Dysregulation of lipid homeostasis causes the deposition of lipids in the form of tiny droplets within foamy macrophages (FMs). In FMs, host-derived lipids aid in survival of various intracellular pathogens leading to sustained infection. In several infectious diseases, the transformation of macrophages into a foamy phenotype is linked to the presence of high IL-10, a potent immune-modulatory cytokine. This review aims to understand the role of IL-10 in the signaling events that are crucial in generation of FMs and highlights how various intracellular pathogens targets the IL-10–FM axis for successful establishment of infections. The review also briefly discusses how the IL-10–FM axis can be a target for developing novel therapeutic strategies to prevent intracellular infections.

Interleukin-10, a potent immune modulator, inhibits innate and adaptive immune responses like phagocytosis, cytokine induction, and Ag presentation by APC-like macrophages and dendritic cells (1, 2). The signaling pathways that mediate the effects of IL-10 are summarized in Fig. 1. APCs recognize pathogen-associated molecular patterns (PAMPs) and secrete proinflammatory cytokines like TNF-α, IFN-γ, etc., which can in turn be regulated by IL-10 in a feedback loop, thereby inhibiting TLR-dependent APC activation (except TLR3) and downstream TLR signaling pathways (3). An example of this is the “superinduction” of LPS-induced proinflammatory cytokines (TNF-α, IL-1β, and IL-12) in type I IFN signaling-deficient macrophages, which are incapable of producing IL-10 via STAT3 phosphorylation (4). Although IL-10 is crucial for controlling host inflammatory response and preserving immune homeostasis, there are also reports suggesting that IL-10, due to its immune-suppressive function, may accelerate the establishment of infectious diseases (3, 5, 6). When expressed in an untimely manner or when its expression is manipulated by the pathogen, IL-10 may inhibit the host’s protective immune responses by blocking the production of proinflammatory cytokines, like TNF-α, IL-12, etc. In addition, IL-10 can 1) directly act on the APCs and interfere with APC functions, 2) inhibit phagocytosis, and/or 3) restrict microbial killing through limiting the production of reactive oxygen species and nitrogen intermediates (2, 6). This may create a favorable condition for the pathogens to establish an infection. The impact of IL-10 in various infections is summarized in Table I.

FIGURE 1.

An overview of IL-10 signaling in macrophages. When IL-10 binds to the receptor complex, JAK1 activates STAT3 and represses proinflammatory cytokines. An IL-10–responsive gene, SOCS3 negatively regulates the IL-10/TYK2/STAT pathway. Inducing IL-10 through TLR activation and adaptor molecules MYD88 and TIRAP activates ERK1/2, p38 MAPK, and NF-κB. IFN-γ inhibits IL-10 production via altering the PI3K/AKT pathway, releasing GSK3, which suppresses IL-10 expression via CREB and AP-1 transcription factors. Thus, p38 MAPK and ERK1/2 pathways leading to IL-10 expression by macrophages are tightly controlled by IFN-γ and IL-10 itself. STAT, signal transducer and activator of transcription; MYD88, myeloid differentiation primary response protein 88; TIRAP, Toll–IL-1 receptor domain–containing adaptor protein; IFN-γR, IFN-γ receptor; GSK-3, glycogen synthase kinase 3; AP-1, activator protein 1; MSK, mitogen- and stress-activated protein kinase; TRAF6, TNF receptor–associated factor 6; RTK, receptor tyrosine kinase; EGFR, epidermal growth factor receptor; SOS, Son of Sevenless; SHC, Src homolog and collagen homolog; GRB2, growth factor receptor-bound protein 2; PPAR-γ, peroxisome proliferator–activated receptor γ; Arg-1, Arginase-1; IRAK, interleukin-1 receptor–associated kinase; TPL2, tumor progression locus 2; TAK1, transforming growth factor β–activated kinase 1; TAB, TAK-1 binding protein; TSC, tuberous sclerosis complex; mTORC1, mammalian target of rapamycin complex 1; IKK, IκB kinase; Rheb, Ras homolog enriched in brain; Mrc1, mannose receptor C type 1; MCP-1, monocyte chemoattractant protein-1.

FIGURE 1.

An overview of IL-10 signaling in macrophages. When IL-10 binds to the receptor complex, JAK1 activates STAT3 and represses proinflammatory cytokines. An IL-10–responsive gene, SOCS3 negatively regulates the IL-10/TYK2/STAT pathway. Inducing IL-10 through TLR activation and adaptor molecules MYD88 and TIRAP activates ERK1/2, p38 MAPK, and NF-κB. IFN-γ inhibits IL-10 production via altering the PI3K/AKT pathway, releasing GSK3, which suppresses IL-10 expression via CREB and AP-1 transcription factors. Thus, p38 MAPK and ERK1/2 pathways leading to IL-10 expression by macrophages are tightly controlled by IFN-γ and IL-10 itself. STAT, signal transducer and activator of transcription; MYD88, myeloid differentiation primary response protein 88; TIRAP, Toll–IL-1 receptor domain–containing adaptor protein; IFN-γR, IFN-γ receptor; GSK-3, glycogen synthase kinase 3; AP-1, activator protein 1; MSK, mitogen- and stress-activated protein kinase; TRAF6, TNF receptor–associated factor 6; RTK, receptor tyrosine kinase; EGFR, epidermal growth factor receptor; SOS, Son of Sevenless; SHC, Src homolog and collagen homolog; GRB2, growth factor receptor-bound protein 2; PPAR-γ, peroxisome proliferator–activated receptor γ; Arg-1, Arginase-1; IRAK, interleukin-1 receptor–associated kinase; TPL2, tumor progression locus 2; TAK1, transforming growth factor β–activated kinase 1; TAB, TAK-1 binding protein; TSC, tuberous sclerosis complex; mTORC1, mammalian target of rapamycin complex 1; IKK, IκB kinase; Rheb, Ras homolog enriched in brain; Mrc1, mannose receptor C type 1; MCP-1, monocyte chemoattractant protein-1.

Close modal
Table I.
Mechanistic overview of IL-10 in various infectious diseases
CategoryInfectious AgentMechanism of Action/SusceptibilityReferences
Virus HIV IL-10–592 “AA” genotype was associated with susceptibility to HIV but not TB in patients from south India. Harishankar et al. (7
HCV IL-10–592 CC genotype was associated with increased susceptibility to HCV infection. Sheneef et al. (8),
Rehermann et al. (9
EBV vIL-10 inhibited proinflammatory cytokines production and impaired NK cells mediated killing of viral particles. Jochum et al. (10
LCMV IL-10 was found to suppress effector Th1 responses and generation of memory CD4+ CD8+ T cells during acute LCMV infection. Tian et al. (11
CMV Expansion of “memory T cells” was increased dramatically in IL-10 knockout mice during CMV infection. Jones et al. (12
Mycobacteria Mycobacterium sp. In Mycobacterium tuberculosis (Mtb) infection, IL-10–deficient mice secreted more IL-12 as compared with wild-type mice. Hickman et al. (13
The sputum of TB patients showed higher levels of IL-10 and increased expression of the Mtb Ag CFP32. Huard et al. (14
Reverse transcriptase PCR analysis of bronchoalveolar lavage (BAL) cells showed higher IL-10 and TGF-β levels in TB patients than other liver diseases. Bonecini-Almeida et al. (15
Infection with Mtb “HN878” strain induced IL-10–secreting regulatory T cell population. Ordway et al. (16
M. bovis strain G18 infection led to higher IL-10 secretion and lower IL-1, IL-6, TNF, and IFN-γ secretion. Jensen et al. (17
BALB/c and C57BL/6 mice showed varied susceptibility to M. avium infection, probably due to differences in their IL-10 activity. Roque et al. (18
Intracellular bacteria Listeria monocytogenes When IL-10 receptor was blocked, mice could survive longer with better pathogen clearance. Silva et al. (19
Salmonella enterica STAT3 signaling enabled IL-10 production and intracellular bacterial multiplication. Jaslow et al. (20
Klebsiella rhinoscleromatis IL-10–deficient BALB/c mice showed fewer specialized foamy macrophages; Mikulicz cells in the lungs as compared with control mice in K. rhinoscleromatis infection. Fevre et al. (21
Orientia tsutsugamushi O. tsutsugamushi infection in macrophages induced type I IFN response and enhanced IL-10 secretion impeding the generation of Ag-specific T-helper 1 type cells and memory T cells. Min et al. (22
Chlamydia trachomatis C. trachomatis infection in PBMCs induced IL-10 and inhibited ERK- and p38 MAPK-dependent proinflammatory cytokines. Du et al. (23
 Paracoccidioides brasiliensis In IL-10–deficient mice, Paracoccidioides infection led to increase in proinflammatory cytokines IFN-γ, TNF-α, and MCP-1 levels. Costa et al. (24
Fungi Candida albicans Type 2 diabetes mellitus patients showed that with the increased IL-10 level, the risk of oral candidiasis in diabetes surged to 40%. Halimi et al. (25
IL-10–deficient mice infected with C. albicans showed better fungal clearance and less severe systemic candidiasis. Vazquez-Torres et al. (26
Histoplasma capsulatum With H. capsulatum infection in HIF-1α knockout mice, CREB-mediated IL-10 secretion was increased. Fecher et al. (27
Parasites Trypanasoma cruzi Mice infected with T. cruzi showed IL-10 secretion, M2 macrophage, and sustained parasitic infection. Ponce et al. (28
Coinfection of T. cruzi strains G and CL in THP-1 cells had lower level of anti-inflammatory cytokines, IL-10, and TGF-β than monoinfection with either strain G or CL. Oliveira et al. (29
Toxoplasma gondii Lectins microneme proteins 1 and 4 (MIC1 and MIC4) bind to TLR4 and augment IL-10 secretion. Blocking of TLR4 prevented TLR4-mediated release of IL-10 in BMDMs. Ricci-Azevedo et al. (30
Leishmania donovani Treatment with anti–IL-10 receptor Ab revealed that inhibition of IL-10 signaling enhanced the inflammatory immune response in mice and rapid killing of L. donovaniMurray et al. (31
Leishmania major L. major infection in IL-10–deficient C57BL/6 mice showed improved infection clearance compared with control mice. Belkaid et al. (32
Plasmodium chabaudi chabaudi P. chabaudi chabaudi infection in IL-10–deficient mice showed exacerbated Th1 immune response. Li et al. (33
CategoryInfectious AgentMechanism of Action/SusceptibilityReferences
Virus HIV IL-10–592 “AA” genotype was associated with susceptibility to HIV but not TB in patients from south India. Harishankar et al. (7
HCV IL-10–592 CC genotype was associated with increased susceptibility to HCV infection. Sheneef et al. (8),
Rehermann et al. (9
EBV vIL-10 inhibited proinflammatory cytokines production and impaired NK cells mediated killing of viral particles. Jochum et al. (10
LCMV IL-10 was found to suppress effector Th1 responses and generation of memory CD4+ CD8+ T cells during acute LCMV infection. Tian et al. (11
CMV Expansion of “memory T cells” was increased dramatically in IL-10 knockout mice during CMV infection. Jones et al. (12
Mycobacteria Mycobacterium sp. In Mycobacterium tuberculosis (Mtb) infection, IL-10–deficient mice secreted more IL-12 as compared with wild-type mice. Hickman et al. (13
The sputum of TB patients showed higher levels of IL-10 and increased expression of the Mtb Ag CFP32. Huard et al. (14
Reverse transcriptase PCR analysis of bronchoalveolar lavage (BAL) cells showed higher IL-10 and TGF-β levels in TB patients than other liver diseases. Bonecini-Almeida et al. (15
Infection with Mtb “HN878” strain induced IL-10–secreting regulatory T cell population. Ordway et al. (16
M. bovis strain G18 infection led to higher IL-10 secretion and lower IL-1, IL-6, TNF, and IFN-γ secretion. Jensen et al. (17
BALB/c and C57BL/6 mice showed varied susceptibility to M. avium infection, probably due to differences in their IL-10 activity. Roque et al. (18
Intracellular bacteria Listeria monocytogenes When IL-10 receptor was blocked, mice could survive longer with better pathogen clearance. Silva et al. (19
Salmonella enterica STAT3 signaling enabled IL-10 production and intracellular bacterial multiplication. Jaslow et al. (20
Klebsiella rhinoscleromatis IL-10–deficient BALB/c mice showed fewer specialized foamy macrophages; Mikulicz cells in the lungs as compared with control mice in K. rhinoscleromatis infection. Fevre et al. (21
Orientia tsutsugamushi O. tsutsugamushi infection in macrophages induced type I IFN response and enhanced IL-10 secretion impeding the generation of Ag-specific T-helper 1 type cells and memory T cells. Min et al. (22
Chlamydia trachomatis C. trachomatis infection in PBMCs induced IL-10 and inhibited ERK- and p38 MAPK-dependent proinflammatory cytokines. Du et al. (23
 Paracoccidioides brasiliensis In IL-10–deficient mice, Paracoccidioides infection led to increase in proinflammatory cytokines IFN-γ, TNF-α, and MCP-1 levels. Costa et al. (24
Fungi Candida albicans Type 2 diabetes mellitus patients showed that with the increased IL-10 level, the risk of oral candidiasis in diabetes surged to 40%. Halimi et al. (25
IL-10–deficient mice infected with C. albicans showed better fungal clearance and less severe systemic candidiasis. Vazquez-Torres et al. (26
Histoplasma capsulatum With H. capsulatum infection in HIF-1α knockout mice, CREB-mediated IL-10 secretion was increased. Fecher et al. (27
Parasites Trypanasoma cruzi Mice infected with T. cruzi showed IL-10 secretion, M2 macrophage, and sustained parasitic infection. Ponce et al. (28
Coinfection of T. cruzi strains G and CL in THP-1 cells had lower level of anti-inflammatory cytokines, IL-10, and TGF-β than monoinfection with either strain G or CL. Oliveira et al. (29
Toxoplasma gondii Lectins microneme proteins 1 and 4 (MIC1 and MIC4) bind to TLR4 and augment IL-10 secretion. Blocking of TLR4 prevented TLR4-mediated release of IL-10 in BMDMs. Ricci-Azevedo et al. (30
Leishmania donovani Treatment with anti–IL-10 receptor Ab revealed that inhibition of IL-10 signaling enhanced the inflammatory immune response in mice and rapid killing of L. donovaniMurray et al. (31
Leishmania major L. major infection in IL-10–deficient C57BL/6 mice showed improved infection clearance compared with control mice. Belkaid et al. (32
Plasmodium chabaudi chabaudi P. chabaudi chabaudi infection in IL-10–deficient mice showed exacerbated Th1 immune response. Li et al. (33

CMV, cytomegalovirus; EBV, Epstein–Barr virus; HCV, hepatitis C virus; HIV, human immunodeficiency virus; LCMV, lymphocytic choriomeningitis virus.

Dysregulated lipid metabolism in macrophages gives rise to lipid bodies amassing either triacyl glycerol–, cholesterol-, or phospholipid-rich droplets, resulting in the foamy appearance (34). Foamy macrophage (FM) biogenesis comprises complex coordinated steps of lipid input, metabolism, storage, and mobilization associated with chronic inflammation and infectious diseases (34). FMs have been well documented in several inflammatory metabolic disorders like atherosclerosis, hyperlipidemia, diabetes, insulin resistance, and obesity; cancer (non-small lung cancer, esophageal xanthoma, papillary renal cell carcinoma); and autoimmune conditions like multiple sclerosis, rheumatic diseases, and systemic lupus erythromatosis (reviewed in references 34 and 35). In addition, FMs are a characteristic feature of several intracellular bacterial and parasitic infections like tuberculosis (TB), leprosy, chlamydiasis, salmonellosis, toxoplasmosis, and infection with Klebsiella sp., Trypansoma cruzi, or Chagas’ disease (21, 36–41). Recently, proinflammatory FMs were shown to develop in a model of extracellular Trypansoma carassii infection in zebrafish (42).

IL-10 plays a crucial role in FM generation (summarized in Fig. 2). This is well demonstrated in il10−/− mice infected with Klebsiella rhinoscleromatis, which had lower frequencies of inflammatory FM known as “Mikulicz cells” and consequently had reduced bacterial burden underpinning 1) the importance of IL-10 in the generation of a foamy phenotype and 2) the fact that FMs serve as sites of bacterial replication and survival (21). IL-10 plays a crucial role in FM generation during TB as well (43). Human monocyte-derived macrophages, when treated with pleural effusion from TB patients, develop a foamy phenotype and acquire immune-suppressive properties like increased expression of IL-10, CD163, and PD-L1 expression and reduced TNF-α secretion (43). However, this phenomenon is not observed upon treatment of monocyte-derived macrophages with IL-10–depleted pleural effusion, underpinning the critical role of IL-10 in the FM differentiation process (43). Additionally, alveolar macrophages as observed in fixed sections of lung biopsies from individuals with untreated TB were found to be foamy in nature, to express M2 markers CD68 and CD163, and to have high levels of surface PD-L1, suggesting that they are immune suppressive (44). FM generation via IL-10 and TLR-mediated mechanisms has been reported in instances of infection by other mycobacteria like Mycobacterium leprae as well (45).

FIGURE 2.

Role of IL-10 in foamy macrophage formation. IL-10 increases CD36 and ACAT cell-surface expression and promotes FM synthesis and lipid body formation via an IL-10/STAT3 axis. Pathogens such as Mtb, M. leprae, and M. avium-containing phagosomes consume these lipid compounds and allow bacilli persistence. By activating TLR2, Chlamydia sp. increase IL-10 cytokine in host macrophages, causing FMs by blocking LXR signaling and limiting cholesterol efflux through ABCA1 and ABCG1 transporters. Protozoan parasites like T. gondi, L. major, and H. capsulatum induce FM via TLR2/TLR4 through IL-10–dependent pathway by upregulating SR-A and CD36, which facilitate uptake of modified LDL and cholesteryl ester accumulation. HLA-DR, human leukocyte Ag – DR isotype; CD163, cluster of differentiation 163; PD-L1, programmed death ligand 1; LB, lipid body; AA, arachidonic acid; LOX, lipoxygenase; COX, cyclooxygenase; LTB4, leukotriene B4; RXR, retinoid X receptor; LXR, liver X receptor; FXR, farnesoid X receptor; FXRE, FXR-responsive elements; miR-33, microRNA-33; SREBP-2, sterol regulatory element-binding protein 2; HMG-CoA reductase, β-hydroxy β-methylglutaryl-CoA reductase; Chol, cholesterol; FA, fatty acid; TG, triglyceride; ABCA1, ATP-binding cassette transporter A1; ABCG1, ATP-binding cassette, subfamily G member 1; ApoA1, apolipoprotein AI; HDL, high-density lipoprotein.

FIGURE 2.

Role of IL-10 in foamy macrophage formation. IL-10 increases CD36 and ACAT cell-surface expression and promotes FM synthesis and lipid body formation via an IL-10/STAT3 axis. Pathogens such as Mtb, M. leprae, and M. avium-containing phagosomes consume these lipid compounds and allow bacilli persistence. By activating TLR2, Chlamydia sp. increase IL-10 cytokine in host macrophages, causing FMs by blocking LXR signaling and limiting cholesterol efflux through ABCA1 and ABCG1 transporters. Protozoan parasites like T. gondi, L. major, and H. capsulatum induce FM via TLR2/TLR4 through IL-10–dependent pathway by upregulating SR-A and CD36, which facilitate uptake of modified LDL and cholesteryl ester accumulation. HLA-DR, human leukocyte Ag – DR isotype; CD163, cluster of differentiation 163; PD-L1, programmed death ligand 1; LB, lipid body; AA, arachidonic acid; LOX, lipoxygenase; COX, cyclooxygenase; LTB4, leukotriene B4; RXR, retinoid X receptor; LXR, liver X receptor; FXR, farnesoid X receptor; FXRE, FXR-responsive elements; miR-33, microRNA-33; SREBP-2, sterol regulatory element-binding protein 2; HMG-CoA reductase, β-hydroxy β-methylglutaryl-CoA reductase; Chol, cholesterol; FA, fatty acid; TG, triglyceride; ABCA1, ATP-binding cassette transporter A1; ABCG1, ATP-binding cassette, subfamily G member 1; ApoA1, apolipoprotein AI; HDL, high-density lipoprotein.

Close modal

Macrophage differentiation occurs in response to environmental cues like cytokines and microbial PAMPs. Broadly and traditionally, macrophages are classified into classically activated proinflammatory M1 and alternatively activated, anti-inflammatory/regulatory M2 phenotypes (46). M1 macrophages are formed in response to IFN-γ and LPS, whereas M2 macrophages arise in the presence of either IL-4 + IL-13 (M2a) or TLR ligands (M2b) or IL-10 (M2c) (47). Transcriptome analysis has helped in identification of a wide spectrum of macrophage categories each having a unique molecular signature (48). Macrophages are therefore highly heterogenous and possess immense plasticity, and their categories appear to exist as a continuum (reviewed in references 46, 47, 49, and 50). Evidence demonstrates that in atherosclerotic plaques, cancer, and infections like tuberculosis, FMs display anti-inflammatory properties akin to those observed in M2-type macrophages, which are regulatory, have reduced capacity for NO generation, are less microbicidal, and promote tissue repair (51–53). Evidence of the link between M2 macrophages and the foamy phenotype comes from studies in which alternative differentiation of macrophages from diabetic patients, in the presence of IL-4, IL-10, or OVA immune complex, leads to increased FM formation compared with classical differentiation in the presence of IFN-γ (53). The induction of the FM is mediated by expression of scavenger receptor SR-A1 and CD36 and endoplasmic reticulum stress-mediated JNK activation and increased peroxisome proliferator-activated receptor-γ (PPAR-γ) expression (53). However, it is important to point out that in a model of extracellular trypanosome infection of zebrafish, the FMs generated were proinflammatory and contributed to susceptibility to infection (42), highlighting the heterogeneity of FM, which can be dependent among other things on the nature of the pathogen: e.g., intracellular versus extracellular. The M2-like nature of FMs promotes establishment of infection in TB and tumor activity in cancer; however, it has been suggested that M2 FMs in atherosclerotic lesions can be antiartherogenic by countering the proinflammatory M1 macrophages (51).

FM formation during infection is affected by IL-10 as well as pathogen-induced alteration of lipid homeostasis pathways. This review focuses on three major aspects: 1) contribution of IL-10 to pathogenesis and FM formation during various infections, 2) pathways involved in FM formation and how the IL-10–FM axis is exploited by various pathogens to favor intracellular survival, and finally, 3) how the IL-10–FM signaling axis can serve as a target for therapeutic interventions in infectious disease.

Macrophages are flexible and often adapt their functions according to environmental stimuli such as stress, infection, disease, etc. Dysregulated lipid metabolism in macrophages causes the production of lipid droplets or lipid bodies, in which they can accumulate triacylglycerol, cholesterol, enzymes like cyclooxygenases 5- and 15-lypoxygenases leukotriene C4-synthase, phospholipase A2, MAP kinases, or other phospholipid-rich droplets, resulting in the foamy appearance of the macrophages (34, 54). Such lipid-rich FMs are exploited by pathogens for a variety of purposes. They can serve as nutrient reservoirs, interfere with host signaling cascades, or help in immune escape (36). A classic example of this is Mycobacterium tuberculosis (Mtb), which efficiently uses host cholesterol to meet its carbon and energy requirements with the help of the protein Mce4 (55). Mtb infection increases the expression of microRNA (miR-33) in macrophages suppressing lipid catabolism via autophagy to preserve intracellular lipid stores (56). Within FMs, Mtb uses host lipids for synthesis of its cell wall and survival during dormancy (3). Interestingly, not just Mtb but also other pathogens promote FM generation, leading to clinical manifestations (54). Various factors and signaling cascades involved in FM formation and how various pathogens manipulate IL-10–FM signaling to favor its intracellular survival are described below and summarized in Table II.

Table II.
Hijacking the IL-10–FM signaling axis by various pathogens to suit its intracellular survival
PathogenCharacteristicsReferences
TLRs 
M. bovis BCG TLR2-deficient mice failed to form foamy macrophage formation during M. bovis BCG infection and showed reduced IL-10 levels. D’Avila et al. (57
M. leprae M. leprae infection in peritoneal macrophages revealed that heterodimerization of TLR2/TLR6 and IL-10 secretion were important for FMs. Mattos et al. (45
M. leprae was phagocytosed by Schwann cells isolated from LL patients and showed higher IL-10 and PGE2 levels were observed along with compromised mycobactericidal function of Schwann cells. Mattos et al. (58
Schwann cells isolated from TLR6 knockout C57BL/6 mice had reduced phagocytosis of M. leprae cells followed by limited capability to generate lipid bodies following infection. Mattos et al. (58
Pro- and anti-inflammatory cytokines 
Nocardia brasiliensis In BMDMs isolated from BALB/C mice, higher IL-10 secretion and foam cell formation were observed postinfection with N. brasiliensisRosas-Taraco et al. (59
Foam cells were developed in bone marrow–derived dendritic cells after N. brasiliensis infection. Meester et al. (60
Chlamydia pneumoniae C. pneumoniae infection favored Nlrp3-induced IL-1β signaling that reduced Gpr109a and ABCA1 expression. This helped in retaining the cholesterol intracellularly, and FM formation occurred. Tumurkhuu et al. (61
C. pneumoniae-induced FM formation inhibited by resveratrol probably via by IL-17A signaling. Di Pietro et al. (62
Mycoplasma pneumoniae A patient suffering from M. pneumoniae–induced cryptogenic organizing pneumonia showed high inflammation and accumulation of FMs in the lungs. Zeidan et al. (63
Transcription factors 
 SARS Cov-2 SARS-CoV-2 virus showed preferentially replication in foam cells than macrophage probably due to high IL-10 and reduced expression of transcription factors IRF1 and IRF1. Eberhardt et al. (64
Mycobacterium tuberculosis Mtb-infected foam cells showed activation of NF-κB signaling. Agarwal et al. (65
In HIF-1α–deficient mice infected with Mtb, BDMDs exhibited reduced FM due to diminished IFN-γ/HIF-1α signaling, potentially influenced by IL-10. Knight et al. (66
Silencing of PPAR-γ and TR4 contributed inhibited Mtb-induced IL-10 FM formation. Mahajan et al. (52
M. bovis BCG TLR2 and PPAR-γ both were important for the formation lipid body in M. bovis BCG infection. Almeida et al. (67
Mycobacterium avium subsp. paratuberculosis (MAP) M. avium, in macrophages exploited miR-129 expression targeting transcription factor Sp2 to modulate lipid metabolism. Wright et al. (68
Chlamydia pneumoniae C. pneumoniae infection induced foam cell formation by upregulating ACAT1 in PPAR-γ–dependent pathway. Mei et al. (69
C. pneumoniae elicited lipid accumulation via TLR2-mediated inhibition of LXR signaling and FM formation. Cao et al. (70
Lipid and lipoprotein transporters 
Mycobacterium tuberculosis Treatment of macrophages from IL-10 knockout mice with TB pleural effusion followed by Mtb infection showed lowered FM formation than control mice, suggesting a IL-10/STAT3–dependent activation of ACAT expression. Genoula et al. (43
M. leprae Costaining of skin biopsy samples from leprae patients showed FM formation, increased in PGE2 expression, and elevated IL-10/IL-12 ratio. Mattos et al. (45
In the skin lesion samples from LL patients, colocalization of lipid markers ApoB and oxLDL with macrophage markers CD209+ CD163+ suggested that IL-10 induced foam cell formation. Montoya et al. (71
M. leprae–infected foamy macrophages in LL lesions have shown oxidized phospholipids as well as expression of genes LDL receptor-related protein 1 (LRP-1) and scavenger receptors SR-A1, and SR-B1. Mattos et al. (37
Leishmania major L. major infection in apoE KO mice exhibited elevated IL-10, fewer foam cells, and increased atherosclerotic lesions. Fernandes et al. (72
Toxoplasma gondii During chronic latent T. gondii infection, TgACAT1 and TgACAT2 localized to endoplasmic reticulum and inhibited ACAT1 and ACAT2 in the host hepatocyte, resulting in FM production. Prandota et al. (73
PathogenCharacteristicsReferences
TLRs 
M. bovis BCG TLR2-deficient mice failed to form foamy macrophage formation during M. bovis BCG infection and showed reduced IL-10 levels. D’Avila et al. (57
M. leprae M. leprae infection in peritoneal macrophages revealed that heterodimerization of TLR2/TLR6 and IL-10 secretion were important for FMs. Mattos et al. (45
M. leprae was phagocytosed by Schwann cells isolated from LL patients and showed higher IL-10 and PGE2 levels were observed along with compromised mycobactericidal function of Schwann cells. Mattos et al. (58
Schwann cells isolated from TLR6 knockout C57BL/6 mice had reduced phagocytosis of M. leprae cells followed by limited capability to generate lipid bodies following infection. Mattos et al. (58
Pro- and anti-inflammatory cytokines 
Nocardia brasiliensis In BMDMs isolated from BALB/C mice, higher IL-10 secretion and foam cell formation were observed postinfection with N. brasiliensisRosas-Taraco et al. (59
Foam cells were developed in bone marrow–derived dendritic cells after N. brasiliensis infection. Meester et al. (60
Chlamydia pneumoniae C. pneumoniae infection favored Nlrp3-induced IL-1β signaling that reduced Gpr109a and ABCA1 expression. This helped in retaining the cholesterol intracellularly, and FM formation occurred. Tumurkhuu et al. (61
C. pneumoniae-induced FM formation inhibited by resveratrol probably via by IL-17A signaling. Di Pietro et al. (62
Mycoplasma pneumoniae A patient suffering from M. pneumoniae–induced cryptogenic organizing pneumonia showed high inflammation and accumulation of FMs in the lungs. Zeidan et al. (63
Transcription factors 
 SARS Cov-2 SARS-CoV-2 virus showed preferentially replication in foam cells than macrophage probably due to high IL-10 and reduced expression of transcription factors IRF1 and IRF1. Eberhardt et al. (64
Mycobacterium tuberculosis Mtb-infected foam cells showed activation of NF-κB signaling. Agarwal et al. (65
In HIF-1α–deficient mice infected with Mtb, BDMDs exhibited reduced FM due to diminished IFN-γ/HIF-1α signaling, potentially influenced by IL-10. Knight et al. (66
Silencing of PPAR-γ and TR4 contributed inhibited Mtb-induced IL-10 FM formation. Mahajan et al. (52
M. bovis BCG TLR2 and PPAR-γ both were important for the formation lipid body in M. bovis BCG infection. Almeida et al. (67
Mycobacterium avium subsp. paratuberculosis (MAP) M. avium, in macrophages exploited miR-129 expression targeting transcription factor Sp2 to modulate lipid metabolism. Wright et al. (68
Chlamydia pneumoniae C. pneumoniae infection induced foam cell formation by upregulating ACAT1 in PPAR-γ–dependent pathway. Mei et al. (69
C. pneumoniae elicited lipid accumulation via TLR2-mediated inhibition of LXR signaling and FM formation. Cao et al. (70
Lipid and lipoprotein transporters 
Mycobacterium tuberculosis Treatment of macrophages from IL-10 knockout mice with TB pleural effusion followed by Mtb infection showed lowered FM formation than control mice, suggesting a IL-10/STAT3–dependent activation of ACAT expression. Genoula et al. (43
M. leprae Costaining of skin biopsy samples from leprae patients showed FM formation, increased in PGE2 expression, and elevated IL-10/IL-12 ratio. Mattos et al. (45
In the skin lesion samples from LL patients, colocalization of lipid markers ApoB and oxLDL with macrophage markers CD209+ CD163+ suggested that IL-10 induced foam cell formation. Montoya et al. (71
M. leprae–infected foamy macrophages in LL lesions have shown oxidized phospholipids as well as expression of genes LDL receptor-related protein 1 (LRP-1) and scavenger receptors SR-A1, and SR-B1. Mattos et al. (37
Leishmania major L. major infection in apoE KO mice exhibited elevated IL-10, fewer foam cells, and increased atherosclerotic lesions. Fernandes et al. (72
Toxoplasma gondii During chronic latent T. gondii infection, TgACAT1 and TgACAT2 localized to endoplasmic reticulum and inhibited ACAT1 and ACAT2 in the host hepatocyte, resulting in FM production. Prandota et al. (73

LL, lepromatous leprosy; TLR, Toll-like receptor.

Toll-like receptors (TLRs) in IL-10–mediated FM formation

In macrophages, the recognition of microbial PAMPs by TLRs triggers signaling events leading to FM formation (summarized in Fig. 2). Mycobacterium bovis Bacillus Calmette-Guérin (BCG) infection in C57BL/6 mice induces TLR2-dependent FM formation (57). The same study also demonstrated that the accumulation of lipid bodies in macrophages was directly linked to the presence of PGE2 and IL-10 (58). M. leprae infection–induced lipid droplet accumulation in mouse peritoneal macrophages was found to be partially dependent on TLR2 and TLR6 (45). Also, culture medium from such infected cells had a high IL-10/IL-12 ratio and could induce a foamy phenotype in uninfected cells (45). M. leprae–infected Schwann cells from lepromatous leprosy patients and have a foamy phenotype dependent on TLR6 but not on TLR2 (58). Additionally, M. leprae–induced lipid droplet biogenesis correlates positively with increased IL-10 and PGE2 and reduced IL-12 and NO, highlighting the M2-like nature of FM and underpinning the importance of IL-10 in conjunction with TLR signaling in the formation of FM (58). These reports suggest that TLR signaling and PGE2 and IL-10 expression act in concert during mycobacterial infection to induce lipid droplet biogenesis and FM formation. The TLR involved, however, can vary depending on the infectious organism and the nature of the macrophage.

Impact of proinflammatory cytokines versus IL-10 on FM formation

FMs are associated with conditions of chronic inflammation ranging from noninfectious atherosclerotic plaques to infectious tuberculous granulomas. The biogenesis and nature of the FM is disease dependent; for example, FMs in tuberculosis accumulate triglycerides, as opposed to FMs in atherosclerosis, which are rich in cholesterol (34). Similarly, the origin of FMs in various infection conditions is also heterogenous and can be influenced by several cytokines. In TB, the generation of host proinflammatory responses against an infection like increase in IFN-γ can drive FM formation (66). IFN-γ–induced HIF-1α signaling drives lipid droplet accumulation in Mtb-infected mouse bone marrow–derived macrophages (BMDMs) as well as a mouse model of in vivo Mtb infection (66). Importantly, Mtb cannot use host lipids from droplets that are formed in the presence of IFN-γ, suggesting that this might be a protective host immune response (66). In another example, Chlamydia pneumoniae, an obligate intracellular pathogen known for accelerating atherosclerosis, induces proinflammatory IL-1β through NLRP3 activation, and this in turn inhibits Gpr109a- and ABCA1-mediated cholesterol efflux, leading to accumulation of cholesterol and formation of foamy macrophages, in which C. pneumoniae can survive and persist (61). This is an excellent example of a pathogen exploiting a feedback mechanism to create a survival niche (61). As opposed to the above examples, pathogens like the actinomycete Nocardia brasiliensis induce an immune-suppressive response characterized by expansion of T regulatory cells and elevated TGF-β and IL-10 in infected mice (59). Also, in vitro N. brasiliensis infection in murine BMDMs and bone marrow–derived dendritic cells promotes intracellular accumulation of lipids and development of a foamy phenotype (74). CFSE-labeled BMDMs and bone marrow–derived dendritic cells when transferred into actinomycetoma lesions of infected mice were positive for staining for lipid droplets (60). Even though there is no direct evidence, it can be speculated that the immune-suppressive environment observed during in vivo N. brasiliensis infection might have a role to play in formation of FM (59, 60). It is worth pointing out that as a contrast to observations in murine models of infection in which IFN-γ was found to play a critical role in FM formation, a positive correlation was found between IL-10 levels and pleural CD14+ lipid-laden cells from TB patients, which could mean that IL-10 either was secreted by FM or was involved in their generation (17). Cell free TB pleural effusion is capable of inducing FM formation in monocyte-derived macrophages from healthy donors; however, IL-10–depleted pleural effusion lost this capacity, underpinning the role of IL-10 in FM formation (43).

Transcription factors in IL-10–mediated FM formation

PPARs are transcription factors that maintain homeostasis between lipids, lipoproteins, and glucose metabolism. PPAR-γ binds to fatty acids and eicosanoids and is involved in modulating lipid biogenesis (74). TR4 (testicular receptor 4) functions as a lipid sensor to induce FM partly by regulating oxLDL receptor CD36 (52, 75). PPAR-γ and TR4 contribute to lipid biogenesis by regulating oxLDL receptor CD36, inhibiting phagolysomsome maturation, increasing IL-10, and enhancing alternate polarization of macrophages, eventually leading to the creation of a foamy niche, which enables intracellular Mtb survival (52). Silencing of PPAR-γ and TR4 results in reduced intracellular Mtb survival, IL-10, and arginase activity and increased NO, IL-6, and TNF-α, demonstrating the critical role of the PPAR-γ/ΤΡ4−IL-10 alternate polarization axis in creating a foamy phenotype conducive to Mtb survival (52). TLR2-induced PPAR-γ was found to be important for the formation of lipid bodies during infection with M. bovis BCG (67). C. pneumoniae infection induces FM formation by upregulating ACAT1 (acetyl-CoA acetyltransferase 1) through a PPAR-γ-dependent pathway (69). Examples of other transcription factors involved in FM formation are the microRNA miR-129–regulated Sp2 in Mycobacterium avium–infected macrophages (68) and the liver X receptor (LXR), a cholesterol efflux regulator, in C. pneumoniae infection (70, 76). However, there is no evidence of the involvement of IL-10 in FM formation mediated either by Sp2 or LXR.

Role of lipoproteins in IL-10–mediated FM formation

The generation of a FM results from breakdown in the balance between the influx and efflux of lipoproteins. Free cholesterol from lipoproteins, endocytosed through cell surface receptors such as CD36, is esterified by the endoplasmic reticulum enzyme ACAT, leading to the formation of lipid droplets inside FMs (77). Evidence for the critical role of ACAT in the formation of lipid droplets comes from studies in which dexamethasone was shown to upregulate ACAT, consequently leading to the formation of FMs (78). Inhibition of ACAT causes accumulation of free cholesterol, leading to the induction of a stress response, resulting in apoptosis (79). Pleural effusion from TB patients was found to be capable of increasing ACAT expression in human monocyte-derived macrophages, leading to an increase in the synthesis of lipid bodies and development of a foamy phenotype in these cells (43). This phenomenon was not observed when IL-10 was depleted from the pleural effusion or when ACAT was inhibited using a specific inhibitor, Sandoz 58-035 (43).

Apolipoprotein E (ApoE), a glycosylated protein, is an important component of triglyceride-rich lipid complexes, which helps in lipoprotein clearance from blood. Apoe−/− mice, due to defective clearance of very-low-density lipoproteins and chylomicrons, develop atherosclerosis (72, 80). Further, atherosclerosis is accelerated in ApoE and IL-10 double knockout mice, demonstrating that the presence of IL-10 is protective during atherosclerosis (81). Conversely, increase in IL-10 in Apoe−/− mice during hypoxic conditions coincided with decreased plaque formation (82). Interestingly, Apoe−/− mice are susceptible to fungal, viral, and bacterial infections, including tuberculosis, due to their inability to mount protective innate and adaptive immune responses (83). Currently, there is no study that directly links IL-10, ApoE and FM during infection. However, evidence from studies on atherosclerosis and the Apoe−/− mouse model warrants further exploration of this link.

In skin lesion biopsies from disseminated lepromatous leprosy patients, ApoB, the major lipoprotein of oxLDL, colocalizes with CD209+CD163+ IL-10–secreting foamy macrophages, which are heavily infected with M. leprae as opposed to self-healing, contained tuberculoid lesions (71). This provides evidence that 1) FM are niches for mycobacterial growth and 2) a link exists between the IL-10–secreting M2-like nature and FMs.

Scavenger receptors (SRs) in IL-10–mediated FM formation

IL-10 promotes cholesterol uptake from modified lipoproteins in macrophages and their transformation into FM by increasing the expression of scavenger receptor SR-B1 (CD36) and scavenger receptor A (SR-A) (75). Membrane proteins like fatty acid translocase (FAT/CD36), MSR1/CD204, and macrophage receptor with collagenous domain (Marco) mediate the uptake of LDL into macrophages, which are then metabolized to triacylglycerides, phospholipids, and cholesterol, leading to the development of a foamy phenotype (84, 85). TB pleural effusion increases CD36 expression and foamy phenotype in healthy macrophages in an IL-10–dependent fashion (43). Foamy M. leprae–infected macrophages in lepromatous leprosy lesions show increased accumulation of cholesterol and higher RNA and protein levels of LDL-R, CD36, SRA-1, SR-B1, and LRP-1 (37). These same lesions were also reported by the same authors to have a high IL-10/IL-12 ratio dependent on TLR2 (45), underpinning the nexus between TLR signaling, IL-10, and scavenger receptor expression, leading to accumulation of lipid droplets in infected macrophages.

IL-10 is an immune function modulator that inhibits infection-induced Th1-type cytokines (3). However, in the chronic phase of infection, elevated IL-10 can limit pathology due to exaggerated inflammation (86). Finding the cellular origins of IL-10 in response to infectious diseases may help to develop immune therapeutics to treat infections and their pathogenic sequelae, such as FM favoring pathogen survival and chronicity (6, 87). Several therapeutics that target IL-10 or FM formation have been proposed to prevent intracellular survival of pathogens, leading to chronic infectious conditions (88, 89).

Mtb induces IL-10 in vivo as well as in vitro and the role of IL-10 in TB has been extensively studied (86, 90). Host-directed therapies targeting IL-10 to improve TB prognosis have produced promising results (91, 92). Blocking IL-10 by administering anti–IL-10 receptor 1 (IL-10R1) during Mtb infection in susceptible CBA/J mice reduced bacterial load and increased survival (93). Administration of IL-10R1 along with BCG vaccination resulted in better bacterial control and survival in CBA/J mice upon subsequent Mtb challenge (92). This was associated with presence of higher frequencies of CD4+ and CD8+ memory cells and reduced Th1/Th17 cytokines (92). Aerosol delivery of peptide inhibitors/analogs targeting either STAT3 or IL-10 receptor α chain (IL-10Ra) in Mtb-infected mice increased NO but decreased arginase activity, ultimately leading to reduced bacillary load (91). Similarly, blockade of IL-10R in Leishmania donovani–infected BALB/c mice resulted in rapid parasite killing, reduced tissue damage and increased responsiveness to antimony chemotherapy (31). Another novel strategy to counteract IL-10 during Mtb infection would be to develop inhibitors that target the 1115 leucine-rich repeat domains of TLR2, which binds to Mtb proteins like PPE18, leading to elevated IL-10 (90).

In TB, lipid metabolic pathways can also be targeted for host-directed therapy to reduce FM production. Statins, which inhibit 3-hydroxy-3-methylglutaryl-CoA reductase and are commonly used for treating atherosclerosis and hyperlipidemia, could potentially be used for TB treatment (94). Administration of simvastatin reduced the duration of TB chemotherapy in Mtb-infected mice (95), and use of lovastatin in in-vitro M. leprae–infected mice diminished cholesterol colocalization to pathogen-containing phagosomes and decreased intracellular survival of M. leprae (37). Statins, e.g., fluvastatin, simvastatin, and atorvastatin, have been found to inhibit TLR4-mediated inflammatory pathways that lead to lipid droplet biogenesis (96). Known antioxidant resveratrol suppresses superoxide anion and IL-17A levels in C. pneumoniae–infected macrophages and inhibits their conversion to FM (62). This impact of resveratrol is overridden by GW9662, a specific PPAR-γ agonist, suggesting that perhaps resveratrol functions via suppression of PPAR-γ–mediated FM formation pathways (62). GW501516, a PPAR-δ agonist that increases cholesterol efflux (97), and GQ-177, a PPAR-γ partial agonist that reduces atherosclerosis in Ldlr−/− mice fed a high diabetogenic diet (97), could also be used to correct metabolic imbalances and FM formation in infections. Similarly, synthetic LXR and retinoid X receptor agonists reduced atherosclerosis in Ldlr−/− and Apoe−/− mice by promoting cholesterol efflux (98, 99).

The study of FM in infection has provided valuable insights into the signaling events associated with the generation of FM. The formation of FM is deeply linked to the nature of the infecting organism, and the pathways can vary substantially. However, within these intricate processes, IL-10 emerges as a pivotal modulator, influencing immune and metabolic regulatory factors. Therefore, the use of immune-based host-directed therapies, like IL-10 receptor Ab (92, 93), represents the next generation of treatments that are being pursued with great interest for developing effective therapeutics to manage infectious diseases and associated clinical pathology (100).

Given the complexity of the immune response, it is advisable to study the cellular and molecular processes of IL-10 and FM together to get a holistic overview. Considering recent advances, it is evident that by expanding our knowledge on IL-10–mediated FM formation, new (and safer) therapeutic targets could be identified for diseases that are often associated with poor clinical outcome such as TB. This insight is further supported by the extensive studies on FMs and their role in disease pathogenesis, notably in atherosclerosis (101, 102), and could serve as an excellent example of how one can exploit the potential of IL-10 as an immune-therapeutic target against diverse pathogenic challenges. The specific molecular events in IL-10–induced FM formation warrant further investigation for a comprehensive understanding.

The authors have no financial conflicts of interest.

We thank Niteen Pathak for help and suggestions. The figures are created using Biorender.com, an illustration tool, in accordance with its term and services.

This work was supported by a research associateship from the Department of Biotechnology of the Government of India (to K.S.). A.A. is supported by the Department of Biotechnology, Govt. of India BioCARe fellowship. The authors gratefully acknowledge the financial support (to S.M.) by Grant JCB/2021/000035 from the Science and Engineering Research Board and the Department of Science and Technology of the Government of India, Grant BT/PR51149/MED/29/1660/2023 from the Department of Biotechnology of the Government of India, Grant 37WS(0020)/2023-24/EMR-II/ASPIRE from the Council of Scientific and Industrial Research of the Government of India, Grant 2021-10087/GTGE/ADHOC-BMS from the Indian Council of Medical Research, and a core grant from Centre for DNA Fingerprinting and Diagnostics by the Department of Biotechnology.

APC

antigen-presenting cell

BCG

Bacillus Calmette-Guérin

BMDM

bone marrow–derived macrophage

FM

foamy macrophage

HIF-1α

hypoxia-inducible factor-1α

LXR

liver X receptor

Mtb

Mycobacterium tuberculosis

oxLDL

oxidized low-density lipoprotein

PAMP

pathogen-associated molecular pattern

PPAR

peroxisome proliferator-activated receptor

SR

scavenger receptor

TB

tuberculosis

1
Saraiva
,
M.
,
A.
O’Garra
.
2010
.
The regulation of IL-10 production by immune cells
.
Nat. Rev. Immunol.
10
:
170
181
.
2
Moore
,
K. W.
,
R.
de Waal Malefyt
,
R. L.
Coffman
,
A.
O’Garra
.
2001
.
Interleukin-10 and the interleukin-10 receptor
.
Annu. Rev. Immunol.
19
:
683
765
.
3
Bhat
,
K. H.
,
S.
Mukhopadhyay
.
2015
.
Macrophage takeover and the host–bacilli interplay during tuberculosis
.
Future Microbiol.
10
:
853
872
.
4
Chang
,
E. Y.
,
B.
Guo
,
S. E.
Doyle
,
G.
Cheng
.
2007
.
Involvement of the type I IFN production and signaling pathway in lipopolysaccharide-induced IL-10 production
.
J. Immunol.
178
:
6705
6709
.
5
McNab
,
F. W.
,
J.
Ewbank
,
R.
Rajsbaum
,
E.
Stavropoulos
,
A.
Martirosyan
,
P. S.
Redford
,
X.
Wu
,
C. M.
Graham
,
M.
Saraiva
,
P.
Tsichlis
, et al
2013
.
TPL-2–ERK1/2 signaling promotes host resistance against intracellular bacterial infection by negative regulation of type I IFN production
.
J. Immunol.
191
:
1732
1743
.
6
Mittal
,
S. K.
,
P. A.
Roche
.
2015
.
Suppression of antigen presentation by IL-10
.
Curr. Opin. Immunol
34
:
22
27
.
7
Harishankar
,
M.
,
H.
Ravikrishnan
,
A.
Ravishankar
,
L. E.
Hanna
,
S.
Swaminathan
,
P.
Selvaraj
,
R.
Bethunaickan
.
2018
.
IL-10 promoter-592 polymorphism may influence susceptibility to HIV infection in South Indian population
.
Curr. HIV Res.
16
:
58
63
.
8
Sheneef
,
A.
,
M. M.
Esmat
,
A. N.
Mohammad
,
A. A.
Mahmoud
,
H. M.
Moghazy
,
A. K.
Noureldin
.
2017
.
Interleukin-10 and interferon gamma gene polymorphisms and hepatitis C virus-related liver cirrhosis risk
.
J. Interferon Cytokine Res.
37
:
175
180
.
9
Rehermann
,
B.
,
M.
Nascimbeni
.
2005
.
Immunology of hepatitis B virus and hepatitis C virus infection
.
Nat. Rev. Immunol.
5
:
215
229
.
10
Jochum
,
S.
,
A.
Moosmann
,
S.
Lang
,
W.
Hammerschmidt
,
R.
Zeidler
.
2012
.
The EBV immunoevasins vIL-10 and BNLF2a protect newly infected B cells from immune recognition and elimination
.
PLoS Pathog.
8
:
e1002704
.
11
Tian
,
Y.
,
S. B.
Mollo
,
L. E.
Harrington
,
A. J.
Zajac
.
2016
.
IL-10 regulates memory T cell development and the balance between Th1 and follicular Th cell responses during an acute viral infection
.
J. Immunol.
197
:
1308
1321
.
12
Jones
,
M.
,
K.
Ladell
,
K. K.
Wynn
,
M. A.
Stacey
,
M. F.
Quigley
,
E.
Gostick
,
D. A.
Price
,
I. R.
Humphreys
.
2010
.
IL-10 restricts memory T cell inflation during cytomegalovirus infection
.
J. Immunol.
185
:
3583
3592
.
13
Hickman
,
S. P.
,
J.
Chan
,
P.
Salgame
.
2002
.
Mycobacterium tuberculosis induces differential cytokine production from dendritic cells and macrophages with divergent effects on naive T cell polarization
.
J. Immunol.
168
:
4636
4642
.
14
Huard
,
R. C.
,
S.
Chitale
,
M.
Leung
,
L. C. O.
Lazzarini
,
H.
Zhu
,
E.
Shashkina
,
S.
Laal
,
M. B.
Conde
,
A. L.
Kritski
,
J. T.
Belisle
, et al
2003
.
The Mycobacterium tuberculosis complex-restricted gene cfp32 encodes an expressed protein that is detectable in tuberculosis patients and is positively correlated with pulmonary interleukin-10
.
Infect. Immun.
71
:
6871
6883
.
15
Bonecini-Almeida
,
M. G.
,
J. L.
Ho
,
N.
Boéchat
,
R. C.
Huard
,
S.
Chitale
,
H.
Doo
,
J.
Geng
,
L.
Rego
,
L. C. O.
Lazzarini
,
A. L.
Kritski
, et al
2004
.
Down-modulation of lung immune responses by interleukin-10 and transforming growth factor β (TGF-β) and analysis of TGF-β receptors I and II in active tuberculosis
.
Infect. Immun.
72
:
2628
2634
.
16
Ordway
,
D.
,
M.
Henao-Tamayo
,
M.
Harton
,
G.
Palanisamy
,
J.
Troudt
,
C.
Shanley
,
R. J.
Basaraba
,
I. M.
Orme
.
2007
.
The hypervirulent Mycobacterium tuberculosis strain HN878 induces a potent TH1 response followed by rapid down-regulation
.
J. Immunol.
179
:
522
531
.
17
Jensen
,
K.
,
J. M.
Stevens
,
E. J.
Glass
.
2019
.
Interleukin 10 knock-down in bovine monocyte-derived macrophages has distinct effects during infection with two divergent strains of Mycobacterium bovis
.
PLoS One
14
:
e0222437
.
18
Roque
,
S.
,
C.
Nobrega
,
R.
Appelberg
,
M.
Correia-Neves
.
2007
.
IL-10 underlies distinct susceptibility of BALB/c and C57BL/6 mice to Mycobacterium avium infection and influences efficacy of antibiotic therapy
.
J. Immunol.
178
:
8028
8035
.
19
Silva
,
R. A.
,
R.
Appelberg
.
2001
.
Blocking the receptor for interleukin 10 protects mice from lethal listeriosis
.
Antimicrob. Agents Chemother.
45
:
1312
1314
.
20
Jaslow
,
S. L.
,
K. D.
Gibbs
,
W. F.
Fricke
,
L.
Wang
,
K. J.
Pittman
,
M. K.
Mammel
,
J. T.
Thaden
,
V. G.
Fowler
,
G. E.
Hammer
,
J. R.
Elfenbein
, et al
2018
.
Salmonella activation of STAT3 signaling by SarA effector promotes intracellular replication and production of IL-10
.
Cell Rep.
23
:
3525
3536
.
21
Fevre
,
C.
,
A. S.
Almeida
,
S.
Taront
,
T.
Pedron
,
M.
Huerre
,
M. C.
Prevost
,
A.
Kieusseian
,
A.
Cumano
,
S.
Brisse
,
P. J.
Sansonetti
,
R.
Tournebize
.
2013
.
A novel murine model of rhinoscleroma identifies Mikulicz cells, the disease signature, as IL-10 dependent derivatives of inflammatory monocytes
.
EMBO Mol. Med.
5
:
516
530
.
22
Min
,
C. K.
,
H. I.
Kim
,
N. Y.
Ha
,
Y.
Kim
,
E. K.
Kwon
,
N. T. H.
Yen
,
J. I.
Youn
,
Y. K.
Jeon
,
K. S.
Inn
,
M. S.
Choi
, et al
2018
.
A type I interferon and IL-10 induced by Orientia tsutsugamushi infection suppresses antigen-specific T cells and their memory responses
.
Front. Immunol.
9
:
2022
.
23
Du
,
K.
,
M.
Zhou
,
Q.
Li
,
X. Z.
Liu
.
2018
.
Chlamydia trachomatis inhibits the production of pro-inflammatory cytokines in human PBMCs through induction of IL-10
.
J. Med. Microbiol.
67
:
240
248
.
24
Costa
,
T. A.
,
S. B.
Bazan
,
C.
Feriotti
,
E. F.
Araújo
,
Ê.
Bassi
,
F. V.
Loures
,
V. L.
Calich
.
2013
.
In pulmonary paracoccidioidomycosis IL-10 deficiency leads to increased immunity and regressive infection without enhancing tissue pathology
.
PLoS Negl. Trop. Dis.
7
:
e2512
.
25
Halimi
,
A.
,
N.
Mortazavi
,
A.
Memarian
,
M.
Zahedi
,
F.
Niknejad
,
A.
Sohrabi
,
S. J.
Sarraf
.
2022
.
The relation between serum levels of interleukin 10 and interferon-gamma with oral candidiasis in type 2 diabetes mellitus patients
.
BMC Endocr. Disord.
22
:
296
.
26
Vazquez-Torres
,
A.
,
J.
Jones-Carson
,
R. D.
Wagner
,
T.
Warner
,
E.
Balish
.
1999
.
Early resistance of interleukin-10 knockout mice to acute systemic candidiasis
.
Infect. Immun.
67
:
670
674
.
27
Fecher
,
R. A.
,
M. C.
Horwath
,
D.
Friedrich
,
J.
Rupp
,
G. S.
Deepe
, Jr
.
2016
.
Inverse correlation between IL-10 and HIF-1α in macrophages infected with Histoplasma capsulatum
.
J. Immunol.
197
:
565
579
.
28
Ponce
,
N. E.
,
L. M.
Sanmarco
,
N.
Eberhardt
,
M. C.
García
,
H. W.
Rivarola
,
R. C.
Cano
,
M. P.
Aoki
.
2016
.
CD73 inhibition shifts cardiac macrophage polarization toward a microbicidal phenotype and ameliorates the outcome of experimental Chagas cardiomyopathy
.
J. Immunol.
197
:
814
823
.
29
Oliveira
,
M. M.
,
C. R.
Bonturi
,
B. R.
Salu
,
M. L. V.
Oliva
,
R. A.
Mortara
,
C. M.
Orikaza
.
2022
.
Modulation of STAT-1, STAT-3, and STAT-6 activities in THP-1 derived macrophages infected with two Trypanosoma cruzi strains
.
Front. Immunol.
13
:
1038332
.
30
Ricci-Azevedo
,
R.
,
F. C.
Mendonça-Natividade
,
A. C.
Santana
,
J. A.
Diniz
,
M. C.
Roque-Barreira
.
2021
.
Microneme proteins 1 and 4 from Toxoplasma gondii induce IL-10 production by macrophages through TLR4 endocytosis
.
Front. Immunol.
12
:
655371
.
31
Murray
,
H. W.
,
C. M.
Lu
,
S.
Mauze
,
S.
Freeman
,
A. L.
Moreira
,
G.
Kaplan
,
R. L.
Coffman
.
2002
.
Interleukin-10 (IL-10) in experimental visceral leishmaniasis and IL-10 receptor blockade as immunotherapy
.
Infect. Immun.
70
:
6284
6293
.
32
Belkaid
,
Y.
,
K. F.
Hoffmann
,
S.
Mendez
,
S.
Kamhawi
,
M. C.
Udey
,
T. A.
Wynn
,
D. L.
Sacks
.
2001
.
The role of interleukin (IL)-10 in the persistence of Leishmania major in the skin after healing and the therapeutic potential of anti-IL-10 receptor antibody for sterile cure
.
J. Exp. Med.
194
:
1497
1506
.
33
Li
,
C.
,
I.
Corraliza
,
J.
Langhorne
.
1999
.
A defect in interleukin-10 leads to enhanced malarial disease in Plasmodium chabaudi chabaudi infection in mice
.
Infect. Immun.
67
:
4435
4442
.
34
Guerrini
,
V.
,
M. L.
Gennaro
.
2019
.
Foam cells: one size doesn’t fit all
.
Trends Immunol.
40
:
1163
1179
.
35
Saka
,
H. A.
,
R.
Valdivia
.
2012
.
Emerging roles for lipid droplets in immunity and host–pathogen interactions
.
Annu. Rev. Cell Dev. Biol.
28
:
411
437
.
36
Peyron
,
P.
,
J.
Vaubourgeix
,
Y.
Poquet
,
F.
Levillain
,
C.
Botanch
,
F.
Bardou
,
M.
Daffé
,
J. F.
Emile
,
B.
Marchou
,
P. J.
Cardona
, et al
2008
.
Foamy macrophages from tuberculous patients’ granulomas constitute a nutrient-rich reservoir for M. tuberculosis persistence
.
PLoS Pathog.
4
:
e1000204
.
37
Mattos
,
K. A.
,
V. C. G.
Oliveira
,
M.
Berrêdo-Pinho
,
J. J.
Amaral
,
L. C. M.
Antunes
,
R. C. N.
Melo
,
C. C. D.
Acosta
,
D. F.
Moura
,
R.
Olmo
,
J.
Han
, et al
2014
.
Mycobacterium leprae intracellular survival relies on cholesterol accumulation in infected macrophages: a potential target for new drugs for leprosy treatment
.
Cell. Microbiol.
16
:
797
815
.
38
Johansen
,
M. D.
,
K.
de Silva
,
K. M.
Plain
,
R. J.
Whittington
,
A. C.
Purdie
.
2019
.
Mycobacterium avium subspecies paratuberculosis is able to manipulate host lipid metabolism and accumulate cholesterol within macrophages
.
Microb. Pathog.
130
:
44
53
.
39
D’Avila
,
H.
,
C. G.
Freire-de-Lima
,
N. R.
Roque
,
L.
Teixeira
,
C.
Barja-Fidalgo
,
A. R.
Silva
,
R. C.
Melo
,
G. A.
Dosreis
,
H. C.
Castro-Faria-Neto
,
P. T.
Bozza
.
2011
.
Host cell lipid bodies triggered by Trypanosoma cruzi infection and enhanced by the uptake of apoptotic cells are associated with prostaglandin E2 generation and increased parasite growth
.
J. Infect. Dis.
204
:
951
961
.
40
Nicolaou
,
G.
,
A. H.
Goodall
,
C.
Erridge
.
2012
.
Diverse bacteria promote macrophage foam cell formation via Toll-like receptor-dependent lipid body biosynthesis
.
J. Atheroscler. Thromb.
19
:
137
148
.
41
Rank
,
R. G.
,
J.
Whittimore
,
A. K.
Bowlin
,
P. B.
Wyrick
.
2011
.
In vivo ultrastructural analysis of the intimate relationship between polymorphonuclear leukocytes and the chlamydial developmental cycle
.
Infect. Immun.
79
:
3291
3301
.
42
Jacobs
,
S. H.
,
E.
Dóró
,
F. R.
Hammond
,
M. E.
Nguyen-Chi
,
G.
Lutfalla
,
G. F.
Wiegertjes
,
M.
Forlenza
.
2021
.
Occurrence of foamy macrophages during the innate response of zebrafish to trypanosome infections
.
eLife
10
:
e64520
.
43
Genoula
,
M.
,
J. L. M.
Franco
,
M.
Dupont
,
D.
Kviatcovsky
,
A.
Milillo
,
P.
Schierloh
,
E. J.
Moraña
,
S.
Poggi
,
D.
Palmero
,
D.
Mata-Espinosa
, et al
2018
.
Formation of foamy macrophages by tuberculous pleural effusions is triggered by the interleukin-10/signal transducer and activator of transcription 3 axis through ACAT upregulation
.
Front. Immunol.
9
:
459
.
44
Hwang
,
S. A.
,
Y.
Ali
,
E.
Fedotova
,
R. L.
Hunter
,
R. E.
Brown
.
2020
.
Morphoproteomics identifies the foamy alveolar macrophage as an M2 phenotype with PD-L1 expression in the early lesion of post-primary tuberculosis: implications for host immune surveillance and therapy
.
Ann. Clin. Lab. Sci.
50
:
429
438
.
45
Mattos
,
K. A.
,
H.
D’Avila
,
L. S.
Rodrigues
,
V. G.
Oliveira
,
E. N.
Sarno
,
G. C.
Atella
,
G. M.
Pereira
,
P. T.
Bozza
,
M. C.
Pessolani
.
2010
.
Lipid droplet formation in leprosy: Toll-like receptor-regulated organelles involved in eicosanoid formation and Mycobacterium leprae pathogenesis
.
J. Leukoc. Biol.
87
:
371
384
.
46
Murray
,
P. J.
,
J. E.
Allen
,
S. K.
Biswas
,
E. A.
Fisher
,
D. W.
Gilroy
,
S.
Goerdt
,
S.
Gordon
,
J. A.
Hamilton
,
L. B.
Ivashkiv
,
T.
Lawrence
, et al
2014
.
Macrophage activation and polarization: nomenclature and experimental guidelines
.
Immunity
41
:
14
20
.
47
Martinez
,
F. O.
,
S.
Gordon
.
2014
.
The M1 and M2 paradigm of macrophage activation: time for reassessment
.
F1000Prime Rep.
6
:
13
.
48
Xue
,
J.
,
S. V.
Schmidt
,
J.
Sander
,
A.
Draffehn
,
W.
Krebs
,
I.
Quester
,
D.
De Nardo
,
T. D.
Gohel
,
M.
Emde
,
L.
Schmidleithner
, et al
2014
.
Transcriptome-based network analysis reveals a spectrum model of human macrophage activation
.
Immunity
40
:
274
288
.
49
Bhattacharya
,
S.
,
A.
Aggarwal
.
2019
.
M2 macrophages and their role in rheumatic diseases
.
Rheumatol. Int.
39
:
769
780
.
50
Orecchioni
,
M.
,
Y.
Ghosheh
,
A. B.
Pramod
,
K.
Ley
.
2019
.
Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS) vs. alternatively activated macrophages
.
Front. Immunol.
10
:
1084
.
51
Lee-Rueckert
,
M.
,
J.
Lappalainen
,
P. T.
Kovanen
,
J. C.
Escola-Gil
.
2022
.
Lipid-laden macrophages and inflammation in atherosclerosis and cancer: an integrative view
.
Front. Cardiovasc. Med.
9
:
777822
.
52
Mahajan
,
S.
,
H. K.
Dkhar
,
V.
Chandra
,
S.
Dave
,
R.
Nanduri
,
A. K.
Janmeja
,
J. N.
Agrewala
,
P.
Gupta
.
2012
.
Mycobacterium tuberculosis modulates macrophage lipid-sensing nuclear receptors PPARγ and TR4 for survival
.
J. Immunol.
188
:
5593
5603
.
53
Oh
,
J.
,
A. E.
Riek
,
S.
Weng
,
M.
Petty
,
D.
Kim
,
M.
Colonna
,
M.
Cella
,
C.
Bernal-Mizrachi
.
2012
.
Endoplasmic reticulum stress controls M2 macrophage differentiation and foam cell formation
.
J. Biol. Chem.
287
:
11629
11641
.
54
Melo
,
R. C. N.
,
A. M.
Dvorak
.
2012
.
Lipid body–phagosome interaction in macrophages during infectious diseases: host defense or pathogen survival strategy?
PLoS Pathog.
8
:
e1002729
.
55
Pandey
,
A. K.
,
C. M.
Sassetti
.
2008
.
Mycobacterial persistence requires the utilization of host cholesterol
.
Proc. Natl. Acad. Sci. U. S. A.
105
:
4376
4380
.
56
Ouimet
,
M.
,
S.
Koster
,
E.
Sakowski
,
B.
Ramkhelawon
,
C.
van Solingen
,
S.
Oldebeken
,
D.
Karunakaran
,
C.
Portal-Celhay
,
F. J.
Sheedy
,
T. D.
Ray
, et al
2016
.
Mycobacterium tuberculosis induces the miR-33 locus to reprogram autophagy and host lipid metabolism
.
Nat. Immunol.
17
:
677
686
.
57
D’Avila
,
H.
,
R. C. N.
Melo
,
G. G.
Parreira
,
E.
Werneck-Barroso
,
H. C.
Castro-Faria-Neto
,
P. T.
Bozza
.
2006
.
Mycobacterium bovis Bacillus Calmette-Guérin induces TLR2-mediated formation of lipid bodies: intracellular domains for eicosanoid synthesis in vivo
.
J. Immunol.
176
:
3087
3097
.
58
Mattos
,
K. A.
,
V. G. C.
Oliveira
,
H.
D’Avila
,
L. S.
Rodrigues
,
R. O.
Pinheiro
,
E. N.
Sarno
,
M. C. V.
Pessolani
,
P. T.
Bozza
.
2011
.
TLR6-driven lipid droplets in Mycobacterium leprae–infected schwann cells: immunoinflammatory platforms associated with bacterial persistence
.
J. Immunol.
187
:
2548
2558
.
59
Rosas-Taraco
,
A. G.
,
A. R.
Perez-Liñan
,
P.
Bocanegra-Ibarias
,
L. I.
Perez-Rivera
,
M. C.
Salinas-Carmona
.
2012
.
Nocardia brasiliensis induces an immunosuppressive microenvironment that favors chronic infection in BALB/c mice
.
Infect. Immun.
80
:
2493
2499
.
60
Meester
,
I.
,
A. G.
Rosas-Taraco
,
M. C.
Salinas-Carmona
.
2014
.
Nocardia brasiliensis induces formation of foamy macrophages and dendritic cells in vitro and in vivo
.
PLoS One
9
:
e100064
.
61
Tumurkhuu
,
G.
,
J.
Dagvadorj
,
R. A.
Porritt
,
T. R.
Crother
,
K.
Shimada
,
E. J.
Tarling
,
E.
Erbay
,
M.
Arditi
,
S.
Chen
.
2018
.
Chlamydia pneumoniae hijacks a host autoregulatory IL-1β loop to drive foam cell formation and accelerate atherosclerosis
.
Cell Metab.
28
:
432
448.e4
.
62
Di Pietro
,
M.
,
F.
De Santis
,
G.
Schiavoni
,
S.
Filardo
,
R.
Sessa
.
2013
.
Resveratrol in Chlamydia pneumoniae–induced foam cell formation and interleukin-17A synthesis
.
J. Biol. Regul. Homeost. Agents
27
:
509
518
.
63
Zeidan
,
B. S.
, Jr
.,
J. K.
Patel
,
A.
Kirk
,
A.
Gonzalez
,
I.
Khan
.
2021
.
Cryptogenic organizing pneumonia secondary to Mycoplasma pneumoniae infection: a case report
.
Cureus
13
:
e20623
.
64
Eberhardt
,
N.
,
M. G.
Noval
,
R.
Kaur
,
L.
Amadori
,
M.
Gildea
,
S.
Sajja
,
D.
Das
,
B.
Cilhoroz
,
O.
Stewart
,
D. M.
Fernandez
, et al
2023
.
SARS-CoV-2 infection triggers pro-atherogenic inflammatory responses in human coronary vessels
.
Nat. Cardiovasc. Res.
2
:
899
916
.
65
Agarwal
,
P.
,
T. W.
Combes
,
F.
Shojaee-Moradie
,
B.
Fielding
,
S.
Gordon
,
V.
Mizrahi
,
F. O.
Martinez
.
2020
.
Foam cells control Mycobacterium tuberculosis infection
.
Front. Microbiol.
11
:
1394
.
66
Knight
,
M.
,
J.
Braverman
,
K.
Asfaha
,
K.
Gronert
,
S.
Stanley
.
2018
.
Lipid droplet formation in Mycobacterium tuberculosis infected macrophages requires IFN-γ/HIF-1α signalling and supports host defense
.
PLoS Pathog.
14
:
e1006874
.
67
Almeida
,
P. E.
,
A. R.
Silva
,
C. M.
Maya-Monteiro
,
D.
Töröcsik
,
H.
D’Avila
,
B.
Dezsö
,
K. G.
Magalhães
,
H. C.
Castro-Faria-Neto
,
L.
Nagy
,
P. T.
Bozza
.
2009
.
Mycobacterium bovis Bacillus Calmette-Guérin infection induces TLR2-dependent peroxisome proliferator-activated receptor γ expression and activation: functions in inflammation, lipid metabolism, and pathogenesis
.
J. Immunol.
183
:
1337
1345
.
68
Wright
,
K.
,
R.
Mizzi
,
K. M.
Plain
,
A. C.
Purdie
,
K.
de Silva
.
2022
.
Mycobacterium avium subsp. paratuberculosis exploits miRNA expression to modulate lipid metabolism and macrophage polarisation pathways during infection
.
Sci. Rep.
12
:
9681
.
69
Mei
,
C. L.
,
P.
He
,
B.
Cheng
,
W.
Liu
,
Y. F.
Wang
,
J. J.
Wan
.
2009
.
Chlamydia pneumoniae induces macrophage-derived foam cell formation via PPARα and PPARγ-dependent pathways
.
Cell. Biol. Int.
33
:
301
308
.
70
Cao
,
F.
,
A.
Castrillo
,
P.
Tontonoz
,
F.
Re
,
G. I.
Byrne
.
2007
.
Chlamydia pneumoniae–induced macrophage foam cell formation is mediated by Toll-like receptor 2
.
Infect. Immun.
75
:
753
759
.
71
Montoya
,
D.
,
D.
Cruz
,
R. M.
Teles
,
D. J.
Lee
,
M. T.
Ochoa
,
S. R.
Krutzik
,
R.
Chun
,
M.
Schenk
,
X.
Zhang
,
B. G.
Ferguson
, et al
2009
.
Divergence of macrophage phagocytic and antimicrobial programs in leprosy
.
Cell Host Microbe
6
:
343
353
.
72
Fernandes
,
L. R.
,
A. C.
Ribeiro
,
M.
Segatto
,
L. F.
Santos
,
J.
Amaral
,
L. R.
Portugal
,
J. I.
Leite
.
2013
.
Leishmania major self-limited infection increases blood cholesterol and promotes atherosclerosis development
.
Cholesterol
2013
:
754580
.
73
Prandota
,
J.
2019
.
Unexplained liver damage, cryptogenic liver cirrhosis, and steatohepatitis may be caused by latent chronic toxoplasmosis
.
J. Gastroenterol. Hepatol. Res.
8
:
2824
2840
.
74
Argmann
,
C. A.
,
C. G.
Sawyez
,
C. J.
McNeil
,
R. A.
Hegele
,
M. W.
Huff
.
2003
.
Activation of peroxisome proliferator-activated receptor gamma and retinoid X receptor results in net depletion of cellular cholesteryl esters in macrophages exposed to oxidized lipoproteins
.
Arterioscler. Thromb. Vasc. Biol.
23
:
475
482
.
75
Xie
,
S.
,
Y. F.
Lee
,
E.
Kim
,
L. M.
Chen
,
J.
Ni
,
L. Y.
Fang
,
S.
Liu
,
S. J.
Lin
,
J. I.
Abe
,
B.
Berk
, et al
2009
.
TR4 nuclear receptor functions as a fatty acid sensor to modulate CD36 expression and foam cell formation
.
Proc. Natl. Acad. Sci. U. S. A.
106
:
13353
13358
.
76
Venkateswaran
,
A.
,
B. A.
Laffitte
,
S. B.
Joseph
,
P. A.
Mak
,
D. C.
Wilpitz
,
P. A.
Edwards
,
P.
Tontonoz
.
2000
.
Control of cellular cholesterol efflux by the nuclear oxysterol receptor LXRα
.
Proc. Natl. Acad. Sci. U. S. A.
97
:
12097
12102
.
77
Getz
,
G. S.
,
C. A.
Reardon
.
2014
.
The mutual interplay of lipid metabolism and the cells of the immune system in relation to atherosclerosis
.
Clin. Lipidol.
9
:
657
671
.
78
Yang
,
L.
,
J. B.
Yang
,
J.
Chen
,
G. Y.
Yu
,
P.
Zhou
,
L.
Lei
,
Z. Z.
Wang
,
C.
Cy Chang
,
X. Y.
Yang
,
T. Y.
Chang
,
B. L.
Li
.
2004
.
Enhancement of human ACAT1 gene expression to promote the macrophage-derived foam cell formation by dexamethasone
.
Cell Res.
14
:
315
323
.
79
Scull
,
C. M.
,
I.
Tabas
.
2011
.
Mechanisms of ER stress-induced apoptosis in atherosclerosis
.
Arterioscler. Thromb. Vasc. Biol.
31
:
2792
2797
.
80
Lo Sasso
,
G.
,
W. K.
Schlage
,
S.
Boué
,
E.
Veljkovic
,
M. C.
Peitsch
,
J.
Hoeng
.
2016
.
The Apoe−/− mouse model: a suitable model to study cardiovascular and respiratory diseases in the context of cigarette smoke exposure and harm reduction
.
J. Transl. Med.
14
:
146
.
81
Caligiuri
,
G.
,
M.
Rudling
,
V.
Ollivier
,
M. P.
Jacob
,
J. B.
Michel
,
G. K.
Hansson
,
A.
Nicoletti
.
2003
.
Interleukin-10 deficiency increases atherosclerosis, thrombosis, and low-density lipoproteins in apolipoprotein E knockout mice
.
Mol. Med.
9
:
10
17
.
82
Kang
,
J. G.
,
H. J.
Sung
,
M. J.
Amar
,
M.
Pryor
,
A. T.
Remaley
,
M. D.
Allen
,
A. C.
Noguchi
,
D. A.
Springer
,
J.
Kwon
,
J.
Chen
, et al
2016
.
Low ambient oxygen prevents atherosclerosis
.
J. Mol. Med. (Berl.)
94
:
277
286
.
83
Martens
,
G. W.
,
M. C.
Arikan
,
J.
Lee
,
F.
Ren
,
T.
Vallerskog
,
H.
Kornfeld
.
2008
.
Hypercholesterolemia impairs immunity to tuberculosis
.
Infect. Immun.
76
:
3464
3472
.
84
Gudgeon
,
J.
,
J. L.
Marín-Rubio
,
M.
Trost
.
2022
.
The role of macrophage scavenger receptor 1 (MSR1) in inflammatory disorders and cancer
.
Front. Immunol.
13
:
1012002
.
85
Vogel
,
A.
,
J. S.
Brunner
,
A.
Hajto
,
O.
Sharif
,
G.
Schabbauer
.
2022
.
Lipid scavenging macrophages and inflammation
.
Biochim. Biophys. Acta Mol. Cell Biol. Lipids
1867
:
159066
.
86
Redford
,
P. S.
,
P. J.
Murray
,
A.
O’Garra
.
2011
.
The role of IL-10 in immune regulation during M. tuberculosis infection
.
Mucosal Immunol.
4
:
261
270
.
87
Saxena
,
A.
,
S.
Khosraviani
,
S.
Noel
,
D.
Mohan
,
T.
Donner
,
A. R.
Hamad
.
2015
.
Interleukin-10 paradox: a potent immunoregulatory cytokine that has been difficult to harness for immunotherapy
.
Cytokine
74
:
27
34
.
88
Brooks
,
D. G.
,
M. J.
Trifilo
,
K. H.
Edelmann
,
L.
Teyton
,
D. B.
McGavern
,
M. B.
Oldstone
.
2006
.
Interleukin-10 determines viral clearance or persistence in vivo
.
Nat. Med.
12
:
1301
1309
.
89
Javadifar
,
A.
,
S.
Rastgoo
,
M.
Banach
,
T.
Jamialahmadi
,
T. P.
Johnston
,
A.
Sahebkar
.
2021
.
Foam cells as therapeutic targets in atherosclerosis with a focus on the regulatory roles of non-coding RNAs
.
Int. J. Mol. Sci.
22
:
2529
.
90
Nair
,
S.
,
P. A.
Ramaswamy
,
S.
Ghosh
,
D. C.
Joshi
,
N.
Pathak
,
I.
Siddiqui
,
P.
Sharma
,
S. E.
Hasnain
,
S. C.
Mande
,
S.
Mukhopadhyay
.
2009
.
The PPE18 of Mycobacterium tuberculosis interacts with TLR2 and activates IL-10 induction in macrophage
.
J. Immunol.
183
:
6269
6281
.
91
Upadhyay
,
R.
,
A.
Sanchez-Hidalgo
,
C. J.
Wilusz
,
A. J.
Lenaerts
,
J.
Arab
,
J.
Yeh
,
K.
Stefanisko
,
N. I.
Tarasova
,
M.
Gonzalez-Juarrero
.
2018
.
Host directed therapy for chronic tuberculosis via intrapulmonary delivery of aerosolized peptide inhibitors targeting the IL-10-STAT3 pathway
.
Sci. Rep.
8
:
16610
.
92
Dwivedi
,
V.
,
S.
Gautam
,
C. A.
Headley
,
T.
Piergallini
,
J. B.
Torrelles
,
J.
Turner
.
2022
.
IL-10 receptor blockade delivered simultaneously with Bacillus Calmette-Guérin vaccination sustains long-term protection against Mycobacterium tuberculosis infection in mice
.
J. Immunol.
208
:
1406
1416
.
93
Beamer
,
G. L.
,
D. K.
Flaherty
,
B. D.
Assogba
,
P.
Stromberg
,
M.
Gonzalez-Juarrero
,
R.
de Waal Malefyt
,
B.
Vesosky
,
J.
Turner
.
2008
.
Interleukin-10 promotes Mycobacterium tuberculosis disease progression in CBA/J mice
.
J. Immunol.
181
:
5545
5550
.
94
Guerra-De-Blas
,
P. D. C.
,
P.
Torres-González
,
M.
Bobadilla-Del-Valle
,
I.
Sada-Ovalle
,
A.
Ponce-De-León-Garduño
,
J.
Sifuentes-Osornio
.
2018
.
Potential effect of statins on mycobacterium tuberculosis infection
.
J. Immunol. Res.
2018
:
7617023
.
95
Dutta
,
N. K.
,
N.
Bruiners
,
M. L.
Pinn
,
M. D.
Zimmerman
,
B.
Prideaux
,
V.
Dartois
,
M. L.
Gennaro
,
P. C.
Karakousis
.
2016
.
Statin adjunctive therapy shortens the duration of TB treatment in mice
.
J. Antimicrob. Chemother.
71
:
1570
1577
.
96
García-Fernández-Bravo
,
I.
,
A.
Torres-Do-Rego
,
A.
López-Farré
,
F.
Galeano-Valle
,
P.
Demelo-Rodriguez
,
L. A.
Alvarez-Sala-Walther
.
2022
.
Undertreatment or overtreatment with statins: where are we?
Front. Cardiovasc. Med.
9
:
808712
.
97
Oliver
,
W. R.
,
J. L.
Shenk
,
M. R.
Snaith
,
C. S.
Russell
,
K. D.
Plunket
,
N. L.
Bodkin
,
M. C.
Lewis
,
D. A.
Winegar
,
M. L.
Sznaidman
,
M. H.
Lambert
, et al
2001
.
A selective peroxisome proliferator-activated receptor δ agonist promotes reverse cholesterol transport
.
Proc. Natl. Acad. Sci. U. S. A.
98
:
5306
5311
.
98
Claudel
,
T.
,
M. D.
Leibowitz
,
C.
Fiévet
,
A.
Tailleux
,
B.
Wagner
,
J. J.
Repa
,
G.
Torpier
,
J. M.
Lobaccaro
,
J. R.
Paterniti
,
D. J.
Mangelsdorf
, et al
2001
.
Reduction of atherosclerosis in apolipoprotein E knockout mice by activation of the retinoid X receptor
.
Proc. Natl. Acad. Sci. U. S. A.
98
:
2610
2615
.
99
Joseph
,
S. B.
,
E.
McKilligin
,
L.
Pei
,
M. A.
Watson
,
A. R.
Collins
,
B. A.
Laffitte
,
M.
Chen
,
G.
Noh
,
J.
Goodman
,
G. N.
Hagger
, et al
2002
.
Synthetic LXR ligand inhibits the development of atherosclerosis in mice
.
Proc. Natl. Acad. Sci. U. S. A.
99
:
7604
7609
.
100
Wallis
,
R. S.
,
A.
O’Garra
,
A.
Sher
,
A.
Wack
.
2023
.
Host-directed immunotherapy of viral and bacterial infections: past, present and future
.
Nat. Rev. Immunol.
23
:
121
133
.
101
Nissen
,
S. E.
,
K.
Wolski
.
2007
.
Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes
.
N. Engl. J. Med.
356
:
2457
2471
.
102
Fang
,
F.
,
C.
Xiao
,
C.
Li
,
X.
Liu
,
S.
Li
.
2023
.
Tuning macrophages for atherosclerosis treatment
.
Regen. Biomater.
10
:
rbac103
.