Skip to main content

MINI REVIEW article

Front. Cell. Infect. Microbiol., 30 September 2020
Sec. Microbes and Innate Immunity
This article is part of the Research Topic Emerging Concepts in Dengue Pathogenesis and Host Innate Immune Response View all 12 articles

Role of Platelet Cytokines in Dengue Virus Infection

  • 1Disease Biology Laboratory, Regional Center for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, India
  • 2School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, India

Platelets are anucleated blood cells derived from bone marrow megakaryocytes and play a crucial role in hemostasis and thrombosis. Platelets contain specialized storage organelles, called alpha-granules, contents of which are rich in cytokines such as C-X-C Motif Chemokine Ligand (CXCL) 1/4/7, (C-C motif) ligand (CCL) 5/3, CXCL8 (also called as interleukin 8, IL-8), and transforming growth factor β (TGF-β). Activation of platelets lead to degranulation and release of contents into the plasma. Platelet activation is a common event in many viral infections including human immunodeficiency virus (HIV), H1N1 influenza, Hepatitis C virus (HCV), Ebola virus (EBV), and Dengue virus (DENV). The cytokines CXCL8, CCL5 (also known as Regulated on Activation, Normal T Expressed and Secreted, RANTES), tumor necrosis factor α (TNF-α), CXCL1/5 and CCL3 released, promote development of a pro-inflammatory state along with the recruitment of other immune cells to the site of infection. Platelets also interact with Monocytes and Neutrophils and facilitate their activation to release different cytokines which further enhances inflammation. Upon activation, platelets also secrete factors such as CXCL4 (also known as platelet factor, PF4), CCL5 and fibrinopeptides which are critical regulators of replication and propagation of several viruses in the host. Studies suggest that CXCL4 can both inhibit as well as enhance HIV1 infection. Data from our lab show that CXCL4 inhibits interferon (IFN) pathway and promotes DENV replication in monocytes in vitro and in patients significantly. Inhibition of CXCL4 mediated signaling results in increased IFN production and suppressed DENV and JEV replication in monocytes. In this review, we discuss the role of platelets in viral disease progression with a focus on dengue infection.

Dengue Infection

Dengue is one of the most prevalent arboviral diseases (Simmons et al., 2012) affecting ~390 million people around the globe (Bhatt et al., 2013) causing an estimated 20000 deaths each year (Simon et al., 2015). Mainly this virus affects the tropical and subtropical regions of the world (Wilder-Smith et al., 2019). India accounts for the largest number of dengue cases, with ~33 million apparent and another 100 million asymptomatic infections occurring annually (Bhatt et al., 2013). Since the 1990s, dengue epidemics have become recurrent in several parts of India, at a rate of 34.81 per million of the population in 2010–2014. Thus, indicating that the number of dengue cases has increased markedly in recent times (Mutheneni et al., 2017). The principle vectors for transmission of the disease are mosquitoes Aedes aegypti and Aedes albopictus (Kraemer et al., 2015). Whereas, non-vector transmission of the virus is also possible through blood transfusion, organ transplantation, needle stick injuries and mucosal splashes (Wilder-Smith et al., 2009; Busch et al., 2016; Sabino et al., 2016). Dengue virus (DENV) is a Flavivirus that belongs to the family Flaviviridae and has four serotypes, DENV1–4. The virus genome consists of a single-strand of positive-polarity RNA which codes for three structural proteins (capsid C, membrane M, and the envelope E) and seven non-structural proteins (NS1, NS2A, NS2B, NS3, NS4A, NS4B, NS5) (Noble and Shi, 2012). The clinical features of the disease range from asymptomatic infection, undifferentiated febrile illness (dengue fever) to severe complication including Dengue Haemorrhagic Fever (DHF) and Dengue Shock Syndrome (DSS), characterized by plasma leakage and coagulopathy (Kalayanarooj, 2011; Simmons et al., 2012). The pathogenesis of severe dengue is the result of a complex interaction between viral and host factors. The four DENV serotypes (DENV 1–4) share homology, but each serotype possesses specific immunoreactivity. Primary infection with one DENV serotype increases the risk of severe dengue upon secondary infection with other DENV serotype. This is because the sub-neutralizing cross-reactive antibodies opsonize mature and immature virus particles, causing infection of mononuclear phagocytes via Fc-receptor (FcR) thus enhancing DENV infection, termed as antibody dependent enhancement (ADE) (Littaua et al., 1990; Dejnirattisai et al., 2010; Schmidt, 2010; Guzman et al., 2013). This is characterized by persistent high fever, thrombocytopenia, event of DHF or DSS and elevated IgG/IgM ratio (Innis et al., 1989; Shu et al., 2003; Prince et al., 2011; Cucunawangsih and Kurniawan, 2015). Complement activation by virus-antibody complexes and T-cell mediated immune response have also been reported in the progression of secondary dengue infection. Independent of primary or secondary infection, platelet activation is the hallmark of dengue infection. Thrombocytopenia (decrease in platelet count) is another common symptom of dengue infection and is associated with hemodynamic instability and progression in severity of dengue fever (Krishnamurti et al., 2001; Schexneider and Reedy, 2005; Mourão et al., 2007; Bozza et al., 2008).

Platelet Functions

The primary and most important role of platelets is to maintain hemostasis (Clemetson, 2012). Platelets are mediators of thrombosis too (Kaplan and Jackson, 2011). Besides, platelets also contribute to non-canonical functions such as immune modulation, atherogenesis, tissue repair and regeneration, angiogenesis, and metastasis (Lindemann et al., 2007; Nurden, 2011; Semple et al., 2011; Eisinger and Langer, 2018; Koupenova et al., 2018; Schlesinger, 2018). Platelets mediate innate immune response and contribute to antimicrobial activity either by releasing antimicrobial proteins or by modulating immune response of other immune cells such as neutrophils and monocytes (Flad and Brandt, 2010). Platelet dysfunction can lead to impaired immune or inflammatory responses and tissue damage. During infection, platelet activation can lead to serious pathophysiological conditions such as Infective Endocarditis and Disseminated Intravascular Coagulation (Beynon et al., 2006; Kitchens, 2009). Platelet activation followed by thrombocytopenia is found to be associated with patients of several infections and sepsis (Akca et al., 2002; Claushuis et al., 2016; Levi, 2016). Platelet activation is the hallmark of many viral infections including dengue, HIV, HCV, H1N1, and Ebola (Geisbert et al., 2003; Chaipan et al., 2006; Afdhal et al., 2008; Assinger et al., 2014; Ojha et al., 2017, 2019).

Platelet Granules and Cytokines

Platelets contain α and dense granules, also known as storage granules. These granules store huge numbers of proteins, RNAs, antimicrobial peptides and growth factors (Manne et al., 2017). α granules are ~500 nm in diameter, round in shape and around 50 granules are present in each platelet (Yadav and Storrie, 2017). Different cytokines, growth factors and antimicrobial peptides present are listed in Table 1. Upon platelet activation the α-granules release these contents into the plasma (Jonnalagadda et al., 2012). The dense granules on the other hand are smaller in size and contain Adenosine nucleotides (ATP, ADP), serotonin, calcium, magnesium and pyrophosphate (Rendu and Brohard-Bohn, 2001).

TABLE 1
www.frontiersin.org

Table 1. Contents of platelet granules.

Immune Function of Platelets

Platelets are modulators of innate immunity and possess both surface and intracellular receptors for detection of pathogens in the bloodstream. Receptors include Toll-like receptors (TLRs), NOD-like receptors (NLR), C-type Lectins and integrins for detection of pathogen associated molecular patterns (PAMPs), and Fc receptors and complement receptors for antibody opsonized pathogens (McDonald and Dunbar, 2019). Platelets express TLR 1–4, 6, 7, and 9 (Hamzeh-Cognasse et al., 2018; McDonald and Dunbar, 2019). On activation the TLRs promote secretion of interferons and proinflammatory cytokines such as IL-6, CXCL8, TNF-α, and CCL5 which further activate other cells resulting in increased inflammation. CD40L, expressed on platelets as a result of activation (Henn et al., 1998), mediates its interaction with CD40 present on B cells, Monocytes, Dendritic cells, Macrophages, endothelial cells and modulate their activity. CD40 and CD40L interaction leads to recruitment of TNF receptor associated factors (TRAFs) which further lead to activation of canonical and non-canonical NF-kB pathways (Bishop et al., 2007). Platelets via CD40L induce B cell isotype switching and augment CD8+ T cell response (Elzey et al., 2003). Activated platelets cause Dendritic cell maturation and activation by direct surface contact through CD40L or through soluble effectors (Czapiga et al., 2004; Hagihara et al., 2004; Martinson et al., 2004). In Dendritic cells CD40 expression tends to increase the expression of MHC class II along with essential co-stimulatory molecules such as CD58, CD80, and CD86. This further leads to improved T cell activation by better presentation of the antigen. Platelets possess an active proteasome and can process and present exogenous antigens which has been shown both in vitro and in vivo using Experimental Cerebral Malaria (ECM) mouse model (Chapman et al., 2012). An increase in surface expression of HLA class 1 has been observed on platelets obtained from patients with Dengue infection (Trugilho et al., 2017). However, the actual mode of presentation of DENV antigen still needs to be investigated.

Recently it has been shown that platelets can differentiate between bacterial LPS isoforms and as a result between pathogens (Berthet et al., 2012). This helps in providing a pathogen specific response by secreting different cytokines. It has been shown that platelets upon activation with thrombin trigger complement activation. Complement factor C3b bound to bacteria gets detected by GPIb receptor on platelet surface resulting in phagocytosis of the bacteria-platelet complex (Verschoor et al., 2011). Platelets also respond to Damage Associated Molecular Patterns (DAMPs) (Fuchs et al., 2011).

Platelets get activated upon interacting with pathogens directly through their surface receptors or indirectly through plasma proteins such as fibrinogen, vWF, complement and IgG (Fitzgerald et al., 2006; Cox et al., 2011). Upon activation by bacterial pathogens platelets release antimicrobial substances such as ROS (reactive oxygen species), antimicrobial peptides, defensins, thrombocidins and proteases (Suzuki et al., 2001; Tang et al., 2002; Trier et al., 2008; Wiesner and Vilcinskas, 2010; Yeaman, 2014). Platelets support phagocytosis and intracellular killing of bacteria by Neutrophils and release of Neutrophil Extracellular Traps (NETs) (Miyabe et al., 2004; Assinger et al., 2011; Kim and Jenne, 2016). In disease conditions such as malaria, platelets have a direct lethal effect on all major Plasmodium species (Kho et al., 2018). Platelets bind with erythrocytes and cause killing of intraerythrocytic Plasmodium along with intracellular accumulation of platelet CXCL4 (McMorran et al., 2012). In patients with mycobacterium tuberculosis, infected platelets along with other immune cells are responsible for pulmonary inflammation and tissue damage leading to morbidity and spread of infection (Fox et al., 2018). Thus, platelets help to fight infection but at times can also help in disease progression.

Immune Response of Platelets During Viral Infections

CXCL4 is highly abundant in platelets. CXCL4 can associate with CCL5 to modulate monocyte functions. CCL3, CCL5, CCL7, CCL17, CXCL1, CXCL5, and CXCL8 are some of the chemokines among the whole pool of chemokines released by platelets which attract leukocytes causing further activation of platelets (Gear and Camerini, 2003). A comparative study reports that CXCL4 in conjunction with M-CSF supports HIV-1 replication in immune cells including monocytes and macrophages. It was found that CXCL4 derived macrophages can be infected with macrophage-tropic HIV-1 that uses either CCR5 or CXCR4 as a co-receptor for viral entry. CXCL4 increases HIV-1 replication in M-CSF-derived human macrophages after virus adsorption on to the cells (Schwartzkopff et al., 2009). CXCL4 has also been identified as a broad spectrum inhibitor of HIV-1 (Auerbach et al., 2012). In another study, it was found that CXCL4 inhibitory effect lies in a concentration range where it exists in monomeric form. As a monomer it binds with the viral envelope protein resulting in inhibition of HIV-1 attachment to the cell surface. But oligomerization initiated upon increasing concentration; the tetrameric or higher-order forms promoted viral replication in vitro (Parker et al., 2016). CXCL4 and beta thromboglobulin (β-TG) have been reported as prognostic markers in Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) infections. A decreased serum CXCL4 and increased β-TG were reported in the study (Poon et al., 2012). CXCL4 is very well-known for its regulatory functions in immune response and inflammation. Role of CXCL4 has been studied in pulmonary influenza infection, and it was found that it helps in protecting the mice from H1N1 virus infection (Guo et al., 2017). CXCL4 up-regulated in HCV induces liver fibrosis both in vitro and in vivo (Zaldivar et al., 2010). CXCL4 also increases replication and propagation of dengue and Japanese encephalitis viruses (JEV) in monocytes by downregulating the type-I interferon production, thus leading to increased viral load (Ojha et al., 2019).

Platelet derived CCL5 and CCL3 have been reported as major HIV-suppressive factors (Cocchi et al., 1995). CCL5 is also reported as inhibitor of Influenza A (Wareing et al., 2004) and HCV infection (Katsounas et al., 2011). CCL5 is reported to be involved in viral lung diseases (Culley et al., 2006). CCL17 is released by activated platelets and functions in further platelet activation in an autocrine manner by binding to its receptor CCR4 present on platelets (Gear and Camerini, 2003). A recent study reported that co-culture of monocyte derived dendritic cells (MoDCs) and HCV infected cells lead to the expression of CCL17 and CCL22 which attract regulatory T cells at the site of infection (Riezu-Boj et al., 2011). Co-culture of MoDCs with HBV transfected cells induces the expression of CCL17 and CCL22 which recruits IL-17 secreting T-cells (Zhang et al., 2020).

Immune Response of Platelets to Dengue Infection

Dengue Infection in Platelets

The presence of DENV has been reported in circulating platelets of dengue patients (Noisakran et al., 2009). Platelet activation and thrombocytopenia are the hallmarks of dengue infection. Platelet activation results in the release of the granular constituents. Platelets from dengue patients present signs of activation, mitochondrial dysfunction and activation of apoptosis caspase cascade resulting in thrombocytopenia. Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) also termed as CD209 has been shown as a critical receptor involved in DENV-dependent platelet activation (Hottz et al., 2013b). DENV directly binds to the surface receptors DC-SIGN, heparan sulfate proteoglycan (HSP) and CLEC-2 (C-type-lectin-like receptor 2) on platelets and replicates inside the platelets (Simon et al., 2015; Sung and Hsieh, 2019). One of the findings suggests that platelets attach to the histones (H2A) present in systemic circulation of the dengue patients, which lead to their activation (Trugilho et al., 2017). Platelets have specialized FcγRII receptors for binding of immunoglobulin coated virus particles. Antibody dependent enhancement of DENV occurs via these FcγRII receptors on platelets (Wang et al., 1995) upon secondary dengue infection. This leads to platelet activation and thrombocytopenia which results into severe dengue diseases, DHF, and DSS.

Immunoregulation of Dengue Infection by Platelets

Platelets are the key players involved in immunoregulation of dengue disease as there is platelet activation upon dengue infection and release of the granular contents. Cytokines are released from alpha granules, and have been shown to play regulatory roles in dengue virus infection. The CCR1-CCL2 axis plays an important role in the pathogenesis of dengue infection whereas CCR1-CCL5 axis was found to show a protective role in dengue infection (Sierra et al., 2014). A study has suggested that low levels of CCL5 and high levels of CXCL8 during early dengue infection could serve as a marker for severe dengue disease (Patra et al., 2019). Upon DENV infection there is activation of endothelial cells leading to increased expression of E-selectin on the endothelial cells. E-selectin and P-selectin helps in platelet adhesion to endothelial cells (Krishnamurti et al., 2002). P-selectin expressed on the surface of activated platelets promote interaction of platelets with monocytes and neutrophils leading to aggregate formation (Onlamoon et al., 2010). The endothelial cells upon dengue infection secrete CXCL8, IL-6, CXCL10, CXCL11, and CCL5 (Avirutnan et al., 1998; Dalrymple and Mackow, 2012; Kelley et al., 2012). Platelets also secrete IL-1β, CXCL8, and CCL5 and contribute to the total cytokine pool. These cytokines together help in increasing the vascular permeability and possess chemoattractant properties which progress inflammation and plasma leakage in vivo thus leading to dengue disease severity (Kelley et al., 2012).

Increased platelet monocyte aggregates have been reported in dengue patients having thrombocytopenia and increased vascular permeability. Platelet binding modulates cytokine release by monocytes in dengue infection. Interaction of platelets isolated from dengue patients with monocytes from healthy individuals lead to the synthesis and secretion of IL-1β, CXCL8, and IL-10 by the monocytes. Also interaction of monocytes with apoptotic platelets leads to secretion of IL-10 through P selectin and phosphatidylserine recognition in platelet-monocyte aggregates. IL-10 secretion requires platelet-monocyte contact but not phagocytosis of platelets by the monocytes. Activated and apoptotic platelets aggregate with monocytes during dengue infection and cause specific cytokine responses contributing to the pathogenesis of dengue. The cytokines IL-1β, CXCL8, and IL-10, released by monocytes in response to interactions with platelets from dengue patients, are frequently observed to be increased in plasma of severe dengue patients (Hottz et al., 2014).

Vascular leakage is one of the hallmarks of DHF, and an increased CCL2 level is reported in DHF. CCL2 leads to disrupted distribution of tight junction protein on the cell membrane of human umbilical vein endothelial cells (HUVEC) leading to increased vascular permeability (Lee et al., 2006). Platelets release IL-1β upon DENV infection by recruiting nucleotide-binding domain leucine rich repeat containing protein (NLRP3) inflammasome and caspase-1 which is highly correlated with increased vascular permeability and activation of innate immunity in dengue infection (Hottz et al., 2013a, 2014; Guabiraba and Ryffel, 2014).

Macrophage migration inhibitory factor (MIF) is a pleiotropic proinflammatory cytokine that mediates several immune responses, serum levels of MIF, IL-6, and IL-10 are reported to be higher in DHF patients as compared to DF patients (Chen et al., 2006). MIF is secreted mainly by T cells, and also by other immune and non-immune cells such as macrophages, endothelial cells, epithelial cells, neutrophils and monocytes (Lai et al., 2020). It is also secreted by platelets (Wirtz et al., 2015). Mif–/– mice show lesser DENV induced inflammation, thrombocytopenia and viral load suggesting it as one of the crucial cytokines involved in dengue pathogenesis (Assunção-Miranda et al., 2010). MIF helps in enhancing dengue viral replication in host cells, it also causes endothelial hyperpermeability upon DENV infection, and also modulates the functions of immune cells. MIF inhibition during dengue infection also leads to decreased TNF-α and IL-6 production which otherwise lead to vascular hyperpermeability. MIF release by neutrophils leads to neutrophil extracellular trap (NET) formation and inflammation which further helps in dengue pathogenesis. It can also be a potential therapeutic target against dengue infection (Lai et al., 2020). In comparison with DENV infection, secretory NS1 protein released upon DENV infection and exogenous NS1 protein stimulation also leads to a partial inflammatory phenotype in platelets. Similar to infected platelets, NS1-stimulated platelets also release the stored cytokines/chemokines CXCL4, CCL5, and MIF but, in contrast, they do not secrete IL-1β. NS1 induces pro-IL-1β synthesis in platelets but does not induce caspase-1 activation for IL-1β processing and secretion, which occurs upon classical DENV infection providing all necessary signals for IL-1β synthesis, caspase-1 activation, and IL-1β release. There is increased caspase-1 activation in platelets stimulated with either ATP, NS1, and ATP, or DENV, but not in platelets stimulated with NS1 alone (Quirino-Teixeira et al., 2020). DENV activated platelets deliver inflammatory signals to monocytes leading to secretion of CXCL8, IL-10, and IL-6, and also show increased accumulation of lipid droplets (LD) in the monocytes after 18 h of interaction. Stimuli-specific-activated platelets can cause phenotypic changes and metabolic reprogramming in monocytes. Activated platelets exposed to DENV in vitro form aggregates with monocytes and signal to LD formation and CXCL8, IL-10, CCL2 and prostaglandin E2 (PGE2) secretion. Pharmacologic inhibition of LD biogenesis prevents PGE2 secretion, but not CXCL8 release, by platelet-monocyte complexes. Mechanistically LD formation in monocytes exposed to DENV-activated platelets is partially dependent on platelet-produced MIF. LD formation is higher in monocytes, which have platelets adhered on their surface, suggesting the importance of adhesion besides paracrine signaling. Activated platelets aggregate with monocytes during DENV infection and lead to LD biogenesis and release of inflammatory mediators (Barbosa-Lima et al., 2020).

CXCL4 in Dengue Infection

CXCL4 is the most abundant chemokine present in platelet α granules, released upon platelet activation (Gleissner et al., 2008). A quantitative proteomic study has investigated the protein content of platelets in dengue patient samples and healthy controls. In dengue patients, activated platelets release a significant amount of CXCL4 in plasma as compared to healthy volunteers (Trugilho et al., 2017). CXCL4 has been identified as a prognostic tool for classifying acute and severe dengue patients (Fragnoud et al., 2015). A comparative study between dengue and leptospirosis (caused by Leptospira bacteria) revealed that out of the 19 biomarkers assessed CXCL4 was the one higher in dengue fever compared to leptospirosis (Conroy et al., 2014). A recent work from our lab also showed that CXCL4 is one of the abundant proteins present in blood plasma upon dengue infection. It also helps in propagating the virus inside monocytes in vitro. Binding of CXCL4 to CXCR3 leads to increased phosphorylation of p38-MAPK and diminished activation of STAT-2 and IRF-9. This decreases the synthesis and secretion of IFN-α by the DENV2 infected monocytes, resulting in 3–4-fold increase in virus replication. Also, blocking CXCL4 using neutralizing CXCL4 antibodies or its receptor CXCR3 through inhibitor AMG487 reversed the above signaling pathway and significantly restored the IFN-α production, thus inhibiting the DENV propagation in monocytes. The study also showed a decrease in the levels of proinflammatory cytokines TNF-α, IL-1β, and IL-6 in monocytes upon treatment of the cells with rhCXCL4, which was rescued upon treating the cells with anti-CXCL4 antibody or antagonist AMG487, concluding that CXCL4 significantly impacts dengue virus replication (Ojha et al., 2019).

Therapy for Dengue

Till date, there is no specific treatment for Dengue infection. Although few studies describe the development of drugs targeting host factors required for DENV propagation. Chloroquine (CQ) has immunomodulatory effects by suppressing release of TNF-α and IL-6 (Tricou et al., 2010). Although the in vitro study showed some promising results but a randomized trial in patients did not show significant reduction in the development of DHF (Savarino et al., 2003). Another drug, Celgosivir is an alkaloid castanospermine derived from the Moreton Bay Chestnut tree showed some inhibitory effects on all four serotypes (Durantel, 2009). Celgosivir was tested in Phase I and II trials as a possible treatment for HIV and hepatitis C infection and was found to be safe (Durantel, 2009; Rathore et al., 2011). In a recent study we have shown that inhibition of CXCL4-CXCR3 interaction by an antagonist AMG487 significantly reduced the replication of DENV and JEV, since both viruses use CXCL4 for their rapid replication in host immune cells in vitro. The AMG487 treatment reduced the JEV infection in mice and increased the mice survivability suggesting the CXCL4-CXCR3 axis as the therapeutic target for prophylaxis of dengue (Ojha et al., 2019).

IFNs play major antiviral roles. IFN-α/β have proved to be of considerable value in some chronic virus infections, particularly hepatitis and papillomavirus infections (Finter, 1994). A study suggests the anti-viral activity of IFN-α and ribavirin as a combination therapy against DENV (Piresde Mello et al., 2018). Studies also suggest that manipulating IL-10, an anti-inflammatory cytokine may serve as an effective antiviral treatment in addition to the development of a safe dengue vaccine (Tsai et al., 2013).

A live attenuated vaccine known as Dengvaxia, developed by Sanofi, was approved by the Food and Drug Administration (FDA) in the United States licensed in May 2019, for use in children 9–16 years old living in an area where dengue is common such as US territories Samoa, Puerto Rico and Virgin Islands. Another live attenuated vaccine, named TetraVax-DV, developed by Butantan, is currently in clinical trial phase 3 in Brazil (Durbin et al., 2011). The Dengue tetravalent vaccine, developed by Panacea Biotec Ltd, is in phase 2 trial in India (TDB Panacea Agreement, 2017).

Author Contributions

PG has conceptualized the approach. All authors wrote the article.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

Afdhal, N., McHutchison, J., Brown, R., Jacobson, I., Manns, M., and Poordad, F. (2008). Thrombocytopenia associated with chronic liver disease. J. Hepatol. 48, 1000–1007. doi: 10.1016/j.jhep.2008.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Akca, S., Haji-Michael, P., de Mendonça, A., Suter, P., Levi, M., and Vincent, J. L. (2002). Time course of platelets counts in critically ill patients. Crit. Care. Med. 30, 753–756. doi: 10.1097/00003246-200204000-00005

PubMed Abstract | CrossRef Full Text | Google Scholar

Assinger, A., Kral, J. B., Yaiw, K. C., Schrottmaier, W. C., Kurzejamska, E., Wang, Y., et al. (2014). Human cytomegalovirus-platelet interaction triggers toll-like receptor 2-dependent proinflammatory and proangiogenic responses. Arterioscler. Thromb. Vasc. 34, 801–809. doi: 10.1161/ATVBAHA.114.303287

PubMed Abstract | CrossRef Full Text | Google Scholar

Assinger, A., Laky, M., Schabbauer, G., Hirschl, A. M., Buchberger, E., Binder, B. R., et al. (2011). Efficient phagocytosis of periodontopathogens by neutrophils requires plasma factors, platelets and TLR2. J. Thromb. Haemost. 9, 799–809. doi: 10.1111/j.1538-7836.2011.04193.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Assunção-Miranda, I., Amaral, F. A., Bozza, F. A., Fagundes, C. T., Sousa, L. P., Souza, D. G., et al. (2010). Contribution of macrophage migration inhibitory factor to the pathogenesis of dengue virus infection. FASEB. J. 24, 218–228. doi: 10.1096/fj.09-139469

PubMed Abstract | CrossRef Full Text | Google Scholar

Auerbach, D. J., Lin, Y., Miao, H., Cimbro, R., Difiore, M. J., Gianolini, M. E., et al. (2012). Identification of the platelet-derived chemokine CXCL4/PF4 as a broad-spectrum HIV-1 inhibitor. Proc. Natl. Acad. Sci. U.S.A. 109, 9569–9574. doi: 10.1073/pnas.1207314109

PubMed Abstract | CrossRef Full Text | Google Scholar

Avirutnan, P., Malasit, P., Seliger, B., Bhakdi, S., and Husmann, M. (1998). Dengue virus infection of human endothelial cells leads to chemokine production, complement activation, and apoptosis. J. Immunol. 161, 6338–6346.

PubMed Abstract | Google Scholar

Barbosa-Lima, G., Hottz, E. D., de Assis, E. F., Liechocki, S., Souza, T., Zimmerman, G. A., et al. (2020). Dengue virus-activated platelets modulate monocyte immunometabolic response through lipid droplet biogenesis and cytokine signaling. J. Leukoc. Biol. 1–14. doi: 10.1002/JLB.4MA0620-658R

PubMed Abstract | CrossRef Full Text | Google Scholar

Berthet, J., Damien, P., Hamzeh-Cognasse, H., Arthaud, C. A., Eyraud, M. A., Zéni, F., et al. (2012). Human platelets can discriminate between various bacterial LPS isoforms via TLR4 signaling and differential cytokine secretion. Clin. Immunol. 145, 189–200. doi: 10.1016/j.clim.2012.09.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Beynon, R. P., Bahl, V. K., and Prendergast, B. D. (2006). Infective endocarditis. BMJ 333, 334–339. doi: 10.1136/bmj.333.7563.334

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatt, S., Gething, P. W., Brady, O. J., Messina, J. P., Farlow, A. W., Moyes, C. L., et al. (2013). The global distribution and burden of dengue. Nature 496, 504–507. doi: 10.1038/nature12060

PubMed Abstract | CrossRef Full Text | Google Scholar

Bishop, G. A., Moore, C. R., Xie, P., Stunz, L. L., and Kraus, Z. J. (2007). TRAF proteins in CD40 signaling. Adv. Exp. Med. Biol. 597, 131–151. doi: 10.1007/978-0-387-70630-6_11

PubMed Abstract | CrossRef Full Text | Google Scholar

Blair, P., and Flaumenhaft, R. (2009). Platelet alpha-granules: basic biology and clinical correlates. Blood. Rev. 23, 177–189. doi: 10.1016/j.blre.2009.04.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Bozza, F. A., Cruz, O. G., Zagne, S. M., Azeredo, E. L., Nogueira, R. M., Assis, E. F., et al. (2008). Multiplex cytokine profile from dengue patients: MIP-1beta and IFN-gamma as predictive factors for severity. BMC. Infect. Dis. 8, 86–97. doi: 10.1186/1471-2334-8-86

PubMed Abstract | CrossRef Full Text | Google Scholar

Busch, M. P., Sabino, E. C., Brambilla, D., Lopes, M. E., Capuani, L., Chowdhury, D., et al. (2016). Duration of dengue viremia in blood donors and relationships between donor viremia, infection incidence and clinical case reports during a large epidemic. J. Infect. Dis. 214, 49–54. doi: 10.1093/infdis/jiw122

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaipan, C., Soilleux, E. J., Simpson, P., Hofmann, H., Gramberg, T., and Marzi, A. (2006). DC-SIGN and CLEC-2 mediate human immunodeficiency virus type 1 capture by platelets. J. Virol. 80, 8951–8960. doi: 10.1128/JVI.00136-06

PubMed Abstract | CrossRef Full Text | Google Scholar

Chapman, L. M., Aggrey, A. A., Field, D. J., Srivastava, K., Ture, S., Yui, K., et al. (2012). Platelets present antigen in the context of MHC class I. J. Immunol. 189, 916–923. doi: 10.4049/jimmunol.1200580

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, L. C., Lei, H. Y., Liu, C. C., Shiesh, S. C., Chen, S. H., Liu, H. S., et al. (2006). Correlation of serum levels of macrophage migration inhibitory factor with disease severity and clinical outcome in dengue patients. Am. J. Trop. Med. Hyg. 74, 142–147. doi: 10.4269/ajtmh.2006.74.142

PubMed Abstract | CrossRef Full Text | Google Scholar

Claushuis, T. A., van Vught, L. A., Scicluna, B. P., Wiewel, M. A., Klein Klouwenberg, P. M., Hoogendijk, A. J., et al. (2016). Thrombocytopenia is associated with a dysregulated host response in critically ill sepsis patients. Blood 127, 3062–3072. doi: 10.1182/blood-2015-11-680744

PubMed Abstract | CrossRef Full Text | Google Scholar

Clemetson, K. J. (2012). Platelets and primary haemostasis. Thromb. Res. 129, 220–224. doi: 10.1016/j.thromres.2011.11.036

PubMed Abstract | CrossRef Full Text | Google Scholar

Cocchi, F., DeVico, A. L., Garzino-Demo, A., Arya, S. K., Gallo, R. C., and Lusso, P. (1995). Identification of RANTES, MIP-1 alpha, and MIP-1 beta as the major HIV-suppressive factors produced by CD8+T cells. Science 270, 1811–1815. doi: 10.1126/science.270.5243.1811

PubMed Abstract | CrossRef Full Text | Google Scholar

Conroy, A. L., Gélvez, M., Hawkes, M., Rajwans, N., Liles, W. C., Villar-Centeno, L. A., et al. (2014). Host biomarkers distinguish dengue from leptospirosis in Colombia: a case-control study. BMC. Infect. Dis. 14:35. doi: 10.1186/1471-2334-14-35

PubMed Abstract | CrossRef Full Text | Google Scholar

Cox, D., Kerrigan, S. W., and Watson, S. P. (2011). Platelets and the innate immune system: mechanisms of bacterial-induced platelet activation. J. Thromb. Haemost. 9, 1097–1107. doi: 10.1111/j.1538-7836.2011.04264.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Cucunawangsih, L. N. P., and Kurniawan, A. (2015). Immunoglobulin G (IgG) to IgM ratio in secondary adult dengue infection using samples from early days of symptoms onset. BMC. Infect. Dis. 15, 276–281. doi: 10.1186/s12879-015-1022-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Culley, F. J., Pennycook, A. M., Tregoning, J. S., Dodd, J. S., Walzl, G., Wells, T. N., et al. (2006). Role of ccl5 (rantes) in viral lung disease. J. Virol. 80, 8151–8157. doi: 10.1128/JVI.00496-06

PubMed Abstract | CrossRef Full Text | Google Scholar

Czapiga, M., Kirk, A. D., and Lekstrom-Himes, J. (2004). Platelets deliver costimulatory signals to antigen-presenting cells: a potential bridge between injury and immune activation. Exp. Hematol. 32, 135–139. doi: 10.1016/j.exphem.2003.11.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Dalrymple, N. A., and Mackow, E. R. (2012). Endothelial cells elicit immune-enhancing responses to dengue virus infection. J. Virol. 86, 6408–6415. doi: 10.1128/JVI.00213-12

PubMed Abstract | CrossRef Full Text | Google Scholar

Dejnirattisai, W., Jumnainsong, A., Onsirisakul, N., Fitton, P., Vasanawathana, S., Limpitikul, W., et al. (2010). Cross-reacting antibodies enhance dengue virus infection in humans. Science 328, 745–748. doi: 10.1126/science.1185181

PubMed Abstract | CrossRef Full Text | Google Scholar

Durantel, D. (2009). Celgosivir, an alpha-glucosidase I inhibitor for the potential treatment of HCV infection. Curr. Opin. Investig. Drugs 10, 860–s870.

PubMed Abstract | Google Scholar

Durbin, A. P., Kirkpatrick, B. D., Pierce, K. K., Schmidt, A. C., and Whitehead, S. S. (2011). Development and clinical evaluation of multiple investigational monovalent DENV vaccines to identify components for inclusion in a live attenuated tetravalent DENV vaccine. Vaccine 29, 7242–7250. doi: 10.1016/j.vaccine.2011.07.023

PubMed Abstract | CrossRef Full Text | Google Scholar

Eisinger, F., and Langer, H. F. (2018). The mutual relation of platelet activation and innate immunity. Hamostaseologie 38, 186–202. doi: 10.1055/s-0038-1669450

PubMed Abstract | CrossRef Full Text | Google Scholar

Elzey, B. D., Tian, J., Jensen, R. J., Swanson, A. K., Lees, J. R., Lentz, S. R., et al. (2003). Platelet-mediated modulation of adaptive immunity.A communication link between innate and adaptive immune compartments. Immunity 19, 9–19. doi: 10.1016/s1074-7613(03)00177-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Finter, N. B. (1994). Cytokines in the treatment of virus infections. Biotherapy 7, 151–159. doi: 10.1007/BF01878481

PubMed Abstract | CrossRef Full Text | Google Scholar

Fitzgerald, J. R., Foster, T. J., and Cox, D. (2006). The interaction of bacterial pathogens with platelets. Nat. Rev. Microbiol. 4, 445–457. doi: 10.1038/nrmicro1425

PubMed Abstract | CrossRef Full Text | Google Scholar

Flad, H. D., and Brandt, E. (2010). Platelet-derived chemokines: pathophysiology and therapeutic aspects. Cell. Mol. Life. Sci. 67, 2363–2386. doi: 10.1007/s00018-010-0306-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fox, K. A., Kirwan, D. E., Whittington, A. M., Krishnan, N., Robertson, B. D., Gilman, R. H., et al. (2018). Platelets regulate pulmonary inflammation and tissue destruction in tuberculosis. Am. J. Respir. Crit. Care. Med. 198, 245–255. doi: 10.1164/rccm.201710-2102OC

PubMed Abstract | CrossRef Full Text | Google Scholar

Fragnoud, R., Flamand, M., Reynier, F., Buchy, P., Duong, V., Pachot, A., et al. (2015). Differential proteomic analysis of virus-enriched fractions obtained from plasma pools of patients with dengue fever or severe dengue. BMC. Infect. Dis. 15, 518–531. doi: 10.1186/s12879-015-1271-7

CrossRef Full Text | Google Scholar

Fuchs, T. A., Bhandari, A. A., and Wagner, D. D. (2011). Histones induce rapid and profound thrombocytopenia in mice. Blood 118, 3708–3714. doi: 10.1182/blood-2011-01-332676

PubMed Abstract | CrossRef Full Text | Google Scholar

Gear, A. R., and Camerini, D. (2003). Platelet chemokines and chemokine receptors: linking hemostasis, inflammation, and host defense. Microcirculation 10, 335–350. doi: 10.1038/sj.mn.7800198

PubMed Abstract | CrossRef Full Text | Google Scholar

Geisbert, T. W., Hensley, L. E., Jahrling, P. B., Larsen, T., Geisbert, J. B., and Paragas, J. (2003). Treatment of Ebola virus infection with a recombinant inhibitor of factor VIIa/tissue factor: a study in rhesus monkeys. Lancet 362, 1953–1958. doi: 10.1016/S0140-6736(03)15012-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Gleissner, C. A., von Hundelshausen, P., and Ley, K. (2008). Platelet chemokines in vascular disease. Arterioscler. Thromb. Vasc. Biol. 28, 1920–1927. doi: 10.1161/ATVBAHA.108.169417

PubMed Abstract | CrossRef Full Text | Google Scholar

Gremmel, T., Frelinger, A. L. III., and Michelson, A. D. (2016). Platelet physiology. Semin. Thromb. Hemost. 42, 191–204. doi: 10.1055/s-0035-1564835

PubMed Abstract | CrossRef Full Text | Google Scholar

Guabiraba, R., and Ryffel, B. (2014). Dengue virus infection: current concepts in immune mechanisms and lessons from murine models. Immunol 141, 143–156 doi: 10.1111/imm.12188

PubMed Abstract | CrossRef Full Text | Google Scholar

Guo, L., Feng, K., Wang, Y. C., Mei, J. J., Ning, R. T., Zheng, H. W., et al. (2017). Critical role of CXCL4 in the lung pathogenesis of influenza (H1N1) respiratory infection. Mucosal. Immunol. 10, 1529–1541. doi: 10.1038/mi.2017.1

PubMed Abstract | CrossRef Full Text | Google Scholar

Guzman, M. G., Alvarez, M., and Halstead, S. B. (2013). Secondary infection as a risk factor for dengue hemorrhagic fever/dengue shock syndrome: an historical perspective and role of antibody-dependent enhancement of infection. Arch. Virol. 158, 1445–1459. doi: 10.1007/s00705-013-1645-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Hagihara, M., Higuchi, A., Tamura, N., Ueda, Y., Hirabayashi, K., and Ikeda, Y. (2004). Platelets, after exposure to a high shear stress, induce IL-10-producing, mature dendritic cells in vitro. J. Immunol. 172, 5297–5303. doi: 10.4049/jimmunol.172.9.5297

PubMed Abstract | CrossRef Full Text | Google Scholar

Hamzeh-Cognasse, H., Berthelot, P., Tardy, B., Pozzetto, B., Bourlet, T., Laradi, S., et al. (2018). Platelet toll-like receptors are crucial sensors of infectious danger moieties. Platelets 29, 533–540. doi: 10.1080/09537104.2018.1445842

PubMed Abstract | CrossRef Full Text | Google Scholar

Heijnen, H., and van der Sluijs, P. (2015). Platelet secretory behaviour: as diverse as the granules … or not?. J. Thromb. Haemost. 13, 2141–2151. doi: 10.1111/jth.13147

PubMed Abstract | CrossRef Full Text | Google Scholar

Henn, V., Slupsky, J. R., Gräfe, M., Anagnostopoulos, I., Förster, R., and Müller-Berghaus, G. (1998). CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells. Nature. 391, 591-594. doi: 10.1038/35393

PubMed Abstract | CrossRef Full Text | Google Scholar

Hottz, E. D., Lopes, J. F., Freitas, C., Valls-de-Souza, R., Oliveira, M. F., Bozza, M. T., et al. (2013a). Platelets mediate increased endothelium permeability in dengue through NLRP3-inflammasome activation. Blood 122, 3405–3414. doi: 10.1182/blood-2013-05-504449

PubMed Abstract | CrossRef Full Text | Google Scholar

Hottz, E. D., Medeiros-de-Moraes, I. M., Vieira-de-Abreu, A., de Assis, E. F., Vals-de-Souza, R., Castro-Faria-Neto, H. C., et al. (2014). Platelet activation and apoptosis modulate monocyte inflammatory responses in dengue. J. Immunol. 193, 1864–1872. doi: 10.4049/jimmunol.1400091

PubMed Abstract | CrossRef Full Text | Google Scholar

Hottz, E. D., Oliveira, M. F., Nunes, P. C., Nogueira, R. M., Valls-de-Souza, R., Da Poian, A. T., et al. (2013b). Dengue induces platelet activation, mitochondrial dysfunction and cell death through mechanisms that involve DC-SIGN and caspases. J. Throm. Haemost. 11, 951–962. doi: 10.1111/jth.12178

PubMed Abstract | CrossRef Full Text | Google Scholar

Innis, B. L., Nisalak, A., Nimmannitya, S., Kusalerdchariya, S., Chongswasdi, V., Suntayakorn, S., et al. (1989). An enzyme-linked immunosorbent assay to characterize dengue infections where dengue and Japanese encephalitis co-circulate. Am. J. Trop. Med. Hyg. 40, 418–427. doi: 10.4269/ajtmh.1989.40.418

PubMed Abstract | CrossRef Full Text | Google Scholar

Jonnalagadda, D., Izu, L. T., and Whiteheart, S. W. (2012). Platelet secretion is kinetically heterogeneous in an agonist-responsive manner. Blood 120, 5209–5216. doi: 10.1182/blood-2012-07-445080

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalayanarooj, S. (2011). Clinical manifestations and management of dengue/DHF/DSS. Trop. Med. Health. 39, 83–87. doi: 10.2149/tmh.2011-S10

PubMed Abstract | CrossRef Full Text | Google Scholar

Kaplan, Z. S., and Jackson, S. P. (2011). The role of platelets in atherothrombosis. Hematol. Am. Soc. Hematol. Educ. Program. 2011, 51–61. doi: 10.1182/asheducation-2011.1.51

PubMed Abstract | CrossRef Full Text | Google Scholar

Karshovska, E., Weber, C., and von Hundelshausen, P. (2013). Platelet chemokines in health and disease. Thromb. Haemost. 110, 894–902. doi: 10.1160/TH13-04-0341

PubMed Abstract | CrossRef Full Text | Google Scholar

Katsounas, A., Schlaak, J. F., and Lempicki, R. A. (2011). CCL5: a double-edged sword in host defense against the hepatitis C virus. Int. Rev. of Immunol. 30, 366–378. doi: 10.3109/08830185.2011.593105

PubMed Abstract | CrossRef Full Text | Google Scholar

Kelley, J. F., Kaufusi, P. H., and Nerurkar, V. R. (2012). Dengue hemorrhagic fever-associated immunomediators induced via maturation of dengue virus nonstructural 4B protein in monocytes modulate endothelial cell adhesion molecules and human microvascular endothelial cells permeability. Virology 422, 326–337. doi: 10.1016/j.virol.2011.10.030

PubMed Abstract | CrossRef Full Text | Google Scholar

Kho, S., Barber, B. E., Johar, E., Andries, B., Poespoprodjo, J. R., Kenangalem, E., et al. (2018). Platelets kill circulating parasites of all major plasmodium species in human malaria. Blood 132, 1332–1344. doi: 10.1182/blood-2018-05-849307

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, S. J., and Jenne, C. N. (2016). Role of platelets in neutrophil extracellular trap (NET) production and tissue injury. Semin. Immunol. 28, 546–554. doi: 10.1016/j.smim.2016.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Kitchens, C. S. (2009). Thrombocytopenia and thrombosis in disseminated intravascular coagulation (DIC). Hematol. Am. Soc. Hematol. Educ. Program 2009, 240–246. doi: 10.1182/asheducation-2009.1.240

PubMed Abstract | CrossRef Full Text | Google Scholar

Koupenova, M., Clancy, L., Corkrey, H. A., and Freedman, J. E. (2018). Circulating platelets as mediators of immunity, inflammation, and thrombosis. Circ. Res. 122, 337–351. doi: 10.1161/CIRCRESAHA.117.310795

PubMed Abstract | CrossRef Full Text | Google Scholar

Kraemer, M. U., Sinka, M. E., Duda, K. A., Mylne, A. Q., Shearer, F. M., Barker, C. M., et al. (2015). The global distribution of the arbovirus vectors aedes aegypti and Ae. albopictus. Elife 4:e08347. doi: 10.7554/eLife.08347

CrossRef Full Text | Google Scholar

Krishnamurti, C., Kalayanarooj, S., Cutting, M. A., Peat, R. A., Rothwell, S. W., Reid, T. J., et al. (2001). Mechanisms of hemorrhage in dengue without circulatory collapse. Am. J. Trop. Med. Hyg. 65, 840–847. doi: 10.4269/ajtmh.2001.65.840

PubMed Abstract | CrossRef Full Text | Google Scholar

Krishnamurti, C., Peat, R. A., Cutting, M. A., and Rothwell, S. W. (2002). Platelet adhesion to dengue-2 virus-infected endothelial cells. Am. J. Trop. Med. Hyg. 66, 435–441. doi: 10.4269/ajtmh.2002.66.435

PubMed Abstract | CrossRef Full Text | Google Scholar

Lai, Y. C., Chao, C. H., and Yeh, T. M. (2020). Roles of macrophage migration inhibitory factor in dengue pathogenesis: from pathogenic factor to therapeutic target. Microorganisms 8, 891–906. doi: 10.3390/microorganisms8060891

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, Y. R., Liu, M. T., Lei, H. Y., Liu, C. C., Wu, J. M., Tung, Y. C., et al. (2006). MCP-1, a highly expressed chemokine in dengue haemorrhagic fever/dengue shock syndrome patients, may cause permeability change, possibly through reduced tight junctions of vascular endothelium cells. J. Gen. Virol. 87, 3623–3630. doi: 10.1099/vir.0.82093-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Levi, M. (2016). Platelets in critical illness. Semin. Thromb. Hemost. 42, 252–257. doi: 10.1055/s-0035-1570080

PubMed Abstract | CrossRef Full Text | Google Scholar

Lindemann, S., Krämer, B., Seizer, P., and Gawaz, M. (2007). Platelets, inflammation and atherosclerosis. J. Thromb. Haemost. 1, 203–211. doi: 10.1111/j.1538-7836.2007.02517.x

CrossRef Full Text | Google Scholar

Littaua, R., Kurane, I., and Ennis, F. A. (1990). Human IgG Fc receptor II mediates antibody-dependent enhancement of dengue virus infection. J. Immunol. 144, 3183–3186.

PubMed Abstract | Google Scholar

Manne, B. K., Xiang, S. C., and Rondina, M. T. (2017). Platelet secretion in inflammatory and infectious diseases. Platelets 28, 155–164. doi: 10.1080/09537104.2016.1240766

PubMed Abstract | CrossRef Full Text | Google Scholar

Martinson, J., Bae, J., Klingemann, H. G., and Tam, Y. (2004). Activated platelets rapidly up-regulate CD40L expression and can effectively mature and activate autologous ex vivo differentiated DC. Cytotherapy 6, 487–497. doi: 10.1080/14653240410005249

PubMed Abstract | CrossRef Full Text | Google Scholar

McDonald, B., and Dunbar, M. (2019). Platelets and intravascular immunity: guardians of the vascular space during bloodstream infections and sepsis. Front. Immunol. 10:2400. doi: 10.3389/fimmu.2019.02400

PubMed Abstract | CrossRef Full Text | Google Scholar

McMorran, B. J., Wieczorski, L., Drysdale, K. E., Chan, J. A., Huang, H. M., Smith, C., et al. (2012). Platelet factor 4 and duffy antigen required for platelet killing of plasmodium falciparum. Science 338, 1348–1351. doi: 10.1126/science.1228892

PubMed Abstract | CrossRef Full Text | Google Scholar

Miyabe, K., Sakamoto, N., Wu, Y. H., Mori, N., and Sakamoto, H. (2004). Effects of platelet release products on neutrophilic phagocytosis and complement receptors. Thromb. Res. 114, 29–36. doi: 10.1016/j.thromres.2004.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Mourão, M. P., Lacerda, M. V., Macedo, V. O., and Santos, J. B. (2007). Thrombocytopenia in patients with dengue virus infection in the Brazilian Amazon. Platelets 18, 605–612. doi: 10.1080/09537100701426604

PubMed Abstract | CrossRef Full Text | Google Scholar

Mutheneni, S. R., Morse, A. P., Caminade, C., and Upadhyayula, S. M. (2017). Dengue burden in India: recent trends and importance of climatic parameters. Emerg. Microbes. Infect. 6:e70. doi: 10.1038/emi.2017.57

PubMed Abstract | CrossRef Full Text | Google Scholar

Noble, C. G., and Shi, P. Y. (2012). Structural biology of dengue virus enzymes: towards rational design of therapeutics. Antiviral Res. 96, 115–126. doi: 10.1016/j.antiviral.2012.09.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Noisakran, S., Chokephaibulkit, K., Songprakhon, P., Onlamoon, N., Hsiao, H. M., Villinger, F., et al. (2009). A re-evaluation of the mechanisms leading to dengue hemorrhagic fever. Ann. N. Y. Acad. Sci. 1171(Suppl. 1), E24–E35. doi: 10.1111/j.1749-6632.2009.05050.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Nurden, A. T. (2011). Platelets, inflammation and tissue regeneration. Thromb. Haemost. 105(Suppl. 1), S13–S33. doi: 10.1160/THS10-11-0720

PubMed Abstract | CrossRef Full Text | Google Scholar

Ojha, A., Bhasym, A., Mukherjee, S., Annarapu, G. K., Bhakuni, T., Akbar, I., et al. (2019). Platelet factor 4 promotes rapid replication and propagation of Dengue and Japanese encephalitis viruses. EBioMedicine 39, 332–347. doi: 10.1016/j.ebiom.2018.11.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Ojha, A., Nandi, D., Batra, H., Singhal, R., Annarapu, G. K., Bhattacharyya, S., et al. (2017). Platelet activation determines the severity of thrombocytopenia in dengue infection. Sci. Rep. 7:41697. doi: 10.1038/srep41697

PubMed Abstract | CrossRef Full Text | Google Scholar

Onlamoon, N., Noisakran, S., Hsiao, H. M., Duncan, A., Villinger, F., Ansari, A. A., et al. (2010). Dengue virus-induced hemorrhage in a nonhuman primate model. Blood 115, 1823–1834. doi: 10.1182/blood-2009-09-242990

PubMed Abstract | CrossRef Full Text | Google Scholar

Parker, Z. F., Rux, A. H., Riblett, A. M., Lee, F. H., Rauova, L., Cines, D. B., et al. (2016). Platelet factor 4 inhibits and enhances HIV-1 infection in a concentration-dependent manner by modulating viral attachment. AIDS Res. Hum. Retroviruses 2, 705–717. doi: 10.1089/AID.2015.0344

PubMed Abstract | CrossRef Full Text | Google Scholar

Patra, G., Mallik, S., Saha, B., and Mukhopadhyay, S. (2019). Assessment of chemokine and cytokine signatures in patients with dengue infection: a hospital-based study in Kolkata, India. Acta. Trop. 190, 73–79. doi: 10.1016/j.actatropica.2018.10.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Piresde Mello, C. P., Drusano, G. L., Rodriquez, J. L., Kaushik, A., and Brown, A. N. (2018). Antiviral effects of clinically-relevant interferon-α and ribavirin regimens against dengue virus in the hollow fiber infection model (HFIM). Viruses 10, 317–328. doi: 10.3390/v10060317

CrossRef Full Text | Google Scholar

Poon, T. C., Pang, R. T., Chan, K. C., Lee, N. L., Chiu, R. W., Tong, Y. K., et al. (2012). Proteomic analysis reveals platelet factor 4 and beta-thromboglobulin as prognostic markers in severe acute respiratory syndrome. Electrophoresis 33, 1894–1900. doi: 10.1002/elps.201200002

PubMed Abstract | CrossRef Full Text | Google Scholar

Prince, H. E., Yeh, C., and Lapé-Nixon, M. (2011). Utility of IgM/IgG ratio and IgG avidity for distinguishing primary and secondary dengue virus infections using sera collected more than 30 days after disease onset. Clin.Vaccine.Immunol. 18, 1951–1956. doi: 10.1128/CVI.05278-11

PubMed Abstract | CrossRef Full Text | Google Scholar

Quirino-Teixeira, A. C., Rozini, S. V., Barbosa-Lima, G., Coelho, D. R., Carneiro, P. H., Mohana-Borges, R., et al. (2020). Inflammatory signaling in dengue-infected platelets requires translation and secretion of nonstructural protein 1. Blood. Adv. 4, 2018–2031. doi: 10.1182/bloodadvances.2019001169

PubMed Abstract | CrossRef Full Text | Google Scholar

Rathore, A. P., Paradkar, P. N., Watanabe, S., Tan, K. H., Sung, C., Connolly, J. E., et al. (2011). Celgosivir treatment misfolds dengue virus NS1 protein, induces cellular pro-survival genes and protects against lethal challenge mouse model. Antiviral. Res. 92, 453–460. doi: 10.1016/j.antiviral.2011.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Rendu, F., and Brohard-Bohn, B. (2001). The platelet release reaction: granules constituents, secretion and functions. Platelets 12, 261–273. doi: 10.1080/09537100120068170

PubMed Abstract | CrossRef Full Text | Google Scholar

Riezu-Boj, J. I., Larrea, E., Aldabe, R., Guembe, L., Casares, N., Galeano, E., et al. (2011). Hepatitis C virus induces the expression of CCL17 and CCL22 chemokines that attract regulatory T cells to the site of infection. J. Hepatol. 54, 422–431. doi: 10.1016/j.jhep.2010.07.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Sabino, E. C., Loureiro, P., Lopes, M. E., Capuani, L., McClure, C., Chowdhury, D., et al. (2016). Transfusion-transmitted dengue and associated clinical symptoms during the 2012 epidemic in Brazil. J. Infect. Dis. 213, 694–702. doi: 10.1093/infdis/jiv326

PubMed Abstract | CrossRef Full Text | Google Scholar

Savarino, A., Boelaert, J. R., Cassone, A., Majori, G., and Cauda, R. (2003). Effects of chloroquine on viral infections: an old drug against today's diseases?. Lancet. Infect. Dis. 3, 722–727. doi: 10.1016/s1473-3099(03)00806-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Schexneider, K. I., and Reedy, E. A. (2005). Thrombocytopenia in dengue fever. Curr. Hematol. Rep. 4, 145–148.

PubMed Abstract | Google Scholar

Schlesinger, M. (2018). Role of platelets and platelet receptors in cancer metastasis. J. Hematol. Oncol. 11, 125–139. doi: 10.1186/s13045-018-0669-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Schmidt, A. C. (2010). Response to dengue fever–the good, the bad, and the ugly? N. Engl. J. Med. 363, 484–487. doi: 10.1056/NEJMcibr1005904

PubMed Abstract | CrossRef Full Text | Google Scholar

Schwartzkopff, F., Grimm, T. A., Lankford, C. S., Fields, K., Wang, J., Brandt, E., et al. (2009). Platelet factor 4 (CXCL4) facilitates human macrophage infection with HIV-1 and potentiates virus replication. Innate. Immun. 15, 368–379. doi: 10.1177/1753425909106171

PubMed Abstract | CrossRef Full Text | Google Scholar

Semple, J. W., Italiano, J. E. Jr, and Freedman, J. (2011). Platelets and the immune continuum. Nat. Rev. Immunol. 11, 264–274. doi: 10.1038/nri2956

PubMed Abstract | CrossRef Full Text | Google Scholar

Shu, P. Y., Chen, L. K., Chang, S. F., Yueh, Y. Y., Chow, L., Chien, L. J., et al. (2003). Comparison of capture immunoglobulin M (IgM) and IgG enzyme-linked immunosorbent assay (ELISA) and nonstructural protein NS1 serotype-specific IgG ELISA for differentiation of primary and secondary dengue virus infections. Clin. Diagn. Lab. Immunol. 10, 622–630. doi: 10.1128/cdli.10.4.622-630.2003

PubMed Abstract | CrossRef Full Text | Google Scholar

Sierra, B., Perez, A. B., Garcia, G., Aguirre, E., Alvarez, M., Gonzalez, D., et al. (2014). Role of CC chemokine receptor 1 and two of its ligands in human dengue infection. Three approaches under the Cuban situation. Microbes. Infect. 16, 40–50. doi: 10.1016/j.micinf.2013.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Simmons, C. P., Farrar, J. J., Nguyen, V., and Wills, B. (2012). Dengue. N. Engl. J. Med. 366, 1423–1432. doi: 10.1056/NEJMra1110265

PubMed Abstract | CrossRef Full Text | Google Scholar

Simon, A. Y., Sutherland, M. R., and Pryzdial, E. L. (2015). Dengue virus binding and replication by platelets. Blood 126, 378–385. doi: 10.1182/blood-2014-09-598029

PubMed Abstract | CrossRef Full Text | Google Scholar

Sung, P. S., and Hsieh, S. L. (2019). CLEC2 and CLEC5A: pathogenic host factors in acute viral infections. Front. Immunol. 10:2867. doi: 10.3389/fimmu.2019.02867

PubMed Abstract | CrossRef Full Text | Google Scholar

Suzuki, K., Sugimura, K., Hasegawa, K., Yoshida, K., Suzuki, A., Ishizuka, K., et al. (2001). Activated platelets in ulcerative colitis enhance the production of reactive oxygen species by polymorphonuclear leukocytes. Scand. J. Gastroenterol. 36, 1301–1306. doi: 10.1080/003655201317097164

PubMed Abstract | CrossRef Full Text | Google Scholar

Tang, Y. Q., Yeaman, M. R., and Selsted, M. E. (2002). Antimicrobial peptides from human platelets. Infect. Immun. 70, 6524–6533. doi: 10.1128/iai.70.12.6524-6533.2002

PubMed Abstract | CrossRef Full Text | Google Scholar

TDB Panacea Agreement (2017). Technology Development Board Enters Into an Agreement With M/s Panacea Biotec Pvt Ltd New Delhi to Complete the Late Stage Development of First Indian Dengue Vaccine. Available online at: http://tdb.gov.in/wp-content/uploads/2017/11/Dengue-Press-Release-14-11-2017.pdf (accessed September 17, 2020).

Google Scholar

Tricou, V., Minh, N. N., Van, T. P., Lee, S. J., Farrar, J., Wills, B., et al. (2010). A randomized controlled trial of chloroquine for the treatment of dengue in Vietnamese adults. PLoS. Negl. Trop. Dis. 4:e785. doi: 10.1371/journal.pntd.0000785

PubMed Abstract | CrossRef Full Text | Google Scholar

Trier, D. A., Gank, K. D., Kupferwasser, D., Yount, N. Y., French, W. J., Michelson, A. D., et al. (2008). Platelet antistaphylococcal responses occur through P2X1 and P2Y12 receptor-induced activation and kinocidin release. Infect. Immun. 76, 5706–5713. doi: 10.1128/IAI.00935-08

PubMed Abstract | CrossRef Full Text | Google Scholar

Trugilho, M., Hottz, E. D., Brunoro, G., Teixeira-Ferreira, A., Carvalho, P. C., Salazar, G. A., et al. (2017). Platelet proteome reveals novel pathways of platelet activation and platelet-mediated immunoregulation in dengue. PLoS Pathog. 13:e1006385. doi: 10.1371/journal.ppat.1006385

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsai, T. T., Chuang, Y. J., Lin, Y. S., Wan, S. W., Chen, C. L., and Lin, C. F. (2013). An emerging role for the anti-inflammatory cytokine interleukin-10 in dengue virus infection. J. Biomed. Sci. 20, 40–48. doi: 10.1186/1423-0127-20-40

PubMed Abstract | CrossRef Full Text | Google Scholar

Verschoor, A., Neuenhahn, M., Navarini, A. A., Graef, P., Plaumann, A., Seidlmeier, A., et al. (2011). A platelet-mediated system for shuttling blood-borne bacteria to CD8α+ dendritic cells depends on glycoprotein GPIb and complement C3. Nat. Immunol. 12, 1194–1201. doi: 10.1038/ni.2140

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S., He, R., Patarapotikul, J., Innis, B. L., and Anderson, R. (1995). Antibody-enhanced binding of dengue-2 virus to human platelets. Virology 213, 254–257. doi: 10.1006/viro.1995.1567

PubMed Abstract | CrossRef Full Text | Google Scholar

Wareing, M. D., Lyon, A. B., Lu, B., Gerard, C., and Sarawar, S. R. (2004). Chemokine expression during the development and resolution of a pulmonary leukocyte response to influenza A virus infection in mice. J. Leukoc. Bio. 76, 886–895. doi: 10.1189/jlb.1203644

PubMed Abstract | CrossRef Full Text | Google Scholar

Wiesner, J., and Vilcinskas, A. (2010). Antimicrobial peptides: the ancient arm of the human immune system. Virulence 1, 440–464. doi: 10.4161/viru.1.5.12983

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilder-Smith, A., Chen, L. H., Massad, E., and Wilson, M. E. (2009). Threat of dengue to blood safety in dengue-endemic countries. Emerg. Infect. Dis. 15, 8–11. doi: 10.3201/eid1501.071097

PubMed Abstract | CrossRef Full Text | Google Scholar

Wilder-Smith, A., Ooi, E. E., Horstick, O., and Wills, B. (2019). Dengue. Lancet 393, 350–363. doi: 10.1016/S0140-6736(18)32560-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Wirtz, T. H., Tillmann, S., Strüßmann, T., Kraemer, S., Heemskerk, J. W., Grottke, O., et al. (2015). Platelet-derived MIF: a novel platelet chemokine with distinct recruitment properties. Atherosclerosis 239, 1–10. doi: 10.1016/j.atherosclerosis.2014.12.039

CrossRef Full Text | Google Scholar

Yadav, S., and Storrie, B. (2017). The cellular basis of platelet secretion: emerging structure/function relationships. Platelets 28, 108–118. doi: 10.1080/09537104.2016.1257786

CrossRef Full Text | Google Scholar

Yeaman, M. R. (2014). Platelets: at the nexus of antimicrobial defence. Nat. Rev. Microbiol. 12, 426–437. doi: 10.1038/nrmicro3269

PubMed Abstract | CrossRef Full Text | Google Scholar

Zaldivar, M. M., Pauels, K., von Hundelshausen, P., Berres, M. L., Schmitz, P., Bornemann, J., et al. (2010). CXC chemokine ligand 4 (Cxcl4) is a platelet-derived mediator of experimental liver fibrosis. Hepatology 51, 1345–1353. doi: 10.1002/hep.23435

CrossRef Full Text | Google Scholar

Zhang, K., Liu, Y., Yang, X., Sun, H., Shu, X., Zhang, Y., et al. (2020). HBV promotes the recruitment of IL-17 secreting T cells via chemokines CCL22 and CCL17. Liver Int. 40, 1327–1338. doi: 10.1111/liv.14438

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: platelets, cytokines, PF4, immune modulation, dengue infection

Citation: Singh A, Bisht P, Bhattacharya S and Guchhait P (2020) Role of Platelet Cytokines in Dengue Virus Infection. Front. Cell. Infect. Microbiol. 10:561366. doi: 10.3389/fcimb.2020.561366

Received: 12 May 2020; Accepted: 31 August 2020;
Published: 30 September 2020.

Edited by:

Binod Kumar, Loyola University Chicago, United States

Reviewed by:

Eugenio D. Hottz, Juiz de Fora Federal University, Brazil
Bhagyaraj Ella, University of Florida, United States

Copyright © 2020 Singh, Bisht, Bhattacharya and Guchhait. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Prasenjit Guchhait, prasenjit@rcb.res.in

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.