Skip to main content
Wiley Open Access Collection logoLink to Wiley Open Access Collection
. 2022 Jul 3;309(1):64–74. doi: 10.1111/imr.13109

Heterogeneity in IgG‐CD16 signaling in infectious disease outcomes*

Joseph C Gonzalez 1,2, Saborni Chakraborty 1, Natalie K Thulin 3, Taia T Wang 1,2,4,
PMCID: PMC9539944  PMID: 35781671

Abstract

In this review, we discuss how IgG antibodies can modulate inflammatory signaling during viral infections with a focus on CD16a‐mediated functions. We describe the structural heterogeneity of IgG antibody ligands, including subclass and glycosylation that impact binding by and downstream activity of CD16a, as well as the heterogeneity of CD16a itself, including allele and expression density. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID‐19).

Keywords: COVID‐19, dengue disease, effector functions, Fc afucosylation, IgG1 antibodies

1. INTRODUCTION

Immunoglobulin G (IgG) antibodies provide a dominant form of defense against infectious diseases. IgGs are composed of two distinct structural domains that enable coupling of the recognition of pathogens to cellular responses including both adaptive and innate immune functions. This is achieved through direct binding of pathogens by the IgG Fab domain to form antigen‐IgG immune complexes. These immune complexes, in turn, direct the inflammatory response elicited during an infection through interactions with Fc gamma receptors (FcγRs) on immune cells. FcγRs are expressed on a variety of immune cells and serve as conduits for crosstalk between the adaptive and innate arms of the immune system (Figure 1A). Depending on the cell type and the specific FcγRs engaged, FcγRs transduce signaling that can escalate or limit the degree of the inflammatory response to an ongoing infection.

FIGURE 1.

FIGURE 1

(A) Type I Fcγ receptors (FcγRs) expression pattern on human white blood cell subsets. + indicates constitutive expression; − indicates no expression; +/− indicates no or low expression; * indicates inducible expression; */− indicates low or inducible expression; # indicates expression depending on FCGR2C allelic status. Classical monocytes are defined as CD14 highCD16 ‐ and non‐classical monocytes are defined as CD14 dimCD16 ++. (B) Binding affinities (association constant of binding [KA]) of four IgG subclasses (IgG1, IgG2, IgG3 and IgG4) to the various type I FcγRs. No color and * indicates that binding was either negligible or not detected to that FcγR. IgG1 AF denotes afucosylated IgG1, whereas IgG1 F is the core fucosylated IgG

Viral infections are controlled through a combination of adaptive and innate pathways, including virus neutralization, IgG Fc‐mediated effector/inflammatory cellular functions and innate antiviral pathways such as the Type I interferon system. 1 , 2 Upon viral infection, IgG‐mediated effector control is initiated when viral particles are bound by reactive antibodies to form high‐valence immune complexes that stabilize the otherwise low‐affinity interactions between IgG Fc and FcγR‐expressing cells. Some viral immune complexes may be neutralizing, thus preventing direct infection of host cells by viral particles. In the absence of neutralizing antibodies, immune complexes form that can both trigger FcγR‐mediated functions and remain infectious. Neutralizing and non‐neutralizing viral immune complexes as well as IgG‐bound viral debris may activate FcγR‐expressing host cells to initiate cellular processes that facilitate the resolution of infections. Clearance of immune complexes formed from viral particles and debris is critical for FcγR‐driven homeostatic mechanisms and persistence of immune complexes can drive prolonged activation of FcγR‐expressing cells leading to inflammatory sequelae and disease. 3 , 4 , 5 Once control of an infection is established and the viral load wanes, decreasing amounts of viral antigen are available to form immune complexes and perpetuate the inflammatory response. Consequently, antibody‐dependent immune activation and inflammation decrease as the antigen is cleared and immune homeostasis is restored.

In this review, we discuss how IgGs can modulate inflammatory signaling during viral infections with a focus on CD16a‐mediated functions. While inflammation is a mechanism required for immune homeostasis and resolution of acute infections, we focus here on two infectious diseases that are driven by pathogenic inflammatory responses during infection. Specifically, we review and discuss the evolving body of literature showing that afucosylated IgG immune complex signaling through CD16a contributes to the overwhelming inflammatory response that is central to the pathogenesis of severe forms of dengue disease and coronavirus disease 2019 (COVID‐19).

2. MECHANISMS GOVERNING HETEROGENEITY IN ANTIBODY EFFECTOR FUNCTIONS IN VIVO

IgG antibody effector functions are determined by the ratio of activating to inhibitory (A/I) signaling transduced through FcγRs on the surface of immune cells following their engagement by immune complexes. The A/I signaling ratio, first articulated by Ravetch and colleagues, governs the maturation of effector cells and is a major determinant of protection mediated by IgG antibodies. 6 , 7 , 8 , 9 A/I signal transduction is impacted by variables within both host IgG Fc and FcγR repertoires. Studies in the last decade have revealed a tremendous amount of heterogeneity in human IgG Fc domain repertoires. 10 , 11 These observations lead to the broader hypothesis that antibody heterogeneity is a dominant driver of divergent effector responses during infection. 12 The cumulative work on this topic shows that specific IgG Fc domain repertoires are predictors of clinical outcomes in some human diseases, yet it also shows that IgG repertoires, alone, do not clearly predict outcomes in many diseases where antibodies are thought to have a critical role in immunity. This invites the hypothesis that the less well studied heterogeneity on the other end of the signaling axis – FcγR expression and FcγR‐mediated effector cell functionality – may be an equally, or more important, driver of clinical outcomes that depend on antibody effector function. 13 FcγR heterogeneity might be particularly relevant to the increased risk for many infectious diseases that is associated with some demographic and health features such as advanced age, obesity, and diabetes, where effector cells are known to be different from healthy adults in distribution and/or functionality. 14 , 15 , 16 , 17 , 18 Studies characterizing IgG Fc repertoires and effector cells from matched donors are needed to truly address the role of antibody effector function in human immunity.

2.1. Regulation of A/I signaling by IgG Fc domain structure

FcγR affinity for a particular IgG antibody is determined in large part by the structural aspects of the Fc domain (Figure 2). The human IgG isotype is comprised of four subclasses: IgG1, IgG2, IgG3, and IgG4 with decreasing abundance from IgG1‐4. Each subclass contains a structurally distinct Fc domain, which has additional structural heterogeneity arising from IgG allotypes, 20 and variable Fc glycosylation. 21 IgG subclass and Fc glycoforms are clear modulators of Fc‐FcγR interactions while the role of specific allotypes is less understood.

FIGURE 2.

FIGURE 2

IgG glycoprotein and N‐linked glycan. IgG domain architecture (PDB 1HZH 19 ) and cartoon representation of IgG1 Fc glycans

Fc glycosylation introduces even greater structural and functional diversity to the Fc domain repertoire. All IgG subclasses contain a highly conserved core N‐linked glycosylation site at Asn‐297 within the CH2 domain of each heavy chain. The core N‐glycan structure is composed of four N‐acetylglucosamine (GlcNac) and three mannose residues arranged in a biantennary formation (Figure 2). As the IgG1 glycoprotein travels through the endoplasmic reticulum and Golgi apparatus, the core N‐glycan structure can be modified by the addition of other sugar residues such as core fucose linked to the innermost GlcNac, an additional bisecting GlcNac linked to the central mannose, as well as galactose and sialic acid residues that can be added to either or both antennae. Not all modifications are wholly independent; for example, sialic acid can only be added to glycans that contain galactose. Nonetheless, the many possible combinations of these modifications contribute to a variety of complex glycans at Asn‐297. The presence or absence of particular residues within these glycans impacts the binding affinity between IgG and Type I and Type II FcγRs. 12 At present, our understanding of the functional diversity conferred on IgG by various Fc glycoforms is largely limited to the most abundant subclass, IgG1. For IgG1, a major regulator of inflammatory FcγR signaling is core fucosylation of the Fc glycan that regulates interactions with the activating FcγR, CD16, as reviewed in greater detail below (Figure 1B). Sialylation of the IgG1 Fc confers the ability of immune complexes to signal through Type II FcγRs. 22 The activity of Type II FcγRs has been demonstrated largely in vivo where they have been observed to mediate diverse functions via interactions with sialylated immune complexes. For example, sialylated IgG ICs promote the anti‐inflammatory activity of therapeutic IVIG in context of autoimmune diseases through the engagement of DC‐SIGN/SIGN‐R1 23 , 24 , 25 , 26 and CD23‐mediated modulation of B cell selection during adaptive responses. 11 , 27 There remains much to learn about the molecular regulation of IgG Fc glycosylation but sex and age are known to correlate with specific patterns of glycosylation, as well as factors that correlate with geographic location; whether these are heritable or not, is not yet clear. 28 , 29 , 30

2.2. Regulation of A/I signaling by FcγRs

There are six human Type I FcγRs including FcγRIIc (CD32c), which is expressed by a minority of individuals. 31 , 32 Type I FcγRs are made up of both activating and inhibitory receptors as determined by their signaling through immunoreceptor tyrosine‐based activation (ITAM) or inhibition (ITIM) motifs, respectively. Type I receptors are activating, except for FcγRIIb (CD32b), which is the sole inhibitory receptor. The extracellular domains of the FcγR alpha chains are variable in their affinities for IgG Fc domains. FcγRI (CD64) is the only high affinity receptor that can bind to monomeric IgG. Co‐expression patterns of FcγR on immune cells shape the direction and magnitude of Fc‐mediated immune activity (Figure 1A,B).

High‐avidity, productive interactions between Fcs within immune complexes and FcγRs on innate cells trigger cell‐type specific signaling cascades that culminate in diverse cellular responses, shaping the innate and adaptive immune landscape. FcγR‐mediated activation of innate immune cells leads to a gamut of cellular responses ranging from those often modeled in in vivo assays such as antibody‐dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP), to the induction of more complex immune programs including cytotoxic degranulation, proinflammatory cytokine and chemokine production, macrophage polarization, enhancement of antigen presentation by APCs, maturation of DCs and maintaining immune homeostasis. 20 , 33 , 34 , 35 Despite the distinct effector roles of the different cellular subsets, all of them signal through ITAM or ITIM motifs via pathways that are fairly conserved and are well reviewed elsewhere. 36 , 37 These motifs are present either in the cytoplasmic tail of the Fc receptors themselves (CD32a,b,c) or on non‐covalently associated ITAM‐containing adaptor proteins FcεR1γ (FcRγ) or CD3ζ (Figure 1A).

Multivalent Fc‐FcγR engagement on the cell surface leads to clustering of the FcγRs and CD45 phosphatase exclusion on the plasma membrane, enabling the activation of the Lyn and/or Fyn and Hgr Src‐family kinases, that then phosphorylate conserved tyrosines (Tyr) in the cytoplasmic ITAM motifs. Phosphorylation of the two Tyrs creates docking sites for Syk kinases through their SH2 domains and leads to activation of these Syk kinases. Subsequently, activated Syk then phosphorylates a plethora of downstream adaptor molecules that propagate further signaling cascades involving phospholipase C‐gamma (PLCγ), phosphoinositide 3‐kinase (PI3K), calcineurin, and Vav‐1 pathways. 37 , 38 , 39 , 40 , 41 , 42 , 43 Many of these pathways coregulate or cooperate in the performance of cell‐specific immune activities. To counterbalance activating signaling through ITAM motifs, most of the innate cell subsets co‐express an inhibitory Fc receptor that signals via the conserved ITIM in its cytoplasmic domain. When activating and inhibitory FcγRs are simultaneously cross‐linked and co‐cluster, the ITAM‐phosphorylating Src kinases also phosphorylate the conserved Tyrs of proximal ITIMs. ITIM phosphorylation promotes non‐covalent binding of tyrosine phosphatases SHP‐1 and SHP2 and the inositol phosphatases SHIP‐1 and SHIP2 via the SH2 domains of the phosphatases. These phosphatases subsequently dephosphorylate the Tyr phosphates on proximal ITAMs and other adaptor molecules, thereby tempering ITAM signaling and any associated immune activation. 44 , 45

Multiple factors contribute to the heterogeneity of FcγR repertoires including single nucleotide polymorphisms (SNP), copy number variation (CNV), and glycosylation patterns of FcγRs. 21 , 31 , 46 , 47 SNPs in the FcγR genes modulate binding affinity to IgG Fc. There are two major CD32a allotypes, R131 and H131 where H131 exhibits greater binding affinity for IgG1 and IgG2. 31 CD16a similarly has two well characterized allotypes, F158 and V158 where the less frequent V158 allotype exhibits higher affinity for IgG compared to F158. 31 , 48 , 49 , 50 , 51 , 52 While the CD16a allele expressed correlates with the clinical response to some therapeutic anti‐tumor monoclonal antibodies, 53 , 54 , 55 , 56 there is limited data supporting a correlation between either CD16 variants and clinical outcomes in infectious diseases. Recent studies suggest that apart from allelic variations, FcγR expression levels can also be highly heterogeneous amongst individuals, possibly explained in part by CNV, and may be a previously underappreciated determinant of antibody signaling outcomes. 13 , 46 , 47 , 57 , 58 The glycan repertoire of FcγRs is also highly heterogeneous owing to the variability in the number of N‐glycosylation sites on the FcγRs and an array of alternative glycoforms that can exist at each site. 59 Structural and biophysical data have shown that the presence or absence of certain glycans at particular amino acid positions of these receptors impacts CD16a binding to IgG, bringing into focus the role of FcγR N‐glycans in shaping effector immune functions. 60 , 61 , 62 , 63 , 64 Elegant profiling studies of FcγRs from primary human NK cells and monocytes have shown that receptor glycosylation profiles vary amongst different immune cells and are also governed by the FcγR allotype of an individual. 60 , 65 , 66 , 67 , 68 More studies are required to gain an understanding of this heterogeneity of the glycan profiles of FcγR and whether they change upon certain activating/inhibitory signals, or between healthy and diseased states.

3. AFUCOSYLATED IGG1‐CD16A SIGNALING AXIS

A major focus of this review is the abundance of recent data that show how heterogeneity within the IgG and FcγR repertoires, and specifically in the afucosylated IgG1‐CD16a signaling axis, impacts the course of some infectious diseases. Specifically, the proinflammatory IgG Fc glycoform lacking a core fucose, here termed “afucosylated”, is enriched on IgG1 in people who progress to severe forms of dengue disease and COVID‐19, the disease caused by SARS‐CoV‐2. 69 , 70 , 71 , 72 , 73 , 74 Afucosylated IgG1 has high affinity for the activating FcγR, CD16a, which mediates diverse inflammatory functions that likely only partly overlap in these two very different viral infectious diseases (Figure 1B).

3.1. IgG1 afucosylation

In healthy adults and children, IgG1 antibodies are largely modified by fucosylated Fc glycoforms with afucosylated glycoforms generally comprising <10% of the IgG population. 29 An increased abundance of afucosylated IgG1 has been observed in a number of clinical settings including fetal/neonatal alloimmunity, acute dengue disease, in people with, or who will develop, severe COVID‐19, and in malaria. 69 , 70 , 71 , 72 , 73 , 74 , 75 , 76 Increased afucosylated glycoforms have also been shown in association with bulk IgG glycans, including glycans on the Fab and Fc from all IgG subclasses, in HIV elite controllers and latent tuberculosis patients. 77 , 78 The genetic or environmental factors that contribute to afucosylated IgG1 production are not yet understood.

3.2. CD16a

Expression of the receptor for afucosylated IgG1, CD16a, is mostly limited to natural killer (NK) cells, monocytes, and macrophages. 79 Recent intriguing studies also suggest that very low levels of CD16a may be expressed on neutrophils and induced on CD4+ and/or CD8+ T cell subsets in disease states such as autoimmunity and chronic infection. 47 , 80 , 81 , 82 , 83 , 84 Within the monocyte lineage, CD16a expression is limited to the non‐classical and intermediate subsets, while the more abundant classical monocyte subset does not express CD16a (Figure 1A). 85 Historically, it has been unclear whether these three monocyte subsets represent distinct developmental trajectories or are part of one linear trajectory, an implication of the latter being that CD16a is a marker of monocyte maturity, similar to NK cells. 86 More recent studies on CD16a strongly support one linear trajectory in which CD16a expression is acquired as CD16a‐ classical monocytes give rise to CD16a + non‐classical monocytes through the transitional CD16a + intermediate subset. 87 , 88 , 89 , 90 Unlike monocytes, a majority of macrophages, including alveolar macrophage, express relatively high levels of CD16a. 91

Functionally, CD16a is the major cytotoxic receptor but also mediates maturation and activation of CD16a‐expressing cells. This modulates autocrine and paracrine immunity, impacting antigen presentation and inflammatory processes including cytokine release and chemotactic cell migration. 74 , 79

4. PATHOGENIC ROLE OF AFUCOSYLATED IGG1‐CD16 SIGNALING IN SEVERE DENGUE DISEASE AND COVID‐19

There are multiple viral and host‐derived factors that determine the trajectory of disease following dengue virus or SARS‐CoV‐2 infections, yet severe disease caused by these viruses is fundamentally the result of inadequate pre‐exiting immunity together with an inflammatory host response during infection that damages tissues and disrupts critical physiologic processes. Dengue viruses and SARS‐CoV‐2 are similar in that they are both enveloped, positive‐sense, single‐stranded RNA viruses; however, dengue viruses are mosquito‐borne and can infect cells in a broad range of organs while SARS‐CoV‐2 is a respiratory virus with a majority of replication thought to take place in the upper and lower respiratory tracts. While hyperinflammation is common to severe forms of disease caused by dengue viruses and SARS‐CoV‐2, severe dengue disease and severe COVID‐19 are distinct and result from very different pathological inflammatory processes. That both diseases are likely mediated in part by an aberrant afucosylated IgG1‐CD16a signaling speaks to the diversity of functions that can arise through engaging this effector pathway.

4.1. Afucosylated IgG1 in dengue disease

There are four serotypes of dengue viruses (DENV), DENV1‐4 and primary infection with one serotype elicits antibodies that are broadly reactive, but that do not neutralize other DENV serotypes. The vast majority of DENV infections result in a subclinical or mild clinical phenotype but a subset of individual's progress to more severe disease. The most severe dengue disease forms, dengue hemorrhagic fever and dengue shock syndrome (DHF/DSS), are characterized by sequelae including thrombocytopenia and plasma leakage due to increased capillary permeability, in some cases resulting in shock (DSS). 92 A majority of DHF/DSS occurs in secondary DENV infections when non‐neutralizing, reactive IgGs are present that modulate the level of infection and the cellular response to infection, through “extrinsic” and “intrinsic” antibody‐dependent enhancement (ADE) mechanisms, respectively. 93 , 94 An aggressive inflammatory response including elevated production of interferon (IFN)‐gamma, tumor necrosis factor (TNF)‐α, interleukin (IL)‐2, IL‐6, IL‐8, and vascular endothelial growth factor (VEGF) 95 likely contribute to capillary permeability and other sequalae of severe disease. These cytokines are thought to derive from virus‐infected monocytes, dendritic cells, platelets, mast cells and T cell subsets. Inherent cellular differences have been associated with cytokine production in both protective and pathologic inflammatory responses during infection. 96

Although the presence of reactive, non‐neutralizing IgG is a risk factor for progression to severe disease, it has been appreciated for several decades that the presence of non‐neutralizing IgG alone does not predict the development of DHF/DSS. This suggests that only specific IgG repertoires, and potentially specific FcγRs, mediate ADE of disease. Recent studies have begun to resolve this important topic in dengue disease pathogenesis. In 2017, our group and others studied a cohort of patients with secondary DENV infection with mild or severe disease. They observed that patients who progressed to DHF/DSS produced anti‐DENV IgG1 with significantly elevated Fc afucosylation over those that did not develop severe disease. 69 An increased frequency of anti‐DENV IgG1 relative to IgG2 was also associated with severe disease. Further, afucosylated IgG from patients with severe disease could activate platelets and trigger platelet loss in an FcγR‐dependent manner, in vivo, consistent with thrombocytopenia characteristic of severe dengue. The identification of afucosylated anti‐DENV IgG1 as a biomarker of severe dengue disease shed light on why only a subset of DENV patients with anti‐DENV IgGs develop severe disease.

In 2020, we further showed that elevated afucosylated anti‐DENV IgG1 in maternal circulation predicted the development of clinically significant disease in their infants during primary dengue infection. 70 This was presumably due to transplacental transfer of the maternal IgG, rendering infants susceptible to ADE of disease during primary infections. This study reproduced our earlier finding that afucosylated anti‐DENV IgGs predicted dengue disease severity. As afucosylated IgG1 has increased affinity for CD16a, the identification of IgG1 afucosylation as a correlate of dengue disease severity implicates CD16a in the pathophysiology of ADE. We assessed the requirement for CD16a in antibody‐dependent DENV infections using engineered human monocytic cell lines (U937) that express different combinations of CD16a and the other low‐affinity activating FcγR, CD32a70. This work showed that CD16a and CD32a had distinct roles in ADE of dengue infection. CD32a mediated the majority of entry by DENV immune complexes (extrinsic ADE), while the presence of CD16a did not modify the ability of DENV immune complexes to enter cells. However, in the presence of both CD16a and CD32a, infection was enhanced through a mechanism that required functional CD16a signaling. Thus, CD16a‐mediated ITAM signaling triggered intrinsic ADE of DENV infection in a mechanism that was shown to require the calcineurin signaling network. Together these data support a functional role for afucosylated anti‐DENV IgG1 in driving severe DENV disease though increased infection in and ITAM signaling of CD16a expressing monocytes (Figure 3). In addition to modulating viral infection in CD16a‐expressing cells, afucosylated DENV immune complexes likely contribute more broadly to the hyperinflammmatory sequelae in severe dengue disease.

FIGURE 3.

FIGURE 3

Model for antibody‐dependent enhancement (ADE) in dengue infection and the roles of activating type I Fcγ receptors (FcγRs) and afucosylated IgG. Viral attachment and entry occurs mostly by CD32a mediated endocytosis after interactions with anti‐E IgG1:dengue ICs. Additionally, high affinity interactions between afucosylated anti‐E IgG1:dengue ICs and CD16a promotes increased ITAM signaling. ITAM signaling triggers downstream calcium flux and activates the calcineurin signaling network. Specific calcineurin inhibitors prevented FcγR‐dependent dengue virus infection

In 2021 Bournazos et al. again demonstrated afucosylated IgG1 as a correlate of severe disease and provided significant additional insights into the kinetics of afucosylated IgG1 production in less severe cases of DENV infection. Using matched patient plasma, Bournazos et al. showed that the abundance of afucosylated IgG1 increases during convalescence after mild primary infection and separately in response to secondary infection. 71 These two independent increases in afucosylated IgG1 drove a generally higher abundance of afucosylated IgG1 in patients with secondary DENV. The timing of these increases in afucosylated IgG1 strongly suggests that production of afucosylated IgG1 is part of the host response to DENV infection. However, only a subset of DENV patients exhibited increased afucosylated IgG1, indicating that some individuals may be more predisposed to produce antibodies with this modification than others. Further investigation into the regulation and mechanism of afucosylated anti‐dengue IgG1 production and activity are required to understand the role of these antibodies in DENV disease pathogenesis.

4.2. Afucosylated IgG1 in severe COVID‐19

As with dengue virus infections, the vast majority of infections with SARS‐CoV‐2 result in subclinical or very mild disease. Severe COVID‐19 arises in part from aberrant activation of cells in the myeloid compartment, triggering a hyperinflammatory cascade. 97 , 98 High concentrations of proinflammatory cytokines, particularly IL‐6 and TNFα are strongly associated with severe disease and poor prognosis. 99 Further, unlike other respiratory viral infections, patients with severe COVID‐19 benefit from steroid treatment and other medications targeting inflammatory pathways, highlighting the underlying hyperinflammatory pathophysiology of this disease.

In 2021, our group described a proinflammatory feature of IgG that correlated with severe COVID‐19: afucosylated IgG1. 72 While this was a familiar correlate of disease severity from dengue disease studies, this finding was unexpected given the distinct pathophysiologies of severe COVID‐19 and dengue disease. Afucosylated IgG1 antibodies against the SARS‐CoV‐2 receptor‐binding domain (RBD) were more abundant in hospitalized COVID‐19 patients compared to both adult COVID‐19 outpatients and seropositive children. This increased abundance of afucosylated IgG1 was predominantly observed in hospitalized male patients from two independent cohorts but did not correlate with age. IgG subclass differences amongst the various groups were also probed as they are known modulators Fc‐FcγR interactions. A significant increase in anti‐RBD IgG3 antibody level was observed in patients admitted to the intensive care unit (ICU). 72 The IgG3 subclass and afucosylated IgG1 are among the most inflammatory IgG structures due to their higher affinities to activating FcγRs, particularly CD16a (Figure 1B). Thus, the Fc signature observed in hospitalized COVID‐19 patients was highly proinflammatory, and IgG from patients with elevated afucosylation (>20%) had 5–6 fold higher affinity to recombinant CD16a as compared to patient IgG that exhibited low afucosylation (<10%). Enhanced binding to CD16a translated to higher inflammatory potential as evidenced by robust production of IL‐6, TNFα, and other proinflammatory factors by healthy donor monocytes stimulated with ICs composed of afucosylated IgG and SARS‐CoV‐2 spike trimer in vitro. A subsequent study by Larsen et al. reproduced the finding of enriched afucosylated IgG1 in sera of severe COVID‐19 patients and proposed an interesting hypothesis that Fc fucosylation is regulated by whether an antigen is presented to the B cell in a membrane‐bound or soluble form. 73 Findings from Hoepel et al. reinforced the proinflammatory capability of afucosylated ICs as they demonstrated that even homeostasis‐promoting human alveolar macrophage could be thoroughly reprogrammed toward a proinflammatory phenotype by stimulation with afucosylated immune complexes. 100

While these studies established that afucosylated IgGs were present in severe COVID‐19, it was not clear whether this was a cause or effect of severe disease. In either case, afucosylated immune complexes might modulate disease severity, yet clarifying the timing of production was important to determining whether Fc afucosylation was an early biomarker of risk in SARS‐CoV‐2 infections. To assess whether elevated afucosylated IgG1 preceded progression to severe symptoms, samples were studied from two independent, longitudinal cohorts of mild COVID‐19 patients, a subset of whom would go on to develop more severe disease. From this analysis, in 2022, our group further reported that elevated IgG1 afucosylation, together with absent or low early neutralizing antibodies was an early predictor of risk for disease progression in mild COVID‐19 patients. 74 Receiver operating curve (ROC) analysis showed that, individually, neutralizing titers and IgG afucosylation were only moderate predictors of disease progression; however, combining the two features enabled robust predictive capacity to identify mild COVID‐19 patients who were at risk for developing severe symptoms (“progressors”). 74 We also observed that ASCs, specifically plasmablasts from “progressors” expressed significantly less of the enzyme α‐1,6‐fucosyltransferase (FUT8) that is responsible for core fucosylation, consistent with increased afucosylated IgG1 in this group. 74

Dysregulation in innate immune effector cell subsets has been shown to correlate with COVID‐19 disease severity. 101 , 102 , 103 , 104 As IgG‐mediated, inflammatory effector cell activity is a function of both cell frequency and FcγR co‐expression by these cells, we went on to perform an in‐depth profiling of peripheral innate immune cells from “progressors” and non‐progressor controls to quantify both cell frequency and FcγR expression. We observed that monocyte distribution within peripheral blood immune cells (PBMCs) was distinct in “progressors” prior to the development of more severe symptoms. Specifically, “progressors” had an increased frequency of CD16a‐expressing immune cells, non‐classical and intermediate monocytes. In addition to being more frequent, non‐classical and intermediate monocytes from “progressors” expressed more surface copies of CD16a protein. Other activating and inhibitory FcγRs were not distinct between “progressors” and controls. ROC analysis showed that, separate from the non‐neutralizing, afucosylated IgG1 correlate of progression to more severe COVID‐19, CD16a expression alone within the peripheral myeloid cell compartment was also strongly predictive of disease progression. 74 Together, these data link the afucosylated IgG1‐CD16a axis to the development of worsening symptoms in COVID‐19.

Although the recruitment of monocytes and other innate immune cells to sites of infection and inflammation is fundamentally a homeostatic immune mechanism, autopsies from patients with COVID‐19 revealed extreme infiltration of lungs thought to result in poor gas exchange, and co‐localization of viral RNA with myeloid cell infiltrates, likely demonstrating an overwhelming inflammatory response to viral antigens. 105 , 106 To study how afucosylated immune complexes might modulate inflammation in the lung, specifically, our group developed a mouse model enabling assessment of human immune complex‐mediated lung inflammation. Administration of human afucosylated immune complexes into the lungs of FcγR‐humanized mice 107 promoted a rapid and robust inflammatory response characterized by neutrophil and monocyte infiltration, as well as the production of numerous proinflammatory soluble factors including IL‐6, TNFα, and myelo‐attractive chemokines. 74 This inflammatory response to afucosylated immune complexes was CD16a dependent and was not present in mice that received fucosylated immune complexes. The absence of virus and infection in this mouse model allows for the characterization of the inflammatory potential of afucosylated IgG1, as well as other human IgG subclasses and glycoforms, unconfounded by viral and antiviral activity alike. While the humanized mouse model used to study how antibody signaling impacts the lung is not a model of COVID‐19 pathogenesis, the inflammatory response within the lung in response to afucosylated IgG1 resembles the inflammatory state of the lungs of severe COVID‐19 patients, namely a persistence of neutrophils and inflammatory monocytes or monocyte‐derived macrophages (Figure 4). 108 , 109 , 110 , 111 , 112 These data collectively demonstrate that the enhanced afucosylated IgG1‐CD16a signaling axis observed in COVID‐19 “progressors” can promote a rapid and robust inflammatory response in vivo that exhibits similarities to findings in severe COVID‐19.

FIGURE 4.

FIGURE 4

Model for immunopathology of severe COVID‐19 mediated by enhanced CD16a:IgG signaling axis. Enhanced signaling via the ITAM domain of CD16a, which in turn leads to increased proinflammatory cytokine production in lung of severe COVID‐19 patients might be achieved through either formation of ICs with persisting viral antigens and afucosylated anti‐SARS‐CoV‐2 IgG1, which have higher affinity for CD16a on effector cells or by recruitment of effector cells, which have an increased expression of CD16a on their surface and thus an increased inflammatory signaling potential. Thus, proinflammatory elements on both sides of the receptor‐ligand axis might contribute to more severe disease

5. CONCLUDING REMARKS AND OUTSTANDING QUESTIONS

The molecular regulation of IgG1 Fc glycosylation, particularly Fc fucosylation is incompletely understood. Although a number of studies have identified genes other than Fut8 that might be involved in core fucosylation, including Ikzf1, Ikzf3, Hnf1a, Hnf4a, and less enigmatic genes involved in fucose biosynthesis, the details of how these genes might participate in the regulation of FUT8 expression in ASCs and the production of afucosylated IgG1 have yet to be fully described. 113 , 114 , 115 , 116 Sex and age are also known to correlate with IgG Fc glycoforms 28 , 29 , 30 and exogenous delivery of sex hormones can impact Fc glycosylation. 117 , 118 A greater understanding of how and why afucosylated IgG1 antibodies are produced in healthy and disease contexts could lead to the development of therapeutic interventions to modulate IgG afucosylation for clinical benefit.

Another key question in regulation of human antibody effector function is the extent to which heterogeneity in FcγRs, as opposed to IgG Fcs, impacts disease outcomes. Is FcγR repertoire heterogeneity a more, less, or equivalently important variable relative to Fc heterogeneity in driving antibody‐mediated disease outcomes (both positive and negative outcomes)? As demonstrated in vivo, the inflammatory potential of afucosylated IgG1 in the lung was mediated almost entirely by CD16a. Further, humans with mild COVID‐19 who progressed to more severe disease had increased expression of CD16a on myeloid cells and this early cellular feature alone predicted progression to more severe disease. 74 These observations point to the question of how CD16a protein expression is regulated within the myeloid compartment. Additional identification of the viral or host‐derived factors responsible for eliciting classical monocyte differentiation or maturation into CD16a‐expressing non‐classical monocytes might also clarify the distinct monocyte response observed in patients who progress to severe COVID‐19. Structural heterogeneity of CD16a imparted by genetics and glycosylation, as well as heterogeneity of CD16a expression by immune effector cells are likely co‐determinants of both protective and hyperinflammatory antibody signaling outcomes in infectious disease and are deserving of further investigation. Ideally, studies would characterize both human FcγR‐expressing effector cells and IgG antibodies from matched donors to clarify how heterogeneity in antibody signaling contributes to infectious disease outcomes.

CONFLICT OF INTEREST

The authors have no conflicts of interest to declare.

ACKNOWLEDGEMENTS

Support was received from Stanford University the Searle Scholars Program and the National Institute of Allergy and Infectious Diseases of the National Institutes of Health under Award Numbers U19 AI111825, R01 AI139119 and 5 T32 AI007290.

Gonzalez JC, Chakraborty S, Thulin NK, Wang TT. Heterogeneity in IgG‐CD16 signaling in infectious disease outcomes. Immunol Rev. 2022;309:64‐74. doi: 10.1111/imr.13109

*This article is part of a series of reviews covering SARS‐CoV‐2 Immunity appearing in Volume 309 of Immunological Reviews.

DATA AVAILABILITY STATEMENT

Data sharing not applicable ‐ no new data generated, or the article describes entirely theoretical research.

REFERENCES

  • 1. Knipe DM, Howley PM. Fields Virology. 6th ed. Wolters Kluwer/Lippincott Williams & Wilkins Health; 2013. [Google Scholar]
  • 2. Thulin NK, Wang TT. The role of Fc gamma receptors in broad protection against influenza viruses. Vaccines (Basel). 2018;6(3):36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Wieland A, Shashidharamurthy R, Kamphorst AO, et al. Antibody effector functions mediated by Fcgamma‐receptors are compromised during persistent viral infection. Immunity. 2015;42(2):367‐378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Warren JS, Ward PA. Immune Complex Diseases. eLS; 2017:1‐9. [Google Scholar]
  • 5. Wang TT, Ravetch JV. Immune complexes: not just an innocent bystander in chronic viral infection. Immunity. 2015;42(2):213‐215. [DOI] [PubMed] [Google Scholar]
  • 6. Kalergis AM, Ravetch JV. Inducing tumor immunity through the selective engagement of activating Fcgamma receptors on dendritic cells. J Exp Med. 2002;195(12):1653‐1659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Dhodapkar KM, Kaufman JL, Ehlers M, et al. Selective blockade of inhibitory Fcgamma receptor enables human dendritic cell maturation with IL‐12p70 production and immunity to antibody‐coated tumor cells. Proc Natl Acad Sci USA. 2005;102(8):2910‐2915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Nimmerjahn F, Ravetch JV. Divergent immunoglobulin g subclass activity through selective Fc receptor binding. Science. 2005;310(5753):1510‐1512. [DOI] [PubMed] [Google Scholar]
  • 9. Nimmerjahn F, Bruhns P, Horiuchi K, Ravetch JV. FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity. 2005;23(1):41‐51. [DOI] [PubMed] [Google Scholar]
  • 10. Mahan AE, Tedesco J, Dionne K, et al. A method for high‐throughput, sensitive analysis of IgG Fc and Fab glycosylation by capillary electrophoresis. J Immunol Methods. 2015;417:34‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Wang TT, Maamary J, Tan GS, et al. Anti‐HA glycoforms drive B cell affinity selection and determine influenza vaccine efficacy. Cell. 2015;162(1):160‐169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Wang TT. IgG Fc glycosylation in human immunity. Curr Top Microbiol Immunol. 2019;63‐75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Kerntke C, Nimmerjahn F, Biburger M. There is (Scientific) strength in numbers: a comprehensive quantitation of Fc gamma receptor numbers on human and murine peripheral blood leukocytes. Front Immunol. 2020;11:118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Friedrich K, Sommer M, Strobel S, et al. Perturbation of the monocyte compartment in human obesity. Front Immunol. 2019;10:1874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Devevre EF, Renovato‐Martins M, Clement K, Sautes‐Fridman C, Cremer I, Poitou C. Profiling of the three circulating monocyte subpopulations in human obesity. J Immunol. 2015;194(8):3917‐3923. [DOI] [PubMed] [Google Scholar]
  • 16. Min D, Brooks B, Wong J, Salomon R, Bao W, Harrisberg B, Twigg SM, Yue DK, McLennan SV Alterations in monocyte CD16 in association with diabetes complications. Mediators Inflamm 2012;2012:649083, 1, 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Seidler S, Zimmermann HW, Bartneck M, Trautwein C, Tacke F. Age‐dependent alterations of monocyte subsets and monocyte‐related chemokine pathways in healthy adults. BMC Immunol. 2010;11:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Dugast AS, Tonelli A, Berger CT, et al. Decreased Fc receptor expression on innate immune cells is associated with impaired antibody‐mediated cellular phagocytic activity in chronically HIV‐1 infected individuals. Virology. 2011;415(2):160‐167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Saphire EO, Parren PW, Pantophlet R, et al. Crystal structure of a neutralizing human IGG against HIV‐1: a template for vaccine design. Science. 2001;293(5532):1155‐1159. [DOI] [PubMed] [Google Scholar]
  • 20. Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to effector functions. Front Immunol. 2014;5:520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Subedi GP, Barb AW. The immunoglobulin G1 N‐glycan composition affects binding to each low affinity Fc gamma receptor. MAbs. 2016;8(8):1512‐1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Pincetic A, Bournazos S, DiLillo DJ, et al. Type I and type II Fc receptors regulate innate and adaptive immunity. Nat Immunol. 2014;15(8):707‐716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Kaneko Y, Nimmerjahn F, Ravetch JV. Anti‐inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science. 2006;313(5787):670‐673. [DOI] [PubMed] [Google Scholar]
  • 24. Anthony RM, Nimmerjahn F, Ashline DJ, Reinhold VN, Paulson JC, Ravetch JV. Recapitulation of IVIG anti‐inflammatory activity with a recombinant IgG Fc. Science. 2008;320(5874):373‐376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Anthony RM, Kobayashi T, Wermeling F, Ravetch JV. Intravenous gammaglobulin suppresses inflammation through a novel T(H)2 pathway. Nature. 2011;475(7354):110‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Washburn N, Schwab I, Ortiz D, et al. Controlled tetra‐Fc sialylation of IVIg results in a drug candidate with consistent enhanced anti‐inflammatory activity. Proc Natl Acad Sci USA. 2015;112(11):E1297‐E1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Maamary J, Wang TT, Tan GS, Palese P, Ravetch JV. Increasing the breadth and potency of response to the seasonal influenza virus vaccine by immune complex immunization. Proc Natl Acad Sci USA. 2017;114(38):10172‐10177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Pucic M, Knezevic A, Vidic J, et al. High throughput isolation and glycosylation analysis of IgG‐variability and heritability of the IgG glycome in three isolated human populations. Mol Cell Proteom. 2011;10(10):1‐15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Bakovic MP, Selman MH, Hoffmann M, et al. High‐throughput IgG Fc N‐glycosylation profiling by mass spectrometry of glycopeptides. J Proteome Res. 2013;12(2):821‐831. [DOI] [PubMed] [Google Scholar]
  • 30. de Haan N, Reiding KR, Driessen G, van der Burg M, Wuhrer M. Changes in healthy human IgG Fc‐glycosylation after birth and during early childhood. J Proteome Res. 2016;15(6):1853‐1861. [DOI] [PubMed] [Google Scholar]
  • 31. Bruhns P, Iannascoli B, England P, et al. Specificity and affinity of human Fcgamma receptors and their polymorphic variants for human IgG subclasses. Blood. 2009;113(16):3716‐3725. [DOI] [PubMed] [Google Scholar]
  • 32. van der Heijden J, Breunis WB, Geissler J, de Boer M, van den Berg TK, Kuijpers TW. Phenotypic variation in IgG receptors by nonclassical FCGR2C alleles. J Immunol. 2012;188(3):1318‐1324. [DOI] [PubMed] [Google Scholar]
  • 33. Vogelpoel LT, Hansen IS, Rispens T, et al. Fc gamma receptor‐TLR cross‐talk elicits pro‐inflammatory cytokine production by human M2 macrophages. Nat Commun. 2014;5:5444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Platzer B, Stout M, Fiebiger E. Antigen cross‐presentation of immune complexes. Front Immunol. 2014;5:140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Wang XY, Wang B, Wen YM. From therapeutic antibodies to immune complex vaccines. NPJ Vaccines. 2019;4:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody‐dependent enhancement. Nat Rev Immunol. 2020;20(10):633‐643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Getahun A, Cambier JC. Of ITIMs, ITAMs, and ITAMis: revisiting immunoglobulin Fc receptor signaling. Immunol Rev. 2015;268(1):66‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Kiefer F, Brumell J, Al‐Alawi N, et al. The Syk protein tyrosine kinase is essential for Fcgamma receptor signaling in macrophages and neutrophils. Mol Cell Biol. 1998;18(7):4209‐4220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Pores‐Fernando AT, Gaur S, Doyon MY, Zweifach A. Calcineurin‐dependent lytic granule exocytosis in NK‐92 natural killer cells. Cell Immunol. 2009;254(2):105‐109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Hall AB, Gakidis MA, Glogauer M, et al. Requirements for Vav guanine nucleotide exchange factors and Rho GTPases in FcgammaR‐ and complement‐mediated phagocytosis. Immunity. 2006;24(3):305‐316. [DOI] [PubMed] [Google Scholar]
  • 41. Utomo A, Cullere X, Glogauer M, Swat W, Mayadas TN. Vav proteins in neutrophils are required for FcgammaR‐mediated signaling to Rac GTPases and nicotinamide adenine dinucleotide phosphate oxidase component p40(phox). J Immunol. 2006;177(9):6388‐6397. [DOI] [PubMed] [Google Scholar]
  • 42. Park YJ, Yoo SA, Kim M, Kim WU. The role of calcium‐calcineurin‐NFAT signaling pathway in health and autoimmune diseases. Front Immunol. 2020;11:195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Ben Mkaddem S, Benhamou M, Monteiro RC. Understanding Fc receptor involvement in inflammatory diseases: from mechanisms to new therapeutic tools. Front Immunol. 2019;10:811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Bournazos S, Wang TT, Dahan R, Maamary J, Ravetch JV. Signaling by antibodies: recent progress. Annu Rev Immunol. 2017;35:285‐311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity. 2006;24(1):19‐28. [DOI] [PubMed] [Google Scholar]
  • 46. Breunis WB, van Mirre E, Geissler J, et al. Copy number variation at the FCGR locus includes FCGR3A, FCGR2C and FCGR3B but not FCGR2A and FCGR2B. Hum Mutat. 2009;30(5):E640‐E650. [DOI] [PubMed] [Google Scholar]
  • 47. Franke L, el Bannoudi H, Jansen DT, et al. Association analysis of copy numbers of FC‐gamma receptor genes for rheumatoid arthritis and other immune‐mediated phenotypes. Eur J Hum Genet. 2016;24(2):263‐270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Koene HR, Kleijer M, Algra J, Roos D, von dem Borne AE, de Haas M. Fc gammaRIIIa‐158V/F polymorphism influences the binding of IgG by natural killer cell Fc gammaRIIIa, independently of the Fc gammaRIIIa‐48L/R/H phenotype. Blood. 1997;90(3):1109‐1114. [PubMed] [Google Scholar]
  • 49. Wu J, Edberg JC, Redecha PB, et al. A novel polymorphism of FcgammaRIIIa (CD16) alters receptor function and predisposes to autoimmune disease. J Clin Invest. 1997;100(5):1059‐1070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Dall'Ozzo S, Andres C, Bardos P, Watier H, Thibault G. Rapid single‐step FCGR3A genotyping based on SYBR Green I fluorescence in real‐time multiplex allele‐specific PCR. J Immunol Methods 2003;277(1–2):185–192, 192. [DOI] [PubMed] [Google Scholar]
  • 51. Mahaweni NM, Olieslagers TI, Rivas IO, et al. A comprehensive overview of FCGR3A gene variability by full‐length gene sequencing including the identification of V158F polymorphism. Sci Rep. 2018;8(1):15983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Liu F, Ding H, Jin X, et al. FCGR3A 158V/F polymorphism and response to frontline R‐CHOP therapy in diffuse large B‐cell lymphoma. DNA Cell Biol. 2014;33(9):616‐623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Ruyssen‐Witrand A, Rouanet S, Combe B, et al. Fcgamma receptor type IIIA polymorphism influences treatment outcomes in patients with rheumatoid arthritis treated with rituximab. Ann Rheum Dis. 2012;71(6):875‐877. [DOI] [PubMed] [Google Scholar]
  • 54. Quartuccio L, Fabris M, Pontarini E, et al. The 158VV Fcgamma receptor 3A genotype is associated with response to rituximab in rheumatoid arthritis: results of an Italian multicentre study. Ann Rheum Dis. 2014;73(4):716‐721. [DOI] [PubMed] [Google Scholar]
  • 55. Mellor JD, Brown MP, Irving HR, Zalcberg JR, Dobrovic A. A critical review of the role of Fc gamma receptor polymorphisms in the response to monoclonal antibodies in cancer. J Hematol Oncol. 2013;6:1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Magnes T, Melchardt T, Hufnagl C, et al. The influence of FCGR2A and FCGR3A polymorphisms on the survival of patients with recurrent or metastatic squamous cell head and neck cancer treated with cetuximab. Pharmacogenomics J. 2018;18(3):474‐479. [DOI] [PubMed] [Google Scholar]
  • 57. Machado LR, Bowdrey J, Ngaimisi E, et al. Copy number variation of Fc gamma receptor genes in HIV‐infected and HIV‐tuberculosis co‐infected individuals in sub‐Saharan Africa. PLoS One. 2013;8(11):e78165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Breunis WB, van Mirre E, Bruin M, et al. Copy number variation of the activating FCGR2C gene predisposes to idiopathic thrombocytopenic purpura. Blood. 2008;111(3):1029‐1038. [DOI] [PubMed] [Google Scholar]
  • 59. Hayes JM, Cosgrave EF, Struwe WB, et al. Glycosylation and Fc receptors. Curr Top Microbiol Immunol. 2014;382:165‐199. [DOI] [PubMed] [Google Scholar]
  • 60. Patel KR, Roberts JT, Subedi GP, Barb AW. Restricted processing of CD16a/Fc gamma receptor IIIa N‐glycans from primary human NK cells impacts structure and function. J Biol Chem. 2018;293(10):3477‐3489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Subedi GP, Barb AW. CD16a with oligomannose‐type N‐glycans is the only "low‐affinity" Fc gamma receptor that binds the IgG crystallizable fragment with high affinity in vitro. J Biol Chem. 2018;293(43):16842‐16850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Ferrara C, Grau S, Jager C, et al. Unique carbohydrate‐carbohydrate interactions are required for high affinity binding between FcgammaRIII and antibodies lacking core fucose. Proc Natl Acad Sci USA. 2011;108(31):12669‐12674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Sondermann P, Huber R, Oosthuizen V, Jacob U. The 3.2‐A crystal structure of the human IgG1 Fc fragment‐Fc gammaRIII complex. Nature. 2000;406(6793):267‐273. [DOI] [PubMed] [Google Scholar]
  • 64. Ferrara C, Stuart F, Sondermann P, Brunker P, Umana P. The carbohydrate at FcgammaRIIIa Asn‐162. An element required for high affinity binding to non‐fucosylated IgG glycoforms. J Biol Chem. 2006;281(8):5032‐5036. [DOI] [PubMed] [Google Scholar]
  • 65. Patel KR, Nott JD, Barb AW. Primary human natural killer cells retain proinflammatory IgG1 at the cell surface and express CD16a glycoforms with donor‐dependent variability. Mol Cell Proteomics. 2019;18(11):2178‐2190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Patel KR, Roberts JT, Barb AW. Allotype‐specific processing of the CD16a N45‐glycan from primary human natural killer cells and monocytes. Glycobiology. 2020;30(7):427‐432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Roberts JT, Patel KR, Barb AW. Site‐specific N‐glycan analysis of antibody‐binding fc gamma receptors from primary human monocytes. Mol Cell Proteomics. 2020;19(2):362‐374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Washburn N, Meccariello R, Duffner J, et al. Characterization of endogenous human FcgammaRIII by mass spectrometry reveals site, allele and sequence specific glycosylation. Mol Cell Proteomics. 2019;18(3):534‐545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Wang TT, Sewatanon J, Memoli MJ, et al. IgG antibodies to dengue enhanced for FcgammaRIIIA binding determine disease severity. Science. 2017;355(6323):395‐398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Thulin NK, Brewer RC, Sherwood R, et al. Maternal anti‐dengue igg fucosylation predicts susceptibility to dengue disease in infants. Cell Rep. 2020;31(6):107642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Bournazos S, Vo HTM, Duong V, et al. Antibody fucosylation predicts disease severity in secondary dengue infection. Science. 2021;372(6546):1102‐1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Chakraborty S, Gonzalez J, Edwards K, et al. Proinflammatory IgG Fc structures in patients with severe COVID‐19. Nat Immunol. 2021;22(1):67‐73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Larsen MD, de Graaf EL, Sonneveld ME, et al. Afucosylated IgG characterizes enveloped viral responses and correlates with COVID‐19 severity. Science. 2021;371(6532):eabc8378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Chakraborty S, Gonzalez JC, Sievers BL, et al. Early non‐neutralizing, afucosylated antibody responses are associated with COVID‐19 severity. Sci Transl Med. 2022;14(635):eabm7853. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Kapur R, Kustiawan I, Vestrheim A, et al. A prominent lack of IgG1‐Fc fucosylation of platelet alloantibodies in pregnancy. Blood. 2014;123(4):471‐480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Larsen MD, Lopez‐Perez M, Dickson EK, et al. Afucosylated Plasmodium falciparum‐specific IgG is induced by infection but not by subunit vaccination. Nat Commun. 2021;12(1):5838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Ackerman ME, Crispin M, Yu X, et al. Natural variation in Fc glycosylation of HIV‐specific antibodies impacts antiviral activity. J Clin Invest. 2013;123(5):2183‐2192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Lu LL, Chung AW, Rosebrock TR, et al. A functional role for antibodies in tuberculosis. Cell. 2016;167(2):433‐443 e414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Bournazos S, Wang TT, Ravetch JV. The role and function of Fcgamma receptors on myeloid cells. Microbiol Spectr. 2016;4(6). doi: 10.1128/microbiolspec.MCHD-0045-2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Golay J, Valgardsdottir R, Musaraj G, Giupponi D, Spinelli O, Introna M. Human neutrophils express low levels of FcgammaRIIIA, which plays a role in PMN activation. Blood. 2019;133(13):1395‐1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Wang Y, Jonsson F. Expression, role, and regulation of neutrophil Fcgamma receptors. Front Immunol. 2019;10:1958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Chauhan AK, Chen C, Moore TL, DiPaolo RJ. Induced expression of FcgammaRIIIa (CD16a) on CD4+ T cells triggers generation of IFN‐gammahigh subset. J Biol Chem. 2015;290(8):5127‐5140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Roy Chowdhury R, Vallania F, Yang Q, et al. A multi‐cohort study of the immune factors associated with M. tuberculosis infection outcomes. Nature. 2018;560(7720):644‐648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bjorkstrom NK, Gonzalez VD, Malmberg KJ, et al. Elevated numbers of Fc gamma RIIIA+ (CD16+) effector CD8 T cells with NK cell‐like function in chronic hepatitis C virus infection. J Immunol. 2008;181(6):4219‐4228. [DOI] [PubMed] [Google Scholar]
  • 85. Ziegler‐Heitbrock L, Ancuta P, Crowe S, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):e74‐e80. [DOI] [PubMed] [Google Scholar]
  • 86. Victor AR, Weigel C, Scoville SD, et al. Epigenetic and posttranscriptional regulation of CD16 expression during Human NK Cell development. J Immunol. 2018;200(2):565‐572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Ong SM, Teng K, Newell E, et al. A novel, five‐marker alternative to CD16‐CD14 gating to identify the three human monocyte subsets. Front Immunol. 2019;10:1761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Wong KL, Tai JJ, Wong WC, et al. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood. 2011;118(5):e16‐e31. [DOI] [PubMed] [Google Scholar]
  • 89. Tak T, Drylewicz J, Conemans L, et al. Circulatory and maturation kinetics of human monocyte subsets in vivo. Blood. 2017;130(12):1474‐1477. [DOI] [PubMed] [Google Scholar]
  • 90. Rogacev KS, Zawada AM, Hundsdorfer J, et al. Immunosuppression and monocyte subsets. Nephrol Dial Transplant. 2015;30(1):143‐153. [DOI] [PubMed] [Google Scholar]
  • 91. Bruggeman CW, Houtzager J, Dierdorp B, et al. Tissue‐specific expression of IgG receptors by human macrophages ex vivo. PLoS One. 2019;14(10):e0223264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Zhao L, Huang X, Hong W, et al. Slow resolution of inflammation in severe adult dengue patients. BMC Infect Dis. 2016;16:291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody‐dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect Dis. 2010;10(10):712‐722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Guzman MG, Alvarez M, Halstead SB. Secondary infection as a risk factor for dengue hemorrhagic fever/dengue shock syndrome: an historical perspective and role of antibody‐dependent enhancement of infection. Arch Virol. 2013;158(7):1445‐1459. [DOI] [PubMed] [Google Scholar]
  • 95. Srikiatkhachorn A, Green S. Markers of dengue disease severity. Curr Top Microbiol Immunol. 2010;338:67‐82. [DOI] [PubMed] [Google Scholar]
  • 96. Friberg H, Beaumier CM, Park S, et al. Protective versus pathologic pre‐exposure cytokine profiles in dengue virus infection. PLoS Negl Trop Dis. 2018;12(12):e0006975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Merad M, Blish CA, Sallusto F, Iwasaki A. The immunology and immunopathology of COVID‐19. Science. 2022;375(6585):1122‐1127. [DOI] [PubMed] [Google Scholar]
  • 98. Merad M, Subramanian A, Wang TT. An aberrant inflammatory response in severe COVID‐19. Cell Host Microbe. 2021;29(7):1043‐1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Del Valle DM, Kim‐Schulze S, Huang HH, et al. An inflammatory cytokine signature predicts COVID‐19 severity and survival. Nat Med. 2020;26(10):1636‐1643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Hoepel W, Chen HJ, Geyer CE, et al. High titers and low fucosylation of early human anti‐SARS‐CoV‐2 IgG promote inflammation by alveolar macrophages. Sci Transl Med. 2021;13(596):1‐16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Wilk AJ, Lee MJ, Wei B, et al. Multi‐omic profiling reveals widespread dysregulation of innate immunity and hematopoiesis in COVID‐19. J Exp Med. 2021;218(8):1‐29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Meizlish ML, Pine AB, Bishai JD, et al. A neutrophil activation signature predicts critical illness and mortality in COVID‐19. Blood Adv. 2021;5(5):1164‐1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Kuchroo M, Huang J, Wong P, et al. Multiscale PHATE identifies multimodal signatures of COVID‐19. Nat Biotechnol. 2022;40:681‐691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Merad M, Martin JC. Pathological inflammation in patients with COVID‐19: a key role for monocytes and macrophages. Nat Rev Immunol. 2020;20(6):355‐362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Valdebenito S, Bessis S, Annane D, et al. COVID‐19 lung pathogenesis in SARS‐CoV‐2 autopsy cases. Front Immunol. 2021;12:735922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Delorey TM, Ziegler CGK, Heimberg G, et al. COVID‐19 tissue atlases reveal SARS‐CoV‐2 pathology and cellular targets. Nature. 2021;595:107‐113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Smith P, DiLillo DJ, Bournazos S, Li F, Ravetch JV. Mouse model recapitulating human Fcgamma receptor structural and functional diversity. Proc Natl Acad Sci USA. 2012;109(16):6181‐6186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Borczuk AC, Salvatore SP, Seshan SV, et al. COVID‐19 pulmonary pathology: a multi‐institutional autopsy cohort from Italy and New York City. Mod Pathol. 2020;33(11):2156‐2168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Schurink B, Roos E, Radonic T, et al. Viral presence and immunopathology in patients with lethal COVID‐19: a prospective autopsy cohort study. Lancet Microbe. 2020;1(7):e290‐e299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Ouwendijk WJD, Raadsen MP, van Kampen JJA, et al. High levels of neutrophil extracellular traps persist in the lower respiratory tract of critically Ill patients with coronavirus disease 2019. J Infect Dis. 2021;223(9):1512‐1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Nathan C. Neutrophils and COVID‐19: nots, NETs, and knots. J Exp Med. 2020;217(9):1‐3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Chen ST, Park MD, Del Valle DM, et al. Shift of lung macrophage composition is associated with COVID‐19 disease severity and recovery. bioRxiv. 2022. doi: 10.1101/2022.01.11.475918 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Lauc G, Huffman JE, Pucic M, et al. Loci associated with N‐glycosylation of human immunoglobulin G show pleiotropy with autoimmune diseases and haematological cancers. PLoS Genet. 2013;9(1):e1003225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Thanabalasingham G, Huffman JE, Kattla JJ, et al. Mutations in HNF1A result in marked alterations of plasma glycan profile. Diabetes. 2013;62(4):1329‐1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Klaric L, Tsepilov YA, Stanton CM, et al. Glycosylation of immunoglobulin G is regulated by a large network of genes pleiotropic with inflammatory diseases. Sci Adv. 2020;6(8):eaax0301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Norton PA, Mehta AS. Expression of genes that control core fucosylation in hepatocellular carcinoma: systematic review. World J Gastroenterol. 2019;25(23):2947‐2960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Mijakovac A, Juric J, Kohrt WM, et al. Effects of estradiol on immunoglobulin G glycosylation: mapping of the downstream signaling mechanism. Front Immunol. 2021;12:680227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Engdahl C, Bondt A, Harre U, et al. Estrogen induces St6gal1 expression and increases IgG sialylation in mice and patients with rheumatoid arthritis: a potential explanation for the increased risk of rheumatoid arthritis in postmenopausal women. Arthritis Res Ther. 2018;20(1):84. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing not applicable ‐ no new data generated, or the article describes entirely theoretical research.


Articles from Immunological Reviews are provided here courtesy of Wiley

RESOURCES