Next Article in Journal
First-in-Class Colchicine-Based Visible Light Photoswitchable Microtubule Dynamics Disrupting Agent
Next Article in Special Issue
miRNA-Mediated Fine Regulation of TLR-Induced M1 Polarization
Previous Article in Journal
Perfusion and Ultrasonication Produce a Decellularized Porcine Whole-Ovary Scaffold with a Preserved Microarchitecture
Previous Article in Special Issue
Beyond Pattern Recognition: TLR2 Promotes Chemotaxis, Cell Adhesion, and Migration in THP-1 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review

1
Department of Cardiology, Wilhelminenspital, 1090 Vienna, Austria
2
Division of Angiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
3
Division of Vascular Surgery, Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
4
Faculty of Biophysics, Biochemistry and Biotechnology, Department of Medical Biotechnology, Jagiellonian University, 30-387 Krakow, Poland
5
Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, 1090 Vienna, Austria
6
Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology, Karl Landsteiner Society, 3100 St. Pölten, Austria
7
Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
8
Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
*
Author to whom correspondence should be addressed.
Cells 2023, 12(14), 1865; https://doi.org/10.3390/cells12141865
Submission received: 15 May 2023 / Revised: 12 July 2023 / Accepted: 14 July 2023 / Published: 16 July 2023

Abstract

:
Toll-like receptors (TLRs) have a critical role in the pathogenesis and disease course of viral infections. The induced pro-inflammatory responses result in the disturbance of the endovascular surface layer and impair vascular homeostasis. The injury of the vessel wall further promotes pro-thrombotic and pro-coagulatory processes, eventually leading to micro-vessel plugging and tissue necrosis. Moreover, TLRs have a direct role in the sensing of viruses and platelet activation. TLR-mediated upregulation of von Willebrand factor release and neutrophil, as well as macrophage extra-cellular trap formation, further contribute to (micro-) thrombotic processes during inflammation. The following review focuses on TLR signaling pathways of TLRs expressed in humans provoking pro-thrombotic responses, which determine patient outcome during viral infections, especially in those with cardiovascular diseases.

1. Toll-like Receptors’ Role in Inflammation and Thrombosis

Toll-like receptors (TLRs) play a major role in the modulation and progression of inflammation as a result of different pathogens such as bacteria, fungi and viruses [1,2]. Once they are synthesized in the endoplasmic reticulum, TLRs are transported to endosomal or plasma membranes [3]. TLRs mediate, as well as propagate, inflammation and, depending on how strongly a TLR is activated, the initiated responses can be beneficial or harmful to the host [4].
Pro-inflammatory diseases increase the rates of thrombo (-embolic) events as a result of increased thrombin generation due to inflammation [5]. Thrombin is the strongest human platelet activator [6] and systemic inflammation plays a major role in atherosclerosis [7]; hence, TLRs also have a direct effect on platelets’ capacity to modulate inflammation [8,9]. Pathways of immuno-thrombosis, initiated by platelet activation following viral sensing, are crucial in the pathogenesis of viral infections [10,11,12]. Herein, TLR-3, TLR-9 and TLR-10 respond to double-stranded (ds) ribonucleic acid (RNA), TLR-7 and -8 recognize single-stranded (ss) RNA and TLR-2 and TLR-4 sense viral envelope glycoproteins [13,14]. However, other TLRs also play a role in viral recognition [13,15]. TLR-3 has been shown to be involved in the development of the immune response to severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) and Middle East respiratory syndrome coronavirus (MERS-CoV) [16]. Increased levels of RNA transcription similar to the profile of TLR-3 were observed on the second day after coronavirus infection, shown in a model of non-sterile inflammation induced by viral injection in mice [16].
This increase in TLR-3 activity enhances downstream activities of toll-interleukin receptor (TIR) domain-containing adaptor protein inducing interferon (TRIF), interferon regulatory factor 3 (IRF3), NF-kB and pro-inflammatory cytokines, all of which can induce pro-coagulatory and pro-thrombotic responses [16]. During viral or bacterial infection type I interferon (IFN), signaling is induced upon detection of pathogens (by pathogen-associated molecular patterns) via pattern recognition receptors (PRRs) [17]. IRF3 is activated immediately after viral infection occurs and is a primary activator of IFN genes and the RANTES chemokine gene, finally resulting in the recruitment of leukocytes to sites of inflammation [18]. IFN signaling induces coagulation by activating the coagulation cascade by increasing the expression of high mobility group box 1 (HMGB1), which is a damage-associated molecular pattern (DAMP), in the bloodstream. In thrombi, HMGB1 is most commonly expressed on platelets [19]. TRIF redirects HMGB1 to activate granulocytes, monocytes, macrophages and dendritic cells, inducing coagulation [20]. Monocytes react to inflammation by expressing tissue factor, which induces the coagulation cascade. NF-kB further increases inflammation by inducing adhesion molecules necessary for leukocyte binding and transmigration [21].
Contrarily, TLR-3 deficient mice had an increased survival when compared with wild-type mice when subjected to influenza A virus by non-sterile inflammation due to intra-nasal inoculation [22]. Unsurprisingly, the TLR-3 deficient mice had higher viral production in their lungs [22]. Mouse models showed that protease-activated receptor (PAR)-2 suppressed TLR-3 signaling and thus contributed to viral infectivity [23]. These findings were contrary to previous reports showing increased susceptibility to non-sterile influenza A virus infection in PAR-2 deficient mice when compared with wild-type mice [24].
Traditionally, platelets have been thought to play a role in the amplification of the coagulation cascade at the site of vascular injury [25]. More recently, platelets have been recognized as drivers of leucocyte-mediated immunity [26,27]. Platelets express Fc gamma receptor IIa, which increases platelet functions, causing them to form platelet–leukocyte aggregates [28]. These aggregates can trap and immobilize pathogens [29]. Platelets can also actively regulate immuno-thrombosis in various diseases such as infection [30], injury as a result of ischemia and/or reperfusion [31], cardiovascular diseases (CVD) [32], sepsis [33] and cancer [34].
TLRs are central to immuno-thrombotic platelet function via expression on or within immune cells (such as platelets, monocytes/macrophages, neutrophil granulocytes, dendritic cells, natural killer cells and cells of adaptive immunity (T- and B- cells)) and thus play a vital role in the first line of protection from injury and infection [9,35,36,37].
TLRs are among the PRRs [38] and can recognize both PAMPs and DAMPs [39]. PAMPs are derived from pathogens [40], while DAMPs are associated with tissue damage, which is endogenous [38]. As such, TLRs are partially responsible for the elimination of viruses [41]. However, it must be noted, that this positive aspect can also have a negative impact on the host as a result of tissue destruction and persistent inflammation, such as in the pathogenesis of coronavirus disease 2019 (COVID-19) [42].

2. TLR Signaling

In humans, 10 types of TLR have been identified that fall into one of two groups based on their expression on different immune cells. The localization of the TLR expression determines the PAMP/DAMP specificity of the TLR [43]. TLRs 3, 7, 8 and 9 are expressed primarily intra-cellularly [44,45], while TLRs 1, 2, 4, 5, 6 and 10 are expressed primarily on the surface of the cell membrane [2,46,47,48]. The site of expression corresponds to their roles [43]. TLRs generally signal by dimerization after ligand binding [49]. The exception is TLR-2, which hetero-dimerizes with TLR-1 or TLR-6 and recognizes tri-acylated (TLR-1) and di-acylated (TLR-6) lipopeptides, respectively [50]. The occurrence of all 10 human TLRs has been described for human platelets [12].
TLRs consist of an extra-cellular leucine-rich repeat (LRR) domain responsible for PAMP and DAMP sensing, a transmembrane helix and a TIR domain located in the cytosol [51]. Downstream signaling involves the adaptor proteins called toll-interleukin-1 receptor resistance (TIR) domain-containing proteins. In humans, these are myeloid differentiation primary response protein 88 (MyD88), MyD88 adaptor-like (MAL) (also called TIR domain-containing adaptor protein (TIRAP)), TRIF (also known as TICAM1) and TRIF-related adaptor molecule (TRAM, also known as TICAM2) and TIR domain sterile alpha and HEAT/Armadillo motif (SARM) [52,53,54]. In contrast to MyD88, MAL, TRIF and TRAM (which have activating functions), SARM has negative regulatory effects on TRIF-dependent signaling [52,53,54,55]. Importantly, various TLRs use different TIR domain-containing adaptor proteins and induce a broad set of intra-cellular signal transduction pathways, which predominantly result in NF-κB activation [54].
In addition, a pro-apoptotic PI3K/AKT/GSK-3β pathway has been described in rats. [56,57].
TLRs initiate immune responses by activating transcription factors of the nuclear factor-κB (NF-κB) and the interferon regulatory factor (IRF) family in both a MyD88-dependent as well as a MyD88-independent manner [54,58,59]. NF-κB causes the production of pro-inflammatory cytokines and chemokines while also participating in inflammasome regulation. It is also critical in regulating inflammatory T cells and innate immune cells [58]. Modulation of TLR-induced pathways in a positive or negative manner can occur via activation of PI3K by different TLR-agonists such as LPS, CpG, flagellin and by-products resulting from viral infection [60]. In humans, all TLR-receptors utilize MyD88-induced signaling to induce inflammatory cytokine production [61,62]. The impact of MyD88 signaling after TLR-4 activation on inflammatory pathways can also be shown for TLR-3, which was initially thought to exclusively use the TRIF pathway [63]. Once these adaptor proteins bind to TLRs, cytosolic signaling complexes are activated. These contain tumor necrosis associated factor (TRAF) and interleukin receptor associated kinase (IRAK) proteins, which activate NF-κB and IRF, a transcription factor. These in turn trigger the production of pro-inflammatory cytokines and type 1 interferons [54,59]. NF-κB is required for interleukin (IL)-6 and tumor necrosis factor (TNF) production, which in turn activates the transcription of NF-κB [61,64]. As a result of this pro-inflammatory cascade, TLR-1/2 and TLR-4 cause increased P-selectin expression on platelets, activation of integrin alpha(IIb)beta(3) and increased production of reactive oxygen species (ROS) [65,66]. Moreover, thrombin generation can be induced by platelet activation via TLR-2 and TLR-4 [67]. Thus, TLRs can be considered to be the drivers behind the activation of pro-inflammatory processes in platelets, as NF-κB is responsible for first procaspase activating compound (PAC-1) and fibrinogen binding as well as adenosine triphosphate (ATP) release as a result of inflammatory and pro-thrombotic stimuli [68].

3. TLRs and Diseases

Viruses, such as severe acute respiratory syndrome corona virus type 2 (SARS-CoV-2), are pro-inflammatory and pro-thrombotic in nature [69]. The cytokine storm induced by SARS-CoV-2 is thought to highly involve TLR signaling [70,71]. Herein, SARS-CoV-2 shares these patho-mechanisms with other viruses; however, distinct differences in the regulation of inflammation and viral persistence can be observed [10,72,73,74]. In addition, SARS-CoV-2 leads to an upregulation of a plethora of TLRs [42,72]. Systemic hyper-inflammation is triggered via the previously outlined mechanism via TLR-2, -4, -6, -7 and -8 [42,72,75]. IL-1β is produced as a result of inflammasome activation and induces IL-6 [76]. High levels of inflammasome activation have been associated with poor outcomes in COVID-19 patients [77]. Moreover, long-lasting inflammatory processes maintaining endothelial dysfunction due to viral persistence might be the underlying cause of thromboembolic events and cardiovascular complications frequently observed in patients suffering from COVID-19 [10,78]. TLR-4 and Nox-2 inhibition is suggested to reduce oxidative stress and platelet-dependent thrombus growth in ex vivo models using the blood of SARS-CoV-2 patients. [79] Moreover, TNF α inhibition also reduces Nox-2-related oxidative stress and platelet activation enhanced by plasma of SARS-CoV-2 patients, thus eliciting signaling pathways in which TLR-4 activation promotes platelet-dependent thrombus growth [79].
There is increasing evidence that TLRs contribute to inflammatory vascular diseases, such as aneurysm formation and different forms of vasculitis [80], and might also be linked to micro- and macro-vascular complications in type 2 diabetes [81,82]. Disease involvement of TLRs is displayed in Table 1.

3.1. TLR-1

TLR-1 signaling involves downstream pathways of MyD88 [54]. Hally et al. showed TLR-1 to be significantly upregulated in platelets of patients with acute myocardial infarction (AMI). Platelets from AMI patients and healthy controls were analyzed and compared via Western blotting. While the role of TLR-1 is poorly characterized in AMI patients, it is likely that, similar to the manner in which TLRs exacerbate inflammation, TLR-1 may increase platelet reactivity and therefore thrombosis during and after AMI and thus present a further method of platelet activation [83].
Furthermore, there is evidence that overstimulation of TLR-1 is involved in the pathogenesis of autoimmune diseases such as diabetes mellitus type 1 (DM1) [84], which in itself induces a pro-thrombotic state [85]. The TLR 1/2 pathway, together with TLR-3-induced signaling, is implicated in the defense against chikungunya virus (CHIKV) infection [15]. CHIKV belongs to the alphavirus genus of Togaviridae and is transmitted by female mosquito Aedes arthropods [86]. The signaling pathways induced by CHIKV sensing involve MyD88 (by TLR 1/2) and NF-κB, as well as TRIF (by TLR-3) and IRF1 [15]. These pathways result in high IL-27 expression [15]. The latter has pleiotropic effects regarding immuno-modulation and may have implications for the pathogenesis of immune thrombocytopenia [87].

3.2. TLR-2

TLR-2 signaling involves downstream pathways of MyD88, TIRAP, TRAM and TRIF [88,89,90].
In addition, SARM is capable of TLR-2 signaling modulation [91].
After dimerization with TLR-1, TLR-2/1 causes platelet activation in a dose-dependent manner when subjected to the TLR-2/1 agonist Pam3CSK4 in both healthy subjects’ and AMI platelets and with and without dual anti-platelet therapy (DAPT, in this case aspirin and clopidogrel or ticagrelor) in in vitro experiments. It has been theorized that, by this mechanism, TLR-2/1 may be involved in the pathogenesis of AMI and could aggravate myocardial ischemia or reperfusion injury and recurrent atherothrombotic events [83].
In the presence of histones, which are released into the circulation during neutrophil extra-cellular trap (NET) formation, thrombin generation is driven by TLR-2- and TLR-4-induced platelet signaling [67]. This enhances platelet activation and promotes further platelet–leukocyte aggregate formation and activation of neutrophils leading to NETosis [10].
Thrombin is the strongest platelet activator and, despite current guideline-driven antiplatelet therapy, thrombin-induced platelet activation still accounts for a considerable and stable platelet aggregate formation [92,93,94].
TLR-2 is also upregulated in patients with abdominal aortic aneurysm (AAA) when compared with healthy individuals; it is currently believed that TLR-2 may be integral at regulating inflammation in the aorta in the context of AAA formation [95].
There is strong evidence that TLRs have a crucial role in the formation of AAA, which are defined as saccular distensions of the abdominal aorta exceeding 30 mm in diameter or 1.5-fold of the regular diameter [96,97,98,99]. The pathogenesis of AAAs is characterized by excessive diapedesis of leukocytes [100], inflammation [101] and the subsequent release of matrix metalloproteinases and elastase from macrophages and lymphocytes that degrade the extra-cellular matrix [102,103] and weaken the vessel wall. While the exact mechanisms that initiate the inflammatory response in the aortic wall have not yet been thoroughly understood, there are hints that TLR activation may be crucial in initiating inflammatory processes, which ultimately lead to AAA formation and atherogenesis [104,105].
There are several studies that have linked TLR-2 and TLR-4 to the formation of AAAs. Yan et al. showed that increased levels of TLR-2 expression were found in human samples of AAA tissue [106]. In addition, the inhibition of TLR-2 in a murine AAA model resulted in a significant reduction in AAA size and TLR-2-deficient mice failed to develop AAAs [106]. Proteins involved in inflammatory downstream signaling pathways, including matrix metalloproteinase and NF-κB, and macrophage recruitment were also significantly reduced in TLR-2-deficient mice [106]. In a TLR-4-deficient murine model of AAA formation, reduced levels of chemokines and interleukins were observed in comparison with a TLR-4-non-deficient murine control group [107].
The role of TLRs for AAA formation was further endorsed by Jabłońska et al. [99], who examined the levels of TLR messenger ribonucleic acid (mRNA) in the blood of AAA patients, healthy volunteers and AAA tissue samples. In the blood, both TLR-2 and TLR-4 mRNA expression was increased in AAA patients compared with control subjects. However, elevated protein levels in serum could only be proven for TLR-4. Compared with the serum levels, TLR-2 expression was increased 20-fold in the AAA specimens [99]. Furthermore, certain polymorphisms in the gene encoding for TLR-2 and TLR-3 were demonstrated to codetermine the risk of AAA formation [98].
Lastly, the knock-out of MyD88, a downstream signaling molecule involved in both the TLR-2 and TLR-4 pathway, reduced both AAA formation and atherosclerosis after angiotensin II infusion in mice predisposed to both disease entities by the knock-out of either apolipoprotein E or low-density lipoprotein receptor (LDL-R) [108]. While in TLR-2 and LDL-R-deficient mice, angiotensin II infusion resulted in AAA formation but not atherosclerosis, both were attenuated in mice deficient in TLR-4 and LDL-R [108].
These findings demonstrate that TLRs and their pro-inflammatory downstream signaling pathways have a crucial role in AAA initiation and formation.
As TLR involvement is crucial in AAA formation, it is not surprising that viral infections such as cytomegalovirus [109] or human immunodeficiency virus (HIV) [110] are discussed to contribute to aneurysm pathophysiology. However, the exact mechanisms and potential novel therapeutic target molecules will need to be identified in future studies.
In viral infections, TLR-2 not only recognizes SARS-COV-2 but is also responsible for sensing the CMV envelope glycoproteins B and H and responding to varicella zoster, vaccinia, Epstein–Barr, hepatitis B and hepatitis C viruses [13,111,112].
Furthermore, Sepehri et al. discussed the upregulated expression of TLR-2 being associated with an increased risk of type 2 diabetes mellitus (DM2). They concluded that, as a result of TLR-2 involvement in activating the innate immune response upon recognition of DAMPs, TLR-2 was responsible for the induction of ROS and inflammatory cytokines, which contributed to the exacerbation of DM2. TLR-2 expression increased in obese patients and correlated with increased serum levels of glucose and free fatty acids. Infections may be considered crucial for the development of DM2 as a result of PAMP-activated TLR-2-initiated pathways and that insulin suppresses TLR-2 expression. These mechanisms shed light on the circulus vitiosus of DM2 [113].
Finally, TLR-2 was shown to have similar involvement in the pathogenesis of auto-immune diseases as TLR-1 (DM1 [84], Graves’ Disease (GD) [114]).

3.3. TLR-3

TLR-3 can be stimulated in endothelial cells by endogenous RNA, which is released as a result of apoptosis and necrosis and causes a pro-inflammatory cellular response [115]. Short, single and double strands of RNA result in an inhibition to neo-angiogenesis [116]. TLR-3 signaling is mediated by TRIF and MyD88, hereby conferring the pro-inflammatory responses [63]. Signaling results in the phosphorylation of Akt, ERK1/2 and p38 MAPK and of the subunit p65 of NF-κB [117]. Najem et al. used a cell-permeant nucleic acid stain to test whether TLR-3 was involved in inflammatory venous thrombosis. Polyinosine polycytidylic acid (poly:C), a synthetic double-stranded RNA analog and TLR-3 ligand were given to wild-type mice after FeCl3 (non-sterile) induced inferior vena cava injury, increasing the size and cellular density of thrombi when compared with TLR-3 knock-out mice. As a result of this stimulation of the TLR-3 in this model of sterile inflammation, an increased production of reactive oxygen species was observed, as well as increased macrophage and neutrophil recruitment in the wild-type mice. These results strongly suggest that TLR-3 stimulation and RNA release, after endothelial injury, are involved in thrombus formation as a result of the pro-inflammatory response, which leads to the recruitment of macrophages and neutrophils to the injury site [118]. TLR-3 seems to play a more promotional role in platelet activation, as opposed to TLR-2 and -4, which react to classic platelet stimulation by thrombin, ADP or arachidonic acid (AA); in in vitro models TLR-3 activation fails to induce platelet aggregation [117]. However, the presence of suboptimal concentrations of AA, ADP and collagen and thrombin TLR-3 activation by synthetic dsRNA analog lead to a platelet aggregation of 60–80%. Thus, TLR-3 may be considered a promoter for platelet activation [117].
In vivo mouse models showed that TLR-3 knock-out mice reduced coagulatory markers when subjected to poly I:C compared with wild-type mice [119]. Thus, the activation of TLR-3 can induce an endothelial pro-coagulatory state, which can influence cellular hemostasis [119]. There is evidence that the activation of protease activated receptor (PAR)-1 in the presence of dsRNA analog induced INF-β expression in murine models that were infected with coxsackievirus B3 (CBV3). However, this induction of INF-β expression was not present when PAR-2 was activated. Thus, it is believed that PAR-2 negatively regulates TLR-3-dependent INF-β expression [23,120,121]. In vitro experiments using mouse models showed that PAR-4 activation increased chemokine expression, while decreasing TLR-3-related NF-κB expression of pro-inflammatory genes [122]. Wild-type mice had lower immune cell numbers, fewer inflammatory mediators in the lung and decreased mortality when compared with PAR-4 knock-out mice [122].
In a cohort of Danish females, the upregulation of TLR-3 was associated with systemic lupus erythematosus (SLE) [123]. The pro-thrombotic state of SLE was, in part, due to the systemic inflammation and increased circulating immune complexes that were modulated by TLRs [124]. Akin to SLE, TLR-3 is believed to be involved in the pathogenesis of DM1 [84].
TLR-3 is essential for anti-viral activity during rhinovirus infection by inducing IL-6, CXCL8 and CCL5 [125]. Moreover, TLR-3 mRNA expression is induced by rhinovirus replication [125]. During infection with the West Nile virus, TLR-3 response accounts for the development of lethal encephalitis [126]. Herein, the breakdown of the blood–brain barrier is mediated by tumor necrosis factor alpha receptor 1 signaling [126]. It may be assumed that inflammation-mediated endothelial dysfunction with glycocalyx disintegration may be crucial in the disruption of the blood–brain barrier [127].

3.4. TLR-4

TLR-4 can be activated by various ligands, including lipopolysaccharides, viral glycoproteins, tenascin-C, fibronectin extra domain A and extra-cellular cold-inducible RNA-binding protein (eCIRP) [13,128,129,130,131,132]. The latter is released during sepsis, tissue ischemia–reperfusion injury, trauma and hemorrhage and acts as an endogenous DAMP [132,133]. TLR-4 can activate MyD88- and TIRAP, as well as TRIF-dependent pathways [134,135] (Figure 1).
These pathways, with cross-talks between them, result in the phosphorylation of MAP kinases and activation of IKK alpha/beta, NEMO, IKKε and TBK1, which lead to the phosphorylation and activation of transcription factors such as NF-κB, IRFs, activator protein 1 (AP-1) and activating transcription factor 2 (ATF2) [54,134,136].
Signaling through TIRAP, but also through MyD88, activates double-stranded RNA (dsRNA)-activated protein kinase PKR, which is upstream of MAPK signaling and results in NF-κB activation [135]. PKR has also been shown to be capable of PI3K/Akt pathway activation during neo-vascularization [137]. In addition, the activated PKR signaling might have a modulatory role, as it can also regulate NRF2 activation, a transcription factor promoting the expression of anti-oxidant enzymes such as heme oxygenase 1 (HO-1) or superoxide dismutase 1 (SOD-1) [138,139].
The adaptor TRAM has been described to bridge to TRIF and so both TRAM- and TRIF-associated signaling leads to IRF3 activation via IKKε and TBK1 [54,140]. The fifth adaptor protein, SARM, has been described to negatively regulate MyD88 and TRIF- mediated TLR-4 signaling [52,141].
TLR-4 has also been considered to play a role in the induction of apoptosis and, most prominently, fibrosis [142]. TLR-4 has been suggested to induce apoptosis via the PI3K/AKT/GSK-3β signaling pathway [57].
Significant evidence exists for the participation of TLR-4 in coagulation by several mechanisms [143]. Among these, TLR-4 can promote endothelial and platelet activation; the latter is also mediated by the internalization of micro-particles [144,145].
Moreover, via NF-κB and AP-1 activation, TLR-4 mediates together with TLR-2 tissue factor (TF) expression on endothelial cells [146]. In monocytes, TF expression mediated by TLR-4 and TLR-6 has been described [147].
The dual nature of TLRs on platelets is evident in TLR-4, which has been shown to both augment and inhibit neutrophil responses such as platelet–neutrophil aggregates, neutrophil extra-cellular trap formation and bacterial trapping in septic patients [148]. When a co-culture of neutrophils and platelets is subjected to TLR-4 agonists, CD62L (L-selectin) expression, phagocytosis and IL-8 secretion are increased, while shedding of CD62L and elastase secretion are decreased. Thus, platelet TLR-4 is responsible for neutrophil responses to pathogens and lipopolysaccharides (LPS) [149]. The latter facilitate the aggregation of platelets and neutrophils and the production of NETs [149]. TLR-4 signaling also mediates platelet–monocyte interactions and is required for P-selectin-induced platelet–monocyte aggregation [26,150]. In addition, TLR-4 induces caspase-1 activation and caspase-11 expression, leading to cellular pyroptosis [151]. Caspase-11-mediated inflammatory responses occur partly via gasdermin D-induced pyroptosis in macrophages, a process involved in the pathogenesis of atherosclerosis [152]. Hence, TLR-4 signaling promotes gasdermin D-induced effects. The latter are mediated via the NF-kB pathway [153]. Gasdermin D and caspase-1 signaling have furthermore been shown to be involved in TLR-4-induced macrophage extra-cellular trap (MET) formation and METosis [133]. METosis is a process wherein monocytes or macrophages release anti-microbial proteins and DNA which form extra-cellular traps [133]. Akin to NETosis, METosis leads to the release of DNA, anti-microbial proteins and histones from monocytes or macrophages, promoting extra-cellular trap formation and being a highly potent activator of immuno-thrombosis [133,154] (see Figure 2).
Tissue-type plasminogen activator (tPA) is a major activator of fibrinolysis; the anti-inflammatory properties of enzymatically inactive (EI) tPA are TLR specific. EI tPA, reduces the pro-inflammatory process in bone marrow-derived macrophages (BMDMs) as a result of LPS activity by blocking BMDMs to some of the TLR specific agonists. This inhibits the expression of TNFα and ILs [155]. A deregulated example of this function occurs in diabetics who have wound healing disorders as a result of increased inflammatory activity, in part due to TLRs [156].
There is evidence that TLR-4 may be solely responsible for fibrinous cardiac remodeling after ischemic events. Mouse models have shown that TLR-4 knock-out mice had no evidence of fibrinous remodeling, even in the presence of fibrin modulators and agonists, whereas wild-type mice experienced typical cardiac remodeling with fibrinous elements after permanent ligation of the left descending coronary artery (sham surgery). Furthermore, knock-out TLR-4 mice had reduced left ventricular remodeling and increased preservation in systolic function [157].
Human models have shown significant upregulation of TLR-4 on platelets in patients after experiencing an acute myocardial infarction. Furthermore, in vitro experiments have demonstrated that healthy and AMI platelets are activated in the presence of high doses of the TLR-4 agonist LPS. Increased activation of TLR-4 is also associated with heart failure following AMI [158].
TLR-4 likely plays a key role in the pathogenesis of Graves’ Disease and may contribute to heart failure in these patients [114].
Similar to TLR-2, TLR-4 is upregulated in AAA patients when compared with healthy patients (please see paragraph concerning TLR-2) [95].
During Dengue virus infection, the flavivirus non-structural protein 1 (NS1) mediates TLR-4-associated cytokine production [159]. NS1, which is a secreted glycoprotein from Dengue, Zika, West Nile, Japanese encephalitis and yellow fever viruses, is implicated in viral replication, immune evasion and vascular leakage [160]. Herein, it contributes also to Dengue hemorrhagic fever and shock [42]. The emergence of hyper-permeability, and in turn tissue edema, is induced by the disruption of the glycocalyx components heparan sulfate, sialic acid and syndecan 1 [160,161,162]. This is mediated via the upregulation of the enzymes sialidases and heparanase contributing to glycocalyx degradation [161,163]. However, pathomechanisms seem to be more distinct in Dengue virus infection than in infection with other flaviviruses [163].
Many viruses cause endoplasmic reticular stress when they use the cell machinery to produce large amounts of viral proteins [164]. These stressed or dying cells release TLR-4 agonists, for example, high mobility group protein 1 (HBGP1). This causes an inflammatory reaction, which can also be observed in obese individuals [165].
TLR-4 in neutrophils also has a key role in NET formation, for example, when recognizing respiratory syncytial virus (RSV) fusion protein [166]. In mice, TLR-4 has also been involved in the reactivation of cytomegalovirus, which has been previously intra-peritoneally injected (as a non-sterile infection/inflammatory model), from latency after LPS stimulation [167]. Interestingly, TLR-4 has also had a role in long-term post-COVID-19 sequelae [168,169]. Herein, S100A8/A9, a calcium-binding protein, stimulates the TLR-4/receptor for advance glycation end-products’ (RAGE) pathway and chronically activates IL-1b, IL-6 and tumor necrosis factor (TNF)-alpha expression [168]. Neuro-inflammation triggered by SARS-CoV-2 spike protein, which was injected intra-cerebroventricularly (non-sterile infection model), which binds to TLR-4, has been discussed to mediate long-term cognitive impairment after COVID-19 [169].

3.5. TLR-5

TLR-5-mediated signaling involves TIRs, MyD88, TIRAP, TRIF and possibly also TRAM [88].
TLR-5 acts as a sensor for the immune system against bacteria by capturing flagellated bacteria. Flagellin, a structural protein of the flagellum, stimulates inflammatory responses and development of adaptive immunity in humans. Once the protein ligand on the flagellum is bound by TLR-5, MyD88 and TRIF are recruited. This leads to NF-κB activation and cytokine secretion and the inflammatory response is induced [170].
Xiao et al. showed that TLR-5 might be associated with decreased GD susceptibility in female subjects in a Chinese Cantonese population. Gene polymorphisms related to TLR-5 in 332 GD patients compared with 351 healthy controls were associated with a decreased risk of GD in women [171].
TLR-5 expression was significantly elevated in patients with severe COVID-19 [72]. However, TLR-5 has been discussed to confer beneficial effects during viral infections such as influenza A and COVID-19 [80,172]. In influenza A infection, activation of the TLR-5 pathway by flagellin has shown a decrease in viral RNA, possibly independent of signaling via type I interferon and IL-22 [172]. Furthermore, TLR-5 seems to have a role in the inhibition of hepatitis B virus [173]. The involvement of TLR-signaling pathways in hepatitis B pathophysiology is supported by the results of a transgenic mouse model with injections of an anti-CD40 agonist, CD40 alpha, which showed the inhibition of HBV replication by induction of inflammatory cytokines [174].
On the other hand, recent results using non-sterile inflammatory models in mice suggest that, during COVID-19, TLR-5 signaling might enhance SARS-CoV-2 infectivity [175].

3.6. TLR-6

TLR-6-mediated signaling involves TIRs, MyD88 and TIRAP [88,135].
TLR-6, along with TLR-4, seems to play a significant role in thrombosis in patients with high levels of LDL. Increased LDL promotes inflammation through oxidative stress. This causes the expression of the pro-coagulatory tissue factor (TF). [176] Owens et al. designed an experiment using animal models, where adding simvastatin reduced the expression of TF. It was further shown that deficiency in TLR-4 and TLR-6 reduced levels of micro-particles in the plasma, reduced expression of TF and reduced coagulation and inflammation in hyper-cholesterolemic mice and monkeys. Thus, the involvement of TLR-6 and -4 may be considered a major contributor to atherosclerosis [147].
The synergistic activation of TLR-2/6 and TLR-9 has been shown to protect mice against non-sterile influenza virus infection in a non-sterile inflammation model in mice [177].

3.7. TLR-7 and -8

Signaling through TLR-7 involves TIRs, MyD88, TRAM and also TIRAP [178,179]. Interestingly, TLR-7 and TLR-9 signaling can be modulated by SARM1, which induces via this pathway apoptosis in neurons [180].
TLR-8 mediated signaling involves TIRs, MyD88 and TIRAP [181].
So far, only TLR-7 has been shown to have significant involvement in monocyte conversion to dendritic cells to support the primary immune response against pathogens. This occurs when TLR-7 induces cytokine production in monocytes and disposal of damaged cells. Chronic TLR-7 stimulation causes monocytes to differentiate into macrophages [182].
Moreover, TLR-7 signaling in plasmacytoid dendritic cells (pDCs) involves the translocation of IRF5 and IRF7 from the cytosol to the nucleus and might herein be involved in the activation of pDCs [183].
Both TLRs 7 and 8 bind single-stranded RNA, thus initiating the immune response against viruses [183,184]. Myocardial cells have been shown to express TLR-7 and -8 when subjected to coxsackie B viruses [185]. This may explain the production mechanism of IL-6, INF-β and TNFα in myocarditis patients and, in part, the chronic aspect of the disease [185], while IL-6 promotes platelet production by acting upon megakaryocytes and hepatocytes (increased release of thrombopoietin) [186]. Highly increased levels of IFN-β have been correlated with thrombocytopenia [187]. Finally, TNFα is considered the causal molecule for platelet hyper-reactivity and the formation of larger thrombi in older humans and aged mouse models [188].
Platelets also play a vital role in the immune response to viruses such as influenza virus type A. Platelets express TLR-7 on their cell surface. Once activated, TLR-7 causes platelets to express alpha granules, P-selectin and CD40L, leading to a platelet-driven pro-thrombotic effect. Driven by TLR-7, platelets can engulf the virus, causing the release of complement factor C3, which stimulates the release of neutrophil DNA, thus promoting the formation of platelet–neutrophil aggregates preceding NET formation [189].
TLR-7 has been reported to be involved in type I IFN induction by Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV) [190]. In SARS-CoV-2-infected patients, a decrease in pDCs was observed, correlating with disease severity [191]. It is suggested that SARS-CoV-2 dampens TLR-7 responses through interaction with neuropilin-1 [191]. In pDCs, TLR-7 induced pathways after viral RNA sensing trigger MyD88-IRAK4-TRAF6 signaling, leading to CXCL10 induction as well as IRF7 phosphorylation, translocation mediating type I and III interferon expression [191].
Genetic polymorphisms of TLR-7 and -8 have been shown to predict susceptibility to CHIKV [192]. However, in hepatitis C virus infection, TLR-7, together with TLR-3 signaling, seems to constitute a protective immune response [193]. This hypothesis was corroborated by a study showing that myocarditis (coxsackie B virus) patients with mutant TLR-3 phenotype had increased viral replication when compared with patients with a normal TLR-3 phenotype, thus showing that genetic differences in TLR-3 together with PAR-2 modulation of INF- β effect the host’s vulnerability to viral cardiomyopathies [23,194].
Pro-coagulant pathways have been described after the recognition of HIV nucleic acids by TLR-7 and -8 on neutrophils, leading to NET production [195].
TLR-7 has been shown to be involved in the pathogenesis of SLE in Japanese females, when gene analysis was performed, compared with a healthy control [196], while TLR-8 has been positively correlated to SLE in a Danish population [123].
TLR-7 has been shown to be involved in the pathogenesis of GD [171], while TLR-8 seems to be involved in the pathogenesis of rheumatoid arthritis [197].
Similar to TLR-1, -2 and -3, TLR-7 mediates inflammation contributing to DM1 [84].

3.8. TLR-9

TLR-9 is integral to inflammation and metabolism. The pathways induced by TLR-9 activation involve TIRs, MyD88 and TIRAP [198]. In addition, signaling through TRIF has also been shown [199]; studies regarding TRAM are missing [88]. Together with TLR-7, TLR-9 can induce apoptosis via SARM1 [180]. As hitherto known, signaling through TLR-9 can result in NF-κB or IRF-7-dependent type I interferon (IFN) pathway activation [200]. Further, PI3Kγ has also had some critical roles in the modulation of immune responses mediated by TLR-9 [60].
Similar to TLR-7 signaling, TLR-9-mediated signaling in pDCs involves translocation of IRF5 and IRF7 from the cytosol to the nucleus [183].
It is known that, in humans, BAD-LAMP (LAMP5) dampens TLR-9-mediated type I IFN production by control of TLR9 sorting in a different endosome subset [201].
TLR-9 is associated with the pathogenesis of non-alcoholic steatosis hepatis (NASH) and is likely a driver for NASH-associated fibrosis, as it has been shown to be expressed in 13.3% of normal liver tissue, 53.3% in mildly fibrotic patients, 80% in cases of cirrhosis and 95% in hepato-cellular carcinoma patients. TLR-9 is activated by circulating mitochondrial DNA, which is increased in obese individuals, metabolic dysfunction-associated fatty liver disease and NASH [202].
Similar to TLRs 7 and 8, TLR-9 is involved not only in the pathogenesis of auto-immune diseases such as SLE (shown in Asian and Danish cohorts) but also in rheumatoid arthritis and multiple sclerosis [123,203,204].
In mice, TLR-9 is involved in non-sterile cytomegalovirus infection, as shown in a model of non-sterile inflammation [205]. Furthermore, TLR-9 senses herpes simplex virus type 1 and 2 and Epstein–Barr virus [206,207,208,209].
MyD88 activation and signaling through IRAK4 suppresses lytic reactivation of Epstein–Barr virus and favors its latency in B-cells [210]. In peripheral T-cell lymphomas, TLR-9 and programmed cell death-ligand 1 (PD-L1) expression are associated with poor survival [211]. Hence, targeting of both TLR-9 and PD-L1 is suggested to induce a sustained anti-tumor immunity [212].
TLR-9 deficient mice have been compared to wild-type mice with regard to their capability of resolving venous thrombosis. The TLR-9-inhibited and -deficient mice were less capable of resolving venous thrombosis after inferior vena cava ligation when compared with wild-type mice. When wild-type mice were subjected to a TLR-9 stimulant, early venous thrombosis resolution was accelerated [213].

3.9. TLR-10

TLR-10 mediated signaling involves MyD88 and possibly also TRIF [214].
TLR-10 seems to be the only human TLR that has an inhibitory function over the innate immune system and inflammation. Its role in this modulatory function within the innate immunity is largely unknown (except an inhibitory effect on TLR-2 responses [215]) and it is assumed that the exact anti-inflammatory properties and the impact on the trained immune response in humans as well as therapeutic options remain to be established [216]. Homo-dimer TLR-10/10 and hetero-dimer TLR-10/2 have been shown to recruit MyD88 [216]. Different ligands are discussed for binding to TLR-10, among those HIV-gp41, in turn promoting IL-8 production and NF-κB activation [217]. Moreover, TLR-10 is able to bind dsRNA in an acidic environment [14]. After recruitment of MyD88, the initiated pathway inhibits the production of interferon regulatory factor-7-dependent type I IFN [14]. In addition, a cross-talk with TLR-3-initiated pathways has been described [14]. Furthermore, Lee et al. have discussed TLR-10 as a relevant viral sensor of innate immunity [218].

4. Discussion

The synergistic patho-mechanisms of inflammation and in consequence disturbance of the endothelial surface layer with altered vascular perfusion, (micro-)thrombosis and tissue edema drive life-threatening complications of viral infections [10,219].
TLRs are a key factor in regulating NET formation, as the activation of TLRs on neutrophils triggers NET release and herein the binding, immobilization and inactivation of viruses [220]. Similarly, TLR- 4-induced METosis leads to the release of nuclear and mitochondrial DNA and histones [133]. The released histones are recognized as DAMP and activate platelets via TLR-2 and -4, leading to thrombin generation [67,221] (see also Figure 1). Moreover, signaling through TLR-2 increases vWF release from alpha granules in megakaryocytes/platelets [222,223] and also from Weibel–Palade bodies in endothelial cells [224]. As recently reviewed, these mechanisms are central to SARS-CoV-2 patho-physiology [10] but can also be observed in other viral infections [220,225,226]. One can assume that viral infections leading to (micro-) thrombotic complications are characterized by a TLR-vWF-NETosis axis, which in itself drives the processes of immuno-thrombosis impairing (micro-)vascular integrity. The disturbance of the latter is an underlying cause of Virchow’s triad impairment and drives tissue hypoxia, leading to organ failure [10]. In this context, rheological changes due to infection and inflammation should also be considered [227]. When exposed to oxidative inflammation, red blood cell membrane fluidity decreases, impairing systemic micro-circulation and, therefore, tissue perfusion [228]. The latter could be shown in different cardiovascular diseases [229,230,231,232], where a chronic inflammatory oxidative stress burden co-exists [233,234,235]. Limitation of oxidative injury might be given by PKR activation, which is known to enhance NRF2-mediated gene expression of anti-oxidant proteins such as SOD-1 and HO-1 [138,139].
This could be a negative feedback loop that limits inflammatory processes mediated by TLRs. However, the exact mechanisms should be studied in different conditions of inflammation, e.g., atherosclerosis or ischemia–reperfusion injury and inhibiting concepts such as cellular conditioning or HO-1 induction [236,237,238,239].
It should also be noted that pathways of immuno-thrombosis induced by TLR signaling contribute to changes in the vascular wall, including atherosclerosis and aneurysm development and progress [240,241].
Anemia as a result of inflammatory processes has been previously recognized and widely discussed [242]. Though a result of multiple causes, anemia can also be driven by chronic TLR-7 and TLR-9 signaling, initiating the differentiation of inflammatory hemophagocytes [243]. The latter are also responsible for thrombo-cytopenia [243]. Moreover, infection with SARS-CoV-2 leads to elevated RBC calcium levels, resulting in higher RBC fragility [244]. In hospitalized COVID-19 patients, anemia is linked to decreased survival [245].
Anemia leads to alterations of platelet function with enhanced monocyte–platelet aggregate formation and P-selectin expression, as observed in patients with DAPT consisting of either aspirin and clopidogrel or aspirin and prasugrel/ticagrelor, respectively [246]. Furthermore, the highest risk of ischemic events has been reported in anemic patients with high on-treatment residual platelet reactivity (HRPR); however, the highest risk of bleeding has been reported in anemic patients without HRPR [247].
Therapeutic possibilities influencing TLR pathways are challenging, since they are limited by side effects through pleiotropic functions. In discussion as potential benefits are TLR agonists, such as TLR-3 agonist poly(I:C), which has been shown to confer anti-viral effects in animals [248]. TLR-9 agonism by oligonucleotides enhances cytokine production and modulates viral response [248]. However, it should be noted that the models of TLR agonism represent a “sterile” inflammatory, which might not depict all processes involved after pathogen-induced signaling.
On the other hand, TLR-7/9 antagonists such as chloroquine, hydroxy-chloroquine and quinacrine have been widely used for the treatment of immune-mediated inflammatory disorders (herein, SLE, rheumatoid arthritis, and Sjögren’s syndrome) [249].
The activation of TLR-7 has been suggested to modulate hepatitis B, herpes simplex and human papillomavirus infections [13]. In SARS-CoV-2 infection, TLR-2/6 agonism by INNA-051 has shown promising results in reducing viral RNA levels in a non-sterile infection/inflammation model in ferrets [250]; mouse models have shown that activation of TLRs 2 and 7 induces pro-coagulatory transcription factor expression in (non-sterile) sepsis-induced coagulopathy, making it a possible therapeutic goal in the future [251].
Table 1. Diseases linked to toll-like receptors. Table showing an overview of diseases discussed in the manuscript and the TLRs involved in their pathogenesis.
Table 1. Diseases linked to toll-like receptors. Table showing an overview of diseases discussed in the manuscript and the TLRs involved in their pathogenesis.
DiseasesTLRs InvolvedReferences
Auto-immune
Graves’ Disease1, 2, 5, 7, 8[114,171]
Multiple Sclerosis9[204]
Rheumatoid Arthritis2, 8, 9[48,91,197,203]
Systemic Lupus Erythematosus3, 7, 8, 9[123,196]
Cardiovascular
Abdominal Aortic Aneurysm2, 4[95,98,99,104,105,106,108]
Acute Myocardial Infarction1, 2, 4[83,157,158]
Atherosclerosis1, 2, 4, 6[104,152,240]
Vasculitis4, 5[80]
Infectious
Chikungunya Virus1, 2, 3, 7, 8[15,192]
Cytomegalovirus2, 4, 7, 9[109,167,205]
Coronavirus Disease 20192, 4, 5, 6, 7, 8[42,72,74,75,168,169,175,250]
Dengue Virus4[159,160]
Epstein Barr Virus2, 7, 9[13,209]
Herpes Simplex Virus 1 and 29[207,208]
Hepatitis B2, 5, 7[13,112,173]
Hepatitis C2, 3, 4, 7[13,111,193]
Human Immunodeficiency Virus7, 8, 10[110,195,217]
Influenza2, 5, 6, 7, 9, 10[22,172,177,179,189,218]
Middle Eastern Respiratory Syndrome3, 7[16,190]
Respiratory Syncytial Virus4[166]
West Nile Virus3[126]
Varicella Zoster2[13]
Metabolic
Diabetes Mellitus Type 11, 2, 3, 4, 7, 9[82,84]
Diabetes Mellitus Type 21, 2, 4[81,82,113]
Non-Alcoholic Steatosis Hepatis9[202]
In general, our review is intended to raise awareness regarding thrombo-inflammatory pathways mediated by TLR responses. This should give opportunities for hypothesis generation in future research. Although we have used as data source the NCBI database PubMed and the herein indexed publications with a broad strategy on used MESH terms, a limitation to our review is its narrative character, which also mirrors the opinion of the authors.
Moreover, we attempted to describe human TLR receptors and signaling pathways; however, knowledge is often limited by the availability of animal-based models.

5. Conclusions

Pathways induced by TLR signaling are complex and can promote beneficial effects such as viral elimination with side effects harming tissue homeostasis [10]. TLR pathways can result in a burst of immuno-thrombosis, resulting in NET and MET production, promoting a pro-inflammatory and pro-thrombotic response destabilizing the equilibrium of vascular and platelet function [148,150]. Moreover, TLR pathways may play an important role in virus reactivation and associated long-term pro-inflammatory responses [167,169].
Patients’ comorbidities and the multi-level effects of viral infections including inflammation-driven pro-thrombotic effects pose therapeutical challenges and the potential for adverse drug interactions without a clear clinical benefit. Further studies to elucidate the cross-talks in TLR signaling with a focus on viral long-term sequelae are warranted.
Pathogen entry causes sensing by TLRs, which in turn activate platelets leading to platelet–leukocyte aggregation [26]. TLR signaling mediates neutrophil and macrophage activation and promotes neutrophil extra-cellular trap (NET) as well as macrophage extra-cellular trap (MET) formation [37,133,150]. NETosis and METosis cause the release of DNA and histones, which, as DAMPs, drive further pro-inflammatory and pro-coagulatory responses via TLRs [133,154]. Immuno-thrombotic processes lead to alterations based on Virchow’s triad, of which endothelial injury with glycocalyx degradation is crucial in patho-physiological processes [10].

Author Contributions

Conceptualization, P.P.W.; Writing—Original Draft Preparation, B.P., M.P. and P.P.W.; Writing—Review and Editing, B.P., C.W.K., C.N., R.K., A.J., M.P., T.G., B.J. and P.P.W.; Supervision, C.W.K. and P.P.W. All authors have contributed substantially to the work. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Vijay, K. Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int. Immunopharmacol. 2018, 59, 391–412. [Google Scholar] [CrossRef] [PubMed]
  2. Agier, J.; Zelechowska, P.; Kozlowska, E.; Brzezinska-Blaszczyk, E. Expression of surface and intracellular Toll-like receptors by mature mast cells. Cent. Eur. J. Immunol. 2016, 41, 333–338. [Google Scholar] [CrossRef] [Green Version]
  3. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef] [PubMed]
  4. Marks, K.E.; Cho, K.; Stickling, C.; Reynolds, J.M. Toll-like Receptor 2 in Autoimmune Inflammation. Immune Netw. 2021, 21, e18. [Google Scholar] [CrossRef] [PubMed]
  5. Reganon, E.; Vila, V.; Martinez-Sales, V.; Vaya, A.; Aznar, J. Inflammation, fibrinogen and thrombin generation in patients with previous myocardial infarction. Haematologica 2002, 87, 740–745, discussion 745. [Google Scholar] [PubMed]
  6. Yun, S.H.; Sim, E.H.; Goh, R.Y.; Park, J.I.; Han, J.Y. Platelet Activation: The Mechanisms and Potential Biomarkers. BioMed Res. Int. 2016, 2016, 9060143. [Google Scholar] [CrossRef] [Green Version]
  7. Kaplan, R.C.; Frishman, W.H. Systemic inflammation as a cardiovascular disease risk factor and as a potential target for drug therapy. Heart Dis. 2001, 3, 326–332. [Google Scholar] [CrossRef]
  8. Vallance, T.M.; Zeuner, M.T.; Williams, H.F.; Widera, D.; Vaiyapuri, S. Toll-like Receptor 4 Signalling and Its Impact on Platelet Function, Thrombosis, and Haemostasis. Mediat. Inflamm. 2017, 2017, 9605894. [Google Scholar] [CrossRef] [Green Version]
  9. Aslam, R.; Speck, E.R.; Kim, M.; Crow, A.R.; Bang, K.W.; Nestel, F.P.; Ni, H.; Lazarus, A.H.; Freedman, J.; Semple, J.W. Platelet Toll-like receptor expression modulates lipopolysaccharide-induced thrombocytopenia and tumor necrosis factor-alpha production in vivo. Blood 2006, 107, 637–641. [Google Scholar] [CrossRef] [Green Version]
  10. Wadowski, P.P.; Panzer, B.; Jozkowicz, A.; Kopp, C.W.; Gremmel, T.; Panzer, S.; Koppensteiner, R. Microvascular Thrombosis as a Critical Factor in Severe COVID-19. Int. J. Mol. Sci. 2023, 24, 2492. [Google Scholar] [CrossRef]
  11. Sartorius, R.; Trovato, M.; Manco, R.; D’Apice, L.; De Berardinis, P. Exploiting viral sensing mediated by Toll-like receptors to design innovative vaccines. NPJ Vaccines 2021, 6, 127. [Google Scholar] [CrossRef] [PubMed]
  12. Hally, K.; Fauteux-Daniel, S.; Hamzeh-Cognasse, H.; Larsen, P.; Cognasse, F. Revisiting Platelets and Toll-like Receptors (TLRs): At the Interface of Vascular Immunity and Thrombosis. Int. J. Mol. Sci. 2020, 21, 6150. [Google Scholar] [CrossRef] [PubMed]
  13. Xagorari, A.; Chlichlia, K. Toll-like receptors and viruses: Induction of innate antiviral immune responses. Open Microbiol. J. 2008, 2, 49–59. [Google Scholar] [CrossRef]
  14. Lee, S.M.; Yip, T.F.; Yan, S.; Jin, D.Y.; Wei, H.L.; Guo, R.T.; Peiris, J.S.M. Recognition of Double-Stranded RNA and Regulation of Interferon Pathway by Toll-like Receptor 10. Front. Immunol. 2018, 9, 516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Valdes-Lopez, J.F.; Fernandez, G.J.; Urcuqui-Inchima, S. Synergistic Effects of Toll-like Receptor 1/2 and Toll-like Receptor 3 Signaling Triggering Interleukin 27 Gene Expression in Chikungunya Virus-Infected Macrophages. Front. Cell Dev. Biol. 2022, 10, 812110. [Google Scholar] [CrossRef] [PubMed]
  16. Totura, A.L.; Whitmore, A.; Agnihothram, S.; Schafer, A.; Katze, M.G.; Heise, M.T.; Baric, R.S. Toll-like Receptor 3 Signaling via TRIF Contributes to a Protective Innate Immune Response to Severe Acute Respiratory Syndrome Coronavirus Infection. mBio 2015, 6, e00638-15. [Google Scholar] [CrossRef] [Green Version]
  17. Ryan, T.A.J.; O’Neill, L.A.J. An Emerging Role for Type I Interferons as Critical Regulators of Blood Coagulation. Cells 2023, 12, 778. [Google Scholar] [CrossRef]
  18. Grandvaux, N.; Servant, M.J.; tenOever, B.; Sen, G.C.; Balachandran, S.; Barber, G.N.; Lin, R.; Hiscott, J. Transcriptional profiling of interferon regulatory factor 3 target genes: Direct involvement in the regulation of interferon-stimulated genes. J. Virol. 2002, 76, 5532–5539. [Google Scholar] [CrossRef] [Green Version]
  19. Vogel, S.; Bodenstein, R.; Chen, Q.; Feil, S.; Feil, R.; Rheinlaender, J.; Schaffer, T.E.; Bohn, E.; Frick, J.S.; Borst, O.; et al. Platelet-derived HMGB1 is a critical mediator of thrombosis. J. Clin. Investig. 2015, 125, 4638–4654. [Google Scholar] [CrossRef] [Green Version]
  20. Ruf, W. TRIF turns the switch for DIC in sepsis. Blood 2020, 135, 1073–1074. [Google Scholar] [CrossRef] [Green Version]
  21. Mussbacher, M.; Salzmann, M.; Brostjan, C.; Hoesel, B.; Schoergenhofer, C.; Datler, H.; Hohensinner, P.; Basilio, J.; Petzelbauer, P.; Assinger, A.; et al. Cell Type-Specific Roles of NF-kappaB Linking Inflammation and Thrombosis. Front. Immunol. 2019, 10, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Le Goffic, R.; Balloy, V.; Lagranderie, M.; Alexopoulou, L.; Escriou, N.; Flavell, R.; Chignard, M.; Si-Tahar, M. Detrimental contribution of the Toll-like receptor (TLR)3 to influenza A virus-induced acute pneumonia. PLoS Pathog. 2006, 2, e53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Weithauser, A.; Bobbert, P.; Antoniak, S.; Bohm, A.; Rauch, B.H.; Klingel, K.; Savvatis, K.; Kroemer, H.K.; Tschope, C.; Stroux, A.; et al. Protease-activated receptor-2 regulates the innate immune response to viral infection in a coxsackievirus B3-induced myocarditis. J. Am. Coll. Cardiol. 2013, 62, 1737–1745. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Khoufache, K.; LeBouder, F.; Morello, E.; Laurent, F.; Riffault, S.; Andrade-Gordon, P.; Boullier, S.; Rousset, P.; Vergnolle, N.; Riteau, B. Protective role for protease-activated receptor-2 against influenza virus pathogenesis via an IFN-gamma-dependent pathway. J. Immunol. 2009, 182, 7795–7802. [Google Scholar] [CrossRef] [Green Version]
  25. Periayah, M.H.; Halim, A.S.; Mat Saad, A.Z. Mechanism Action of Platelets and Crucial Blood Coagulation Pathways in Hemostasis. Int. J. Hematol. Oncol. Stem Cell Res. 2017, 11, 319–327. [Google Scholar]
  26. Dib, P.R.B.; Quirino-Teixeira, A.C.; Merij, L.B.; Pinheiro, M.B.M.; Rozini, S.V.; Andrade, F.B.; Hottz, E.D. Innate immune receptors in platelets and platelet-leukocyte interactions. J. Leukoc. Biol. 2020, 108, 1157–1182. [Google Scholar] [CrossRef]
  27. Rossaint, J.; Margraf, A.; Zarbock, A. Role of Platelets in Leukocyte Recruitment and Resolution of Inflammation. Front. Immunol. 2018, 9, 2712. [Google Scholar] [CrossRef] [Green Version]
  28. Huang, Z.Y.; Chien, P.; Indik, Z.K.; Schreiber, A.D. Human platelet FcgammaRIIA and phagocytes in immune-complex clearance. Mol. Immunol. 2011, 48, 691–696. [Google Scholar] [CrossRef] [Green Version]
  29. Yeaman, M.R. Platelets in defense against bacterial pathogens. Cell. Mol. Life Sci. 2010, 67, 525–544. [Google Scholar] [CrossRef] [Green Version]
  30. Portier, I.; Campbell, R.A. Role of Platelets in Detection and Regulation of Infection. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 70–78. [Google Scholar] [CrossRef]
  31. Burkard, P.; Vogtle, T.; Nieswandt, B. Platelets in Thrombo-Inflammation: Concepts, Mechanisms, and Therapeutic Strategies for Ischemic Stroke. Hamostaseologie 2020, 40, 153–164. [Google Scholar] [CrossRef] [PubMed]
  32. von Hundelshausen, P.; Weber, C. Platelets as immune cells: Bridging inflammation and cardiovascular disease. Circ. Res. 2007, 100, 27–40. [Google Scholar] [CrossRef] [PubMed]
  33. Assinger, A.; Schrottmaier, W.C.; Salzmann, M.; Rayes, J. Platelets in Sepsis: An Update on Experimental Models and Clinical Data. Front. Immunol. 2019, 10, 1687. [Google Scholar] [CrossRef]
  34. Olsson, A.K.; Cedervall, J. The pro-inflammatory role of platelets in cancer. Platelets 2018, 29, 569–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Kabelitz, D. Expression and function of Toll-like receptors in T lymphocytes. Curr. Opin. Immunol. 2007, 19, 39–45. [Google Scholar] [CrossRef] [PubMed]
  36. Dolganiuc, A.; Garcia, C.; Kodys, K.; Szabo, G. Distinct Toll-like receptor expression in monocytes and T cells in chronic HCV infection. World J. Gastroenterol. 2006, 12, 1198–1204. [Google Scholar] [CrossRef]
  37. Prince, L.R.; Whyte, M.K.; Sabroe, I.; Parker, L.C. The role of TLRs in neutrophil activation. Curr. Opin. Pharmacol. 2011, 11, 397–403. [Google Scholar] [CrossRef]
  38. Roh, J.S.; Sohn, D.H. Damage-Associated Molecular Patterns in Inflammatory Diseases. Immune Netw. 2018, 18, e27. [Google Scholar] [CrossRef]
  39. Tang, D.; Kang, R.; Coyne, C.B.; Zeh, H.J.; Lotze, M.T. PAMPs and DAMPs: Signal 0s that spur autophagy and immunity. Immunol. Rev. 2012, 249, 158–175. [Google Scholar] [CrossRef]
  40. Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef]
  41. Lester, S.N.; Li, K. Toll-like receptors in antiviral innate immunity. J. Mol. Biol. 2014, 426, 1246–1264. [Google Scholar] [CrossRef]
  42. Khanmohammadi, S.; Rezaei, N. Role of Toll-like receptors in the pathogenesis of COVID-19. J. Med. Virol. 2021, 93, 2735–2739. [Google Scholar] [CrossRef] [PubMed]
  43. Chaturvedi, A.; Pierce, S.K. How location governs toll-like receptor signaling. Traffic 2009, 10, 621–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Nishiya, T.; Kajita, E.; Miwa, S.; Defranco, A.L. TLR3 and TLR7 are targeted to the same intracellular compartments by distinct regulatory elements. J. Biol. Chem. 2005, 280, 37107–37117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Valencia Pacheco, G.J.; Pinzon Herrera, F.; Cruz Lopez, J.J.; Vera Gamboa Ldel, C.; Pavia Ruiz, N.; Santos Rivero, A.; Sanchez Lugo, S.; Puerto, F. Expression and activation of intracellular receptors TLR7, TLR8 and TLR9 in peripheral blood monocytes from HIV-infected patients. Colomb. Medica 2013, 44, 92–99. [Google Scholar] [CrossRef]
  46. Miyake, K. Innate immune sensing of pathogens and danger signals by cell surface Toll-like receptors. Semin. Immunol. 2007, 19, 3–10. [Google Scholar] [CrossRef]
  47. Fore, F.; Budipranama, M.; Destiawan, R.A. TLR10 and Its Role in Immunity. Handb. Exp. Pharmacol. 2022, 276, 161–174. [Google Scholar] [CrossRef]
  48. Shotorbani, S.S.; Su, Z.L.; Xu, H.X. Toll-like receptors are potential therapeutic targets in rheumatoid arthritis. World J. Biol. Chem. 2011, 2, 167–172. [Google Scholar] [CrossRef]
  49. Jin, M.S.; Lee, J.O. Structures of the toll-like receptor family and its ligand complexes. Immunity 2008, 29, 182–191. [Google Scholar] [CrossRef] [Green Version]
  50. Buwitt-Beckmann, U.; Heine, H.; Wiesmuller, K.H.; Jung, G.; Brock, R.; Akira, S.; Ulmer, A.J. Toll-like receptor 6-independent signaling by diacylated lipopeptides. Eur. J. Immunol. 2005, 35, 282–289. [Google Scholar] [CrossRef]
  51. Ve, T.; Williams, S.J.; Kobe, B. Structure and function of Toll/interleukin-1 receptor/resistance protein (TIR) domains. Apoptosis 2015, 20, 250–261. [Google Scholar] [CrossRef] [PubMed]
  52. Carty, M.; Goodbody, R.; Schroder, M.; Stack, J.; Moynagh, P.N.; Bowie, A.G. The human adaptor SARM negatively regulates adaptor protein TRIF-dependent Toll-like receptor signaling. Nat. Immunol. 2006, 7, 1074–1081. [Google Scholar] [CrossRef] [PubMed]
  53. Schilling, D.; Thomas, K.; Nixdorff, K.; Vogel, S.N.; Fenton, M.J. Toll-like receptor 4 and Toll-IL-1 receptor domain-containing adapter protein (TIRAP)/myeloid differentiation protein 88 adapter-like (Mal) contribute to maximal IL-6 expression in macrophages. J. Immunol. 2002, 169, 5874–5880. [Google Scholar] [CrossRef] [PubMed]
  54. Akira, S.; Takeda, K. Toll-like receptor signalling. Nat. Rev. Immunol. 2004, 4, 499–511. [Google Scholar] [CrossRef] [PubMed]
  55. O’Neill, L.A. DisSARMing Toll-like receptor signaling. Nat. Immunol. 2006, 7, 1023–1025. [Google Scholar] [CrossRef]
  56. Zhang, X.; Jiang, W.; Zhou, A.L.; Zhao, M.; Jiang, D.R. Inhibitory effect of oxymatrine on hepatocyte apoptosis via TLR4/PI3K/Akt/GSK-3beta signaling pathway. World J. Gastroenterol. 2017, 23, 3839–3849. [Google Scholar] [CrossRef]
  57. Zhang, X.; Jiang, D.; Jiang, W.; Zhao, M.; Gan, J. Role of TLR4-Mediated PI3K/AKT/GSK-3beta Signaling Pathway in Apoptosis of Rat Hepatocytes. BioMed Res. Int. 2015, 2015, 631326. [Google Scholar] [CrossRef] [Green Version]
  58. Ernst, O.; Vayttaden, S.J.; Fraser, I.D.C. Measurement of NF-kappaB Activation in TLR-Activated Macrophages. Methods Mol. Biol. 2018, 1714, 67–78. [Google Scholar] [CrossRef]
  59. Rubio, D.; Xu, R.H.; Remakus, S.; Krouse, T.E.; Truckenmiller, M.E.; Thapa, R.J.; Balachandran, S.; Alcami, A.; Norbury, C.C.; Sigal, L.J. Crosstalk between the type 1 interferon and nuclear factor kappa B pathways confers resistance to a lethal virus infection. Cell Host Microbe 2013, 13, 701–710. [Google Scholar] [CrossRef] [Green Version]
  60. Lima, B.H.F.; Marques, P.E.; Gomides, L.F.; Mattos, M.S.; Kraemer, L.; Queiroz-Junior, C.M.; Lennon, M.; Hirsch, E.; Russo, R.C.; Menezes, G.B.; et al. Converging TLR9 and PI3Kgamma signaling induces sterile inflammation and organ damage. Sci. Rep. 2019, 9, 19085. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. El-Zayat, S.R.; Sibaii, H.; Mannaa, F.A. Toll-like receptors activation, signaling, and targeting: An overview. Bull. Natl. Res. Cent. 2019, 43, 187. [Google Scholar] [CrossRef] [Green Version]
  63. Teixeira, H.S.; Zhao, J.; Kazmierski, E.; Kinane, D.F.; Benakanakere, M.R. TLR3-Dependent Activation of TLR2 Endogenous Ligands via the MyD88 Signaling Pathway Augments the Innate Immune Response. Cells 2020, 9, 1910. [Google Scholar] [CrossRef] [PubMed]
  64. Van Quickelberghe, E.; De Sutter, D.; van Loo, G.; Eyckerman, S.; Gevaert, K. A protein-protein interaction map of the TNF-induced NF-kappaB signal transduction pathway. Sci. Data 2018, 5, 180289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Wadowski, P.P.; Weikert, C.; Pultar, J.; Lee, S.; Eichelberger, B.; Koppensteiner, R.; Lang, I.M.; Panzer, S.; Gremmel, T. Ticagrelor Inhibits Toll-like and Protease-Activated Receptor Mediated Platelet Activation in Acute Coronary Syndromes. Cardiovasc. Drugs Ther. 2020, 34, 53–63. [Google Scholar] [CrossRef] [Green Version]
  66. Cognasse, F.; Nguyen, K.A.; Damien, P.; McNicol, A.; Pozzetto, B.; Hamzeh-Cognasse, H.; Garraud, O. The Inflammatory Role of Platelets via Their TLRs and Siglec Receptors. Front. Immunol. 2015, 6, 83. [Google Scholar] [CrossRef] [Green Version]
  67. Semeraro, F.; Ammollo, C.T.; Morrissey, J.H.; Dale, G.L.; Friese, P.; Esmon, N.L.; Esmon, C.T. Extracellular histones promote thrombin generation through platelet-dependent mechanisms: Involvement of platelet TLR2 and TLR4. Blood 2011, 118, 1952–1961. [Google Scholar] [CrossRef] [Green Version]
  68. Damien, P.; Cognasse, F.; Payrastre, B.; Spinelli, S.L.; Blumberg, N.; Arthaud, C.A.; Eyraud, M.A.; Phipps, R.P.; McNicol, A.; Pozzetto, B.; et al. NF-kappaB Links TLR2 and PAR1 to Soluble Immunomodulator Factor Secretion in Human Platelets. Front. Immunol. 2017, 8, 85. [Google Scholar] [CrossRef] [Green Version]
  69. Sriram, K.; Insel, P.A. Inflammation and thrombosis in COVID-19 pathophysiology: Proteinase-activated and purinergic receptors as drivers and candidate therapeutic targets. Physiol. Rev. 2021, 101, 545–567. [Google Scholar] [CrossRef]
  70. Sheahan, T.; Morrison, T.E.; Funkhouser, W.; Uematsu, S.; Akira, S.; Baric, R.S.; Heise, M.T. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog. 2008, 4, e1000240. [Google Scholar] [CrossRef] [Green Version]
  71. Zhou, H.; Zhao, J.; Perlman, S. Autocrine interferon priming in macrophages but not dendritic cells results in enhanced cytokine and chemokine production after coronavirus infection. mBio 2010, 1, e00219-10. [Google Scholar] [CrossRef] [Green Version]
  72. Zheng, M.; Karki, R.; Williams, E.P.; Yang, D.; Fitzpatrick, E.; Vogel, P.; Jonsson, C.B.; Kanneganti, T.D. TLR2 senses the SARS-CoV-2 envelope protein to produce inflammatory cytokines. Nat. Immunol. 2021, 22, 829–838. [Google Scholar] [CrossRef]
  73. Vercellotti, G.M. Effects of viral activation of the vessel wall on inflammation and thrombosis. Blood Coagul. Fibrinolysis 1998, 9 (Suppl. 2), S3–S6. [Google Scholar] [PubMed]
  74. Sariol, A.; Perlman, S. SARS-CoV-2 takes its Toll. Nat. Immunol. 2021, 22, 801–802. [Google Scholar] [CrossRef] [PubMed]
  75. Manik, M.; Singh, R.K. Role of toll-like receptors in modulation of cytokine storm signaling in SARS-CoV-2-induced COVID-19. J. Med. Virol. 2022, 94, 869–877. [Google Scholar] [CrossRef] [PubMed]
  76. Tosato, G.; Jones, K.D. Interleukin-1 induces interleukin-6 production in peripheral blood monocytes. Blood 1990, 75, 1305–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Rodrigues, T.S.; de Sa, K.S.G.; Ishimoto, A.Y.; Becerra, A.; Oliveira, S.; Almeida, L.; Goncalves, A.V.; Perucello, D.B.; Andrade, W.A.; Castro, R.; et al. Inflammasomes are activated in response to SARS-CoV-2 infection and are associated with COVID-19 severity in patients. J. Exp. Med. 2021, 218, e20201707. [Google Scholar] [CrossRef] [PubMed]
  78. Wadowski, P.P.; Piechota-Polanczyk, A.; Andreas, M.; Kopp, C.W. Cardiovascular Disease Management in the Context of Global Crisis. Int. J. Environ. Res. Public Health 2022, 20, 689. [Google Scholar] [CrossRef]
  79. Carnevale, R.; Cammisotto, V.; Bartimoccia, S.; Nocella, C.; Castellani, V.; Bufano, M.; Loffredo, L.; Sciarretta, S.; Frati, G.; Coluccia, A.; et al. Toll-like Receptor 4-Dependent Platelet-Related Thrombosis in SARS-CoV-2 Infection. Circ. Res. 2023, 132, 290–305. [Google Scholar] [CrossRef]
  80. Deng, J.; Ma-Krupa, W.; Gewirtz, A.T.; Younge, B.R.; Goronzy, J.J.; Weyand, C.M. Toll-like receptors 4 and 5 induce distinct types of vasculitis. Circ. Res. 2009, 104, 488–495. [Google Scholar] [CrossRef]
  81. Badr, R.E.; Salama, M.I.; Abd-Elmaogood, A.K.; Eldeib, A.E.M. Toll-like receptor 2 expression on monocytes and microvascular complications in type 2 diabetic patients. Diabetes Metab. Syndr. 2019, 13, 1299–1302. [Google Scholar] [CrossRef]
  82. Jialal, I.; Kaur, H. The Role of Toll-like Receptors in Diabetes-Induced Inflammation: Implications for Vascular Complications. Curr. Diabetes Rep. 2012, 12, 172–179. [Google Scholar] [CrossRef] [PubMed]
  83. Hally, K.E.; La Flamme, A.C.; Larsen, P.D.; Harding, S.A. Platelet Toll-like receptor (TLR) expression and TLR-mediated platelet activation in acute myocardial infarction. Thromb. Res. 2017, 158, 8–15. [Google Scholar] [CrossRef] [PubMed]
  84. Lien, E.; Zipris, D. The role of Toll-like receptor pathways in the mechanism of type 1 diabetes. Curr. Mol. Med. 2009, 9, 52–68. [Google Scholar] [CrossRef] [PubMed]
  85. Picard, F.; Adjedj, J.; Varenne, O. Diabetes Mellitus, a prothrombotic disease. Ann. Cardiol. Angeiol. 2017, 66, 385–392. [Google Scholar] [CrossRef] [PubMed]
  86. Schwameis, M.; Buchtele, N.; Wadowski, P.P.; Schoergenhofer, C.; Jilma, B. Chikungunya vaccines in development. Hum. Vaccines Immunother. 2016, 12, 716–731. [Google Scholar] [CrossRef]
  87. Hassan, T.; Abdel Rahman, D.; Raafat, N.; Fathy, M.; Shehab, M.; Hosny, A.; Fawzy, R.; Zakaria, M. Contribution of interleukin 27 serum level to pathogenesis and prognosis in children with immune thrombocytopenia. Medicine 2022, 101, e29504. [Google Scholar] [CrossRef]
  88. Lannoy, V.; Cote-Biron, A.; Asselin, C.; Rivard, N. TIRAP, TRAM, and Toll-like Receptors: The Untold Story. Mediat. Inflamm. 2023, 2023, 2899271. [Google Scholar] [CrossRef]
  89. Stack, J.; Doyle, S.L.; Connolly, D.J.; Reinert, L.S.; O’Keeffe, K.M.; McLoughlin, R.M.; Paludan, S.R.; Bowie, A.G. TRAM is required for TLR2 endosomal signaling to type I IFN induction. J. Immunol. 2014, 193, 6090–6102. [Google Scholar] [CrossRef] [Green Version]
  90. Petnicki-Ocwieja, T.; Chung, E.; Acosta, D.I.; Ramos, L.T.; Shin, O.S.; Ghosh, S.; Kobzik, L.; Li, X.; Hu, L.T. TRIF mediates Toll-like receptor 2-dependent inflammatory responses to Borrelia burgdorferi. Infect. Immun. 2013, 81, 402–410. [Google Scholar] [CrossRef] [Green Version]
  91. Thwaites, R.S.; Unterberger, S.; Chamberlain, G.; Gray, H.; Jordan, K.; Davies, K.A.; Harrison, N.A.; Sacre, S. Expression of sterile-alpha and armadillo motif containing protein (SARM) in rheumatoid arthritis monocytes correlates with TLR2-induced IL-1beta and disease activity. Rheumatology 2021, 60, 5843–5853. [Google Scholar] [CrossRef]
  92. Wadowski, P.P.; Eichelberger, B.; Kopp, C.W.; Pultar, J.; Seidinger, D.; Koppensteiner, R.; Lang, I.M.; Panzer, S.; Gremmel, T. Disaggregation Following Agonist-Induced Platelet Activation in Patients on Dual Antiplatelet Therapy. J. Cardiovasc. Transl. Res. 2017, 10, 359–367. [Google Scholar] [CrossRef] [Green Version]
  93. Panzer, B.; Wadowski, P.P.; Huber, K.; Panzer, S.; Gremmel, T. Protease-activated receptor-mediated platelet aggregation in patients with type 2 diabetes on potent P2Y(12) inhibitors. Diabet. Med. 2022, 39, e14868. [Google Scholar] [CrossRef]
  94. Wadowski, P.P.; Pultar, J.; Weikert, C.; Eichelberger, B.; Panzer, B.; Huber, K.; Lang, I.M.; Koppensteiner, R.; Panzer, S.; Gremmel, T. Protease-activated receptor-mediated platelet aggregation in acute coronary syndrome patients on potent P2Y(12) inhibitors. Res. Pract. Thromb. Haemost. 2019, 3, 383–390. [Google Scholar] [CrossRef] [Green Version]
  95. Jablonska, A.; Zagrapan, B.; Neumayer, C.; Klinger, M.; Eilenberg, W.; Nanobachvili, J.; Paradowska, E.; Brostjan, C.; Huk, I. TLR2 2029C/T and TLR3 1377C/T and -7C/A Polymorphisms Are Associated with the Occurrence of Abdominal Aortic Aneurysm. J. Immunol. 2020, 204, 2900–2909. [Google Scholar] [CrossRef] [PubMed]
  96. Haque, K.; Bhargava, P. Abdominal Aortic Aneurysm. Am. Fam. Physician 2022, 106, 165–172. [Google Scholar] [PubMed]
  97. Kessler, V.; Klopf, J.; Eilenberg, W.; Neumayer, C.; Brostjan, C. AAA Revisited: A Comprehensive Review of Risk Factors, Management, and Hallmarks of Pathogenesis. Biomedicines 2022, 10, 94. [Google Scholar] [CrossRef] [PubMed]
  98. Jablonska, A.; Neumayer, C.; Bolliger, M.; Gollackner, B.; Klinger, M.; Paradowska, E.; Nanobachvili, J.; Huk, I. Analysis of host Toll-like receptor 3 and RIG-I-like receptor gene expression in patients with abdominal aortic aneurysm. J. Vasc. Surg. 2018, 68, 39S–46S. [Google Scholar] [CrossRef]
  99. Jablonska, A.; Neumayer, C.; Bolliger, M.; Burghuber, C.; Klinger, M.; Demyanets, S.; Nanobachvili, J.; Huk, I. Insight into the expression of toll-like receptors 2 and 4 in patients with abdominal aortic aneurysm. Mol. Biol. Rep. 2020, 47, 2685–2692. [Google Scholar] [CrossRef]
  100. Treska, V.; Kocova, J.; Boudova, L.; Neprasova, P.; Topolcan, O.; Pecen, L.; Tonar, Z. Inflammation in the wall of abdominal aortic aneurysm and its role in the symptomatology of aneurysm. Cytokines Cell. Mol. Ther. 2002, 7, 91–97. [Google Scholar] [CrossRef]
  101. Klopf, J.; Brostjan, C.; Neumayer, C.; Eilenberg, W. Neutrophils as Regulators and Biomarkers of Cardiovascular Inflammation in the Context of Abdominal Aortic Aneurysms. Biomedicines 2021, 9, 1236. [Google Scholar] [CrossRef] [PubMed]
  102. Reilly, J.M.; Brophy, C.M.; Tilson, M.D. Characterization of an elastase from aneurysmal aorta which degrades intact aortic elastin. Ann. Vasc. Surg. 1992, 6, 499–502. [Google Scholar] [CrossRef] [PubMed]
  103. Newman, K.M.; Malon, A.M.; Shin, R.D.; Scholes, J.V.; Ramey, W.G.; Tilson, M.D. Matrix metalloproteinases in abdominal aortic aneurysm: Characterization, purification, and their possible sources. Connect. Tissue Res. 1994, 30, 265–276. [Google Scholar] [CrossRef]
  104. Mullick, A.E.; Tobias, P.S.; Curtiss, L.K. Modulation of atherosclerosis in mice by Toll-like receptor 2. J. Clin. Investig. 2005, 115, 3149–3156. [Google Scholar] [CrossRef] [Green Version]
  105. Aoyama, N.; Suzuki, J.; Ogawa, M.; Watanabe, R.; Kobayashi, N.; Hanatani, T.; Ashigaki, N.; Sekinishi, A.; Izumi, Y.; Isobe, M. Toll-like receptor-2 plays a fundamental role in periodontal bacteria-accelerated abdominal aortic aneurysms. Circ. J. 2013, 77, 1565–1573. [Google Scholar] [CrossRef] [Green Version]
  106. Yan, H.; Cui, B.; Zhang, X.; Fu, X.; Yan, J.; Wang, X.; Lv, X.; Chen, Z.; Hu, Z. Antagonism of toll-like receptor 2 attenuates the formation and progression of abdominal aortic aneurysm. Acta Pharm. Sin. B 2015, 5, 176–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Lai, C.H.; Wang, K.C.; Lee, F.T.; Tsai, H.W.; Ma, C.Y.; Cheng, T.L.; Chang, B.I.; Yang, Y.J.; Shi, G.Y.; Wu, H.L. Toll-like Receptor 4 Is Essential in the Development of Abdominal Aortic Aneurysm. PLoS ONE 2016, 11, e0146565. [Google Scholar] [CrossRef] [Green Version]
  108. Owens, A.P., 3rd; Rateri, D.L.; Howatt, D.A.; Moore, K.J.; Tobias, P.S.; Curtiss, L.K.; Lu, H.; Cassis, L.A.; Daugherty, A. MyD88 deficiency attenuates angiotensin II-induced abdominal aortic aneurysm formation independent of signaling through Toll-like receptors 2 and 4. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2813–2819. [Google Scholar] [CrossRef] [Green Version]
  109. Jablonska, A.; Zagrapan, B.; Paradowska, E.; Neumayer, C.; Eilenberg, W.; Brostjan, C.; Klinger, M.; Nanobachvili, J.; Huk, I. Abdominal aortic aneurysm and virus infection: A potential causative role for cytomegalovirus infection? J. Med. Virol. 2021, 93, 5017–5024. [Google Scholar] [CrossRef]
  110. Hogh, J.; Pham, M.H.C.; Knudsen, A.D.; Thudium, R.F.; Gelpi, M.; Sigvardsen, P.E.; Fuchs, A.; Kuhl, J.T.; Afzal, S.; Nordestgaard, B.G.; et al. HIV infection is associated with thoracic and abdominal aortic aneurysms: A prospective matched cohort study. Eur. Heart J. 2021, 42, 2924–2931. [Google Scholar] [CrossRef]
  111. Shehata, M.A.; Abou El-Enein, A.; El-Sharnouby, G.A. Significance of toll-like receptors 2 and 4 mRNA expression in chronic hepatitis C virus infection. Egypt. J. Immunol. 2006, 13, 141–152. [Google Scholar]
  112. Li, Q.; Wang, J.; Islam, H.; Kirschning, C.; Lu, H.; Hoffmann, D.; Dittmer, U.; Lu, M. Hepatitis B virus particles activate B cells through the TLR2-MyD88-mTOR axis. Cell Death Dis. 2021, 12, 34. [Google Scholar] [CrossRef] [PubMed]
  113. Sepehri, Z.; Kiani, Z.; Nasiri, A.A.; Kohan, F. Toll-like receptor 2 and type 2 diabetes. Cell. Mol. Biol. Lett. 2016, 21, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Polak, A.; Grywalska, E.; Klatka, J.; Rolinski, J.; Matyjaszek-Matuszek, B.; Klatka, M. Toll-like Receptors-2 and -4 in Graves’ Disease-Key Players or Bystanders? Int. J. Mol. Sci. 2019, 20, 4732. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Kariko, K.; Ni, H.; Capodici, J.; Lamphier, M.; Weissman, D. mRNA is an endogenous ligand for Toll-like receptor 3. J. Biol. Chem. 2004, 279, 12542–12550. [Google Scholar] [CrossRef] [Green Version]
  116. Cho, W.G.; Albuquerque, R.J.; Kleinman, M.E.; Tarallo, V.; Greco, A.; Nozaki, M.; Green, M.G.; Baffi, J.Z.; Ambati, B.K.; De Falco, M.; et al. Small interfering RNA-induced TLR3 activation inhibits blood and lymphatic vessel growth. Proc. Natl. Acad. Sci. USA 2009, 106, 7137–7142. [Google Scholar] [CrossRef]
  117. D’Atri, L.P.; Etulain, J.; Rivadeneyra, L.; Lapponi, M.J.; Centurion, M.; Cheng, K.; Yin, H.; Schattner, M. Expression and functionality of Toll-like receptor 3 in the megakaryocytic lineage. J. Thromb. Haemost. 2015, 13, 839–850. [Google Scholar] [CrossRef]
  118. Najem, M.; Rys, R.; Laurance, S.; Couturaud, F.; Blostein, M.D.; Lemarié, C.A. TLR3 promotes venous thrombosis through neutrophil recruitment. Rev. Mal. Respir. 2021, 38, 580. [Google Scholar] [CrossRef]
  119. Shibamiya, A.; Hersemeyer, K.; Schmidt Woll, T.; Sedding, D.; Daniel, J.M.; Bauer, S.; Koyama, T.; Preissner, K.T.; Kanse, S.M. A key role for Toll-like receptor-3 in disrupting the hemostasis balance on endothelial cells. Blood 2009, 113, 714–722. [Google Scholar] [CrossRef] [Green Version]
  120. Posma, J.J.; Grover, S.P.; Hisada, Y.; Owens, A.P., 3rd; Antoniak, S.; Spronk, H.M.; Mackman, N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 13–24. [Google Scholar] [CrossRef]
  121. Antoniak, S.; Mackman, N. Multiple roles of the coagulation protease cascade during virus infection. Blood 2014, 123, 2605–2613. [Google Scholar] [CrossRef] [Green Version]
  122. Tatsumi, K.; Schmedes, C.M.; Houston, E.R.; Butler, E.; Mackman, N.; Antoniak, S. Protease-activated receptor 4 protects mice from Coxsackievirus B3 and H1N1 influenza A virus infection. Cell. Immunol. 2019, 344, 103949. [Google Scholar] [CrossRef]
  123. Laska, M.J.; Troldborg, A.; Hansen, B.; Stengaard-Pedersen, K.; Junker, P.; Nexo, B.A.; Voss, A. Polymorphisms within Toll-like receptors are associated with systemic lupus erythematosus in a cohort of Danish females. Rheumatology 2014, 53, 48–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Al-Homood, I.A. Thrombosis in systemic lupus erythematosus: A review article. ISRN Rheumatol. 2012, 2012, 428269. [Google Scholar] [CrossRef] [Green Version]
  125. Hewson, C.A.; Jardine, A.; Edwards, M.R.; Laza-Stanca, V.; Johnston, S.L. Toll-like receptor 3 is induced by and mediates antiviral activity against rhinovirus infection of human bronchial epithelial cells. J. Virol. 2005, 79, 12273–12279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wang, T.; Town, T.; Alexopoulou, L.; Anderson, J.F.; Fikrig, E.; Flavell, R.A. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat. Med. 2004, 10, 1366–1373. [Google Scholar] [CrossRef] [PubMed]
  127. Mayhan, W.G. Cellular mechanisms by which tumor necrosis factor-alpha produces disruption of the blood-brain barrier. Brain Res. 2002, 927, 144–152. [Google Scholar] [CrossRef]
  128. Tsan, M.F.; Gao, B. Endogenous ligands of Toll-like receptors. J. Leukoc. Biol. 2004, 76, 514–519. [Google Scholar] [CrossRef]
  129. Midwood, K.; Sacre, S.; Piccinini, A.M.; Inglis, J.; Trebaul, A.; Chan, E.; Drexler, S.; Sofat, N.; Kashiwagi, M.; Orend, G.; et al. Tenascin-C is an endogenous activator of Toll-like receptor 4 that is essential for maintaining inflammation in arthritic joint disease. Nat. Med. 2009, 15, 774–780. [Google Scholar] [CrossRef]
  130. Malara, A.; Gruppi, C.; Abbonante, V.; Cattaneo, D.; De Marco, L.; Massa, M.; Iurlo, A.; Gianelli, U.; Balduini, C.L.; Tira, M.E.; et al. EDA fibronectin-TLR4 axis sustains megakaryocyte expansion and inflammation in bone marrow fibrosis. J. Exp. Med. 2019, 216, 587–604. [Google Scholar] [CrossRef]
  131. Roberts, A.L.; Mavlyutov, T.A.; Perlmutter, T.E.; Curry, S.M.; Harris, S.L.; Chauhan, A.K.; McDowell, C.M. Fibronectin extra domain A (FN-EDA) elevates intraocular pressure through Toll-like receptor 4 signaling. Sci. Rep. 2020, 10, 9815. [Google Scholar] [CrossRef]
  132. Qiang, X.; Yang, W.L.; Wu, R.; Zhou, M.; Jacob, A.; Dong, W.; Kuncewitch, M.; Ji, Y.; Yang, H.; Wang, H.; et al. Cold-inducible RNA-binding protein (CIRP) triggers inflammatory responses in hemorrhagic shock and sepsis. Nat. Med. 2013, 19, 1489–1495. [Google Scholar] [CrossRef] [Green Version]
  133. Lee, Y.; Reilly, B.; Tan, C.; Wang, P.; Aziz, M. Extracellular CIRP Induces Macrophage Extracellular Trap Formation Via Gasdermin D Activation. Front. Immunol. 2021, 12, 780210. [Google Scholar] [CrossRef]
  134. Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef] [Green Version]
  135. Horng, T.; Barton, G.M.; Medzhitov, R. TIRAP: An adapter molecule in the Toll signaling pathway. Nat. Immunol. 2001, 2, 835–841. [Google Scholar] [CrossRef] [PubMed]
  136. Lee, I.T.; Shih, R.H.; Lin, C.C.; Chen, J.T.; Yang, C.M. Role of TLR4/NADPH oxidase/ROS-activated p38 MAPK in VCAM-1 expression induced by lipopolysaccharide in human renal mesangial cells. Cell Commun. Signal 2012, 10, 33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Zhu, M.; Liu, X.; Wang, S.; Miao, J.; Wu, L.; Yang, X.; Wang, Y.; Kang, L.; Li, W.; Cui, C.; et al. PKR promotes choroidal neovascularization via upregulating the PI3K/Akt signaling pathway in VEGF expression. Mol. Vis. 2016, 22, 1361–1374. [Google Scholar]
  138. Vivarini, A.C.; Calegari-Silva, T.C.; Saliba, A.M.; Boaventura, V.S.; Franca-Costa, J.; Khouri, R.; Dierckx, T.; Dias-Teixeira, K.L.; Fasel, N.; Barral, A.M.P.; et al. Systems Approach Reveals Nuclear Factor Erythroid 2-Related Factor 2/Protein Kinase R Crosstalk in Human Cutaneous Leishmaniasis. Front. Immunol. 2017, 8, 1127. [Google Scholar] [CrossRef] [Green Version]
  139. Liddell, J.R. Are Astrocytes the Predominant Cell Type for Activation of Nrf2 in Aging and Neurodegeneration? Antioxidants 2017, 6, 65. [Google Scholar] [CrossRef] [Green Version]
  140. Solis, M.; Romieu-Mourez, R.; Goubau, D.; Grandvaux, N.; Mesplede, T.; Julkunen, I.; Nardin, A.; Salcedo, M.; Hiscott, J. Involvement of TBK1 and IKKepsilon in lipopolysaccharide-induced activation of the interferon response in primary human macrophages. Eur. J. Immunol. 2007, 37, 528–539. [Google Scholar] [CrossRef] [PubMed]
  141. Carlsson, E.; Ding, J.L.; Byrne, B. SARM modulates MyD88-mediated TLR activation through BB-loop dependent TIR-TIR interactions. Biochim. Biophys. Acta 2016, 1863, 244–253. [Google Scholar] [CrossRef]
  142. Yang, L.; Seki, E. Toll-like receptors in liver fibrosis: Cellular crosstalk and mechanisms. Front. Physiol. 2012, 3, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Schattner, M. Platelet TLR4 at the crossroads of thrombosis and the innate immune response. J. Leukoc. Biol. 2019, 105, 873–880. [Google Scholar] [CrossRef]
  144. Krikun, G.; Trezza, J.; Shaw, J.; Rahman, M.; Guller, S.; Abrahams, V.M.; Lockwood, C.J. Lipopolysaccharide appears to activate human endometrial endothelial cells through TLR-4-dependent and TLR-4-independent mechanisms. Am. J. Reprod. Immunol. 2012, 68, 233–237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Jerez-Dolz, D.; Torramade-Moix, S.; Palomo, M.; Moreno-Castano, A.; Lopez-Vilchez, I.; Hernandez, R.; Badimon, J.J.; Zafar, M.U.; Diaz-Ricart, M.; Escolar, G. Internalization of microparticles by platelets is partially mediated by toll-like receptor 4 and enhances platelet thrombogenicity. Atherosclerosis 2020, 294, 17–24. [Google Scholar] [CrossRef]
  146. Yang, X.; Li, L.; Liu, J.; Lv, B.; Chen, F. Extracellular histones induce tissue factor expression in vascular endothelial cells via TLR and activation of NF-kappaB and AP-1. Thromb. Res. 2016, 137, 211–218. [Google Scholar] [CrossRef]
  147. Owens, A.P., 3rd; Passam, F.H.; Antoniak, S.; Marshall, S.M.; McDaniel, A.L.; Rudel, L.; Williams, J.C.; Hubbard, B.K.; Dutton, J.A.; Wang, J.; et al. Monocyte tissue factor-dependent activation of coagulation in hypercholesterolemic mice and monkeys is inhibited by simvastatin. J. Clin. Investig. 2012, 122, 558–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Clark, S.R.; Ma, A.C.; Tavener, S.A.; McDonald, B.; Goodarzi, Z.; Kelly, M.M.; Patel, K.D.; Chakrabarti, S.; McAvoy, E.; Sinclair, G.D.; et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat. Med. 2007, 13, 463–469. [Google Scholar] [CrossRef]
  149. Hally, K.E.; Bird, G.K.; La Flamme, A.C.; Harding, S.A.; Larsen, P.D. Platelets modulate multiple markers of neutrophil function in response to in vitro Toll-like receptor stimulation. PLoS ONE 2019, 14, e0223444. [Google Scholar] [CrossRef] [Green Version]
  150. D’Mello, C.; Almishri, W.; Liu, H.; Swain, M.G. Interactions between Platelets and Inflammatory Monocytes Affect Sickness Behavior in Mice with Liver Inflammation. Gastroenterology 2017, 153, 1416–1428.e2. [Google Scholar] [CrossRef]
  151. Napier, B.A.; Brubaker, S.W.; Sweeney, T.E.; Monette, P.; Rothmeier, G.H.; Gertsvolf, N.A.; Puschnik, A.; Carette, J.E.; Khatri, P.; Monack, D.M. Complement pathway amplifies caspase-11-dependent cell death and endotoxin-induced sepsis severity. J. Exp. Med. 2016, 213, 2365–2382. [Google Scholar] [CrossRef] [PubMed]
  152. Jiang, M.; Sun, X.; Liu, S.; Tang, Y.; Shi, Y.; Bai, Y.; Wang, Y.; Yang, Q.; Yang, Q.; Jiang, W.; et al. Caspase-11-Gasdermin D-Mediated Pyroptosis Is Involved in the Pathogenesis of Atherosclerosis. Front. Pharmacol. 2021, 12, 657486. [Google Scholar] [CrossRef]
  153. Wang, Y.; Zhu, X.; Yuan, S.; Wen, S.; Liu, X.; Wang, C.; Qu, Z.; Li, J.; Liu, H.; Sun, L.; et al. TLR4/NF-kappaB Signaling Induces GSDMD-Related Pyroptosis in Tubular Cells in Diabetic Kidney Disease. Front. Endocrinol. 2019, 10, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Han, T.; Tang, H.; Lin, C.; Shen, Y.; Yan, D.; Tang, X.; Guo, D. Extracellular traps and the role in thrombosis. Front. Cardiovasc. Med. 2022, 9, 951670. [Google Scholar] [CrossRef] [PubMed]
  155. Das, L.; Azmoon, P.; Banki, M.A.; Mantuano, E.; Gonias, S.L. Tissue-type plasminogen activator selectively inhibits multiple toll-like receptors in CSF-1-differentiated macrophages. PLoS ONE 2019, 14, e0224738. [Google Scholar] [CrossRef] [PubMed]
  156. Kircheis, R.; Planz, O. The Role of Toll-like Receptors (TLRs) and Their Related Signaling Pathways in Viral Infection and Inflammation. Int. J. Mol. Sci. 2023, 24, 6701. [Google Scholar] [CrossRef] [PubMed]
  157. Riad, A.; Jager, S.; Sobirey, M.; Escher, F.; Yaulema-Riss, A.; Westermann, D.; Karatas, A.; Heimesaat, M.M.; Bereswill, S.; Dragun, D.; et al. Toll-like receptor-4 modulates survival by induction of left ventricular remodeling after myocardial infarction in mice. J. Immunol. 2008, 180, 6954–6961. [Google Scholar] [CrossRef] [Green Version]
  158. Satoh, M.; Shimoda, Y.; Maesawa, C.; Akatsu, T.; Ishikawa, Y.; Minami, Y.; Hiramori, K.; Nakamura, M. Activated toll-like receptor 4 in monocytes is associated with heart failure after acute myocardial infarction. Int. J. Cardiol. 2006, 109, 226–234. [Google Scholar] [CrossRef]
  159. Modhiran, N.; Watterson, D.; Blumenthal, A.; Baxter, A.G.; Young, P.R.; Stacey, K.J. Dengue virus NS1 protein activates immune cells via TLR4 but not TLR2 or TLR6. Immunol. Cell Biol. 2017, 95, 491–495. [Google Scholar] [CrossRef] [Green Version]
  160. Weinbaum, S.; Cancel, L.M.; Fu, B.M.; Tarbell, J.M. The Glycocalyx and Its Role in Vascular Physiology and Vascular Related Diseases. Cardiovasc. Eng. Technol. 2021, 12, 37–71. [Google Scholar] [CrossRef]
  161. Tang, T.H.; Alonso, S.; Ng, L.F.; Thein, T.L.; Pang, V.J.; Leo, Y.S.; Lye, D.C.; Yeo, T.W. Increased Serum Hyaluronic Acid and Heparan Sulfate in Dengue Fever: Association with Plasma Leakage and Disease Severity. Sci. Rep. 2017, 7, 46191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Suwarto, S.; Sasmono, R.T.; Sinto, R.; Ibrahim, E.; Suryamin, M. Association of Endothelial Glycocalyx and Tight and Adherens Junctions with Severity of Plasma Leakage in Dengue Infection. J. Infect. Dis. 2017, 215, 992–999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. 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] [PubMed] [Green Version]
  164. He, B. Viruses, endoplasmic reticulum stress, and interferon responses. Cell Death Differ. 2006, 13, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Shenderov, K.; Riteau, N.; Yip, R.; Mayer-Barber, K.D.; Oland, S.; Hieny, S.; Fitzgerald, P.; Oberst, A.; Dillon, C.P.; Green, D.R.; et al. Cutting edge: Endoplasmic reticulum stress licenses macrophages to produce mature IL-1beta in response to TLR4 stimulation through a caspase-8- and TRIF-dependent pathway. J. Immunol. 2014, 192, 2029–2033. [Google Scholar] [CrossRef] [Green Version]
  166. Funchal, G.A.; Jaeger, N.; Czepielewski, R.S.; Machado, M.S.; Muraro, S.P.; Stein, R.T.; Bonorino, C.B.; Porto, B.N. Respiratory syncytial virus fusion protein promotes TLR-4-dependent neutrophil extracellular trap formation by human neutrophils. PLoS ONE 2015, 10, e0124082. [Google Scholar] [CrossRef] [Green Version]
  167. Cook, C.H.; Trgovcich, J.; Zimmerman, P.D.; Zhang, Y.; Sedmak, D.D. Lipopolysaccharide, tumor necrosis factor alpha, or interleukin-1beta triggers reactivation of latent cytomegalovirus in immunocompetent mice. J. Virol. 2006, 80, 9151–9158. [Google Scholar] [CrossRef] [Green Version]
  168. Holms, R.D. Long COVID (PASC) Is Maintained by a Self-Sustaining Pro-Inflammatory TLR4/RAGE-Loop of S100A8/A9 > TLR4/RAGE Signalling, Inducing Chronic Expression of IL-1b, IL-6 and TNFa: Anti-Inflammatory Ezrin Peptides as Potential Therapy. Immuno 2022, 2, 512–533. [Google Scholar] [CrossRef]
  169. Fontes-Dantas, F.L.; Fernandes, G.G.; Gutman, E.G.; De Lima, E.V.; Antonio, L.S.; Hammerle, M.B.; Mota-Araujo, H.P.; Colodeti, L.C.; Araujo, S.M.B.; Froz, G.M.; et al. SARS-CoV-2 Spike protein induces TLR4-mediated long-term cognitive dysfunction recapitulating post-COVID-19 syndrome in mice. Cell Rep. 2023, 42, 112189. [Google Scholar] [CrossRef]
  170. Yoon, S.I.; Kurnasov, O.; Natarajan, V.; Hong, M.; Gudkov, A.V.; Osterman, A.L.; Wilson, I.A. Structural basis of TLR5-flagellin recognition and signaling. Science 2012, 335, 859–864. [Google Scholar] [CrossRef] [Green Version]
  171. Xiao, W.; Liu, Z.; Lin, J.; Xiong, C.; Li, J.; Wu, K.; Ma, Y.; Gong, Y.; Liu, Z. Association of TLR4 and TLR5 gene polymorphisms with Graves’ disease in Chinese Cantonese population. Hum. Immunol. 2014, 75, 609–613. [Google Scholar] [CrossRef] [PubMed]
  172. Georgel, A.F.; Cayet, D.; Pizzorno, A.; Rosa-Calatrava, M.; Paget, C.; Sencio, V.; Dubuisson, J.; Trottein, F.; Sirard, J.C.; Carnoy, C. Toll-like receptor 5 agonist flagellin reduces influenza A virus replication independently of type I interferon and interleukin 22 and improves antiviral efficacy of oseltamivir. Antivir. Res. 2019, 168, 28–35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Isogawa, M.; Robek, M.D.; Furuichi, Y.; Chisari, F.V. Toll-like receptor signaling inhibits hepatitis B virus replication in vivo. J. Virol. 2005, 79, 7269–7272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Kimura, K.; Kakimi, K.; Wieland, S.; Guidotti, L.G.; Chisari, F.V. Activated intrahepatic antigen-presenting cells inhibit hepatitis B virus replication in the liver of transgenic mice. J. Immunol. 2002, 169, 5188–5195. [Google Scholar] [CrossRef] [Green Version]
  175. Ruffin, M.; Bigot, J.; Calmel, C.; Mercier, J.; Givelet, M.; Oliva, J.; Pizzorno, A.; Rosa-Calatrava, M.; Corvol, H.; Balloy, V.; et al. Flagellin From Pseudomonas aeruginosa Modulates SARS-CoV-2 Infectivity in Cystic Fibrosis Airway Epithelial Cells by Increasing TMPRSS2 Expression. Front. Immunol. 2021, 12, 714027. [Google Scholar] [CrossRef]
  176. Owens, A.P., 3rd; Mackman, N. Sources of tissue factor that contribute to thrombosis after rupture of an atherosclerotic plaque. Thromb. Res. 2012, 129 (Suppl. 2), S30–S33. [Google Scholar] [CrossRef] [Green Version]
  177. Tuvim, M.J.; Gilbert, B.E.; Dickey, B.F.; Evans, S.E. Synergistic TLR2/6 and TLR9 activation protects mice against lethal influenza pneumonia. PLoS ONE 2012, 7, e30596. [Google Scholar] [CrossRef]
  178. Shevlin, E.; Miggin, S.M. The TIR-domain containing adaptor TRAM is required for TLR7 mediated RANTES production. PLoS ONE 2014, 9, e107141. [Google Scholar] [CrossRef] [Green Version]
  179. Piao, W.; Shirey, K.A.; Ru, L.W.; Lai, W.; Szmacinski, H.; Snyder, G.A.; Sundberg, E.J.; Lakowicz, J.R.; Vogel, S.N.; Toshchakov, V.Y. A Decoy Peptide that Disrupts TIRAP Recruitment to TLRs Is Protective in a Murine Model of Influenza. Cell Rep. 2015, 11, 1941–1952. [Google Scholar] [CrossRef] [Green Version]
  180. Mukherjee, P.; Winkler, C.W.; Taylor, K.G.; Woods, T.A.; Nair, V.; Khan, B.A.; Peterson, K.E. SARM1, Not MyD88, Mediates TLR7/TLR9-Induced Apoptosis in Neurons. J. Immunol. 2015, 195, 4913–4921. [Google Scholar] [CrossRef] [Green Version]
  181. Nilsen, K.E.; Skjesol, A.; Frengen Kojen, J.; Espevik, T.; Stenvik, J.; Yurchenko, M. TIRAP/Mal Positively Regulates TLR8-Mediated Signaling via IRF5 in Human Cells. Biomedicines 2022, 10, 1476. [Google Scholar] [CrossRef]
  182. Gamrekelashvili, J.; Kapanadze, T.; Sablotny, S.; Ratiu, C.; Dastagir, K.; Lochner, M.; Karbach, S.; Wenzel, P.; Sitnow, A.; Fleig, S.; et al. Notch and TLR signaling coordinate monocyte cell fate and inflammation. Elife 2020, 9, e57007. [Google Scholar] [CrossRef]
  183. Cohen, P. The TLR and IL-1 signalling network at a glance. J. Cell Sci. 2014, 127, 2383–2390. [Google Scholar] [CrossRef] [Green Version]
  184. Diebold, S.S. Recognition of viral single-stranded RNA by Toll-like receptors. Adv. Drug Deliv. Rev. 2008, 60, 813–823. [Google Scholar] [CrossRef]
  185. Triantafilou, K.; Orthopoulos, G.; Vakakis, E.; Ahmed, M.A.; Golenbock, D.T.; Lepper, P.M.; Triantafilou, M. Human cardiac inflammatory responses triggered by Coxsackie B viruses are mainly Toll-like receptor (TLR) 8-dependent. Cell. Microbiol. 2005, 7, 1117–1126. [Google Scholar] [CrossRef] [PubMed]
  186. Senchenkova, E.Y.; Komoto, S.; Russell, J.; Almeida-Paula, L.D.; Yan, L.S.; Zhang, S.; Granger, D.N. Interleukin-6 mediates the platelet abnormalities and thrombogenesis associated with experimental colitis. Am. J. Pathol. 2013, 183, 173–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Ferroni, P.; Roselli, M.; Diodati, A.; Casciani, C.U.; Gazzaniga, P.P. Effects on platelet function by human interferon-beta in carcinoma patients. Anticancer Res. 1994, 14, 2779–2784. [Google Scholar] [PubMed]
  188. Davizon-Castillo, P.; McMahon, B.; Aguila, S.; Bark, D.; Ashworth, K.; Allawzi, A.; Campbell, R.A.; Montenont, E.; Nemkov, T.; D’Alessandro, A.; et al. TNF-alpha-driven inflammation and mitochondrial dysfunction define the platelet hyperreactivity of aging. Blood 2019, 134, 727–740. [Google Scholar] [CrossRef]
  189. 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] [Green Version]
  190. Channappanavar, R.; Fehr, A.R.; Zheng, J.; Wohlford-Lenane, C.; Abrahante, J.E.; Mack, M.; Sompallae, R.; McCray, P.B., Jr.; Meyerholz, D.K.; Perlman, S. IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J. Clin. Investig. 2019, 129, 3625–3639. [Google Scholar] [CrossRef]
  191. van der Sluis, R.M.; Cham, L.B.; Gris-Oliver, A.; Gammelgaard, K.R.; Pedersen, J.G.; Idorn, M.; Ahmadov, U.; Hernandez, S.S.; Cemalovic, E.; Godsk, S.H.; et al. TLR2 and TLR7 mediate distinct immunopathological and antiviral plasmacytoid dendritic cell responses to SARS-CoV-2 infection. EMBO J. 2022, 41, e109622. [Google Scholar] [CrossRef] [PubMed]
  192. Dutta, S.K.; Tripathi, A. Association of toll-like receptor polymorphisms with susceptibility to chikungunya virus infection. Virology 2017, 511, 207–213. [Google Scholar] [CrossRef] [PubMed]
  193. Kayesh, M.E.H.; Kohara, M.; Tsukiyama-Kohara, K. Toll-like Receptor Response to Hepatitis C Virus Infection: A Recent Overview. Int. J. Mol. Sci. 2022, 23, 5475. [Google Scholar] [CrossRef]
  194. Gorbea, C.; Makar, K.A.; Pauschinger, M.; Pratt, G.; Bersola, J.L.; Varela, J.; David, R.M.; Banks, L.; Huang, C.H.; Li, H.; et al. A role for Toll-like receptor 3 variants in host susceptibility to enteroviral myocarditis and dilated cardiomyopathy. J. Biol. Chem. 2010, 285, 23208–23223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Saitoh, T.; Komano, J.; Saitoh, Y.; Misawa, T.; Takahama, M.; Kozaki, T.; Uehata, T.; Iwasaki, H.; Omori, H.; Yamaoka, S.; et al. Neutrophil extracellular traps mediate a host defense response to human immunodeficiency virus-1. Cell Host Microbe 2012, 12, 109–116. [Google Scholar] [CrossRef] [Green Version]
  196. Kawasaki, A.; Furukawa, H.; Kondo, Y.; Ito, S.; Hayashi, T.; Kusaoi, M.; Matsumoto, I.; Tohma, S.; Takasaki, Y.; Hashimoto, H.; et al. TLR7 single-nucleotide polymorphisms in the 3’ untranslated region and intron 2 independently contribute to systemic lupus erythematosus in Japanese women: A case-control association study. Arthritis Res. Ther. 2011, 13, R41. [Google Scholar] [CrossRef] [Green Version]
  197. Santos-Sierra, S. Targeting Toll-like Receptor (TLR) Pathways in Inflammatory Arthritis: Two Better Than One? Biomolecules 2021, 11, 1291. [Google Scholar] [CrossRef]
  198. Javmen, A.; Szmacinski, H.; Lakowicz, J.R.; Toshchakov, V.Y. Blocking TIR Domain Interactions in TLR9 Signaling. J. Immunol. 2018, 201, 995–1006. [Google Scholar] [CrossRef] [Green Version]
  199. Volpi, C.; Fallarino, F.; Pallotta, M.T.; Bianchi, R.; Vacca, C.; Belladonna, M.L.; Orabona, C.; De Luca, A.; Boon, L.; Romani, L.; et al. High doses of CpG oligodeoxynucleotides stimulate a tolerogenic TLR9-TRIF pathway. Nat. Commun. 2013, 4, 1852. [Google Scholar] [CrossRef] [Green Version]
  200. Chen, H.C.; Zhan, X.; Tran, K.K.; Shen, H. Selectively targeting the toll-like receptor 9 (TLR9)--IRF 7 signaling pathway by polymer blend particles. Biomaterials 2013, 34, 6464–6472. [Google Scholar] [CrossRef] [Green Version]
  201. Combes, A.; Camosseto, V.; N’Guessan, P.; Arguello, R.J.; Mussard, J.; Caux, C.; Bendriss-Vermare, N.; Pierre, P.; Gatti, E. BAD-LAMP controls TLR9 trafficking and signalling in human plasmacytoid dendritic cells. Nat. Commun. 2017, 8, 913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Shepard, C.R. TLR9 in MAFLD and NASH: At the Intersection of Inflammation and Metabolism. Front. Endocrinol. 2021, 11, 613639. [Google Scholar] [CrossRef] [PubMed]
  203. Fischer, A.; Abdollahi-Roodsaz, S.; Bohm, C.; Niederreiter, B.; Meyer, B.; Yau, A.C.Y.; Lonnblom, E.; Joosten, L.A.B.; Koenders, M.; Lehmann, C.H.K.; et al. The involvement of Toll-like receptor 9 in the pathogenesis of erosive autoimmune arthritis. J. Cell. Mol. Med. 2018, 22, 4399–4409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Zhou, Y.; Fang, L.; Peng, L.; Qiu, W. TLR9 and its signaling pathway in multiple sclerosis. J. Neurol. Sci. 2017, 373, 95–99. [Google Scholar] [CrossRef]
  205. Varani, S.; Cederarv, M.; Feld, S.; Tammik, C.; Frascaroli, G.; Landini, M.P.; Soderberg-Naucler, C. Human cytomegalovirus differentially controls B cell and T cell responses through effects on plasmacytoid dendritic cells. J. Immunol. 2007, 179, 7767–7776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Zyzak, J.; Mitkiewicz, M.; Leszczynska, E.; Reniewicz, P.; Moynagh, P.N.; Siednienko, J. HSV-1/TLR9-Mediated IFNbeta and TNFalpha Induction Is Mal-Dependent in Macrophages. J. Innate Immun. 2020, 12, 387–398. [Google Scholar] [CrossRef]
  207. Krug, A.; Luker, G.D.; Barchet, W.; Leib, D.A.; Akira, S.; Colonna, M. Herpes simplex virus type 1 activates murine natural interferon-producing cells through toll-like receptor 9. Blood 2004, 103, 1433–1437. [Google Scholar] [CrossRef] [Green Version]
  208. Lund, J.; Sato, A.; Akira, S.; Medzhitov, R.; Iwasaki, A. Toll-like receptor 9-mediated recognition of Herpes simplex virus-2 by plasmacytoid dendritic cells. J. Exp. Med. 2003, 198, 513–520. [Google Scholar] [CrossRef]
  209. Fiola, S.; Gosselin, D.; Takada, K.; Gosselin, J. TLR9 contributes to the recognition of EBV by primary monocytes and plasmacytoid dendritic cells. J. Immunol. 2010, 185, 3620–3631. [Google Scholar] [CrossRef] [Green Version]
  210. Jordi, M.; Marty, J.; Mordasini, V.; Lunemann, A.; McComb, S.; Bernasconi, M.; Nadal, D. IRAK4 is essential for TLR9-induced suppression of Epstein-Barr virus BZLF1 transcription in Akata Burkitt’s lymphoma cells. PLoS ONE 2017, 12, e0186614. [Google Scholar] [CrossRef] [Green Version]
  211. Qian, J.; Meng, H.; Lv, B.; Wang, J.; Lu, Y.; Li, W.; Zhao, S. TLR9 expression is associated with PD-L1 expression and indicates a poor prognosis in patients with peripheral T-cell lymphomas. Pathol. Res. Pract. 2020, 216, 152703. [Google Scholar] [CrossRef]
  212. Fernandez-Rodriguez, L.; Cianciaruso, C.; Bill, R.; Trefny, M.P.; Klar, R.; Kirchhammer, N.; Buchi, M.; Festag, J.; Michel, S.; Kohler, R.H.; et al. Dual TLR9 and PD-L1 targeting unleashes dendritic cells to induce durable antitumor immunity. J. Immunother. Cancer 2023, 11, e006714. [Google Scholar] [CrossRef]
  213. Henke, P.K.; Mitsuya, M.; Luke, C.E.; Elfline, M.A.; Baldwin, J.F.; Deatrick, K.B.; Diaz, J.A.; Sood, V.; Upchurch, G.R.; Wakefield, T.W.; et al. Toll-like receptor 9 signaling is critical for early experimental deep vein thrombosis resolution. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 43–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Su, S.B.; Tao, L.; Deng, Z.P.; Chen, W.; Qin, S.Y.; Jiang, H.X. TLR10: Insights, controversies and potential utility as a therapeutic target. Scand. J. Immunol. 2021, 93, e12988. [Google Scholar] [CrossRef] [PubMed]
  215. Oosting, M.; Cheng, S.C.; Bolscher, J.M.; Vestering-Stenger, R.; Plantinga, T.S.; Verschueren, I.C.; Arts, P.; Garritsen, A.; van Eenennaam, H.; Sturm, P.; et al. Human TLR10 is an anti-inflammatory pattern-recognition receptor. Proc. Natl. Acad. Sci. USA 2014, 111, E4478–E4484. [Google Scholar] [CrossRef] [PubMed]
  216. Fore, F.; Indriputri, C.; Mamutse, J.; Nugraha, J. TLR10 and Its Unique Anti-Inflammatory Properties and Potential Use as a Target in Therapeutics. Immune Netw. 2020, 20, e21. [Google Scholar] [CrossRef] [PubMed]
  217. Henrick, B.M.; Yao, X.D.; Zahoor, M.A.; Abimiku, A.; Osawe, S.; Rosenthal, K.L. TLR10 Senses HIV-1 Proteins and Significantly Enhances HIV-1 Infection. Front. Immunol. 2019, 10, 482. [Google Scholar] [CrossRef]
  218. Lee, S.M.; Kok, K.H.; Jaume, M.; Cheung, T.K.; Yip, T.F.; Lai, J.C.; Guan, Y.; Webster, R.G.; Jin, D.Y.; Peiris, J.S. Toll-like receptor 10 is involved in induction of innate immune responses to influenza virus infection. Proc. Natl. Acad. Sci. USA 2014, 111, 3793–3798. [Google Scholar] [CrossRef] [PubMed]
  219. Wadowski, P.P.; Jilma, B.; Kopp, C.W.; Ertl, S.; Gremmel, T.; Koppensteiner, R. Glycocalyx as Possible Limiting Factor in COVID-19. Front. Immunol. 2021, 12, 607306. [Google Scholar] [CrossRef]
  220. Hong, W.; Yang, J.; Zou, J.; Bi, Z.; He, C.; Lei, H.; He, X.; Li, X.; Alu, A.; Ren, W.; et al. Histones released by NETosis enhance the infectivity of SARS-CoV-2 by bridging the spike protein subunit 2 and sialic acid on host cells. Cell. Mol. Immunol. 2022, 19, 577–587. [Google Scholar] [CrossRef]
  221. Ligi, D.; Lo Sasso, B.; Giglio, R.V.; Maniscalco, R.; DellaFranca, C.; Agnello, L.; Ciaccio, M.; Mannello, F. Circulating histones contribute to monocyte and MDW alterations as common mediators in classical and COVID-19 sepsis. Crit. Care 2022, 26, 260. [Google Scholar] [CrossRef] [PubMed]
  222. Singh, B.; Biswas, I.; Bhagat, S.; Surya Kumari, S.; Khan, G.A. HMGB1 facilitates hypoxia-induced vWF upregulation through TLR2-MYD88-SP1 pathway. Eur. J. Immunol. 2016, 46, 2388–2400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Carestia, A.; Kaufman, T.; Rivadeneyra, L.; Landoni, V.I.; Pozner, R.G.; Negrotto, S.; D’Atri, L.P.; Gomez, R.M.; Schattner, M. Mediators and molecular pathways involved in the regulation of neutrophil extracellular trap formation mediated by activated platelets. J. Leukoc. Biol. 2016, 99, 153–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  224. Into, T.; Kanno, Y.; Dohkan, J.; Nakashima, M.; Inomata, M.; Shibata, K.; Lowenstein, C.J.; Matsushita, K. Pathogen recognition by Toll-like receptor 2 activates Weibel-Palade body exocytosis in human aortic endothelial cells. J. Biol. Chem. 2007, 282, 8134–8141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Tsujii, N.; Nogami, K.; Yoshizawa, H.; Hayakawa, M.; Isonishi, A.; Matsumoto, M.; Shima, M. Influenza-associated thrombotic microangiopathy with unbalanced von Willebrand factor and a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 levels in a heterozygous protein S-deficient boy. Pediatr. Int. 2016, 58, 926–929. [Google Scholar] [CrossRef] [PubMed]
  226. Djamiatun, K.; van der Ven, A.J.; de Groot, P.G.; Faradz, S.M.; Hapsari, D.; Dolmans, W.M.; Sebastian, S.; Fijnheer, R.; de Mast, Q. Severe dengue is associated with consumption of von Willebrand factor and its cleaving enzyme ADAMTS-13. PLoS Negl. Trop. Dis. 2012, 6, e1628. [Google Scholar] [CrossRef] [Green Version]
  227. Bester, J.; Swanepoel, A.C.; Windberger, U. Editorial: Pathological Changes in Erythrocytes During Inflammation and Infection. Front. Physiol. 2022, 13, 943114. [Google Scholar] [CrossRef]
  228. Maruyama, T.; Hieda, M.; Mawatari, S.; Fujino, T. Rheological Abnormalities in Human Erythrocytes Subjected to Oxidative Inflammation. Front. Physiol. 2022, 13, 837926. [Google Scholar] [CrossRef]
  229. Wadowski, P.P.; Schorgenhofer, C.; Rieder, T.; Ertl, S.; Pultar, J.; Serles, W.; Sycha, T.; Mayer, F.; Koppensteiner, R.; Gremmel, T.; et al. Microvascular rarefaction in patients with cerebrovascular events. Microvasc. Res. 2022, 140, 104300. [Google Scholar] [CrossRef]
  230. Wadowski, P.P.; Kautzky-Willer, A.; Gremmel, T.; Koppensteiner, R.; Wolf, P.; Ertl, S.; Weikert, C.; Schorgenhofer, C.; Jilma, B. Sublingual microvasculature in diabetic patients. Microvasc. Res. 2020, 129, 103971. [Google Scholar] [CrossRef] [PubMed]
  231. Wadowski, P.P.; Steinlechner, B.; Zimpfer, D.; Schloglhofer, T.; Schima, H.; Hulsmann, M.; Lang, I.M.; Gremmel, T.; Koppensteiner, R.; Zehetmayer, S.; et al. Functional capillary impairment in patients with ventricular assist devices. Sci. Rep. 2019, 9, 5909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  232. Wadowski, P.P.; Hulsmann, M.; Schorgenhofer, C.; Lang, I.M.; Wurm, R.; Gremmel, T.; Koppensteiner, R.; Steinlechner, B.; Schwameis, M.; Jilma, B. Sublingual functional capillary rarefaction in chronic heart failure. Eur. J. Clin. Investig. 2018, 48, e12869. [Google Scholar] [CrossRef] [PubMed]
  233. Pouvreau, C.; Dayre, A.; Butkowski, E.G.; de Jong, B.; Jelinek, H.F. Inflammation and oxidative stress markers in diabetes and hypertension. J. Inflamm. Res. 2018, 11, 61–68. [Google Scholar] [CrossRef] [Green Version]
  234. Carvalho, C.; Moreira, P.I. Oxidative Stress: A Major Player in Cerebrovascular Alterations Associated to Neurodegenerative Events. Front. Physiol. 2018, 9, 806. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  235. Yang, X.; Li, Y.; Li, Y.; Ren, X.; Zhang, X.; Hu, D.; Gao, Y.; Xing, Y.; Shang, H. Oxidative Stress-Mediated Atherosclerosis: Mechanisms and Therapies. Front. Physiol. 2017, 8, 600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. Alonso-Pineiro, J.A.; Gonzalez-Rovira, A.; Sanchez-Gomar, I.; Moreno, J.A.; Duran-Ruiz, M.C. Nrf2 and Heme Oxygenase-1 Involvement in Atherosclerosis Related Oxidative Stress. Antioxidants 2021, 10, 1463. [Google Scholar] [CrossRef] [PubMed]
  237. Andreas, M.; Schmid, A.I.; Doberer, D.; Schewzow, K.; Weisshaar, S.; Heinze, G.; Bilban, M.; Moser, E.; Wolzt, M. Heme arginate improves reperfusion patterns after ischemia: A randomized, placebo-controlled trial in healthy male subjects. J. Cardiovasc. Magn. Reson. 2012, 14, 55. [Google Scholar] [CrossRef] [Green Version]
  238. Vartanian, K.B.; Stevens, S.L.; Marsh, B.J.; Williams-Karnesky, R.; Lessov, N.S.; Stenzel-Poore, M.P. LPS preconditioning redirects TLR signaling following stroke: TRIF-IRF3 plays a seminal role in mediating tolerance to ischemic injury. J. Neuroinflammation 2011, 8, 140. [Google Scholar] [CrossRef] [Green Version]
  239. Loboda, A.; Damulewicz, M.; Pyza, E.; Jozkowicz, A.; Dulak, J. Role of Nrf2/HO-1 system in development, oxidative stress response and diseases: An evolutionarily conserved mechanism. Cell. Mol. Life Sci. 2016, 73, 3221–3247. [Google Scholar] [CrossRef] [Green Version]
  240. Li, B.; Xia, Y.; Hu, B. Infection and atherosclerosis: TLR-dependent pathways. Cell. Mol. Life Sci. 2020, 77, 2751–2769. [Google Scholar] [CrossRef] [Green Version]
  241. Tang, A.T.; Choi, J.P.; Kotzin, J.J.; Yang, Y.; Hong, C.C.; Hobson, N.; Girard, R.; Zeineddine, H.A.; Lightle, R.; Moore, T.; et al. Endothelial TLR4 and the microbiome drive cerebral cavernous malformations. Nature 2017, 545, 305–310. [Google Scholar] [CrossRef] [Green Version]
  242. Means, R.T., Jr. The anaemia of infection. Baillieres Best Pract. Res. Clin. Haematol. 2000, 13, 151–162. [Google Scholar] [CrossRef] [PubMed]
  243. Akilesh, H.M.; Buechler, M.B.; Duggan, J.M.; Hahn, W.O.; Matta, B.; Sun, X.; Gessay, G.; Whalen, E.; Mason, M.; Presnell, S.R.; et al. Chronic TLR7 and TLR9 signaling drives anemia via differentiation of specialized hemophagocytes. Science 2019, 363, eaao5213. [Google Scholar] [CrossRef]
  244. Bouchla, A.; Kriebardis, A.G.; Georgatzakou, H.T.; Fortis, S.P.; Thomopoulos, T.P.; Lekkakou, L.; Markakis, K.; Gkotzias, D.; Panagiotou, A.; Papageorgiou, E.G.; et al. Red Blood Cell Abnormalities as the Mirror of SARS-CoV-2 Disease Severity: A Pilot Study. Front. Physiol. 2021, 12, 825055. [Google Scholar] [CrossRef] [PubMed]
  245. Bellmann-Weiler, R.; Lanser, L.; Barket, R.; Rangger, L.; Schapfl, A.; Schaber, M.; Fritsche, G.; Woll, E.; Weiss, G. Prevalence and Predictive Value of Anemia and Dysregulated Iron Homeostasis in Patients with COVID-19 Infection. J. Clin. Med. 2020, 9, 2429. [Google Scholar] [CrossRef] [PubMed]
  246. Wadowski, P.P.; Kopp, C.W.; Koppensteiner, R.; Lang, I.M.; Pultar, J.; Lee, S.; Weikert, C.; Panzer, S.; Gremmel, T. Decreased platelet inhibition by P2Y12 receptor blockers in anaemia. Eur. J. Clin. Investig. 2018, 48, e12861. [Google Scholar] [CrossRef]
  247. Giustino, G.; Kirtane, A.J.; Baber, U.; Genereux, P.; Witzenbichler, B.; Neumann, F.J.; Weisz, G.; Maehara, A.; Rinaldi, M.J.; Metzger, C.; et al. Impact of Anemia on Platelet Reactivity and Ischemic and Bleeding Risk: From the Assessment of Dual Antiplatelet Therapy with Drug-Eluting Stents Study. Am. J. Cardiol. 2016, 117, 1877–1883. [Google Scholar] [CrossRef]
  248. Averett, D.R.; Fletcher, S.P.; Li, W.; Webber, S.E.; Appleman, J.R. The pharmacology of endosomal TLR agonists in viral disease. Biochem. Soc. Trans. 2007, 35, 1468–1472. [Google Scholar] [CrossRef] [Green Version]
  249. Sun, S.; Rao, N.L.; Venable, J.; Thurmond, R.; Karlsson, L. TLR7/9 antagonists as therapeutics for immune-mediated inflammatory disorders. Inflamm. Allergy Drug Targets 2007, 6, 223–235. [Google Scholar] [CrossRef]
  250. Proud, P.C.; Tsitoura, D.; Watson, R.J.; Chua, B.Y.; Aram, M.J.; Bewley, K.R.; Cavell, B.E.; Cobb, R.; Dowall, S.; Fotheringham, S.A.; et al. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model. EBioMedicine 2021, 63, 103153. [Google Scholar] [CrossRef]
  251. Williams, B.; Neder, J.; Cui, P.; Suen, A.; Tanaka, K.; Zou, L.; Chao, W. Toll-like receptors 2 and 7 mediate coagulation activation and coagulopathy in murine sepsis. J. Thromb. Haemost. 2019, 17, 1683–1693. [Google Scholar] [CrossRef] [PubMed]
Figure 1. TLR-4-mediated pathways. Abbreviations: AKT—protein kinase B, ATF2—activating transcription factor 2, AP-1—activator protein 1, Bax/Bcl-2 ratio—regulator protein ratio known to be responsible for apoptosis, GSK3ß—glycogen synthase kinase 3 beta, HO-1—heme oxygenase 1, IKKα/β—IκB kinase alpha/beta, IKK ε—IκB kinase epsilon, IRAK—interleukin receptor associated kinase, IRFs—interferon regulator factor, TAK—transforming growth factor-β-activated kinase, TBK1—TANK binding kinase, TIR—toll interleukin receptor, TIRAP—TIR domain-containing adaptor protein, TRAM—TRIF-related adaptor molecule, TRAF—tumor necrosis associated factor, TRIF—TIR domain-containing adaptor protein inducing interferon, MAPK—mitogen-activated protein kinases, MyD88—myeloid differentiation primary response protein 88, NEMO—NF-kappa-B essential modulator/inhibitor of nuclear factor kappa-B kinase subunit gamma, NF-κB—nuclear factor k light chain enhancer of activated B cells, NRF2—nuclear factor erythroid-2-related factor 2, PKR—double-stranded RNA-dependent protein kinase, PI3K—phosphoinositide 3-kinase, ROS—reactive oxygen species, SARM—selective androgen receptor modulators, SOD1—superoxide dismutase 1.
Figure 1. TLR-4-mediated pathways. Abbreviations: AKT—protein kinase B, ATF2—activating transcription factor 2, AP-1—activator protein 1, Bax/Bcl-2 ratio—regulator protein ratio known to be responsible for apoptosis, GSK3ß—glycogen synthase kinase 3 beta, HO-1—heme oxygenase 1, IKKα/β—IκB kinase alpha/beta, IKK ε—IκB kinase epsilon, IRAK—interleukin receptor associated kinase, IRFs—interferon regulator factor, TAK—transforming growth factor-β-activated kinase, TBK1—TANK binding kinase, TIR—toll interleukin receptor, TIRAP—TIR domain-containing adaptor protein, TRAM—TRIF-related adaptor molecule, TRAF—tumor necrosis associated factor, TRIF—TIR domain-containing adaptor protein inducing interferon, MAPK—mitogen-activated protein kinases, MyD88—myeloid differentiation primary response protein 88, NEMO—NF-kappa-B essential modulator/inhibitor of nuclear factor kappa-B kinase subunit gamma, NF-κB—nuclear factor k light chain enhancer of activated B cells, NRF2—nuclear factor erythroid-2-related factor 2, PKR—double-stranded RNA-dependent protein kinase, PI3K—phosphoinositide 3-kinase, ROS—reactive oxygen species, SARM—selective androgen receptor modulators, SOD1—superoxide dismutase 1.
Cells 12 01865 g001
Figure 2. Platelets as drivers of leukocyte-mediated immunity and immuno-thrombosis via toll-like receptors (TLRs).
Figure 2. Platelets as drivers of leukocyte-mediated immunity and immuno-thrombosis via toll-like receptors (TLRs).
Cells 12 01865 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

Panzer, B.; Kopp, C.W.; Neumayer, C.; Koppensteiner, R.; Jozkowicz, A.; Poledniczek, M.; Gremmel, T.; Jilma, B.; Wadowski, P.P. Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review. Cells 2023, 12, 1865. https://doi.org/10.3390/cells12141865

AMA Style

Panzer B, Kopp CW, Neumayer C, Koppensteiner R, Jozkowicz A, Poledniczek M, Gremmel T, Jilma B, Wadowski PP. Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review. Cells. 2023; 12(14):1865. https://doi.org/10.3390/cells12141865

Chicago/Turabian Style

Panzer, Benjamin, Christoph W. Kopp, Christoph Neumayer, Renate Koppensteiner, Alicja Jozkowicz, Michael Poledniczek, Thomas Gremmel, Bernd Jilma, and Patricia P. Wadowski. 2023. "Toll-like Receptors as Pro-Thrombotic Drivers in Viral Infections: A Narrative Review" Cells 12, no. 14: 1865. https://doi.org/10.3390/cells12141865

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