Hostname: page-component-78c5997874-4rdpn Total loading time: 0 Render date: 2024-10-28T05:16:09.381Z Has data issue: false hasContentIssue false

Thrombocytopenia in dengue infection: mechanisms and a potential application

Published online by Cambridge University Press:  14 October 2024

Ahmad Suhail Khazali
Affiliation:
Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM) Cawangan Perlis, Arau, Perlis, Malaysia
Waqiyuddin Hilmi Hadrawi
Affiliation:
Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
Fatimah Ibrahim
Affiliation:
Department of Biomedical Engineering, Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia Center for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
Shatrah Othman
Affiliation:
Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
Nurshamimi Nor Rashid*
Affiliation:
Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia Center for Innovation in Medical Engineering (CIME), Faculty of Engineering, Universiti Malaya, Kuala Lumpur, Malaysia
*
Corresponding author: Nurshamimi Nor Rashid; Email: [email protected]
Rights & Permissions [Opens in a new window]

Abstract

Thrombocytopenia is a common symptom and one of the warning signs of dengue virus (DENV) infection. Platelet depletion is critical as it may lead to other severe dengue symptoms. Understanding the molecular events of this condition during dengue infection is challenging because of the multifaceted factors involved in DENV infection and the dynamics of the disease progression. Platelet levels depend on the balance between platelet production and platelet consumption or clearance. Megakaryopoiesis and thrombopoiesis, two interdependent processes in platelet production, are hampered during dengue infection. Conversely, platelet elimination via platelet activation, apoptosis and clearance processes are elevated. Together, these anomalies contribute to thrombocytopenia in dengue patients. Targeting the molecular events of dengue-mediated thrombocytopenia shows great potential but still requires further investigation. Nonetheless, the application of new knowledge in this field, such as immature platelet fraction analysis, may facilitate physicians in monitoring the progression of the disease.

Type
Review
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2024. Published by Cambridge University Press

Introduction

Dengue is a mosquito-borne disease common in tropical and subtropical countries. Although dengue infection is usually non-life threatening, progression into the severe form of the disease could be fatal because of serious complications associated with severe dengue. In 2009, the World Health Organization (WHO) revised the characterization of dengue infection into two main categories: non-severe dengue and severe dengue (Ref. 1). The non-severe dengue is further divided into two subcategories: dengue without warning signs and dengue with warning signs. This revised characterization aims to facilitate clinicians in diagnosing patients with warning signs and severe dengue. The symptoms associated with these categories are summarized in Figure 1. Owing to the lack of antiviral drugs or vaccines for dengue, current treatments aim to alleviate the symptoms (Refs Reference Jasamai2, Reference Obi3). Therefore, clinicians must constantly monitor disease progression and apply appropriate treatment based on the patient's condition according to the guidelines provided by the WHO. Increased haematocrit concurrent with a rapid decline in platelet count, or thrombocytopenia, is one of the warning signs in dengue infection (Fig. 1). The level of thrombocytopenia may correlate with the severity of the disease (Refs Reference Faridah4, Reference Mourao5), and severe thrombocytopenia usually precedes the onset of the critical phase of the disease (Ref. Reference van de Weg6). Additionally, severe thrombocytopenia in dengue patients could be the precursor to other severe dengue complications, such as plasma leakage and bleeding (Refs Reference Mourao5, Reference Makroo7). Dengue virus (DENV) has also been shown to activate platelets and other cells, causing the release of inflammatory cytokines that increase endothelium permeability (Ref. Reference Malavige and Ogg8). Furthermore, DENV impairs platelet function, leading to endothelial dysfunction (Ref. Reference de Azeredo, Monteiro and de-Oliveira Pinto9). However, conflicting reports exist where haemorrhaging was not observed in some patients with severe thrombocytopenia (Ref. Reference Chaudhary10). Alterations in platelet level and activation status may also cause coagulation and fibrinolysis abnormalities, as reported here (Refs Reference de Azeredo, Monteiro and de-Oliveira Pinto9, Reference Adane and Getawa11). These findings signify the direct and indirect effects of platelet dysfunction in dengue. This review will focus on the mechanisms of thrombocytopenia during dengue infection and discuss a potential application based on this knowledge.

Figure 1. Classification of dengue by the WHO. This information is based on 2009 WHO guideline (Ref. 1). The present work focuses on one of the symptoms of dengue with warning signs which is thrombocytopenia.

Thrombocytes

Platelets, or thrombocytes, are colourless cell fragments produced by megakaryocytes through a process called thrombopoiesis. A normal platelet count ranges from 150 000 to 350 000 per microlitre of blood, but because of their small size, they make up just a tiny fraction of the blood volume (Ref. Reference Scott and Fong12). Nonetheless, platelets are metabolically active and contain several functional organelles, including the endoplasmic reticulum, Golgi apparatus and mitochondria (Ref. Reference Yun13). They also possess a wide range of surface receptors, adhesion molecules and granules (Ref. Reference Yadav and Storrie14).

Platelet count is an important medical parameter, where a reduction in platelet count in the blood (thrombocytopenia) can be typically observed in numerous medical conditions such as viral infections, haematologic malignancies, autoimmune disorders and side effects of medications (Ref. Reference Izak and Bussel15). During dengue infection, the platelet count could drop to less than 30 000 per microlitre of blood (Refs Reference Ojha16, Reference Pothapregada, Kamalakannan and Thulasingam17). A recent meta-analysis study found that thrombocytopenia is one of the two factors that could serve as independent predictive markers of severe dengue (Ref. Reference Thach18).

Platelets are vital in blood coagulation to prevent blood loss during vessel injury. They play paramount roles in maintaining the structural integrity of the blood vessels, where the depletion of platelets has been shown to cause leakage because of microvessel disruption (Ref. Reference Ho-Tin-Noe, Demers and Wagner19). Platelets also safeguard and support the semi-permeability of the vessels by physically filling the gaps and secreting growth factors and cytokines to promote endothelial growth and maintain the barrier function of resting endothelium (Ref. Reference Ho-Tin-Noe, Demers and Wagner19). Conversely, activated platelets have been shown to secrete various inflammation mediators that can disrupt vessel integrity (Ref. Reference Malavige and Ogg8). Thus, platelet level and activation status are critical in dengue progression, and aberrant platelet parameters may increase the risk of severe dengue.

Mechanisms of thrombocytopenia in dengue infection

The mechanisms of dengue-mediated thrombocytopenia in patients still eluded researchers, but significant progress has been made in recent years. In short, dengue infection may disrupt platelet production in the bone marrow and/or expedite platelet clearance, causing thrombocytopenia. This review will be split into three main sections. In the first section (‘Dengue reduces platelet production’), we will focus on the effects of dengue infection on megakaryopoiesis and thrombopoiesis. In the second section (‘Dengue increases platelet activation and clearance’), we will discuss the effects of dengue infection on platelet activation and clearance. Lastly, we will discuss an application of the knowledge in this research area.

Dengue reduces platelet production

Megakaryopoiesis and thrombopoiesis

Megakaryocytes are large haematopoietic cells ranging from 20 to 100 μm. The name ‘mega’ (large) – ‘karyo’ (nucleus) reflects their appearance, with a large and multilobulated nucleus that encompasses most of their granular cytoplasm. The nuclei of mature megakaryocytes are characterized as hyperploid, with an average ploidy of 16 N DNA, but could go up to 128 N (Ref. Reference Noetzli, French and Machlus20). Megakaryocytes are specialized cells that serve as the precursor in platelet biogenesis, where the granulated cytoplasm will pinch off, releasing up to 104 platelets (Ref. Reference Pang, Weiss and Poncz21).

Megakaryopoiesis, or the process of producing mature megakaryocytes, is orchestrated by several factors that play different roles at different stages of megakaryopoiesis. It begins with haematopoietic stem cells (HSCs) differentiating into common myeloid progenitors (CMPs). CMPs will then differentiate into megakaryocyte–erythrocyte progenitors (MEPs). MEPs will continue to differentiate along the megakaryocytic lineage to produce megakaryocyte progenitors (MKPs) or megakaryoblasts and eventually form mature megakaryocytes to produce platelets (Refs Reference Geddis22, Reference Couldwell and Machlus23).

Thrombopoietin (TPO) is a key factor that binds to the c-Mpl receptor to activate several signalling pathways involving Janus kinase, signal transducer and activator of transcription protein 3 and 5 (STAT3 and STAT5), p38 mitogen-activated protein kinase (p38 MAPK), extracellular signal-regulated kinase, phosphoinositide-3 kinase (PI3K) and protein kinase B (AKT) to initiate megakaryopoiesis (Ref. Reference Kaushansky24). Activation of these signalling pathways increases the expression of transcription factors such as GATA1, FOG1, FLI1, MYB and nuclear factor erythroid 2 (NF-E2), which upregulate genes crucial for megakaryocytes such as CD41, CD42b and CD61 (Refs Reference Noetzli, French and Machlus20, Reference Kaushansky24). TPO also increases the number of MKP cells, induces polyploidy and promotes megakaryocyte maturation (Ref. Reference Kaushansky24).

In addition to TPO, various cytokines such as the interleukin family (IL-1b, IL-3 and IL-6) work in synergy with TPO to promote the proliferation of MKP cells (Ref. Reference Noetzli, French and Machlus20). IL-1b has also been shown to upregulate the gene and protein levels of TPO, c-Jun, c-Fos, GATA-1 and NF-E2 in a dose-dependent manner, which likely serves as the basis for thrombocytosis during inflammation (Ref. Reference Chuen25).

Some cytokines and factors influence megakaryopoiesis independent of TPO-mediated signalling. For example, IL-1a induces proplatelet shedding into the bone marrow during megakaryocyte maturation (Ref. Reference Noetzli, French and Machlus20). C-C motif ligand 5 (CCL5) or regulated on activation, normal T cell expressed and secreted chemokine (RANTES) can also increase megakaryocyte ploidy and platelet production through CCR5 signalling (Ref. Reference Machlus26). A study has identified an enzyme, tyrosyl-tRNA synthetase variant (YRSACT), that can directly convert HSCs into MKPs to enhance megakaryopoiesis and thrombopoiesis of human induced-pluripotent cells that lack TPO signalling in vitro and in vivo (Ref. Reference Kanaji27).

Thrombopoiesis is the process of platelet formation and is highly dependent on the development of mature megakaryocytes during megakaryopoiesis (Ref. Reference Severin, Ghevaert and Mazharian28). During this process, cytoskeletal proteins, membrane and granulated cytoplasm of mature megakaryocytes undergo extensive remodelling and form pseudopodial projections called proplatelets (Ref. Reference Geddis22). The proplatelets, characterized by long, thin shafts with swelling at the tip, are then released into the blood vessels (Ref. Reference Kaushansky29). The production of proplatelets requires intracellular factors such as the NF-E2 transcription factor and extracellular factors such as estradiol (Ref. Reference Nagata30) and shear force (Ref. Reference Thon31). TPO, in addition to its central role in megakaryopoiesis, is also a crucial factor in thrombopoiesis (Ref. Reference Kaushansky24). As previously mentioned, TPO-mediated signalling elevates the level of the NF-E2 transcription factor, which is paramount in megakaryocyte maturation (Ref. Reference Noetzli, French and Machlus20). NF-E2 is also paramount in thrombopoiesis, as knocking out this factor resulted in severe thrombocytopenia because of impaired thrombopoiesis but no effects on megakaryopoiesis, as the megakaryocytes in these mice, though morphologically comparable with normal megakaryocytes, were unable to generate proplatelet extensions (Ref. Reference Lecine32). Thrombopoiesis and its critical factors and regulators have been extensively reviewed elsewhere (Refs Reference Noetzli, French and Machlus20, Reference Noh33, Reference Tijssen and Ghevaert34).

Dengue impairs megakaryopoiesis and thrombopoiesis

Several viruses can disrupt megakaryocyte proliferation and maturation. Herpesviruses such as human cytomegalovirus and human herpes simplex virus impair megakaryopoiesis by causing apoptosis (Ref. Reference Raadsen35). Pathogenic Hantaan orthohantavirus has been reported to infect and replicate in mature megakaryocytes by hijacking CD61 surface protein (Ref. Reference Lutteke36).

DENV causes platelet reduction in patients, beginning typically on day 2 before the onset of the critical phase and persisting until days 6–7 (Ref. 1). One mechanism of thrombocytopenia is megakaryocyte infection and death. DENV has been shown to efficiently infect human megakaryocyte cell lines, primary human megakaryocytes or progenitors and megakaryocytes in humanized mice (Refs Reference Vogt37, Reference Noisakran38, Reference Lahon, Arya and Banerjea39).

MEG-01 is one of the most common megakaryoblastic leukaemia cell lines in thrombocyte studies. Lahon et al. reported an effective DENV infection and replication in TPO-treated MEG-01 cells, compromising the PI3K/AKT/mTOR pathway that is essential in megakaryocyte survival and maturation (Ref. Reference Lahon, Arya and Banerjea39). DENV infection also caused significant cell death and repressed the expression of megakaryopoiesis-related transcription factors, namely GATA-1, GATA-2, NF-E2 and mature megakaryocyte marker CD61 (Ref. Reference Lahon, Arya and Banerjea39). Another study verified the susceptibility of MEG-01 cells to DENV infection, but reported a reduced DENV replication in phorbol-12-myristate-13-acetate (PMA) pre-treated MEG-01 cells when compared with control and MEG-01 cells treated with PMA after DENV infection (Ref. Reference Banerjee40). In this study, Banerjee et al. showed that viral RNA copy number was significantly low in PMA-pre-treated cells at 2 dpi compared with control and PMA treatment post-infection samples. The authors stated that these PMA-differentiated MEG-01 cells were refractory against DENV infection/replication (Ref. Reference Banerjee40). However, this statement contradicts several reports showing high DENV infection in mature megakaryocytes (Refs Reference Vogt37, Reference Noisakran38, Reference Attatippaholkun41). A plausible explanation for the lower DENV replication in PMA-pre-treated cells could be because of PMA activity that activates protein kinase C (PKC), which in turn, inhibits DENV NS5 and suppresses DENV replication (Ref. Reference Noppakunmongkolchai42). In contrast, PMA treatment post-DENV infection substantially increased DENV replication in MEG-01 cells (Refs Reference Banerjee40, Reference Kaur43). Similar observations were reported in K562 cells treated with PMA after DENV infection (Ref. Reference Kaur43). PMA treatment post-DENV infection significantly increased DENV replication and infectious progeny production without influencing viral entry. In this study, PMA suppressed cellular reactive oxygen species (ROS) production by upregulating antioxidant NFE2L2 transcription factor to allow virus replication (Ref. Reference Kaur43). It is also possible that the maturation process with highly active transcriptional and translational machineries inadvertently create an intracellular milieu suitable for viral replication and virion production (Ref. Reference Singh44).

In addition to cell lines, primary CD34+ MKPs obtained from umbilical cord blood are also prone to DENV infection, leading to disrupted colony formation and elevated apoptosis (Ref. Reference Basu45). Similarly, another study reported that umbilical cord blood cells were highly susceptible to DENV infection, and these CD34+ cells serve as the reservoir of viral progenies (Ref. Reference Vats46). Interestingly, these progenies were mostly latent.

In a humanized mouse model, a higher percentage of DENV infection in mature CD41a+/− + CD42b+ megakaryocytes (35%) was observed when compared with immature CD41a+ + CD42b megakaryocytes (1.5%) (Ref. Reference Vogt37). However, the number of mature human megakaryocytes in this model was greatly diminished (Ref. Reference Vogt37). Nevertheless, this finding is consistent with another study where DENV displayed a selective tropism for CD42-expressing MEG-01 cells compared with CD42 or CD41+ cells (Ref. Reference Attatippaholkun41). Another study, using patient samples and rhesus monkeys, showed CD61+ cells were susceptible to DENV infection (Ref. Reference Noisakran38).

In addition to impaired megakaryopoiesis, DENV infection can hamper thrombopoiesis by significantly reducing proplatelet formation in PMA-treated MEG-01 cells (Ref. Reference Banerjee40). The exact mechanism of this effect is currently unclear but may involve the NF-E2 transcription factor, a crucial thrombopoiesis factor. DENV infection markedly reduced NF-E2 protein expression in mature MEG-01 cells (Ref. Reference Lahon, Arya and Banerjea39). Another potential mechanism may involve the sphingosine 1-phosphate receptor (S1pr). S1pr1, one of the sphingosine-1-phosphate receptor subtypes, has been studied in dengue infection settings but mostly on its roles in regulating vascular permeability where sphingosine 1-phosphate (S1P) levels were found markedly reduced in acute dengue patients (Ref. Reference Gomes47). S1pr1 was reported to be crucial in platelet production, where knocking out of S1pr1 in mice resulted in severe thrombocytopenia concomitant with significant reductions in proplatelet formation and proplatelet shedding (Ref. Reference Zhang48). Thus, the S1P–S1pr1 axis may also be implicated in dengue-mediated thrombopoiesis impairment.

A study unravelled a novel role of DENV NS3 protease in cleaving GrpE protein homologue 1 (GrpEL1), a cochaperone of mitochondrial heat shock protein 70 (mtHsp70) (Ref. Reference Gandikota49). GrpEL1 and its orthologue, GrpEL2, form a subcomplex that is pivotal in maintaining the stability of nucleotide exchange factor for mtHsp70, thus modulating critical mtHsp70 functions such as importing mitochondrial pre-protein into the matrix (Ref. Reference Srivastava50). Overexpression of NS3 protease reduced GrpEL1 protein level in vitro. More importantly, GrpEL1 protein level was significantly reduced in dengue patient samples, especially in dengue haemorrhagic fever (DHF) and dengue shock syndrome (DSS) patients, which correlated with thrombocytopenia level in these patients (Ref. Reference Gandikota49). Reduced GrpEL1 compromised mitochondria health and functions, leading to thrombocytopenia (Ref. Reference Gandhi51). Another form of DENV protease, NS2B-NS3 protease, which localized to the nucleus, could cleave erythroid differentiation regulatory factor 1 (EDRF1) transcription factor (Ref. Reference Gandhi51). EDRF1 is critical in platelet formation as it regulates the levels of GATA1 and spectrin, a cytoskeletal protein essential for membrane re-organization during platelet biogenesis (Ref. Reference Patel-Hett52). Overexpression of NS2B-NS3 protease significantly diminished EDRF1 and GATA1 protein levels. Similarly, EDRF1 level was also reduced in dengue patients, especially in thrombocytopenic dengue haemorrhage patients and dengue shock syndrome patients (Ref. Reference Gandhi51). Thus, NS3 contributes to thrombocytopenia by cleaving EDRF1 to impair thrombopoiesis, and cleaving GrpEL1 to cause mitochondrial dysfunction and disrupt platelet formation.

In short, DENV can infect megakaryocytes at different stages of cell maturation and impair platelet production, leading to reduced platelet formation.

Dengue increases platelet activation and clearance

Platelet activation

The roles of platelets during haemostasis and thrombosis events begin with a series of activation processes that include platelet adhesion, network extension and thrombus formation involving multiple factors, as summarized in Table 1 (Ref. Reference Thomas and Storey53). Briefly, in the event of high shear or vascular injury, nearby platelets will be exposed to von Willebrand factor (vWF) on the vessel wall (Ref. Reference Freedman54). The vWF will serve as a bridge to connect platelet glycoprotein (GP) Ib receptor complex on the platelet's membrane with the collagen layer, allowing platelet GPVI receptors to adhere directly onto the collagen matrix for a strong adhesion (Ref. Reference Estevez and Du55). The vWF also facilitates the activation of circulating platelets by connecting the GP IIb/IIIa receptor on the activated platelets to the GP Ib on the circulating platelets to activate them. Bound fibrinogen will strengthen the crosslink of the GP IIb/IIIa receptor on both activated platelets (Ref. Reference Jennings56). The activated platelets undergo structural changes with the formation of pseudopodia and initiate the release of platelet granules (Ref. Reference Jennings56). The released granules contain chemokines and cytokines to signal other circulating platelets to interact with the adhering platelets and form extensions of activated platelet network (Ref. Reference Herter, Rossaint and Zarbock57).

Table 1. Factors in platelet activation and aggregation

Platelets contain α- and dense granules. α-Granules are the most abundant secretory organelles in platelets, and they contain adhesive proteins, cytokines and chemokines crucial for platelet adhesiveness and thrombus formation (Ref. Reference Heijnen and van der Sluijs65). These chemokines such as RANTES, CXCL1, platelets factor 4 (PF4 or CXCL4) and IL-6 interact with platelet surface receptors and immune cells (Ref. Reference Smith66). RANTES (CCL5) binds to chemokine receptors CCR1, CCR3 and CCR5 on platelets to stimulate other platelets and recruit monocyte to the inflamed endothelium (Ref. Reference Gear and Camerini67). PF4 also facilitates platelet adhesion and mediates leucocyte interaction (Ref. Reference Fox68).

Dense granules, on the other hand, contain small molecules such as adenosine diphosphate (ADP), adenosine triphosphate (ATP), calcium ions serotonin and phosphates (Ref. Reference Heijnen and van der Sluijs65). ADP is crucial in platelet activation through its interaction with two G-protein-coupled receptors (P2Y1 and P2Y12) to initiate platelet aggregation and provide a feedback mechanism to increase the secretion of thromboxane A2 (TxA2) and other agonists (Ref. Reference Offermanns69). Studies showed that the absence of P2Y12 in humans resulted in haemorrhage, and lacking P2Y1 in mice prolonged the bleeding time (Ref. Reference Gachet70). In addition to procoagulant activity, ADP also mediates the release of TxA2 where the interaction of TxA2 with TxA2 receptor (TP) attracts other platelets to bind to the adherent platelets, forming a stable thrombus (Ref. Reference Jennings56). Thrombin plays essential roles in plug formation through protease-activated receptor 1 and 4 (PAR-1 and PAR-4) that are coupled to Gq and G12/G13 proteins, respectively (Ref. Reference Chen71). In addition, hormones such as serotonin activate platelets through 5-hydroxytryptamine 2A (5HT-2A) receptors and promote platelet aggregation (Ref. Reference Marcinkowska72).

Platelet death and clearance

Platelets generally have a life span of 5–10 days before clearance from the body (Ref. Reference Tanaka, Bolliger, Hemmings and Egan73) via several mechanisms such as platelet apoptosis, antibody-mediated phagocytosis in the spleen, removal by the Kupffer cells in the liver via lectin-glycan recognition or massive platelet release because of blood loss (Refs Reference Grozovsky, Hoffmeister and Falet74, Reference Quach, Chen and Li75). Ageing platelets express a higher level of phosphatidylserine (PS), a type of phospholipid that activates apoptosis (Ref. Reference Hou76). Other pro- and anti-apoptotic factors such as Bak, Bax and Bcl-XL are also involved in platelet apoptosis (Ref. Reference Quach, Chen and Li75). Thrombin-activated platelets undergo metabolic exhaustion marked by mitochondrial depolarization, accumulation of ROS and ATP depletion, followed by platelet dysfunctional and disintegration, calpain-activation and eventually platelet fragmentation (death) (Ref. Reference Kim77). Activated and apoptotic platelets are also exposed to clearance by macrophages. Desialylation of platelet via neuraminidase or vWF action induces platelet clearance and acute thrombocytopenia (Ref. Reference Quach, Chen and Li75). Platelets are also cleared from the body through platelet plug clearance (Ref. Reference Ojha16).

Dengue increases platelet activation and apoptosis

Invading pathogens such as bacteria, parasites and viruses activates platelets via pattern recognition receptors such as Toll-like receptors (TLRs) and C-type lectin receptors (CLRs), complement Fc receptors (FcR), major histocompatibility complex (MHC) class I and many more (Refs Reference Attatippaholkun41, Reference Li, Li and Ni78, Reference Fard79). Excessive platelet activation and apoptosis, marked by elevated P-selectin and annexin V level, respectively, was observed in dengue patients during the febrile and defervescence phases, causing thrombocytopenia in these patients when compared with healthy controls, non-dengue febrile patients and non-thrombocytopenic dengue patients (Ref. Reference Hottz64). Similarly, increased platelet activation concurrent with thrombocytopenia was observed in dengue patients during the critical phase (days 4–6) (Ref. Reference Ojha16). Dengue-induced thrombocytopenia was replicated in vivo when mice injected with DENV2 suffered thrombocytopenia concomitant with elevated platelet activation (Ref. Reference Masri63). Several mechanisms for platelet activation and death mediated by the DENV are summarized below.

DENV NS1. Following infection, DENV hijacks the translational machinery to produce new dengue proteins such as dengue non-structural 1 (NS1) protein and new virions. NS1 binding to TLR4 on primary human platelets enhanced platelet activation marked by enhanced P-selectin expression, causing platelet apoptosis (Ref. Reference Chao58) and the released of granule-stored chemokines such as RANTES, macrophage migration inhibitory factor (MIF) and PF4 (Ref. Reference Quirino-Teixeira80). Full-length NS1 and all NS1 domains, especially the wing domain, could activate platelets, but only the full-length NS1 could induce platelet aggregation in the presence of platelet agonists (Ref. Reference Garcia-Larragoiti81). The increased P-selectin expression on NS1-activated platelets promoted platelet adherence to endothelial cells and macrophages, compromising endothelial permeability and increasing platelet phagocytosis, respectively (Ref. Reference Chao58). DENV NS1 also amplified thrombo-inflammatory responses and induced the synthesis of pro-IL-1β but not its secretion (Ref. Reference Quirino-Teixeira80). IL-1β secretion from NS1-activated platelets was mediated by nucleotide-binding domain leucine-rich repeat-containing protein (NLRP3)-caspase-1 inflammasome (Ref. Reference Quirino-Teixeira80). More importantly, the secretion of pro-inflammatory cytokines such as IL-1β and MIF contributed to vascular permeability and tissue injury (Ref. Reference Pan82). DENV NS1 also sensitized the platelets to aggregation where exposure to DENV NS1 alone could not stimulate platelet aggregation, but co-treatment of NS1 with subthreshold levels of platelet agonists markedly increased platelet aggregation which contributed to thrombocytopenia (Refs Reference Chao58, Reference Garcia-Larragoiti81).

DENV envelope protein. Dengue can infect cells via two CLRs, specifically DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin) and CLEC-2 (C-type lectin-like-receptor). The N-glycans on the DENV envelope protein can bind to DC-SIGN, causing virus adsorption onto the plasma membrane (Ref. Reference Alen83). The binding of DENV to DC-SIGN was shown to increase platelet activation, cause mitochondria dysfunction and trigger caspase-dependent apoptosis (Ref. Reference Hottz64).

As previously mentioned, co-stimulation of NS1-activated platelet with suboptimal ATP concentration upregulated IL-1β secretion, possibly through NLRP3 inflammasome (Ref. Reference Quirino-Teixeira80). Similarly, DENV envelope domain III (EIII) also activated platelet, and caused thrombocytopenia in vivo primarily through NLRP3-mediated pyroptosis (Ref. Reference Lien84). EIII also impaired clotting time, and interestingly, inhibition of this response pathway via NLRP3 inhibition or EIII blocking could significantly improve the conditions (Ref. Reference Lien84).

Other factors. Proteomics analysis comparing platelets from dengue patients with healthy controls showed 167 differentially abundant proteins, with most of the proteins involved in ‘antigen processing and presentation’ and ‘platelet activation’ processes (Ref. Reference Trugilho85). As expected, P-selectin, PF4 and RANTES were among the highly upregulated factors in dengue (Ref. Reference Trugilho85). Histones H2A and HLA class I were also upregulated in dengue-infected platelets. The binding of circulating histone H2A to the TLR4 receptor on platelets caused platelet activation that could be completely blocked by anti-histone H2A monoclonal antibody (Ref. Reference Trugilho85).

CLEC-2 also plays a pivotal role in platelet activation. Elevated CD62p and CD63 expression after DENV infection was abolished in human platelets treated with CLEC2-mAb and clec2−/− mouse platelets (Ref. Reference Sung, Huang and Hsieh86). More importantly, the study reported that activated platelets via CLEC2 produced extracellular vesicles that activated neutrophils and macrophages via CLEC5A and TLR2 and contributed to the formation of neutrophil extracellular traps (NETs). NET level in the plasma was reported to inversely correlate with platelet count and positively correlate with P-selectin expression during days 7–13 of disease in acute dengue patients (Ref. Reference Garishah87).

Mast cell-derived serotonin was reported to be elevated during dengue infection in vitro and in vivo and activated the platelets through the 5HT-2 receptor, which was then demonstrated to be phagocytosed (Ref. Reference Masri63). Inhibiting the 5HT-2 receptor reduced dengue-induced thrombocytopenia and platelet activation (Ref. Reference Masri63). However, although the roles of serotonin in platelet activation, aggregation and phagocytosis were sufficiently established in vivo (Ref. Reference Masri63), an earlier metabolomic study showed a significant reduction of serotonin in dengue high fever patients compared with dengue fever patients, especially during the febrile and defervescence phases (Ref. Reference Cui88). The study also proposed that reduced serotonin coupled with elevated interferon gamma may serve as a biomarker of severe dengue. The conflicting reports on serotonin levels post-dengue infection could be because of the different hosts where, as opposed to rodents, mast cells are not the primary source of serotonin in humans (Ref. Reference de Mast and de Groot89). Thus, the role of serotonin in dengue and dengue-mediated thrombocytopenia requires further investigation.

Another important mechanism of platelet activation is through antibody-dependent enhancement or ADE. This mechanism involves the interaction between virus-immunoglobulin (Ig) G complexes and FcR-bearing cells (Ref. Reference Koupenova, Livada and Morrell90). Unlike other factors, ADE is primarily active during secondary DENV infection as the IgG antibody secreted during the first infection binds to the second-generation virus and permits faster and more severe infections via FcR expressed on immune cells and platelets (Ref. Reference Teo91). A previous study showed that immature dengue virion becomes highly infectious in the presence of anti-prM antibodies that facilitate viral entry and enhance the intracellular processing of prM to M protein (Ref. Reference Rodenhuis-Zybert92). Another study demonstrated that DHF/DSS patients showed elevated afucosylated IgGs with strong affinities towards FcγRIIA and FcγRIIIA receptors expressed on the platelets, leading to platelet reduction. The level of afucosylated IgGs correlated with thrombocytopenia severity in these patients and was a significant risk factor for thrombocytopenia (Ref. Reference Wang93). As FcRs are expressed on immune cells, it was postulated that thrombocytopenia observed in this study could be because of enhance platelet activation, platelet sequestration and/or antibody-dependent cell cytotoxicity (Ref. Reference Wang93). Additionally, afucosylated IgGs could also serve as a prognostic factor of dengue disease severity (Ref. Reference Bournazos94).

Dengue increases platelet clearance

The activated and apoptotic platelets are subsequently cleared from the body in several ways. Phagocytosis of apoptotic platelets by macrophages in acute and early convalescence dengue patients was reported to be 2.5–3.5 times higher than the platelets from healthy controls (Ref. Reference Alonzo95). Phagocytosis was substantially reduced when these patient-derived platelets were pre-treated with D89E mutant protein that masked the PS on the apoptotic platelets (Ref. Reference Alonzo95). Phagocytosis of DENV2-activated platelets by primary human monocytes was also reported in vitro (Ref. Reference Ojha16).

The vWF could also mediate platelet clearance. vWF, elevated in thrombocytopenic dengue patients, increased platelet desialylation, thus exposing the platelet to clearance via Ashwell–Morell receptor-mediated pathway (Ref. Reference Riswari60). DENV-activated platelets also showed high levels of bound complement factor C3 and IgG, suggesting elevated platelet lysis, clearance and increased thrombus formation that caused platelet depletion in the plasma (Refs Reference Ojha16, Reference Wang93). Immune-mediated platelet clearance and lysis during dengue infection have been reviewed here (Ref. Reference Hottz96).

The events and processes described in the sections ‘Dengue reduces platelet production’ and ‘Dengue increases platelet activation and clearance’ are not mutually exclusive and may simultaneously occur to exacerbate dengue symptoms. Figure 2 summarizes these events. Since the pathophysiology and progression of dengue fever vary among patients, it is challenging to determine the predominant mechanism for thrombocytopenia in patients. Nonetheless, research in this field has improved disease management. An emerging application of knowledge in this field is immature platelet fraction (IPF), which will be briefly discussed in the next section.

Figure 2. Summary of DENV-mediated thrombocytopenia. DENV causes thrombocytopenia in several ways. (1) DENV impairs megakaryopoiesis. DENV infects and causes apoptosis of megakaryocytes and the progenitor cells. DENV also prevents megakaryocyte maturation. (2) DENV impairs thrombopoiesis by reducing megakaryocytes and interfering with platelet formation. (3) Secondary DENV infection causes elevated platelet activation via IgG–FcR connection in the ADE process. (4) DENV increases platelet activation. (5) DENV causes cytokines, chemokines and other factors to be released from infected platelets and nearby endothelial cells. (6) DENV infects and causes platelet apoptosis. (7) ADE, elevated platelet activation and secretion of various factors cause the platelets to coagulate. (8) Coagulated platelets and apoptotic platelets are cleared from the circulation by phagocytes. Regular arrows (→) in green indicate stimulatory modifications. Blunt-ended arrows (˧) in red indicate inhibitory modifications.

Potential application: IPF% as a predictive tool for platelet recovery in dengue patient

Thus far, there is no treatment against dengue, but targeting the molecular factors involved in dengue-induced thrombocytopenia could prevent or reverse this complication, and could be vital in managing dengue progression to severe dengue (Refs Reference Masri63, Reference Lien84, Reference Sung, Huang and Hsieh86). There have been considerable advances in platelet study that could be applied to address platelet abnormality in dengue.

IPF is an automated measurement of reticulated platelet levels. These reticulated platelets are nascent platelets with punctate and coarse condensations. They still contain the mRNA that can be stained with nucleic acid binding dyes and quantified (Ref. Reference Briggs97). Essentially, this parameter reflects the rate of thrombopoiesis that is upregulated in response to elevated platelet consumption or clearance (Ref. Reference Briggs97).

Normal IPF% is in the range of 1.1–6.1%, with a mean of 3.4%, but in hyperdestructive thrombocytopenic patients, the IPF% could elevate to 22.3% in autoimmune thrombocytopenic purpura patients and 17.2% in thrombotic thrombocytopenic purpura patients (Ref. Reference Briggs97). In patients with hypoproductive conditions such as aplastic anaemia and chemotherapy-induced bone marrow toxicity, IPF% levels remain unchanged, corroborating the utility of this parameter in evaluating thrombopoiesis activity (Ref. Reference Jeon98). In dengue patients, IPF% increases to varying degrees (Refs Reference Dadu99, Reference Looi100).

In dengue, IPF may serve as an indicator of platelet recovery from dengue-induced thrombocytopenia. In a study conducted on 32 dengue patients, more than 84% of the patients showed platelet count recovery 24 h after reaching the IPF peak (Ref. Reference Dadu99). Similarly, another study showed an inverse correlation between IPF% and platelet count where IPF% was high and platelet count was low during the early phase of the disease (days 2–9). Platelet count rebounded 24–48 h after peak IPF% (Ref. Reference Looi100). This inverse correlation was less pronounced in severe dengue patients, where platelet recovery was slightly delayed (Ref. Reference Looi100).

Another important observation was severe dengue patients had a significant elevation of IPF% on days 3–5 after the onset of fever compared with non-severe dengue patients (Ref. Reference Looi100). Another study reported a similar trend where severe dengue patients had higher IPF% when compared with non-severe dengue patients (Ref. Reference Yasuda101). The observed changes in this parameter suggest that platelet consumption or clearance could be the primary underlying mechanism of dengue-induced thrombocytopenia.

In short, IPF% may serve as a useful indicator for platelet recovery as this parameter signals the body's response to platelet death and clearance because of DENV infection. Platelet recovery is typically observed within 24–48 h post-peak IPF% in most dengue patients. Thus, this parameter may aid clinicians in monitoring the progress of the disease. Nonetheless, further studies are needed to determine the utility of this parameter in dengue management as well as to verify the correlation between IPF% and dengue severity.

Conclusion

In conclusion, the DENV can infect megakaryocytes and their progenitor cells, causing cell death to reduce the rate of platelet production. DENV also induces platelet activation, leading to platelet apoptosis, clearance and phagocytosis. These non-mutually exclusive events complicate the efforts to prevent or treat this complication in dengue. However, there has been significant progress in understanding and treating thrombocytopenia, which will benefit dengue patients. IPF, for example, could be further investigated as a prognostic tool to determine platelet recovery in dengue patients.

Author contributions

A. S. K. designed, performed the literature search and prepared the manuscript. W. H. H. performed the literature search and prepared the manuscript. F. I. reviewed the manuscript. S. O. supervised W. H. H. and edited the manuscript. N. N. R. supervised W. H. H., reviewed and approved the manuscript. All authors read and approved the final manuscript.

Funding statement

This study was financially supported by the Ministry of Higher Education, Malaysia through Fundamental Research Grant Scheme FRGS/1/2020/SKK0/UM/02/28. The funding agency was not involved in any way in this study.

Competing interests

None.

References

World Health O (2009) Dengue guidelines for diagnosis, treatment, prevention and control: new edition. Geneva: World Health Organization.Google Scholar
Jasamai, M et al. (2019) Current prevention and potential treatment options for dengue infection. Journal of Pharmaceutical Science 22, 440456.Google ScholarPubMed
Obi, JO et al. (2021) Current trends and limitations in dengue antiviral research. Tropical Medicine & Infectious Disease 6, 180.CrossRefGoogle ScholarPubMed
Faridah, IN et al. (2022) Dynamic changes of platelet and factors related dengue haemorrhagic fever: a retrospective study in Indonesian. Diagnostics (Basel) 12, 950.CrossRefGoogle ScholarPubMed
Mourao, MP et al. (2007) Thrombocytopenia in patients with dengue virus infection in the Brazilian Amazon. Platelets 18, 605612.CrossRefGoogle ScholarPubMed
van de Weg, CA et al. (2012) Evaluation of the 2009 WHO dengue case classification in an Indonesian pediatric cohort. American Journal of Tropical Medicine & Hygiene 86, 166170.CrossRefGoogle Scholar
Makroo, RN et al. (2007) Role of platelet transfusion in the management of dengue patients in a tertiary care hospital. Asian Journal of Transfusion Science 1, 47.CrossRefGoogle ScholarPubMed
Malavige, GN and Ogg, GS (2017) Pathogenesis of vascular leak in dengue virus infection. Immunology 151, 261269.CrossRefGoogle ScholarPubMed
de Azeredo, EL, Monteiro, RQ and de-Oliveira Pinto, LM (2015) Thrombocytopenia in dengue: interrelationship between virus and the imbalance between coagulation and fibrinolysis and inflammatory mediators. Mediators of Inflammation 2015, 313842.CrossRefGoogle ScholarPubMed
Chaudhary, R et al. (2006) Transfusion support to dengue patients in a hospital based blood transfusion service in north India. Transfusion and Apheresis Science 35, 239244.CrossRefGoogle Scholar
Adane, T and Getawa, S (2021) Coagulation abnormalities in dengue fever infection: a systematic review and meta-analysis. PLoS Neglected Tropical Disease 15, e0009666.CrossRefGoogle ScholarPubMed
Scott, AS and Fong, E (2016) Body Structures and Functions. USA: Cengage Learning.Google Scholar
Yun, SH et al. (2016) Platelet activation: the mechanisms and potential biomarkers. Biomed Research International 2016, 9060143.CrossRefGoogle ScholarPubMed
Yadav, S and Storrie, B (2017) The cellular basis of platelet secretion: emerging structure/function relationships. Platelets 28, 108118.CrossRefGoogle Scholar
Izak, M and Bussel, JB (2014) Management of thrombocytopenia. F1000Prime Reports 6, 45.CrossRefGoogle ScholarPubMed
Ojha, A et al. (2017) Platelet activation determines the severity of thrombocytopenia in dengue infection. Scientific Reports 7, 41697.CrossRefGoogle ScholarPubMed
Pothapregada, S, Kamalakannan, B and Thulasingam, M (2015) Role of platelet transfusion in children with bleeding in dengue fever. Journal of Vector Borne Disease 52, 304308.CrossRefGoogle ScholarPubMed
Thach, TQ et al. (2021) Predictive markers for the early prognosis of dengue severity: a systematic review and meta-analysis. PLoS Neglected Tropical Disease 15, e0009808.CrossRefGoogle ScholarPubMed
Ho-Tin-Noe, B, Demers, M and Wagner, DD (2011) How platelets safeguard vascular integrity. Journal of Thrombosis and Haemostasis 9(Suppl 1), 5665.CrossRefGoogle ScholarPubMed
Noetzli, LJ, French, SL and Machlus, KR (2019) New insights into the differentiation of megakaryocytes from hematopoietic progenitors. Arterioscler, Thrombosis and Vascular Biology 39, 12881300.CrossRefGoogle ScholarPubMed
Pang, L, Weiss, MJ and Poncz, M (2005) Megakaryocyte biology and related disorders. Journal of Clinical Investigation 115, 33323338.CrossRefGoogle ScholarPubMed
Geddis, AE (2009) The regulation of proplatelet production. Haematologica 94, 756759.CrossRefGoogle ScholarPubMed
Couldwell, G and Machlus, KR (2019) Modulation of megakaryopoiesis and platelet production during inflammation. Thrombosis Research 179, 114120.CrossRefGoogle Scholar
Kaushansky, K (2005) The molecular mechanisms that control thrombopoiesis. Journal of Clinical Investigation 115, 33393347.CrossRefGoogle ScholarPubMed
Chuen, CK et al. (2004) Interleukin-1beta up-regulates the expression of thrombopoietin and transcription factors c-Jun, c-Fos, GATA-1, and NF-E2 in megakaryocytic cells. Journal of Laboratory and Clinical Medicine 143, 7588.CrossRefGoogle ScholarPubMed
Machlus, KR et al. (2016) CCL5 derived from platelets increases megakaryocyte proplatelet formation. Blood 127, 921926.CrossRefGoogle ScholarPubMed
Kanaji, T et al. (2018) Tyrosyl-tRNA synthetase stimulates thrombopoietin-independent hematopoiesis accelerating recovery from thrombocytopenia. Proceeding of the National Academy of Sciences of the USA 115, E8228E8E35.CrossRefGoogle ScholarPubMed
Severin, S, Ghevaert, C and Mazharian, A (2010) The mitogen-activated protein kinase signaling pathways: role in megakaryocyte differentiation. Journal of Thrombosis and Haemostasis 8, 1726.CrossRefGoogle ScholarPubMed
Kaushansky, K (2008) Historical review: megakaryopoiesis and thrombopoiesis. Blood 111, 981986.CrossRefGoogle ScholarPubMed
Nagata, Y et al. (2003) Proplatelet formation of megakaryocytes is triggered by autocrine-synthesized estradiol. Genes & Development 17, 28642869.CrossRefGoogle ScholarPubMed
Thon, JN et al. (2010) Cytoskeletal mechanics of proplatelet maturation and platelet release. Journal of Cell Biology 191, 861874.CrossRefGoogle Scholar
Lecine, P et al. (1998) Mice lacking transcription factor NF-E2 provide in vivo validation of the proplatelet model of thrombocytopoiesis and show a platelet production defect that is intrinsic to megakaryocytes. Blood 92, 16081616.CrossRefGoogle ScholarPubMed
Noh, JY (2021) Megakaryopoiesis and platelet biology: roles of transcription factors and emerging clinical implications. International Journal of Molecular Science 22, 9615.CrossRefGoogle ScholarPubMed
Tijssen, MR and Ghevaert, C (2013) Transcription factors in late megakaryopoiesis and related platelet disorders. Journal of Thrombosis & Haemostasis 11, 593604.CrossRefGoogle ScholarPubMed
Raadsen, M et al. (2021) Thrombocytopenia in virus infections. Journal of Clinical Medicine 10, 877.CrossRefGoogle ScholarPubMed
Lutteke, N et al. (2010) Switch to high-level virus replication and HLA class I upregulation in differentiating megakaryocytic cells after infection with pathogenic hantavirus. Virology 405, 7080.CrossRefGoogle Scholar
Vogt, MB et al. (2019) Dengue viruses infect human megakaryocytes, with probable clinical consequences. PLoS Neglected Tropical Diseases 13, e0007837.CrossRefGoogle Scholar
Noisakran, S et al. (2012) Infection of bone marrow cells by dengue virus in vivo. Experimental Hematology 40, 250259, e4.CrossRefGoogle Scholar
Lahon, A, Arya, RP and Banerjea, AC (2021) Dengue virus dysregulates master transcription factors and PI3K/AKT/mTOR signaling pathway in megakaryocytes. Frontiers in Cellular and Infection Microbiology 11, 715208.CrossRefGoogle ScholarPubMed
Banerjee, A et al. (2020) Dengue virus infection impedes megakaryopoiesis in MEG-01 cells where the virus envelope protein interacts with the transcription factor TAL-1. Scientific Reports 10, 19587.CrossRefGoogle Scholar
Attatippaholkun, N et al. (2018) Selective tropism of dengue virus for human glycoprotein Ib. Scientific Reports 8, 2688.CrossRefGoogle Scholar
Noppakunmongkolchai, W et al. (2016) Inhibition of protein kinase C promotes dengue virus replication. Virology Journal 13, 35.CrossRefGoogle ScholarPubMed
Kaur, J et al. (2021) Replication of dengue virus in K562-megakaryocytes induces suppression in the accumulation of reactive oxygen species. Frontiers of Microbiology 12, 784070.CrossRefGoogle ScholarPubMed
Singh, K et al. (2022) Transcriptional and translational dynamics of Zika and dengue virus infection. Viruses 14, 1418.CrossRefGoogle ScholarPubMed
Basu, A et al. (2008) Dengue 2 virus inhibits in vitro megakaryocytic colony formation and induces apoptosis in thrombopoietin-inducible megakaryocytic differentiation from cord blood CD34+ cells. FEMS Immunology and Medical Microbiology 53, 4651.CrossRefGoogle ScholarPubMed
Vats, A et al. (2021) Evidence that hematopoietic stem cells in human umbilical cord blood is infectable by dengue virus: proposing a vertical transmission candidate. Heliyon 7, e06785.CrossRefGoogle Scholar
Gomes, L et al. (2014) Sphingosine 1-phosphate in acute dengue infection. PLoS ONE 9, e113394.CrossRefGoogle ScholarPubMed
Zhang, L et al. (2012) A novel role of sphingosine 1-phosphate receptor S1pr1 in mouse thrombopoiesis. Journal of Experimental Medicine 209, 21652181.CrossRefGoogle ScholarPubMed
Gandikota, C et al. (2020) Mitochondrial import of dengue virus NS3 protease and cleavage of GrpEL1, a cochaperone of mitochondrial Hsp70. Journal of Virology 94, e01178-20.CrossRefGoogle ScholarPubMed
Srivastava, S et al. (2017) Regulation of mitochondrial protein import by the nucleotide exchange factors GrpEL1 and GrpEL2 in human cells. Journal of Biological Chemistry 292, 1807518090.CrossRefGoogle Scholar
Gandhi, L et al. (2023) Differential localization of dengue virus protease affects cell homeostasis and triggers to thrombocytopenia. iScience 26, 107024.CrossRefGoogle Scholar
Patel-Hett, S et al. (2011) The spectrin-based membrane skeleton stabilizes mouse megakaryocyte membrane systems and is essential for proplatelet and platelet formation. Blood 118, 16411652.CrossRefGoogle ScholarPubMed
Thomas, MR and Storey, RF (2015) The role of platelets in inflammation. Thrombosis & Haemostasis 114, 449458.Google ScholarPubMed
Freedman, JE (2005) Molecular regulation of platelet-dependent thrombosis. Circulation 112, 27252734.CrossRefGoogle Scholar
Estevez, B and Du, X (2017) New concepts and mechanisms of platelet activation signaling. Physiology (Bethesda) 32, 162177.Google Scholar
Jennings, LK (2009) Mechanisms of platelet activation: need for new strategies to protect against platelet-mediated atherothrombosis. Thrombosis & Haemostasis 102, 248257.Google Scholar
Herter, JM, Rossaint, J and Zarbock, A (2014) Platelets in inflammation and immunity. Journal of Thrombosis & Haemostasis 12, 17641775.CrossRefGoogle Scholar
Chao, CH et al. (2019) Dengue virus nonstructural protein 1 activates platelets via Toll-like receptor 4, leading to thrombocytopenia and hemorrhage. PLoS Pathogen 15, e1007625.CrossRefGoogle ScholarPubMed
Djamiatun, K et al. (2012) Severe dengue is associated with consumption of von Willebrand factor and its cleaving enzyme ADAMTS-13. PLoS Neglected Tropical Disease 6, e1628.CrossRefGoogle ScholarPubMed
Riswari, SF et al. (2019) Desialylation of platelets induced by von Willebrand factor is a novel mechanism of platelet clearance in dengue. PLoS Pathogen 15, e1007500.CrossRefGoogle ScholarPubMed
Oliveira, ES et al. (2018) Increased levels of Txa(2) induced by dengue virus infection in IgM positive individuals is related to the mild symptoms of dengue. Viruses 10, 104.CrossRefGoogle Scholar
Fiestas Solorzano, VE et al. (2021) Different profiles of cytokines, chemokines and coagulation mediators associated with severity in Brazilian patients infected with dengue virus. Viruses 13, 1789.CrossRefGoogle ScholarPubMed
Masri, MFB et al. (2019) Peripheral serotonin causes dengue virus-induced thrombocytopenia through 5HT(2) receptors. Blood 133, 23252337.CrossRefGoogle ScholarPubMed
Hottz, ED et al. (2013) Dengue induces platelet activation, mitochondrial dysfunction and cell death through mechanisms that involve DC-SIGN and caspases. Journal of Thrombosis & Haemostasis 11, 951962.CrossRefGoogle Scholar
Heijnen, H and van der Sluijs, P (2015) Platelet secretory behaviour: as diverse as the granules … or not? Journal of Thrombosis & Haemostasis 13, 21412151.CrossRefGoogle Scholar
Smith, CW (2022) Release of alpha-granule contents during platelet activation. Platelets 33, 491502.CrossRefGoogle ScholarPubMed
Gear, AR and Camerini, D (2003) Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense. Microcirculation (New York, N.Y.: 1994) 10, 335350.CrossRefGoogle Scholar
Fox, JM et al. (2018) CXCL4/platelet factor 4 is an agonist of CCR1 and drives human monocyte migration. Scientific Reports 8, 9466.CrossRefGoogle Scholar
Offermanns, S (2006) Activation of platelet function through G protein-coupled receptors. Circulation Research 99, 12931304.CrossRefGoogle Scholar
Gachet, C (2012) P2y(12) receptors in platelets and other hematopoietic and non-hematopoietic cells. Purinergic Signalling 8, 609619.CrossRefGoogle ScholarPubMed
Chen, WF et al. (2013) Platelet protease-activated receptor (PAR)4, but not PAR1, associated with neutral sphingomyelinase responsible for thrombin-stimulated ceramide-NF-kappaB signaling in human platelets. Haematologica 98, 793801.CrossRefGoogle Scholar
Marcinkowska, M et al. (2022) Exploring the antiplatelet activity of serotonin 5-HT(2A) receptor antagonists bearing 6-fluorobenzo[d]isoxazol-3-yl)propyl) motif as potential therapeutic agents in the prevention of cardiovascular diseases. Biomedicine & Pharmacotherapy 145, 112424.CrossRefGoogle ScholarPubMed
Tanaka, KA and Bolliger, D (2019) Transfusion and coagulation therapy. In Hemmings, HC and Egan, TD (eds), Pharmacology and Physiology for Anesthesia: Foundations and Clinical Application, 2nd Edn. USA: Elsevier, pp. 849869.CrossRefGoogle Scholar
Grozovsky, R, Hoffmeister, KM and Falet, H (2010) Novel clearance mechanisms of platelets. Current of Opinion Hematology 17, 585589.CrossRefGoogle ScholarPubMed
Quach, ME, Chen, W and Li, R (2018) Mechanisms of platelet clearance and translation to improve platelet storage. Blood 131, 15121521.CrossRefGoogle Scholar
Hou, Y et al. (2015) Platelets in hemostasis and thrombosis: novel mechanisms of fibrinogen-independent platelet aggregation and fibronectin-mediated protein wave of hemostasis. Journal of Biomedical Research 29, 437444.Google ScholarPubMed
Kim, OV et al. (2019) Fatal dysfunction and disintegration of thrombin-stimulated platelets. Haematologica 104, 18661878.CrossRefGoogle Scholar
Li, C, Li, J and Ni, H (2020) Crosstalk between platelets and microbial pathogens. Frontiers in Immunology 11, 1962.CrossRefGoogle Scholar
Fard, MB et al. (2021) Thrombosis in COVID-19 infection: role of platelet activation-mediated immunity. Thrombosis Journal 19, 59.CrossRefGoogle ScholarPubMed
Quirino-Teixeira, AC et al. (2020) Inflammatory signaling in dengue-infected platelets requires translation and secretion of nonstructural protein 1. Blood Advances 4, 20182031.CrossRefGoogle ScholarPubMed
Garcia-Larragoiti, N et al. (2021) Platelet activation and aggregation response to dengue virus nonstructural protein 1 and domains. Journal of Thrombosis & Haemostasis 19, 25722582.CrossRefGoogle ScholarPubMed
Pan, P et al. (2019) Dengue virus infection activates interleukin-1beta to induce tissue injury and vascular leakage. Frontiers in Microbiology 10, 2637.CrossRefGoogle ScholarPubMed
Alen, MM et al. (2012) Crucial role of the N-glycans on the viral E-envelope glycoprotein in DC-SIGN-mediated dengue virus infection. Antiviral Research 96, 280287.CrossRefGoogle ScholarPubMed
Lien, TS et al. (2021) Exposure of platelets to dengue virus and envelope protein domain III induces NLRP3 inflammasome-dependent platelet cell death and thrombocytopenia in mice. Frontiers in Immunology 12, 616394.CrossRefGoogle ScholarPubMed
Trugilho, MRO et al. (2017) Platelet proteome reveals novel pathways of platelet activation and platelet-mediated immunoregulation in dengue. PLoS Pathogen 13, e1006385.CrossRefGoogle Scholar
Sung, PS, Huang, TF and Hsieh, SL (2019) Extracellular vesicles from CLEC2-activated platelets enhance dengue virus-induced lethality via CLEC5A/TLR2. Nature Communication 10, 2402.CrossRefGoogle Scholar
Garishah, FM et al. (2021) Neutrophil extracellular traps in dengue are mainly generated NOX-independently. Frontiers in Immunology 12, 629167.CrossRefGoogle ScholarPubMed
Cui, L et al. (2016) Serum metabolomics reveals serotonin as a predictor of severe dengue in the early phase of dengue fever. PLoS Neglected Tropical Diseases 10, e0004607.CrossRefGoogle ScholarPubMed
de Mast, Q and de Groot, PG (2019) Serotonin, key to thrombocytopenia in dengue? Blood 133, 22492250.CrossRefGoogle Scholar
Koupenova, M, Livada, AC and Morrell, CN (2022) Platelet and megakaryocyte roles in innate and adaptive immunity. Circulation Research 130, 288308.CrossRefGoogle ScholarPubMed
Teo, A et al. (2023) Understanding antibody-dependent enhancement in dengue: are afucosylated IgG1s a concern? PLoS Pathogen 19, e1011223.CrossRefGoogle Scholar
Rodenhuis-Zybert, IA et al. (2010) Immature dengue virus: a veiled pathogen? PLoS Pathogen 6, e1000718.CrossRefGoogle ScholarPubMed
Wang, TT et al. (2017) IgG antibodies to dengue enhanced for FcgammaRIIIA binding determine disease severity. Science (New York, N.Y.) 355, 395398.CrossRefGoogle Scholar
Bournazos, S et al. (2021) Antibody fucosylation predicts disease severity in secondary dengue infection. Science (New York, N.Y.) 372, 11021105.CrossRefGoogle ScholarPubMed
Alonzo, MT et al. (2012) Platelet apoptosis and apoptotic platelet clearance by macrophages in secondary dengue virus infections. Journal of Infectious Disease 205, 13211329.CrossRefGoogle ScholarPubMed
Hottz, E et al. (2011) Platelets in dengue infection. Drug Discovery Today: Disease Mechanisms 8, e33–e8.Google Scholar
Briggs, C et al. (2004) Assessment of an immature platelet fraction (IPF) in peripheral thrombocytopenia. British Journal Haematology 126, 9399.CrossRefGoogle Scholar
Jeon, K et al. (2020) Immature platelet fraction: a useful marker for identifying the cause of thrombocytopenia and predicting platelet recovery. Medicine (Baltimore) 99, e19096.CrossRefGoogle ScholarPubMed
Dadu, T et al. (2014) Evaluation of the immature platelet fraction as an indicator of platelet recovery in dengue patients. International Journal of Lab Hematology 36, 499504.CrossRefGoogle ScholarPubMed
Looi, KW et al. (2021) Evaluation of immature platelet fraction as a marker of dengue fever progression. International Journal of Infectious Diseases 110, 187194.CrossRefGoogle ScholarPubMed
Yasuda, I et al. (2021) Unique characteristics of new complete blood count parameters, the immature platelet fraction and the immature platelet fraction count, in dengue patients. PLoS ONE 16, e0258936.CrossRefGoogle ScholarPubMed
Figure 0

Figure 1. Classification of dengue by the WHO. This information is based on 2009 WHO guideline (Ref. 1). The present work focuses on one of the symptoms of dengue with warning signs which is thrombocytopenia.

Figure 1

Table 1. Factors in platelet activation and aggregation

Figure 2

Figure 2. Summary of DENV-mediated thrombocytopenia. DENV causes thrombocytopenia in several ways. (1) DENV impairs megakaryopoiesis. DENV infects and causes apoptosis of megakaryocytes and the progenitor cells. DENV also prevents megakaryocyte maturation. (2) DENV impairs thrombopoiesis by reducing megakaryocytes and interfering with platelet formation. (3) Secondary DENV infection causes elevated platelet activation via IgG–FcR connection in the ADE process. (4) DENV increases platelet activation. (5) DENV causes cytokines, chemokines and other factors to be released from infected platelets and nearby endothelial cells. (6) DENV infects and causes platelet apoptosis. (7) ADE, elevated platelet activation and secretion of various factors cause the platelets to coagulate. (8) Coagulated platelets and apoptotic platelets are cleared from the circulation by phagocytes. Regular arrows (→) in green indicate stimulatory modifications. Blunt-ended arrows (˧) in red indicate inhibitory modifications.