Immunomodulatory responses of toll like receptors against 2019nCoV

Year & Volume - Issue: 
Authors: 
Amany Sayed Maghraby
Article type: 
CID: 
e0202
PDF File: 
Abstract: 
The present review discusses the immune signals via toll like receptors (TLRs) against 2019nCoV. We researched using different database, up to June 18th, 2020. All the included articles were published in English language. The outcome of this review, that some TLRs agonists or antagonists are progressed as drugs to combat and down regulating TLRs immune signals respectively. TLRs 3 and 4 recognized 2019nCoV spike protein through immune and molecular signals that leading to immune stimulation of pro-inflammatory cytokines and even the immune fever. While the TLRs7 and 8 recognized single-stranded ribonucleic acids (ssRNAs) leading to elevation of the tumour necrosis factor α (TNF-α), interleukin (IL)-6 and -12 levels. TLRs agonists or antagonists utilized as immunotherapeutic targets against 2019nCoV via TLRs signals. Chloroquine and hydroxychloroquine; the approval compounds for 2019nCoV therapy can be inhibiting the class II major histocompatibility complex molecules expression and antigen presentation and even immune suppressions of the pro-inflammatory cytokines profile.
Cite as: 
Maghraby AS. Immunomodulatory responses of toll like receptors against 2019nCoV. Russian Open Medical Journal 2021; 10: e0202.

Introduction

In December 2019, a novel severe acute respiratory syndrome (SARS)-like coronavirus (CoV) designated 2019nCoV broke up in Wuhan, China. It was causing approximately 60,000 cases and over 1,350 deaths [1]. In January 2020, World Health Organization (WHO) identified the new coronavirus as a novel coronavirus “2019 nCoV” and the disease is COVID-19 [2]. 2019nCoV are positive-stranded ribonucleic acid (RNA) viruses. According to sequence identity of the spike protein (S protien) or the non-structural proteins (ns protein), CoVs are categorized into Alpha-CoV, Beta-CoV, Gamma-CoV, and Delta-CoV. The CoVs infection transmitted from birds to mammals [3-6]. Approximately 10-20% of common cold cases annually are caused by CoVs [7-8]. Human CoV (h CoV) accompanying illness is frequently self-limited in immune competent patients and causing severe respiratory tract inflammation in the adolescent and elderlies [4, 9]; unfortunately, 2019nCov infections may be asymptomatic in the early stage until severe pneumonia, dyspnea, renal insufficiency, and even death [2]; However, Huang et al., [10] reported that the severity of 2019nCoV disease is associated with the clinical features and cytokine profile [10]. Chen et al., [11] verified the results of Huang et al., [10]; Nevertheless, enhancement of IL-2R and IL-6 levels in sera from 2019nCoV infected patients, estimated the extreme seriousness of the disease [11]; Additionally, a biopsy from dead 2019nCoV patient declared interstitial mononuclear inflammatory infiltrates in both lungs, dominated by lymphocytes and over activation of T cells accounted [12].

The innate immunity is self-possessed of several patterns of recognition receptors (PRRs) to recognize pathogen-associated molecular patterns (PAMPs); the PRRs involved toll like receptors (TLRs) were discovered 30 years ago interpreting the immune signals of innate immunity, inflammation and cytokines profile [13-14].

TLR1 to TLR10 were signified on primary bronchial epithelial cells, upon TLRs stimulation by specific ligands causing activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB); furthermore, increased levels of IL-8, amplified the epithelial cell responsiveness, resulting in altered immune inflammatory responses [15]. On the human alveolar macrophages (MQ); the level of expression of TLRs 3, 5 and 9 was low while TLRs 1, 2, 4, 7 and 8 was high [16].

TLR4 is expressed on lung endothelial cells and is crucial for capillary concealment and neutrophil recruitment after systemic administration of lipopolysaccharide (LPS) [17].

TLR3-deficient mice showed reduction in the lung inflammatory responses [18]. Also, Le Goffic et al [19] demonstrated the immunological role of TLR3 deficiency in Influenza A virus infected mice. They perceived that the survival rates had increased in spite of elevated the viral burden in lungs, reduction in pro-inflammatory cytokines and decreasing in the number of CD8+ T lymphocytes in the bronchoalveolar airspace [19]. Respiratory syncytial virus modulates TLR3 up-regulation, leads to priming the lung epithelial cells for successive exposures to extracellular double-stranded RNA (dsRNA).

There are two dependent pathways for TLRs signals. For instance; TLR3 signals via TIR-domain-containing adaptor protein, inducing interferon beta (TRIF), while TLR7/8/9 rely on myeloid differentiation factor-88 (MyD88) [15, 20-21]. In the TRIF-dependent pathway, the ubiquitin E3 ligase tumour necrosis factor receptor-associated factor 3 (TRAF3) is recruited and hence activates TANK-binding kinase-1 (TBK1) and Inhibitor-κB kinase ε (IKKε) [22]. Activated TBK1 and IKKε phosphorylate interferon (IFN) regulatory factor (IRF) transcription factors IRF3 and IRF7 to drive the expression of type I IFNs [23].

In the MyD88-dependent pathway, TRAF6 is attached to the MyD88 signal program, resulting in the stimulation of the kinase complex composed of IKKα and IKKβ. Activated IKK complex influences NF-κB activation, which induces the expression of pro inflammatory cytokines [6]. Furthermore, Myd88 also initiates IFN-α by immune promoting IKKα-dependent IRF7 phosphorylation, that is crucial for the antiviral survival in plasmacytoid dendritic cells (pDCs) [24-25]. TLR7 has been associated with stimulation of IFN type-1 which is essential for evacuation of viruses as the SARS CoV. TLR7-positive plamacytoid cells are copiously in lung tissue [26-28]. For example, upon activation of the TLR7 pathway by the influenza virus, pDC have the potential to assemble type I IFN [29]. TLR4 deficient mice exhibit immune suppression in IL-12 level and mononuclear cells infiltration leading to the immune reduction in viral clearance [30].

 

TLRs immune signals

The damage-associated molecular patterns (DAMPs) originated from the host damaged tissues acting as dangerous signals or ligands to release certain intracellular molecules, some of which specifically enhancing TLRs (as high mobility group box 1 (HMGB1), hyaluronan and heat shock proteins) [31-34]. TLRs signals are implicated in secretion of tumour necrosis factor α (TNF-α), interleukin (IL)-8, macrophage inflammatory proteins (MIP)-1α, -1β, RANTES (regulated upon activation, normal T cell expressed and presumably secreted), growth-regulated oncogene (GRO)-α, -β and -γ, IL-6, IL-5 and transforming growth factor (TGF)-β which are essential for promoting the immune influx of neutrophils, eosinophils, monocytes, natural killer (NK) cells, MQ and dendritic cells (DCs) to evade the pathogen [35-37]. In addition, TLRs immune signals are elaborated in of anti-microbial substances production (as defensins, lysozyme, nitric oxide and IL-37) in the lower respiratory tract [13]. TLRs involved proteins clan in recognition and initiating an appropriate response up against pathogenic attacks [38] as mucins which is the protein component of mucus and its expression is induced either directly or indirectly by TLRs signals [35, 39].

In the lower respiratory tract; TLR3 is expressed on the luminal and basal sides, whereas TLRs 2, 6 and 1 on basolateral side. TLR6 are plenteous. Whereas; TLRs 2, 4, 5 and 10 along with TLRs 3, 7 and 9 in the lower levels are on both cell surface as well as intracellular compartments [40]. TLR4 expressed in the lung endothelial cells is essential for neutrophil recruitment [41] and capillary sequestration [13, 17] while, TLRs 7 and 9 are highly expressed by lung pDCs that downregulate the allergic response. Alveolar MQ express TLRs 3, 5 and 9 and higher levels of TLRs 1, 2, 4, 7 and 8 [13, 16, 28, 42].

 

TLRs\ 2019 nCoV immune recognitions

2019nCoV are recognized by the three hereditary immune cytoplasmic receptors: toll-like receptors (TLRs), retinoic acid-inducible gene I-like (RIG-I) receptors (RLRs), and nucleotide-binding oligomerization domain-like receptors (NLRs) [43-46]. Immune responses to viral infection are negotiated through TLRs 3, 7, 8, 9, 4 and 2 signals. Each TLR is contributing differently depending on the virus, cell type and infection model examined [47-48]. TLRs7 and 8 detect GU-rich and AU-rich single-stranded ribonucleic acid (ssRNA) sequences of RNA viruses [47-48]. TLRs7 and 8 used as endosomal PRRs for a number of ssRNA viruses as coronaviruses [48-52].

Moreover; TLR7 signaling is well characterized in human and mice for type I IFN production by pDCs in response to ssRNA viral stimuli [48]. TLR-3 may recognize the virus intermediates during intracellular replication dsRNA. The realization of pathogen associated molecular patterns (PAMPs) as viral RNA, particles, or danger signals via endosomal RNA receptors, TLRs 3 and 7 and the cytosolic RNA sensor, RIG-I/MDA5 (melanoma differentiation-​associated gene 5). This recognition leads to stimulation of the downstream signaling cascade NF-κB and IRF3 that influence the expression of type I IFN and IL-1β, -6, -18, and TNF-α that activates the janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway that initiate the transcription of IFN type 1 [53-55].

Interestingly; sometimes, accessory proteins of CoV during replication can interfere with TLR3 immune signal and prevent TLR3 stimulation; therefore, CoV evade the immune house. Only TLRs7 and -8 recognize ssRNA viral genomic. Nonetheless, 2019nCoV infects lung alveolar epithelial cells through S-protein containing a ligand binding domain with the angiotensin-converting enzyme II (ACE2) [56]. The TLRs 3 and 4 recognize S-protein and leads to activation of pro-inflammatory cytokines [2, 57-58]. The TLRs7 and 8 recognize SARS-CoV-ssRNAs leading to elevation of the TNF-α, IL-6 and IL-12 levels [58-59].

2019nCoV immune fever is the most common symptom of 2019-nCoV infection. It is resulted from the binding of 2019nCoV with the TLRs causing the release of pro-IL-1b that is cleaved by caspase-1, followed by inflammasome activation and release of active mature IL-1b that is a mediator of lung inflammation, fever and fibrosis [60]. The TLR4 expression is resulted due to the immune fever [61] and identifies S-protein via MyD88, this introduction will provoke the stimulation of NF-κB transcription factors and the pathogen-activated protein kinases (MAPKs) pathway to induce proinflammatory proteins [2, 62-63].

 

TLRs\2019 nCoV immunotherapy

However, various antiviral drugs have no productive role compared with standard guideline care [64]. TLRs agonists or antagonists utilized as immunotherapeutic targets to up and down regulate TLRs immune responses respectively; Therefore, TLRs are being directed in a dual way by small molecule ligands (agonists) development which have the ability to bind with TLRs and augment host immune capabilities; On the other hand, TLRs act as adjuvants in vaccine therapies and by inhibiting TLRs through specified antibodies, small molecule inhibitors, oligopeptides and by improving endogenous anti-TLR molecules. Also, negative regulation of signal pathways involving TLRs are crucial as a novel therapeutic strategy [65].

 

2019nCoV Immunotheraby by TLRs agonists

Our review summarizes some essential TLRs agonist: Mycobacterium bovis BCG Tokyo 172 (SMP-105). Which is a TLR2 agonist from Mycobacterium bovis containing mycolic acids, and peptidoglycans. SMP-105 can be served as an immune adjuvant as anti-inflammatory agent. The mode of action of SMP-105 starts by upregulating NF-κβ in a TLR2 dependent and TLR4 independent manner resulting in the deficiency of TNF-α and IL-6 levels [66].

Lipoprotein 1 and Lipopeptide-CGP40774 are TLR2 agonists capable of managing asthma and airway inflammation by inhibiting immunoglobulin E (IgE), IL-4 and IL-13 levels [67-68].

Rintatolimod is a TLR3 agonist being investigated for treatment of acute respiratory syndrome, influenza, hepatitis infections and cancer. It regulates the cellular RNase L enzyme levels in patients [69].

Another one is LPS that is a potent functional TLR4 ligand [70]. The LPS stimulating action is initiated by binding the disulfide high-mobility group protein B1 (HMGB1) at low nanomolar avidity with the TLR4 co-receptor myeloid differentiation factor 2 (MD-2), in an analogous way to LPS but at different position [71].

Poly-γ-glutamic acid (γ-PGA) is a TLR4 agonist seems to cause downregulation of Th2 cytokines, airway inflammation and eosinophilia while elevation the manifestation of co-stimulatory molecules, CD80, CD86 and CD40 on dendritic cells [72].

ER803022 is a synthetic TLR4 agonist, it has immune potential against asthma in murine models in a TLR-dependent MyD88 upregulation and IL-12/IFN-γ profile [73].

Diebold et al. [48] have stated that the ssRNA of influenza virus is a TLR7 agonist, and TLR7 has a preference for a particular RNA motif that is uridine. In addition, adenosine- and uridine-rich oligoribonucleotides (ORNs) are capable of stimulating TLR8, without affecting TLR7, while guanosine-rich ORNs activate TLRs7 and 8-dependent signaling [48]. In quest of TLRs 7 and 8 agonists, a range of chemical modifications was done to natural ORN ligands to augment their selectivity for TLR7 and/or TLR8 and stability against nucleases [74-77]. In 2015, Tanji et al. [78] stated for the first time, that TLR8 recognizes the ssRNA degradation products at two different sites are uridine and short ORNs such as UG and UUG. Uridine alone is not capable of stimulating TLR8 in the absence of the ORNs at the second site. The ORN binding with the second site proliferates the affinity of uridine at the first site; Therefore, it leads to stimulation of TLR8 [78]. TLR7 also recognizes the degraded form of ssRNA guanosine binds with the first site while the oligonucleotide (polyU) binds with the second site, and the conjunction of these two products activates the dimerization of TLR7 in a synergistic way [79]. Up regulating TLR7 not only suppresses Th2-mediated airway inflammation but also suppresses the manifestation of IgE titre; Additionally, it seriously inhibits IL-17 and IL-13 production through IL-10 mediated pathway [80]; Therefore, TLR7 is designed to cure airway diseases such as asthma and allergic rhinitis. Heil et al. [47] investigated the effect of ssRNAs on the immune stimulation the APCs via TLRs7 and 8. They proved that guanosine (G) and uridine (U)-rich oligonucleotides enhance the production of TNFα by human PBMCs and that ssRNAs are the natural ligands of TLRs 7 and 8 [47].

TLRs7 and 8 respond to imidazoquinoline compounds. These synthetic compounds have been stated to mediate the TLRs7 and 8 inflammatory responses [53].

Imiquimod is a clinically approved TLR7 agonist against asthma and viral induced lung and airway infection. Imiquimod augments the IFN-α level via TLR7–MYD88 dependent pathway.

Resiquimod (R-848) is a TLR7 and TLR8 agonist enhance IFN-α, IL-12 and TNF-α production while reducing lung eosinophilia and airway inflammation models [81-82]. A synthetic TLR7 agonist SA-2 regulates T cell production through IFN-α, IL-27 and IL-10 [83]. ANA773 is a tiny molecule TLR7 agonist has anti-viral immune potential [84].

Asthma exacerbation is treated by CpG-ODN, which is a TLR9 agonist [85-86]. CpG-ODN interacts with TLR9 present on leukocytes, B cells and T cells and stimulates the secretion of IL-6, IFN-γ, IL-12 and CD4 T lymphocytes, successively stimulating Th1 type of inflammatory response along with enhanced IL-12 and IL-10 production [87]. CpG-ODN also suppresses Th2 cytokine, airway eosinophilia, IgE levels, and bronchial hyperreactivity in vitro and in vivo models [88-90]. Indoleamine 2, 3-dioxygenase, IMO-2055, MO-2125 and IMO-2134 TLR9 agonists [91-92] are revealed to have anti-inflammatory and anti-allergic responses (Table 1).

 

Table 1. 2019nCoV immunotherapy by TLRs agonists

Agonist

TLR

Reference

SNIP-105

TLR2

68

CGP40774

TLR2

6, 82

Rintatolimod

TLR3

72

lipopolysaccharide

TLR4

115

y-PGA

TLR4

56

ER803022

TLR4

11

ssRNA

TLRs7 &8

39, 24

Imiquimod

TLR 7

35, 75

Resiquimod

TLRs7&8

35, 75

SA-2

TLR7

47

ANA773

TLR7

10

CpG-ODN

TLR9

105,91

IMO-205 5, MO-2125 and IMO-2134

TLR9

1, 73

TLR, toll like receptor; y-PGA, poly-y-glutamic acid; LPS, lipopolysaccharide.

 

2019nCoV Immunotherapy by TLR antagonists

This review gives an outline of some natural or synthetic TLRs antagonists (Table 2). A small-molecule TLR4-specific antagonist (P5779) prevents HMGB1-MD-2 interaction, protects mice from influenza virus-induced lethality and downregulate pro inflammatory cytokine gene expression in the lungs [93]. Targeting TLR4 with small-molecule antagonists, like TAK-242, or with anti-TLR4-specific antibodies is considered as protection against influenza virus infections [94-95].

 

Table 2. 2019nCoV immunotherapy by TLRS antagonists

Antagonists

TLR

References

P5779

TLR4

97

TAK-242

TLR4

77, 78

RAGE

TLR4

3

RS-LPS

TLR4

38

N1-0101

TLR4

67

1A6

TLR4

120

Resveratrol

TLRs 3&4

121, 122

Eritoran TM (E5564)

TLR4

97, 98: 88.

OPN-305

TLR2

41

Capsazepine

TLR3

62

R837

TLR7

40

TLR, toll like receptor.

 

A TLR4 is the receptor for advanced glycation end products (RAGE) antagonists, further supports that the HMGB1/RAGE/TLR4-axis is central in the pathogenesis of influenza infections [96]. Disulfide HMGB1-TLR4 stimulation induces a substantial production of proinflammatory cytokines both in vivo and in vitro [97]. The clinical end results of murine influenza infection indicated that it can be improved by TLR4-specific antagonists.

A TLR4 antagonist, under acylated form of rhodobacter sphaeroides LPS (RS-LPS) when inhaled, reduce the symptoms of asthma by inhibiting eosinophilia and lymphocytosis, also by limiting the levels of Th2 cytokines and lower airway hyper responsiveness [98].

RS-LPS acts as a TLR4 antagonist by suppressing the induction of IkBz (gene NFKBIZ, a member of the NF-kB family, play a consequential role in functioning of epithelial cells) in bronchial epithelial cells, consequently leading to inhibition of contribution of pro-inflammatory cytokines in a coculture based study of bronchial epithelial cells and monocytes stimulated with house dust mite mix (HDM) [99].

One of the rare synthesized antibodies that target TLRs is NI-0101, which is specific to TLR4 and blocks TLR4 dimerization. It has the potency to minimize cytokine secretion and averts flu and its symptoms in ex vivo and in vivo models. This drug has accomplished phase I clinical trial successfully and was proved to be a safe, low-risk and non-toxic drug at various concentrations [100].

1A6 is anti-TLR4 monoclonal antibody effectively in minimizing inflammatory disorders and has real role in lung injury models [101]. Lipid A analogues specifically targets TLR4 signals and has become potential therapeutic tool against airway diseases.

Additionally, eritoran is the TLR4-blocking compound ameliorated murine influenza-induced lung injury by inhibiting the cytokine storm. It has been proved to prevent high mobility group box 1 (HMGB1)-mediated TLR4-dependent signaling in vitro, and to suppress extracellular HMGB1 release in vivo by preventing necroptotic cell death in respiratory epithelial cells [93, 102]. Eritoran TM (E5564) is a TLR4 antagonist that affects TLR4/MD-2/LPS complex formation which is the most advanced TLR agonist [103]. A fully humanized IgG4 monoclonal TLR2 specific antibody, OPN-305 blocks TLR2-mediated pro-inflammatory cytokine production [104]. R837 is TLR7 antagonist is that plays a critical role in relaxing the airway passage through release of nitric oxide in a TLR7 dependent manner in vitro as well as in vivo models [105].

Capsazepine and its analogues have been considered as TLR3 inhibitors and they have shown to repress the production of pro-inflammatory cytokines such as IL-8 and TNF-α in asthmatic patients [106].

Resveratrol suppresses TLR4 expression [107] and activated SIRT1 to inhibit HMGB1/TLR4/ MyD88/NF-κB signaling [108]. Apart from synthesized compounds, a natural product known as Resveratrol that is found in grapes and peanuts down regulates TLR3 and one of its adapter molecule (TRIF) and assigns a defensive mechanism against asthma development [109].

Via TLRs signals the immunomodulatory effects of chloroquine (CQ) and hydroxychloroquine (HCQ) synergize their antiviral effects in the treatment of 2019nCoV [110]. The bioimmune action of CQ and HCQ depends on changing the intracellular pH, TLR 7, TLR 9 and cGAS response of interferon genes. Whereas; CQ/HCQ causes irregular obligation between TLRs 7 and 9 and their RNA/DNA ligands, due to changing of pH towards the basic, in the cellular endosomal environment, which suppressed TLR signaling [111-113]. The approaching of STING pathway (TLR and cGAS response of interferon genes) results in the attenuation of pro-inflammatory cytokines [114]. Noteworthy; the activation of TLRs 7 and 9 by their ligands requires the low pH of endosomes and lysosomes and an active autophagic pathway, the CQ/HCQ, through vesicle alkalinization, may inhibit this interaction and TLR signaling, in turn inhibiting the inflammatory response [115]. Matrix metalloproteinases (MMPs) are a family of zinc endopeptidases that regulate inflammation and tissue repair at several levels. On the contrary; CQ has been shown to downregulate MMP-9 expression through the inhibition of TLR9 signaling [116]. In particular, HCQ can enhance the intracellular pH and inhibit lysosomal activity in APCs. Meanwhile, due to the altered pH of endosomes and interrupted binding between TLR7 and TLR9 and their RNA/DNA ligands, TLRs signal are suppressed by administration of HCQ [111-112].

Remdesivir is a broad spectrum anti-viral drug that has shown to inhibit SARS-CoV-2, in vitro and in vivo. In absence of any effective treatment for SARS-CoV-2 infection (COVID-19), remdesivir has been tried for a compassionate use in severe COVID-19. Remdesivir hold promises as treatment for 2019 nCoV supported by a recent study on 2019nCoV [117] and multiple research studies on SARS and MERS-CoVs [118-120] and a single clinical report [121]. The Chinese government has started a clinical trial using remdesivir for treating 2019nCoV patients with mild to severe symptoms (NCT04252664, NCT04257656). Finally, the current situation also provides an opportunity to evolve innovative treatments to target infectious diseases [1].

 

2019nCoV Immunotherapy by TLRs-vaccine

Enayatkhania et al, [122] have testified and prognosticated NOM recombinant protein against HLA-A11:01 and TLR4 receptors. The vaccine could be considered as a possible vaccine candidate against 2019nCoV by stimulating cellular and humoral immune responses.

 

Conclusion

In this paper, we researched a literature (over 300 ones) using different database as the PubMed, up to June 18th, 2020. We used a specific key word as: COVID19, toll like receptors and COVID19, TLRs immunotherapy, antiviral compounds etc. All the included articles were published in English language. Our studies on the immune signals via the toll like receptors (TLRs) against 2019nCoV. The outcome of this review, that some TLRs agonists or antagonists are utilized as immunotherapeutic targets against 2019nCoV. The TLRs 3 and 4 recognize S-protein and leads to activation of pro-inflammatory cytokines [2, 57-58]. 2019nCoV- immune fever is the most common symptom of 2019nCoV infection resulted from the binding of 2019nCoV with the TLRs causing the release of pro-IL-1b. It is cleaved by caspase-1, followed by inflammasome activation and release of active mature IL-1b that is a mediator of lung inflammation, fever and fibrosis [60]. Via TLRs signals, the CQ and HCQ were inhibiting the immune responses. The bioimmune action of CQ and HCQ is causing irregular obligation between TLRs 7 and 9 and their RNA/DNA ligands, depends on changing the intracellular pH modifying the intracellular pH towards the basic, in the cellular endosomal environment. Recently in 2020, Enayatkhania, et al, [122] have testified NOM recombinant protein against HLA-A11:01 and TLR4 receptors and prognosticated that it could be considered as a possible vaccine candidate against 2019nCoV. Substantially; in 2019nCoV- immune fever; the high light must be focused on TLRs immunotherapy as an innate immune innovative alternative to some antivirals resist drugs by utilizing TLRs agonists or antagonists to up and down regulate TLRs immune responses respectively

 

Acknowledgments

The author thanks the National Research Center of Egypt for all supporting.

 

Conflict of interest

No competing financial interests exist

 

Ethical approval

Not required.

 

Funding Information

No funding

References: 
  1. Tse LV, Meganck RM, Graham RL, Baric RS. The current and future state of vaccines, antivirals and gene therapies against emerging coronaviruses. Front Microbiol 2020; 11: 658. https://doi.org/10.3389/fmicb.2020.00658.
  2. Li G, Fan Y, Lai Y, Han T, Li Z, Zhou P, et al. Coronavirus infections and immune responses. J Med Virol 2020; 92(4): 424-432. https://doi.org/10.1002/jmv.25685.
  3. Perlman S, Netland J. Coronaviruses post-SARS: update on replication and pathogenesis. Nat Rev Microbiol 2009; 7(6): 439-450. https://doi.org/10.1038/nrmicro2147.
  4. Woo P, Lau SKP, Huang Y, Yuen KY. Coronavirus diversity, phylogeny and interspecies jumping. Exp Biol Med (Maywood) 2009; 234(10): 1117-1127. https://doi.org/10.3181/0903-mr-94.
  5. Peck KM, Burch CL, Heise MT, Baric RS. Coronavirus host range expansion and middle east respiratory syndrome coronavirus emergence: biochemical mechanisms and evolutionary perspectives. Annu Rev Virol 2015; 2(1): 95-117. https://doi.org/10.1146/annurev-virology-100114-055029.
  6. Raj VS, Mou H, Smits SL, Dekkers DH, Müller MA, Dijkman R, et al. Dipeptidyl dipeptidase 4 is a functional receptor for the emerging human coronavirus-EMC. Nature 2013; 495(7440): 251-254. https://doi.org/10.1038/nature12005.
  7. McIntosh K, Kapikian AZ, Turner HC, Hartley JW, Parrott RH, Chanock RM. Seroepidemiologic studies of coronavirus infection in adults and children. Am J Epidemiol 1970; 91(6): 585-592. https://doi.org/10.1093/oxfordjournals.aje.a121171.
  8. Cabeça TK, Granato C, Bellei N. Epidemiological and clinical features of human coronavirus infections among different subsets of patients. Influenza Other Respir Viruses 2013; 7(6): 1040-1047. https://doi.org/10.1111/irv.12101.
  9. Lau SK, Woo PC, Yip CC, Tse H, Tsoi H, Cheng VC, et al. Coronavirus HKU1 and other coronavirus infections in Hong Kong. J Clin Microbiol 2006; 44(6): 2063-2071. https://doi.org/10.1128/jcm.02614-05.
  10. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020; 395(10223): 497-506. https://doi.org/10.1016/S0140-6736(20)30183-5.
  11. Chen Y, Liu Q, Guo D. Emerging coronaviruses: genome structure, replication, and pathogenesis. J Med Virol 2020; 92(4): 418-423. https://doi.org/10.1002/jmv.25681.
  12. Xu Z, Shi L, Wang Y, Wang Y, Zhang J, Huang L, et al. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir Med 2020; 8: 420-422. https://doi.org/10.1016/S2213-2600(20)30076-X.
  13. Lafferty EI, Qureshi ST, Schnare M. The role of toll-like receptors in acute and chronic lung inflammation. J Inflamm (Lond) 2010; 7: 57. https://doi.org/10.1186/1476-9255-7-57.
  14. Patinote C, Karroum NB, Moarbess G, Cirnat N, Kassab I, Bonnet PA, et al. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem 2020; 193: 112238. https://doi.org/10.1016/j.ejmech.2020.112238.
  15. Sha Q, Truong-Tran AQ, Plitt JR, Beck LA, Schleimer RP. Activation of airway epithelial cells by toll-like receptor agonists. Am J Respir Cell Mol Biol 2004; 31(3): 358-364. https://doi.org/10.1165/rcmb.2003-0388oc.
  16. Hoppstädter J, Diesel B, Zarbock R, Breinig T, Monz D, Koch M, et al. Differential cell reaction upon toll-like receptor 4 and 9 activation in human alveolar and lung interstitial macrophages. Respir Res 2010; 11(1): 124. https://doi.org/10.1186/1465-9921-11-124.
  17. Andonegui G, Bonder CS, Green F, Mullaly SC, Zbytnuik L, Raharjo E, et al. Endothelium-derived toll-like receptor-4 is the key molecule in LPS-induced neutrophil sequestration into lungs. J Clin Invest 2003; 111(7): 1011-1020. https://doi.org/10.1172/JCI16510.
  18. Wang Q, Miller DJ, Bowman ER, Nagarkar DR, Schneider D, Zhao Y, et al. MDA5 and TLR3 initiate pro-inflammatory signaling pathways leading to rhinovirus-induced airways inflammation and hyperresponsiveness. PLoS Pathog 2011; 7(5): e1002070. https://doi.org/10.1371/journal.ppat.1002070.
  19. Le Goffic R, Balloy V, Lagranderie M, Alexopoulou L, Escriou N, Flavell R, et al. Detrimental contribution of the toll like receptor (TLR)3 to influenza A virus-induced acute pneumonia. PLoS Pathog 2006; 2(6): e53. https://doi.org/10.1371/journal.ppat.0020053.
  20. Groskreutz DJ, Monick MM, Powers LS, Yarovinsky TO, look DC, Hunninghake GW, et al. Respiratory syncytial virus induces TLR3 protein and protein kinase R, leading to increased double-stranded RNA responsiveness in airway epithelial cells. J Immunol 2006; 176(3): 1733-1740. https://doi.org/10.4049/jimmunol.176.3.1733.
  21. Gay NJ, Symmons MF, Gangloff M, Bryant CE. Assembly and localization of toll like receptor signalling complexes. Nat Rev Immunol 2014; 14(8): 546-558. https://doi.org/10.1038/nri3713.
  22. Saha SK, Pietras EM, He JQ, Kang JR, Liu SY, Oganesyan G, et al. Regulation of antiviral responses by a direct and specific interaction between TRAF3 and Cardif. EMBO J 2006; 25(14): 3257-3263. https://doi.org/10.1038/sj.emboj.7601220.
  23. Fitzgerald KA, McWhirter SM, Faia KL, Rowe DC, Latz E, Golenbock DT, et al. IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway. Nat Immunol 2003; 4(5): 491-496. https://doi.org/10.1038/ni921
  24. Honda K, Ohba Y, Yanai H, Negishi H, Mizutani T, Takaoka A, et al. Spatiotemporal regulation of MyD88 IRF 7 signalling for robust type I interferon induction. Nature 2005; 434(7036): 1035-1040. https://doi.org/10.1038/nature03547.
  25. Wang L, Zhao J, Ren J, Hall KH, Moorman JP, Yao ZQ, et al. Protein phosphatase 1 abrogates IRF7-mediated type I IFN response in antiviral immunity. Eur J Immunol 2016; 46(10): 2409-2419. https://doi.org/10.1002/eji.201646491.
  26. Roberts A, Lamirande EW, Vogel L, Jackson JP, Paddock CD, Guarner J, et al. Animal models and vaccines for SARS-CoV infection. Virus Res 2008; 133(1): 20-32. https://doi.org/10.1016/j.virusres.2007.03.025.
  27. Sheahan T, Morrison TE, Funkhouser W, Uematsu S, Akira S, Baric RS, et al. MyD88 is required for protection from lethal infection with a mouse-adapted SARS-CoV. PLoS Pathog 2008; 4(12): e100024. https://doi.org/10.1371/journal.ppat.1000240.
  28. Plantinga M, Hammad H, Lambrecht BN. Origin and functional specializations of DC subsets in the lung. Eur J Immunol 2010; 40(8): 2112-2118. https://doi.org/10.1002/eji.201040562.
  29. [29] Di Domizio J, Blum A, Gallagher-Gambarelli M, Molens JP, Chaperol L, Plumas J. TLR7 stimulation in human plasmacytoid dendritic cells leads to the induction of early IFN-inducible genes in the absence of type I IFN. Blood 2009; 114(9): 1794-1802. https://doi.org/10.1182/blood-2009-04-216770.
  30. Akira S. Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell 2006; 124(4): 783-801. https://doi.org/10.1016/j.cell.2006.02.015.
  31. Erridge C. Endogenous ligands of TLR2 and TLR4: agonists or assistants? J Leukoc Biol 2010; 87(6): 989-999. https://doi.org/10.1189/jlb.1209775.
  32. Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell 2010; 140(6): 805-820. https://doi.org/10.1016/j.cell.2010.01.022.
  33. Yamamoto M, Takeda K. Current views of toll-like receptor signaling pathways. Gastroenterol Res Pract 2010; 2010: 240365. https://doi.org/10.1155/2010/240365.
  34. Yang L, Seki E. Toll-like receptors in liver fibrosis: cellular crosstalk and mechanisms. Front Physiol 2012; 3: 138. https://doi.org/10.3389/fphys.2012.00138.
  35. McClure R, Massari P. TLR-dependent human mucosal epithelial cell responses to microbial pathogens. Front Immunol 2014; 5: 386. https://doi.org/10.3389/fimmu.2014.00386.
  36. Ohkuni T, Kojima T, Ogasawara N, Masaki T, Fuchimoto J, Kamekura R, et al. Poly(I:C) reduces expression of JAM-A and induces secretion of IL-8 and TNF-α via distinct NF-κB pathways in human nasal epithelial cells. Toxicol Appl Pharmacol 2011; 250(1): 29-38. https://doi.org/10.1016/j.taap.2010.09.023.
  37. Wu CA, Peluso JJ, Zhu L, Lingenheld EG, Walker ST, Puddington L. Bronchial epithelial cells produce IL-5: implications for local immune responses in the airways. Cell Immunol 2010; 264(1): 32-41. https://doi.org/10.1016/j.cellimm.2010.04.008.
  38. Arora S, Ahmad S, Irshad R, Goyal Y, Rafat S, Siddiqui N, et al. TLRs in pulmonary diseases. Life Sci 2019; 233: 116671. https://doi.org/10.1016/j.lfs.2019.116671.
  39. Ryu JH, Yoo JY, Kim MJ, Hwang SG, Ahn KC, Ryu JC, et al. Distinct TLR-mediated pathways regulate house dust mite–induced allergic disease in the upper and lower airways. J Allergy Clin Immunol 2013; 131(2): 549-561. https://doi.org/10.1016/j.jaci.2012.07.050.
  40. Ioannidis I, Ye F, McNally B, Willette M, Flaño E. TLR expression and induction of type I and type III interferons in primary airway epithelial cells. J Virol 2013; 87(6): 3261-3270. https://doi.org/10.1128/jvi.01956-12.
  41. Koller B, Kappler M, Latzin P, Gaggar A, Schreiner M, Takyar S, et al. TLR expression on neutrophils at the pulmonary site of infection: TLR1/TLR2-mediated up-regulation of TLR5 expression in cystic fibrosis lung disease. J Immunol 2008; 181(4): 2753-2763. https://doi.org/10.4049/jimmunol.181.4.2753.
  42. Bautista MV, Chen Y, Ivanova VS, Rahimi MK, Watson AM, Rose MC. IL-8 regulates mucin gene expression at the post transcriptional level in lung epithelial cells. J Immunol 2009; 183(3): 2159-2166. https://doi.org/10.4049/jimmunol.0803022.
  43. Nelemans T, Kikkert M. Viral innate immune evasion and the pathogenesis of emerging RNA virus infections. Viruses 2019; 11(10): 961. https://doi.org/10.3390/v11100961.
  44. Fehr AR, Channappanavar R, Perlman S. Middle East respiratory syndrome: emergence of a pathogenic human coronavirus. Annu Rev Med 2017; 68: 387-399. https://doi.org/10.1146/annurev-med-051215-031152.
  45. Newton AH, Cardani A, Braciale TJ. The host immune response in respiratory virus infection: balancing virus clearance and immunopathology. Semin Immunopathol 2016; 38(4): 471-482. https://doi.org/10.1007/s00281-016-0558-0.
  46. Channappanavar R, Zhao J, Perlman S. T cell-mediated immune response to respiratory coronaviruses. Immunol Res 2014; 59(1): 118-128. https://doi.org/10.1007/s12026-014-8534-z.
  47. Heil F, Hemmi H, Hochrein H, Ampenberger F, Kirschning C, Akira S, et al. Species-specific recognition of single-stranded RNA via toll-like receptors 7 and 8. Science 2004; 303(5663): 1526-1529. https://doi.org/10.1126/science.1093620.
  48. Diebold SS, Kaisho T, Hemmi H, Akira S, Reis e Sousa C. Innate antiviral responses by means of TLR7-mediated recognition of single-stranded RNA. Science 2004; 303(5663): 1529-1153. https://doi.org/10.1126/science.1093616.
  49. Melchjorsen J, Jensen SB, Malmgaard L, Rasmussen SB, Weber F, Bowie AG, et al. Activation of innate defense against a paramyxovirus is mediated by RIG-I and TLR7 and TLR8 in a cell-type-specific manner. J Virol 2005; 79(20): 12944-12951. https://doi.org/10.1128/jvi.79.20.12944-12951.2005.
  50. Brown JN, Kohler JJ, Coberley CR, Sleasman JW, Goodenow MM. HIV-1 activates macrophages independent of toll-like receptors. PloS One 2008; 3(12): e3664. https://doi.org/10.1371/journal.pone.0003664.
  51. Beignon AS, McKenna K, Skoberne M, Manches O, DaSilva I, Kavanagh DG, et al. Endocytosis of HIV-1 activates plasmacytoid dendritic cells via toll-like receptor-viral RNA interactions. J Clin Invest 2005; 115(11): 3265-3275. https://doi.org/10.1172/jci26032.
  52. Triantafilou K, Orthopoulos G, Vakakis E, Ahmed MA, Golenbock DT, Lepper PM, et al. Human cardiac inflammatory responses triggered by Coxsackie B viruses are mainly toll like receptor (TLR) 8-dependent. Cell Microbiol 2005; 7(8): 1117-1126. https://doi.org/10.1111/j.1462-5822.2005.00537.x.
  53. Cutler DJ, MacIntyre AC, Tett SE. Pharmacokinetics and cellular uptake of 4-aminoquinoline antimalarials. Agents Actions Suppl 1988; 24: 142-157. https://doi.org/10.1007/978-3-0348-9160-8_13.
  54. de Wit E, Van Doremalen N, Falzarano D, Munster VJ. SARS and MERS: Recent insights into emerging coronaviruses. Nat Rev Microbiol 2016; 14(8): 523-534. https://doi.org/10.1038/nrmicro.2016.81.
  55. Jamilloux Y, Henry T, Belot A, Viel S, Fauter M, El Jammal T, et al. Should we stimulate or suppress immune responses in COVID-19? Cytokine and anti-cytokine interventions. Autoimmun Rev 2020; 19(7): 102567. https://doi.org/10.1016/j.autrev.2020.102567.
  56. Yan R, Zhang Y, Li Y, Xia L, Guo Y, Zhou Q. Structural basis for the recognition of the SARS-CoV-2 by full-length human ACE2. Science 2020; 367(6485): 1444-1448. https://doi.org/10.1126/science.abb2762
  57. Tartey S. Takeuchi O. Pathogen recognition and toll-like receptor targeted therapeutics in innate immune cells. Int Rev Immunol 2017; 36(2): 57-73. https://doi.org/10.1080/08830185.2016.1261318 .
  58. Feng G, Zheng KI, Yan QQ, Rios RS, Targher G, Byrne CD, et al. COVID-19 and liver dysfunction: Current insights and emergent therapeutic strategies. J Clin Transl Hepatol 2020; 8(1): 18-24. https://doi.org/10.14218/jcth.2020.00018.
  59. Russell B, Charlotte M, Gincy G, Santaolalla A, Cope A, Papa S, et al. Associations between immune-suppressive and stimulating drugs and novel COVID-19. Ecancermedicalscience 2020; 14: 1022. https://doi.org/10.3332/ecancer.2020.1022.
  60. Conti P, Ronconi G, Caraffa A, Gallenga CE, Ross R, Frydas I, et al. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by COVID-19: anti-inflammatory strategies. J Biol Regul Homeost Agents 2020; 34(2): 327-331. https://doi.org/10.23812/conti-e.
  61. Zhao W, An H, Zhou J, Xu H, Yu Y, Cao X. Hyperthermia differentially regulates TLR4 and TLR2-mediated innate immune response. Immunol Lett 2007; 108(2): 137-142. https://doi.org/10.1016/j.imlet.2006.11.008 .
  62. Rabi FA, Al Zoubi MS, Kasasbeh GA, Salameh DM, Al-Nasser AD. SARS-CoV-2 and coronavirus disease 2019: what we know so far. Pathogens 2020; 9(3): 231. https://doi.org/10.3390/pathogens9030231.
  63. Totura AL, Baric RS. SARS coronavirus pathogenesis: host innate immune responses and viral antagonism of interferon. Curr Opin Virol 2012; 2(3): 264-275. https://doi.org/10.1016/j.coviro.2012.04.004 .
  64. Cao B, Wang Y, Wen D, Wen L, Wang J, Fan G, et al. A trial of lopinavir-ritonavir in adults hospitalized with severe COVID-19. N Engl J 2020; 382(19): 1787-1799. https://doi.org/10.1056/nejmoa2001282.
  65. Jezierska A, Kolosova IA, Verin AD. Toll like receptors signaling pathways as a target for therapeutic interventions. Curr Signal Transduct Ther 2011; 6(3): 428-440. https://doi.org/10.2174/157436211797483930.
  66. Murata M, Sato T, Miyauchi M, Koga E, Aoki M, Kimura T, Kashiwazaki Y. SMP-105, cell-wall skeleton purified from Mycobacterium bovis BCG Tokyo 172, activates innate immunity through TLR2/MyD88 pathway and prevents tumor metastasis into draining lymph nodes. Cancer Res 2007; 67(9 Suppl): 3554. https://cancerres.aacrjournals.org/content/67/9_Supplement/3554.
  67. Aumeunier A, Grela F, Ramadan A, Pham Van L, Bardel E, Gomez Alcala A, et al. Systemic toll-like receptor stimulation suppresses experimental allergic asthma and autoimmune diabetes in NOD mice. PLoS One 2010; 5(7): e11484. https://doi.org/10.1371/journal.pone.0011484.
  68. Revets H, Pynaert G, Grooten J, De Baetselier P. Lipoprotein I. TLR2/4 ligand modulates Th2-driven allergic immune responses. J Immunol 2005; 174(2): 1097-1103. https://doi.org/10.4049/jimmunol.174.2.1097.
  69. Overton ET, Goepfert PA, Cunningham P, Carter WA, Horvath J, Young D, Intranasal seasonal influenza vaccine and a TLR-3 agonist, rintatolimod, induced cross-reactive IgA antibody formation against avian H5N1 and H7N9 influenza HA in humans. Vaccine 2014; 32(42): 5490-5495. https://doi.org/10.1016/j.vaccine.2014.07.078.
  70. Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Aspects Med 2014; 40: 1-116. https://doi.org/10.1016/j.mam.2014.05.001.
  71. Yang H, Wang H, Ju Z, Ragab AA, Lundbäck P, Long W, et al. MD-2 is required for disulfide HMGB1-dependent TLR4 signaling. J Exp Med 2015; 212(1): 5–14. https://doi.org/10.1084/jem.20141318.
  72. Lee K, Kim SH, Yoon HJ, Paik DJ, Kim JM, Youn J. Bacillus-derived poly-gamma-glutamic acid attenuates allergic airway inflammation through a toll-like receptor-4-dependent pathway in a murine model of asthma. Clin Exp Allergy 2011; 41(8): 1143–1156. https://doi.org/10.1111/j.1365-2222.2011.03792.x.
  73. Bortolatto J, Borducchi E, Rodriguez D, Keller AC, Faquim-Mauro E, Bortoluci KR, Toll-like receptor 4 agonists adsorbed to aluminium hydroxide adjuvant attenuate ovalbumin-specific allergic airway disease: role of MyD88 adaptor molecule and interleukin-12/interferon-gamma axis. Clin Exp Allergy 2008; 38(10): 1668-1679. https://doi.org/10.1111/j.1365-2222.2008.03036.x.
  74. Diebold SS, Massacrier C, Akira S, Paturel C, Morel Y, Reis e Sousa C. Nucleic acid agonists for toll-like receptor 7 are defined by the presence of uridine ribonucleotides. Eur J Immunol 2006; 36(12): 3256-3267. https://doi.org/10.1002/eji.200636617.
  75. Forsbach A, Nemorin JG, Montino C, Muller C, Samulowitz U, Vicari AP, et al. Identification of RNA sequence motifs stimulating sequence-specific TLR8-dependent immune responses. J Immunol 2008; 180(6): 3729-3738. https://doi.org/10.4049/jimmunol.180.6.3729.
  76. Lan T, Kandimalla ER, Yu D, Bhagat L, Li Y, Wang D, et al. Stabilized immune modulatory RNA compounds as agonists of toll-like receptors 7 and 8. Proc Natl Acad Sci 2007; 104(34): 13750-13755. https://doi.org/10.1073/pnas.0706059104.
  77. Lan T, Dai M, Wang D, Zhu FG, Kandimalla ER, Agrawal S. Toll-like receptor 7 selective synthetic oligoribonucleotide agonists: synthesis and structure-activity relationship studies. J Med Chem 2009; 52: 6871-6879. https://doi.org/10.1021/jm901145s.
  78. Tanji H, Ohto U, Shibata T, Taoka M, Yamauchi Y, Isobe T, et al. Toll-like receptor 8 senses degradation products of single-stranded RNA. Nat Struct Mol Biol 2015; 22(2): 109-115. https://doi.org/10.1038/nsmb.2943.
  79. Zhang Z, Ohto U, Shibata T, Krayukhina E, Taoka M, Yamauchi Y, et al. Structural analysis reveals that toll-like receptor 7 is a dual receptor for guanosine and single-stranded RNA. Immunity 2016; 45(4): 737-748. https://doi.org/10.1016/j.immuni.2016.09.011.
  80. Drake MG, Kaufman EH, Fryer AD, Jacoby DB. The therapeutic potential of toll-like receptor 7 stimulation in asthma. Inflamm Allergy Drug Targets 2012; 11(6): 484-491. https://doi.org/10.2174/187152812803589967.
  81. Gorden KB, Gorski KS, Gibson SJ, Kedl RM, Kieper WC, Qiu X, et al. Synthetic TLR agonists reveal functional differences between human TLR7 and TLR8. J Immunol 2005; 174(3): 1259-1268. https://doi.org/10.4049/jimmunol.174.3.1259.
  82. Peng G, Guo Z, Kiniwa Y, Voo KS, Peng W, Fu T, et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science 2005; 309(5739): 1380-1384. https://doi.org/10.1126/science.1113401.
  83. Isobe Y, Kurimoto A, Tobe M, Hashimoto K, Nakamura T, Norimura K, et al. Synthesis and biological evaluation of novel 9 substituted-8-hydroxyadenine derivatives as potent interferon inducers. J Med Chem 2006; 49(6): 2088-2095. https://doi.org/10.1021/jm051089s.
  84. Bergmann JF, de Bruijne J, Hotho DM, de Knegt RJ, Boonstra A, Weegink CJ, et al. Randomised clinical trial: anti-viral activity of ANA773, an oral inducer of endogenous interferons acting via TLR7, in chronic HCV. Aliment Pharmacol Ther 2011; 34(4): 443-453. https://doi.org/10.1111/j.1365-2036.2011.04745.x.
  85. Vollmer J, Krieg AM. Immunotherapeutic applications of CpG oligodeoxynucleotide TLR9 agonists. Adv Drug Deliv Rev 2009; 61(3): 195-204. https://doi.org/10.1016/j.addr.2008.12.008.
  86. Senti G, Johansen P, Haug S, Bull C, Gottschaller C, Müller P, et al. Use of A-type CpG oligodeoxy nucleotides as an adjuvant in allergen-specific immunotherapy in humans: a phase I/IIa clinical trial. Clin Exp Allergy 2009; 39(4): 562-570. https://doi.org/10.1111/j.1365-2222.2008.03191.x.
  87. Gupta GK, Agrawal DK. CpG oligodeoxynucleotides as TLR9 agonists: therapeutic application in allergy and asthma. BioDrugs 2010; 24(4): 225-235. https://doi.org/10.2165/11536140-000000000-00000.
  88. Serebrisky D, Teper AA, Huang CK, Lee SY, Zhang TF, Schofield BH, et al. CpG oligodeoxynucleotides can reverse Th2-associated allergic airway responses and alter the B7.1/B7.2 expressions in a murine model of asthma. J Immunol 2000; 165(10): 5906-5912. https://doi.org/10.4049/jimmunol.165.10.5906.
  89. Banerjee B, Kelly KJ, Fink JN, Henderson JD Jr, Bansal NK, Kurup VP. B. Modulation of airway inflammation by immunostimulatory CpG oligodeoxynucleotides in a murine model of allergic aspergillosis. Infect Immunol 2004; 72(10): 6087-6094. https://doi.org/10.1128/iai.72.10.6087-6094.2004.
  90. Chiang DJ, Ye YL, Chen WL, Lee YL, Hsu NY, Chiang BL. Ribavirin or CpG DNA sequence-modulated dendritic cells decrease the IgE level and airway inflammation. Am J Respir Crit Care Med 2003; 168(5): 575-580. https://doi.org/10.1164/rccm.2205005.
  91. Agrawal S, Kandimalla ER. Synthetic agonists of toll like receptors 7, 8 and 9. Biochem Soc Trans 2007; 35(Pt 6): 1461-1467. https://doi.org/10.1042/bst0351461.
  92. Panter G, Kuznik A, Jerala R. Therapeutic applications of nucleic acids as ligands for toll-like receptors. Curr Opin Mol Ther 2009; 11(2): 133-145. https://pubmed.ncbi.nlm.nih.gov/19330719/.
  93. [93] Shirey KA, Lai W, Scott AJ, Lipsky M, Mistry P, Pletneva LM, et al. The TLR4 antagonist eritoran protects mice from lethal influenza infection. Nature 2013; 497(7450): 498-502. https://doi.org/10.1038/nature12118.
  94. Perrin-Cocon L, Aublin-Gex A, Sestito SE, Shirey KA, Patel MC, André P, et al. TLR4 antagonist FP7 inhibits LPS-induced cytokine production and glycolytic reprogramming in dendritic cells, and protects mice from lethal influenza infection. Sci Rep 2017; 7: 40791. https://doi.org/10.1038/srep40791.
  95. Piao W, Shirey KA, Ru LW, Lai W, Szmacinski H, Snyder GA, et al. A decoy peptide that disrupts TIRAP recruitment to TLRs is protective in a murine model of influenza. Cell Rep 2015; 11(12): 1941-1952. https://doi.org/10.1016/j.celrep.2015.05.035.
  96. Andersson Ulf, William O, Kevin J. Extracellular HMGB1: a therapeutic target in severe pulmonary inflammation including COVID 19? Mol Med 2020; 26(1): 42. https://doi.org/10.1186/s10020-020-00172-4.
  97. Yang H, Hreggvidsdottir HS, Palmblad K, Wang H, Ochani M, Li J, et al. A critical cysteine is required for HMGB1 binding to toll-like receptor 4 and activation of macrophage cytokine release. Proc Natl Acad Sci USA 2010; 107(26): 11942-11947. https://doi.org/10.1073/pnas.1003893107.
  98. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite allergen induces asthma via toll-like receptor 4 triggering of airway structural cells. Nat Med 2009; 15(4): 410-416. https://doi.org/10.1038/nm.1946.
  99. Sundaram K, Mitra S, Gavrilin MA, Wewers MD. House dust mite allergens and the induction of monocyte interleukin 1beta production that triggers an I kappa Bzeta-dependent granulocyte macrophage colony-stimulating factor release from human lung epithelial cells. Am J Respir Cell Mol Biol 2015; 53(3): 400-411. https://doi.org/10.1165/rcmb.2014-0370oc.
  100. Monnet E, Lapeyre G, Poelgeest EV, Jacqmin P, Graaf K, Reijers J, et al. Evidence of NI-0101 pharmacological activity, an anti-TLR4 antibody, in a randomized phase I dose escalation study in healthy volunteers receiving LPS. Clin Pharmacol Ther 2017; 101(2): 200-208. https://doi.org/10.1002/cpt.522.
  101. Ungaro R, Fukata M, Hsu D, Hernandez Y, Breglio K, Chen A, et al. A novel toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol Gastrointest Liver Physiol 2009; 296(6): G1167-G1179. https://doi.org/10.1152/ajpgi.90496.2008.
  102. Shirey KA, Lai W, Patel MC, Pletneva LM, Pang C, Kurt-Jones E, et al. Novel strategies for targeting innate immune responses to influenza. Mucosal Immunol 2016; 9(5): 1173-1182. https://doi.org/10.1038/mi.2015.141.
  103. Savov JD, Brass DM, Lawson BL, McElvania-Tekippe E, Walker JK, Schwartz DA. Toll-like receptor 4 antagonist (E5564) prevents the chronic airway response to inhaled lipopolysaccharide. Am J Physiol Lung Cell Mol Physiol 2005; 289(2): L329-L337. https://doi.org/10.1152/ajplung.00014.2005.
  104. Hennessy E, Parker AE, O’Neill L. Targeting toll-like receptors: emerging therapeutics? Nat Rev Drug Discov 2010; 9(4): 293-307. https://doi.org/10.1038/nrd3203.
  105. Hemmi H, Kaisho T, Takeuchi O, Sato S, Sanjo H, Hoshino K, et al. Small anti-viral compounds activate immune cells via the TLR7 MyD88-dependent signaling pathway. Nat Immunol 2002; 3(2): 196-200. https://doi.org/10.1038/ni758.
  106. Mahmutovic-Persson I, Johansson M, Brandelius A, Calvén J, Bjermer L, Yudina Y, et al. Capacity of capsazepinoids to relax human small airways and inhibit TLR3-induced TSLP and IFN beta production in diseased bronchial epithelial cells. Int Immunopharmacol 2012; 13(3): 292-300. https://doi.org/10.1016/j.intimp.2012.04.007.
  107. Yang Y, Li S, Yang Q, Shi Y, Zheng M, Liu Y, et al. Resveratrol reduces the proinflammatory effects and lipopolysaccharide- induced expression of HMGB1 and TLR4 in RAW264.7 cells. Cell Physiol Biochem 2014; 33(5): 1283-1292. https://doi.org/10.1159/000358696.
  108. Le K, Chibaatar Daliv E, Wu S, Qian F, Ali AI, Yu D, et al. SIRT1-regulated HMGB1 release is partially involved in TLR4 signal transduction: a possible anti- neuroinflammatory mechanism of resveratrol in neonatal hypoxic-ischemic brain injury. Int Immunopharmacol 2019; 75: 105779. https://doi.org/10.1016/j.intimp.2019.105779.
  109. Zang N, Xie X, Deng Y, Wu S, Wang L, Peng C, et al. Resveratrol-mediated gamma interferon reduction prevents airway inflammation and airway hyperresponsiveness in respiratory syncytial virus-infected immunocompromised mice. J Virol 2011; 85(24): 13061-13068. https://doi.org/10.1128/jvi.05869-11.
  110. Schrezenmeier E, Dörner T. Mechanisms of action of hydroxychloroquine and chloroquine: implications for rheumatology. Nat Rev Rheumatol 2020; 16(3): 155-166. https://doi.org/10.1038/s41584-020-0372-x.
  111. Kuznik A, Bencina M, Svajger U, Jeras M, Rozman B, Jerala R. Mechanism of endosomal TLR inhibition by antimalarial drugs and imidazoquinolines. J Immunol 2011; 186(8): 4794-4804. https://doi.org/10.4049/jimmunol.1000702.
  112. Ewald SE, Lee BL, Lau L, Wickliffe KE, Shi GP, Chapman HA, et al. The ectodomain of toll-like receptor 9 is cleaved to generate a functional receptor. Nature 2008; 456(7222): 658-662. https://doi.org/10.1038/nature07405.
  113. Zhou D, Dai SM, Tong Q. COVID-19: a recommendation to examine the effect of hydroxychloroquine in preventing infection and progression. J Antimicro Chemother 2020; 75(7): 1667-1670. https://doi.org/10.1093/jac/dkaa114.
  114. van den Borne BE, Dijkmans BA, de Rooij HH, le Cessie S, Verweij CL. Chloroquine and hydroxychloroquine equally affect tumor necrosis factor-alpha, interleukin 6, and interferon-gamma production by peripheral blood mononuclear cells. J Rheumatol 1997; 24(1): 55-60. https://pubmed.ncbi.nlm.nih.gov/9002011.
  115. Saitoh S, Miyake K. Regulatory molecules required for nucleotide-sensing toll like receptors. Immunol Rev 2009; 22(1): 32-43. https://doi.org/10.1111/j.1600-065x.2008.00729.x.
  116. Lim EJ, Lee SH, Lee JG, Chin BR, Bae YS, Kim JR, et al. Activation of toll-like receptor-9 induces matrix metalloproteinase-9 expression through Akt and tumor necrosis factor-alpha signaling. FEBS Lett 2006; 580(18): 4533-4538. https://doi.org/10.1016/j.febslet.2006.06.100.
  117. Wang M, Cao R, Zhang L, Yang X, Liu J, Xu M, et al. Remdesivir and chloroquine effectively inhibit the recently emerged novel coronavirus (2019-nCoV) in vitro. Cell Res 2020; 30(3): 269-271. https://doi.org/10.1038/s41422-020-0282-0.
  118. Sheahan TP, Sims AC, Graham RL, Menachery VD, Gralinski LE, Case JB, et al. Broad-spectrum antiviral GS-5734 inhibits both epidemic and zoonotic coronaviruses. Sci Transl Med 2017; 9(396): eaal3653. https://doi.org/10.1126/scitranslmed.aal3653.
  119. Sheahan TP, Sims AC, Leist SR, Schäfer A, Won J, Brown AJ, et al. Comparative therapeutic efficacy of remdesivir and combination lopinavir, ritonavir, and interferon beta against MERS-CoV. Nat Commun 2020; 11(1): 222. https://doi.org/10.1038/s41467-019-13940-6.
  120. de Wit E, Feldmann F, Cronin J, Jordan R, Okumura A, Thomas T, et al. Prophylactic and therapeutic remdesivir (GS-5734) treatment in the rhesus macaque model of MERS-CoV infection. Proc Natl Acad Sci U S A 2020; 117(12): 6771-6776. https://doi.org/10.1073/pnas.1922083117.
  121. Holshue ML, DeBolt C, Lindquist S, Lofy KH, Wiesman J, Bruce H, et al. First case of 2019 novel coronavirus in the United States. N Engl J Med 2020; 382(10): 929-936. https://doi.org/10.1056/nejmoa2001191.
  122. Enayatkhania M, Hasaniazad M, Faezi S, Guklani H, Davoodian P, Ahmadi N, et al. Reverse vaccinology approach to design a novel multi-epitope vaccine candidate against COVID-19: an in-silico study. J Biomol Struct Dyn 2020; 39(8): 2857-2872. https://doi.org/10.1080/07391102.2020.1756411.
About the Authors: 

Amany Sayed Maghraby – Professor, Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Division, Immune- and Bio-markers for Infection Group, the Center of Excellence for Advanced Sciences, National Research Center, Cairo, Egypt. http://orcid.org/0000-0002-9723-5011

Received 26 Novemberl 2020, Revised 23 February 2021, Accepted 3 April 2021 
© 2020, Maghraby A.S.   
© 2020, Russian Open Medical Journal 
Correspondence to Amany Sayed Maghraby. Address: El Buhouth Street, Dokki, Cairo 12622, Egypt. E-mail: as.maghraby@nrc.sci.eg.

DOI: 
10.15275/rusomj.2021.0202
Author(s):