ABSTRACT
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages’ inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
KEYWORDS: macrophages, macrophage polarization, tissue microenvironment, innate immunity, M1, M2, M1/M2, coagulation
INTRODUCTION
Macrophages are among the first immune cells to respond to foreign invaders, damaged tissues, or tumor cells (1–3). Macrophages are responsible for phagocytizing and killing pathogens, clearing dead cells, and healing damaged tissue (1–4).
Based on their origin, macrophages are differentiated into tissue-resident macrophages (TRMs) or monocyte-derived macrophages (MDMs). TRMs originate during embryonic development and proliferate in the tissue (Fig. 1.1) (5, 6). On the other hand, MDMs are generated from peripheral blood monocytes which are recruited into damaged and/or infected tissue (Fig. 1.2 and 1.3) (7, 8). Macrophages exhibit plasticity by polarizing into a continuum of pro- (M1-like) and anti-inflammatory (M2-like) phenotypes upon exposure to microenvironmental cues (Fig. 1.4) (9, 10). Macrophages display receptors that recognize these cues in the TME, which ultimately induce polarization and modulate M1-like microbicidal and M2-like repair responses (Fig. 1.4) (9, 11–16).
Fig 1.
Tissue microenvironment signals induce M1 and M2 macrophage polarization on a spectrum. (Step 1) In the initial response to infection and/or damage, tissue-resident macrophages (TRMɸs) serve as sentinels by sensing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) to stimulate CCL2 for monocyte recruitment. (Step 2) Monocytes extravasate into the tissue where (Step 3) Macrophage-colony stimulating factor (M-CSF) differentiates monocytes into naive (M0) monocyte-derived macrophages (MDMɸs). (Step 4) Factors including PAMPs, DAMPs, and cytokines produced by TH1 and TH2 cells then induce M1 and M2 polarization on a spectrum. IFN-γ, LPS, and GM-CSF induce M1-like polarization leading to a signaling cascade that enables transcription factors STAT1, activator protein-1 (AP-1), NFκB, IRF3, and IRF5 to activate proinflammatory genes. Cytokines and chemokines produced by M1s activate CD8+ cytotoxic T cells (Tc), TH1, natural killer (NK) cells, and neutrophils (PMN). Virus-infected macrophages can also present viral antigens on their major histocompatibility complex-I (MHC-I) to TC cells, resulting in granzyme release that induces apoptosis. Macrophages infected with bacteria or fungi can present their antigen on MHC-II to TH1 cells resulting in IFN-γ secretion. IFN-γ promotes phagocytosis and upregulates iNOS to produce NOs degrading the pathogen. IL-4, IL-13, immune complexes, and glucocorticoids induce M2-like polarization. For M2-like polarization, signaling cascades activate transcription factors IRF4, STAT3, STAT6, PPARɑ, and CCAAT/enhancer-binding protein beta (C/EBPβ) to induce anti-inflammatory genes. Cytokines and chemokines produced by M2s dampen M1s, NKs, and PMNs inflammatory response. Furthermore, these factors activate TH2 and regulatory T cells (Treg) to secrete cytokines to suppress phagocytosis, ultimately resulting in pathogen survival and persistence. (Step 5) At the same time, endothelial cells produce IL-6, which stimulates hepatocytes to produce Fg and infiltrate into the tissue to form fibrin clots by proteolytic activity of thrombin for tissue repair.
M1-like macrophages generally secrete proinflammatory cytokines, produce high amounts of reactive nitrogen and oxygen species for antimicrobial and tumoricidal responses, activate inflammatory T-helper-1 (TH1), and promote robust phagocytic ability (Fig. 1.4). By contrast, M2-like generally resolve inflammatory responses, by phagocytizing apoptotic cells, promoting tissue repair and remodeling, as well as inducing anti-parasitic responses (Fig. 1.4) (4, 9, 15, 17, 18). Based on changes in the TME, macrophage polarization is a highly dynamic process and can switch between different phenotypes beyond the M1-M2 dichotomy (10, 19). With opposing activities of killing and repairing, it is critical to tightly regulate M1-M2 polarization to prevent disease since dysfunctional M1-M2 polarization is associated with adverse pathologies (20). For example, sustained M1-like polarization is implicated in inflammatory diseases such as acute rheumatoid arthritis, systemic lupus erythematosus, inflammatory bowel diseases, atherosclerosis, and even exacerbating SARS-CoV-2 infection (COVID-19) (21–23). Furthermore, excessive M2-like polarization is associated with cancers and asthma and promotes bacterial and fungal infections (24, 25). Interestingly, macrophage response plays a critical role in responding to vascular and tissue injury by interacting with coagulation factors to carefully orchestrate inflammation and wound healing (26). Thus, macrophage dysfunction can contribute to promoting coagulopathy-associated diseases, especially hemophilic and thrombotic diseases (26). Therefore, there is a need to better understand how macrophage polarization is modulated by coagulation factors in both wound healing and coagulopathies, which this review will discuss.
Taking advantage of macrophage plasticity, therapies have aimed at repolarizing macrophages between M1s and M2s to generate immunotherapeutic strategies to improve clinical outcomes in favor of the patients (27). However, to optimize this therapeutic approach, it is crucial to understand the mechanisms of how the tissue microenvironment (TME) during diseases drives macrophage polarization toward clinically unfavorable outcomes. In this review, we will discuss how infections and coagulation modulate macrophage polarization in the TME. Thus, this review will provide a better understanding of the dynamics of macrophage polarization within TMEs in diseases, which is important in the future development of therapies.
INDUCTION OF MACROPHAGE POLARIZATION
TRMs are immune sentinels that detect pathogen-associated (PAMPs) and damage-associated (DAMPs) molecular patterns. Upon recognition of PAMPs and DAMPs, TRMs produce cytokines and chemokines such as CXCL1 and CXCL2 for neutrophil recruitment and CCL2 for monocyte infiltration to damaged/infected tissue (Fig. 1.1) (15, 28–30). During acute inflammation, monocytes are recruited from the bone marrow into affected tissues (Fig. 1.2), which then differentiate into MDMs upon exposure to macrophage-colony stimulating factor (M-CSF/CSF-1) (Fig. 1.3) (31). In this altered TME, both TRMs and MDMs are exposed to cues that then spatiotemporally polarize these macrophages to a spectrum of phenotypes tailored to specific functions (Fig. 1.4). Upon receiving these cues, intracellular signaling cascades will alter the transcriptional programming, promoting specific metabolic and enzymatic pathways needed for M1-like proinflammatory or M2-like anti-inflammatory response. To understand this polarization, we discuss how the spectrum of phenotypes is induced and regulated and the challenges of describing macrophage polarization.
Induction of M1 macrophages
M1-like macrophages generally have a proinflammatory activation pattern where they (i) secrete inflammatory cytokines and chemokines; (ii) produce reactive oxygen (ROS) and nitrogen species (RNS); and (iii) promote antigen presentation to activate TH1 response for anti-tumorigenic and antimicrobial activity (9, 10, 15). Depending on the stimulus in the TME, macrophages can polarize toward M1s displaying a spectrum of proinflammatory phenotypes (32). Discoveries made from in vitro studies using human and murine macrophage-like cell lines and ex vivo studies using bone marrow-derived macrophages (BMDMs), murine peritoneal macrophages, and human macrophages from peripheral blood mononuclear cells led to the realization that different inducers can polarize macrophages on a spectrum of M1-like phenotypes (32). Each activating inducer and its corresponding murine and human markers are described in specific detail in the nomenclature and experimental guidelines by Murray et al (32).
It has been established that stimulation of cultured murine and human peripheral blood macrophages with interferon-γ (IFN-γ) exert a clear-cut M1 polarization mediated by signal transducers and activators of transcription 1 (STAT1) and interferon regulatory factors (IRF)-1, -5, and -8 signaling (32–34). Specifically in murine macrophages, IFN-γ binding to IFNGR receptor activates JAK1/JAK2 to promote STAT1 binding to gamma interferon activation site (GAS) elements in the promoter of the Nos2 gene (35–37). This pathway leads to M1s expressing a murine-specific marker inducible nitric oxide synthase (iNOS), which metabolizes macrophages’ arginine to nitric oxides (NOs) to kill the phagocytized pathogen (35–37). In addition to NO production, all macrophages express the NADPH oxidase complex (NOX2), which catalyzes superoxide production upon IFN-γ activation (38, 39). The generation of these superoxides is important for producing secondary products, such as hydrogen peroxide and hydroxyl radicals, which possess stronger bactericidal effects (38). For this to occur, activation of NOX2 requires translocation of subunits p40phox, p47phox, p67phox, and Rac from the cytosol to the integral membrane heterodimer Flavocytochrome b558 (38). Flavocytochrome b558 consists of two membrane subunits gp91phox and p22phox that mediates electron transfer from NADPH to molecular oxygen to produce superoxides (38). In murine and human macrophages, IFN-γ promotes trafficking of gp91phox and p22phox to the plasma membrane through the endocytic pathway resulting in increased superoxide production (39). Aside from iNOS and NOX activation, IFN-γ activates STAT1 and NF-κB in separate pathways to also produce inflammatory cytokines and chemokines including CXCL9-11, CCL5, interleukin (IL)−1β, IL-6, IL-12, IL-18, IL-23, and TNF-ɑ (9, 10, 15).
M1s are activated by the interaction of the toll-like receptor (TLR)-4 and co-receptor CD14 with lipopolysaccharide (LPS; Gram-negative bacterial cell wall component), which then induces nuclear factor-kappa B (NF-κB), activating protein-1 (AP-1), and interferon regulatory factor (IRF)−3 (Fig. 1.4) (10). Furthermore, TLR-4 signaling can also induce IRF3, which then releases IFN-ɑ and -β (40–43). Much like exposure to IFN-γ, stimulation with LPS alone or in combination with IFN-γ will also produce these proinflammatory cytokines and chemokines (15). Common with both inducers is NF-κB activation, which is a key transcription factor of M1 polarization and induces TNF-ɑ, IL-1β, IL-6, IL-12, cyclooxygenase-2 (COX-2), hypoxia-inducible factor 1ɑ (HIF-1ɑ), and suppressor of cytokine signaling 3 (SOCS3) (41, 42). These secreted or expressed factors are known to inhibit production of cytokines, chemokines, and transcription factors that promote M2 polarization (9, 10).
Also, granulocyte macrophage-colony stimulating factor (GM-CSF), produced by fibroblasts, endothelial cells, recruited innate immune cells, and T cells, can shift macrophages toward the M1-like phenotype as inflammation or infection progresses (31). In response to infection or tissue damage, both M-CSF and GM-CSF induce macrophage differentiation (31). During homeostasis, M-CSF is ubiquitously produced by tissue-resident cells to differentiate newly recruited monocytes into macrophages upon initial infection and tissue injury (31). As inflammatory conditions prolong, GM-CSF levels then quickly increases to stimulate recruitment, survival, and activation of myeloid cells, particularly M1 polarization (31, 44). GM-CSF is recognized by macrophages’ GM-CSF receptor (CSF-2R), which activates IRF5 to drive the secretion of IL-12 and IL-23, promoting inflammatory TH1 and TH17 cell response (45). In an inflamed TME, GM-CSF plays a role in regulating macrophage antimicrobial defense and wound healing (31, 46–48). For example, in an inflammatory bowel disease mouse model, it was shown that the group 3 innate lymphoid cells were the major producers of GM-CSF, leading to induce M1 polarization and bactericidal response against Citrobacter rodentium while suppressing M2 pro-fibrotic response (48).
Induction of M2 macrophages
In contrast to M1s, M2-like macrophages are critical for resolving inflammation by (i) suppressing proinflammatory response; (ii) activating anti-inflammatory TH2 cells; (iii) clearing apoptotic cells from damaged tissue; and (iv) repairing and remodeling tissue to restore homeostasis (49). Moreover, based on stimuli in the TME, macrophages can polarize to diverse M2-like phenotypes with different functions in suppressing proinflammatory response to promote wound healing (Fig. 1.4) (49, 50).
Much like how IFN-γ induces M1s, IL-4 and/or IL-13 exert an antagonistic effect on IFN-γ to promote M2 polarization mediated by STAT6 signaling (32, 51). STAT6 activation, in turn, then induces the expression of CD206 (Mannose receptor C-type 1, Mrc1) and CD163 (scavenger receptor for hemoglobin and haptoglobin) on both human and mouse macrophages (10, 52). Furthermore, mouse genetic studies investigating M2 polarization found expression of murine-specific M2 markers such as Chitinase 3-like 3 (Chi3l3, Ym1; also known as eosinophil chemotactic factor-lymphocyte), Arginase-1 (Arg1), and Resistin-like ɑ (Retnla, Fizz1) (25, 53, 54). In addition, STAT6 cooperates with transcriptional factor KLF4 to antagonize the M1-mediated STAT1 signaling pathway, thereby activating anti-inflammatory genes and leading to the production of IL-10, transforming growth factor (TGF)-β, CCL17-18, CCL22, and CCL24 (10, 15, 52). This activation leads macrophages to have a wound-healing phenotype with the ability to promote endocytic activity, cell growth, and tissue repair by secreting collagen precursors, stimulating fibroblasts, and secreting platelet-derived growth factor (PDGF) (49, 50). They also produce high levels of Arginase-1 (Arg1), competing with iNOS for arginine to produce polyamines needed for tissue repair (49).
Interestingly, M2-like polarization can also be induced by a combination of immune complexes with LPS, TLR, and IL-1R agonists leading to a subset that expresses both pro- and anti-inflammatory markers, such as iNOS, Arg1, CD86, CD68, MHCII, and CD163 (52, 55). In addition, they also produce anti-inflammatory IL-10 and CCL1 to suppress inflammation by promoting TH2 and Treg cell responses (52). M2s induced by immune complexes are found to have immunoregulatory effects with protective and pathogenic roles depending on TME (52, 55). For example, a study showed that macrophages induced with both LPS and IgG antibodies resulted in M2 macrophages with pro- and anti-inflammatory profiles (56, 57). These specific M2-like macrophages were shown to have a role in preventing excessive fibrosis ex vivo and in vivo, thereby reducing tissue injury in a myocardial ischemia in vivo model (56, 57).
M2-like macrophages can also display a pro-resolving phenotype induced by glucocorticoids (GCs) (52). GCs, including cortisol, dexamethasone, and prednisone, have a potent ability to suppress proinflammatory responses through nuclear glucocorticoid receptors (GR) in macrophages (58). GCs, with their lipophilic structure, penetrate the plasma membrane and bind to the cytosolic GR forming a GC/GR complex that will then translocate into the nucleus (59). This complex will then bind to GC response elements (GREs) in the promoter regions of GC-associated genes that promote anti-inflammatory response (59). To inhibit pro-inflammatory genes, the GC/GR complex can either bind to “negative” GREs upstream of pro-inflammatory genes or directly interact (or tether) with transcription factors to prevent activation (59). Through this mechanism, GCs inhibit transcription of M1-producing cytokine genes Il1b, Il6, Il12, and tnf and downregulate iNOS and COX-2 in both human and mouse macrophages (59, 60). GCs are found to induce M2 polarization similar to macrophages treated with IL-4 and IL-13, which enable IL-10 and TGF-β production and upregulates both mannose and CD163 receptor (59). Together, this allows these macrophages to resolve inflammation by promoting phagocytosis of apoptotic cells during tissue repair (59).
In summary, it is critical to understand the TMEs and their diverse signals to further dissect macrophage polarization, function, and their role in disease outcome, since dysregulated polarization may result in exacerbated pathologies.
Challenges to describing macrophage polarization
Recently, researchers in the macrophage biology field acknowledged the challenges to describing macrophage polarization due to the lack of consensus on standardizing how to define polarization states and their resulting phenotypes in vitro, ex vivo, and in vivo experiments (32). First, the widespread use of the M1/M2 dichotomy or “classical”/“alternative” activation over decades and to this day pushes the oversimplification that macrophage polarization occurs on these two extreme ends of the spectrum, where in reality it does not. “Classical” and “alternative” activation terms first encompassed from in vitro studies that demonstrated IFN-γ and IL-4 differentially polarize macrophages to pro- or anti-inflammatory phenotypes, respectively (61, 62). Several years later, it was discovered that peritoneal macrophages isolated from C57BL/6 mice (mice with elevated proinflammatory T-helper-1 (TH1) response) produced higher bactericidal NOs when stimulated with either IFN-γ or LPS in vitro (63). By contrast, macrophages isolated from BALB/C mice (mice with higher anti-inflammatory TH2 response) yielded low amounts of NOs when stimulated with IFN-γ and/or LPS (63). Importantly, it was found that murine peritoneal macrophages from these two different strains showed opposite metabolic pathways on arginine: where macrophages from C57BL/6 mice metabolize arginine to NOs via iNOS, while macrophages from BALB/C mice metabolize arginine to ornithine via Arg1. Due to these observations, the M1-M2 dichotomy was initially proposed, which mirrored the TH1/TH2 responses in that TH1 cells’ production of IFN-γ promotes M1s, while TH2 cells produce IL-4 to polarize M2s (63). Although now it is known that sources of IFN-γ and IL-4 can come from other immune cells, especially innate lymphoid cells (64).
Further advancement through in vitro studies on murine and human macrophages with different stimuli revealed diverse phenotypes that led to the development of subsets, such as M2a, M2b, M2c, and M2d to name a few examples (52, 65). However, this nomenclature has generated inconsistencies and confusion on which cell surface, intracellular, and secreted marker bins macrophages into these subsets. In addition, it is difficult to determine whether macrophages binned into these subsets are terminally differentiated into these subsets and whether they are indeed functionally distinct from each other. To address the pitfalls in describing macrophage polarization, a group of experts in the macrophage biology field met at the International Congress of Immunology in 2013 and provided nomenclature and experimental guidelines, which are summarized in reference (32).
Although in vitro studies have contributed to defining M1 and M2 phenotypes, the major challenge is that activation signatures defined in vitro can be modulated by mixed signals that are overlooked in vivo. Therefore, translating in vitro results to in vivo mouse and human disease models is problematic due to the complexity of factors in the TME that polarize macrophages beyond the M1 and M2 extremes. Considering that macrophages display plasticity, changes in the TME will cause macrophages to transition into intermediate states between M1s and M2s. Thus, the limitation of the simple bipolar M1-M2 terminology is that these two states do not exist alone in TMEs. Hence, in this review, we describe M1s and M2s as general activation patterns and acknowledge that a continuum of phenotypes exists in vivo and in human disease studies.
MACROPHAGE POLARIZATION DURING INFECTIONS
During infections, macrophages enhance their antimicrobial responses using their repertoire of pattern recognition receptors (PRRs) to engage with pathogens directly, or indirectly with opsonins, inducing M1 polarization and phagocytosis (Fig. 1.4) (66). Macrophages then amplify their antimicrobial response by producing highly reactive ROS and RNS, activating TH1 and TH17 cells via antigen presentation, and secreting proinflammatory cytokines to promote immune cell recruitment (15, 24, 67). Due to M1’s antimicrobial responses, some pathogens have developed immune evasion mechanisms that manipulate macrophage polarization and function, thereby preventing phagocytosis and killing to promote colonization and infection (25).
Viral infections
Viruses also take advantage of M1-M2 polarization to promote their virulence and infection (Fig. 2). When macrophages encounter viruses, macrophages polarize to M1s, which leads to activating IRFs, producing type I interferons (IFN-ɑ and -β), promoting natural killer cells (NK) and T cell activation (10, 68). In addition, M1s utilize intracellular RIG-I-Like receptors (RLRs) to recognize viral RNA, leading to the induction of IFNs and IFN-stimulated genes (69). For example, in a mouse West Nile Virus infection model, upon RLRs recognition of viral RNAs, M1 programming is activated to control the infection (70).
Fig 2.
Pathogens can modulate macrophage polarization leading to microbial clearance or survival. Bacteria, viruses, and fungi species can promote macrophages toward (A) M1 or (B) M2 polarization on a continuum affecting the outcome of infection.
Although M1s are essential in acute infection, excessive M1 function by prolonged inflammatory cytokine secretion results in immune hyperactivation, tissue damage, organ failure, viral persistence, and death (71). During the COVID-19 pandemic, in most cases of SARS-CoV-2 infections, patients were able to clear the infection, which correlated with the presence of IFNs, IL-1β, IL-6, and TNF-ɑ (23, 72). Contrarily, hospitalized patients with excessively high levels of IL-6 and TNF-ɑ in serum correlated with severe disease, including complications of pneumonia and death (72). It has been speculated that the hyperinflammatory signature of the airway myeloid cells promotes an excessive M1 function, which perpetuates immune cell recruitment and lung inflammation, leading to severe disease (73). Since various immune cells produce these cytokines, it is difficult to discern if M1s are solely responsible for manifesting cytokine release syndrome (termed as “cytokine storms”).
Viruses can also modulate multiple macrophage polarization states to promote viral dissemination. Viruses such as Hepatitis B, Hepatitis C, and Epstein-Barr Virus (EBV) can induce M2 polarization (Fig. 2B) (74–77). Human cytomegalovirus (HCMVs) is able to establish latency after initial infection (78). HCMVs target monocytes for viral entry and promote monocyte survival by differentiating them into hybrid M1/M2 macrophages (78, 79). This atypical M1 polarization is characterized by expressing an M1 transcriptional profile but also upregulating anti-inflammatory factors such as IL-1R antagonists and IL-10. This hybrid phenotype results in pathogen clearance reduction, promoting viral entry, intracellular survival, and transendothelial migration (79). Another study further confirmed that different HCMV strains induced a PI3K-SHIP1-Akt signaling pathway in monocytes, resulting in a hybrid M1/M2 phenotype (80).
A way for the host to orchestrate M1/M2 response during viral infection is by sending signals that reduce M1 inflammatory cytokine secretion while promoting M2 functions in repairing damaged tissue to restore homeostasis. During the repair phase, apoptotic cell clearance (efferocytosis) by macrophages activates anti-inflammatory programming, which is triggered by degradation products of apoptotic cells (ACs) within phagolysosomes (81). This orchestration into M2s is critical to prevent inflammation-induced tissue damage. Interestingly, a study found that M2s that engulfed SARS-CoV-2-infected-ACs reduced expression of genes associated with tissue remodeling and immunoregulatory functions including ccl18, cd206, mmp9, pparg, and cd163, but increased expression and robust secretion of IL-6 and IL-1β, which promotes inflammation (81). Of importance, CD206 (Mrc1), a mannose receptor on M2s, was decreased on these specific macrophages and impaired efferocytosis, which may provide an explanation to viral persistence in severe COVID-19 (81).
Moreover, the presence of anti-inflammatory mediators during acute viral infection renders the host susceptible to developing secondary bacterial infections (82). Recently, a study identified that excessive induction of the anti-inflammatory CYP450 lipid metabolite activates the peroxisome proliferator-activated receptor (PPAR) via transcription factor PPARɑ (83, 84). This signaling dampened TLR stimulation, leading to suppression of NFκB-mediated response in a mouse model of secondary S. aureus lung infection post-influenza (83, 84). Furthermore, PPARɑ activation promoted M2 polarization and correlated with inhibiting S. aureus clearance (84). These findings demonstrate that viruses actively capitalize on both M1s and M2s to promote persistent infection.
Bacterial infections
M1 targets pathogens during acute phase of infection by phagocytosing and degrading bacteria through ROS and RNS within phagosomes (Fig. 1.4). Specific pathogens are known to induce M1 polarization including E. coli, Salmonella typhi, Helicobacter pylori, Enterococcus faecalis, Staphylococcus aureus, Listeria monocytogenes, Mycobacterium tuberculosis, and Mycobacterium ulcerans (Fig. 2A) (85–91).
LPS is among the virulence factors that influence macrophage polarization, which is a major component of the outer membrane of gram-negative bacteria that is recognized by TLR-4 and its coreceptor CD14 and MD-2 (92). Pathogens such as E. coli, S. typhi, and H. pylori possess LPS, which activates NF-κB, induces proinflammatory cytokine production, promotes phagocytosis, and enhances intracellular pathogen degradation through TLR-4/CD14 signaling (88, 93, 94). In an uncomplicated urinary tract infection mouse model, LPS from uropathogenic E. coli (UPEC) upregulates expression of CD14 for binding on macrophages to induce antibacterial responses via NFκB and p38 mitogen-activated protein kinase resulting in decreased UPEC burden in bladders (95). In the same study, UPEC infection was exacerbated in CD14-deficient mice bladders, which was associated with a decrease in CD14-dependent pathways including immune cell trafficking, proinflammatory cytokine production in macrophages, and IL-17 signaling (95). Thus, CD14 as a TLR-4 coreceptor is pivotal in amplifying M1-like macrophage response.
In an active infection in the TME, invasive pathogens have distinct ways to modify the host extracellular matrix (ECM) by (i) producing tissue-degrading enzymes called invasins; (ii) hijacking host proteolytic systems; or (iii) capitalizing on host immune cells’ production of proteases to facilitate migration and colonization throughout the tissue (96, 97). This ECM degradation enables macrophages to both recognize pathogens through its PRRs and sense physical alterations in the ECM through their mechanosensitive ion channels (MSICs) (96–99). Interestingly, a mechanosensitive ion channel, Piezo1, expressed on mouse macrophages, senses these alterations in the ECM stiffness, which leads to the increasing influx of Ca2+ into macrophages to promote M1-like responses, particularly inducing IFN-γ production and iNOS upregulation (98). Another group demonstrated that E. coli infection-induced TLR-4 signaling increased Piezo1 activity in mouse macrophages ex vivo and in vivo (100). LPS stimulation by E. coli infection triggered the assembly of TLR-4 and Piezo1 as a coreceptor to induce Ca2+ influx leading to cytoskeleton remodeling for phagocytosis, ROS production, and M1-like bactericidal response through the CaMKII-Mst1/2-Rac1 signaling axis (100). Deficiency in Piezo1 promotes wound healing M2-like macrophages (98). This suggests that MSICs during bacterial infection play a role in coordinating with TLRs in modulating macrophage polarization; however, this needs further evaluation.
Although several gram-negative bacterial species have LPS and other virulence features that generally promote M1 polarization through TLRs signaling, some species, particularly H. pylori and Francisella tularensis, can evade the innate immune response by displaying structural variations of LPS and flagellin that impede recognition by TLR-2, -4, and -5 on macrophages (101–103). The structure of LPS consists of a polysaccharide O-antigen chain, a core oligosaccharide, and lipid A, which serves to anchor LPS to the plasma membrane (104). Unlike E. coli LPS structure, the biosynthesis of H. pylori O-antigen leads to a unique variation called the Lewis system antigens, which has been shown to subvert host immune response (101, 105). Thus, it is this alteration in the O-antigen that causes H. pylori LPS to antagonize TLR-4 preventing NF-κB activation in human macrophages (106, 107). Although this pathogen was able to be phagocytosed by macrophages, H. pylori is still highly resistant to killing within phagosomes by preventing phagosome maturation, utilizing bacterial Arg1 to outcompete macrophages’ iNOS for arginine, and inducing mitochondrial-associated apoptosis (108–111).
Another evasion strategy that bacteria utilize is to drive macrophages toward M2 polarization (Fig. 2B) (112). For example, S. enterica serovar Typhimurium, a foodborne pathogen, produces the effector protein, SteE, to induce M2 polarization (112, 113). SteE activates STAT3, which increases IL-10 levels resulting in bacterial intracellular replication in vitro and increased bacterial burden in a murine model (112, 113). Another pathogen, F. tularensis, a causative agent of the zoonotic disease tularemia, promotes M2 polarization through TLR-2 signaling to induce Arg1, allowing bacterial replication inside macrophages (114). This specific pathogen has an atypical LPS structure that renders it inert instead of serving as a potent endotoxin due to its inability to interact with host TLR-4 and its coreceptors CD14 and MD-2 (103, 115). Despite failure to induce TLR-4 signaling, F. tularensis was still able to be phagocytosed by macrophages’ mannose receptors which then formed phagosomes containing the pathogen (116). In addition, once inside the cell, F. tularensis was able to upregulate microRNA-155 to downregulate TLR adapter protein MyD88 and phosphatase SHIP-1, consequently suppressing downstream proinflammatory response in human macrophages (117). In addition, this pathogen expresses acid phosphatases that appear to enhance intracellular survival by selectively dephosphorylating the p40phox and p47phox subunits of the NADPH oxidase complex, inhibiting ROS production (118). Furthermore, F. tularensis possess the ability to selectively destabilize host mRNA encoding proinflammatory IL-1β, IL-6, and CXCL1 once the pathogen escaped from phagosomes into the cytosol of infected macrophages (119).
Fungal infections
During fungal infections, macrophages display various PRRs that are capable of recognizing β-glucans, zymosans, chitin, and ɑ-mannans on fungal cell walls to induce fungicidal response (120). M1s utilize C-type lectin receptors (CLRs), such as Dectin-1 and Dectin-2, to bind to β-glucans of fungal pathogens and induce ROS production (121, 122). In addition, recognition of fungal cell wall components by cell surface receptors such as TLR-2, TLR-4, TLR-5, Dectin-1, Dectin-2, and Mincle results in the induction of downstream activation of caspase recruitment domain-containing protein 9 (CARD9) (123, 124), which is essential in M1 activation and for induction of vaccine-mediated immunity against Cryptococcus neoformans, a causative agent of pulmonary cryptococcosis (125). Furthermore, CARD9 is also involved in activating NF-κB downstream of Retinoic acid-inducible gene I receptor (RIG-I) (126). In animal studies, CARD9 deficiency skewed macrophages to M2s, leading to decreased NO production and resulted in inability to control replication and dissemination even after vaccination against C. neoformans (125).
PPRs collaboration is critical for triggering macrophage’s inflammatory response against fungi. In the case of Histoplasma capsulatum, this fungus is recognized by both macrophage complement receptor-3 (CR3) and Dectin-1 (127). This interaction induces macrophage secretion of TNF and IL-6, facilitating host responses against disseminated histoplasmosis (127). During Candida albicans infection, CLRs Dectin-1, -2, and Mincle collaborate and activate cytokine production by inflammatory monocytes (128).
As an evasion mechanism upon phagocytosis, fungal pathogens such as C. neoformans, Talaromyces marneffei, and Candida spp. can rewire macrophages to skew toward M2s for their survival and dissemination (Fig. 2B) (129–133). For instance, an ex vivo and in vivo study found that C. neoformans enhanced its virulence by secreting CPL1 and binding to TLR-4 (132). This interaction promoted M2 polarization by propagating IL-4 signaling, which upregulated Arg1 allowing C. neoformans to replicate within macrophages (132). Furthermore, an animal study showed that oral antibiotic treatment induced overgrowth of gut commensal Candida species (129). Interestingly, the fungal overgrowth in the gut had systemic effects, resulting in increasing PGE2 plasma concentrations, which then polarized alveolar macrophages to M2s and exacerbated allergen-induced airway inflammation (129). Some reports have shown that fungi can produce PGE2 (134, 135); however, this report does not assess whether the increased levels of PGE2 is due to fungal production (134, 135) or host cells’ PGE2 production in response to the Candida overgrowth (136).
Reprogramming of metabolic pathways during macrophage polarization is key to address the energetic and anabolic needs to respond to infections. To support M1s microbicidal functions, M1s elevates glycolytic metabolism by increasing pentose phosphate pathway activity that results in the production of ROS for superoxide burst (130). It was found that the melanin on the surface of Aspergillus fumigatus spores activates HIF1ɑ-dependent glycolysis in macrophages, resulting in increased phagocytosis and pathogen clearance (137).
THE ROLE OF HEMOSTASIS AND COAGULATION IN MODULATING MACROPHAGE POLARIZATION
Role of hemostasis in tissue injury and repair
Physical trauma and infection can cause tissue injury that then initiates hemostasis, which is a mechanism that leads to the cessation of excessive bleeding from damaged blood vessels (138). Hemostasis is a complex regulated process that employs platelet adhesion and production of pro- and anti-coagulant factors, fibrinolytic proteins, chemokines, cytokines, and growth factors necessary for arresting hemorrhage and promoting tissue repair (138, 139). This process is divided into four stages: (i) vasoconstriction; (ii) platelet adhesion and aggregation; (iii) coagulation cascade activation leading to fibrin clotting (thrombosis) (Fig. 3A through C); and (iv) fibrinolysis resulting in fibrin clot dissolution (Fig. 3D) (139). Immediately after initial injury to blood vessels, the damaged vascular endothelium secretes endothelin to trigger vasoconstriction (139). At this stage, activated endothelial cells produce chemoattractants, cytokines, and adhesion molecules, such as collagen and von Willebrand Factor, that augment recruitment, adhesion, and aggregation of platelets, neutrophils, monocytes, and macrophages at the injury site (139). Mediators secreted by platelets from the damaged vessel initiate activation of the coagulation cascade that ultimately lead to fibrin formation that serve as a temporary plug for wound healing (140). To get this fibrin formation, hepatocytes in the liver first produce fibrinogen (Fg) as one of the prominent acute phase proteins that circulate in the blood to the damaged site (Fig. 1.5) (141). Through multiple triggers at these sites, a complex cascade of reactions containing coagulation factors becomes activated through the intrinsic pathway (by contact activation from damaged blood vessels) and the extrinsic pathway (induced by tissue damage) (Fig. 3A and B), which ultimately promotes proteolytic activity of thrombin to polymerize Fg into fibrin clots (Fig. 3C) (142).
Fig 3.
Vascular and tissue injury activates the coagulation cascade leading to fibrin clot formation. (A) Vascular damage activates the intrinsic pathway of the coagulation cascade by proteolytic cleavage of FXII, FXI, and FIX. Activation of FVIII produces FVIIIa to form a complex with FIXa (FIXa:FVIIIa) to subsequently cleave FX into FXa. (B) Tissue injury initiates the extrinsic pathway of the coagulation cascade by activating FIII (Tissue Factor) and FVII to generate the FIIIa:FVIIa complex. The FIIIa:FVIIa complex cleaves FX to produce FXa. (C) Both FXa and FVa activate FII (prothrombin) into FIIa (thrombin). Thrombin proteolytically cleaves soluble Fg fibrinopeptides to form fibrin fibers. FXIIIa crosslinks fibrin fibers to stabilize fibrin clots. (D) For fibrinolysis to occur, urokinase- (uPA) and tissue-plasminogen activator (tPA) activate plasminogen (PG) into plasmin to degrade fibrin clots generating fibrin degradation products. To halt the fibrinolytic process, uPA and tPA are inhibited by plasminogen activator inhibitor-I (PAI-I).
The destruction of tissue leads to alterations in the endothelial cells (ECs) and the ECM that signals monocytes and macrophages to be recruited and adhere to these injury sites (143). At these sites, macrophages serve as key regulators in the tissue healing process by properly amplifying and dampening inflammation, tissue repair/remodeling, removing apoptotic cells, and restoring tissue homeostasis (49, 144, 145). Upon initial exposure to IL-1β, IL-6, IL-8, and TNF-ɑ released by platelets, macrophages upregulate expression of tissue factor (TF) on its cell surface, which is critical in initiating the coagulation cascade (146). At the same time, macrophages itself release proinflammatory cytokines, such as IL-1, IL-6, and TNF-ɑ, to increase immune cell recruitment, upregulate adhesion molecules (i.e., selectins and integrins) to interact with ECs for transmigration into damaged tissue and promote procoagulant factors (26). Subsequently, thrombin released by platelets cleaves G-protein-coupled protease-activated receptors (PARs)-1, -3, and -4 on macrophages to drive proinflammatory M1-like responses (26). Once fibrin clots are formed, macrophages produce tissue factor pathway inhibitors to suppress TF-induced coagulation and eventually clear procoagulant factors to prevent excessive clot formation (26, 147). Since diverse functions are driven by macrophage polarization, dysregulation of these functions modulated by coagulation factors can lead to hemophilic or thrombotic diseases (26). Therefore, it is critical that macrophage function is regulated to prevent the development of these diseases. Due of the complexity of the coagulation cascade, this review will highlight a few of the selected coagulation factors and regulators that been shown to be involved in macrophage polarization.
Tissue injury-induced coagulation activation modulates macrophage polarization for tissue repair
The coagulation cascade is directly involved in modulating macrophage response to orchestrate inflammatory responses for wound healing (148). Macrophages express receptors, such as protease-activated receptors (PARs), TLRs, and integrins, that recognize different coagulation factors (148). Initial vascular injury induces platelets to release thrombin that activates PAR-1 expressed on macrophages resulting in the production of proinflammatory cytokines (148). In this intrinsic pathway, activated FXII produced by damaged vessels appears to have a role in inducing M1 polarization through the urokinase plasminogen activator receptor (uPAR) to secrete TNF-ɑ, IL-1β, IL-6, and IL-6 (149). Further down the cascade, FVIII plays an important role in macrophage differentiation and polarization (150, 151). Deficiency in FVIII or FIX led to prolonged bleeding in patients with hemophilia due to the inability to clot (152). Interestingly, it was observed that the majority of monocytes from the peripheral blood of hemophilia patients failed to differentiate into macrophages upon exposure to either M-CSF or GM-CSF due to low expression of CSF-1 and CSF-2 receptors (150). From those monocytes that could differentiate into macrophages in these patients, they had significant reduction in hemoglobin-haptoglobin receptor CD163 and Tie2 angiopoietin receptor, which are expressed on M2-like macrophages responsible for tissue regeneration (150). In addition, high levels of adipokine leptin detected in blood of hemophiliacs has been shown to inhibit this M2-like regenerative phenotype in human THP-1 monocyte cells (150). Another study using human monocyte-derived macrophages showed that the use of recombinant FVIII Fc fusion protein, not FVIII, uniquely skews macrophages toward an M2-like repair response (151). Together, this suggests that FVIII may favor M2 polarization to suppress proinflammatory response and enhance profibrotic and tissue repair in vitro and ex vivo. However, in vivo studies are needed to validate and further elucidate FVIII role in macrophage polarization.
During tissue injury, platelet-derived thrombin recognized by PAR-1 on macrophages triggers TF production (153). This creates a positive feedback loop where the FIIIa:FVIIa complex binds to macrophages’ PAR-2 resulting in M1-like responses including ROS and proinflammatory cytokine production (154). Interestingly, it has been shown that type 1 interferon (IFN) signaling is required for macrophages’ TF production, which induced caspase-11 to activate inflammasomes for pyroptosis (inflammatory cell death) (155, 156). Inflammasome activation by anti-viral/bacterial type 1 IFN- or LPS-induced signaling leads to the release of microvesicles containing TFs, triggering coagulation (156, 157). These findings suggest an important link that bacterial and viral sepsis can trigger excessive inflammatory M1-like responses leading to disseminated intravascular coagulation (DIC), a life-threatening condition observed in patients with severe COVID-19 and bacterial infections (155, 158–161).
Macrophages express receptors and integrins that recognize thrombin, Fg, and fibrin, these three factors can also modulate macrophage polarization to promote proper wound healing. Although not limited to these receptors, TLR-4, Mac-1 (ɑMβ2 or CD11b/CD18), and CD11c/CD18 (ɑXβ2) on macrophages can recognize Fg and fibrin (14, 162–165). In a study using murine and human macrophage cell lines, soluble Fg interaction with TLR-4 stimulated proinflammatory chemokines, specifically macrophage inflammatory protein (MIP)-1ɑ, -1β, -2, and monocyte chemoattractant protein-1 (MCP-1) (166). Interestingly, in an in vivo study that used mice expressing Fg with a mutated γ chain (Fgγ390-396A) showed that macrophages, monocytes, and neutrophils failed to interact with the mutated Fg, resulting in a major defect in the inflammatory response upon S. aureus peritoneal infection and leading to inability to rapidly clear the infection (14, 167). Although both Fg and fibrin are important for promoting antimicrobial host response, there is a need to elucidate the differential role of soluble Fg versus fibrin on macrophage response. To distinguish the effects of Fg and fibrin on host response, Prasad and colleagues generated FgAEK mice that display normal levels of circulating Fg but lack the ability to form fibrin clots due to a germ-line mutation in the Fg’s Aɑ chain thrombin cleavage site (168). They found these mice were unable to clear S. aureus in the peritoneal cavity within the first hour similar to Fg-deficient mice, suggesting the importance of fibrin polymer in inducing antimicrobial response during acute response (168). However, the exact mechanism of how soluble Fg interacts with immune cells, specifically macrophages, in vivo and its antimicrobial response to prolonged infection to other pathogens and tissue injury warrants further studies.
An ex vivo study showed that soluble Fg-induced murine BMDMs to upregulate iNOS and produce proinflammatory cytokines, particularly TNF-ɑ, MCP-1, CXCL9, MIP-1ɑ, MIP-2, and CCL2 (169). Importantly, fibrin outcompeted soluble Fg to promote an M2-like anti-inflammatory phenotype by upregulating Arg1 and secreting IL-10 (169). From this study, it is unclear whether soluble Fg and fibrin favors interactions with TLR-4, Mac-1, or CD11c/CD18 on macrophages. Factors such as tethering properties, protein conformation, and binding site presentation of soluble Fg and fibrin can influence specific interaction with macrophage receptors and integrins, which may subsequently drive distinct pro- or anti-inflammatory intracellular signaling pathways. Thus, it is important to elucidate the mechanisms of how Fg and fibrin can differentially alter macrophage polarization and intracellular signaling pathways.
Fibrinolysis and RAMPs promote M2-like macrophages to restore tissue homeostasis
During the tissue repair and restoration process, M2-like macrophages play an important role in resolving inflammation, repairing, remodeling, and restoring damaged tissue. Besides cytokines, PAMPs, DAMPs, and macrophages can also be polarized by resolution-associated molecular patterns (RAMPs) and coagulation factors secreted from damaged tissue (170). During tissue repair, dying cells release RAMPs, playing a role in transitioning M1s to M2s. Dying cells that release heat shock protein (HSP)-10, HSP27, binding immunoglobulin protein (BiP), and annexin A1 promote autocrine IL-10 production polarizing macrophages toward M2 (171). In addition, phosphatidylserine flipped to the dying cells’ surface activates Mer tyrosine kinase (MerTK) receptors on macrophages and drives M2-like responses such as efferocytosis (172). These RAMPs contribute to their role in promoting anti-inflammatory M2 response [discussed in detail by Koncz et al. (173)].
In Alzheimer’s disease, multiple sclerosis, and experimental autoimmune encephalomyelitis mouse models, multiomic profiling of fibrin-induced microglia revealed that fibrin-CD11b signaling promoted proinflammatory oxidative stress and type 1 IFN response, which led to neuronal dysfunction (174). Furthermore, genetic deletion of Fg binding motif to CD11b in these diseased mice models reduced microglial neurodegenerative signatures, suggesting that targeting excess fibrin can favor clinical outcomes for patients with neurodegenerative or autoimmune diseases (174). As part of fibrin clot dissolution, CCR2+ M1-like murine macrophages possess the ability to clear fibrin and fibrin degradation products (FDPs) via endocytosis and lysosomal degradation dependent on plasminogen (PG) and plasmin activity (175). Interestingly, when fibrin gels were subcutaneously implanted in PG-deficient mice, there was a reduced presence of M1 macrophages with uptake of extravascular fibrin (175). Furthermore, they found that fibrin degradation by M1s via this plasmin-dependent endocytic mechanism, even in the absence of all three Fg receptors Mac-1, intercellular adhesion molecule-1 (ICAM-1), and mannose receptor (MR) (175). Based on this study, these M1 macrophages express their cell-surface PG receptor for PG binding, which then gets activated by cell-surface uPA and tPA (175). This leads to plasmin to partially degrade fibrin extracellularly and enable endocytosis of these FDPs by M1 macrophages to fully undergo complete degradation within the lysosomes (175). Other in vitro and in vivo mouse studies also reported that PG is essential in promoting macrophages’ phagocytosis of apoptotic thymocytes, hepatocytes, and alveolar neutrophils, which may be explained by upregulating expression of receptors that recognize RAMPs from dying cells (170, 176–178). Hence, this suggests that PG has an important role in resolving inflammation by initiating M1 macrophages to degrade fibrin both extra- and intracellularly and promote efferocytosis.
Since fibrin serves as a provisional ECM before being replaced by collagen, it may be possible that PG stimulates M1s to initiate the efferocytosis and fibrin degradation before transitioning into different M2 phenotypes to complete ECM remodeling and restoration. RAMPs released from dying cells can polarize macrophages toward different M2 phenotypes (170). In the cell proliferation phase, wound-healing macrophages elevate the production of growth factors PDGF, vascular endothelial growth factor (VEGF), and TGF-β to promote cell proliferation, granulation tissue/ECM formation, and angiogenesis (179). At the same time, neutrophils that phagocytize apoptotic cells induce another subset of M2 macrophages to produce IL-10 for suppressing proinflammatory activities (180). In the remodeling phase, pro-resolving macrophages are activated, allowing the production of matrix metalloproteases (MMPs) to remodel the ECM and restore tissue homeostasis (52). Together, a proper orchestration of macrophage polarization by coagulation factors is critical for tissue homeostasis since dysregulated polarization may result in exacerbated inflammation, resulting in chronic inflammation and promoting the onset of inflammatory diseases including atherosclerosis, diabetes, autoimmune diseases, and coagulopathy-associated diseases (87, 181–186).
PERSPECTIVES AND CONCLUSIONS
Macrophage polarization has emerged as a key feature in gaining a better understanding of how macrophages promote or resolve infections, inflammation, and cancers. One of the challenges to understanding macrophage function is that the description of macrophage polarization is disputed, oversimplified, and confusing due to the lack of nomenclature and experimental standardization. This is an issue that has been persistently brought to attention among macrophage and phagocyte biologists for at least two decades with great debate (32, 65, 187, 188). The use of in vitro and ex vivo studies with macrophage-like cell lines, murine bone marrow-derived or peritoneal macrophages, and human macrophages isolated from peripheral blood allowed researchers to stimulate these macrophages with various inducers generating different phenotypes (32). Since each laboratory varies in defining macrophage activation and polarization states, it is important to provide reporting standards for these studies such as (i) describing the source and differentiation method of macrophages; (ii) defining the activator with the recommended M(inducer) notation (i.e., M(IFN-γ), M(LPS), M(IL-4), M(GCs), etc.); (iii) reporting activation conditions; and (iv) stating the method of analysis and readout (32). While in vitro models demonstrate induction of discrete macrophage polarization states (M1 vs M2), in vivo disease models have revealed that polarization occurs on a continuum with mixed phenotypes due to the complex TME. Therefore, it is difficult to translate in vitro and ex vivo characterization to in vivo mouse models and human diseases due to other underlying factors that can also polarize diverse macrophage populations in these scenarios. Hence, there is a great need to go beyond just detection for the presence or absence of markers to characterize macrophages in complex TMEs. A proposed solution is to elucidate the interplay of epigenetics and metabolism in regulating and characterizing macrophage polarization. Recent studies and literature reviews shed a light on the epigenetics and metabolism on driving macrophage polarization and function marking advancements in this direction (189–193). The challenge to studying this is that it requires the use of multiple specialized techniques and instruments with its own set of limitations and difficult technical expertise. One group addressed this obstacle by developing a novel triomics (epigenetics, metabolomics, and transcriptomics) method using mass spectrometry on IFN-γ- or IL-4-stimulated BMDMs to simultaneously analyze metabolites, histone modifications, and protein expression all in a single sample (194). This novel method is one of many tools that can be applied to in vivo and clinical samples, thereby generating detailed and meaningful characterization and functions of macrophages in different diseases. While there is much progress in understanding macrophage polarization, there is still the need to further develop approaches to better understand how the host tissue microenvironment in inflammation, infections, and cancers affect macrophage functions.
Specifically, an area that needs to be investigated further is how the activation of the coagulation cascade upon tissue injury influences macrophage functions during infections, since it has been reported that a major component of this cascade, Fg and its polymerized form fibrin, can modulate macrophage polarization and antimicrobial responses to infections and insertion of medical devices (65, 195, 196). This is relevant in clinical settings, specifically when using implanted biomaterials, such as urinary catheters. These implants induce tissue damage, which activates the coagulation cascade, resulting in Fg recruitment and its polymerization to fibrin clot by thrombin proteolytic activity (197, 198). In catheter-associated urinary tract infections (CAUTIs), urinary catheterization elicits bladder inflammation in both mice and humans, promoting Fg/fibrin accumulation and recruitment of neutrophils and macrophages into the catheterized bladder (199–205). Importantly, in humans and mice, Fg and fibrin are used as platforms for biofilm formation on the catheters and bladders by diverse uropathogens including E. coli, E. faecalis, C. albicans, K. pneumonia, P. aeruginosa, S. aureus, and Acinetobacter baumannii (199–205). Furthermore, CAUTI studies performed on mice with fibrinolytic deficiencies revealed that mutated soluble Fg decreased microbial burden while excessive fibrin accumulation enhanced microbial colonization (198). The fact that catheterization allows different microbes to infect is puzzling given the strong immune cell recruitment, indicating that the immune response generated is unable to control the infection. Since Fg and fibrin are accumulated in the catheterized bladder, they could play a role in macrophage polarization. It is known that macrophages integrins (Mac-1 [ɑMβ2] and ɑXβ2) and TLR-4 interact with Fg, resulting in M1 polarization and proinflammatory responses (14, 162, 167). Oppositely to Fg, fibrin promotes M2 polarization and prevents M1 polarization by Fg, LPS, and IFN-γ (169). This differential polarization could be explained by differences in the tethering properties, receptor or integrin engagement, and protein conformation between Fg and fibrin. Coagulation factors that promote fibrin formation could also indirectly induce M2 polarization in other diseases such as cancer (206, 207). For example, it was reported that higher activity of FX, a plasma protein that activates prothrombin into thrombin for fibrin formation, dampened antitumor response by inducing M2 polarization in cancer mouse models (206, 207). Interestingly, a study found that chemical inhibition of FX using rivaroxaban in myeloid cells reprograms M2 tumor-associated macrophages to M1 that promote antitumor immunity (207). Therefore, it is important to further study the role of Fg and fibrin on macrophage polarization in different disease and infection models to better understand and develop reprogramming strategies.
A major challenge is to predict how the complex inflammatory microenvironments will affect macrophage polarization and their clinical associations with disease pathologies. Since macrophage polarization occurs on a spectrum, there is a major need to elucidate mixed M1/M2 phenotypes. Although great strides have been made in developing novel strategies targeting macrophage polarization, still these therapeutics have shown mixed results in different disease models. This could be explained by how other factors in the TME can affect both macrophage and other immune cell responses. Therefore, understanding how the TME drives the plasticity of macrophage polarization by modulating pro- and anti-inflammatory phenotypes can provide an avenue to developing viable therapeutic strategies in human diseases.
ACKNOWLEDGMENTS
This work was done in the Flores-Mireles laboratory and funded by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) grant R01DK128805 (to A.L.F.-M. and A.M.M.), Diversity supplement R01DK12880501-A1S1 (to A.M.M.), and a Good Venture Foundation (Open Philanthropy) grant (to A.L.F.-M. and A.M.M.).
Figures were created using BioRender.
Biographies
Armando M. Marrufo graduated with his Ph.D. in the Biological Sciences at the University of Notre Dame, where under mentorship of Dr. Ana Flores-Mireles his research elucidated how the urinary catheterized bladder environment induced anti-inflammatory macrophage polarization to promote catheter-associated urinary tract infections. Prior to his graduate studies, he completed his Bachelor of Science degree in Biological Sciences at Texas A&M University in College Station, Texas, where his first research experience focused on elucidating the mechanism of how bacteriophages induce lysis of gram-negative bacteria. He then went on to complete his Master of Science degree in Biomedical Sciences at the University of North Texas Health Science Center in Fort Worth, Texas, where his research aimed to utilize natural killer cells to target inhibitory ligands on triple-negative breast cancer cells.
Anna Lidia Flores-Mireles did her B.S. in Marine Biology and M.Sc. in Marine Biotechnology in Mexico. She did a PhD in Microbiology at Cornell University under Dr. Steve Winans' supervision. Then, she did her postdoctoral research at Washington University School of Medicine under the supervision of Dr. Scott Hultgren and Dr. Michael Caparon. In 2018, she became the Janet C. and Jeffrey A. Hawk Assistant Professor of Biological Sciences at the University of Notre Dame. Her laboratory is working on understanding how catheter-induced bladder inflammation renders the host susceptible to catheter-associated urinary tract infections and subsequent systemic dissemination. By utilizing a comprehensive approach, combining microbial genetics, transgenic mice, clinical samples, material modification, pathogen and host transcriptomics and proteomics as well as immunological, histological, microscopic, and pharmacological techniques, her lab goals are to develop novel antibiotic-sparing therapies against these infections.
Contributor Information
Ana Lidia Flores-Mireles, Email: afloresm@nd.edu.
Karen M. Ottemann, University of California at Santa Cruz Department of Microbiology and Environmental Toxicology, Santa Cruz, California, USA
REFERENCES
- 1. Gordon S. 2008. Elie Metchnikoff: father of natural immunity. Eur J Immunol 38:3257–3264. doi: 10.1002/eji.200838855 [DOI] [PubMed] [Google Scholar]
- 2. Pandey S, Kant S, Khawary M, Tripathi D. 2022. Macrophages in microbial pathogenesis: commonalities of defense evasion mechanisms. Infect Immun 90:e0029121. doi: 10.1128/IAI.00291-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Watanabe S, Alexander M, Misharin AV, Budinger GRS. 2019. The role of macrophages in the resolution of inflammation. J Clin Invest 129:2619–2628. doi: 10.1172/JCI124615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Su Y, Gao J, Kaur P, Wang Z. 2020. Neutrophils and macrophages as targets for development of nanotherapeutics in inflammatory diseases. Pharmaceutics 12:1222. doi: 10.3390/pharmaceutics12121222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Epelman S, Lavine KJ, Randolph GJ. 2014. Origin and functions of tissue macrophages. Immunity 41:21–35. doi: 10.1016/j.immuni.2014.06.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Italiani P, Boraschi D. 2014. From monocytes to M1/M2 macrophages: phenotypical vs functional differentiation. Front Immunol 5:514. doi: 10.3389/fimmu.2014.00514 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Nielsen MC, Andersen MN, Møller HJ. 2020. Monocyte isolation techniques significantly impact the phenotype of both isolated monocytes and derived macrophages in vitro. Immunology 159:63–74. doi: 10.1111/imm.13125 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Yang J, Zhang L, Yu C, Yang XF, Wang H. 2014. Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res 2:1. doi: 10.1186/2050-7771-2-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Murray PJ. 2017. Macrophage polarization. Annu Rev Physiol 79:541–566. doi: 10.1146/annurev-physiol-022516-034339 [DOI] [PubMed] [Google Scholar]
- 10. Sica A, Mantovani A. 2012. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 122:787–795. doi: 10.1172/JCI59643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F, Rodewald H-R. 2015. Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518:547–551. doi: 10.1038/nature13989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Ley K, Pramod AB, Croft M, Ravichandran KS, Ting JP. 2016. How mouse macrophages sense what is going on. Front Immunol 7:204. doi: 10.3389/fimmu.2016.00204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Chawla A. 2010. Control of macrophage activation and function by PPARs. Circ Res 106:1559–1569. doi: 10.1161/CIRCRESAHA.110.216523 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Flick MJ, Du X, Witte DP, Jirousková M, Soloviev DA, Busuttil SJ, Plow EF, Degen JL. 2004. Leukocyte engagement of fibrin(ogen) via the integrin receptor alphaMbeta2/Mac-1 is critical for host inflammatory response in vivo. J Clin Invest 113:1596–1606. doi: 10.1172/JCI20741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. 2004. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 25:677–686. doi: 10.1016/j.it.2004.09.015 [DOI] [PubMed] [Google Scholar]
- 16. Hirano S, Kanno S. 2015. Macrophage receptor with collagenous structure (MARCO) is processed by either macropinocytosis or endocytosis-autophagy pathway. PLOS ONE 10:e0142062. doi: 10.1371/journal.pone.0142062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Ross EA, Devitt A, Johnson JR. 2021. Macrophages: the good, the bad, and the gluttony. Front Immunol 12:708186. doi: 10.3389/fimmu.2021.708186 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Lemke G. 2019. How macrophages deal with death. Nat Rev Immunol 19:539–549. doi: 10.1038/s41577-019-0167-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Liu SX, Gustafson HH, Jackson DL, Pun SH, Trapnell C. 2020. Trajectory analysis quantifies transcriptional plasticity during macrophage polarization. Sci Rep 10:12273. doi: 10.1038/s41598-020-68766-w [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wang N, Liang H, Zen K. 2014. Molecular mechanisms that influence the macrophage M1-M2 polarization balance. Front Immunol 5:614. doi: 10.3389/fimmu.2014.00614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Cutolo M, Campitiello R, Gotelli E, Soldano S. 2022. The role of M1/M2 macrophage polarization in rheumatoid arthritis synovitis. Front Immunol 13:867260. doi: 10.3389/fimmu.2022.867260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Peng Y, Zhou M, Yang H, Qu R, Qiu Y, Hao J, Bi H, Guo D. 2023. Regulatory mechanism of M1/M2 macrophage polarization in the development of autoimmune diseases. Mediators Inflamm 2023:8821610. doi: 10.1155/2023/8821610 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Wang Chau C, Sugimura R. 2022. Locked in a pro-inflammatory state. Elife 11. doi: 10.7554/eLife.80699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Liu J, Geng X, Hou J, Wu G. 2021. New insights into M1/M2 macrophages: key modulators in cancer progression. Cancer Cell Int. 21:389. doi: 10.1186/s12935-021-02089-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Labonte AC, Tosello-Trampont AC, Hahn YS. 2014. The role of macrophage polarization in infectious and inflammatory diseases. Mol Cells 37:275–285. doi: 10.14348/molcells.2014.2374 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Wilhelm G, Mertowska P, Mertowski S, Przysucha A, Strużyna J, Grywalska E, Torres K. 2023. The crossroads of the coagulation system and the immune system: interactions and connections. Int J Mol Sci 24:12563. doi: 10.3390/ijms241612563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Degboé Y, Poupot R, Poupot M. 2022. Repolarization of unbalanced macrophages: unmet medical need in chronic inflammation and cancer. Int J Mol Sci 23:1496. doi: 10.3390/ijms23031496 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Gray JI, Farber DL. 2022. Tissue-resident immune cells in humans. Annu Rev Immunol 40:195–220. doi: 10.1146/annurev-immunol-093019-112809 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wu Y, Hirschi KK. 2020. Tissue-resident macrophage development and function. Front Cell Dev Biol 8:617879. doi: 10.3389/fcell.2020.617879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Schiwon M, Weisheit C, Franken L, Gutweiler S, Dixit A, Meyer-Schwesinger C, Pohl J-M, Maurice NJ, Thiebes S, Lorenz K, Quast T, Fuhrmann M, Baumgarten G, Lohse MJ, Opdenakker G, Bernhagen J, Bucala R, Panzer U, Kolanus W, Gröne H-J, Garbi N, Kastenmüller W, Knolle PA, Kurts C, Engel DR. 2014. Crosstalk between sentinel and helper macrophages permits neutrophil migration into infected uroepithelium. Cell 156:456–468. doi: 10.1016/j.cell.2014.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Ushach I, Zlotnik A. 2016. Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage. J Leukoc Biol 100:481–489. doi: 10.1189/jlb.3RU0316-144R [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M, Mantovani A, Martinez FO, Mege J-L, Mosser DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA. 2014. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41:14–20. doi: 10.1016/j.immuni.2014.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Hu X, Ivashkiv LB. 2009. Cross-regulation of signaling pathways by interferon-gamma: implications for immune responses and autoimmune diseases. Immunity 31:539–550. doi: 10.1016/j.immuni.2009.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Waddell SJ, Popper SJ, Rubins KH, Griffiths MJ, Brown PO, Levin M, Relman DA. 2010. Dissecting interferon-induced transcriptional programs in human peripheral blood cells. PLoS ONE 5:e9753. doi: 10.1371/journal.pone.0009753 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Gao J, Morrison DC, Parmely TJ, Russell SW, Murphy WJ. 1997. An interferon-gamma-activated site (GAS) is necessary for full expression of the mouse iNOS gene in response to interferon-gamma and lipopolysaccharide. J Biol Chem 272:1226–1230. doi: 10.1074/jbc.272.2.1226 [DOI] [PubMed] [Google Scholar]
- 36. Rath M, Müller I, Kropf P, Closs EI, Munder M. 2014. Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front Immunol 5:532. doi: 10.3389/fimmu.2014.00532 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Blanchette J, Jaramillo M, Olivier M. 2003. Signalling events involved in interferon-gamma-inducible macrophage nitric oxide generation. Immunology 108:513–522. doi: 10.1046/j.1365-2567.2003.01620.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Sumimoto H. 2008. Structure, regulation and evolution of nox-family NADPH oxidases that produce reactive oxygen species. Febs J 275:3249–3277. doi: 10.1111/j.1742-4658.2008.06488.x [DOI] [PubMed] [Google Scholar]
- 39. Casbon A-J, Long ME, Dunn KW, Allen L-AH, Dinauer MC. 2012. Effects of IFN-γ on intracellular trafficking and activity of macrophage NADPH oxidase flavocytochrome b558. J Leukoc Biol 92:869–882. doi: 10.1189/jlb.0512244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Zanoni I, Granucci F. 2013. Role of CD14 in host protection against infections and in metabolism regulation. Front Cell Infect Microbiol 3:32. doi: 10.3389/fcimb.2013.00032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Tugal D, Liao X, Jain MK. 2013. Transcriptional control of macrophage polarization. Arterioscler Thromb Vasc Biol 33:1135–1144. doi: 10.1161/ATVBAHA.113.301453 [DOI] [PubMed] [Google Scholar]
- 42. Liu T, Zhang L, Joo D, Sun S-C. 2017. Sun, NF-κB signaling in inflammation. Sig Transduct Target Ther 2:17023. doi: 10.1038/sigtrans.2017.23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Platanias LC. 2005. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat Rev Immunol 5:375–386. doi: 10.1038/nri1604 [DOI] [PubMed] [Google Scholar]
- 44. Van Overmeire E, Stijlemans B, Heymann F, Keirsse J, Morias Y, Elkrim Y, Brys L, Abels C, Lahmar Q, Ergen C, Vereecke L, Tacke F, De Baetselier P, Van Ginderachter JA, Laoui D. 2016. M-CSF and GM-CSF receptor signaling differentially regulate monocyte maturation and macrophage polarization in the tumor microenvironment. Cancer Res. 76:35–42. doi: 10.1158/0008-5472.CAN-15-0869 [DOI] [PubMed] [Google Scholar]
- 45. Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, Hussell T, Feldmann M, Udalova IA. 2011. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol 12:231–238. doi: 10.1038/ni.1990 [DOI] [PubMed] [Google Scholar]
- 46. Griseri T, Arnold IC, Pearson C, Krausgruber T, Schiering C, Franchini F, Schulthess J, McKenzie BS, Crocker PR, Powrie F. 2015. Granulocyte macrophage colony-stimulating factor-activated eosinophils promote interleukin-23 driven chronic colitis. Immunity 43:187–199. doi: 10.1016/j.immuni.2015.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Pearson C, Thornton EE, McKenzie B, Schaupp A-L, Huskens N, Griseri T, West N, Tung S, Seddon BP, Uhlig HH, Powrie F. 2016. ILC3 GM-CSF production and mobilisation orchestrate acute intestinal inflammation. Elife 5:e10066. doi: 10.7554/eLife.10066 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Castro-Dopico T, Fleming A, Dennison TW, Ferdinand JR, Harcourt K, Stewart BJ, Cader Z, Tuong ZK, Jing C, Lok LSC, Mathews RJ, Portet A, Kaser A, Clare S, Clatworthy MR. 2020. GM-CSF calibrates macrophage defense and wound healing programs during intestinal infection and inflammation. Cell Rep 32:107857. doi: 10.1016/j.celrep.2020.107857 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. 2018. The role of macrophages in acute and chronic wound healing and interventions to promote pro-wound healing phenotypes. Front Physiol 9:419. doi: 10.3389/fphys.2018.00419 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Yao Y, Xu XH, Jin L. 2019. Macrophage polarization in physiological and pathological pregnancy. Front Immunol 10:792. doi: 10.3389/fimmu.2019.00792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Rutschman R, Lang R, Hesse M, Ihle JN, Wynn TA, Murray PJ. 2001. Cutting edge: stat6-dependent substrate depletion regulates nitric oxide production. J Immunol 166:2173–2177. doi: 10.4049/jimmunol.166.4.2173 [DOI] [PubMed] [Google Scholar]
- 52. Rőszer T. 2015. Understanding the mysterious M2 macrophage through activation markers and effector mechanisms. Mediators Inflamm 2015:816460. doi: 10.1155/2015/816460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Raes G, De Baetselier P, Noël W, Beschin A, Brombacher F, Hassanzadeh Gh G. 2002. Differential expression of FIZZ1 and Ym1 in alternatively versus classically activated macrophages. J Leukoc Biol 71:597–602. [PubMed] [Google Scholar]
- 54. Raes G, Van den Bergh R, De Baetselier P, Ghassabeh GH, Scotton C, Locati M, Mantovani A, Sozzani S. 2005. Arginase-1 and Ym1 are markers for murine, but not human, alternatively activated myeloid cells. J Immunol 174:6561. doi: 10.4049/jimmunol.174.11.6561 [DOI] [PubMed] [Google Scholar]
- 55. Wang LX, Zhang SX, Wu HJ, Rong XL, Guo J. 2019. M2b macrophage polarization and its roles in diseases. J Leukoc Biol 106:345–358. doi: 10.1002/JLB.3RU1018-378RR [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Yue Y, Huang S, Li H, Li W, Hou J, Luo L, Liu Q, Wang C, Yang S, Lv L, Shao J, Wu Z. 2020. M2b macrophages protect against myocardial remodeling after ischemia/reperfusion injury by regulating kinase activation of platelet-derived growth factor receptor of cardiac fibroblast. Ann Transl Med 8:1409–1409. doi: 10.21037/atm-20-2788 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Yue Y, Yang X, Feng K, Wang L, Hou J, Mei B, Qin H, Liang M, Chen G, Wu Z. 2017. M2b macrophages reduce early reperfusion injury after myocardial ischemia in mice: a predominant role of inhibiting apoptosis via A20. Int J Cardiol 245:228–235. doi: 10.1016/j.ijcard.2017.07.085 [DOI] [PubMed] [Google Scholar]
- 58. Oakley RH, Cidlowski JA. 2013. The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease. J Allergy Clin Immunol 132:1033–1044. doi: 10.1016/j.jaci.2013.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Ehrchen JM, Roth J, Barczyk-Kahlert K. 2019. More than suppression: glucocorticoid action on monocytes and macrophages. Front Immunol 10:2028. doi: 10.3389/fimmu.2019.02028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Di Rosa M, Radomski M, Carnuccio R, Moncada S. 1990. Glucocorticoids inhibit the induction of nitric oxide synthase in macrophages. Biochem Biophys Res Commun 172:1246–1252. doi: 10.1016/0006-291x(90)91583-e [DOI] [PubMed] [Google Scholar]
- 61. Stein M, Keshav S, Harris N, Gordon S. 1992. Interleukin 4 potently enhances murine macrophage mannose receptor activity: a marker of alternative immunologic macrophage activation. J Exp Med 176:287–292. doi: 10.1084/jem.176.1.287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nathan CF, Murray HW, Wiebe ME, Rubin BY. 1983. Identification of interferon-gamma as the lymphokine that activates human macrophage oxidative metabolism and antimicrobial activity. J Exp Med 158:670–689. doi: 10.1084/jem.158.3.670 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. 2000. M-1/M-2 macrophages and the Th1/Th2 paradigm1. J Immunol 164:6166–6173. doi: 10.4049/jimmunol.164.12.6166 [DOI] [PubMed] [Google Scholar]
- 64. Wang X, Peng H, Tian Z. 2019. Innate lymphoid cell memory. Cell Mol Immunol 16:423–429. doi: 10.1038/s41423-019-0212-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Martinez FO, Gordon S. 2014. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep 6:13. doi: 10.12703/P6-13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Takeuchi O, Akira S. 2010. Pattern recognition receptors and inflammation. Cell 140:805–820. doi: 10.1016/j.cell.2010.01.022 [DOI] [PubMed] [Google Scholar]
- 67. Guerriero JL. 2019. Macrophages: their untold story in T cell activation and function. Int Rev Cell Mol Biol 342:73–93. doi: 10.1016/bs.ircmb.2018.07.001 [DOI] [PubMed] [Google Scholar]
- 68. Lester SN, Li K. 2014. Toll-like receptors in antiviral innate immunity. J Mol Biol 426:1246–1264. doi: 10.1016/j.jmb.2013.11.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Rehwinkel J, Gack MU. 2020. RIG-I-like receptors: their regulation and roles in RNA sensing. Nat Rev Immunol 20:537–551. doi: 10.1038/s41577-020-0288-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Stone AEL, Green R, Wilkins C, Hemann EA, Gale M. 2019. RIG-I-like receptors direct inflammatory macrophage polarization against West Nile virus infection. Nat Commun 10:3649. doi: 10.1038/s41467-019-11250-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Fajgenbaum DC, June CH. 2020. Cytokine storm. N Engl J Med 383:2255–2273. doi: 10.1056/NEJMra2026131 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Del Valle DM, Kim-Schulze S, Huang H-H, Beckmann ND, Nirenberg S, Wang B, Lavin Y, Swartz TH, Madduri D, Stock A, et al. 2020. An inflammatory cytokine signature predicts COVID-19 severity and survival. Nat Med 26:1636–1643. doi: 10.1038/s41591-020-1051-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Szabo PA, Dogra P, Gray JI, Wells SB, Connors TJ, Weisberg SP, Krupska I, Matsumoto R, Poon MML, Idzikowski E, Morris SE, Pasin C, Yates AJ, Ku A, Chait M, Davis-Porada J, Guo XV, Zhou J, Steinle M, Mackay S, Saqi A, Baldwin MR, Sims PA, Farber DL. 2021. Longitudinal profiling of respiratory and systemic immune responses reveals myeloid cell-driven lung inflammation in severe COVID-19. Immunity 54:797–814. doi: 10.1016/j.immuni.2021.03.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Xiang X, Wu Y, Lv X-Q, Xu R-Q, Liu Y, Pan S-H, He M, Lai G-Q. 2022. Hepatitis B virus infection promotes M2 polarization of macrophages by upregulating the expression of B7X in vivo and in vitro. Viral Immunol 35:597–608. doi: 10.1089/vim.2022.0029 [DOI] [PubMed] [Google Scholar]
- 75. Bility MT, Cheng L, Zhang Z, Luan Y, Li F, Chi L, Zhang L, Tu Z, Gao Y, Fu Y, Niu J, Wang F, Su L. 2014. Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: induction of human-specific liver fibrosis and M2-like macrophages. PLoS Pathog 10:e1004032. doi: 10.1371/journal.ppat.1004032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Bility MT, Nio K, Li F, McGivern DR, Lemon SM, Feeney ER, Chung RT, Su L. 2016. Chronic hepatitis C infection-induced liver fibrogenesis is associated with M2 macrophage activation. Sci Rep 6:39520. doi: 10.1038/srep39520 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Zhang B, Miao T, Shen X, Bao L, Zhang C, Yan C, Wei W, Chen J, Xiao L, Sun C, Du J, Li Y. 2020. EB virus-induced ATR activation accelerates nasopharyngeal carcinoma growth via M2-type macrophages polarization. Cell Death Dis 11:742. doi: 10.1038/s41419-020-02925-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Griffiths P, Reeves M. 2021. Pathogenesis of human cytomegalovirus in the immunocompromised host. Nat Rev Microbiol 19:759–773. doi: 10.1038/s41579-021-00582-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Chan G, Bivins-Smith ER, Smith MS, Smith PM, Yurochko AD. 2008. Transcriptome analysis reveals human cytomegalovirus reprograms monocyte differentiation toward an M1 macrophage. J Immunol 181:698–711. doi: 10.4049/jimmunol.181.1.698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Cojohari O, Mahmud J, Altman AM, Peppenelli MA, Miller MJ, Chan GC. 2020. Human cytomegalovirus mediates unique monocyte-to-macrophage differentiation through the PI3K/SHIP1/Akt signaling network. Viruses 12:652. doi: 10.3390/v12060652 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Salina ACG, Dos-Santos D, Rodrigues TS, Fortes-Rocha M, Freitas-Filho EG, Alzamora-Terrel DL, Castro IMS, Fraga da Silva TFC, de Lima MHF, Nascimento DC, et al. 2022. Efferocytosis of SARS-CoV-2-infected dying cells impairs macrophage anti-inflammatory functions and clearance of apoptotic cells. Elife 11:e74443. doi: 10.7554/eLife.74443 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Chen WH, Toapanta FR, Shirey KA, Zhang L, Giannelou A, Page C, Frieman MB, Vogel SN, Cross AS. 2012. Potential role for alternatively activated macrophages in the secondary bacterial infection during recovery from influenza. Immunol Lett 141:227–234. doi: 10.1016/j.imlet.2011.10.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Tam VC, Suen R, Treuting PM, Armando A, Lucarelli R, Gorrochotegui-Escalante N, Diercks AH, Quehenberger O, Dennis EA, Aderem A, Gold ES. 2020. PPARα exacerbates necroptosis, leading to increased mortality in postinfluenza bacterial superinfection. Proc Natl Acad Sci U S A 117:15789–15798. doi: 10.1073/pnas.2006343117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Lucarelli R, Gorrochotegui-Escalante N, Taddeo J, Buttaro B, Beld J, Tam V. 2022. Eicosanoid-activated PPARα inhibits NFκB-dependent bacterial clearance during post-influenza superinfection. Front Cell Infect Microbiol 12:881462. doi: 10.3389/fcimb.2022.881462 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Shaughnessy LM, Swanson JA. 2007. The role of the activated macrophage in clearing Listeria monocytogenes infection. Front Biosci 12:2683–2692. doi: 10.2741/2364 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Ehrt S, Schnappinger D, Bekiranov S, Drenkow J, Shi S, Gingeras TR, Gaasterland T, Schoolnik G, Nathan C. 2001. Reprogramming of the macrophage transcriptome in response to interferon-gamma and Mycobacterium tuberculosis: signaling roles of nitric oxide synthase-2 and phagocyte oxidase. J Exp Med 194:1123–1140. doi: 10.1084/jem.194.8.1123 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Kiszewski AE, Becerril E, Aguilar LD, Kader ITA, Myers W, Portaels F, Hernàndez Pando R. 2006. The local immune response in ulcerative lesions of Buruli disease. Clin Exp Immunol 143:445–451. doi: 10.1111/j.1365-2249.2006.03020.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Wang H, Xie Z, Yang F, Wang Y, Jiang H, Huang X, Zhang Y. 2022. Salmonella enterica serovar Typhi influences inflammation and autophagy in macrophages. Braz J Microbiol 53:525–534. doi: 10.1007/s42770-022-00719-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Quiding-Järbrink M, Raghavan S, Sundquist M. 2010. Enhanced M1 macrophage polarization in human Helicobacter pylori-associated atrophic gastritis and in vaccinated mice. PLoS One 5:e15018. doi: 10.1371/journal.pone.0015018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. 2020. The role of macrophages in Staphylococcus aureus infection. Front Immunol 11:620339. doi: 10.3389/fimmu.2020.620339 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Zou J, Shankar N. 2015. Roles of TLR/MyD88/MAPK/NF-κB signaling pathways in the regulation of phagocytosis and proinflammatory cytokine expression in response to E. faecalis infection. PLoS ONE 10:e0136947. doi: 10.1371/journal.pone.0136947 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Kim SJ, Kim HM. 2017. Dynamic lipopolysaccharide transfer cascade to TLR4/Md2 complex via LBP and CD14. BMB Rep 50:55–57. doi: 10.5483/bmbrep.2017.50.2.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Hoshino K, Takeuchi O, Kawai T, Sanjo H, Ogawa T, Takeda Y, Takeda K, Akira S. 1999. Cutting edge: toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J Immunol 162:3749–3752. doi: 10.4049/jimmunol.162.7.3749 [DOI] [PubMed] [Google Scholar]
- 94. Ito N, Tsujimoto H, Ueno H, Xie Q, Shinomiya N. 2020. Helicobacter pylori-mediated immunity and signaling transduction in gastric cancer. J Clin Med 9:3699. doi: 10.3390/jcm9113699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Carey AJ, Sullivan MJ, Duell BL, Crossman DK, Chattopadhyay D, Brooks AJ, Tan CK, Crowley M, Sweet MJ, Schembri MA, Ulett GC. 2016. Uropathogenic Escherichia coli engages CD14-dependent signaling to enable bladder-macrophage-dependent control of acute urinary tract infection. J Infect Dis 213:659–668. doi: 10.1093/infdis/jiv424 [DOI] [PubMed] [Google Scholar]
- 96. Steukers L, Glorieux S, Vandekerckhove AP, Favoreel HW, Nauwynck HJ. 2012. Diverse microbial interactions with the basement membrane barrier. Trends Microbiol 20:147–155. doi: 10.1016/j.tim.2012.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Tomlin H, Piccinini AM. 2018. A complex interplay between the extracellular matrix and the innate immune response to microbial pathogens. Immunology 155:186–201. doi: 10.1111/imm.12972 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Atcha H, Jairaman A, Holt JR, Meli VS, Nagalla RR, Veerasubramanian PK, Brumm KT, Lim HE, Othy S, Cahalan MD, Pathak MM, Liu WF. 2021. Mechanically activated ion channel Piezo1 modulates macrophage polarization and stiffness sensing. Nat Commun 12:3256. doi: 10.1038/s41467-021-23482-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Singh B, Fleury C, Jalalvand F, Riesbeck K. 2012. Human pathogens utilize host extracellular matrix proteins laminin and collagen for adhesion and invasion of the host. FEMS Microbiol Rev 36:1122–1180. doi: 10.1111/j.1574-6976.2012.00340.x [DOI] [PubMed] [Google Scholar]
- 100. Geng J, Shi Y, Zhang J, Yang B, Wang P, Yuan W, Zhao H, Li J, Qin F, Hong L, Xie C, Deng X, Sun Y, Wu C, Chen L, Zhou D. 2021. TLR4 signalling via Piezo1 engages and enhances the macrophage mediated host response during bacterial infection. Nat Commun 12:3519. doi: 10.1038/s41467-021-23683-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Sijmons D, Guy AJ, Walduck AK, Ramsland PA. 2022. Helicobacter pylori and the role of lipopolysaccharide variation in innate immune evasion. Front Immunol 13:868225. doi: 10.3389/fimmu.2022.868225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Sanders CJ, Yu Y, Moore DA III, Williams IR, Gewirtz AT. 2006. Humoral immune response to flagellin requires T cells and activation of innate immunity. J Immunol 177:2810–2818. doi: 10.4049/jimmunol.177.5.2810 [DOI] [PubMed] [Google Scholar]
- 103. Barker JH, Weiss J, Apicella MA, Nauseef WM. 2006. Basis for the failure of Francisella tularensis lipopolysaccharide to prime human polymorphonuclear leukocytes. Infect Immun 74:3277–3284. doi: 10.1128/IAI.02011-05 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Raetz CRH, Whitfield C. 2002. Lipopolysaccharide endotoxins. Annu Rev Biochem 71:635–700. doi: 10.1146/annurev.biochem.71.110601.135414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Lamarque D, Moran AP, Szepes Z, Delchier JC, Whittle BJ. 2000. Cytotoxicity associated with induction of nitric oxide synthase in rat duodenal epithelial cells in vivo by lipopolysaccharide of Helicobacter pylori: inhibition by superoxide dismutase. Br J Pharmacol 130:1531–1538. doi: 10.1038/sj.bjp.0703468 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Lepper PM, Triantafilou M, Schumann C, Schneider EM, Triantafilou K. 2005. Lipopolysaccharides from Helicobacter pylori can act as antagonists for toll-like receptor 4. Cell Microbiol 7:519–528. doi: 10.1111/j.1462-5822.2005.00482.x [DOI] [PubMed] [Google Scholar]
- 107. Yokota S, Ohnishi T, Muroi M, Tanamoto K, Fujii N, Amano K. 2007. Highly-purified Helicobacter pylori LPS preparations induce weak inflammatory reactions and utilize toll-like receptor 2 complex but not toll-like receptor 4 complex. FEMS Microbiol Immunol 51:140–148. doi: 10.1111/j.1574-695X.2007.00288.x [DOI] [PubMed] [Google Scholar]
- 108. Chaturvedi R, Cheng Y, Asim M, Bussière FI, Xu H, Gobert AP, Hacker A, Casero RA, Wilson KT. 2004. Induction of polyamine oxidase 1 by Helicobacter pylori causes macrophage apoptosis by hydrogen peroxide release and mitochondrial membrane depolarization. J Biol Chem 279:40161–40173. doi: 10.1074/jbc.M401370200 [DOI] [PubMed] [Google Scholar]
- 109. Deen NS, Gong L, Naderer T, Devenish RJ, Kwok T. 2015. Analysis of the relative contribution of phagocytosis, LC3-associated phagocytosis, and canonical autophagy during Helicobacter pylori infection of macrophages. Helicobacter 20:449–459. doi: 10.1111/hel.12223 [DOI] [PubMed] [Google Scholar]
- 110. Augusto AC, Miguel F, Mendonça S, Pedrazzoli J Jr, Gurgueira SA. 2007. Oxidative stress expression status associated to Helicobacter pylori virulence in gastric diseases. Clin Biochem 40:615–622. doi: 10.1016/j.clinbiochem.2007.03.014 [DOI] [PubMed] [Google Scholar]
- 111. Abadi ATB. 2017. Strategies used by Helicobacter pylori to establish persistent infection. World J Gastroenterol 23:2870–2882. doi: 10.3748/wjg.v23.i16.2870 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Thiriot JD, Martinez-Martinez YB, Endsley JJ, Torres AG. 2020. Hacking the host: exploitation of macrophage polarization by intracellular bacterial pathogens. Pathog Dis 78:ftaa009. doi: 10.1093/femspd/ftaa009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Brodsky IE. 2020. JAK-ing into M1/M2 polarization steErs Salmonella-containing macrophages away from immune attack to promote bacterial persistence. Cell Host Microbe 27:3–5. doi: 10.1016/j.chom.2019.12.007 [DOI] [PubMed] [Google Scholar]
- 114. Shirey KA, Cole LE, Keegan AD, Vogel SN. 2008. Francisella tularensis live vaccine strain induces macrophage alternative activation as a survival mechanism. J Immunol 181:4159–4167. doi: 10.4049/jimmunol.181.6.4159 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Ancuta P, Pedron T, Girard R, Sandström G, Chaby R. 1996. Inability of the Francisella tularensis lipopolysaccharide to mimic or to antagonize the induction of cell activation by endotoxins. Infect Immun 64:2041–2046. doi: 10.1128/iai.64.6.2041-2046.1996 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Schulert GS, Allen L-AH. 2006. Differential infection of mononuclear phagocytes by Francisella tularensis: role of the macrophage mannose receptor. J Leukoc Biol 80:563–571. doi: 10.1189/jlb.0306219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Bandyopadhyay S, Long ME, Allen L-AH. 2014. Differential expression of microRNAs in Francisella tularensis-infected human macrophages: MiR-155-dependent downregulation of MyD88 inhibits the inflammatory response. PLOS ONE 9:e109525. doi: 10.1371/journal.pone.0109525 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Mohapatra NP, Soni S, Rajaram MVS, Dang PM-C, Reilly TJ, El-Benna J, Clay CD, Schlesinger LS, Gunn JS. 2010. Francisella acid phosphatases inactivate the NADPH oxidase in human phagocytes. J Immunol 184:5141–5150. doi: 10.4049/jimmunol.0903413 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Bauler TJ, Chase JC, Wehrly TD, Bosio CM. 2014. Virulent Francisella tularensis destabilize host mRNA to rapidly suppress inflammation. J Innate Immun 6:793–805. doi: 10.1159/000363243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Leopold Wager CM, Wormley FL. 2014. Classical versus alternative macrophage activation: the Ying and the Yang in host defense against pulmonary fungal infections. Mucosal Immunol 7:1023–1035. doi: 10.1038/mi.2014.65 [DOI] [PubMed] [Google Scholar]
- 121. Underhill DM, Rossnagle E, Lowell CA, Simmons RM. 2005. Dectin-1 activates Syk tyrosine kinase in a dynamic subset of macrophages for reactive oxygen production. Blood 106:2543–2550. doi: 10.1182/blood-2005-03-1239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Brown GD, Gordon S. 2001. Immune recognition. A new receptor for beta-glucans. Nature 413:36–37. doi: 10.1038/35092620 [DOI] [PubMed] [Google Scholar]
- 123. Strasser D, Neumann K, Bergmann H, Marakalala MJ, Guler R, Rojowska A, Hopfner K-P, Brombacher F, Urlaub H, Baier G, Brown GD, Leitges M, Ruland J. 2012. Syk kinase-coupled C-type lectin receptors engage protein kinase C-δ to elicit Card9 adaptor-mediated innate immunity. Immunity 36:32–42. doi: 10.1016/j.immuni.2011.11.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Wang Y, Zhang D, Hou Y, Shen S, Wang T. 2020. The adaptor protein CARD9, from fungal immunity to tumorigenesis. Am J Cancer Res 10:2203–2225. [PMC free article] [PubMed] [Google Scholar]
- 125. Campuzano A, Castro-Lopez N, Martinez AJ, Olszewski MA, Ganguly A, Leopold Wager C, Hung C-Y, Wormley FL. 2020. CARD9 is required for classical macrophage activation and the induction of protective immunity against pulmonary cryptococcosis. mBio 11. doi: 10.1128/mBio.03005-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Poeck H, Bscheider M, Gross O, Finger K, Roth S, Rebsamen M, Hannesschläger N, Schlee M, Rothenfusser S, Barchet W, Kato H, Akira S, Inoue S, Endres S, Peschel C, Hartmann G, Hornung V, Ruland J. 2010. Recognition of RNA virus by RIG-I results in activation of CARD9 and inflammasome signaling for interleukin 1 beta production. Nat Immunol 11:63–69. doi: 10.1038/ni.1824 [DOI] [PubMed] [Google Scholar]
- 127. Huang J-H, Lin C-Y, Wu S-Y, Chen W-Y, Chu C-L, Brown GD, Chuu C-P, Wu-Hsieh BA. 2015. CR3 and dectin-1 collaborate in macrophage cytokine response through association on lipid rafts and activation of Syk-JNK-AP-1 pathway. PLoS Pathog 11:e1004985. doi: 10.1371/journal.ppat.1004985 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Thompson A, Davies LC, Liao C-T, da Fonseca DM, Griffiths JS, Andrews R, Jones AV, Clement M, Brown GD, Humphreys IR, Taylor PR, Orr SJ. 2019. The protective effect of inflammatory monocytes during systemic C. albicans infection is dependent on collaboration between C-type lectin-like receptors. PLoS Pathog 15:e1007850. doi: 10.1371/journal.ppat.1007850 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Kim Y-G, Udayanga KGS, Totsuka N, Weinberg JB, Núñez G, Shibuya A. 2014. Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi-induced PGE2. Cell Host Microbe 15:95–102. doi: 10.1016/j.chom.2013.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Russell DG, Huang L, VanderVen BC. 2019. Immunometabolism at the interface between macrophages and pathogens. Nat Rev Immunol 19:291–304. doi: 10.1038/s41577-019-0124-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Stuckey PV, Santiago-Tirado FH. 2023. Fungal mechanisms of intracellular survival: what can we learn from bacterial pathogens? Infect Immun 91:e0043422. doi: 10.1128/iai.00434-22 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Dang EV, Lei S, Radkov A, Volk RF, Zaro BW, Madhani HD. 2022. Secreted fungal virulence effector triggers allergic inflammation via TLR4. Nature 608:161–167. doi: 10.1038/s41586-022-05005-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Wei W, Ning C, Huang J, Wang G, Lai J, Han J, He J, Zhang H, Liang B, Liao Y, Le T, Luo Q, Li Z, Jiang J, Ye L, Liang H. 2021. Talaromyces marneffei promotes M2-like polarization of human macrophages by downregulating SOCS3 expression and activating the TLR9 pathway . Virulence 12:1997–2012. doi: 10.1080/21505594.2021.1958470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Noverr MC, Phare SM, Toews GB, Coffey MJ, Huffnagle GB. 2001. Pathogenic yeasts Cryptococcus neoformans and Candida albicans produce immunomodulatory prostaglandins. Infect Immun 69:2957–2963. doi: 10.1128/IAI.69.5.2957-2963.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Noverr MC, Toews GB, Huffnagle GB. 2002. Production of prostaglandins and leukotrienes by pathogenic fungi. Infect Immun 70:400–402. doi: 10.1128/IAI.70.1.400-402.2002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Gagliardi MC, Teloni R, Mariotti S, Bromuro C, Chiani P, Romagnoli G, Giannoni F, Torosantucci A, Nisini R. 2010. Endogenous PGE2 promotes the induction of human Th17 responses by fungal ss-glucan. J Leukoc Biol 88:947–954. doi: 10.1189/jlb.0310139 [DOI] [PubMed] [Google Scholar]
- 137. Gonçalves SM, Duarte-Oliveira C, Campos CF, Aimanianda V, Ter Horst R, Leite L, Mercier T, Pereira P, Fernández-García M, Antunes D, et al. 2020. Phagosomal removal of fungal melanin reprograms macrophage metabolism to promote antifungal immunity. Nat Commun 11:2282. doi: 10.1038/s41467-020-16120-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Smith SA, Travers RJ, Morrissey JH. 2015. How it all starts: initiation of the clotting cascade. Crit Rev Biochem Mol Biol 50:326–336. doi: 10.3109/10409238.2015.1050550 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Periayah MH, Halim AS, Mat Saad AZ. 2017. Mechanism action of platelets and crucial blood coagulation pathways in hemostasis. Int J Hematol Oncol Stem Cell Res 11:319–327. [PMC free article] [PubMed] [Google Scholar]
- 140. Semple JW, Italiano JE, Freedman J. 2011. Platelets and the immune continuum. Nat Rev Immunol 11:264–274. doi: 10.1038/nri2956 [DOI] [PubMed] [Google Scholar]
- 141. Vilar R, Fish RJ, Casini A, Neerman-Arbez M. 2020. Fibrin(ogen) in human disease: both friend and foe. Haematologica 105:284–296. doi: 10.3324/haematol.2019.236901 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Göbel K, Eichler S, Wiendl H, Chavakis T, Kleinschnitz C, Meuth SG. 2018. The coagulation factors fibrinogen, thrombin, and factor XII in inflammatory disorders-a systematic review. Front Immunol 9:1731. doi: 10.3389/fimmu.2018.01731 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Cohen HB, Mosser DM. 2013. Extrinsic and intrinsic control of macrophage inflammatory responses. J Leukoc Biol 94:913–919. doi: 10.1189/jlb.0413236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Goren I, Allmann N, Yogev N, Schürmann C, Linke A, Holdener M, Waisman A, Pfeilschifter J, Frank S. 2009. A transgenic mouse model of inducible macrophage depletion: effects of diphtheria toxin-driven lysozyme M-specific cell lineage ablation on wound inflammatory, angiogenic, and contractive processes. Am J Pathol 175:132–147. doi: 10.2353/ajpath.2009.081002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Li S, Li B, Jiang H, Wang Y, Qu M, Duan H, Zhou Q, Shi W. 2013. Macrophage depletion impairs corneal wound healing after autologous transplantation in mice. PLOS ONE 8:e61799. doi: 10.1371/journal.pone.0061799 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Chen Y, Zhong H, Zhao Y, Luo X, Gao W. 2020. Role of platelet biomarkers in inflammatory response. Biomark Res 8:28. doi: 10.1186/s40364-020-00207-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Petit L, Lesnik P, Dachet C, Moreau M, Chapman MJ. 1999. Tissue factor pathway inhibitor is expressed by human monocyte–derived macrophages. Arterioscler Thromb Vasc Biol 19:309–315. doi: 10.1161/01.ATV.19.2.309 [DOI] [PubMed] [Google Scholar]
- 148. Antoniak S. 2018. The coagulation system in host defense. Res Pract Thromb Haemost 2:549–557. doi: 10.1002/rth2.12109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Vorlova S, Koch M, Manthey HD, Cochain C, Busch M, Chaudhari SM, Stegner D, Yepes M, Lorenz K, Nolte MW, Nieswandt B, Zernecke A. 2017. Coagulation factor XII induces pro-inflammatory cytokine responses in macrophages and promotes atherosclerosis in mice. Thromb Haemost 117:176–187. doi: 10.1160/TH16-06-0466 [DOI] [PubMed] [Google Scholar]
- 150. Knowles LM, Lessig D, Bernard M, Schwarz EC, Eichler H, Pilch J. 2018. Macrophage polarization is impaired in hemophilia. Blood 132:2499–2499. doi: 10.1182/blood-2018-99-117111 [DOI] [Google Scholar]
- 151. Kis-Toth K, Rajani GM, Simpson A, Henry KL, Dumont J, Peters RT, Salas J, Loh C. 2018. Recombinant factor VIII Fc fusion protein drives regulatory macrophage polarization. Blood Adv 2:2904–2916. doi: 10.1182/bloodadvances.2018024497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Franchini M, Mannucci PM. 2012. Past, present and future of hemophilia: a narrative review. Orphanet J Rare Dis 7:24. doi: 10.1186/1750-1172-7-24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Grover SP, Mackman N. 2018. Tissue factor. Arterioscler Thromb Vasc Biol 38:709–725. doi: 10.1161/ATVBAHA.117.309846 [DOI] [PubMed] [Google Scholar]
- 154. Cunningham MA, Romas P, Hutchinson P, Holdsworth SR, Tipping PG. 1999. Tissue factor and factor viia receptor/ligand interactions induce proinflammatory effects in macrophages. Blood 94:3413–3420. doi: 10.1182/blood.V94.10.3413.422k24_3413_3420 [DOI] [PubMed] [Google Scholar]
- 155. Yang X, Cheng X, Tang Y, Qiu X, Wang Z, Fu G, Wu J, Kang H, Wang J, Wang H, Chen F, Xiao X, Billiar TR, Lu B. 2020. The role of type 1 interferons in coagulation induced by gram-negative bacteria. Blood 135:1087–1100. doi: 10.1182/blood.2019002282 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Wu C, Lu W, Zhang Y, Zhang G, Shi X, Hisada Y, Grover SP, Zhang X, Li L, Xiang B, Shi J, Li X-A, Daugherty A, Smyth SS, Kirchhofer D, Shiroishi T, Shao F, Mackman N, Wei Y, Li Z. 2019. Inflammasome activation triggers blood clotting and host death through pyroptosis. Immunity 50:1401–1411. doi: 10.1016/j.immuni.2019.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Hagar JA, Powell DA, Aachoui Y, Ernst RK, Miao EA. 2013. Cytoplasmic LPS activates caspase-11: implications in TLR4-independent endotoxic shock. Science 341:1250–1253. doi: 10.1126/science.1240988 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Iba T, Levy JH, Levi M. 2022. Viral-induced inflammatory coagulation disorders: preparing for another epidemic. Thromb Haemost 122:8–19. doi: 10.1055/a-1562-7599 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Guervilly C, Bonifay A, Burtey S, Sabatier F, Cauchois R, Abdili E, Arnaud L, Lano G, Pietri L, Robert T, Velier M, Papazian L, Albanese J, Kaplanski G, Dignat-George F, Lacroix R. 2021. Dissemination of extreme levels of extracellular vesicles: tissue factor activity in patients with severe COVID-19. Blood Adv 5:628–634. doi: 10.1182/bloodadvances.2020003308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Popescu NI, Lupu C, Lupu F. 2022. Disseminated Intravascular coagulation and its immune mechanisms. Blood 139:1973–1986. doi: 10.1182/blood.2020007208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Unar A, Bertolino L, Patauner F, Gallo R, Durante-Mangoni E. 2023. Pathophysiology of disseminated intravascular coagulation in sepsis: a clinically focused overview. Cells 12:2120. doi: 10.3390/cells12172120 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Smiley ST, King JA, Hancock WW. 2001. Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 4. J Immunol 167:2887–2894. doi: 10.4049/jimmunol.167.5.2887 [DOI] [PubMed] [Google Scholar]
- 163. Loike JD, Sodeik B, Cao L, Leucona S, Weitz JI, Detmers PA, Wright SD, Silverstein SC. 1991. CD11c/CD18 on neutrophils recognizes a domain at the N terminus of the A alpha chain of fibrinogen. Proc Natl Acad Sci U S A 88:1044–1048. doi: 10.1073/pnas.88.3.1044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Nham SU. 1999. Characteristics of fibrinogen binding to the domain of CD11c, an alpha subunit of p150,95. Biochem Biophys Res Commun 264:630–634. doi: 10.1006/bbrc.1999.1564 [DOI] [PubMed] [Google Scholar]
- 165. Wright SD, Weitz JI, Huang AJ, Levin SM, Silverstein SC, Loike JD. 1988. Complement receptor type three (CD11b/CD18) of human polymorphonuclear leukocytes recognizes fibrinogen. Proc Natl Acad Sci U S A 85:7734–7738. doi: 10.1073/pnas.85.20.7734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Smiley ST, King JA, Hancock WW. 2001. Fibrinogen stimulates macrophage chemokine secretion through toll-like receptor 41. J Immunol 167:2887–2894. doi: 10.4049/jimmunol.167.5.2887 [DOI] [PubMed] [Google Scholar]
- 167. Flick MJ, Du X, Degen JL. 2004. Fibrin(ogen)-alpha M beta 2 interactions regulate leukocyte function and innate immunity in vivo. Exp Biol Med (Maywood) 229:1105–1110. doi: 10.1177/153537020422901104 [DOI] [PubMed] [Google Scholar]
- 168. Prasad JM, Gorkun OV, Raghu H, Thornton S, Mullins ES, Palumbo JS, Ko Y-P, Höök M, David T, Coughlin SR, Degen JL, Flick MJ. 2015. Mice expressing a mutant form of fibrinogen that cannot support fibrin formation exhibit compromised antimicrobial host defense. Blood 126:2047–2058. doi: 10.1182/blood-2015-04-639849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Hsieh JY, Smith TD, Meli VS, Tran TN, Botvinick EL, Liu WF. 2017. Differential regulation of macrophage inflammatory activation by fibrin and fibrinogen. Acta Biomater 47:14–24. doi: 10.1016/j.actbio.2016.09.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Koncz G, Jenei V, Tóth M, Váradi E, Kardos B, Bácsi A, Mázló A. 2023. Damage-mediated macrophage polarization in sterile inflammation. Front Immunol 14:1169560. doi: 10.3389/fimmu.2023.1169560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Chuang Y, Hung ME, Cangelose BK, Leonard JN. 2016. Regulation of the IL-10-driven macrophage phenotype under incoherent stimuli. Innate Immun 22:647–657. doi: 10.1177/1753425916668243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Zizzo G, Hilliard BA, Monestier M, Cohen PL. 2012. Efficient clearance of early apoptotic cells by human macrophages requires M2c polarization and MerTK induction. J Immunol 189:3508–3520. doi: 10.4049/jimmunol.1200662 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Koncz G, Jenei V, Tóth M, Váradi E, Kardos B, Bácsi A, Mázló A. 2023. Damage-mediated macrophage polarization in sterile inflammation. Front Immunol 14:1169560. doi: 10.3389/fimmu.2023.1169560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Mendiola AS, Yan Z, Dixit K, Johnson JR, Bouhaddou M, Meyer-Franke A, Shin M-G, Yong Y, Agrawal A, MacDonald E, Muthukumar G, Pearce C, Arun N, Cabriga B, Meza-Acevedo R, Alzamora MDPS, Zamvil SS, Pico AR, Ryu JK, Krogan NJ, Akassoglou K. 2023. Defining blood-induced microglia functions in neurodegeneration through multiomic profiling. Nat Immunol 24:1173–1187. doi: 10.1038/s41590-023-01522-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Motley MP, Madsen DH, Jürgensen HJ, Spencer DE, Szabo R, Holmbeck K, Flick MJ, Lawrence DA, Castellino FJ, Weigert R, Bugge TH. 2016. A CCR2 macrophage endocytic pathway mediates extravascular fibrin clearance in vivo. Blood 127:1085–1096. doi: 10.1182/blood-2015-05-644260 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Das R, Ganapathy S, Settle M, Plow EF. 2014. Plasminogen promotes macrophage phagocytosis in mice. Blood 124:679–688. doi: 10.1182/blood-2014-01-549659 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Kawao N, Nagai N, Tamura Y, Horiuchi Y, Okumoto K, Okada K, Suzuki Y, Umemura K, Yano M, Ueshima S, Kaji H, Matsuo O. 2012. Urokinase-type plasminogen activator and plasminogen mediate activation of macrophage phagocytosis during liver repair in vivo. Thromb Haemost 107:749–759. doi: 10.1160/TH11-08-0567 [DOI] [PubMed] [Google Scholar]
- 178. Vago JP, Sugimoto MA, Lima KM, Negreiros-Lima GL, Baik N, Teixeira MM, Perretti M, Parmer RJ, Miles LA, Sousa LP. 2019. Plasminogen and the plasminogen receptor, Plg-R(KT), regulate macrophage phenotypic, and functional changes. Front Immunol 10:1458. doi: 10.3389/fimmu.2019.01458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Murray PJ, Wynn TA. 2011. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol 11:723–737. doi: 10.1038/nri3073 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Filardy AA, Pires DR, Nunes MP, Takiya CM, Freire-de-Lima CG, Ribeiro-Gomes FL, DosReis GA. 2010. Proinflammatory clearance of apoptotic neutrophils induces an IL-12(Low)IL-10(high) regulatory phenotype in macrophages. J Immunol 185:2044–2050. doi: 10.4049/jimmunol.1000017 [DOI] [PubMed] [Google Scholar]
- 181. Liu YC, Zou XB, Chai YF, Yao YM. 2014. Macrophage polarization in inflammatory diseases. Int J Biol Sci 10:520–529. doi: 10.7150/ijbs.8879 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Hou P, Fang J, Liu Z, Shi Y, Agostini M, Bernassola F, Bove P, Candi E, Rovella V, Sica G, Sun Q, Wang Y, Scimeca M, Federici M, Mauriello A, Melino G. 2023. Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis 14:691. doi: 10.1038/s41419-023-06206-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Kraakman MJ, Murphy AJ, Jandeleit-Dahm K, Kammoun HL. 2014. Macrophage polarization in obesity and type 2 diabetes: weighing down our understanding of macrophage function? Front Immunol 5:470. doi: 10.3389/fimmu.2014.00470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Sharifiaghdam M, Shaabani E, Faridi-Majidi R, De Smedt SC, Braeckmans K, Fraire JC. 2022. Macrophages as a therapeutic target to promote diabetic wound healing. Mol Ther 30:2891–2908. doi: 10.1016/j.ymthe.2022.07.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Saradna A, Do DC, Kumar S, Fu QL, Gao P. 2018. Macrophage polarization and allergic asthma. Transl Res 191:1–14. doi: 10.1016/j.trsl.2017.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Yang S, Zhao M, Jia S. 2023. Macrophage: key player in the pathogenesis of autoimmune diseases. Front Immunol 14:1080310. doi: 10.3389/fimmu.2023.1080310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Mosser DM, Edwards JP. 2008. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 8:958–969. doi: 10.1038/nri2448 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Murray PJ, Wynn TA. 2011. Obstacles and opportunities for understanding macrophage polarization. J Leukoc Biol 89:557–563. doi: 10.1189/jlb.0710409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Baardman J, Licht I, de Winther MPJ, Van den Bossche J. 2015. Metabolic-epigenetic crosstalk in macrophage activation. Epigenomics 7:1155–1164. doi: 10.2217/epi.15.71 [DOI] [PubMed] [Google Scholar]
- 190. Noe JT, Rendon BE, Geller AE, Conroy LR, Morrissey SM, Young LEA, Bruntz RC, Kim EJ, Wise-Mitchell A, Barbosa de Souza Rizzo M, Relich ER, Baby BV, Johnson LA, Affronti HC, McMasters KM, Clem BF, Gentry MS, Yan J, Wellen KE, Sun RC, Mitchell RA. 2021. Lactate supports a metabolic-epigenetic link in macrophage polarization. Sci Adv 7:eabi8602. doi: 10.1126/sciadv.abi8602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191. Chen S, Yang J, Wei Y, Wei X. 2020. Epigenetic regulation of macrophages: from homeostasis maintenance to host defense. Cell Mol Immunol 17:36–49. doi: 10.1038/s41423-019-0315-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192. Wculek SK, Dunphy G, Heras-Murillo I, Mastrangelo A, Sancho D. 2022. Metabolism of tissue macrophages in homeostasis and pathology. Cell Mol Immunol 19:384–408. doi: 10.1038/s41423-021-00791-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Chang CY, Armstrong D, Corry DB, Kheradmand F. 2023. Alveolar macrophages in lung cancer: opportunities challenges. Front Immunol 14:1268939. doi: 10.3389/fimmu.2023.1268939 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Sowers ML, Tang H, Singh VK, Khan A, Mishra A, Restrepo BI, Jagannath C, Zhang K. 2022. Multi-OMICs analysis reveals metabolic and epigenetic changes associated with macrophage polarization. J Biol Chem 298:102418. doi: 10.1016/j.jbc.2022.102418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Mills CD. 2012. M1 and M2 macrophages: oracles of health and disease. Crit Rev Immunol 32:463–488. doi: 10.1615/critrevimmunol.v32.i6.10 [DOI] [PubMed] [Google Scholar]
- 196. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. 2019. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. alternatively activated macrophages. Front Immunol 10:1084. doi: 10.3389/fimmu.2019.01084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Palta S, Saroa R, Palta A. 2014. Overview of the coagulation system. Indian J Anaesth 58:515–523. doi: 10.4103/0019-5049.144643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198. Molina JJ, Kohler KN, Gager C, Andersen MJ, Wongso E, Lucas ER, Paik A, Xu W, Donahue DL, Bergeron K, Klim A, Caparon MG, Hultgren SJ, Desai A, Ploplis VA, Flick MJ, Castellino FJ, Flores-Mireles AL. 2024. Fibrinolytic-deficiencies predispose hosts to septicemia from a catheter-associated UTI. Nat Commun 15:2704. doi: 10.1038/s41467-024-46974-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Flores-Mireles AL, Walker JN, Bauman TM, Potretzke AM, Schreiber HL, Park AM, Pinkner JS, Caparon MG, Hultgren SJ, Desai A. 2016. Fibrinogen release and deposition on urinary catheters placed during urological procedures. J Urol 196:416–421. doi: 10.1016/j.juro.2016.01.100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200. Walker JN, Flores-Mireles AL, Pinkner CL, Schreiber HL 4th, Joens MS, Park AM, Potretzke AM, Bauman TM, Pinkner JS, Fitzpatrick JAJ, Desai A, Caparon MG, Hultgren SJ. 2017. Catheterization alters bladder ecology to potentiate Staphylococcus aureus infection of the urinary tract. Proc Natl Acad Sci U S A 114:E8721–E8730. doi: 10.1073/pnas.1707572114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Rousseau M, Goh HMS, Holec S, Albert ML, Williams RB, Ingersoll MA, Kline KA. 2016. Bladder catheterization increases susceptibility to infection that can be prevented by prophylactic antibiotic treatment. JCI Insight 1:e88178. doi: 10.1172/jci.insight.88178 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. Di Venanzio G, Flores-Mireles AL, Calix JJ, Haurat MF, Scott NE, Palmer LD, Potter RF, Hibbing ME, Friedman L, Wang B, Dantas G, Skaar EP, Hultgren SJ, Feldman MF. 2019. Urinary tract colonization is enhanced by a plasmid that regulates uropathogenic Acinetobacter baumannii chromosomal genes. Nat Commun 10:2763. doi: 10.1038/s41467-019-10706-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. La Bella AA, Andersen MJ, Gervais NC, Molina JJ, Molesan A, Stuckey PV, Wensing L, Nobile CJ, Shapiro RS, Santiago-Tirado FH, Flores-Mireles AL. 2023. The catheterized bladder environment promotes Efg1- and Als1-dependent Candida albicans infection. Sci Adv 9:eade7689. doi: 10.1126/sciadv.ade7689 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Guiton PS, Hannan TJ, Ford B, Caparon MG, Hultgren SJ. 2013. Enterococcus faecalis overcomes foreign body-mediated inflammation to establish urinary tract infections. Infect Immun 81:329–339. doi: 10.1128/IAI.00856-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Guiton PS, Hung CS, Hancock LE, Caparon MG, Hultgren SJ. 2010. Enterococcal biofilm formation and virulence in an optimized murine model of foreign body-associated urinary tract infections. Infect Immun 78:4166–4175. doi: 10.1128/IAI.00711-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Zhang Y, Feng J, Fu H, Liu C, Yu Z, Sun Y, She X, Li P, Zhao C, Liu Y, Liu T, Liu Q, Liu Q, Li G, Wu M. 2018. Coagulation factor X regulated by CASC2c recruited macrophages and induced M2 polarization in glioblastoma multiforme. Front Immunol 9:1557. doi: 10.3389/fimmu.2018.01557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Graf C, Wilgenbus P, Pagel S, Pott J, Marini F, Reyda S, Kitano M, Macher-Göppinger S, Weiler H, Ruf W. 2019. Myeloid cell–synthesized coagulation factor X dampens antitumor immunity. Sci Immunol 4:eaaw8405. doi: 10.1126/sciimmunol.aaw8405 [DOI] [PMC free article] [PubMed] [Google Scholar]