Open access peer-reviewed chapter

Macrophage Polarization in Viral Infectious Diseases: Confrontation with the Reality

Written By

Perla Abou Atmeh, Soraya Mezouar and Jean-Louis Mège

Submitted: 01 April 2022 Reviewed: 27 June 2022 Published: 19 July 2022

DOI: 10.5772/intechopen.106083

From the Edited Volume

Macrophages - Celebrating 140 Years of Discovery

Edited by Vijay Kumar

Chapter metrics overview

124 Chapter Downloads

View Full Metrics

Abstract

The role of macrophages in viral infections is well documented. Their activation status also called macrophage polarization categorized by the dichotomy of M1 and M2 phenotype remained poorly investigated. Recent studies have shown the complexity of macrophage polarization in response to viral infection and the limits of its use in infected individuals. The aim of this chapter is to reappraise the concept of macrophage polarization in viral infectious diseases, which are more complicated than the models of macrophage-virus interaction. If this concept has been largely used to describe activation status of myeloid cells in experimental conditions, it has to be assessed in light of high-throughput technologies at molecular and phenotypic levels. We update knowledge on macrophage polarization in viral infectious diseases with a special attention for severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection leading to coronavirus disease (COVID-19). Hence, we propose an overview of the concept of macrophages as targets for therapeutic intervention in viral infectious disease. Finally, we tempted to focus our approach on patient investigation restricting the use of in vitro experiments and animal models to mechanistic questions.

Keywords

  • macrophages
  • myeloid cells
  • polarization
  • viral diseases

1. Introduction

First described in 1882 by Ilya Mechnikov, macrophages or “phagocytes” are distributed widely in the body where they acquire specific tissue identities and functions [1]. Macrophages are key effectors of tissue homeostasis contributing to wound healing and tissue repair. When tissue homeostasis is altered, monocytes are recruited from blood to tissues where they differentiate into macrophages. These latter are critical components of innate and adaptive immunity and contribute to inflammation and host defense.

During infection, macrophages represent the first cells on the battle front. They can act as scavengers by engulfing and destroying pathogens or altered host cells, alert the immune system through the secretion of lipid mediators, cytokines, and chemokine; or present antigens to T lymphocytes [2]. The expression of lectins, scavenger receptors, and immunoglobulin receptors enables macrophage phagocytosis, antibody-dependent cell phagocytosis (ADCP) and cytotoxicity (ADCC) [3]. They are also equipped with pattern recognition receptors (PRRs) that following their stimulation lead to the activation of transcription factors, the release of toxic mediators (reactive oxygen intermediates, proteases, inflammatory molecules) [3]. Because of their ability to promote adaptive immune response to virus via antibody release and CD8 T cell activation, macrophages can contribute to the cure of viral infections.

According to their function in pathological conditions, macrophages are considered as activated or alternatively activated also referred to as M1 and M2 polarization phenotype, respectively. The polarization of macrophages is a concept introduced to describe the features of myeloid cell activation and to classify them in functional categories (Figure 1), according to initially reported polarization of immune response into Th1 and Th2 types [4, 5, 6]. The M1 macrophages are induced by Th1 cytokines such as interferon (IFN)-γ and/or lipopolysaccharide (LPS) and present a pro-inflammatory phenotype. They are characterized by the secretion of inflammatory cytokines including tumor necrosis factor (TNF), interleukin (IL)-1α, IL-1β, IL-6, IL-12, IL-23, and also the expression of several markers including CD80, CD86, and CD68. The M2 macrophages are likely more heterogeneous: they have been classified into four categories, M2a, M2b, M2c, and M2d, depending on the stimulus [7, 8]. As described in Table 1, macrophages stimulated by IL-4 or IL-13 lead to an M2a profile associating the expression of CD206, IL-1 receptor type 2 (IL1-R2) and arginase and the secretion of transforming growth factor (TGF)-β and IL-10. M2b macrophages are induced by immune complexes, Toll-like receptor (TLR) ligands, or IL-1β. This profile is associated with the secretion of inflammatory cytokines (TNF, IL-1β, IL-6), IL-10, and the chemokine CCL1. M2c macrophages are induced by IL-10, TGF-β, or glucocorticoids; they expressed CD163 and CD206 and exhibit anti-inflammatory activity through the secretion of IL-10 and TGF-β. Finally, M2d polarization is initiated by TLRs (TLR2, TLR4, TLR7, and TLR9) or adenosine receptor ligands (A2A) with the secretion of vascular endothelial growth factor (VEGF) supporting pro-angiogenic and pro-tumoral functions [9, 10, 11]. It is becoming evident that these categories of activation states are an over-simplification of the diversity of macrophage activation modes. We have tried to reappraise the concept of macrophage polarization by associating the type of polarization with the agonist [12]. This approach allows to propose several activation profiles with LPS, IFN-γ, or their combination instead of a unique M1 activation state [13, 14]. In clinical situations, it has been shown that tumor-associated macrophages (TAMs) are clearly specialized cell populations in which polarization and functions are related: M1-like and M2-like TAMs have anti-tumoral and pro-tumoral activities, respectively [15]. In other clinical situations including infectious diseases, such functional dichotomy is rarely observed [7], and the functional role of macrophage polarization remains an exception.

Figure 1.

In vitro and in vivo macrophage polarization markers and protein secretion.

StimuliMarker expressionCytokine productionChemokine productionFunctions
M1IFN-γ, LPS, GM-CSF, TNFCD68, CD86, CD80, MHC-II, IL1R, TLR2, TLR4, iNOS, SOCS3, IL-12high/IL-10low, IL-6, TNFTNF, IL-1β, IL-6, IL-12, IL-23CCL10, CCL11, CCL5, CCl8, CCL9, CCL2, CCL3, CCL4
  • Pro-inflammation

  • Microbicidal

  • Tumor resistance

M2aIL-4, IL-13CD163, CD206, MHC-II, TGM2, IL-1R II, scavenger receptors, Arg-1IL-10, TGF-β, IL-1raCCL17, CCL22, CCL24
  • Anti-inflammatory

  • Wound healing

M2bLPS + immune complexe, IL-1 β, immune complexes, TLRsCCL1, CD86, IL-6, IL-10high/IL-12low, TNF, MHC-IIIL-1β, IL-6, IL-10, TNFCCL1
  • Immunoregulation

  • Tumor progression

  • Promoting infection

M2cGlucocorticoids, IL-10, TGF-βCD163, TLR8, TLR1, CD206IL-10, TGF-βCCR2
  • Phagocytosis

  • Immunosuppression

  • Tissue remodeling

M2dIL-6, adenosine, leukocyte inhibitory factor, TLRsVEGFIL-10, TGF-β,
TNF, IL-12
CCL5, CXCL10, CXCL16
  • Angiogenesis

  • Tumor progression

Table 1.

In vitro macrophages polarization sub-types.

IFN: interferon, LPS: polysaccharide; GM-CSF: granulocyte macrophage colony stimulating factor; TLR: Toll like receptor; MHC: major histocompatibility complex; iNOS: inducible nitric oxide; Arg: arginase; TNF: tumor necrosis factor; IL: interleukin; TGF: transforming growth factor; VEGF: vascular endothelial growth factor.

The mechanisms of macrophage polarization have been the object of a broad literature. It appears now that the metabolism of macrophages is different according to their polarization. Hence, M1 macrophages exhibit increased glycolysis and broken tricarboxylic acid (TCA) cycle, leading to the accumulation of succinate and citrate. In contrast, M2 macrophages have intact TCA cycle leading to the generation of adenosine triphosphate (ATP) [16, 17]. It is becoming evident that the polarization of macrophages is determined by transcriptional changes, as shown by using new technologies [4]. In aortic macrophages studied in vivo by scRNA-seq, Chang et al. identified three classes of macrophages: resident-like, inflammatory, and a final group with strong expression of triggering receptor expressed on myeloid cells 2 (TREM2) [18]. Interestingly, M2 markers are found in the inflammatory macrophage population, suggesting that the traditional classification of macrophage polarization does not fully reflect the diversity of the in vivo macrophage populations (Figure 1). Quantitative mass spectrometry imaging has also been proposed to investigate macrophage polarization in situ. It enabled the cartography of functional macrophage population and the visualization of their distribution in normal and pathological tissues [19, 20, 21, 22]. In addition, macrophage polarization requires dynamic and reversible epigenomic marks at enhancers and promoters of signal responsive genes [4]. The epigenetic mechanisms of M2 polarization reveal the role of histone methylation and acetylation. Hence, the overexpression of DNA methyltransferase 3B or the loss of histone deacetylase-3 (HDAC3) promotes M2 phenotype. The histone demethylase JMJD3 (lysine demethylase 6B, KDM6B) is activated by IL-4 and binds M2 genes, leading to repress M1 inflammatory program. In contrast, IFN-γ increases chromatin accessibility [4, 23]. Finally, polarization and functional responses of macrophages are influenced by differential expression of microRNAs: the literature has reported miRs specialized in M1 polarization and miRs increasing M2-like responses [24].

Is the specialization of M1/M2-like macrophages evolutionary conserved? Two ancient molecular mechanisms, inducible nitric oxide synthase 2 (iNOS2) and arginase (Arg), characterize macrophage polarization. If the ability to produce large amounts of nitric oxide in response to microbial agonists, a hallmark of M1 macrophages, has emerged with vertebrates, the Arg pathway is described in both prokaryotes and eukaryotes [25]. The cytosolic and mitochondrial Args are encoded by two genes, and it has been shown that the duplication of Arg gene occurred after the separation of vertebrates and invertebrates [26]. Hence, it is likely that macrophage polarization occurs during early vertebrate evolution [27].

The aim of this chapter is to update the knowledge about macrophage polarization in viral infections with a special focus on severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. We are deciding to analyze papers reporting infections in humans and to restrict animal models for specific questions. The analysis of the literature reveals the heterogeneous definition of macrophage polarization and the frequently inappropriate use of this concept. It is the reason why the final paragraph will describe how macrophage polarization can be studied in patients (Table 2) and to propose some recommendations for investigating infected patients.

M1 macrophageM2 macrophage
Marker expressionCD68, CD86, CD80CD68, CD86, CD80, CD163, CD206
CytokineTNF, IL-1β, IL-6, IL-12, IL-23, IL-18IL-10, TGF-β, TNF, VEGF, IL-1β
FunctionsPro-inflammatory and anti-tumoralPro- and/or anti-inflammatory

Table 2.

In vivo polarization of macrophage sub-types.

TNF: tumor necrosis factor; IL: interleukin; TGF: transforming growth factor; VEGF: vascular endothelial growth factor.

Advertisement

2. Viral infectious diseases in humans

As obligate intracellular pathogens, viruses require host cells to replicate. Innate defenses are essentials to block or inhibit cell infection, to eliminate virus from infected cells, and to alert cells the adaptive immunity. This crucial stage is regulated by myeloid cells including, dendritic cells, and mainly by macrophages. The early response is based on [1] the recognition of pathogen-associated molecular patterns (PAMPs) by PRRs, [2] the elimination of foreign agents [3] and the activation of type I and II IFNs, and interferon-stimulated gene (ISG), which have broad-spectrum antiviral activity [28, 29]. Although this defense is effective, a complete cure of the infection requires mounting adaptive immune response that is controlled to prevent immune pathogenicity. Macrophage-mediated immune response can be circumvented or hijacked by the virus to allow its replication and persistence in the host.

The first studies highlighting the interaction between myeloid cells and viruses date back to the end of 1970s [30, 31]. In a general point of view, macrophages polarize into M1 following a contact with viruses, which induces a pro-inflammatory response while the phenotype M2 is often found at late stages of infection [32]. It has been reported that virulent strains of viruses will “bias” the M1 polarization profile of macrophages into M2. In contrast, attenuated viruses lead to an M2 polarization profile M2 [33]. M1-activated macrophages play a key role in the elimination of viruses through several strategies, including the secretion of reactive species [34], secretion of antiviral cytokines [35], or activation of other immune cells such as T cells and natural killers [36, 37]. Hence, some viruses are able to counteract M1 macrophages in order to obtain an environment favorable to their replication. Indeed, some viruses are able to inhibit production of nitric oxide [38, 39, 40, 41] or pro-inflammatory cytokines [42, 43, 44], suppress antigen presentation [45, 46, 47, 48], or simply modulate the signaling pathways associated with polarization by inducing M2 phenotype [8, 49, 50, 51]. We previously addressed the role of macrophage polarization in bacterial infections [7, 52]; the aim of the present talk is to assess the role of macrophage polarization in viral infections by selecting typical viral infections.

2.1 Hepatitis viruses

Five viruses, known as hepatitis A, B (HBV), C (HCV), D, and E, are responsible for the majority of acute and chronic hepatitis. HBV infection is responsible of acute self-resolving disease in adults but not in early life. Hepatic macrophages whatever their origin are involved in host response to HBV. HBV antigen and nucleic acids are detected in monocytes and macrophages from patients with hepatitis. The interaction of HBV with macrophages including hepatic macrophages may affect macrophage polarization by suppressing M1 polarization and promoting M2 polarization [53]. M1-associated cytokines are likely protective as shown by higher risk of HBV reactivation and hepatotoxicity in patients with anti-TNF treatment given for inflammatory diseases [54]. During the progression of HBV-related disease (from mild chronic hepatitis B to decompensated cirrhosis), M2-type monocytes expressing CD163 and CD206 are increased, whereas the frequency of cells expressing M1 markers decreases. Monocytes and Kupffer cells expressing an M2 profile are predicting a poor clinical outcome [55]. The non-protective effect of M2-type polarization is illustrated by the observation that IL-10 gene promoter polymorphisms are associated with HBV progression [44]. The measurement of soluble CD163 and soluble mannose receptor may be a pertinent approach of follow-up of patients with chronic hepatitis. Both markers are released during liver damage and are associated with M2 polarization and fibrosis; their levels are reduced after antiviral treatment [56]. The relationship between chronic evolution of hepatitis B, fibrosis, and infiltration of liver by M2-like macrophages has been demonstrated in a humanized mouse model of infection [57].

HCV is transmitted between adults and is responsible for a high percentage of chronic infections with a major risk of liver cirrhosis and hepatocellular carcinoma [58]. The co-culture of monocyte-derived macrophages with HCV-infected hepatocytes induces M2 surface markers. TGF-β produced by these polarized macrophages activates hepatic stellate cells, leading to fibrosis. However, monocytes and macrophages do not seem completely polarized at the cytokine level [59]. Cell-free virus or exosome-packaged HCV induces the differentiation of monocytes into macrophages with M2 phenotype and non-polarized cytokine production under the control of TLR7/8. Interestingly, TLR7/8 is overexpressed in pro-fibrotic monocytes from chronic HCV-infected patients [60]. It has been also shown that HCV core protein inhibits phagocytosis activity of M1 and M2 macrophages and CD4+ T cell activation induced by M1 macrophages but promotes that induced by M2 macrophages [61]. M1 and M2 macrophages generated from chronic HCV patients lose their phenotypic characteristics, suggesting that chronic HCV infection is rather associated with an impaired polarization than a reprogramming of macrophages [62]. Finally, in biopsies of HCV-infected patients, it has been shown that non-infected cells such as Kupffer cells are a source of IFNs, demonstrating the interplay between hepatic cells [63]. Taken together, these data suggest that HBV and HCV share the ability to interfere with M1-type macrophage polarization, thus accounting for viral persistence, hepatic fibrosis, and evolution to carcinoma.

2.2 Human immunodeficiency virus (HIV)

The permissivity of monocytes and macrophages to HIV depends on their differentiation stage, polarization status, and tissue location [64]. The M1/M2 polarization of macrophages impacts the steps of viral cycle including entry, reverse transcription, transcription, and posttranscription. As the level of inflammatory cytokines is high in the early stage of HIV-1 infection [65, 66], it is likely that M1-like macrophages play a role in HIV infection. The entry of R5 and R5/X4 HIV in M1-like macrophages is decreased because of cytokine-mediated downregulation of CD4 and CCR5. In contrast, M1 cytokines such as TNF increase viral transcription. In M2 macrophages elicited by IL-4/IL-13 and/or IL-10-mediated deactivated macrophages, both entry and replication of HIV-1 are decreased [64]. The implication of TNF in M1 polarization of HIV-infected macrophages is debated [67]. Hence, it is likely that M1 macrophages enable the formation of viral reservoirs early in the disease. At later stages, an M2 shift of macrophages is observed. At the onset of acquired immunodeficiency syndrome (AIDS), deactivated macrophages predominate via enhanced clearance of apoptotic cells, which is known to promote M2-like macrophages [68]. Severe evolution of HIV infection is associated with elevated IL-10 levels, but not IL-4 levels, suggesting that AIDS is characterized by IL-10-mediated M2-like phenotype [69]. These findings have been confirmed in acute and chronic HIV and SIV infections. Hence, at the beginning of the infection, the central nervous system, heart, and blood vessels exhibit M1-like macrophages, whereas M2-like macrophages are observed in later responses [32]. It is likely that CD163+ M2 macrophages play a protective role in SIV-infected macaques through their anti-inflammatory functions [70]. It has been proposed that the persistence of initial activation in patients with chronic infection and successful antiviral therapy is correlated to non-AIDS complications such neurocognitive disorder and cardiovascular dysfunctions [71]. Unfortunately, few markers have been investigated in these studies, and it is likely that more precise data will be necessary to reanalyze the polarization of macrophages in HIV infection [71, 72].

2.3 Flavivirus

Flavivirus is responsible for infections essentially dominated by dengue virus (DENV) and Zika virus (ZIKV). First, DENV infection presents a large spectrum of clinical presentations from moderate symptoms to classical dengue and hemorrhagic dengue. Monocytes and macrophages are involved in the infection pathogenesis. Macrophage-colony-stimulating factor (M-CSF)-differentiated macrophages (M2-like macrophages) are poorly sensitive to DENV infection. In contrast, granulocyte macrophage CSF (GM-CSF)-differentiated macrophages (M1-like macrophages) are highly susceptible to DENV infection with high release of cytokines and activation of NLRP3 inflammasome [73]. Nevertheless, some biomarkers such as soluble CD163, known to be associated with M2 polarization of macrophages, seem predictive of severe dengue [74]. In pediatric dengue patients compared with healthy individuals, the number of M2-like macrophages is increased with decreased number of M1-like macrophages. In dengue patients with bleeding trend, both macrophage subsets are decreased and are associated with decreased platelet count [48]. Second, ZIKV is associated with numerous cases of microcephaly and/or central nervous system malformations. ZIKV infects myeloid cells and has a tropism for placenta including maternal and fetal tissues. It has been shown that ZIKV replicates in both placenta macrophages, also named Hofbauer cells, and trophoblasts [75]. Two lineages of ZIKV, African and Asian, have been described, and it has been shown that they exhibit differences in pathogenicity despite close sequence homology [76, 77]. While the Asian strain of ZIKV elicits an expansion of non-classical monocytes from healthy donors and M2-skewed immunosuppressive program, African strain promotes a M1 program [78].

2.4 Cytomegalovirus (CMV)

Human CMV (hCMV) uses TLR2 and intracytosoplasmic sensors to invade monocytes and macrophages [79]. In monocytes, hCMV stimulates a transcriptomic program in which M1 genes are enriched [80]. On the other hand, a product of hCMV genome, UL111A gene, encodes functional homologs of human IL-10 during both productive and latent phases of CMV infection [81]. In CD14+ monocytes, the viral IL-10 induces M2c phenotype associating increased expression of CD163 and CD14 and downregulation of HLA-DR. The viral IL-10 also upregulates heme oxygenase 1 (HO-1), a driver of phenotype shift to M2 macrophages [82] known to also down-modulate M1-associated cytokines and poorly stimulate CD4+ T cells [83]. We hypothesize that CMV triggers an M1 program in monocytes and that the release of viral or human IL-10 leads bystander monocytes to be reprogrammed toward an M2 phenotype. The polarization of monocytes in response to hCMV is likely necessary for their differentiation into macrophages. Recently, it has been shown that hCMV stimulates the expression of M1 and M2 markers in monocytes and activates PI3K-Akt axis, leading to caspase 3 activation [84]. CMV susceptibility is dependent on polarization of myeloid cells. M1-like macrophages are more resistant to CMV than M2-like macrophages, and it is likely that this resistance is related to the ability of M1-like macrophages to induce IFN-γ production by natural killer (NK) cells [85]. This hypothesis is supported by two other studies. Although hCMV susceptibility is higher in M2-like macrophages, productive and persistent viral infection is observed in both M1- and M2-like macrophages. Infected M1- and M2-like macrophages are efficient in stimulating proliferation of autologous T cells from hCMV-seropositive donors [86]. The susceptibility of M2 macrophages is optimal in the early phase of hCMV infection, whereas, in the late phase, macrophage activation necessary for viral replication is dependent on the activation of mammalian target of rapamycin (mTORC)1 complex, as confirmed with experiments including rapamycin [87].

2.5 Influenza virus

Four influenza virus genera (A, B, C, and D) belonging to orthomyxoviridae are responsible for flu, a seasonal respiratory epidemics, or pandemics, such as the “Great Influenza” pandemics of 1918 [88]. The severity of influenza pneumonia depends on host susceptibility and strain diversity and can lead to acute respiratory distress syndrome and lethality. These latter complications are associated with uncontrolled inflammatory response. Numerous evidences show that macrophages including alveolar macrophages are involved in the pathophysiology of influenza virus infections via a direct viral infection or the overproduction of cytokines. This is emphasized by the observation that over-pathogenic strains of influenza virus productively infect monocytes [89]. Animal models demonstrate that macrophage reprogramming is critical in outcome of influenza virus infections. Hence, GM-CSF protects from mortality and morbidity and redirects responses of alveolar macrophages from M1-like to M-2-like activation. This finding was unexpected because GM-CSF is known as an M1 inducer and depresses arginase, a canonical marker of M2-like status [90]. The inactivation of NOS (nitric oxide synthase) 2 and IFN-γ favors M2 reprogramming and improves outcome of viral infection [91]. In contrast, the treatment of macrophages with baicalin that possesses antiviral properties stimulated an M1 phenotype shift associated with activation of IFN pathway and inhibition of influenza virus replication [92].

The evidences of macrophage polarization in humans infected with influenza virus are scarce. In one study of patients from the 2009 to 2010 pandemics, monocytes have been reported as a marker of severity independently of viral load [93]. Monocytes from patients with severe infection exhibit increased expression of M1 markers and TNF production and a down-modulation of CD163, an M2 marker. Murine models of influenza virus infection also show high proportion of recruited M1 monocytes and decreased number of resident M2 alveolar macrophages, confirming that the severity of influenza virus infection is associated with macrophage reprogramming toward an M1 phenotype [93].

2.6 Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)

SARS-CoV-2, a strain of the coronavirus family, causes coronavirus disease-2019 (COVID-19) characterized in the most severe cases by an increased production of cytokines including IL-1α, IL-1β, IL-6, IL-7, TNF, type I and II IFN, CCL2, CCL3, and CXCL10 [94, 95]. This cytokine storm was initially observed in influenza syndrome that occurs after systemic infection and immunotherapy [96] before it was extended to describe immune response in COVID-19 patients [97]. To better understand the pathophysiology of this emergent disease, the researchers focused on the response of myeloid cells such as macrophages or dendritic cells. Macrophages are permissive to SARS-CoV-2 infection but no viral replication is observed in vitro [98, 99]. Interestingly, the production of inflammatory cytokines and chemokines is observed after SARS-CoV-2 infection of macrophages but not after infection of monocyte-derived dendritic cells [98, 100]. The release of TNF, IL-1β, IL-10, IFN-α/β, and IL-6 by infected-macrophages leads to type I IFN-immune response, suggesting a protective role against viral infection [100].

The polarization of macrophages in COVID-19 has also been investigated. Alveolar macrophages respond differently to infection depending on their polarization status. After SARS-CoV-2 infection, M1 macrophages are associated with viral spreading, whereas M2 polarization induces virus degradation and infection limitation. Indeed, macrophages from human ACE2 transgenic mice present an increased infection rate after in vitro treatment by IFN-γ or LPS compared with IL-4 treatment [101]. These results are controversial because the SARS-CoV-2 infection of M1 and M2 macrophages from the THP-1 cell line is similar [99]. The gene expression study of lung alveoli from COVID-19 patients reveals different macrophage patterns depending on their polarization profile. The gene expression study of lung alveoli from COVID-19 patients shows that highly inflammatory macrophages are mostly found in patients with severe COVID-19 [102, 103]. Thus, the polarization of macrophages during infection by SARS-CoV-2 is suggested as determining the severity of the disease, even if involved molecular mechanisms remain unexplored to date.

Advertisement

3. Macrophage polarization and treatment of viral infections

The role of deregulated immune response in pathogenesis of viral infections such as COVID-19 pandemics justifies the use of drugs that target host response and exhibit anti-infective properties. Hence, chloroquine and hydroxychloroquine are known for their antiviral and immunomodulatory effects, which lead to propose these molecules in the treatment of SARS-Cov2 infection [104]. Beyond the debate about the efficiency of chloroquine and hydroxychloroquine, their immunomodulatory properties affect the macrophage polarization. It is established that treatment with chloroquine can reverse the polarization of TAMs from M2 to M1 phenotype in tumor models [105]. Similarly, chloroquine and hydroxychloroquine interfere with LPS-mediated M1 polarization of macrophages [106]. The combination of hydroxychloroquine and azithromycin is interesting since this latter molecule is known to induce M2 macrophage polarization [106, 107]. Ivermectin, a macrocyclic lactone known for its antiparasitic effect, has an anti-inflammatory effect promoting M2 polarization of macrophages without effect on viral load; it has been proposed to limit the inflammation of respiratory tract and to improve COVID-19 outcome [108]. Remdesivir, an adenosine analog, reduces inflammatory gene expression and has been largely used in COVID-19 treatment [109].

As mentioned above, M1 polarization of macrophages is a determining phenotype against viral infections. The artificial induction of macrophage M1 polarization may be an interesting adjuvant to antiviral treatment for non-COVID19 infectious diseases. Baicaline has been proposed to limit influenza virus infection via the M1 polarization of macrophages, thus activating their antiviral function via the IFN signaling pathway [92, 110]. It is important to note that viral infections can lead to hyperactivation of macrophages, leading to an excessive inflammatory response known as macrophage activation syndrome. In this context, anti-inflammatory molecules such as tofacitinib, anti-IL1R, or IL-6R have been clinically tested, particularly for COVID-19 [111, 112].

Advertisement

4. Conclusion and perspectives

As the first line of defense, macrophages represent key immune cells against viral infections. In general, the antiviral response is mediated by a pro-inflammatory response of polarized M1 macrophages. Some viruses are able to counteract the antiviral response of macrophages by modulating their polarization by switching the M1 phenotype to M2 phenotype. It should also be noted that viruses such as influenza and SARS-CoV-2 are capable of modulating an M1 over-polarization of macrophages responsible for severe diseases. Interestingly, modulation of macrophage polarization has been investigated as a therapeutic strategy. However, the in vivo polarization profile remains more intricate compared with in vitro situations. This can be explained by the techniques classically used for investigating macrophage polarization (gene or protein expression) whose limitation is that the observed signals result from a mixture of diverse cells. Hopes for quantitative mass spectrometry imaging as a tissue-level investigative tool remain unanswered to date. Thus, clarifying the tools for investigating macrophage polarization in clinical settings and the associated molecular mechanisms are key steps in the development of therapies in viral infections.

Advertisement

Acknowledgments

Soraya Mezouar was supported by a “Fondation pour la Recherche Médicale” postdoctoral fellowship (reference: SPF20151234951). This work was supported by the French Government under the “Investissements d’avenir” (Investments for the future) program managed by the “Agence Nationale de la Recherche” (reference: 10-IAHU-03).

Advertisement

Author contributions

P.A.A, S.M., and J.L.M. conceived and wrote the manuscript.

Advertisement

Declaration of interest

The authors declare no competing interests.

References

  1. 1. Lavin Y, Mortha A, Rahman A, Merad M. Regulation of macrophage development and function in peripheral tissues. Nature Reviews. Immunology. 2015;15(12):731-744
  2. 2. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nature Reviews. Immunology. 2011;11(11):723-737
  3. 3. Kumar V. Phagocytosis: Phenotypically simple yet a mechanistically complex process: Phagocytosis is a very complex but crucial process playing a pivotal role in embryonic development and host defense to maintain immune homeostasis. International Reviews of Immunology. 2020;39(3):118-150
  4. 4. Locati M, Curtale G, Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annual Review of Pathology. 2020;15:123-147
  5. 5. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: Time for reassessment. F1000prime Reports. 2014;6:13
  6. 6. Mills CD, Ley K. M1 and M2 macrophages: The chicken and the egg of immunity. Journal of Innate Immunity. 2014;6(6):716-726
  7. 7. Benoit M, Desnues B, Mege JL. Macrophage polarization in bacterial infections. Journal of Immunology. 2008;181(6):3733-3739
  8. 8. Wang L, Zhang S, Wu H, Rong X, Guo J. M2b macrophage polarization and its roles in diseases. Journal of Leukocyte Biology. 2019;106(2):345-358
  9. 9. Nikovics K, Favier AL. Macrophage identification in situ. Biomedicines. 2021;9(10):1393
  10. 10. Pinhal-Enfield G, Ramanathan M, Hasko G, Vogel SN, Salzman AL, Boons GJ, et al. An Angiogenic switch in macrophages involving synergy between toll-like receptors 2, 4, 7, and 9 and adenosine A2A receptors. The American Journal of Pathology. 2003;163(2):711-721
  11. 11. Ferrante CJ, Pinhal-Enfield G, Elson G, Cronstein BN, Hasko G, Outram S, et al. The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Rα) signaling. Inflammation. 2013;36(4):921-931
  12. 12. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity. 2014;41(1):14-20
  13. 13. Murray PJ. Macrophage polarization. Annual Review of Physiology. 2017;79(1):541-566
  14. 14. Ouedraogo R, Daumas A, Capo C, Mege JL, Textoris J. Whole-cell MALDI-TOF mass spectrometry is an accurate and rapid method to analyze different modes of macrophage activation. Journal of Visualized Experiments. 2013;82:50926
  15. 15. Wu K, Lin K, Li X, Yuan X, Xu P, Ni P, et al. Redefining tumor-associated macrophage subpopulations and functions in the tumor microenvironment. Frontiers in Immunology. 2020;11:1731
  16. 16. O’Neill LAJ. A broken Krebs cycle in macrophages. Immunity. 2015;42(3):393-394
  17. 17. O’Neill LAJ, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nature Reviews. Immunology. 2016;16(9):553-565
  18. 18. Chang Z, Wang Y, Liu C, Smith W, Kong L. Natural products for regulating macrophages M2 polarization. Current Stem Cell Research & Therapy. 2020;15(7):559-569
  19. 19. Unsihuay D, Mesa Sanchez D, Laskin J. Quantitative mass spectrometry imaging of biological systems. Annual Review of Physical Chemistry. 2021;72:307-329
  20. 20. Holzlechner M, Strasser K, Zareva E, Steinhäuser L, Birnleitner H, Beer A, et al. In situ characterization of tissue-resident immune cells by MALDI mass spectrometry imaging. Journal of Proteome Research. 2017;16(1):65-76
  21. 21. Roussel M, Bartkowiak T, Irish JM. Picturing polarized myeloid phagocytes and regulatory cells by mass cytometry. Methods in Molecular Biology (Clifton, N.J.). 2019;1989:217-226
  22. 22. Porta Siegel T, Hamm G, Bunch J, Cappell J, Fletcher JS, Schwamborn K. Mass spectrometry imaging and integration with other imaging modalities for greater molecular understanding of biological tissues. Molecular Imaging and Biology. 2018;20(6):888-901
  23. 23. Foster SL, Hargreaves DC, Medzhitov R. Gene-specific control of inflammation by TLR-induced chromatin modifications. Nature. 2007;447(7147):972-978
  24. 24. Zhong Y, Yi C. MicroRNA-720 suppresses M2 macrophage polarization by targeting GATA3. Bioscience Reports. 2016;36(4):e00363
  25. 25. Edholm ES, Rhoo KH, Robert J. Evolutionary aspects of macrophages polarization. Results and Problems in Cell Differentiation. 2017;62:3-22
  26. 26. Dzik JM. Evolutionary roots of arginase expression and regulation. Frontiers in Immunology. 2014;5:544
  27. 27. Hodgkinson J, Grayfer L, Belosevic M. Biology of bony fish macrophages. Biology. 2015;4(4):881-906
  28. 28. Thompson MR, Kaminski JJ, Kurt-Jones EA, Fitzgerald KA. Pattern recognition receptors and the innate immune response to viral infection. Viruses. 2011;3(6):920-940
  29. 29. Feng J, Wickenhagen A, Turnbull ML, Rezelj VV, Kreher F, Tilston-Lunel NL, et al. Interferon-stimulated gene (ISG)-expression screening reveals the specific antibunyaviral activity of ISG20. Jung JU, editor. Journal of Virology. 2018;92(13):e02140-e02117
  30. 30. Wardley RC, Wilkinson PJ. The growth of virulent African swine fever virus in pig monocytes and macrophages. The Journal of General Virology. 1978;38(1):183-186
  31. 31. Gazzolo L, Moscovici C, Moscovici MG. Persistence of avian oncoviruses in chicken macrophages. Infection and Immunity. 1979;23(2):294-297
  32. 32. Burdo TH, Walker J, Williams KC. Macrophage polarization in AIDS: Dynamic Interface between anti-viral and anti-inflammatory macrophages during acute and chronic infection. Journal of Clinical and Cellular Immunology. 2015;6(3):333
  33. 33. Ferrer MF, Thomas P, López Ortiz AO, Errasti AE, Charo N, Romanowski V, et al. Junin virus triggers macrophage activation and modulates polarization according to viral strain pathogenicity. Frontiers in Immunology. 2019;10:2499
  34. 34. Camini FC, da Silva Caetano CC, Almeida LT, de Brito Magalhães CL. Implications of oxidative stress on viral pathogenesis. Archives of Virology. 2017;162(4):907-917
  35. 35. Arango Duque G, Descoteaux A. Macrophage cytokines: Involvement in immunity and infectious diseases. Frontiers in Immunology. 2014;5(491):491. Available from: http://journal.frontiersin.org/article/10.3389/fimmu.2014.00491/abstract
  36. 36. Guerriero JL. Macrophages. In: International Review of Cell and Molecular Biology [Internet]. Elsevier; 2019. pp. 73-93. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1937644818300698
  37. 37. Zwirner NW, Domaica CI, Fuertes MB. Regulatory functions of NK cells during infections and cancer. Journal of Leukocyte Biology. 2021;109(1):185-194
  38. 38. Kao YJ, Piedra PA, Larsen GL, Colasurdo GN. Induction and regulation of nitric oxide synthase in airway epithelial cells by respiratory syncytial virus. American Journal of Respiratory and Critical Care Medicine. 2001;163(2):532-539
  39. 39. Santiago-Olivares C, Rivera-Toledo E, Gómez B. Nitric oxide production is downregulated during respiratory syncytial virus persistence by constitutive expression of arginase 1. Archives of Virology. 2019;164(9):2231-2241
  40. 40. Granja AG, Sabina P, Salas ML, Fresno M, Revilla Y. Regulation of inducible nitric oxide synthase expression by viral A238L-mediated inhibition of p65/RelA acetylation and p300 transactivation. Journal of Virology. 2006;80(21):10487-10496
  41. 41. Odkhuu E, Komatsu T, Naiki Y, Koide N, Yokochi T. Sendai virus C protein inhibits lipopolysaccharide-induced nitric oxide production through impairing interferon-β signaling. International Immunopharmacology. 2014;23(1):267-272
  42. 42. Bradley JH, Harrison A, Corey A, Gentry N, Gregg RK. Ebola virus secreted glycoprotein decreases the anti-viral immunity of macrophages in early inflammatory responses. Cellular Immunology. 2018;324:24-32
  43. 43. Zhu W, Banadyga L, Emeterio K, Wong G, Qiu X. The roles of Ebola virus soluble glycoprotein in replication, pathogenesis, and countermeasure development. Viruses. 2019;11(11):999
  44. 44. McElroy AK, Nichol ST. Rift Valley fever virus inhibits a pro-inflammatory response in experimentally infected human monocyte derived macrophages and a pro-inflammatory cytokine response may be associated with patient survival during natural infection. Virology. 2012;422(1):6-12
  45. 45. Hurtado C, Bustos MJ, Granja AG, de León P, Sabina P, López-Viñas E, et al. The African swine fever virus lectin EP153R modulates the surface membrane expression of MHC class I antigens. Archives of Virology. 2011;156(2):219-234
  46. 46. Rehm KE, Connor RF, Jones GJB, Yimbu K, Roper RL. Vaccinia virus A35R inhibits MHC class II antigen presentation. Virology. 2010;397(1):176-186
  47. 47. Rehm KE, Connor RF, Jones GJB, Yimbu K, Mannie MD, Roper RL. Vaccinia virus decreases major histocompatibility complex (MHC) class II antigen presentation, T-cell priming, and peptide association with MHC class II. Immunology. 2009;128(3):381-392
  48. 48. Schaefer MR, Wonderlich ER, Roeth JF, Leonard JA, Collins KL. HIV-1 Nef targets MHC-I and CD4 for degradation via a final common β-COP–dependent pathway in T cells. Hope TJ, editor. PLoS Pathogens. 2008;4(8):e1000131
  49. 49. Wilson HM. SOCS proteins in macrophage polarization and function. Frontiers in Immunology. 2014;5:357. Available from: http://journal.frontiersin.org/article/10.3389/fimmu.2014.00357/abstract
  50. 50. Kwon Y, Meyer K, Peng G, Chatterjee S, Hoft DF, Ray R. Hepatitis C virus E2 envelope glycoprotein induces an Immunoregulatory phenotype in macrophages. Hepatology. 2019;69(5):1873-1884
  51. 51. Ouyang P, Rakus K, van Beurden SJ, Westphal AH, Davison AJ, Gatherer D, et al. IL-10 encoded by viruses: A remarkable example of independent acquisition of a cellular gene by viruses and its subsequent evolution in the viral genome. The Journal of General Virology. 2014;95(2):245-262
  52. 52. Mezouar S, Mege JL. New tools for studying macrophage polarization: Application to bacterial infections. In: Prakash H, editor. Macrophages [Internet]. London, UK: IntechOpen; 2021. Available from: https://www.intechopen.com/books/macrophages/new-tools-for-studying-macrophage-polarization-application-to-bacterial-infections
  53. 53. Faure-Dupuy S, Delphin M, Aillot L, Dimier L, Lebossé F, Fresquet J, et al. Hepatitis B virus-induced modulation of liver macrophage function promotes hepatocyte infection. Journal of Hepatology. 2019;71(6):1086-1098
  54. 54. Temel T, Cansu DÜ, Korkmaz C, Kaşifoğlu T, Özakyol A. The long-term effects of anti-TNF-α agents on patients with chronic viral hepatitis C and B infections. International Journal of Rheumatic Diseases. 2015;18(1):40-45
  55. 55. Liang J, Long Z, Zhang Y, Wang J, Chen X, Liu X, et al. Chloride intercellular channel 3 (CLIC-3) suppression-mediated macrophage polarization: A potential indicator of poor prognosis of hepatitis B virus-related acute-on-chronic liver failure. Immunology and Cell Biology. 2022:imcb.12542
  56. 56. Laursen TL, Wong GLH, Kazankov K, Sandahl T, Møller HJ, Hamilton-Dutoit S, et al. Soluble CD163 and mannose receptor associate with chronic hepatitis B activity and fibrosis and decline with treatment: sCD163 and sMR in chronic hepatitis B. Journal of Gastroenterology and Hepatology. 2018;33(2):484-491
  57. 57. Bility MT, Cheng L, Zhang Z, Luan Y, Li F, Chi L, et al. Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: Induction of human-specific liver fibrosis and M2-like macrophages. Walker CM, editor. PLoS Pathogens. 2014;10(3):e1004032
  58. 58. Scheel TKH, Rice CM. Understanding the hepatitis C virus life cycle paves the way for highly effective therapies. Nature Medicine. 2013;19(7):837-849
  59. 59. Saha B, Kodys K, Szabo G. Hepatitis C virus-induced monocyte differentiation into polarized M2 macrophages promotes stellate cell activation via TGF-β. Cellular and Molecular Gastroenterology and Hepatology. 2016;2(3):302-316.e8
  60. 60. Saha B, Kodys K, Adejumo A, Szabo G. Circulating and exosome-packaged hepatitis C single-stranded RNA induce monocyte differentiation via TLR7/8 to polarized macrophages and fibrocytes. Journal of immunology (Baltimore, Md. : 1950). 2017;198(5):1974-1984
  61. 61. Hou Z, Zhang J, Han Q, Su C, Qu J, Xu D, et al. Hepatitis B virus inhibits intrinsic RIG-I and RIG-G immune signaling via inducing miR146a. Scientific Reports. 2016;6(1):26150
  62. 62. Ahmed F, Ibrahim A, Cooper CL, Kumar A, Crawley AM. Chronic hepatitis C virus infection impairs M1 macrophage differentiation and contributes to CD8+ T-cell dysfunction. Cell. 2019;8(4):E374
  63. 63. Park SH, Rehermann B. Immune responses to HCV and other hepatitis viruses. Immunity. 2014;40(1):13-24
  64. 64. Herbein G, Varin A. The macrophage in HIV-1 infection: From activation to deactivation? Retrovirology. 2010;7(1):33
  65. 65. Lafeuillade A, Tamalet C, Pellegrino P, Tourres C, Yahi N, Vignoli C, et al. High viral burden in lymph nodes during early stages of HIV-1 infection. AIDS (London, England). 1993;7(11):1527-1528
  66. 66. Emilie D, Peuchmaur M, Maillot MC, Crevon MC, Brousse N, Delfraissy JF, et al. Production of interleukins in human immunodeficiency virus-1-replicating lymph nodes. The Journal of Clinical Investigation. 1990;86(1):148-159
  67. 67. Porcheray F, Samah B, Léone C, Dereuddre-Bosquet N, Gras G. Macrophage activation and human immunodeficiency virus infection: HIV replication directs macrophages towards a pro-inflammatory phenotype while previous activation modulates macrophage susceptibility to infection and viral production. Virology. 2006;349(1):112-120
  68. 68. Ogden CA, Pound JD, Batth BK, Owens S, Johannessen I, Wood K, et al. Enhanced apoptotic cell clearance capacity and B cell survival factor production by IL-10-activated macrophages: Implications for Burkitt’s lymphoma. Journal of Immunology (Baltimore, Md.: 1950). 2005;174(5):3015-3023
  69. 69. Sandanger Ø, Ryan L, Bohnhorst J, Iversen AC, Husebye H, Halaas Ø, et al. IL-10 enhances MD-2 and CD14 expression in monocytes and the proteins are increased and correlated in HIV-infected patients. Journal of Immunology (Baltimore, Md.: 1950). 2009;182(1):588-595
  70. 70. Yearley JH, Pearson C, Shannon RP, Mansfield KG. Phenotypic variation in myocardial macrophage populations suggests a role for macrophage activation in SIV-associated cardiac disease. AIDS Research and Human Retroviruses. 2007;23(4):515-524
  71. 71. Akiyama H, Jalloh S, Park S, Lei M, Mostoslavsky G, Gummuluru S. Expression of HIV-1 intron-containing RNA in microglia induces inflammatory responses. Journal of Virology. 2020;95(5):e01386-20
  72. 72. Lv T, Cao W, Li T. HIV-related immune activation and inflammation: Current understanding and strategies. Journal of Immunology Research. 2021;2021:7316456
  73. 73. Wu MF, Chen ST, Yang AH, Lin WW, Lin YL, Chen NJ, et al. CLEC5A is critical for dengue virus-induced inflammasome activation in human macrophages. Blood. 2013;121(1):95-106
  74. 74. Rahman K, Desai C, Iyer SS, Thorn NE, Kumar P, Liu Y, et al. Loss of junctional adhesion molecule a promotes severe steatohepatitis in mice on a diet high in saturated fat, fructose, and cholesterol. Gastroenterology. 2016;151(4):733-746.e12
  75. 75. Quicke KM, Bowen JR, Johnson EL, McDonald CE, Ma H, O’Neal JT, et al. Zika virus infects human placental macrophages. Cell Host & Microbe. 2016;20(1):83-90
  76. 76. Wang L, Valderramos SG, Wu A, Ouyang S, Li C, Brasil P, et al. From mosquitos to humans: Genetic evolution of Zika virus. Cell Host & Microbe. 2016;19(5):561-565
  77. 77. Miner JJ, Diamond MS. Zika virus pathogenesis and tissue tropism. Cell Host & Microbe. 2017;21(2):134-142
  78. 78. Foo SS, Chen W, Chan Y, Bowman JW, Chang LC, Choi Y, et al. Asian Zika virus strains target CD14+ blood monocytes and induce M2-skewed immunosuppression during pregnancy. Nature Microbiology. 2017;2(11):1558-1570
  79. 79. Baasch S, Ruzsics Z, Henneke P. Cytomegaloviruses and macrophages-friends and foes from early on? Frontiers in Immunology. 2020;11:793
  80. 80. Chan G, Bivins-Smith ER, Smith MS, Smith PM, Yurochko AD. Transcriptome analysis reveals human cytomegalovirus reprograms monocyte differentiation toward an M1 macrophage. Journal of Immunology (Baltimore, Md.: 1950). 2008;181(1):698-711
  81. 81. Jenkins C, Abendroth A, Slobedman B. A novel viral transcript with homology to human interleukin-10 is expressed during latent human cytomegalovirus infection. Journal of Virology. 2004;78(3):1440-1447
  82. 82. Naito Y, Takagi T, Higashimura Y. Heme oxygenase-1 and anti-inflammatory M2 macrophages. Archives of Biochemistry and Biophysics. 2014;564:83-88
  83. 83. Avdic S, Cao JZ, McSharry BP, Clancy LE, Brown R, Steain M, et al. Human cytomegalovirus interleukin-10 polarizes monocytes toward a deactivated M2c phenotype to repress host immune responses. Journal of Virology. 2013;87(18):10273-10282
  84. 84. Cojohari O, Mahmud J, Altman AM, Peppenelli MA, Miller MJ, Chan GC. Human cytomegalovirus mediates unique monocyte-to-macrophage differentiation through the PI3K/SHIP1/Akt signaling network. Viruses. 2020;12(6):E652
  85. 85. Romo N, Magri G, Muntasell A, Heredia G, Baía D, Angulo A, et al. Natural killer cell-mediated response to human cytomegalovirus-infected macrophages is modulated by their functional polarization. Journal of Leukocyte Biology. 2011;90(4):717-726
  86. 86. Bayer C, Varani S, Wang L, Walther P, Zhou S, Straschewski S, et al. Human cytomegalovirus infection of M1 and M2 macrophages triggers inflammation and autologous T-cell proliferation. Journal of Virology. 2013;87(1):67-79
  87. 87. Poglitsch M, Weichhart T, Hecking M, Werzowa J, Katholnig K, Antlanger M, et al. CMV late phase-induced mTOR activation is essential for efficient virus replication in polarized human macrophages. American Journal of Transplantation: Official Journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2012;12(6):1458-1468
  88. 88. Vemula S, Zhao J, Liu J, Wang X, Biswas S, Hewlett I. Current approaches for diagnosis of influenza virus infections in humans. Viruses. 2016;8(4):96
  89. 89. Roberts JMK, Simbiken N, Dale C, Armstrong J, Anderson DL. Tolerance of honey bees to Varroa mite in the absence of deformed wing virus. Viruses. 2020;12(5):575
  90. 90. Halstead ES, Umstead TM, Davies ML, Kawasawa YI, Silveyra P, Howyrlak J, et al. GM-CSF overexpression after influenza a virus infection prevents mortality and moderates M1-like airway monocyte/macrophage polarization. Respiratory Research. 2018;19(1):3
  91. 91. Sun K, Metzger DW. Inhibition of pulmonary antibacterial defense by interferon-γ during recovery from influenza infection. Nature Medicine. 2008;14(5):558-564
  92. 92. Geng P, Zhu H, Zhou W, Su C, Chen M, Huang C, et al. Baicalin inhibits influenza a virus infection via promotion of M1 macrophage polarization. Frontiers in Pharmacology. 2020;11:01298
  93. 93. Cole SL, Dunning J, Kok WL, Benam KH, Benlahrech A, Repapi E, et al. M1-like monocytes are a major immunological determinant of severity in previously healthy adults with life-threatening influenza. JCI Insight. 2017;2(7):e91868
  94. 94. Lucas C, Wong P, Klein J, Castro TBR, Silva J, Sundaram M, et al. Longitudinal analyses reveal immunological misfiring in severe COVID-19. Nature. 2020;584(7821):463-469
  95. 95. Arunachalam PS, Wimmers F, Mok CKP, Perera RAPM, Scott M, Hagan T, et al. Systems biological assessment of immunity to mild versus severe COVID-19 infection in humans. Science. 2020;369(6508):1210-1220
  96. 96. Fajgenbaum DC, June CH. Cytokine Storm. The New England Journal of Medicine. 2020;383(23):2255-2273
  97. 97. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, et al. COVID-19: Consider cytokine storm syndromes and immunosuppression. Lancet (London, England). 2020;395(10229):1033-1034
  98. 98. Yang D, Chu H, Hou Y, Chai Y, Shuai H, Lee ACY, et al. Attenuated interferon and proinflammatory response in SARS-CoV-2-infected human dendritic cells is associated with viral antagonism of STAT1 phosphorylation. The Journal of Infectious Diseases. 2020;222(5):734-745
  99. 99. Boumaza A, Gay L, Mezouar S, Bestion E, Diallo AB, Michel M, et al. Monocytes and macrophages, targets of severe acute respiratory syndrome coronavirus 2: The clue for coronavirus disease 2019 immunoparalysis. The Journal of Infectious Diseases. 2021;224(3):395-406
  100. 100. Zheng J, Wang Y, Li K, Meyerholz DK, Allamargot C, Perlman S. Severe acute respiratory syndrome coronavirus 2-induced immune activation and death of monocyte-derived human macrophages and dendritic cells. The Journal of Infectious Diseases. 2021;223(5):785-795
  101. 101. Lv J, Wang Z, Qu Y, Zhu H, Zhu Q, Tong W, et al. Distinct uptake, amplification, and release of SARS-CoV-2 by M1 and M2 alveolar macrophages. Cell Discovery. 2021;7(1):24
  102. 102. Liao M, Liu Y, Yuan J, Wen Y, Xu G, Zhao J, et al. Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19. Nature Medicine. 2020;26(6):842-844
  103. 103. Channappanavar R, Fehr AR, Vijay R, Mack M, Zhao J, Meyerholz DK, et al. Dysregulated type I interferon and inflammatory monocyte-macrophage responses cause lethal pneumonia in SARS-CoV-infected mice. Cell Host & Microbe. 2016;19(2):181-193
  104. 104. Meyerowitz EA, Vannier AGL, Friesen MGN, Schoenfeld S, Gelfand JA, Callahan MV, et al. Rethinking the role of hydroxychloroquine in the treatment of COVID-19. The FASEB Journal. 2020;34(5):6027-6037
  105. 105. Chen D, Xie J, Fiskesund R, Dong W, Liang X, Lv J, et al. Chloroquine modulates antitumor immune response by resetting tumor-associated macrophages toward M1 phenotype. Nature Communications. 2018;9(1):873
  106. 106. Vitte J, Michel M, Mezouar S, Diallo AB, Boumaza A, Mege JL, et al. Immune modulation as a therapeutic option during the SARS-CoV-2 outbreak: The case for antimalarial aminoquinolines. Frontiers in Immunology. 2020;11:2159
  107. 107. Haydar D, Cory TJ, Birket SE, Murphy BS, Pennypacker KR, Sinai AP, et al. Azithromycin polarizes macrophages to an M2 phenotype via inhibition of the STAT1 and NF-κB signaling pathways. Journal of Immunology. 2019;203(4):1021-1030
  108. 108. Melo GD, Lazarini F, Larrous F, Feige L, Kornobis E, Levallois S, et al. Attenuation of clinical and immunological outcomes during SARS-CoV-2 infection by ivermectin. EMBO Molecular Medicine. 2021;13(8). Available from: https://onlinelibrary.wiley.com/doi/10.15252/emmm.202114122
  109. 109. Yin L, Zhao H, Zhang H, Li Y, Dong Y, Ju H, et al. Remdesivir alleviates acute kidney injury by inhibiting the activation of NLRP3 inflammasome. Frontiers in Immunology. 2021;12:652446
  110. 110. Li R, Wang L. Baicalin inhibits influenza virus a replication via activation of type-I IFN signaling by reducing miR-146a. Molecular Medicine Reports. 2019. Available from: http://www.spandidos-publications.com/10.3892/mmr.2019.10743
  111. 111. Napoli C, Benincasa G, Criscuolo C, Faenza M, Liberato C, Rusciano M. Immune reactivity during COVID-19: Implications for treatment. Immunology Letters. 2021;231:28-34
  112. 112. Mahmudpour M, Roozbeh J, Keshavarz M, Farrokhi S, Nabipour I. COVID-19 cytokine storm: The anger of inflammation. Cytokine. 2020;133:155151

Written By

Perla Abou Atmeh, Soraya Mezouar and Jean-Louis Mège

Submitted: 01 April 2022 Reviewed: 27 June 2022 Published: 19 July 2022