Next Article in Journal
Vaccination History, Body Mass Index, Age, and Baseline Gene Expression Predict Influenza Vaccination Outcomes
Previous Article in Journal
Analysis of Exosomal MicroRNA Dynamics in Response to Rhinovirus Challenge in a Longitudinal Case-Control Study of Asthma
Previous Article in Special Issue
Rational Engineering of a Sub-Picomolar HIV-1 Blocker
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CXCL10 Chemokine: A Critical Player in RNA and DNA Viral Infections

by
Noha Mousaad Elemam
1,2,
Iman Mamdouh Talaat
1,2,3 and
Azzam A. Maghazachi
1,2,*
1
Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
2
Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
3
Pathology Department, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
*
Author to whom correspondence should be addressed.
Viruses 2022, 14(11), 2445; https://doi.org/10.3390/v14112445
Submission received: 29 September 2022 / Revised: 31 October 2022 / Accepted: 1 November 2022 / Published: 3 November 2022
(This article belongs to the Special Issue Chemokines and Chemokine Receptors in Viral Infection 2024)

Abstract

:
Chemokines constitute a group of small, secreted proteins that regulate leukocyte migration and contribute to their activation. Chemokines are crucial inflammatory mediators that play a key role in managing viral infections, during which the profile of chemokine expression helps shape the immune response and regulate viral clearance, improving clinical outcome. In particular, the chemokine ligand CXCL10 and its receptor CXCR3 were explored in a plethora of RNA and DNA viral infections. In this review, we highlight the expression profile and role of the CXCL10/CXCR3 axis in the host defense against a variety of RNA and DNA viral infections. We also discuss the interactions among viruses and host cells that trigger CXCL10 expression, as well as the signaling cascades induced in CXCR3 positive cells.

1. Role of Chemokines during Viral Infections

Chemokines are small, secreted molecules that enhance cell interaction via G-protein-coupled receptors [1]. The chemokine-encoding genes in humans are located on chromosomes 4 and 17 [2]. In addition to being active as monomers, during an immune response, chemokines form homodimers and heterodimers, which result in a robust immunological response [3,4]. A general classification of chemokines is based on the structure of the two cysteine residues closest to the N terminus, which can be juxtaposed (CC) or separated by a single amino acid (CXC) or by three amino acids (CX3C). Chemokines that lack the first cysteine residue are known as XC chemokines [5,6]. Others classified chemokines into two primary categories according to their functions: those engaged in homeostasis and those involved in inflammation. Chemokine biological effects include many processes, such as angiogenesis regulation, tumor development, and invasion [7].
Further, chemokine receptors can be categorized into conventional (cCKRs) and atypical [8]. Conventional receptors can heterodimerize with other cCKRs, membrane proteins, and atypical chemokine receptors in addition to existing as homodimers and oligomers [9,10]. It should be noted that chemokines may also operate as natural antagonists rather than inducing activating signals when they attach to their appropriate receptors [11]. On the other hand, atypical chemokine receptors (ACKRs) are structurally linked to cCKRs when they bind to chemokines with a high affinity. However, because the intracellular signaling motifs are absent or altered, they do not activate the same signaling pathways as cCKRs [12,13]. In other words, the perturbation of ACKRs does not activate the G-protein signaling pathway but instead internalizes the chemokines to be degraded through the recruitment of β-arrestins [8]. Functionally, atypical chemokine receptors are known to regulate the localization, distribution, and abundance of chemokines and are considered proficient scavengers, thus modulating chemokine concentrations and bioavailability [14]. Moreover, ACKRs prevent unnecessary desensitization upon excessive exposure to chemokine ligands [15,16,17,18].
Several physiological processes, including cell proliferation and angiogenesis, as well as pathological conditions, including inflammation, neoplasia, autoimmunity, and infections (viral, bacterial, or parasitic), are controlled by binding chemokines to their cognate receptors [19]. During viral infections, chemokines are released as a result of a series of events [20], connecting several components [21] that boost the innate and adaptive immune systems which promote cell migration towards the sites of infection [22]. Nevertheless, viruses attempt to elude the immune system by molecular mimicry [23].

2. Structure and Function of CXCL10

Three chemokines are known to be interferon (IFN)-induced angiostatic CXC chemokines. These include monokine induced by interferon (MIG/CXCL9), interferon gamma-induced protein 10 (IP-10/CXCL10), and interferon-inducible T-cell chemoattractant (I-TAC/CXCL11). The chemokine CXCL10 binds CXCR3, which is present in T helper lymphocytes, natural killer (NK) cells, dendritic cells (DCs), macrophages, and B cells [24,25,26,27]. CXCR3 has two different isoforms, CXCR3-A and CXCR3-B, that possess several functions. For example, upon the binding of CXCL10 to isoform A, chemotaxis and proliferation are induced [28,29], while binding to the B isoform results in inhibiting cell migration and proliferation [29,30]. The atypical chemokine receptor ACKR2 is known to bind most inflammatory CC chemokine members [31] and is activated by the agonist CXCL10. Chevigné et al. described a novel aspect of CXCL10 regulation [32]. In an inflammatory status, CXCL10 could be secreted by various immune cells such as neutrophils, eosinophils, and monocytes, as well as epithelial cells, endothelial cells, and fibroblasts upon IFN-γ triggering [33,34,35]. Consequently, activated Th1 lymphocytes, monocytes, and NK cells would migrate towards the sites of inflammation in order to trigger the release of pro-inflammatory cytokines and chemokines. CXCL10 does not have the structural domain ELR “Glu-Leu-Arg” tripeptide motif, which is present in certain CXC chemokines and which might have anti-angiogenic along with anti-tumor properties [30,36,37,38,39]. The gene coding for CXCL10 is localized on chromosome 4 at band q21, and its translation produces a 12 kDa protein that has 2 internal disulfide cross-bridges [34]. Upon cleavage, a 10 kDa protein is generated and secreted with 4 conserved cysteine residues in the N-terminal [34]. The structure of CXCL10 exhibits a typical chemokine fold consisting of a three-stranded β sheet overlaid by an α helix with a number of the receptor binding residues located in the associated loops stabilized by the disulfide bonds [40]. The transcription of CXCL10 is regulated by various external stimuli, including cytokines such as IFN-γ and bacterial components such as lipopolysaccharides (LPS).
CXCL10 possesses an upstream regulatory sequence that contains several critical regulatory elements for nuclear factor-κB (NF-κB) and interferon-stimulated response element (ISRE), as well as sites for the binding of proteins such as heat shock (HS) factors [41]. Cytokines such as tumor necrosis factor (TNF-α) and IFN-γ induce the expression of CXCL10 through NF-κB and ISRE present in the promoter of CXCL10 [42,43,44]. This could lead to the activation of protein kinase C and increase the intracellular Ca2+ mobilization [45,46]. Since it is an inflammatory chemokine, CXCL10 was associated with multiple disorders, including infectious diseases, autoimmune diseases, and cancer [18,47,48,49,50]. Furthermore, it exacerbates inflammation, causing tissue damage [47]. Moreover, CXCL10 was expressed within tissues following viral infections, suggesting an essential role for this chemokine in host defense by contributing to the lymphocyte activation, migration, and infiltration of specific T cell subsets within the sites of infection [51].

3. Role of CXCL10 in RNA Viral Infections

CXCL10 could be protective or pathogenic in viral infections depending on the host’s immune status and the type of viral infection [52]. CXCL10 has been implicated in multiple RNA viral infections, including rhinovirus, respiratory syncytial virus (RSV), coxsackie virus, hepatitis C virus (HCV), Ebola, dengue, and equine infectious anemia virus [33,53,54,55,56,57,58,59,60]. It is also involved in various models of respiratory viral infection, including Sendai, influenza, corona, and adenoviruses, as well as neurogenic viral reactions where the expression and role of CXCL10 have been reported [61,62,63,64,65,66]. This highlights the significance of CXCL10 expression in multiple viral infections that could aid in viral clearance or contribute to disease pathogenesis.
The absence of CXCL10 using antibodies or knockout mouse models resulted in increased viral titers and reduced T cell infiltration within the brains of mice infected with a murine hepatitis virus (MHV), leading to increased mortality [67,68]. CXCL10 also affects the migration of CD8+ T cells in the liver of adenovirus infected mice [69,70]. These studies indicate that CXCL10 is crucial in host defense and the development of a protective T cell response during viral infections. Furthermore, CXCL10 has been shown to induce the migration of T cells and NK cells following viral infections [71,72,73,74], where it attracts NK cells into the CNS post MHV infection [51]. Moreover, CXCL10 limits the spread of infection, contributing to early host defense and associated with a direct anti-viral effect against several RNA viruses [75,76]. For instance, CXCL10 blocked the entry and replication of the dengue virus by inhibiting its binding to the cell surface receptors [77]. Additionally, NK cell activation and migration caused an increase in IFN-γ production, which stimulates CXCL10 expression in cardiomyocytes and other resident myocardial cells in coxsackievirus B3 viral infection. This led to an inhibition of viral replication at an early stage in order to prevent cardiomyocyte damage and improve cardiac function [78].
Previous studies reported a synergy among double-stranded RNA (dsRNA) and IFN-γ-induced signaling in regulating CXCL10 production. As mentioned earlier, pro-inflammatory mediators and IFN-γ increased CXCL10 expression by a number of cell types, including keratinocytes, macrophages, endothelial cells, smooth muscle cells, and epithelial cell fibroblasts and astrocytes [79,80,81,82,83,84,85,86,87,88]. Majumder et al. reported that IFN-γ and TNF-α could promote CXCL10 production in human fibrosarcoma cell lines via binding the p48 complexes and the signal transducer and activator of transcription 1 (STAT-1) to ISRE site in CXCL10 promoter [89]. In another study, Oslund et al. determined that IFN-γ and influenza virus were able to synergistically induce CXCL10 in human airway epithelial cells through dsRNA-induced signaling both in vitro and in vivo [90]. Furthermore, the 735 bp proximal region of the CXCL10 promoter was the cause of this induction as it contains ISRE and NF-κB transcription factor binding sites. Following influenza virus infection, CD8+ T cell and NK cell migration towards the airway epithelial milieu was significantly affected by the ability of the airway epithelial cells to dramatically upregulate CXCL10 [90]. Other dsRNA-mediated pathways have also been identified in the upregulation of CXCL10, including retinoic acid-inducible gene I (RIG-I) and protein kinase R (PKR), which further augment the anti-viral signal by the activation of type I IFN production [91,92,93].
CXCL10 was found to have a protective function during certain RNA viral infections such as the severe acute respiratory syndrome coronavirus (SARS-CoV) and Epstein-Barr virus (EBV) [94,95,96]. On the other hand, CXCL10 was found to promote the infection caused by the human immunodeficiency virus (HIV) by stimulating the virus replication in macrophages and lymphocytes [97]. In short, the expression of CXCL10 was differentially associated with clinical symptoms in multiple infections. For example, a high expression of CXCL10 occurred before the development of clinical symptoms during HIV infection and murine retroviruses [98], and CXCL10 levels were positively correlated with organ damage and pathogen burden in HCV and HIV infections [99,100]. Moreover, persistent high levels of CXCL10 were linked to the failure of highly active antiretroviral therapy (HAART) in HIV-infected patients [101], as anti-retroviral treatment decreased the plasma levels of CXCL10, indicating its vital role in disease pathogenesis [97]. Additionally, the elevation of CXCL10 levels was reported for at least two weeks after disease onset, while corticosteroid therapy caused a reduction in the plasma CXCL10 levels, during SARS infection [102,103]. The chemokine receptor CXCR3 was highly expressed on viral-specific stem-like CD8+ T cells. CXCL10 regulates the persistence and heterogeneity of CD8+ T cells in the spleens of mice infected with lymphocytic choriomeningitis virus (LCMV). Moreover, functional CD8+ T cell responses were found to be greater in Cxcl10−/− mice and were associated with a lower viral count [104].
Among various chemokines, CXCR3 has been studied in viral hepatitis as it plays a significant role in the recruitment of T cells into the peripheral inflammatory sites [105,106]. The RNA virus, hepatitis A virus (HAV), is known to cause severe liver injury accompanied by the release of inflammatory mediators. HAV-infected hepatocytes produced multiple chemokines, including CXCL10. Moreover, CXCL10 levels were significantly increased in the serum of patients infected with HAV [107]. The CXCL10 production was reduced subsequent to inhibiting the signaling molecules associated with RIG-I-like receptor (RLR), such as mitochondrial antiviral signaling protein and interferon regulatory factor 3 (IRF3), in HAV-infected cells [107]. Similar to HAV, HCV-infected cells produced CXCL10 independent of type I or III IFNs [108], regulated by the expression of interferon regulatory factor 3 (IRF3) and NF-κB [109]. Additionally, CXCR3 is involved in the recruitment of effector CD8+ T cells and CD4+ T helper cells into HCV-infected livers [110,111,112], and the expression of CXCR3 in the liver or the peripheral blood is correlated with the severity of hepatic inflammation in HCV infection [113,114,115,116]. Similarly, the Kernow-p6 hepatitis E virus (HEV) strain was reported to stimulate the release of CXCL10 in enterocytes and infected hepatocytes [117,118,119,120]. Further, patients with HEV genotype three infections (clade efg) suffer from severe clinical representations, accompanied by higher CXCL10 serum levels and liver necro-inflammatory activity [121].
In chronic HCV infection, CXCL10 contributed to the development of necroinflammation and liver fibrosis. Moreover, the intrahepatic production of IFN-γ promoted CXCL10 expression by sinusoidal endothelium and hepatocytes, thereby triggering the recruitment of CXCR3+ T cells. CXCL10 plasma levels were significantly higher in HCV-infected patients with advanced fibrosis [122]. Additionally, CXCL10 levels were associated with the extrahepatic manifestations observed with this viral infection. For instance, CXCL10 levels were much higher in patients with HCV-associated cryoglobulinemia compared with those patients with autoimmune thyroiditis [122]. Additionally, high CXCL10 levels present in cases of HCV-associated cryoglobulinemia were associated with the presence of another extrahepatic manifestation, i.e., active vasculitis. Therefore, targeting CXCL10 and its receptor CXCR3 was suggested for treating HCV patients. Examples of such an approach include antibodies blocking the interaction between CXCR3 and its ligands, which reduced chronic liver inflammation and damage via the impairment of the infiltration of the nonspecific T cells [123]. This therapeutic strategy could be evaluated primarily for the non-responders to current anti-HCV therapy. This and other studies highlighted the potential use of CXCL10 as a prognostic marker of HCV clearance and successful therapy [122].
CXCL10 is also known to play a critical role in the host defense against infection caused by the John Howard Mueller (JHM) strain of mouse hepatitis virus (JHMV). Early expression of CXCL10 was observed to promote the attraction of CXCR3+ T cells into the central nervous system, which subsequently aids in limiting viral replication [124]. On the other hand, chronic CXCL10 expression contributed to the neuroinflammation and demyelination associated with JHMV infection. This was attributed to the attraction of CD4+ T cells that amplify neuroinflammation through the IFN induction of chemokine release. Therefore, the CXCL10/CXCR3 signaling pathway may influence glial biology and may repair virally induced neurologic diseases [124].
CXCL10 is a key player in respiratory syndromes. Its level was higher in the plasma and bronchial alveolar lavage fluid (BALF), which correlated with the disease severity [125,126,127]. Elevated CXCL10 levels were reported in patients with SARS or Middle East respiratory syndrome (MERS) as early as 2–3 days post-infection [61,103,128,129,130,131]. This increase was found to reduce the proliferation of myeloid progenitor cells, triggering lymphopenia as observed in coronavirus disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection [131]. Additionally, CXCL10 was reported to trigger T cells apoptosis and lymphopenia seen in SARS-CoV-, MERS-, and SARS-CoV-2-infected patients, leading to T lymphocyte malfunction and worsening clinical outcome [132]. Previous studies using ex vivo human lung tissue explants demonstrated that the inoculation of SARS-CoV and SARS-CoV-2 induced the expression of various chemokines and cytokines, particularly CXCL10 [133].
Similar to other respiratory diseases, high CXCL10 levels in COVID-19 patients showed a strong correlation with disease severity [134,135]. As mentioned earlier, CXCL10 levels were increased in SARS-CoV-2 infected patients, especially those with acute respiratory distress syndrome (ARDS) [136,137]. Consequently, the use of CXCL10 as an independent predictor for COVID-19 progression was suggested since it was correlated with ARDS in critically ill patients and severe cases [132,138,139]. This was further supported in COVID-19 patients requiring intensive care unit (ICU) admission, who exhibited higher levels of CXCL10 than patients with mild infection [138,140,141,142,143]. Furthermore, higher mortality was reported in COVID-19 patients with a dramatic increase in CXCL10 levels compared with severe and mild COVID-19 patients [136,144,145,146]. In this regard, symptomatic COVID-19 patients showed higher levels of CXCL10 compared with convalescent cases [134]. Interestingly, CXCL10 levels were reported to be significantly reduced upon improving clinical outcomes in hospitalized COVID-19 patients [137]. Consequently, several studies reported CXCL10 as an excellent predictive biomarker of patient outcomes in COVID-19 [137,141]. CXCL10 was also identified to be a potential biomarker to predict left ventricular dysfunction in multisystemic inflammatory syndrome (MIS-C) patients, occurring post-exposure to COVID-19 infection [147].
Intriguingly, across all three coronaviruses (SARS-CoV, MERS-CoV, and SARS-CoV-2), CXCL10 was identified as a crucial contributor to pulmonary pathogenesis. CXCL10 was associated with lung injury and the activation of the toll-like receptor 4 (TLR4) signaling pathway [148,149]. Although not an ELR chemokine, CXCL10 induced neutrophil infiltration into the lungs, triggering further production of this chemokine, coupled with the release of oxidative burst by neutrophils. Eventually, this worsens the inflammatory lung state, causing progression to ARDS [150,151]. Therefore, antibodies against CXCL10 could be a potential and promising therapeutic approach in ARDS, as previously described in the influenza A virus subtype (H1N1) mouse model [150,152]. Furthermore, corticosteroids, the anti-inflammatory agents used in COVID-19 infection, were found to reduce the levels of multiple chemokines, including CXCL10, along with stimulating the NF-κB and activator protein-1 (AP-1) signaling pathways [153].
IL-6 is one of the cytokines released during COVID-19-mediated cytokine storm [154,155]. During this storm, high IL-6 levels act on lung endothelial cells, causing an increase in their permeability for serum proteins and the infiltration of inflammatory cells. This leads to an uncontrolled excessive immune response, such as in severe COVID-19 cases [154,155]. In addition, CXCL10 was recognized as a fundamental chemokine acting as a chemoattractant for monocytes, macrophages, and dendritic cells, as well as NK and T cells during COVID-19 infection. Upon the entry of SARS-CoV-2 into the lung epithelium, it triggers cytokine and chemokine production, which include IL-6 and CXCL10. One of the causes for the increased production of CXCL10 is IL-6, which also leads to an increase in the infiltration of CXCR3+ macrophages, the main producers of IL-6. These observations highlight the vicious cycle between CXCL10, IL-6, and macrophages in the lungs of COVID-19 patients, which could regulate the onset, maintenance, and progression of cytokine storms during COVID-19 infection [156].
As in respiratory coronavirus infections, CXCL10 showed a significant increase during measles virus (MeV) infection, which may distinguish MeV from other diseases such as rubella virus (RuV), parvovirus B19 (B19V), human herpesvirus 6 (HHV6), EBV, and human cytomegalovirus (HCMV). Another common factor was the high level of CXCL10 in hospitalized MeV patients compared with non-hospitalized ones, and higher CXCL10 level was observed in cases of primary infection compared with re-infected cases [157,158]. This demonstrates persistent inflammation and robust viral replication that might aggravate the clinical course of primary infection [159,160]. Interestingly, CXCL10 levels were associated with the serological stages of MeV infection. For instance, CXCL10 production was increased early after infection and reached its peak with the appearance of MeV-specific IgM antibodies, which later declined [157]. Another interesting finding during MeV infection is the association between mortality due to MeV infection and high CXCL10 levels in children. This further supports the link between CXCL10 and the clinical severity of MeV infection. CXCL10 and CCL5 were highly expressed in vitro and in vivo during RSV infection. However, CXCL10 was found to have a protective role by decreasing viral load and disease pathogenesis [161]. This was mediated via the promotion of leukocyte recruitment and the trafficking of dendritic cells (DCs). Furthermore, CXCL10 is stimulated by type I IFN in CXCR3+ DCs, triggering myeloid DC maturation [161].
CXCL10 expression was higher in Zika virus (ZIKV)-infected individuals. This expression could be mediated through the induction of IFN-γ signaling pathway by the ZIKV NS5 protein. Previously, CXCL10 was discovered to be a biomarker of several bacterial infections [162]. Similarly, CXCL10 was identified as a biomarker of acute ZIKV infection and a predictor of disease severity [163]. A summary of the role of CXCL10 in RNA viral infections is shown in Figure 1.

4. Effects of CXCL10 during DNA Viral Infections

As illustrated in Figure 2, CXCL10 is implicated in various DNA viral infections. For instance, CXCL10 was found to be a critical player in the innate defense against vaccinia virus infection by recruiting and activating NK cells [75]. In varicella-zoster virus (VZV) infection, upregulated CXCL10 expression was demonstrated along with the infiltration of CXCR3+ cells towards the infected dorsal root ganglia. Consequently, CXCL10 could be a distinct chemokine that attracts inflammatory cells, specifically CXCR3+ cells causing ganglionitis [164].
According to several studies, CXCL10 is implicated in hepatitis B virus (HBV) infection, including a study where the polymorphism G-201A in the promoter of the CXCL10 gene predicted susceptibility to chronic HBV infection [52]. During chronic infection, serum and intrahepatic levels of CXCL10 were elevated and correlated with HBV DNA and alanine aminotransferase (ALT) enzyme levels, coupled with progressive liver disease [165,166]. Other studies confirmed that CXCR3 ligands such as CXCL9, CXCL10, and CXCL11 are highly increased along with ALT in acute hepatitis patients, resulting from the release of IFN-α and IFN-γ by plasmacytoid DCs and NK cells, respectively, in response to HBV infection [167,168]. Furthermore, these three chemokines were decreased with reduced HBV DNA during acute infection [169]. In contrast, high CXCL10/IP-10 and CXCL9/Mig levels during HBV infection were correlated with severe liver disease. Subsequently, their neutralization was linked to the preservation of antiviral effects including less tissue damage [170]. Zhou et al. demonstrated that the upregulation of CXCL10 was dose-dependent through the activation of NF-κB, thereby enhancing the infiltration of peripheral leukocytes into the liver of HBV infection [171]. Consequently, blocking CXCL10 could reduce the recruitment of lymphocytes and improve the disease severity [71,172]. In HIV and HBV co-infected patients, mRNA levels for CXCL10 and CXCR3 were correlated with liver fibrosis. It has been suggested that the HIV infection of hepatocytes in the presence of HBV could promote inflammation and flux of CXCL10 production, thus promoting the recruitment of activated CXCR3+ cells towards the liver that could cause liver fibrosis [173].
CXCL10 is crucial in host defense against multiple neurotropic viruses, including herpes simplex virus-1 (HSV-1) [174]. It was previously reported that lack of either CXCL10 or its receptor CXCR3 could impair the mobilization of functional CD8+ effector memory (TEM) and CD8+ residual memory (TRM) cells within latently infected trigeminal ganglia following HSV-1 reactivation [175]. In contrast, increasing the levels of CXCL10 in latently HSV-1 infected CXCL10-deficient mice significantly restored the number of local antiviral CD8+ TEM and CD8+ TRM cells associated with protection against recurrent ocular herpes. These findings demonstrate that CXCL10/CXCR3 axis is crucial for CD8+ T cell immunity, which protects against recurrent herpesvirus infection [175]. The chemokine receptor CXCR3 is highly expressed on activated CD4+, CD8+ T cells, and NK cells at the sites of HSV-1 replication [176,177,178,179,180]. Additionally, during HSV-1 infection, CXCL10 was one of the first and most abundantly expressed chemokines, indicating that it may be essential for the coordinated immune response against HSV-1. Further, HSV-1 viral burden was elevated in CXCR3-deficient (CXCR3−/−) and CXCL10-deficient (CXCL10−/−) animals compared with wild types [174,177]. In this regard, CXCL10−/− mice exhibited impaired NK and HSV-1-specific CD8+ T cell activation, which increased their susceptibility to HSV-1 infection [174].
Unlike HSV-1, CXCL10 was found to promote the infection caused by stimulating virus replication in immune cells during herpes simplex virus type 2 (HSV-2) infection [181]. The expression of CXCL9 and CXCL10 was increased in the cervical tissues of mice infected with HSV-2 [182,183,184]. At the same time, other studies reported an upregulation of CXCL10 and CXCL11 during HSV-2 infection [182,185,186]. Moreover, the HSV-2 protein infected-cell polypeptide 4 (ICP4) was suggested to be the critical viral component that stimulated the production of CXCL9, CXCL10, and CXCL11 [186].

5. Role of CXCL10 in Oncolytic Viruses

Multiple strains of oncolytic viruses such as vesicular stomatitis virus (VSV), HSV-2, maraba virus, chikungunya virus, and reolysin have been reported to drive the high expression of chemokines in infected tumors [187,188,189,190]. For instance, the intratumoral administration of oncolytic HSV-2 was associated with high CXCL10 expression, promoting the attraction of adoptively transferred T cells towards treated lesions [190]. Among many other cytokines and chemokines, CXCL10 was tested as an immune stimulant in the genomes of oncolytic viruses, to enhance the anti-tumor activity in several cancer studies [187]. Moreover, VSV was identified in promoting inflammatory tumor cell killing via the release of chemokines, including CXCL10, which trigger the infiltration of T effector cells into the tumor growth sites [191]. Another example was the oncolytic adenovirus with an inserted CXCL10 gene (Adv-CXCL10), which enhanced the recruitment of CXCR3+ T cells into the colon tumor microenvironment corroborated with increasing the efficacy of PD-1 antibody [192]. These results were translated into a clinical trial using an oncolytic adenovirus (NG-641) expressing CXCL9, CXCL10, and IFN-α, which is currently under investigation (NCT04053283) [193,194].

6. Conclusions

An impaired immune response is a critical hallmark in the development and progression of viral infections. Notably, the role of chemokines during viral infections has been explored and analyzed during the context of many RNA and DNA viral infections. CXCL10 was found to mediate its inflammatory activity through CXCR3, which is mainly expressed on T cells, NK cells, macrophages, and dendritic cells. Several studies report the significance of CXCL10 and its chemokine receptor CXCR3 in host defense and viral clearance, while other reports demonstrate its role in disease pathogenesis. Moreover, CXCL10 was suggested to be a potential diagnostic and prognostic marker in several viral infections such as COVID-19. Further, CXCL10 has a potential role as an anti-tumor agent mediated by oncolytic viruses. Therefore, targeting CXCL10 could be a possible therapeutic modality against many viral infections, as well as against tumor development. Further in-depth understanding of the chemokine system may allow the development of such therapeutics against viral infections, which could be beneficial in preventing opportunistic microbes.

Author Contributions

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

Funding

This research was funded by the Terry Fox Foundation (MISC051) and by the Sharjah Research Academy (2101090289).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Maghazachi, A.M.A.; Al-Aoukaty, A. Chemokines activate natural killer cells through heterotrimeric G-proteins: Implications for the treatment of AIDS and cancer. FASEB J. 1998, 12, 913–924. [Google Scholar] [CrossRef] [PubMed]
  2. Nomiyama, H.; Osada, N.; Yoshie, O. The evolution of mammalian chemokine genes. Cytokine Growth Factor Rev. 2010, 21, 253–262. [Google Scholar] [CrossRef] [PubMed]
  3. Proudfoot, A.E.; Uguccioni, M. Modulation of Chemokine Responses: Synergy and Cooperativity. Front. Immunol. 2016, 7, 183. [Google Scholar] [CrossRef] [Green Version]
  4. Mellado, M.; Rodríguez-Frade, J.M.; Vila-Coro, A.J.; Fernández, S.; Martín de Ana, A.; Jones, D.R.; Torán, J.L.; Martínez, A.C. Chemokine receptor homo-or heterodimerization activates distinct signaling pathways. EMBO J. 2001, 20, 2497–2507. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Murphy, P.M.; Baggiolini, M.; Charo, I.F.; Hébert, C.A.; Horuk, R.; Matsushima, K.; Miller, L.H.; Oppenheim, J.J.; Power, C.A. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol. Rev. 2000, 52, 145–176. [Google Scholar] [PubMed]
  6. Kennedy, J.; Kelner, G.S.; Kleyensteuber, S.; Schall, T.J.; Weiss, M.C.; Yssel, H.; Schneider, P.V.; Cocks, B.G.; Bacon, K.B.; Zlotnik, A. Molecular cloning and functional characterization of human lymphotactin. J. Immunol. 1995, 155, 203–209. [Google Scholar] [PubMed]
  7. Hughes, C.E.; Nibbs, R.J.B. A guide to chemokines and their receptors. FEBS J. 2018, 285, 2944–2971. [Google Scholar] [CrossRef]
  8. Stone, M.J.; Hayward, J.A.; Huang, C.; Huma, E.Z.; Sanchez, J. Mechanisms of Regulation of the Chemokine-Receptor Network. Int. J. Mol. Sci. 2017, 18, 342. [Google Scholar] [CrossRef] [Green Version]
  9. Wilson, S.; Wilkinson, G.; Milligan, G. The CXCR1 and CXCR2 receptors form constitutive homo- and heterodimers selectively and with equal apparent affinities. J. Biol. Chem. 2005, 280, 28663–28674. [Google Scholar] [CrossRef] [Green Version]
  10. Sohy, D.; Yano, H.; de Nadai, P.; Urizar, E.; Guillabert, A.; Javitch, J.A.; Parmentier, M.; Springael, J.Y. Hetero-oligomerization of CCR2, CCR5, and CXCR4 and the protean effects of “selective” antagonists. J. Biol. Chem. 2009, 284, 31270–31279. [Google Scholar] [CrossRef]
  11. Petkovic, V.; Moghini, C.; Paoletti, S.; Uguccioni, M.; Gerber, B. Eotaxin-3/CCL26 is a natural antagonist for CC chemokine receptors 1 and 5. A human chemokine with a regulatory role. J. Biol. Chem. 2004, 279, 23357–23363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Nibbs, R.J.; Graham, G.J. Immune regulation by atypical chemokine receptors. Nat. Rev. Immunol. 2013, 13, 815–829. [Google Scholar] [CrossRef] [PubMed]
  13. Bachelerie, F.; Ben-Baruch, A.; Burkhardt, A.M.; Combadiere, C.; Farber, J.M.; Graham, G.J.; Horuk, R.; Sparre-Ulrich, A.H.; Locati, M.; Luster, A.D.; et al. International Union of Basic and Clinical Pharmacology. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol. Rev. 2013, 66, 1–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Cardona, A.E.; Sasse, M.E.; Liu, L.; Cardona, S.M.; Mizutani, M.; Savarin, C.; Hu, T.; Ransohoff, R.M. Scavenging roles of chemokine receptors: Chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues. Blood 2008, 112, 256–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fukuma, N.; Akimitsu, N.; Hamamoto, H.; Kusuhara, H.; Sugiyama, Y.; Sekimizu, K. A role of the Duffy antigen for the maintenance of plasma chemokine concentrations. Biochem. Biophys. Res. Commun. 2003, 303, 137–139. [Google Scholar] [CrossRef]
  16. Lee, J.S.; Frevert, C.W.; Wurfel, M.M.; Peiper, S.C.; Wong, V.A.; Ballman, K.K.; Ruzinski, J.T.; Rhim, J.S.; Martin, T.R.; Goodman, R.B. Duffy antigen facilitates movement of chemokine across the endothelium in vitro and promotes neutrophil transmigration in vitro and in vivo. J. Immunol. 2003, 170, 5244–5251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Pruenster, M.; Mudde, L.; Bombosi, P.; Dimitrova, S.; Zsak, M.; Middleton, J.; Richmond, A.; Graham, G.J.; Segerer, S.; Nibbs, R.J.; et al. The Duffy antigen receptor for chemokines transports chemokines and supports their promigratory activity. Nat. Immunol. 2009, 10, 101–108. [Google Scholar] [CrossRef] [PubMed]
  18. Elemam, N.M.; Khalil, B.A.; Maghazachi, A.A. Chemokines and chemokine receptors. In Encyclopedia of Infection and Immunity; Rezaei, N., Ed.; Elsevier: Oxford, UK, 2022; pp. 193–205. [Google Scholar]
  19. Kleist, A.B.; Getschman, A.E.; Ziarek, J.J.; Nevins, A.M.; Gauthier, P.A.; Chevigné, A.; Szpakowska, M.; Volkman, B.F. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem. Pharmacol. 2016, 114, 53–68. [Google Scholar] [CrossRef] [Green Version]
  20. Salazar-Mather, T.P.; Hokeness, K.L. Cytokine and chemokine networks: Pathways to antiviral defense. Curr. Top. Microbiol. Immunol. 2006, 303, 29–46. [Google Scholar] [CrossRef]
  21. Decalf, J.R.M.; Fernandes, S.; Longman, R.; Ahloulay, M.; Audat, F.O.; Lefrerre, F.O.; Rice, C.M.; Pol, S.; Albert, M.L. Plasmacytoid dendritic cells initiate a complex chemokine and cytokine network and are a viable drug target in chronic HCV patients. J. Exp. Med. 2007, 204, 2423–2437. [Google Scholar] [CrossRef]
  22. Mahalingam, S.; Friedland, J.S.; Heise, M.T.; Rulli, N.E.; Meanger, J.; Lidbury, B.A. Chemokines and viruses: Friends or foes? Trends Microbiol. 2003, 11, 383–391. [Google Scholar] [CrossRef]
  23. Murphy, P.M. Viral exploitation and subversion of the immune system through chemokine mimicry. Nat. Immunol. 2001, 2, 116–122. [Google Scholar] [CrossRef] [PubMed]
  24. Loetscher, M.; Loetscher, P.; Brass, N.; Meese, E.; Moser, B. Lymphocyte-specific chemokine receptor CXCR3: Regulation, chemokine binding and gene localization. Eur. J. Immunol. 1998, 28, 3696–3705. [Google Scholar] [CrossRef]
  25. Qin, S.; Rottman, J.B.; Myers, P.; Kassam, N.; Weinblatt, M.; Loetscher, M.; Koch, A.E.; Moser, B.; Mackay, C.R. The chemokine receptors CXCR3 and CCR5 mark subsets of T cells associated with certain inflammatory reactions. J. Clin. Investig. 1998, 101, 746–754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Sallusto, F.; Lenig, D.; Mackay, C.R.; Lanzavecchia, A. Flexible programs of chemokine receptor expression on human polarized T helper 1 and 2 lymphocytes. J. Exp. Med. 1998, 187, 875–883. [Google Scholar] [CrossRef]
  27. Wang, L.; Knudsen, E.; Jin, Y.; Gessani, S.; Maghazachi, A.A. Lysophospholipids and chemokines activate distinct signal transduction pathways in T helper 1 and T helper 2 cells. Cell Signal. 2004, 16, 991–1000. [Google Scholar] [CrossRef]
  28. Kelsen, S.G.; Aksoy, M.O.; Yang, Y.; Shahabuddin, S.; Litvin, J.; Safadi, F.; Rogers, T.J. The chemokine receptor CXCR3 and its splice variant are expressed in human airway epithelial cells. Am. J. Physiol.-Lung Cell. Mol. Physiol. 2004, 287, L584–L591. [Google Scholar] [CrossRef]
  29. Lasagni, L.; Francalanci, M.; Annunziato, F.; Lazzeri, E.; Giannini, S.; Cosmi, L.; Sagrinati, C.; Mazzinghi, B.; Orlando, C.; Maggi, E.; et al. An alternatively spliced variant of CXCR3 mediates the inhibition of endothelial cell growth induced by IP-10, Mig, and I-TAC, and acts as functional receptor for platelet factor 4. J. Exp. Med. 2003, 197, 1537–1549. [Google Scholar] [CrossRef]
  30. Bodnar, R.J.; Yates, C.C.; Wells, A. IP-10 blocks vascular endothelial growth factor-induced endothelial cell motility and tube formation via inhibition of calpain. Circ. Res. 2006, 98, 617–625. [Google Scholar] [CrossRef]
  31. Bonecchi, R.; Locati, M.; Galliera, E.; Vulcano, M.; Sironi, M.; Fra, A.M.; Gobbi, M.; Vecchi, A.; Sozzani, S.; Haribabu, B.; et al. Differential Recognition and Scavenging of Native and Truncated Macrophage-Derived Chemokine (Macrophage-Derived Chemokine/CC Chemokine Ligand 22) by the D6 Decoy Receptor. J. Immunol. 2004, 172, 4972. [Google Scholar] [CrossRef]
  32. Chevigné, A.; Janji, B.; Meyrath, M.; Reynders, N.; D’Uonnolo, G.; Uchański, T.; Xiao, M.; Berchem, G.; Ollert, M.; Kwon, Y.-J.; et al. CXCL10 Is an Agonist of the CC Family Chemokine Scavenger Receptor ACKR2/D6. Cancers 2021, 13, 1054. [Google Scholar] [CrossRef] [PubMed]
  33. Dyer, K.D.; Percopo, C.M.; Fischer, E.R.; Gabryszewski, S.J.; Rosenberg, H.F. Pneumoviruses infect eosinophils and elicit MyD88-dependent release of chemoattractant cytokines and interleukin-6. Blood 2009, 114, 2649–2656. [Google Scholar] [CrossRef] [PubMed]
  34. Luster, A.D.; Ravetch, J.V. Biochemical characterization of a gamma interferon-inducible cytokine (IP-10). J. Exp. Med. 1987, 166, 1084–1097. [Google Scholar] [CrossRef]
  35. Lo, B.K.; Yu, M.; Zloty, D.; Cowan, B.; Shapiro, J.; McElwee, K.J. CXCR3/ligands are significantly involved in the tumorigenesis of basal cell carcinomas. Am. J. Pathol. 2010, 176, 2435–2446. [Google Scholar] [CrossRef] [Green Version]
  36. Angiolillo, A.L.; Sgadari, C.; Taub, D.D.; Liao, F.; Farber, J.M.; Maheshwari, S.; Kleinman, H.K.; Reaman, G.H.; Tosato, G. Human interferon-inducible protein 10 is a potent inhibitor of angiogenesis in vivo. J. Exp. Med. 1995, 182, 155–162. [Google Scholar] [CrossRef] [Green Version]
  37. Persano, L.; Crescenzi, M.; Indraccolo, S. Anti-angiogenic gene therapy of cancer: Current status and future prospects. Mol. Aspects Med. 2007, 28, 87–114. [Google Scholar] [CrossRef]
  38. Belperio, J.A.; Keane, M.P.; Arenberg, D.A.; Addison, C.L.; Ehlert, J.E.; Burdick, M.D.; Strieter, R.M. CXC chemokines in angiogenesis. J. Leukoc. Biol. 2000, 68, 1–8. [Google Scholar] [CrossRef]
  39. Strieter, R.M.; Polverini, P.J.; Kunkel, S.L.; Arenberg, D.A.; Burdick, M.D.; Kasper, J.; Dzuiba, J.; Van Damme, J.; Walz, A.; Marriott, D.; et al. The functional role of the ELR motif in CXC chemokine-mediated angiogenesis. J. Biol. Chem. 1995, 270, 27348–27357. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Swaminathan, G.J.; Holloway, D.E.; Colvin, R.A.; Campanella, G.K.; Papageorgiou, A.C.; Luster, A.D.; Acharya, K.R. Crystal structures of oligomeric forms of the IP-10/CXCL10 chemokine. Structure 2003, 11, 521–532. [Google Scholar] [CrossRef]
  41. Ohmori, Y.; Hamilton, T.A. Cooperative interaction between interferon (IFN) stimulus response element and kappa B sequence motifs controls IFN gamma- and lipopolysaccharide-stimulated transcription from the murine IP-10 promoter. J. Biol. Chem. 1993, 268, 6677–6688. [Google Scholar] [CrossRef]
  42. Majumder, S.; Zhou, L.Z.; Chaturvedi, P.; Babcock, G.; Aras, S.; Ransohoff, R.M. Regulation of human IP-10 gene expression in astrocytoma cells by inflammatory cytokines. J. Neurosci. Res. 1998, 54, 169–180. [Google Scholar] [CrossRef]
  43. Varley, C.L.; Armitage, S.; Hassanshahiraviz, G.; Dickson, A.J. Regulation of the C-X-C chemokine, mob-1, gene expression in primary rat hepatocytes. Cytokine 2003, 23, 64–75. [Google Scholar] [CrossRef]
  44. Romagnani, P.; Lazzeri, E.; Lasagni, L.; Mavilia, C.; Beltrame, C.; Francalanci, M.; Rotondi, M.; Annunziato, F.; Maurenzig, L.; Cosmi, L.; et al. IP-10 and Mig production by glomerular cells in human proliferative glomerulonephritis and regulation by nitric oxide. J. Am. Soc. Nephrol. 2002, 13, 53–64. [Google Scholar] [CrossRef] [PubMed]
  45. Han, B.; Logsdon, C.D. Cholecystokinin induction of mob-1 chemokine expression in pancreatic acinar cells requires NF-kappaB activation. Am. J. Physiol. 1999, 277, C74–C82. [Google Scholar] [CrossRef] [PubMed]
  46. Han, B.; Logsdon, C.D. CCK stimulates mob-1 expression and NF-kappaB activation via protein kinase C and intracellular Ca(2+). Am. J. Physiol. Cell Physiol. 2000, 278, C344–C351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Lee, E.Y.; Lee, Z.H.; Song, Y.W. CXCL10 and autoimmune diseases. Autoimmun. Rev. 2009, 8, 379–383. [Google Scholar] [CrossRef]
  48. Liu, M.; Guo, S.; Stiles, J.K. The emerging role of CXCL10 in cancer (Review). Oncol. Lett. 2011, 2, 583–589. [Google Scholar] [CrossRef] [Green Version]
  49. Mee, J.B.; Johnson, C.M.; Morar, N.; Burslem, F.; Groves, R.W. The psoriatic transcriptome closely resembles that induced by interleukin-1 in cultured keratinocytes: Dominance of innate immune responses in psoriasis. Am. J. Pathol. 2007, 171, 32–42. [Google Scholar] [CrossRef] [Green Version]
  50. Elemam, N.M.; Hannawi, S.; Maghazachi, A.A. Role of chemokines and chemokine receptors in rheumatoid arthritis. Immunotargets Ther. 2020, 9, 43–56. [Google Scholar] [CrossRef] [Green Version]
  51. Trifilo, M.J.; Montalto-Morrison, C.; Stiles, L.N.; Hurst, K.R.; Hardison, J.L.; Manning, J.E.; Masters, P.S.; Lane, T.E. CXC chemokine ligand 10 controls viral infection in the central nervous system: Evidence for a role in innate immune response through recruitment and activation of natural killer cells. J. Virol. 2004, 78, 585–594. [Google Scholar] [CrossRef]
  52. Deng, G.; Zhou, G.; Zhang, R.; Zhai, Y.; Zhao, W.; Yan, Z.; Deng, C.; Yuan, X.; Xu, B.; Dong, X.; et al. Regulatory polymorphisms in the promoter of CXCL10 gene and disease progression in male hepatitis B virus carriers. Gastroenterology 2008, 134, 716–726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Haeberle, H.A.; Kuziel, W.A.; Dieterich, H.J.; Casola, A.; Gatalica, Z.; Garofalo, R.P. Inducible expression of inflammatory chemokines in respiratory syncytial virus-infected mice: Role of MIP-1alpha in lung pathology. J. Virol. 2001, 75, 878–890. [Google Scholar] [CrossRef] [Green Version]
  54. Tripp, R.A.; Jones, L.; Anderson, L.J. Respiratory syncytial virus G and/or SH glycoproteins modify CC and CXC chemokine mRNA expression in the BALB/c mouse. J. Virol. 2000, 74, 6227–6229. [Google Scholar] [CrossRef] [Green Version]
  55. Schneider, D.; Ganesan, S.; Comstock, A.T.; Meldrum, C.A.; Mahidhara, R.; Goldsmith, A.M.; Curtis, J.L.; Martinez, F.J.; Hershenson, M.B.; Sajjan, U. Increased cytokine response of rhinovirus-infected airway epithelial cells in chronic obstructive pulmonary disease. Am. J. Respir. Crit. Care Med. 2010, 182, 332–340. [Google Scholar] [CrossRef]
  56. Mihm, S.; Schweyer, S.; Ramadori, G. Expression of the chemokine IP-10 correlates with the accumulation of hepatic IFN-gamma and IL-18 mRNA in chronic hepatitis C but not in hepatitis B. J. Med. Virol. 2003, 70, 562–570. [Google Scholar] [CrossRef]
  57. Mahanty, S.; Gupta, M.; Paragas, J.; Bray, M.; Ahmed, R.; Rollin, P.E. Protection from lethal infection is determined by innate immune responses in a mouse model of Ebola virus infection. Virology 2003, 312, 415–424. [Google Scholar] [CrossRef] [Green Version]
  58. Nightingale, Z.D.; Patkar, C.; Rothman, A.L. Viral replication and paracrine effects result in distinct, functional responses of dendritic cells following infection with dengue 2 virus. J. Leukoc. Biol. 2008, 84, 1028–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Warke, R.V.; Becerra, A.; Zawadzka, A.; Schmidt, D.J.; Martin, K.J.; Giaya, K.; Dinsmore, J.H.; Woda, M.; Hendricks, G.; Levine, T.; et al. Efficient dengue virus (DENV) infection of human muscle satellite cells upregulates type I interferon response genes and differentially modulates MHC I expression on bystander and DENV-infected cells. J. Gen. Virol. 2008, 89, 1605–1615. [Google Scholar] [CrossRef] [PubMed]
  60. Covaleda, L.; Fuller, F.J.; Payne, S.L. EIAV S2 enhances pro-inflammatory cytokine and chemokine response in infected macrophages. Virology 2010, 397, 217–223. [Google Scholar] [CrossRef] [Green Version]
  61. Glass, W.G.; Subbarao, K.; Murphy, B.; Murphy, P.M. Mechanisms of host defense following severe acute respiratory syndrome-coronavirus (SARS-CoV) pulmonary infection of mice. J. Immunol. 2004, 173, 4030–4039. [Google Scholar] [CrossRef]
  62. Tsunoda, I.; Lane, T.E.; Blackett, J.; Fujinami, R.S. Distinct roles for IP-10/CXCL10 in three animal models, Theiler’s virus infection, EAE, and MHV infection, for multiple sclerosis: Implication of differing roles for IP-10. Mult. Scler. 2004, 10, 26–34. [Google Scholar] [CrossRef] [PubMed]
  63. Christensen, J.E.; de Lemos, C.; Moos, T.; Christensen, J.P.; Thomsen, A.R. CXCL10 is the key ligand for CXCR3 on CD8+ effector T cells involved in immune surveillance of the lymphocytic choriomeningitis virus-infected central nervous system. J. Immunol. 2006, 176, 4235–4243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Stiles, L.N.; Hosking, M.P.; Edwards, R.A.; Strieter, R.M.; Lane, T.E. Differential roles for CXCR3 in CD4+ and CD8+ T cell trafficking following viral infection of the CNS. Eur. J. Immunol. 2006, 36, 613–622. [Google Scholar] [CrossRef] [PubMed]
  65. De Lemos, C.; Christensen, J.E.; Nansen, A.; Moos, T.; Lu, B.; Gerard, C.; Christensen, J.P.; Thomsen, A.R. Opposing effects of CXCR3 and CCR5 deficiency on CD8+ T cell-mediated inflammation in the central nervous system of virus-infected mice. J. Immunol. 2005, 175, 1767–1775. [Google Scholar] [CrossRef] [Green Version]
  66. Li, H.; Gang, Z.; Yuling, H.; Luokun, X.; Jie, X.; Hao, L.; Li, W.; Chunsong, H.; Junyan, L.; Mingshen, J.; et al. Different neurotropic pathogens elicit neurotoxic CCR9-or neurosupportive CXCR3-expressing microglia. J. Immunol. 2006, 177, 3644–3656. [Google Scholar] [CrossRef] [Green Version]
  67. Gandhi, K.S.; McKay, F.C.; Diefenbach, E.; Crossett, B.; Schibeci, S.D.; Heard, R.N.; Stewart, G.J.; Booth, D.R.; Arthur, J.W. Novel approaches to detect serum biomarkers for clinical response to interferon-β treatment in multiple sclerosis. PLoS ONE 2010, 5, e10484. [Google Scholar] [CrossRef] [Green Version]
  68. Treacy, O.; Ryan, A.E.; Heinzl, T.; O’Flynn, L.; Cregg, M.; Wilk, M.; Odoardi, F.; Lohan, P.; O’Brien, T.; Nosov, M.; et al. Adenoviral transduction of mesenchymal stem cells: In vitro responses and in vivo immune responses after cell transplantation. PLoS ONE 2012, 7, e42662. [Google Scholar] [CrossRef] [Green Version]
  69. Rubin, S.M. Management of multiple sclerosis: An overview. Dis. Month 2013, 59, 253–260. [Google Scholar] [CrossRef]
  70. Iwakura, Y.; Ishigame, H. The IL-23/IL-17 axis in inflammation. J. Clin. Investig. 2006, 116, 1218–1222. [Google Scholar] [CrossRef] [Green Version]
  71. Kakimi, K.; Lane, T.E.; Wieland, S.; Asensio, V.C.; Campbell, I.L.; Chisari, F.V.; Guidotti, L.G. Blocking chemokine responsive to gamma-2/interferon (IFN)-gamma inducible protein and monokine induced by IFN-gamma activity in vivo reduces the pathogenetic but not the antiviral potential of hepatitis B virus-specific cytotoxic T lymphocytes. J. Exp. Med. 2001, 194, 1755–1766. [Google Scholar] [CrossRef]
  72. Loetscher, M.; Gerber, B.; Loetscher, P.; Jones, S.A.; Piali, L.; Clark-Lewis, I.; Baggiolini, M.; Moser, B. Chemokine receptor specific for IP10 and mig: Structure, function, and expression in activated T-lymphocytes. J. Exp. Med. 1996, 184, 963–969. [Google Scholar] [CrossRef] [PubMed]
  73. Arai, K.; Liu, Z.X.; Lane, T.; Dennert, G. IP-10 and Mig facilitate accumulation of T cells in the virus-infected liver. Cell. Immunol. 2002, 219, 48–56. [Google Scholar] [CrossRef]
  74. Maghazachi, A.A.; Skålhegg, B.S.; Rolstad, B.; Al-Aoukaty, A. Interferon-inducible protein-10 and lymphotactin induce the chemotaxis and mobilization of intracellular calcium in natural killer cells through pertussis toxin-sensitive and-insensitive heterotrimeric G-proteins. FASEB J. 1997, 11, 765–774. [Google Scholar] [CrossRef] [PubMed]
  75. Mahalingam, S.; Farber, J.M.; Karupiah, G. The interferon-inducible chemokines MuMig and Crg-2 exhibit antiviral activity In vivo. J. Virol. 1999, 73, 1479–1491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Cole, A.M.; Ganz, T.; Liese, A.M.; Burdick, M.D.; Liu, L.; Strieter, R.M. Cutting edge: IFN-inducible ELR- CXC chemokines display defensin-like antimicrobial activity. J. Immunol. 2001, 167, 623–627. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Chen, J.P.; Lu, H.L.; Lai, S.L.; Campanella, G.S.; Sung, J.M.; Lu, M.Y.; Wu-Hsieh, B.A.; Lin, Y.L.; Lane, T.E.; Luster, A.D.; et al. Dengue virus induces expression of CXC chemokine ligand 10/IFN-gamma-inducible protein 10, which competitively inhibits viral binding to cell surface heparan sulfate. J. Immunol. 2006, 177, 3185–3192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Yuan, J.; Liu, Z.; Lim, T.; Zhang, H.; He, J.; Walker, E.; Shier, C.; Wang, Y.; Su, Y.; Sall, A.; et al. CXCL10 inhibits viral replication through recruitment of natural killer cells in coxsackievirus B3-induced myocarditis. Circ. Res. 2009, 104, 628–638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Lombardi, A.; Cantini, G.; Mello, T.; Francalanci, M.; Gelmini, S.; Cosmi, L.; Santarlasci, V.; Degl’Innocenti, S.; Luciani, P.; Deledda, C.; et al. Molecular mechanisms underlying the pro-inflammatory synergistic effect of tumor necrosis factor alpha and interferon gamma in human microvascular endothelium. Eur. J. Cell Biol. 2009, 88, 731–742. [Google Scholar] [CrossRef]
  80. Dhillon, N.K.; Peng, F.; Ransohoff, R.M.; Buch, S. PDGF synergistically enhances IFN-γ-induced expression of CXCL10 in blood-derived macrophages: Implications for HIV dementia. J. Immunol. 2007, 179, 2722. [Google Scholar] [CrossRef] [Green Version]
  81. Hardaker, E.L.; Bacon, A.M.; Carlson, K.; Roshak, A.K.; Foley, J.J.; Schmidt, D.B.; Buckley, P.T.; Comegys, M.; Panettieri, R.A., Jr.; Sarau, H.M.; et al. Regulation of TNF-alpha- and IFN-gamma-induced CXCL10 expression: Participation of the airway smooth muscle in the pulmonary inflammatory response in chronic obstructive pulmonary disease. FASEB J. 2004, 18, 191–193. [Google Scholar] [CrossRef]
  82. Kanda, N.; Watanabe, S. Substance P enhances the production of interferon-induced protein of 10 kDa by human keratinocytes in synergy with interferon-gamma. J. Investig. Dermatol. 2002, 119, 1290–1297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Loos, T.; Dekeyzer, L.; Struyf, S.; Schutyser, E.; Gijsbers, K.; Gouwy, M.; Fraeyman, A.; Put, W.; Ronsse, I.; Grillet, B.; et al. TLR ligands and cytokines induce CXCR3 ligands in endothelial cells: Enhanced CXCL9 in autoimmune arthritis. Lab. Investig. 2006, 86, 902–916. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Brentano, F.; Schorr, O.; Gay, R.E.; Gay, S.; Kyburz, D. RNA released from necrotic synovial fluid cells activates rheumatoid arthritis synovial fibroblasts via Toll-like receptor 3. Arthritis Rheum. 2005, 52, 2656–2665. [Google Scholar] [CrossRef] [PubMed]
  85. Cheng, G.; Nazar, A.S.; Shin, H.S.; Vanguri, P.; Shin, M.L. IP-10 gene transcription by virus in astrocytes requires cooperation of ISRE with adjacent kappaB site but not IRF-1 or viral transcription. J. Interferon Cytokine Res. 1998, 18, 987–997. [Google Scholar] [CrossRef] [PubMed]
  86. Lebre, M.C.; van der Aar, A.M.; van Baarsen, L.; van Capel, T.M.; Schuitemaker, J.H.; Kapsenberg, M.L.; de Jong, E.C. Human keratinocytes express functional Toll-like receptor 3, 4, 5, and 9. J. Investig. Dermatol. 2007, 127, 331–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Morris, G.E.; Parker, L.C.; Ward, J.R.; Jones, E.C.; Whyte, M.K.; Brightling, C.E.; Bradding, P.; Dower, S.K.; Sabroe, I. Cooperative molecular and cellular networks regulate Toll-like receptor-dependent inflammatory responses. FASEB J. 2006, 20, 2153–2155. [Google Scholar] [CrossRef] [Green Version]
  88. Taima, K.; Imaizumi, T.; Yamashita, K.; Ishikawa, A.; Fujita, T.; Yoshida, H.; Takanashi, S.; Okumura, K.; Satoh, K. Expression of IP-10/CXCL10 is upregulated by double-stranded RNA in BEAS-2B bronchial epithelial cells. Respiration 2006, 73, 360–364. [Google Scholar] [CrossRef]
  89. Majumder, S.; Zhou, L.Z.; Chaturvedi, P.; Babcock, G.; Aras, S.; Ransohoff, R.M. p48/STAT-1alpha-containing complexes play a predominant role in induction of IFN-gamma-inducible protein, 10 kDa (IP-10) by IFN-gamma alone or in synergy with TNF-alpha. J. Immunol. 1998, 161, 4736–4744. [Google Scholar]
  90. Oslund, K.L.; Zhou, X.; Lee, B.; Zhu, L.; Duong, T.; Shih, R.; Baumgarth, N.; Hung, L.-Y.; Wu, R.; Chen, Y. Synergistic Up-Regulation of CXCL10 by Virus and IFN γ in Human Airway Epithelial Cells. PLoS ONE 2014, 9, e100978. [Google Scholar] [CrossRef] [Green Version]
  91. Carpentier, P.A.; Williams, B.R.; Miller, S.D. Distinct roles of protein kinase R and toll-like receptor 3 in the activation of astrocytes by viral stimuli. Glia 2007, 55, 239–252. [Google Scholar] [CrossRef]
  92. Imaizumi, T.; Kumagai, M.; Taima, K.; Fujita, T.; Yoshida, H.; Satoh, K. Involvement of retinoic acid-inducible gene-I in the IFN-{gamma}/STAT1 signalling pathway in BEAS-2B cells. Eur. Respir. J. 2005, 25, 1077–1083. [Google Scholar] [CrossRef] [PubMed]
  93. Berghäll, H.; Sirén, J.; Sarkar, D.; Julkunen, I.; Fisher, P.B.; Vainionpää, R.; Matikainen, S. The interferon-inducible RNA helicase, mda-5, is involved in measles virus-induced expression of antiviral cytokines. Microbes Infect. 2006, 8, 2138–2144. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, J.; Subbarao, K. The immunobiology of SARS*. Annu. Rev. Immunol. 2007, 25, 443–472. [Google Scholar] [CrossRef] [PubMed]
  95. Hsieh, Y.H.; Chen, C.W.; Schmitz, S.F.; King, C.C.; Chen, W.J.; Wu, Y.C.; Ho, M.S. Candidate genes associated with susceptibility for SARS-coronavirus. Bull. Math. Biol. 2010, 72, 122–132. [Google Scholar] [CrossRef]
  96. Sgadari, C.; Angiolillo, A.L.; Cherney, B.W.; Pike, S.E.; Farber, J.M.; Koniaris, L.G.; Vanguri, P.; Burd, P.R.; Sheikh, N.; Gupta, G.; et al. Interferon-inducible protein-10 identified as a mediator of tumor necrosis in vivo. Proc. Natl. Acad. Sci. USA 1996, 93, 13791–13796. [Google Scholar] [CrossRef] [Green Version]
  97. Lane, B.R.; King, S.R.; Bock, P.J.; Strieter, R.M.; Coffey, M.J.; Markovitz, D.M. The C-X-C chemokine IP-10 stimulates HIV-1 replication. Virology 2003, 307, 122–134. [Google Scholar] [CrossRef] [Green Version]
  98. Peterson, K.E.; Robertson, S.J.; Portis, J.L.; Chesebro, B. Differences in cytokine and chemokine responses during neurological disease induced by polytropic murine retroviruses Map to separate regions of the viral envelope gene. J. Virol. 2001, 75, 2848–2856. [Google Scholar] [CrossRef] [Green Version]
  99. Roe, B.; Coughlan, S.; Hassan, J.; Grogan, A.; Farrell, G.; Norris, S.; Bergin, C.; Hall, W.W. Elevated serum levels of interferon- gamma -inducible protein-10 in patients coinfected with hepatitis C virus and HIV. J. Infect. Dis. 2007, 196, 1053–1057. [Google Scholar] [CrossRef] [Green Version]
  100. Lagging, M.; Romero, A.I.; Westin, J.; Norkrans, G.; Dhillon, A.P.; Pawlotsky, J.-M.; Zeuzem, S.; von Wagner, M.; Negro, F.; Schalm, S.W.; et al. IP-10 predicts viral response and therapeutic outcome in difficult-to-treat patients with HCV genotype 1 infection. Hepatology 2006, 44, 1617–1625. [Google Scholar] [CrossRef]
  101. Stylianou, E.; Aukrust, P.; Bendtzen, K.; Müller, F.; Frøland, S.S. Interferons and interferon (IFN)-inducible protein 10 during highly active anti-retroviral therapy (HAART)-possible immunosuppressive role of IFN-alpha in HIV infection. Clin. Exp. Immunol. 2000, 119, 479–485. [Google Scholar] [CrossRef]
  102. Lam, C.W.; Chan, M.H.; Wong, C.K. Severe acute respiratory syndrome: Clinical and laboratory manifestations. Clin. Biochem. Rev. 2004, 25, 121–132. [Google Scholar] [PubMed]
  103. Wong, C.K.; Lam, C.W.; Wu, A.K.; Ip, W.K.; Lee, N.L.; Chan, I.H.; Lit, L.C.; Hui, D.S.; Chan, M.H.; Chung, S.S.; et al. Plasma inflammatory cytokines and chemokines in severe acute respiratory syndrome. Clin. Exp. Immunol. 2004, 136, 95–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ozga, A.J.; Chow, M.T.; Lopes, M.E.; Servis, R.L.; Di Pilato, M.; Dehio, P.; Lian, J.; Mempel, T.R.; Luster, A.D. CXCL10 chemokine regulates heterogeneity of the CD8+ T cell response and viral set point during chronic infection. Immunity 2022, 55, 82–97.e88. [Google Scholar] [CrossRef] [PubMed]
  105. Kang, W.; Shin, E.C. Clinical implications of chemokines in acute and chronic hepatitis C virus infection. Yonsei Med. J. 2011, 52, 871–878. [Google Scholar] [CrossRef] [Green Version]
  106. Zeremski, M.; Petrovic, L.M.; Talal, A.H. The role of chemokines as inflammatory mediators in chronic hepatitis C virus infection. J. Viral Hepat. 2007, 14, 675–687. [Google Scholar] [CrossRef]
  107. Sung, P.S.; Hong, S.-H.; Lee, J.; Park, S.-H.; Yoon, S.K.; Chung, W.J.; Shin, E.-C. CXCL10 is produced in hepatitis A virus-infected cells in an IRF3-dependent but IFN-independent manner. Sci. Rep. 2017, 7, 6387. [Google Scholar] [CrossRef] [Green Version]
  108. Brownell, J.; Wagoner, J.; Lovelace, E.S.; Thirstrup, D.; Mohar, I.; Smith, W.; Giugliano, S.; Li, K.; Crispe, I.N.; Rosen, H.R.; et al. Independent, parallel pathways to CXCL10 induction in HCV-infected hepatocytes. J. Hepatol. 2013, 59, 701–708. [Google Scholar] [CrossRef] [Green Version]
  109. Brownell, J.; Bruckner, J.; Wagoner, J.; Thomas, E.; Loo, Y.M.; Gale, M., Jr.; Liang, T.J.; Polyak, S.J. Direct, interferon-independent activation of the CXCL10 promoter by NF-κB and interferon regulatory factor 3 during hepatitis C virus infection. J. Virol. 2014, 88, 1582–1590. [Google Scholar] [CrossRef] [Green Version]
  110. Murai, M.; Yoneyama, H.; Harada, A.; Yi, Z.; Vestergaard, C.; Guo, B.; Suzuki, K.; Asakura, H.; Matsushima, K. Active participation of CCR5+ CD8+ T lymphocytes in the pathogenesis of liver injury in graft-versus-host disease. J. Clin. Investig. 1999, 104, 49–57. [Google Scholar] [CrossRef]
  111. Curbishley, S.M.; Eksteen, B.; Gladue, R.P.; Lalor, P.; Adams, D.H. CXCR 3 activation promotes lymphocyte transendothelial migration across human hepatic endothelium under fluid flow. Am. J. Pathol. 2005, 167, 887–899. [Google Scholar] [CrossRef] [Green Version]
  112. Ajuebor, M.N.; Hogaboam, C.M.; Le, T.; Proudfoot, A.E.; Swain, M.G. CCL3/MIP-1alpha is pro-inflammatory in murine T cell-mediated hepatitis by recruiting CCR1-expressing CD4+ T cells to the liver. Eur. J. Immunol. 2004, 34, 2907–2918. [Google Scholar] [CrossRef] [PubMed]
  113. Apolinario, A.; Majano, P.L.; Alvarez-Pérez, E.; Saez, A.; Lozano, C.; Vargas, J.; García-Monzón, C. Increased expression of T cell chemokines and their receptors in chronic hepatitis C: Relationship with the histological activity of liver disease. Am. J. Gastroenterol. 2002, 97, 2861–2870. [Google Scholar] [CrossRef] [PubMed]
  114. Harvey, C.E.; Post, J.J.; Palladinetti, P.; Freeman, A.J.; Ffrench, R.A.; Kumar, R.K.; Marinos, G.; Lloyd, A.R. Expression of the chemokine IP-10 (CXCL10) by hepatocytes in chronic hepatitis C virus infection correlates with histological severity and lobular inflammation. J. Leukoc. Biol. 2003, 74, 360–369. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Helbig, K.J.; Ruszkiewicz, A.; Semendric, L.; Harley, H.A.J.; McColl, S.R.; Beard, M.R. Expression of the CXCR3 ligand I-TAC by hepatocytes in chronic hepatitis C and its correlation with hepatic inflammation. Hepatology 2004, 39, 1220–1229. [Google Scholar] [CrossRef]
  116. Zeremski, M.; Petrovic, L.M.; Chiriboga, L.; Brown, Q.B.; Yee, H.T.; Kinkhabwala, M.; Jacobson, I.M.; Dimova, R.; Markatou, M.; Talal, A.H. Intrahepatic levels of CXCR3-associated chemokines correlate with liver inflammation and fibrosis in chronic hepatitis C. Hepatology 2008, 48, 1440–1450. [Google Scholar] [CrossRef] [Green Version]
  117. Yin, X.; Li, X.; Ambardekar, C.; Hu, Z.; Lhomme, S.; Feng, Z. Hepatitis E virus persists in the presence of a type III interferon response. PLoS Pathog. 2017, 13, e1006417. [Google Scholar] [CrossRef] [Green Version]
  118. Sayed, I.M.; Verhoye, L.; Cocquerel, L.; Abravanel, F.; Foquet, L.; Montpellier, C.; Debing, Y.; Farhoudi, A.; Wychowski, C.; Dubuisson, J.; et al. Study of hepatitis E virus infection of genotype 1 and 3 in mice with humanised liver. Gut 2017, 66, 920–929. [Google Scholar] [CrossRef]
  119. Marion, O.; Lhomme, S.; Nayrac, M.; Dubois, M.; Pucelle, M.; Requena, M.; Migueres, M.; Abravanel, F.; Peron, J.M.; Carrere, N.; et al. Hepatitis E virus replication in human intestinal cells. Gut 2020, 69, 901. [Google Scholar] [CrossRef]
  120. Devhare, P.; Madiyal, M.; Mukhopadhyay, C.; Shetty, S.; Shastry, S. Interplay between Hepatitis E Virus and Host Cell Pattern Recognition Receptors. Int. J. Mol. Sci. 2021, 22, 9259. [Google Scholar] [CrossRef]
  121. Peeters, M.; Schenk, J.; De Somer, T.; Roskams, T.; Locus, T.; Klamer, S.; Subissi, L.; Suin, V.; Delwaide, J.; Stärkel, P.; et al. Viral clade is associated with severity of symptomatic genotype 3 Hepatitis E virus infections in Belgium, 2010–2018. J. Hepatol. 2022. [Google Scholar] [CrossRef]
  122. Ferrari, S.M.; Fallahi, P.; Ruffilli, I.; Elia, G.; Ragusa, F.; Paparo, S.R.; Patrizio, A.; Mazzi, V.; Colaci, M.; Giuggioli, D.; et al. Immunomodulation of CXCL10 Secretion by Hepatitis C Virus: Could CXCL10 Be a Prognostic Marker of Chronic Hepatitis C? J. Immunol. Res. 2019, 2019, 5878960. [Google Scholar] [CrossRef] [PubMed]
  123. Larrubia, J.R.; Benito-Martínez, S.; Calvino, M.; Sanz-de-Villalobos, E.; Parra-Cid, T. Role of chemokines and their receptors in viral persistence and liver damage during chronic hepatitis C virus infection. World J. Gastroenterol. 2008, 14, 7149–7159. [Google Scholar] [CrossRef] [PubMed]
  124. Skinner, D.; Marro, B.S.; Lane, T.E. Chemokine CXCL10 and coronavirus-induced neurologic disease. Viral Immunol. 2019, 32, 25–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Hayney, M.S.; Henriquez, K.M.; Barnet, J.H.; Ewers, T.; Champion, H.M.; Flannery, S.; Barrett, B. Serum IFN-γ-induced protein 10 (IP-10) as a biomarker for severity of acute respiratory infection in healthy adults. J. Clin. Virol. 2017, 90, 32–37. [Google Scholar] [CrossRef] [PubMed]
  126. Almansa, R.; Sanchez-Garcia, M.; Herrero, A.; Calzada, S.; Roig, V.; Barbado, J.; Rico, L.; Bobillo, F.; Eiros, J.M.; Iglesias, V.; et al. Host response cytokine signatures in viral and nonviral acute exacerbations of chronic obstructive pulmonary disease. J. Interferon Cytokine Res. 2011, 31, 409–413. [Google Scholar] [CrossRef]
  127. Quint, J.K.; Donaldson, G.C.; Goldring, J.J.; Baghai-Ravary, R.; Hurst, J.R.; Wedzicha, J.A. Serum IP-10 as a biomarker of human rhinovirus infection at exacerbation of COPD. Chest 2010, 137, 812–822. [Google Scholar] [CrossRef] [Green Version]
  128. Chen, J.; Lau, Y.F.; Lamirande, E.W.; Paddock, C.D.; Bartlett, J.H.; Zaki, S.R.; Subbarao, K. Cellular immune responses to severe acute respiratory syndrome coronavirus (SARS-CoV) infection in senescent BALB/c mice: CD4+ T cells are important in control of SARS-CoV infection. J. Virol. 2010, 84, 1289–1301. [Google Scholar] [CrossRef] [Green Version]
  129. Cameron, M.J.; Ran, L.; Xu, L.; Danesh, A.; Bermejo-Martin, J.F.; Cameron, C.M.; Muller, M.P.; Gold, W.L.; Richardson, S.E.; Poutanen, S.M.; et al. Interferon-mediated immunopathological events are associated with atypical innate and adaptive immune responses in patients with severe acute respiratory syndrome. J. Virol. 2007, 81, 8692–8706. [Google Scholar] [CrossRef] [Green Version]
  130. Tynell, J.; Westenius, V.; Rönkkö, E.; Munster, V.J.; Melén, K.; Österlund, P.; Julkunen, I. Middle East respiratory syndrome coronavirus shows poor replication but significant induction of antiviral responses in human monocyte-derived macrophages and dendritic cells. J. Gen. Virol. 2016, 97, 344–355. [Google Scholar] [CrossRef]
  131. Zhou, J.; Chu, H.; Li, C.; Wong, B.H.; Cheng, Z.S.; Poon, V.K.; Sun, T.; Lau, C.C.; Wong, K.K.; Chan, J.Y.; et al. Active replication of Middle East respiratory syndrome coronavirus and aberrant induction of inflammatory cytokines and chemokines in human macrophages: Implications for pathogenesis. J. Infect. Dis. 2014, 209, 1331–1342. [Google Scholar] [CrossRef]
  132. Jiang, Y.; Xu, J.; Zhou, C.; Wu, Z.; Zhong, S.; Liu, J.; Luo, W.; Chen, T.; Qin, Q.; Deng, P. Characterization of cytokine/chemokine profiles of severe acute respiratory syndrome. Am. J. Respir. Crit. Care Med. 2005, 171, 850–857. [Google Scholar] [CrossRef] [PubMed]
  133. Chu, H.; Chan, J.F.; Wang, Y.; Yuen, T.T.; Chai, Y.; Hou, Y.; Shuai, H.; Yang, D.; Hu, B.; Huang, X.; et al. Comparative replication and immune activation profiles of SARS-CoV-2 and SARS-CoV in human lungs: An ex vivo study with implications for the pathogenesis of COVID-19. Clin. Infect. Dis. 2020, 71, 1400–1409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Chi, Y.; Ge, Y.; Wu, B.; Zhang, W.; Wu, T.; Wen, T.; Liu, J.; Guo, X.; Huang, C.; Jiao, Y.; et al. Serum cytokine and chemokine profile in relation to the severity of coronavirus disease 2019 in China. J. Infect. Dis. 2020, 222, 746–754. [Google Scholar] [CrossRef] [PubMed]
  135. Elemam, N.M.; Hammoudeh, S.; Salameh, L.; Mahboub, B.; Alsafar, H.; Talaat, I.M.; Habib, P.; Siddiqui, M.; Hassan, K.O.; Al-Assaf, O.Y.; et al. Identifying immunological and clinical predictors of COVID-19 severity and sequelae by mathematical modeling. Front. Immunol. 2022, 13, 865845. [Google Scholar] [CrossRef] [PubMed]
  136. Hue, S.; Beldi-Ferchiou, A.; Bendib, I.; Surenaud, M.; Fourati, S.; Frapard, T.; Rivoal, S.; Razazi, K.; Carteaux, G.; Delfau-Larue, M.H.; et al. Uncontrolled Innate and Impaired Adaptive Immune Responses in Patients with COVID-19 Acute Respiratory Distress Syndrome. Am. J. Respir. Crit. Care Med. 2020, 202, 1509–1519. [Google Scholar] [CrossRef] [PubMed]
  137. Gudowska-Sawczuk, M.; Mroczko, B. What is currently known about the role of CXCL10 in SARS-CoV-2 infection? Int. J. Mol. Sci. 2022, 23, 3673. [Google Scholar] [CrossRef]
  138. Yang, Y.; Shen, C.; Li, J.; Yuan, J.; Wei, J.; Huang, F.; Wang, F.; Li, G.; Li, Y.; Xing, L.; et al. Plasma IP-10 and MCP-3 levels are highly associated with disease severity and predict the progression of COVID-19. J. Allergy Clin. Immunol. 2020, 146, 119–127.e114. [Google Scholar] [CrossRef]
  139. Altara, R.; Manca, M.; Brandão, R.D.; Zeidan, A.; Booz, G.W.; Zouein, F.A. Emerging importance of chemokine receptor CXCR3 and its ligands in cardiovascular diseases. Clin. Sci. 2016, 130, 463–478. [Google Scholar] [CrossRef]
  140. Huang, C.; Wang, Y.; Li, X.; Ren, L.; Zhao, J.; Hu, Y.; Zhang, L.; Fan, G.; Xu, J.; Gu, X.; et al. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet 2020, 395, 497–506. [Google Scholar] [CrossRef] [Green Version]
  141. Lorè, N.I.; De Lorenzo, R.; Rancoita, P.M.V.; Cugnata, F.; Agresti, A.; Benedetti, F.; Bianchi, M.E.; Bonini, C.; Capobianco, A.; Conte, C.; et al. CXCL10 levels at hospital admission predict COVID-19 outcome: Hierarchical assessment of 53 putative inflammatory biomarkers in an observational study. Mol. Med. 2021, 27, 129. [Google Scholar] [CrossRef]
  142. Wang, J.; Xu, Y.; Zhang, X.; Wang, S.; Peng, Z.; Guo, J.; Jiang, H.; Liu, J.; Xie, Y.; Wang, J.; et al. Leptin correlates with monocytes activation and severe condition in COVID-19 patients. J. Leukoc. Biol. 2021, 110, 9–20. [Google Scholar] [CrossRef] [PubMed]
  143. Kwon, J.S.; Kim, J.Y.; Kim, M.C.; Park, S.Y.; Kim, B.N.; Bae, S.; Cha, H.H.; Jung, J.; Kim, M.J.; Lee, M.J.; et al. Factors of severity in patients with COVID-19: Cytokine/chemokine concentrations, viral load, and antibody responses. Am. J. Trop. Med. Hyg. 2020, 103, 2412–2418. [Google Scholar] [CrossRef] [PubMed]
  144. Xu, Z.S.; Shu, T.; Kang, L.; Wu, D.; Zhou, X.; Liao, B.W.; Sun, X.L.; Zhou, X.; Wang, Y.Y. Temporal profiling of plasma cytokines, chemokines and growth factors from mild, severe and fatal COVID-19 patients. Signal Transduct. Target. Ther. 2020, 5, 100. [Google Scholar] [CrossRef] [PubMed]
  145. Yang, L.; Han, Y.; Nilsson-Payant, B.E.; Gupta, V.; Wang, P.; Duan, X.; Tang, X.; Zhu, J.; Zhao, Z.; Jaffré, F.; et al. A human pluripotent stem cell-based platform to study SARS-CoV-2 tropism and model virus infection in human cells and organoids. Cell Stem Cell 2020, 27, 125–136.e127. [Google Scholar] [CrossRef] [PubMed]
  146. Blanco-Melo, D.; Nilsson-Payant, B.E.; Liu, W.C.; Uhl, S.; Hoagland, D.; Møller, R.; Jordan, T.X.; Oishi, K.; Panis, M.; Sachs, D.; et al. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 2020, 181, 1036–1045.e1039. [Google Scholar] [CrossRef] [PubMed]
  147. Başar, E.Z.; Sönmez, H.E.; Uzuner, H.; Karadenizli, A.; Güngör, H.S.; Akgün, G.; Yetimakman, A.F.; Öncel, S.; Babaoğlu, K. CXCL10/IP10 as a Biomarker Linking Multisystem Inflammatory Syndrome and Left Ventricular Dysfunction in Children with SARS-CoV-2. J. Clin. Med. 2022, 11, 1416. [Google Scholar] [CrossRef]
  148. Imai, Y.; Kuba, K.; Neely, G.G.; Yaghubian-Malhami, R.; Perkmann, T.; van Loo, G.; Ermolaeva, M.; Veldhuizen, R.; Leung, Y.H.; Wang, H.; et al. Identification of oxidative stress and Toll-like receptor 4 signaling as a key pathway of acute lung injury. Cell 2008, 133, 235–249. [Google Scholar] [CrossRef]
  149. Barrenschee, M.; Lex, D.; Uhlig, S. Effects of the TLR2 agonists MALP-2 and Pam3Cys in isolated mouse lungs. PLoS ONE 2010, 5, e13889. [Google Scholar] [CrossRef] [Green Version]
  150. Ichikawa, A.; Kuba, K.; Morita, M.; Chida, S.; Tezuka, H.; Hara, H.; Sasaki, T.; Ohteki, T.; Ranieri, V.M.; dos Santos, C.C.; et al. CXCL10-CXCR3 enhances the development of neutrophil-mediated fulminant lung injury of viral and nonviral origin. Am. J. Respir. Crit. Care Med. 2013, 187, 65–77. [Google Scholar] [CrossRef] [Green Version]
  151. Neville, L.F.; Mathiak, G.; Bagasra, O. The immunobiology of interferon-gamma inducible protein 10 kD (IP-10): A novel, pleiotropic member of the C-X-C chemokine superfamily. Cytokine Growth Factor Rev. 1997, 8, 207–219. [Google Scholar] [CrossRef]
  152. Wang, W.; Yang, P.; Zhong, Y.; Zhao, Z.; Xing, L.; Zhao, Y.; Zou, Z.; Zhang, Y.; Li, C.; Li, T.; et al. Monoclonal antibody against CXCL-10/IP-10 ameliorates influenza A (H1N1) virus induced acute lung injury. Cell Res. 2013, 23, 577–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Cinatl, J., Jr.; Michaelis, M.; Morgenstern, B.; Doerr, H.W. High-dose hydrocortisone reduces expression of the pro-inflammatory chemokines CXCL8 and CXCL10 in SARS coronavirus-infected intestinal cells. Int. J. Mol. Med. 2005, 15, 323–327. [Google Scholar] [CrossRef] [PubMed]
  154. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  155. Tanaka, T.; Narazaki, M.; Kishimoto, T. Immunotherapeutic implications of IL-6 blockade for cytokine storm. Immunotherapy 2016, 8, 959–970. [Google Scholar] [CrossRef] [PubMed]
  156. Coperchini, F.; Chiovato, L.; Rotondi, M. Interleukin-6, CXCL10 and infiltrating macrophages in COVID-19-related cytokine storm: Not one for all but all for one! Front. Immunol. 2021, 12, 668507. [Google Scholar] [CrossRef] [PubMed]
  157. Semmler, G.; Griebler, H.; Aberle, S.W.; Stiasny, K.; Richter, L.; Holzmann, H.; Weseslindtner, L. Elevated CXCL10 serum levels in measles virus primary infection and reinfection correlate with the serological stage and hospitalization status. J. Infect. Dis. 2020, 222, 2030–2034. [Google Scholar] [CrossRef]
  158. Lin, W.W.; Nelson, A.N.; Ryon, J.J.; Moss, W.J.; Griffin, D.E. Plasma cytokines and chemokines in zambian children with measles: Innate responses and association with HIV-1 coinfection and in-hospital mortality. J. Infect. Dis. 2017, 215, 830–839. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Moss, W.J. Measles. Lancet 2017, 390, 2490–2502. [Google Scholar] [CrossRef]
  160. Mina, M.J.; Metcalf, C.J.; de Swart, R.L.; Osterhaus, A.D.; Grenfell, B.T. Long-term measles-induced immunomodulation increases overall childhood infectious disease mortality. Science 2015, 348, 694–699. [Google Scholar] [CrossRef] [Green Version]
  161. Lindell, D.M.; Lane, T.E.; Lukacs, N.W. CXCL10/CXCR3-mediated responses promote immunity to respiratory syncytial virus infection by augmenting dendritic cell and CD8(+) T cell efficacy. Eur. J. Immunol. 2008, 38, 2168–2179. [Google Scholar] [CrossRef] [Green Version]
  162. Liu, M.; Guo, S.; Hibbert, J.M.; Jain, V.; Singh, N.; Wilson, N.O.; Stiles, J.K. CXCL10/IP-10 in infectious diseases pathogenesis and potential therapeutic implications. Cytokine Growth Factor Rev. 2011, 22, 121–130. [Google Scholar] [CrossRef] [PubMed]
  163. Naveca, F.G.; Pontes, G.S.; Chang, A.Y.; Silva, G.; Nascimento, V.A.D.; Monteiro, D.; Silva, M.S.D.; Abdalla, L.F.; Santos, J.H.A.; Almeida, T.A.P.; et al. Analysis of the immunological biomarker profile during acute Zika virus infection reveals the overexpression of CXCL10, a chemokine linked to neuronal damage. Mem. Inst. Oswaldo Cruz 2018, 113, e170542. [Google Scholar] [CrossRef] [PubMed]
  164. Steain, M.; Gowrishankar, K.; Rodriguez, M.; Slobedman, B.; Abendroth, A. Upregulation of CXCL10 in human dorsal root ganglia during experimental and natural varicella-zoster virus infection. J. Virol. 2011, 85, 626–631. [Google Scholar] [CrossRef] [Green Version]
  165. Dunn, C.; Brunetto, M.; Reynolds, G.; Christophides, T.; Kennedy, P.T.; Lampertico, P.; Das, A.; Lopes, A.R.; Borrow, P.; Williams, K.; et al. Cytokines induced during chronic hepatitis B virus infection promote a pathway for NK cell-mediated liver damage. J. Exp. Med. 2007, 204, 667–680. [Google Scholar] [CrossRef] [Green Version]
  166. Bayard, F.; Godon, O.; Nalpas, B.; Costentin, C.; Zhu, R.; Soussan, P.; Vallet-Pichard, A.; Fontaine, H.; Mallet, V.; Pol, S.; et al. T-cell responses to hepatitis B splice-generated protein of hepatitis B virus and inflammatory cytokines/chemokines in chronic hepatitis B patients. ANRS study: HB EP 02 HBSP-FIBRO. J. Viral Hepat. 2012, 19, 872–880. [Google Scholar] [CrossRef] [PubMed]
  167. Fisicaro, P.; Valdatta, C.; Boni, C.; Massari, M.; Mori, C.; Zerbini, A.; Orlandini, A.; Sacchelli, L.; Missale, G.; Ferrari, C. Early kinetics of innate and adaptive immune responses during hepatitis B virus infection. Gut 2009, 58, 974–982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Yoshio, S.; Sugiyama, M.; Shoji, H.; Mano, Y.; Mita, E.; Okamoto, T.; Matsuura, Y.; Okuno, A.; Takikawa, O.; Mizokami, M.; et al. Indoleamine-2,3-dioxygenase as an effector and an indicator of protective immune responses in patients with acute hepatitis B. Hepatology 2016, 63, 83–94. [Google Scholar] [CrossRef] [Green Version]
  169. Yoshio, S.; Mano, Y.; Doi, H.; Shoji, H.; Shimagaki, T.; Sakamoto, Y.; Kawai, H.; Matsuda, M.; Mori, T.; Osawa, Y.; et al. Cytokine and chemokine signatures associated with hepatitis B surface antigen loss in hepatitis B patients. JCI Insight 2018, 3, 122268. [Google Scholar] [CrossRef]
  170. Kakimi, K.; Lane, T.E.; Chisari, F.V.; Guidotti, L.G. Cutting edge: Inhibition of hepatitis B virus replication by activated NK T cells does not require inflammatory cell recruitment to the liver. J. Immunol. 2001, 167, 6701–6705. [Google Scholar] [CrossRef] [Green Version]
  171. Zhou, Y.; Wang, S.; Ma, J.-W.; Lei, Z.; Zhu, H.-F.; Lei, P.; Yang, Z.-S.; Zhang, B.; Yao, X.-X.; Shi, C.; et al. Hepatitis B virus protein X-induced expression of the CXC chemokine IP-10 is mediated through activation of NF-kB and increases migration of leukocytes. J. Biol. Chem. 2010, 285, 12159–12168. [Google Scholar] [CrossRef] [Green Version]
  172. Sitia, G.; Isogawa, M.; Kakimi, K.; Wieland, S.F.; Chisari, F.V.; Guidotti, L.G. Depletion of neutrophils blocks the recruitment of antigen-nonspecific cells into the liver without affecting the antiviral activity of hepatitis B virus-specific cytotoxic T lymphocytes. Proc. Natl. Acad. Sci. USA 2002, 99, 13717–13722. [Google Scholar] [CrossRef] [PubMed]
  173. Singh, K.P.; Zerbato, J.M.; Zhao, W.; Braat, S.; Deleage, C.; Tennakoon, G.S.; Mason, H.; Dantanarayana, A.; Rhodes, A.; Rhodes, J.W.; et al. Intrahepatic CXCL10 is strongly associated with liver fibrosis in HIV-Hepatitis B co-infection. PLoS Pathog. 2020, 16, e1008744. [Google Scholar] [CrossRef] [PubMed]
  174. Wuest, T.R.; Carr, D.J.J. Dysregulation of CXCR3 signaling due to CXCL10 deficiency impairs the antiviral response to herpes simplex virus 1 infection. J. Immunol. 2008, 181, 7985. [Google Scholar] [CrossRef] [Green Version]
  175. Srivastava, R.; Khan, A.A.; Chilukuri, S.; Syed, S.A.; Tran, T.T.; Furness, J.; Bahraoui, E.; BenMohamed, L. CXCL10/CXCR3-dependent mobilization of herpes simplex virus-specific CD8+ T(EM) and CD8+ T(RM) cells within infected tissues allows efficient protection against recurrent herpesvirus infection and disease. J. Virol. 2017, 91, e00278-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Carr, D.J.; Tomanek, L. Herpes simplex virus and the chemokines that mediate the inflammation. Curr. Top. Microbiol. Immunol. 2006, 303, 47–65. [Google Scholar] [CrossRef] [Green Version]
  177. Wickham, S.; Lu, B.; Ash, J.; Carr, D.J. Chemokine receptor deficiency is associated with increased chemokine expression in the peripheral and central nervous systems and increased resistance to herpetic encephalitis. J. Neuroimmunol. 2005, 162, 51–59. [Google Scholar] [CrossRef]
  178. Barbi, J.; Oghumu, S.; Lezama-Davila, C.M.; Satoskar, A.R. IFN-gamma and STAT1 are required for efficient induction of CXC chemokine receptor 3 (CXCR3) on CD4+ but not CD8+ T cells. Blood 2007, 110, 2215–2216. [Google Scholar] [CrossRef] [Green Version]
  179. Cook, W.J.; Kramer, M.F.; Walker, R.M.; Burwell, T.J.; Holman, H.A.; Coen, D.M.; Knipe, D.M. Persistent expression of chemokine and chemokine receptor RNAs at primary and latent sites of herpes simplex virus 1 infection. Virol. J. 2004, 1, 5. [Google Scholar] [CrossRef] [Green Version]
  180. O’Garra, A.; McEvoy, L.M.; Zlotnik, A. T-cell subsets: Chemokine receptors guide the way. Curr. Biol. 1998, 8, R646–R649. [Google Scholar] [CrossRef] [Green Version]
  181. Sin, J.; Kim, J.J.; Pachuk, C.; Satishchandran, C.; Weiner, D.B. DNA vaccines encoding interleukin-8 and RANTES enhance antigen-specific Th1-type CD4+ T-cell-mediated protective immunity against herpes simplex virus type 2 in vivo. J. Virol. 2000, 74, 11173–11180. [Google Scholar] [CrossRef] [Green Version]
  182. Thapa, M.; Carr Daniel, J.J. Herpes simplex virus type 2-induced mortality following genital infection is blocked by anti-tumor necrosis factor alpha antibody in CXCL10-deficient mice. J. Virol. 2008, 82, 10295–10301. [Google Scholar] [CrossRef] [PubMed]
  183. Nakanishi, Y.; Lu, B.; Gerard, C.; Iwasaki, A. CD8+ T lymphocyte mobilization to virus-infected tissue requires CD4+ T-cell help. Nature 2009, 462, 510–513. [Google Scholar] [CrossRef] [Green Version]
  184. Thapa, M.; Welner, R.S.; Pelayo, R.; Carr, D.J.J. CXCL9 and CXCL10 expression are critical for control of genital herpes simplex virus type 2 infection through mobilization of HSV-specific CTL and NK cells to the nervous system. J. Immunol. 2008, 180, 1098. [Google Scholar] [CrossRef] [Green Version]
  185. Lind, L.; Studahl, M.; Persson Berg, L.; Eriksson, K. CXCL11 production in cerebrospinal fluid distinguishes herpes simplex meningitis from herpes simplex encephalitis. J. Neuroinflamm. 2017, 14, 134. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Zhang, M.; Deng, X.; Guan, X.; Geng, L.; Fu, M.; Zhang, B.; Chen, R.; Hu, H.; Hu, K.; Zhang, D.; et al. Herpes simplex virus type 2 infection-induced expression of CXCR3 ligands promotes CD4+ T cell migration and is regulated by the viral immediate-early protein ICP4. Front. Immunol. 2018, 9, 32–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Pol, J.G.; Workenhe, S.T.; Konda, P.; Gujar, S.; Kroemer, G. Cytokines in oncolytic virotherapy. Cytokine Growth Factor Rev. 2020, 56, 4–27. [Google Scholar] [CrossRef] [PubMed]
  188. Abraham, R.; Mudaliar, P.; Padmanabhan, A.; Sreekumar, E. Induction of Cytopathogenicity in Human Glioblastoma Cells by Chikungunya Virus. PLoS ONE 2013, 8, e75854. [Google Scholar] [CrossRef]
  189. Carew, J.S.; Espitia, C.M.; Zhao, W.; Mita, M.M.; Mita, A.C.; Nawrocki, S.T. Oncolytic reovirus inhibits angiogenesis through induction of CXCL10/IP-10 and abrogation of HIF activity in soft tissue sarcomas. Oncotarget 2017, 8, 49. [Google Scholar] [CrossRef] [Green Version]
  190. Fu, X.; Rivera, A.; Tao, L.; Zhang, X. An HSV-2 based oncolytic virus can function as an attractant to guide migration of adoptively transferred T cells to tumor sites. Oncotarget 2014, 6, 2. [Google Scholar] [CrossRef] [Green Version]
  191. Eckert, E.C.; Nace, R.A.; Tonne, J.M.; Evgin, L.; Vile, R.G.; Russell, S.J. Generation of a Tumor-Specific Chemokine Gradient Using Oncolytic Vesicular Stomatitis Virus Encoding CXCL9. Mol. Therapy-Oncol. 2020, 16, 63–74. [Google Scholar] [CrossRef]
  192. Li, X.; Lu, M.; Yuan, M.; Ye, J.; Zhang, W.; Xu, L.; Wu, X.; Hui, B.; Yang, Y.; Wei, B.; et al. CXCL10-armed oncolytic adenovirus promotes tumor-infiltrating T-cell chemotaxis to enhance anti-PD-1 therapy. Oncoimmunology 2022, 11, 2118210. [Google Scholar] [CrossRef] [PubMed]
  193. Ji, Q.; Wu, Y.; Albers, A.; Fang, M.; Qian, X. Strategies for Advanced Oncolytic Virotherapy: Current Technology Innovations and Clinical Approaches. Pharmaceutics 2022, 14, 1811. [Google Scholar] [CrossRef] [PubMed]
  194. Champion, B.R.; Besneux, M.; Patsalidou, M.; Silva, A.; Zonca, M.; Marino, N.; Genova, G.D.; Illingworth, S.; Fedele, S.; Slater, L.; et al. Abstract 5013: NG-641: An oncolytic T-SIGn virus targeting cancer-associated fibroblasts in the stromal microenvironment of human carcinomas. Cancer Res. 2019, 79, 5013. [Google Scholar] [CrossRef]
Figure 1. Role of CXCL10 during RNA viral infections. ARDS: acute respiratory distress syndrome, CNS: central nervous system, HAV: hepatitis A virus, HCV: hepatitis C virus, HIV: human immunodeficiency virus, LCMV: lymphocytic choriomeningitis virus, MERS-CoV: middle east respiratory syndrome coronavirus, MeV: measles virus, MHV: murine hepatitis virus, RSV: respiratory syncytial virus, SARS-CoV: severe acute respiratory syndrome coronavirus, SARS-CoV-2: severe acute respiratory syndrome coronavirus 2.
Figure 1. Role of CXCL10 during RNA viral infections. ARDS: acute respiratory distress syndrome, CNS: central nervous system, HAV: hepatitis A virus, HCV: hepatitis C virus, HIV: human immunodeficiency virus, LCMV: lymphocytic choriomeningitis virus, MERS-CoV: middle east respiratory syndrome coronavirus, MeV: measles virus, MHV: murine hepatitis virus, RSV: respiratory syncytial virus, SARS-CoV: severe acute respiratory syndrome coronavirus, SARS-CoV-2: severe acute respiratory syndrome coronavirus 2.
Viruses 14 02445 g001
Figure 2. Role of CXCL10 during DNA viral infections. HBV: hepatitis B virus, HSV: herpes simplex virus, VZV: varicella-zoster virus.
Figure 2. Role of CXCL10 during DNA viral infections. HBV: hepatitis B virus, HSV: herpes simplex virus, VZV: varicella-zoster virus.
Viruses 14 02445 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

Elemam, N.M.; Talaat, I.M.; Maghazachi, A.A. CXCL10 Chemokine: A Critical Player in RNA and DNA Viral Infections. Viruses 2022, 14, 2445. https://doi.org/10.3390/v14112445

AMA Style

Elemam NM, Talaat IM, Maghazachi AA. CXCL10 Chemokine: A Critical Player in RNA and DNA Viral Infections. Viruses. 2022; 14(11):2445. https://doi.org/10.3390/v14112445

Chicago/Turabian Style

Elemam, Noha Mousaad, Iman Mamdouh Talaat, and Azzam A. Maghazachi. 2022. "CXCL10 Chemokine: A Critical Player in RNA and DNA Viral Infections" Viruses 14, no. 11: 2445. https://doi.org/10.3390/v14112445

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