1,523
Views
0
CrossRef citations to date
0
Altmetric
Immunotherapy - Other

From defense to offense: Modulating toll-like receptors to combat arbovirus infections

ORCID Icon, , , &
Article: 2306675 | Received 08 Sep 2023, Accepted 14 Jan 2024, Published online: 23 Jan 2024

ABSTRACT

Arboviruses are a significant threat to global public health, with outbreaks occurring worldwide. Toll-like receptors (TLRs) play a crucial role in the innate immune response against these viruses by recognizing pathogen-associated molecular patterns and initiating an inflammatory response. Significantly, TLRs commonly implicated in the immune response against viral infections include TLR2, TLR4, TLR6, TLR3, TLR7, and TLR8; limiting or allowing them to replicate and spread within the host. Modulating TLRs has emerged as a promising approach to combat arbovirus infections. This review summarizes recent advances in TLR modulation as a therapeutic target in arbovirus infections. Studies have shown that the activation of TLRs can enhance the immune response against arbovirus infections, leading to increased viral clearance and protection against disease. Conversely, inhibition of TLRs can reduce the excessive inflammation and tissue damage associated with arbovirus infection. Modulating TLRs represents a potential therapeutic strategy to combat arbovirus infections.

Introduction

Toll-like receptors

Toll-like receptors (TLRs) were first discovered in 1997 and the discovery was a significant breakthrough in immunology, as it provided insights into how the immune system recognizes and responds to pathogens.Citation1 Hoffmann and his colleagues were studying the innate immune system of fruit flies (Drosophila melanogaster) when they found that a gene called Toll, which had previously been known for its role in embryonic development in fruit flies, also played a role in the immune response.Citation1 Observation of the mutations in the Toll gene resulted in increased susceptibility to fungal infections in fruit flies and this led to the investigation of Toll function in the innate immune response of other organisms, including humans.Citation2 Although toll-like receptors are highly expressed on innate immune cells such as macrophages and neutrophils, they are also present in certain nonimmune cells including epithelial cells.Citation3 These receptors recognize pathogen-associated molecular patterns (PAMPs), such as viral structural proteins, lipopolysaccharides, and flagellin, which are present on the surface of many types of bacteria and viruses.Citation4 The activation of TLRs triggered an immune response that included the production of cytokines, chemokines, and other inflammatory mediators.Citation5 The discovery of TLRs opened new avenues for understanding the mechanisms underlying innate immunity and paved the way for the development of new strategies for combating infectious diseases.

In humans, 10 known TLRs have been identified so far, and classified into two groups namely transmembrane and intracellular TLRs. The former comprises TLR1, TLR2, TLR4, TLR5, TLR6 and TLR10 whilst the latter consists of TLR3, TLR7, TLR8 and TLR9.Citation6 Each receptor has a distinct role in the recognition and response to different types of pathogens.Citation7 TLRs function by recognizing specific PAMPs that are present on the surface of various pathogens, such as bacteria, viruses, and fungi.Citation8

TLRs on the cell surface predominantly recognize membrane components of the said microbes. To illustrate, TLR2 forms dimers with TLR1 or TLR6 to recognize a diverse array of PAMPs including peptidoglycans, lipoproteins, mannan, and fungal zymosan.Citation9 On the other hand, intracellular TLRs in the endosomes recognize nucleic acid from viruses and bacteria, in addition to self-nucleic acids from damaged cells known as damage-associated molecular patterns (DAMPs).Citation10 provides an overview of the natural ligands associated with the respective TLRs.

Table 1. The natural ligands associated with each TLRs.

When a TLR recognizes a PAMP, it triggers a signaling cascade within the immune cell that leads to the activation of transcription factors and the production of cytokines and chemokines, which help to coordinate an immune response against the invading pathogen.Citation19 TLRs are made up of several different structural domains, including a transmembrane domain, an extracellular domain, and an intracellular signaling domain.Citation20 The extracellular domain of the TLR is responsible for recognizing and binding to PAMPs.Citation21 This domain is composed of leucine-rich repeats (LRRs), which form a horseshoe-shaped structure that is thought to interact with the PAMPs.Citation22

The intracellular signaling domain of the TLR is responsible for initiating an immune response once the TLR has bound to a PAMP.Citation19 This domain is composed of a Toll/interleukin-1 receptor (TIR) domain, which interacts with downstream signaling molecules to activate immune cells and trigger the release of cytokines.Citation23 In addition to the LRRs and TIR domain, some TLRs also contain additional domains that can help to enhance their activity. For example, TLR4 has a co-receptor called MD-2 that can help to bind to lipopolysaccharide (LPS), a component of the outer membrane of Gram-negative bacteria.Citation24 As previously mentioned, TLRs also play a role in the recognition of endogenous molecules that are released by damaged or stressed cells, known as DAMPs.Citation21 This recognition can lead to inflammation and tissue repair processes.Citation25

TLRs work by using adaptor molecules to initiate downstream signaling cascades. The signaling cascades initiated by TLRs and their adaptor molecules are complex and involve multiple steps. There are several different adaptor molecules that can be used by TLRs, but the two main ones are MyD88 and TRIF.Citation26 When a TLR recognizes a PAMP on the surface of a pathogen, it undergoes a conformational change that allows it to recruit an adaptor molecule to its intracellular signaling domain.Citation27,Citation28 The adaptor molecule then interacts with downstream signaling molecules, leading to the activation of transcription factors and the expression of genes involved in the immune response.Citation27,Citation28 The adaptor molecule MyD88 is used by most TLRs, apart from TLR3, which uses the adaptor molecule TRIF.Citation25 Once recruited to the TLR, MyD88 interacts with the interleukin-1 receptor-associated kinase (IRAK) family of proteins, leading to their activation and subsequent phosphorylation of the transcription factor NF-κB. This results in the expression of pro-inflammatory cytokines, such as interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α).Citation27–30

In contrast, TRIF recruits another downstream signaling molecule called TRAF3, which leads to the activation of interferon regulatory factor 3 (IRF3) and the expression of type I interferons (IFN-α and IFN-β).Citation31 These interferons play a crucial role in the antiviral immune response by inducing the expression of genes involved in viral clearance and the activation of immune cells.Citation32 In addition to MyD88 and TRIF, there are several other adaptor molecules that can be used by TLRs, such as TRAM and TIRAP.Citation33 These adaptor molecules can also interact with downstream signaling molecules to activate various immune responses.Citation33 Overall, TLRs are important components of the innate immune system, helping to identify and respond to a wide range of potential pathogens and maintain tissue homeostasis.

Arboviruses

Viruses, including arboviruses, are small obligate intracellular parasites, relying on host cells for multiplication and subsequent spread of infection within the host. The seven stages of viral replication are attachment, penetration, uncoating, replication, assembly, maturation, and release. This interaction with host cells inevitably triggers the activation of TLRs via the various viral PAMPs at different stages.Citation34 Arboviruses are RNA viruses that are transmitted to humans and other animals by arthropods, such as mosquitoes, ticks, and sandflies.Citation35 The term “arbovirus” is derived from “arthropod-borne virus.” Arboviruses can cause a range of illnesses in humans, from mild febrile illnesses to severe neurological diseases.Citation36 Examples of arboviruses that affect humans include the dengue virus (DENV), Yellow Fever virus (YFV), Zika virus (ZIKV), West Nile virus (WNV), and Chikungunya virus (CHIKV).Citation37 The geographic distribution of arboviruses is determined by the distribution of the arthropod vectors that transmit them. DENV from the family Flaviviridae is a positive single-strand RNA. It is transmitted by Aedes mosquitoes and is endemic in tropical and subtropical regions around the world, particularly in Southeast Asia, the Western Pacific, and the Americas.Citation38 DENV can cause a range of clinical symptoms, from mild febrile illness to severe dengue hemorrhagic fever and dengue shock syndrome, which can be fatal.Citation38 Dengue virus infections are caused by any of the four serotypes (DENV 1–4) of the virus. However, when someone gets infected by a certain serotype, they will only gain lifelong immunity to that specific serotype and not to others.Citation39

ZIKV is another single-stranded positive-sense RNA virus that belongs to the family Flaviviridae.Citation40 This virus has evolved into two genotypes, namely the African and Asian genotypes.Citation41 It is transmitted by Aedes mosquitoes and has been responsible for recent outbreaks in the Americas, Africa, and Asia.Citation34 ZIKV infection is usually mild and self-limiting, but it can cause neurological complications, such as Guillain-Barré syndrome and congenital Zika syndrome, which can result in severe birth defects in newborns.Citation42 CHIKV is an enveloped, positive single-stranded RNA virus from the family Togaviridae.Citation43 Three major genotypes have been identified which are West African, East/Central/South African (ECSA), and Asian genotypes.Citation44 It is transmitted by Aedes mosquitoes and is endemic in Africa, Asia, and the Indian subcontinent.Citation45 In recent years, CHIKV has caused large outbreaks in the Caribbean and South America.Citation45 CHIKV infection can cause severe joint pain, fever, rash, and in some cases, long-term joint pain, and disability.Citation45 WNV from the family Flaviviridae is an enveloped, positive single-strand RNA, and is transmitted by Culex mosquitoes and is found in Africa, Europe, the Middle East, and North America.Citation46,Citation47 WNV infection is usually asymptomatic or mild, but in some cases, it can cause severe neurological diseases, such as encephalitis and meningitis.Citation47 Belonging to the family Flaviviridae, Yellow fever virus (YFV) shares the characteristic of being an enveloped, single-stranded positive RNA virus.Citation48 Thus far, 7 genotypes have been described including five African lineages and two South American.Citation49 It is transmitted by Aedes and Haemagogus mosquitoes and is endemic in tropical regions of Africa and South America.Citation50 YFV infection can cause a range of clinical symptoms, from mild febrile illness to severe hemorrhagic fever and liver failure, which can be fatal.Citation50

The geographical distribution and impact of arboviruses vary depending on the specific virus and its transmission cycle, the distribution of the arthropod vector, and the susceptibility of the human population.Citation51 There are several factors that have contributed to the spread of arboviruses by their vectors, such as globalization and increased travel, urbanization and population growth, climate change, and changes in land use.Citation51 Various measures have been taken to control the spread of arboviruses and their vectors including vector control, vaccine and therapeutic development, surveillance, and early warning systems, public education, and behavior change as well as research and innovation.Citation52 Despite these measures, the continued emergence and spread of arboviruses highlight the need for sustained investment in research, surveillance, and public health interventions to address this ongoing global health threat.

Methods

Literature was searched in Web of Science, PubMed, and Google Scholar, using the combinations (benefitting from Boolean operators: AND, OR) of the following search terms: TLR, arbovirus, DENV, ZIKV, WNV, CHIKV, YFV, innate immunity, antiviral, agonist, and antagonist. The references were checked from all sources to find eligible articles. The reference lists of all articles published in peer-reviewed journals selected were reviewed 31,108 not published in English were excluded, all the duplicates were excluded, and a total of 72,347 references were identified. Among these eligible references, only 22 summarized or investigated the pathway system of toll-like receptors, 17 discussed the general information about arboviruses, 19 offered detailed research addressing the interplay between innate immunity on DENV, 16 on ZIKV, 9 on WNV, 14 on CHIKV, and 8 on YFV. Also, 13 publications discussed innate immunity in general, 7 on its perspective to develop antivirals, 13 studies on existing agonists, and 13 on existing antagonists. (). A total of 123 high-quality research results published in authoritative journals were summarized and preferred cited in the review. In addition to searching sources in Web of Science, PubMed, and Google Scholar, we actively tracked the latest updates on outbreaks and case progress by regularly visiting the websites of reputable organizations such as the World Health Organization (WHO), the United States Centers for Disease Control and Prevention (CDC), and the Ministry of Health, Malaysia (MOH).

Figure 1. Flowchart of database search, screening, and selection process. This figure depicts the systematic approach used to identify, screen, and select relevant studies for inclusion in the review manuscript.

Figure 1. Flowchart of database search, screening, and selection process. This figure depicts the systematic approach used to identify, screen, and select relevant studies for inclusion in the review manuscript.

The relationship between TLR and arbovirus infection

TLRs elicit immune responses during viral infections by activating their respective signaling pathways which promotes the production and release of inflammatory cytokines and chemokines.Citation53 Despite their important role in producing the protective effect of immune defenses, exaggerated activation may lead to undesirable and harmful effects such as in West Nile Virus, where TLR3-dependent inflammation lead to lethal encephalitis.Citation54

Previous research has established that during dengue virus infection, the signaling pathways of TLR2 and TLR6 were up-regulated via the presence of DENV non-structural protein 1 (NS1), which is essential for virus replication. This in turn increases the IL-6 and TNF-α production by peripheral blood mononuclear cells (PBMC) which is attributed to increased vascular permeability and leakage.Citation55 On the contrary, another study suggested that instead of the aforementioned TLR2 and TLR6, TLR4 is the receptor that triggers the inflammatory cytokine release in the presence of NS1.Citation56 Additionally, studies have shown that TLR3,Citation57–60 TLR7,Citation57,Citation58,Citation61–63 and TLR8Citation57,Citation64–66 are involved in the recognition of DENV RNA and the activation of downstream signaling pathways, leading to the production of antiviral cytokines and type I interferons (IFN). IL-8 and distinct IFNα/β release are triggered after viral recognition in human monocytic cells by TLR3 following endosomal acidification.Citation57 Strong signaling through TLR3 and RIG-1, but not Mda5, was experienced by skin fibroblasts infected with DENV-2, leading to over-expression of IFNβ, TNFα, defensin 5 (HB5), and β-defensin 2 (HβD2).Citation58 Moreover, DENV-infected fibroblasts displayed enhanced nuclear translocation of IFN regulatory factor 3 (IRF3) but not interferon regulatory factor 7 (IRF7) compared to mock-infected fibroblasts.Citation58 The TLR-induced response was specifically characterized by increases in myeloid DC subset activation together with increased serum levels of CXCL-10 and IL-1Ra.Citation59 An experimental indication that poly (I:C) may be a promising immunomodulatory drug against DENV infection and might be useful for clinical prophylaxis is that TLR3 activation is efficient in preventing DENV2 replication via IFN-β.Citation60 It is likely that certain genotypes of TLR7 and 8 polymorphisms contribute to elevated dengue/chikungunya virus loads in patients who are also infected with those diseases.Citation61 Uncleaved prM-containing immature particles may help to control the course of dengue virus infection by functioning as a carrier to the endolysosome-localized TLR7 sensor.Citation62 As a first line of innate defense against DENV infection in vivo, TLR7‘s RNA helicase-mediated sensing works in a tissue-dependent manner.Citation63

However, some studies have suggested that DENV can also evade or modulate the TLR pathway to promote viral replication and evasion of the host immune response. For example, DENV can induce the expression of suppressor of cytokine signaling (SOCS) proteins, which can inhibit the TLR signaling pathway and the production of antiviral cytokines.Citation67 One of the mechanisms used by DENV involves the expression of viral proteins that can interfere with the TLR signaling pathway. For example, DENV non-structural protein 4B (NS4B) has been shown to inhibit the TLR3-mediated activation of interferon regulatory factor 3 (IRF3) and the production of type I interferons.Citation68 DENV NS1 can also inhibit the TLR3-mediated induction of interferon-stimulated genes (ISGs) and the production of type I interferons.Citation69 In addition, the DENV envelope (E) protein has been shown to inhibit TLR4-mediated activation of NF-κB and the production of pro-inflammatory cytokines, such as IL-6 and TNF-α.Citation70 These viral proteins may interfere with the TLR pathway to allow for increased viral replication and evasion of the host immune response.

In the case of ZIKV infection, TLR3 and TLR7 have been found to recognize the viral RNA, initiating a signaling cascade that leads to the production of pro-inflammatory cytokines and interferons.Citation71,Citation72 TLR3 recognizes double-stranded RNA, which is a viral replication intermediate, while TLR7 recognizes single-stranded RNA, such as that produced during viral transcription. Recent studies suggest that TLRs may also play a role in the pathogenesis of ZIKV infection, with aberrant activation of TLR signaling pathways contributing to disease severity.Citation73 The mechanisms by which ZIKV proteins interact with TLR pathways are complex and not fully understood. The NS5 protein of ZIKV has been shown to inhibit TLR3-mediated signaling, which is involved in the innate immune response to viruses.Citation74 This inhibition is thought to help the virus evade detection by the host immune system. The NS3 protein of ZIKV has also been shown to interact with TLR3, TLR7, and TLR8.Citation75 This interaction leads to the activation of the NF-κB and IRF3 pathways, which are involved in the production of pro-inflammatory cytokines and type I interferons. These cytokines and interferons are important for the clearance of viral infections. On the other hand, the ZIKV envelope protein (E protein) has been shown to activate the TLR2 pathway, leading to the induction of pro-inflammatory cytokines.Citation76 This activation is thought to contribute to the immune response to ZIKV infection and can help limit viral replication.Citation76

WNV is known to activate TLR pathways in infected cells. The viral envelope protein (E protein) of WNV has been shown to activate the TLR3 pathway, leading to the production of interferons and other pro-inflammatory cytokines.Citation77 These cytokines play a role in the host’s immune response to viral infection, limiting viral replication. The WNV NS5 protein has been shown to interact with the TLR3 pathway, inhibiting the production of type I interferons.Citation78 This is thought to help the virus evade detection by the host immune system, and contribute to the pathogenesis of WNV infection. The WNV NS4B protein has also been shown to interact with the TLR3 pathway, inhibiting the activation of IRF3, a transcription factor involved in the production of type I interferons.Citation79 This interaction is thought to contribute to the ability of WNV to evade the host immune response and establish infection.

Research has shown that the CHIKV E2 protein interacts with TLR2, leading to the production of pro-inflammatory cytokines such as IL-6 and TNF-α.Citation80 In addition, CHIKV has been shown to activate the TLR4 pathway, leading to the production of pro-inflammatory cytokines and type I interferons.Citation81 CHIKV may “hijack” the TLR pathways in infected cells to evade the host immune response.Citation82 For example, the CHIKV nsP2 protein has been shown to interact with the TLR3 pathway, inhibiting the production of type I interferons.Citation83 This is thought to help the virus evade detection by the host immune system and contribute to the pathogenesis of CHIKV infection. Other CHIKV proteins, including nsP3 and E2, have been shown to interact with the TLR7/8 pathway, which plays a role in the recognition of single-stranded RNA viruses.Citation84 This interaction may contribute to the immune response to CHIKV infection.

YFV envelope protein E is the main protein responsible for the activation of TLRs. The current Yellow Fever vaccine utilizes a weakened form of the yellow fever virus, which can trigger various TLRs and generate a well-balanced Th1/Th2 response in dendritic cells of mice. Notably, when TLR2 is absent in knockout mice, there is a significant enhancement in Th1 responses, coupled with reduced production of IL-6 and IL-12.Citation85 Additionally, the YFV E protein has been shown to activate the TLR3 pathway, leading to the production of interferon-beta (IFN-β). It appears that the YFV E protein can activate TLRs directly without the need for accessory molecules.Citation86 The activation of TLRs by the YFV E protein seems to occur through the recognition of PAMPs present in the virus, such as viral RNA or glycoproteins. This leads to the activation of downstream signaling pathways, including the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways.Citation86 Overall, it appears that the YFV E protein can hijack or navigate TLR pathways to its liking, leading to the production of pro-inflammatory cytokines and IFN-β.Citation87 This can help the virus to evade host immune responses and establish infection in the host.

Understanding the mechanisms by which DENV, ZIKV, WNV, CHIKV, and YFV can evade or modulate the TLR pathway (as represented in ) may help in the development of new strategies to prevent or treat DENV infection.

Figure 2. Evasion and modulation of TLR pathway by the arboviruses. This figure illustrates the intricate mechanisms by which these arboviruses evade or modulate the TLR pathway during infection. Also included are the natural ligands for respective TLRs.

Figure 2. Evasion and modulation of TLR pathway by the arboviruses. This figure illustrates the intricate mechanisms by which these arboviruses evade or modulate the TLR pathway during infection. Also included are the natural ligands for respective TLRs.

TLR agonists as adjuvants for arbovirus vaccines

Many vaccines contain a component called adjuvants that increases the efficacy of the vaccine by stimulating the immune response more effectively. These include mineral salts, emulsions, and immune potentiators such as TLR agonists.Citation88 TLR agonists trigger the TLRs to activate the innate immune response and promote immunity against infectious diseases, cancer, and other conditions.Citation89 TLR agonists have been investigated as potential vaccine adjuvants, as they can enhance the immune response to the vaccine antigen and improve vaccine efficacy.Citation90 In addition, TLR agonists have been studied as potential therapies for cancer and other diseases.Citation91 For example, TLR7 and TLR9 agonists have shown promise in the treatment of some cancers (e.g. melanoma and lymphomas), as they can activate immune cells to recognize and attack tumor cells.Citation92,Citation93 This extends to previous studies having explored the usage of TLR9 agonist as monotherapy for skin cancers and hematologic malignancies, as well as in combination with other therapies such as radiotherapy and certain chemotherapies.Citation75 Imiquimod (IMQ), a TLR7 agonist demonstrated this by increasing the expression of autophagy-related genes and autophagosomes, leading to cell death in melanoma cells.Citation74 The innate immune response toward cancer and viruses exhibits parallels in their mechanisms, focusing on the recognition and removal of abnormal or potentially harmful cells- specifically, malignant cells and viral pathogens. Consistent with the immune response toward cancer, TLR-assisted cell death of the virus-infected cells, may restrict further virus multiplication in the host.Citation94 TLR agonists have also been investigated for their potential use in treating autoimmune and inflammatory disorders.Citation95

There are TLR agonists that have been approved to treat medical conditions. Imiquimod is a TLR7 agonist that is approved for the topical treatment of certain skin conditions, including genital warts and actinic keratoses.Citation95,Citation96 Resiquimod is a TLR7 and TLR8 agonist that is approved for the topical treatment of external genital and perianal warts caused by human papillomavirus (HPV).Citation97 Bacillus Calmette-Guérin (BCG) is a vaccine that contains a TLR2 and TLR4 agonist and is used to prevent tuberculosis.Citation98 MEDI9197 is a TLR7 agonist that is being studied as a potential treatment for advanced solid tumors.Citation99 VTX-2337 is a TLR8 agonist that is being studied as a potential treatment for various types of cancer.Citation100

Vaccines are an effective way to prevent arbovirus infections, and several arbovirus vaccines are currently available. However, some of these vaccines have limitations, such as a lack of efficacy against all strains of the virus, the need for multiple doses, and the risk of adverse effects.Citation101 One strategy to overcome these limitations is to use adjuvants. TLR agonists are one type of adjuvant that has been investigated for use in arbovirus vaccines.Citation102–104 TLR agonists can enhance the immune response to arbovirus vaccines by activating innate immune cells, such as dendritic cells, to produce cytokines and chemokines that promote the activation of antigen-specific T cells and B cells.Citation105 TLR agonists can also enhance the production of antibodies against the vaccine antigen, which is an important component of the immune response to arboviruses.Citation105

Several TLR agonists have been studied as adjuvants for arbovirus vaccines. For example, TLR9, TLR2/1, and TLR7/8 agonists have been shown to enhance the immune response to the live-attenuated YFV vaccine 17D (YF-17D). A phase 3 observer-blind, placebo-controlled randomized study involving 900 participants took place at 11 sites across the US from February 2018 to May 2019. The administration of YF-17D vaccine and TAK-003, whether sequentially or concomitantly, proved to be immunogenic and well-tolerated.Citation85 In addition, a combination of TLR4 agonists and saponin adjuvants have been shown to enhance the immune response to the WNV vaccine in Syrian hamsters.Citation106 The use of TLR agonists as adjuvants for arbovirus vaccines has several potential benefits. First, TLR agonists can enhance the efficacy of existing vaccines, which could lead to better protection against arbovirus infections. Second, TLR agonists can reduce the number of vaccine doses needed, which could improve vaccine coverage and compliance. Third, TLR agonists could be used to develop new vaccines against arboviruses that are currently difficult to vaccinate against.

However, there are also some potential challenges and risks associated with the use of TLR agonists as adjuvants for arbovirus vaccines. For example, some TLR agonists can cause inflammation and adverse reactions, and the optimal dose and timing of TLR agonist administration may need to be carefully considered to avoid these effects.Citation107 The use of TLR agonists as adjuvants for arbovirus vaccines is an active area of research that holds promise for improving the prevention and control of arbovirus infections. Further studies are needed to fully understand the potential benefits and risks of TLR agonists as adjuvants for arbovirus vaccines and to develop safe and effective TLR agonist-based vaccine formulations.

TLR antagonists as therapeutic agents for arbovirus infection

TLR antagonists are substances that inhibit the activity of TLRs to modulate the immune response to infectious agents by blocking the activation of TLRs.Citation108 By inhibiting the activity of TLRs, TLR antagonists can reduce inflammation and tissue damage that can occur as a result of an excessive or dysregulated immune response to infection. TLR antagonists can also potentially reduce the severity of infections caused by certain pathogens, such as bacteria and viruses.

There are several different types of TLR antagonists that have been developed and studied for various applications. Small molecule inhibitors are synthetic compounds that bind to TLRs and block their activity. Small molecule inhibitors have been developed for various TLRs, including TLR4 and TLR7, and they have been studied for their potential to treat infectious diseases, inflammatory disorders, and cancer.Citation109 Antibodies that target TLR2 and TLR4 have been developed and studied for their potential to treat sepsis, a life-threatening condition that can occur as a result of an overwhelming immune response to infection.Citation110 In particular, eritoran tetrasodium (E5564) is suggested to completely block cellular activation by LPS via TLR4 signaling, hence reducing the following production of pro-inflammatory cytokines and chemokines.Citation110 Some natural compounds, such as curcumin and epigallocatechin gallate (EGCG), have been shown to have TLR antagonistic activity.Citation111,Citation112 These compounds have been studied for their potential to treat inflammatory disorders and cancer by way of cell death induction including apoptosis and autophagy.Citation111,Citation112 This can be briefly illustrated by EGCG inhibiting NF-κB activity through the TLR4 signaling pathway and activating caspases, resulting in the apoptosis of epidermoid carcinoma cells.Citation111

Currently, there are no TLR antagonists that have been approved by regulatory agencies for the treatment of medical conditions. However, several TLR antagonists have been developed and studied in preclinical and clinical trials for various applications. A TLR4 antagonist called eritoran tetrasodium has been studied in clinical trials for the treatment of sepsis.Citation113 Although early clinical trials showed promising results, larger phase III clinical trials failed to demonstrate a significant improvement in patient outcomes.Citation113 Another TLR4 antagonist called TAK-242 has been studied in preclinical and clinical trials for the treatment of various inflammatory conditions, such as rheumatoid arthritisCitation114 and Crohn’s disease.Citation115

TLR7 antagonists have also been developed and studied for the treatment of viral infections, such as hepatitis B and C. One TLR7 antagonist called GS-9620 has shown promising results in preclinical and clinical trials for the treatment of chronic hepatitis B infection.Citation116 While TLR antagonists have shown promise in preclinical and early clinical trials for the treatment of various medical conditions, more research is needed to fully understand their efficacy, safety, and potential side effects. Further clinical trials are needed to determine whether TLR antagonists can effectively treat various diseases. A TLR7 antagonist, IRS661, was found to reduce IFN-I and other cytokines in the lung by acting through pDCs and monocytes, which resulted in a reduction of inflammation and its intensity.Citation117 Moreover, inhibiting TLR7 demonstrated a reduction in mortality and inflammation in the mouse model’s infected lung, specifically by diminishing neutrophil recruitment and monocyte-produced chemo-attractants, even in the absence of IFN-I signaling.Citation117

A TLR4 antagonist, eritoran (also known as E5564), was found to block pulmonary pathology, clinical symptoms, cytokine and oxidized phospholipid expression, influenza-induced mortality in mice, and lower viral titers.Citation118 It was also discovered that eritoran-mediated protection required CD14 and TLR2, as CD14 directly bound eritoran and prevented ligand binding to MD2.Citation118 Eritoran in addition to another TLR4 antagonist, Rhodobacter sphaeroides lipopolysaccharide, attached to a hydrophobic region in MD-2 and dose-dependently inhibited RSV F-protein-mediated TLR4 activity in HEK293T-TLR4-CD14-MD-2 transfectants through TNF-mediated signaling.Citation119

Convinced by the inhibitory effects of TLR antagonists against other viruses, they have the potential to revolutionize the treatment of arbovirus infections. Further research is needed to utilize TLR antagonists for the treatment of arbovirus infections in humans and to fully understand the efficacy, safety, and potential side effects of TLR antagonists as therapeutic agents for arbovirus infections. Clinical trials are needed to determine whether TLR antagonists can be effective treatments for arbovirus infections in humans.

Challenges and future direction for TLR-based strategies against arbovirus infection

The use of TLR-based strategies against arboviral and other viral infections has shown promising results in preclinical and clinical studies (), but several challenges need to be addressed before their clinical application. One of the main challenges is the complexity of arboviral infections. Arboviruses have complex life cycles, involving both the insect and mammalian hosts, and they have evolved to evade the host’s immune response.Citation120 Different arboviral species have different mechanisms of infection and immune evasion, making it difficult to develop a universal TLR-based strategy. Therefore, it is essential to develop a detailed understanding of the immune response to specific arboviruses and identify the PAMPs recognized by TLRs to design effective TLR-based strategies.

Table 2. TLR-based strategies against arboviral and other viral infections in preclinical and clinical studies.

Another challenge is the potential side effects of TLR agonists. TLR agonists are potent immune stimulators and can induce systemic inflammation and cytokine storms, which can cause tissue damage and lead to severe side effects.Citation121 Therefore, it is important to identify safe and effective TLR agonists and determine the appropriate dose and route of administration. The use of TLR-based strategies in combination with other treatments, such as antiviral drugs or monoclonal antibodies, may enhance their efficacy. However, the optimal combination and sequence of treatments need to be determined, and potential drug interactions and side effects must be considered.

The development of TLR-based strategies also requires appropriate animal models to evaluate their efficacy and safety. Animal models that mimic human arboviral infections are essential to understand the immune response and testing TLR-based strategies. However, the availability of appropriate animal models for arboviral infections is limited.Citation122 Future directions for the development of TLR-based strategies against arboviral infections include the identification of novel TLR agonists and the development of TLR agonist-based vaccines. The use of adjuvants, such as TLR agonists, in vaccines, can enhance the immune response and increase the efficacy of vaccines. Therefore, TLR agonist-based vaccines may provide a promising approach to the prevention of arboviral infections.

Conclusions

In summary, TLR-based strategies hold promise as a potential approach to combat arboviral infections. However, several challenges need to be addressed, including the complexity of arboviral infections, potential side effects of TLR agonists, optimal combination and sequence of treatments, appropriate animal models, and identification of novel TLR agonists. Future research should focus on addressing these challenges to develop safe and effective TLR-based strategies against arboviral infections.

Acknowledgments

We would like to express our deepest gratitude to the staff of the Department of Medical Microbiology, Faculty of Medicine, Universiti Malaya, and Tropical Infectious Diseases Research and Education Centre (TIDREC), Universiti Malaya for providing the necessary facilities and resources in bringing this review to fruition. We acknowledge the funding from the Ministry of Higher Education, Malaysia for niche area research under the Higher Institution Centre of Excellence (HICoE) program (MO002-2019 & TIDREC-2023).

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The work was supported by the Ministry of Higher Education, Malaysia for niche area research under the Higher Institution Centre of Excellence (HICoE) program [MO002-2019, TIDREC-2023].

References

  • Lemaitre B, Reichhart JM, Hoffmann JA. Drosophila host defense: differential induction of antimicrobial peptide genes after infection by various classes of microorganisms. Proc Natl Acad Sci. 1997;94:14614–12. doi:10.1073/pnas.94.26.14614.
  • Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The dorsoventral regulatory gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell. 1996;86(6):973–83. doi:10.1016/S0092-8674(00)80172-5.
  • Kawasaki T, Kawai T. Toll-like receptor signaling pathways. Front Immunol. 2014 Sep 25;5:461. doi:10.3389/fimmu.2014.00461.
  • Tsan MF, Gao B. Review: pathogen-associated molecular pattern contamination as putative endogenous ligands of toll-like receptors. J Endotoxin Res. 2007;13(1):6–14. doi:10.1177/0968051907078604.
  • Roeder A, Kirschning CJ, Rupec RA, Schaller M, Weindl G, Korting HC. Toll-like receptors as key mediators in innate antifungal immunity. Med Mycol. 2004;42(6):485–98. doi:10.1080/13693780400011112.
  • Mukherjee S, Huda S, Sinha Babu SP. Toll-like receptor polymorphism in host immune response to infectious diseases: a review. Scand J Immunol. 2019;90(1):e12771. doi:10.1111/sji.12771.
  • Rice M, Nicol A, Nuovo GJ. The differential expression of toll like receptors and RIG-1 in the placenta of neonates with in utero infections. Ann Diagn Pathol. 2023;62:152080. doi:10.1016/j.anndiagpath.2022.152080.
  • Kawai T, Akira S. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int Immunol. 2009;21(4):317–37. doi:10.1093/intimm/dxp017.
  • Takeuchi O, Hoshino K, Kawai T, Sanjo H, Takada H, Ogawa T, Takeda K, Akira S. Differential roles of TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall components. Immunity. 1999;11(4):443–51. doi:10.1016/s1074-7613(00)80119-3.
  • Yu L, Wang L, Chen S. Endogenous toll-like receptor ligands and their biological significance. J Cell Mol Med. 2010;14(11):2592–2603. doi:10.1111/j.1582-4934.2010.01127.x.
  • Takeda K, Takeuchi O, Akira S. Recognition of lipopeptides by toll-like receptors. J Endotoxin Res. 2002;8(6):459–463. doi:10.1177/09680519020080060101.
  • Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, Wilson CB, Schroeder L, Aderem A. The repertoire for pattern recognition of pathogens by the innate immune system is defined by cooperation between toll-like receptors. Proc Natl Acad Sci USA. 2000;97(25):13766–71. doi:10.1073/pnas.250476497.
  • Hajjar AM, O’Mahony DS, Ozinsky A, Underhill DM, Aderem A, Klebanoff SJ, Wilson CB. Cutting edge: functional interactions between toll-like receptor (TLR) 2 and TLR1 or TLR6 in response to phenol-soluble modulin. J Immunol. 2001;166(1):15–19. doi:10.4049/jimmunol.166.1.15.
  • Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801. doi:10.1016/j.cell.2006.02.015.
  • Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on toll-like receptors. Nat Immunol. 2010;11(5):373–384. doi:10.1038/ni.1863.
  • Karikó K, Ni H, Capodici J, Lamphier M, Weissman D. mRNA is an endogenous ligand for toll-like receptor 3. J Biol Chem. 2004;279(13):12542–50. doi:10.1074/jbc.M310175200.
  • Sioud M. Innate sensing of self and non-self RNAs by toll-like receptors. Trends Mol Med. 2006;12(4):167–176. doi:10.1016/j.molmed.2006.02.004.
  • Ivanov S, Dragoi AM, Wang X, Dallacosta C, Louten J, Musco G, Sitia G, Yap GS, Wan Y, Biron CA. et al. A novel role for HMGB1 in TLR9-mediated inflammatory responses to CpG-DNA. Blood. 2007;110(6):1970–81. doi:10.1182/blood-2006-09-044776.
  • Boehme KW, Compton T. Innate sensing of viruses by toll-like receptors. J Virol. 2004;78(15):7867–73. doi:10.1128/JVI.78.15.7867-7873.2004.
  • Botos I, Segal DM, Davies DR. The structural biology of toll-like receptors. Structure. 2011;19(4):447–59. doi:10.1016/j.str.2011.02.004.
  • Gao D, Li W. Structures and recognition modes of toll‐like receptors. Proteins. 2017;85(1):3–9. doi:10.1002/prot.25179.
  • Bell JK, Mullen GE, Leifer CA, Mazzoni A, Davies DR, Segal DM. Leucine-rich repeats and pathogen recognition in toll-like receptors. Trends Immunol. 2003;24(10):528–33. doi:10.1016/S1471-4906(03)00242-4.
  • Narayanan KB, Park HH. Toll/interleukin-1 receptor (TIR) domain-mediated cellular signaling pathways. Apoptosis. 2015;20(2):196–209. doi:10.1007/s10495-014-1073-1.
  • Park SH, Kyeong MS, Hwang Y, Ryu SY, Han SB, Kim Y. Inhibition of LPS binding to MD-2 co-receptor for suppressing TLR4-mediated expression of inflammatory cytokine by 1-dehydro-10-gingerdione from dietary ginger. Biochem Biophys Res Commun. 2012;419:735–40. doi:10.1016/j.bbrc.2012.02.091.
  • Rani M, Nicholson SE, Zhang Q, Schwacha MG. Damage-associated molecular patterns (DAMPs) released after burn are associated with inflammation and monocyte activation. Burns. 2017;43(2):297–303. doi:10.1016/j.burns.2016.10.001.
  • Brieger A, Rink L, Haase H. Differential regulation of TLR-dependent MyD88 and TRIF signaling pathways by free zinc ions. J Immunol. 2013;191(4):1808–17. doi:10.4049/jimmunol.1301261.
  • Akira S, Sato S. Toll-like receptors and their signaling mechanisms. Scand J Infect Dis. 2003;35(9):555–62. doi:10.1080/00365540310015683.
  • Baccala R, Gonzalez-Quintial R, Lawson BR, Stern ME, Kono DH, Beutler B, Theofilopoulos AN. Sensors of the innate immune system: their mode of action. Nat Rev Rheumatol. 2009;5(8):448–56. doi:10.1038/nrrheum.2009.136.
  • Brown J, Wang H, Hajishengallis GN, Martin M. TLR-signaling networks: an integration of adaptor molecules, kinases, and cross-talk. J Dent Res. 2011;90(4):417–27. doi:10.1177/0022034510381264.
  • Campbell GR, Rawat P, Spector SA. Pacritinib inhibition of IRAK1 blocks aberrant TLR8 signalling by SARS-CoV-2 and HIV-1-derived RNA. J Innate Immun. 2023;15(1):96–106. doi:10.1159/000525292.
  • Honda K, Takaoka A, Taniguchi T. Type I interferon gene induction by the interferon regulatory factor family of transcription factors. Immunity. 2006;25(3):349–60. doi:10.1016/j.immuni.2006.08.009.
  • Tian Y, Wang ML, Zhao J. Crosstalk between autophagy and type I interferon responses in innate antiviral immunity. Viruses. 2019;11:132. doi:10.3390/v11020132.
  • Yamamoto M, Takeda K, Akira S. TIR domain-containing adaptors define the specificity of TLR signaling. Mol Immunol. 2004;40(12):861–8. doi:10.1016/j.molimm.2003.10.006.
  • Lester SN, Li K. Toll-like receptors in antiviral innate immunity. J Mol Biol. 2014;426(6):1246–1264. doi:10.1016/j.jmb.2013.11.024.
  • Chamberlain RW. Arboviruses, the arthropod-borne animal viruses. In: Maramorosch K. editor. Insect viruses. Current topics in microbiology and immunology. Vol. 42. Berlin, Heidelberg: Springer; 1968. pp. 38–58.
  • Patterson J, Sammon M, Garg M. Dengue, Zika and chikungunya: emerging arboviruses in the New World. West J Emerg Med. 2016;17:671–9. doi:10.5811/westjem.2016.9.30904.
  • Boga JA, Alvarez-Arguelles ME, Rojo-Alba S, Rodríguez M, de Oña M, Melón S. Simultaneous detection of Dengue virus, Chikungunya virus, Zika virus, Yellow fever virus and West Nile virus. J Virol Methods. 2019;268:53–5.
  • Yang X, Quam MB, Zhang T, Sang S. Global burden for dengue and the evolving pattern in the past 30 years. J Travel Med. 2021. doi:10.1093/jtm/taab146.
  • Rothman AL. Immunity to dengue virus: a tale of original antigenic sin and tropical cytokine storms. Nat Rev Immunol. 2011 Jul 15;11(8):532–543. doi:10.1038/nri3014.
  • Wang A, Thurmond S, Islas L, Hui K, Hai R. Zika virus genome biology and molecular pathogenesis. Emerg Microbes Infect. 2017 Mar 22;6(3):e13. doi:10.1038/emi.2016.141.
  • Bernardo-Menezes LC, Agrelli A, Oliveira ASLE, Moura RR, Crovella S, Brandão LAC. An overview of Zika virus genotypes and their infectivity. Rev Soc Bras Med Trop. 2022 Sep 30;55:e02632022. doi:10.1590/0037-8682-0263-2022.
  • Shen S, Shi J, Wang J, Tang S, Wang H, Hu Z, Deng F. Phylogenetic analysis revealed the central roles of two African countries in the evolution and worldwide spread of Zika virus. Virol Sin. 2016;31(2):118–30. doi:10.1007/s12250-016-3774-9.
  • Schwartz O, Albert ML. Biology and pathogenesis of chikungunya virus. Nat Rev Microbiol. 2010;8(7):491–500. doi:10.1038/nrmicro2368.
  • Caglioti C, Lalle E, Castilletti C, Carletti F, Capobianchi MR, Bordi L. Chikungunya virus infection: an overview. New Microbiol. 2013;36:211–27.
  • Chen R, Puri V, Fedorova N, Lin D, Hari KL, Jain R, Rodas JD, Das SR, Shabman RS, Weaver SC. et al. Comprehensive genome scale phylogenetic study provides new insights on the global expansion of chikungunya virus. J Virol. 2016;90(23):10600–11. doi:10.1128/JVI.01166-16.
  • Brinton MA. Replication cycle and molecular biology of the West Nile virus. Viruses. 2013 Dec 27;6(1):13–53. doi:10.3390/v6010013.
  • Zeller HG, Schuffenecker I. West Nile virus: an overview of its spread in Europe and the Mediterranean basin in contrast to its spread in the Americas. Eur J Clin Microbiol Infect Dis. 2004;23(3):147–56. doi:10.1007/s10096-003-1085-1.
  • Beasley DW, McAuley AJ, Bente DA. Yellow fever virus: genetic and phenotypic diversity and implications for detection, prevention and therapy. Ant Res. 2015;115:48–70.
  • Gardner CL, Ryman KD. Yellow fever: a reemerging threat. Clin Lab Med. 2010;30(1):237–260. doi:10.1016/j.cll.2010.01.001.
  • Couto-Lima D, Madec Y, Bersot MI, Campos SS, Motta MD, Santos FB, Vazeille M, Vasconcelos PF, Lourenço-de-Oliveira R, Failloux AB. Potential risk of re-emergence of urban transmission of yellow fever virus in Brazil facilitated by competent Aedes populations. Sci Rep. 2017;7(1):1–2. doi:10.1038/s41598-017-05186-3.
  • Gould EA, Higgs S. Impact of climate change and other factors on emerging arbovirus diseases. Trans R Soc Trop Med Hyg. 2009;103(2):109–21. doi:10.1016/j.trstmh.2008.07.025.
  • Tambo E, El Dessouky AG, Khater EI. Innovative preventive and resilience approaches against Aedes-linked vector-borne arboviral diseases threat and epidemics burden in gulf council countries. Oman Med J. 2019. doi:10.5001/omj.2019.73.
  • Xagorari A, Chlichlia K. Toll-like receptors and viruses: induction of innate antiviral immune responses. Open Microbiol J. 2008;2:49–59. doi:10.2174/1874285800802010049.
  • Wang T, Town T, Alexopoulou L, Anderson JF, Fikrig E, Flavell RA. Toll-like receptor 3 mediates West Nile virus entry into the brain causing lethal encephalitis. Nat Med. 2004;10(12):1366–1373. doi:10.1038/nm1140.
  • Gagnon SJ, Mori M, Kurane I, Green S, Vaughn DW, Kalayanarooj S, Suntayakorn S, Ennis FA, Rothman AL. Cytokine gene expression and protein production in peripheral blood mononuclear cells of children with acute dengue virus infections. J Med Virol. 2002;67(1):41–6. doi:10.1002/jmv.2190.
  • Modhiran N, Watterson D, Blumenthal A, Baxter AG, Young PR, Stacey KJ. Dengue virus NS1 protein activates immune cells via TLR4 but not TLR2 or TLR6. Immunol Cell Biol. 2017;95(5):491–495. doi:10.1038/icb.2017.5.
  • Tsai YT, Chang SY, Lee CN, Kao CL. Human TLR3 recognizes dengue virus and modulates viral replication in vitro. Cell Microbiol. 2009;11(4):604–15. doi:10.1111/j.1462-5822.2008.01277.x.
  • Bustos-Arriaga J, García-Machorro J, León-Juárez M, García-Cordero J, Santos-Argumedo L, Flores-Romo L, Méndez-Cruz AR, Juárez-Delgado FJ, Cedillo-Barrón L. Activation of the innate immune response against DENV in normal non-transformed human fibroblasts. PloS Negl Trop Dis. 2011. doi:10.1371/journal.pntd.0001420.
  • Sariol CA, Martínez MI, Rivera F, Rodríguez IV, Pantoja P, Abel K, Arana T, Giavedoni L, Hodara V, White LJ. et al. Decreased dengue replication and an increased anti-viral humoral response with the use of combined toll-like receptor 3 and 7/8 agonists in macaques. PloS One. 2011. doi:10.1371/journal.pone.0019323.
  • Liang Z, Wu S, Li Y, He L, Wu M, Jiang L, Feng L, Zhang P, Huang X. Activation of toll-like receptor 3 impairs the dengue virus serotype 2 replication through induction of IFN-β in cultured hepatoma cells. PloS One. 2011. doi:10.1371/journal.pone.0023346.
  • Sengupta S, Mukherjee S, Bhattacharya N, Tripathi A. Differential genotypic signatures of toll-like receptor polymorphisms among dengue-chikungunya mono-and co-infected Eastern Indian patients. Eur J Clin Microbiol Infect Dis. 2021;40:1369–81.
  • Décembre E, Assil S, Hillaire ML, Dejnirattisai W, Mongkolsapaya J, Screaton GR, Davidson AD, Dreux M. Sensing of immature particles produced by dengue virus infected cells induces an antiviral response by plasmacytoid dendritic cells. PloS Pathog. 2014. doi:10.1371/journal.ppat.1004434.
  • Perry ST, Prestwood TR, Lada SM, Benedict CA, Shresta S. Cardif-mediated signaling controls the initial innate response to dengue virus in vivo. J Virol. 2009;83(16):8276–81. doi:10.1128/JVI.00365-09.
  • Kayesh ME, Kitab B, Sanada T, Hayasaka D, Morita K, Kohara M, Tsukiyama-Kohara K. Susceptibility and initial immune response of Tupaia belangeri cells to dengue virus infection. Infect Genet Evol. 2017;51:203–10. doi:10.1016/j.meegid.2017.04.003.
  • Luo H, Winkelmann ER, Fernandez-Salas I, Li L, Mayer SV, Danis-Lozano R, Sanchez-Casas RM, Vasilakis N, Tesh R, Barrett AD. et al. Zika, dengue and yellow fever viruses induce differential anti-viral immune responses in human monocytic and first trimester trophoblast cells. Ant Res. 2018;151:55–62.
  • Kayesh ME, Tsukiyama-Kohara K. Mammalian animal models for dengue virus infection: a recent overview. Arch Virol. 2022;167(1):31–44. doi:10.1007/s00705-021-05298-2.
  • Estrada-Jiménez T, Millan-Perez Pena L, Flores-Mendoza L, Sedeño-Monge V, Santos-López G, Rosas-Murrieta N, Reyes-Carmona S, Terán-Cabanillas E, Hernández J, Herrera-Camacho I. et al. Upregulation of the suppressors of cytokine signaling 1 and 3 is associated with arrest of phosphorylated-STAT1 nuclear importation and reduced innate response in dengue virus-infected macrophages. Viral Immunol. 2016;29(2):95–104. doi:10.1089/vim.2014.0136.
  • Latanova A, Starodubova E, Karpov V. Flaviviridae nonstructural proteins: the role in molecular mechanisms of triggering inflammation. Viruses. 2022. doi:10.3390/v14081808.
  • Kao YS, Wang LC, Chang PC, Lin HM, Lin YS, Yu CY, Chen CC, Lin CF, Yeh TM, Wan SW. et al. Negative regulation of type I interferon signaling by integrin-linked kinase permits dengue virus replication. PloS Pathog. 2023. doi:10.1371/journal.ppat.1011241.
  • Udawatte DJ, Lang DM, Currier JR, Medin CL, Rothman AL. Dengue virus downregulates TNFR1-and TLR3-stimulated NF-κB activation by targeting RIPK1. Front Cell Infect Microbiol. 2022. doi:10.3389/fcimb.2022.926036.
  • Plociennikowska A, Frankish J, Moraes T, Del Prete D, Kahnt F, Acuna C, Slezak M, Binder M, Bartenschlager R. TLR3 activation by Zika virus stimulates inflammatory cytokine production which dampens the antiviral response induced by RIG-I-like receptors. J Virol. 2021. doi:10.1128/jvi.01050-20.
  • Santos CN, Magalhães LS, Fonseca AB, Bispo AJ, Porto RL, Alves JC, Dos Santos CA, de Carvalho JV, da Silva AM, Teixeira MM. et al. Association between genetic variants in TREM1, CXCL10, IL4, CXCL8 and TLR7 genes with the occurrence of congenital Zika syndrome and severe microcephaly. Sci Rep. 2023. doi:10.1038/s41598-023-30342-3.
  • Haque A, Akçeşme FB, Pant AB. A review of Zika virus: hurdles toward vaccine development and the way forward. Antivir Ther. 2018;23(4):285–93. doi:10.3851/IMP3215.
  • Lundberg R, Melén K, Westenius V, Jiang M, Österlund P, Khan H, Vapalahti O, Julkunen I, Kakkola L. Zika virus non-structural protein NS5 inhibits the RIG-I pathway and interferon lambda 1 promoter activation by targeting IKK epsilon. Viruses. 2019. doi:10.3390/v11111024.
  • Gim E, Shim DW, Hwang I, Shin OS, Yu JW. Zika virus impairs host NLRP3-mediated inflammasome activation in an NS3-dependent manner. Immune Netw. 2019 23;19(6). doi:10.4110/in.2019.19.e40.
  • Hernández-Sarmiento LJ, Valdés-López JF, Urcuqui-Inchima S. American-Asian-and African lineages of Zika virus induce differential pro-inflammatory and interleukin 27-dependent antiviral responses in human monocytes. Virus Res. 2023. doi:10.1016/j.virusres.2023.199040.
  • Daffis S, Samuel MA, Suthar MS, Gale JM, Diamond MS. Toll-like receptor 3 has a protective role against West Nile virus infection. J Virol. 2008;82(21):10349–58. doi:10.1128/JVI.00935-08.
  • Laurent-Rolle M, Boer EF, Lubick KJ, Wolfinbarger JB, Carmody AB, Rockx B, Liu W, Ashour J, Shupert WL, Holbrook MR. et al. The NS5 protein of the virulent West Nile virus NY99 strain is a potent antagonist of type I interferon-mediated JAK-STAT signaling. J Virol. 2010;84(7):3503–15. doi:10.1128/JVI.01161-09.
  • Xie G, Luo H, Tian B, Mann B, Bao X, McBride J, Tesh R, Barrett AD, Wang T. A West Nile virus NS4B-P38G mutant strain induces cell intrinsic innate cytokine responses in human monocytic and macrophage cells. Vaccine. 2015;33(7):869–78. doi:10.1016/j.vaccine.2014.12.056.
  • Segato-Vendrameto CZ, Zanluca C, Zucoloto AZ, Zaninelli TH, Bertozzi MM, Saraiva-Santos T, Ferraz CR, Staurengo-Ferrari L, Badaro-Garcia S, Manchope MF. et al. Chikungunya virus and its envelope protein E2 induce hyperalgesia in mice: inhibition by anti-E2 monoclonal antibodies and by targeting TRPV1. Cells. 2023. doi:10.3390/cells12040556.
  • Lani R, Teoh BT, Sam SS, AbuBakar S, Hassandarvish P. Fisetin modulates toll-like receptor-mediated innate antiviral response in Chikungunya virus-infected hepatocellular carcinoma Huh7 cells. Immuno. 2022;2(4):703–19. doi:10.3390/immuno2040043.
  • Petitdemange C, Wauquier N, Vieillard V. Control of immunopathology during chikungunya virus infection. J Allergy Clin Immunol. 2015;135(4):846–55. doi:10.1016/j.jaci.2015.01.039.
  • Bouraï M, Lucas-Hourani M, Gad HH, Drosten C, Jacob Y, Tafforeau L, Cassonnet P, Jones LM, Judith D, Couderc T. et al. Mapping of Chikungunya virus interactions with host proteins identified nsP2 as a highly connected viral component. J Virol. 2012;86(6):3121–34. doi:10.1128/JVI.06390-11.
  • Amaral MP, Coirada FC, de Souza Apostolico J, Tomita N, Fernandes ER, Souza HF, Chura-Chambi RM, Morganti L, Boscardin SB, Rosa DS. Prime-boost with Chikungunya virus E2 envelope protein combined with poly (I: C) induces specific humoral and cellular immune responses. Curr Res Immunol. 2021;2:23–31.
  • Tricou V, Essink B, Ervin JE, Turner M, Escudero I, Rauscher M, Brose M, Lefevre I, Borkowski A, Wallace D. Immunogenicity and safety of concomitant and sequential administration of yellow fever YF-17D vaccine and tetravalent dengue vaccine candidate TAK-003: a phase 3 randomized, controlled study. PloS Negl Trop Dis. 2023;17:e0011124. doi:10.1371/journal.pntd.0011124.
  • Cong Y, McArthur MA, Cohen M, Jahrling PB, Janosko KB, Josleyn N, Kang K, Zhang T, Holbrook MR. Characterization of yellow fever virus infection of human and non-human primate antigen presenting cells and their interaction with CD4+ T cells. Plos Neglect Trop Dis. 2016. doi:10.1371/journal.pntd.0004709.
  • Van Tol S, Hage A, Giraldo MI, Bharaj P, Rajsbaum R. The TRIMendous role of TRIMs in virus–host interactions. Vaccines. 2017. doi:10.3390/vaccines5030023.
  • Facciolà A, Visalli G, Laganà A, Di Pietro A. An overview of vaccine adjuvants: Current evidence and future perspectives. Vaccines (Basel). 2022 May 22;10(5):819. doi:10.3390/vaccines10050819.
  • Makkouk A, Abdelnoor AM. The potential use of toll-like receptor (TLR) agonists and antagonists as prophylactic and/or therapeutic agents. Immunopharmacol Immunotoxicol. 2009;31(3):331–8. doi:10.1080/08923970902802926.
  • Nicholls EF, Madera L, Hancock RE. Immunomodulators as adjuvants for vaccines and antimicrobial therapy. Annal New York Acad Sci. 2010;1213:46–61.
  • Connolly DJ, O’Neill LA. New developments in toll-like receptor targeted therapeutics. Curr Opin Pharmacol. 2012;12(4):510–8. doi:10.1016/j.coph.2012.06.002.
  • Cho JH, Lee HJ, Ko HJ, Yoon BI, Choe J, Kim KC, Hahn TW, Han JA, Choi SS, Jung YM. et al. The TLR7 agonist imiquimod induces anti-cancer effects via autophagic cell death and enhances anti-tumoral and systemic immunity during radiotherapy for melanoma. Oncotarget. 2017;8(15):24932–48. doi:10.18632/oncotarget.15326.
  • Krieg AM. Toll-like receptor 9 (TLR9) agonists in the treatment of cancer. Oncogene. 2008;27(2):161–7. doi:10.1038/sj.onc.1210911.
  • Dunne A, Marshall NA, Mills KH. TLR-based therapeutics. Curr Opin Pharmacol. 2011;11:404–11.
  • Yuan J, Ni G, Wang T, Mounsey K, Cavezza S, Pan X, Liu X. Genital warts treatment: Beyond imiquimod. Hum Vaccin Immunother. 2018;14(7):1815–9. doi:10.1080/21645515.2018.1445947.
  • Shoimer I, Rosen N, Muhn C. Current management of actinic keratoses. Skin Ther Let. 2010;15:5–7.
  • Meyer T, Surber C, French LE, Stockfleth E. Resiquimod, a topical drug for viral skin lesions and skin cancer. Expert Opin Investig Drugs. 2013;22(1):149–59. doi:10.1517/13543784.2013.749236.
  • Pasco ST, Anguita J. Lessons from Bacillus Calmette-Guerin: harnessing trained immunity for vaccine development. Cells. 2020. doi:10.3390/cells9092109.
  • Siu L, Brody J, Gupta S, Marabelle A, Jimeno A, Munster P, Grilley-Olson J, Rook AH, Hollebecque A, Wong RK. et al. Safety and clinical activity of intratumoral MEDI9197 alone and in combination with durvalumab and/or palliative radiation therapy in patients with advanced solid tumors. J Immunother Cancer. 2020. doi:10.1136/jitc-2020-001095.
  • Frega G, Wu Q, Le Naour J, Vacchelli E, Galluzzi L, Kroemer G, Kepp O. Trial watch: experimental TLR7/TLR8 agonists for oncological indications. Oncoimmunology. 2020. doi:10.1080/2162402X.2020.1796002.
  • Shoushtari M, Roohvand F, Salehi-Vaziri M, Arashkia A, Bakhshi H, Azadmanesh K. Adenovirus vector-based vaccines as forefront approaches in fighting the battle against flaviviruses. Hum Vaccin Immunother. 2022;18(5). doi:10.1080/21645515.2022.2079323.
  • Antonelli AC, Almeida VP, de Castro FO, Silva JM, Pfrimer IA, Cunha-Neto E, Maranhão AQ, Brígido MM, Resende RO, Bocca AL. et al. In silico construction of a multiepitope Zika virus vaccine using immunoinformatics tools. Sci Rep. 2022. doi:10.1038/s41598-021-03990-6.
  • DeFilippis VR. Chikungunya virus vaccines: platforms, progress, and challenges. In: Heise M. editor. Chikungunya virus. Current topics in microbiology and immunology, Vol. 435. Springer: Cham; 2019. doi:10.1007/82_2019_175.
  • Van Hoeven N, Joshi SW, Nana GI, Bosco-Lauth A, Fox C, Bowen RA, Clements DE, Martyak T, Parks DE, Baldwin S. et al. A novel synthetic TLR-4 agonist adjuvant increases the protective response to a clinical-stage West Nile virus vaccine antigen in multiple formulations. PloS One. 2016. doi:10.1371/journal.pone.0149610.
  • Gnjatic S, Sawhney NB, Bhardwaj N. Toll-like receptor agonists: are they good adjuvants? Cancer J. 2010;16:382–91.
  • Van Hoeven N, Wiley S, Gage E, Fiore-Gartland A, Granger B, Gray S, Fox C, Clements DE, Parks DE, Winram S. et al. A combination of TLR-4 agonist and saponin adjuvants increases antibody diversity and protective efficacy of a recombinant west Nile virus antigen. npj Vaccines. 2018;3(1):39. doi:10.1038/s41541-018-0077-1.
  • Huang L, Ge X, Liu Y, Li H, Zhang Z. The role of toll-like receptor agonists and their nanomedicines for tumor immunotherapy. Pharmaceutics. 2022. doi:10.3390/pharmaceutics14061228.
  • Lin E, Freedman JE, Beaulieu LM. Innate immunity and toll‐like receptor antagonists: a potential role in the treatment of cardiovascular diseases. Cardiovasc Ther. 2009;27(2):117–23. doi:10.1111/j.1755-5922.2009.00077.x.
  • Hong-Geller E, Chaudhary A, Lauer S. Targeting toll-like receptor signaling pathways for design of novel immune therapeutics. Curr Drug Discov Technol. 2008;5(1):29–38. doi:10.2174/157016308783769441.
  • Leon CG, Tory R, Jia J, Sivak O, Wasan KM. Discovery and development of toll-like receptor 4 (TLR4) antagonists: a new paradigm for treating sepsis and other diseases. Pharm Res. 2008;25(8):1751–61. doi:10.1007/s11095-008-9571-x.
  • Sharifi-Rad M, Pezzani R, Redaelli M, Zorzan M, Imran M, Ahmed Khalil A, Salehi B, Sharopov F, Cho WC, Sharifi-Rad J. Preclinical activities of epigallocatechin gallate in signaling pathways in cancer. Molecules. 2020. doi:10.3390/molecules25030467.
  • Boyanapalli SS, Kong AN. “Curcumin, the king of spices”: epigenetic regulatory mechanisms in the prevention of cancer, neurological, and inflammatory diseases. Curr Pharmacol Rep. 2015;1(2):129–39. doi:10.1007/s40495-015-0018-x.
  • Opal SM, Laterre PF, Francois B, LaRosa SP, Angus DC, Mira JP, Wittebole X, Dugernier T, Perrotin D, Tidswell M. et al. Effect of eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: the ACCESS randomized trial. JAMA. 2013;309(11):1154–62. doi:10.1001/jama.2013.2194.
  • Karami J, Farhadi E, Delbandi AA, Shekarabi M, Tahmasebi MN, Vaziri AS, Akhtari M, Mousavi MJ, Jamshidi A, Mahmoudi M. Evaluation of TAK-242 (resatorvid) effects on inflammatory status of fibroblast-like synoviocytes in rheumatoid arthritis and trauma patients. Iran J Allergy Asthma Immunol. 2021. doi:10.18502/ijaai.v20i4.6955.
  • Tam JS, Coller JK, Hughes PA, Prestidge CA, Bowen JM. Toll-like receptor 4 (TLR4) antagonists as potential therapeutics for intestinal inflammation. Indian J Gastroenterol. 2021;40(1):5–21. doi:10.1007/s12664-020-01114-y.
  • Lanford RE, Guerra B, Chavez D, Giavedoni L, Hodara VL, Brasky KM, Fosdick A, Frey CR, Zheng J, Wolfgang G. et al. GS-9620, an oral agonist of toll-like receptor-7, induces prolonged suppression of hepatitis B virus in chronically infected chimpanzees. Gastroenterology. 2013;144:1508–17.
  • Rappe JC, Finsterbusch K, Crotta S, Mack M, Priestnall SL, Wack A. A TLR7 antagonist restricts interferon-dependent and-independent immunopathology in a mouse model of severe influenza. J Experiment Med. 2021. doi:10.1084/jem.20201631.
  • Shirey KA, Lai W, Scott AJ, Lipsky M, Mistry P, Pletneva LM, Karp CL, McAlees J, Gioannini TL, Weiss J. et al. The TLR4 antagonist eritoran protects mice from lethal influenza infection. Nature. 2013;497(7450):498–502. doi:10.1038/nature12118.
  • Rallabhandi P, Phillips RL, Boukhvalova MS, Pletneva LM, Shirey KA, Gioannini TL, Weiss JP, Chow JC, Hawkins LD, Vogel SN. et al. Respiratory syncytial virus fusion protein-induced toll-like receptor 4 (TLR4) signaling is inhibited by the TLR4 antagonists rhodobacter sphaeroides lipopolysaccharide and eritoran (E5564) and requires direct interaction with MD-2. MBio. 2012. doi:10.1128/mbio.00218-12.
  • Sexton NR, Ebel GD. Effects of arbovirus multi-host life cycles on dinucleotide and codon usage patterns. Viruses. 2019. doi:10.3390/v11070643.
  • Tang XD, Ji TT, Dong JR, Feng H, Chen FQ, Chen X, Zhao HY, Chen DK, Ma WT. Pathogenesis and treatment of cytokine storm induced by infectious diseases. Int J Mol Sci. 2021. doi:10.3390/ijms222313009.
  • Marín-Lopez A, Calvo-Pinilla E, Moreno S, Utrilla-Trigo S, Nogales A, Brun A, Fikrig E, Ortego J. Modeling arboviral infection in mice lacking the interferon alpha/beta receptor. Viruses. 2019. doi:10.3390/v11010035.