Next Article in Journal
Multivariate Assessment of Thyroid, Lipid, and Inflammatory Profiles by HBV Status and Viral Load: Age- and Sex-Specific Findings
Previous Article in Journal
Depletion of Caspase-12 Alleviates Retinal Degeneration in Aged BALB/c Mice Following Systemic Neonatal Infection by Murine Cytomegalovirus (MCMV)
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanisms of Thromboinflammation in Viral Infections—A Narrative Review

by
Viviane Lima Batista
1,†,
Jenniffer Ramos Martins
2,†,
Celso Martins Queiroz-Junior
3,
Eugenio Damaceno Hottz
4,
Mauro Martins Teixeira
2 and
Vivian Vasconcelos Costa
3,*
1
Department of Microbiology, Federal University of Minas Gerais, Belo Horizonte 30161-970, Brazil
2
Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte 30161-970, Brazil
3
Department of Morphology, Federal University of Minas Gerais, Belo Horizonte 30161-970, Brazil
4
Department of Immunothrombosis, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2025, 17(9), 1207; https://doi.org/10.3390/v17091207
Submission received: 29 July 2025 / Revised: 21 August 2025 / Accepted: 31 August 2025 / Published: 3 September 2025
(This article belongs to the Section Human Virology and Viral Diseases)

Abstract

The circulatory and immune systems function in close coordination to maintain homeostasis and act as a frontline defense against infections. However, under certain conditions, this interaction becomes dysregulated, leading to thromboinflammation, a pathological process marked by the concurrent and excessive activation of coagulation, inflammation, and endothelial dysfunction. During viral infections, this phenomenon can markedly worsen clinical outcomes. Evidence indicates that viruses such as dengue, chikungunya, influenza, and SARS-CoV can trigger thromboinflammatory responses involving platelet activation, the release of procoagulant and pro-inflammatory mediators, and the formation of thrombi within blood vessels. While this response may initially help contain viral dissemination, in cases of high viremia it can progress to disseminated intravascular coagulation (DIC), hemorrhage, and multiple organ failure. This review compiles current evidence on thromboinflammatory mechanisms induced by arboviral and respiratory viruses and examines how these processes contribute to diseases’ pathogenesis and clinical severity.

1. From Homeostasis to Thromboinflammation: Mechanisms and Implications

The mammalian circulatory system performs essential biological functions, including oxygen delivery and defense against pathogens. It relies on a sophisticated hemostatic surveillance mechanism capable of rapidly sealing injuries to blood vessel walls, thereby preventing excessive blood loss and preserving the organism’s integrity [1,2]. Blood coagulation is a complex process in which platelets play a central role, working in concert with fibrin to form clots and prevent hemorrhage [3,4].
Platelets are small, anucleate, discoid cell fragments measuring approximately 2–3 μm in diameter, primarily responsible for regulating hemostasis. Beyond this role, they are essential in wound healing, angiogenesis, inflammation, innate immunity, and pathological thrombosis. They originate from the cytoplasm of megakaryocytes (MKs) located in the bone marrow [5,6], and the lungs [7]. Platelets play a critical role in maintaining vascular integrity, acting as sentinels for the detection of vessel injury. Upon vascular damage, they are rapidly recruited to the affected site through a tightly regulated process involving specific receptors’ interactions with components of the extracellular matrix [3,4,8,9]. Furthermore, during this process, activated platelets release the contents of their dense granules, including adenosine diphosphate (ADP) and serotonin, and their alpha granules, which contain inflammatory and coagulation factors, and express P-selectin on their surface, thereby amplifying the hemostatic response [3,4,8,9]. Activation of the coagulation cascade occurs in parallel with platelet activation. This process culminates in the formation of fibrin strands, a phenomenon known as secondary coagulation, which stabilizes the platelet plug. As extensively described by Engelmann et al. (2013) and Furie et al. (2008), this mechanism involves a series of reactions that lead to fibrin’s deposition and the amplification of the thromboinflammatory response [3,4].
The immune and hemostatic (coagulation) systems maintain a finely tuned balance, and their interaction is highly complex [4,10,11]. Under physiological conditions, this interplay is known as Immunothrombosis, in which immune cells and activated platelets, together with the coagulation system, act in concert to form blood clots. Conversely, when this response becomes dysregulated, it can progress to thromboinflammation, defined as the mutual amplification of thrombosis, inflammation, and vascular dysfunction [12]. This condition is a hallmark of several coagulopathies, disorders affecting blood coagulation that can lead to uncontrolled thrombosis and, in severe cases, multiple organ failure and death [10]. The concept of thromboinflammation is comprehensively reviewed by Waltraud C. et al. (2024) [11].
Thromboinflammation plays a crucial role in the pathogenesis of several diseases, including stroke [12], acute myocardial infarction [13], and preeclampsia [14]. In the context of viral infections, thromboinflammation is associated with the worsening of dengue [15], chikungunya [16], zika virus [17], yellow fever [18], influenza [19], and SARS-CoV [20]. This suggests that activation of the coagulation cascade during these infections has evolved in parallel with the immune system to help limit viral spread. However, acute viremia can trigger disseminated intravascular coagulation and/or hemorrhagic events, thereby worsening the clinical outcome and increasing mortality. In the following sections, we describe the thromboinflammatory mechanisms associated with each of these viruses.

2. Thromboinflammatory Mechanisms Induced by Arboviruses

Arboviruses comprise a diverse group of viruses transmitted by vectors, primarily arthropods such as mosquitoes. Among the most important vectors are Aedes aegypti and Aedes albopictus, which are responsible for the spread of several endemic diseases across multiple regions worldwide. Chikungunya and dengue are among the most prevalent and clinically significant arboviral diseases with systemic involvement [21].

2.1. Dengue

Dengue is an acute systemic viral infection caused by four genetically and antigenically related viruses, known as serotypes 1–4 (DENV-1 to DENV-4), each grouped into their respective genotypes [22]. These arboviruses belong to the genus Orthoflavivirus. Clinically, the disease is classified as dengue with or without warning signs, or as severe dengue (SD) [23]. Severe dengue is characterized by increased vascular permeability and a cytokine storm, followed by shock, which can be fatal [22,24,25,26]. Several severe manifestations of dengue are driven by hemostatic dysfunction and thromboinflammation through mechanisms involving (1) thrombocytopenia [27]; (2) formation of platelet–leukocyte aggregates [28]; (3) vascular and endothelial dysfunction [29]; and (4) coagulation abnormalities [30] (Figure 1 and Table 1).

2.1.1. Dengue-Induced Thrombocytopenia

Thrombocytopenia is a common manifestation in dengue patients, occurring in both mild and severe cases, although its underlying mechanisms are not yet fully understood. Evidence suggests that it may result from decreased platelet production in the bone marrow and/or enhanced destruction and clearance in the peripheral blood [32]. Additionally, recent studies have shown that the lung is also a site of platelet biogenesis, and pulmonary injury may contribute to the development of thrombocytopenia [7,33]. In this context, dengue virus has been detected in the lung tissue of patients with severe disease, as revealed by post-mortem biopsy analyses [34]. Furthermore, several studies have demonstrated a positive association between platelet activation and thrombocytopenia during dengue infection [27,35,36,37].
Dengue virus (DENV) may impair platelet formation by interfering with megakaryopoiesis [38,39,40,41,42]. This occurs through direct infection of bone marrow megakaryocytes, activating the Notch signaling pathway, which is associated with suppression of megakaryopoiesis in human cells [43,44]. Moreover, DENV-infected megakaryocytes reprogram their transcriptome, modulating the expression of genes transferred to platelets during infection, including interferon-stimulated genes (ISGs), potentially affecting both antiviral responses [45] and thrombopoiesis [46,47]. Platelets derived from DENV-infected megakaryocytes may exhibit impaired aggregation, as indicated by a reduced expression of surface markers such as CD41, CD42a, and CD61. The downregulation of these markers is linked to defective platelet adhesion and contributes to the thrombocytopenia observed in dengue infection [39]. Another key aspect is that megakaryocytes undergo an endomitotic cell cycle, characterized by an S phase followed by aborted mitotic entry, during which anaphase and cytokinesis are entirely bypassed, resulting in irregular chromosomal segregation [38,48]. Evidence suggests that DENV suppresses megakaryocyte polyploidization through a mechanism that is not yet fully understood, impairing proper platelet formation and contributing to the thrombocytopenia characteristic of the infection [38].
Several studies have demonstrated that platelets harboring high numbers of DENV genome copies become hyperactivated, as evidenced by elevated P-selectin expression and increased PAC-1 antibody binding on their surface. These platelets also display enhanced deposition of complement C3 and IgG on their membrane factors, linked to platelet lysis and clearance [27,36,49]. DENV-2-activated platelets markedly promote clot formation, while phagocytic cells, including monocytes, macrophages, dendritic cells, and neutrophils, rapidly engulf these platelets in an activation-dependent manner, thereby contributing to the thrombocytopenia observed in dengue [27,50,51]. In addition, these platelets exhibit mitochondrial dysfunction and classical apoptotic features, including mitochondrial depolarization, activation of caspases-9 and -3, and increased phosphatidylserine exposure, the primary “eat me” signal recognized by macrophages [52]. Evidence also indicates that platelet clearance in the spleen is a key event in DENV-induced thrombocytopenia. Furthermore, serotonin released by mast cells during infection has been shown to play a pivotal role in platelet activation, thereby exacerbating hemostatic dysfunction [53]. Additional mechanisms contributing to dengue-associated thrombocytopenia have been comprehensively reviewed by Quirino-Teixeira (2022) and Khazali et al. (2024) [32,54].
Collectively, these findings indicate that DENV-induced thrombocytopenia results from a multifactorial process involving multiple organs and physiological pathways, including modulation of transcription factors and disruption of key stages in platelet production.

2.1.2. Platelet–Leukocyte Aggregate Formation Mediated by P-Selectin

Platelet–leukocyte aggregates play a pivotal role in the pathogenesis of thromboinflammation and in the vascular complications observed in severe dengue cases [52]. Activated platelets amplify the inflammatory cascade by releasing pro-inflammatory and procoagulant mediators, including neutrophil-activating peptide 2 (NAP-2) and platelet-activating factor (PAF), thereby contributing to the exacerbation of inflammation and the thrombotic events associated with the disease [55]. Furthermore, both platelets and endothelial cells translocate P-selectin to their surface upon activation. This vascular adhesion molecule mediates interactions between activated platelets, endothelial cells, and leukocytes expressing P-selectin glycoprotein ligand-1 (PSGL-1), its primary receptor in vivo [56,57]. Engagement of P-selectin with PSGL-1 triggers the activation of β2 integrins on leukocytes, which in turn promotes their interaction with multiple ligands, such as platelet GPIbα, ICAM-2, and fibrinogen in the presence of thrombin, ultimately leading to full integrin activation [58].
Clinical evidence indicates that the formation of platelet–leukocyte aggregates, particularly platelet–monocyte aggregates (PMAs), is a frequent and dynamic event in dengue virus-infected patients, occurring in a biphasic pattern between days 6–8 and 11–16 after fever’s onset [59]. One study reported a significant increase in PMAs in dengue patients compared to healthy controls, with a positive correlation between platelet P-selectin expression and aggregate frequency [52]. PMAs were particularly pronounced in patients with thrombocytopenia and clinical signs of vascular leakage, such as hemoconcentration, hypoalbuminemia, and postural hypotension. Furthermore, PMA frequency negatively correlated with both platelet counts and serum albumin levels. Functionally, monocytes exposed to platelets from dengue patients secreted a range of pro- and anti-inflammatory cytokines, including IL-1β, IL-8, IL-10, and MCP-1, whereas platelets from healthy donors induced only MCP-1 production. These findings indicate that PMA formation contributes to the exacerbated inflammatory response and plays a key role in the pathophysiology of severe dengue [52]. The platelet–monocyte interaction is recognized as a key mechanism linking thrombosis and inflammation. Accordingly, PMAs are elevated in various thromboinflammatory diseases and correlate with disease severity [60,61,62].
In addition to monocytes, neutrophils act as key sentinel cells during dengue virus (DENV) infection. Evidence shows that DENV can activate neutrophils through serotype-independent mechanisms involving the C-type lectin receptor CLEC5A and Toll-like receptor 2 (TLR2), leading to NLRP3 inflammasome’s activation and subsequent release of neutrophil extracellular traps (NETs) and inflammatory cytokines [63,64,65]. Furthermore, DENV activates platelets via the CLEC2 receptor, promoting their aggregation with neutrophils and the release of extracellular vesicles that enhance NET formation and inflammatory mediator production through CLEC5A and TLR2 signaling in neutrophils and macrophages. Simultaneous blockade of CLEC5A and TLR2 inhibits NET formation, reduces inflammation, and markedly decreases DENV-induced systemic vascular permeability. These findings highlight the critical roles of the CLEC2 and CLEC5A/TLR2 pathways in platelet–leukocyte interactions and support the development of therapeutic strategies targeting immune modulation and tissue protection in severe dengue [64].
A recent study by Böer et al. (2024) evaluated the monocyte-to-lymphocyte ratio (MLR), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio (PLR) as potential prognostic inflammatory biomarkers in dengue patients. In a retrospective cohort of 193 patients treated in Brazil between 2012 and 2013, cases of SD were significantly associated with elevated MLR and reduced PLR, suggesting that an increased MLR combined with a decreased PLR may indicate a higher risk of progression to severe disease [66]. Additionally, platelets can interact with various lymphocyte populations, including T cells, B cells, and NK cells, via P-selectin- and integrin-mediated binding [67]. However, the role of platelet–lymphocyte aggregates in dengue’s pathogenesis remains incompletely understood. The dynamics of platelet–leukocyte aggregates have been further reviewed by Amin and colleagues (2025) [68] (Table 1).
P-selectin acts as a central mediator of thromboinflammation. In a sickle cell anemia model, blockade of P-selectin using an anti-CD62P monoclonal antibody reduced platelet–neutrophil aggregates and attenuated lung injury in mice [69]. Furthermore, studies in P-selectin-deficient (P-/-) mice have demonstrated that the absence of this molecule decreases formation of platelet–leukocyte aggregate and, consequently, development of thrombus in models of deep vein thrombosis [70,71]. Collectively, these findings highlight the critical role of P-selectin in leukocyte aggregation and thrombus formation.

2.1.3. Endothelial Dysfunction

Severe dengue is characterized by a transient increase in vascular permeability due to endothelial dysfunction, predominantly occurring during the critical phase, which typically lasts 24–48 h [72,73]. Disruption of the endothelial barrier leads to excessive fluid leakage, a phenomenon known as hyperpermeability, clinically manifested as vascular leakage and fluid’s accumulation in surrounding tissues. As a result, arterial and pulse pressures decrease, compromising tissue perfusion and potentially causing organ dysfunction [73]. The pathogenesis of endothelial dysfunction in dengue is multifactorial, arising from complex interactions between viral factors and the host immune response. Among viral components, non-structural protein 1 (NS1), secreted during DENV infection, plays a central role. NS1 alone can induce endothelial hyperpermeability in vitro and vascular leakage in vivo, independently of viral infection. Moreover, NS1 degrades the endothelial glycocalyx-like layer (EGL) in vitro via activation of endothelial sialidases and the cathepsin L/heparanase pathway [74]. Additionally, NS1 enhances vascular permeability through Toll-like receptor 4 (TLR4) activation, leading to inflammatory cytokine production [75], and it stimulates endothelial cells to secrete macrophage migration inhibitory factor (MIF), which increases permeability via autophagy-mediated mechanisms [76]. Multiple cell types respond to NS1 by producing pro-inflammatory mediators, including TNF, IL-1β, matrix metalloproteinases (MMPs), vascular endothelial growth factor (VEGF), and platelets themselves, which collectively disrupt the endothelial barrier and enhance vascular permeability during DENV infection [75,77]. Moreover, various cell types, including platelets, respond to NS1 stimulation by producing pro-inflammatory mediators such as TNF, IL-1β, matrix metalloproteinases (MMPs), and vascular endothelial growth factor (VEGF) [78,79]. These molecules collectively contribute to endothelial barrier disruption and the increased vascular permeability observed during DENV infection [77,80,81]. Additionally, direct DENV infection of endothelial cells modulates surface adhesion molecules, including ICAM-1, VCAM-1, and E-selectin, facilitating leukocyte recruitment to the activated endothelium [82,83] (Table 1).
Mast cells play a central role in the pathogenesis of severe dengue, primarily through direct activation by DENV, which triggers the release of newly synthesized mediators such as histamine, mast cell-specific proteases, eicosanoids (including leukotrienes and prostaglandins), cytokines, and chemokines. These mediators act directly on the vascular endothelium, compromising its integrity and function [84]. In vitro studies have shown that human mast cells can be permissive to DENV infection in an antibody-dependent manner via the FcγRII receptor [85,86]. Dengue patients exhibit elevated histamine levels in plasma and urine, as well as increased concentrations of VEGF and mast cell-derived proteases [84,87,88]. The release of these granule-stored mediators promotes endothelial activation, and mast cell-derived chymase has been shown to contribute to vascular leakage in mouse models of DENV infection [89]. DENV-induced mast cell activation also enhances the production of chemokines such as CCL3, CCL4, and CCL5 [90], along with pro-inflammatory cytokines including IL-6, IL-1β, and TNF [89,91]. Importantly, TNF released by infected mast cells acts as a potent activator of endothelial cells, playing a central role in the pathogenesis of SD [86,91,92].

2.1.4. Abnormalities in Coagulation

DENV infection is frequently associated with hemostatic disturbances, primarily driven by the host immune response. This response triggers simultaneous activation of coagulation and fibrinolysis pathways, leading to a thromboinflammatory state and multifactorial coagulopathy, particularly in severe cases [30,93,94]. Patients with severe dengue often present with thrombocytopenia, a prolonged prothrombin time (PT) and activated partial thromboplastin time (aPTT), and elevated plasma D-dimer levels, reflecting systemic hemostatic activation [95,96].
During convalescence, gradual normalization of hemostatic parameters reflects the restoration of hemostatic balance, with the degree of dysfunction correlating directly with disease severity [97]. Wills et al. (2002) reported marked reductions in anticoagulant proteins C, S, and antithrombin III in children with dengue, likely resulting from vascular leakage. Elevated levels of tissue factor (TF), thrombomodulin, and PAI-1 further indicate activation of the endothelium, platelets, and monocytes [37]. Systemic inflammation contributes to coagulopathy, as evidenced by Avila-Aguero et al. (2004), who found that fever, spontaneous bleeding, and a positive tourniquet test were associated with increased IL-6, IL-8, TNF, and coagulation activation [95]. In vitro, IL-6 increases endothelial permeability via MEK/ERK signaling [98], and animal models link elevations in IL-6 and IL-8 to increased D-dimer levels and thrombotic risk [99,100]. Elevated D-dimer reflects excessive coagulation followed by fibrinolysis and is associated with endothelial dysfunction, shock severity, and mortality in children with dengue shock syndrome [101]. Recently, Das et al. (2024) demonstrated that DENV NS1 downregulates coagulation factors I, V, X, and XIII through ERK-mediated transcriptional repression of HNF4α in Huh7 cells overexpressing NS1 [102]. The role of TF in hemorrhagic fevers has also been highlighted in Ebola virus infection: Geisbert et al. (2003) reported increased TF expression in monocytes and macrophages of infected primates, and its inhibition reduced lethality [103,104]. Normally absent from circulation, TF is a potent initiator of coagulation and represents a critical interface between inflammation and hemostasis [105,106].
In dengue, increased TF expression in monocytes from patients with severe disease has been reported, contributing to coagulopathy and thrombocytopenia [37,106]. Additionally, Huerta-Zepeda et al. (2008) observed upregulation of TF and protease-activated receptor-1 (PAR-1) in DENV-activated endothelial cells [107]. These findings suggest that coagulopathy in DENV infection results from a combination of immune activation, systemic inflammation, and endothelial dysfunction. Understanding the underlying molecular mechanisms, including the roles of NS1 and TF, may facilitate the development of targeted therapies to prevent the hemorrhagic and thrombotic complications associated with severe dengue.

2.1.5. Thromboinflammation in Dengue: Pregnancy Context

Although most cases of SD are characterized by the thromboinflammatory mechanisms previously described, certain groups develop less common yet clinically significant complications. Pregnant women represent a particularly vulnerable population due to physiological and immunological changes during gestation that impair their ability to mount effective immune responses [108]. Furthermore, hormonal adaptations, such as progesterone-induced increases in body temperature and estrogen-mediated water retention, may enhance attractiveness to mosquitoes, thereby facilitating DENV transmission [108,109].
In this population, thromboinflammation plays a critical role, increasing the risk of adverse outcomes for both mother and fetus [110]. Although rare, vertical transmission of DENV, most frequently reported in endemic areas and associated with high viremia, occurs predominantly during the peripartum period [111,112,113]. Maternal thromboinflammatory manifestations, including thrombocytopenia, plasma leakage, and bleeding tendency, can compromise placental circulation, resulting in restrictions in fetal growth, low birth weight, or stillbirth. Increased vascular permeability during infection facilitates viral passage across the placental barrier, while capillary leakage syndrome induces ischemia, further impairing fetal development [113,114,115,116,117].
Studies suggest that placental hypoxia may induce trophoblastic epithelial damage, chorangiosis, and inflammatory changes in the decidua or chorion–decidua, thereby negatively impacting fetal growth [118,119]. Newborns from mothers with acute DENV infection may present with conditions ranging from mild febrile illness accompanied by thrombocytopenia to severe outcomes such as intracranial hemorrhage and neonatal death [120]. Systematic reviews and meta-analyses confirm that maternal dengue increases the risk of spontaneous abortion, preterm birth, and low birth weight [118,121].

3. Chikungunya

Chikungunya virus (CHIKV) is an Alphavirus belonging to the Togaviridae family, transmitted by Aedes aegypti and Aedes albopictus mosquitoes, which are also vectors for DENV [122,123]. CHIKV infection can be asymptomatic or symptomatic, with the latter commonly presenting with high fever, headache, fatigue, nausea, rash, and, most notably, intense arthralgia, a hallmark feature of the disease [122]. The chronic form of the disease, first described in 1979 and subsequently reported in later outbreaks, is defined by persistent or recurrent joint pain lasting weeks or months after the acute phase [124]. CHIKV exhibits tropism for joints and muscle tissues, triggering an inflammatory response in synovial joints, often leading to disabling pain and edema [125,126,127,128]. Chronic arthralgia usually affects the same areas involved in the acute phase and may mimic rheumatoid arthritis [124,128].
The immune and thromboinflammatory mechanisms driving persistent inflammation and musculoskeletal symptoms in CHIKV infection remain poorly understood. Recent evidence indicates that endothelial cells play a central role in vascular alterations in CHIKV-infected mice, where infection induces production of nitric oxide (NO) and reactive oxygen species (ROS) and activates the NF-κB pathway, suggesting that CHIKV complications may extend beyond the joints [16]. ROS are highly reactive oxygen-derived molecules, including superoxide anion, hydroxyl radical, and hydrogen peroxide [129]. At physiological levels, ROS contribute to redox signaling, cell proliferation, differentiation, and angiogenesis. However, excessive ROS production overwhelms cellular antioxidant defenses, resulting in oxidative stress and cell death [130].
Previous evidence also suggests that platelets may contribute to musculoskeletal symptoms. Electron microscopy images showed that CHIKV directly associates with human platelets and can be trapped within platelet aggregates. During this interaction, some platelets displayed degranulation and lysis [131,132]. Similarly, rabbit platelets exposed to CHIKV for 24 h showed significant degranulation and lysis, suggesting that CHIKV may induce platelet activation and cell damage in multiple models [131,132]. Additionally, increased expression of platelet P-selectin and elevated levels of soluble P-selectin have been observed in patients with Chikungunya [133]. The same study reported elevated plasma levels of 12-HETE, a metabolite of arachidonic acid. 12-HETE is known to modulate vascular and joint inflammation in both sterile and infectious diseases. In rheumatoid arthritis, increased neutrophil infiltration, platelet activation, and 12-HETE synthesis have been documented [134]. Given that rheumatoid arthritis-like symptoms may occur in chronic CHIKV infection, 12-HETE’s elevation could be linked to platelet activation. Supporting this, Quintanilha et al. (2022) showed that patients who developed chronic joint and muscle pain had higher platelet activation in the acute phase [133].
Vascular complications have also been reported in CHIKV infection [135]. A case report described deep vein thrombosis in a CHIKV-infected patient [136]. Another study assessed D-dimer levels in 141 PCR-confirmed CHIKV patients and found that 63.8% had elevated D-dimer levels, suggesting an association with activation of coagulation [17]. D-dimers are fibrin degradation fragments generated during fibrinolysis, and their elevation serves as a biomarker of clot formation and breakdown in thrombotic events [137]. Despite these findings, thromboinflammatory events during CHIKV infection remain underexplored. Given the established role of platelets in vascular pathology in other viral diseases, further investigation is needed to better understand their contribution to the pathogenesis of Chikungunya virus infection (Table 1).

4. Influenza

The influenza virus belongs to the Orthomyxoviridae family and is a single-stranded RNA virus with zoonotic potential, responsible for epidemics that cause approximately 5 million severe respiratory infections in humans each year [138]. It is primarily transmitted through respiratory droplets and contact with contaminated surfaces. Clinical manifestations range from asymptomatic or mild infections to severe forms that may progress to viral pneumonia, respiratory failure, and death, particularly in high-risk groups such as the elderly or individuals with comorbidities [139]. Influenza is the causative agent of flu and is classified into four types: A (associated with global pandemics), B, C, and D [140]. Influenza A virus (IAV) is of major global importance due to its high genetic variability and remarkable adaptability. It can infect not only humans but also a wide variety of animal species, including birds, pigs, horses, bats, and others [141]. IAV is classified based on its surface glycoproteins, hemagglutinin (HA) and neuraminidase (NA), resulting in subtypes such as H1N1, H3N2, H5N1, and H7N9. Among these, H1N1 and H3N2 are the most prevalent and widely disseminated among humans [142,143].
The virus enters host cells via endocytosis after its hemagglutinin binds to sialic acid on the surface of epithelial cells in the respiratory tract [144] and is recognized by Toll-like receptors (TLRs), which activate key immune cells such as macrophages, neutrophils, and dendritic cells [145]. Furthermore, the virus can be detected in circulation, where it is internalized by platelets via TLR7, leading to the release of complement C3. This, in turn, stimulates neutrophils to release their DNA, resulting in a highly prothrombotic process and increasing the risk of cardiovascular events. In parallel, platelets release GM-CSF, which favors neutrophil survival and the formation of platelet–neutrophil clusters. Consequently, these platelets become dysfunctional or reduced in number, impairing their ability to contain inflammation and further favoring viral spread [146]. This recognition leads to the production of pro-inflammatory cytokines, including TNF, IL-6, IL-8, and IFN-α/β, resulting in symptoms such as fever, cough, rhinorrhea, sore throat, myalgia and, in severe cases, acute respiratory distress syndrome (ARDS) and pulmonary failure [147]. This intense inflammatory response can cause tissue damage, particularly to the pulmonary endothelium, making it more susceptible to dysfunction. The pulmonary endothelium actively regulates key processes involved in lung homeostasis, such as angiogenesis, vasomotor tone, regulation of vascular permeability to macromolecules and leukocytes, blood coagulation, and the production of cytokines and adhesion molecules. These functions, however, may be impaired during ARDS [148].
Endothelial dysfunction during influenza infection is a complex event involving multiple mechanisms, including the following: (I) Increased expression of adhesion molecules associated with endothelial activation (e.g., VCAM-1 and ICAM-1), which contributes to enhanced leukocyte and platelet adhesion to the endothelium. These molecules bind to very late antigen-4 (VLA-4) and CD11/CD18 integrins, thereby facilitating leukocytes’ migration and adhesion to the endothelial surface [149]. (II) Reduced production of NO, a key molecule generated by nitric oxide synthase (NOS) activity. NO exerts inhibitory effects on platelet activation via signaling pathways, and its decreased availability is directly associated with enhanced platelet activation and the promotion of a procoagulant state [150]. (III) Excessive upregulation of TF, a protein involved in initiating the coagulation cascade following vascular injury. Aberrant expression of TF triggers a robust activation of the coagulation cascade, leading to thrombus formation, as previously reported in murine infections with IAV/H1N1 [151]. (IV) Exposure of extracellular matrix proteins (e.g., collagen). Under normal conditions, extracellular matrix proteins are found beneath the vascular endothelium; however, endothelial injury leads to their exposure to circulating blood, promoting interactions with platelets that facilitate their adhesion and activation. This process, in turn, amplifies both the inflammatory response and the coagulation cascade, as demonstrated in vitro experiments using human platelets [152].
Platelets play a crucial role in immunological homeostasis through both direct and indirect interactions with various immune cell types, such as leukocytes. However, their role during viral infections, particularly in the context of IAV, remains incompletely understood. Several case studies have demonstrated a correlation between influenza virus infection and thrombocytopenia. For example, children with severe influenza A and B infections exhibited a significantly reduced platelet distribution width (PDW) compared to those with non-severe disease [153]. Moreover, PDW has been associated with disease severity in patients with IAV-induced ARDS [148]. Platelets can engulf the virus via binding to sialoglycans, followed by neuraminidase-mediated removal of sialic acids, which facilitates hepatic clearance of platelets. This mechanism has been observed through the internalization of IAV in vitro. In vivo experiments using IAV/H1N1-infected animal models [154] indicated a severe disease phenotype in which viral load was inversely correlated with platelet count during the acute phase, highlighting an intrinsic role of IAV in the development of thrombocytopenia. A recent study demonstrated that to circumvent the deleterious effects of thrombocytopenia, hematopoietic stem cells (HSCs) can detect inflammatory signals and, independently of thrombopoietin, increase megakaryocyte production to restore platelet levels [155]. A potential mechanism of thrombocytopenia in experimental IAV infection is pulmonary microvascular thrombosis [156]. In this model, activation or inhibition of protease-activated receptor 4 (PAR4), a thrombin receptor, exacerbated or recovered, respectively, lung injury and mortality, which was also mitigated by genetic deletion or pharmacological inhibition of integrin αIIb/β3. Integrin αIIb/β3 is a surface receptor found on platelets and is responsible for fibrinogen-mediated platelet adhesion and aggregation, facilitating the formation of platelet aggregates and thrombi. In a study, PAC-1 (an antibody that binds to the active conformation of αIIbβ3 integrin) was used to assess platelet activation during H1N1 infection, and a significant increase was observed in infected patients, consistent with platelets in a hyperactivated state, inducing an inflammatory response (IL-6 production) and leukocyte/endothelial activation and increasing the risk of thrombosis and organ failure [157]. Thrombin was also directly involved in platelet activation in in vitro assays in which human platelets were stimulated with H1N1 virus in the presence of IAV immune serum [158]. Thrombin is a key enzyme in the coagulation cascade; it cleaves fibrinogen into fibrin, leading to clot formation.
Although respiratory viruses such as IAV are known to impair lung function and can cause thrombocytopenia, the deleterious effects of infection on platelet precursor cells are still poorly understood. Megakaryocytes, platelet precursors, were recently found to be localized in the lungs, where they play an active role in thrombopoiesis [159]. IAV infection can lead to the development of ARDS, which is associated with a robust activation of the inflammatory response and the release of pro-inflammatory mediators [160]. Together, these events disrupt endothelial homeostasis, promoting platelet and leukocyte adhesion, platelet activation, activation of coagulation pathways, and clot formation, thereby exacerbating the thrombotic and inflammatory response [156]. These mechanisms may result in venous or arterial thrombosis [161,162], potentially leading to severe complications such as pulmonary embolism, stroke, or myocardial infarction. Although seasonal vaccines are available for different viral strains [163], severe infections still occur due to both viral and host-related factors. Understanding the underlying mechanisms is crucial for developing effective therapeutic strategies to manage thromboinflammatory complications associated with severe IAV infection and other respiratory diseases (Figure 2).

5. SARS-CoV-2

Coronaviruses (CoVs) belong to the Coronaviridae family, Orthocoronavirinae subfamily, and Betacoronavirus genus. They are associated with the development of severe acute respiratory syndrome (SARS) in infected individuals who progress to the most severe forms of the disease, characterized by atypical pneumonia, acute lung injury, fibrosis, and death [165]. Due to their high transmissibility, an epidemic caused by SARS-CoV-1 emerged between 2002 and 2003 in Guangdong Province, China, resulting in at least 770 deaths [166]. In late 2019, the appearance of SARS-CoV-2 in Wuhan, China, led to a global pandemic with immeasurable social and economic consequences. The disease caused by this novel coronavirus, termed COVID-19, presents a broad clinical spectrum ranging from asymptomatic infection to severe respiratory distress and multiorgan failure, which has caused millions of deaths worldwide [167]. Importantly, SARS-CoV-2 pathogenesis extends beyond severe acute respiratory syndrome, manifesting as a systemic disease involving vascular and hematological components [168].
SARS-CoV-2, the causative agent of COVID-19, is a positive-sense single-stranded RNA virus that shares 79% nucleotide sequence similarity with SARS-CoV [169]. The structure of SARS-CoV-2 is highly characteristic, consisting of nucleocapsid (N), membrane (M), envelope (E), and spike (S) proteins. The spike protein is responsible for binding to its receptor, angiotensin-converting enzyme 2 (ACE2), which is widely expressed on cells of the respiratory tract [170,171]. Once infection is established, viral replication occurs within the respiratory tract, triggering the recruitment of immune cells to the site of infection, production of pro-inflammatory cytokines and chemokines, increased permeability across the pulmonary endothelium, and consequent tissue damage mechanisms that are closely associated with SARS pathogenesis [172,173].
In addition to the pulmonary involvement induced by infection, coagulation abnormalities have emerged as significant contributors to the morbidity and mortality associated with the disease. Hypercoagulable states have been frequently observed in patients infected with SARS-CoV-2. Decreased levels of circulating platelets have been associated with more severe disease outcomes [174]. Furthermore, thrombotic events such as venous and arterial thromboembolism have been correlated with increased mortality among infected individuals [175]. Consistently, microvascular thrombosis was ten times higher in pulmonary histopathological studies from patients who expired from COVID-19 than IAV pneumonia [176]. This section discusses thromboinflammation mechanisms related to SARS-CoV 2 (Figure 2).
Thrombocytopenia can be one of the outcomes of SARS-CoV-2 infection and is associated with disease progression and severity [165]. However, the reduction in platelet count is multifactorial and may involve various mechanisms linked to a complex host–pathogen interaction. Furthermore, SARS-CoV-2 infection triggers an immune response that increases pro-inflammatory cytokine levels, which interfere with CD34+ hematopoietic stem cells, impairing megakaryopoiesis and leading to platelet depletion [167]. Interestingly, SARS-CoV-2 has been detected in megakaryocytes (from both the bone marrow and lungs) and in the platelets from surviving and non-surviving COVID-19 patients, indicating direct infection of these cells [177]. Additionally, transmission electron microscopy analysis revealed the presence of viral particles within bone marrow megakaryocytes from thrombocytopenic and ICU-admitted patients, along with a reduction in these cells, suggesting a direct relationship with the thrombocytopenia observed in clinical findings [178,179].
Regarding the inflammatory response in COVID-19, it is well established that viral infection promotes the recruitment and activation of immune cells, such as macrophages, neutrophils, and dendritic cells, as part of the host defense mechanism. This process triggers the release of several inflammatory mediators, including IL-1β, IL-6, TNF, IFN-γ, IL-12, IL-15, and IL-18 [166]. The heightened inflammatory environment contributes to platelet activation, which may also occur through formation of immune complexes involving IgG binding to the spike protein S1 and subsequent crosslinking of the FcγRII receptor on platelets [180]. Platelet activation is further evidenced by increased P-selectin expression and the formation of platelet–leukocyte aggregates [181,182], mechanisms that enhance thrombus formation and decrease circulating platelet counts. In patients with severe COVID-19 requiring mechanical ventilation, endothelial damage and morphological alterations in pulmonary capillaries have also been reported, leading to platelet activation, aggregation, and increased consumption [183]. A recent study identified the platelet enzyme 12-lipoxygenase (12-LOX) as a protective factor during SARS-CoV-2 infection. The deficiency of 12-LOX was associated with exacerbated inflammation and worse disease outcomes. Expressed in platelets, 12-LOX regulates arachidonic acid metabolism and generates bioactive lipid mediators, including 12-HETE and 12-HETrE, which modulate both platelet activation and inflammatory signaling. In mice lacking 12-LOX, SARS-CoV-2 infection triggered an exacerbated pulmonary response characterized by increased leukocyte infiltration [184]. Additionally, platelets from patients with community-acquired pneumonia, with or without COVID-19, release chemokines, PAI-1, and CD40 ligand. Enhanced release of P-selectin, CD40 ligand, PAI-1, and CCL5 mediated via GPVI was linked to a higher risk of requiring invasive or non-invasive ventilation and/or mortality in COVID-19 patients [185].
Several studies have demonstrated that platelet activation and the formation of platelet–monocyte aggregates are closely associated with disease severity in patients with COVID-19. In severe cases, platelets promote TF expression by monocytes through signaling pathways mediated by P-selectin and integrin αIIb/β3 [186]. Besides inducing TF, P-selectin and integrin-mediated adhesion induces the secretion of inflammatory cytokines and chemokines by the monocyte [187]. Interestingly, monocyte activation was amplified by TF signaling, which induced the secretion of IL-1β and TNF-α in this model [187]. In addition to promoting leukocyte adhesion, activated platelets release mediators that favor thrombosis and inflammation, such as soluble CD40 ligand (sCD40L). Activated platelets also produce lipid mediators such as thromboxane A2 (TXA2) [188] and PAF [179], which are directly associated with vasoconstriction and the formation of platelet aggregates, contributing to a hypercoagulable state, thrombus formation, and heightened inflammation. When bound to endothelial cells, sCD40L induces the expression of adhesion molecules (e.g., ICAM-1, VCAM-1, and E-selectin), which are involved in leukocyte recruitment [189,190]. Recently, a combination of platelet and endothelial cell transcriptomics revealed the S100A8/A9 gene product myeloid-related protein (MRP) 8/14 as a main platelet-derived mediator inducing endothelial thromboinflammation in severe COVID-19 [191].
SARS-CoV-2 infection activates an inflammatory response characterized by the production of several pro-inflammatory mediators that can disrupt crucial pathways, including thrombotic and fibrinolytic processes, as well as those that regulate endothelial tight junctions [192]. This imbalance and inflammatory stimulation promote the recruitment of inflammatory cells to the site of infection, leading to increased endothelial permeability and consequent vascular leakage. It has been demonstrated that neutrophils recruited to the tissue produce NETs in response to infection, which contribute to the formation of platelet–neutrophil aggregates and thrombotic events in COVID-19 patients [193,194,195].
In COVID-19, endothelial dysfunction due to vascular damage or inflammation exposes vWF, promoting platelet activation, adhesion, and aggregation [196]. The inflammatory environment promotes dysregulation between plasminogen activator inhibitor-1 (PAI-1) and the fibrinolytic system, both of which are key regulators of clot formation and dissolution [197]. Fibrinogen is a plasma glycoprotein with a fundamental role in blood coagulation. Upon conversion to fibrin by thrombin, it contributes to clot formation and provides the structural framework for platelet aggregates, supporting platelet adhesion to the vascular endothelium. Under physiological conditions, this process facilitates vascular repair and prevents bleeding [188]. In the context of COVID-19, negative regulation of the fibrinolytic pathway due to elevated PAI-1, the main inhibitor of fibrinolysis, leads to excessive production of fibrinogen, promoting a prothrombotic hypercoagulable state and endothelial dysfunction via thrombus formation. These thrombi can develop in both large vessels and the microvasculature [189].
Hypercoagulability has been associated with increased fibrin deposition in COVID-19 patients [20,190]. During fibrinolysis, when fibrin clots are degraded by plasmin, D-dimer fragments are released. Elevated levels of D-dimers are indicative of hypercoagulability and excessive activation of the coagulation cascade, making them an important biomarker for COVID-19 severity [198,199]. Understanding these interactions is crucial for unraveling the mechanisms behind the severe thrombotic outcomes that affect many patients with COVID-19. Platelet activation and platelet–leukocyte aggregates’ formation significantly contribute to an increased risk of thrombotic events and vascular complications.
Understanding these events is crucial for predicting clinical outcomes, as the symptoms of respiratory viral infections often overlap [200]. Both IAV and SARS-CoV-2 infections upregulate endothelial adhesion molecules, such as VCAM-1 and ICAM-1, which promote platelet–neutrophil aggregation and stimulate the formation of neutrophil extracellular traps (NETs). These cellular interactions enhance the release of pro-inflammatory cytokines, including IL-6 and TNF, amplifying the acute inflammatory response. This excessive inflammation contributes to tissue damage and can ultimately progress to acute respiratory distress syndrome (ARDS).

6. The Evidence of Thromboinflammation and Post-Acute Viral Sequelae

Beyond the acute phase of COVID-19, increasing evidence highlights thromboinflammation as a central mechanism in the post-acute sequelae of SARS-CoV-2 infection (long COVID), a condition marked by persistent or newly emerging symptoms after resolution of the acute illness. Several studies report that, months after infection, patients continue to display elevated endothelial activation markers, such as von Willebrand factor, along with fibrinolysis-resistant microclots, indicating sustained hypofibrinolysis and ongoing vascular dysfunction [201,202]. Increased fibrinogen deposition has also been detected in the lungs and brain of COVID-19 patients, correlating with disease severity and serving as a potential biomarker for long COVID [20,203,204]. Furthermore, fibrin can directly bind the SARS-CoV-2 spike protein, generating pro-inflammatory clots that intensify systemic thromboinflammation and contribute to COVID-19-related neuropathology [20].
This prothrombotic state is further reinforced by persistent activation of both the classical and alternative complement pathways, leading to dysregulated formation of the terminal C5b-9 complex, which exacerbates tissue injury and endothelial dysfunction [205]. At the cellular level, patients up to 12 months post-infection display monocyte–platelet aggregates and platelet hyper-reactivity, indicating long-lasting platelet activation [205]. Another key mechanism involves the persistence of SARS-CoV-2 RNA and proteins in various tissues, including the small intestine, even in mild cases often accompanied by reactivation of latent herpesviruses such as Epstein–Barr virus (EBV) and cytomegalovirus (CMV) [206,207]. This phenomenon parallels the behavior of persistent neurotropic viruses and may sustain chronic low-grade inflammation, neuroinflammation, and prolonged clinical symptoms [206,207]. Collectively, the convergence of viral persistence, latent virus reactivation, and ongoing thromboinflammatory activation establishes a plausible pathophysiological axis underlying long COVID.
Similar mechanisms are also observed in DENV infection, in which endothelial dysfunction and direct viral effects contribute to neurological complications. In the central nervous system (CNS), cases of ischemic or hemorrhagic stroke and encephalitis have been reported [208,209]. In the peripheral nervous system, manifestations include long thoracic nerve palsy, abducens nerve palsy, facial palsy, brachial neuritis, myositis, hypokalemic paralysis, and Guillain-Barré syndrome. Ocular complications, including maculopathy, optic neuropathy, and subconjunctival and vitreous hemorrhages, have also been described [210,211].
Encephalopathy is the most frequent neurological manifestation of dengue. Laboratory tests often reveal marked elevations in serum transaminases, hyponatremia, and hypoxia. In Thailand, for example, encephalopathy was reported in nearly half of the children with neurological manifestations associated with DENV infection [212,213,214]. The pathophysiology appears to be multifactorial, involving cerebral edema, hypoxia, and systemic endothelial dysfunction [212,213,214] largely resulting from the thromboinflammatory response triggered by the virus. Another rare but significant complication is ischemic and/or hemorrhagic stroke, reported in patients with confirmed DENV infection [215,216,217,218]. Although the specific mechanisms are not yet fully elucidated, there is evidence that thromboinflammation plays a central role [210]. Studies indicate that interactions among platelets, regulatory T cells, and endothelial cells contribute to thrombus formation, inflammatory activation, and tissue injury, with the CD84 molecule identified as a key mediator at the platelet–T cell interface during ischemic stroke [219].
Clinical manifestations characterized by tremors, rigidity, and bradykinesia have also been reported in patients with DENV infection [220,221]. These cases are classified as viral parkinsonism, a term used to describe syndromes that mimic Parkinson’s disease in association with infectious processes. The mechanism by which DENV triggers these manifestations is not yet fully understood. Evidence suggests that the virus can cross the blood–brain barrier via cytokine-induced increased permeability or through the direct action of viral proteins, such as NS1 [222]. Recent studies have shown that DENV NS1 protein modulates microRNA expression in human cerebral microvascular endothelial cells, compromising endothelial barrier integrity [222]. Moreover, NS1 promotes degradation of the endothelial glycocalyx (EGL), a membrane-associated network of proteoglycans and glycoproteins lining the vascular endothelium, through sialic acid cleavage and the release of heparan sulfate proteoglycans, resulting in increased endothelial permeability [74].
Viral infection-associated parkinsonism is characterized by the presence of at least two of the following symptoms: tremor, bradykinesia, rigidity, and postural instability. Movement disorders related to DENV infection have been widely described in the literature [223]. Furthermore, recent studies indicate that long-term neurological complications associated with viral infections are becoming increasingly frequent, highlighting the need for research aimed at developing therapeutic strategies for these patients.

7. Antithrombotic Strategies in Viral Infections: Anticoagulation and Antiplatelet Therapy

The recognition of thromboinflammation as a central component of COVID-19 pathophysiology has spurred extensive research into therapeutic strategies aimed at mitigating the disease’s prothrombotic effects. High-quality randomized clinical trials have demonstrated the efficacy and safety of anticoagulant therapy in hospitalized COVID-19 patients [224,225,226]. Among the agents investigated, heparin is notable due to its widespread use across various regimens and routes of administration. Studies indicate that both low-molecular-weight heparin (LMWH) and unfractionated heparin are associated with improved clinical outcomes. Therapeutic-dose LMWH has been linked to increased survival until hospital discharge, as well as a reduced need for cardiovascular and respiratory support [227]. In addition to systemic administration, alternative approaches such as nebulized heparin have shown efficacy in patients hospitalized with secondary COVID-19 pneumonia [228]. Previous evidence also indicates that inhaled heparin can improve pulmonary coagulopathy and reduce the need for mechanical ventilation ARDS [229]. Its mechanism of action involves inactivating thrombin and factor Xa via interaction with antithrombin, thereby interrupting the coagulation cascade [230]. A recent meta-analysis further reinforces the role of anticoagulation in the context of COVID-19 [231].
Beyond anticoagulant therapies, antiplatelet agents such as aspirin have also been investigated. Experimental and observational studies suggest that aspirin is associated with a reduced incidence of ARDS and improved survival in cases of acute lung injury [232]. As a widely available and low-cost drug, aspirin has been shown to decrease thrombotic events and attenuate in vitro platelet hyperactivity in patients with SARS-CoV-2 infection [233]. Clinical studies have further reported that aspirin use is associated with a lower risk of 28-day in-hospital mortality [234]. However, findings remain somewhat controversial: the RECOVERY trial indicated that, although aspirin did not reduce overall mortality, it was associated with a modest increase in 28-day hospital discharge rates [235].
Meta-analyses indicate that patients with severe COVID-19 have a significantly higher risk of acute cerebrovascular events, heart failure, and myocardial infarction compared to patients with non-severe disease [236]. In this context, P2Y12 receptor antagonists such as clopidogrel, prasugrel, and ticagrelor, widely used in the treatment of ischemic stroke and acute coronary syndrome, have been proposed as potential therapeutic strategies [232,237]. A review conducted by Bikdeli and colleagues (2020) compiled the main drugs with potential roles in modulating COVID-19-associated thromboinflammation, highlighting the importance of investigating antithrombotic agents in this setting [232].
On the other hand, the use of antiplatelet agents in patients with dengue remains controversial due to the increased risk of bleeding associated with thrombocytopenia. A cohort study involving adults with laboratory-confirmed dengue evaluated individuals who were previously receiving aspirin or clopidogrel for cardiovascular diseases [238]. The study compared patients who continued therapy during the thrombocytopenic phase with those who suspended it. The results indicated that therapy’s interruption did not increase the risk of cardiovascular or cerebrovascular events, while continuation of therapy did not worsen the clinical course. Therefore, decisions regarding antiplatelet therapy should be individualized, considering both the bleeding risk and the patient’s underlying cardiovascular condition [238].
In the management of dengue-induced thrombocytopenia, a narrative review suggested the use of thrombopoietin receptor (TPO-R) agonists, such as eltrombopag and romiplostim, the latter approved by the FDA in 2018 for idiopathic thrombocytopenia [239]. These drugs stimulate platelet production by activating thrombopoietin receptors on megakaryocytes and their precursor cells, thereby increasing platelets’ release into circulation. Evidence indicates potential efficacy of these agents in severe cases of thrombocytopenia associated with dengue virus infection [240,241,242,243]. Additionally, transcriptomic and bioinformatic analyses using the Connectivity Map (CMap) have identified potential drug repurposing strategies, including enalapril, traditionally indicated for hypertension, heart failure, and left ventricular dysfunction [244,245]. These agents act as angiotensin-converting enzyme (ACE) inhibitors, reducing angiotensin II and aldosterone levels, thereby lowering peripheral resistance and increasing renin activity. In murine models, enalapril and losartan reduced viral uptake and interleukin-1 production by macrophages during dengue infection, suggesting additional anti-inflammatory and antiviral effects [246].
In the context of influenza-induced pneumonia, a murine study demonstrated that clopidogrel reduced platelet activation and the formation of platelet–leukocyte aggregates [247]. However, this effect alone was insufficient to decrease mortality. Notably, when clopidogrel was combined with oseltamivir, a significant improvement in disease outcomes and survival was observed in the animal model, indicating a synergistic effect between the drugs. These findings suggest that in severe influenza pneumonia, the combination of platelet activation blockade with clopidogrel and viral replication inhibition with oseltamivir may constitute an effective therapeutic strategy [247].

8. Conclusions

Thromboinflammation represents a critical interface between the immune and hemostatic systems during viral infections, playing a major role in the pathogenesis and clinical severity of diseases such as dengue, chikungunya, influenza, and COVID-19. A detailed understanding of the molecular and cellular mechanisms driving this dysregulated response is essential for developing more effective therapeutic strategies. Interventions targeting platelet activation, inflammation, and coagulation may provide promising approaches to reduce the morbidity and mortality associated with these infections. Ultimately, advancing our knowledge of virus-induced thromboinflammation could enable more targeted therapies and improve the clinical management of affected patients.

9. Methodology

Bibliographic searches for relevant electronic sources were carried out throughout the duration of this study. The Medline/PubMed database (https://pubmed.ncbi.nlm.nih.gov/, accessed on 10 June 2025) and the CAPES Periodicals Portal (Coordination for the Improvement of Higher Education Personnel—CAPES/MEC) (https://www.periodicos.capes.gov.br, accessed on 10 June 2025) were used for data retrieval. Original research articles, review papers, and book chapters in English were selected according to their relevance to the study’s subject matter. The search strategy included the following keywords: thromboinflammation; endothelial dysfunction; thrombocytopenia; viral infection; platelet activation; influenza; chikungunya; SARS-CoV-2; dengue. persistence of platelet activation; neurological sequelae; viral parkinsonism.
Table 1. Thromboinflammatory pathways in different viral infections.
Table 1. Thromboinflammatory pathways in different viral infections.
Thromboinflammatory PathwaysDengueChikungunyaInfluenzaSARS-CoV-2
Platelet activation
  • Thrombocytopenia [27]
  • Platelet activation with increased P-selectin expression and PF4 release [28,36]
  • Formation of platelet–leukocyte aggregates [59,60]
  • Platelet activation with increased P-selectin [133]
  • Platelet activation with increased P-selectin expression [156,157]
  • Thrombocytopenia [165]
  • Platelet activation with increased P-selectin expression and PF4 release [175,184]
  • Formation of platelet–leukocyte aggregates [186].
  • Persistent platelet activation [205]
Endothelial dysfunction
  • Endothelial activation [73,75]
  • Increased vascular permeability [74]
  • Modulation of ICAM-1 and VCAM-1 [29]
  • Endothelial injury associated with arthritis and chronic inflammation [16]
  • Endothelial activation
  • Modulation of ICAM-1 and VCAM-1 [151]
  • Endothelial activation [19]
  • Modulation of ICAM-1 and VCAM-1 [189,190]
  • Increased vascular permeability [189,190]
Abnormalities in coagulation
  • Increased D-dimer levels [95,96,97]
  • Prolonged activated partial thromboplastin time (aPTT) [95]
  • Prolonged prothrombin time [96]
  • Increased expression of tissue factor in monocytes [106].
  • Hemorrhage and hypovolemic shock [30]
  • Increased D-dimer levels [17]
  • Rare cases of deep vein thrombosis [136]
  • Increased expression of tissue factor [157]
  • Increased D-dimer levels [198,199]
  • Fibrinogen deposition in tissues [20,203,204]
  • Venous and arterial thrombosis [196]
  • Systemic thromboembolic complications [175]

Author Contributions

V.L.B.: Conceptualization, writing (original draft), review and editing, J.R.M.: Conceptualization, writing (original draft), review and editing; C.M.Q.-J.: Review and editing and writing; E.D.H.: Review. M.M.T.: Review and editing, project administration, funding acquisition. V.V.C.: Conceptualization, funding acquisition, project administration, resources, supervision, validation, writing (original draft, review and editing). All authors have read and agreed to the published version of the manuscript.

Funding

This work received financial support from the National Institute of Science and Technology in Dengue and Host-microorganism Interaction (INCT dengue, CNPq, Brazil process 408527/2024-2), a program funded by the Brazilian National Science Council (CNPq, Brazil process 465425/2014-3) and Minas Gerais Foundation for Science (FAPEMIG, Brazil process 25036/2014-3), and from Rede de Pesquisa em Imunobiológicos e Biofármacos para terapias avançadas e inovadoras (ImunoBioFar), provided by FAPEMIG under process RED-00202-22, 9568-1 and FAPEMIG processes APQ-04650-23 and APQ-02618-23.

Acknowledgments

Thanks to L’Oréal-UNESCO-ABC ‘Para Mulheres na Ciência’ prize, granted to VVC.

Conflicts of Interest

The authors declare no competing financial interests or personal relationships that could have influenced the work reported in this article.

Abbreviations

The following abbreviations are used in this manuscript:
aPTTActivated partial thromboplastin time
ACE2Angiotensin-converting enzyme 2
ADPAdenosine diphosphate
ARDSRespiratory distress syndrome
CHIKVChikungunya virus
DENVDengue virus
DHFDengue hemorrhagic fever
DICDisseminated intravascular coagulation
EGLEndothelial glycocalyx-like layer
GPGlycoprotein
GPIbαGlycoprotein Ib alpha chain
HAHemagglutinin
HSCsHematopoietic stem cells
IAVInfluenza A virus
ICAM-1Intercellular adhesion molecule-1
ISGsInterferon-stimulated genes
MIFMigration inhibitory factors
MCP-1Monocyte chemoattractant protein –1
MKsMegakaryocytes
MLRMonocyte-to-lymphocyte ratio
MMPsMatrix metalloproteinases
MRPMyeloid-related protein
NANeuraminidase
NAP-2Neutrophil-activating peptide 2
NETsNeutrophil extracellular traps
NLRNeutrophil-to-lymphocyte ratio
NONitric oxide
NS1Non-structural protein 1
PAFPlatelet-activating factor
PAI-1Plasminogen activator inhibitor-1
PARProtease-activated receptor
PDWPlatelet distribution width
PF-4Platelet factor 4
PLRPlatelet-to-lymphocyte ratio
PMAsPlatelet–monocyte aggregates
PSGL-1P-selectin glycoprotein ligand-1
PTProthrombin time
ROSReactive oxygen species
SARS Severe acute respiratory syndrome
sCD40LSoluble CD40 ligand
SDSevere dengue
TFTissue factor
TLRToll-like receptor
TXA2Thromboxane A2
VCAM-1Vascular cell adhesion molecule-1
VEGFVascular endothelial growth factor
VLA-4Very late antigen-4

References

  1. Loof, T.G.; Mörgelin, M.; Johansson, L.; Oehmcke, S.; Olin, A.I.; Dickneite, G.; Norrby-Teglund, A.; Theopold, U.; Herwald, H. Coagulation, an Ancestral Serine Protease Cascade, Exerts a Novel Function in Early Immune Defense. Blood 2011, 118, 2589–2598. [Google Scholar] [CrossRef]
  2. Loof, T.G.; Schmidt, O.; Herwald, H.; Theopold, U. Coagulation Systems of Invertebrates and Vertebrates and Their Roles in Innate Immunity: The Same Side of Two Coins? J. Innate Immun. 2011, 3, 34–40. [Google Scholar] [CrossRef] [PubMed]
  3. Furie, B.; Furie, B.C. Mechanisms of Thrombus Formation. N. Engl. J. Med. 2008, 359, 938–949. [Google Scholar] [CrossRef] [PubMed]
  4. Engelmann, B.; Massberg, S. Thrombosis as an Intravascular Effector of Innate Immunity. Nat. Rev. Immunol. 2013, 13, 34–45. [Google Scholar] [CrossRef]
  5. Thon, J.N.; Italiano, J.E. Platelets: Production, Morphology and Ultrastructure. In Antiplatelet Agents; Springer: Berlin, Heidelberg, 2012; pp. 3–22. [Google Scholar]
  6. Asquith, N.L.; Carminita, E.; Camacho, V.; Rodriguez-Romera, A.; Stegner, D.; Freire, D.; Becker, I.C.; Machlus, K.R.; Khan, A.O.; Italiano, J.E. The Bone Marrow Is the Primary Site of Thrombopoiesis. Blood 2024, 143, 272–278. [Google Scholar] [CrossRef]
  7. Lefrançais, E.; Ortiz-Muñoz, G.; Caudrillier, A.; Mallavia, B.; Liu, F.; Sayah, D.M.; Thornton, E.E.; Headley, M.B.; David, T.; Coughlin, S.R.; et al. The Lung Is a Site of Platelet Biogenesis and a Reservoir for Haematopoietic Progenitors. Nature 2017, 544, 105–109. [Google Scholar] [CrossRef]
  8. Massberg, S.; Gawaz, M.; Grüner, S.; Schulte, V.; Konrad, I.; Zohlnhöfer, D.; Heinzmann, U.; Nieswandt, B. A Crucial Role of Glycoprotein VI for Platelet Recruitment to the Injured Arterial Wall In Vivo. J. Exp. Med. 2003, 197, 41–49. [Google Scholar] [CrossRef] [PubMed]
  9. Ruggeri, Z.M. Mechanisms Initiating Platelet Thrombus Formation. Thromb. Haemost. 1997, 78, 611–616. [Google Scholar] [CrossRef] [PubMed]
  10. Almskog, L.M.; Ågren, A. Thromboinflammation vs. Immunothrombosis: Strategies for Overcoming Anticoagulant Resistance in COVID-19 and Other Hyperinflammatory Diseases. Is ROTEM Helpful or Not? Front. Immunol. 2025, 16, 1599639. [Google Scholar] [CrossRef] [PubMed]
  11. Schrottmaier, W.C.; Assinger, A. The Concept of Thromboinflammation. Hamostaseologie 2024, 44, 021–030. [Google Scholar] [CrossRef] [PubMed]
  12. Dhanesha, N.; Patel, R.B.; Doddapattar, P.; Ghatge, M.; Flora, G.D.; Jain, M.; Thedens, D.; Olalde, H.; Kumskova, M.; Leira, E.C.; et al. PKM2 Promotes Neutrophil Activation and Cerebral Thromboinflammation: Therapeutic Implications for Ischemic Stroke. Blood 2022, 139, 1234–1245. [Google Scholar] [CrossRef] [PubMed]
  13. Polzin, A.; Benkhoff, M.; Thienel, M.; Barcik, M.; Mourikis, P.; Shchurovska, K.; Helten, C.; Ehreiser, V.; Zhe, Z.; von Wulffen, F.; et al. Long-Term FXa Inhibition Attenuates Thromboinflammation after Acute Myocardial Infarction and Stroke by Platelet Proteome Alteration. J. Thromb. Haemost. 2025, 23, 668–683. [Google Scholar] [CrossRef] [PubMed]
  14. Singh, K.K.; Gupta, A.; Forstner, D.; Guettler, J.; Ahrens, M.S.; Prakasan Sheeja, A.; Fatima, S.; Shamkeeva, S.; Lia, M.; Dathan-Stumpf, A.; et al. LMWH Prevents Thromboinflammation in the Placenta via HBEGF-AKT Signaling. Blood Adv. 2024, 8, 4756–4766. [Google Scholar] [CrossRef] [PubMed]
  15. Leal de Azeredo, E.; Solórzano, V.E.F.; de Oliveira, D.B.; Marinho, C.F.; de Souza, L.J.; da Cunha, R.V.; Damasco, P.V.; Kubelka, C.F.; De-Oliveira-Pinto, L.M. Increased Circulating Procoagulant and Anticoagulant Factors as TF and TFPI According to Severity or Infecting Serotypes in Human Dengue Infection. Microbes Infect. 2017, 19, 62–68. [Google Scholar] [CrossRef]
  16. Oliveira-Neto, J.T.d.; Souza, J.d.P.; Rodrigues, D.; Machado, M.R.; Alves, J.V.; Barros, P.R.; Bressan, A.F.; Silva, J.F.; Costa, T.J.; Costa, R.M.; et al. Acute Chikungunya Infection Induces Vascular Dysfunction by Directly Disrupting Redox Signaling in Endothelial Cells. Cells 2024, 13, 1770. [Google Scholar] [CrossRef]
  17. Ramacciotti, E.; Agati, L.B.; Aguiar, V.C.R.; Wolosker, N.; Guerra, J.C.; de Almeida, R.P.; Alves, J.C.; Lopes, R.D.; Wakefield, T.W.; Comerota, A.J.; et al. Zika and Chikungunya Virus and Risk for Venous Thromboembolism. Clin. Appl. Thromb. 2019, 25, 1076029618821184. [Google Scholar] [CrossRef] [PubMed]
  18. Bailey, A.L.; Kang, L.-I.; de Assis Barros D’Elia Zanella, L.G.F.; Silveira, C.G.T.; Ho, Y.-L.; Foquet, L.; Bial, G.; McCune, B.T.; Duarte-Neto, A.N.; Thomas, A.; et al. Consumptive Coagulopathy of Severe Yellow Fever Occurs Independently of Hepatocellular Tropism and Massive Hepatic Injury. Proc. Natl. Acad. Sci. USA 2020, 117, 32648–32656. [Google Scholar] [CrossRef]
  19. Mackman, N.; Grover, S.P.; Antoniak, S. Tissue Factor Expression, Extracellular Vesicles, and Thrombosis after Infection with the Respiratory Viruses Influenza A Virus and Coronavirus. J. Thromb. Haemost. 2021, 19, 2652–2658. [Google Scholar] [CrossRef]
  20. Ryu, J.K.; Yan, Z.; Montano, M.; Sozmen, E.G.; Dixit, K.; Suryawanshi, R.K.; Matsui, Y.; Helmy, E.; Kaushal, P.; Makanani, S.K.; et al. Fibrin Drives Thromboinflammation and Neuropathology in COVID-19. Nature 2024, 633, 905–913. [Google Scholar] [CrossRef] [PubMed]
  21. Huang, Y.-J.S.; Higgs, S.; Vanlandingham, D.L. Emergence and Re-Emergence of Mosquito-Borne Arboviruses. Curr. Opin. Virol. 2019, 34, 104–109. [Google Scholar] [CrossRef] [PubMed]
  22. Guzman, M.G.; Gubler, D.J.; Izquierdo, A.; Martinez, E.; Halstead, S.B. Dengue Infection. Nat. Rev. Dis. Prim. 2016, 2, 16055. [Google Scholar] [CrossRef] [PubMed]
  23. WHO. Dengue: Guidelines for Diagnosis, Treatment, Prevention and Control; World Health Organization: Geneva, Switzerland, 2009. [Google Scholar]
  24. OMS. Recommendations for Treatment. Psychiatr. News 2009, 41, 29. [Google Scholar] [CrossRef]
  25. Bhatt, S.; Gething, P.W.; Brady, O.J.; Messina, J.P.; Farlow, A.W.; Moyes, C.L.; Drake, J.M.; Brownstein, J.S.; Hoen, A.G.; Sankoh, O.; et al. The Global Distribution and Burden of Dengue. Nature 2013, 496, 504–507. [Google Scholar] [CrossRef]
  26. Khetarpal, N.; Khanna, I. Dengue Fever: Causes, Complications, and Vaccine Strategies. J. Immunol. Res. 2016, 2016, 6803098. [Google Scholar] [CrossRef]
  27. Ojha, A.; Nandi, D.; Batra, H.; Singhal, R.; Annarapu, G.K.; Bhattacharyya, S.; Seth, T.; Dar, L.; Medigeshi, G.R.; Vrati, S.; et al. Platelet Activation Determines the Severity of Thrombocytopenia in Dengue Infection. Sci. Rep. 2017, 7, 41697. [Google Scholar] [CrossRef] [PubMed]
  28. Barbosa-Lima, G.; Hottz, E.D.; de Assis, E.F.; Liechocki, S.; Souza, T.M.L.; Zimmerman, G.A.; Bozza, F.A.; Bozza, P.T. Dengue Virus-Activated Platelets Modulate Monocyte Immunometabolic Response through Lipid Droplet Biogenesis and Cytokine Signaling. J. Leukoc. Biol. 2020, 108, 1293–1306. [Google Scholar] [CrossRef]
  29. Syenina, A.; Saron, W.A.A.; Jagaraj, C.J.; Bibi, S.; Arock, M.; Gubler, D.J.; Rathore, A.P.S.; Abraham, S.N.; St John, A.L. Th1-Polarized, Dengue Virus-Activated Human Mast Cells Induce Endothelial Transcriptional Activation and Permeability. Viruses 2020, 12, 1379. [Google Scholar] [CrossRef]
  30. Adane, T.; Getawa, S. Coagulation Abnormalities in Dengue Fever Infection: A Systematic Review and Meta-Analysis. PLoS Negl. Trop. Dis. 2021, 15, e0009666. [Google Scholar] [CrossRef]
  31. Rocha, F. Created in BioRender. Figure 1: Thromboinflammatory Mechanisms Are Induced by Arboviruses. BioRender. 2025. Available online: https://BioRender.com/daeu735 (accessed on 10 June 2025).
  32. Quirino-Teixeira, A.C.; Andrade, F.B.; Pinheiro, M.B.M.; Rozini, S.V.; Hottz, E.D. Platelets in Dengue Infection: More than a Numbers Game. Platelets 2022, 33, 176–183. [Google Scholar] [CrossRef]
  33. Yang, M.; Xiao, D.W.; Li, K.K.H.; Li, C.K.; Chik, K.W.; Fok, T.F. Lung Damage May Induce Thrombocytopenia. Blood 2004, 104, 3026. [Google Scholar] [CrossRef]
  34. Moragas, L.J.; Arruda, L.V.; Oliveira, L.d.L.S.; Alves, F.d.A.V.; Salomão, N.G.; da Silva, J.F.R.; Basílio-de-Oliveira, C.A.; Basílio-de-Oliveira, R.P.; Mohana-Borges, R.; Azevedo, C.G.; et al. Detection of Viral Antigen and Inflammatory Mediators in Fatal Pediatric Dengue: A Study on Lung Immunopathogenesis. Front. Immunol. 2025, 16, 1487284. [Google Scholar] [CrossRef]
  35. Michels, M.; Alisjahbana, B.; de Groot, P.G.; Indrati, A.R.; Fijnheer, R.; Puspita, M.; Dewi, I.M.W.; van de Wijer, L.; de Boer, E.M.S.; Roest, M.; et al. Platelet Function Alterations in Dengue Are Associated with Plasma Leakage. Thromb. Haemost. 2014, 112, 352–362. [Google Scholar] [CrossRef]
  36. Hottz, E.D.; Oliveira, M.F.; Nunes, P.C.G.; Nogueira, R.M.R.; Valls-de-Souza, R.; Da Poian, A.T.; Weyrich, A.S.; Zimmerman, G.A.; Bozza, P.T.; Bozza, F.A. Dengue Induces Platelet Activation, Mitochondrial Dysfunction and Cell Death through Mechanisms That Involve DC-SIGN and Caspases. J. Thromb. Haemost. 2013, 11, 951–962. [Google Scholar] [CrossRef] [PubMed]
  37. Wills, B.A.; Oragui, E.E.; Stephens, A.C.; Daramola, O.A.; Dung, N.M.; Loan, H.T.; Chau, N.V.; Chambers, M.; Stepniewska, K.; Farrar, J.J.; et al. Coagulation Abnormalities in Dengue Hemorrhagic Fever: Serial Investigations in 167 Vietnamese Children with Dengue Shock Syndrome. Clin. Infect. Dis. 2002, 35, 277–285. [Google Scholar] [CrossRef] [PubMed]
  38. Banerjee, A.; Tripathi, A.; Duggal, S.; Banerjee, A.; Vrati, S. Dengue Virus Infection Impedes Megakaryopoiesis in MEG-01 Cells Where the Virus Envelope Protein Interacts with the Transcription Factor TAL-1. Sci. Rep. 2020, 10, 19587. [Google Scholar] [CrossRef] [PubMed]
  39. Vogt, M.B.; Lahon, A.; Arya, R.P.; Spencer Clinton, J.L.; Rico-Hesse, R. Dengue Viruses Infect Human Megakaryocytes, with Probable Clinical Consequences. PLoS Negl. Trop. Dis. 2019, 13, e0007837. [Google Scholar] [CrossRef]
  40. Sridharan, A.; Chen, Q.; Tang, K.F.; Ooi, E.E.; Hibberd, M.L.; Chen, J. Inhibition of Megakaryocyte Development in the Bone Marrow Underlies Dengue Virus-Induced Thrombocytopenia in Humanized Mice. J. Virol. 2013, 87, 11648–11658. [Google Scholar] [CrossRef]
  41. Kaur, J.; Rawat, Y.; Sood, V.; Periwal, N.; Rathore, D.K.; Kumar, S.; Kumar, N.; Bhattacharyya, S. Replication of Dengue Virus in K562-Megakaryocytes Induces Suppression in the Accumulation of Reactive Oxygen Species. Front. Microbiol. 2022, 12, 784070. [Google Scholar] [CrossRef] [PubMed]
  42. Clark, K.B.; Noisakran, S.; Onlamoon, N.; Hsiao, H.-M.; Roback, J.; Villinger, F.; Ansari, A.A.; Perng, G.C. Multiploid CD61+ Cells Are the Pre-Dominant Cell Lineage Infected during Acute Dengue Virus Infection in Bone Marrow. PLoS ONE 2012, 7, e52902. [Google Scholar] [CrossRef]
  43. La Russa, V.F.; Innis, B.L. 11 Mechanisms of Dengue Virus-Induced Bone Marrow Suppression. Baillieres. Clin. Haematol. 1995, 8, 249–270. [Google Scholar] [CrossRef]
  44. Poirault-Chassac, S.; Six, E.; Catelain, C.; Lavergne, M.; Villeval, J.-L.; Vainchenker, W.; Lauret, E. Notch/Delta4 Signaling Inhibits Human Megakaryocytic Terminal Differentiation. Blood 2010, 116, 5670–5678. [Google Scholar] [CrossRef] [PubMed]
  45. Campbell, R.A.; Schwertz, H.; Hottz, E.D.; Rowley, J.W.; Manne, B.K.; Washington, A.V.; Hunter-Mellado, R.; Tolley, N.D.; Christensen, M.; Eustes, A.S.; et al. Human Megakaryocytes Possess Intrinsic Antiviral Immunity through Regulated Induction of IFITM3. Blood 2019, 133, 2013–2026. [Google Scholar] [CrossRef]
  46. Morodomi, Y.; Kanaji, S.; Sullivan, B.M.; Zarpellon, A.; Orje, J.N.; Won, E.; Shapiro, R.; Yang, X.-L.; Ruf, W.; Schimmel, P.; et al. Inflammatory Platelet Production Stimulated by Tyrosyl-TRNA Synthetase Mimicking Viral Infection. Proc. Natl. Acad. Sci. USA 2022, 119, e2212659119. [Google Scholar] [CrossRef]
  47. Pozner, R.G.; Ure, A.E.; Jaquenod de Giusti, C.; D’Atri, L.P.; Italiano, J.E.; Torres, O.; Romanowski, V.; Schattner, M.; Gómez, R.M. Junín Virus Infection of Human Hematopoietic Progenitors Impairs In Vitro Proplatelet Formation and Platelet Release via a Bystander Effect Involving Type I IFN Signaling. PLoS Pathog. 2010, 6, e1000847. [Google Scholar] [CrossRef] [PubMed]
  48. Ravid, K.; Lu, J.; Zimmet, J.M.; Jones, M.R. Roads to Polyploidy: The Megakaryocyte Example. J. Cell. Physiol. 2002, 190, 7–20. [Google Scholar] [CrossRef] [PubMed]
  49. Trugilho, M.R.d.O.; Hottz, E.D.; Brunoro, G.V.F.; Teixeira-Ferreira, A.; Carvalho, P.C.; Salazar, G.A.; Zimmerman, G.A.; Bozza, F.A.; Bozza, P.T.; Perales, J. Platelet Proteome Reveals Novel Pathways of Platelet Activation and Platelet-Mediated Immunoregulation in Dengue. PLOS Pathog. 2017, 13, e1006385. [Google Scholar] [CrossRef]
  50. Wan, S.-W.; Yang, Y.-W.; Chu, Y.-T.; Lin, C.-F.; Chang, C.-P.; Yeh, T.-M.; Anderson, R.; Lin, Y.-S. Anti-Dengue Virus Nonstructural Protein 1 Antibodies Contribute to Platelet Phagocytosis by Macrophages. Thromb. Haemost. 2016, 115, 646–656. [Google Scholar] [CrossRef]
  51. Alonzo, M.T.G.; Lacuesta, T.L.V.; Dimaano, E.M.; Kurosu, T.; Suarez, L.C.; Mapua, C.A.; Akeda, Y.; Matias, R.R.; Kuter, D.J.; Nagata, S.; et al. Platelet Apoptosis and Apoptotic Platelet Clearance by Macrophages in Secondary Dengue Virus Infections. J. Infect. Dis. 2012, 205, 1321–1329. [Google Scholar] [CrossRef]
  52. 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.; Weyrich, A.S.; Zimmerman, G.A.; Bozza, F.A.; Bozza, P.T. Platelet Activation and Apoptosis Modulate Monocyte Inflammatory Responses in Dengue. J. Immunol. 2014, 193, 1864–1872. [Google Scholar] [CrossRef] [PubMed]
  53. Masri, M.F.B.; Mantri, C.K.; Rathore, A.P.S.; St. John, A.L. Peripheral Serotonin Causes Dengue Virus–Induced Thrombocytopenia through 5HT2 Receptors. Blood 2019, 133, 2325–2337. [Google Scholar] [CrossRef] [PubMed]
  54. Khazali, A.S.; Hadrawi, W.H.; Ibrahim, F.; Othman, S.; Nor Rashid, N. Thrombocytopenia in Dengue Infection: Mechanisms and a Potential Application. Expert Rev. Mol. Med. 2024, 26, e26. [Google Scholar] [CrossRef] [PubMed]
  55. Peshkova, A.D.; Saliakhutdinova, S.M.; Sounbuli, K.; Selivanova, Y.A.; Andrianova, I.A.; Khabirova, A.I.; Litvinov, R.I.; Weisel, J.W. The Differential Formation and Composition of Leukocyte-Platelet Aggregates Induced by Various Cellular Stimulants. Thromb. Res. 2024, 241, 109092. [Google Scholar] [CrossRef]
  56. Furie, B.; Furie, B.C. Role of Platelet P-Selectin and Microparticle PSGL-1 in Thrombus Formation. Trends Mol. Med. 2004, 10, 171–178. [Google Scholar] [CrossRef]
  57. Moore, K.L.; Stults, N.L.; Diaz, S.; Smith, D.F.; Cummings, R.D.; Varki, A.; McEver, R.P. Identification of a Specific Glycoprotein Ligand for P-Selectin (CD62) on Myeloid Cells. J. Cell Biol. 1992, 118, 445–456. [Google Scholar] [CrossRef] [PubMed]
  58. Xu, T.; Zhang, L.; Geng, Z.H.; Wang, H.-B.; Wang, J.-T.; Chen, M.; Geng, J.-G. P-Selectin Cross-Links PSGL-1 and Enhances Neutrophil Adhesion to Fibrinogen and ICAM-1 in a Src Kinase-Dependent, but GPCR-Independent Mechanism. Cell Adh. Migr. 2007, 1, 115–123. [Google Scholar] [CrossRef] [PubMed]
  59. Tsai, J.-J.; Jen, Y.-H.; Chang, J.-S.; Hsiao, H.-M.; Noisakran, S.; Perng, G.C. Frequency Alterations in Key Innate Immune Cell Components in the Peripheral Blood of Dengue Patients Detected by FACS Analysis. J. Innate Immun. 2011, 3, 530–540. [Google Scholar] [CrossRef] [PubMed]
  60. Furman, M.I.; Benoit, S.E.; Barnard, M.R.; Valeri, C.R.; Borbone, M.L.; Becker, R.C.; Hechtman, H.B.; Michelson, A.D. Increased Platelet Reactivity and Circulating Monocyte-Platelet Aggregates in Patients with Stable Coronary Artery Disease. J. Am. Coll. Cardiol. 1998, 31, 352–358. [Google Scholar] [CrossRef] [PubMed]
  61. Allen, N.; Barrett, T.J.; Guo, Y.; Nardi, M.; Ramkhelawon, B.; Rockman, C.B.; Hochman, J.S.; Berger, J.S. Circulating Monocyte-Platelet Aggregates Are a Robust Marker of Platelet Activity in Cardiovascular Disease. Atherosclerosis 2019, 282, 11–18. [Google Scholar] [CrossRef]
  62. Passacquale, G.; Vamadevan, P.; Pereira, L.; Hamid, C.; Corrigall, V.; Ferro, A. Monocyte-Platelet Interaction Induces a Pro-Inflammatory Phenotype in Circulating Monocytes. PLoS ONE 2011, 6, e25595. [Google Scholar] [CrossRef]
  63. Lien, T.-S.; Chan, H.; Sun, D.-S.; Wu, J.-C.; Lin, Y.-Y.; Lin, G.-L.; Chang, H.-H. Exposure of Platelets to Dengue Virus and Envelope Protein Domain III Induces Nlrp3 Inflammasome-Dependent Platelet Cell Death and Thrombocytopenia in Mice. Front. Immunol. 2021, 12, 616394. [Google Scholar] [CrossRef]
  64. Sung, P.-S.; Huang, T.-F.; Hsieh, S.-L. Extracellular Vesicles from CLEC2-Activated Platelets Enhance Dengue Virus-Induced Lethality via CLEC5A/TLR2. Nat. Commun. 2019, 10, 2402. [Google Scholar] [CrossRef]
  65. Garishah, F.M.; Rother, N.; Riswari, S.F.; Alisjahbana, B.; Overheul, G.J.; van Rij, R.P.; van der Ven, A.; van der Vlag, J.; de Mast, Q. Neutrophil Extracellular Traps in Dengue Are Mainly Generated NOX-Independently. Front. Immunol. 2021, 12, 629167. [Google Scholar] [CrossRef]
  66. Böer, L.M.; Junqueira, I.C.; Nascimento, T.C.d.; Guilarde, A.O.; Féres, V.C.d.R.; Alcântara, K.C.d. Monocyte-Lymphocyte, Neutrophil-Lymphocyte, and Platelet-Lymphocyte Ratios as Inflammatory Biomarkers of Clinical Dengue Severity. Biosci. J. 2024, 40, e40038. [Google Scholar] [CrossRef]
  67. Li, N.; Ji, Q.; Hjemdahl, P. Platelet–Lymphocyte Conjugation Differs between Lymphocyte Subpopulations. J. Thromb. Haemost. 2006, 4, 874–881. [Google Scholar] [CrossRef] [PubMed]
  68. Amin, A.; Nikdoust, F.; Khorram, S.; Marashi, S.M.; Ghanavati, P.; Ameri, F.; Akbarzadeh, A.; Hasanvand, A.; Khodakarim, N. Dengue Virus Infection: How Platelet-Leukocyte Crosstalk Shapes Thrombotic Events and Inflammation. Mol. Biol. Rep. 2025, 52, 119. [Google Scholar] [CrossRef]
  69. Vats, R.; Brzoska, T.; Bennewitz, M.F.; Jimenez, M.A.; Pradhan-Sundd, T.; Tutuncuoglu, E.; Jonassaint, J.; Gutierrez, E.; Watkins, S.C.; Shiva, S.; et al. Platelet Extracellular Vesicles Drive Inflammasome–IL-1β–Dependent Lung Injury in Sickle Cell Disease. Am. J. Respir. Crit. Care Med. 2020, 201, 33–46. [Google Scholar] [CrossRef] [PubMed]
  70. Sullivan, V.; Hawley, A.; Farris, D.; Knipp, B.; Varga, A.; Wrobleski, S.; Thanapron, P.; Eagleton, M.; Myers, D.; Fowlkes, J.; et al. Decrease in Fibrin Content of Venous Thrombi in Selectin-Deficient Mice. J. Surg. Res. 2003, 109, 1–7. [Google Scholar] [CrossRef]
  71. Yokoyama, S.; Ikeda, H.; Haramaki, N.; Yasukawa, H.; Murohara, T.; Imaizumi, T. Platelet P-Selectin Plays an Important Role in Arterial Thrombogenesis by Forming Large Stable Platelet-Leukocyte Aggregates. J. Am. Coll. Cardiol. 2005, 45, 1280–1286. [Google Scholar] [CrossRef]
  72. Lee, T.H.; Lee, L.K.; Lye, D.C.; Leo, Y.S. Current Management of Severe Dengue Infection. Expert Rev. Anti. Infect. Ther. 2017, 15, 67–78. [Google Scholar] [CrossRef] [PubMed]
  73. Yacoub, S.; Wertheim, H.; Simmons, C.P.; Screaton, G.; Wills, B. Microvascular and Endothelial Function for Risk Prediction in Dengue: An Observational Study. Lancet 2015, 385, S102. [Google Scholar] [CrossRef] [PubMed]
  74. Puerta-Guardo, H.; Glasner, D.R.; Harris, E. Dengue Virus NS1 Disrupts the Endothelial Glycocalyx, Leading to Hyperpermeability. PLOS Pathog. 2016, 12, e1005738. [Google Scholar] [CrossRef]
  75. Modhiran, N.; Watterson, D.; Muller, D.A.; Panetta, A.K.; Sester, D.P.; Liu, L.; Hume, D.A.; Stacey, K.J.; Young, P.R. Dengue Virus NS1 Protein Activates Cells via Toll-like Receptor 4 and Disrupts Endothelial Cell Monolayer Integrity. Sci. Transl. Med. 2015, 7, 304ra142. [Google Scholar] [CrossRef]
  76. Chen, H.-R.; Chuang, Y.-C.; Chao, C.-H.; Yeh, T.-M. Macrophage Migration Inhibitory Factor Induces Vascular Leakage via Autophagy. Biol. Open 2015, 4, 244–252. [Google Scholar] [CrossRef]
  77. Chen, H.-R.; Chuang, Y.-C.; Lin, Y.-S.; Liu, H.-S.; Liu, C.-C.; Perng, G.-C.; Yeh, T.-M. Dengue Virus Nonstructural Protein 1 Induces Vascular Leakage through Macrophage Migration Inhibitory Factor and Autophagy. PLoS Negl. Trop. Dis. 2016, 10, e0004828. [Google Scholar] [CrossRef]
  78. Quirino-Teixeira, A.C.; Rozini, S.V.; Barbosa-Lima, G.; Coelho, D.R.; Carneiro, P.H.; Mohana-Borges, R.; Bozza, P.T.; Hottz, E.D. Inflammatory Signaling in Dengue-Infected Platelets Requires Translation and Secretion of Nonstructural Protein 1. Blood Adv. 2020, 4, 2018–2031. [Google Scholar] [CrossRef] [PubMed]
  79. Chao, C.-H.; Wu, W.-C.; Lai, Y.-C.; Tsai, P.-J.; Perng, G.-C.; Lin, Y.-S.; Yeh, T.-M. Dengue Virus Nonstructural Protein 1 Activates Platelets via Toll-like Receptor 4, Leading to Thrombocytopenia and Hemorrhage. PLOS Pathog. 2019, 15, e1007625. [Google Scholar] [CrossRef] [PubMed]
  80. Assunção-Miranda, I.; Amaral, F.A.; Bozza, F.A.; Fagundes, C.T.; Sousa, L.P.; Souza, D.G.; Pacheco, P.; Barbosa-Lima, G.; Gomes, R.N.; Bozza, P.T.; et al. Contribution of Macrophage Migration Inhibitory Factor to the Pathogenesis of Dengue Virus Infection. FASEB J. 2010, 24, 218–228. [Google Scholar] [CrossRef]
  81. Chuang, Y.-C.; Lei, H.-Y.; Liu, H.-S.; Lin, Y.-S.; Fu, T.-F.; Yeh, T.-M. Macrophage Migration Inhibitory Factor Induced by Dengue Virus Infection Increases Vascular Permeability. Cytokine 2011, 54, 222–231. [Google Scholar] [CrossRef]
  82. Anderson, R.; Wang, S.; Osiowy, C.; Issekutz, A.C. Activation of Endothelial Cells via Antibody-Enhanced Dengue Virus Infection of Peripheral Blood Monocytes. J. Virol. 1997, 71, 4226–4232. [Google Scholar] [CrossRef] [PubMed]
  83. Peyrefitte, C.N.; Pastorino, B.; Grau, G.E.; Lou, J.; Tolou, H.; Couissinier-Paris, P. Dengue Virus Infection of Human Microvascular Endothelial Cells from Different Vascular Beds Promotes Both Common and Specific Functional Changes. J. Med. Virol. 2006, 78, 229–242. [Google Scholar] [CrossRef] [PubMed]
  84. Furuta, T.; Murao, L.A.; Lan, N.T.P.; Huy, N.T.; Huong, V.T.Q.; Thuy, T.T.; Tham, V.D.; Nga, C.T.P.; Ha, T.T.N.; Ohmoto, Y.; et al. Association of Mast Cell-Derived VEGF and Proteases in Dengue Shock Syndrome. PLoS Negl. Trop. Dis. 2012, 6, e1505. [Google Scholar] [CrossRef]
  85. Brown, M.G.; King, C.A.; Sherren, C.; Marshall, J.S.; Anderson, R. A Dominant Role for FcγRII in Antibody-Enhanced Dengue Virus Infection of Human Mast Cells and Associated CCL5 Release. J. Leukoc. Biol. 2006, 80, 1242–1250. [Google Scholar] [CrossRef] [PubMed]
  86. King, C.A.; Marshall, J.S.; Alshurafa, H.; Anderson, R. Release of Vasoactive Cytokines by Antibody-Enhanced Dengue Virus Infection of a Human Mast Cell/Basophil Line. J. Virol. 2000, 74, 7146–7150. [Google Scholar] [CrossRef]
  87. Phan, D.T.; Ha, N.T.; Thuc, L.T.; Diet, N.H.; Phu, L.V.; Ninh, L.Y.; An, V.T. Some Changes in Immunity and Blood in Relation to Clinical States of Dengue Hemorrhagic Fever Patients in Vietnam. Haematologia 1991, 24, 13–21. [Google Scholar] [PubMed]
  88. Tuchinda, M.; Dhorranintra, B.; Tuchinda, P. Histamine Content in 24-Hour Urine in Patients with Dengue Haemorrhagic Fever. Southeast Asian J. Trop. Med. Public Health 1977, 8, 80–83. [Google Scholar]
  89. St John, A.L. Influence of Mast Cells on Dengue Protective Immunity and Immune Pathology. PLoS Pathog. 2013, 9, e1003783. [Google Scholar] [CrossRef]
  90. King, C.A.; Anderson, R.; Marshall, J.S. Dengue Virus Selectively Induces Human Mast Cell Chemokine Production. J. Virol. 2002, 76, 8408–8419. [Google Scholar] [CrossRef] [PubMed]
  91. Brown, M.G.; Hermann, L.L.; Issekutz, A.C.; Marshall, J.S.; Rowter, D.; Al-Afif, A.; Anderson, R. Dengue Virus Infection of Mast Cells Triggers Endothelial Cell Activation. J. Virol. 2011, 85, 1145–1150. [Google Scholar] [CrossRef]
  92. Brown, M.G.; McAlpine, S.M.; Huang, Y.Y.; Haidl, I.D.; Al-Afif, A.; Marshall, J.S.; Anderson, R. RNA Sensors Enable Human Mast Cell Anti-Viral Chemokine Production and IFN-Mediated Protection in Response to Antibody-Enhanced Dengue Virus Infection. PLoS ONE 2012, 7, e34055. [Google Scholar] [CrossRef] [PubMed]
  93. Kannan, A.; Narayanan, K.; Sasikumar, S.; Philipose, J.; Surendran, S. Coagulopathy in Dengue Fever Patients. Int. J. Res. Med. Sci. 2014, 2, 1070. [Google Scholar] [CrossRef]
  94. Wills, B.; Van Ngoc, T.; Van, N.T.H.; Thuy, T.T.T.; Thuy, T.T.N.; Dung, N.M.; Diet, T.V.; Van Vinh Chau, N.; Trung, D.T.; Farrar, J. Hemostatic Changes in Vietnamese Children with Mild Dengue Correlate with the Severity of Vascular Leakage Rather than Bleeding. Am. J. Trop. Med. Hyg. 2009, 81, 638–644. [Google Scholar] [CrossRef]
  95. Avila-Aguero, M.L.; Avila-Aguero, C.R.; Um, S.L.; Soriano-Fallas, A.; Cañas-Coto, A.; Yan, S.B. Systemic Host Inflammatory and Coagulation Response in the Dengue Virus Primo-Infection. Cytokine 2004, 27, 173–179. [Google Scholar] [CrossRef]
  96. Ho, T.-S.; Wang, S.-M.; Lin, Y.-S.; Liu, C.-C. Clinical and Laboratory Predictive Markers for Acute Dengue Infection. J. Biomed. Sci. 2013, 20, 75. [Google Scholar] [CrossRef] [PubMed]
  97. Huang, Y.-H.; Liu, C.-C.; Wang, S.-T.; Lei, H.-Y.; Liu, H.-S.; Lin, Y.-S.; Wu, H.-L.; Yeh, T.-M. Activation of Coagulation and Fibrinolysis during Dengue Virus Infection. J. Med. Virol. 2001, 63, 247–251. [Google Scholar] [CrossRef] [PubMed]
  98. Alsaffar, H.; Martino, N.; Garrett, J.P.; Adam, A.P. Interleukin-6 Promotes a Sustained Loss of Endothelial Barrier Function via Janus Kinase-Mediated STAT3 Phosphorylation and de Novo Protein Synthesis. Am. J. Physiol. Physiol. 2018, 314, C589–C602. [Google Scholar] [CrossRef] [PubMed]
  99. Levi, M.; ten Cate, H. Disseminated Intravascular Coagulation. N. Engl. J. Med. 1999, 341, 586–592. [Google Scholar] [CrossRef]
  100. Trotta, R.F.; Diehl, L.F.; Shorr, A.F.; Hanzel, G.S.; Alkins, S.A. D-Dimer Assay Predicts Mortality in Critically Ill Patients without Disseminated Intravascular Coagulation or Venous Thromboembolic Disease. Intensive Care Med. 1999, 25, 207–210. [Google Scholar] [CrossRef]
  101. Nurnaningsih; Sunbanu, S.E.; Rusmawatiningtyas, D.; Arguni, E.; Makrufardi, F.; Kumara, I.F. Disseminated Intravascular Coagulation Initial Score as a Predictor of Mortality in Children with Dengue Shock Syndrome: A Retrospective Cohort Study. Ann. Med. Surg. 2022, 79, 103890. [Google Scholar] [CrossRef]
  102. Das, S.; Mallik, M.H.; Chattopadyay, P.; Mallick, S.; Karmakar, D.; Ghora, S.; Begum, F.; Chatterjee, B.; Thagriki, D.S.; Srivastava, A.K.; et al. Dengue Virus NS1 Leads to Downregulation of HNF4 Alpha in Liver Cells Resulting in a Decrease in Coagulation Factors I, V, X, and XIII, Contributing to Coagulopathy. J. Virol. 2024, 98, e01418-24. [Google Scholar] [CrossRef]
  103. Geisbert, T.W.; Hensley, L.E.; Jahrling, P.B.; Larsen, T.; Geisbert, J.B.; Paragas, J.; Young, H.A.; Fredeking, T.M.; Rote, W.E.; Vlasuk, G.P. Treatment of Ebola Virus Infection with a Recombinant Inhibitor of Factor VIIa/Tissue Factor: A Study in Rhesus Monkeys. Lancet 2003, 362, 1953–1958. [Google Scholar] [CrossRef]
  104. Geisbert, T.W.; Young, H.A.; Jahrling, P.B.; Davis, K.J.; Kagan, E.; Hensley, L.E. Mechanisms Underlying Coagulation Abnormalities in Ebola Hemorrhagic Fever: Overexpression of Tissue Factor in Primate Monocytes/Macrophages Is a Key Event. J. Infect. Dis. 2003, 188, 1618–1629. [Google Scholar] [CrossRef] [PubMed]
  105. Francischetti, I.M.B.; Seydel, K.B.; Monteiro, R.Q. Blood Coagulation, Inflammation, and Malaria. Microcirculation 2008, 15, 81–107. [Google Scholar] [CrossRef]
  106. de Azeredo, E.L.; Kubelka, C.F.; Alburquerque, L.M.; Barbosa, L.S.; Damasco, P.V.; Ávila, C.A.L.; Motta-Castro, A.R.C.; da Cunha, R.V.; Monteiro, R.Q. Tissue Factor Expression on Monocytes from Patients with Severe Dengue Fever. Blood Cells Mol. Dis. 2010, 45, 334–335. [Google Scholar] [CrossRef]
  107. Huerta-Zepeda, A.; Cabello-Gutiérrez, C.; Cime-Castillo, J.; Monroy-Martínez, V.; Manjarrez-Zavala, M.E.; Gutiérrez-Rodríguez, M.; Izaguirre, R.; Ruiz-Ordaz, B. Crosstalk between Coagulation and Inflammation during Dengue Virus Infection. Thromb. Haemost. 2008, 99, 936–943. [Google Scholar] [CrossRef] [PubMed]
  108. Georgakopoulou, V.; Taskou, C.; Sarantaki, A.; Spandidos, D.; Gourounti, K.; Chaniotis, D.; Beloukas, A. Vector-borne Infectious Diseases in Pregnancy in the Era of Climate Change: A Focus on Mosquito- and Tick-borne Pathogens (Review). Exp. Ther. Med. 2025, 30, 174. [Google Scholar] [CrossRef] [PubMed]
  109. Steward, K.; Raja, A. Physiology, Ovulation and Basal Body Temperature; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  110. Goicochea-Ríos, E.d.S.; Otiniano, N.M.; Rojas-Infantas, L.d.C.; Ocaña-Gutiérrez, V.R.; Gómez-Goicochea, N.I. Dengue Infection during Pregnancy and the Occurrence of Pathological Neonatal Outcome: A Systematic Review and Meta-Analysis. F1000Research 2025, 13, 1523. [Google Scholar] [CrossRef] [PubMed]
  111. Chen, L.H.; Wilson, M.E. Update on Non-Vector Transmission of Dengue: Relevant Studies with Zika and Other Flaviviruses. Trop. Dis. Travel Med. Vaccines 2016, 2, 15. [Google Scholar] [CrossRef]
  112. Kerdpanich, A.; Watanaveeradej, V.; Samakoses, R.; Chumnanvanakij, S.; Chulyamitporn, T.; Sumeksri, P.; Vuthiwong, C.; Kounruang, C.; Nisalak, A.; Endy, T. Perinatal Dengue Infection. Southeast Asian J. Trop. Med. Public Health 2001, 32, 488–493. [Google Scholar]
  113. Basurko, C.; Matheus, S.; Hildéral, H.; Everhard, S.; Restrepo, M.; Cuadro-Alvarez, E.; Lambert, V.; Boukhari, R.; Duvernois, J.-P.; Favre, A.; et al. Estimating the Risk of Vertical Transmission of Dengue: A Prospective Study. Am. J. Trop. Med. Hyg. 2018, 98, 1826–1832. [Google Scholar] [CrossRef]
  114. Machain-Williams, C.; Raga, E.; Baak-Baak, C.M.; Kiem, S.; Blitvich, B.J.; Ramos, C. Maternal, Fetal, and Neonatal Outcomes in Pregnant Dengue Patients in Mexico. BioMed Res. Int. 2018, 2018, 9643083. [Google Scholar] [CrossRef] [PubMed]
  115. Mubashir, M.; Ahmed, K.S.; Mubashir, H.; Quddusi, A.; Farooq, A.; Ahmed, S.I.; Jamil, B.; Qureshi, R. Dengue and Malaria Infections in Pregnancy. Wien. Klin. Wochenschr. 2020, 132, 188–196. [Google Scholar] [CrossRef]
  116. Paixão, E.S.; Teixeira, M.G.; Costa, M.d.C.N.; Rodrigues, L.C. Dengue during Pregnancy and Adverse Fetal Outcomes: A Systematic Review and Meta-Analysis. Lancet Infect. Dis. 2016, 16, 857–865. [Google Scholar] [CrossRef] [PubMed]
  117. Vouga, M.; Chiu, Y.-C.; Pomar, L.; de Meyer, S.V.; Masmejan, S.; Genton, B.; Musso, D.; Baud, D.; Stojanov, M. Dengue, Zika and Chikungunya during Pregnancy: Pre- and Post-Travel Advice and Clinical Management. J. Travel Med. 2019, 26, taz077. [Google Scholar] [CrossRef] [PubMed]
  118. Brar, R.; Sikka, P.; Suri, V.; Singh, M.P.; Suri, V.; Mohindra, R.; Biswal, M. Maternal and Fetal Outcomes of Dengue Fever in Pregnancy: A Large Prospective and Descriptive Observational Study. Arch. Gynecol. Obstet. 2021, 304, 91–100. [Google Scholar] [CrossRef]
  119. Nunes, P.C.G.; Daumas, R.P.; Sánchez-Arcila, J.C.; Nogueira, R.M.R.; Horta, M.A.P.; dos Santos, F.B. 30 Years of Fatal Dengue Cases in Brazil: A Review. BMC Public Health 2019, 19, 329. [Google Scholar] [CrossRef]
  120. Hariyanto, H.; Yahya, C.Q.; Wibowo, P.; Tampubolon, O.E. Management of Severe Dengue Hemorrhagic Fever and Bleeding Complications in a Primigravida Patient: A Case Report. J. Med. Case Rep. 2016, 10, 357. [Google Scholar] [CrossRef]
  121. Thaithumyanon, P.; Thisyakorn, U.; Deerojnawong, J.; Innis, B.L. Dengue Infection Complicated by Severe Hemorrhage and Vertical Transmission in a Parturient Woman. Clin. Infect. Dis. 1994, 18, 248–249. [Google Scholar] [CrossRef] [PubMed]
  122. Kril, V.; Aïqui-Reboul-Paviet, O.; Briant, L.; Amara, A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu. Rev. Virol. 2021, 8, 327–347. [Google Scholar] [CrossRef] [PubMed]
  123. Coffey, L.; Failloux, A.-B.; Weaver, S. Chikungunya Virus–Vector Interactions. Viruses 2014, 6, 4628–4663. [Google Scholar] [CrossRef] [PubMed]
  124. van Aalst, M.; Nelen, C.M.; Goorhuis, A.; Stijnis, C.; Grobusch, M.P. Long-Term Sequelae of Chikungunya Virus Disease: A Systematic Review. Travel Med. Infect. Dis. 2017, 15, 8–22. [Google Scholar] [CrossRef] [PubMed]
  125. Sourisseau, M.; Schilte, C.; Casartelli, N.; Trouillet, C.; Guivel-Benhassine, F.; Rudnicka, D.; Sol-Foulon, N.; Roux, K.L.; Prevost, M.-C.; Fsihi, H.; et al. Characterization of Reemerging Chikungunya Virus. PLoS Pathog. 2007, 3, e89. [Google Scholar] [CrossRef]
  126. Couderc, T.; Chrétien, F.; Schilte, C.; Disson, O.; Brigitte, M.; Guivel-Benhassine, F.; Touret, Y.; Barau, G.; Cayet, N.; Schuffenecker, I.; et al. A Mouse Model for Chikungunya: Young Age and Inefficient Type-I Interferon Signaling Are Risk Factors for Severe Disease. PLoS Pathog. 2008, 4, e29. [Google Scholar] [CrossRef] [PubMed]
  127. Matusali, G.; Colavita, F.; Bordi, L.; Lalle, E.; Ippolito, G.; Capobianchi, M.R.; Castilletti, C. Tropism of the Chikungunya Virus. Viruses 2019, 11, 175. [Google Scholar] [CrossRef]
  128. Ganu, M.A.; Ganu, A.S. Post-Chikungunya Chronic Arthritis--Our Experience with DMARDs over Two Year Follow Up. J. Assoc. Physicians India 2011, 59, 83–86. [Google Scholar]
  129. Lauridsen, C. From Oxidative Stress to Inflammation: Redox Balance and Immune System. Poult. Sci. 2019, 98, 4240–4246. [Google Scholar] [CrossRef] [PubMed]
  130. Panieri, E.; Santoro, M.M. ROS Signaling and Redox Biology in Endothelial Cells. Cell. Mol. Life Sci. 2015, 72, 3281–3303. [Google Scholar] [CrossRef] [PubMed]
  131. Bryce Larke, R.P.; Wheelock, E.F. Stabilization of Chikungunya Virus Infectivity by Human Blood Platelets. J. Infect. Dis. 1970, 122, 523–531. [Google Scholar] [CrossRef]
  132. Chernesky, M.A.; Larke, R.P.B. Contrasting Effects of Rabbit and Human Platelets on Chikungunya Virus Infectivity. Can. J. Microbiol. 1977, 23, 1237–1244. [Google Scholar] [CrossRef] [PubMed]
  133. Gomes de Azevedo-Quintanilha, I.; Campos, M.M.; Teixeira Monteiro, A.P.; Dantas do Nascimento, A.; Calheiros, A.S.; Oliveira, D.M.; Dias, S.S.G.; Soares, V.C.; Santos, J.d.C.; Tavares, I.; et al. Increased Platelet Activation and Platelet-Inflammasome Engagement during Chikungunya Infection. Front. Immunol. 2022, 13, 958820. [Google Scholar] [CrossRef] [PubMed]
  134. Duchez, A.-C.; Boudreau, L.H.; Naika, G.S.; Bollinger, J.; Belleannée, C.; Cloutier, N.; Laffont, B.; Mendoza-Villarroel, R.E.; Lévesque, T.; Rollet-Labelle, E.; et al. Platelet Microparticles Are Internalized in Neutrophils via the Concerted Activity of 12-Lipoxygenase and Secreted Phospholipase A 2-IIA. Proc. Natl. Acad. Sci. USA 2015, 112, E3564–E3573. [Google Scholar] [CrossRef]
  135. Alvarez, M.F.; Bolívar-Mejía, A.; Rodriguez-Morales, A.J.; Ramirez-Vallejo, E. Cardiovascular Involvement and Manifestations of Systemic Chikungunya Virus Infection: A Systematic Review. F1000Research 2017, 6, 390. [Google Scholar] [CrossRef] [PubMed]
  136. Marques, M.A.; Adami de Sá, F.P.; Lupi, O.; Brasil, P.; von Ristow, A. Trombose Venosa Profunda e Vírus Chicungunha. J. Vasc. Bras. 2017, 16, 60–62. [Google Scholar] [CrossRef]
  137. He, X.; Balati, A.; Wang, W.; Wang, H.; Zhang, B.; Li, C.; Yu, D.; Guo, S.; Zeng, H. Association of Thrombocytopenia and D-Dimer Elevation with in-Hospital Mortality in Acute Aortic Dissection. Ann. Med. 2025, 57, 2478477. [Google Scholar] [CrossRef]
  138. Iuliano, A.D.; Roguski, K.M.; Chang, H.H.; Muscatello, D.J.; Palekar, R.; Tempia, S.; Cohen, C.; Gran, J.M.; Schanzer, D.; Cowling, B.J.; et al. Estimates of Global Seasonal Influenza-Associated Respiratory Mortality: A Modelling Study. Lancet 2018, 391, 1285–1300. [Google Scholar] [CrossRef] [PubMed]
  139. Troeger, C.; Forouzanfar, M.; Rao, P.C.; Khalil, I.; Brown, A.; Swartz, S.; Fullman, N.; Mosser, J.; Thompson, R.L.; Reiner, R.C.; et al. Estimates of the Global, Regional, and National Morbidity, Mortality, and Aetiologies of Lower Respiratory Tract Infections in 195 Countries: A Systematic Analysis for the Global Burden of Disease Study 2015. Lancet Infect. Dis. 2017, 17, 1133–1161. [Google Scholar] [CrossRef] [PubMed]
  140. Krammer, F.; Smith, G.J.D.; Fouchier, R.A.M.; Peiris, M.; Kedzierska, K.; Doherty, P.C.; Palese, P.; Shaw, M.L.; Treanor, J.; Webster, R.G.; et al. Influenza. Nat. Rev. Dis. Prim. 2018, 4, 3. [Google Scholar] [CrossRef]
  141. Hill, N.J.; Runstadler, J.A. A Bird’s Eye View of Influenza A Virus Transmission: Challenges with Characterizing Both Sides of a Co-Evolutionary Dynamic. Integr. Comp. Biol. 2016, 56, 304–316. [Google Scholar] [CrossRef] [PubMed]
  142. Bailey, E.S.; Choi, J.Y.; Fieldhouse, J.K.; Borkenhagen, L.K.; Zemke, J.; Zhang, D.; Gray, G.C. The Continual Threat of Influenza Virus Infections at the Human–Animal Interface. Evol. Med. Public Health 2018, 2018, 192–198. [Google Scholar] [CrossRef] [PubMed]
  143. Wu, N.C.; Wilson, I.A. Influenza Hemagglutinin Structures and Antibody Recognition. Cold Spring Harb. Perspect. Med. 2020, 10, a038778. [Google Scholar] [CrossRef] [PubMed]
  144. Du, R.; Cui, Q.; Rong, L. Competitive Cooperation of Hemagglutinin and Neuraminidase during Influenza A Virus Entry. Viruses 2019, 11, 458. [Google Scholar] [CrossRef]
  145. Rago, F.; Melo, E.M.; Miller, L.M.; Duray, A.M.; Batista Felix, F.; Vago, J.P.; de Faria Gonçalves, A.P.; Angelo, A.L.P.M.; Cassali, G.D.; de Gaetano, M.; et al. Treatment with Lipoxin A4 Improves Influenza A Infection Outcome, Induces Macrophage Reprogramming, Anti-Inflammatory and pro-Resolutive Responses. Inflamm. Res. 2024, 73, 1903–1918. [Google Scholar] [CrossRef]
  146. Koupenova, M.; Corkrey, H.A.; Vitseva, O.; Manni, G.; Pang, C.J.; Clancy, L.; Yao, C.; Rade, J.; Levy, D.; Wang, J.P.; et al. The Role of Platelets in Mediating a Response to Human Influenza Infection. Nat. Commun. 2019, 10, 1780. [Google Scholar] [CrossRef]
  147. Chen, Y.; Shi, Y.; Dou, L.; Liu, Y.; Zhang, J. Relationship of Influenza Virus to Inflammatory Factors and Immune Function in Elderly Patients with COPD: A Retrospective Analysis. Technol. Health Care 2025, 33, 09287329251317307. [Google Scholar] [CrossRef]
  148. Fang, X.; Gao, J.; Zhang, Z.; Yang, X.; Wang, Q.; Wang, J. Platelet Distribution Width in Patients with Influenza A Virus-Induced Acute Respiratory Distress Syndrome: An Old Indicator with Promising Clinical Application. Diagn. Microbiol. Infect. Dis. 2025, 111, 116657. [Google Scholar] [CrossRef]
  149. Videm, V.; Albrigtsen, M. Soluble ICAM-1 and VCAM-1 as Markers of Endothelial Activation. Scand. J. Immunol. 2008, 67, 523–531. [Google Scholar] [CrossRef] [PubMed]
  150. Guglielmini, G.; Berardi, E.; Messina, F.; Marcotullio, M.C.; Gresele, P. Effects of 3,5,4′-Tri-[4-(Nitrooxy)Butanoyl]Oxy Resveratrol, a New Nitric Oxide-Releasing Derivative of Resveratrol, on Platelet Activation. Pharmacol. Rep. 2025, 77, 729–738. [Google Scholar] [CrossRef]
  151. Antoniak, S.; Tatsumi, K.; Hisada, Y.; Milner, J.J.; Neidich, S.D.; Shaver, C.M.; Pawlinski, R.; Beck, M.A.; Bastarache, J.A.; Mackman, N. Tissue Factor Deficiency Increases Alveolar Hemorrhage and Death in Influenza A Virus-infected Mice. J. Thromb. Haemost. 2016, 14, 1238–1248. [Google Scholar] [CrossRef] [PubMed]
  152. Jang, J.; Yu, H.; Oh, E.B.; Park, J.W.; Kim, S.; Kim, T.; Sohn, J.; Jin, B.-R.; Chang, T.-S. Targeting NADPH Oxidase with APX-115: Suppression of Platelet Activation and Thrombotic Response. Antioxid. Redox Signal. 2025, 43, 4–6. [Google Scholar] [CrossRef] [PubMed]
  153. Zou, S.; Mohtar, S.H.; Othman, R.; Hassan, R.M.; Liang, K.; Lei, D.; Xu, B. Platelet Distribution Width as an Useful Indicator of Influenza Severity in Children. BMC Infect. Dis. 2024, 24, 9. [Google Scholar] [CrossRef] [PubMed]
  154. Jansen, A.J.G.; Spaan, T.; Low, H.Z.; Di Iorio, D.; van den Brand, J.; Tieke, M.; Barendrecht, A.; Rohn, K.; van Amerongen, G.; Stittelaar, K.; et al. Influenza-Induced Thrombocytopenia Is Dependent on the Subtype and Sialoglycan Receptor and Increases with Virus Pathogenicity. Blood Adv. 2020, 4, 2967–2978. [Google Scholar] [CrossRef]
  155. Rommel, M.G.E.; Walz, L.; Fotopoulou, F.; Kohlscheen, S.; Schenk, F.; Miskey, C.; Botezatu, L.; Krebs, Y.; Voelker, I.M.; Wittwer, K.; et al. Influenza A Virus Infection Instructs Hematopoiesis to Megakaryocyte-Lineage Output. Cell Rep. 2022, 41, 111447. [Google Scholar] [CrossRef] [PubMed]
  156. Lê, V.B.; Schneider, J.G.; Boergeling, Y.; Berri, F.; Ducatez, M.; Guerin, J.-L.; Adrian, I.; Errazuriz-Cerda, E.; Frasquilho, S.; Antunes, L.; et al. Platelet Activation and Aggregation Promote Lung Inflammation and Influenza Virus Pathogenesis. Am. J. Respir. Crit. Care Med. 2015, 191, 804–819. [Google Scholar] [CrossRef] [PubMed]
  157. Rondina, M.T.; Brewster, B.; Grissom, C.K.; Zimmerman, G.A.; Kastendieck, D.H.; Harris, E.S.; Weyrich, A.S. In Vivo Platelet Activation in Critically Ill Patients with Primary 2009 Influenza A(H1N1). Chest 2012, 141, 1490–1495. [Google Scholar] [CrossRef]
  158. Boilard, E.; Paré, G.; Rousseau, M.; Cloutier, N.; Dubuc, I.; Lévesque, T.; Borgeat, P.; Flamand, L. Influenza Virus H1N1 Activates Platelets through FcγRIIA Signaling and Thrombin Generation. Blood 2014, 123, 2854–2863. [Google Scholar] [CrossRef]
  159. Pariser, D.N.; Hilt, Z.T.; Ture, S.K.; Blick-Nitko, S.K.; Looney, M.R.; Cleary, S.J.; Roman-Pagan, E.; Saunders, J.; Georas, S.N.; Veazey, J.; et al. Lung Megakaryocytes Are Immune Modulatory Cells. J. Clin. Investig. 2021, 131, e137377. [Google Scholar] [CrossRef]
  160. de Jong, M.D.; Simmons, C.P.; Thanh, T.T.; Hien, V.M.; Smith, G.J.D.; Chau, T.N.B.; Hoang, D.M.; Van Vinh Chau, N.; Khanh, T.H.; Dong, V.C.; et al. Fatal Outcome of Human Influenza A (H5N1) Is Associated with High Viral Load and Hypercytokinemia. Nat. Med. 2006, 12, 1203–1207. [Google Scholar] [CrossRef] [PubMed]
  161. Avnon, L.S.; Munteanu, D.; Smoliakov, A.; Jotkowitz, A.; Barski, L. Thromboembolic Events in Patients with Severe Pandemic Influenza A/H1N1. Eur. J. Intern. Med. 2015, 26, 596–598. [Google Scholar] [CrossRef] [PubMed]
  162. Obi, A.T.; Tignanelli, C.J.; Jacobs, B.N.; Arya, S.; Park, P.K.; Wakefield, T.W.; Henke, P.K.; Napolitano, L.M. Empirical Systemic Anticoagulation Is Associated with Decreased Venous Thromboembolism in Critically Ill Influenza A H1N1 Acute Respiratory Distress Syndrome Patients. J. Vasc. Surg. Venous Lymphat. Disord. 2019, 7, 317–324. [Google Scholar] [CrossRef] [PubMed]
  163. Wen, F.; Liu, S.; Zhou, L.; Zhu, Y.; Wang, W.; Wei, M.; Xu, X.; Liu, Y.; Shuai, Q.; Yu, J.; et al. Immunogenicity and Safety of 1 versus 2 Doses of Quadrivalent-Inactivated Influenza Vaccine in Children Aged 3–8 Years with or without Previous Influenza Vaccination Histories. Hum. Vaccin. Immunother. 2025, 21, 2468074. [Google Scholar] [CrossRef] [PubMed]
  164. Rocha, F. Created in BioRender. Figure 2: Respiratory Virus-Induced Thromboinflammation. BioRender. 2025. Available online: https://BioRender.com/flkjij2 (accessed on 10 June 2025).
  165. Yang, X.; Yang, Q.; Wang, Y.; Wu, Y.; Xu, J.; Yu, Y.; Shang, Y. Thrombocytopenia and Its Association with Mortality in Patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1469–1472. [Google Scholar] [CrossRef]
  166. Gonçalves, J.J.; da Mata, C.P.S.M.; Lourenço, A.A.; Ribeiro, Á.L.; Ferreira, G.M.; Fraga-Silva, T.F.d.C.; de Souza, F.M.; Almeida, V.E.S.; Batista, I.A.; D‘Avila-Mesquita, C.; et al. Timeline Kinetics of Systemic and Airway Immune Mediator Storm for Comprehensive Analysis of Disease Outcome in Critically Ill COVID-19 Patients. Front. Immunol. 2022, 13, 903903. [Google Scholar] [CrossRef]
  167. Chae, D.-H.; Park, H.S.; Kim, K.-M.; Yu, A.; Park, J.H.; Oh, M.-K.; Choi, S.W.; Ryu, J.; Dunbar, C.E.; Yoo, H.M.; et al. SARS-CoV-2 Pseudovirus Dysregulates Hematopoiesis and Induces Inflammaging of Hematopoietic Stem and Progenitor Cells. Exp. Mol. Med. 2025, 57, 616–627. [Google Scholar] [CrossRef]
  168. de Maistre, E.; Savard, P.; Guinot, P.-G. COVID-19 and the Concept of Thrombo-Inflammation: Review of the Relationship between Immune Response, Endothelium and Coagulation. J. Clin. Med. 2023, 12, 7245. [Google Scholar] [CrossRef] [PubMed]
  169. Lu, R.; Zhao, X.; Li, J.; Niu, P.; Yang, B.; Wu, H.; Wang, W.; Song, H.; Huang, B.; Zhu, N.; et al. Genomic Characterisation and Epidemiology of 2019 Novel Coronavirus: Implications for Virus Origins and Receptor Binding. Lancet 2020, 395, 565–574. [Google Scholar] [CrossRef] [PubMed]
  170. Lan, J.; Ge, J.; Yu, J.; Shan, S.; Zhou, H.; Fan, S.; Zhang, Q.; Shi, X.; Wang, Q.; Zhang, L.; et al. Structure of the SARS-CoV-2 Spike Receptor-Binding Domain Bound to the ACE2 Receptor. Nature 2020, 581, 215–220. [Google Scholar] [CrossRef] [PubMed]
  171. Wang, Q.; Zhang, Y.; Wu, L.; Niu, S.; Song, C.; Zhang, Z.; Lu, G.; Qiao, C.; Hu, Y.; Yuen, K.-Y.; et al. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell 2020, 181, 894–904.e9. [Google Scholar] [CrossRef] [PubMed]
  172. Bos, L.D.J.; Ware, L.B. Acute Respiratory Distress Syndrome: Causes, Pathophysiology, and Phenotypes. Lancet 2022, 400, 1145–1156. [Google Scholar] [CrossRef]
  173. Mendes, S.; Guimarães, L.C.; Costa, P.A.C.; Fernandez, C.C.; Figueiredo, M.M.; Teixeira, M.M.; dos Santos, R.A.S.; Guimarães, P.P.G.; Frézard, F. Intranasal Liposomal Angiotensin-(1-7) Administration Reduces Inflammation and Viral Load in the Lungs during SARS-CoV-2 Infection in K18-HACE2 Transgenic Mice. Antimicrob. Agents Chemother. 2024, 68, e00835-24. [Google Scholar] [CrossRef]
  174. Tang, N.; Li, D.; Wang, X.; Sun, Z. Abnormal Coagulation Parameters Are Associated with Poor Prognosis in Patients with Novel Coronavirus Pneumonia. J. Thromb. Haemost. 2020, 18, 844–847. [Google Scholar] [CrossRef]
  175. Middeldorp, S.; Coppens, M.; van Haaps, T.F.; Foppen, M.; Vlaar, A.P.; Müller, M.C.A.; Bouman, C.C.S.; Beenen, L.F.M.; Kootte, R.S.; Heijmans, J.; et al. Incidence of Venous Thromboembolism in Hospitalized Patients with COVID-19. J. Thromb. Haemost. 2020, 18, 1995–2002. [Google Scholar] [CrossRef] [PubMed]
  176. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in Covid-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef]
  177. Zhu, A.; Real, F.; Capron, C.; Rosenberg, A.R.; Silvin, A.; Dunsmore, G.; Zhu, J.; Cottoignies-Callamarte, A.; Massé, J.-M.; Moine, P.; et al. Infection of Lung Megakaryocytes and Platelets by SARS-CoV-2 Anticipate Fatal COVID-19. Cell. Mol. Life Sci. 2022, 79, 365. [Google Scholar] [CrossRef]
  178. Garcia, C.; Vardon-Bounes, F.; Compagnon, B.; Guilbeau-Frugier, C.; Voisin, S.; Rieu, J.-B.; De Mas, V.; Payrastre, B.; Ribes, A. Detection of SARS-CoV-2 in Bone Marrow Megakaryocytes and Elevated Emperipolesis in COVID-19 Patients with Thrombocytopenia. J. Thromb. Haemost. 2025, 23, 2327–2334. [Google Scholar] [CrossRef] [PubMed]
  179. Antonopoulou, S.; Petsini, F.; Detopoulou, M.; Theoharides, T.C.; Demopoulos, C.A. Is There an Interplay between the SARS-CoV-2 Spike Protein and Platelet-Activating Factor? BioFactors 2022, 48, 1271–1283. [Google Scholar] [CrossRef]
  180. Petry, J.; Shoykhet, M.; Weiser, T.; Griesbaum, L.; Bashiri Dezfouli, A.; Verschoor, A.; Wollenberg, B. SARS-CoV-2 S1 Protein Induces IgG-Mediated Platelet Activation and Is Prevented by 1.8-Cineole. Biomed. Pharmacother. 2025, 187, 118100. [Google Scholar] [CrossRef] [PubMed]
  181. Zuchtriegel, G.; Uhl, B.; Puhr-Westerheide, D.; Pörnbacher, M.; Lauber, K.; Krombach, F.; Reichel, C.A. Platelets Guide Leukocytes to Their Sites of Extravasation. PLOS Biol. 2016, 14, e1002459. [Google Scholar] [CrossRef] [PubMed]
  182. Eichhorn, T.; Weiss, R.; Huber, S.; Ebeyer-Masotta, M.; Mostageer, M.; Emprechtinger, R.; Knabl, L.; Knabl, L.; Würzner, R.; Weber, V. Expression of Tissue Factor and Platelet/Leukocyte Markers on Extracellular Vesicles Reflect Platelet–Leukocyte Interaction in Severe COVID-19. Int. J. Mol. Sci. 2023, 24, 16886. [Google Scholar] [CrossRef]
  183. Rapkiewicz, A.V.; Mai, X.; Carsons, S.E.; Pittaluga, S.; Kleiner, D.E.; Berger, J.S.; Thomas, S.; Adler, N.M.; Charytan, D.M.; Gasmi, B.; et al. Megakaryocytes and Platelet-Fibrin Thrombi Characterize Multi-Organ Thrombosis at Autopsy in COVID-19: A Case Series. eClinicalMedicine 2020, 24, 100434. [Google Scholar] [CrossRef]
  184. Andrade, A.C.; Lacasse, E.; Dubuc, I.; Gudimard, L.; Puhm, F.; Queiroz, C.; Allaeys, I.; Prunier, J.; Dumais, É.; Flamand, N.; et al. Loss of Platelet 12-Lipoxygenase Aggravates the Severity of Sars-Cov-2 Infection. Blood 2023, 142, 3938. [Google Scholar] [CrossRef]
  185. Chouchane, O.; Léopold, V.; Michels, E.H.A.; de Brabander, J.; van Linge, C.C.A.; Klarenbeek, A.M.; Wiersinga, W.J.; van ‘t Veer, C.; van der Poll, T. Differential Platelet Protein Release Profiles in Community-Acquired Pneumonia and COVID-19. ERJ Open Res. 2025, 11, 00863–02024. [Google Scholar] [CrossRef] [PubMed]
  186. Hottz, E.D.; Azevedo-Quintanilha, I.G.; Palhinha, L.; Teixeira, L.; Barreto, E.A.; Pão, C.R.R.; Righy, C.; Franco, S.; Souza, T.M.L.; Kurtz, P.; et al. Platelet Activation and Platelet-Monocyte Aggregate Formation Trigger Tissue Factor Expression in Patients with Severe COVID-19. Blood 2020, 136, 1330–1341. [Google Scholar] [CrossRef]
  187. Hottz, E.D.; Martins-Gonçalves, R.; Palhinha, L.; Azevedo-Quintanilha, I.G.; de Campos, M.M.; Sacramento, C.Q.; Temerozo, J.R.; Soares, V.C.; Dias, S.S.G.; Teixeira, L.; et al. Platelet-Monocyte Interaction Amplifies Thromboinflammation through Tissue Factor Signaling in COVID-19. Blood Adv. 2022, 6, 5085–5099. [Google Scholar] [CrossRef]
  188. Baycan, O.F. Plasminogen Activator Inhibitor-1 Levels as an Indicator of Severity and Mortality for COVID-19. North. Clin. Istanb. 2022, 10, 1–9. [Google Scholar] [CrossRef]
  189. Panigada, M.; Bottino, N.; Tagliabue, P.; Grasselli, G.; Novembrino, C.; Chantarangkul, V.; Pesenti, A.; Peyvandi, F.; Tripodi, A. Hypercoagulability of COVID-19 Patients in Intensive Care Unit: A Report of Thromboelastography Findings and Other Parameters of Hemostasis. J. Thromb. Haemost. 2020, 18, 1738–1742. [Google Scholar] [CrossRef] [PubMed]
  190. Fox, S.E.; Akmatbekov, A.; Harbert, J.L.; Li, G.; Quincy Brown, J.; Vander Heide, R.S. Pulmonary and Cardiac Pathology in African American Patients with COVID-19: An Autopsy Series from New Orleans. Lancet Respir. Med. 2020, 8, 681–686. [Google Scholar] [CrossRef] [PubMed]
  191. Barrett, T.J.; Cornwell, M.; Myndzar, K.; Rolling, C.C.; Xia, Y.; Drenkova, K.; Biebuyck, A.; Fields, A.T.; Tawil, M.; Luttrell-Williams, E.; et al. Platelets Amplify Endotheliopathy in COVID-19. Sci. Adv. 2021, 7, eabh2434. [Google Scholar] [CrossRef] [PubMed]
  192. Al-Tamimi, A.O.; Yusuf, A.M.; Jayakumar, M.N.; Ansari, A.W.; Elhassan, M.; AbdulKarim, F.; Kannan, M.; Halwani, R.; Ahmad, F. SARS-CoV-2 Infection Induces Soluble Platelet Activation Markers and PAI-1 in the Early Moderate Stage of COVID-19. Int. J. Lab. Hematol. 2022, 44, 712–721. [Google Scholar] [CrossRef]
  193. Middleton, E.A.; He, X.-Y.; Denorme, F.; Campbell, R.A.; Ng, D.; Salvatore, S.P.; Mostyka, M.; Baxter-Stoltzfus, A.; Borczuk, A.C.; Loda, M.; et al. Neutrophil Extracellular Traps Contribute to Immunothrombosis in COVID-19 Acute Respiratory Distress Syndrome. Blood 2020, 136, 1169–1179. [Google Scholar] [CrossRef] [PubMed]
  194. Wang, J.; Li, Q.; Yin, Y.; Zhang, Y.; Cao, Y.; Lin, X.; Huang, L.; Hoffmann, D.; Lu, M.; Qiu, Y. Excessive Neutrophils and Neutrophil Extracellular Traps in COVID-19. Front. Immunol. 2020, 11, 2063. [Google Scholar] [CrossRef] [PubMed]
  195. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical Features of Patients Infected with 2019 Novel Coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef]
  196. Arce, N.A.; Cao, W.; Brown, A.K.; Legan, E.R.; Wilson, M.S.; Xu, E.-R.; Berndt, M.C.; Emsley, J.; Zhang, X.F.; Li, R. Activation of von Willebrand Factor via Mechanical Unfolding of Its Discontinuous Autoinhibitory Module. Nat. Commun. 2021, 12, 2360. [Google Scholar] [CrossRef]
  197. Goshua, G.; Pine, A.B.; Meizlish, M.L.; Chang, C.-H.; Zhang, H.; Bahel, P.; Baluha, A.; Bar, N.; Bona, R.D.; Burns, A.J.; et al. Endotheliopathy in COVID-19-Associated Coagulopathy: Evidence from a Single-Centre, Cross-Sectional Study. Lancet Haematol. 2020, 7, e575–e582. [Google Scholar] [CrossRef] [PubMed]
  198. Terra, P.O.C.; Donadel, C.D.; Oliveira, L.C.; Menegueti, M.G.; Auxiliadora-Martins, M.; Calado, R.T.; De Santis, G.C. Neutrophil-to-lymphocyte Ratio and D-dimer Are Biomarkers of Death Risk in Severe COVID-19: A Retrospective Observational Study. Health Sci. Rep. 2022, 5, e514. [Google Scholar] [CrossRef] [PubMed]
  199. Zhang, L.; Yan, X.; Fan, Q.; Liu, H.; Liu, X.; Liu, Z.; Zhang, Z. D-dimer Levels on Admission to Predict In-hospital Mortality in Patients with Covid-19. J. Thromb. Haemost. 2020, 18, 1324–1329. [Google Scholar] [CrossRef] [PubMed]
  200. Asseri, A.A.; Al-Qahtani, S.M.; Alzaydani, I.A.; Al-Jarie, A.; Alyazidi, N.S.; Alrmelawi, A.A.; Alqahtani, A.M.; Alsulayyim, R.S.; Alzailaie, A.K.; Abdullah, D.M.; et al. Clinical and Epidemiological Characteristics of Respiratory Syncytial Virus, SARS-CoV-2 and Influenza Paediatric Viral Respiratory Infections in Southwest Saudi Arabia. Ann. Med. 2025, 57, 2445791. [Google Scholar] [CrossRef] [PubMed]
  201. Fogarty, H.; Townsend, L.; Morrin, H.; Ahmad, A.; Comerford, C.; Karampini, E.; Englert, H.; Byrne, M.; Bergin, C.; O’Sullivan, J.M.; et al. Persistent Endotheliopathy in the Pathogenesis of Long COVID Syndrome. J. Thromb. Haemost. 2021, 19, 2546–2553. [Google Scholar] [CrossRef]
  202. Pretorius, E.; Vlok, M.; Venter, C.; Bezuidenhout, J.A.; Laubscher, G.J.; Steenkamp, J.; Kell, D.B. Persistent Clotting Protein Pathology in Long COVID/Post-Acute Sequelae of COVID-19 (PASC) Is Accompanied by Increased Levels of Antiplasmin. Cardiovasc. Diabetol. 2021, 20, 172. [Google Scholar] [CrossRef]
  203. Taquet, M.; Skorniewska, Z.; Hampshire, A.; Chalmers, J.D.; Ho, L.-P.; Horsley, A.; Marks, M.; Poinasamy, K.; Raman, B.; Leavy, O.C.; et al. Acute Blood Biomarker Profiles Predict Cognitive Deficits 6 and 12 Months after COVID-19 Hospitalization. Nat. Med. 2023, 29, 2498–2508. [Google Scholar] [CrossRef]
  204. Lee, M.-H.; Perl, D.P.; Nair, G.; Li, W.; Maric, D.; Murray, H.; Dodd, S.J.; Koretsky, A.P.; Watts, J.A.; Cheung, V.; et al. Microvascular Injury in the Brains of Patients with Covid-19. N. Engl. J. Med. 2021, 384, 481–483. [Google Scholar] [CrossRef] [PubMed]
  205. Cervia-Hasler, C.; Brüningk, S.C.; Hoch, T.; Fan, B.; Muzio, G.; Thompson, R.C.; Ceglarek, L.; Meledin, R.; Westermann, P.; Emmenegger, M.; et al. Persistent Complement Dysregulation with Signs of Thromboinflammation in Active Long Covid. Science 2024, 383, eadg7942. [Google Scholar] [CrossRef]
  206. Stein, S.R.; Ramelli, S.C.; Grazioli, A.; Chung, J.-Y.; Singh, M.; Yinda, C.K.; Winkler, C.W.; Sun, J.; Dickey, J.M.; Ylaya, K.; et al. SARS-CoV-2 Infection and Persistence in the Human Body and Brain at Autopsy. Nature 2022, 612, 758–763. [Google Scholar] [CrossRef] [PubMed]
  207. Gaebler, C.; Wang, Z.; Lorenzi, J.C.C.; Muecksch, F.; Finkin, S.; Tokuyama, M.; Cho, A.; Jankovic, M.; Schaefer-Babajew, D.; Oliveira, T.Y.; et al. Evolution of Antibody Immunity to SARS-CoV-2. Nature 2021, 591, 639–644. [Google Scholar] [CrossRef]
  208. Garg, R.K.; Rizvi, I.; Ingole, R.; Jain, A.; Malhotra, H.S.; Kumar, N.; Batra, D. Cortical Laminar Necrosis in Dengue Encephalitis—A Case Report. BMC Neurol. 2017, 17, 79. [Google Scholar] [CrossRef]
  209. Sahu, R.; Verma, R.; Jain, A.; Garg, R.K.; Singh, M.K.; Malhotra, H.S.; Sharma, P.K.; Parihar, A. Neurologic Complications in Dengue Virus Infection. Neurology 2014, 83, 1601–1609. [Google Scholar] [CrossRef]
  210. Kulkarni, R.; Pujari, S.; Gupta, D. Neurological Manifestations of Dengue Fever. Ann. Indian Acad. Neurol. 2021, 24, 693–702. [Google Scholar] [CrossRef]
  211. Sanjay, S.; Wagle, A.M.; Au Eong, K.G. Optic Neuropathy Associated with Dengue Fever. Eye 2008, 22, 722–724. [Google Scholar] [CrossRef] [PubMed]
  212. George, R.; Lam, S.K. Dengue Virus Infection--the Malaysian Experience. Ann. Acad. Med. Singap. 1997, 26, 815–819. [Google Scholar]
  213. Murthy, J.M.K. Neurological Complications of Dengue Infection. Neurol. India 2010, 58, 581. [Google Scholar] [CrossRef]
  214. Pancharoen, C.; Thisyakorn, U. Neurological Manifestations in Dengue Patients. Southeast Asian J. Trop. Med. Public Health 2001, 32, 341–345. [Google Scholar] [PubMed]
  215. Liou, L.-M.; Lan, S.-H.; Lai, C.-L. Dengue Fever with Ischemic Stroke. Neurologist 2008, 14, 40–42. [Google Scholar] [CrossRef] [PubMed]
  216. Mathew, S.; Pandian, J.D. Stroke in Patients with Dengue. J. Stroke Cerebrovasc. Dis. 2010, 19, 253–256. [Google Scholar] [CrossRef]
  217. Munoz, X.; Muñoz, X.; Obach, V.; Hurtado, B.; de Frutos, P.G.; Chamorro, A.; Sala, N. Association of Specific Haplotypes of GAS6 Gene with Stroke. Thromb. Haemost. 2007, 98, 406–412. [Google Scholar] [CrossRef] [PubMed]
  218. Verma, R.; Sahu, R.; Singh, A.; Atam, V. Dengue Infection Presenting as Ischemic Stroke: An Uncommon Neurological Manifestation. Neurol. India 2013, 61, 317. [Google Scholar] [CrossRef] [PubMed]
  219. Schuhmann, M.K.; Stoll, G.; Bieber, M.; Vögtle, T.; Hofmann, S.; Klaus, V.; Kraft, P.; Seyhan, M.; Kollikowski, A.M.; Papp, L.; et al. CD84 Links T Cell and Platelet Activity in Cerebral Thrombo-Inflammation in Acute Stroke. Circ. Res. 2020, 127, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  220. Azmin, S.; Sahathevan, R.; Suehazlyn, Z.; Law, Z.K.; Rabani, R.; Nafisah, W.Y.; Tan, H.J.; Norlinah, M.I. Post-Dengue Parkinsonism. BMC Infect. Dis. 2013, 13, 179. [Google Scholar] [CrossRef]
  221. Hopkins, H.K.; Traverse, E.M.; Barr, K.L. Viral Parkinsonism: An Underdiagnosed Neurological Complication of Dengue Virus Infection. PLoS Negl. Trop. Dis. 2022, 16, e0010118. [Google Scholar] [CrossRef] [PubMed]
  222. Apoorva; Kumar, A.; Singh, S.K. Dengue Virus NS1 Hits Hard at the Barrier Integrity of Human Cerebral Microvascular Endothelial Cells via Cellular MicroRNA Dysregulations. Tissue Barriers 2024, 2424628. [Google Scholar] [CrossRef]
  223. Srivastava, N.K.; Pandey, N.; Makani, J.; Hussain, I.; Dhiman, N.R.; Kumar, A.; Joshi, D. A Comprehensive and Critical Narrative Review on Movement Disorders Linked through Dengue Virus Based Infection. Neurol. Sci. 2025, 46, 4193–4207. [Google Scholar] [CrossRef]
  224. Chow, J.H.; Khanna, A.K.; Kethireddy, S.; Yamane, D.; Levine, A.; Jackson, A.M.; McCurdy, M.T.; Tabatabai, A.; Kumar, G.; Park, P.; et al. Aspirin Use Is Associated with Decreased Mechanical Ventilation, Intensive Care Unit Admission, and In-Hospital Mortality in Hospitalized Patients with Coronavirus Disease 2019. Anesth. Analg. 2021, 132, 930–941. [Google Scholar] [CrossRef] [PubMed]
  225. Mazloomzadeh, S.; Khaleghparast, S.; Ghadrdoost, B.; Mousavizadeh, M.; Baay, M.R.; Noohi, F.; Sharifnia, H.; Ahmadi, A.; Tavan, S.; Malekpour Alamdari, N.; et al. Effect of Intermediate-Dose vs Standard-Dose Prophylactic Anticoagulation on Thrombotic Events, Extracorporeal Membrane Oxygenation Treatment, or Mortality Among Patients with COVID-19 Admitted to the Intensive Care Unit. JAMA 2021, 325, 1620. [Google Scholar] [CrossRef]
  226. Sivaloganathan, H.; Ladikou, E.E.; Chevassut, T. COVID-19 Mortality in Patients on Anticoagulants and Antiplatelet Agents. Br. J. Haematol. 2020, 190, e192. [Google Scholar] [CrossRef] [PubMed]
  227. ATTACC Investigators; ACTIV-4a Investigators; REMAP-CAP Investigators; Lawler, P.R.; Goligher, E.C.; Berger, J.S.; Neal, M.D.; McVerry, B.J.; Nicolau, J.C.; Gong, M.N.; et al. Therapeutic Anticoagulation with Heparin in Noncritically Ill Patients with Covid-19. N. Engl. J. Med. 2021, 385, 790–802. [Google Scholar] [CrossRef] [PubMed]
  228. Gong, J.; Nie, N.; Jiang, M.; Yang, X.; Wang, Q.; Deng, J.; Kang, J.; Li, X.; Zhang, L.; Zhang, Y.; et al. Efficacy and Safety of Combined Nebulization of Unfractionated Heparin, Acetylcysteine, Budesonide and Ipratropium Bromide in Hospitalised Patients with COVID-19 Pneumonia: A Randomized Controlled Clinical Trial. BMC Pulm. Med. 2025, 25, 346. [Google Scholar] [CrossRef]
  229. Camprubí-Rimblas, M.; Tantinyà, N.; Bringué, J.; Guillamat-Prats, R.; Artigas, A. Anticoagulant Therapy in Acute Respiratory Distress Syndrome. Ann. Transl. Med. 2018, 6, 36. [Google Scholar] [CrossRef] [PubMed]
  230. Esmon, C.T. Targeting Factor Xa and Thrombin: Impact on Coagulation and Beyond. Thromb. Haemost. 2014, 111, 625–633. [Google Scholar] [CrossRef] [PubMed]
  231. Lother, S.A.; Tennenhouse, L.; Rabbani, R.; Abou-Setta, A.M.; Askin, N.; Turgeon, A.F.; Murthy, S.; Houston, B.L.; Houston, D.S.; Mendelson, A.A.; et al. The Association of Antiplatelet Agents with Mortality among Patients with Non–COVID-19 Community-Acquired Pneumonia: A Systematic Review and Meta-Analysis. Res. Pract. Thromb. Haemost. 2024, 8, 102526. [Google Scholar] [CrossRef]
  232. Bikdeli, B.; Madhavan, M.V.; Jimenez, D.; Chuich, T.; Dreyfus, I.; Driggin, E.; Nigoghossian, C.D.; Ageno, W.; Madjid, M.; Guo, Y.; et al. COVID-19 and Thrombotic or Thromboembolic Disease: Implications for Prevention, Antithrombotic Therapy, and Follow-Up. J. Am. Coll. Cardiol. 2020, 75, 2950–2973. [Google Scholar] [CrossRef]
  233. Manne, B.K.; Denorme, F.; Middleton, E.A.; Portier, I.; Rowley, J.W.; Stubben, C.; Petrey, A.C.; Tolley, N.D.; Guo, L.; Cody, M.; et al. Platelet Gene Expression and Function in Patients with COVID-19. Blood 2020, 136, 1317–1329. [Google Scholar] [CrossRef]
  234. Chow, J.H.; Rahnavard, A.; Gomberg-Maitland, M.; Chatterjee, R.; Patodi, P.; Yamane, D.P.; Levine, A.R.; Davison, D.; Hawkins, K.; Jackson, A.M.; et al. Association of Early Aspirin Use with In-Hospital Mortality in Patients with Moderate COVID-19. JAMA Netw. Open 2022, 5, e223890. [Google Scholar] [CrossRef]
  235. RECOVERY Collaborative Group. Aspirin in Patients Admitted to Hospital with COVID-19 (RECOVERY): A Randomised, Controlled, Open-Label, Platform Trial. Lancet 2022, 399, 143–151. [Google Scholar] [CrossRef]
  236. Siepmann, T.; Sedghi, A.; Simon, E.; Winzer, S.; Barlinn, J.; de With, K.; Mirow, L.; Wolz, M.; Gruenewald, T.; Schroettner, P.; et al. Increased Risk of Acute Stroke among Patients with Severe COVID-19: A Multicenter Study and Meta-analysis. Eur. J. Neurol. 2021, 28, 238–247. [Google Scholar] [CrossRef] [PubMed]
  237. Biswas, M.; Sawajan, N.; Rungrotmongkol, T.; Sanachai, K.; Ershadian, M.; Sukasem, C. Pharmacogenetics and Precision Medicine Approaches for the Improvement of COVID-19 Therapies. Front. Pharmacol. 2022, 13, 835136. [Google Scholar] [CrossRef] [PubMed]
  238. Chia, P.Y.; Htun, H.L.; Leo, Y.S.; Lye, D.C. Safety of Temporary Interruption of Antiplatelet Therapy in Dengue Fever with Thrombocytopenia. J. Infect. 2021, 82, 270–275. [Google Scholar] [CrossRef]
  239. Roy, S. Thrombopoietin Receptor Agonists: Can These Be the Future Answer to the Deadly Thrombocytopenia in Dengue Fever? Cureus 2019, 11, e4361. [Google Scholar] [CrossRef]
  240. Aboulafia, D.; Vishnu, P. Long-Term Safety and Efficacy of Romiplostim for Treatment of Immune Thrombocytopenia. J. Blood Med. 2016, 7, 99–106. [Google Scholar] [CrossRef]
  241. Mitchell, W.B.; Pinheiro, M.P.; Boulad, N.; Kaplan, D.; Edison, M.N.; Psaila, B.; Karpoff, M.; White, M.J.; Josefsson, E.C.; Kile, B.T.; et al. Effect of Thrombopoietin Receptor Agonists on the Apoptotic Profile of Platelets in Patients with Chronic Immune Thrombocytopenia. Am. J. Hematol. 2014, 89, E228–E234. [Google Scholar] [CrossRef]
  242. Kim, Y.-K.; Lee, S.-S.; Jeong, S.-H.; Ahn, J.-S.; Yang, D.-H.; Lee, J.-J.; Kim, H.-J. Efficacy and Safety of Eltrombopag in Adult Refractory Immune Thrombocytopenia. Blood Res. 2015, 50, 19. [Google Scholar] [CrossRef] [PubMed]
  243. Bussel, J.B.; Cheng, G.; Saleh, M.N.; Psaila, B.; Kovaleva, L.; Meddeb, B.; Kloczko, J.; Hassani, H.; Mayer, B.; Stone, N.L.; et al. Eltrombopag for the Treatment of Chronic Idiopathic Thrombocytopenic Purpura. N. Engl. J. Med. 2007, 357, 2237–2247. [Google Scholar] [CrossRef]
  244. Ayaz, S.I.; Sharkey, C.M.; Kwiatkowski, G.M.; Wilson, S.S.; John, R.S.; Tolomello, R.; Mahajan, A.; Millis, S.; Levy, P.D. Intravenous Enalaprilat for Treatment of Acute Hypertensive Heart Failure in the Emergency Department. Int. J. Emerg. Med. 2016, 9, 28. [Google Scholar] [CrossRef] [PubMed]
  245. Todd, P.A.; Goa, K.L. Enalapril. Drugs 1992, 43, 346–381. [Google Scholar] [CrossRef] [PubMed]
  246. Hernández-Fonseca, J.P.; Durán, A.; Valero, N.; Mosquera, J. Losartan and Enalapril Decrease Viral Absorption and Interleukin 1 Beta Production by Macrophages in an Experimental Dengue Virus Infection. Arch. Virol. 2015, 160, 2861–2865. [Google Scholar] [CrossRef] [PubMed]
  247. Pulavendran, S.; Rudd, J.M.; Maram, P.; Thomas, P.G.; Akhilesh, R.; Malayer, J.R.; Chow, V.T.K.; Teluguakula, N. Combination Therapy Targeting Platelet Activation and Virus Replication Protects Mice against Lethal Influenza Pneumonia. Am. J. Respir. Cell Mol. Biol. 2019, 61, 689–701. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Thromboinflammatory mechanisms are induced by arboviruses. (A) Dengue. DENV triggers thromboinflammation through direct infection of bone marrow megakaryocytes, leading to cellular dysfunction and the release of immature, dysfunctional platelets. These platelets undergo premature activation and apoptosis. In peripheral blood, the virus can directly activate platelets and promote their interaction with leukocytes, enhancing clearance and contributing to thrombocytopenia. Both the virus and its NS1 protein also target endothelial cells, inducing activation, increased vascular permeability, and barrier disruption. In tissues, DENV activates resident mast cells, which release VEGF, histamines, proteases, and eicosanoids, amplifying inflammation and endothelial dysfunction. These events collectively disturb vascular homeostasis and coagulation. Clinically, severe dengue is characterized by prolonged PT and aPTT, elevated D-dimer levels (indicating fibrinolysis and risk of deep vein thrombosis), and increased expression of tissue factor and PAI-1, reflecting early coagulation cascade activation. (B) Chikungunya. The thromboinflammatory pathways triggered by CHIKV remain less defined. The virus exhibits tropism for joints, where chronic disease is linked to persistent synovial inflammation driven by nitric oxide production and NF-κB activation, leading to symptoms resembling rheumatoid arthritis. During the acute phase, CHIKV promotes platelet activation, P-selectin expression, and the formation of platelet–leukocyte aggregates, which may contribute to joint pain. Activated platelets also release 12-HETE, a pro-inflammatory lipid mediator also found in rheumatoid arthritis. These mechanisms may underline the elevated D-dimer levels observed in acute infection and the increased risk of thrombotic events, such as deep vein thrombosis, reported in some patients. Created in Biorender [31].
Figure 1. Thromboinflammatory mechanisms are induced by arboviruses. (A) Dengue. DENV triggers thromboinflammation through direct infection of bone marrow megakaryocytes, leading to cellular dysfunction and the release of immature, dysfunctional platelets. These platelets undergo premature activation and apoptosis. In peripheral blood, the virus can directly activate platelets and promote their interaction with leukocytes, enhancing clearance and contributing to thrombocytopenia. Both the virus and its NS1 protein also target endothelial cells, inducing activation, increased vascular permeability, and barrier disruption. In tissues, DENV activates resident mast cells, which release VEGF, histamines, proteases, and eicosanoids, amplifying inflammation and endothelial dysfunction. These events collectively disturb vascular homeostasis and coagulation. Clinically, severe dengue is characterized by prolonged PT and aPTT, elevated D-dimer levels (indicating fibrinolysis and risk of deep vein thrombosis), and increased expression of tissue factor and PAI-1, reflecting early coagulation cascade activation. (B) Chikungunya. The thromboinflammatory pathways triggered by CHIKV remain less defined. The virus exhibits tropism for joints, where chronic disease is linked to persistent synovial inflammation driven by nitric oxide production and NF-κB activation, leading to symptoms resembling rheumatoid arthritis. During the acute phase, CHIKV promotes platelet activation, P-selectin expression, and the formation of platelet–leukocyte aggregates, which may contribute to joint pain. Activated platelets also release 12-HETE, a pro-inflammatory lipid mediator also found in rheumatoid arthritis. These mechanisms may underline the elevated D-dimer levels observed in acute infection and the increased risk of thrombotic events, such as deep vein thrombosis, reported in some patients. Created in Biorender [31].
Viruses 17 01207 g001
Figure 2. Respiratory virus-induced thromboinflammation. (A) Influenza virus. Influenza promotes endothelial dysfunction by upregulating adhesion molecules (VCAM-1, ICAM-1) and reducing nitric oxide (NO) production, a process linked to platelet activation and a procoagulant state. The infection also induces tissue factor (TF) expression, enhancing thrombus formation. Additionally, influenza alters platelet morphology by reducing platelets’ volume and size distribution, changes that correlate with disease severity and acute respiratory distress syndrome (ARDS). (B) SARS-CoV-2. SARS-CoV-2 can infect megakaryocytes and has been detected in the bone marrow of animal models, suggesting a mechanism for infection-associated thrombocytopenia. The virus also drives platelet activation and the formation of platelet–leukocyte aggregates, particularly with neutrophils, promoting neutrophil extracellular trap (NET) release and thrombosis. Elevated von Willebrand factor and D-dimer levels further indicate a hypercoagulable state. These events converge to cause endothelial dysfunction, characterized by increased adhesion molecule expression, and sustain the systemic thromboinflammatory response observed in COVID-19. Created in Biorender [164].
Figure 2. Respiratory virus-induced thromboinflammation. (A) Influenza virus. Influenza promotes endothelial dysfunction by upregulating adhesion molecules (VCAM-1, ICAM-1) and reducing nitric oxide (NO) production, a process linked to platelet activation and a procoagulant state. The infection also induces tissue factor (TF) expression, enhancing thrombus formation. Additionally, influenza alters platelet morphology by reducing platelets’ volume and size distribution, changes that correlate with disease severity and acute respiratory distress syndrome (ARDS). (B) SARS-CoV-2. SARS-CoV-2 can infect megakaryocytes and has been detected in the bone marrow of animal models, suggesting a mechanism for infection-associated thrombocytopenia. The virus also drives platelet activation and the formation of platelet–leukocyte aggregates, particularly with neutrophils, promoting neutrophil extracellular trap (NET) release and thrombosis. Elevated von Willebrand factor and D-dimer levels further indicate a hypercoagulable state. These events converge to cause endothelial dysfunction, characterized by increased adhesion molecule expression, and sustain the systemic thromboinflammatory response observed in COVID-19. Created in Biorender [164].
Viruses 17 01207 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Batista, V.L.; Martins, J.R.; Queiroz-Junior, C.M.; Hottz, E.D.; Teixeira, M.M.; Costa, V.V. Mechanisms of Thromboinflammation in Viral Infections—A Narrative Review. Viruses 2025, 17, 1207. https://doi.org/10.3390/v17091207

AMA Style

Batista VL, Martins JR, Queiroz-Junior CM, Hottz ED, Teixeira MM, Costa VV. Mechanisms of Thromboinflammation in Viral Infections—A Narrative Review. Viruses. 2025; 17(9):1207. https://doi.org/10.3390/v17091207

Chicago/Turabian Style

Batista, Viviane Lima, Jenniffer Ramos Martins, Celso Martins Queiroz-Junior, Eugenio Damaceno Hottz, Mauro Martins Teixeira, and Vivian Vasconcelos Costa. 2025. "Mechanisms of Thromboinflammation in Viral Infections—A Narrative Review" Viruses 17, no. 9: 1207. https://doi.org/10.3390/v17091207

APA Style

Batista, V. L., Martins, J. R., Queiroz-Junior, C. M., Hottz, E. D., Teixeira, M. M., & Costa, V. V. (2025). Mechanisms of Thromboinflammation in Viral Infections—A Narrative Review. Viruses, 17(9), 1207. https://doi.org/10.3390/v17091207

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop