Next Article in Journal
Targeting Prostate Cancer Cells Using Anti-Sortilin and Anti-Syndecan-1 Antibody Drug Conjugates
Previous Article in Journal
Genetic Biomarkers for Statin-Induced Myopathy
Previous Article in Special Issue
Pathophysiology of Pulmonary Arterial Hypertension: Focus on Vascular Endothelium as a Potential Therapeutic Target
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Toll-like Receptors and Viral Infections in the Pathogenesis and Progression of Pulmonary Arterial Hypertension—A Narrative Review

by
Agnieszka Styczeń
1,
Martyna Krysa
1,
Paulina Mertowska
2,*,
Ewelina Grywalska
2,
Tomasz Urbanowicz
3,
Maciej Krasiński
4,
Malwina Grobelna
5,
Weronika Topyła-Putowska
1,
Mansur Rahnama-Hezavah
6 and
Michał Tomaszewski
1
1
Department of Cardiology, Medical University of Lublin, Jaczewskiego 8, 20-093 Lublin, Poland
2
Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
3
Cardiac Surgery and Transplantology Department, Poznań University of Medical Sciences, 1/2 Dluga Street, 61-848 Poznań, Poland
4
Department of Obstetrics and Gynaecology, St Mary’s Hospital, Imperial College Healthcare Trust, London W2 1NY, UK
5
Department of Vascular, Endovascular Surgery, Angiology and Phlebology, Poznan University of Medical Science, 1/2 Długa Street, 61-848 Poznań, Poland
6
Chair and Department of Oral Surgery, Medical University of Lublin, 20-093 Lublin, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(22), 11143; https://doi.org/10.3390/ijms262211143
Submission received: 29 September 2025 / Revised: 8 November 2025 / Accepted: 14 November 2025 / Published: 18 November 2025

Abstract

Aberrant activation of innate immunity promotes the development of pulmonary arterial hypertension (PAH); however, the role of pattern recognition by Toll-like receptors (TLRs) within the pulmonary vasculature remains unclear. To consolidate knowledge (as of June 2025) about TLRs and their interactions with viruses in PAH and to identify therapeutic implications. A narrative review of experimental and clinical studies investigating ten TLRs in the context of the pulmonary vascular microenvironment and viral infections. Activation of TLR1/2, TLR4, TLR5/6, TLR7/8, and TLR9 converges on the MyD88–NF-κB/IL-6 axis, thereby enhancing endothelial-mesenchymal transition, smooth muscle proliferation, oxidative stress, thrombosis, and maladaptive inflammation, ultimately increasing pulmonary vascular resistance. Conversely, TLR3, through TRIF–IFN-I, preserves endothelial integrity and inhibits vascular remodeling; its downregulation correlates with PAH severity, and poly (I:C) restitution has been shown to improve hemodynamics and right ventricular function. HIV-1, EBV, HCV, endogenous retrovirus K, and SARS-CoV-2 infections modulate TLR circuits, either amplifying pro-remodeling cascades or attenuating protective pathways. The “TLR rheostat” is shaped by polymorphisms, ligand biochemistry, compartmentalization, and biomechanical forces. The balance between MyD88-dependent signaling and the TRIF–IFN-I axis determines the trajectory of PAH. Prospective therapeutic strategies may include TLR3 agonists, MyD88/NF-κB inhibitors, modulation of IL-6, and combination approaches integrating antiviral therapy with targeted immunomodulation in a precision approach.

1. Introduction

Pulmonary arterial hypertension (PAH) is a rare, progressive disorder characterized by elevated pulmonary arterial pressure and pulmonary vascular resistance, ultimately leading to right ventricular failure and death if left untreated. Despite significant advances in pharmacological therapies targeting endothelial dysfunction, vasoconstriction, and smooth muscle cell proliferation, the pathogenesis of PAH remains incompletely understood, and its prognosis continues to be poor [1,2,3,4].
Recent evidence suggests that chronic inflammation, immune dysregulation, and aberrant innate immune signaling play crucial roles in the initiation and progression of PAH [5,6,7,8]. Among the principal components of the innate immune system, Toll-like receptors (TLRs)—a family of pattern recognition receptors (PRRs)—have emerged as pivotal mediators linking environmental and microbial stimuli to vascular remodeling, endothelial dysfunction, and immune cell infiltration within the pulmonary circulation [9,10,11,12]. TLRs recognize conserved molecular motifs derived from pathogens (pathogen-associated molecular patterns, PAMPs) or endogenous danger signals (damage-associated molecular patterns, DAMPs), thereby initiating downstream signaling cascades that culminate in the production of cytokines, chemokines, and interferons (Table 1) [9,13,14].
In humans, ten TLRs (TLR1–TLR10) are expressed. The role of specific receptors in pulmonary vascular disease remains an active area of investigation. Several preclinical studies have demonstrated the upregulation of TLR2, TLR3, TLR4, and TLR9 in lungs and vascular cells in experimental PAH models [15,16,17,18]. Functionally, TLR activation contributes to the core pathological mechanisms of PAH—including endothelial cell apoptosis, proliferation of pulmonary arterial smooth muscle cells (PASMCs), oxidative stress, and sustained inflammation—thereby driving vascular remodeling and hemodynamic deterioration [6,19,20]. In contrast, certain receptors, most notably TLR3, exert protective effects by modulating interferon signaling and preserving endothelial integrity, underscoring the receptor-specific balance within TLR networks that may ultimately shape disease trajectory and therapeutic opportunities [21,22].
Table 1. Summary of ligands recognized by Toll-like receptors (TLR1–TLR10): pathogen-associated molecular patterns (PAMPs) and endogenously derived danger signals (DAMPs) and their significance in immunopathology [23,24,25,26,27,28,29,30,31,32,33,34].
Table 1. Summary of ligands recognized by Toll-like receptors (TLR1–TLR10): pathogen-associated molecular patterns (PAMPs) and endogenously derived danger signals (DAMPs) and their significance in immunopathology [23,24,25,26,27,28,29,30,31,32,33,34].
TLRPAMPDAMP
TLR1/2Triacylated bacterial lipopeptides (e.g., Pam3CSK4; TLR1/2 heterodimer).No specific, reliable DAMPs for TLR1 itself; DAMP signaling usually via TLR2-dependent heterodimers.
TLR2Gram(+) bacteria: lipopeptides, lipoteichoic acid, peptidoglycan; zymosan (fungi); often as TLR2/6.HMGB1, hyaluronan fragments, biglycan, HSP;
TLR3dsRNA viral; agonist: poly(I:C).Endogenous dsRNA/exosomal RNA from necrotic cancer cells/tissues.
TLR4LPS (Gram−); classic endotoxin receptor.HMGB1, S100A8/A9, heme, ECM fragments (e.g., hyaluronan), and fibronectin-EDA are the TLRs with the most significant number of proposed DAMPs (some of which are still under discussion).
TLR5Bacterial flagellin.There are no widely accepted endogenous DAMPs for humans.
TLR6/2Diacylated bacterial lipopeptides (e.g., Pam2CSK4; TLR2/6 heterodimer).As for TLR2, HMGB1/ECM/HSP has been reported, but data for TLR6 alone are sparse.
TLR7viral ssRNA (GU-rich); agonists: imidazoquinolines.Endogenous miRNAs (e.g., miR-21, let-7, miR-154-5p); RNA–LL37 complexes facilitating self-RNA recognition.
TLR8Viral ssRNA; R848 and other small molecules.Extracellular miRNAs (e.g., miR-21/miR-29a in tumor exosomes); RNA–LL37 complexes activating TLR8 in neutrophils.
TLR9Unmethylated CpG bacterial/viral DNA; agonist: CpG ODN.mtDNA (also oxidized) and DNA–LL37 complexes facilitating the recognition of one’s own DNA.
TLR10Function/ligands remain ambiguous; lipopeptides are suggested (often as TLR2/10); the role is more likely to be modulatory/inhibitory.No reliably confirmed DAMPs; receptor still considered “orphan”.
Abbreviations: TLR—Toll-like receptor, PAMP—Pathogen-associated molecular pattern, DAMP—Danger-associated molecular pattern, HMGB1—High mobility group box one protein, HSP—Heat shock protein, dsRNA—Double-stranded RNA, poly(I:C)—Polyinosinic: polycytidylic acid (synthetic dsRNA analog), LPS—Lipopolysaccharide, S100A8/A9—S100 calcium-binding proteins A8 and A9 (also known as calprotectin), ECM—Extracellular matrix, ssRNA—Single-stranded RNA, miRNA (miR)—MicroRNA, LL37—Human cathelicidin antimicrobial peptide (LL-37), R848—Resiquimod (synthetic imidazoquinoline, TLR7/8 agonist), CpG ODN—CpG oligodeoxynucleotide (synthetic unmethylated CpG DNA), mtDNA—Mitochondrial DNA.
Furthermore, the interaction between TLR activation and viral infections has garnered attention as a potential trigger in the development of PAH, particularly among predisposed individuals. Human viruses such as HIV, Epstein–Barr virus, and SARS-CoV-2 can enhance pulmonary vascular remodeling and immune activation through direct and indirect mechanisms involving TLR pathways [17,34,35,36,37,38]. In predisposed hosts (e.g., with polymorphisms in TLR/adapter genes, attenuated IFN-I response, autoimmune diseases, or environmental exposures), viral PAMPs (ssRNA/dsRNA, CpG DNA, envelope proteins) and secondary DAMPs released from damaged tissues (mtDNA, HMGB1, NETs) activate TLR2/4 on endothelium and smooth muscle cells, as well as TLR3/7/8/9 on antigen-presenting cells and adventitia. This results in the activation of the MyD88–NF-κB/IL-6 axis and, in context, the TRIF–IFN-I axis, which promotes endothelial barrier dysfunction, endothelial-mesenchymal transition, PASMC proliferation, fibrotic remodeling, thromboinflammatory microangiopathy, and sustained leukocyte recruitment. Additionally, virally derived or host miRNAs carried in extracellular vesicles can allosterically amplify TLR7/8/9 signaling. The reactivation of latent viruses (e.g., EBV) under conditions of chronic inflammation can act as a secondary trigger, sustaining a pathological positive feedback loop. Accumulating evidence indicates that the TLR–virus axis represents an important, modifiable hub in PAH pathobiology, which may account for the heterogeneity of clinical phenotypes and differences in treatment response [17,34,35,36,37,38].
In this narrative review, we aim to synthesize current knowledge on the role of Toll-like receptors in the pathogenesis and progression of pulmonary arterial hypertension. We further highlight the contribution of viral infections to TLR-mediated immune responses in PAH, with particular emphasis on the translational relevance of TLRs as potential biomarkers and therapeutic targets.

2. The Role of Toll-like Receptors in the Pathogenesis and Progression of Pulmonary Arterial Hypertension

2.1. TLR1 and TLR2

TLR1 and TLR2 constitute the functional TLR1/2/6/10 subfamily encoded on chromosome 4, in which heterodimerization-dependent signaling at the plasma membrane enables recognition of a broad spectrum of microbial lipopeptides [39,40]. TLR1 (CD281) is a 786-amino acid transmembrane glycoprotein containing 19 LRR repeats, abundantly expressed in the spleen, kidney, and lung of adults and—at the cellular level—on B and T lymphocytes (CD4+), monocytes/macrophages, selected dendritic cell subpopulations, astrocytes, microglia, and platelets; relatively lower expression is observed in the fetal brain and liver [17,41,42,43]. This receptor preferentially binds triacylated bacterial lipopeptides and some parasitic proteins, in the absence of reliably confirmed endogenous ligands; after ligand binding, it most often acts as a TLR1/2 heterodimer, initiating the MyD88→NF-κB cascade and inducing proinflammatory cytokines. In the context of barrier physiology, TLR1 is involved in sensing the gut microbiota and recognizing Borrelia burgdorferi surface lipoproteins, which determines an effective response to the etiological agent of Lyme disease [44,45]. TLR2 (CD282), which shares a similar structure (784 amino acids, 19 LRRs), is present on B and T lymphocytes—including Tregs (CD4+CD25+)—neutrophils, monocytes/macrophages, mast cells, myeloid dendritic cells, Schwann cells, and microglia. It recognizes complex bacterial lipo- and glycopeptides, the lipoteichoic acid of Gram-positive bacteria, and fungal β-glucans. In addition, Hsp70, gp96, and signals from necrotic cells have been described as potential DAMPs [46,47,48,49]. Functionally, TLR2 acts cooperatively with other receptors: in association with TLR1, it preferentially recognizes triacylated lipopeptides, whereas with TLR6, it preferentially detects diacylated lipopetides. In both cases, receptor activation leads to the recruitment of MyD88, subsequent activation of NF-κB, and the expression of inflammatory mediators, forming a key module of innate pathogen recognition and integration of environmental signals into the effector response [46,47,48,49].
In a rat model of monocrotaline-induced pulmonary hypertension (MCT-PAH), macrophage infiltration in the lung tissue was observed, accompanied by impaired necroptosis dependent on receptor-interacting protein kinase-3 (RIPK3) and activation of the associated TLR pathway [50]. In 2023, Lian et al. attempted to better explain the role of single Toll-like receptors in the development and progression of PAH induced in Sprague-Dawley rats. The animals were administered monocrotaline subcutaneously twice at an interval of 7 days, at a dose adjusted to the animal’s body weight (20 mg/kg b.w.). The study examined the characteristics of infiltrating cells in the immune system and sought potential biomarkers for PAH using a comprehensive bioinformatics approach. Critical central genes were identified. The bioinformatics tool CIBERSORT was employed to assess the characteristics of infiltrating immune cells. Subsequently, the correlation between central genes and immune cells was examined using Pearson analysis [51].
Lian et al. demonstrated that the expression of TLR1 genes in rats with MT-PAH was reduced in comparison to the control group. Moreover, this expression correlated positively with the number of neutrophils and monocytes. On the other hand, a negative correlation was observed between TLR1 gene expression and the number of resting CD4+ T lymphocytes. This test confirmed that the TLR1 receptor plays a pivotal role in the functioning of the innate immune system. It is also worth noting that, as a representative of the TLR family, it participates in the recognition of a wide range of pathogens. The positive correlation between TLR1 expression and the number of monocytes and macrophages suggests that TLR1 may contribute to the development of PAH by regulating neutrophil- and monocyte-related immune responses, as well as the progression of subsequent inflammatory states [51] (Table 2).
Overall, TLR2 plays a significant role in the pathogenesis of pulmonary arterial hypertension by participating in the activation of endothelial cells and vascular smooth muscle cells, thereby contributing to vascular remodeling, and facilitating the development of the inflammatory response. Preclinical studies have shown that TLR2 activation is associated with reduced apoptosis and proliferation of PASMC, increased production of growth factors (e.g., TGF-beta, PDGF), as well as the induction of fibroblasts and extracellular matrix deposition. Excessive activation of TLR2 in animal models leads to oxidative stress, increased expression of adhesion molecules (ICAM-1, VCAM-1)—which facilitates leukocyte migration—and endothelial dysfunction (impaired permeability and impaired nitric oxide production). Nevertheless, it is essential to emphasize that the most crucial pathophysiological mechanism by which TLR2 activation contributes to the development of PAH is the induction of chronic inflammation. The induction occurs via the activation of the NF-kB pathway and the production of proinflammatory cytokines, including IL-1, IL-6, and TNF-alpha, as accompanied by the recruitment of macrophages and neutrophils, and the ability to maintain the already initiated chronic inflammation in the pulmonary vessels. Furthermore, enhanced TLR2 activity correlates with the severity of the clinical condition and with the deterioration of the right ventricular function of the animals. Therefore, modifying the activity of this receptor may in the future become a potential therapeutic target in the treatment of PAH [15,42,52,53] (Table 2).
Notably, Broen et al. demonstrated that a rare polymorphism in the TLR2 gene is associated with the systemic sclerosis phenotype and enhances the production of inflammatory mediators. The described trial revealed that among patients with systemic sclerosis, the rare TLR2 Pro631His variant is associated with antitopoisomerase positivity (the SSc diffuse manifestation) and the development of PAH. Additionally, this variant has an impact on TLR-2-mediated cellular responses. Specifically, monocyte-derived dendritic cells carrying the Pro63His variant cause the excessive production of proinflammatory cytokines (IL-5 and NRF-alpha), which is a consequence of TLR2 activation. However, more research is required on this topic to clarify the significance of TLR2 in the pathogenesis of both SSc and PAH [43].

2.2. TLR3

TLR3, a transmembrane glycoprotein also known as CD283, consists of 904 amino acids and contains 23 LRRs. The genes encoding TLR3 are located on chromosome 4. This receptor is located intracellularly in various cell types, including B lymphocytes and dendritic cells. The intracellular organelles in which the cell membrane TLR3 is expressed are the endoplasmic reticulum, phagosomes, and early endosomes. Exogenous ligands for TLR3 are viral or synthetic double-stranded RNA and polycytidylic acid, whereas mRNA constitutes the TLR3 endogenous ligand [54,55]. TLR3 is a unique receptor in the entire family of TLR due to its activation occurring independently of the MyD88 protein (Figure 1). The further signaling cascade uses the adaptor protein TIR-domain-containing adapter-inducing interferon-β (TRIF). In the division of all TLR3 into subfamilies, this receptor constitutes a separate subfamily on its own [56,57,58,59].
In the human body, the highest expression of TLR3 is found in the cells of the pancreas and the placenta. In relation to the cardiovascular system, activators of TLR3 have been shown to affect human vascular cells [54,55]. It has been observed that TLR3 plays a protective role against the development of atherosclerotic lesions in a mouse model of the disease [60,61]. Additionally, stimulation of TLR3 signaling is associated with ischemic preconditioning-induced protection against brain ischemia. It has also been demonstrated that the TLR is responsible for the attenuation of reactive astrogliosis, a phenomenon observed in cases of central nervous system ischemia [62,63,64]. TLR3 is also a promising target for antiviral drugs, as its activation in the final phase of the immune response leads to the production of substantial amounts of interferon-1 subtypes [28,65,66].
Evidence shows that TLR3 plays a protective role in the pathogenesis of pulmonary arterial hypertension. In a 2019 clinical study, Farkas et al. hypothesized that the expression of TLR3 is an essential process for maintaining endothelial cell homeostasis and protection, and the activation of TLR3 with drugs is a potential therapeutic target in the treatment of pulmonary arterial hypertension [21]. The study was conducted using a mouse model of pulmonary arterial hypertension induced by chronic hypoxia and the active substance SU5416, and also examined fragments of human lung tissue. It demonstrated a significantly reduced expression of the TLR3 receptor in the endothelial cells of vascular occlusive changes, typical of PAH. Moreover, decreased expression of PAH in the lung tissue of animals was observed. This phenomenon resulted in a marked exacerbation of the disease and enhanced endothelial cell apoptosis in mice exposed to chronic hypoxia and treated with the SU5416 substance. In addition, it was found that in the endothelium of plexiform and concentric vascular lesions, there was a significant loss of TLR3 expression. At the same time, in the control group of patients, endothelial cells showed strong expression of this receptor. It should also be emphasized that the study confirmed reduced expression of TLR3 in endothelial cells in cultured pulmonary artery cells (PAEC) and in lung tissue with induced pulmonary arterial hypertension. These findings led to the conclusion that the loss of TLR3 expression leads to pulmonary vascular remodeling, resulting in a more intensive vascular remodeling process [21].
The next stage of the study assessed the intensity of the caspase-dependent apoptosis process of pulmonary artery endothelium cells. The study group demonstrated an increased number of apoptotic PAEC intimal cells, accompanied by reduced TLR3 expression. The increased intensity of the apoptosis process in cells characterized by reduced TLR3 expression was also confirmed in vitro. It is worth noting that the study also showed that reduced TLR3 expression results in increased expression of genes for IL-6 and endothelin-1, which consequently promotes a gene expression profile that favors increased vascular remodeling, endothelial dysfunction, and the development of local inflammation [21].
Farkas D. et al. also examined the effect of the dsRNA substance PolyC (I:C), a TLR3 agonist, on the functioning of PAEC. It has been demonstrated that treatment with this substance prevents the development of PAH, inhibits its progression, and reduces the severity of right ventricular failure. Moreover, it diminishes the intensity of proliferation and remodeling of the pulmonary artery wall, while simultaneously limiting its apoptosis. Furthermore, the administration of Poly (I:C) did not intensify local inflammation. In summary, the study demonstrated for the first time that reduced TLR3 expression is present in the lung tissue and endothelial cells of patients with PAH, both in vitro and in vivo, which leads to increased cell susceptibility to apoptosis and the development of PAH. Moreover, both the dsRNA of the substance PolyC (I:C)—polyinosinic/polycytidylic acid and the TLR3 agonist increase TLR3 gene expression and reduce the severity of existing PAH. This protective effect of TLR3 up-regulation is most likely mediated by the induction of interleukin-10. Based on the entire study, a key conclusion was drawn that the expression of TLR3 is a crucial element in maintaining PAEC homeostasis. Therefore, it is necessary to conduct further research to identify active substances that restore appropriate TLR3 expression, which may represent a new and effective therapeutic strategy for treating PAH, complementing existing methods [21,67].
In 2023, Bhagwani A.R. et al. attempted to supplement the existing knowledge about TLR3 in the pathogenesis of PAH. The researchers aimed to elucidate the roles of the TLR3 receptor and p53 protein in the clonal expansion and pathogenesis of PAH by regulating bone morphogenetic protein type 2 (BMPR2) signaling. The study demonstrated reduced gene expression of the p53 protein in PAEC, which exacerbated existing pulmonary arterial hypertension. Additionally, it was observed that reducing p53 degradation resulted in the induction of TLR3 and BMPR2 and the abolition of clonal expansion of EC, thus alleviating the course of PAH. In addition, the substance Poly(I:C) was responsible for the increased binding of interferon regulatory factor (IRF3) to the BMPR2 promoter, thereby reducing the severity of pulmonary arterial hypertension in mice with activated p53 protein. This phenomenon was not observed in animals with TLR3 downregulation [22].

2.3. TLR4

The genes encoding TLR4, also known as CD284, are located on chromosome 9, and the entire receptor is composed of 839 amino acids [68]. At the histological level, TLR4 is situated outside the cell on various cell types, including neutrophils, T lymphocytes, monocytes/macrophages, mast cells, myeloid dendritic cells, intestinal epithelial cells, and breast cancer cells [69,70,71]. Ligands for this particular transmembrane glycoprotein include many chemical substances, the most important of which, and simultaneously the only mentioned exogenous ligand, is lipopolysaccharide, which is found in all Gram-negative bacteria and some Gram-positive bacteria. Endogenous ligands for TLR4 include fragments of heparan sulfate, hyaluronic acid, fibrinogen, various opioid drugs, heat shock proteins, and nickel [72,73,74]. Among all TLRs, TLR4 constitutes a distinct subfamily. This receptor is also an exception among all receptors, as it is the only one that can transmit a signal in the pathway using the MyD88 protein, both with and without its participation (Figure 2) [69,70,75].
In the human body, TLR4 plays a pivotal role due to its proven involvement in the pathogenesis of numerous infectious and neoplastic diseases. It has been demonstrated that stimulation of TLR4 with lipopolysaccharide or its natural derivatives activates its robust anti-cancer activity through the production of pro-inflammatory cytokines and type 1 interferon [76,77].
TLR4 has also been shown to have a significant role in the occurrence of long-term adverse effects of opioid drugs. Activation of TLR4 results in the release of substances that modulate the current inflammatory state, including IL-1β and TNF-α. The constant presence of these substances in the human body most likely causes a decrease in the effectiveness of opioid treatment after some time. This, in turn, leads to the development of tolerance, allodynia, and hyperalgesia [78,79]. For a long time, scientific studies have also highlighted the role of TLR4 in both the pathogenesis and development of neurodegenerative diseases, such as Parkinson’s disease, Alzheimer’s disease, and Huntington’s disease [80,81,82].
TLR4 is a critical mediator of innate immunity and vascular homeostasis. Its dysregulation has been increasingly implicated in the pathogenesis of pulmonary arterial hypertension, where chronic inflammation, vascular remodeling, and thrombosis play key roles. In the pulmonary circulation, TLR4 is expressed not only on immune cells but also on endothelial cells, pulmonary artery smooth muscle cells (PASMCs), and platelets—key cellular players in the development of PAH [19,69,72,83,84,85,86,87].
Activation of TLR4 by exogenous ligands (e.g., lipopolysaccharide) or DAMPs such as high-mobility group box one protein (HMGB1), fibronectin, or hyaluronic acid, triggers signaling cascades via MyD88 and TRIF, converging on NF-κB, AP-1, and interferon regulatory factors, and inducing pro-inflammatory cytokines such as IL-6 and TNF-α [24,88].
In pulmonary arterial endothelial cells (PAECs), TLR4 activation promotes increased permeability, endothelial-to-mesenchymal transition (EndMT), and oxidative stress, leading to vascular dysfunction and remodeling. Inhibition of TLR4 in experimental models has been shown to restore endothelial barrier integrity and reduce pro-inflammatory signaling [24,88].
In PASMCs, TLR4 regulates redox balance via the Nox1/Nox4–ROS axis. Hypoxic conditions suppress TLR4 expression and enhance NADPH oxidase activity, promoting ROS generation, PASMC proliferation, and vessel wall thickening. TLR4-deficient mice spontaneously develop PAH-like features, including increased right ventricular systolic pressure (RVSP) and medial hypertrophy of pulmonary arteries [87,89].
Platelet-expressed TLR4 contributes to thromboinflammation by enhancing the release of vasoactive substances, including serotonin, thromboxane A2, and growth factors (e.g., PDGF, TGF-β). TLR4-deficient platelets exhibit attenuated aggregation and reduced release of inflammatory mediators, resulting in milder PAH severity in animal models [90,91].
HMGB1–TLR4 signaling has also been shown to suppress bone morphogenetic protein receptor 2 (BMPR2) expression, a hallmark of PAH pathophysiology. This inhibition reduces Smad1/5/8 phosphorylation and Id1 expression, promoting PASMC proliferation. Targeted inhibition of the HMGB1–TLR4 axis restores BMPR2 signaling and attenuates pulmonary vascular remodeling [92,93].
Furthermore, gut-derived ligands such as LPS and trimethylamine-N-oxide (TMAO) can activate TLR4 in the lung, linking gut dysbiosis to pulmonary vascular inflammation via the systemic HMGB1–TLR4 axis [94,95].
Pharmacologic modulation of TLR4 offers promise. Cinnamaldehyde, a natural TLR4 inhibitor, alleviates hypoxia-induced PAH in vivo by suppressing the TLR4/NF-κB/HIF-1α pathway, reducing right ventricular hypertrophy and collagen deposition [52,89]. However, systemic TLR4 blockade may carry immunological risks; future strategies should prioritize ligand- or cell-specific targeting.

2.4. TLR5 and TLR6

TLR5 (CD285) is a receptor whose coding genes are located on chromosome 1. Its exact location in human body cells has not been determined so far. However, it has been demonstrated that TLR5 is located on the outer surface of cells, including B lymphocytes, regulatory lymphocytes, macrophages, and dendritic cells. TLR5 consists of 20 LRRs, and the entire receptor is composed of 858 amino acids. TLR5 is a separate subfamily among all TLRs [96]. Activation of TLR5 occurs through the MyD88 protein pathway, and the pathogen-associated molecular pattern for this described transmembrane glycoprotein is flagellin, a component of the flagella of Gram-negative bacteria, which serves as an extrinsic ligand for the receptor. The endogenous ligands for TLR5 are still unknown [97,98,99,100]. It has also been demonstrated that aberrant functioning of TLR5 is associated with the onset of inflammatory diseases, including inflammatory bowel disease and rheumatoid arthritis, as well as with the development of neoplastic diseases, such as breast, cervical, endometrial, gastric, and ovarian cancer [101,102,103,104,105,106,107].
The TLR5-Nox4 signaling pathway has been proven to be a key element in the development of atherosclerosis. It has been observed that the flagellin-TLR-5-Nox4 axis stimulates the movement of vascular smooth muscle cells and, consequently, initiates the formation of atherosclerotic plaque. It was shown that the flagellin-TLR5-Nox4 axis enhances inflammatory processes in the endothelium by activating the NF-kB factor, inducing the expression of proinflammatory cytokines such as IL-6, IL-8, RANTES (Regulated on Activation, Normal T-cell Expressed and Secreted), and the adhesion factor ICAM-1 (intercellular adhesion molecule-1). It was also noted that in mice lacking Nox4, both of the processes mentioned above were significantly less intense [108,109,110,111,112].
To date, there are no dedicated functional studies in PAH models that precisely determine the role of TLR5 in the pathogenesis and progression of this disease (Table 3). Transcriptomic analysis (using a mouse model with pulmonary hypertension) showed a significant induction of TLR5 expression in the lung tissue of patients with PAH. TLR5 recognizes flagellin in bacteria, activating the MyD88 →IRAK→NF-kB pathway, which leads to the release of cytokines such as TNF-alpha, IL-6, and IL-8. On this basis, it can be assumed that it thereby stimulates the formation of a local inflammatory state, analogously to TLR2 and TLR4, which are responsible for the recruitment of leukocytes, oxidative stress, and the phenomenon of vascular remodeling. As TLR5 may play a pro-inflammatory and remodulatory role, further studies, both in vivo and in vitro, should be conducted to assess the potential for intervention in PAH therapy by modulating TLR5 function [113,114,115].
The genes encoding TLR6 are located on chromosome 4. In the human body, TLR6 (CD286) is situated extracellularly in cells such as B lymphocytes, monocytes/macrophages, and mast cells. This receptor consists of 19 LRRs and 796 amino acids. TLR6, similarly to TLR5, is activated via the MyD88 protein pathway, which stimulates the nuclear factor NF-kB. The exogenous ligands for TLR6 are multiple diacyl lipopeptides, while endogenous ligands for TLR6 have not been discovered yet. TLR6 belongs to the TLR1/2/6/10 subfamily of receptors [116,117].
It has been proven that A 359T>C single-nucleotide polymorphism in the structure of the extracellular LRR of TLR6 predisposes to Legionnaires’ Disease development [118]. Moreover, there are assumptions that the Ser249Pro polymorphism in TLR may be associated with a greater incidence of asthma bronchialis in some populations [119].
Grote K. et al. attempted to determine the role of this transmembrane glycoprotein in the functioning of the vascular system. They conducted studies showing a significant participation of the TLR2/TLR6 complex in the process of angiogenesis. It was observed that lipopolysaccharide, a macrophage-activating lipopeptide of 2 kDa (MALP-2), commonly found in bacteria, stimulated the formation of new vessels through a pathway mediated by the TLR2/TLR6 complex, the activation of which causes the release of granulocyte-macrophage colony-stimulating factor (GM-CSF). Stimulation of angiogenesis, involving the described complex, was confirmed to occur both in vitro and in vivo [120].
No scientific papers have been published to explain the importance of TLR6 in PAH in direct clinical trials. Only transcriptional RNA-seq analyses are available in an animal model of PAH induced, for example, by monocrotaline. These studies demonstrate that stimulation of the TLR6/TLR2 heterodimer in PAMSCs by typical bacterial ligands activates the NF-κB pathway, promoting the production of IL-1β, IL-6, and TNF-α, which are crucially involved in pulmonary vascular remodeling. Although there are no direct studies describing TLR6 activity in animals with PAH, there are strong indirect indications that activation of this receptor promotes vascular proliferation and remodeling, the increase of endothelin-1 levels, and the development of local inflammation in PAH. Due to the dynamic growth of knowledge on PAH, further research is also necessary in this field to evaluate TLR6 as a potential therapeutic target in PAH, which could be achieved by antagonizing signals in the TLR2/TLR6 heterodimer and NF-κB axis (Table 3) [15,42].

2.5. TLR7 and TLR8

The genes TLR7 and TLR8, which are subsequent transmembrane glycoproteins, are located on the X chromosome in the human body. TLR7 (CD287) is located intracellularly in structures such as lysosomes, endoplasmic reticulum, and phagosomes; however, the precise location of TLR8 (CD288) inside the cell remains unknown [121,122]. Both TLR7 and TLR8 are composed of 25 LRRs. Both receptors have a similar number of amino acids—TLR consists of 1049 amino acids, while TLR8 is composed of 1041 amino acids. TLR7 and TLR8, together with TLR9, form one of the six subfamilies of TLRs, distinguished by amino acid sequence homology. The single-stranded DNA of viruses constitutes an extrinsic ligand for both TLR7 and TLR8, while their intrinsic ligands have not been identified so far. Activation of the signaling cascade of TLR7 and TLR8 receptors occurs using the MyD88 protein [121,122,123,124,125]. TLR7 is present on B lymphocytes and plasmacytoid dendritic cells, and when activated, it produces signaling using interferon-gamma. In contrast, TLR8 induces NF-κB-dependent cytokines in myeloid cells (Figure 3) [126,127,128,129,130,131].
In the human vascular system, TLR7 and TLR8 act as “guardians” of immunity, selectively recognizing subsets of RNA sequences crucial for an effective response against viruses [132,133]. It has also been proven that constant activation of TLR7/8 results in excessive production of proinflammatory signals, often pathological, as exemplified by a vast “cytokine storm” in the course of viral pneumonia, usually leading to the patients’ death. It has been noted that the abnormal production of interferon, mediated by TLR9 activation, can trigger a reaction exhibiting the characteristics of an autoimmune relationship, as evidenced by the repolarization of T helper lymphocytes and the autoreactive expansion of B lymphocytes [127,134]. Moreover, the local application of the TLR7/TLR8 receptor agonist resiquimod (R848) has been observed to lead to the development of autoimmune disorders similar to those observed in systemic lupus erythematosus, which is characterized by excessive production of pro-inflammatory cytokines, the production of autoantibodies, and multi-organ damage [134,135].
Yeh F.-C. et al. conducted a study using rats in 2023 to clarify the role of autoimmune phenomena induced by stimulation of TLR7 and 8 in the perpetuation of pulmonary artery endothelial damage and vascular dysfunction caused by the VEGF antagonist Sugen 5416 (SU5416) in the initial stages and subsequent stages of progression of pulmonary vascular disease in rats [136]. As is well known, SU5416 is a factor that induces endothelial cell apoptosis—a transient but specific pathophysiology underlying the phenomenon of pulmonary vascular remodeling. In the present study, SU5416 was used in combination with other factors that could potentially influence the exacerbation of pulmonary artery remodeling, such as pneumonectomy, chronic hypoxia, stimulation with hypoxia-inducible factor-1α, the presence of a vasoactive stressor, and the phenomenon of T cell exhaustion. All the described phenomena were aimed at generating experimental PAH in vivo [136].
The study used male rats weighing between 180 g and 200 g. The animals were given a single subcutaneous injection of SU5416, adjusting the dose of the substance to the animal’s body weight (20 mg/kg b.w.). Then, the rats were administered the TLR7/8 agonist R840 (R-SU) externally three times a week for a total of 5 weeks. After 5 weeks, the animals developed pulmonary hypertension with a phenotype associated with a severe course, including a significant increase in mPAP and significant right ventricular hypertrophy. In histological preparations of the lung tissue of 5W R-SU rats, obstructive proliferation of pulmonary vessels was found. Furthermore, histologically, extensive perivascular infiltration of immune cells was observed in the examined tissue. Neither SU5416 nor R848 was able to induce the pathomechanisms responsible for the development of PAH. Infiltrations of immune cells were only evident in rodents exposed to two substances simultaneously, using the ‘double hit’ principle, located around vascular changes in the lungs, as well as in the enlarged lymphoid tissue associated with the bronchi (Figure 3) [135].
Immunohistochemical preparations revealed a focal accumulation of CD103+ cells at the epidermal-dermal junction of the ears of rats treated externally with R848. Secondly, the animals showed increased expression of mRNA for interferons IFNA1 and IFNB1, as well as mRNA for TLR7 and TLR8 in the spleen, indicating the involvement of pDCs (perivascular dendritic cells) due to the activation of TLR7 and TLR8. It is believed that autoimmune processes stimulated by TLR7 activity demonstrate the action of SU5416-sustaining destructive stimuli for pulmonary blood vessels, which ultimately lead to vascular remodeling of an obliterative nature [135,136].
The study also included a group of rats that were given a prolonged 7-week exposure to SU5416, after which heart of the animals was imaged using MRI (BioSpec 9.4 T) at fixed time points—after 2, after 5, and at the time point after 7 weeks (maintained for another 2 weeks without R848). It is worth emphasizing that in rats, the extended 7-week control point protocol revealed proliferation of pulmonary arteries and increased mPAP at a similar level to the group of animals subjected to a 5-week exposure to SU5416. Data obtained from MRI studies were analyzed, noting a successive decrease in right ventricular efficiency and progression of PAH, which was expressed by progressive dilatation of the right ventricular cavity and hypertrophy of the right ventricular muscle, as well as by impairment of right ventricular function (e.g., reduced right ventricular ejection fraction—RVEF) and loss of ventricular-arterial coupling [135].
In R-SU rats, autoimmune reactions analogous to those observed in patients diagnosed with SLE were observed, with involvement of numerous internal organs, including glomerulonephritis and splenomegaly [137]. A significantly increased expression of IFNA1/IFNAB1 and TLR7 and TLR8 was observed in the spleen cells of R-SU rats, which is evidence of the animals’ response to substance R848 [136]. Repolarization towards Th17 lymphocytes was observed in the peripheral blood of the studied animals, characterized by an increased ratio of Th17 to regulatory T lymphocytes. This phenomenon is attributed to the activation of TLR7 and TLR8 and is an expression of the loss of autoregulatory mechanisms and the development of autoimmune reactions. They have been described in relation to the degree of severity and prognosis in different PAH groups [137,138,139]. It is worth emphasizing that significantly increased titers of autoantibodies were detected in the plasma of R-SU rats, including antinuclear antibodies, antibodies against double-stranded DNA, and anticentromere antibodies. Autoantibody production is a common immunopathological mechanism observed in patients with PAH. The study results also emphasized that activation of TLR7/8 receptors leads to fatal complications of SLE, such as malignant arterial hypertension and systemic vasculitis in female rats, in which the observed greater susceptibility to the development of autoimmune diseases induced by TLR7 activation may be related to the dose effect—the gene expression of the biallelic X chromosome [140,141,142].

2.6. TLR9

TLR9 (CD289), composed of 1032 amino acids, is a receptor located on chromosome 3. In its structure, TLR9 contains 25 tandem leucine-rich repeats. Histologically, TLR9 is situated inside the cell, in structures such as lysosomes, endoplasmic reticulum, and phagosomes [143]. The exogenous ligands for TLR9 are bacterial and viral CpG DNAs, whereas the internal ligands are antibody-chromatin complexes. The mechanism of action of the described transmembrane glycoprotein consists of detecting molecules associated with damage to the body’s own tissues (DAMPs), which come from inside the cell [144,145]. Although the role of Toll-like receptors in the pathogenesis of cardiovascular diseases is the subject of numerous scientific studies, it remains largely unexplained. Several years ago, it was discovered that tissue injury inducing the release of nuclear or mitochondrial CpG (5′—C—phosphate—G—3′) DNA can stimulate the TLR9 receptor as a DAMP [146,147,148,149,150,151,152,153,154,155,156,157,158]. Moreover, it has been suggested that Toll-like receptor 9, by sensing mitochondrial DNA that has been previously excised and transported to the cytosol, may play a “gatekeeper” function at the border between the vascular system and the immune system [149].
Recent studies have highlighted the importance of mitochondrial DNA production and subsequent inflammation, moderated by TLR9, as key factors in the pathogenesis of various cardiovascular diseases [150,151,152,153]. It has also been proven that episodes of acute left ventricular heart failure, caused by TLR9-mediated inflammatory reactions due to the release of mtDNA (mitochondrial DNA) damaged due to hemodynamic stress, have been observed in mice with transverse aortic stenosis [153]. It has been noted that circulating damaged mtDNA activates TLR9 and is partially responsible for the increase in blood pressure in rats with spontaneous hypertension [152]. More recent studies also emphasize the role of TLR9 activation and subsequent stimulation of IL-6 as a proinflammatory cytokine in the migration and proliferation of smooth muscle cells in the endothelium of pulmonary arteries in rats [154].
The TLR9 receptor has been identified as a pro-inflammatory receptor in relation to the pathogenesis of PAH, as it is responsible for activating NF-κB, IL-6, and subsequent inflammatory processes in animal models. In a 2021 study by Tomohito Ishikawa et al., it was hypothesized that mtDNA could activate TLR9, thereby stimulating the accumulation of inflammatory cells and IL-6 production in the lungs of rats, leading to the progression of pulmonary arterial hypertension. The study was conducted on 165 adult male rats (Sprague Dawley) weighing between 200 and 250 g, in which pulmonary arterial hypertension was induced with a single subcutaneous injection of monocrotaline adjusted for the animal’s body weight (60 mg/kg b.w.). Three types of study protocols were used: a preventive protocol, a short-term reversal protocol, and an extended reversal protocol. The tested rats were divided into two groups: one randomly administered a selective TLR9 antagonist, E6446 (provided by Eisai Inc., São Paulo, Brazil), and the other a non-selective quinoline agonist, most closely related to TLR9 among all Toll-like receptors, chloroquine (Sigma-Aldrich, Saint Louis, MO, USA). The study then assessed the effect of TLR9 activation on the value of hemodynamic parameters in the pulmonary circulation, vascular remodeling, and survival of rats [155]. In rats administered monocrotaline, a significant increase in the concentration of mtDNA markers in blood serum, activation of TLR9, and increased levels of IL-6 mRNA in lung tissue were observed approximately 14 days after injection of the active substance. Moreover, animals exposed to monocrotaline exhibited altered hemodynamic parameters in the pulmonary circulation. On the 21st day after the administration of the alkaloid, an increase in the value of the right ventricular systolic pressure, increased total peripheral resistance, and more intensive remodeling of pulmonary vessels, together with the activation of macrophages, were observed [155].
In the preventive protocol, administration (from −3 to 21 days after monocrotaline injection) of a selective or nonselective TLR9 inhibitor significantly reduced the increase in right ventricular systolic pressure and total pulmonary vascular resistance index, as well as vascular remodeling, and macrophage accumulation at day 21. These inhibitors also significantly reduced NF-κB activation and IL-6 mRNA levels to a similar extent. In the short-term reversal protocol, E6446 treatment (days 14–17 after monocrotaline injection) virtually normalized NF-κB activation and IL-6 mRNA levels and reduced macrophage accumulation. In an extended reversal protocol, E6446 treatment (days 14–24 after monocrotaline injection) reversed total pulmonary vascular resistance index and vascular remodeling and further increased survival rates in monocrotaline-exposed rats [155].
After completing the study, Ishikawa et al. formulated three essential conclusions. Firstly, activation of TLR9 participates in the pathogenesis of monocrotaline-induced pulmonary arterial hypertension in rats via activation of the NF-κB-IL-6 pathway. Secondly, long-term inhibition of TLR9 activity not only impedes progression but also reverses the pathophysiological phenomena observed in the lungs and reduces the expression of mRNA of the main proinflammatory interleukin in the pathogenesis of PAH, i.e., IL-6. Thirdly, reduced TLR9 activity leads to prolonged survival in rats exposed to the alkaloid described [81]. During the trial, it was also noted that vascular stress plays a key role in the pathogenesis and maintenance of perivascular inflammation, which is dependent on NF-κB-IL-6, as well as in the phenomenon of vascular remodeling in rats with PAH. In conclusion, the inhibition of TLR9 activity is a promising therapeutic target for the future in the treatment of this chronically progressive, incurable disease. However, the extrapolation of the studies conducted in the rat model to the human model of pulmonary arterial hypertension remains somewhat controversial, and further studies are needed to clarify the way in which hemodynamic stress induces mtDNA production in the lungs, and to determine the exact cell type stimulating TLR9 activation (Figure 4) [155,156].
The role of endothelial TLR9 in the development of PAH induced by hemoglobin (Hb)-induced tissue injury has also been studied [155]. It is widely believed that pulmonary vascular inflammation plays a crucial role in mediating the development and progression of PAH-related hemolytic anemia. However, the mechanisms by which hemoglobin located in the extracellular compartment promotes vascular inflammation, thereby leading to PAH, have not yet been fully explained. It is hypothesized that free cellular Hb may influence the progression of PAH due to its vigorous chemical reaction with NO, which results in reduced NO bioavailability and pulmonary artery vasoconstriction [157,158,159,160]. In clinical conditions, the release of hemoglobin as a result of hemolysis may occur in various diseases, including severe sepsis, hemolytic anemia [160,161,162,163], extracorporeal membrane oxygenation (ECMO), and chronic renal replacement therapy (CRRT) [161]. It is worth emphasizing that excessive amounts of released hemoglobin and peroxides stimulate the local microenvironment, which is rich in DAMPs, thereby activating TLRs and potentially initiating or exacerbating the progression and clinical symptoms of PAH [162,163].
Loomis et al. [154] attempted to prove the hypothesis that hemoglobin-induced lipid peroxidation and subsequent endothelial cell injury are associated with the activation of TLR9 located in the endothelium, leading to IL-6-mediated proliferation of smooth muscle cells in the pulmonary arteries. The research group consisted of male and female mice weighing between 20 and 25 g, which were subjected to jugular vein cannulation and then underwent surgery. The test assessed the effect of hemoglobin on endothelial cells (mPAEC) and smooth muscle cells (mPASMC) of the pulmonary arteries of the studied animals. Substances inhibiting TLR9 and IL-6 were then used, as well as Hb and heme-binding proteins, i.e., haptoglobin and hemopexin. The role of Hb in vivo was also examined using myeloid differentiation cells typical of the primary response 88 and mice that do not express TLR9. In animals, IL-6 concentration was measured in peripheral blood serum, TLR9 expression was assessed based on the mRNA concentration for this receptor in plasma, and systolic pressure in the right heart chamber was measured by direct puncture of this cavity with the chest closed [154].
The study showed increased expression of IL-6 (1.75 ± 0.3-fold; p = 0.04) and a significant increase in TLR9 expression (60%; p = 0.01). In contrast, in mice lacking TLR9, a lower level of IL-6 expression was noted in the lung tissue compared to wild-type cohorts. It was also demonstrated that Hb in its oxidized form is responsible for inducing lipid peroxidation. In mice co-cultured with activated hemoglobin endothelium, significantly higher concentrations of nucleic acids, more intense multiplication of pulmonary artery smooth muscle cells, and significantly higher concentrations of lactate dehydrogenase in blood serum were observed. The study proved that the proliferative activity of mouse PASMC was increased (13 ± 1%; p = 0.01) during co-culture with activated Hb mPAEC (Figure 4) [154].
Summarizing, Hb stimulates lipid peroxidation of cell membranes, which may be the initial step in a locally limited inflammatory response mediated by TLR9, resulting in the presentation of an activated PASMC phenotype and ultimately leading to the development of pulmonary arterial hypertension. The process of lipid peroxidation leads to the release of nucleic acids by cell death or their excision from damaged mitochondrial DNA. Then, the phenomenon of acid endocytosis occurs, which activates TLR9 on adjacent endothelial cells. This leads to the upregulation of IL-6, promoting the proliferation and growth of SMC with a phenotype characteristic of pulmonary vascular disease. The toxicity of Hb and heme is attributed to their prooxidant properties, which cause endothelial cell damage and apoptosis by inducing the peroxidation process [154,164,165,166,167].

2.7. TLR10

TLR10 (CD290) is the least described receptor in medical literature in relation to all 10 receptors found in humans. It is known that chromosome 4 is the site where the genes encoding TLR10 are situated. This transmembrane glycoprotein is composed of 811 amino acids and 19 LRRs. Histologically, TLR10 is located outside the cell; however, its precise location remains a subject of research. It is most likely present on B lymphocytes, monocytes/macrophages, and intestinal epithelial cells. Based on amino acid homology, TLR10 belongs to the TLR2/TLR6/TLR10 subfamily of receptors. The exogenous ligands for TLR10 are triacylated lipopeptides, and induction of signal transduction from TLR10 occurs via the MyD88 protein. The endogenous ligands for TLR10 remain unknown [168,169,170,171,172,173,174,175,176]. Recently, it has been discovered that other potential ligands for TLR10 may include Borrelia burgdorferi, Listeria monocytogenes, and Helicobacter pylori LPS (TLR2/10), as well as HIV-1 gp41 and H1N1/H5N1 [177,178,179].
There are currently no scientific studies available on the potential involvement of TLR10 in cardiovascular diseases and its role in the pathogenesis of PAH. Hypothetically, since activation of the TLR10 receptor has anti-inflammatory effects, this receptor could play a protective role in the vascular system. Activation of TLR10 could therefore result in reduced endothelial cell damage, reduced proliferation, and reduced local inflammation in the vessels. Further clinical trials are undeniably needed in this area.

3. The Role of Viral Infections in the Development and Progression of Pulmonary Arterial Hypertension

Viral infections are increasingly recognized as a “second hit” that initiates or perpetuates cascades leading to PAH. In genetically or environmentally predisposed individuals, infections—including HIV-1, EBV, HCV, SARS-CoV-2, and reactivation of endogenous retroviruses—can affect the pulmonary vascular wall both directly and indirectly. Direct effects include dysfunction and apoptosis of endothelial cells (PAEC), endothelial-to-mesenchymal transition (EndMT), and smooth muscle cell remodeling (PASMC). Indirectly, viral PAMPs and secondary DAMPs (e.g., mtDNA, HMGB1) activate Toll-like receptors and their adaptors, switching the response from the protective TRIF-IFN-I axis (typical of TLR3) to the proinflammatory MyD88-NF-κB/IL-6 pathways—including TLR2/4/7/8/9. This results in persistent inflammation, oxidative stress, immunothrombosis, and disruption of the BMPR2-Smad axis, which together drive vascular remodeling and increased pulmonary resistance. Viral infections also modulate adaptive immunity, promoting Th17 polarization, T cell exhaustion, the activation of plasmacytoid dendritic cells (pDCs), and type I interferon production, as well as—in some phenotypes—the development of autoantibodies. Preclinical data suggest that TLR3 agonism—e.g., poly(I:C)—may be protective, whereas TLR7/8/9 activation enhances remodeling and autoimmunity. In the present chapter, we synthesize the epidemiological and mechanistic evidence linking individual viruses to PAH, map the virus-TLR-vessel interface, and assess translational implications: biomarkers (e.g., IFN-I/IL-6 signatures, circulating mtDNA), risk stratification, and therapeutic targets (i.e., antiviral therapies, TLR modulation, targeting the IL-6 axis, and restoring BMPR2 signaling) [35,37,180,181].

3.1. Epstein–Barr Virus (EBV) Infection

Epstein–Barr virus (EBV) is a lymphotropic herpesvirus that persists in the human body in a latent form, undergoing periodic reactivation. Its ability to infect and transform B lymphocytes, together with its capacity to maintain persistent latency, makes it one of the best-characterized oncogenic factors implicated in the pathogenesis of lymphomas and epithelial cancers [182,183,184,185]. Following primary infection of epithelial cells, EBV colonizes B lymphocytes, where its genome remains latent. The IFI16 protein enables the reactivation and long-term persistence of the virus in host cells. Although infection is typically asymptomatic, in immunocompromised individuals it promotes the development of tumors and the deregulation of the immune response [183,186,187].
In the context of PAH, EBV is considered a potential cofactor that initiates or exacerbates vascular remodeling. Viral reactivation leads to the release of viral PAMPs and secondary DAMPs, which activate TLR and inflammatory pathways in the vascular wall. This leads to the chronic activation of NF-κB, increased cytokine production—including IL-6, endothelial dysfunction, endothelial-to-mesenchymal transition (EndMT), the proliferation of PASMCs, and immunothrombotic phenomena, which are processes that lead to increased vascular resistance in the pulmonary circulation [188,189,190,191].
Observations from studies on idiopathic pulmonary fibrosis indicate a significant role of EBV in modulating inflammatory and fibrotic processes. Treatment with Ganciclovir has been shown to attenuate disease progression in patients with advanced disease [192]. Calabrese et al. identified the presence of herpesvirus genomes in 40% of the study population in lung biopsies of patients with idiopathic pulmonary fibrosis, with EBV accounting for 59.1% of the positive cases. The presence of the virus correlated with pulmonary arterial intimal thickening and increased expression of TGF-β, i.e., markers of remodeling and a proinflammatory vascular microarchitecture [193,194]. The collected data suggest that EBV may modulate the phenotype and dynamics of PAH, particularly in patients with concomitant interstitial lung disease or immunosuppression. This requires further translational research focused on:
  • Identification of target cells and TLR pathways activated by EBV in the lung.
  • Validation of biomarkers, such as plasma EBV DNA and IL-6/TGF-β signatures.
  • Assessment of the therapeutic potential of strategies combining antiviral treatment with modulation of the innate immune response and blocking the pro-remodeling cytokine and factor axes.
Therefore, EB appears to be a factor capable of modulating the course of PAH by influencing inflammation and pulmonary vascular remodeling, thereby opening new avenues for diagnosis and treatment [188,189,190,191,192,193,194].

3.2. Human Immunodeficiency Virus (HIV) Infection

PAH is a rare (approximately 0.5%) but serious complication of HIV infection [195,196,197]. The association of HIV and the development of pulmonary hypertension was first described in 1987 by Kim and Factor [198]. Given the high and still increasing prevalence of HIV infection—estimated at 40.8 million people worldwide according to WHO data from 2024—HIV may become one of the leading causes of pulmonary arterial hypertension globally [199].
Modern antiretroviral treatment based on HAART (highly active antiretroviral therapy) and the effective treatment of opportunistic infections significantly extend the survival period of HIV-infected patients [200,201,202]. As a result, late complications, including PAH, are being observed with increasing frequency [203]. The pathogenic mechanism underlying the development of pulmonary arterial hypertension in the course of HIV infection remains unclear. HIV and its associated proteins are not present in the vascular endothelium [35,204,205,206]. An indirect role of HIV in the pathogenesis of pulmonary hypertension has been postulated. HIV stimulates monocytes, macrophages, and lymphocytes to secrete proinflammatory cytokines associated with the development of idiopathic pulmonary hypertension: IL-1, IL-6, TNF, and platelet-derived growth factor [35,207,208]. Viral proteins—such as negative factor (Nef), Tat protein, and glycoprotein 120 (gp-120)—can initiate vascular oxidative stress, endothelial damage, and the proliferation and migration of smooth muscle cells, leading to the development of PAH [209,210,211,212].
No correlation has been demonstrated between the severity of pulmonary arterial hypertension and the severity of HIV infection, the degree of immunodeficiency, or the number of CD4+ T lymphocytes [213,214]. In lung biopsies with complex plexiform lesions, the presence of HIV was not detected. This observation suggests that the virus may also exert indirect effects, through the induction of inflammatory process and growth factors, thereby acting as a trigger mechanism in predisposed patients [35,215,216].

3.3. Hepatitis C Virus (HCV) Infection

Hepatitis C virus (HCV) belongs to the Flaviviridae family and is a small, enveloped, positive-strand RNA virus. In humans, it is responsible for the development of chronic hepatitis C, which in some cases leads to cirrhosis, liver failure, and hepatocellular carcinoma; it is also associated with some lymphomas [217,218,219]. According to data from 2022, HCV infection affects approximately 50 million people worldwide (0.6% of the adult population), with 1.5 million new cases annually. Despite the availability of effective antiviral therapies, mortality rates remain high, primarily due to complications of cirrhosis and HCC [220,221,222,223]. The pathophysiological links between HCV infection and the development of PAH are not clearly defined; however, experimental data indicate the involvement of nonstructural proteins of the virus. The NS5A protein has been shown to activate the STAT3 axis, a key mechanism that promotes pulmonary artery smooth muscle cell (PASMC) proliferation and disrupts endothelial homeostasis. In contrast, other NS3–NS5A/B proteins induce COX-2 expression and increase prostaglandin E2 production, thereby fostering a proinflammatory environment conducive to vascular remodeling [224,225,226,227,228,229]. These mechanisms may link chronic HCV infection with the activation of innate inflammatory responses, including the TLR–NF-κB–IL-6 axis, perpetuating the inflammatory-proliferative process in the pulmonary artery wall.
The introduction of direct-acting antiviral drugs (DAAs) has revolutionized the treatment of HCV. An example is sofosbuvir, a nucleotide inhibitor of HCV RNA polymerase, which effectively inhibits viral replication [230]. Interferon-α remains used in some therapeutic regimens. However, clinical reports indicate that both interferon-α and sofosbuvir may induce or exacerbate PAH in some cases. A study by Savale et al. demonstrated that interferon-α-induced PAH has a relatively mild clinical and hemodynamic phenotype, characterized by low right atrial pressure, a moderate increase in pulmonary vascular resistance, normal cardiac output, and only slightly reduced exercise tolerance [227]. Renard et al. described cases of PAH development or progression during sofosbuvir treatment, which was attributed to a rapid decrease in vasodilatory mediators following HCV eradication and secondary activation of the STAT3 axis, potentially unmasking latent or previously stable PAH [224]. HCV infection may therefore promote the development of PAH by activating the STAT3 and COX-2/PGE2 pathways, leading to increased PASMC proliferation, endothelial dysfunction, and chronic inflammation. At the same time, selected antiviral therapies may modify the hemodynamic course in specific situations. This requires further translational research aimed at identifying biomarkers predisposing to PAH in HCV, monitoring patients during antiviral treatment, and developing therapeutic strategies that simultaneously control HCV infection and minimize the risk of pulmonary circulation disorders [224,225,226,227,228,229].

3.4. Human Endogenous Retrovirus K (HERV-K) Infection

Human endogenous retrovirus K (HERV-K) is one of the most active and conserved members of the HERV family, which are remnants of ancient retroviral infections integrated into the human genome and transmitted vertically through successive generations [231,232,233,234,235]. Although HERVs lack classical replication capacity, they retain the potential for transcription and translation, and their expression can be reactivated by cellular stress, epigenetic disruption, or exogenous infections [236,237,238]. HERV-K has been associated with testicular malignancies, melanoma, and neurodegenerative diseases, among others, indicating its pathogenic potential [239,240,241]. In the context of PAH, the observations of Saito et al. are significant. They demonstrated increased HERV copy numbers, particularly HERV-K, in the lungs of patients with PAH. Analyses also revealed increased expression of transcripts encoding HERV-K dUTPase and HERV-K(II) isoforms in the absence of detectable exogenous virus infection [242]. Mechanistically, recombinant HERV-K dUTPase induced expression of the SAMHD1 protein, which regulates deoxynucleotide metabolism and participates in the antiviral response, in both monocytes and PAECs. This, in turn, resulted in pronounced proinflammatory activation of monocytes, cytokine expression, and endothelial cell apoptosis, directly promoting endothelial dysfunction and initiating vascular remodeling [242].
In animal models, administration of HERV-K dUTPase to rats resulted in the development of a PAH phenotype, characterized by morphological and inflammatory changes in the pulmonary vessels and hemodynamic disturbances, including increased pulmonary arterial pressure [242]. These results suggest that reactivation of endogenous HERV-K sequences may itself generate PAH-like pathology, independent of exogenous virus infection. The collected data indicate that HERV-K may influence the development of PAH through synergistic mechanisms: direct endothelial damage associated with PAEC apoptosis, chronic inflammatory activation of monocytes, and the perpetuation of a proinflammatory-proliferative microenvironment, as well as potential interactions with innate immune sensors such as TLRs and NLRs, which may recognize HERV transcription products as danger signals. This results in increased NF-κB signaling, IL-6 induction, and activation of pathways leading to PASMC proliferation and immunothrombosis. In light of these observations, HERV-K appears not only as a biomarker of active pulmonary vascular remodeling but also as a potential translational target. Directions for further research should include strategies to limit HERV transcription (e.g., epigenetic modulation), neutralization of their protein products, or inhibition of secondary inflammatory pathways induced by HERV-K reactivation. Although knowledge in this area is still limited, available evidence indicates that HERV-K may be an important endogenous factor modulating the pathophysiology of PAH [243,244,245,246,247].

3.5. SARS-CoV-2 Infection

The COVID-19 pandemic caused by SARS-CoV-2 has compounded the already known susceptibility of the pulmonary vascular bed to inflammatory and thrombotic injuries and revealed a particular vulnerability of patients with previously diagnosed PAH/PH. This population has been reported to exhibit a higher risk of hospitalization, right ventricular decompensation, and death, with in-hospital mortality rates reaching 25–43% in selected registries [247,248,249,250,251,252,253,254]. Increasing evidence also points to persistent vascular sequelae following COVID-19, including secondary forms of pulmonary hypertension in convalescent patients, especially after severe pneumonia or ARDS [255,256,257,258]. At a mechanistic level, SARS-CoV-2 engages innate immune pathways relevant to pulmonary vascular remodeling: viral structural proteins (E and—in specific experimental systems—S) activate TLR2, while S can also trigger TLR4 in a context-dependent manner; in parallel, viral RNA activates endosomal TLR3/7/8. These inputs converge on MyD88→NF-κB and TRIF→IRF3/7 cascades, driving IL-6/TNF and type-I interferon responses, oxidative stress, and secondary HIF-1α activation—signaling that sustains a pro-inflammatory/pro-proliferative vascular phenotype [259,260,261,262]. SARS-CoV-2–induced endothelial dysfunction is well documented: endotheliitis, barrier disruption, immunothrombosis, and angiogenesis culminate in thrombotic microangiopathy and increased right-ventricular afterload; ACE2 down-regulation further contributes to RAAS imbalance, vasoconstriction, and capillary leak—hallmarks overlapping with PH/PAH pathogenesis and initiating EndMT-driven intimal/adventitial remodeling [263,264]. Notably, despite this overlap, direct experimental proof that SARS-CoV-2-evoked TLR signaling causally perturbs canonical PAH pathways (e.g., BMPR2) remains limited; conversely, TLR3 activation can upregulate BMPR2 and ameliorate experimental PH, whereas TLR4 is implicated in pulmonary vascular homeostasis and its loss favors remodeling—underscoring context dependence [19,22].
The basis for these phenomena is extensive damage to the pulmonary endothelium, accompanied by endotheliitis, immunothrombosis, and vascular wall remodeling—processes crucial for the pathogenesis of PH/PAH [188,265,266,267]. SARS-CoV-2 enters cells via ACE2, which promotes dysregulation of the RAAS axis (ACE2 deficiency and relative predominance of angiotensin II), vasoconstriction, capillary leakage, and a prothrombotic state [268,269,270]. In parallel, thrombotic microangiopathy develops, characterized by generalized microvascular thrombosis, which impairs perfusion and increases right ventricular afterload [271,272,273,274]. Histopathological and imaging studies confirm generalized endothelial activation, inflammatory infiltrates, and vascular barrier damage, which initiate EndMT and remodeling of the intima and adventitia [275,276,277,278]. The molecular mechanisms involve innate immune pathways (TLR activation by viral PAMPs and secondary DAMPs), NF-κB/AP-1 cascades with IL-6/TNF-α production, oxidative stress, and the secondary activation of HIF-1α—a constellation of signals that perpetuates the proinflammatory and proliferative phenotype of the pulmonary vasculature [260,279,280,281,282].
The acute phase may result in the development or exacerbation of chronic PH. Phenotypes secondary to interstitial lung disease and post-ARDS fibrosis, as well as forms associated with chronic hypoxemia and persistent impairment of vascular reserve, are observed after COVID-19. In some echocardiographic series, features of PH were detected in up to 70% of patients several months after recovery, particularly in older individuals with obesity and prolonged desaturation [257,282,283,284]. In patients with baseline PAH/CTEPH, SARS-CoV-2 infection more often leads to acute right ventricular failure and worse short-term outcomes, reflecting the cumulative effects of microthrombosis, vasoconstriction, and increased pulmonary resistance [285,286,287,288,289].
From a therapeutic perspective, continued PAH-specific therapy (endothelin receptor antagonists, PDE-5 inhibitors, analogs/prostanoids) is recommended during the acute phase of COVID-19 to reduce the risk of decompensation. Inpatient management should be individualized and include oxygen therapy, diuretic therapy, and hemodynamic support according to the patient’s profile [290,291,292]. The issue of reinitiating or intensifying PAH-specific therapy after infection remains ambiguous. It requires evaluation in prospective studies, as do algorithms for monitoring convalescent patients for the development of new-onset PH (biomarkers of RV overload, echocardiogram, and eligibility for hemodynamic testing) [293,294]. In summary, SARS-CoV-2 represents both an acute stressor on the lung endothelium and a potential long-term driver of pulmonary vascular disease; mechanistic, translational, and clinical studies are needed to precisely define the risk trajectory and optimize therapeutic and surveillance strategies in this vulnerable population.

4. Conclusions

The collected evidence indicates that the balance of TLRs largely determines the course and prognosis of PAH—specifically, the balance between the proinflammatory MyD88–NF-κB/IL-6 axis and the protective TRIF–IFN–α axis. Activation of most receptors (TLR1/2, 4, 5/6, 7/8, 9) amplifies inflammation, oxidative stress, immunothrombosis, and vascular wall remodeling, thereby increasing pulmonary vascular resistance. In contrast, TLR3 exerts an opposing effect by stabilizing the endothelium and inhibiting remodeling, and its dysfunction is associated with more severe disease. Viral infections and reactivations (HIV-1, EBV, HCV, HERV-K, SARS-CoV-2) further tilt this balance toward pro-remodeling cascades in genetically or immunologically predisposed individuals. Taken together, these observations suggest that more effective therapeutic strategies should go beyond vasodilation alone and target modulation of the innate immunity of the vascular wall—enhancing TLR3/TRIF signaling and selectively suppressing the overactive MyD88–NF-κB/IL-6 axis (including through IL-6 modulation), combined with antiviral therapy if necessary. Precise patient selection based on simple, clinically available indicators (e.g., IFN-I/IL-6 signatures or markers of mitochondrial damage) and methodological caution will be crucial, as non-selective TLR blockade may carry a risk of infection. This therapeutic approach, based on TLR and viral biology, offers a real chance to slow vascular remodeling and improve PAH treatment outcomes.

Author Contributions

Conceptualization, A.S., M.K. (Martyna Krysa), P.M., E.G., T.U. and M.K. (Maciej Krasiński); M.G., M.R.-H., W.T.-P. and M.T.; methodology, A.S., M.K. (Martyna Krysa) and P.M.; software, A.S., M.K. (Martyna Krysa) and P.M.; validation, E.G., T.U., M.K. (Martyna Krysa) and M.T.; formal analysis, A.S., M.K. (Martyna Krysa) and P.M.; investigation, A.S., M.K.(Martyna Krysa) and P.M.; resources, E.G., T.U., M.K. (Maciej Krasiński) and M.T.; data curation, E.G. and M.T.; writing—original draft preparation, A.S., M.K. (Martyna Krysa), P.M., E.G., T.U., M.K. (Maciej Krasiński), M.G., M.R.-H., W.T.-P. and M.T.; writing—review and editing, A.S., M.K. (Martyna Krysa) and P.M.; visualization, P.M.; supervision, E.G. and M.T.; project administration, E.G. and M.T.; funding acquisition, E.G., T.U. and M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Medical University of Lublin DS640. The APC was funded by Poznan University of Medical Sciences.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

This article is a review and does not involve the generation or analysis of new primary data. All data supporting the statements and conclusions of this work are contained within the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
AbbreviationExpansion
ACE2angiotensin-converting enzyme 2
ANAantinuclear antibodies
AP-1Activator Protein-1
ARDSacute respiratory distress syndrome
BMPR2bone morphogenetic protein receptor type 2
CD281–CD290CD designations corresponding to TLR1–TLR10
CNScentral nervous system
COX-2cyclooxygenase-2
CpG DNAunmethylated CpG motif–containing DNA
CpG ODNCpG oligodeoxynucleotide
CRRTcontinuous renal replacement therapy
CTEPHchronic thromboembolic pulmonary hypertension
DAAdirect-acting antiviral
DAMPdamage-associated molecular pattern
dsDNAdouble-stranded DNA
dsRNAdouble-stranded RNA
EBVEpstein–Barr virus
ECendothelial cell(s)
ECMextracellular matrix
ECMOextracorporeal membrane oxygenation
E6446selective TLR9 antagonist
EndMTendothelial-to-mesenchymal transition
ET-1endothelin-1
GM-CSFgranulocyte–macrophage colony-stimulating factor
gp120HIV-1 envelope glycoprotein 120
gp96endoplasmin/GRP94 (heat-shock protein HSP90B1)
HAARThighly active antiretroviral therapy
Hbhemoglobin
HCChepatocellular carcinoma
HCVhepatitis C virus
HERV-Khuman endogenous retrovirus K
HIF-1αhypoxia-inducible factor-1 alpha
HIV-1human immunodeficiency virus type 1
HMGB1high-mobility group box 1
HSPheat-shock protein
Hsp70heat-shock protein 70
ICAM-1intercellular adhesion molecule-1
Id1inhibitor of DNA binding 1
IFNinterferon (general)
IFN-Itype I interferons
IFN-αinterferon-alpha
IFN-βinterferon-beta
IFN-γinterferon-gamma
IFNA1interferon alpha 1 gene
IFNB1interferon beta 1 gene
IL-1interleukin-1
IL-6interleukin-6
IL-8interleukin-8
IRAKinterleukin-1 receptor–associated kinase
IRF3interferon regulatory factor 3
LDHlactate dehydrogenase
LL-37human cathelicidin antimicrobial peptide
LPSlipopolysaccharide
MALP-2macrophage-activating lipopeptide-2 (TLR2/6 agonist)
miRNAmicroRNA
MRImagnetic resonance imaging
mtDNAmitochondrial DNA
MyD88myeloid differentiation primary response 88
NOX (NADPH oxidase)nicotinamide adenine dinucleotide phosphate oxidase system
NETsneutrophil extracellular traps
NF-κBnuclear factor kappa-B
NLRNOD-like receptor
NOnitric oxide
Nox1NOX family isoform 1
Nox4NOX family isoform 4
NS3nonstructural protein 3 (HCV)
NS5Anonstructural protein 5A (HCV)
NS5Bnonstructural protein 5B (HCV)
NefHIV-1 negative factor protein
PAMPpathogen-associated molecular pattern
PAECpulmonary artery endothelial cell(s)
PAHpulmonary arterial hypertension
PASMCpulmonary arterial smooth muscle cell(s)
PDE-5phosphodiesterase type 5
PDGFplatelet-derived growth factor
PGE2prostaglandin E2
PHpulmonary hypertension
poly(I:C)polyinosinic:polycytidylic acid (synthetic dsRNA; TLR3 agonist)
p53tumor protein p53
pDCplasmacytoid dendritic cell(s)
RAASrenin–angiotensin–aldosterone system
R848resiquimod (TLR7/8 agonist)
R840imidazoquinoline TLR7/8 agonist (experimental)
RANTES (CCL5)Regulated upon Activation, Normal T-cell Expressed and Secreted
RIPK3receptor-interacting protein kinase 3
RNA-seqRNA sequencing
ROSreactive oxygen species
RVright ventricle
RVEFright ventricular ejection fraction
RVSPright ventricular systolic pressure
SARS-CoV-2severe acute respiratory syndrome coronavirus 2
SLEsystemic lupus erythematosus
Smad1/5/8SMAD proteins downstream of BMP/TGF-β signaling
SMCsmooth muscle cell(s)
ssRNAsingle-stranded RNA
STAT3signal transducer and activator of transcription 3
SScsystemic sclerosis
SU5416VEGFR inhibitor used to induce experimental PAH
TatHIV trans-activator of transcription protein
TGF-βtransforming growth factor-beta
Th17T helper 17 cells
TIRToll/Interleukin-1 receptor (domain)
TLRToll-like receptor(s)
TMAOtrimethylamine-N-oxide
TNFtumor necrosis factor
TNF-αtumor necrosis factor-alpha
TPVRtotal pulmonary vascular resistance (index)
TRIFTIR-domain–containing adapter-inducing interferon-β
Tregregulatory T cell(s)
TXA2thromboxane A2
VCAM-1vascular cell adhesion molecule-1
VEGFvascular endothelial growth factor
VEGFRvascular endothelial growth factor receptor
WHOWorld Health Organization
mPAPmean pulmonary arterial pressure

References

  1. Montani, D.; Günther, S.; Dorfmüller, P.; Perros, F.; Girerd, B.; Garcia, G.; Jaïs, X.; Savale, L.; Artaud-Macari, E.; Price, L.C.; et al. Pulmonary Arterial Hypertension. Orphanet J. Rare Dis. 2013, 8, 97. [Google Scholar] [CrossRef]
  2. Boucly, A.; Gerges, C.; Savale, L.; Jaïs, X.; Jevnikar, M.; Montani, D.; Sitbon, O.; Humbert, M. Pulmonary Arterial Hypertension. Presse Med. 2023, 52, 104168. [Google Scholar] [CrossRef] [PubMed]
  3. Manek, G.; Bhardwaj, A. Pulmonary Hypertension. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  4. Rosenkranz, S.; Howard, L.S.; Gomberg-Maitland, M.; Hoeper, M.M. Systemic Consequences of Pulmonary Hypertension and Right-Sided Heart Failure. Circulation 2020, 141, 678–693. [Google Scholar] [CrossRef] [PubMed]
  5. Funk-Hilsdorf, T.C.; Behrens, F.; Grune, J.; Simmons, S. Dysregulated Immunity in Pulmonary Hypertension: From Companion to Composer. Front. Physiol. 2022, 13, 819145. [Google Scholar] [CrossRef] [PubMed]
  6. Ge, Q.; Zhang, T.; Yu, J.; Lu, X.; Xiao, S.; Zhang, T.; Qing, T.; Xiao, Z.; Zeng, L.; Luo, L. A New Perspective on Targeting Pulmonary Arterial Hypertension: Programmed Cell Death Pathways (Autophagy, Pyroptosis, Ferroptosis). Biomed. Pharmacother. 2024, 181, 117706. [Google Scholar] [CrossRef]
  7. DeVaughn, H.; Rich, H.E.; Shadid, A.; Vaidya, P.K.; Doursout, M.-F.; Shivshankar, P. Complement Immune System in Pulmonary Hypertension-Cooperating Roles of Circadian Rhythmicity in Complement-Mediated Vascular Pathology. Int. J. Mol. Sci. 2024, 25, 12823. [Google Scholar] [CrossRef]
  8. Zhang, Y.; Li, X.; Li, S.; Zhou, Y.; Zhang, T.; Sun, L. Immunotherapy for Pulmonary Arterial Hypertension: From the Pathogenesis to Clinical Management. Int. J. Mol. Sci. 2024, 25, 8427. [Google Scholar] [CrossRef]
  9. Goulopoulou, S.; McCarthy, C.G.; Webb, R.C. Toll-like Receptors in the Vascular System: Sensing the Dangers Within. Pharmacol. Rev. 2016, 68, 142–167. [Google Scholar] [CrossRef]
  10. Li, D.; Wu, M. Pattern Recognition Receptors in Health and Diseases. Signal Transduct. Target. Ther. 2021, 6, 291. [Google Scholar] [CrossRef]
  11. Wang, R.; Lan, C.; Benlagha, K.; Camara, N.O.S.; Miller, H.; Kubo, M.; Heegaard, S.; Lee, P.; Yang, L.; Forsman, H.; et al. The Interaction of Innate Immune and Adaptive Immune System. MedComm 2024, 5, e714. [Google Scholar] [CrossRef]
  12. Chen, Y.-H.; Wu, K.-H.; Wu, H.-P. Unraveling the Complexities of Toll-like Receptors: From Molecular Mechanisms to Clinical Applications. Int. J. Mol. Sci. 2024, 25, 5037. [Google Scholar] [CrossRef] [PubMed]
  13. Chen, R.; Zou, J.; Chen, J.; Zhong, X.; Kang, R.; Tang, D. Pattern Recognition Receptors: Function, Regulation and Therapeutic Potential. Signal Transduct. Target. Ther. 2025, 10, 216. [Google Scholar] [CrossRef] [PubMed]
  14. Tanekhy, M. The Role of Toll-like Receptors in Innate Immunity and Infectious Diseases of Teleost. Aquac. Res. 2016, 47, 1369–1391. [Google Scholar] [CrossRef]
  15. Bhagwani, A.; Thompson, A.A.R.; Farkas, L. When Innate Immunity Meets Angiogenesis—The Role of Toll-Like Receptors in Endothelial Cells and Pulmonary Hypertension. Front. Med. 2020, 7, 352. [Google Scholar] [CrossRef] [PubMed]
  16. Arora, S.; Ahmad, S.; Irshad, R.; Goyal, Y.; Rafat, S.; Siddiqui, N.; Dev, K.; Husain, M.; Ali, S.; Mohan, A.; et al. TLRs in Pulmonary Diseases. Life Sci. 2019, 233, 116671. [Google Scholar] [CrossRef]
  17. George, P.M.; Badiger, R.; Shao, D.; Edwards, M.R.; Wort, S.J.; Paul-Clark, M.J.; Mitchell, J.A. Viral Toll Like Receptor Activation of Pulmonary Vascular Smooth Muscle Cells Results in Endothelin-1 Generation; Relevance to Pathogenesis of Pulmonary Arterial Hypertension. Biochem. Biophys. Res. Commun. 2012, 426, 486–491. [Google Scholar] [CrossRef]
  18. Lafferty, E.I.; Qureshi, S.T.; Schnare, M. The Role of Toll-like Receptors in Acute and Chronic Lung Inflammation. J. Inflamm. 2010, 7, 57. [Google Scholar] [CrossRef]
  19. Ma, L.; Ambalavanan, N.; Liu, H.; Sun, Y.; Jhala, N.; Bradley, W.E.; Dell’Italia, L.J.; Michalek, S.; Wu, H.; Steele, C.; et al. TLR4 Regulates Pulmonary Vascular Homeostasis and Remodeling via Redox Signaling. Front. Biosci. 2016, 21, 397–409. [Google Scholar] [CrossRef]
  20. Pullamsetti, S.S.; Savai, R.; Seeger, W.; Goncharova, E.A. Translational Advances in the Field of Pulmonary Hypertension.From Cancer Biology to New Pulmonary Arterial Hypertension Therapeutics. Targeting Cell Growth and Proliferation Signaling Hubs. Am. J. Respir. Crit. Care Med. 2017, 195, 425–437. [Google Scholar] [CrossRef]
  21. Farkas, D.; Thompson, A.A.R.; Bhagwani, A.R.; Hultman, S.; Ji, H.; Kotha, N.; Farr, G.; Arnold, N.D.; Braithwaite, A.; Casbolt, H.; et al. Toll-like Receptor 3 is a Therapeutic Target for Pulmonary Hypertension. Am. J. Respir. Crit. Care Med. 2019, 199, 199–210. [Google Scholar] [CrossRef]
  22. Bhagwani, A.R.; Ali, M.; Piper, B.; Liu, M.; Hudson, J.; Kelly, N.; Bogamuwa, S.; Yang, H.; Londino, J.D.; Bednash, J.S.; et al. A P53-TLR3 Axis Ameliorates Pulmonary Hypertension by Inducing BMPR2 via IRF3. iScience 2023, 26, 105935. [Google Scholar] [CrossRef]
  23. Willcocks, S.; Offord, V.; Seyfert, H.-M.; Coffey, T.J.; Werling, D. Species-Specific PAMP Recognition by TLR2 and Evidence for Species-Restricted Interaction with Dectin-1. J. Leukoc. Biol. 2013, 94, 449–458. [Google Scholar] [CrossRef]
  24. Piccinini, A.M.; Midwood, K.S. DAMPening Inflammation by Modulating TLR Signalling. Mediat. Inflamm. 2010, 2010, 672395. [Google Scholar] [CrossRef]
  25. Ma, M.; Jiang, W.; Zhou, R. DAMPs and DAMP-Sensing Receptors in Inflammation and Diseases. Immunity 2024, 57, 752–771. [Google Scholar] [CrossRef]
  26. Kawai, T.; Ikegawa, M.; Ori, D.; Akira, S. Decoding Toll-like Receptors: Recent Insights and Perspectives in Innate Immunity. Immunity 2024, 57, 649–673. [Google Scholar] [CrossRef] [PubMed]
  27. Curtin, J.F.; Liu, N.; Candolfi, M.; Xiong, W.; Assi, H.; Yagiz, K.; Edwards, M.R.; Michelsen, K.S.; Kroeger, K.M.; Liu, C.; et al. HMGB1 Mediates Endogenous TLR2 Activation and Brain Tumor Regression. PLoS Med. 2009, 6, e1000010. [Google Scholar] [CrossRef] [PubMed]
  28. Hsieh, M.L.; Nishizaki, D.; Adashek, J.J.; Kato, S.; Kurzrock, R. Toll-like Receptor 3: A Double-Edged Sword. Biomark. Res. 2025, 13, 32. [Google Scholar] [CrossRef] [PubMed]
  29. Chen, B.; Di, B. Endogenous Ligands of TLR4 in Microglia: Potential Targets for Related Neurological Diseases. Curr. Drug Targets 2024, 25, 953–970. [Google Scholar] [CrossRef]
  30. Kim, J.; Ha, S.; Son, M.; Kim, D.; Kim, M.-J.; Kim, B.; Kim, D.; Chung, H.Y.; Chung, K.W. TLR7 Activation by miR-21 Promotes Renal Fibrosis by Activating the pro-Inflammatory Signaling Pathway in Tubule Epithelial Cells. Cell Commun. Signal. 2023, 21, 215. [Google Scholar] [CrossRef]
  31. Fabbri, M.; Paone, A.; Calore, F.; Galli, R.; Gaudio, E.; Santhanam, R.; Lovat, F.; Fadda, P.; Mao, C.; Nuovo, G.J.; et al. MicroRNAs Bind to Toll-like Receptors to Induce Prometastatic Inflammatory Response. Proc. Natl. Acad. Sci. USA 2012, 109, E2110–E2116. [Google Scholar] [CrossRef]
  32. Rebetz, J.; Cederholm, H.; McGauran, D.; Moore, E.; Chi, C.; Tabak, K.; Allhorn, M.; Olsson, M.L.; Egesten, A.; Semple, J.W.; et al. Mitochondrial DNA via Recipient TLR9 Acts as a Potent First-Hit in Murine Transfusion-Related Acute Lung Injury (TRALI). Blood 2025, 146, 2479–2490. [Google Scholar] [CrossRef]
  33. Rodrigues, C.R.; Balachandran, Y.; Aulakh, G.K.; Singh, B. TLR10: An Intriguing Toll-Like Receptor with Many Unanswered Questions. J. Innate Immun. 2024, 16, 96–104. [Google Scholar] [CrossRef]
  34. Almodovar, S.; Hsue, P.Y.; Morelli, J.; Huang, L.; Flores, S.C. Pathogenesis of HIV-Associated Pulmonary Hypertension. Proc. Am. Thorac. Soc. 2011, 8, 308–312. [Google Scholar] [CrossRef] [PubMed]
  35. Butrous, G. Global Landscape of Infection-Induced Pulmonary Hypertension. Infect. Dis. Rep. 2025, 17, 35. [Google Scholar] [CrossRef] [PubMed]
  36. Cool, C.D.; Voelkel, N.F.; Bull, T. Viral Infection and Pulmonary Hypertension: Is There an Association? Expert Rev. Respir. Med. 2011, 5, 207–216. [Google Scholar] [CrossRef] [PubMed]
  37. Castillo-Galán, S.; Parra, V.; Cuenca, J. Unraveling the Pathogenesis of Viral-Induced Pulmonary Arterial Hypertension: Possible New Therapeutic Avenues with Mesenchymal Stromal Cells and Their Derivatives. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2025, 1871, 167519. [Google Scholar] [CrossRef]
  38. Wang, D.; Gomes, M.T.; Mo, Y.; Prohaska, C.C.; Zhang, L.; Chelvanambi, S.; Clauss, M.A.; Zhang, D.; Machado, R.F.; Gao, M.; et al. Human Endogenous Retrovirus, SARS-CoV-2, and HIV Promote PAH via Inflammation and Growth Stimulation. Int. J. Mol. Sci. 2023, 24, 7472. [Google Scholar] [CrossRef]
  39. Nie, L.; Cai, S.-Y.; Shao, J.-Z.; Chen, J. Toll-Like Receptors, Associated Biological Roles, and Signaling Networks in Non-Mammals. Front. Immunol. 2018, 9, 1523. [Google Scholar] [CrossRef]
  40. Mertowski, S.; Lipa, P.; Morawska, I.; Niedźwiedzka-Rystwej, P.; Bębnowska, D.; Hrynkiewicz, R.; Grywalska, E.; Roliński, J.; Załuska, W. Toll-Like Receptor as a Potential Biomarker in Renal Diseases. Int. J. Mol. Sci. 2020, 21, 6712. [Google Scholar] [CrossRef]
  41. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q15399/entry (accessed on 11 September 2025).
  42. Xiao, G.; Zhuang, W.; Wang, T.; Lian, G.; Luo, L.; Ye, C.; Wang, H.; Xie, L. Transcriptomic Analysis Identifies Toll-like and Nod-like Pathways and Necroptosis in Pulmonary Arterial Hypertension. J. Cell Mol. Med. 2020, 24, 11409–11421. [Google Scholar] [CrossRef]
  43. Broen, J.C.A.; Bossini-Castillo, L.; van Bon, L.; Vonk, M.C.; Knaapen, H.; Beretta, L.; Rueda, B.; Hesselstrand, R.; Herrick, A.; Worthington, J.; et al. A Rare Polymorphism in the Gene for Toll-like Receptor 2 is Associated with Systemic Sclerosis Phenotype and Increases the Production of Inflammatory Mediators. Arthritis Rheum. 2012, 64, 264–271. [Google Scholar] [CrossRef] [PubMed]
  44. Takeda, K.; Akira, S. Toll-like Receptors. Curr. Protoc. Immunol. 2015, 109, 14.12.1–14.12.10. [Google Scholar] [CrossRef] [PubMed]
  45. de Kivit, S.; Tobin, M.C.; Forsyth, C.B.; Keshavarzian, A.; Landay, A.L. Regulation of Intestinal Immune Responses through TLR Activation: Implications for Pro- and Prebiotics. Front. Immunol. 2014, 5, 60. [Google Scholar] [CrossRef] [PubMed]
  46. UniProt. Available online: https://www.uniprot.org/uniprotkb/O60603/entry (accessed on 11 September 2025).
  47. Jin, M.S.; Kim, S.E.; Heo, J.Y.; Lee, M.E.; Kim, H.M.; Paik, S.-G.; Lee, H.; Lee, J.-O. Crystal Structure of the TLR1-TLR2 Heterodimer Induced by Binding of a Tri-Acylated Lipopeptide. Cell 2007, 130, 1071–1082. [Google Scholar] [CrossRef]
  48. Sabroe, I.; Dower, S.K.; Whyte, M.K.B. The Role of Toll-like Receptors in the Regulation of Neutrophil Migration, Activation, and Apoptosis. Clin. Infect. Dis. 2005, 41 (Suppl. S7), S421–S426. [Google Scholar] [CrossRef]
  49. Sallusto, F.; Lanzavecchia, A. The Instructive Role of Dendritic Cells on T-Cell Responses. Arthritis Res. 2002, 4 (Suppl. S3), S127–S132. [Google Scholar] [CrossRef]
  50. Ma, D.; Wang, X.; Liu, X.; Li, Z.; Liu, J.; Cao, J.; Wang, G.; Guo, Y.; Zhao, S. Macrophage Infiltration Initiates RIP3/MLKL-Dependent Necroptosis in Paclitaxel-Induced Neuropathic Pain. Mediat. Inflamm. 2022, 2022, 1567210. [Google Scholar] [CrossRef]
  51. Lian, G.; You, J.; Lin, W.; Gao, G.; Xu, C.; Wang, H.; Luo, L. Bioinformatics Analysis of the Immune Cell Infiltration Characteristics and Correlation with Crucial Diagnostic Markers in Pulmonary Arterial Hypertension. BMC Pulm. Med. 2023, 23, 300. [Google Scholar] [CrossRef]
  52. Adu-Amankwaah, J.; Shi, Y.; Song, H.; Ma, Y.; Liu, J.; Wang, H.; Yuan, J.; Sun, K.; Hu, Q.; Tan, R. Signaling Pathways and Targeted Therapy for Pulmonary Hypertension. Signal Transduct. Target. Ther. 2025, 10, 207. [Google Scholar] [CrossRef]
  53. Wang, J.; Uddin, M.N.; Wang, R.; Gong, Y.; Wu, Y. Comprehensive Analysis and Validation of Novel Immune and Vascular Remodeling Related Genes Signature Associated with Drug Interactions in Pulmonary Arterial Hypertension. Front. Genet. 2022, 13, 922213. [Google Scholar] [CrossRef]
  54. UniProt. Available online: https://www.uniprot.org/uniprotkb/O15455/entry (accessed on 11 September 2025).
  55. Kim, J.H.; Song, D.H.; Youn, S.-J.; Kim, J.W.; Cho, G.; Kim, S.C.; Lee, H.; Jin, M.S.; Lee, J.-O. Crystal Structures of Mono- and Bi-Specific Diabodies and Reduction of Their Structural Flexibility by Introduction of Disulfide Bridges at the Fv Interface. Sci. Rep. 2016, 6, 34515. [Google Scholar] [CrossRef]
  56. Jiang, Z.; Mak, T.W.; Sen, G.; Li, X. Toll-like Receptor 3-Mediated Activation of NF-kappaB and IRF3 Diverges at Toll-IL-1 Receptor Domain-Containing Adapter Inducing IFN-Beta. Proc. Natl. Acad. Sci. USA 2004, 101, 3533–3538. [Google Scholar] [CrossRef]
  57. Chen, Y.; Lin, J.; Zhao, Y.; Ma, X.; Yi, H. Toll-like Receptor 3 (TLR3) Regulation Mechanisms and Roles in Antiviral Innate Immune Responses. J. Zhejiang Univ. Sci. B 2021, 22, 609–632. [Google Scholar] [CrossRef]
  58. Han, B.; Zhang, C.; Wang, X.; Song, H.; Zhang, L.; Li, T.; He, J.; Zhao, H. The Functional Mechanisms of Toll-Like Receptor 3 and Its Implications in Digestive System Tumors. FBL 2023, 28, 297. [Google Scholar] [CrossRef]
  59. Kawai, T.; Akira, S. TLR Signaling. Cell Death Differ. 2006, 13, 816–825. [Google Scholar] [CrossRef]
  60. Li, B.; Xia, Y.; Hu, B. Infection and Atherosclerosis: TLR-Dependent Pathways. Cell Mol. Life Sci. 2020, 77, 2751–2769. [Google Scholar] [CrossRef] [PubMed]
  61. Ishibashi, M.; Sayers, S.; D’Armiento, J.M.; Tall, A.R.; Welch, C.L. TLR3 Deficiency Protects against Collagen Degradation and Medial Destruction in Murine Atherosclerotic Plaques. Atherosclerosis 2013, 229, 52–61. [Google Scholar] [CrossRef]
  62. Pan, L.; Zhu, W.; Li, Y.; Xu, X.; Guo, L.; Lu, Q.; Wang, J. Astrocytic Toll-Like Receptor 3 is Associated with Ischemic Preconditioning- Induced Protection against Brain Ischemia in Rodents. PLoS ONE 2014, 9, e99526. [Google Scholar] [CrossRef] [PubMed]
  63. Cole, J.E.; Navin, T.J.; Cross, A.J.; Goddard, M.E.; Alexopoulou, L.; Mitra, A.T.; Davies, A.H.; Flavell, R.A.; Feldmann, M.; Monaco, C. Unexpected Protective Role for Toll-like Receptor 3 in the Arterial Wall. Proc. Natl. Acad. Sci. USA 2011, 108, 2372–2377. [Google Scholar] [CrossRef] [PubMed]
  64. Squillace, S.; Salvemini, D. Toll-like Receptor-Mediated Neuroinflammation: Relevance for Cognitive Dysfunctions. Trends Pharmacol. Sci. 2022, 43, 726–739. [Google Scholar] [CrossRef]
  65. Negishi, H.; Osawa, T.; Ogami, K.; Ouyang, X.; Sakaguchi, S.; Koshiba, R.; Yanai, H.; Seko, Y.; Shitara, H.; Bishop, K.; et al. A Critical Link between Toll-like Receptor 3 and Type II Interferon Signaling Pathways in Antiviral Innate Immunity. Proc. Natl. Acad. Sci. USA 2008, 105, 20446–20451. [Google Scholar] [CrossRef] [PubMed]
  66. Yu, R.; Zhu, B.; Chen, D. Type I Interferon-Mediated Tumor Immunity and Its Role in Immunotherapy. Cell. Mol. Life Sci. 2022, 79, 191. [Google Scholar] [CrossRef] [PubMed]
  67. Kopeć, G.; Skride, A.; Ereminiene, E.; Simkova, I.; Enache, R.; Samarzija, M.; Salobir, B.; Jansa, P. Emerging Therapies and New Directions in the Treatment of Pulmonary Arterial Hypertension. Pol. Heart J. 2025, 83, 18–26. [Google Scholar] [CrossRef] [PubMed]
  68. UniProt. Available online: https://www.uniprot.org/uniprotkb/O00206/entry (accessed on 12 September 2025).
  69. Kim, H.-J.; Kim, H.; Lee, J.-H.; Hwangbo, C. Toll-like Receptor 4 (TLR4): New Insight Immune and Aging. Immun. Ageing 2023, 20, 67. [Google Scholar] [CrossRef]
  70. Duan, T.; Du, Y.; Xing, C.; Wang, H.Y.; Wang, R.-F. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front. Immunol. 2022, 13, 812774. [Google Scholar] [CrossRef]
  71. Li, J.; Yang, F.; Wei, F.; Ren, X. The Role of Toll-like Receptor 4 in Tumor Microenvironment. Oncotarget 2017, 8, 66656–66667. [Google Scholar] [CrossRef]
  72. Oda, M.; Yamamoto, H.; Kawakami, T. Maintenance of Homeostasis by TLR4 Ligands. Front. Immunol. 2024, 15, 1286270. [Google Scholar] [CrossRef]
  73. Wei, J.; Zhang, Y.; Li, H.; Wang, F.; Yao, S. Toll-like Receptor 4: A Potential Therapeutic Target for Multiple Human Diseases. Biomed. Pharmacother. 2023, 166, 115338. [Google Scholar] [CrossRef]
  74. Mansouri, A.; Akthar, I.; Miyamoto, A. TLR2 and TLR4 Bridge Physiological and Pathological Inflammation in the Reproductive System. Commun. Biol. 2025, 8, 1008. [Google Scholar] [CrossRef]
  75. Qin, C.; Zhang, B.; Zhang, L.; Zhang, Z.; Wang, L.; Tang, L.; Li, S.; Yang, Y.; Yang, F.; Zhang, P.; et al. MyD88-Dependent Toll-like Receptor 4 Signal Pathway in Intervertebral Disc Degeneration. Exp. Ther. Med. 2016, 12, 611–618. [Google Scholar] [CrossRef]
  76. Chettab, K.; Fitzsimmons, C.; Novikov, A.; Denis, M.; Phelip, C.; Mathé, D.; Choffour, P.A.; Beaumel, S.; Fourmaux, E.; Norca, P.; et al. A Systemically Administered Detoxified TLR4 Agonist Displays Potent Antitumor Activity and an Acceptable Tolerance Profile in Preclinical Models. Front. Immunol. 2023, 14, 1066402. [Google Scholar] [CrossRef]
  77. Richert, I.; Berchard, P.; Abbes, L.; Novikov, A.; Chettab, K.; Vandermoeten, A.; Dumontet, C.; Karanian, M.; Kerzerho, J.; Caroff, M.; et al. A TLR4 Agonist Induces Osteosarcoma Regression by Inducing an Antitumor Immune Response and Reprogramming M2 Macrophages to M1 Macrophages. Cancers 2023, 15, 4635. [Google Scholar] [CrossRef] [PubMed]
  78. Gabr, M.M.; Saeed, I.; Miles, J.A.; Ross, B.P.; Shaw, P.N.; Hollmann, M.W.; Parat, M.-O. Interaction of Opioids with TLR4—Mechanisms and Ramifications. Cancers 2021, 13, 5274. [Google Scholar] [CrossRef] [PubMed]
  79. Li, Y.; Sun, L.; Zhou, Q.; Lee, A.J.; Wang, L.; Zhang, R.; Wang, S. Effects of Opioid Drugs on Immune Function in Cancer Patients. Biomed. Pharmacother. 2024, 175, 116665. [Google Scholar] [CrossRef] [PubMed]
  80. Dabi, Y.T.; Ajagbe, A.O.; Degechisa, S.T. Toll-like Receptors in Pathogenesis of Neurodegenerative Diseases and Their Therapeutic Potential. Immun. Inflamm. Dis. 2023, 11, e839. [Google Scholar] [CrossRef]
  81. Zhang, W.; Xiao, D.; Mao, Q.; Xia, H. Role of Neuroinflammation in Neurodegeneration Development. Signal Transduct. Target. Ther. 2023, 8, 267. [Google Scholar] [CrossRef]
  82. Li, L.; Acioglu, C.; Heary, R.F.; Elkabes, S. Role of Astroglial Toll-like Receptors (TLRs) in Central Nervous System Infections, Injury and Neurodegenerative Diseases. Brain Behav. Immun. 2021, 91, 740–755. [Google Scholar] [CrossRef]
  83. Fu, Y.; Gong, T.; Loughran, P.A.; Li, Y.; Billiar, T.R.; Liu, Y.; Wen, Z.; Fan, J. Roles of TLR4 in Macrophage Immunity and Macrophage-Pulmonary Vascular/Lymphatic Endothelial Cell Interactions in Sepsis. Commun. Biol. 2025, 8, 469. [Google Scholar] [CrossRef]
  84. Stierschneider, A.; Wiesner, C. Shedding Light on the Molecular and Regulatory Mechanisms of TLR4 Signaling in Endothelial Cells under Physiological and Inflamed Conditions. Front. Immunol. 2023, 14, 1264889. [Google Scholar] [CrossRef]
  85. Sun, Z. Platelet TLR4, a Critical Link in Pulmonary Arterial Hypertension. Circ. Res. 2014, 114, 1551–1553. [Google Scholar] [CrossRef]
  86. Dai, J.; Chen, H.; Fang, J.; Wu, S.; Jia, Z. Vascular Remodeling: The Multicellular Mechanisms of Pulmonary Hypertension. Int. J. Mol. Sci. 2025, 26, 4265. [Google Scholar] [CrossRef]
  87. Ye, Y.; Xu, Q.; Wuren, T. Inflammation and Immunity in the Pathogenesis of Hypoxic Pulmonary Hypertension. Front. Immunol. 2023, 14, 1162556. [Google Scholar] [CrossRef]
  88. Peri, F.; Calabrese, V. Toll-like Receptor 4 (TLR4) Modulation by Synthetic and Natural Compounds: An Update. J. Med. Chem. 2014, 57, 3612–3622. [Google Scholar] [CrossRef]
  89. Zhang, J.; Zhang, W.; Yang, Z.; Fan, B.; Wang, C.; Tian, Z. Cinnamaldehyde Alleviates Pulmonary Hypertension by Affecting Vascular Remodeling through the TLR4/NF-kB/HIF-1a Pathway. Clin. Exp. Hypertens. 2025, 47, 2486829. [Google Scholar] [CrossRef] [PubMed]
  90. 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] [PubMed]
  91. Bauer, E.M.; Chanthaphavong, R.S.; Sodhi, C.P.; Hackam, D.J.; Billiar, T.R.; Bauer, P.M. Genetic Deletion of Toll-like Receptor 4 on Platelets Attenuates Experimental Pulmonary Hypertension. Circ. Res. 2014, 114, 1596–1600. [Google Scholar] [CrossRef] [PubMed]
  92. Wang, J.; Tian, X.-T.; Peng, Z.; Li, W.-Q.; Cao, Y.-Y.; Li, Y.; Li, X.-H. HMGB1/TLR4 Promotes Hypoxic Pulmonary Hypertension via Suppressing BMPR2 Signaling. Vasc. Pharmacol. 2019, 117, 35–44. [Google Scholar] [CrossRef]
  93. Grinnan, D.; Farkas, L. A Novel Peptide for Immunomodulation in Pulmonary Arterial Hypertension. Am. J. Respir. Crit. Care Med. 2019, 199, 1460–1461. [Google Scholar] [CrossRef]
  94. Huang, L.; Zhang, H.; Liu, Y.; Long, Y. The Role of Gut and Airway Microbiota in Pulmonary Arterial Hypertension. Front. Microbiol. 2022, 13, 929752. [Google Scholar] [CrossRef]
  95. Wedgwood, S.; Gerard, K.; Halloran, K.; Hanhauser, A.; Monacelli, S.; Warford, C.; Thai, P.N.; Chiamvimonvat, N.; Lakshminrusimha, S.; Steinhorn, R.H.; et al. Intestinal Dysbiosis and the Developing Lung: The Role of Toll-Like Receptor 4 in the Gut-Lung Axis. Front. Immunol. 2020, 11, 357. [Google Scholar] [CrossRef]
  96. UniProt. Available online: https://www.uniprot.org/uniprotkb/O60602/entry (accessed on 12 September 2025).
  97. Feuillet, V.; Medjane, S.; Mondor, I.; Demaria, O.; Pagni, P.P.; Galán, J.E.; Flavell, R.A.; Alexopoulou, L. Involvement of Toll-like Receptor 5 in the Recognition of Flagellated Bacteria. Proc. Natl. Acad. Sci. USA 2006, 103, 12487–12492. [Google Scholar] [CrossRef]
  98. Mizel, S.B.; West, A.P.; Hantgan, R.R. Identification of a Sequence in Human Toll-like Receptor 5 Required for the Binding of Gram-Negative Flagellin. J. Biol. Chem. 2003, 278, 23624–23629. [Google Scholar] [CrossRef] [PubMed]
  99. Amemiya, K.; Dankmeyer, J.L.; Bernhards, R.C.; Fetterer, D.P.; Waag, D.M.; Worsham, P.L.; DeShazer, D. Activation of Toll-Like Receptors by Live Gram-Negative Bacterial Pathogens Reveals Mitigation of TLR4 Responses and Activation of TLR5 by Flagella. Front. Cell. Infect. Microbiol. 2021, 11, 745325. [Google Scholar] [CrossRef] [PubMed]
  100. Tallant, T.; Deb, A.; Kar, N.; Lupica, J.; de Veer, M.J.; DiDonato, J.A. Flagellin Acting via TLR5 is the Major Activator of Key Signaling Pathways Leading to NF-κB and Proinflammatory Gene Program Activation in Intestinal Epithelial Cells. BMC Microbiol. 2004, 4, 33. [Google Scholar] [CrossRef] [PubMed]
  101. Buschard, K.; Krogvold, L.; Pociot, F.; Gerling, I.; Thea, R.; Dahl-Jørgensen, K.; Hansen, C.H.F. TLR5 Influences the Development of Type 1 Diabetes. BMJ Open Diabetes Res. Care 2025, 13, e005111. [Google Scholar] [CrossRef]
  102. Feng, S.; Zhang, C.; Chen, S.; He, R.; Chao, G.; Zhang, S. TLR5 Signaling in the Regulation of Intestinal Mucosal Immunity. J. Inflamm. Res. 2023, 16, 2491–2501. [Google Scholar] [CrossRef]
  103. Chamberlain, N.D.; Vila, O.M.; Volin, M.V.; Volkov, S.; Pope, R.M.; Swedler, W.; Mandelin, A.M.; Shahrara, S. TLR5; a Novel and Unidentified Inflammatory Mediator in Rheumatoid Arthritis That Correlates with Disease Activity Score and Joint TNF-α Levels. J. Immunol. 2012, 189, 475–483. [Google Scholar] [CrossRef]
  104. Vijay, K. Toll-like Receptors in Immunity and Inflammatory Diseases: Past, Present, and Future. Int. Immunopharmacol. 2018, 59, 391–412. [Google Scholar] [CrossRef]
  105. Wang, K.; Huang, H.; Zhan, Q.; Ding, H.; Li, Y. Toll-like Receptors in Health and Disease. MedComm 2024, 5, e549. [Google Scholar] [CrossRef]
  106. Cai, Z.; Sanchez, A.; Shi, Z.; Zhang, T.; Liu, M.; Zhang, D. Activation of Toll-Like Receptor 5 on Breast Cancer Cells by Flagellin Suppresses Cell Proliferation and Tumor Growth. Cancer Res. 2011, 71, 2466–2475. [Google Scholar] [CrossRef]
  107. McGinty, M.T.; Putelo, A.M.; Kolli, S.H.; Feng, T.-Y.; Dietl, M.R.; Hatzinger, C.N.; Bajgai, S.; Poblete, M.K.; Azar, F.N.; Mohammad, A.; et al. TLR5 Signaling Causes Dendritic Cell Dysfunction and Orchestrates Failure of Immune Checkpoint Therapy against Ovarian Cancer. Cancer Immunol. Res. 2025, 13, 696–711. [Google Scholar] [CrossRef]
  108. Kim, J.; Yoo, J.-Y.; Suh, J.M.; Park, S.; Kang, D.; Jo, H.; Bae, Y.S. The Flagellin-TLR5-Nox4 Axis Promotes the Migration of Smooth Muscle Cells in Atherosclerosis. Exp. Mol. Med. 2019, 51, 78. [Google Scholar] [CrossRef]
  109. Kim, J.; Seo, M.; Kim, S.K.; Bae, Y.S. Flagellin-Induced NADPH Oxidase 4 Activation is Involved in Atherosclerosis. Sci. Rep. 2016, 6, 25437. [Google Scholar] [CrossRef] [PubMed]
  110. Ellenbroek, G.H.J.M.; van Puijvelde, G.H.M.; Anas, A.A.; Bot, M.; Asbach, M.; Schoneveld, A.; van Santbrink, P.J.; Foks, A.C.; Timmers, L.; Doevendans, P.A.; et al. Leukocyte TLR5 Deficiency Inhibits Atherosclerosis by Reduced Macrophage Recruitment and Defective T-Cell Responsiveness. Sci. Rep. 2017, 7, 42688. [Google Scholar] [CrossRef] [PubMed]
  111. Shi, D.; Zhao, S.; Jiang, W.; Zhang, C.; Liang, T.; Hou, G. TLR5: A Prognostic and Monitoring Indicator for Triple-Negative Breast Cancer. Cell Death Dis. 2019, 10, 954. [Google Scholar] [CrossRef] [PubMed]
  112. Pfirschke, C.; Garris, C.; Pittet, M.J. Common TLR5 Mutations Control Cancer Progression. Cancer Cell 2015, 27, 1–3. [Google Scholar] [CrossRef]
  113. Rohm, I.; Grün, K.; Müller, L.M.; Bäz, L.; Förster, M.; Schrepper, A.; Kretzschmar, D.; Pistulli, R.; Yilmaz, A.; Bauer, R.; et al. Cellular Inflammation in Pulmonary Hypertension: Detailed Analysis of Lung and Right Ventricular Tissue, Circulating Immune Cells and Effects of a Dual Endothelin Receptor Antagonist. Clin. Hemorheol. Microcirc. 2019, 73, 497–522. [Google Scholar] [CrossRef]
  114. Sakamachi, Y.; Wiley, E.; Solis, A.; Johnson, C.G.; Meng, X.; Hussain, S.; Lipinski, J.H.; O’Dwyer, D.N.; Randall, T.; Malphurs, J.; et al. Toll-Like-Receptor 5 Protects against Pulmonary Fibrosis by Reducing Lung Dysbiosis. bioRxiv 2024. bioRxiv:2024.04.30.591719. [Google Scholar] [CrossRef]
  115. Oh, S.; Jang, A.Y.; Chae, S.; Choi, S.; Moon, J.; Kim, M.; Spiekerkoetter, E.; Zamanian, R.T.; Yang, P.C.; Hwang, D.; et al. Comparative Analysis on the Anti-Inflammatory/Immune Effect of Mesenchymal Stem Cell Therapy for the Treatment of Pulmonary Arterial Hypertension. Sci. Rep. 2021, 11, 2012. [Google Scholar] [CrossRef]
  116. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9Y2C9/entry (accessed on 12 September 2025).
  117. Jang, T.-H.; Park, H.H. Crystal Structure of TIR Domain of TLR6 Reveals Novel Dimeric Interface of TIR-TIR Interaction for Toll-like Receptor Signaling Pathway. J. Mol. Biol. 2014, 426, 3305–3313. [Google Scholar] [CrossRef]
  118. Misch, E.A.; Verbon, A.; Prins, J.M.; Skerrett, S.J.; Hawn, T.R. A TLR6 Polymorphism is Associated with Increased Risk of Legionnaires’ Disease. Genes Immun. 2013, 14, 420–426. [Google Scholar] [CrossRef]
  119. Hoffjan, S.; Stemmler, S.; Parwez, Q.; Petrasch-Parwez, E.; Arinir, U.; Rohde, G.; Reinitz-Rademacher, K.; Schultze-Werninghaus, G.; Bufe, A.; Epplen, J.T. Evaluation of the Toll-like Receptor 6 Ser249Pro Polymorphism in Patients with Asthma, Atopic Dermatitis and Chronic Obstructive Pulmonary Disease. BMC Med. Genet. 2005, 6, 34. [Google Scholar] [CrossRef]
  120. Grote, K.; Petri, M.; Liu, C.; Jehn, P.; Spalthoff, S.; Kokemüller, H.; Luchtefeld, M.; Tschernig, T.; Krettek, C.; Haasper, C.; et al. Toll-like Receptor 2/6-Dependent Stimulation of Mesenchymal Stem Cells Promotes Angiogenesis by Paracrine Factors. Eur. Cell Mater. 2013, 26, 66–79; discussion 79. [Google Scholar] [CrossRef]
  121. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9NYK1/entry (accessed on 12 September 2025).
  122. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9NR97/entry (accessed on 12 September 2025).
  123. Rathinam, V.A.K.; Fitzgerald, K.A. Innate Immune Sensing of DNA Viruses. Virology 2011, 411, 153–162. [Google Scholar] [CrossRef]
  124. Bello, M.B.; Usman, D. Identification of Single Stranded RNA Fragments Recognised by TLR7/8 in Dengue and Other Mosquito-Borne Flaviviral Genomes: A Bioinformatics Approach. Microbe 2025, 7, 100406. [Google Scholar] [CrossRef]
  125. Soleiman-Meigooni, S.; Yarahmadi, A.; Kheirkhah, A.-H.; Afkhami, H. Recent Advances in Different Interactions between Toll-like Receptors and Hepatitis B Infection: A Review. Front. Immunol. 2024, 15, 1363996. [Google Scholar] [CrossRef] [PubMed]
  126. Wen, L.; Zhang, B.; Wu, X.; Liu, R.; Fan, H.; Han, L.; Zhang, Z.; Ma, X.; Chu, C.-Q.; Shi, X. Toll-like Receptors 7 and 9 Regulate the Proliferation and Differentiation of B Cells in Systemic Lupus Erythematosus. Front. Immunol. 2023, 14, 1093208. [Google Scholar] [CrossRef]
  127. Sakata, K.; Nakayamada, S.; Miyazaki, Y.; Kubo, S.; Ishii, A.; Nakano, K.; Tanaka, Y. Up-Regulation of TLR7-Mediated IFN-α Production by Plasmacytoid Dendritic Cells in Patients With Systemic Lupus Erythematosus. Front. Immunol. 2018, 9, 1957. [Google Scholar] [CrossRef] [PubMed]
  128. Sakref, C.; Saby, A.; Rodriguez, C.; Ardin, M.; Moudombi, L.; Doffin, A.-C.; Gobbini, E.; Voissiere, A.; Besson, L.; Laoubi, L.; et al. Type III Interferon Primes pDCs for TLR7 Activation and Antagonizes Immune Suppression Mediated by TGF-β and PGE2. Nat. Commun. 2025, 16, 3045. [Google Scholar] [CrossRef]
  129. Lee, J.; Hayashi, M.; Lo, J.-F.; Fearns, C.; Chu, W.-M.; Luo, Y.; Xiang, R.; Chuang, T.-H. NF-κB Activation Primes Cells to a pro-Inflammatory Polarized Response to a TLR7 Agonist. Biochem. J. 2009, 421, 301–310. [Google Scholar] [CrossRef]
  130. Hamerman, J.A.; Pottle, J.; Ni, M.; He, Y.; Zhang, Z.-Y.; Buckner, J.H. Negative Regulation of TLR Signaling in Myeloid Cells—Implications for Autoimmune Diseases. Immunol. Rev. 2016, 269, 212–227. [Google Scholar] [CrossRef]
  131. Cervantes, J.L.; Weinerman, B.; Basole, C.; Salazar, J.C. TLR8: The Forgotten Relative Revindicated. Cell Mol. Immunol. 2012, 9, 434–438. [Google Scholar] [CrossRef]
  132. Iwasaki, A.; Pillai, P.S. Innate Immunity to Influenza Virus Infection. Nat. Rev. Immunol. 2014, 14, 315–328. [Google Scholar] [CrossRef] [PubMed]
  133. Jurk, M.; Heil, F.; Vollmer, J.; Schetter, C.; Krieg, A.M.; Wagner, H.; Lipford, G.; Bauer, S. Human TLR7 or TLR8 Independently Confer Responsiveness to the Antiviral Compound R-848. Nat. Immunol. 2002, 3, 499. [Google Scholar] [CrossRef] [PubMed]
  134. de Groot, N.G.; Bontrop, R.E. COVID-19 Pandemic: Is a Gender-Defined Dosage Effect Responsible for the High Mortality Rate among Males? Immunogenetics 2020, 72, 275–277. [Google Scholar] [CrossRef] [PubMed]
  135. Yokogawa, M.; Takaishi, M.; Nakajima, K.; Kamijima, R.; Fujimoto, C.; Kataoka, S.; Terada, Y.; Sano, S. Epicutaneous Application of Toll-like Receptor 7 Agonists Leads to Systemic Autoimmunity in Wild-Type Mice: A New Model of Systemic Lupus Erythematosus. Arthritis Rheumatol. 2014, 66, 694–706. [Google Scholar] [CrossRef]
  136. Yeh, F.-C.; Chen, C.-N.; Xie, C.-Y.; Baxan, N.; Zhao, L.; Ashek, A.; Sabrin, F.; Lawrie, A.; Wilkins, M.; Zhao, L. TLR7/8 Activation Induces Autoimmune Vasculopathy and Causes Severe Pulmonary Arterial Hypertension. Eur. Respir. J. 2023, 62, 2300204. [Google Scholar] [CrossRef]
  137. Hubert, F.-X.; Voisine, C.; Louvet, C.; Heslan, M.; Josien, R. Rat Plasmacytoid Dendritic Cells Are an Abundant Subset of MHC Class II+ CD4+CD11b-OX62- and Type I IFN-Producing Cells That Exhibit Selective Expression of Toll-like Receptors 7 and 9 and Strong Responsiveness to CpG. J. Immunol. 2004, 172, 7485–7494. [Google Scholar] [CrossRef]
  138. Hautefort, A.; Girerd, B.; Montani, D.; Cohen-Kaminsky, S.; Price, L.; Lambrecht, B.N.; Humbert, M.; Perros, F. T-Helper 17 Cell Polarization in Pulmonary Arterial Hypertension. Chest 2015, 147, 1610–1620. [Google Scholar] [CrossRef]
  139. Seki, N.; Tsujimoto, H.; Tanemura, S.; Ishigaki, S.; Takei, H.; Sugahara, K.; Yoshimoto, K.; Akiyama, M.; Kaneko, Y.; Chiba, K.; et al. Th17/IL-17A Axis is Critical for Pulmonary Arterial Hypertension (PAH) in Systemic Sclerosis (SSc): SSc Patients with High Levels of Serum IL-17A Exhibit Reduced Lung Functions and Increased Prevalence of PAH. Cytokine 2024, 176, 156534. [Google Scholar] [CrossRef]
  140. Huertas, A.; Phan, C.; Bordenave, J.; Tu, L.; Thuillet, R.; Le Hiress, M.; Avouac, J.; Tamura, Y.; Allanore, Y.; Jovan, R.; et al. Regulatory T Cell Dysfunction in Idiopathic, Heritable and Connective Tissue-Associated Pulmonary Arterial Hypertension. Chest 2016, 149, 1482–1493. [Google Scholar] [CrossRef]
  141. Robles-Vera, I.; Visitación, N.D.L.; Toral, M.; Sánchez, M.; Gómez-Guzmán, M.; O’valle, F.; Jiménez, R.; Duarte, J.; Romero, M. Toll-like Receptor 7-Driven Lupus Autoimmunity Induces Hypertension and Vascular Alterations in Mice. J. Hypertens. 2020, 38, 1322–1335. [Google Scholar] [CrossRef] [PubMed]
  142. Margery-Muir, A.A.; Bundell, C.; Nelson, D.; Groth, D.M.; Wetherall, J.D. Gender Balance in Patients with Systemic Lupus Erythematosus. Autoimmun. Rev. 2017, 16, 258–268. [Google Scholar] [CrossRef] [PubMed]
  143. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9NR96/entry (accessed on 12 September 2025).
  144. Bauer, S.; Wagner, H. Bacterial CpG-DNA Licenses TLR9. Curr. Top. Microbiol. Immunol. 2002, 270, 145–154. [Google Scholar] [CrossRef] [PubMed]
  145. Ohto, U.; Shibata, T.; Tanji, H.; Ishida, H.; Krayukhina, E.; Uchiyama, S.; Miyake, K.; Shimizu, T. Structural Basis of CpG and Inhibitory DNA Recognition by Toll-like Receptor 9. Nature 2015, 520, 702–705. [Google Scholar] [CrossRef]
  146. Ding, Z.; Liu, S.; Wang, X.; Khaidakov, M.; Dai, Y.; Mehta, J.L. Oxidant Stress in Mitochondrial DNA Damage, Autophagy and Inflammation in Atherosclerosis. Sci. Rep. 2013, 3, 1077. [Google Scholar] [CrossRef]
  147. Suresh, M.V.; Thomas, B.; Dolgachev, V.A.; Sherman, M.A.; Goldberg, R.; Johnson, M.; Chowdhury, A.; Machado-Aranda, D.; Raghavendran, K. Toll-Like Receptor-9 (TLR9) Is Requisite for Acute Inflammatory Response and Injury Following Lung Contusion. Shock 2016, 46, 412–419. [Google Scholar] [CrossRef]
  148. Tsuji, N.; Tsuji, T.; Ohashi, N.; Kato, A.; Fujigaki, Y.; Yasuda, H. Role of Mitochondrial DNA in Septic AKI via Toll-Like Receptor 9. J. Am. Soc. Nephrol. 2016, 27, 2009–2020. [Google Scholar] [CrossRef]
  149. Schumacker, P.T.; Gillespie, M.N.; Nakahira, K.; Choi, A.M.K.; Crouser, E.D.; Piantadosi, C.A.; Bhattacharya, J. Mitochondria in Lung Biology and Pathology: More than Just a Powerhouse. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 306, L962–L974. [Google Scholar] [CrossRef]
  150. Garcia-Martinez, I.; Santoro, N.; Chen, Y.; Hoque, R.; Ouyang, X.; Caprio, S.; Shlomchik, M.J.; Coffman, R.L.; Candia, A.; Mehal, W.Z. Hepatocyte Mitochondrial DNA Drives Nonalcoholic Steatohepatitis by Activation of TLR9. J. Clin. Investig. 2016, 126, 859–864. [Google Scholar] [CrossRef]
  151. Han, S.J.; Li, H.; Kim, M.; Shlomchik, M.J.; Lee, H.T. Kidney Proximal Tubular TLR9 Exacerbates Ischemic Acute Kidney Injury. J. Immunol. 2018, 201, 1073–1085. [Google Scholar] [CrossRef]
  152. McCarthy, C.G.; Wenceslau, C.F.; Goulopoulou, S.; Ogbi, S.; Baban, B.; Sullivan, J.C.; Matsumoto, T.; Webb, R.C. Circulating Mitochondrial DNA and Toll-like Receptor 9 Are Associated with Vascular Dysfunction in Spontaneously Hypertensive Rats. Cardiovasc. Res. 2015, 107, 119–130. [Google Scholar] [CrossRef]
  153. Oka, T.; Hikoso, S.; Yamaguchi, O.; Taneike, M.; Takeda, T.; Tamai, T.; Oyabu, J.; Murakawa, T.; Nakayama, H.; Nishida, K.; et al. Mitochondrial DNA That Escapes from Autophagy Causes Inflammation and Heart Failure. Nature 2012, 485, 251–255. [Google Scholar] [CrossRef]
  154. Loomis, Z.; Eigenberger, P.; Redinius, K.; Lisk, C.; Karoor, V.; Nozik-Grayck, E.; Ferguson, S.K.; Hassell, K.; Nuss, R.; Stenmark, K.; et al. Hemoglobin Induced Cell Trauma Indirectly Influences Endothelial TLR9 Activity Resulting in Pulmonary Vascular Smooth Muscle Cell Activation. PLoS ONE 2017, 12, e0171219. [Google Scholar] [CrossRef]
  155. Ishikawa, T.; Abe, K.; Takana-Ishikawa, M.; Yoshida, K.; Watanabe, T.; Imakiire, S.; Hosokawa, K.; Hirano, M.; Hirano, K.; Tsutsui, H. Chronic Inhibition of Toll-Like Receptor 9 Ameliorates Pulmonary Hypertension in Rats. J. Am. Heart Assoc. 2021, 10, e019247. [Google Scholar] [CrossRef] [PubMed]
  156. Abe, K.; Shinoda, M.; Tanaka, M.; Kuwabara, Y.; Yoshida, K.; Hirooka, Y.; McMurtry, I.F.; Oka, M.; Sunagawa, K. Haemodynamic Unloading Reverses Occlusive Vascular Lesions in Severe Pulmonary Hypertension. Cardiovasc. Res. 2016, 111, 16–25. [Google Scholar] [CrossRef] [PubMed]
  157. Gladwin, M.T. Revisiting the Hyperhemolysis Paradigm. Blood 2015, 126, 695–696. [Google Scholar] [CrossRef] [PubMed]
  158. Gladwin, M.T.; Vichinsky, E. Pulmonary Complications of Sickle Cell Disease. N. Engl. J. Med. 2008, 359, 2254–2265. [Google Scholar] [CrossRef]
  159. Kato, G.J.; Taylor, J.G. Pleiotropic Effects of Intravascular Hemolysis on Vascular Homeostasis. Br. J. Haematol. 2010, 148, 690–701. [Google Scholar] [CrossRef]
  160. Kato, G.J. Novel Small Molecule Therapeutics for Sickle Cell Disease: Nitric Oxide, Carbon Monoxide, Nitrite, and Apolipoprotein A-I. Hematol. Am. Soc. Hematol. Educ. Program 2008, 2008, 186–192. [Google Scholar] [CrossRef]
  161. Haase, M.; Bellomo, R.; Haase-Fielitz, A. Novel Biomarkers, Oxidative Stress, and the Role of Labile Iron Toxicity in Cardiopulmonary Bypass-Associated Acute Kidney Injury. J. Am. Coll. Cardiol. 2010, 55, 2024–2033. [Google Scholar] [CrossRef]
  162. Lisk, C.; Kominsky, D.; Ehrentraut, S.; Bonaventura, J.; Nuss, R.; Hassell, K.; Nozik-Grayck, E.; Irwin, D.C. Hemoglobin-Induced Endothelial Cell Permeability is Controlled, in Part, via a Myeloid Differentiation Primary Response Gene-88-Dependent Signaling Mechanism. Am. J. Respir. Cell Mol. Biol. 2013, 49, 619–626. [Google Scholar] [CrossRef] [PubMed]
  163. Silva, G.; Jeney, V.; Chora, A.; Larsen, R.; Balla, J.; Soares, M.P. Oxidized Hemoglobin is an Endogenous Proinflammatory Agonist That Targets Vascular Endothelial Cells. J. Biol. Chem. 2009, 284, 29582–29595. [Google Scholar] [CrossRef] [PubMed]
  164. Shaver, C.M.; Upchurch, C.P.; Janz, D.R.; Grove, B.S.; Putz, N.D.; Wickersham, N.E.; Dikalov, S.I.; Ware, L.B.; Bastarache, J.A. Cell-Free Hemoglobin: A Novel Mediator of Acute Lung Injury. Am. J. Physiol. Lung Cell Mol. Physiol. 2016, 310, L532–L541. [Google Scholar] [CrossRef] [PubMed]
  165. Schaer, D.J.; Vinchi, F.; Ingoglia, G.; Tolosano, E.; Buehler, P.W. Haptoglobin, Hemopexin, and Related Defense Pathways-Basic Science, Clinical Perspectives, and Drug Development. Front. Physiol. 2014, 5, 415. [Google Scholar] [CrossRef]
  166. Schaer, D.J.; Buehler, P.W. Cell-Free Hemoglobin and Its Scavenger Proteins: New Disease Models Leading the Way to Targeted Therapies. Cold Spring Harb. Perspect. Med. 2013, 3, a013433. [Google Scholar] [CrossRef]
  167. Smith, A.; McCulloh, R.J. Hemopexin and Haptoglobin: Allies against Heme Toxicity from Hemoglobin Not Contenders. Front. Physiol. 2015, 6, 187. [Google Scholar] [CrossRef]
  168. UniProt. Available online: https://www.uniprot.org/uniprotkb/Q9BXR5/entry (accessed on 12 September 2025).
  169. 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]
  170. 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]
  171. 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]
  172. Nyman, T.; Stenmark, P.; Flodin, S.; Johansson, I.; Hammarström, M.; Nordlund, P. The Crystal Structure of the Human Toll-like Receptor 10 Cytoplasmic Domain Reveals a Putative Signaling Dimer. J. Biol. Chem. 2008, 283, 11861–11865. [Google Scholar] [CrossRef]
  173. Guan, Y.; Ranoa, D.R.E.; Jiang, S.; Mutha, S.K.; Li, X.; Baudry, J.; Tapping, R.I. Human TLRs 10 and 1 Share Common Mechanisms of Innate Immune Sensing but Not Signaling. J. Immunol. 2010, 184, 5094–5103. [Google Scholar] [CrossRef]
  174. Chuang, T.; Ulevitch, R.J. Identification of hTLR10: A Novel Human Toll-like Receptor Preferentially Expressed in Immune Cells. Biochim. Biophys. Acta 2001, 1518, 157–161. [Google Scholar] [CrossRef]
  175. Hornung, V.; Rothenfusser, S.; Britsch, S.; Krug, A.; Jahrsdörfer, B.; Giese, T.; Endres, S.; Hartmann, G. Quantitative Expression of Toll-like Receptor 1-10 mRNA in Cellular Subsets of Human Peripheral Blood Mononuclear Cells and Sensitivity to CpG Oligodeoxynucleotides. J. Immunol. 2002, 168, 4531–4537. [Google Scholar] [CrossRef] [PubMed]
  176. Regan, T.; Nally, K.; Carmody, R.; Houston, A.; Shanahan, F.; Macsharry, J.; Brint, E. Identification of TLR10 as a Key Mediator of the Inflammatory Response to Listeria Monocytogenes in Intestinal Epithelial Cells and Macrophages. J. Immunol. 2013, 191, 6084–6092. [Google Scholar] [CrossRef] [PubMed]
  177. Nagashima, H.; Iwatani, S.; Cruz, M.; Jiménez Abreu, J.A.; Uchida, T.; Mahachai, V.; Vilaichone, R.; Graham, D.Y.; Yamaoka, Y. Toll-like Receptor 10 in Helicobacter Pylori Infection. J. Infect. Dis. 2015, 212, 1666–1676. [Google Scholar] [CrossRef] [PubMed]
  178. Xia, P.; Wu, Y.; Lian, S.; Yan, L.; Meng, X.; Duan, Q.; Zhu, G. Research Progress on Toll-like Receptor Signal Transduction and Its Roles in Antimicrobial Immune Responses. Appl. Microbiol. Biotechnol. 2021, 105, 5341–5355. [Google Scholar] [CrossRef]
  179. Neuper, T.; Frauenlob, T.; Sarajlic, M.; Posselt, G.; Wessler, S.; Horejs-Hoeck, J. TLR2, TLR4 and TLR10 Shape the Cytokine and Chemokine Release of H. Pylori-Infected Human DCs. Int. J. Mol. Sci. 2020, 21, 3897. [Google Scholar] [CrossRef]
  180. Pullamsetti, S.S.; Savai, R.; Janssen, W.; Dahal, B.K.; Seeger, W.; Grimminger, F.; Ghofrani, H.A.; Weissmann, N.; Schermuly, R.T. Inflammation, Immunological Reaction and Role of Infection in Pulmonary Hypertension. Clin. Microbiol. Infect. 2011, 17, 7–14. [Google Scholar] [CrossRef]
  181. Tomaszewski, M.; Bębnowska, D.; Hrynkiewicz, R.; Dworzyński, J.; Niedźwiedzka-Rystwej, P.; Kopeć, G.; Grywalska, E. Role of the Immune System Elements in Pulmonary Arterial Hypertension. J. Clin. Med. 2021, 10, 3757. [Google Scholar] [CrossRef]
  182. Cao, Y.; Xie, L.; Shi, F.; Tang, M.; Li, Y.; Hu, J.; Zhao, L.; Zhao, L.; Yu, X.; Luo, X.; et al. Targeting the Signaling in Epstein–Barr Virus-Associated Diseases: Mechanism, Regulation, and Clinical Study. Signal Transduct. Target. Ther. 2021, 6, 15. [Google Scholar] [CrossRef]
  183. Huang, W.; Bai, L.; Tang, H. Epstein-Barr Virus Infection: The Micro and Macro Worlds. Virol. J. 2023, 20, 220. [Google Scholar] [CrossRef]
  184. Silva, J.D.M.; Alves, C.E.d.C.; Pontes, G.S. Epstein-Barr Virus: The Mastermind of Immune Chaos. Front. Immunol. 2024, 15, 1297994. [Google Scholar] [CrossRef]
  185. Kliszczewska, E.; Jarzyński, A.; Boguszewska, A.; Pasternak, J.; Polz-Dacewicz, M. Epstein-Barr Virus—Pathogenesis, Latency and Cancers. J. Pre-Clin. Clin. Res. 2017, 11, 142–146. [Google Scholar] [CrossRef]
  186. Pisano, G.; Roy, A.; Ahmed Ansari, M.; Kumar, B.; Chikoti, L.; Chandran, B. Interferon-γ-Inducible Protein 16 (IFI16) is Required for the Maintenance of Epstein-Barr Virus Latency. Virol. J. 2017, 14, 221. [Google Scholar] [CrossRef]
  187. Ansari, M.A.; Singh, V.V.; Dutta, S.; Veettil, M.V.; Dutta, D.; Chikoti, L.; Lu, J.; Everly, D.; Chandran, B. Constitutive Interferon-Inducible Protein 16-Inflammasome Activation during Epstein-Barr Virus Latency I, II, and III in B and Epithelial Cells. J. Virol. 2013, 87, 8606–8623, Erratum in J. Virol. 2017, 91, e01519-17. [Google Scholar] [CrossRef] [PubMed]
  188. Kurakula, K.; Smolders, V.F.E.D.; Tura-Ceide, O.; Jukema, J.W.; Quax, P.H.A.; Goumans, M.-J. Endothelial Dysfunction in Pulmonary Hypertension: Cause or Consequence? Biomedicines 2021, 9, 57. [Google Scholar] [CrossRef] [PubMed]
  189. Hashimoto, T.; Sakata, Y.; Fukushima, K.; Maeda, T.; Arita, Y.; Shioyama, W.; Nakaoka, Y.; Hori, Y.; Morii, E.; Aozasa, K.; et al. Pulmonary Arterial Hypertension Associated with Chronic Active Epstein-Barr Virus Infection. Intern. Med. 2011, 50, 119–124. [Google Scholar] [CrossRef] [PubMed]
  190. Misaki, Y.; Minakata, D.; Ibe, T.; Gomyo, A.; Yoshimura, K.; Kimura, S.; Nakamura, Y.; Kawamura, M.; Kawamura, S.; Takeshita, J.; et al. Chronic Active Epstein-Bar Virus Infection Complicated by Pulmonary Artery Hypertension. J. Infect. Chemother. 2023, 29, 212–218. [Google Scholar] [CrossRef]
  191. Fukuda, Y.; Momoi, N.; Akaihata, M.; Nagasawa, K.; Mitomo, M.; Aoyagi, Y.; Endoh, K.; Hosoya, M. Pulmonary Arterial Hypertension Associated with Chronic Active Epstein-Barr Virus Infection. Pediatr. Int. 2015, 57, 731–734. [Google Scholar] [CrossRef]
  192. Egan, J.J.; Adamali, H.I.; Lok, S.S.; Stewart, J.P.; Woodcock, A.A. Ganciclovir Antiviral Therapy in Advanced Idiopathic Pulmonary Fibrosis: An Open Pilot Study. Pulm. Med. 2011, 2011, 240805. [Google Scholar] [CrossRef]
  193. Calabrese, F.; Kipar, A.; Lunardi, F.; Balestro, E.; Perissinotto, E.; Rossi, E.; Nannini, N.; Marulli, G.; Stewart, J.P.; Rea, F. Herpes Virus Infection is Associated with Vascular Remodeling and Pulmonary Hypertension in Idiopathic Pulmonary Fibrosis. PLoS ONE 2013, 8, e55715. [Google Scholar] [CrossRef] [PubMed]
  194. Duckworth, A.; Longhurst, H.J.; Paxton, J.K.; Scotton, C.J. The Role of Herpes Viruses in Pulmonary Fibrosis. Front. Med. 2021, 8, 704222. [Google Scholar] [CrossRef] [PubMed]
  195. Savale, L.; Lador, F.; Jais, X.; Montani, D.; Simonneau, G.; Humbert, M.; Sitbon, O. HIV-related pulmonary arterial hypertension. Rev. Mal. Respir. 2012, 29, 491–500. [Google Scholar] [CrossRef] [PubMed]
  196. Mahajan, G.; Barjatya, H.C.; Bhakar, B.L.; Gothwal, S.K.; Jangir, T. To Estimate Prevalence of Pulmonary Arterial Hypertension in HIV Patients and Its Association with CD4 Cell Count. Clin. Epidemiol. Glob. Health 2024, 25, 101479. [Google Scholar] [CrossRef]
  197. Șoldea, S.; Iovănescu, M.; Berceanu, M.; Mirea, O.; Raicea, V.; Beznă, M.C.; Rocșoreanu, A.; Donoiu, I. Cardiovascular Disease in HIV Patients: A Comprehensive Review of Current Knowledge and Clinical Implications. Int. J. Mol. Sci. 2025, 26, 1837. [Google Scholar] [CrossRef]
  198. Kim, K.K.; Factor, S.M. Membranoproliferative Glomerulonephritis and Plexogenic Pulmonary Arteriopathy in a Homosexual Man with Acquired Immunodeficiency Syndrome. Hum. Pathol. 1987, 18, 1293–1296. [Google Scholar] [CrossRef]
  199. HIV Data and Statistics. Available online: https://www.who.int/teams/global-hiv-hepatitis-and-stis-programmes/hiv/strategic-information/hiv-data-and-statistics (accessed on 12 September 2025).
  200. Tseng, A.; Seet, J.; Phillips, E.J. The Evolution of Three Decades of Antiretroviral Therapy: Challenges, Triumphs and the Promise of the Future. Br. J. Clin. Pharmacol. 2015, 79, 182–194. [Google Scholar] [CrossRef]
  201. Masters, M.C.; Krueger, K.M.; Williams, J.L.; Morrison, L.; Cohn, S.E. Beyond One Pill, Once Daily: Current Challenges of Antiretroviral Therapy Management in the United States. Expert Rev. Clin. Pharmacol. 2019, 12, 1129–1143. [Google Scholar] [CrossRef]
  202. Woldegeorgis, B.Z.; Asgedom, Y.S.; Habte, A.; Kassie, G.A.; Badacho, A.S. Highly Active Antiretroviral Therapy is Necessary but Not Sufficient. A Systematic Review and Meta-Analysis of Mortality Incidence Rates and Predictors among HIV-Infected Adults Receiving Treatment in Ethiopia, a Surrogate Study for Resource-Poor Settings. BMC Public Health 2024, 24, 1735. [Google Scholar] [CrossRef]
  203. Kumar, A.; Mahajan, A.; Salazar, E.A.; Pruitt, K.; Guzman, C.A.; Clauss, M.A.; Almodovar, S.; Dhillon, N.K. Impact of Human Immunodeficiency Virus on Pulmonary Vascular Disease. Glob. Cardiol. Sci. Pract. 2021, 2021, e202112. [Google Scholar] [CrossRef]
  204. Chan, S.Y.; Loscalzo, J. Pathogenic Mechanisms of Pulmonary Arterial Hypertension. J. Mol. Cell Cardiol. 2008, 44, 14–30. [Google Scholar] [CrossRef] [PubMed]
  205. Bigna, J.J.R.; Sime, P.S.D.; Koulla-Shiro, S. HIV Related Pulmonary Arterial Hypertension: Epidemiology in Africa, Physiopathology, and Role of Antiretroviral Treatment. AIDS Res. Ther. 2015, 12, 36. [Google Scholar] [CrossRef] [PubMed]
  206. Prakash, P.; Swami Vetha, B.S.; Chakraborty, R.; Wenegieme, T.-Y.; Masenga, S.K.; Muthian, G.; Balasubramaniam, M.; Wanjalla, C.N.; Hinton, A.O.; Kirabo, A.; et al. HIV-Associated Hypertension: Risks, Mechanisms, and Knowledge Gaps. Circ. Res. 2024, 134, e150–e175. [Google Scholar] [CrossRef] [PubMed]
  207. Palakeel, J.J.; Ali, M.; Chaduvula, P.; Chhabra, S.; Lamsal Lamichhane, S.; Ramesh, V.; Opara, C.O.; Khan, F.Y.; Kabiraj, G.; Kauser, H.; et al. An Outlook on the Etiopathogenesis of Pulmonary Hypertension in HIV. Cureus 2022, 14, e27390. [Google Scholar] [CrossRef]
  208. Singh, T.; Motes, A.; Vinan-Vega, M.; Nugent, K. Pulmonary Arterial Hypertension in Human Immunodeficiency Virus Infections. Southwest Respir. Crit. Care Chron. 2024, 12, 12–18. [Google Scholar] [CrossRef]
  209. Chandran, S.; Adler, M.; Chen, L.; Kaur, S.; Dhillon, N.K. HIV-Tat and Vascular Endothelium: Implications in the HIV Associated Brain, Heart, and Lung Complications. Front. Immunol. 2025, 16, 1621338. [Google Scholar] [CrossRef]
  210. Kovacs, L.; Khakina, B.N.; de Chantemèle, E.J.B. Role of HIV-Encoded Proteins in Cardiovascular Disease. Am. J. Physiol. Cell Physiol. 2025, 329, C592–C598. [Google Scholar] [CrossRef]
  211. Anand, A.R.; Rachel, G.; Parthasarathy, D. HIV Proteins and Endothelial Dysfunction: Implications in Cardiovascular Disease. Front. Cardiovasc. Med. 2018, 5, 185. [Google Scholar] [CrossRef]
  212. Butrous, G. Human Immunodeficiency Virus–Associated Pulmonary Arterial Hypertension. Circulation 2015, 131, 1361–1370. [Google Scholar] [CrossRef]
  213. Henriques-Forsythe, M.; Annangi, S.; Farber, H.W. Prevalence and Hospital Discharge Status of Human Immunodeficiency Virus–Associated Pulmonary Arterial Hypertension in the United States. Pulm. Circ. 2015, 5, 506–512. [Google Scholar] [CrossRef] [PubMed]
  214. Porter, K.M.; Walp, E.R.; Elms, S.C.; Raynor, R.; Mitchell, P.O.; Guidot, D.M.; Sutliff, R.L. Human Immunodeficiency Virus-1 Transgene Expression Increases Pulmonary Vascular Resistance and Exacerbates Hypoxia-Induced Pulmonary Hypertension Development. Pulm. Circ. 2013, 3, 58–67. [Google Scholar] [CrossRef] [PubMed]
  215. Marecki, J.C.; Cool, C.D.; Parr, J.E.; Beckey, V.E.; Luciw, P.A.; Tarantal, A.F.; Carville, A.; Shannon, R.P.; Cota-Gomez, A.; Tuder, R.M.; et al. HIV-1 Nef Is Associated with Complex Pulmonary Vascular Lesions in SHIV-Nef–Infected Macaques. Am. J. Respir. Crit. Care Med. 2006, 174, 437–445. [Google Scholar] [CrossRef]
  216. Garcia, A.K.; Almodovar, S. The Intersection of HIV and Pulmonary Vascular Health: From HIV Evolution to Vascular Cell Types to Disease Mechanisms. J. Vasc. Dis. 2024, 3, 174–200. [Google Scholar] [CrossRef]
  217. Morozov, V.A.; Lagaye, S. Hepatitis C Virus: Morphogenesis, Infection and Therapy. World J. Hepatol. 2018, 10, 186–212. [Google Scholar] [CrossRef]
  218. Suhail, M.; Sohrab, S.S.; Kamal, M.A.; Azhar, E.I. Role of Hepatitis C Virus in Hepatocellular Carcinoma and Neurological Disorders: An Overview. Front. Oncol. 2022, 12, 913231. [Google Scholar] [CrossRef]
  219. Topi, S.; Gaxhja, E.; Charitos, I.A.; Colella, M.; Santacroce, L. Hepatitis C Virus: History and Current Knowledge. Gastroenterol. Insights 2024, 15, 676–707. [Google Scholar] [CrossRef]
  220. Mazumder, H.; Hossain, M.F.; Shrestha, P.; Mahmud, S.; Husain, M.; Ahmed, R. Prevalence and Associated Risk Factors of Current Hepatitis C Infection among U.S. General Population and Injection Drug Users Aged 20–59 Years: NHANES 2009–2018. PLoS ONE 2024, 19, e0309345. [Google Scholar] [CrossRef]
  221. Stroffolini, T.; Stroffolini, G. Prevalence and Modes of Transmission of Hepatitis C Virus Infection: A Historical Worldwide Review. Viruses 2024, 16, 1115. [Google Scholar] [CrossRef]
  222. Huang, D.Q.; Terrault, N.A.; Tacke, F.; Gluud, L.L.; Arrese, M.; Bugianesi, E.; Loomba, R. Global Epidemiology of Cirrhosis—Aetiology, Trends and Predictions. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 388–398. [Google Scholar] [CrossRef]
  223. Kwong, A.; Kim, W.R.; Mannalithara, A.; Heo, N.-Y.; Udompap, P.; Kim, D. Decreasing Mortality and Disease Severity in Hepatitis C Patients Awaiting Liver Transplantation in the United States. Liver Transpl. 2018, 24, 735–743. [Google Scholar] [CrossRef]
  224. Renard, S.; Borentain, P.; Salaun, E.; Benhaourech, S.; Maille, B.; Darque, A.; Bregigeon, S.; Colson, P.; Laugier, D.; Gaubert, M.R.; et al. Severe Pulmonary Arterial Hypertension in Patients Treated for Hepatitis C With Sofosbuvir. Chest 2016, 149, e69–e73. [Google Scholar] [CrossRef]
  225. Tsuchiya, H.; Kioka, H.; Ozu, K.; Ohtani, T.; Yamaguchi, O.; Yazaki, Y.; Yamauchi-Takihara, K.; Sakata, Y. Interferon Therapy Exacerbated Pulmonary Hypertension in a Patient with Hepatitis C Virus Infection: Pathogenic Interplay among Multiple Risk Factors. Intern. Med. 2017, 56, 1061–1065. [Google Scholar] [CrossRef]
  226. Doherty, M.; Bhindi, A.; Kilpin, M.; Jeganathan, V.; Fisher, L.; Chimunda, T. A Case of Pulmonary Arterial Hypertension Secondary to Hepatitis C Direct Antiviral Treatment with Harvoni® (Ledipasvir/Sofosbuvir Fixed-Dose Combination). Aust. Crit. Care 2020, 33, S37. [Google Scholar] [CrossRef]
  227. Savale, L.; Sattler, C.; Günther, S.; Montani, D.; Chaumais, M.-C.; Perrin, S.; Jaïs, X.; Seferian, A.; Jovan, R.; Bulifon, S.; et al. Pulmonary Arterial Hypertension in Patients Treated with Interferon. Eur. Respir. J. 2014, 44, 1627–1634. [Google Scholar] [CrossRef] [PubMed]
  228. Gong, G.; Waris, G.; Tanveer, R.; Siddiqui, A. Human Hepatitis C Virus NS5A Protein Alters Intracellular Calcium Levels, Induces Oxidative Stress, and Activates STAT-3 and NF-Kappa B. Proc. Natl. Acad. Sci. USA 2001, 98, 9599–9604. [Google Scholar] [CrossRef] [PubMed]
  229. Paulin, R.; Courboulin, A.; Meloche, J.; Mainguy, V.; Dumas de la Roque, E.; Saksouk, N.; Côté, J.; Provencher, S.; Sussman, M.A.; Bonnet, S. Signal Transducers and Activators of Transcription-3/Pim1 Axis Plays a Critical Role in the Pathogenesis of Human Pulmonary Arterial Hypertension. Circulation 2011, 123, 1205–1215. [Google Scholar] [CrossRef] [PubMed]
  230. Guedj, J.; Pang, P.S.; Denning, J.; Rodriguez-Torres, M.; Lawitz, E.; Symonds, W.; Perelson, A.S. Analysis of Hepatitis C Viral Kinetics during Administration of Two Nucleotide Analogues: Sofosbuvir (GS-7977) and GS-0938. Antivir. Ther. 2014, 19, 211–220. [Google Scholar] [CrossRef]
  231. Garcia-Montojo, M.; Doucet-O’Hare, T.; Henderson, L.; Nath, A. Human Endogenous Retrovirus-K (HML-2): A Comprehensive Review. Crit. Rev. Microbiol. 2018, 44, 715–738. [Google Scholar] [CrossRef]
  232. Lavie, L.; Medstrand, P.; Schempp, W.; Meese, E.; Mayer, J. Human Endogenous Retrovirus Family HERV-K(HML-5): Status, Evolution, and Reconstruction of an Ancient Betaretrovirus in the Human Genome. J. Virol. 2004, 78, 8788–8798. [Google Scholar] [CrossRef]
  233. Contreras-Galindo, R.; Kaplan, M.H.; Dube, D.; Gonzalez-Hernandez, M.J.; Chan, S.; Meng, F.; Dai, M.; Omenn, G.S.; Gitlin, S.D.; Markovitz, D.M. Human Endogenous Retrovirus Type K (HERV-K) Particles Package and Transmit HERV-K–Related Sequences. J. Virol. 2015, 89, 7187–7201. [Google Scholar] [CrossRef] [PubMed]
  234. Simula, E.R.; Jasemi, S.; Cossu, D.; Fais, M.; Cossu, I.; Chessa, V.; Canu, M.; Sechi, L.A. Human Endogenous Retroviruses as Novel Therapeutic Targets in Neurodegenerative Disorders. Vaccines 2025, 13, 415. [Google Scholar] [CrossRef] [PubMed]
  235. Johanning, G.L.; Malouf, G.G.; Zheng, X.; Esteva, F.J.; Weinstein, J.N.; Wang-Johanning, F.; Su, X. Expression of Human Endogenous Retrovirus-K is Strongly Associated with the Basal-like Breast Cancer Phenotype. Sci. Rep. 2017, 7, 41960. [Google Scholar] [CrossRef]
  236. Dou, A.; Xu, J.; Zhou, C. The Relationship between HERVs and Exogenous Viral Infections: A Focus on the Value of HERVs in Disease Prediction and Treatment. Virulence 2025, 16, 2523888. [Google Scholar] [CrossRef]
  237. Mantovani, F.; Kitsou, K.; Magiorkinis, G. HERVs: Expression Control Mechanisms and Interactions in Diseases and Human Immunodeficiency Virus Infection. Genes 2024, 15, 192. [Google Scholar] [CrossRef]
  238. Zhang, M.; Zheng, S.; Liang, J.Q. Transcriptional and Reverse Transcriptional Regulation of Host Genes by Human Endogenous Retroviruses in Cancers. Front. Microbiol. 2022, 13, 946296. [Google Scholar] [CrossRef]
  239. Stricker, E.; Peckham-Gregory, E.C.; Scheurer, M.E. HERVs and Cancer—A Comprehensive Review of the Relationship of Human Endogenous Retroviruses and Human Cancers. Biomedicines 2023, 11, 936. [Google Scholar] [CrossRef]
  240. Costa, B.; Vale, N. Exploring HERV-K (HML-2) Influence in Cancer and Prospects for Therapeutic Interventions. Int. J. Mol. Sci. 2023, 24, 14631. [Google Scholar] [CrossRef]
  241. Kozubek, P.; Kuźniar, J.; Czaja, M.; Sitka, H.; Kochman, U.; Leszek, J. Human Endogenous Retroviruses and Their Putative Role in Pathogenesis of Alzheimer’s Disease, Inflammation, and Senescence. Biomedicines 2025, 13, 59. [Google Scholar] [CrossRef]
  242. Saito, T.; Miyagawa, K.; Chen, S.-Y.; Tamosiuniene, R.; Wang, L.; Sharpe, O.; Samayoa, E.; Harada, D.; Moonen, J.-R.A.J.; Cao, A.; et al. Upregulation of Human Endogenous Retrovirus-K is Linked to Immunity and Inflammation in Pulmonary Arterial Hypertension. Circulation 2017, 136, 1920–1935. [Google Scholar] [CrossRef]
  243. Culley, M.K.; Chan, S.Y. Human Endogenous Retrovirus K and Pulmonary Arterial Hypertension. Circulation 2017, 136, 1936–1938. [Google Scholar] [CrossRef]
  244. Zhou, X.; Tian, W.; Gu, S.; Rabinovitch, M.; Nicolls, M.R.; Snyder, M.P. Microbiome–Immune Interaction in Pulmonary Arterial Hypertension: What Have We Missed? Research 2025, 8, 669. [Google Scholar] [CrossRef] [PubMed]
  245. Taylor, S.; Isobe, S.; Cao, A.; Contrepois, K.; Benayoun, B.A.; Jiang, L.; Wang, L.; Melemenidis, S.; Ozen, M.O.; Otsuki, S.; et al. Endogenous Retroviral Elements Generate Pathologic Neutrophils in Pulmonary Arterial Hypertension. Am. J. Respir. Crit. Care Med. 2022, 206, 1019–1034. [Google Scholar] [CrossRef] [PubMed]
  246. Otsuki, S.; Saito, T.; Taylor, S.; Li, D.; Moonen, J.-R.; Marciano, D.P.; Harper, R.L.; Cao, A.; Wang, L.; Ariza, M.E.; et al. Monocyte-Released HERV-K dUTPase Engages TLR4 and MCAM Causing Endothelial Mesenchymal Transition. JCI Insight 2021, 6, e146416. [Google Scholar] [CrossRef] [PubMed]
  247. Harper, R.L.; Zhou, X.; Marciano, D.P.; Cao, A.; Wang, L.; Chen, G.; Adil, M.S.; Zhou, W.; Maguire, P.; Deivanayagam, S.; et al. Altered Maturation and Activation State of Circulating Monocytes is Associated with Their Enhanced Recruitment in Pulmonary Arterial Hypertension. Respir. Res. 2025, 26, 148. [Google Scholar] [CrossRef]
  248. Hinojosa, W.; Cristo-Ropero, M.J.; Cruz-Utrilla, A.; Segura de la Cal, T.; López-Medrano, F.; Salguero-Bodes, R.; Pérez-Olivares, C.; Navarro, B.; Ochoa, N.; Arribas Ynsurriaga, F.; et al. The Impact of COVID-19 Pandemic on Pulmonary Hypertension: What Have We Learned? Pulm. Circ. 2022, 12, e12142. [Google Scholar] [CrossRef]
  249. Farha, S.; Heresi, G.A. COVID-19 and Pulmonary Arterial Hypertension: Early Data and Many Questions. Ann. Am. Thorac. Soc. 2020, 17, 1528–1530. [Google Scholar] [CrossRef]
  250. Bertici, N.S.; Dobrescu, L.; Pislaru, L.; Marincu, I. Controversies over the Impact of the COVID-19 Pandemic on Patients with Pulmonary Arterial Hypertension. Eur. Respir. J. 2021, 58, PA3587. [Google Scholar] [CrossRef]
  251. Tudoran, C.; Tudoran, M.; Lazureanu, V.E.; Marinescu, A.R.; Pop, G.N.; Pescariu, A.S.; Enache, A.; Cut, T.G. Evidence of Pulmonary Hypertension after SARS-CoV-2 Infection in Subjects without Previous Significant Cardiovascular Pathology. J. Clin. Med. 2021, 10, 199. [Google Scholar] [CrossRef]
  252. Soliman, Y.M.A.; Elkorashy, R.I.M.; Aziz, A.A.; Abdelnaby, A.; Magdy, S. Impact and Predictors of Outcome of COVID-19 in Pulmonary Hypertension Patients. Egypt. J. Bronchol. 2022, 16, 56. [Google Scholar] [CrossRef]
  253. Charif, F.; Dakroub, F.; Bou Akl, I.; Kularatne, M.; Montani, D. Pulmonary Arterial Hypertension and COVID-19: Piecing the Puzzle. Respir. Med. Res. 2023, 84, 101053. [Google Scholar] [CrossRef] [PubMed]
  254. Mamzer, A.; Waligora, M.; Kopec, G.; Ptaszynska-Kopczynska, K.; Kurzyna, M.; Darocha, S.; Florczyk, M.; Mroczek, E.; Mularek-Kubzdela, T.; Smukowska-Gorynia, A.; et al. Impact of the COVID-19 Pandemic on Pulmonary Hypertension Patients: Insights from the BNP-PL National Database. Int. J. Environ. Res. Public Health 2022, 19, 8423. [Google Scholar] [CrossRef] [PubMed]
  255. Al-Jahdhami, I.; Al-Mawali, A.; Bennji, S.M. Respiratory Complications after COVID-19. Oman Med. J. 2022, 37, e343. [Google Scholar] [CrossRef]
  256. Ambardar, S.R.; Hightower, S.L.; Huprikar, N.A.; Chung, K.K.; Singhal, A.; Collen, J.F. Post-COVID-19 Pulmonary Fibrosis: Novel Sequelae of the Current Pandemic. J. Clin. Med. 2021, 10, 2452. [Google Scholar] [CrossRef]
  257. Cueto-Robledo, G.; Porres-Aguilar, M.; Puebla-Aldama, D.; del Pilar Barragan-Martinez, M.; Jurado-Hernández, M.Y.; García-César, M.; Rojas, M.B.T.; García-Treminio, C.; Roldan-Valadez, E. Severe Pulmonary Hypertension: An Important Sequel After Severe Post-Acute COVID-19 Pneumonia. Curr. Probl. Cardiol. 2022, 47, 101004. [Google Scholar] [CrossRef] [PubMed]
  258. Oronsky, B.; Larson, C.; Hammond, T.C.; Oronsky, A.; Kesari, S.; Lybeck, M.; Reid, T.R. A Review of Persistent Post-COVID Syndrome (PPCS). Clin. Rev. Allergy Immunol. 2023, 64, 66–74. [Google Scholar] [CrossRef]
  259. 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]
  260. Khan, S.; Shafiei, M.S.; Longoria, C.; Schoggins, J.W.; Savani, R.C.; Zaki, H. SARS-CoV-2 Spike Protein Induces Inflammation via TLR2-Dependent Activation of the NF-κB Pathway. eLife 2021, 10, e68563. [Google Scholar] [CrossRef]
  261. Halajian, E.A.; LeBlanc, E.V.; Gee, K.; Colpitts, C.C. Activation of TLR4 by Viral Glycoproteins: A Double-Edged Sword? Front. Microbiol. 2022, 13, 1007081. [Google Scholar] [CrossRef]
  262. Asano, T.; Boisson, B.; Onodi, F.; Matuozzo, D.; Moncada-Velez, M.; Maglorius Renkilaraj, M.R.L.; Zhang, P.; Meertens, L.; Bolze, A.; Materna, M.; et al. X-Linked Recessive TLR7 Deficiency in ~1% of Men under 60 Years Old with Life-Threatening COVID-19. Sci. Immunol. 2021, 6, eabl4348. [Google Scholar] [CrossRef]
  263. Varga, Z.; Flammer, A.J.; Steiger, P.; Haberecker, M.; Andermatt, R.; Zinkernagel, A.S.; Mehra, M.R.; Schuepbach, R.A.; Ruschitzka, F.; Moch, H. Endothelial Cell Infection and Endotheliitis in COVID-19. Lancet 2020, 395, 1417–1418. [Google Scholar] [CrossRef]
  264. Ackermann, M.; Verleden, S.E.; Kuehnel, M.; Haverich, A.; Welte, T.; Laenger, F.; Vanstapel, A.; Werlein, C.; Stark, H.; Tzankov, A.; et al. Pulmonary Vascular Endothelialitis, Thrombosis, and Angiogenesis in COVID-19. N. Engl. J. Med. 2020, 383, 120–128. [Google Scholar] [CrossRef]
  265. Ejikeme, C.; Safdar, Z. Exploring the Pathogenesis of Pulmonary Vascular Disease. Front. Med. 2024, 11, 1402639. [Google Scholar] [CrossRef] [PubMed]
  266. Jadaun, P.K.; Chatterjee, S. COVID-19 and Dys-Regulation of Pulmonary Endothelium: Implications for Vascular Remodeling. Cytokine Growth Factor Rev. 2022, 63, 69–77. [Google Scholar] [CrossRef] [PubMed]
  267. Haberecker, M.; Schwarz, E.I.; Steiger, P.; Frontzek, K.; Scholkmann, F.; Zeng, X.; Höller, S.; Moch, H.; Varga, Z. Autopsy-Based Pulmonary and Vascular Pathology: Pulmonary Endotheliitis and Multi-Organ Involvement in COVID-19 Associated Deaths. Respiration 2022, 101, 155–165. [Google Scholar] [CrossRef] [PubMed]
  268. Ramos, S.G.; da Cruz Rattis, B.A.; Ottaviani, G.; Celes, M.R.N.; Dias, E.P. ACE2 Down-Regulation May Act as a Transient Molecular Disease Causing RAAS Dysregulation and Tissue Damage in the Microcirculatory Environment Among COVID-19 Patients. Am. J. Pathol. 2021, 191, 1154–1164. [Google Scholar] [CrossRef]
  269. Bourgonje, A.R.; Abdulle, A.E.; Timens, W.; Hillebrands, J.; Navis, G.J.; Gordijn, S.J.; Bolling, M.C.; Dijkstra, G.; Voors, A.A.; Osterhaus, A.D.; et al. Angiotensin-converting Enzyme 2 (ACE2), SARS-CoV-2 and the Pathophysiology of Coronavirus Disease 2019 (COVID-19). J. Pathol. 2020, 251, 228–248. [Google Scholar] [CrossRef]
  270. Gagliardi, S.; Hotchkin, T.; Tibebe, H.; Hillmer, G.; Marquez, D.; Izumi, C.; Chang, J.; Diggs, A.; Ezaki, J.; Suzuki, Y.J.; et al. The Renin–Angiotensin System Modulates SARS-CoV-2 Entry via ACE2 Receptor. Viruses 2025, 17, 1014. [Google Scholar] [CrossRef]
  271. Tiwari, N.R.; Phatak, S.; Sharma, V.R.; Agarwal, S.K. COVID-19 and Thrombotic Microangiopathies. Thromb. Res. 2021, 202, 191–198. [Google Scholar] [CrossRef]
  272. Safak, S.; Aksoy, E.; Dirim, A.B.; Demir, E.; Garayeva, N.; Oto, O.A.; Artan, A.S.; Yazici, H.; Besisik, S.; Turkmen, A. Successful Treatment of a COVID-19 Patient with Thrombotic Microangiopathy. Clin. Kidney J. 2021, 14, 1287–1288. [Google Scholar] [CrossRef]
  273. Baykara, Y.; Sevgi, K.; Akgun, Y. COVID-19 Microangiopathy: Insights into plasma exchange as a therapeutic strategy. Rev. Bras. Hematol. Hemoter. 2025, 47, 103963. [Google Scholar] [CrossRef]
  274. Smadja, D.M.; Mentzer, S.J.; Fontenay, M.; Laffan, M.A.; Ackermann, M.; Helms, J.; Jonigk, D.; Chocron, R.; Pier, G.B.; Gendron, N.; et al. COVID-19 is a Systemic Vascular Hemopathy: Insight for Mechanistic and Clinical Aspects. Angiogenesis 2021, 24, 755–788. [Google Scholar] [CrossRef] [PubMed]
  275. Chen, P.-Y.; Schwartz, M.A.; Simons, M. Endothelial-to-Mesenchymal Transition, Vascular Inflammation, and Atherosclerosis. Front. Cardiovasc. Med. 2020, 7, 53. [Google Scholar] [CrossRef]
  276. Zhang, L.; Tang, C.; Zhang, M.; Tong, X.; Xie, Y.; Yan, R.; Wang, X.; Zhang, X.; Liu, D.; Li, S. Single Cell Meta-Analysis of EndMT and EMT State in COVID-19. Front. Immunol. 2022, 13, 976512. [Google Scholar] [CrossRef]
  277. Ciszewski, W.M.; Woźniak, L.A.; Sobierajska, K. Diverse Roles of SARS-CoV-2 Spike and Nucleocapsid Proteins in EndMT Stimulation through the TGF-β-MRTF Axis Inhibited by Aspirin. Cell Commun. Signal 2024, 22, 296. [Google Scholar] [CrossRef]
  278. Baldassarro, V.A.; Alastra, G.; Cescatti, M.; Quadalti, C.; Lorenzini, L.; Giardino, L.; Calzà, L. SARS-CoV-2-Related Peptides Induce Endothelial-to-Mesenchymal Transition in Endothelial Capillary Cells Derived from Different Body Districts: Focus on Membrane (M) Protein. Cell Tissue Res. 2024, 397, 241–262. [Google Scholar] [CrossRef]
  279. Yamada, T.; Takaoka, A. Innate Immune Recognition against SARS-CoV-2. Inflamm. Regen. 2023, 43, 7. [Google Scholar] [CrossRef]
  280. Asaba, C.N.; Ekabe, C.J.; Ayuk, H.S.; Gwanyama, B.N.; Bitazar, R.; Bukong, T.N. Interplay of TLR4 and SARS-CoV-2: Unveiling the Complex Mechanisms of Inflammation and Severity in COVID-19 Infections. JIR 2024, 17, 5077–5091. [Google Scholar] [CrossRef]
  281. Xu, S.; Ilyas, I.; Weng, J. Endothelial Dysfunction in COVID-19: An Overview of Evidence, Biomarkers, Mechanisms and Potential Therapies. Acta Pharmacol. Sin. 2023, 44, 695–709. [Google Scholar] [CrossRef]
  282. Aljadah, M.; Khan, N.; Beyer, A.M.; Chen, Y.; Blanker, A.; Widlansky, M.E. Clinical Implications of COVID-19-Related Endothelial Dysfunction. JACC Adv. 2024, 3, 101070. [Google Scholar] [CrossRef]
  283. Oliveira, R.K.F.; Nyasulu, P.S.; Iqbal, A.A.; Hamdan Gul, M.; Ferreira, E.V.M.; Leclair, J.W.; Htun, Z.M.; Howard, L.S.; Mocumbi, A.O.; Bryant, A.J.; et al. Cardiopulmonary Disease as Sequelae of Long-Term COVID-19: Current Perspectives and Challenges. Front. Med. 2022, 9, 1041236. [Google Scholar] [CrossRef] [PubMed]
  284. Kauppert, C.A.; Dvorak, I.; Kollert, F.; Heinemann, F.; Jörres, R.A.; Pfeifer, M.; Budweiser, S. Pulmonary Hypertension in Obesity-Hypoventilation Syndrome. Respir. Med. 2013, 107, 2061–2070. [Google Scholar] [CrossRef] [PubMed]
  285. Montani, D.; Certain, M.-C.; Weatherald, J.; Jaïs, X.; Bulifon, S.; Noel-Savina, E.; Nieves, A.; Renard, S.; Traclet, J.; Bouvaist, H.; et al. COVID-19 in Patients with Pulmonary Hypertension: A National Prospective Cohort Study. Am. J. Respir. Crit. Care Med. 2022, 206, 573–583. [Google Scholar] [CrossRef] [PubMed]
  286. Riou, M.; Coste, F.; Meyer, A.; Enache, I.; Talha, S.; Charloux, A.; Reboul, C.; Geny, B. Mechanisms of Pulmonary Vasculopathy in Acute and Long-Term COVID-19: A Review. Int. J. Mol. Sci. 2024, 25, 4941. [Google Scholar] [CrossRef]
  287. Eroume, À. Egom, E.; Shiwani, H.A.; Nouthe, B. From Acute SARS-CoV-2 Infection to Pulmonary Hypertension. Front. Physiol. 2022, 13, 1023758. [Google Scholar] [CrossRef]
  288. Ciurzyński, M.; Kurzyna, M.; Kopeć, G.; Błaszczak, P.; Chrzanowski, Ł.; Kamiński, K.; Mizia-Stec, K.; Mularek-Kubzdela, T.; Biederman, A.; Zieliński, D.; et al. An Expert Opinion of the Polish Cardiac Society Working Group on Pulmonary Circulation on Screening for Chronic Thromboembolic Pulmonary Hypertension Patients after Acute Pulmonary Embolism: Update. Pol. Heart J. 2022, 80, 723–732. [Google Scholar] [CrossRef]
  289. Taha, H.A.; Elshafey, B.I.; Abdullah, T.M.; Salem, H.A. Study of Pulmonary Hypertension in Post-COVID-19 Patients by Transthoracic Echocardiography. Egypt. J. Bronchol. 2023, 17, 27. [Google Scholar] [CrossRef]
  290. Puk, O.; Nowacka, A.; Smulewicz, K.; Mocna, K.; Bursiewicz, W.; Kęsy, N.; Kwiecień, J.; Wiciński, M. Pulmonary Artery Targeted Therapy in Treatment of COVID-19 Related ARDS. Literature Review. Biomed. Pharmacother. 2022, 146, 112592. [Google Scholar] [CrossRef]
  291. Badagliacca, R.; Sciomer, S.; Petrosillo, N. Endothelin Receptor Antagonists for Pulmonary Arterial Hypertension and COVID-19: Friend or Foe? J. Heart Lung Transplant. 2020, 39, 729–730. [Google Scholar] [CrossRef]
  292. Nuche, J.; Pérez-Olivares, C.; de la Cal, T.S.; López-Guarch, C.J.; Ynsaurriaga, F.A.; Subías, P.E. Clinical Course of COVID-19 in Pulmonary Arterial Hypertension Patients. Rev. Española Cardiol. 2020, 73, 775–778. [Google Scholar] [CrossRef]
  293. Pęksa, J.W. Diagnosis and Treatment of Pulmonary Hypertension—Based on the 2022 ESC Guidelines. Lek. POZ 2023, 9, 7–15. [Google Scholar]
  294. Humbert, M.; Kovacs, G.; Hoeper, M.M.; Badagliacca, R.; Berger, R.M.F.; Brida, M.; Carlsen, J.; Coats, A.J.S.; Escribano-Subias, P.; Ferrari, P.; et al. 2022 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension: Developed by the Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS). Endorsed by the International Society for Heart and Lung Transplantation (ISHLT) and the European Reference Network on Rare Respiratory Diseases (ERN-LUNG). Eur. Heart J. 2022, 43, 3618–3731. [Google Scholar] [CrossRef]
Figure 1. TLR3 in PAH—signaling, vascular-neurological functions, endothelial phenotype, mediators, interventions, and the TLR3–p53–BMPR2 axis (based on [21,22,28,54,55,56,57,58,59,60,61,62,63,64,65,66,67]). Abbreviations: TLR3—Toll-like receptor 3, MyD88—Myeloid differentiation primary response 88, MyD88-independent—Signaling that does not require MyD88, TRIF—TIR-domain–containing adaptor-inducing interferon-β, TIR—Toll/Interleukin-1 receptor (domain), IFN-β—Interferon beta, IFN—interferons, CNS—Central nervous system, PAH—Pulmonary arterial hypertension, EC—Endothelial cell(s), PAEC—Pulmonary artery endothelial cell(s), IL-6—Interleukin-6, poly(I:C)—Polyinosinic: polycytidylic acid (synthetic dsRNA analog; TLR3 agonist, BMPR2—Bone morphogenetic protein receptor type 2, p53—Tumor protein p53, IRF3—Interferon regulatory factor 3, SU5416—Vascular endothelial growth factor receptor (VEGFR) inhibitor used to induce experimental PAH.
Figure 1. TLR3 in PAH—signaling, vascular-neurological functions, endothelial phenotype, mediators, interventions, and the TLR3–p53–BMPR2 axis (based on [21,22,28,54,55,56,57,58,59,60,61,62,63,64,65,66,67]). Abbreviations: TLR3—Toll-like receptor 3, MyD88—Myeloid differentiation primary response 88, MyD88-independent—Signaling that does not require MyD88, TRIF—TIR-domain–containing adaptor-inducing interferon-β, TIR—Toll/Interleukin-1 receptor (domain), IFN-β—Interferon beta, IFN—interferons, CNS—Central nervous system, PAH—Pulmonary arterial hypertension, EC—Endothelial cell(s), PAEC—Pulmonary artery endothelial cell(s), IL-6—Interleukin-6, poly(I:C)—Polyinosinic: polycytidylic acid (synthetic dsRNA analog; TLR3 agonist, BMPR2—Bone morphogenetic protein receptor type 2, p53—Tumor protein p53, IRF3—Interferon regulatory factor 3, SU5416—Vascular endothelial growth factor receptor (VEGFR) inhibitor used to induce experimental PAH.
Ijms 26 11143 g001
Figure 2. Key aspects of TLR4 involvement in pulmonary arterial hypertension (PAH)—based on expression, signaling mechanisms, cellular targets, vascular consequences, and translational implications (based on [24,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95]). Abbreviations: PAH—Pulmonary arterial hypertension, PAEC—Pulmonary artery endothelial cells, PASMC—Pulmonary artery smooth muscle cells, LPS—Lipopolysaccharide, HMGB1—High mobility group box 1 protein, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, EndMT—Endothelial-to-mesenchymal transition, ROS—Reactive oxygen species, Nox1/Nox4—NADPH oxidase isoforms 1 and 4, NADPH oxidase—Nicotinamide adenine dinucleotide phosphate oxidase, TXA2—Thromboxane A2, PDGF—Platelet-derived growth factor, TGF-β—Transforming growth factor beta, BMPR2—Bone morphogenetic protein receptor type 2, Smad1/5/8—SMAD family transcription factors 1, 5, and 8, Id1—Inhibitor of DNA binding 1, TMAO—Trimethylamine-N-oxide, HIF-1α—Hypoxia-inducible factor 1-alpha, ↑ means increase, ↓ means decrease, → denotes a relationship, a causal connection, or a transition to the next piece of information.
Figure 2. Key aspects of TLR4 involvement in pulmonary arterial hypertension (PAH)—based on expression, signaling mechanisms, cellular targets, vascular consequences, and translational implications (based on [24,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95]). Abbreviations: PAH—Pulmonary arterial hypertension, PAEC—Pulmonary artery endothelial cells, PASMC—Pulmonary artery smooth muscle cells, LPS—Lipopolysaccharide, HMGB1—High mobility group box 1 protein, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, EndMT—Endothelial-to-mesenchymal transition, ROS—Reactive oxygen species, Nox1/Nox4—NADPH oxidase isoforms 1 and 4, NADPH oxidase—Nicotinamide adenine dinucleotide phosphate oxidase, TXA2—Thromboxane A2, PDGF—Platelet-derived growth factor, TGF-β—Transforming growth factor beta, BMPR2—Bone morphogenetic protein receptor type 2, Smad1/5/8—SMAD family transcription factors 1, 5, and 8, Id1—Inhibitor of DNA binding 1, TMAO—Trimethylamine-N-oxide, HIF-1α—Hypoxia-inducible factor 1-alpha, ↑ means increase, ↓ means decrease, → denotes a relationship, a causal connection, or a transition to the next piece of information.
Ijms 26 11143 g002
Figure 3. Key Observations on TLR7/8 Activation in Experimental Pulmonary Arterial Hypertension (PAH) (based on [121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142]). Abbreviations: PAH—Pulmonary arterial hypertension, SU5416—Vascular endothelial growth factor receptor (VEGFR) inhibitor used to induce experimental PAH, s.c.—Subcutaneous, R848 (R-SU)—Resiquimod, synthetic imidazoquinoline and TLR7/8 agonist, HIF-1α—Hypoxia-inducible factor 1-alpha, mPAP—Mean pulmonary arterial pressure, RV—Right ventricle, MRI—Magnetic resonance imaging, RVEF—Right ventricular ejection fraction, IFNA1—Interferon alpha 1, IFNB1—Interferon beta 1, pDC—Plasmacytoid dendritic cells, IFN—Interferon, SLE—Systemic lupus erythematosus, ANA—Antinuclear antibodies, anti-dsDNA—Anti–double-stranded DNA antibodies, Th17—T helper 17 cells, Treg—Regulatory T cells, ↑ means increase; ↓ means decrease.
Figure 3. Key Observations on TLR7/8 Activation in Experimental Pulmonary Arterial Hypertension (PAH) (based on [121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142]). Abbreviations: PAH—Pulmonary arterial hypertension, SU5416—Vascular endothelial growth factor receptor (VEGFR) inhibitor used to induce experimental PAH, s.c.—Subcutaneous, R848 (R-SU)—Resiquimod, synthetic imidazoquinoline and TLR7/8 agonist, HIF-1α—Hypoxia-inducible factor 1-alpha, mPAP—Mean pulmonary arterial pressure, RV—Right ventricle, MRI—Magnetic resonance imaging, RVEF—Right ventricular ejection fraction, IFNA1—Interferon alpha 1, IFNB1—Interferon beta 1, pDC—Plasmacytoid dendritic cells, IFN—Interferon, SLE—Systemic lupus erythematosus, ANA—Antinuclear antibodies, anti-dsDNA—Anti–double-stranded DNA antibodies, Th17—T helper 17 cells, Treg—Regulatory T cells, ↑ means increase; ↓ means decrease.
Ijms 26 11143 g003
Figure 4. TLR9 in pulmonary arterial hypertension (PAH). (based on [144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167]). Abbreviations: PAH—Pulmonary arterial hypertension, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, IL-6—Interleukin-6, DAMP—Danger-associated molecular pattern, PAMP—Pathogen-associated molecular pattern, mtDNA—Mitochondrial DNA, CpG DNA—Cytosine-phosphate-guanine DNA motifs (unmethylated bacterial/viral DNA), MyD88—Myeloid differentiation primary response 88, AP-1—Activator protein 1, MCT-PAH—Monocrotaline-induced pulmonary arterial hypertension, RVSP—Right ventricular systolic pressure, TPVR—Total pulmonary vascular resistance, E6446—Selective TLR9 antagonist, Hb—Hemoglobin, EC—Endothelial cells, PASMC—Pulmonary artery smooth muscle cells, LDH—Lactate dehydrogenase, ↑ means increase, ↓ means decrease, → denotes a relationship, a causal connection, or a transition to the next piece of information.
Figure 4. TLR9 in pulmonary arterial hypertension (PAH). (based on [144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167]). Abbreviations: PAH—Pulmonary arterial hypertension, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, IL-6—Interleukin-6, DAMP—Danger-associated molecular pattern, PAMP—Pathogen-associated molecular pattern, mtDNA—Mitochondrial DNA, CpG DNA—Cytosine-phosphate-guanine DNA motifs (unmethylated bacterial/viral DNA), MyD88—Myeloid differentiation primary response 88, AP-1—Activator protein 1, MCT-PAH—Monocrotaline-induced pulmonary arterial hypertension, RVSP—Right ventricular systolic pressure, TPVR—Total pulmonary vascular resistance, E6446—Selective TLR9 antagonist, Hb—Hemoglobin, EC—Endothelial cells, PASMC—Pulmonary artery smooth muscle cells, LDH—Lactate dehydrogenase, ↑ means increase, ↓ means decrease, → denotes a relationship, a causal connection, or a transition to the next piece of information.
Ijms 26 11143 g004
Table 2. Comparison of the role of TLR1 and TLR2 in the regulation of inflammatory response, vascular remodeling, and hemodynamics in the pathogenesis of pulmonary arterial hypertension (PAH) (based on [39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]).
Table 2. Comparison of the role of TLR1 and TLR2 in the regulation of inflammatory response, vascular remodeling, and hemodynamics in the pathogenesis of pulmonary arterial hypertension (PAH) (based on [39,40,41,42,43,44,45,46,47,48,49,50,51,52,53]).
AspectTLR1TLR2
Regulation/expression in PAH (pre-clinical models)↓ Expression in the lungs of rats with MT-PAH; positive correlation with neutrophil and monocyte counts, negative correlation with resting CD4+ lymphocytes↑ Lung expression in MT-PAH; macrophage infiltration, activation of the TLR–RIPK3 axis, and dysregulation of TLR/NLR pathways promote progression of vascular remodeling
Compartments/target cells (inference from data)Modulation of the innate component (neutrophils, monocytes) and, indirectly, T lymphocytes; role in sensing pathogenic stimuliEndothelial cells (EC), pulmonary artery smooth muscle (PASMC), fibroblasts, leukocytes; high activity in the pulmonary vascular microenvironment
Signal pathsMainly MyD88 → NF-κB, induction of inflammatory mediators (inferred from TLR1/2 function)MyD88 → NF-κB; induction of IL-1, IL-6, TNF-α; association with RIPK3 and interactions with NLRs
Impact on key mechanisms for PAHIndirectly enhancing innate effector cell recruitment; potentially driving neutrophil/monocyte-dependent inflammatory responses↓ PASMC apoptosis; ↑ PASMC proliferation; ↑ TGF-β/PDGF; fibroblast activation and ECM deposition; ↑ oxidative stress; ↑ ICAM-1/VCAM-1 (facilitated leukocyte migration); EC dysfunction (permeability, ↓ NO)
Vascular/hemodynamic consequencesPotential involvement in the initiation and maintenance of inflammation affecting vascular remodelingIncreased vascular remodeling and inflammation; correlation with severity and deterioration of right ventricular function in animal models
Biomarkers and therapeutic implicationsCandidate immunoinfiltration biomarker (requires translational validation)Potential biomarker of inflammatory activity; modification of TLR2 activity considered a therapeutic target in PAH
Abbreviations: PAH—Pulmonary arterial hypertension, MT-PAH—Monocrotaline-induced pulmonary arterial hypertension (preclinical rat model), RIPK3—Receptor-interacting protein kinase 3, NLR—NOD-like receptor, EC—Endothelial cells, PASMC—Pulmonary artery smooth muscle cells, MyD88—Myeloid differentiation primary response 88, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, IL-1—Interleukin-1, IL-6—Interleukin-6, TNF-α—Tumor necrosis factor-alpha, TGF-β—Transforming growth factor-beta, PDGF—Platelet-derived growth factor, ECM—Extracellular matrix, ICAM-1—Intercellular adhesion molecule-1, VCAM-1—Vascular cell adhesion molecule-1, NO—Nitric oxide; ↑ means increase; ↓ means decrease.
Table 3. Comparative aspects of TLR5 and TLR6 in PAH (based on [15,42,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120]).
Table 3. Comparative aspects of TLR5 and TLR6 in PAH (based on [15,42,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120]).
AspectTLR5TLR6 (with TLR2)
Status of evidence in PAHNo dedicated functional studies have been conducted in PAH models; however, transcriptomic analyses indicate a significant induction of TLR5 in the lungs of patients and mice with PH.No direct clinical/animal studies in PAH; available RNA-seq analyses and data from stimulation of the TLR2/6 heterodimer in PASMCs.
Ligand(s)/recognitionBacterial flagellin.The TLR2/6 heterodimer recognizes diacylated bacterial lipopeptides (e.g., MALP-2).
Canonical signaling pathwayMyD88 → IRAK → NF-κB → TNF-α, IL-6, IL-8.MyD88 → NF-κB (via TLR2/6 heterodimer) → IL-1β, IL-6, TNF-α.
Cellular/vascular effects (inferred)Induction of inflammation similar to TLR2/TLR4: leukocyte recruitment, oxidative stress, vascular wall remodeling.Promotion of proliferation and vascular remodeling, an increase in endothelin-1, and persistence of inflammation in the pulmonary microenvironment.
Angiogenesis/repairNo data available.The TLR2/6 complex enhances angiogenesis (via GM-CSF) both in vitro and in vivo, supporting blood flow, immune cell recruitment, and tissue regeneration (e.g., in the liver).
Genetic associationsNo reports in the cited material.TLR6 A359T>C: predisposition to Legionnaires’ disease; Ser249Pro: possible association with asthma in selected populations.
Implications for PAHPotential pro-inflammatory and pro-remodeling role; requires verification in in vivo/in vitro studies.Potential therapeutic target: antagonism of signaling via the TLR2/6 → NF-κB axis to reduce inflammation and remodeling.
Abbreviations: PAH—Pulmonary arterial hypertension, PH—Pulmonary hypertension, RNA-seq—RNA sequencing, PASMC—Pulmonary artery smooth muscle cells, MyD88—Myeloid differentiation primary response 88, IRAK—Interleukin-1 receptor–associated kinase, NF-κB—Nuclear factor kappa-light-chain-enhancer of activated B cells, TNF-α—Tumor necrosis factor alpha, IL-6—Interleukin-6, IL-8—Interleukin-8, IL-1β—Interleukin-1 beta, Endothelin-1—Endothelin-1, GM-CSF—Granulocyte-macrophage colony-stimulating factor, MALP-2—Macrophage-activating lipopeptide-2, → denotes a relationship, a causal connection, or a transition to the next piece of information.
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

Styczeń, A.; Krysa, M.; Mertowska, P.; Grywalska, E.; Urbanowicz, T.; Krasiński, M.; Grobelna, M.; Topyła-Putowska, W.; Rahnama-Hezavah, M.; Tomaszewski, M. The Role of Toll-like Receptors and Viral Infections in the Pathogenesis and Progression of Pulmonary Arterial Hypertension—A Narrative Review. Int. J. Mol. Sci. 2025, 26, 11143. https://doi.org/10.3390/ijms262211143

AMA Style

Styczeń A, Krysa M, Mertowska P, Grywalska E, Urbanowicz T, Krasiński M, Grobelna M, Topyła-Putowska W, Rahnama-Hezavah M, Tomaszewski M. The Role of Toll-like Receptors and Viral Infections in the Pathogenesis and Progression of Pulmonary Arterial Hypertension—A Narrative Review. International Journal of Molecular Sciences. 2025; 26(22):11143. https://doi.org/10.3390/ijms262211143

Chicago/Turabian Style

Styczeń, Agnieszka, Martyna Krysa, Paulina Mertowska, Ewelina Grywalska, Tomasz Urbanowicz, Maciej Krasiński, Malwina Grobelna, Weronika Topyła-Putowska, Mansur Rahnama-Hezavah, and Michał Tomaszewski. 2025. "The Role of Toll-like Receptors and Viral Infections in the Pathogenesis and Progression of Pulmonary Arterial Hypertension—A Narrative Review" International Journal of Molecular Sciences 26, no. 22: 11143. https://doi.org/10.3390/ijms262211143

APA Style

Styczeń, A., Krysa, M., Mertowska, P., Grywalska, E., Urbanowicz, T., Krasiński, M., Grobelna, M., Topyła-Putowska, W., Rahnama-Hezavah, M., & Tomaszewski, M. (2025). The Role of Toll-like Receptors and Viral Infections in the Pathogenesis and Progression of Pulmonary Arterial Hypertension—A Narrative Review. International Journal of Molecular Sciences, 26(22), 11143. https://doi.org/10.3390/ijms262211143

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