Next Article in Journal
Transformation of Cellulose via Two-Step Carbonization to Conducting Carbonaceous Particles and Their Outstanding Electrorheological Performance
Next Article in Special Issue
Could a Lower Toll-like Receptor (TLR) and NF-κB Activation Due to a Changed Charge Distribution in the Spike Protein Be the Reason for the Lower Pathogenicity of Omicron?
Previous Article in Journal
Antitumoral Activity of a CDK9 PROTAC Compound in HER2-Positive Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toll-like Receptor Response to Hepatitis C Virus Infection: A Recent Overview

by
Mohammad Enamul Hoque Kayesh
1,*,
Michinori Kohara
2 and
Kyoko Tsukiyama-Kohara
3,*
1
Department of Microbiology and Public Health, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Barishal 8210, Bangladesh
2
Department of Microbiology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
3
Transboundary Animal Diseases Centre, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(10), 5475; https://doi.org/10.3390/ijms23105475
Submission received: 21 April 2022 / Revised: 11 May 2022 / Accepted: 13 May 2022 / Published: 13 May 2022
(This article belongs to the Special Issue The Role of Toll-Like Receptors (TLR) in Infection and Inflammation)

Abstract

:
Hepatitis C virus (HCV) infection remains a major global health burden, causing chronic hepatitis, cirrhosis, and hepatocellular carcinoma. Toll-like receptors (TLRs) are evolutionarily conserved pattern recognition receptors that detect pathogen-associated molecular patterns and activate downstream signaling to induce proinflammatory cytokine and chemokine production. An increasing number of studies have suggested the importance of TLR responses in the outcome of HCV infection. However, the exact role of innate immune responses, including TLR response, in controlling chronic HCV infection remains to be established. A proper understanding of the TLR response in HCV infection is essential for devising new therapeutic approaches against HCV infection. In this review, we discuss the progress made in our understanding of the host innate immune response to HCV infection, with a particular focus on the TLR response. In addition, we discuss the mechanisms adopted by HCV to avoid immune surveillance mediated by TLRs.

1. Introduction

Hepatitis C virus (HCV) infection is a major global health burden [1,2]. HCV infection frequently causes chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma (HCC) [3]. According to the World Health Organization, 58 million people worldwide are chronically infected with HCV, with approximately 1.5 million new infections occurring each year [4]. HCV is an enveloped, positive-sense, single-stranded RNA virus belonging to the genus Hepacivirus and the family Flaviviridae [5]. HCV has a ∼10-kb long genome, encoding a large polyprotein of approximately 3000 amino acids that is processed by host and viral proteases into three structural (core, E1, and E2) and seven non-structural (NS) proteins (p7, NS2, NS3, NS4A, NS4B, NS5A, and NS5B) [6]. HCV has high genetic diversity resulting in seven major genotypes and more than 60 subtypes [7].
The innate immune system, an essential component of host immunity, plays a key role in the initial detection of invading pathogens, including viruses, and subsequently activates adaptive immunity, thereby playing an important role in the early control of viral infection [8,9,10,11]. The host innate immune response is activated upon microbial invasion and detection of evolutionarily conserved structures found on pathogens, called pathogen-associated molecular patterns (PAMPs), by germ-line-encoded pattern recognition receptors (PRRs) [12]. PRRs also recognize molecules released by damaged cells, known as damage-associated molecular patterns [13]. Different PRRs, including Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs), AIM2-like receptors (ALRs), C-type lectin receptors (CLRs), and intracellular DNA sensors such as cGAS, are key innate immune components that recognize viral components such as viral nucleic acids and proteins [14,15]. However, TLRs are the most widely characterized PRRs, constituting key components of innate immunity, and are involved in the early interaction with PAMPs of invading microbes [16].
TLRs are evolutionarily conserved type I transmembrane proteins that contain a conserved structure of an N-terminal ectodomain of leucine-rich repeats, a single transmembrane domain, and a cytosolic Toll/interleukin (IL)-1 receptor (TIR) domain [11,17]. The cytosolic TIR domain is responsible for the activation of downstream signaling, and TLR signaling pathways are regulated by TIR domain-containing cytosolic adaptor proteins such as myeloid differentiation factor 88 (MyD88), MyD88 adaptor-like (MAL or TIRAP), TIR-domain-containing adaptor protein inducing interferon (IFN)-β (TRIF or TICAM1), TRIF-related adaptor molecule (TRAM or TICAM2), and sterile α- and armadillo-motif-containing protein (SARM) [8,18,19]. The adaptor protein MyD88 is used by nearly all TLR signaling pathways, except TLR3 [20]. Members of each TLR family have similar functions across species [21,22]. TLRs are encoded by a large gene family, and different organisms appear to encode a certain number of TLRs. For example, the TLR family comprises 10 members (TLR1–TLR10) in humans and 12 members (TLR1–TLR9 and TLR11–TLR13) in mice [23]. TLRs can be localized either on the cell surface, such as TLRs 1, 2, 4, 5, 6, and 10, or in intracellular compartments (e.g., the endoplasmic reticulum, endosome, lysosome, or endolysosome) such as TLRs 3, 7, 8, and 9 [23,24,25]. TLR1, TLR2, TLR4, TLR5, and TLR6 play pivotal roles in viral protein recognition [26]. To recognize viral double-stranded RNA, single-stranded RNA, and DNA, the membrane proteins TLR3, TLR7/8, and TLR9 are used, respectively [27,28,29,30].
TLR-induced innate immune responses appear to be a prerequisite for the generation of most adaptive immune responses and play a central role in shaping such responses [25,31]. To provide protection against invading microbes, TLR responses ultimately lead to the induction of IFNs, cytokines, and chemokines via several distinct signaling pathways, which is very important in limiting the spread of infection [14,31,32]. TLR agonists appear to be potential immunomodulators for treating infections and play a significant role in modulating immunotherapeutic effects [33,34,35,36]. However, the TLR response may not always be beneficial to the host, but a dysregulated response may lead to immune-mediated pathology rather than protection [37,38,39,40]. Therefore, a proper understanding of the TLR response in any infection, including HCV infection, is critical. Against this background, we discuss here the current progress made in our understanding of the host TLR response in HCV infection and the mechanisms adopted by HCV to avoid TLR-mediated immune surveillance, which may help in devising new therapeutic or preventive strategies.

2. TLR Response to HCV Infection

A complex interplay between the IFN system and viral countermeasures exists in HCV infections [41]. During viral replication, HCV PAMPs can be recognized as non-self by PRRs, leading to the activation of innate and adaptive immune responses [42]. TLRs are important PRRs that recognize PAMPs present in HCV [43]. TLRs are key triggering molecules for cytokine production, and TLR signaling pathways provide a link between innate and acquired immunity [15,44]. Different TLRs have been found to interact with HCV proteins and nucleic acids. It has been reported that HCV is sensed by TLR3 through the detection of dsRNA intermediates in infected hepatoma cells, which may activate the TLR3-signaling cascade and lead to the production of type I and II IFNs, expression of interferon-stimulated genes, and proinflammatory cytokines limiting HCV replication [45]. Induction of TLR2 and TLR4 expression and modulation of the proinflammatory response by HCV proteins have been reported in Raji cells and peripheral blood mononuclear cells (PBMCs) [46,47]. Several other studies also found increased expression of TLR2 and TLR4 mRNA in chronic hepatitis patients compared to controls [48,49]. However, TLR4 mRNA expression was downregulated in cirrhotic patients when compared to chronic hepatitis patients [48]. Hypo-responsiveness to TLR ligands has been reported in patients with chronic HCV infection [50]. Differential expression of TLRs has been reported in HCV-infected patients [51,52], where increased expression of tumor necrosis factor (TNF)-α, IL-6, and IL-12 p35 in PBMCs was also reported [52]. It has been observed that NS5A can activate the promoter of the TLR4 gene in both hepatocytes and B-cells, and enhanced TLR4 expression induces the induction of IFN-β and IL-6 production in human B-cells [46].
The HCV core and NS3 proteins play significant roles in HCV pathogenesis; it has been shown that they can trigger proinflammatory cytokine production in monocytes, inhibit myeloid dendritic cell accessory cell functions, and provide immunoinhibitory effects via IL-10 induction [53]. Other studies have reported that HCV viral proteins, including HCV core and NS3, could be recognized by TLR2, triggering the activation of inflammatory cells [54,55]. Impaired recognition of HCV core and NS3 proteins caused by the R753Q SNP in TLR2 has been reported [55]. HCV core and NS3 proteins may enhance the activity of IL-1 receptor-associated kinase (IRAK), phosphorylation of p38, extracellular regulated (ERK), and c-jun N-terminal (JNK) kinases and induce the activation of activator protein 1 (AP-1) [54]. In an in vitro study, Chang et al. reported an association between TLR1 and TLR6 in the TLR2-mediated activation of macrophages by HCV core and NS3 proteins [56]. While the involvement of TLR2 in sensing HCV core protein has been observed previously, infectious virions or enveloped HCV-like particles do not activate TLR2 [57]. However, another study showed that HCV core protein can activate TLR2 with decreased IL-6 production by human antigen-presenting cells after subsequent stimulation with TLR2 and TLR4 ligands [58]. HCV core protein may affect pDCs by reducing TLR9-triggered IFN-α as well as TNF-α and IL-10 production [59].
HCV core and NS3 antigens induce TLR1-, TLR2-, and TLR6-mediated inflammatory responses in corneal epithelial cells [60]. Compared to healthy controls, upregulation of TLR2 and TLR4 expression in peripheral monocytes was also observed in patients with chronic hepatitis C, with or without HIV coinfection [61]. An association between TLR4 signaling and the outcome of acute hepatitis C has also been reported [62]. Activation of the TLR3/TRIF signaling pathway by HCV NS5B, a viral RNA-dependent RNA polymerase, has been reported [63]. In addition to TLR3, HCV RNA can also be recognized by RIG-I, triggering the production of multiple cytokines, including type I IFN [42]. In a murine replicon model, the antiviral role of TLR4 activation in suppressing HCV replication was demonstrated [64]. An association between TLR4 gene polymorphisms and chronic HCV infection has also been reported in a Saudi Arabian population [65], suggesting a putative role of TLR4 in HCV infection. However, a larger genome-wide association study is required to validate these associations.
As HCV has an RNA genome, it is likely that TLR7 plays a role in the immune response against HCV infection. Both TLR3 and TLR7 have been suggested to coordinate protective immunity against HCV infection [45,66,67]. A decreased expression of TLR3 and TLR7 mRNA has been reported in chronic HCV patients with a decreased IFN-α expression compared to healthy controls [68,69,70]. An earlier study reported significantly elevated expression of TLR3 in individuals who spontaneously cleared the virus [71], suggesting a protective role of TLR3 in HCV genotype 3 infection. TLR7 and TLR8 were also found to be elevated in patients with liver cirrhosis [71]. A previous study reported that impaired TLR3- and TLR7/8-mediated cytokine responses may contribute to aggressive HCV recurrence after liver transplantation [72], also indicating the association of these molecules with HCV infection. However, more extensive in vivo studies are required to understand their use in protective immune responses against HCV infections.
It has been reported that TLR7 can sense HCV RNA in exosomes released from infected hepatocytes, inducing type I IFN response [73,74]. An in vitro study demonstrated that TLR7 can induce HCV immunity not only by IFN induction but also via an IFN-independent mechanism [75]. The antiviral roles of TLR7 and TLR8 have also been suggested in HCV infection [76]. In a previous study, it was shown that HCV could be recognized and inhibited by TLR7 and TLR8 via TNF-α production [77]. HCV genomic RNA-induced TLR7- and TLR8-mediated anti-HCV immune responses have been reported in various antigen-presenting cells [78]. Polymorphisms in TLR7 and/or TLR8 genes have been shown to modulate HCV infection outcomes [79,80,81], suggesting an association between these molecules and HCV infection. The TLR9 rs187084 C allele was reported to be associated with spontaneous virus clearance in women, suggesting the sex-specific effects of TLR9 promoter variants on spontaneous clearance in HCV infection and implying the role of the DNA sensor TLR9 in natural immunity against HCV infection [82]. In a recent study, an association between TLR9 gene polymorphisms and the outcome of the HCV-specific cell-mediated immune response was reported [83], indicating a putative role of TLR9 in HCV infection. A suitable small animal model is still lacking for HCV infection, which is essential for a proper understanding of the TLR response to HCV infection. Tree shrews appear to be a promising animal model for several important viral infections in humans, including hepatitis C virus [84], and show a higher degree of genetic similarity to primates than to rodents [85,86]. In an earlier study, we showed that HCV could trigger innate immune responses in the livers of chronically infected tree shrews, with significant induction of intrahepatic TLR3, TLR7, and TLR8 mRNA [87]. For simplicity, the findings obtained in different studies are shown in Figure 1, without indication of cell type/system, and highlight that various TLRs are implicated in HCV infection, which may influence viral pathogenesis. Therefore, a complete understanding of the TLR response in HCV infection is critical for designing new and successful therapeutic or preventive interventions.

3. Inhibition of Innate Immune Response by HCV Infection

The host has evolved multifaceted innate immune mechanisms to sense and counteract HCV infection. However, the success of innate IFN response in inhibiting HCV infection remains low. In a large proportion of patients, HCV persistence has been observed for decades despite the expression of hundreds of interferon-stimulated genes, indicating the inability of the IFN system to clear HCV infection [30,88,89]. In addition, HCV has developed multiple strategies for innate immune regulation, including proteolytic cleavage of molecules that play key roles in the induction of the IFN response, changes in IFN-induced effector proteins, interference with the function of CD8+ T cells, and immune escape in T- and B-cell epitopes [90]. HCV encodes several proteins, including core, NS3/4A, NS4B, and NS5A, which play active roles in inhibiting the innate immune response [42,91,92,93]. HCV frameshift (F) protein, which is expressed by a translational ribosomal frameshift [94], has also been suggested to play a role in the immune evasion mechanism [92].
NS3/4A plays a central role in HCV pathogenesis by cleaving several host proteins [95,96,97]. HCV NS3/4A has the potential to cleave TRIF and impair the TLR3-dependent signaling pathway [45,93,98]. It has also been reported that NS3/4A protease can disrupt RIG-I signaling by cleavage or delocalization of IFN-β promoter stimulator 1, also known as mitochondrial antiviral signaling protein (MAVS), preventing downstream activation of IRF-3 and IFN-β induction [93,98,99,100,101,102]. Notably, cleavage of MAVS by NS3/4A has been reported in the infected human liver, demonstrating the importance of this cleavage in HCV infection in vivo [101,103]. HCV proteins may interfere with IFN-induced intracellular signaling, which could be an important mechanism for viral persistence and treatment resistance [104]. HCV core may inhibit the IFN-signaling pathway by interfering with the Janus kinase/signal transducer and activator of the transcription pathway [105,106,107]. Several studies have reported the inhibition or degradation of STAT1 by HCV core protein [104,108,109,110,111], which may inhibit the JAK/STAT-signaling pathway of the host response. Reduced levels of STAT2 phosphorylation caused by HCV core proteins have also been reported [112,113].
Several studies have shown the implication of HCV core protein in the activation of the NF-κB pathway for inducing inflammatory response [114,115,116,117]; however, an implication of HCV core protein in the suppression of the NF-κB pathway has also been reported [118]. STAT3 has been found to be downregulated in HCV-infected livers and in Huh7 cells [119], which may favor viral replication. Impaired TLR4 signaling in HCV-infected dendritic cells has been previously demonstrated [120]. It has been reported that NS4B can interact with the stimulator of IFN genes (STING), which may cause inhibition of downstream signaling [91,121,122]. HCV NS4A, NS4B, and NS5A may inhibit IFN-β induction, contributing to the persistence of this virus [63]. NS5A can directly bind to MyD88, a major adaptor molecule in TLR signaling, inhibiting the activation of TLR-mediated cytokine production [123]. NS5A can induce IL-8 expression, associated with the interruption of IFN-α [124]. NS5A also blocks the antiviral activity of 2′–5′ oligoadenylate synthetase (2′–5′ OAS) [125]. It has been reported that HCV may utilize NS5A and E2 to inhibit PKR-mediated antiviral defense [126,127,128]. A previous study also suggested a role for NS5A in the inhibition of the IFN response that is activated by HCV via RIG-I and MDA5 [129]. NS5A may interact with nucleosome assembly protein 1-like 1 (NAP1L1), a nuclear-cytoplasmic chaperone, which may downregulate genes essential for innate immunity, such as RIG-I- and TLR3-mediated responses [130].
Chronic HCV infection results in impaired TLR response in pDCs as well as impaired activation of naive CD4 T cells, with reduced activation marker (HLA-DR) and cytokine (IFN-α) expression upon R-848 stimulation [131]. Samrat et al. reported that HCV core and F protein can induce poor T-cell responses, resulting in low granzyme B expression by CD4+ and CD8+ T cells [92]. It has been reported that HCV p7 protein has an immune evasion function, which may suppress antiviral IFN function by counteracting IFN-inducible protein 6-16 (IFI6-16) [132]. Based on these findings, it is assumed that HCV and its proteins play a crucial role in inhibiting or suppressing the host innate immune response (Figure 2) by different known and unknown mechanisms; therefore, a clear understanding of immune inhibition or evasion is critical for devising new therapeutic and preventive strategies to control HCV infection.

4. Potential of TLR Agonists as Immunomodulators

A large number of viruses have been shown to trigger innate immunity via TLRs, which have been found important in the outcome of many viral infections [133,134,135], suggesting manipulating the TLR response could serve as a therapeutic avenue against viral infections. Although there is a great therapeutic success against HCV infection with newly approved drugs, direct-acting antivirals (DAAs) [136], cirrhosis and HCC have been reported in patients following viral clearance [137]. Moreover, current HCV treatment approaches are not effective in preventing recurrent infections. Notably, high treatment cost has restricted its use in economically weak countries. Therefore, there is an urgent need to develop an alternative therapeutic approach as well as an effective, safe, and affordable vaccine against HCV. There is a growing interest in the use of TLR agonists as vaccine adjuvants [138], which are capable of stimulating innate and adaptive immune responses, thereby improving vaccine efficacy. Recently, TLR agonists have received much attention as immunomodulators with the ability to induce the production of IFN, proinflammatory cytokines, and chemokines and have been found to be promising against many viral infections, including hepatitis B virus and human immunodeficiency virus type 1 (HIV-1) [35,139,140,141]. Manipulating TLR response has also been found to be promising against SARS-CoV-2 infection. In a ferret model, it has been shown that the injection of TLR2/6 agonist INNA-051 significantly reduced SARS-CoV-2 viral RNA levels in the nose and throat [142]. Additionally, it has been proposed that TLR agonists, including imiquimod, an immune stimulator of TLR7, could serve as an effective therapeutic approach in the early stages of COVID-19 [143].
Chronic HCV infection induces weak cellular immune responses against viral antigens, and viral clearance after acute hepatitis or therapy requires strong and multispecific antiviral CD4+ and CD8+ T-cell responses [144,145]. TLR agonists may play an important role in enhancing immunotherapeutic effects [33,34,35,138,146]. The use of TLR agonists as immunomodulators to enhance the immune response in chronic HCV infection could be of great interest for the control of HCV infection. Isatoribine, an agonist of TLR7, showed dose-dependent changes in immunologic biomarkers and antiviral effects against HCV infection [147]. The antiviral activity of the synthetic TLR7 agonist was shown to be associated with the stimulation of antiviral genes, such as IRF7, but not with the activation of the IFN-responsive STAT-1 transcription factor [75]. A previous study demonstrated that oral administration of resiquimod, a TLR 7/8 agonist, transiently reduced viral levels but was associated with adverse effects similar to IFN-α [148]. Another study showed that TLR3/4/7/8/9 agonists could induce anti-HCV activity in PBMC supernatants, correlating with IFN-α and the IFN-induced antiviral biomarker 2′,5′-oligoadenylate synthase induction. However, TLR4 and TLR8 agonists induce the proinflammatory cytokines IL-1β and TNF-α at concentrations similar to those inducing antiviral activity, raising concerns regarding adverse side effects [149]. In a randomized clinical trial, oral administration of ANA773, a prodrug of the TLR7 agonist, resulted in an IFN-dependent response leading to a significant decrease in serum HCV RNA levels, with mild to moderate adverse events [150].
It has been reported that TLR7/9 agonists may enhance the inhibition of infectivity and IFN-α production by pDCs, suggesting pDCs could serve as a drug target against HCV infection [151]. It also suggests the possibility of using TLR7/9 agonists in HCV vaccine development. An earlier study showed that although a hepatitis C viral-like particle vaccine adjuvanted with TLR2 agonists, R4Pam2Cys and E8Pam2Cys, induced higher antibody titers in mice, it did not induce stronger NAb responses compared to vaccines without adjuvants [152], suggesting the usefulness of TLR agonists as vaccine adjuvants for HCV vaccines. Using appropriate adjuvants in HCV vaccine candidates, the induction of a strong T- and B-cell immune response may be enhanced [153]. In an in vitro study with an HCV-infected hepatoma cell line, Huh7.5, Dominguez-Molina et al. showed that TLR agonists can enhance antiviral pDCs function primarily through IFN-α production against HCV infection [151].

5. Conclusions

From the currently available data, it is understood that there is a differential expression of TLRs in HCV infection. Moreover, TLR3 and TLR7 may play a protective role against HCV infection. However, the exact role of the TLR response to HCV infection requires further extensive study, which also requires a suitable small animal model. HCV has developed multiple strategies to inhibit the innate immune response toward establishing a chronic infection, which also requires additional investigation for developing new therapeutic approaches. As there are mixed effects, an extensive study is still required to select the best-suited TLR agonist for use as a vaccine adjuvant for HCV vaccine candidates. Overall, a clear understanding of TLR interactions in HCV infection is critical for providing new therapeutic and preventive approaches to fight the disease, including TLR agonist-adjuvanted HCV vaccines.

Author Contributions

Conceptualization, M.E.H.K., M.K. and K.T.-K.; writing—original draft preparation, M.E.H.K. and K.T.-K.; writing—review and editing, M.E.H.K., M.K. and K.T.-K.; All authors have read and agreed to the published version of the manuscript.

Funding

Grant from the Tokyo Metropolitan Government for drug development targeting liver cirrhosis.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mohd Hanafiah, K.; Groeger, J.; Flaxman, A.D.; Wiersma, S.T. Global epidemiology of hepatitis C virus infection: New estimates of age-specific antibody to HCV seroprevalence. Hepatology 2013, 57, 1333–1342. [Google Scholar] [CrossRef] [PubMed]
  2. Lavanchy, D. The global burden of hepatitis C. Liver Int. 2009, 29 (Suppl. S1), 74–81. [Google Scholar] [CrossRef] [PubMed]
  3. Saito, I.; Miyamura, T.; Ohbayashi, A.; Harada, H.; Katayama, T.; Kikuchi, S.; Watanabe, Y.; Koi, S.; Onji, M.; Ohta, Y.; et al. Hepatitis C virus infection is associated with the development of hepatocellular carcinoma. Proc. Natl. Acad. Sci. USA 1990, 87, 6547–6549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. World Health Organization (WHO). Hepatitis C. Updated on 27 July 2021. 2021. Available online: https://www.who.int/news-room/fact-sheets/detail/hepatitis-c (accessed on 25 March 2022).
  5. Choo, Q.L.; Kuo, G.; Weiner, A.J.; Overby, L.R.; Bradley, D.W.; Houghton, M. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science 1989, 244, 359–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Moradpour, D.; Penin, F.; Rice, C.M. Replication of hepatitis C virus. Nat. Rev. Microbiol. 2007, 5, 453–463. [Google Scholar] [CrossRef]
  7. Smith, D.B.; Bukh, J.; Kuiken, C.; Muerhoff, A.S.; Rice, C.M.; Stapleton, J.T.; Simmonds, P. Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: Updated criteria and genotype assignment web resource. Hepatology 2014, 59, 318–327. [Google Scholar] [CrossRef] [Green Version]
  8. Takeda, K.; Akira, S. Toll-like receptors in innate immunity. Int. Immunol. 2005, 17, 1–14. [Google Scholar] [CrossRef]
  9. Takeuchi, O.; Akira, S. Innate immunity to virus infection. Immunol. Rev. 2009, 227, 75–86. [Google Scholar] [CrossRef]
  10. Zuniga, E.I.; Macal, M.; Lewis, G.M.; Harker, J.A. Innate and Adaptive Immune Regulation During Chronic Viral Infections. Annu. Rev. Virol. 2015, 2, 573–597. [Google Scholar] [CrossRef] [Green Version]
  11. Medzhitov, R.; Janeway, C., Jr. Innate immunity. N. Engl. J. Med. 2000, 343, 338–344. [Google Scholar] [CrossRef]
  12. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Amarante-Mendes, G.P.; Adjemian, S.; Branco, L.M.; Zanetti, L.C.; Weinlich, R.; Bortoluci, K.R. Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front. Immunol. 2018, 9, 2379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kawai, T.; Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011, 34, 637–650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Akira, S.; Uematsu, S.; Takeuchi, O. Pathogen recognition and innate immunity. Cell 2006, 124, 783–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Akira, S.; Takeda, K.; Kaisho, T. Toll-like receptors: Critical proteins linking innate and acquired immunity. Nat. Immunol. 2001, 2, 675–680. [Google Scholar] [CrossRef]
  17. Fitzgerald, K.A.; Kagan, J.C. Toll-like Receptors and the Control of Immunity. Cell 2020, 180, 1044–1066. [Google Scholar] [CrossRef]
  18. Narayanan, K.B.; Park, H.H. Toll/interleukin-1 receptor (TIR) domain-mediated cellular signaling pathways. Apoptosis 2015, 20, 196–209. [Google Scholar] [CrossRef]
  19. O’Neill, L.A.; Bowie, A.G. The family of five: TIR-domain-containing adaptors in Toll-like receptor signalling. Nat. Rev. Immunol. 2007, 7, 353–364. [Google Scholar] [CrossRef]
  20. Lee, M.S.; Kim, Y.J. Signaling pathways downstream of pattern-recognition receptors and their cross talk. Annu. Rev. Biochem. 2007, 76, 447–480. [Google Scholar] [CrossRef]
  21. Kaisho, T.; Akira, S. Toll-like receptor function and signaling. J. Allergy Clin. Immunol. 2006, 117, 979–987. [Google Scholar] [CrossRef]
  22. Akira, S.; Takeda, K. Toll-like receptor signalling. Nat. Rev. Immunol. 2004, 4, 499–511. [Google Scholar] [CrossRef] [PubMed]
  23. Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. O’Neill, L.A.; Golenbock, D.; Bowie, A.G. The history of Toll-like receptors-redefining innate immunity. Nat. Rev. Immunol. 2013, 13, 453–460. [Google Scholar] [CrossRef] [PubMed]
  25. Chaturvedi, A.; Pierce, S.K. How location governs toll-like receptor signaling. Traffic 2009, 10, 621–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Prinz, M.; Heikenwalder, M.; Schwarz, P.; Takeda, K.; Akira, S.; Aguzzi, A. Prion pathogenesis in the absence of Toll-like receptor signalling. EMBO Rep. 2003, 4, 195–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Kawai, T.; Akira, S. Toll-like receptor and RIG-I-like receptor signaling. Ann. N. Y. Acad. Sci. 2008, 1143, 1–20. [Google Scholar] [CrossRef] [PubMed]
  28. Alexopoulou, L.; Holt, A.C.; Medzhitov, R.; Flavell, R.A. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 2001, 413, 732–738. [Google Scholar] [CrossRef]
  29. Diebold, S.S.; 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, 1529–1531. [Google Scholar] [CrossRef]
  30. Heim, M.H.; Thimme, R. Innate and adaptive immune responses in HCV infections. J. Hepatol. 2014, 61, S14–S25. [Google Scholar] [CrossRef] [Green Version]
  31. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820. [Google Scholar] [CrossRef] [Green Version]
  32. Iwasaki, A.; Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 2004, 5, 987–995. [Google Scholar] [CrossRef] [PubMed]
  33. Mullins, S.R.; Vasilakos, J.P.; Deschler, K.; Grigsby, I.; Gillis, P.; John, J.; Elder, M.J.; Swales, J.; Timosenko, E.; Cooper, Z.; et al. Intratumoral immunotherapy with TLR7/8 agonist MEDI9197 modulates the tumor microenvironment leading to enhanced activity when combined with other immunotherapies. J. Immunother. Cancer 2019, 7, 244. [Google Scholar] [CrossRef] [PubMed]
  34. Surendran, N.; Simmons, A.; Pichichero, M.E. TLR agonist combinations that stimulate Th type I polarizing responses from human neonates. Innate Immun. 2018, 24, 240–251. [Google Scholar] [CrossRef] [PubMed]
  35. Kayesh, M.E.H.; Kohara, M.; Tsukiyama-Kohara, K. Toll-Like Receptor Response to Hepatitis B Virus Infection and Potential of TLR Agonists as Immunomodulators for Treating Chronic Hepatitis B: An Overview. Int. J. Mol. Sci. 2021, 22, 10462. [Google Scholar] [CrossRef]
  36. Owen, A.M.; Fults, J.B.; Patil, N.K.; Hernandez, A.; Bohannon, J.K. TLR Agonists as Mediators of Trained Immunity: Mechanistic Insight and Immunotherapeutic Potential to Combat Infection. Front. Immunol. 2020, 11, 622614. [Google Scholar] [CrossRef]
  37. Yokota, S.; Okabayashi, T.; Fujii, N. The battle between virus and host: Modulation of Toll-like receptor signaling pathways by virus infection. Mediat. Inflamm. 2010, 2010, 184328. [Google Scholar] [CrossRef] [Green Version]
  38. Huang, B.; Zhao, J.; Unkeless, J.C.; Feng, Z.H.; Xiong, H. TLR signaling by tumor and immune cells: A double-edged sword. Oncogene 2008, 27, 218–224. [Google Scholar] [CrossRef] [Green Version]
  39. Ebermeyer, T.; Cognasse, F.; Berthelot, P.; Mismetti, P.; Garraud, O.; Hamzeh-Cognasse, H. Platelet Innate Immune Receptors and TLRs: A Double-Edged Sword. Int. J. Mol. Sci. 2021, 22, 7894. [Google Scholar] [CrossRef]
  40. Zheng, M.; Karki, R.; Williams, E.P.; Yang, D.; Fitzpatrick, E.; Vogel, P.; Jonsson, C.B.; Kanneganti, T.D. TLR2 senses the SARS-CoV-2 envelope protein to produce inflammatory cytokines. Nat. Immunol. 2021, 22, 829–838. [Google Scholar] [CrossRef]
  41. Metz, P.; Reuter, A.; Bender, S.; Bartenschlager, R. Interferon-stimulated genes and their role in controlling hepatitis C virus. J. Hepatol. 2013, 59, 1331–1341. [Google Scholar] [CrossRef] [Green Version]
  42. Horner, S.M.; Gale, M., Jr. Regulation of hepatic innate immunity by hepatitis C virus. Nat. Med. 2013, 19, 879–888. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Yang, D.R.; Zhu, H.Z. Hepatitis C virus and antiviral innate immunity: Who wins at tug-of-war? World J. Gastroenterol. 2015, 21, 3786–3800. [Google Scholar] [CrossRef] [PubMed]
  44. Kawai, T.; Akira, S. The role of pattern-recognition receptors in innate immunity: Update on Toll-like receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
  45. Wang, N.; Liang, Y.; Devaraj, S.; Wang, J.; Lemon, S.M.; Li, K. Toll-like receptor 3 mediates establishment of an antiviral state against hepatitis C virus in hepatoma cells. J. Virol. 2009, 83, 9824–9834. [Google Scholar] [CrossRef] [Green Version]
  46. Machida, K.; Cheng, K.T.; Sung, V.M.; Levine, A.M.; Foung, S.; Lai, M.M. Hepatitis C virus induces toll-like receptor 4 expression, leading to enhanced production of beta interferon and interleukin-6. J. Virol. 2006, 80, 866–874. [Google Scholar] [CrossRef] [Green Version]
  47. Feldmann, G.; Nischalke, H.D.; Nattermann, J.; Banas, B.; Berg, T.; Teschendorf, C.; Schmiegel, W.; Duhrsen, U.; Halangk, J.; Iwan, A.; et al. Induction of interleukin-6 by hepatitis C virus core protein in hepatitis C-associated mixed cryoglobulinemia and B-cell non-Hodgkin’s lymphoma. Clin. Cancer Res. 2006, 12, 4491–4498. [Google Scholar] [CrossRef] [Green Version]
  48. Shehata, M.A.; Abou El-Enein, A.; El-Sharnouby, G.A. Significance of toll-like receptors 2 and 4 mRNA expression in chronic hepatitis C virus infection. Egypt J. Immunol. 2006, 13, 141–152. [Google Scholar]
  49. Wang, J.P.; Zhang, Y.; Wei, X.; Li, J.; Nan, X.P.; Yu, H.T.; Li, Y.; Wang, P.Z.; Bai, X.F. Circulating Toll-like receptor (TLR) 2, TLR4, and regulatory T cells in patients with chronic hepatitis C. APMIS 2010, 118, 261–270. [Google Scholar] [CrossRef]
  50. Chung, H.; Watanabe, T.; Kudo, M.; Chiba, T. Correlation between hyporesponsiveness to Toll-like receptor ligands and liver dysfunction in patients with chronic hepatitis C virus infection. J. Viral Hepat. 2011, 18, e561–e567. [Google Scholar] [CrossRef]
  51. He, Q.; Graham, C.S.; Durante Mangoni, E.; Koziel, M.J. Differential expression of toll-like receptor mRNA in treatment non-responders and sustained virologic responders at baseline in patients with chronic hepatitis C. Liver Int. 2006, 26, 1100–1110. [Google Scholar] [CrossRef]
  52. Sato, K.; Ishikawa, T.; Okumura, A.; Yamauchi, T.; Sato, S.; Ayada, M.; Matsumoto, E.; Hotta, N.; Oohashi, T.; Fukuzawa, Y.; et al. Expression of Toll-like receptors in chronic hepatitis C virus infection. J. Gastroenterol. Hepatol. 2007, 22, 1627–1632. [Google Scholar] [CrossRef] [PubMed]
  53. Dolganiuc, A.; Kodys, K.; Kopasz, A.; Marshall, C.; Do, T.; Romics, L., Jr.; Mandrekar, P.; Zapp, M.; Szabo, G. Hepatitis C virus core and nonstructural protein 3 proteins induce pro- and anti-inflammatory cytokines and inhibit dendritic cell differentiation. J. Immunol. 2003, 170, 5615–5624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Dolganiuc, A.; Oak, S.; Kodys, K.; Golenbock, D.T.; Finberg, R.W.; Kurt-Jones, E.; Szabo, G. Hepatitis C core and nonstructural 3 proteins trigger toll-like receptor 2-mediated pathways and inflammatory activation. Gastroenterology 2004, 127, 1513–1524. [Google Scholar] [CrossRef]
  55. Brown, R.A.; Gralewski, J.H.; Eid, A.J.; Knoll, B.M.; Finberg, R.W.; Razonable, R.R. R753Q single-nucleotide polymorphism impairs toll-like receptor 2 recognition of hepatitis C virus core and nonstructural 3 proteins. Transplantation 2010, 89, 811–815. [Google Scholar] [CrossRef] [PubMed]
  56. Chang, S.; Dolganiuc, A.; Szabo, G. Toll-like receptors 1 and 6 are involved in TLR2-mediated macrophage activation by hepatitis C virus core and NS3 proteins. J. Leukoc. Biol. 2007, 82, 479–487. [Google Scholar] [CrossRef]
  57. Hoffmann, M.; Zeisel, M.B.; Jilg, N.; Paranhos-Baccala, G.; Stoll-Keller, F.; Wakita, T.; Hafkemeyer, P.; Blum, H.E.; Barth, H.; Henneke, P.; et al. Toll-like receptor 2 senses hepatitis C virus core protein but not infectious viral particles. J. Innate Immun. 2009, 1, 446–454. [Google Scholar] [CrossRef] [Green Version]
  58. Chung, H.; Watanabe, T.; Kudo, M.; Chiba, T. Hepatitis C virus core protein induces homotolerance and cross-tolerance to Toll-like receptor ligands by activation of Toll-like receptor 2. J. Infect. Dis. 2010, 202, 853–861. [Google Scholar] [CrossRef] [Green Version]
  59. Dolganiuc, A.; Chang, S.; Kodys, K.; Mandrekar, P.; Bakis, G.; Cormier, M.; Szabo, G. Hepatitis C virus (HCV) core protein-induced, monocyte-mediated mechanisms of reduced IFN-alpha and plasmacytoid dendritic cell loss in chronic HCV infection. J. Immunol. 2006, 177, 6758–6768. [Google Scholar] [CrossRef] [Green Version]
  60. Rajalakshmy, A.R.; Malathi, J.; Madhavan, H.N. HCV core and NS3 proteins mediate toll like receptor induced innate immune response in corneal epithelium. Exp. Eye Res. 2014, 128, 117–128. [Google Scholar] [CrossRef]
  61. Riordan, S.M.; Skinner, N.A.; Kurtovic, J.; Locarnini, S.; McIver, C.J.; Williams, R.; Visvanathan, K. Toll-like receptor expression in chronic hepatitis C: Correlation with pro-inflammatory cytokine levels and liver injury. Inflamm. Res. 2006, 55, 279–285. [Google Scholar] [CrossRef]
  62. Chen Yi Mei, S.L.; Burchell, J.; Skinner, N.; Millen, R.; Matthews, G.; Hellard, M.; Dore, G.J.; Desmond, P.V.; Sundararajan, V.; Thompson, A.J.; et al. Toll-like Receptor Expression and Signaling in Peripheral Blood Mononuclear Cells Correlate with Clinical Outcomes in Acute Hepatitis C Virus Infection. J. Infect. Dis. 2016, 214, 739–747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Moriyama, M.; Kato, N.; Otsuka, M.; Shao, R.X.; Taniguchi, H.; Kawabe, T.; Omata, M. Interferon-beta is activated by hepatitis C virus NS5B and inhibited by NS4A, NS4B, and NS5A. Hepatol. Int. 2007, 1, 302–310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Broering, R.; Wu, J.; Meng, Z.; Hilgard, P.; Lu, M.; Trippler, M.; Szczeponek, A.; Gerken, G.; Schlaak, J.F. Toll-like receptor-stimulated non-parenchymal liver cells can regulate hepatitis C virus replication. J. Hepatol. 2008, 48, 914–922. [Google Scholar] [CrossRef] [PubMed]
  65. Al-Qahtani, A.A.; Al-Anazi, M.R.; Al-Zoghaibi, F.; Abdo, A.A.; Sanai, F.M.; Khan, M.Q.; Albenmousa, A.; Al-Ashgar, H.I.; Al-Ahdal, M.N. The association of toll-like receptor 4 polymorphism with hepatitis C virus infection in Saudi Arabian patients. Biomed. Res. Int. 2014, 2014, 357062. [Google Scholar] [CrossRef]
  66. Zhang, Y.L.; Guo, Y.J.; Bin, L.; Sun, S.H. Hepatitis C virus single-stranded RNA induces innate immunity via Toll-like receptor 7. J. Hepatol. 2009, 51, 29–38. [Google Scholar] [CrossRef]
  67. Li, K.; Li, N.L.; Wei, D.; Pfeffer, S.R.; Fan, M.; Pfeffer, L.M. Activation of chemokine and inflammatory cytokine response in hepatitis C virus-infected hepatocytes depends on Toll-like receptor 3 sensing of hepatitis C virus double-stranded RNA intermediates. Hepatology 2012, 55, 666–675. [Google Scholar] [CrossRef]
  68. Motavaf, M.; Noorbakhsh, F.; Alavian, S.M.; Sharifi, Z. Distinct Toll-like Receptor 3 and 7 Expression in Peripheral Blood Mononuclear Cells from Patients with Chronic Hepatitis C Infection. Hepat. Mon. 2014, 14, e16421. [Google Scholar] [CrossRef] [Green Version]
  69. Mohammed, K.I.; Adel, L.A.; Ali-Eldin, F.A.; Eladawy, S. Expression of Toll like receptors 3 & 7 in peripheral blood from patients with chronic hepatitis C virus infection and their correlation with interferon-alpha. Egypt J. Immunol. 2013, 20, 13–22. [Google Scholar]
  70. Suef, R.A.; Mohamed, E.E.M.; Mansour, M.T.M.; Weigand, K.; Farag, M.M.S. Differential expression of viral pathogen-associated molecular pattern receptors mRNA in Egyptian chronic hepatitis C virus patients. Egypt. J. Med. Hum. Genet. 2021, 22, 13. [Google Scholar] [CrossRef]
  71. Firdaus, R.; Biswas, A.; Saha, K.; Mukherjee, A.; Pal, F.; Chaudhuri, S.; Chandra, A.; Konar, A.; Sadhukhan, P.C. Modulation of TLR 3, 7 and 8 expressions in HCV genotype 3 infected individuals: Potential correlations of pathogenesis and spontaneous clearance. Biomed. Res. Int. 2014, 2014, 491064. [Google Scholar] [CrossRef]
  72. Howell, J.; Sawhney, R.; Skinner, N.; Gow, P.; Angus, P.; Ratnam, D.; Visvanathan, K. Toll-like receptor 3 and 7/8 function is impaired in hepatitis C rapid fibrosis progression post-liver transplantation. Am. J. Transplant. 2013, 13, 943–953. [Google Scholar] [CrossRef] [PubMed]
  73. Takahashi, K.; Asabe, S.; Wieland, S.; Garaigorta, U.; Gastaminza, P.; Isogawa, M.; Chisari, F.V. Plasmacytoid dendritic cells sense hepatitis C virus-infected cells, produce interferon, and inhibit infection. Proc. Natl. Acad. Sci. USA 2010, 107, 7431–7436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Dreux, M.; Garaigorta, U.; Boyd, B.; Decembre, E.; Chung, J.; Whitten-Bauer, C.; Wieland, S.; Chisari, F.V. Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell Host Microbe 2012, 12, 558–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Lee, J.; Wu, C.C.; Lee, K.J.; Chuang, T.H.; Katakura, K.; Liu, Y.T.; Chan, M.; Tawatao, R.; Chung, M.; Shen, C.; et al. Activation of anti-hepatitis C virus responses via Toll-like receptor 7. Proc. Natl. Acad. Sci. USA 2006, 103, 1828–1833. [Google Scholar] [CrossRef] [Green Version]
  76. Wang, C.H.; Eng, H.L.; Lin, K.H.; Chang, C.H.; Hsieh, C.A.; Lin, Y.L.; Lin, T.M. TLR7 and TLR8 gene variations and susceptibility to hepatitis C virus infection. PLoS ONE 2011, 6, e26235. [Google Scholar] [CrossRef] [Green Version]
  77. Lee, J.; Tian, Y.; Chan, S.T.; Kim, J.Y.; Cho, C.; Ou, J.H. TNF-alpha Induced by Hepatitis C Virus via TLR7 and TLR8 in Hepatocytes Supports Interferon Signaling via an Autocrine Mechanism. PLoS Pathog. 2015, 11, e1004937. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, Y.; El-Far, M.; Dupuy, F.P.; Abdel-Hakeem, M.S.; He, Z.; Procopio, F.A.; Shi, Y.; Haddad, E.K.; Ancuta, P.; Sekaly, R.P.; et al. HCV RNA Activates APCs via TLR7/TLR8 While Virus Selectively Stimulates Macrophages Without Inducing Antiviral Responses. Sci. Rep. 2016, 6, 29447. [Google Scholar] [CrossRef] [Green Version]
  79. Fakhir, F.Z.; Lkhider, M.; Badre, W.; Alaoui, R.; Meurs, E.F.; Pineau, P.; Ezzikouri, S.; Benjelloun, S. Genetic variations in toll-like receptors 7 and 8 modulate natural hepatitis C outcomes and liver disease progression. Liver Int. 2018, 38, 432–442. [Google Scholar] [CrossRef]
  80. Wang, C.H.; Eng, H.L.; Lin, K.H.; Liu, H.C.; Chang, C.H.; Lin, T.M. Functional polymorphisms of TLR8 are associated with hepatitis C virus infection. Immunology 2014, 141, 540–548. [Google Scholar] [CrossRef]
  81. El-Bendary, M.; Neamatallah, M.; Elalfy, H.; Besheer, T.; Elkholi, A.; El-Diasty, M.; Elsareef, M.; Zahran, M.; El-Aarag, B.; Gomaa, A.; et al. The association of single nucleotide polymorphisms of Toll-like receptor 3, Toll-like receptor 7 and Toll-like receptor 8 genes with the susceptibility to HCV infection. Br. J. Biomed. Sci. 2018, 75, 175–181. [Google Scholar] [CrossRef]
  82. Fischer, J.; Weber, A.N.R.; Bohm, S.; Dickhofer, S.; El Maadidi, S.; Deichsel, D.; Knop, V.; Klinker, H.; Moller, B.; Rasenack, J.; et al. Sex-specific effects of TLR9 promoter variants on spontaneous clearance of HCV infection. Gut 2017, 66, 1829–1837. [Google Scholar] [CrossRef] [PubMed]
  83. Abdelwahab, S.F.; Hamdy, S.; Osman, A.M.; Zakaria, Z.A.; Galal, I.; Sobhy, M.; Hashem, M.; Allam, W.R.; Abdel-Samiee, M.; Rewisha, E.; et al. Association of the polymorphism of the Toll-like receptor (TLR)-3 and TLR-9 genes with hepatitis C virus-specific cell-mediated immunity outcomes among Egyptian health-care workers. Clin. Exp. Immunol. 2021, 203, 3–12. [Google Scholar] [CrossRef] [PubMed]
  84. Kayesh, M.E.H.; Sanada, T.; Kohara, M.; Tsukiyama-Kohara, K. Tree Shrew as an Emerging Small Animal Model for Human Viral Infection: A Recent Overview. Viruses 2021, 13, 1641. [Google Scholar] [CrossRef] [PubMed]
  85. Fan, Y.; Huang, Z.Y.; Cao, C.C.; Chen, C.S.; Chen, Y.X.; Fan, D.D.; He, J.; Hou, H.L.; Hu, L.; Hu, X.T.; et al. Genome of the Chinese tree shrew. Nat. Commun. 2013, 4, 1426. [Google Scholar] [CrossRef] [Green Version]
  86. Sanada, T.; Tsukiyama-Kohara, K.; Shin, I.T.; Yamamoto, N.; Kayesh, M.E.H.; Yamane, D.; Takano, J.I.; Shiogama, Y.; Yasutomi, Y.; Ikeo, K.; et al. Construction of complete Tupaia belangeri transcriptome database by whole-genome and comprehensive RNA sequencing. Sci. Rep. 2019, 9, 12372. [Google Scholar] [CrossRef]
  87. Kayesh, M.E.H.; Ezzikouri, S.; Sanada, T.; Chi, H.; Hayashi, Y.; Rebbani, K.; Kitab, B.; Matsuu, A.; Miyoshi, N.; Hishima, T.; et al. Oxidative Stress and Immune Responses during Hepatitis C Virus Infection in Tupaia belangeri. Sci. Rep. 2017, 7, 9848. [Google Scholar] [CrossRef]
  88. Sarasin-Filipowicz, M.; Oakeley, E.J.; Duong, F.H.; Christen, V.; Terracciano, L.; Filipowicz, W.; Heim, M.H. Interferon signaling and treatment outcome in chronic hepatitis C. Proc. Natl. Acad. Sci. USA 2008, 105, 7034–7039. [Google Scholar] [CrossRef] [Green Version]
  89. Asselah, T.; Bieche, I.; Narguet, S.; Sabbagh, A.; Laurendeau, I.; Ripault, M.P.; Boyer, N.; Martinot-Peignoux, M.; Valla, D.; Vidaud, M.; et al. Liver gene expression signature to predict response to pegylated interferon plus ribavirin combination therapy in patients with chronic hepatitis C. Gut 2008, 57, 516–524. [Google Scholar] [CrossRef] [Green Version]
  90. Thimme, R.; Binder, M.; Bartenschlager, R. Failure of innate and adaptive immune responses in controlling hepatitis C virus infection. FEMS Microbiol. Rev. 2012, 36, 663–683. [Google Scholar] [CrossRef] [Green Version]
  91. Ding, Q.; Cao, X.; Lu, J.; Huang, B.; Liu, Y.J.; Kato, N.; Shu, H.B.; Zhong, J. Hepatitis C virus NS4B blocks the interaction of STING and TBK1 to evade host innate immunity. J. Hepatol. 2013, 59, 52–58. [Google Scholar] [CrossRef]
  92. Samrat, S.K.; Vedi, S.; Singh, S.; Li, W.; Kumar, R.; Agrawal, B. Immunization with Recombinant Adenoviral Vectors Expressing HCV Core or F Proteins Leads to T Cells with Reduced Effector Molecules Granzyme B and IFN-gamma: A Potential New Strategy for Immune Evasion in HCV Infection. Viral Immunol. 2015, 28, 309–324. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Li, K.; Foy, E.; Ferreon, J.C.; Nakamura, M.; Ferreon, A.C.; Ikeda, M.; Ray, S.C.; Gale, M., Jr.; Lemon, S.M. Immune evasion by hepatitis C virus NS3/4A protease-mediated cleavage of the Toll-like receptor 3 adaptor protein TRIF. Proc. Natl. Acad. Sci. USA 2005, 102, 2992–2997. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Xu, Z.; Choi, J.; Lu, W.; Ou, J.H. Hepatitis C virus f protein is a short-lived protein associated with the endoplasmic reticulum. J. Virol. 2003, 77, 1578–1583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Oshiumi, H.; Miyashita, M.; Matsumoto, M.; Seya, T. A distinct role of Riplet-mediated K63-Linked polyubiquitination of the RIG-I repressor domain in human antiviral innate immune responses. PLoS Pathog. 2013, 9, e1003533. [Google Scholar] [CrossRef] [Green Version]
  96. Morikawa, K.; Lange, C.M.; Gouttenoire, J.; Meylan, E.; Brass, V.; Penin, F.; Moradpour, D. Nonstructural protein 3-4A: The Swiss army knife of hepatitis C virus. J. Viral Hepat. 2011, 18, 305–315. [Google Scholar] [CrossRef]
  97. Kang, X.; Chen, X.; He, Y.; Guo, D.; Guo, L.; Zhong, J.; Shu, H.B. DDB1 is a cellular substrate of NS3/4A protease and required for hepatitis C virus replication. Virology 2013, 435, 385–394. [Google Scholar] [CrossRef] [Green Version]
  98. Rehermann, B. Hepatitis C virus versus innate and adaptive immune responses: A tale of coevolution and coexistence. J. Clin. Investig. 2009, 119, 1745–1754. [Google Scholar] [CrossRef] [Green Version]
  99. Foy, E.; Li, K.; Sumpter, R., Jr.; Loo, Y.M.; Johnson, C.L.; Wang, C.; Fish, P.M.; Yoneyama, M.; Fujita, T.; Lemon, S.M.; et al. Control of antiviral defenses through hepatitis C virus disruption of retinoic acid-inducible gene-I signaling. Proc. Natl. Acad. Sci. USA 2005, 102, 2986–2991. [Google Scholar] [CrossRef] [Green Version]
  100. Sklan, E.H.; Charuworn, P.; Pang, P.S.; Glenn, J.S. Mechanisms of HCV survival in the host. Nat. Rev. Gastroenterol. Hepatol. 2009, 6, 217–227. [Google Scholar] [CrossRef]
  101. Loo, Y.M.; Owen, D.M.; Li, K.; Erickson, A.K.; Johnson, C.L.; Fish, P.M.; Carney, D.S.; Wang, T.; Ishida, H.; Yoneyama, M.; et al. Viral and therapeutic control of IFN-beta promoter stimulator 1 during hepatitis C virus infection. Proc. Natl. Acad. Sci. USA 2006, 103, 6001–6006. [Google Scholar] [CrossRef] [Green Version]
  102. Meylan, E.; Curran, J.; Hofmann, K.; Moradpour, D.; Binder, M.; Bartenschlager, R.; Tschopp, J. Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature 2005, 437, 1167–1172. [Google Scholar] [CrossRef] [PubMed]
  103. Bellecave, P.; Sarasin-Filipowicz, M.; Donze, O.; Kennel, A.; Gouttenoire, J.; Meylan, E.; Terracciano, L.; Tschopp, J.; Sarrazin, C.; Berg, T.; et al. Cleavage of mitochondrial antiviral signaling protein in the liver of patients with chronic hepatitis C correlates with a reduced activation of the endogenous interferon system. Hepatology 2010, 51, 1127–1136. [Google Scholar] [CrossRef] [PubMed]
  104. Blindenbacher, A.; Duong, F.H.; Hunziker, L.; Stutvoet, S.T.; Wang, X.; Terracciano, L.; Moradpour, D.; Blum, H.E.; Alonzi, T.; Tripodi, M.; et al. Expression of hepatitis c virus proteins inhibits interferon alpha signaling in the liver of transgenic mice. Gastroenterology 2003, 124, 1465–1475. [Google Scholar] [CrossRef]
  105. Bode, J.G.; Ludwig, S.; Ehrhardt, C.; Albrecht, U.; Erhardt, A.; Schaper, F.; Heinrich, P.C.; Haussinger, D. IFN-alpha antagonistic activity of HCV core protein involves induction of suppressor of cytokine signaling-3. FASEB J. 2003, 17, 488–490. [Google Scholar] [CrossRef] [PubMed]
  106. Basu, A.; Meyer, K.; Ray, R.B.; Ray, R. Hepatitis C virus core protein modulates the interferon-induced transacting factors of Jak/Stat signaling pathway but does not affect the activation of downstream IRF-1 or 561 gene. Virology 2001, 288, 379–390. [Google Scholar] [CrossRef] [Green Version]
  107. Stone, A.E.; Mitchell, A.; Brownell, J.; Miklin, D.J.; Golden-Mason, L.; Polyak, S.J.; Gale, M.J., Jr.; Rosen, H.R. Hepatitis C virus core protein inhibits interferon production by a human plasmacytoid dendritic cell line and dysregulates interferon regulatory factor-7 and signal transducer and activator of transcription (STAT) 1 protein expression. PLoS ONE 2014, 9, e95627. [Google Scholar] [CrossRef] [Green Version]
  108. Lin, W.; Choe, W.H.; Hiasa, Y.; Kamegaya, Y.; Blackard, J.T.; Schmidt, E.V.; Chung, R.T. Hepatitis C virus expression suppresses interferon signaling by degrading STAT1. Gastroenterology 2005, 128, 1034–1041. [Google Scholar] [CrossRef]
  109. Lin, W.; Kim, S.S.; Yeung, E.; Kamegaya, Y.; Blackard, J.T.; Kim, K.A.; Holtzman, M.J.; Chung, R.T. Hepatitis C virus core protein blocks interferon signaling by interaction with the STAT1 SH2 domain. J. Virol. 2006, 80, 9226–9235. [Google Scholar] [CrossRef] [Green Version]
  110. Heim, M.H.; Moradpour, D.; Blum, H.E. Expression of hepatitis C virus proteins inhibits signal transduction through the Jak-STAT pathway. J. Virol. 1999, 73, 8469–8475. [Google Scholar] [CrossRef] [Green Version]
  111. Duong, F.H.; Filipowicz, M.; Tripodi, M.; La Monica, N.; Heim, M.H. Hepatitis C virus inhibits interferon signaling through up-regulation of protein phosphatase 2A. Gastroenterology 2004, 126, 263–277. [Google Scholar] [CrossRef]
  112. Luquin, E.; Larrea, E.; Civeira, M.P.; Prieto, J.; Aldabe, R. HCV structural proteins interfere with interferon-alpha Jak/STAT signalling pathway. Antivir. Res. 2007, 76, 194–197. [Google Scholar] [CrossRef] [PubMed]
  113. Hosui, A.; Ohkawa, K.; Ishida, H.; Sato, A.; Nakanishi, F.; Ueda, K.; Takehara, T.; Kasahara, A.; Sasaki, Y.; Hori, M.; et al. Hepatitis C virus core protein differently regulates the JAK-STAT signaling pathway under interleukin-6 and interferon-gamma stimuli. J. Biol. Chem. 2003, 278, 28562–28571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Yoshida, H.; Kato, N.; Shiratori, Y.; Otsuka, M.; Maeda, S.; Kato, J.; Omata, M. Hepatitis C virus core protein activates nuclear factor kappa B-dependent signaling through tumor necrosis factor receptor-associated factor. J. Biol. Chem. 2001, 276, 16399–16405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. You, L.R.; Chen, C.M.; Lee, Y.H. Hepatitis C virus core protein enhances NF-kappaB signal pathway triggering by lymphotoxin-beta receptor ligand and tumor necrosis factor alpha. J. Virol. 1999, 73, 1672–1681. [Google Scholar] [CrossRef] [Green Version]
  116. Marusawa, H.; Hijikata, M.; Chiba, T.; Shimotohno, K. Hepatitis C virus core protein inhibits Fas-and tumor necrosis factor alpha-mediated apoptosis via NF-kappaB activation. J. Virol. 1999, 73, 4713–4720. [Google Scholar] [CrossRef] [Green Version]
  117. Kato, N.; Yoshida, H.; Ono-Nita, S.K.; Kato, J.; Goto, T.; Otsuka, M.; Lan, K.; Matsushima, K.; Shiratori, Y.; Omata, M. Activation of intracellular signaling by hepatitis B and C viruses: C-viral core is the most potent signal inducer. Hepatology 2000, 32, 405–412. [Google Scholar] [CrossRef]
  118. Nguyen, H.; Sankaran, S.; Dandekar, S. Hepatitis C virus core protein induces expression of genes regulating immune evasion and anti-apoptosis in hepatocytes. Virology 2006, 354, 58–68. [Google Scholar] [CrossRef] [Green Version]
  119. Larrea, E.; Aldabe, R.; Molano, E.; Fernandez-Rodriguez, C.M.; Ametzazurra, A.; Civeira, M.P.; Prieto, J. Altered expression and activation of signal transducers and activators of transcription (STATs) in hepatitis C virus infection: In vivo and in vitro studies. Gut 2006, 55, 1188–1196. [Google Scholar] [CrossRef]
  120. Agaugue, S.; Perrin-Cocon, L.; Andre, P.; Lotteau, V. Hepatitis C lipo-Viro-particle from chronically infected patients interferes with TLR4 signaling in dendritic cell. PLoS ONE 2007, 2, e330. [Google Scholar] [CrossRef] [Green Version]
  121. Nitta, S.; Sakamoto, N.; Nakagawa, M.; Kakinuma, S.; Mishima, K.; Kusano-Kitazume, A.; Kiyohashi, K.; Murakawa, M.; Nishimura-Sakurai, Y.; Azuma, S.; et al. Hepatitis C virus NS4B protein targets STING and abrogates RIG-I-mediated type I interferon-dependent innate immunity. Hepatology 2013, 57, 46–58. [Google Scholar] [CrossRef]
  122. Yi, G.; Wen, Y.; Shu, C.; Han, Q.; Konan, K.V.; Li, P.; Kao, C.C. Hepatitis C Virus NS4B Can Suppress STING Accumulation To Evade Innate Immune Responses. J. Virol. 2016, 90, 254–265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Abe, T.; Kaname, Y.; Hamamoto, I.; Tsuda, Y.; Wen, X.; Taguwa, S.; Moriishi, K.; Takeuchi, O.; Kawai, T.; Kanto, T.; et al. Hepatitis C virus nonstructural protein 5A modulates the toll-like receptor-MyD88-dependent signaling pathway in macrophage cell lines. J. Virol. 2007, 81, 8953–8966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Polyak, S.J.; Khabar, K.S.; Paschal, D.M.; Ezelle, H.J.; Duverlie, G.; Barber, G.N.; Levy, D.E.; Mukaida, N.; Gretch, D.R. Hepatitis C virus nonstructural 5A protein induces interleukin-8, leading to partial inhibition of the interferon-induced antiviral response. J. Virol. 2001, 75, 6095–6106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Horner, S.M.; Gale, M., Jr. Intracellular innate immune cascades and interferon defenses that control hepatitis C virus. J. Interferon Cytokine Res. 2009, 29, 489–498. [Google Scholar] [CrossRef] [Green Version]
  126. Gale, M.J., Jr.; Korth, M.J.; Tang, N.M.; Tan, S.L.; Hopkins, D.A.; Dever, T.E.; Polyak, S.J.; Gretch, D.R.; Katze, M.G. Evidence that hepatitis C virus resistance to interferon is mediated through repression of the PKR protein kinase by the nonstructural 5A protein. Virology 1997, 230, 217–227. [Google Scholar] [CrossRef] [Green Version]
  127. Taylor, D.R.; Shi, S.T.; Romano, P.R.; Barber, G.N.; Lai, M.M. Inhibition of the interferon-inducible protein kinase PKR by HCV E2 protein. Science 1999, 285, 107–110. [Google Scholar] [CrossRef]
  128. Noguchi, T.; Satoh, S.; Noshi, T.; Hatada, E.; Fukuda, R.; Kawai, A.; Ikeda, S.; Hijikata, M.; Shimotohno, K. Effects of mutation in hepatitis C virus nonstructural protein 5A on interferon resistance mediated by inhibition of PKR kinase activity in mammalian cells. Microbiol. Immunol. 2001, 45, 829–840. [Google Scholar] [CrossRef]
  129. Hiet, M.S.; Bauhofer, O.; Zayas, M.; Roth, H.; Tanaka, Y.; Schirmacher, P.; Willemsen, J.; Grunvogel, O.; Bender, S.; Binder, M.; et al. Control of temporal activation of hepatitis C virus-induced interferon response by domain 2 of nonstructural protein 5A. J. Hepatol. 2015, 63, 829–837. [Google Scholar] [CrossRef] [Green Version]
  130. Cevik, R.E.; Cesarec, M.; Da Silva Filipe, A.; Licastro, D.; McLauchlan, J.; Marcello, A. Hepatitis C Virus NS5A Targets Nucleosome Assembly Protein NAP1L1 to Control the Innate Cellular Response. J. Virol. 2017, 91, e00880-17. [Google Scholar] [CrossRef] [Green Version]
  131. Yonkers, N.L.; Rodriguez, B.; Milkovich, K.A.; Asaad, R.; Lederman, M.M.; Heeger, P.S.; Anthony, D.D. TLR ligand-dependent activation of naive CD4 T cells by plasmacytoid dendritic cells is impaired in hepatitis C virus infection. J. Immunol. 2007, 178, 4436–4444. [Google Scholar] [CrossRef] [Green Version]
  132. Qi, H.; Chu, V.; Wu, N.C.; Chen, Z.; Truong, S.; Brar, G.; Su, S.Y.; Du, Y.; Arumugaswami, V.; Olson, C.A.; et al. Systematic identification of anti-interferon function on hepatitis C virus genome reveals p7 as an immune evasion protein. Proc. Natl. Acad. Sci. USA 2017, 114, 2018–2023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Carty, M.; Bowie, A.G. Recent insights into the role of Toll-like receptors in viral infection. Clin. Exp. Immunol. 2010, 161, 397–406. [Google Scholar] [CrossRef] [PubMed]
  134. Patel, M.C.; Shirey, K.A.; Pletneva, L.M.; Boukhvalova, M.S.; Garzino-Demo, A.; Vogel, S.N.; Blanco, J.C. Novel drugs targeting Toll-like receptors for antiviral therapy. Future Virol. 2014, 9, 811–829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Mifsud, E.J.; Tan, A.C.; Jackson, D.C. TLR Agonists as Modulators of the Innate Immune Response and Their Potential as Agents Against Infectious Disease. Front. Immunol. 2014, 5, 79. [Google Scholar] [CrossRef]
  136. Li, G.; De Clercq, E. Current therapy for chronic hepatitis C: The role of direct-acting antivirals. Antivir. Res. 2017, 142, 83–122. [Google Scholar] [CrossRef]
  137. Chinchilla-Lopez, P.; Qi, X.; Yoshida, E.M.; Mendez-Sanchez, N. The Direct-Acting Antivirals for Hepatitis C Virus and the Risk for Hepatocellular Carcinoma. Ann. Hepatol. 2017, 16, 328–330. [Google Scholar] [CrossRef]
  138. Kayesh, M.E.H.; Kohara, M.; Tsukiyama-Kohara, K. An Overview of Recent Insights into the Response of TLR to SARS-CoV-2 Infection and the Potential of TLR Agonists as SARS-CoV-2 Vaccine Adjuvants. Viruses 2021, 13, 2302. [Google Scholar] [CrossRef]
  139. Lucifora, J.; Bonnin, M.; Aillot, L.; Fusil, F.; Maadadi, S.; Dimier, L.; Michelet, M.; Floriot, O.; Ollivier, A.; Rivoire, M.; et al. Direct antiviral properties of TLR ligands against HBV replication in immune-competent hepatocytes. Sci. Rep. 2018, 8, 5390. [Google Scholar] [CrossRef] [Green Version]
  140. Niu, C.; Li, L.; Daffis, S.; Lucifora, J.; Bonnin, M.; Maadadi, S.; Salas, E.; Chu, R.; Ramos, H.; Livingston, C.M.; et al. Toll-like receptor 7 agonist GS-9620 induces prolonged inhibition of HBV via a type I interferon-dependent mechanism. J. Hepatol. 2018, 68, 922–931. [Google Scholar] [CrossRef]
  141. Martinsen, J.T.; Gunst, J.D.; Hojen, J.F.; Tolstrup, M.; Sogaard, O.S. The Use of Toll-Like Receptor Agonists in HIV-1 Cure Strategies. Front. Immunol. 2020, 11, 1112. [Google Scholar] [CrossRef]
  142. Proud, P.C.; Tsitoura, D.; Watson, R.J.; Chua, B.Y.; Aram, M.J.; Bewley, K.R.; Cavell, B.E.; Cobb, R.; Dowall, S.; Fotheringham, S.A.; et al. Prophylactic intranasal administration of a TLR2/6 agonist reduces upper respiratory tract viral shedding in a SARS-CoV-2 challenge ferret model. EBioMedicine 2021, 63, 103153. [Google Scholar] [CrossRef] [PubMed]
  143. Angelopoulou, A.; Alexandris, N.; Konstantinou, E.; Mesiakaris, K.; Zanidis, C.; Farsalinos, K.; Poulas, K. Imiquimod—A toll like receptor 7 agonist—Is an ideal option for management of COVID 19. Environ. Res. 2020, 188, 109858. [Google Scholar] [CrossRef] [PubMed]
  144. Missale, G.; Bertoni, R.; Lamonaca, V.; Valli, A.; Massari, M.; Mori, C.; Rumi, M.G.; Houghton, M.; Fiaccadori, F.; Ferrari, C. Different clinical behaviors of acute hepatitis C virus infection are associated with different vigor of the anti-viral cell-mediated immune response. J. Clin. Investig. 1996, 98, 706–714. [Google Scholar] [CrossRef] [PubMed]
  145. Thimme, R.; Oldach, D.; Chang, K.M.; Steiger, C.; Ray, S.C.; Chisari, F.V. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J. Exp. Med. 2001, 194, 1395–1406. [Google Scholar] [CrossRef]
  146. Hennessy, E.J.; Parker, A.E.; O’Neill, L.A. Targeting Toll-like receptors: Emerging therapeutics? Nat. Rev. Drug Discov. 2010, 9, 293–307. [Google Scholar] [CrossRef]
  147. Horsmans, Y.; Berg, T.; Desager, J.P.; Mueller, T.; Schott, E.; Fletcher, S.P.; Steffy, K.R.; Bauman, L.A.; Kerr, B.M.; Averett, D.R. Isatoribine, an agonist of TLR7, reduces plasma virus concentration in chronic hepatitis C infection. Hepatology 2005, 42, 724–731. [Google Scholar] [CrossRef]
  148. Pockros, P.J.; Guyader, D.; Patton, H.; Tong, M.J.; Wright, T.; McHutchison, J.G.; Meng, T.C. Oral resiquimod in chronic HCV infection: Safety and efficacy in 2 placebo-controlled, double-blind phase IIa studies. J. Hepatol. 2007, 47, 174–182. [Google Scholar] [CrossRef]
  149. Thomas, A.; Laxton, C.; Rodman, J.; Myangar, N.; Horscroft, N.; Parkinson, T. Investigating Toll-like receptor agonists for potential to treat hepatitis C virus infection. Antimicrob. Agents Chemother. 2007, 51, 2969–2978. [Google Scholar] [CrossRef] [Green Version]
  150. Bergmann, J.F.; de Bruijne, J.; Hotho, D.M.; de Knegt, R.J.; Boonstra, A.; Weegink, C.J.; van Vliet, A.A.; van de Wetering, J.; Fletcher, S.P.; Bauman, L.A.; 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, 443–453. [Google Scholar] [CrossRef]
  151. Dominguez-Molina, B.; Machmach, K.; Perales, C.; Tarancon-Diez, L.; Gallego, I.; Sheldon, J.L.; Leal, M.; Domingo, E.; Ruiz-Mateos, E. Toll-Like Receptor 7 (TLR-7) and TLR-9 Agonists Improve Hepatitis C Virus Replication and Infectivity Inhibition by Plasmacytoid Dendritic Cells. J. Virol. 2018, 92, e01219-18. [Google Scholar] [CrossRef] [Green Version]
  152. Christiansen, D.; Earnest-Silveira, L.; Chua, B.; Boo, I.; Drummer, H.E.; Grubor-Bauk, B.; Gowans, E.J.; Jackson, D.C.; Torresi, J. Antibody Responses to a Quadrivalent Hepatitis C Viral-Like Particle Vaccine Adjuvanted with Toll-Like Receptor 2 Agonists. Viral Immunol. 2018, 31, 338–343. [Google Scholar] [CrossRef] [PubMed]
  153. Sepulveda-Crespo, D.; Resino, S.; Martinez, I. Innate Immune Response against Hepatitis C Virus: Targets for Vaccine Adjuvants. Vaccines 2020, 8, 313. [Google Scholar] [CrossRef] [PubMed]
Figure 1. TLR response to HCV infection. Red arrows indicate the induction/activation of components of TLR signaling by HCV or its proteins; black lines indicate the inhibition of the host innate immune response or inhibition of HCV replication, as appropriate.
Figure 1. TLR response to HCV infection. Red arrows indicate the induction/activation of components of TLR signaling by HCV or its proteins; black lines indicate the inhibition of the host innate immune response or inhibition of HCV replication, as appropriate.
Ijms 23 05475 g001
Figure 2. An overview of the mechanism of the host innate immune response inhibition by HCV and its proteins.
Figure 2. An overview of the mechanism of the host innate immune response inhibition by HCV and its proteins.
Ijms 23 05475 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kayesh, M.E.H.; Kohara, M.; Tsukiyama-Kohara, K. Toll-like Receptor Response to Hepatitis C Virus Infection: A Recent Overview. Int. J. Mol. Sci. 2022, 23, 5475. https://doi.org/10.3390/ijms23105475

AMA Style

Kayesh MEH, Kohara M, Tsukiyama-Kohara K. Toll-like Receptor Response to Hepatitis C Virus Infection: A Recent Overview. International Journal of Molecular Sciences. 2022; 23(10):5475. https://doi.org/10.3390/ijms23105475

Chicago/Turabian Style

Kayesh, Mohammad Enamul Hoque, Michinori Kohara, and Kyoko Tsukiyama-Kohara. 2022. "Toll-like Receptor Response to Hepatitis C Virus Infection: A Recent Overview" International Journal of Molecular Sciences 23, no. 10: 5475. https://doi.org/10.3390/ijms23105475

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

Article Metrics

Back to TopTop