
.
2007
;
5
(
suppl 1
):
305
-
309
.
11.
Zhang
H
,
Nimmer
PM
,
Tahir
SK
, et al
.
Bcl-2 family proteins are essential for platelet survival
.
Cell Death Differ
.
2007
;
14
(
5
):
943
-
951
.
12.
Kodama
T
,
Takehara
T
,
Hikita
H
, et al
.
BH3-only activator proteins Bid and Bim are dispensable for Bak/Bax-dependent thrombocyte apoptosis induced by Bcl-xL deficiency: molecular requisites for the mitochondrial pathway to apoptosis in platelets
.
J Biol Chem
.
2011
;
286
(
16
):
13905
-
13913
.
13.
Debrincat
MA
,
Pleines
I
,
Lebois
M
, et al
.
BCL-2 is dispensable for thrombopoiesis and platelet survival
.
Cell Death Dis
.
2015
;
6
(
4
):
e1721
.
14.
Print
CG
,
Loveland
KL
,
Gibson
L
, et al
.
Apoptosis regulator bcl-w is essential for spermatogenesis but appears otherwise redundant
.
Proc Natl Acad Sci USA
.
1998
;
95
(
21
):
12424
-
12431
.
15.
Vandenberg
CJ
,
Cory
S
.
ABT-199, a new Bcl-2-specific BH3 mimetic, has in vivo efficacy against aggressive Myc-driven mouse lymphomas without provoking thrombocytopenia
.
Blood
.
2013
;
121
(
12
):
2285
-
2288
.
16.
Tao
ZF
,
Hasvold
L
,
Wang
L
, et al
.
Discovery of a potent and selective BCL-XL Inhibitor with in vivo activity
.
ACS Med Chem Lett
.
2014
;
5
(
10
):
1088
-
1093
.
17.
Wagner
KU
,
Claudio
E
,
Rucker
EB
III
, et al
.
Conditional deletion of the Bcl-x gene from erythroid cells results in hemolytic anemia and profound splenomegaly
.
Development
.
2000
;
127
(
22
):
4949
-
4958
.
18.
Oltersdorf
T
,
Elmore
SW
,
Shoemaker
AR
, et al
.
An inhibitor of Bcl-2 family proteins induces regression of solid tumours
.
Nature
.
2005
;
435
(
7042
):
677
-
681
.
19.
Josefsson
EC
,
James
C
,
Henley
KJ
, et al
.
Megakaryocytes possess a functional intrinsic apoptosis pathway that must be restrained to survive and produce platelets
.
J Exp Med
.
2011
;
208
(
10
):
2017
-
2031
.
20.
Kelly
PN
,
White
MJ
,
Goschnick
MW
, et al
.
Individual and overlapping roles of BH3-only proteins Bim and Bad in apoptosis of lymphocytes and platelets and in suppression of thymic lymphoma development
.
Cell Death Differ
.
2010
;
17
(
10
):
1655
-
1664
.
21.
Mutlu
A
,
Gyulkhandanyan
AV
,
Freedman
J
,
Leytin
V
.
Activation of caspases-9, -3 and -8 in human platelets triggered by BH3-only mimetic ABT-737 and calcium ionophore A23187: caspase-8 is activated via bypass of the death receptors
.
Br J Haematol
.
2012
;
159
(
5
):
565
-
571
.
22.
Josefsson
EC
,
Burnett
DL
,
Lebois
M
, et al
.
Platelet production proceeds independently of the intrinsic and extrinsic apoptosis pathways
.
Nat Commun
.
2014
;
5
:
3455
.
23.
Dasgupta
SK
,
Abdel-Monem
H
,
Niravath
P
, et al
.
Lactadherin and clearance of platelet-derived microvesicles
.
Blood
.
2009
;
113
(
6
):
1332
-
1339
.
24.
Berger
G
,
Hartwell
DW
,
Wagner
DD
.
P-Selectin and platelet clearance
.
Blood
.
1998
;
92
(
11
):
4446
-
4452
.
25.
Schoenwaelder
SM
,
Yuan
Y
,
Josefsson
EC
, et al
.
Two distinct pathways regulate platelet phosphatidylserine exposure and procoagulant function
.
Blood
.
2009
;
114
(
3
):
663
-
666
.
26.
van Kruchten
R
,
Mattheij
NJ
,
Saunders
C
, et al
.
Both TMEM16F-dependent and TMEM16F-independent pathways contribute to phosphatidylserine exposure in platelet apoptosis and platelet activation
.
Blood
.
2013
;
121
(
10
):
1850
-
1857
.
27.
Suzuki
J
,
Umeda
M
,
Sims
PJ
,
Nagata
S
.
Calcium-dependent phospholipid scrambling by TMEM16F
.
Nature
.
2010
;
468
(
7325
):
834
-
838
.
28.
Fujii
T
,
Sakata
A
,
Nishimura
S
,
Eto
K
,
Nagata
S
.
TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets
.
Proc Natl Acad Sci USA
.
2015
;
112
(
41
):
12800
-
12805
.
29.
Brown
SB
,
Clarke
MC
,
Magowan
L
,
Sanderson
H
,
Savill
J
.
Constitutive death of platelets leading to scavenger receptor-mediated phagocytosis. A caspase-independent cell clearance program
.
J Biol Chem
.
2000
;
275
(
8
):
5987
-
5996
.
30.
Minamikawa
T
,
Williams
DA
,
Bowser
DN
,
Nagley
P
.
Mitochondrial permeability transition and swelling can occur reversibly without inducing cell death in intact human cells
.
Exp Cell Res
.
1999
;
246
(
1
):
26
-
37
.
31.
Bergmeier
W
,
Burger
PC
,
Piffath
CL
, et al
.
Metalloproteinase inhibitors improve the recovery and hemostatic function of in vitro-aged or -injured mouse platelets
.
Blood
.
2003
;
102
(
12
):
4229
-
4235
.
32.
Harrington
WJ
,
Minnich
V
,
Hollingsworth
JW
,
Moore
CV
.
Demonstration of a thrombocytopenic factor in the blood of patients with thrombocytopenic purpura
.
J Lab Clin Med
.
1951
;
38
(
1
):
1
-
10
.
33.
Ballem
PJ
,
Segal
GM
,
Stratton
JR
,
Gernsheimer
T
,
Adamson
JW
,
Slichter
SJ
.
Mechanisms of thrombocytopenia in chronic autoimmune thrombocytopenic purpura. Evidence of both impaired platelet production and increased platelet clearance
.
J Clin Invest
.
1987
;
80
(
1
):
33
-
40
.
34.
Becker
BH
,
Miller
JL
.
Effects of an antiplatelet glycoprotein Ib antibody on hemostatic function in the guinea pig
.
Blood
.
1989
;
74
(
2
):
690
-
694
.
35.
Cadroy
Y
,
Hanson
SR
,
Kelly
AB
, et al
.
Relative antithrombotic effects of monoclonal antibodies targeting different platelet glycoprotein-adhesive molecule interactions in nonhuman primates
.
Blood
.
1994
;
83
(
11
):
3218
-
3224
.
36.
Bergmeier
W
,
Rackebrandt
K
,
Schröder
W
,
Zirngibl
H
,
Nieswandt
B
.
Structural and functional characterization of the mouse von Willebrand factor receptor GPIb-IX with novel monoclonal antibodies
.
Blood
.
2000
;
95
(
3
):
886
-
893
.
37.
Cauwenberghs
N
,
Meiring
M
,
Vauterin
S
, et al
.
Antithrombotic effect of platelet glycoprotein Ib-blocking monoclonal antibody Fab fragments in nonhuman primates
.
Arterioscler Thromb Vasc Biol
.
2000
;
20
(
5
):
1347
-
1353
.
38.
Provan
D
,
Stasi
R
,
Newland
AC
, et al
.
International consensus report on the investigation and management of primary immune thrombocytopenia
.
Blood
.
2010
;
115
(
2
):
168
-
186
.
39.
Segal
JB
,
Powe
NR
.
Prevalence of immune thrombocytopenia: analyses of administrative data
.
J Thromb Haemost
.
2006
;
4
(
11
):
2377
-
2383
.
40.
Soslau
G
,
Giles
J
.
The loss of sialic acid and its prevention in stored human platelets
.
Thromb Res
.
1982
;
26
(
6
):
443
-
455
.
41.
Choi
SI
,
Simone
JV
,
Jorney
LJ
.
Neuraminidase-induced thrombocytopenia in rats
.
Br J Haematol
.
1972
;
22
(
1
):
93
-
101
.
42.
Grewal
PK
,
Uchiyama
S
,
Ditto
D
, et al
.
The Ashwell receptor mitigates the lethal coagulopathy of sepsis
.
Nat Med
.
2008
;
14
(
6
):
648
-
655
.
43.
Rumjantseva
V
,
Grewal
PK
,
Wandall
HH
, et al
.
Dual roles for hepatic lectin receptors in the clearance of chilled platelets
.
Nat Med
.
2009
;
15
(
11
):
1273
-
1280
.
44.
Jansen
AJ
,
Josefsson
EC
,
Rumjantseva
V
, et al
.
Desialylation accelerates platelet clearance after refrigeration and initiates GPIbα metalloproteinase-mediated cleavage in mice
.
Blood
.
2012
;
119
(
5
):
1263
-
1273
.
45.
Li
J
,
van der Wal
DE
,
Zhu
G
, et al
.
Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia
.
Nat Commun
.
2015
;
6
(
1
):
7737
.
46.
Deng
W
,
Xu
Y
,
Chen
W
, et al
.
Platelet clearance via shear-induced unfolding of a membrane mechanoreceptor
.
Nat Commun
.
2016
;
7
:
12863
.
47.
Grewal
PK
.
The Ashwell-Morell receptor
.
Methods Enzymol
.
2010
;
479
:
223
-
241
.
48.
Sørensen
AL
,
Rumjantseva
V
,
Nayeb-Hashemi
S
, et al
.
Role of sialic acid for platelet life span: exposure of beta-galactose results in the rapid clearance of platelets from the circulation by asialoglycoprotein receptor-expressing liver macrophages and hepatocytes
.
Blood
.
2009
;
114
(
8
):
1645
-
1654
.
49.
Li
Y
,
Fu
J
,
Ling
Y
, et al
.
Sialylation on O-glycans protects platelets from clearance by liver Kupffer cells
.
Proc Natl Acad Sci USA
.
2017
;
114
(
31
):
8360
-
8365
.
50.
Lodish
HF
.
Recognition of complex oligosaccharides by the multi-subunit asialoglycoprotein receptor
.
Trends Biochem Sci
.
1991
;
16
(
10
):
374
-
377
.
51.
Grozovsky
R
,
Begonja
AJ
,
Liu
K
, et al
.
The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling
.
Nat Med
.
2015
;
21
(
1
):
47
-
54
.
52.
Josefsson
EC
,
Gebhard
HH
,
Stossel
TP
,
Hartwig
JH
,
Hoffmeister
KM
.
The macrophage alphaMbeta2 integrin alphaM lectin domain mediates the phagocytosis of chilled platelets
.
J Biol Chem
.
2005
;
280
(
18
):
18025
-
18032
.
53.
Hoffmeister
KM
,
Felbinger
TW
,
Falet
H
, et al
.
The clearance mechanism of chilled blood platelets
.
Cell
.
2003
;
112
(
1
):
87
-
97
.
54.
Casari
C
,
Du
V
,
Wu
YP
, et al
.
Accelerated uptake of VWF/platelet complexes in macrophages contributes to VWD type 2B-associated thrombocytopenia
.
Blood
.
2013
;
122
(
16
):
2893
-
2902
.
55.
Yan
R
,
Chen
M
,
Ma
N
, et al
.
Glycoprotein Ibα clustering induces macrophage-mediated platelet clearance in the liver
.
Thromb Haemost
.
2015
;
113
(
1
):
107
-
117
.
56.
Wandall
HH
,
Hoffmeister
KM
,
Sørensen
AL
, et al
.
Galactosylation does not prevent the rapid clearance of long-term, 4 degrees C-stored platelets
.
Blood
.
2008
;
111
(
6
):
3249
-
3256
.
57.
Gangarosa
EJ
,
Landerman
NS
,
Rosch
PJ
,
Herndon
EG
Jr
.
Hematologic complications arising during ristocetin therapy; relation between dose and toxicity
.
N Engl J Med
.
1958
;
259
(
4
):
156
-
161
.
58.
Sanders
WE
,
Read
MS
,
Reddick
RL
,
Garris
JB
,
Brinkhous
KM
.
Thrombotic thrombocytopenia with von Willebrand factor deficiency induced by botrocetin. An animal model
.
Lab Invest
.
1988
;
59
(
4
):
443
-
452
.
59.
Lillicrap
D
.
von Willebrand disease: advances in pathogenetic understanding, diagnosis, and therapy
.
Blood
.
2013
;
122
(
23
):
3735
-
3740
.
60.
Tischer
A
,
Madde
P
,
Moon-Tasson
L
,
Auton
M
.
Misfolding of vWF to pathologically disordered conformations impacts the severity of von Willebrand disease
.
Biophys J
.
2014
;
107
(
5
):
1185
-
1195
.
61.
Sanders
WE
Jr
,
Reddick
RL
,
Nichols
TC
,
Brinkhous
KM
,
Read
MS
.
Thrombotic thrombocytopenia induced in dogs and pigs. The role of plasma and platelet vWF in animal models of thrombotic thrombocytopenic purpura
.
Arterioscler Thromb Vasc Biol
.
1995
;
15
(
6
):
793
-
800
.
62.
Nascimbene
A
,
Neelamegham
S
,
Frazier
OH
,
Moake
JL
,
Dong
JF
.
Acquired von Willebrand syndrome associated with left ventricular assist device
.
Blood
.
2016
;
127
(
25
):
3133
-
3141
.
63.
Webster
ML
,
Sayeh
E
,
Crow
M
, et al
.
Relative efficacy of intravenous immunoglobulin G in ameliorating thrombocytopenia induced by antiplatelet GPIIbIIIa versus GPIbalpha antibodies
.
Blood
.
2006
;
108
(
3
):
943
-
946
.
64.
Zeng
Q
,
Zhu
L
,
Tao
L
, et al
.
Relative efficacy of steroid therapy in immune thrombocytopenia mediated by anti-platelet GPIIbIIIa versus GPIbα antibodies
.
Am J Hematol
.
2012
;
87
(
2
):
206
-
208
.
65.
Peng
J
,
Ma
SH
,
Liu
J
, et al
.
Association of autoantibody specificity and response to intravenous immunoglobulin G therapy in immune thrombocytopenia: a multicenter cohort study
.
J Thromb Haemost
.
2014
;
12
(
4
):
497
-
504
.
66.
Quach
ME
,
Dragovich
MA
,
Chen
W
, et al
.
Fc-independent immune thrombocytopenia via mechanomolecular signaling in platelets
.
Blood
.
2018
;
131
(
7
):
787
-
796
.
67.
Maurer
E
,
Tang
C
,
Schaff
M
, et al
.
Targeting platelet GPIbβ reduces platelet adhesion, GPIb signaling and thrombin generation and prevents arterial thrombosis
.
Arterioscler Thromb Vasc Biol
.
2013
;
33
(
6
):
1221
-
1229
.
68.
Liang
X
,
Syed
AK
,
Russell
SR
,
Ware
J
,
Li
R
.
Dimerization of glycoprotein Ibα is not sufficient to induce platelet clearance
.
J Thromb Haemost
.
2016
;
14
(
2
):
381
-
386
.
69.
Shrimpton
CN
,
Borthakur
G
,
Larrucea
S
,
Cruz
MA
,
Dong
JF
,
López
JA
.
Localization of the adhesion receptor glycoprotein Ib-IX-V complex to lipid rafts is required for platelet adhesion and activation
.
J Exp Med
.
2002
;
196
(
8
):
1057
-
1066
.
70.
Jin
W
,
Inoue
O
,
Tamura
N
, et al
.
A role for glycosphingolipid-enriched microdomains in platelet glycoprotein Ib-mediated platelet activation
.
J Thromb Haemost
.
2007
;
5
(
5
):
1034
-
1040
.
71.
Kroll
MH
,
Hellums
JD
,
McIntire
LV
,
Schafer
AI
,
Moake
JL
.
Platelets and shear stress
.
Blood
.
1996
;
88
(
5
):
1525
-
1541
.
72.
Zhang
W
,
Deng
W
,
Zhou
L
, et al
.
Identification of a juxtamembrane mechanosensitive domain in the platelet mechanosensor glycoprotein Ib-IX complex
.
Blood
.
2015
;
125
(
3
):
562
-
569
.
73.
Bergmeier
W
,
Piffath
CL
,
Cheng
G
, et al
.
Tumor necrosis factor-α-converting enzyme (ADAM17) mediates GPIbalpha shedding from platelets in vitro and in vivo
.
Circ Res
.
2004
;
95
(
7
):
677
-
683
.
74.
Gardiner
EE
,
Karunakaran
D
,
Shen
Y
,
Arthur
JF
,
Andrews
RK
,
Berndt
MC
.
Controlled shedding of platelet glycoprotein (GP)VI and GPIb-IX-V by ADAM family metalloproteinases
.
J Thromb Haemost
.
2007
;
5
(
7
):
1530
-
1537
.
75.
Tao
Y
,
Zhang
X
,
Liang
X
,
Zang
J
,
Mo
X
,
Li
R
.
Structural basis for the specific inhibition of glycoprotein Ibα shedding by an inhibitory antibody
.
Sci Rep
.
2016
;
6
(
1
):
24789
.
76.
Li
S
,
Wang
Z
,
Liao
Y
, et al
.
The glycoprotein Ibalpha-von Willebrand factor interaction induces platelet apoptosis
.
J Thromb Haemost
.
2010
;
8
(
2
):
341
-
350
.
77.
Li
Z
,
Delaney
MK
,
O’Brien
KA
,
Du
X
.
Signaling during platelet adhesion and activation
.
Arterioscler Thromb Vasc Biol
.
2010
;
30
(
12
):
2341
-
2349
.
78.
Murphy
S
,
Gardner
FH
.
Platelet storage at 22 degrees C; metabolic, morphologic, and functional studies
.
J Clin Invest
.
1971
;
50
(
2
):
370
-
377
.
79.
Holme
S
,
Moroff
G
,
Murphy
S
;
Biomedical Excellence for Safer Transfusion Working Party of the International Society of Blood Transfusion
.
A multi-laboratory evaluation of in vitro platelet assays: the tests for extent of shape change and response to hypotonic shock
.
Transfusion
.
1998
;
38
(
1
):
31
-
40
.
80.
Snyder
EL
,
Hezzey
A
,
Katz
AJ
,
Bock
J
.
Occurrence of the release reaction during preparation and storage of platelet concentrates
.
Vox Sang
.
1981
;
41
(
3
):
172
-
177
.
81.
Seghatchian
J
,
Krailadsiri
P
.
The platelet storage lesion
.
Transfus Med Rev
.
1997
;
11
(
2
):
130
-
144
.
82.
Li
J
,
Xia
Y
,
Bertino
AM
,
Coburn
JP
,
Kuter
DJ
.
The mechanism of apoptosis in human platelets during storage
.
Transfusion
.
2000
;
40
(
11
):
1320
-
1329
.
83.
Shiri
R
,
Yari
F
,
Ahmadinejad
M
,
Vaeli
S
,
Tabatabaei
MR
.
The caspase-3 inhibitor (peptide Z-DEVD-FMK) affects the survival and function of platelets in platelet concentrate during storage
.
Blood Res
.
2014
;
49
(
1
):
49
-
53
.
84.
Catani
MV
,
Gasperi
V
,
Evangelista
D
,
Finazzi Agrò
A
,
Avigliano
L
,
Maccarrone
M
.
Anandamide extends platelets survival through CB(1)-dependent Akt signaling
.
Cell Mol Life Sci
.
2010
;
67
(
4
):
601
-
610
.
85.
Zhuang
Y
,
Ren
G
,
Li
H
, et al
.
In vitro properties of apheresis platelet during extended storage in plasma treated with anandamide
.
Transfus Apheresis Sci
.
2014
;
51
(
1
):
58
-
64
.
86.
Bradley
AJ
,
Read
BL
,
Levin
E
,
Devine
DV
.
Small-molecule complement inhibitors cannot prevent the development of the platelet storage lesion
.
Transfusion
.
2008
;
48
(
4
):
706
-
714
.
87.
Perrotta
PL
,
Perrotta
CL
,
Snyder
EL
.
Apoptotic activity in stored human platelets
.
Transfusion
.
2003
;
43
(
4
):
526
-
535
.
88.
Leaver
HA
,
Schou
AC
,
Rizzo
MT
,
Prowse
CV
.
Calcium-sensitive mitochondrial membrane potential in human platelets and intrinsic signals of cell death
.
Platelets
.
2006
;
17
(
6
):
368
-
377
.
89.
Perales Villarroel
JP
,
Figueredo
R
,
Guan
Y
, et al
.
Increased platelet storage time is associated with mitochondrial dysfunction and impaired platelet function
.
J Surg Res
.
2013
;
184
(
1
):
422
-
429
.
90.
Hayashi
T
,
Tanaka
S
,
Hori
Y
,
Hirayama
F
,
Sato
EF
,
Inoue
M
.
Role of mitochondria in the maintenance of platelet function during in vitro storage
.
Transfus Med
.
2011
;
21
(
3
):
166
-
174
.
91.
Hartley
PS
,
Savill
J
,
Brown
SB
.
The death of human platelets during incubation in citrated plasma involves shedding of CD42b and aggregation of dead platelets
.
Thromb Haemost
.
2006
;
95
(
1
):
100
-
106
.
92.
Michelson
AD
,
Adelman
B
,
Barnard
MR
,
Carroll
E
,
Handin
RI
.
Platelet storage results in a redistribution of glycoprotein Ib molecules. Evidence for a large intraplatelet pool of glycoprotein Ib
.
J Clin Invest
.
1988
;
81
(
6
):
1734
-
1740
.
93.
Kostelijk
EH
,
Folman
CC
,
Gouwerok
CW
,
Kramer
CM
,
Verhoeven
AJ
,
de Korte
D
.
Increase in glycocalicin levels in platelet concentrates stored in plasma or synthetic medium for 8 days: comparison with other platelet activation markers
.
Vox Sang
.
2000
;
79
(
1
):
21
-
26
.
94.
Canault
M
,
Duerschmied
D
,
Brill
A
, et al
.
p38 mitogen-activated protein kinase activation during platelet storage: consequences for platelet recovery and hemostatic function in vivo
.
Blood
.
2010
;
115
(
9
):
1835
-
1842
.
95.
Liang
X
,
Russell
SR
,
Estelle
S
, et al
.
Specific inhibition of ectodomain shedding of glycoprotein Ibα by targeting its juxtamembrane shedding cleavage site
.
J Thromb Haemost
.
2013
;
11
(
12
):
2155
-
2162
.
96.
Murphy
S
,
Gardner
FH
.
Effect of storage temperature on maintenance of platelet viability--deleterious effect of refrigerated storage
.
N Engl J Med
.
1969
;
280
(
20
):
1094
-
1098
.
97.
van der Wal
DE
,
Du
VX
,
Lo
KS
,
Rasmussen
JT
,
Verhoef
S
,
Akkerman
JW
.
Platelet apoptosis by cold-induced glycoprotein Ibα clustering
.
J Thromb Haemost
.
2010
;
8
(
11
):
2554
-
2562
.
98.
Badlou
BA
,
Spierenburg
G
,
Ulrichts
H
,
Deckmyn
H
,
Smid
WM
,
Akkerman
JW
.
Role of glycoprotein Ibalpha in phagocytosis of platelets by macrophages
.
Transfusion
.
2006
;
46
(
12
):
2090
-
2099
.
99.
van der Wal
DE
,
Gitz
E
,
Du
VX
, et al
.
Arachidonic acid depletion extends survival of cold-stored platelets by interfering with the [glycoprotein Ibα--14-3-3ζ] association
.
Haematologica
.
2012
;
97
(
10
):
1514
-
1522
.
100.
Chen
W
,
Druzak
SA
,
Wang
Y
, et al
.
Refrigeration-induced binding of von Willebrand factor facilitates fast clearance of refrigerated platelets
.
Arterioscler Thromb Vasc Biol
.
2017
;
37
(
12
):
2271
-
2279
.
Sign in via your Institution