Next Article in Journal
Translation Efficiency and Degradation of ER-Associated mRNAs Modulated by ER-Anchored poly(A)-Specific Ribonuclease (PARN)
Next Article in Special Issue
Living with Yourself: Innate Lymphoid Cell Immunometabolism
Previous Article in Journal
STIM Protein-NMDA2 Receptor Interaction Decreases NMDA-Dependent Calcium Levels in Cortical Neurons
Previous Article in Special Issue
Metabolic Reprogramming in Mitochondria of Myeloid Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathogens MenTORing Macrophages and Dendritic Cells: Manipulation of mTOR and Cellular Metabolism to Promote Immune Escape

Department of Parasitology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
*
Author to whom correspondence should be addressed.
Cells 2020, 9(1), 161; https://doi.org/10.3390/cells9010161
Submission received: 16 December 2019 / Revised: 1 January 2020 / Accepted: 7 January 2020 / Published: 9 January 2020
(This article belongs to the Special Issue Metabolic Reprogramming in Innate Immune Cell Fate and Function)

Abstract

:
Myeloid cells, including macrophages and dendritic cells, represent an important first line of defense against infections. Upon recognition of pathogens, these cells undergo a metabolic reprogramming that supports their activation and ability to respond to the invading pathogens. An important metabolic regulator of these cells is mammalian target of rapamycin (mTOR). During infection, pathogens use host metabolic pathways to scavenge host nutrients, as well as target metabolic pathways for subversion of the host immune response that together facilitate pathogen survival. Given the pivotal role of mTOR in controlling metabolism and DC and macrophage function, pathogens have evolved strategies to target this pathway to manipulate these cells. This review seeks to discuss the most recent insights into how pathogens target DC and macrophage metabolism to subvert potential deleterious immune responses against them, by focusing on the metabolic pathways that are known to regulate and to be regulated by mTOR signaling including amino acid, lipid and carbohydrate metabolism, and autophagy.

1. Introduction

Immune cells from the myeloid lineage, such as dendritic cells (DCs) and macrophages, form an important first line of defense against invading pathogens. DCs have superior antigen presenting capacities and are crucial for the initiation of adaptive immune responses by controlling priming and differentiation of T cell responses. Macrophages on the other hand, are tissue resident cells that excel in clearing of invading pathogens and infected cells (pro-inflammatory or M1 macrophages) and restoring tissue homeostasis (anti-inflammatory or M2 macrophages). Macrophages as well as DCs express a wide range of pattern recognition receptors (PPRs) and other sensors that allow them to directly or indirectly detect the presence of unwanted intruders. This will initiate a cellular activation program that enables them to directly fight the infectious agent, recruit other immune cells, or initiate adaptive immune responses. Recent work has revealed that DC and macrophage activation status is intrinsically linked to metabolic reprogramming [1,2]. In both DC and macrophages, sensing of pathogens, in particular bacteria, is accompanied by a shift toward glycolytic metabolism, the use of the pentose phosphate pathway (PPP), increased translation, as well and altered activity of the TCA cycle. These metabolic changes have been shown to be essential not only for supporting the increased bioenergetic and biosynthetic demand of DC and macrophage activation, but also for generation of metabolites that affect the epigenetic and transcriptional landscapes of these cells that are integral to their functional properties and fate [2,3,4].
Despite the fact that multicellular organisms have developed elegant immunological strategies to combat and counteract infections, pathogens often still manage to successfully infect their hosts. This can in part be attributed to their ability to evade, modulate, and/or suppress these same immune responses [5,6,7]. In this respect, apart from host immune cells using their metabolism to initiate host defense responses, also pathogens are able to exploit these host metabolic pathways for their own benefit. On the one hand, they tap into these pathways to acquire nutrients and macromolecules that directly promote their own survival and replication [8,9,10,11,12,13,14,15]. On the other hand, it is now becoming clear that pathogens also manipulate metabolic pathways of host innate immune cells, to modulate, dampen, or evade the immune responses to further enhance their chances of survival.
A central regulator of cellular metabolism of DC and macrophages is the nutrient sensor mammalian target of rapamycin (mTOR). mTOR consists of two complexes; mTORC1 and mTORC2. mTORC2 regulates cytoskeletal dynamics and activates Akt, a kinase that regulates mTORC1 activity. mTORC1 itself acts as a metabolic rheostat that controls a wide range of cellular processes according to nutrient and energy availability [16]. For example, mTORC1 promotes anabolic processes such as protein translation and represses autophagy under high nutrient availability. Moreover, mTORC1 regulates several metabolic functions of the cell, such as lipid, amino acid, and carbohydrate metabolism in response to a range of stimuli, including growth factors and immune cell activation cues such pathogens and cytokines [17]. This puts mTORC1 as central node in regulation of cellular homeostasis, immune cell function, and innate antimicrobial responses. Given the central role for mTOR in regulating cellular metabolism, it may come as no surprise that pathogens have evolved mechanisms that specifically manipulate mTOR signaling and associated metabolic pathways in order to evade host innate immune responses. Notably, this modulation of mTOR is not only limited to innate immune cells. It is well-known that modulation of mTOR signaling also has major effects on the adaptive immunity such as on the balance between effector versus memory T cells not only in the context of infection, but also autoimmunity and cancer, which is covered by excellent recent reviews [18,19,20]. In the current review we focus on the innate immune response, by seeking to provide an overview and to discuss the most recent insights into how pathogens target DC and macrophage metabolism to manipulate the immune response. Specifically, we will focus on direct modulation of mTOR signaling itself as well as the metabolic pathways that are known to regulate and to be regulated by the mTOR pathway including amino acid, lipid and carbohydrate metabolism, and autophagy.

2. Direct Targeting of mTOR Complexes by Pathogens

Pathogens have evolved means to directly modulate mTOR signaling to affect DC and macrophage metabolism and function. An elegant example comes from Brugia malayi, an extracellular helminth parasite that causes lymphatic filariasis [21]. Live B. malayi microfilariae affect human monocyte-derived DCs by inducing cell death, impairing their ability to make IL-12 and IL-10 and reducing their CD4+ T cell-activating capacity [22]. Consistent with an important role for mTORC1 in supporting IL-12 and IL-10 production, microfilariae of B. malayi were found to inhibit mTORC1, by means of secretion of a rapamycin-homolog [22]. Likewise, Leishmania major, a parasite that grows in macrophages (in this study the B10R bone marrow-derived macrophage cell line), can cleave mTOR via the protease GP63 which leads to inactivation of mTORC1 and the inhibition of host translation. This inhibition of host translation decreased type I IFN production and inducible nitric oxide synthase (iNOS) expression and consequently enhances the survival of the parasite in a BALB/c mouse model [23].
Instead of inhibiting or degrading mTOR, mTOR can also be relocated to modulate its function. Human cytomegalovirus (HCMV) relocalizes mTOR to the site of viral replication during infection of human foreskin fibroblasts (HFFs)/U373-MG cells (a human glioblastoma-astrocytoma cell line). In the presence of amino acids, mTOR activity correlates with a perinuclear localization, while during amino acid depletion mTOR disperses and loses its activity. HCMV induces perinuclear relocalization of mTOR, mediated through dynein, to maintain mTORC1 activity even in an amino acid scarce environment [24,25]. HCMV is able to infect both macrophages and DCs, but the metabolic and immunological consequences for these cells of HCMV-driven constitutive mTOR activation remain to be determined [26,27].
Also mTORC2 is directly targeted by pathogens. Poxviruses, including vaccinia virus (VacV), were found to target both mTORC1 and mTORC2 in Phorbol 12-myristate 13-acetate (PMA) differentiated Tohoku Hospital Pediatrics-1 (THP-1) cells (a human monocyte cell line) by sequestering raptor (part of mTORC1) and rictor (part of mTORC2) through the structural protein F17, while retaining the phosphorylation of S6K by mTOR. This results in relocation of mTOR to the Golgi where it inhibits cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) activation and hence the induction of interferon-stimulated genes (IRGs). This enables these viruses to block cytosolic sensing, while retaining mTOR-dependent activation of translation [28,29]. mTORC2 signaling is also targeted by HIV-1 to manipulate macrophage migration. The accessory NEF protein of HIV-1 interacts with POTE ankyrin domain family E (POTEE) that enables NEF to directly bind and activate mTORC2. This activation in turn activates Akt and PKC-α leading to the upregulation of M2 markers and increased migration and invasion of HIV-1 infected PMA differentiated THP-1 cells, that may thereby serve as carriers of HIV-1 across mucosal barriers to help disseminate HIV-1 from primary sites of infection [30]. Overall, both intra- and extracellular pathogens are able to subvert immune responses of DCs and macrophages by directly targeting mTORC1 and/or mTORC2 (Figure 1).

3. Targeting of Amino Acid Metabolism by Pathogens

mTOR is a key sensor of amino acids and its activation is dependent on sufficient availability of certain amino acids. Several studies have shown that in innate immune cells, such as macrophages, amino acid availability and their metabolism are intricately linked to cellular activation and polarization in an mTOR signaling-dependent manner [31,32]. It is well-known that amino acid metabolism can be subject to manipulation by pathogens to modulate the immune system for their own survival [33,34]. In this section we focus on the role of a number of key amino acids in innate immune responses against pathogens, how pathogens have devised ways to target amino acid-driven modulation of innate immunity, and how mTOR plays into this (Table 1, Figure 2A).

3.1. Tryptophan

Tryptophan is an essential amino acid that is catabolized by three different enzymes: tryptophan 2,3-dioxygenase (TDO), indoleamine 2,3-dioxygenase (IDO), and indoleamine 2,3-dioxygenase-2 (IDO2). IDO, in particular, is associated with infection and induced by IFN type I/II production, prostaglandins as well as microbial components [35,36]. Because pathogens are either tryptophan autotrophs (fully depending on host tryptophan), such as T. gondii and the mouse chlamydia strain, or at least benefit from host tryptophan for their development or growth, the host immune response upregulates IDO, decreasing its availability to pathogens [37,38,39]. Additionally, the metabolites arising from tryptophan degradation have antimicrobial effects themselves, thereby aiding in host-defense [40]. Notwithstanding, pathogens have evolved strategies to evade this host defense mechanism. This is illustrated by M. tuberculosis, Francisella tularensis, and Chlamydia trachomatis that are able to synthesize tryptophan de novo or from precursors, to become less vulnerable to this host defense mechanism [9,41].
On the other hand, pathogens might also benefit from IDO expression as long term expression of this enzyme can result in immune suppression. It has been described that the induction of IDO by innate immune cells decreases the pool of effector T cells, while increasing the amount of regulatory T cells [42,43]. In addition, IDO expression results in an anti-inflammatory profile in macrophages and DCs [34,44,45,46]. Given the immune suppressive effect of IDO, some pathogens actively stimulate its expression. HCMV and HPV induce the expression of IDO to dampen pro-inflammatory immune responses. HCMV actively manipulates human monocyte-derived DCs to express IDO by expressing a IL-10 homolog, cmvIL-10, encoded by the gene UL111A [47,48]. Likewise, the HPV16 oncoprotein E7 expressed under control of the keratin 14 promotor in a murine skin graft model induces IDO expression in Langerin negative dermal dendritic cells which mediates immune regulation [49].
Amino acid starvation, in this case tryptophan depletion, leads to activation of a stress response mediated by general control non-repressible 2 (GCN2), a kinase activated by amino acid deprivation that reduces global protein synthesis, and concomitantly to inhibition of the mTOR pathway [50,51,52,53]. This integrated stress response has already been described to regulate T-cell suppression in response to tryptophan depletion and to affect primary murine macrophage and DC cytokine production [34,43,54]. It is worth noting that, apart from tryptophan itself, breakdown products of tryptophan, for example kynurenine and serotonin, are also possible activators of mTORC1 [55,56]. Additionally, kynurenines have been implicated in activating the aryl hydrocarbon receptor (AhR) in T cells which contributes to the generation of regulatory T cells, but also to the capacity of DCs (in this case GM-CSF differentiated bone marrow-derived DCs (GM-DCs)) to generate regulatory T cells [57,58]. Other tryptophan metabolites (e.g., serotonin, kynurenic acid, NAD+, indoles) have been implicated in inflammation as well and could therefore potentially be important during infection [59]. Thus, both the depletion of tryptophan and the generation of its breakdown products can affect mTOR activation and/or contribute to the manipulation of the immune response. While the initiation of the stress response may contribute to the aforementioned anti-inflammatory profiles, GCN2 activation/mTOR inhibition may also be host-protective by activating autophagy which in macrophages can contribute to the killing of intracellular pathogens and to enhance antigen presentation to CD4+ and CD8+ T cells by DCs [53,60,61].
Taken together, tryptophan metabolism has an antimicrobial effect in acute infections, whereas it leads to immunosuppression in chronic infections, in part through the GCN2 and mTOR pathways, which impacts innate immune function. A better evaluation of this balance between immune regulation and host-protection will be required in different infectious settings to fully decipher the role of IDO in host-pathogen interplay.

3.2. L-Arginine

L-arginine is a substrate of both nitric oxide synthase (NOS) and arginase [62,63]. The main product of NOS is NO, which is an essential microbicidal metabolite of the innate immune system [64]. The inducible form of NOS (iNOS) is not constitutively expressed, but highly induced upon macrophage activation by bacterial products, such as lipopolysaccharide (LPS) and lipoteichoic acid (LTA), and type I cytokines, including IFNγ. Arginase on the other hand, is primarily induced by type 2 cytokines such as IL-4 and IL-13. Arginase expression metabolizes L-arginine to yield L-ornithine and urea that can be used for the production of polyamines (Figure 3) and thus competes with iNOS for L-arginine [65,66]. The balance in iNOS versus arginase activity and thus the production of NO and polyamines proves to be a target for pathogens to modulate the immune response.
Pathogens modulate this balance to favor arginase over iNOS expression for two main reasons: to reduce NO production and to generate polyamines which can be used for the growth of the pathogen and immune modulation. Multiple pathogens, including Trypanosoma sps, Leishmania spp, Toxoplasma gondii, Salmonella typhimurium, Helicobacter pylori, Streptococcus pneumonia, Candida albicans, M. tuberculosis, and Schistosoma mansoni induce host arginases in mouse macrophages [65,66,67,68,69,70]. Moreover, Leishmania spp, Plasmodium spp, C. albicans, S. mansoni, and H. pylori (RocF) additionally express their own arginase enzyme to favor polyamine over NO production [65,71,72,73,74,75,76], suggesting that this is a common strategy for pathogens to escape from the deleterious effects of NO. In addition, polyamine synthesis itself has been shown to enhance anti-inflammatory alternative activation (AA) program in macrophages, because of their ability to favor mitochondrial respiration which underpins this type of macrophages polarization [77]. While promotion of such an AA phenotype may prevent direct clearance of pathogens, AA macrophages are important players in tissue repair, thereby serving an important host protective role against tissue damage inflicted by these pathogens.
Not only bacteria and parasites, but also viruses exploit host L-arginine metabolism. Viruses mainly use L-arginine metabolism to enhance translation and increase replication efficiency. Because of the strong positive charge of polyamines, they easily bind to the viral DNA/RNA and are able to accelerate viral protein translation [65,78]. For this reason, multiple viruses manipulate the polyamine pathway by encoding (parts of) the polyamine synthesis pathway and some viruses, at least HCV, induce arginase 1 expression. In the case of HCV, the induction of arginase 1 modulates the immune response: HCV induces arginase 1 expression in CD33+ PBMCs which decreased production of IFNγ by co-cultured human NK cells. This suppression of IFNγ production in the NK cells was due to the inhibition of mTOR through the depletion of L-arginine by the arginase [79]. Interestingly, L-arginine is not only depleted by pathogen-driven host arginase expression, but also by arginine deiminase (ADI). This enzyme is expressed by the parasite Giardia duodenalis and L-arginine depletion by this enzyme has been shown to modulate cytokine expression and the phenotype of human monocyte-derived DCs probably via mTORC1 inhibition [80].
L-arginine is one of the amino acids known to stimulate mTOR activity, and its absence reduces mTOR activity and affects innate immune cells, as illustrated by HCV and G. duodenalis [81,82,83]. This mTOR inhibition resembles the stress response described for tryptophan. Indeed, L-arginine withdrawal from T-cells results in anti-inflammatory responses via GNC2 activation and mTOR inhibition [34,84,85]. From what has been observed for HCV and G. duodenalis, it is possible that this stress response also regulates innate immune cell function. Additionally, mTOR has been shown to increase iNOS translation in RAW264.7, peritoneal macrophages, and GM-DCs [86,87]. Therefore, it is conceivable that mTOR inhibition because of pathogen-driven depletion of L-arginine, leads to a reduced iNOS expression which further limits NO production. In conclusion, arginase expression is induced by a variety of pathogens in order to increase polyamine production and suppress NO production, but murine macrophages and mouse models are overrepresented in these studies and the role of mTOR has not yet been fully eluted.

3.3. Asparagine

Certain bacteria express an asparaginase, including S. typhimurium and H. pylori [88,89,90,91,92]. In the case of S. typhimurium, asparaginase expression leads to the inhibition of T cell activation and has been linked to the inhibition of mTOR signaling and autophagy [90]. There are also indications that pathogen-derived asparaginases or modulation of asparagine metabolism may affect macrophage function. For instance, Erwinia asparaginase, a bacterial derived oncolytic drug has been described to block pro-inflammatory macrophage responses (inhibition of phagocytosis, pro-inflammatory cytokine production, and MHC-II expression in ANA-1 and RAW264.7 murine macrophages). This effect appears to be dependent on inhibition of autophagy as a consequence of sustained mTOR signaling, which may be related to the stimulatory effects of aspartate on mTOR activity [93,94]. Asparaginases are also known to have glutaminase activity, but to what extent this additional enzymatic activity affects macrophage biology remains to be addressed.
In summary, active manipulation of amino acid metabolism is a strategy widely used by different classes of pathogens to modulate and suppress host innate immune responses. In addition to the amino acids described here, others, such as glutamine and glycine, also play important roles in immune responses [95,96]. Whether pathogens target their metabolism too, to modulate immune responses is still on open question. As illustrated above, active depletion of amino acids by pathogens appears to be a common strategy to compromise innate immune function by reducing mTOR activity. However, it is worth noting that some pathogens such as HCMV and S. typhimurium, that rely on host cell mTOR signaling for effective replication, are able to keep mTOR and/or its downstream targets activated even under amino acid-deprived conditions [24,97,98,99,100]. This illustrates that pathogens have developed multiple ways to calibrate host immune cell amino acid metabolism and mTOR signaling that together allow for immune evasion without compromising their own replication.

4. Targeting of Lipid Metabolism by Pathogens

mTOR signaling is an important driver of fatty acid (FA) and lipid biosynthesis. Lipids are the major structural component of cellular membranes as well as lipid droplets (LDs). In addition, FAs, particularly when they are polyunsaturated (PUFA), can serve as second messengers and signaling molecules in immune responses. Lipid metabolism in host cells can be modulated and hijacked by pathogens to modify immune responses, which is discussed in this section (Table 1, Figure 2B) [101,102].

4.1. Lipid Droplets

LDs are lipid-rich organelles that regulate the storage and hydrolysis of neutral lipids. In the context of immune responses these organelles play several important roles: they help control infection by serving as a docking site for IFN-induced proteins, they are a source of immune modulatory PUFAs such as prostaglandins E2 (PGE2), and of fatty acids that can be used to fuel Oxphos, and they play a role in cross-presentation [103]. LDs in the host are formed rapidly in response to various infectious agents following PRR signaling [104,105,106]. Given their role in aiding cross-presentation and IFN-signaling, their formation is thought to contribute to the host defense particularly in the context of viral infection [107,108,109].
However, LDs are also important sites of PGE2 synthesis, which is known to downregulate pro-inflammatory phenotypes and responses, predominantly in innate immune cells, thereby compromising resistance to multiple infections [106,109,110,111]. For example, T. cruzi benefits from the formation of LDs and the subsequent PGE2 synthesis since the inhibition of COX-2 by aspirin and NS-398-inhibited LD biogenesis in infected macrophages, suppressed the PGE2 production induced by T. cruzi, and diminished the enhancement on parasite replication [112]. Similarly, HCMV, HIV, and M. leprae infection induce LD formation and the production of PGE2 from arachidonic acid. In HIV infection, PGE2 has been linked to reduced antigen-stimulated lymphocyte proliferation, while PGE2 in HCMV infection promotes successful replication. In M. leprae infection, this synthesis is associated with LD formation in Schwann cells, reducing the killing ability of those cells [113,114,115,116,117,118].
In this light, it may come as no surprise that pathogens have been described to actively modulate LD formation and PGE2 production. This is exemplified by S. typhimurium that stimulates LD formation by secreting the secreted effector protein SseJ and by Chlamydia pneumoniae and T. cruzi that are able to accumulate LDs in their vacuoles [104]. Furthermore, the Salmonella pathogenicity island 2 protein C (SpiC) upregulates cyclooxygenase 2 (COX-2) and PGE2 synthesis via the ERK1/2 signal transduction pathway in RAW264.7 macrophages, leading to elevated IL-10 expression [60]. COX-2 upregulation and PGE2 synthesis were observed to a greater extent when murine bone marrow-derived macrophages (BMDMs) and GM-DCs were infected with viable S. typhimurium, both in vitro and in vivo, suggesting that indeed the pathogen itself modulates the production of PGE2 [119]. On top of that, human pathogenic fungi also produce PGE2 and accumulate LDs themselves which has been associated with increased virulence [120,121,122]. These examples support the idea that pathogens are able to hijack the regulation of LD and PGE2 formation to promote immune evasion. However, especially in the case of viruses, in particular HCV and rotaviruses, this might not always be for immunological reasons, but rather to enhance replication efficiency by using LDs as sites of replication and for the assembly of nascent virions [104,123]. Flaviviruses, including Dengue, induce autophagy of LDs, termed lipophagy, to enhance replication efficiency [124]. Dengue induces lipophagy by activating adenosine 5′-monophosphate-activated protein kinase (AMPK), which inhibits mTOR and hence activates autophagy [125].
As already alluded to, mTOR signaling is important for LD formation. The activation of mTOR leads to an upregulation of fatty acid binding proteins (FABPs) that transport free fatty acids (FFAs) to various organelles, and of enzymes involved in triacylglycerol (TAG) synthesis. Consistent with this, M. tuberculosis and T. gondii have been shown to promote mTORC1-dependent TAG accumulation in human macrophages or other cells that contributes to the LD formation [102,126]. Moreover, mannan and peptidoglycan induce COX-2 expression in human polymorphonuclear leukocytes via PRRs and the activation of the PI3K-Akt-mTOR pathway, implicating mTOR signaling not only in LD formation, but also in COX-2 expression that is essential for the manipulation of the host immune response by pathogens [127].
Interestingly, LD formation is also increased by the inhibition of mTORC1 in an autophagy-dependent manner. Autophagy induction is an alternative for increasing TAG levels by increasing the pool of FFAs of which a portion is immediately re-esterified to form TAGs. These TAGs are subsequently packaged into new LDs [128]. Indeed, during L. amazonensis infection of BALB/c macrophages, autophagy in macrophages was associated with increased LD formation, PGE2 synthesis, and increased parasite load [129]. Additionally, rapamycin treatment induces LD formation in the yeast Saccharomyces cerevisiae [130]. This shows that both activation and inhibition of mTOR signaling can result in the formation of LDs. Whether this results in structurally distinct LDs with different immunological functions requires further study [103,131]. In this context it is interesting to note that M. leprae induces LDs that seem to mainly consist of cholesterol and cholesterol esters, while in M. tuberculosis infection LD formation is primarily the result of TAG accumulation [132]. It is tempting to speculate that different pathogens promote the formation of LDs via distinct mechanisms to promote LD formation with functions tailored to their needs.
From what has been discussed above, we may conclude that pathogens can often benefit from the formation of LDs and PGE2 production. mTOR is involved in the formation of LDs and possibly gives rise to functionally different LDs depending on whether mTOR is activated or not. The different functions and their implications in infection is an area that should be explored in more detail to better understand the role of LDs in pathogen–host interactions.

4.2. Membranes

Lipids and fatty acids are an abundant component of membranes and are important for signaling transduction after receptor activation, for example PRRs [133,134]. These receptors are often recruited to and activated in lipid rafts, of which the lipid composition shapes PRR-driven signaling. There are examples of pathogens that are able to modulate lipid composition in either the cytosol or the membranes of target cells in order to alter immune responses. West Nile virus (WNV), for instance, has been shown to redistribute intracellular cholesterol to replication sites, thereby reducing cholesterol levels at the plasma membrane which leads to disruption in antiviral Jak-STAT signaling [135]. Likewise, L. major alters macrophage (BALB/c-derived peritoneal macrophages) CD40 signalosome composition by depleting cholesterol, resulting in the induction of IL-10 production rather than IL-12 [136]. This alteration is regulated by TNF receptor associated factor 6 (TRAF6) and spleen tyrosine kinase (SYK) which both have been shown to regulate mTORC1 signaling, suggesting that mTORC1 signaling is involved in the altered CD40 signalosome composition by L. major [137,138]. Interestingly, cholesterol activates mTORC1 at the lysosomal surface, which indicates that alterations in cholesterol levels elicited by pathogens could, in addition to membrane composition, alter mTORC1 activation [139]. Since pathogens, predominantly viruses, induce membrane formation for their own replication and/or budding, and membrane formation is stimulated via lipid synthesis by mTOR, it is possible that immune regulation through pathogen-driven modifications of membrane composition to modulate signaling and immune responses are more common than currently known.

5. Targeting of Carbohydrate Metabolism by Pathogens

Breakdown of carbohydrates, in particular glucose, plays a central metabolic role in supporting activation and function of immune cells by serving as a rapid source of ATP as well as of carbons to fuel anabolic processes [140,141]. The shift toward anabolism and glycolysis upon macrophage and DC activation is supported by the activation of HIF-1α via mTOR [142]. The involvement of these regulators in glycolysis makes them targets for pathogens aiming to prevent or exploit this shift (Table 1, Figure 2C). For instance, F. tularensis prevents the glycolytic shift by downregulating HIF-1α expression and the subsequent upregulation of glycolytic enzyme PFKFB3 (the gene encoding 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3; it catalyzes the synthesis of fructose-2,6-bisphosphate), which is thought to be driven by its capsule that mimics carbohydrates and outcompetes host sugars that are normally used as substrates in aerobic glycolysis. This downregulation of glycolysis ensures optimal replication and results in impaired pro-inflammatory cytokine production in primary macrophages [143]. Pathogens can also interfere with glycolysis by depleting host cells from glucose. S. typhimurium, for instance, inhibits glycolysis in murine BMDMs by depleting intracellular glucose [144]. Interestingly, this depletion triggers NLRP3 inflammasome signaling which is potentially detrimental for the pathogen. However, this inflammasome activation can be inhibited by S. typhimurium by reducing citrate and reactive oxygen species (ROS) levels [145].
C. albicans exploits glucose metabolism by depleting extracellular glucose, instead of intracellular glucose levels. When C. albicans exits the macrophage (murine BMDMs and PMA differentiated THP-1 cells) it induces glucose uptake and glycolysis through the transcription activators Tye7 and Gal4, which coincides with macrophage restimulation and upregulation of glycolysis. As a consequence, in the absence of glucose these macrophages fail to upregulate glycolysis which leads to the death of the macrophages, thereby evading immune clearance [146]. L. infantum does not prevent the glycolytic shift, but instead promotes glucose oxidation in the mitochondria of murine BMDMs via activation of sirtuin 1 (SIRT1), liver kinase B1 (LKB1), and downstream AMPK, and subsequently via inhibition of mTOR activity. SIRT1 activity is driven by the alteration in intracellular NAD+/NADH ratios. As L. infantum is an NAD+ auxotroph, it is likely that depletion of NAD+ underlies the ability of L. infantum to activate this signaling route [147]. AMPK activation in macrophages has been shown to prevent the acquisition of an inflammatory profile, which would facilitate survival and proliferation of L. infantum [148]. In contrast to the aforementioned pathogens, L. pneumophila promotes a Warburg-like metabolism in human monocyte-derived macrophages, which is a consequence of mitochondrial fragmentation and important for bacterial replication [149]. L. pneumophila actively sustains mTOR activity (through activation of PI3K by the effector proteins Dot/Icm), which may contribute to the promotion of glycolysis [150,151,152]. These examples illustrate that carbohydrate metabolism, in particular glycolysis, is strongly connected to mTOR signaling and that they are a target for various pathogens to modulate the immune response.

6. Targeting of Other mTOR-Controlled Processes by Pathogens

Pathogens do not only target mTOR signaling to alter DC and macrophage metabolism but also to hijack and take advantage of several other processes that are under control of mTOR, such as autophagy and translation, to promote immune evasion. Manipulation of these processes by pathogens are discussed here (Table 1, Figure 2D).

6.1. Autophagy

Autophagy is an essential cellular process that is involved in preventing nutritional starvation and in the defense against pathogens [153]. In innate immunity, autophagy is used in several processes including the control of cytokine production, the degradation and killing of bacteria, and the (cross) presentation of foreign peptides by DCs to induce adaptive immune responses [154]. mTOR is a major regulator of autophagy and because of the role of autophagy in innate immunity, pathogens often modulate the mTOR pathway to affect autophagy [155].
Viruses and intracellular bacteria predominantly suppress autophagy directly or by stimulating mTOR activity, to avoid being degraded and/or since they rely on host anabolic metabolism for their replication [156,157,158,159,160,161]. Following infection of murine BMDMs with S. typhimurium, for example, targets AMPK, SIRT1, and LKB1 to lysosomes for degradation, thereby preventing inhibition of mTOR and activation of autophagy even though ATP and NAD+ are depleted [98]. H. pylori inhibits autophagy as well. Cytotoxin-associated-gene A (CagA) is a critical virulence factor of H. pylori that mediates the activation of mTOR and thus the observed inhibition of autophagy [162].
Nonetheless, some pathogens benefit from the induction of autophagy and thus inhibit mTOR rather than activating it. These pathogens include flaviviruses and coxsackievirus that probably use vesicles arising from autophagy as sites of replication, to liberate nutrients and/or to protect host cells from nutrient starvation-induced death [163,164,165,166]. T. gondii and L. donovani are also inducers of autophagy, but both parasites stimulate autophagy independent of mTOR in order to maintain mTOR activation [167,168]. L. donovani even regulates autophagy in a time-dependent manner in PMA differentiated THP-1 cells. In the early stages of infection mTOR is activated and autophagy is decreased, while later in infection autophagy is activated through mTOR-independent mechanisms, possibly to ensure nutrient supply needed to sustain an ongoing infection [169].
Given the important role for autophagy in shaping innate immune cell function, regulation of autophagy by pathogens also has immunological consequences. By stimulating mTOR, HIV-1 can use the inhibition of autophagy to diminish cross-presentation in infected human monocyte-derived DCs, which enables transfer of infection to neighboring cells such as CD4+ T cells from infected DCs [170,171]. In summary, autophagy is both inhibited by pathogens to evade pathogen degradation and immune recognition as well as stimulated to serve as the nutrient source to sustain replication and infection, illustrating that this process is also targeted by pathogens to promote infection.

6.2. Host Translation

Various pathogens modulate the induction of translation by mTOR to favor the translation of their mRNAs instead of those of the host, to liberate amino acids or to inhibit cytokine responses [172,173,174,175,176]. For example, T. gondii activates mTORC1 to increase translation of mTOR-sensitive transcripts, including those encoding proteins for mitochondrial biogenesis and function. The type I and III strains of T. gondii associate with mitochondria during infection and this association impacts cytokine responses (including increased pro-inflammatory IL-6 and CCL5, but also anti-inflammatory IL-10 production) during in vitro infection of BMDMs with T. gondii and ex vivo in peritoneal exudate cells (PECs) isolated from infected C57BL/6 mice. Therefore, the mitochondrial association and translation of mitochondrial transcripts via mTOR could be a possible route for the manipulation of the host immune response by T. gondii [177,178].
On the other hand, L. major cleaves mTOR to inhibit host translation which prevents for the deleterious effects of type I IFN production and iNOS expression [23]. Also, viruses have been shown to inhibit host-translation to suppress anti-viral cytokine production. The vhs protein of herpes simplex virus (HSV)-1 (encoded by the UL41 gene), for instance, mediates host translation shutoff in the early stage of the virus replication cycle. This shutoff leads to less production of innate cytokines IL-1β, IL-8, and MIP-1 by macrophages [179]. Moreover, mTORC1 inhibition results in the downregulation of the antiviral interferon-induced transmembrane (IFITM) proteins, especially IFITM3. This downregulation of IFITM proteins has already been shown to promote influenza virus A infection in HeLa cells and HFFs [180]. Interestingly, viruses employing internal ribosome entry sites (IRESs) for translation of their mRNAs may benefit from the inhibition of mTORC1, since those viruses do not depend on mTOR-driven cap-dependent translation [23,161,181,182,183].

6.3. Innate Cytokines

Apart from autophagy and translation, mTOR regulates cytokine production in innate immune cells; in particular the balance between pro-inflammatory IL-12 and anti-inflammatory IL-10 expression. DCs and macrophages require mTORC1 signaling for IL-10 expression, which suppresses IL-12 production [17,184]. Various pathogens exploit this reciprocal link by elevating IL-10 production. Different cell wall components of Staphylococcus aureus can activate toll-like receptor 2 (TLR2) signaling, of which some lead to pro-(TNFα production) and others to anti-inflammatory (IL-10 production) responses by human PBMCs. Specifically, IL-10 production after TLR2 activation is dependent on mTOR signaling illustrating that different moieties of pathogens can be used to induce IL-10 production via mTOR signaling [185]. Likewise, blocking of TLR2 or mTOR attenuated IL-10 production induced by the parasite L. donovani in PMA differentiated THP-1 cells while rapamycin decreased IL-10 levels in cornea of Pseudomonas aeruginosa-infected BALB/c mice [186,187]. Of note, in spite of the decreased IL-10 levels during P. aeruginosa infection, rapamycin treatment increased bacterial loads, suggesting that mTOR dependent processes, other than its role in IL-10 production, are more important in controlling P. aeruginosa replication. These examples provide support for the notion that exposure to pathogens can lead to mTOR driven IL-10 expression which could contribute to immune suppression. However, to what extent this mTOR-IL-10 axis is actively exploited by pathogens to promote immune evasion remains to be established.

7. Conclusions

In summary, there is a growing body of literature showing that pathogens have evolved elaborate means to directly or indirectly manipulate mTOR signaling to alter DC and macrophage metabolism and function for their own benefit. From these studies it becomes evident that the consequences of stimulating or inhibiting mTOR signaling by pathogens in terms of immune evasion and pathogen persistence and replication are highly context-dependent. This depends on the cell type and the pathogen involved as well as on the stage of infection and at which level mTOR signaling and metabolism is targeted. In addition, it is also a reflection of the fact that mTOR has a myriad of functions ranging from controlling cellular metabolism to regulating protein expression and autophagy.
Despite this complexity, a clear picture is emerging that a vast range of pathogens, from viruses to multicellular parasites, have developed ways to alter mTOR signaling in host DCs and macrophages to shape their metabolism and immune functions to evade immune responses. Nonetheless, many of the mechanisms through which pathogens target and exploit mTOR signaling and host immune cell metabolism for the control of immune cell function remains to be uncovered. Moreover, to what extent certain changes in activity of mTOR signaling and associated metabolic pathways in response to pathogens are a reflection of manipulation by pathogens, a deliberate host response or the sum of the two combined, is often difficult to define and tease apart. Finally, since mTOR also plays a central role in regulating the functional properties of cells of the adaptive immune system, it is likely that some of the immunomodulatory effects induced by pathogens are not only the result of manipulation of mTOR signaling in innate but also adaptive immune cells [188,189,190]. Addressing these issues will not only contribute to the better understanding of the fundamental role of mTOR signaling and metabolism in interplay between host and pathogen, but also has the potential to identify metabolic vulnerabilities of pathogens that can be exploited and targeted in therapeutic settings to treat infections.

Author Contributions

Conceptualization, B.E. and L.V.N.; writing—original draft preparation, L.V.N.; writing—review and editing, B.E.; visualization, L.V.N.; supervision, B.E.; funding acquisition, B.E. All authors have read and agree to the published version of the manuscript.

Funding

This work is supported by an LUMC Fellowship and VIDI grant (#91719349) by NWO to B.E.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wculek, S.K.; Khouili, S.C.; Priego, E.; Heras-Murillo, I.; Sancho, D. Metabolic Control of Dendritic Cell Functions: Digesting Information. Front. Immunol. 2019, 10, 775. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. O’Neill, L.A.J.; Pearce, E.J. Immunometabolism governs dendritic cell and macrophage function. J. Exp. Med. 2016, 213, 15–23. [Google Scholar] [CrossRef] [PubMed]
  3. Cameron, A.M.; Lawless, S.J.; Pearce, E.J. Metabolism and acetylation in innate immune cell function and fate. Sem. Immunol. 2016, 28, 408–416. [Google Scholar] [CrossRef] [PubMed]
  4. Pearce, E.J.; Everts, B. Dendritic cell metabolism. Nat. Rev. Immunol. 2015, 15, 18–29. [Google Scholar] [CrossRef] [Green Version]
  5. Finlay, B.B.; McFadden, G. Anti-immunology: Evasion of the host immune system by bacterial and viral pathogens. Cell 2006, 124, 767–782. [Google Scholar] [CrossRef] [Green Version]
  6. Collette, J.R.; Lorenz, M.C. Mechanisms of immune evasion in fungal pathogens. Curr. Opin. Microbiol. 2011, 14, 668–675. [Google Scholar] [CrossRef]
  7. Schmid-Hempel, P. Immune defence, parasite evasion strategies and their relevance for “macroscopic phenomena” such as virulence. Philos. Trans. R. Soc. B Biol. Sci. 2009, 364, 85–98. [Google Scholar] [CrossRef] [Green Version]
  8. Freyberg, Z.; Harvill, E.T. Pathogen manipulation of host metabolism: A common strategy for immune evasion. PLoS Pathog. 2017, 13, e1006669. [Google Scholar] [CrossRef] [Green Version]
  9. Olive, A.J.; Sassetti, C.M. Metabolic crosstalk between host and pathogen: Sensing, adapting and competing. Nat. Rev. Microbiol. 2016, 14, 221–234. [Google Scholar] [CrossRef]
  10. Moreno-Altamirano, M.M.B.; Kolstoe, S.E.; Sánchez-García, F.J. Virus Control of Cell Metabolism for Replication and Evasion of Host Immune Responses. Front. Cell Infect. Microbiol. 2019, 9, 95. [Google Scholar] [CrossRef]
  11. Martin, S.; Saha, B.; Riley, J.L. The battle over mTOR: An emerging theatre in host-pathogen immunity. PLoS Pathog. 2012, 8, e1002894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Brunton, J.; Steele, S.; Ziehr, B.; Moorman, N.; Kawula, T. Feeding uninvited guests: mTOR and AMPK set the table for intracellular pathogens. PLoS Pathog. 2013, 9, e1003552. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Osada-Oka, M.; Goda, N.; Saiga, H.; Yamamoto, M.; Takeda, K.; Ozeki, Y.; Yamaguchi, T.; Soga, T.; Tateishi, Y.; Miura, K.; et al. Metabolic adaptation to glycolysis is a basic defense mechanism of macrophages for Mycobacterium tuberculosis infection. Int. Immunol. 2019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Eisenreich, W.; Heesemann, J.; Rudel, T.; Goebel, W. Metabolic host responses to infection by intracellular bacterial pathogens. Front. Cell Infect. Microbiol. 2013, 3, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Shea-Donohue, T.; Qin, B.; Smith, A. Parasites, nutrition, immune responses and biology of metabolic tissues. Parasite Immunol. 2017, 39, e12422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Laplante, M.; Sabatini, D.M. mTOR Signaling in Growth Control and Disease. Cell 2012, 149, 274–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Weichhart, T.; Hengstschläger, M.; Linke, M. Regulation of innate immune cell function by mTOR. Nat. Rev. Immunol. 2015, 15, 599–614. [Google Scholar] [CrossRef]
  18. Liu, Y.; Zhang, D.-T.; Liu, X.-G. mTOR Signaling in T Cell Immunity and Autoimmunity. Int. Rev. Immunol. 2015, 34, 50–66. [Google Scholar] [CrossRef]
  19. Keating, R.; McGargill, M.A. mTOR Regulation of Lymphoid Cells in Immunity to Pathogens. Front. Immunol. 2016, 7, 180. [Google Scholar] [CrossRef]
  20. Guri, Y.; Nordmann, T.M.; Roszik, J. mTOR at the Transmitting and Receiving Ends in Tumor Immunity. Front. Immunol. 2018, 9, 578. [Google Scholar] [CrossRef]
  21. Schroeder, J.H.; McCarthy, D.; Szestak, T.; Cook, D.A.; Taylor, M.J.; Craig, A.G.; Lawson, C.; Lawrence, R.A. Brugia malayi microfilariae adhere to human vascular endothelial cells in a C3-dependent manner. PLoS Negl. Trop. Dis. 2017, 11, e0005592. [Google Scholar] [CrossRef]
  22. Narasimhan, P.B.; Bennuru, S.; Meng, Z.; Cotton, R.N.; Elliott, K.R.; Ganesan, S.; McDonald-Fleming, R.; Veenstra, T.D.; Nutman, T.B.; Semnani, R.T. Microfilariae of Brugia malayi Inhibit the mTOR Pathway and Induce Autophagy in Human Dendritic Cells. Infect. Immun. 2016, 84, 2463–2472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Jaramillo, M.; Gomez, M.A.; Larsson, O.; Shio, M.T.; Topisirovic, I.; Contreras, I.; Luxenburg, R.; Rosenfeld, A.; Colina, R.; McMaster, R.W.; et al. Leishmania repression of host translation through mTOR cleavage is required for parasite survival and infection. Cell Host Microbe 2011, 9, 331–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Clippinger, A.J.; Maguire, T.G.; Alwine, J.C. Human cytomegalovirus infection maintains mTOR activity and its perinuclear localization during amino acid deprivation. J. Virol. 2011, 85, 9369–9376. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Clippinger, A.J.; Alwine, J.C. Dynein mediates the localization and activation of mTOR in normal and human cytomegalovirus-infected cells. Genes Dev. 2012, 26, 2015–2026. [Google Scholar] [CrossRef] [Green Version]
  26. Bayer, C.; Varani, S.; Wang, L.; Walther, P.; Zhou, S.; Straschewski, S.; Bachem, M.; Söderberg-Naucler, C.; Mertens, T.; Frascaroli, G. Human cytomegalovirus infection of M1 and M2 macrophages triggers inflammation and autologous T-cell proliferation. J. Virol. 2013, 87, 67–79. [Google Scholar] [CrossRef] [Green Version]
  27. Gredmark-Russ, S.; Söderberg-Nauclér, C. Dendritic cell biology in human cytomegalovirus infection and the clinical consequences for host immunity and pathology. Virulence 2012, 3, 621–634. [Google Scholar] [CrossRef] [Green Version]
  28. Meade, N.; King, M.; Munger, J.; Walsh, D. mTOR Dysregulation by Vaccinia Virus F17 Controls Multiple Processes with Varying Roles in Infection. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  29. Meade, N.; Furey, C.; Li, H.; Verma, R.; Chai, Q.; Rollins, M.G.; DiGiuseppe, S.; Naghavi, M.H.; Walsh, D. Poxviruses Evade Cytosolic Sensing through Disruption of an mTORC1-mTORC2 Regulatory Circuit. Cell 2018, 174, 1143–1157. [Google Scholar] [CrossRef] [Green Version]
  30. Vekariya, U.; Saxena, R.; Singh, P.; Rawat, K.; Kumar, B.; Kumari, S.; Agnihotri, S.K.; Kaur, S.; Sachan, R.; Nazir, A.; et al. HIV-1 Nef-POTEE; A novel interaction modulates macrophage dissemination via mTORC2 signaling pathway. Life Sci. 2018, 214, 158–166. [Google Scholar] [CrossRef]
  31. Yoon, B.R.; Oh, Y.-J.; Kang, S.W.; Lee, E.B.; Lee, W.-W. Role of SLC7A5 in Metabolic Reprogramming of Human Monocyte/Macrophage Immune Responses. Front. Immunol. 2018, 9, 53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kimura, T.; Nada, S.; Takegahara, N.; Okuno, T.; Nojima, S.; Kang, S.; Ito, D.; Morimoto, K.; Hosokawa, T.; Hayama, Y.; et al. Polarization of M2 macrophages requires Lamtor1 that integrates cytokine and amino-acid signals. Nat. Commun. 2016, 7, 13130. [Google Scholar] [CrossRef] [PubMed]
  33. Wu, G. Amino acids: Metabolism, functions, and nutrition. Amino Acids 2009, 37, 1–17. [Google Scholar] [CrossRef] [PubMed]
  34. McGaha, T.L.; Huang, L.; Lemos, H.; Metz, R.; Mautino, M.; Prendergast, G.C.; Mellor, A.L. Amino acid catabolism: A pivotal regulator of innate and adaptive immunity. Immunol. Rev. 2012, 249, 135. [Google Scholar] [CrossRef] [Green Version]
  35. Zelante, T.; Fallarino, F.; Bistoni, F.; Puccetti, P.; Romani, L. Indoleamine 2,3-dioxygenase in infection: The paradox of an evasive strategy that benefits the host. Microbes Infect. 2009, 11, 133–141. [Google Scholar] [CrossRef]
  36. Friedman, M. Analysis, Nutrition, and Health Benefits of Tryptophan. Int. J. Tryptophan Res. 2018, 11, 1178646918802282. [Google Scholar] [CrossRef] [Green Version]
  37. Passalacqua, K.D.; Charbonneau, M.-E.; O’Riordan, M.X.D. Bacterial Metabolism Shapes the Host-Pathogen Interface. Microbiol. Spectr. 2016, 4. [Google Scholar] [CrossRef] [Green Version]
  38. Qualls, J.E.; Murray, P.J. Immunometabolism within the tuberculosis granuloma: Amino acids, hypoxia, and cellular respiration. Semin. Immunopathol. 2016, 38, 139–152. [Google Scholar] [CrossRef] [Green Version]
  39. Majumdar, T.; Sharma, S.; Kumar, M.; Hussain, M.A.; Chauhan, N.; Kalia, I.; Sahu, A.K.; Rana, V.S.; Bharti, R.; Haldar, A.K.; et al. Tryptophan-kynurenine pathway attenuates β-catenin-dependent pro-parasitic role of STING-TICAM2-IRF3-IDO1 signalosome in Toxoplasma gondii infection. Cell Death Dis. 2019, 10, 161. [Google Scholar] [CrossRef] [Green Version]
  40. Nino-Castro, A.; Abdullah, Z.; Popov, A.; Thabet, Y.; Beyer, M.; Knolle, P.; Domann, E.; Chakraborty, T.; Schmidt, S.V.; Schultze, J.L. The IDO1-induced kynurenines play a major role in the antimicrobial effect of human myeloid cells against Listeria monocytogenes. Innate Immune 2014, 20, 401–411. [Google Scholar] [CrossRef]
  41. Zhang, Y.J.; Reddy, M.C.; Ioerger, T.R.; Rothchild, A.C.; Dartois, V.; Schuster, B.M.; Trauner, A.; Wallis, D.; Galaviz, S.; Huttenhower, C.; et al. Tryptophan biosynthesis protects mycobacteria from CD4 T-cell-mediated killing. Cell 2013, 155, 1296–1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Munn, D.H.; Mellor, A.L. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013, 34, 137–143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mellor, A.L.; Munn, D.H. Ido expression by dendritic cells: Tolerance and tryptophan catabolism. Nat. Rev. Immunol. 2004, 4, 762–774. [Google Scholar] [CrossRef]
  44. Wang, X.F.; Wang, H.S.; Wang, H.; Zhang, F.; Wang, K.F.; Guo, Q.; Zhang, G.; Cai, S.H.; Du, J. The role of indoleamine 2,3-dioxygenase (IDO) in immune tolerance: Focus on macrophage polarization of THP-1 cells. Cell Immunol. 2014, 289, 42–48. [Google Scholar] [CrossRef] [PubMed]
  45. Poormasjedi-Meibod, M.-S.; Jalili, R.B.; Hosseini-Tabatabaei, A.; Hartwell, R.; Ghahary, A. Immuno-regulatory function of indoleamine 2,3 dioxygenase through modulation of innate immune responses. PLoS ONE 2013, 8, e71044. [Google Scholar] [CrossRef] [PubMed]
  46. Pallotta, M.T.; Orabona, C.; Volpi, C.; Vacca, C.; Belladonna, M.L.; Bianchi, R.; Servillo, G.; Brunacci, C.; Calvitti, M.; Bicciato, S.; et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-term tolerance by dendritic cells. Nat. Immunol. 2011, 12, 870–878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Avdic, S.; McSharry, B.P.; Slobedman, B. Modulation of dendritic cell functions by viral IL-10 encoded by human cytomegalovirus. Front. Microbiol. 2014, 5, 337. [Google Scholar] [CrossRef] [PubMed]
  48. Raftery, M.J.; Wieland, D.; Gronewald, S.; Kraus, A.A.; Giese, T.; Schönrich, G. Shaping Phenotype, Function, and Survival of Dendritic Cells by Cytomegalovirus-Encoded IL-10. J. Immunol. 2004, 173, 3383–3391. [Google Scholar] [CrossRef] [Green Version]
  49. Mittal, D.; Kassianos, A.J.; Tran, L.S.; Bergot, A.S.; Gosmann, C.; Hofmann, J.; Blumenthal, A.; Leggatt, G.R.; Frazer, I.H. Indoleamine 2,3-dioxygenase activity contributes to local immune suppression in the skin expressing human papillomavirus oncoprotein e7. J. Investig Dermatol. 2013, 133, 2686–2694. [Google Scholar] [CrossRef] [Green Version]
  50. Metz, R.; Rust, S.; DuHadaway, J.B.; Mautino, M.R.; Munn, D.H.; Vahanian, N.N.; Link, C.J.; Prendergast, G.C. IDO inhibits a tryptophan sufficiency signal that stimulates mTOR: A novel IDO effector pathway targeted by D-1-methyl-tryptophan. Oncoimmunology 2012, 1, 1460–1468. [Google Scholar] [CrossRef] [Green Version]
  51. Battu, S.; Minhas, G.; Mishra, A.; Khan, N. Amino Acid Sensing via General Control Nonderepressible-2 Kinase and Immunological Programming. Front. Immunol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  52. Castilho, B.A.; Shanmugam, R.; Silva, R.C.; Ramesh, R.; Himme, B.M.; Sattlegger, E. Keeping the eIF2 alpha kinase Gcn2 in check. Biochim. Biophys. Acta 2014, 1843, 1948–1968. [Google Scholar] [CrossRef] [Green Version]
  53. Tattoli, I.; Sorbara, M.T.; Vuckovic, D.; Ling, A.; Soares, F.; Carneiro, L.A.; Yang, C.; Emili, A.; Philpott, D.J.; Girardin, S.E. Amino acid starvation induced by invasive bacterial pathogens triggers an innate host defense program. Cell Host Microbe 2012, 11, 563–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ravishankar, B.; Liu, H.; Shinde, R.; Chaudhary, K.; Xiao, W.; Bradley, J.; Koritzinsky, M.; Madaio, M.P.; McGaha, T.L. The amino acid sensor GCN2 inhibits inflammatory responses to apoptotic cells promoting tolerance and suppressing systemic autoimmunity. Proc. Natl. Acad. Sci. USA 2015, 112, 10774–10779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Perl, A.; Hanczko, R.; Lai, Z.W.; Oaks, Z.; Kelly, R.; Borsuk, R.; Asara, J.M.; Phillips, P.E. Comprehensive metabolome analyses reveal N-acetylcysteine-responsive accumulation of kynurenine in systemic lupus erythematosus: Implications for activation of the mechanistic target of rapamycin. Metabolomics 2015, 11, 1157–1174. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Osawa, Y.; Kanamori, H.; Seki, E.; Hoshi, M.; Ohtaki, H.; Yasuda, Y.; Ito, H.; Suetsugu, A.; Nagaki, M.; Moriwaki, H.; et al. L-tryptophan-mediated enhancement of susceptibility to nonalcoholic fatty liver disease is dependent on the mammalian target of rapamycin. J. Biol. Chem. 2011, 286, 34800–34808. [Google Scholar] [CrossRef] [Green Version]
  57. Nguyen, N.T.; Kimura, A.; Nakahama, T.; Chinen, I.; Masuda, K.; Nohara, K.; Fujii-Kuriyama, Y.; Kishimoto, T. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc. Natl. Acad. Sci. USA 2010, 107, 19961–19966. [Google Scholar] [CrossRef] [Green Version]
  58. Mezrich, J.D.; Fechner, J.H.; Zhang, X.; Johnson, B.P.; Burlingham, W.J.; Bradfield, C.A. An interaction between kynurenine and the aryl hydrocarbon receptor can generate regulatory T cells. J. Immunol. 2010, 185, 3190–3198. [Google Scholar] [CrossRef] [Green Version]
  59. Sorgdrager, F.J.H.; Naudé, P.J.W.; Kema, I.P.; Nollen, E.A.; Deyn, P.P.D. Tryptophan Metabolism in Inflammaging: From Biomarker to Therapeutic Target. Front. Immunol. 2019, 10, 2565. [Google Scholar] [CrossRef]
  60. Uchiya, K.-I.; Groisman, E.A.; Nikai, T. Involvement of Salmonella pathogenicity island 2 in the up-regulation of interleukin-10 expression in macrophages: Role of protein kinase A signal pathway. Infect. Immun. 2004, 72, 1964–1973. [Google Scholar] [CrossRef] [Green Version]
  61. Tsalikis, J.; Croitoru, D.O.; Philpott, D.J.; Girardin, S.E. Nutrient sensing and metabolic stress pathways in innate immunity. Cell Microbiol. 2013, 15, 1632–1641. [Google Scholar] [CrossRef]
  62. McRae, M.P. Therapeutic Benefits of l-Arginine: An Umbrella Review of Meta-analyses. J. Chiropr. Med. 2016, 15, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Popovic, P.J.; Zeh, H.J., 3rd; Ochoa, J.B. Arginine and immunity. J Nutr. 2007, 137, 1681S–1686S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Fang, F.C. Perspectives series: Host/pathogen interactions. Mechanisms of nitric oxide-related antimicrobial activity. J. Clin. Investig. 1997, 99, 2818–2825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Das, P.; Lahiri, A.; Lahiri, A.; Chakravortty, D. Modulation of the arginase pathway in the context of microbial pathogenesis: A metabolic enzyme moonlighting as an immune modulator. PLoS Pathog. 2010, 6, e1000899. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Gogoi, M.; Datey, A.; Wilson, K.T.; Chakravortty, D. Dual role of arginine metabolism in establishing pathogenesis. Curr. Opin. Microbiol. 2016, 29, 43–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. El Kasmi, K.C.; Qualls, J.E.; Pesce, J.T.; Smith, A.M.; Thompson, R.W.; Henao-Tamayo, M.; Basaraba, R.J.; König, T.; Schleicher, U.; Koo, M.S.; et al. Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat. Immunol. 2008, 9, 1399–1406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Holzmuller, P.; Geiger, A.; Nzoumbou-Boko, R.; Pissarra, J.; Hamrouni, S.; Rodrigues, V.; Dauchy, F.A.; Lemesre, J.L.; Vincendeau, P.; Bras-Gonçalves, R. Trypanosomatid Infections: How Do Parasites and Their Excreted–Secreted Factors Modulate the Inducible Metabolism of l-Arginine in Macrophages? Front. Immunol. 2018, 9, 778. [Google Scholar] [CrossRef]
  69. Kropf, P.; Fuentes, J.M.; Fähnrich, E.; Arpa, L.; Herath, S.; Weber, V.; Soler, G.; Celada, A.; Modolell, M.; Müller, I. Arginase and polyamine synthesis are key factors in the regulation of experimental leishmaniasis in vivo. FASEB J. 2005, 19, 1000–1002. [Google Scholar] [CrossRef]
  70. Duque-Correa, M.A.; Kühl, A.A.; Rodriguez, P.C.; Zedler, U.; Schommer-Leitner, S.; Rao, M.; Weiner, J.; Hurwitz, R.; Qualls, J.E.; Kosmiadi, G.A.; et al. Macrophage arginase-1 controls bacterial growth and pathology in hypoxic tuberculosis granulomas. Proc. Natl. Acad. Sci. USA 2014, 111, E4024–E4032. [Google Scholar] [CrossRef] [Green Version]
  71. Badirzadeh, A.; Taheri, T.; Taslimi, Y.; Abdossamadi, Z.; Heidari-Kharaji, M.; Gholami, E.; Sedaghat, B.; Niyyati, M.; Rafati, S. Arginase activity in pathogenic and non-pathogenic species of Leishmania parasites. PLoS Negl. Trop. Dis. 2017, 11, e0005774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. da Silva, M.F.L.; Floeter-Winter, L.M. Arginase in Leishmania. Subcell. Biochem. 2014, 74, 103–117. [Google Scholar] [CrossRef]
  73. Boitz, J.M.; Gilroy, C.A.; Olenyik, T.D.; Paradis, D.; Perdeh, J.; Dearman, K.; Davis, M.J.; Yates, P.A.; Li, Y.; Riscoe, M.K.; et al. Arginase Is Essential for Survival of Leishmania donovani Promastigotes but Not Intracellular Amastigotes. Infect. Immun. 2017, 85, e00554-16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Gobert, A.P.; McGee, D.J.; Akhtar, M.; Mendz, G.L.; Newton, J.C.; Cheng, Y.; Mobley, H.L.; Wilson, K.T. Helicobacter pylori arginase inhibits nitric oxide production by eukaryotic cells: A strategy for bacterial survival. Proc. Natl. Acad. Sci. USA 2001, 98, 13844–13849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Chaturvedi, R.; Asim, M.; Lewis, N.D.; Algood, H.M.; Cover, T.L.; Kim, P.Y.; Wilson, K.T. L-Arginine Availability Regulates Inducible Nitric Oxide Synthase-Dependent Host Defense against Helicobacter pylori. Infect. Immun. 2007, 75, 4305–4315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Jiménez-López, C.; Collette, J.R.; Brothers, K.M.; Shepardson, K.M.; Cramer, R.A.; Wheeler, R.T.; Lorenz, M.C. Candida albicans induces arginine biosynthetic genes in response to host-derived reactive oxygen species. Eukaryot. Cell 2013, 12, 91–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Puleston, D.J.; Buck, M.D.; Geltink, R.I.; Kyle, R.L.; Caputa, G.; O’Sullivan, D.; Cameron, A.M.; Castoldi, A.; Musa, Y.; Kabat, A.M.; et al. Polyamines and eIF5A Hypusination Modulate Mitochondrial Respiration and Macrophage Activation. Cell Metab. 2019, 30, 352–363.e8. [Google Scholar] [CrossRef] [Green Version]
  78. Raniga, K.; Liang, C. Interferons: Reprogramming the Metabolic Network against Viral Infection. Viruses 2018, 10, 36. [Google Scholar] [CrossRef] [Green Version]
  79. Goh, C.C.; Roggerson, K.M.; Lee, H.-C.; Golden-Mason, L.; Rosen, H.R.; Hahn, Y.S. Hepatitis C Virus-Induced Myeloid-Derived Suppressor Cells Suppress NK Cell IFN-γ Production by Altering Cellular Metabolism via Arginase-1. J. Immunol. 2016, 196, 2283–2292. [Google Scholar] [CrossRef] [Green Version]
  80. Banik, S.; Renner Viveros, P.; Seeber, F.; Klotz, C.; Ignatius, R.; Aebischer, T. Giardia duodenalis arginine deiminase modulates the phenotype and cytokine secretion of human dendritic cells by depletion of arginine and formation of ammonia. Infect. Immun. 2013, 81, 2309–2317. [Google Scholar] [CrossRef] [Green Version]
  81. Ban, H.; Shigemitsu, K.; Yamatsuji, T.; Haisa, M.; Nakajo, T.; Takaoka, M.; Nobuhisa, T.; Gunduz, M.; Tanaka, N.; Naomoto, Y. Arginine and Leucine regulate p70 S6 kinase and 4E-BP1 in intestinal epithelial cells. Int. J. Mol. Med. 2004, 13, 537–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Chantranupong, L.; Scaria, S.M.; Saxton, R.A.; Gygi, M.P.; Shen, K.; Wyant, G.A.; Wang, T.; Harper, J.W.; Gygi, S.P.; Sabatini, D.M. The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway. Cell 2016, 165, 153–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Bar-Peled, L.; Sabatini, D.M. Regulation of mTORC1 by amino acids. Trends Cell Biol. 2014, 24, 400–406. [Google Scholar] [CrossRef] [PubMed]
  84. Averous, J.; Lambert-Langlais, S.; Mesclon, F.; Carraro, V.; Parry, L.; Jousse, C.; Bruhat, A.; Maurin, A.C.; Pierre, P.; Proud, C.G.; et al. GCN2 contributes to mTORC1 inhibition by leucine deprivation through an ATF4 independent mechanism. Sci. Rep. 2016, 6, 27698. [Google Scholar] [CrossRef]
  85. Fletcher, M.; Ramirez, M.E.; Sierra, R.A.; Raber, P.; Thevenot, P.; Al-Khami, A.A.; Sanchez-Pino, D.; Hernandez, C.; Wyczechowska, D.D.; Ochoa, A.C.; et al. l-Arginine Depletion Blunts Antitumor T-cell Responses by Inducing Myeloid-Derived Suppressor Cells. Cancer Res. 2015, 75, 275–283. [Google Scholar] [CrossRef] [Green Version]
  86. Shaheen, Z.R.; Naatz, A.; Corbett, J.A. CCR5-Dependent Activation of mTORC1 Regulates Translation of Inducible NO Synthase and COX-2 during Encephalomyocarditis Virus Infection. J. Immunol. 2015, 195, 4406–4414. [Google Scholar] [CrossRef] [Green Version]
  87. Everts, B.; Amiel, E.; Huang, S.C.; Smith, A.M.; Chang, C.H.; Lam, W.Y.; Redmann, V.; Freitas, T.C.; Blagih, J.; Van Der Windt, G.J.; et al. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat. Immunol. 2014, 15, 323–332. [Google Scholar] [CrossRef] [Green Version]
  88. Kullas, A.L.; McClelland, M.; Yang, H.J.; Tam, J.W.; Torres, A.; Porwollik, S.; Mena, P.; McPhee, J.B.; Bogomolnaya, L.; Andrews-Polymenis, H.; et al. L-asparaginase II produced by Salmonella typhimurium inhibits T cell responses and mediates virulence. Cell Host Microbe 2012, 12, 791–798. [Google Scholar] [CrossRef] [Green Version]
  89. Shibayama, K.; Takeuchi, H.; Wachino, J.-I.; Mori, S.; Arakawa, Y. Biochemical and pathophysiological characterization of Helicobacter pylori asparaginase. Microbiol. Immunol. 2011, 55, 408–417. [Google Scholar] [CrossRef]
  90. Torres, A.; Luke, J.D.; Kullas, A.L.; Kapilashrami, K.; Botbol, Y.; Koller, A.; Tonge, P.J.; Chen, E.I.; Macian, F.; van der Velden, A.W. Asparagine deprivation mediated by Salmonella asparaginase causes suppression of activation-induced T cell metabolic reprogramming. J. Leukoc. Biol. 2016, 99, 387–398. [Google Scholar] [CrossRef] [Green Version]
  91. Cachumba, J.J.M.; Antunes, F.A.F.; Peres, G.F.D.; Brumano, L.P.; Santos, J.C.D.; Da Silva, S.S. Current applications and different approaches for microbial l-asparaginase production. Braz. J. Microbiol. 2016, 47, 77–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hofreuter, D.; Novik, V.; Galán, J.E. Metabolic diversity in Campylobacter jejuni enhances specific tissue colonization. Cell Host Microbe 2008, 4, 425–433. [Google Scholar] [CrossRef] [Green Version]
  93. Song, P.; Wang, Z.; Zhang, X.; Fan, J.; Li, Y.; Chen, Q.; Wang, S.; Liu, P.; Luan, J.; Ye, L.; et al. The role of autophagy in asparaginase-induced immune suppression of macrophages. Cell Death Dis. 2017, 8, e2721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Rabinovich, S.; Adler, L.; Yizhak, K.; Sarver, A.; Silberman, A.; Agron, S.; Stettner, N.; Sun, Q.; Brandis, A.; Helbling, D.; et al. Diversion of aspartate in ASS1-deficient tumours fosters de novo pyrimidine synthesis. Nature 2015, 527, 379–383. [Google Scholar] [CrossRef] [PubMed]
  95. Cruzat, V.; Macedo Rogero, M.; Noel Keane, K.; Curi, R.; Newsholme, P. Glutamine: Metabolism and Immune Function, Supplementation and Clinical Translation. Nutrients 2018, 10, 1564. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Stunault, M.I.; Bories, G.; Guinamard, R.R.; Ivanov, S. Metabolism Plays a Key Role during Macrophage Activation. Mediat. Inflamm. 2018, 2018, 1–10. [Google Scholar] [CrossRef] [PubMed]
  97. Tattoli, I.; Philpott, D.J.; Girardin, S.E. The bacterial and cellular determinants controlling the recruitment of mTOR to the Salmonella-containing vacuole. Biol. Open 2012, 1, 1215–1225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ganesan, R.; Hos, N.J.; Gutierrez, S.; Fischer, J.; Stepek, J.M.; Daglidu, E.; Krönke, M.; Robinson, N. Salmonella Typhimurium disrupts Sirt1/AMPK checkpoint control of mTOR to impair autophagy. PLoS Pathog. 2017, 13, e1006227. [Google Scholar] [CrossRef] [Green Version]
  99. Kudchodkar, S.B.; Del Prete, G.Q.; Maguire, T.G.; Alwine, J.C. AMPK-mediated inhibition of mTOR kinase is circumvented during immediate-early times of human cytomegalovirus infection. J. Virol. 2007, 81, 3649–3651. [Google Scholar] [CrossRef] [Green Version]
  100. Kudchodkar, S.B.; Yu, Y.; Maguire, T.G.; Alwine, J.C. Human cytomegalovirus infection induces rapamycin-insensitive phosphorylation of downstream effectors of mTOR kinase. J. Virol. 2004, 78, 11030–11039. [Google Scholar] [CrossRef] [Green Version]
  101. Laplante, M.; Sabatini, D.M. An emerging role of mTOR in lipid biosynthesis. Curr. Biol. 2009, 19, R1046–R1052. [Google Scholar] [CrossRef] [Green Version]
  102. Guerrini, V.; Prideaux, B.; Blanc, L.; Bruiners, N.; Arrigucci, R.; Singh, S.; Ho-Liang, H.P.; Salamon, H.; Chen, P.Y.; Lakehal, K.; et al. Storage lipid studies in tuberculosis reveal that foam cell biogenesis is disease-specific. PLoS Pathog. 2018, 14, e1007223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. den Brok, M.H.; Raaijmakers, T.K.; Collado-Camps, E.; Adema, G.J. Lipid Droplets as Immune Modulators in Myeloid Cells. Trends Immunol. 2018, 39, 380–392. [Google Scholar] [CrossRef] [PubMed]
  104. Saka, H.A.; Valdivia, R. Emerging roles for lipid droplets in immunity and host-pathogen interactions. Annu. Rev. Cell Dev. Biol. 2012, 28, 411–437. [Google Scholar] [CrossRef] [PubMed]
  105. Wilson, S.K.; Knoll, L.J. Patatin-like phospholipases in microbial infections with emerging roles in fatty acid metabolism and immune regulation by Apicomplexa. Mol. Microbiol. 2018, 107, 34–46. [Google Scholar] [CrossRef] [PubMed]
  106. D’Avila, H.; Freire-de-Lima, C.G.; Roque, N.R.; Teixeira, L.; Barja-Fidalgo, C.; Silva, A.R.; Melo, R.C.; DosReis, G.A.; Castro-Faria-Neto, H.C.; Bozza, P.T. 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. 2011, 204, 951–961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Steer, S.A.; Corbett, J.A. The Role and Regulation of COX-2 during Viral Infection. Viral Immunol. 2003, 16, 447–460. [Google Scholar] [CrossRef] [PubMed]
  108. Sander, W.J.; O’Neill, H.G.; Pohl, C.H. Prostaglandin E2 as a Modulator of Viral Infections. Front. Physiol. 2017, 8. [Google Scholar] [CrossRef] [Green Version]
  109. Vallochi, A.L.; Teixeira, L.; Oliveira K da, S.; Maya-Monteiro, C.M.; Bozza, P.T. Lipid Droplet, a Key Player in Host-Parasite Interactions. Front. Immunol. 2018, 9, 1022. [Google Scholar] [CrossRef]
  110. Libbing, C.L.; McDevitt, A.R.; Azcueta, R.-M.P.; Ahila, A.; Mulye, M. Lipid Droplets: A Significant but Understudied Contributor of Host–Bacterial Interactions. Cells 2019, 8, 354. [Google Scholar] [CrossRef] [Green Version]
  111. Agard, M.; Asakrah, S.; Morici, L.A. PGE (2) suppression of innate immunity during mucosal bacterial infection. Front. Cell Infect. Microbiol. 2013, 3, 45. [Google Scholar] [CrossRef] [Green Version]
  112. Bozza, P.T.; Payne, J.L.; Morham, S.G.; Langenbach, R.; Smithies, O.; Weller, P.F. Leukocyte lipid body formation and eicosanoid generation: Cyclooxygenase-independent inhibition by aspirin. Proc. Natl. Acad. Sci. USA 1996, 93, 11091–11096. [Google Scholar] [CrossRef] [Green Version]
  113. Raulin, J. Lipids and retroviruses. Lipids 2000, 35, 123–130. [Google Scholar] [CrossRef]
  114. Mattos, K.A.; Oliveira, V.G.; D’Avila, H.; Rodrigues, L.S.; Pinheiro, R.O.; Sarno, E.N.; Pessolani, M.C.; Bozza, P.T. TLR6-driven lipid droplets in Mycobacterium leprae-infected Schwann cells: Immunoinflammatory platforms associated with bacterial persistence. J. Immunol. 2011, 187, 2548–2558. [Google Scholar] [CrossRef] [Green Version]
  115. Rodríguez-Sánchez, I.; Munger, J. Meal for Two: Human Cytomegalovirus-Induced Activation of Cellular Metabolism. Viruses 2019, 11, 273. [Google Scholar] [CrossRef] [Green Version]
  116. Foley, P.; Kazazi, F.; Biti, R.; Sorrell, T.C.; Cunningham, A.L. HIV infection of monocytes inhibits the T-lymphocyte proliferative response to recall antigens, via production of eicosanoids. Immunology 1992, 75, 391–397. [Google Scholar]
  117. Castellano, P.; Prevedel, L.; Valdebenito, S.; Eugenin, E.A. HIV infection and latency induce a unique metabolic signature in human macrophages. Sci. Rep. 2019, 9, 3941. [Google Scholar] [CrossRef] [Green Version]
  118. Yu, Y.; Maguire, T.G.; Alwine, J.C. Human cytomegalovirus infection induces adipocyte-like lipogenesis through activation of sterol regulatory element binding protein 1. J. Virol. 2012, 86, 2942–2949. [Google Scholar] [CrossRef] [Green Version]
  119. Bowman, C.C.; Bost, K.L. Cyclooxygenase-2-mediated prostaglandin E2 production in mesenteric lymph nodes and in cultured macrophages and dendritic cells after infection with Salmonella. J. Immunol. 2004, 172, 2469–2475. [Google Scholar] [CrossRef]
  120. Chakraborty, T.; Thuer, E.; Heijink, M.; Tóth, R.; Bodai, L.; Vágvölgyi, C.; Giera, M.; Gabaldón, T.; Gácser, A. Eicosanoid biosynthesis influences the virulence of Candida parapsilosis. Virulence 2018, 9, 1019–1035. [Google Scholar] [CrossRef] [Green Version]
  121. Murphy, D.J. The dynamic roles of intracellular lipid droplets: From archaea to mammals. Protoplasma 2012, 249, 541–585. [Google Scholar] [CrossRef] [PubMed]
  122. Singh, A.; Del Poeta, M. Lipid signalling in pathogenic fungi. Cell. Microbiol. 2011, 13, 177–185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Crawford, S.E.; Desselberger, U. Lipid droplets form complexes with viroplasms and are crucial for rotavirus replication. Curr. Opin. Virol. 2016, 19, 11–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Leier, H.C.; Messer, W.B.; Tafesse, F.G. Lipids and pathogenic flaviviruses: An intimate union. PLoS Pathog. 2018, 14, e1006952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Jordan, T.X.; Randall, G. Dengue Virus Activates the AMP Kinase-mTOR Axis to Stimulate a Proviral Lipophagy. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Hu, X.; Binns, D.; Reese, M.L. The coccidian parasites and dysregulate mammalian lipid droplet biogenesis. J. Biol. Chem. 2017, 292, 11009–11020. [Google Scholar] [CrossRef] [Green Version]
  127. Fernández, N.; González, A.; Valera, I.; Alonso, S.; Crespo, M.S. Mannan and peptidoglycan induce COX-2 protein in human PMN via the mammalian target of rapamycin. Eur. J. Immunol. 2007, 37, 2572–2582. [Google Scholar] [CrossRef]
  128. Olzmann, J.A.; Carvalho, P. Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol. 2019, 20, 137–155. [Google Scholar] [CrossRef]
  129. Pinheiro, R.O.; Nunes, M.P.; Pinheiro, C.S.; D’Avila, H.; Bozza, P.T.; Takiya, C.M.; Côrte-Real, S.; Freire-de-Lima, C.G.; DosReis, G.A. Induction of autophagy correlates with increased parasite load of Leishmania amazonensis in BALB/c but not C57BL/6 macrophages. Microbes Infect. 2009, 11, 181–190. [Google Scholar] [CrossRef]
  130. Madeira, J.B.; Masuda, C.A.; Maya-Monteiro, C.M.; Matos, G.S.; Montero-Lomelí, M.; Bozaquel-Morais, B.L. TORC1 inhibition induces lipid droplet replenishment in yeast. Mol. Cell Biol. 2015, 35, 737–746. [Google Scholar] [CrossRef] [Green Version]
  131. Menon, D.; Singh, K.; Pinto, S.M.; Nandy, A.; Jaisinghani, N.; Kutum, R.; Dash, D.; Prasad, T.K.; Gandotra, S. Quantitative Lipid Droplet Proteomics Reveals Mycobacterium tuberculosis Induced Alterations in Macrophage Response to Infection. ACS Infect. Dis. 2019, 5, 559–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Mattos, K.A.; Lara, F.A.; Oliveira, V.G.; Rodrigues, L.S.; D’Avila, H.; Melo, R.C.; Manso, P.P.; Sarno, E.N.; Bozza, P.T.; Pessolani, M.C. Modulation of lipid droplets by Mycobacterium leprae in Schwann cells: A putative mechanism for host lipid acquisition and bacterial survival in phagosomes. Cell Microbiol. 2011, 13, 259–273. [Google Scholar] [CrossRef] [PubMed]
  133. Schumann, J. The Impact of Macrophage Membrane Lipid Composition on Innate Immune Response Mechanisms; IntechOpen: Rijeka, Croatia, 2012. [Google Scholar] [CrossRef] [Green Version]
  134. Olsson, S.; Sundler, R. The role of lipid rafts in LPS-induced signaling in a macrophage cell line. Mol. Immunol. 2006, 43, 607–612. [Google Scholar] [CrossRef] [PubMed]
  135. Mackenzie, J.M.; Khromykh, A.A.; Parton, R.G. Cholesterol manipulation by West Nile virus perturbs the cellular immune response. Cell Host Microbe 2007, 2, 229–239. [Google Scholar] [CrossRef] [Green Version]
  136. Rub, A.; Dey, R.; Jadhav, M.; Kamat, R.; Chakkaramakkil, S.; Majumdar, S.; Mukhopadhyaya, R.; Saha, B. Cholesterol depletion associated with Leishmania major infection alters macrophage CD40 signalosome composition and effector function. Nat. Immunol. 2009, 10, 273–280. [Google Scholar] [CrossRef]
  137. Linares, J.F.; Duran, A.; Yajima, T.; Pasparakis, M.; Moscat, J.; Diaz-Meco, M.T. K63 polyubiquitination and activation of mTOR by the p62-TRAF6 complex in nutrient-activated cells. Mol. Cell 2013, 51, 283–296. [Google Scholar] [CrossRef] [Green Version]
  138. Yin, H.; Zhou, H.; Kang, Y.; Zhang, X.; Duan, X.; Alnabhan, R.; Liang, S.; Scott, D.A.; Lamont, R.J.; Shang, J.; et al. Syk negatively regulates TLR4-mediated IFNβ and IL-10 production and promotes inflammatory responses in dendritic cells. Biochim. Biophys. Acta 2016, 1860, 588–598. [Google Scholar] [CrossRef] [Green Version]
  139. Castellano, B.M.; Thelen, A.M.; Moldavski, O.; Feltes, M.; Van Der Welle, R.E.; Mydock-McGrane, L.; Jiang, X.; Van Eijkeren, R.J.; Davis, O.B.; Louie, S.M.; et al. Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science 2017, 355, 1306–1311. [Google Scholar] [CrossRef] [Green Version]
  140. Ganeshan, K.; Chawla, A. Metabolic regulation of immune responses. Annu. Rev. Immunol. 2014, 32, 609–634. [Google Scholar] [CrossRef] [Green Version]
  141. Loftus, R.M.; Finlay, D.K. Immunometabolism: Cellular Metabolism Turns Immune Regulator. J. Biol. Chem. 2016, 291, 1–10. [Google Scholar] [CrossRef] [Green Version]
  142. Kelly, B.; O’Neill, L.A.J. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015, 25, 771–784. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Wyatt, E.V.; Diaz, K.; Griffin, A.J.; Rasmussen, J.A.; Crane, D.D.; Jones, B.D.; Bosio, C.M. Metabolic Reprogramming of Host Cells by Virulent Francisella tularensis for Optimal Replication and Modulation of Inflammation. J. Immunol. 2016, 196, 4227–4236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Sanman, L.E.; Qian, Y.; Eisele, N.A.; Ng, T.M.; van der Linden, W.A.; Monack, D.M.; Weerapana, E.; Bogyo, M. Disruption of glycolytic flux is a signal for inflammasome signaling and pyroptotic cell death. Elife 2016, 5, e13663. [Google Scholar] [CrossRef] [PubMed]
  145. Wynosky-Dolfi, M.A.; Snyder, A.G.; Philip, N.H.; Doonan, P.J.; Poffenberger, M.C.; Avizonis, D.; Zwack, E.E.; Riblett, A.M.; Hu, B.; Strowig, T.; et al. Oxidative metabolism enables Salmonella evasion of the NLRP3 inflammasome. J. Exp. Med. 2014, 211, 653–668. [Google Scholar] [CrossRef] [Green Version]
  146. Tucey, T.M.; Verma, J.; Harrison, P.F.; Snelgrove, S.L.; Lo, T.L.; Scherer, A.K.; Barugahare, A.A.; Powell, D.R.; Wheeler, R.T.; Hickey, M.J.; et al. Glucose Homeostasis Is Important for Immune Cell Viability during Candida Challenge and Host Survival of Systemic Fungal Infection. Cell Metab. 2018, 27, 988–1006. [Google Scholar] [CrossRef]
  147. Moreira, D.; Rodrigues, V.; Abengozar, M.; Rivas, L.; Rial, E.; Laforge, M.; Li, X.; Foretz, M.; Viollet, B.; Estaquier, J.; et al. Leishmania infantum modulates host macrophage mitochondrial metabolism by hijacking the SIRT1-AMPK axis. PLoS Pathog. 2015, 11, e1004684. [Google Scholar] [CrossRef] [Green Version]
  148. Sag, D.; Carling, D.; Stout, R.D.; Suttles, J. Adenosine 5′-Monophosphate-Activated Protein Kinase Promotes Macrophage Polarization to an Anti-Inflammatory Functional Phenotype. J. Immunol. 2008, 181, 8633–8641. [Google Scholar] [CrossRef]
  149. Escoll, P.; Song, O.R.; Viana, F.; Steiner, B.; Lagache, T.; Olivo-Marin, J.C.; Impens, F.; Brodin, P.; Hilbi, H.; Buchrieser, C. Legionella pneumophila Modulates Mitochondrial Dynamics to Trigger Metabolic Repurposing of Infected Macrophages. Cell Host Microbe 2017, 22, 302–316. [Google Scholar] [CrossRef]
  150. Abshire, C.F.; Dragoi, A.-M.; Roy, C.R.; Ivanov, S.S. MTOR-Driven Metabolic Reprogramming Regulates Legionella pneumophila Intracellular Niche Homeostasis. PLoS Pathog. 2016, 12, e1006088. [Google Scholar] [CrossRef]
  151. Ivanov, S. The tug-of-war over MTOR in Legionella infections. Microb. Cell 2017, 4, 67–68. [Google Scholar] [CrossRef]
  152. Prevost, M.S.; Pinotsis, N.; Dumoux, M.; Hayward, R.D.; Waksman, G. The Legionella effector WipB is a translocated Ser/Thr phosphatase that targets the host lysosomal nutrient sensing machinery. Sci. Rep. 2017, 7, 9450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Ravanan, P.; Srikumar, I.F.; Talwar, P. Autophagy: The spotlight for cellular stress responses. Life Sci. 2017, 188, 53–67. [Google Scholar] [CrossRef] [PubMed]
  154. Gurney, M.; Muralidhar, G.; Linton, P.-J. Autophagy in the Immune System. Autophagy Health Dis. 2013, 30, 41–55. [Google Scholar] [CrossRef]
  155. Jung, C.H.; Ro, S.-H.; Cao, J.; Otto, N.M.; Kim, D.-H. mTOR regulation of autophagy. FEBS Lett. 2010, 584, 1287–1295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Tattoli, I.; Sorbara, M.T.; Yang, C.; Tooze, S.A.; Philpott, D.J.; Girardin, S.E. Listeria phospholipases subvert host autophagic defenses by stalling pre-autophagosomal structures. EMBO J. 2013, 32, 3066–3078. [Google Scholar] [CrossRef] [Green Version]
  157. Abdel-Nour, M.; Tsalikis, J.; Kleinman, D.; Girardin, S.E. The emerging role of mTOR signalling in antibacterial immunity. Immunol. Cell Biol. 2014, 92, 346–353. [Google Scholar] [CrossRef]
  158. Choy, A.; Dancourt, J.; Mugo, B.; O’Connor, T.J.; Isberg, R.R.; Melia, T.J.; Roy, C.R. The Legionella effector RavZ inhibits host autophagy through irreversible Atg8 deconjugation. Science 2012, 338, 1072–1076. [Google Scholar] [CrossRef] [Green Version]
  159. Casanova, J.E. Bacterial Autophagy: Offense and Defense at the Host–Pathogen Interface. Cell. Mol. Gastroenterol. Hepatol. 2017, 4, 237–243. [Google Scholar] [CrossRef] [Green Version]
  160. Le Sage, V.; Cinti, A.; Amorim, R.; Mouland, A.J. Adapting the Stress Response: Viral Subversion of the mTOR Signaling Pathway. Viruses 2016, 8. [Google Scholar] [CrossRef]
  161. Buchkovich, N.J.; Yu, Y.; Zampieri, C.A.; Alwine, J.C. The TORrid affairs of viruses: Effects of mammalian DNA viruses on the PI3K–Akt–mTOR signalling pathway. Nat. Rev. Microbiol. 2008, 6, 266–275. [Google Scholar] [CrossRef] [Green Version]
  162. Li, N.; Tang, B.; Jia, Y.P.; Zhu, P.; Zhuang, Y.; Fang, Y.; Li, Q.; Wang, K.; Zhang, W.J.; Guo, G.; et al. Helicobacter pylori CagA Protein Negatively Regulates Autophagy and Promotes Inflammatory Response via c-Met-PI3K/Akt-mTOR Signaling Pathway. Front. Cell. Infect. Microbiol. 2017, 7. [Google Scholar] [CrossRef] [PubMed]
  163. McLean, J.E.; Wudzinska, A.; Datan, E.; Quaglino, D.; Zakeri, Z. Flavivirus NS4A-induced autophagy protects cells against death and enhances virus replication. J. Biol. Chem. 2011, 286, 22147–22159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Liang, Q.; Luo, Z.; Zeng, J.; Chen, W.; Foo, S.S.; Lee, S.A.; Ge, J.; Wang, S.; Goldman, S.A.; Zlokovic, B.V.; et al. Zika Virus NS4A and NS4B Proteins Deregulate Akt-mTOR Signaling in Human Fetal Neural Stem Cells to Inhibit Neurogenesis and Induce Autophagy. Cell Stem Cell 2016, 19, 663–671. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Zhou, Y.; Geng, P.; Liu, Y.; Wu, J.; Qiao, H.; Xie, Y.; Yin, N.; Chen, L.; Lin, X.; Liu, Y.; et al. Rotavirus-encoded virus-like small RNA triggers autophagy by targeting IGF1R via the PI3K/Akt/mTOR pathway. Biochim. Biophys. Acta Mol. Basis Dis. 2018, 1864, 60–68. [Google Scholar] [CrossRef] [PubMed]
  166. Shi, Y.; He, X.; Zhu, G.; Tu, H.; Liu, Z.; Li, W.; Han, S.; Yin, J.; Peng, B.; Liu, W. Coxsackievirus A16 elicits incomplete autophagy involving the mTOR and ERK pathways. PLoS ONE 2015, 10, e0122109. [Google Scholar] [CrossRef] [Green Version]
  167. Wang, Y.; Weiss, L.M.; Orlofsky, A. Host Cell Autophagy Is Induced by Toxoplasma gondii and Contributes to Parasite Growth. J. Biol. Chem. 2009, 284, 1694. [Google Scholar] [CrossRef] [Green Version]
  168. Wang, Y.; Weiss, L.M.; Orlofsky, A. Intracellular parasitism with Toxoplasma gondii stimulates mTOR-dependent host cell growth despite impaired signaling to S6K1 and 4E-BP1. Cell Microbiol. 2009, 11, 983. [Google Scholar] [CrossRef] [Green Version]
  169. Thomas, S.A.; Nandan, D.; Kass, J.; Reiner, N.E. Countervailing, time-dependent effects on host autophagy promotes intracellular survival of Leishmania. J. Biol. Chem. 2018, 293, 2617–2630. [Google Scholar] [CrossRef] [Green Version]
  170. Blanchet, F.P.; Moris, A.; Nikolic, D.S.; Lehmann, M.; Cardinaud, S.; Stalder, R.; Garcia, E.; Dinkins, C.; Leuba, F.; Wu, L.; et al. Human immunodeficiency virus-1 inhibition of immunoamphisomes in dendritic cells impairs early innate and adaptive immune responses. Immunity 2010, 32, 654–669. [Google Scholar] [CrossRef] [Green Version]
  171. Nardacci, R.; Ciccosanti, F.; Marsella, C.; Ippolito, G.; Piacentini, M.; Fimia, G.M. Role of autophagy in HIV infection and pathogenesis. J. Intern. Med. 2017, 281, 422–432. [Google Scholar] [CrossRef]
  172. Alwine, J.C. Modulation of host cell stress responses by human cytomegalovirus. Curr. Top. Microbiol. Immunol. 2008, 325, 263–279. [Google Scholar] [CrossRef] [PubMed]
  173. Shives, K.D.; Massey, A.R.; May, N.A.; Morrison, T.E.; Beckham, J.D. 4EBP-Dependent Signaling Supports West Nile Virus Growth and Protein Expression. Viruses 2016, 8, 287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Joubert, P.-E.; Stapleford, K.; Guivel-Benhassine, F.; Vignuzzi, M.; Schwartz, O.; Albert, M.L. Inhibition of mTORC1 Enhances the Translation of Chikungunya Proteins via the Activation of the MnK/eIF4E Pathway. PLoS Pathog. 2015, 11, e1005091. [Google Scholar] [CrossRef] [PubMed]
  175. Shives, K.D.; Beatman, E.L.; Chamanian, M.; O’Brien, C.; Hobson-Peters, J.; Beckham, J.D. West nile virus-induced activation of mammalian target of rapamycin complex 1 supports viral growth and viral protein expression. J. Virol. 2014, 88, 9458–9471. [Google Scholar] [CrossRef] [Green Version]
  176. De Leon, J.A.; Qiu, J.; Nicolai, C.J.; Counihan, J.L.; Barry, K.C.; Xu, L.; Lawrence, R.E.; Castellano, B.M.; Zoncu, R.; Nomura, D.K.; et al. Positive and Negative Regulation of the Master Metabolic Regulator mTORC1 by Two Families of Legionella pneumophila Effectors. Cell Rep. 2017, 21, 2031–2038. [Google Scholar] [CrossRef] [Green Version]
  177. Leroux, L.P.; Lorent, J.; Graber, T.E.; Chaparro, V.; Masvidal, L.; Aguirre, M.; Fonseca, B.D.; van Kempen, L.C.; Alain, T.; Larsson, O.; et al. The Protozoan Parasite Toxoplasma gondii Selectively Reprograms the Host Cell Translatome. Infect. Immun. 2018, 86. [Google Scholar] [CrossRef] [Green Version]
  178. Pernas, L.; Adomako-Ankomah, Y.; Shastri, A.J.; Ewald, S.E.; Treeck, M.; Boyle, J.P.; Boothroyd, J.C. Toxoplasma effector MAF1 mediates recruitment of host mitochondria and impacts the host response. PLoS Biol. 2014, 12, e1001845. [Google Scholar] [CrossRef] [Green Version]
  179. Suzutani, T.; Nagamine, M.; Shibaki, T.; Ogasawara, M.; Yoshida, I.; Daikoku, T.; Nishiyama, Y.; Azuma, M. The role of the UL41 gene of herpes simplex virus type 1 in evasion of non-specific host defence mechanisms during primary infection. J. Gen. Virol. 2000, 81, 1763–1771. [Google Scholar] [CrossRef]
  180. Shi, G.; Ozog, S.; Torbett, B.E.; Compton, A.A. mTOR inhibitors lower an intrinsic barrier to virus infection mediated by IFITM3. Proc. Natl. Acad. Sci. USA 2018, 115, E10069–E10078. [Google Scholar] [CrossRef] [Green Version]
  181. Beretta, L.; Gingras, A.C.; Svitkin, Y.V.; Hall, M.N.; Sonenberg, N. Rapamycin blocks the phosphorylation of 4E-BP1 and inhibits cap-dependent initiation of translation. EMBO J. 1996, 15, 658–664. [Google Scholar] [CrossRef]
  182. Li, Y.; Fang, L.; Zhou, Y.; Tao, R.; Wang, D.; Xiao, S. Porcine Reproductive and Respiratory Syndrome Virus Infection Induces both eIF2α Phosphorylation-Dependent and -Independent Host Translation Shutoff. J. Virol. 2018, 92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Luo, Q.; Zhang, L.; Wei, F.; Fang, Q.; Bao, F.; Mi, S.; Li, N.; Wang, C.; Liu, Y.; Tu, C. mTORC1 Negatively Regulates the Replication of Classical Swine Fever Virus Through Autophagy and IRES-Dependent Translation. iScience 2018, 3, 87–101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Katholnig, K.; Linke, M.; Pham, H.; Hengstschläger, M.; Weichhart, T. Immune responses of macrophages and dendritic cells regulated by mTOR signalling. Biochem. Soc. Trans. 2013, 41, 927–933. [Google Scholar] [CrossRef] [PubMed]
  185. Peres, A.G.; Stegen, C.; Li, J.; Xu, A.Q.; Levast, B.; Surette, M.G.; Cousineau, B.; Desrosiers, M.; Madrenas, J. Uncoupling of pro- and anti-inflammatory properties of Staphylococcus aureus. Infect. Immun. 2015, 83, 1587–1597. [Google Scholar] [CrossRef] [Green Version]
  186. Cheekatla, S.S.; Aggarwal, A.; Naik, S. mTOR signaling pathway regulates the IL-12/IL-10 axis in Leishmania donovani infection. Med. Microbiol. Immunol. 2012, 201, 37–46. [Google Scholar] [CrossRef]
  187. Foldenauer, M.E.B.; McClellan, S.A.; Berger, E.A.; Hazlett, L.D. Mammalian target of rapamycin regulates IL-10 and resistance to Pseudomonas aeruginosa corneal infection. J. Immunol. 2013, 190, 5649–5658. [Google Scholar] [CrossRef] [Green Version]
  188. Araki, K.; Turner, A.P.; Shaffer, V.O.; Gangappa, S.; Keller, S.A.; Bachmann, M.F.; Larsen, C.P.; Ahmed, R. mTOR regulates memory CD8 T-cell differentiation. Nature 2009, 460, 108–112. [Google Scholar] [CrossRef] [Green Version]
  189. Lai, Z.W.; Kelly, R.; Winans, T.; Marchena, I.; Shadakshari, A.; Yu, J.; Dawood, M.; Garcia, R.; Tily, H.; Francis, L.; et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: A single-arm, open-label, phase 1/2 trial. Lancet 2018, 391, 1186–1196. [Google Scholar] [CrossRef]
  190. Fajgenbaum, D.C.; Langan, R.A.; Japp, A.S.; Partridge, H.L.; Pierson, S.K.; Singh, A.; Arenas, D.J.; Ruth, J.R.; Nabel, C.S.; Stone, K.; et al. Identifying and targeting pathogenic PI3K/AKT/mTOR signaling in IL-6-blockade-refractory idiopathic multicentric Castleman disease. J. Clin. Investig. 2019, 130, 4451–4463. [Google Scholar] [CrossRef]
Figure 1. Strategies that pathogens use to directly target mTOR.
Figure 1. Strategies that pathogens use to directly target mTOR.
Cells 09 00161 g001
Figure 2. Overview of metabolic targets through which pathogens are known to modulate DC and macrophage functions. In (AD), the key strategies through which different pathogens modulate (A) amino acid metabolism, (B) lipid metabolism, (C) carbohydrate metabolism and (D) other mTOR-controlled processes in DCs and macrophages to promote immune evasion are indicated Blue = pathogens involved; Red = inhibition by pathogens; Green = stimulation by pathogens..
Figure 2. Overview of metabolic targets through which pathogens are known to modulate DC and macrophage functions. In (AD), the key strategies through which different pathogens modulate (A) amino acid metabolism, (B) lipid metabolism, (C) carbohydrate metabolism and (D) other mTOR-controlled processes in DCs and macrophages to promote immune evasion are indicated Blue = pathogens involved; Red = inhibition by pathogens; Green = stimulation by pathogens..
Cells 09 00161 g002
Figure 3. L-arginine is metabolized by iNOS and Arginase, creating competition between pathogen clearance and survival by modulating immune responses through the production of NO and polyamines. The dashed arrow represents a process only observed in trypanosomatid parasites.
Figure 3. L-arginine is metabolized by iNOS and Arginase, creating competition between pathogen clearance and survival by modulating immune responses through the production of NO and polyamines. The dashed arrow represents a process only observed in trypanosomatid parasites.
Cells 09 00161 g003
Table 1. Overview of the effects induced by the pathogens described in this review.
Table 1. Overview of the effects induced by the pathogens described in this review.
Direct Targeting mTOR
EffectPathogenReferences
Induction of autophagy by secreting a rapamycin homolog and inhibiting mTOR leading to impaired human monocyte-derived DC functionB. malayi[22]
Cleavage of mTOR via the protease GP63 in B10R macrophages leading to decreased type I IFN production and expression of iNOSL. major[23]
Relocating mTOR to maintain mTORC1 activation in HFFs and U373-MG cellsHCMV[24,25]
Sequestering of raptor and rictor in PMA differentiated THP-1 cells leading to mTOR relocalization and inhibition of cGAS-STING activation and induction of IRGsPoxviruses (including VacV)[28,29]
Direct interaction with mTORC2 to modulate macrophage phenotype and migration (in this case PMA differentiated THP-1 cells)HIV-1[30]
Amino Acid Metabolism
EffectPathogenReferences
Synthesize tryptophan thereby counteracting IDO depletion of tryptophanM. tuberculosis, F. Tularensis, C. trachomatis[9,41]
Expresses a IL-10 homolog (cmvIL-10) that induces IDO in human monocyte-derived DCsHCMV[47,48]
Induction of IDO in langerin negative dermal dendritic cellsHPV[49]
Induction of host arginases in mouse macrophagesTrypanosoma sps, Leishmania sps, T. gondii, S. typhimurium, H. pylori, S. pneumonia, C. albicans, M. tuberculosis and S. mansoni[65,66,67,68,69,70]
Expression of pathogen arginaseLeishmania sps, Plasmodium sps, C. albicans, S. mansoni and H. pylori[65,71,72,73,74,75,76]
Arginase 1 induction in CD33+ PBMCs mediates L-arginine depletion leading to mTOR inhibition and decreased IFNy production in co-cultured NK cellsHCV[79]
Depletion of L-arginine by expression of arginine deiminase modulates cytokine production and phenotype of human monocyte-derived DCs via the inhibition of mTORC1G. duodenalis[80]
Asparaginase expression leading to asparagine depletion and dampening of immune responses in T cells and macrophages (ANA-1 and RAW264.7 cells)Expression: multiple pathogens including H. pylori and S. typhimurium
Dampening immune response: S. typhimurium, Erwinia asparaginase
[88,89,90,91,92]
[90,93]
Lipid Metabolism
EffectPathogenReferences
Induction of LD formation and PGE2 synthesis for successful replication and modulation of the immune (reduction of antigen-stimulated lymphocyte replication, reduction of killing ability infected cells)T. cruzi, M. leprae, HMCV, HIV[112,113,114,115,116,117,118]
Active stimulation of LD formation via SseJS. Typhimurium[104]
Accumulation of LDs in pathogen vacuolesC. pneumoniae, T. cruzi[104]
Upregulation COX-2 and PGE2 synthesis in RAW264.7 cells and murine BMDMs and BMDCsS. typhimurium[60,119]
Production of PGE2 and accumulation of LDs increasing virulenceHuman pathogenic fungi[120,121,122]
Induction of LDs to enhance replication efficiency and the assembly of nascent virionsViruses, including HCV and Rotaviruses[104,123]
mTOR inhibition to induce autophagy of LDs to enhance replication efficiencyFlaviviruses, including Dengue[124,125]
Promoting mTORC1-dependent TAG accumulation in human macrophages that contributes to LD formationM. tuberculosis, T. gondii[102,126]
Inducing LDs and PGE2 synthesis via autophagy in BALB/c macrophagesL. amazonensis[129]
Rapamycin induces LD formationS. cerevisiae[130]
Reducing cholesterol levels at the plasma membrane of Vero cells which disrupts Jak-STAT signalingWNV[135]
Altered CD40 signalosome in BALB/c derived peritoneal macrophages by depleting cholesterol leading to IL-10 productionL. major[136]
Carbohydrate Metabolism
EffectPathogenReferences
Prevents glycolytic shift in primary macrophages by downregulating HIF1αF. Tularensis[143]
Depleting intracellular glucose to inhibit glycolysis in murine BMDMsS. Typhimurium[144,145]
Depleting extracellular glucose leading to the death of restimulated macrophages (murine BMDMs and PMA differentiated THP-1 cells)C. albicans[146]
Promotion of glucose oxidation in murine BMDMs through activation of SIRT1, LKB1 and AMPK increasing survival and proliferationL. infantum[147]
Promotes glycolysis (Warburg like metabolism) in human monocyte-derived macrophagesL. pneumophila[149,150,151,152]
Autophagy
EffectPathogenReferences
Inhibition by stimulating the PI3K-Akt-mTOR pathwayViruses in general[160,161]
Targeting AMPK, SIRT1 and LKB1 for degradation in murine BMDMsS. typhimurium[98]
Activation of mTOR (via CagA) and inhibition of autophagyH. pylori[162]
Induction of autophagy to increase sites of replication, to liberate nutrients and/or to protect host cell deathFlaviviruses (including Zika), coxsackievirus[163,164,165,166]
Induction of autophagy independent of mTOR which contributes to parasite growth.T. gondii[167,168]
Inhibition of autophagy by stimulating mTOR early in infection in PMA differentiated THP-1 cells. Induction of autophagy during later stages of infection, regulated independent of mTOR.L. donovani[169]
Inhibiting autophagy by stimulating mTOR to decrease cross-presentation and enhance spreading of infection.HIV-1[170,171]
Host Translation
EffectPathogenReferences
Activates mTORC1 to promote translation of proteins for mitochondrial biogenesis and function, possibly to modulate the innate immune response (in BMDMs and PECs)T. gondii[177,178]
Cleavage of mTOR via the protease GP63 in B10R macrophages to prevent IFN type I production and iNOS translationL. major[23]
Mediates shut off of host translation which leads to a decreased translation of innate cytokines in U937 cellsHSV-1[179]
Shut off of host translation to favor replication of own genome and to downregulate IFITM proteinsViruses relying on IRES-dependent translation[23,161,180,181,182,183]
Innate Cytokines
EffectPathogenReferences
Cell wall moieties induce the production of IL-10 via TLR2 and mTOR signaling in human PBMCsS. aureus[185]
TLR2 and mTOR dependent IL-10 production in PMA differentiated THP-1 cellsL. donovani[186]
Rapamycin decreases IL-10 production in cornea of infected Balb/c miceP. aeruginosa[187]

Share and Cite

MDPI and ACS Style

Nouwen, L.V.; Everts, B. Pathogens MenTORing Macrophages and Dendritic Cells: Manipulation of mTOR and Cellular Metabolism to Promote Immune Escape. Cells 2020, 9, 161. https://doi.org/10.3390/cells9010161

AMA Style

Nouwen LV, Everts B. Pathogens MenTORing Macrophages and Dendritic Cells: Manipulation of mTOR and Cellular Metabolism to Promote Immune Escape. Cells. 2020; 9(1):161. https://doi.org/10.3390/cells9010161

Chicago/Turabian Style

Nouwen, Lonneke V., and Bart Everts. 2020. "Pathogens MenTORing Macrophages and Dendritic Cells: Manipulation of mTOR and Cellular Metabolism to Promote Immune Escape" Cells 9, no. 1: 161. https://doi.org/10.3390/cells9010161

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop