Next Article in Journal
Identification of Specific microRNAs in Adipose Tissue Affected by Lipedema
Next Article in Special Issue
Extracellular Vesicles: A Novel Diagnostic Tool and Potential Therapeutic Approach for Equine Osteoarthritis
Previous Article in Journal
Transcriptomic Remodeling Occurs During Cambium Activation and Xylem Cell Development in Taxodium ascendens
Previous Article in Special Issue
L-gulono-γ-lactone Oxidase, the Key Enzyme for L-Ascorbic Acid Biosynthesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment

by
Shiying Silvia Zheng
1,2 and
José Sail Perdomo
3,*
1
Haematology Research Unit, St. George and Sutherland Clinical Campuses, School of Medicine & Health, University of New South Wales, Kogarah, NSW 2217, Australia
2
Department of Haematology, St. George Hospital, Kogarah, NSW 2217, Australia
3
Haematology Research Group, Central Clinical School, Faculty Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
*
Author to whom correspondence should be addressed.
Curr. Issues Mol. Biol. 2024, 46(11), 11942-11956; https://doi.org/10.3390/cimb46110709
Submission received: 16 September 2024 / Revised: 18 October 2024 / Accepted: 22 October 2024 / Published: 24 October 2024
(This article belongs to the Special Issue Latest Review Papers in Molecular Biology 2024)

Abstract

:
Immune thrombocytopenia (ITP) is an autoimmune disease in which platelet autoantibodies play a significant role in its pathogenesis. Regulatory T cell dysfunction and T cell-mediated cytotoxicity also contribute to thrombocytopenia. Current therapies are directed towards immune suppression and modulation as well as stimulation of platelet production with thrombopoietin receptor agonists. Additional mechanisms of the pathogenesis of ITP have been suggested by recent experimental data. One of these processes, known as desialylation, involves antibody-induced removal of terminal sialic acid residues on platelet surface glycoproteins, leading to hepatic platelet uptake and thrombocytopenia. Apoptosis, or programmed platelet death, may also contribute to the pathogenesis of ITP. The extent of the impact of desialylation and apoptosis on ITP, the relative proportion of patients affected, and the role of antibody specificity are still the subject of investigation. This review will discuss both historical and new evidence of the influence of desialylation and apoptosis in the pathogenesis of ITP, with an emphasis on the clinical implications of these developments. Further understanding of both platelet desialylation and apoptosis might change current clinical practice and improve patient outcomes.

1. Introduction

Immune thrombocytopenia (ITP) is an acquired autoimmune disease characterised by enhanced platelet destruction and impaired platelet production. Patients can present with no known predisposing condition (primary ITP) or due to an associated disorder such as malignancy, infection, and autoimmune disease (secondary ITP) [1]. Drug-induced ITP is also well recognised [2]. There are three phases of the disease: newly diagnosed (<3 months), persistent (3–12 months), and chronic (>12 months) [3]. ITP affects both children and adults. Adult ITP is typically chronic. Only about 20–30% of cases achieve long-term remission, and many patients require long-term therapy. Primary ITP comprises approximately 80% of all adult ITP cases [4]. On the other hand, paediatric ITP is often triggered by viral infections and is usually acute and self-limiting, with 80% of children experiencing spontaneous remission. In children, treatment is often unnecessary [1,5]. ITP is a heterogeneous disease. Some patients require multiple lines of therapy, while others are managed by observation only. As a result, the quality of life for patients is affected to different degrees.
In addition to its clinical heterogeneity, ITP is a complex disease, and no single proposed mechanism can fully explain its pathogenesis. Early and current data suggest that autoimmunity is responsible for causing thrombocytopenia [6,7,8,9]. Over the past decades, further evidence has emerged, showing that the dysregulation of the immune system is multifactorial. In addition to the production of antiplatelet antibodies by B and plasma cells, regulatory T cell dysfunction and cytotoxic T cell-mediated platelet destruction also contribute to the pathogenesis of ITP. More recent advances further demonstrate that autoimmunity shortens platelet survival through the modification of platelet surface glycans and may hasten platelet programmed cell death.
This review will focus on the key aspects of recent research regarding platelet desialylation and apoptosis, their relevance to ITP pathology, and the diagnostic and treatment implications.

2. Platelet Biogenesis and the Regulation of Lifespan

Platelets are derived from megakaryocytes, which reside in the bone marrow and to a lesser extent in the blood and lungs. It is estimated that 1011 platelets are generated each day in healthy adults [10,11]. Thrombopoietin (TPO) is the primary growth factor that drives thrombopoiesis. TPO stimulates the differentiation of haematopoietic stem cells into megakaryocytic precursors and their proliferation and maturation [12,13]. TPO is synthesised primarily in the liver [14], but expression in kidney and bone marrow has been demonstrated [15]. TPO binds to its c-Mpl receptors on haematopoietic stem cells and platelets [14,16], which provides a mechanism for the regulation of circulating TPO levels [17]. During thrombocytosis, more platelets, and therefore more c-Mpl receptors, are available to bind and internalise TPO. This reduces the amount of free TPO, thus decreasing both megakaryopoiesis and platelet formation. In thrombocytopenic states, for example, due to myelosuppressive therapy, less TPO is internalised by platelets and more TPO is available to bind to megakaryocytes, stimulating thrombopoiesis [18]. Curiously, TPO levels are not significantly increased in ITP compared to non-immune thrombocytopenia. This could be due to the markedly increased platelet turnover rate despite the thrombocytopenic state in ITP [19].

2.1. Platelet Desialylation and the Regulation of Lifespan

Another model of TPO regulation is by controlling the TPO mRNA expression. In the last decade, a mechanism of TPO mRNA regulation was described. In a mouse model, Grozovsky and colleagues showed that aged platelets, by losing their terminal sialic acid groups attached to glycoproteins (desialylation), became recognisable by the hepatic Ashwell–Morell receptors (AMR) [20]. Glycoprotein (GP) Ibα, a highly expressed membrane-bound receptor on platelets, has an extracellular domain with an abundance of N-linked glycans (Figure 1) [21,22]. Hence, GPIbα is thought to be a major target of desialylation. The recognition of desialylated GPs by hepatocytes leads to the hepatic internalisation of platelets, which stimulates Janus kinase 2 (JAK2) and activates signal transduction and transcription 3 (STAT3). This signalling drives hepatic TPO mRNA expression (Figure 2). As a result, platelet production is increased, and platelet mass is restored [20]. In fact, a link between desialylation and platelet survival has been known for decades [23] (Table 1).

2.2. Platelet Apoptosis and Platelet Lifespan

Unlike desialylation (Table 1), the role of apoptosis was only described more recently (Table 2). In the steady state, human platelet lifespan is approximately ten days [39,40]. Intrinsic apoptosis, as triggered by non-receptor-driven events, is probably the most important regulator of platelet life span [41,42,43]. Two seminal studies showed that proteins of the BCL2 family are the key players in this pathway [42,44]. BCL-XL is a pro-survival protein that sustains platelet survival by restraining the pro-death activity of the “multidomain killers” BAD, BAK1, and BAX [41]. In the cytosol, 14-3-3ζ exists in complexes with the pro-apoptotic proteins, controlling their activity by maintaining phosphorylation [45]. It has been hypothesised that, as platelets age, they gradually lose BCL-XL due to degradation. However, BAK1 is degraded more slowly than BCL-XL, eventually reaching a point when the proapoptotic activity can no longer be restrained [42]. Free BAX transits to the mitochondria and induces mitochondrial membrane permeabilization, cytochrome c release, and activation of caspases, which results in platelet apoptosis [41] (Figure 3). Utilising a potent BCL-XL antagonist ABT-737 and 111indium-labelled platelets whole body scintigraphy in an animal model, Zhang and colleagues identified the liver as the site of clearance of apoptotic platelets [44].
It is still unclear if platelet apoptosis and desialylation are mechanistically linked during platelet ageing. Using AMR knockout mice, a preliminary report showed that desialylated platelets are more prone to apoptosis [46]. However, the induction of platelet apoptosis did not induce platelet desialylation [46]. It is possible that the platelet ageing process involves the gradual loss of sialic acid in the circulation, followed by enhanced responsiveness to apoptotic stimuli, which result in the liberation of the pro-death proteins BAK1 and BAX and the eventual platelet apoptosis [46]. Nevertheless, stronger evidence is needed to definitively correlate desialylation with apoptosis in the regulation of platelet lifespan.
Table 2. Evidence for platelet apoptosis.
Table 2. Evidence for platelet apoptosis.
CommentReference; Year
Nucleus is not required for apoptosis[47]; 1994
Apoptosis-like events associated with platelet activation[48]; 1999
Anti-platelet antibodies modulate caspase activity and regulate platelet lifespan in mice[49]; 2002
Apoptosis associated with shortened platelet survival in rabbits[50]; 2004
Anti GPIIb antibody induces platelet apoptosis in mice[51]; 2006
Thrombin induces platelet apoptosis[52,53]; 2007, 2006
Apoptosis program controls platelet lifespan[42]; 2007
Cold storage leads to platelet apoptosis[54]; 2010
Apoptotic platelets observed in paediatric patients with ITP[55]; 2012
Platelet apoptosis in adult ITP[56,57]; 2018, 2016
The presence of anti-platelet antibodies predicts apoptosis in ITP[38]; 2022

3. Immune Thrombocytopenia

3.1. Presentation

The main symptoms and signs of ITP are bleeding, ranging from minor petechiae and purpure to severe haemorrhage, such as gastrointestinal and intracranial bleeding. The severity generally correlates with the degree of thrombocytopenia and the patient’s age. Older patients with a persistently low platelet count of under 30 × 109/L are at a higher risk of bleeding [58]. Notable exceptions are patients with antiplatelet autoantibodies with platelet function inhibitory effects. Occasionally, these antibodies interfere with fibrinogen binding to platelet GPIIb/IIIa or von Willebrand factor (vWF) binding to GPIb/IX receptors, resulting in severe bleeding phenotypes from acquired Glanzmann’s thrombasthenia or Bernard–Soulier syndrome, respectively [59]. These patients have severe bleeding symptoms regardless of their platelet counts. Unexpectedly, patients with ITP are also more susceptible to both venous and arterial thromboembolism [60]. In addition, patients with ITP frequently suffer from fatigue, impaired quality of life [61], and complications related to treatments [62]. Unfortunately, the mortality rates are consistently higher amongst ITP patients than in the general population [63], ranging from 1.3- to 2.2-fold [62,64,65]. Therefore, ITP is a complex disease, and understanding its pathogenesis is of importance in order to improve the current management.

3.2. Pathogenesis

3.2.1. Fc-Dependent Pathway

It was the seminal Harrington–Hollingsworth experiment of self-infusion of ITP plasma, which led to the discovery of a humoral factor accountable for mediating platelet destruction [6,7]. Subsequently, Shulman identified that such a factor could be adsorbed by platelets and was associated with immunoglobulin G (IgG) [8]. IgM and IgA antiplatelet antibodies in the absence of IgG have been described but are rather uncommon in adult ITP [11]. In contrast, IgM may play a dominant role in acute paediatric ITP [66]. These autoantibodies are commonly directed against the platelet membrane GP complexes, and GPIIb/IIIa and GPIb/IX are most frequently targeted [67,68]. Sole specificity against GPIaIIa and GPVI antibodies is infrequent [69,70,71]. Regarding antibodies against platelet GPV, which can bind to the GPV subunit independently of the GPIb/IX complex [72], in a recent evaluation, Porcelijn reported that 5% of the 60 patients with ITP tested for anti-platelet antibodies had sole anti-GPV antibodies [73]. In fact, antibodies against more than one GP receptor are much more common in ITP [68,71].
These autoantibodies play a significant role in ITP pathogenesis. One of the most widely accepted mechanisms of platelet destruction is that opsonised platelets are phagocytosed by splenic and/or hepatic macrophages of the reticuloendothelial system via their Fc-gamma receptors (FcγR) [11,74]. Hence, this antibody-mediated platelet clearance is called the Fc-dependent pathway. Like other autoimmune diseases, ITP signifies the loss of self-tolerance. Self-reactive T cells are believed to be one of the central causes of perpetuating autoimmunity, as B cells’ ability to produce antibodies is supported by antigen-specific T cells. Studies on CD4+FOXP3+ T cells or T regulatory cells (Treg) have shown both reduction [75,76,77] and dysfunction of Treg [77] in patients with untreated ITP. Moreover, in patients with active ITP, the percentage of T-helper (Th) 17 cells is significantly higher than normal controls [78]. Interestingly, T cell clonality has been reported and implicated in treatment failure [79,80,81]. Large prospective trials are required to determine how clinicians can use this information to tailor ITP treatment strategies.

3.2.2. Fc-Independent Pathway

Platelet Desialylation
Fc-independent platelet clearance has also been strongly suggested for anti-GPIbα antibodies-mediated ITP. Using monoclonal antibodies in a mouse model of ITP, Nieswandt and colleagues first demonstrated that removing the Fc fragment of anti-GPIbα monoclonal antibodies did not prevent thrombocytopenia, whereas the opposite was noted for anti-GPIIb/IIIa antibodies, denoting the Fc independence of anti-GPIbα antibodies in causing thrombocytopenia [82]. Since then, there have been further advances in the understanding of this alternative platelet clearance pathway. In 2003, Hoffmeister and colleagues observed that short-term (2 h) platelet cooling to 4 °C resulted in GPIb receptor clustering and rapid clearance of platelets by Mac-1 receptor-expressing Kupffer cells in the liver [83,84]. From 2009 to 2012, the same group discovered that the sialidase neuraminidase-1 (NEU1) was translocated to the platelet surface following longer-term (≥48 h) refrigeration of platelets. NEU1 mediates removal of terminal sialic acid residues on GPIbα, leading to platelet clearance by hepatocytes via the Ashwell–Morell receptors (AMR) [29,30,85].
More recently, Li and colleagues reported the same FcγR-independent model of platelet clearance by anti-GPIbα antibodies. Using both monoclonal antibodies and plasma from patients with ITP, they reported platelet activation secondary to GPIbα antibodies. This led to NEU1 translocation to platelet surface and platelet desialylation, as well as the resultant platelet removal by AMR on hepatocytes and hence, thrombocytopenia. This finding indicated that detection of platelet desialylation in patients with ITP might have potential diagnostic and therapeutic implications. In vivo evidence suggested that sialidase inhibitors may attenuate thrombocytopenia in ITP secondary to anti-GPIbα antibodies, while splenectomy, which reduces Fc-dependent phagocytosis of opsonised platelets, may be of little benefit for these patients [32]. However, this animal model using monoclonal antibodies may not be directly relevant to human ITP due to the lack of platelet surface FcγRIIa on murine platelets [86] and the polyclonal nature of autoantibodies from primary ITP [87]. In fact, when Cantoni et al. examined the antibody specificities and the location of platelet clearance using indium-111-labelled platelets of 93 patients with ITP, no association between antibody specificities and the site of platelet sequestration was found [88]. These observations are tempered by the fact that only four patients had sole GPIb/IX antibodies. Of those, two patients had splenic uptake while the other two had hepatic and mixed platelet sequestration patterns, respectively [88]. In a later study, Amini and colleagues examined 53 patients and reported an association between the presence of anti-GPIbα and hepatic sequestration only in patients with a platelet count under 50 × 109/L [89]. Again, there were only four patients with anti-GPIbα antibodies without concomitant GPIIb/IIIa antibodies. In fact, when GPV antibodies were excluded, there was only one patient with sole anti-GPIbα antibodies [89].
Whether anti-GPIbα antibodies induce platelet desialylation and hepatic sequestration might be dose-dependent. In a murine model, Morodomi and co-workers reported that high-dose intravenous administration of monoclonal anti-mouse GPIbα antibodies led to platelet clearance by both the spleen and liver, while low-dose subcutaneous administration only led to splenic uptake [90]. Furthermore, the discrepancy between Li’s and Cantoni’s findings may also be because the true extent to which desialylation was involved in ITP pathology was unknown at the time. Recent research demonstrated that antibodies against GPIIb/IIIa are also capable of inducing platelet and megakaryocyte desialylation [35,36,37,38]. Unlike previous reports linking antibody-mediated platelet activation to desialylation [32], these latest findings did not find evidence of platelet activation [36,37,38]. Instead, when FcγR is inhibited by monoclonal antibody IV.3, desialylation is reduced [36,37]. Hence, anti-GPIIb/IIIa antibodies potentially signal through FcγRIIa to drive neuraminidase surface translocation and platelet desialylation. More importantly, these studies also confirmed the effectiveness of neuraminidase inhibitor oseltamivir in reducing platelet destruction in vivo in the setting of anti-GPIIb/IIIa antibodies-mediated ITP [37,38], providing further evidence that both anti-GPIb/IX and anti-GPIIb/IIIa antibodies may lead to platelet desialylation.
Platelet apoptosis
Platelet apoptosis has also been implicated in the pathogenic processes of ITP. In 2006, Leytin and colleagues reported that anti-GPIIb antibody injection in mice induced thrombocytopenia, platelet caspase-3 activation, enhanced phosphatidylserine (PS) exposure and mitochondrial inner transmembrane potential (ΔΨm) depolarisation. Capsase-3, PS exposure and ΔΨm depolarisation are markers of platelet apoptosis [51]. Leytin also demonstrated that treatment with IVIg prior to antibody injection ameliorated the degree of thrombocytopenia, reduced caspase-3 activation and PS exposure but not ΔΨm depolarisation. In 2012, Winkler and colleagues reported similar findings in paediatric patients with ITP [55]. However, the number of patients was small (20) and no correlation with antibody specificities was made.
More recently, Qiao and co-workers showed in vitro data of reduced expression of BCL-XL and increased BAX in platelets treated with ITP plasma [91]. The researchers employed Western blot and quantitative PCR techniques to evaluate mRNA extracted from washed donor platelets treated from patients with ITP and control plasma. The results indicated a lower mRNA and protein expression of BCL-XL but an increased mRNA and protein expression of BAX in platelets treated with plasma from patients with ITP when compared to control plasma [91]. However, as platelets lack nuclei, the authors did not describe the mechanism of the changes in RNA level. Possible explanations include alterations in RNA splicing of immature RNAs [92] and modification of mRNA degradation rates [93]. Nevertheless, the imbalanced expression of BCL-XL and BAX at the protein level provided additional evidence of the loss of proapoptotic restrain in ITP. Again, antibody specificity was not examined in this study.
In addition to desialylation, platelet cold storage also leads to platelet apoptosis upon rewarming [54]. Like desialylation, cold storage induced platelet apoptosis may be a GPIbα driven event. Upon rewarming, the GPIbα receptor clusters, which leads to phosphorylation of its cytoplasmic domains. These phosphorylated sites have strong affinity for the above-mentioned adaptor protein 14-3-3ζ [94]. Consequently, BAX is released from 14-3-3ζ, dephosphorylated and becomes active, which leads to platelet apoptosis [54,95]. Other events, including binding of ligands such as vWF [96] and thrombin stimulation [52,53] may also result in platelet apoptosis [97].
Furthermore, both desialylated and apoptotic platelets are removed by the liver [32,44,85]. To date, a mechanistic link between platelet desialylation and apoptosis is still not fully explored (Table 1 and Table 2). From the ITP perspective, anti-GPIbα antibodies were reported to cause platelet desialylation like cold storage [32]. Yet, Leytin et al. found that monoclonal anti-GPIIb antibodies caused platelet apoptosis in a murine ITP model [51]. Whether there is an association between intrinsic apoptosis and ITP caused by anti-GPIbα and/or anti-GPIIbIIIa antibodies is still unclear. Our group analysed the differential impact of these two antibody subtypes on platelet fate, focusing on platelet desialylation and apoptosis. In contrast to Li and colleagues’ report, we found a greater proportion of patients’ sera with anti-GPIIb/IIIa antibodies led to positive neuraminidase surface translocation, while a significant number of sera from patients with sole anti-GPIbα antibodies caused platelet apoptosis [38]. Although the number of patients with sole antibody positivity was small (n = 9 for anti-GPIIb/IIIa, n = 5 for anti-GPIbα), similar findings of anti-GPIIb/IIIa antibodies indicate a greater ability to induce desialylation [35] and for anti-GPIbα to induce apoptosis [57] have been described (Table 2). Therefore, not only does the presence of platelet antibodies predict platelet desialylation and apoptosis, but the actual antibody specificity may also have implications on the ultimate platelet clearance pathway. This raises the potential of ITP treatment individualisation if such findings can be confirmed in future collaborative prospective evaluations.
T cell mediated cytotoxicity
Since not all ITP plasma-induced thrombocytopenia in healthy volunteers is from the famous Harrington experiments [6], and only about 50% of ITP patients have detectable anti-platelet antibodies [98], non-antibody-mediated mechanisms of ITP should be highlighted. Specifically, autoreactive CD8+ cytotoxic T cells causing platelet lysis have been reported [99,100,101]. Interestingly, Zhao et al. found a negative correlation with the detectability of platelet antibodies and the presence of platelet lysis by cytotoxic T cells, which suggested that T cell-mediated cytotoxicity could be a dominant mechanism in those patients without detectable antibodies [101]. Recently, it was shown that cytotoxic CD8+ T cells from patients with ITP induced platelet desialylation and apoptosis [102]. This may be a result of platelet activation through T cell receptor-mediated cytotoxic granule release [103]. Moreover, CD8+ cytotoxic T cells may also affect platelet production in ITP by megakaryocytes by paradoxically increasing megakaryopoiesis and decreasing apoptosis [104]. Changes in the steady state of apoptosis in megakaryocytes seem to correlate with lower platelet production [105].

4. Therapeutic Implications

Treatment can be divided into first-line [steroids, intravenous immunoglobulin (IVIg), and anti-Rh(D) (anti-D)], second-line [TPO receptor agonists (TPO-RA), rituximab, splenectomy] and subsequent therapies [4,106]. The currently available therapies are largely targeting the Fc-dependent pathway (Figure 4). Agents such as corticosteroids [107,108], rituximab [109,110], and cytotoxics (for example, vinca alkaloids and azathioprine [111,112]) are used to reduce antibody production. The Bruton kinase inhibitor rilzabrutinib is a promising novel anti-B cell treatment [113]. IVIg and anti-D are thought to inhibit antibody-mediated platelet clearance by saturating the FcR on macrophages [11,114]. Various combination therapies targeting this pathway have been reported [115,116]. Other novel treatments, such as anti-CD38 monoclonal antibodies to direct against plasma cells and neonatal Fc receptor inhibitors to prevent anti-platelet antibody recycling, are currently in clinical trials [117].
The dysregulation of the immune system is multifactorial, and various components of immunity are involved in perpetuating the disease. The underlying pathogenic stimuli of primary ITP often remain unknown [118]. Other mechanisms, such as megakaryocyte injury [119,120,121], reduced proplatelet formation [122,123], and complement activation [124,125,126], have implications in ITP treatment. This includes the current use of TPO RA eltrombopag [127], romiplostim [128,129], and avatrombopag [130]. Regarding complement inhibition, the Phase 1 results of a selective inhibitor sutimlimab, have recently been published: five of the 12 patients who received treatment had a durable platelet count response while on sutimlimab, which seemed to be well tolerated [131].
Oseltamivir phosphate is commonly used to treat influenza A and B with a good safety profile. It is a prodrug that, after absorption, is hydrolysed by hepatic esterases to the active form, oseltamivir carboxylate [132]. It inhibits influenza virus neuraminidase, an enzyme known to cleave the budding viral progeny from its cellular envelop attachment point (neuraminic acid) just prior to release [133]. Humans have four sialidases/neuraminidases that are homologues of viral neuraminidase, and the activity may be reduced by oseltamivir [134].
Interestingly, even before the systemic studies of ITP antibody-induced platelet desialylation, there were case reports of oseltamivir improving platelet counts in patients with ITP [33,135,136,137]. The first report, published in 2010, described a 13-year-old patient with chronic ITP who was hospitalised and treated with oseltamivir for H1N1 infection. The platelet count increased unexpectedly from 32 × 109/L to over 500 × 109/L [135]. In the second case published in 2015, a 69-year-old female with chronic ITP with anti-GPIb-IX antibodies was successfully managed with oseltamivir [33]. The third case described a 46-year-old female with secondary ITP from HIV infection. She was given oseltamivir for a presumed influenza infection. This was followed by normalisation of platelet count to 213 × 109/L from a pre-treatment level of 9 × 109/L [136]. The fourth case also reported a 51-year-old man with HIV infection on antiviral treatment and refractory ITP. His platelet count responded to combination therapy with romiplostim and oseltamivir [137]. Surprisingly, case number four also demonstrated platelet function recovery upon oseltamivir treatment using PAC1 and P-Selectin binding [137]. This is congruent with a later publication by our research group [36]. We described a patient with chronic ITP and acquired Glanzmann thrombasthenia. Not only did this patient’s antibodies inhibit platelet function, but they also led to neuraminidase translocation to the platelet surface and desialylation [36]. These findings indicate that certain anti-platelet antibodies potentially cause both platelet desialylation and platelet function inhibition.
Since the availability of more robust scientific data, systematic studies have been conducted in patients with ITP. Revilla and co-workers studied 35 patients with ITP and selected 10 of these patients with evidence of platelet desialylation for oseltamivir treatment [34]. Unlike the animal data, oseltamivir monotherapy was ineffective as none of the patients on oseltamivir-only treatment (n = 4) responded. However, when oseltamivir was given in combination with TPO-RA and/or immunosuppressive therapy, four out of the six patients attained a response. In contrast to the experimental data from Marini et al. [37] and our group [38], all four responders had sole anti-GPIbα antibodies [34]. No solid conclusions can be drawn from this study due to the low number of patients with single antibody specificity.
In 2021, the results of a multicentre, randomised, open-label trial using oseltamivir in combination with dexamethasone in newly diagnosed adult ITP were published [138]. The authors reported significantly improved initial response rate at day 14 in the oseltamivir group, defined as platelet count between 30 and 100 × 109/L without clinically significant bleeding. The 6-month response rate was also higher in this group. However, the 12-month sustained response is not significantly different. In addition, although the duration of response was 9 months in the oseltamivir treatment group and 5 months in the control, this did not reach statistical significance. Such results may be due to the lack of power in the study. Patients with detectable anti-GPIb-IX antibodies had a poorer initial response in the dexamethasone group, but no significant difference was noted in the dexamethasone plus oseltamivir arm. The desialylation status was not examined. A sufficiently powered, well-designed, double-blind randomised clinical trial is needed to examine the potential role of oseltamivir in managing patients with newly diagnosed and refractory ITP. Its potential role in paediatric ITP and its use as a single therapy to minimise immunosuppressive and other steroid side effects are yet to be explored in clinical trials.

5. Conclusions

Since the initial use of splenectomy as the only treatment for ITP in the early 20th century [139], significant progress has been made in understanding the pathogenesis of ITP in recent years. This includes advances into the intricate interplay between B and T cells, as well as in the understanding of platelet antigen and autoantibody interaction. This multifaceted aspect of ITP pathology is connected to one of the most significant challenges in caring for patients with ITP: the lack of treatment individualisation. Clearly defining the platelet clearance pathway, such as desialylation and platelet apoptosis, will have important clinical implications. Patients with different autoantibodies could potentially be directed towards specific therapies. In this regard, platelet antibody testing and measuring platelet desialylation/apoptosis levels could prove clinically helpful in the treatment of ITP. Hence, the characterisation of novel mechanisms will not only contribute to the understanding of ITP’s pathogenesis but also potentially guide the development of new diagnostic tests and targeted therapies. Continued dedication to the exploration of personalised treatment should be a priority. Specifically, collaborative, high-powered studies are required to investigate treatments using neuraminidase and/or apoptosis inhibitors in randomised prospective, multicentre ITP clinical trials.

Author Contributions

S.S.Z. drafted the manuscript. J.S.P. reviewed and drafted the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

SSZ received the NSW Ministry of Health Ph.D. Scholarship.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were generated in this review.

Acknowledgments

This work is part of the Doctoral Thesis of S.S.Z.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Cines, D.B.; Bussel, J.B.; Liebman, H.A.; Luning Prak, E.T. The ITP syndrome: Pathogenic and clinical diversity. Blood 2009, 113, 6511–6521. [Google Scholar] [CrossRef] [PubMed]
  2. Chong, B.H.; Choi, P.Y.; Khachigian, L.; Perdomo, J. Drug-induced immune thrombocytopenia. Hematol. Oncol. Clin. N. Am. 2013, 27, 521–540. [Google Scholar] [CrossRef]
  3. Provan, D.; Stasi, R.; Newland, A.C.; Blanchette, V.S.; Bolton-Maggs, P.; Bussel, J.B.; Chong, B.H.; Cines, D.B.; Gernsheimer, T.B.; Godeau, B.; et al. International consensus report on the investigation and management of primary immune thrombocytopenia. Blood 2010, 115, 168–186. [Google Scholar] [CrossRef]
  4. Lambert, M.P.; Gernsheimer, T.B. Clinical updates in adult immune thrombocytopenia. Blood 2017, 129, 2829–2835. [Google Scholar] [CrossRef]
  5. Verissimo, V.; Carter, T.; Wright, H.; Robertson, J.; Osborn, M.; Bradbeer, P.; Sabesan, V.; Saxon, B.; Barbaro, P.; Crighton, G.; et al. Australian and New Zealand consensus guideline for paediatric newly diagnosed immune thrombocytopaenia endorsed by Australian New Zealand Children’s Haematology and Oncology Group. J. Paediatr. Child Health 2023, 59, 711–717. [Google Scholar] [CrossRef] [PubMed]
  6. Harrington, W.J.; Minnich, V.; Hollingsworth, J.W.; Moore, C.V. Demonstration of a thrombocytopenic factor in the blood of patients with thrombocytopenic purpura. J. Lab. Clin. Med. 1951, 38, 1–10. [Google Scholar]
  7. Schwartz, R.S. Immune thrombocytopenic purpura—From agony to agonist. N. Engl. J. Med. 2007, 357, 2299–2301. [Google Scholar] [CrossRef] [PubMed]
  8. Shulman, N.R.; Marder, V.J.; Weinrach, R.S. Similarities between known antiplatelet antibodies and the factor responsible for thrombocytopenia in idiopathic purpura. Physiologic, serologic and isotopic studies. Ann. N. Y. Acad. Sci. 1965, 124, 499–542. [Google Scholar] [CrossRef] [PubMed]
  9. Hollenhorst, M.A.; Al-Samkari, H.; Kuter, D.J. Markers of autoimmunity in immune thrombocytopenia: Prevalence and prognostic significance. Blood Adv. 2019, 3, 3515–3521. [Google Scholar] [CrossRef]
  10. Grozovsky, R.; Giannini, S.; Falet, H.; Hoffmeister, K.M. Regulating billions of blood platelets: Glycans and beyond. Blood 2015, 126, 1877–1884. [Google Scholar] [CrossRef]
  11. Cines, D.B.; Cuker, A.; Semple, J.W. Pathogenesis of immune thrombocytopenia. Presse Med. 2014, 43, e49–e59. [Google Scholar] [CrossRef] [PubMed]
  12. Broudy, V.C.; Lin, N.L.; Kaushansky, K. Thrombopoietin (c-mpl ligand) acts synergistically with erythropoietin, stem cell factor, and interleukin-11 to enhance murine megakaryocyte colony growth and increases megakaryocyte ploidy in vitro. Blood 1995, 85, 1719–1726. [Google Scholar] [CrossRef] [PubMed]
  13. Debili, N.; Wendling, F.; Katz, A.; Guichard, J.; Breton-Gorius, J.; Hunt, P.; Vainchenker, W. The Mpl-ligand or thrombopoietin or megakaryocyte growth and differentiative factor has both direct proliferative and differentiative activities on human megakaryocyte progenitors. Blood 1995, 86, 2516–2525. [Google Scholar] [CrossRef] [PubMed]
  14. Broudy, V.C.; Lin, N.L.; Sabath, D.F.; Papayannopoulou, T.; Kaushansky, K. Human platelets display high-affinity receptors for thrombopoietin. Blood 1997, 89, 1896–1904. [Google Scholar] [CrossRef]
  15. Sungaran, R.; Markovic, B.; Chong, B.H. Localization and regulation of thrombopoietin mRNa expression in human kidney, liver, bone marrow, and spleen using in situ hybridization. Blood 1997, 89, 101–107. [Google Scholar] [CrossRef] [PubMed]
  16. Li, J.; Xia, Y.; Kuter, D.J. Interaction of thrombopoietin with the platelet c-mpl receptor in plasma: Binding, internalization, stability and pharmacokinetics. Br. J. Haematol. 1999, 106, 345–356. [Google Scholar] [CrossRef]
  17. Deutsch, V.R.; Tomer, A. Megakaryocyte development and platelet production. Br. J. Haematol. 2006, 134, 453–466. [Google Scholar] [CrossRef]
  18. Kuter, D.J.; Rosenberg, R.D. The reciprocal relationship of thrombopoietin (c-Mpl ligand) to changes in the platelet mass during busulfan-induced thrombocytopenia in the rabbit. Blood 1995, 85, 2720–2730. [Google Scholar] [CrossRef] [PubMed]
  19. Kuter, D.J.; Gernsheimer, T.B. Thrombopoietin and platelet production in chronic immune thrombocytopenia. Hematol. Oncol. Clin. N. Am. 2009, 23, 1193–1211. [Google Scholar] [CrossRef]
  20. Grozovsky, R.; Begonja, A.J.; Liu, K.; Visner, G.; Hartwig, J.H.; Falet, H.; Hoffmeister, K.M. The Ashwell-Morell receptor regulates hepatic thrombopoietin production via JAK2-STAT3 signaling. Nat. Med. 2015, 21, 47–54. [Google Scholar] [CrossRef]
  21. Berndt, M.C.; Gregory, C.; Kabral, A.; Zola, H.; Fournier, D.; Castaldi, P.A. Purification and preliminary characterization of the glycoprotein Ib complex in the human platelet membrane. Eur. J. Biochem. 1985, 151, 637–649. [Google Scholar] [CrossRef] [PubMed]
  22. Zhang, W.; Deng, W.; Zhou, L.; Xu, Y.; Yang, W.; Liang, X.; Wang, Y.; Kulman, J.D.; Zhang, X.F.; Li, R. Identification of a juxtamembrane mechanosensitive domain in the platelet mechanosensor glycoprotein Ib-IX complex. Blood 2015, 125, 562–569. [Google Scholar] [CrossRef] [PubMed]
  23. Greenberg, J.; Packham, M.A.; Cazenave, J.P.; Reimers, H.J.; Mustard, J.F. Effects on platelet function of removal of platelet sialic acid by neuraminidase. Lab. Investig. 1975, 32, 476–484. [Google Scholar] [PubMed]
  24. Eto, K.; Kunishima, S. Linkage between the mechanisms of thrombocytopenia and thrombopoiesis. Blood 2016, 127, 1234–1241. [Google Scholar] [CrossRef]
  25. Judson, P.A.; Anstee, D.J.; Clamp, J.R. Isolation and characterization of the major oligosaccharide of human platelet membrane glycoprotein GPIb. Biochem. J. 1982, 205, 81–90. [Google Scholar] [CrossRef]
  26. Madoff, M.A.; Ebbe, S.; Baldini, M. Sialic acid of human blood platelets. J. Clin. Investig. 1964, 43, 870–877. [Google Scholar] [CrossRef]
  27. Karpatkin, S.; Shulman, S. Asialo platelets enhance thrombopoiesis. Trans. Assoc. Am. Physicians 1980, 93, 244–250. [Google Scholar]
  28. Kotzé, H.F.; van Wyk, V.; Badenhorst, P.N.; Heyns, A.D.; Roodt, J.P.; Lötter, M.G. Influence of platelet membrane sialic acid and platelet-associated IgG on ageing and sequestration of blood platelets in baboons. Thromb. Haemost. 1993, 70, 676–680. [Google Scholar] [CrossRef]
  29. Sorensen, A.L.; Rumjantseva, V.; Nayeb-Hashemi, S.; Clausen, H.; Hartwig, J.H.; Wandall, H.H.; Hoffmeister, K.M. 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, 1645–1654. [Google Scholar] [CrossRef]
  30. Jansen, A.J.; Josefsson, E.C.; Rumjantseva, V.; Liu, Q.P.; Falet, H.; Bergmeier, W.; Cifuni, S.M.; Sackstein, R.; von Andrian, U.H.; Wagner, D.D.; et al. Desialylation accelerates platelet clearance after refrigeration and initiates GPIbalpha metalloproteinase-mediated cleavage in mice. Blood 2012, 119, 1263–1273. [Google Scholar] [CrossRef]
  31. Li, J.; Callum, J.L.; Lin, Y.; Zhou, Y.; Zhu, G.; Ni, H. Severe platelet desialylation in a patient with glycoprotein Ib/IX antibody-mediated immune thrombocytopenia and fatal pulmonary hemorrhage. Haematologica 2014, 99, e61–e63. [Google Scholar] [CrossRef] [PubMed]
  32. Li, J.; van der Wal, D.E.; Zhu, G.; Xu, M.; Yougbare, I.; Ma, L.; Vadasz, B.; Carrim, N.; Grozovsky, R.; Ruan, M.; et al. Desialylation is a mechanism of Fc-independent platelet clearance and a therapeutic target in immune thrombocytopenia. Nat. Commun. 2015, 6, 7737. [Google Scholar] [CrossRef] [PubMed]
  33. Shao, L.; Wu, Y.; Zhou, H.; Qin, P.; Ni, H.; Peng, J.; Hou, M. Successful treatment with oseltamivir phosphate in a patient with chronic immune thrombocytopenia positive for anti-GPIb/IX autoantibody. Platelets 2015, 26, 495–497. [Google Scholar] [CrossRef]
  34. Revilla, N.; Corral, J.; Miñano, A.; Mingot-Castellano, M.E.; Campos, R.M.; Velasco, F.; Gonzalez, N.; Galvez, E.; Berrueco, R.; Fuentes, I.; et al. Multirefractory primary immune thrombocytopenia; targeting the decreased sialic acid content. Platelets 2019, 30, 743–751. [Google Scholar] [CrossRef] [PubMed]
  35. Grodzielski, M.; Goette, N.P.; Glembotsky, A.C.; Constanza Baroni Pietto, M.; Mendez-Huergo, S.P.; Pierdominici, M.S.; Montero, V.S.; Rabinovich, G.A.; Molinas, F.C.; Heller, P.G.; et al. Multiple concomitant mechanisms contribute to low platelet count in patients with immune thrombocytopenia. Sci. Rep. 2019, 9, 2208. [Google Scholar] [CrossRef] [PubMed]
  36. Zheng, S.S.; Perdomo, J.S.; Leung, H.H.L.; Yan, F.; Chong, B.H. Acquired Glanzmann thrombasthenia associated with platelet desialylation. J. Thromb. Haemost. 2020, 18, 714–721. [Google Scholar] [CrossRef] [PubMed]
  37. Marini, I.; Zlamal, J.; Faul, C.; Holzer, U.; Hammer, S.; Pelzl, L.; Bethge, W.; Althaus, K.; Bakchoul, T. Autoantibody-mediated desialylation impairs human thrombopoiesis and platelet lifespan. Haematologica 2021, 106, 196–207. [Google Scholar] [CrossRef]
  38. Zheng, S.S.; Ahmadi, Z.; Leung, H.H.L.; Wong, R.; Yan, F.; Perdomo, J.S.; Chong, B.H. Antiplatelet antibody predicts platelet desialylation and apoptosis in immune thrombocytopenia. Haematologica 2022, 107, 2195–2205. [Google Scholar] [CrossRef]
  39. Leeksma, C.H.W.; Cohen, J.A. Determination of the Life of Human Blood Platelets using Labelled Diisopropylfluorophosphonate. Nature 1955, 175, 552–553. [Google Scholar] [CrossRef]
  40. Josefsson, E.C.; Dowling, M.R.; Lebois, M.; Kile, B.T. Chapter 3—The Regulation of Platelet Life Span. In Platelets, 3rd ed.; Michelson, A.D., Ed.; Academic Press: Cambridge, MA, USA, 2013; pp. 51–65. [Google Scholar]
  41. Kile, B.T. The role of apoptosis in megakaryocytes and platelets. Br. J. Haematol. 2014, 165, 217–226. [Google Scholar] [CrossRef]
  42. Mason, K.D.; Carpinelli, M.R.; Fletcher, J.I.; Collinge, J.E.; Hilton, A.A.; Ellis, S.; Kelly, P.N.; Ekert, P.G.; Metcalf, D.; Roberts, A.W.; et al. Programmed anuclear cell death delimits platelet life span. Cell 2007, 128, 1173–1186. [Google Scholar] [CrossRef] [PubMed]
  43. McArthur, K.; Chappaz, S.; Kile, B.T. Apoptosis in megakaryocytes and platelets: The life and death of a lineage. Blood 2018, 131, 605–610. [Google Scholar] [CrossRef] [PubMed]
  44. Zhang, H.; Nimmer, P.M.; Tahir, S.K.; Chen, J.; Fryer, R.M.; Hahn, K.R.; Iciek, L.A.; Morgan, S.J.; Nasarre, M.C.; Nelson, R.; et al. Bcl-2 family proteins are essential for platelet survival. Cell Death Differ. 2007, 14, 943–951. [Google Scholar] [CrossRef] [PubMed]
  45. Gardino, A.K.; Yaffe, M.B. 14-3-3 proteins as signaling integration points for cell cycle control and apoptosis. Semin. Cell Dev. Biol. 2011, 22, 688–695. [Google Scholar] [CrossRef]
  46. Grozovsky, R.; Fraser, C.; Hoffmeister, K.M.; Sarosiek, K.; Giannini, S. Desialylation and Apoptosis Crosstalk to Modulate Platelet Clearance. Blood 2019, 134, 1055. [Google Scholar] [CrossRef]
  47. Jacobson, M.D.; Burne, J.F.; Raff, M.C. Programmed cell death and Bcl-2 protection in the absence of a nucleus. EMBO J. 1994, 13, 1899–1910. [Google Scholar] [CrossRef]
  48. Wolf, B.B.; Goldstein, J.C.; Stennicke, H.R.; Beere, H.; Amarante-Mendes, G.P.; Salvesen, G.S.; Green, D.R. Calpain functions in a caspase-independent manner to promote apoptosis-like events during platelet activation. Blood 1999, 94, 1683–1692. [Google Scholar] [CrossRef]
  49. Piguet, P.F.; Vesin, C. Modulation of platelet caspases and life-span by anti-platelet antibodies in mice. Eur. J. Haematol. 2002, 68, 253–261. [Google Scholar] [CrossRef] [PubMed]
  50. Rand, M.L.; Wang, H.; Bang, K.W.A.; Poon, K.S.V.; Packham, M.A.; Freedman, J. Procoagulant surface exposure and apoptosis in rabbit platelets: Association with shortened survival and steady-state senescence. J. Thromb. Haemost. 2004, 2, 651–659. [Google Scholar] [CrossRef]
  51. Leytin, V.; Mykhaylov, S.; Starkey, A.F.; Allen, D.J.; Lau, H.; Ni, H.; Semple, J.W.; Lazarus, A.H.; Freedman, J. Intravenous immunoglobulin inhibits anti-glycoprotein IIb-induced platelet apoptosis in a murine model of immune thrombocytopenia. Br. J. Haematol. 2006, 133, 78–82. [Google Scholar] [CrossRef]
  52. Leytin, V.; Allen, D.J.; Mykhaylov, S.; Lyubimov, E.; Freedman, J. Thrombin-triggered platelet apoptosis. J. Thromb. Haemost. 2006, 4, 2656–2663. [Google Scholar] [CrossRef] [PubMed]
  53. Lopez, J.J.; Salido, G.M.; Gomez-Arteta, E.; Rosado, J.A.; Pariente, J.A. Thrombin induces apoptotic events through the generation of reactive oxygen species in human platelets. J. Thromb. Haemost. 2007, 5, 1283–1291. [Google Scholar] [CrossRef]
  54. van der Wal, D.E.; Du, V.X.; Lo, K.S.; Rasmussen, J.T.; Verhoef, S.; Akkerman, J.W. Platelet apoptosis by cold-induced glycoprotein Ibalpha clustering. J. Thromb. Haemost. 2010, 8, 2554–2562. [Google Scholar] [CrossRef] [PubMed]
  55. Winkler, J.; Kroiss, S.; Rand, M.L.; Azzouzi, I.; Annie Bang, K.W.; Speer, O.; Schmugge, M. Platelet apoptosis in paediatric immune thrombocytopenia is ameliorated by intravenous immunoglobulin. Br. J. Haematol. 2012, 156, 508–515. [Google Scholar] [CrossRef]
  56. Goette, N.P.; Glembotsky, A.C.; Lev, P.R.; Grodzielski, M.; Contrufo, G.; Pierdominici, M.S.; Espasandin, Y.R.; Riveros, D.; García, A.J.; Molinas, F.C.; et al. Platelet Apoptosis in Adult Immune Thrombocytopenia: Insights into the Mechanism of Damage Triggered by Auto-Antibodies. PLoS ONE 2016, 11, e0160563. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, M.; Yan, R.; Zhou, K.; Li, X.; Zhang, Y.; Liu, C.; Jiang, M.; Ye, H.; Meng, X.; Pang, N.; et al. Akt-mediated platelet apoptosis and its therapeutic implications in immune thrombocytopenia. Proc. Natl. Acad. Sci. USA 2018, 115, E10682-e10691. [Google Scholar] [CrossRef]
  58. Cohen, Y.C.; Djulbegovic, B.; Shamai-Lubovitz, O.; Mozes, B. The bleeding risk and natural history of idiopathic thrombocytopenic purpura in patients with persistent low platelet counts. Arch. Intern. Med. 2000, 160, 1630–1638. [Google Scholar] [CrossRef]
  59. Deckmyn, H.; De Reys, S. Functional effects of human antiplatelet antibodies. Semin. Thromb. Hemost. 1995, 21, 46–59. [Google Scholar] [CrossRef]
  60. Doobaree, I.U.; Nandigam, R.; Bennett, D.; Newland, A.; Provan, D. Thromboembolism in adults with primary immune thrombocytopenia: A systematic literature review and meta-analysis. Eur. J. Haematol. 2016, 97, 321–330. [Google Scholar] [CrossRef]
  61. Hill, Q.A.; Newland, A.C. Fatigue in immune thrombocytopenia. Br. J. Haematol. 2015, 170, 141–149. [Google Scholar] [CrossRef]
  62. Portielje, J.E.; Westendorp, R.G.; Kluin-Nelemans, H.C.; Brand, A. Morbidity and mortality in adults with idiopathic thrombocytopenic purpura. Blood 2001, 97, 2549–2554. [Google Scholar] [CrossRef] [PubMed]
  63. Frederiksen, H.; Maegbaek, M.L.; Norgaard, M. Twenty-year mortality of adult patients with primary immune thrombocytopenia: A Danish population-based cohort study. Br. J. Haematol. 2014, 166, 260–267. [Google Scholar] [CrossRef] [PubMed]
  64. Norgaard, M.; Jensen, A.O.; Engebjerg, M.C.; Farkas, D.K.; Thomsen, R.W.; Cha, S.; Zhao, S.; Sorensen, H.T. Long-term clinical outcomes of patients with primary chronic immune thrombocytopenia: A Danish population-based cohort study. Blood 2011, 117, 3514–3520. [Google Scholar] [CrossRef] [PubMed]
  65. Schoonen, W.M.; Kucera, G.; Coalson, J.; Li, L.; Rutstein, M.; Mowat, F.; Fryzek, J.; Kaye, J.A. Epidemiology of immune thrombocytopenic purpura in the General Practice Research Database. Br. J. Haematol. 2009, 145, 235–244. [Google Scholar] [CrossRef] [PubMed]
  66. Schmidt, D.E.; Heitink-Polle, K.M.J.; Porcelijn, L.; van der Schoot, C.E.; Vidarsson, G.; Bruin, M.C.A.; de Haas, M. Anti-platelet antibodies in childhood immune thrombocytopenia: Prevalence and prognostic implications. J. Thromb. Haemost. 2020, 18, 1210–1220. [Google Scholar] [CrossRef]
  67. Brighton, T.A.; Evans, S.; Castaldi, P.A.; Chesterman, C.N.; Chong, B.H. Prospective evaluation of the clinical usefulness of an antigen-specific assay (MAIPA) in idiopathic thrombocytopenic purpura and other immune thrombocytopenias. Blood 1996, 88, 194–201. [Google Scholar] [CrossRef]
  68. Al-Samkari, H.; Rosovsky, R.P.; Karp Leaf, R.S.; Smith, D.B.; Goodarzi, K.; Fogerty, A.E.; Sykes, D.B.; Kuter, D.J. A modern reassessment of glycoprotein-specific direct platelet autoantibody testing in immune thrombocytopenia. Blood Adv. 2019, 4, 9–18. [Google Scholar] [CrossRef]
  69. He, R.; Reid, D.M.; Jones, C.E.; Shulman, N.R. Spectrum of Ig classes, specificities, and titers of serum antiglycoproteins in chronic idiopathic thrombocytopenic purpura. Blood 1994, 83, 1024–1032. [Google Scholar] [CrossRef]
  70. Kiefel, V.; Santoso, S.; Kaufmann, E.; Mueller-Eckhardt, C. Autoantibodies against platelet glycoprotein Ib/IX: A frequent finding in autoimmune thrombocytopenic purpura. Br. J. Haematol. 1991, 79, 256–262. [Google Scholar] [CrossRef]
  71. Vollenberg, R.; Jouni, R.; Norris, P.A.A.; Burg-Roderfeld, M.; Cooper, N.; Rummel, M.J.; Bein, G.; Marini, I.; Bayat, B.; Burack, R.; et al. Glycoprotein V is a relevant immune target in patients with immune thrombocytopenia. Haematologica 2019, 104, 1237–1243. [Google Scholar] [CrossRef]
  72. Garner, S.F.; Campbell, K.; Metcalfe, P.; Keidan, J.; Huiskes, E.; Dong, J.F.; Lopez, J.A.; Ouwehand, W.H. Glycoprotein V: The predominant target antigen in gold-induced autoimmune thrombocytopenia. Blood 2002, 100, 344–346. [Google Scholar] [CrossRef] [PubMed]
  73. Porcelijn, L.; Huiskes, E.; Oldert, G.; Schipperus, M.; Zwaginga, J.J.; de Haas, M. Detection of platelet autoantibodies to identify immune thrombocytopenia: State of the art. Br. J. Haematol. 2018, 182, 423–426. [Google Scholar] [CrossRef] [PubMed]
  74. Johnsen, J. Pathogenesis in immune thrombocytopenia: New insights. Hematol. Am. Soc. Hematol. Educ. Program 2012, 2012, 306–312. [Google Scholar] [CrossRef]
  75. Stasi, R.; Cooper, N.; Del Poeta, G.; Stipa, E.; Laura Evangelista, M.; Abruzzese, E.; Amadori, S. Analysis of regulatory T-cell changes in patients with idiopathic thrombocytopenic purpura receiving B cell-depleting therapy with rituximab. Blood 2008, 112, 1147–1150. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, B.; Zhao, H.; Poon, M.C.; Han, Z.; Gu, D.; Xu, M.; Jia, H.; Yang, R.; Han, Z.C. Abnormality of CD4+CD25+ regulatory T cells in idiopathic thrombocytopenic purpura. Eur. J. Haematol. 2007, 78, 139–143. [Google Scholar] [CrossRef]
  77. Ling, Y.; Cao, X.; Yu, Z.; Ruan, C. Circulating dendritic cells subsets and CD4+Foxp3+ regulatory T cells in adult patients with chronic ITP before and after treatment with high-dose dexamethasome. Eur. J. Haematol. 2007, 79, 310–316. [Google Scholar] [CrossRef]
  78. Zhang, J.; Ma, D.; Zhu, X.; Qu, X.; Ji, C.; Hou, M. Elevated profile of Th17, Th1 and Tc1 cells in patients with immune thrombocytopenic purpura. Haematologica 2009, 94, 1326–1329. [Google Scholar] [CrossRef]
  79. Stasi, R.; Del Poeta, G.; Stipa, E.; Evangelista, M.L.; Trawinska, M.M.; Cooper, N.; Amadori, S. Response to B-cell-depleting therapy with rituximab reverts the abnormalities of T-cell subsets in patients with idiopathic thrombocytopenic purpura. Blood 2007, 110, 2924–2930. [Google Scholar] [CrossRef]
  80. Zhang, H.; Zhang, B.M.; Guo, X.; Xu, L.; You, X.; West, R.B.; Bussel, J.B.; Zehnder, J.L. Blood transcriptome and clonal T-cell correlates of response and non-response to eltrombopag therapy in a cohort of patients with chronic immune thrombocytopenia. Haematologica 2020, 105, e129–e132. [Google Scholar] [CrossRef]
  81. Fogarty, P.F.; Rick, M.E.; Zeng, W.; Risitano, A.M.; Dunbar, C.E.; Bussel, J.B. T cell receptor VB repertoire diversity in patients with immune thrombocytopenia following splenectomy. Clin. Exp. Immunol. 2003, 133, 461–466. [Google Scholar] [CrossRef]
  82. Nieswandt, B.; Bergmeier, W.; Rackebrandt, K.; Gessner, J.E.; Zirngibl, H. Identification of critical antigen-specific mechanisms in the development of immune thrombocytopenic purpura in mice. Blood 2000, 96, 2520–2527. [Google Scholar] [CrossRef] [PubMed]
  83. Hoffmeister, K.M.; Felbinger, T.W.; Falet, H.; Denis, C.V.; Bergmeier, W.; Mayadas, T.N.; von Andrian, U.H.; Wagner, D.D.; Stossel, T.P.; Hartwig, J.H. The clearance mechanism of chilled blood platelets. Cell 2003, 112, 87–97. [Google Scholar] [CrossRef] [PubMed]
  84. Hoffmeister, K.M.; Josefsson, E.C.; Isaac, N.A.; Clausen, H.; Hartwig, J.H.; Stossel, T.P. Glycosylation restores survival of chilled blood platelets. Science 2003, 301, 1531–1534. [Google Scholar] [CrossRef]
  85. Rumjantseva, V.; Grewal, P.K.; Wandall, H.H.; Josefsson, E.C.; Sorensen, A.L.; Larson, G.; Marth, J.D.; Hartwig, J.H.; Hoffmeister, K.M. Dual roles for hepatic lectin receptors in the clearance of chilled platelets. Nat. Med. 2009, 15, 1273–1280. [Google Scholar] [CrossRef]
  86. Qiao, J.; Al-Tamimi, M.; Baker, R.I.; Andrews, R.K.; Gardiner, E.E. The platelet Fc receptor, FcgammaRIIa. Immunol. Rev. 2015, 268, 241–252. [Google Scholar] [CrossRef] [PubMed]
  87. Chan, H.; Moore, J.C.; Finch, C.N.; Warkentin, T.E.; Kelton, J.G. The IgG subclasses of platelet-associated autoantibodies directed against platelet glycoproteins IIb/IIIa in patients with idiopathic thrombocytopenic purpura. Br. J. Haematol. 2003, 122, 818–824. [Google Scholar] [CrossRef]
  88. Cantoni, S.; Carpenedo, M.; Nichelatti, M.; Sica, L.; Rossini, S.; Milella, M.; Popescu, C.; Cairoli, R. Clinical relevance of antiplatelet antibodies and the hepatic clearance of platelets in patients with immune thrombocytopenia. Blood 2016, 128, 2183–2185. [Google Scholar] [CrossRef]
  89. Amini, S.N.; Nelson, V.S.; Porcelijn, L.; Netelenbos, T.; Zwaginga, J.J.; de Haas, M.; Schipperus, M.R.; Kapur, R. The interplay between GPIb/IX antibodies, platelet hepatic sequestration, and TPO levels in patients with chronic ITP. Blood Adv. 2023, 7, 1066–1069. [Google Scholar] [CrossRef]
  90. Morodomi, Y.; Kanaji, S.; Won, E.; Ruggeri, Z.M.; Kanaji, T. Mechanisms of anti-GPIbα antibody–induced thrombocytopenia in mice. Blood 2020, 135, 2292–2301. [Google Scholar] [CrossRef]
  91. Qiao, J.; Liu, Y.; Li, D.; Wu, Y.; Li, X.; Yao, Y.; Niu, M.; Fu, C.; Li, H.; Ma, P.; et al. Imbalanced expression of Bcl-xL and Bax in platelets treated with plasma from immune thrombocytopenia. Immunol. Res. 2016, 64, 604–609. [Google Scholar] [CrossRef]
  92. Nassa, G.; Giurato, G.; Cimmino, G.; Rizzo, F.; Ravo, M.; Salvati, A.; Nyman, T.A.; Zhu, Y.; Vesterlund, M.; Lehtiö, J.; et al. Splicing of platelet resident pre-mRNAs upon activation by physiological stimuli results in functionally relevant proteome modifications. Sci. Rep. 2018, 8, 498. [Google Scholar] [CrossRef] [PubMed]
  93. Mills, E.W.; Green, R.; Ingolia, N.T. Slowed decay of mRNAs enhances platelet specific translation. Blood 2017, 129, e38–e48. [Google Scholar] [CrossRef] [PubMed]
  94. Feng, S.; Christodoulides, N.; Resendiz, J.C.; Berndt, M.C.; Kroll, M.H. Cytoplasmic domains of GpIbalpha and GpIbbeta regulate 14-3-3zeta binding to GpIb/IX/V. Blood 2000, 95, 551–557. [Google Scholar] [CrossRef]
  95. George, N.M.; Targy, N.; Evans, J.J.; Zhang, L.; Luo, X. Bax contains two functional mitochondrial targeting sequences and translocates to mitochondria in a conformational change- and homo-oligomerization-driven process. J. Biol. Chem. 2010, 285, 1384–1392. [Google Scholar] [CrossRef]
  96. Li, S.; Wang, Z.; Liao, Y.; Zhang, W.; Shi, Q.; Yan, R.; Ruan, C.; Dai, K. The glycoprotein Ibalpha-von Willebrand factor interaction induces platelet apoptosis. J. Thromb. Haemost. 2010, 8, 341–350. [Google Scholar] [CrossRef] [PubMed]
  97. Yuan, Y.; Zhang, W.; Yan, R.; Liao, Y.; Zhao, L.; Ruan, C.; Du, X.; Dai, K. Identification of a novel 14-3-3zeta binding site within the cytoplasmic domain of platelet glycoprotein Ibalpha that plays a key role in regulating the von Willebrand factor binding function of glycoprotein Ib-IX. Circ. Res. 2009, 105, 1177–1185. [Google Scholar] [CrossRef]
  98. Vrbensky, J.R.; Moore, J.E.; Arnold, D.M.; Smith, J.W.; Kelton, J.G.; Nazy, I. The sensitivity and specificity of platelet autoantibody testing in immune thrombocytopenia: A systematic review and meta-analysis of a diagnostic test. J. Thromb. Haemost. 2019, 17, 787–794. [Google Scholar] [CrossRef]
  99. Olsson, B.; Andersson, P.O.; Jernas, M.; Jacobsson, S.; Carlsson, B.; Carlsson, L.M.; Wadenvik, H. T-cell-mediated cytotoxicity toward platelets in chronic idiopathic thrombocytopenic purpura. Nat. Med. 2003, 9, 1123–1124. [Google Scholar] [CrossRef] [PubMed]
  100. Zhang, F.; Chu, X.; Wang, L.; Zhu, Y.; Li, L.; Ma, D.; Peng, J.; Hou, M. Cell-mediated lysis of autologous platelets in chronic idiopathic thrombocytopenic purpura. Eur. J. Haematol. 2006, 76, 427–431. [Google Scholar] [CrossRef]
  101. Zhao, C.; Li, X.; Zhang, F.; Wang, L.; Peng, J.; Hou, M. Increased cytotoxic T-lymphocyte-mediated cytotoxicity predominant in patients with idiopathic thrombocytopenic purpura without platelet autoantibodies. Haematologica 2008, 93, 1428–1430. [Google Scholar] [CrossRef]
  102. Qiu, J.; Liu, X.; Li, X.; Zhang, X.; Han, P.; Zhou, H.; Shao, L.; Hou, Y.; Min, Y.; Kong, Z.; et al. CD8+ T cells induce platelet clearance in the liver via platelet desialylation in immune thrombocytopenia. Sci. Rep. 2016, 6, 27445. [Google Scholar] [CrossRef] [PubMed]
  103. Malik, A.; Sayed, A.A.; Han, P.; Tan, M.M.H.; Watt, E.; Constantinescu-Bercu, A.; Cocker, A.T.H.; Khoder, A.; Saputil, R.C.; Thorley, E.V.; et al. The role of CD8+ T cell clones in immune thrombocytopenia. Blood 2023, 141, 2417–2429. [Google Scholar] [CrossRef] [PubMed]
  104. Li, S.; Wang, L.; Zhao, C.; Li, L.; Peng, J.; Hou, M. CD8+ T cells suppress autologous megakaryocyte apoptosis in idiopathic thrombocytopenic purpura. Br. J. Haematol. 2007, 139, 605–611. [Google Scholar] [CrossRef] [PubMed]
  105. Vrbensky, J.R.; Nazy, I.; Toltl, L.J.; Ross, C.; Ivetic, N.; Smith, J.W.; Kelton, J.G.; Arnold, D.M. Megakaryocyte apoptosis in immune thrombocytopenia. Platelets 2018, 29, 729–732. [Google Scholar] [CrossRef]
  106. Choi, P.Y.; Merriman, E.; Bennett, A.; Enjeti, A.K.; Tan, C.W.; Goncalves, I.; Hsu, D.; Bird, R. Consensus guidelines for the management of adult immune thrombocytopenia in Australia and New Zealand. Med. J. Aust. 2022, 216, 43–52. [Google Scholar] [CrossRef] [PubMed]
  107. Fujisawa, K.; Tani, P.; Piro, L.; McMillan, R. The effect of therapy on platelet-associated autoantibody in chronic immune thrombocytopenic purpura. Blood 1993, 81, 2872–2877. [Google Scholar] [CrossRef] [PubMed]
  108. Wang, L.; Xu, L.; Hao, H.; Jansen, A.J.G.; Liu, G.; Li, H.; Liu, X.; Zhao, Y.; Peng, J.; Hou, M. First line treatment of adult patients with primary immune thrombocytopenia: A real-world study. Platelets 2020, 31, 55–61. [Google Scholar] [CrossRef] [PubMed]
  109. Cragg, M.S.; Walshe, C.A.; Ivanov, A.O.; Glennie, M.J. The biology of CD20 and its potential as a target for mAb therapy. Curr. Dir. Autoimmun. 2005, 8, 140–174. [Google Scholar] [CrossRef] [PubMed]
  110. Arnold, D.M.; Vrbensky, J.R.; Karim, N.; Smith, J.W.; Liu, Y.; Ivetic, N.; Kelton, J.G.; Nazy, I. The effect of rituximab on anti-platelet autoantibody levels in patients with immune thrombocytopenia. Br. J. Haematol. 2017, 178, 302–307. [Google Scholar] [CrossRef]
  111. Szczepanik, A.B.; Sikorska, A.; Slomkowski, M.; Konopka, L. The use of vinca alkaloids in preparation for splenectomy of corticosteroid refractory chronic immune thrombocytopenic purpura patients. Int. J. Lab. Hematol. 2007, 29, 347–351. [Google Scholar] [CrossRef]
  112. Quiquandon, I.; Fenaux, P.; Caulier, M.T.; Pagniez, D.; Huart, J.J.; Bauters, F. Re-evaluation of the role of azathioprine in the treatment of adult chronic idiopathic thrombocytopenic purpura: A report on 53 cases. Br. J. Haematol. 1990, 74, 223–228. [Google Scholar] [CrossRef] [PubMed]
  113. Kuter, D.J.; Efraim, M.; Mayer, J.; Trněný, M.; McDonald, V.; Bird, R.; Regenbogen, T.; Garg, M.; Kaplan, Z.; Tzvetkov, N.; et al. Rilzabrutinib, an Oral BTK Inhibitor, in Immune Thrombocytopenia. N. Engl. J. Med. 2022, 386, 1421–1431. [Google Scholar] [CrossRef] [PubMed]
  114. Ware, R.E.; Zimmerman, S.A. Anti-D: Mechanisms of action. Semin. Hematol. 1998, 35, 14–22. [Google Scholar] [PubMed]
  115. Bradbury, C.A.; Pell, J.; Hill, Q.; Bagot, C.; Cooper, N.; Ingram, J.; Breheny, K.; Kandiyali, R.; Rayment, R.; Evans, G.; et al. Mycophenolate Mofetil for First-Line Treatment of Immune Thrombocytopenia. N. Engl. J. Med. 2021, 385, 885–895. [Google Scholar] [CrossRef]
  116. Choi, P.Y.; Roncolato, F.; Badoux, X.; Ramanathan, S.; Ho, S.J.; Chong, B.H. A novel triple therapy for ITP using high-dose dexamethasone, low-dose rituximab, and cyclosporine (TT4). Blood 2015, 126, 500–503. [Google Scholar] [CrossRef]
  117. Kuter, D.J. Novel therapies for immune thrombocytopenia. Br. J. Haematol. 2022, 196, 1311–1328. [Google Scholar] [CrossRef]
  118. Semple, J.W.; Provan, D. The immunopathogenesis of immune thrombocytopenia: T cells still take center-stage. Curr. Opin. Hematol. 2012, 19, 357–362. [Google Scholar] [CrossRef]
  119. Dameshek, W.; Miller, E.B. The megakaryocytes in idiopathic thrombocytopenic purpura, a form of hypersplenism. Blood 1946, 1, 27–50. [Google Scholar] [CrossRef]
  120. Stahl, C.P.; Zucker-Franklin, D.; McDonald, T.P. Incomplete antigenic cross-reactivity between platelets and megakaryocytes: Relevance to ITP. Blood 1986, 67, 421–428. [Google Scholar] [CrossRef]
  121. Houwerzijl, E.J.; Blom, N.R.; van der Want, J.J.; Esselink, M.T.; Koornstra, J.J.; Smit, J.W.; Louwes, H.; Vellenga, E.; de Wolf, J.T. Ultrastructural study shows morphologic features of apoptosis and para-apoptosis in megakaryocytes from patients with idiopathic thrombocytopenic purpura. Blood 2004, 103, 500–506. [Google Scholar] [CrossRef]
  122. Iraqi, M.; Perdomo, J.; Yan, F.; Choi, P.Y.; Chong, B.H. Immune thrombocytopenia: Antiplatelet autoantibodies inhibit proplatelet formation by megakaryocytes and impair platelet production in vitro. Haematologica 2015, 100, 623–632. [Google Scholar] [CrossRef] [PubMed]
  123. Lev, P.R.; Grodzielski, M.; Goette, N.P.; Glembotsky, A.C.; Espasandin, Y.R.; Pierdominici, M.S.; Contrufo, G.; Montero, V.S.; Ferrari, L.; Molinas, F.C.; et al. Impaired proplatelet formation in immune thrombocytopenia: A novel mechanism contributing to decreased platelet count. Br. J. Haematol. 2014, 165, 854–864. [Google Scholar] [CrossRef]
  124. Peerschke, E.I.; Andemariam, B.; Yin, W.; Bussel, J.B. Complement activation on platelets correlates with a decrease in circulating immature platelets in patients with immune thrombocytopenic purpura. Br. J. Haematol. 2010, 148, 638–645. [Google Scholar] [CrossRef]
  125. Najaoui, A.; Bakchoul, T.; Stoy, J.; Bein, G.; Rummel, M.J.; Santoso, S.; Sachs, U.J. Autoantibody-mediated complement activation on platelets is a common finding in patients with immune thrombocytopenic purpura (ITP). Eur. J. Haematol. 2012, 88, 167–174. [Google Scholar] [CrossRef]
  126. Peerschke, E.I.; Panicker, S.; Bussel, J. Classical complement pathway activation in immune thrombocytopenia purpura: Inhibition by a novel C1s inhibitor. Br. J. Haematol. 2016, 173, 942–945. [Google Scholar] [CrossRef]
  127. Stasi, R. Eltrombopag for the treatment of idiopathic thrombocytopenic purpura. Expert. Rev. Hematol. 2008, 1, 145–152. [Google Scholar] [CrossRef] [PubMed]
  128. Kuter, D.J. New thrombopoietic growth factors. Blood 2007, 109, 4607–4616. [Google Scholar] [CrossRef]
  129. Cines, D.B.; Gernsheimer, T.; Wasser, J.; Godeau, B.; Provan, D.; Lyons, R.; Altomare, I.; Wang, X.; Lopez, A. Integrated analysis of long-term safety in patients with chronic immune thrombocytopaenia (ITP) treated with the thrombopoietin (TPO) receptor agonist romiplostim. Int. J. Hematol. 2015, 102, 259–270. [Google Scholar] [CrossRef] [PubMed]
  130. Bussel, J.B.; Kuter, D.J.; Aledort, L.M.; Kessler, C.M.; Cuker, A.; Pendergrass, K.B.; Tang, S.; McIntosh, J. A randomized trial of avatrombopag, an investigational thrombopoietin-receptor agonist, in persistent and chronic immune thrombocytopenia. Blood 2014, 123, 3887–3894. [Google Scholar] [CrossRef]
  131. Broome, C.M.; Roth, A.; Kuter, D.J.; Scully, M.; Smith, R.; Wang, J.; Reuter, C.; Hobbs, W.E.; Daak, A.A.A. Safety and Efficacy of Classical Complement Pathway Inhibition with Sutimlimab in Chronic Immune Thrombocytopenia. Blood Adv. 2023, 7, 987–996. [Google Scholar] [CrossRef]
  132. Davies, B.E. Pharmacokinetics of oseltamivir: An oral antiviral for the treatment and prophylaxis of influenza in diverse populations. J. Antimicrob. Chemother. 2010, 65 (Suppl. 2), ii5–ii10. [Google Scholar] [CrossRef] [PubMed]
  133. Moscona, A. Neuraminidase inhibitors for influenza. N. Engl. J. Med. 2005, 353, 1363–1373. [Google Scholar] [CrossRef] [PubMed]
  134. Li, C.Y.; Yu, Q.; Ye, Z.Q.; Sun, Y.; He, Q.; Li, X.M.; Zhang, W.; Luo, J.; Gu, X.; Zheng, X.; et al. A nonsynonymous SNP in human cytosolic sialidase in a small Asian population results in reduced enzyme activity: Potential link with severe adverse reactions to oseltamivir. Cell Res. 2007, 17, 357–362. [Google Scholar] [CrossRef] [PubMed]
  135. Alioglu, B.; Tasar, A.; Ozen, C.; Selver, B.; Dallar, Y. An experience of oseltamivir phosphate (tamiflu) in a pediatric patient with chronic idiopathic thrombocytopenic purpura: A case report. Pathophysiol. Haemost. Thromb. 2010, 37, 55–58. [Google Scholar] [CrossRef] [PubMed]
  136. Bigot, P.; Auffret, M.; Gautier, S.; Weinborn, M.; Ettahar, N.K.; Coupe, P. Unexpected platelets elevation in a patient with idiopathic thrombocytopenia treated with oseltamivir for influenza infection. Fundam. Clin. Pharmacol. 2016, 30, 483–485. [Google Scholar] [CrossRef]
  137. Álvarez-Román, M.T.; Rivas Pollmar, M.I.; Bernardino, J.I.; Lozano, M.L.; Martín-Salces, M.; Fernández-Bello, I.; Jiménez-Yuste, V.; Butta, N.V. Thrombopoietin receptor agonists in conjunction with oseltamivir for immune thrombocytopenia. AIDS 2016, 30, 1141–1142. [Google Scholar] [CrossRef]
  138. Sun, L.; Wang, J.; Shao, L.; Yuan, C.; Zhao, H.; Li, D.; Wang, Z.; Han, P.; Yu, Y.; Xu, M.; et al. Dexamethasone plus oseltamivir versus dexamethasone in treatment-naive primary immune thrombocytopenia: A multicentre, randomised, open-label, phase 2 trial. Lancet Haematol. 2021, 8, e289–e298. [Google Scholar] [CrossRef]
  139. Stasi, R.; Newland, A.C. ITP: A historical perspective. Br. J. Haematol. 2011, 153, 437–450. [Google Scholar] [CrossRef]
Figure 1. Structure of GPIb-IX-V complex. The GPIbα subunits have a heavily O and N-linked glycosylated at the N-terminus and at the mucin-like macroglycopeptide domain [24,25]. vWF, von Willebrand factor.
Figure 1. Structure of GPIb-IX-V complex. The GPIbα subunits have a heavily O and N-linked glycosylated at the N-terminus and at the mucin-like macroglycopeptide domain [24,25]. vWF, von Willebrand factor.
Cimb 46 00709 g001
Figure 2. Mechanism of TPO regulation in the liver. Aged platelets lose their terminal sialic acid residues. The desialylated platelets are recognised by AMR and removed by hepatocytes, which activates the JAK2/STAT3 pathway and results in increased TPO mRNA and protein production.
Figure 2. Mechanism of TPO regulation in the liver. Aged platelets lose their terminal sialic acid residues. The desialylated platelets are recognised by AMR and removed by hepatocytes, which activates the JAK2/STAT3 pathway and results in increased TPO mRNA and protein production.
Cimb 46 00709 g002
Figure 3. Regulation of platelet lifespan via the intrinsic apoptosis pathway. BCL2 pro-survival protein family restrains BAK and BAX activities by maintaining phosphorylation (p). Platelet senescence resulted in BCL-2 degradation, BAK and BAX dephosphorylation, which causes mitochondrial outer membrane permeabilization and initiates the apoptotic cascade.
Figure 3. Regulation of platelet lifespan via the intrinsic apoptosis pathway. BCL2 pro-survival protein family restrains BAK and BAX activities by maintaining phosphorylation (p). Platelet senescence resulted in BCL-2 degradation, BAK and BAX dephosphorylation, which causes mitochondrial outer membrane permeabilization and initiates the apoptotic cascade.
Cimb 46 00709 g003
Figure 4. Treatments targeting the Fc-dependent pathway. BTK, Bruton kinase; FcRn, neonatal Fc receptor; IVIg, intravenous immunoglobulin.
Figure 4. Treatments targeting the Fc-dependent pathway. BTK, Bruton kinase; FcRn, neonatal Fc receptor; IVIg, intravenous immunoglobulin.
Cimb 46 00709 g004
Table 1. Emergence of desialylation in platelet biology.
Table 1. Emergence of desialylation in platelet biology.
CommentReference; Year
Sialic acid is present on platelets; can be cleaved with neuraminidase[26]; 1964
Desialylated rabbit platelets are rapidly cleared in vivo; no significant changes to platelet function in vitro[23]; 1975
Injection of desialylated platelets induced platelet production. Authors suggested that thrombopoiesis may be regulated by uptake of desialylated platelets[27]; 1980
Sialic acid removal shortens platelet lifespan in primates[28]; 1993
Platelets lacking sialic acid are recognised by asialoglycoprotein receptors[29]; 2009
Cold storage leads to platelet desialylation[30]; 2012
Platelet desialylation by anti GPIb/IX antibody[31]; 2014
Hepatic Ashwell–Morell receptor binds and removes desialylated platelets[20]; 2015
Anti GPIbα, not anti GPIIb/IIIa antibodies, induced desialylation and hepatic platelet uptake in mice[32]; 2015
ITP patient with anti GPIb/IX antibodies successfully treated with oseltamivir[33]; 2015
TPO-RAs in combination with oseltamivir induced sustained platelet production in patients with anti GPIb antibodies[34]; 2019
Plasma from patients with ITP affected the sialylation pattern of control platelets[35]; 2019
Desialylation induced by anti GPIIb/IIIa antibodies and is FcγRIIa-dependent[36,37]; 2020, 2021
Destruction of human platelets induced by anti-GPIIb/IIIa antibodies was prevented with oseltamivir in a humanised mouse model of ITP[37,38]; 2021, 2022
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zheng, S.S.; Perdomo, J.S. Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment. Curr. Issues Mol. Biol. 2024, 46, 11942-11956. https://doi.org/10.3390/cimb46110709

AMA Style

Zheng SS, Perdomo JS. Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment. Current Issues in Molecular Biology. 2024; 46(11):11942-11956. https://doi.org/10.3390/cimb46110709

Chicago/Turabian Style

Zheng, Shiying Silvia, and José Sail Perdomo. 2024. "Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment" Current Issues in Molecular Biology 46, no. 11: 11942-11956. https://doi.org/10.3390/cimb46110709

APA Style

Zheng, S. S., & Perdomo, J. S. (2024). Desialylation and Apoptosis in Immune Thrombocytopenia: Implications for Pathogenesis and Treatment. Current Issues in Molecular Biology, 46(11), 11942-11956. https://doi.org/10.3390/cimb46110709

Article Metrics

Back to TopTop