Abstract
Metabolic flexibility is key for the function of myeloid cells. Arginine metabolism is integral to the regulation of myeloid cell responses. Nitric oxide (NO) production from arginine is vital for the antimicrobial and pro-inflammatory responses. Conversely, the arginase 1 (ARG1)-dependent switch between the branch of NO production and polyamine synthesis downregulates inflammation and promotes recovery of tissue homeostasis. Creatine metabolism is key for energy supply and proline metabolism is required for collagen synthesis. Myeloid ARG1 also regulates extracellular arginine availability and T cell responses in parasitic diseases and cancer. Cancer, surgery, sepsis and persistent inflammation in chronic inflammatory diseases, such as neuroinflammatory diseases or arthritis, are associated with dysregulation of arginine metabolism in myeloid cells. Here, we review current knowledge on arginine metabolism in different myeloid cell types, such as macrophages, neutrophils, microglia, osteoclasts, tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs) and myeloid-derived suppressor cells (MDSCs). A deeper understanding of the function of arginine metabolism in myeloid cells will improve our knowledge on the pathology of several diseases and may set the platform for novel therapeutic applications.
Keywords: Arginine metabolism, Myeloid cells, Infection, Inflammation, Cancer, Sepsis-induced immune paralysis, Surgery
Introduction
Myeloid cells have multiple functions including elimination of microbes, antigen presentation, cytokine secretion, T cell activation, resolution of inflammation and tissue repair [1]. The functional versatility of myeloid cells is enabled by their substantial metabolic plasticity. Particularly arginine metabolism is highly flexible and plays a key role in the modulation of immune function [2]. Almost 100 years after the discovery of the urea cycle in 1932 [3], the importance of arginine metabolism in the regulation of innate and adaptive immune responses is still an area of interest. Here, we review current knowledge on arginine metabolism and its role in myeloid cell function in the context of infection, sepsis, chronic inflammatory disease, cancer and surgery.
Myeloid cells
The myeloid cell compartment constitutes a major pillar of innate immunity. It comprises granulocytes, monocytes, dendritic cells, tissue macrophages and mast cells [4]. Granulocytes, monocytes, and dendritic cells derive from bone marrow hematopoietic stem and progenitor cells, while most tissue-resident macrophages, such as microglia, Kupffer cells or Langerhans cells, originate from embryonic precursors of the yolk sac or the fetal liver [1]. Tissue-resident myeloid cells have long life spans and perform homeostatic functions [1]. In contrast, bone marrow-derived myeloid cells are mobilized upon trauma or infection [1, 5]. Neutrophils are the first line of defense against pathogens. They are rapidly recruited to infected sites, and eliminate microbes through phagocytosis, antimicrobial substances, reactive oxygen species (ROS) and formation of neutrophil extracellular traps (NETs) [6, 7]. Tissue-resident macrophages are replenished by bone marrow monocyte-derived macrophages during life [4]. Upon infection or trauma, monocytes infiltrate affected tissues and transform to inflammatory macrophages, which contribute to elimination of pathogens [4]. Moreover, macrophages are central to mediating resolution of inflammation by clearing dead cells, which triggers restoration of tissue integrity [8, 9]. Dendritic cells play a unique role in bridging the innate and adaptive immune responses [10]. They are localized at sites of possible pathogen entry in order to rapidly detect and phagocytose microbes, and to stimulate T cells through antigen presentation, thereby priming the adaptive immune response [10]. Eosinophils respond to helminth infection and allergen exposure by releasing positively charged proteins stored in cytoplasmic granules [5]. Basophils are the rarest and least studied granulocytes. They are implicated in allergic reactions, immunity against parasitic infections, autoimmunity and tissue restoration [11]. Metabolic circuits regulating immune responses, including arginine metabolism, are mostly studied in monocytes / macrophages due to their relative abundance and easy isolation and culture, while in some myeloid cell types, such as eosinophils or mast cells, knowledge on cell metabolic regulation is scarce.
Arginine metabolism in myeloid cells
Arginine metabolism presents substantial differences between different cell types and species. In humans, arginine is synthesized in the intestinal-renal axis. Citrulline is produced in enterocytes from glutamate or proline, and taken up by the kidney, where it is converted to arginine via argininosuccinate through the function of argininosuccinate synthase (ASS1) and argininosuccinate lyase (ASL) (Fig. 1a). However, under extreme conditions, such as critical illness or maximal growth during infancy, arginine becomes an essential amino acid and must be obtained through the diet [12]. Interestingly, macrophages acquire the ability to produce arginine from citrulline upon inflammatory activation [13, 14].
Fig. 1.
Arginine metabolism in myeloid cells. a. Arginine biosynthesis. Arginine is synthesized in humans in the intestinal-renal axis. Citrulline is produced by glutamate or proline in enterocytes and taken up by the kidney, where it is transformed to argininosuccinate through argininosuccinate synthase 1 (ASS1), which is subsequently metabolized to arginine by argininosuccinate lyase (ASL) [2]. b. Arginine metabolism in myeloid cells. Arginine is transported into the cells via CAT-2B and metabolized to ornithine through the function of arginases 1 or 2 (ARG1, ARG2). Ornithine is then converted to putrescine through the function of ornithine decarboxylase 1 (ODC1) [2, 15, 16]. Putrescine is further converted through spermidine synthase (SRM) to spermidine, which is metabolized by spermine synthase (SMS) to spermine [2]. Spermine can be converted back to spermidine via oxidation. Also, polyamines undergo acetylation through spermidine/spermine-N1-acetyltransferase (SAT1) to N1-acetyl-spermidine and N1-acetyl-spermine, which are then oxidized via polyamine oxidase (PAOX) to their respective precursors [19]. Upon inflammation, arginine is diverted towards the production of citrulline and nitric oxide (NO) through inducible nitric oxide synthase (iNOS) [2, 17]. Citrulline can be also converted to ornithine by laccase domain-containing 1 (LACC1), the expression of which increases under inflammatory conditions [20]. Moreover, ornithine can be metabolized to proline through L-glutamate 5-semialdehyde and 1-pyrroline 5-carboxylate (P5C) [2]. Furthermore, arginine can be metabolized to creatine through the production of guanidinoacetate and the action of glycine amidinotransferase (GATM) and guanidinoacetate N-methyltransferase (GAMT) [2]. The figure was created with BioRender.com
Circulating arginine from the diet and/or endogenous synthesis is transported into the cells mostly by the solute carrier 7 family (SLC7) members cationic amino acid transporters CAT-1 and CAT-2 [15, 16]. Macrophages mainly express CAT-2B (encoded by Slc7a2), which is induced upon classical or alternative activation, and exhibits high affinity for arginine [15, 16]. The importance of arginine metabolism in macrophage immune responses is demonstrated in Slc7a2−/− mice, which have reduced antimicrobial resistance [16]. Intracellular arginine is either used for protein synthesis or metabolized [17, 18]. It can be converted by arginases 1 or 2 (ARG1, ARG2) to ornithine, which is then metabolized via ornithine decarboxylase 1 (ODC1) to putrescine. The latter is a substrate of spermidine synthase (SRM) producing spermidine, which is further converted to spermine by spermine synthase (SMS). Spermine can be converted back to spermidine via oxidation. Furthermore, polyamines are acetylated by spermidine/spermine-N1-acetyltransferase (SAT1) to N1-acetyl-spermidine and N1-acetyl-spermine, which can be oxidized by polyamine oxidase (PAOX) to regenerate their precursor molecules [2, 18, 19]. Ornithine is further metabolized to proline through L-glutamate 5-semialdehyde and 1-pyrroline 5-carboxylate (P5C) [2, 18]. Arginine is also metabolized via L-Arginine:glycine amidinotransferase (GATM) using glycine to ornithine and guanidinoacetate, the latter being further metabolized to creatine through the action of guanidinoacetate N-methyltransferase (GAMT) [2] (Fig. 1b). Polyamine metabolism, proline synthesis and creatine synthesis contribute to tissue homeostasis or promote tissue recovery after injury or infection.
Trauma and pathogens trigger shunting of arginine to production of citrulline and nitric oxide (NO) through the function of the inducible nitric oxide synthase (iNOS) [2, 13, 17] (Fig. 2a). In prolonged inflammation, due to arginine shortage, macrophages acquire the ability of arginine biosynthesis by converting citrulline via ASS1 to argininosuccinate, which is further metabolized via ASL to arginine [13]. Finally, citrulline can be also converted to ornithine by laccase domain-containing 1 (LACC1), the expression of which increases upon inflammation in macrophages [20] (Fig. 1b).
Fig. 2.
The function of arginine metabolism in macrophage inflammatory responses. a. Changes in arginine metabolism in the course of inflammation. In the early inflammatory response, iNOS is induced [2]. ASS1 is upregulated to sustain iNOS function [13]. Later, ARG1 is upregulated to limit iNOS function [17, 56] and shift arginine metabolism toward production of polyamines, which may contribute to termination of inflammation [2, 100, 101]. b. Pathways of arginine metabolism regulating macrophage immune responses. Bacteria, LPS and IFNγ induce iNOS, resulting in production of NO, which is toxic for bacteria [13, 37]. NO also disrupts oxidative phosphorylation [44] and inhibits inflammasome activation in macrophages [47, 48]. Induction of the ASS1- and ASL-mediated argininosuccinate shunt sustains the function of iNOS through production of arginine [13]. ARG1 competes with iNOS for arginine, thereby inhibiting its function [17, 38, 57, 62]. ARG1 expression is induced by different pro- and anti-inflammatory stimuli, such as by LPS, IL-10, IL-4, TGF-β and helminth antibodies [56, 57, 65, 67, 69, 72]. ARG1 is mostly dependent on extracellular arginine, thereby regulating arginine availability to other cells, such as T cells [17, 18, 76]. Ornithine, the product of ARG1, is the precursor of polyamines, which inhibit inflammation and contribute to tissue remodeling [2, 92, 100, 101, 134]. The figure was created with BioRender.com
Numerous studies have addressed the roles of iNOS and ARG1 in macrophage polarization [2, 17]. The latter refers to the differential response of macrophages to in vitro stimulation with the Toll-Like Receptor 4 (TLR4) agonist lipopolysaccharide (LPS) and interferon-γ (IFNγ) or the type 2 immunity-related cytokines Interleukin 4 (IL-4) and IL-13 [21]. LPS or IFNγ induce classical activation hallmarked by production of inflammatory cytokines, ROS, a strong induction of iNOS expression and shift of glucose metabolism to glycolysis (Warburg-like effect) [21]. IL-4 induces alternative activation, characterized by elevated expression of scavenger receptors, matrix remodeling factors, and a strong induction of ARG1 expression [21]. Classical activation is considered to represent the pro-inflammatory response of macrophages to infection or trauma, while alternative activation is generally associated with type 2 immune responses (response to parasite infections, allergies, asthma), and reparative, wound healing responses [21]. However, in vitro macrophage polarization does not reflect the in vivo complexity, which is characterized by location-, origin- and function-related heterogeneity of macrophages driven by exposure to multiple local and systemic signals [21, 22]. Despite the limited relevance of in vitro macrophage polarization to the in vivo reality and pathology of inflammatory disease, it has been a useful tool in order to dissect the role of cell metabolism, and arginine metabolism in particular, in macrophages.
Myeloid arginine metabolism in healthy conditions versus disease
Resident myeloid cells, including microglia (macrophage-like cells of the brain), Kupffer cells (macrophages of the liver), lung macrophages and Langerhans cells (skin macrophages), preserve tissue homeostasis by removing dead cells and debris and maintaining tissue architecture through extracellular matrix deposition [1]. In healthy conditions, polyamine, proline and creatine metabolism are sustained at relatively low levels, while citrulline is not produced. However, upon infection, trauma or disease, all pathways of arginine metabolism, i.e. citrulline, polyamine, creatine and proline production, are upregulated in an orchestrated time-dependent manner to support the antimicrobial function of myeloid cells, supply energy, facilitate phagocytosis, and promote tissue recovery.
The role of iNOS in macrophage inflammatory responses
Bacterial infection or stimulation with LPS, IFNγ and/or cytokines triggers iNOS expression in human and mouse macrophages [13, 23–26]. In contrast to the relatively short promoter region of the murine Nos2 gene [27, 28], the human NOS2 gene is regulated by a classical enhancer region, located between 5 and 6 kb upstream of the transcription site, containing functional binding sites for nuclear factor kappa B (NFκB) and Signal transducer and activator of transcription 1 (STAT1) [29, 30]. Regulation of iNOS mostly occurs at the transcriptional level, but post-transcriptional modifications may also alter its expression [31].
In murine macrophages, iNOS generates large amounts (at the micromolar range) of NO, while production of NO in human macrophages is under debate [13, 23–25, 32, 33]. Unlike inflammatory macrophages, neutrophils produce little NO [34]. NO is toxic and can directly kill bacteria but also suppresses neutrophil recruitment [13, 35]. NOS deficient mice are highly susceptible to Mycobacterium tuberculosis (Mtb) infection and develop severe lung and spleen pathology [24, 36]. However, NO is not always sufficient to restrain bacterial infection [24, 25]. For instance, NO produced by human macrophages does not sufficiently limit mycobacterial replication or survival [25]. Aside from its function in elimination of bacteria, NO exerts numerous functions within macrophages and their surrounding microenvironment [37].
As inflammation progresses, consumption of arginine used for NO synthesis in macrophages as well as arginine consumption by T cells, leads to arginine shortage. Arginine availability is required for iNOS expression and function [38]. Under conditions of limited arginine availability, macrophages regenerate arginine to sustain NOS production throughout the course of the inflammatory response [13]. Citrulline, which is exported from macrophages at early stages of inflammation, is imported at later stages to sustain NO production via activating ASS1/ASL-dependent arginine synthesis [13]. The importance of arginine regeneration in macrophages is evidenced by the fact that mice bearing hematopoietic cell-specific ASS1 deficiency are more sensitive to Mtb infection compared to wild-type control mice [13]. Accordingly, myeloid deficiency of ASS1 or ASL increases the Mycobacterium bovis bacillus, Calmette-Guerin and Mtb infection burden [39]. The aspartate-argininosuccinate shunt also affects the tricarboxylic acid (TCA) cycle flux through the production of fumarate, which accumulates in proinflammatory macrophages [40].
In macrophages, NO disrupts the function of the electron transport chain (ETC) through inhibition of the catalytic NADH binding module of complex I and complex IV (cytochrome c oxidase) [23, 41–44]. Moreover, NO regulates TCA cycle function by decreasing pyruvate dehydrogenase (PDH) and aconitase (ACO2) activity, reducing succinate and itaconate levels and increasing citrate [23, 44]. NO-mediated reduction of PDH and ACO2 enzymatic activities leads to a lower NADH/NAD+ ratio, which in turn promotes the conversion of complex I to its inactive form due to substrate deprivation [44]. Furthermore, NO may cause cysteine nitrosylation of numerous proteins, including enzymes involved in the TCA cycle, oxidative phosphorylation, glycolysis, gluconeogenesis and fatty acid metabolism; NO may thus regulate several metabolic reactions [45]. Consequently, NO downregulates mitochondrial respiration in inflammatory macrophages and mediates reprograming of macrophage cell metabolism from oxidative phosphorylation to glycolysis [14, 44, 46]. Accordingly, inhibition of arginine regeneration through the aspartate-argininosuccinate shunt inhibits NO production and inflammatory activation of macrophages and enhances oxidative phosphorylation [14] (Fig. 2b). Moreover, iNOS inhibition not only increases mitochondrial respiration in LPS + IFNγ treated macrophages, but also enhances M2-like polarization after a subsequent IL-4 treatment [43]. An NO-driven metabolic switch towards glycolysis also occurs in inflammatory dendritic cells [46]. Nevertheless, NO deficiency does not hinder upregulation of glycolysis in inflammatory macrophages suggesting implication of NO-independent mechanisms in the glycolytic shift of the cells [23].
Genetic deletion or pharmacological inhibition of iNOS suppress disease in a mouse psoriasis model due to reduced IL-1α production by skin macrophages [33]. However, iNOS deficient macrophages produce higher amounts of IL-1β and IL-6 than wild-type macrophages upon LPS stimulation, while production of TNF and IL-10 is not affected [23, 35, 44]. NO-mediated S-nitrosylation of NLRP3 prevents inflammasome activation thereby reducing IL-1β production [47]. NO may also synergize with itaconate to block NLRP3 inflammasome activation [48]. Through repression of IL-1 signaling, NO inhibits neutrophil recruitment after Mtb infection [35, 47]. Moreover, NO inhibits NF-κB activity thereby preventing overt inflammation upon Mtb infection [49] (Fig. 2b). By contrast, NO was proposed to stabilize HIF-1α [49], which in turn promotes IL-1β production [50]. Collectively, these data demonstrate the complexity of NO-mediated regulation of inflammatory pathways in macrophages.
iNOS requires tetrahydrobiopterin (BH4) as a cofactor to produce NO [23]. Interestingly, iNOS and BH4 do not always share common effects. For instance, BH4 deficient macrophages have enhanced control of mycobacterial infection, while iNOS deficient macrophages present reduced responses against mycobacteria [24]. The differential impact of BH4 and iNOS deficiency on macrophage function following Mtb infection is reflected by distinct changes in the transcriptomic landscape of macrophages [24]. However, NOS enzymes can also function independently of BH4 and NO production through their reductase activity producing superoxide [51]. This is of particular relevance for human macrophages, which have low amounts of BH4 [24].
The role of arginases in myeloid cells
Inflammation comes with collateral damage of the host tissue. NO is toxic not only for bacteria, but also for host cells including macrophages themselves. As a means to restrain NO production, macrophages upregulate arginases [17, 52] (Fig. 2a, b). For instance, macrophages in granulomas in human tuberculosis express both, iNOS and ARG1 [53]. Arg1 expression is controlled by an enhancer region 3 kb upstream of the transcription site. PU.1, STAT6, and C/EBP bind at the enhancer region and promote Arg1 expression [54, 55]. Gram-negative bacteria or LPS trigger ARG1 expression in macrophages [56, 57]. Mycobacterium-infected macrophages produce IL-6, IL-10 and granulocyte colony-stimulating factor (G-CSF) that sustain ARG1 expression in an autocrine and paracrine manner via STAT3 signaling [52, 58]. In alternatively activated macrophages, STAT6 signaling promotes Arg1 expression [58]. Moreover, IL-10 can synergistically with LPS induce ARG1 expression in macrophages [59]. LPS also triggers negative feedback mechanisms, such as the expression of the transcription factor Fra-1 (Fos-related antigen 1), to suppress macrophage ARG1 expression [60].
Many studies suggest that ARG1 may compete with iNOS for arginine thereby limiting iNOS function [17, 38, 57, 61–63]. The temporal pattern of iNOS and ARG1 expression reflects their function during the course of inflammation: iNOS protein levels peak at 12 h, whereas ARG1 protein expression increases 24 h after induction of M1-like polarization [56] (Fig. 2a). Mechanistically, it was shown that glycolysis-derived lactate mediates lactylation of histone lysine residues in the Arg1 gene promoter thereby increasing ARG1 expression in M1-like macrophages [56]. Consequently, ARG1-deficient inflammatory macrophages produce higher amounts of NO, and myeloid ARG1 deficiency decreases the lung bacterial load during Mtb infection [52]. ARG1 preferentially uses arginine imported from the extracellular milieu rather than arginine synthesized in macrophages thereby regulating arginine availability to neighboring cells [13, 64] (Fig. 2b).
During parasite infection, activated T cells and eosinophils induce ARG1 expression in macrophages through production of IL-4 and IL-13 and activation of STAT6 [55, 65, 66] (Fig. 2b). However, other cytokines involved in resolution of inflammation and wound healing do also increase ARG1 expression: for instance, TGF-β increases ARG1 in macrophages and dendritic cells [67, 68] and IL-10 upregulates the expression of the IL-4 receptor α (IL-4Rα) thereby acting synergistically with IL-4 to promote ARG1 expression [69, 70]. In contrast, TNF inhibits IL-4-induced ARG1 expression and TNF deficiency in Leishmania major infected mice leads to ARG1 hyperexpression in myeloid cells, impaired NO production and increased lethality [62]. Moreover, neutrophils may promote ARG1 expression in macrophages upon parasite infection, leading to reduced helminth survival due to arginine depletion [71]. Helminth-specific antibodies can also induce ARG1 in macrophages that subsequently promotes the trapping of parasite larvae [72]. However, some parasites, such as Leishmania, induce ARG1-driven arginine metabolism in macrophages and dendritic cells in order to hijack myeloid polyamine production and thereby to support their own intracellular growth [73, 74]. Despite these evolutional adaptations serving parasite survival, ARG1-expressing macrophages are in general designed to fight parasite infection by walling off toxic worm eggs through collagen deposition enabling their disposal [17]. The pro-fibrotic effect of ARG1 may be due to promotion of proline synthesis, which is required for collagen production [75]. Yet, mice with myeloid ARG1 deficiency display increased collagen deposition upon Schistosoma mansoni (S. mansoni) infection [65], indicating the complex regulation of fibrosis in the setting of type 2 immunity.
L-arginine is unconditionally needed for T cell activation [76]. The expression of arginase isoforms in T cells appears to be context-dependent [18, 77]. For instance, while arginine is mainly metabolized by ARG2 in tumor-associated T cells [18], it is metabolized by ARG1 in lung CD4+ T cells upon influenza infection [77]. High arginase activity in myeloid cells results in extracellular arginine depletion causing T cell hyporesponsiveness [76, 78, 79] (Fig. 2b). Consequently, S. mansoni-infected mice with myeloid-specific ARG1 deficiency develop lethal non-resolving T cell-driven inflammation, suggesting that myeloid ARG1 controls Th2-dependent responses by depriving T cells from arginine [17, 65, 78, 80]. However, the effects of myeloid ARG1 are tissue-, disease- and context-dependent. For instance, macrophage ARG1 deficiency does not alter T cell-driven asthma pathogenesis in mice [81]. Moreover, inhibition of both ARG1 and iNOS exacerbates lung granuloma pathology and the bacterial burden in Mtb infected mice, possibly due to uncontrolled Th1 cell responses [82], while S. mansoni-induced ARG1 exacerbates lung inflammation upon co-infection with Mtb [83].
In contrast to cytosolic ARG1, the function of mitochondrial ARG2 in myeloid cells has received less attention [59]. Similarly to ARG1 expression, ARG2 expression is upregulated in human and mouse macrophages upon LPS treatment [59]. ARG2 increases mitochondrial respiration by promoting succinate dehydrogenase (complex II) activity, thereby downregulating succinate levels and consequently Hypoxia Inducible Factor 1 Subunit Alpha (HIF-1a) and IL-1β expression, and promoting an anti-inflammatory phenotype in macrophages [59, 84, 85]. However, proinflammatory effects of ARG2 for instance through increase of mitochondrial ROS, were also reported [86].
In conclusion, during tissue repair or parasite infection, whereby synthesis of extracellular matrix and overall anabolism is of paramount importance, it seems counterintuitive that macrophages upregulate arginine catabolism, while also increasing its uptake. This seeming paradox can be explained by the necessity of 1. arginine depletion by ARG1 to dampen iNOS activity and thereby avoid toxic NO overproduction, 2. deprivation of T cells from arginine thereby controlling T cell responses, and 3. production of arginine metabolites, such as polyamines and proline, which may contribute to regulation of inflammation and tissue remodeling (Fig. 2a, b).
The role of polyamines in regulation of inflammatory responses
Production of polyamines (putrescine, spermidine and spermine) starts with ornithine decarboxylation, i.e. the rate-limiting step of polyamine synthesis [19]. In inflammatory macrophages, ornithine derives from both arginine via ARG1 and ARG2 and citrulline via LACC1 [2, 20]. LACC1 deficient mice have exacerbated inflammation upon S. Typhimurium infection, which is reversed by ornithine treatment [20]. ODC1 myeloid deficiency increases gastric and colonic inflammation upon Helicobacter pylori and Citrobacter rodentium infection, respectively, while putrescine reverses these effects [87, 88]. Moreover, bacterial putrescine increases the abundance of anti-inflammatory macrophages and promotes mucosal homeostasis in the intestine of mice with dextran sulfate sodium (DSS)-induced colitis [89]. Similar to ARG1, the anti-inflammatory effect of ODC1 is associated with inhibition of iNOS [88, 90]. Mechanistically, a driver of polyamine synthesis in macrophages is mammalian target of rapamycin complex 1 (mTORC1) [91].
Polyamines dampen macrophage inflammatory responses and promote macrophage alternative activation through several mechanisms [20, 92, 93]. Spermidine can be transformed to hypusine by the deoxyhypusine synthase (DHPS). Hypusine is an uncommon amino acid present only in a single protein, the translation factor eukaryotic initiation factor 5A (eIF5A), and is crucial for eIF5A function. Hypusination of eIF5A increases the expression of mitochondrial proteins participating in the TCA cycle and oxidative phosphorylation, and promotes alternative activation of macrophages [94]. Spermidine also binds to mitochondrial trifunctional protein (MTP), promoting fatty acid oxidation, mitochondrial function and anti-tumoral activity of T cells [95]. Furthermore, polyamines promote autophagy, a homeostatic degradation and recycling mechanism of intracellular components, involved in cell survival and longevity [96, 97], which also impedes macrophage inflammatory responses and promotes alternative activation [98]. In addition, intracellular spermine inhibits K+ efflux-dependent NLRP3 inflammasome activation upon Edwardsiella piscicida infection [99].
Polyamine metabolism is also implicated in efferocytosis, i.e. phagocytosis of apoptotic cells, a process that contributes to resolution of inflammation. Polyamine levels increase in efferocytic macrophages, mainly via import [100, 101]. Putrescine promotes efferocytosis by increasing the expression of the guanosine triphosphate (GTP)-exchange factor (GEF) Dbl, which in turn activates Rac1 and actin polymerization enabling efferocytosis [101]. Consistently, ODC1 deficiency impairs continual efferocytosis and resolution of atherosclerosis, while putrescine restrains atherosclerosis [101]. Apoptotic cells release polyamines, which are uptaken by efferocytic macrophages, in parts via Rac1-dependent endocytic import [100, 102]. Received polyamines contribute to transcriptional reprogramming of macrophages leading to suppression of proinflammatory responses [100, 102]. Polyamines also promote wound healing and tissue remodeling [103–105]. In addition, ornithine released by arginase-expressing macrophages can be uptaken by fibroblasts and used for proline synthesis, which is required for collagen deposition [106].
The role of proline metabolism in myeloid cell responses
Proline metabolism is upregulated in pro-fibrotic states [107] and collagen synthesis depends on proline availability [108]. Nevertheless, the role of proline metabolism in macrophages is poorly understood. A recent study revealed that Prolidase or Peptidase D (PEPD), which mediates collagen turnover by degrading proline-containing dipeptides and is expressed in adipose tissue macrophages, regulates adipose tissue fibrosis. PEPD expression and activity is reduced in the adipose tissue of obese humans and mice and its pharmacological inhibition or genetic deletion promotes adipose tissue fibrosis and insulin resistance. Interestingly, PEPD is secreted by adipose tissue macrophages, and extracellular PEPD increases inflammation and fibrosis in the adipose tissue by activating Epidermal Growth Factor Receptor (EGFR) signaling. Reduced prolidase activity in the adipose tissue correlates with increased circulating PEPD levels, which serve as a biomarker of adipose tissue inflammation and fibrosis [109]. Furthermore, proline metabolism is implicated in trained innate immunity in lung macrophages. The expression of the rate-limiting enzyme of proline biosynthesis, glutaminase (GLS), is upregulated in trained macrophages in an allergic asthma mouse model with respiratory virus infection and early life allergen sensitization. Inhibition of proline synthesis suppresses trained macrophages and the development of allergic asthma, while proline supplementation recovers trained macrophages [110]. Finally, proline catabolism in Candida albicans cells was shown to be essential for its hyphal growth in phagosomes of engulfing macrophages, a process that C. albicans uses to escape killing by macrophages [111].
The role of creatine metabolism in macrophages
ATP-mediated energy supply is highly dependent on creatine metabolism [112]. Creatine is primarily synthesized in the liver and kidneys [113]. It is phosphorylated to creatine phosphate, which serves as a phosphate donor in the conversion of ADP to ATP, and thus supplies locally and acutely energy [114]. In macrophages, creatine is uptaken through the SLC6A8 transporter and phosphorylated by the cytosolic creatine kinase (CK-B) [115]. Exogenous creatine dampens IFNγ-triggered inflammatory responses, while it increases IL-4-induced responses, possibly through promoting ATP-dependent chromatin remodeling processes [115]. GATM expression is upregulated upon IL-4 treatment and is required for IL-4-induced responses [116]. In contrast, GATM expression is suppressed upon LPS treatment but does not affect proinflammatory macrophage responses [116]. In microglia, cyclocreatine, a membrane-transpassing creatine analogue, promotes microglial protective functions, suppresses autophagy and improves Aβ plaque-associated neurite destruction in a model of Alzheimer’s disease (AD) [117]. Mechanistically, CK-B is mobilized and interacts with F-actin at nascent phagosomes promoting actin polymerization and phagocytosis by increasing ATP supply in phagocytic macrophages [118]. This makes creatine metabolism crucial for the clearing function of macrophages.
The role of arginine metabolism in sepsis-induced immunodeficiency
Sepsis is a dysregulated systemic inflammatory and immune response to a disseminated infection that leads to immune paralysis and organ dysfunction [119]. It is manifested by a cytokine storm, i.e. derailed production of pro- and anti-inflammatory cytokines, accompanied or followed by immune paralysis with prominent lymphopenia and impaired T cell numbers and function [119]. The mechanisms underlying sepsis-induced immunossuppression are not entirely understood. Given the role of arginine in T cell function, reduced arginine availability during sepsis may contribute to immune dysfunction. Despite the increased arginine release from catabolic protein breakdown, arginine plasma concentration is reduced in sepsis due to decreased de novo arginine synthesis and its increased consumption through myeloid ARG1 and iNOS [120, 121]. Consequently, limited arginine availability in sepsis may lead to impaired T cell proliferation and function and related immunosuppression [122]. Therapeutic citrulline administration increasing arginine plasma levels, restores T cell mitochondrial function and proliferation and reduces sepsis-induced immunosuppression [123]. Especially patients with persistent immunosuppressed inflammatory catabolic state (PICS) may benefit from arginine supplementation [124]. Hence, restoring arginine levels may be useful against sepsis-induced lymphopenia and immune paralysis.
Myeloid arginine metabolism in chronic inflammatory diseases
Microglia-mediated neuroinflammation
Microglia are resident immune cells of the central nervous system (CNS). They derive from embryonic yolk sac precursors and can self-renew. They perform various functions, including phagocytosis, synaptic pruning during neuronal development and inflammatory reactions to infection or injury [125]. In the healthy mouse brain, ARG1 is expressed in microglia of certain brain regions, such as the basal forebrain [126]. ARG1 expression is induced by IL-4Rα and STAT6 signaling in microglia and promotes their phagocytic capacity [126–128]. Microglia-specific ARG1 deficiency results in impaired neural maturation in the hippocampus, which consequently causes cognitive deficits in mice [126]. In mouse models of AD, a neurodegenerative disease characterized by deposition of amyloid beta peptide (Aβ), an early toxic event in AD pathogenesis, ARG1+ microglia mediate Aβ plaque clearance, while ARG1 overexpression in the CNS reduces inflammation and hippocampal atrophy and ameliorates disease [129, 130]. Moreover, microglia may release ARG1, which catabolizes extracellular arginine and reduces arginine levels in the brain [131]. However, inhibition of arginine catabolism with ARG1 blockers reduces microglial inflammation and mitigates AD-like pathology [131]. As in macrophages, arginine metabolism in microglia is reprogramed depending on the inflammatory state and phagocytic activity [126, 132]. For instance, upon demyelination, microglial cells highly express iNOS, while at later stages of remyelination they acquire an iNOS−/ARG1+ phenotype [132]. iNOS may promote citrullination of myelin, which impairs remyelination and increases neuroinflammation [133]. Hence, the switch from iNOS to ARG1 is essential for resolution of neuroinflammation and restoration of neural tissue integrity.
Upregulation of ARG1 expression leads to increased polyamine production [134]. Alterations in brain polyamine metabolism are associated with neurodegenerative disease pathology [135, 136]. Inflammation upregulates ODC1 expression in microglia and neurons [137]. In turn, spermidine counteracts microglia-mediated neuroinflammation and exerts neuroprotective effects [138, 139]. Spermidine administration increases expression of genes related to phagocytosis in microglia and reduces Aβ pathology in an AD mouse model [138]. Moreover, administration of spermidine to mice with experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, the most prevalent demyelinating disease in humans, diminishes the potential of macrophages for T cell activation and ameliorates disease severity [139].
Arthritis
Osteoclasts are multinucleated giant cells derived from myeloid precursors. They regulate bone homeostasis through bone resorption, as opposed to bone-forming osteoblasts. Osteoclast maturation is driven by receptor activator of nuclear factor kappa-Β ligand (RANKL) and macrophage colony stimulating factor (M-CSF). Excessive osteoclast activation is linked to pathologic conditions, such as arthritis and osteoporosis [140]. Rheumatoid arthritis patients have increased arginine levels in the serum suggesting alterations in arginine metabolism [141]. L-arginine supplementation restrains inflammatory bone loss in experimental mouse arthritis and inhibits osteoclastogenesis in an ARG1-dependent manner tilting energy metabolism of inflammatory osteoclasts from glycolysis to oxidative phosphorylation [60, 141]. Conversely, arginine depletion by application of recombinant ARG1 also inhibits RANKL-driven osteoclastogenesis and ameliorates arthritis in mice [142]. These data suggest that both, supra- and hypophysiological amounts of arginine impair osteoclastogenesis. Hypoxia may induce iNOS expression in osteoclasts and promote osteoclastogenesis through NO-mediated effects [143, 144]. In accordance, iNOS deficient mice were reported to have bone abnormalities associated with reduced osteoclast-mediated bone resorption [145, 146]. NO also promotes osteoblast differentiation, as shown by ASL deficiency-mediated NO depletion [147]. Finally, protein citrullination by peptidylarginine deiminases (PAD) is essential for osteoclast differentiation and anti-citrullinated protein/peptide antibodies (ACPAs) are associated with bone loss and are therefore important diagnostic biomarkers in rheumatoid arthritis [148].
The role of myeloid arginine metabolism in cancer
Arginine metabolism in tumor-associated macrophages and tumor-associated neutrophils
Tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) infiltrate solid tumors, and control tumor growth and metastasis through various mechanisms, including T cell suppression, secretion of growth factors such as TGF-β, promotion of angiogenesis and matrix remodeling [149, 150]. Collagen taken up by TAMs is a rich source of arginine, which is further metabolized by ARG1 and iNOS [151–153]. Granulocyte–macrophage colony-stimulating factor (GM-CSF) produced by tumor cells and lactic acid synergistically induce ARG1 expression in TAMs [154]. The effect of GM-CSF was abrogated by inhibition of STAT3 and p38 MAPK, despite the fact that GM-CSF mainly signals via STAT5 [154, 155]. Moreover, the induction of ARG1 expression by lactic acid in combination with GM-CSF was mediated through the cAMP-cAMP response element–binding protein (CREB) pathway [154]. ARG1 expression generally correlates with the immunosuppressive function of TAMs and is dependent on arginine availability [154, 156]. For instance in glioma tumors, arginine deprivation reduces ARG1 expression in tumor-associated macrophages/microglia and enhances their proinflammatory and phagocytic function [156].
Similar to M2-like macrophages, ARG1-expressing TAMs and TANs deprive T cells from arginine thereby suppressing their anti-tumor effects [157, 158]. Extracellular L-arginine is required for the expression of T cell antigen receptor ζ chain (CD3ζ) and T cell proliferation [159, 160]. Consequently, ARG1 inhibition enhances the effects of immunotherapy, increases tumor-infiltrating CD8+ T cell and natural killer (NK) cell populations and reduces tumor growth in mice [161–163]. TANs also express ARG1 but in contrast to TAMs, do not metabolize arginine; instead they exocytose ARG1 in granules, which consumes arginine and thereby suppresses T and NK cell function [164–166]. Not only ARG1 but also iNOS mediates TAM immunosuppressive effects through production of peroxynitrite, which causes nitration and nitrosylation of the TCR and its co-receptor CD8 at tyrosine residues, thereby disrupting TCR signaling [167].
Tumor cells are auxotrophic for arginine [168]. They use arginine for protein synthesis and iNOS-mediated catabolism; in fact, tumor iNOS expression correlates with increased tumor cell proliferation, migration and metastasis [169–171]. Hence, competition of tumor cells with TAMs for arginine may also limit tumor growth [157]. In fact, to overcome arginine shortage, tumor cells can acquire the ability to upregulate arginine biosynthesis through the argininosuccinate shunt [172].
Moreover, TAMs produce high amounts of polyamines, which are associated with both, malignancy and immunosuppression [173]. TAM-derived polyamines may be taken up by tumor cells and promote their proliferation [173]. On the other hand, polyamines buffer the low intracellular pH in TAMs to support their survival in the acidic environment of solid tumors [174]. Consequently, simultaneous blockage of synthesis and uptake of polyamines reduces the myeloid/T cell ratio, decreases tumor growth in a CD8+ T cell-dependent manner and increases survival of tumor-bearing mice [174–176]. Finally, TAMs can produce creatine, which is taken up by tumor cells through the SLC6A8 transporter to promote their survival, while inhibition of creatine transport hinders tumor growth [177].
Arginine metabolism in myeloid-derived suppressor cells
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of rather immature, immunosuppressive myeloid cells, which are particularly abundant in cancer and chronic inflammatory diseases [178, 179]. Increased amounts of MDSCs correlate with poor prognosis in cancer patients [180]. They are classified into polymorphonuclear-(PMN)-MDSCs and monocytic-(M-)MDSCs [149, 181]. However, it remains unclear whether MDSCs are indeed a distinct immature myeloid cell type, as initially described, or merely myeloid cells, which gained immunosuppressive function in response to local conditions [182]. Arginine metabolism plays a pivotal role in the immunosuppressive function of MDSCs. ARG1 is highly expressed in human PMN-MDSCs thereby depriving T cells from arginine [183–185]. This restrains T cell proliferation and impairs T cell receptor CD3ζ chain expression, thereby suppressing Τ cell function [184]. While mouse MDSCs deplete arginine via its uptake and intracellular metabolism, human MDSCs mainly release arginase [186]. MDSCs may also secrete ARG1 within extracellular vesicles [187]. Consequently, released ARG1 decreases arginine levels in the local microenvironment impairing T cell function [78]. Accordingly, in cancer patients, arginase activity in PBMCs is strongly upregulated, while arginine amounts in plasma and expression of the CD3ζ TCR chain are diminished [188]. T cell-deriving cytokines, such as IL-4 and IL-10, can induce ARG1 expression in MDSCs [189]. Mechanistically, cyclooxygenase 1 and 2 (COX-1 and −2)–dependent PGE2 production by tumor cells promotes ARG1 expression in MDSCs in a paracrine fashion [190]. Moreover, β2 adrenergic signaling via STAT3 drives Arg1 gene expression in MDSCs [191, 192]. Interestingly, administration of the pegylated form of the catabolic enzyme arginase I (peg-Arg I) blocked T cell proliferation and function, and induced MDSC accumulation through an eIF2a-dependent mechanism leading to increased tumor growth [193]. ARG1+ MDSCs release polyamines, which promote an immunosuppressive phenotype of dendritic cells via indoleamine 2,3-dioxygenase 1 (IDO1) activation [67]. However, MDSCs also inhibit CD8+ T cell function through direct cell-to-cell contact and NO-mediated nitration of TCR proteins causing TCR conformational changes [194]. Concluding, arginine metabolism is key for the immunosuppressive function of MDSCs. Ongoing clinical studies hence test its therapeutic targeting in cancer [195].
Myeloid arginine metabolism in surgery
Surgical stress causes postoperative immunological changes including MDSC expansion and suppression of NK cell function, which in case of cancer surgery increases the risk of postoperative metastatic disease [196–201]. ARG1-expressing MDSCs lead to post-operative systemic depletion of arginine [202]. Consequently, arginine-deprived conditions lead to impaired NK cell proliferation, cytotoxicity and IFNγ production [203]. Perioperative arginine supplementation was shown to improve recovery, mitigate postoperative infections, accelerate NK cell recovery and reduce post-operative metastases [200, 201, 204].
Concluding remarks
NO production and the argininosuccinate shunt promote inflammation and anti-microbial responses. In contrast, polyamine and proline metabolism is associated with anti-inflammatory and tissue-restorative responses. At the junction of these branches of arginine metabolism lies ARG1, which regulates the balance between pro- and anti-inflammatory myeloid responses. Dysregulation of this balance leads to non-resolving inflammation and tissue destruction as is the case in chronic inflammatory diseases, or to immunosuppression in cancer. Since arginine metabolism plays a critical role in myeloid cell reprograming, the therapeutic potential of targeting certain immuno-regulatory nodes in arginine metabolism may be leveraged to treat diseases, which are driven by dysregulated immune function.
Author contributions
VIA and EK wrote the manuscript and made the figures, TC edited the manuscript.
Funding
Open Access funding enabled and organized by Projekt DEAL. The research of the authors is supported by funds from the Deutsche Forschungsgemeinschaft (AL 1686/6–1 and SFB-TRR 205 project A07 to VIA; SFB-TRR 127 project A03 to TC), the Saxon State Ministry of Science, Culture and Tourism-SMWK (Unterstützung profilbestimmender Struktureinheiten der TU Dresden, to T.C.) and the Deutsche Krebshilfe (Exzellenzförderprogramm für etablierte Wissenschaftlerinnen und Wissenschaftler to T.C.).
Data availability
No original data was generated for this review article.
Declarations
Conflict of interest
The authors declare no competing interests.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- 1.Ng LG, Liu Z, Kwok I, Ginhoux F (2023) Origin and heterogeneity of tissue myeloid cells: a focus on GMP-Derived monocytes and neutrophils. Annu Rev Immunol 41:375–404 [DOI] [PubMed] [Google Scholar]
- 2.Marti ILAA, Reith W (2021) Arginine-dependent immune responses. Cell Mol Life Sci 78:5303–5324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Krebs H, Henseleit K (1932) Untersuchungen über die Harnstoffbildung im Tierkörper. Klin Wochenschr 11:757–759 [Google Scholar]
- 4.Bassler K, Schulte-Schrepping J, Warnat-Herresthal S, Aschenbrenner AC, Schultze JL (2019) The myeloid cell compartment-cell by cell. Annu Rev Immunol 37:269–293 [DOI] [PubMed] [Google Scholar]
- 5.Gurtner A, Crepaz D, Arnold IC (2023) Emerging functions of tissue-resident eosinophils. J Exp Med 220:e20221435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Mayadas TN, Cullere X, Lowell CA (2014) The multifaceted functions of neutrophils. Annu Rev Pathol 9:181–218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mitroulis I, Alexaki VI, Kourtzelis I, Ziogas A, Hajishengallis G, Chavakis T (2015) Leukocyte integrins: role in leukocyte recruitment and as therapeutic targets in inflammatory disease. Pharmacol Ther 147:123–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Fullerton JN, Gilroy DW (2016) Resolution of inflammation: a new therapeutic frontier. Nat Rev Drug Discov 15:551–567 [DOI] [PubMed] [Google Scholar]
- 9.Kourtzelis I, Hajishengallis G, Chavakis T (2020) Phagocytosis of apoptotic cells in resolution of inflammation. Front Immunol 11:553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis e Sousa C (2021) Dendritic Cells Revisited. Annu Rev Immunol 39:131–66 [DOI] [PubMed] [Google Scholar]
- 11.Miyake K, Ito J, Karasuyama H (2022) Role of basophils in a broad spectrum of disorders. Front Immunol 13:902494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Tomlinson C, Rafii M, Sgro M, Ball RO, Pencharz P (2011) Arginine is synthesized from proline, not glutamate, in enterally fed human preterm neonates. Pediatr Res 69:46–50 [DOI] [PubMed] [Google Scholar]
- 13.Qualls JE, Subramanian C, Rafi W, Smith AM, Balouzian L, DeFreitas AA, Shirey KA, Reutterer B, Kernbauer E, Stockinger S, Decker T, Miyairi I, Vogel SN, Salgame P, Rock CO, Murray PJ (2012) Sustained generation of nitric oxide and control of mycobacterial infection requires argininosuccinate synthase 1. Cell Host Microbe 12:313–323 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Jha AK, Huang SC, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, Chmielewski K, Stewart KM, Ashall J, Everts B, Pearce EJ, Driggers EM, Artyomov MN (2015) Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 42:419–430 [DOI] [PubMed] [Google Scholar]
- 15.Jungnickel KEJ, Parker JL, Newstead S (2018) Structural basis for amino acid transport by the CAT family of SLC7 transporters. Nat Commun 9:550 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Thompson RW, Pesce JT, Ramalingam T, Wilson MS, White S, Cheever AW, Ricklefs SM, Porcella SF, Li L, Ellies LG, Wynn TA (2008) Cationic amino acid transporter-2 regulates immunity by modulating arginase activity. PLoS Pathog 4:e1000023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Murray PJ (2016) Amino acid auxotrophy as a system of immunological control nodes. Nat Immunol 17:132–139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Geiger R, Rieckmann JC, Wolf T, Basso C, Feng Y, Fuhrer T, Kogadeeva M, Picotti P, Meissner F, Mann M, Zamboni N, Sallusto F, Lanzavecchia A (2016) L-Arginine modulates T cell metabolism and enhances survival and anti-tumor activity. Cell 167(829–42):e13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sanchez-Jimenez F, Medina MA, Villalobos-Rueda L, Urdiales JL (2019) Polyamines in mammalian pathophysiology. Cell Mol Life Sci 76:3987–4008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Wei Z, Oh J, Flavell RA, Crawford JM (2022) LACC1 bridges NOS2 and polyamine metabolism in inflammatory macrophages. Nature 609:348–353 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, Locati M, Mantovani A, Martinez FO, Mege JL, Mosser DM, Natoli G, Saeij JP, Schultze JL, Shirey KA, Sica A, Suttles J, Udalova I, van Ginderachter JA, Vogel SN, Wynn TA (2014) Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41:14–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chavakis T, Alexaki VI, Ferrante AW Jr (2023) Macrophage function in adipose tissue homeostasis and metabolic inflammation. Nat Immunol 24:757–766 [DOI] [PubMed] [Google Scholar]
- 23.Bailey JD, Diotallevi M, Nicol T, McNeill E, Shaw A, Chuaiphichai S, Hale A, Starr A, Nandi M, Stylianou E, McShane H, Davis S, Fischer R, Kessler BM, McCullagh J, Channon KM, Crabtree MJ (2019) Nitric oxide modulates metabolic remodeling in inflammatory macrophages through TCA cycle regulation and itaconate accumulation. Cell Rep 28(218–30):e7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.McNeill E, Stylianou E, Crabtree MJ, Harrington-Kandt R, Kolb AL, Diotallevi M, Hale AB, Bettencourt P, Tanner R, O’Shea MK, Matsumiya M, Lockstone H, Muller J, Fletcher HA, Greaves DR, McShane H, Channon KM (2018) Regulation of mycobacterial infection by macrophage Gch1 and tetrahydrobiopterin. Nat Commun 9:5409 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Jung JY, Madan-Lala R, Georgieva M, Rengarajan J, Sohaskey CD, Bange FC, Robinson CM (2013) The intracellular environment of human macrophages that produce nitric oxide promotes growth of mycobacteria. Infect Immun 81:3198–3209 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Seim GL, Britt EC, John SV, Yeo FJ, Johnson AR, Eisenstein RS, Pagliarini DJ, Fan J (2019) Two-stage metabolic remodelling in macrophages in response to lipopolysaccharide and interferon-gamma stimulation. Nat Metab 1:731–742 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Goldring CE, Reveneau S, Algarte M, Jeannin JF (1996) In vivo footprinting of the mouse inducible nitric oxide synthase gene: inducible protein occupation of numerous sites including Oct and NF-IL6. Nucleic Acids Res 24:1682–1687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Xie QW, Kashiwabara Y, Nathan C (1994) Role of transcription factor NF-kappa B/Rel in induction of nitric oxide synthase. J Biol Chem 269:4705–4708 [PubMed] [Google Scholar]
- 29.Guo Z, Shao L, Du Q, Park KS, Geller DA (2007) Identification of a classic cytokine-induced enhancer upstream in the human iNOS promoter. FASEB J 21:535–542 [DOI] [PubMed] [Google Scholar]
- 30.Taylor BS, de Vera ME, Ganster RW, Wang Q, Shapiro RA, Morris SM Jr, Billiar TR, Geller DA (1998) Multiple NF-kappaB enhancer elements regulate cytokine induction of the human inducible nitric oxide synthase gene. J Biol Chem 273:15148–15156 [DOI] [PubMed] [Google Scholar]
- 31.Rodriguez-Pascual F, Hausding M, Ihrig-Biedert I, Furneaux H, Levy AP, Forstermann U, Kleinert H (2000) Complex contribution of the 3’-untranslated region to the expressional regulation of the human inducible nitric-oxide synthase gene. Involvement of the RNA-binding protein HuR. J Biol Chem 275:26040–26049 [DOI] [PubMed] [Google Scholar]
- 32.Gross TJ, Kremens K, Powers LS, Brink B, Knutson T, Domann FE, Philibert RA, Milhem MM, Monick MM (2014) Epigenetic silencing of the human NOS2 gene: rethinking the role of nitric oxide in human macrophage inflammatory responses. J Immunol 192:2326–2338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhong J, Scholz T, Yau ACY, Guerard S, Huffmeier U, Burkhardt H, Holmdahl R (2018) Mannan-induced Nos2 in macrophages enhances IL-17-driven psoriatic arthritis by innate lymphocytes. Sci Adv 4:eaas9864 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Padgett EL, Pruett SB (1995) Rat, mouse and human neutrophils stimulated by a variety of activating agents produce much less nitrite than rodent macrophages. Immunology 84:135–141 [PMC free article] [PubMed] [Google Scholar]
- 35.Mishra BB, Lovewell RR, Olive AJ, Zhang G, Wang W, Eugenin E, Smith CM, Phuah JY, Long JE, Dubuke ML, Palace SG, Goguen JD, Baker RE, Nambi S, Mishra R, Booty MG, Baer CE, Shaffer SA, Dartois V, McCormick BA, Chen X, Sassetti CM (2017) Nitric oxide prevents a pathogen-permissive granulocytic inflammation during tuberculosis. Nat Microbiol 2:17072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.MacMicking JD, North RJ, LaCourse R, Mudgett JS, Shah SK, Nathan CF (1997) Identification of nitric oxide synthase as a protective locus against tuberculosis. Proc Natl Acad Sci U S A 94:5243–5248 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Palmieri EM, McGinity C, Wink DA, McVicar DW (2020) Nitric oxide in macrophage immunometabolism: hiding in plain sight. Metabolites 10:429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lee J, Ryu H, Ferrante RJ, Morris SM Jr, Ratan RR (2003) Translational control of inducible nitric oxide synthase expression by arginine can explain the arginine paradox. Proc Natl Acad Sci U S A 100:4843–4848 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Lange SM, McKell MC, Schmidt SM, Zhao J, Crowther RR, Green LC, Bricker RL, Arnett E, Kohler SE, Schlesinger LS, Setchell KDR, Qualls JE (2019) l-Arginine synthesis from l-Citrulline in myeloid cells drives host defense against mycobacteria in vivo. J Immunol 202:1747–1754 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Hooftman A, Peace CG, Ryan DG, Day EA, Yang M, McGettrick AF, Yin M, Montano EN, Huo L, Toller-Kawahisa JE, Zecchini V, Ryan TAJ, Bolado-Carrancio A, Casey AM, Prag HA, Costa ASH, De Los SG, Ishimori M, Wallace DJ, Venuturupalli S, Nikitopoulou E, Frizzell N, Johansson C, Von Kriegsheim A, Murphy MP, Jefferies C, Frezza C, O’Neill LAJ (2023) Macrophage fumarate hydratase restrains mtRNA-mediated interferon production. Nature 615:490–498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chouchani ET, Methner C, Nadtochiy SM, Logan A, Pell VR, Ding S, James AM, Cocheme HM, Reinhold J, Lilley KS, Partridge L, Fearnley IM, Robinson AJ, Hartley RC, Smith RA, Krieg T, Brookes PS, Murphy MP (2013) Cardioprotection by S-nitrosation of a cysteine switch on mitochondrial complex I. Nat Med 19:753–759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Clementi E, Brown GC, Feelisch M, Moncada S (1998) Persistent inhibition of cell respiration by nitric oxide: crucial role of S-nitrosylation of mitochondrial complex I and protective action of glutathione. Proc Natl Acad Sci U S A 95:7631–7636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Van den Bossche J, Baardman J, Otto NA, van der Velden S, Neele AE, van den Berg SM, Luque-Martin R, Chen HJ, Boshuizen MC, Ahmed M, Hoeksema MA, de Vos AF, de Winther MP (2016) Mitochondrial dysfunction prevents repolarization of inflammatory macrophages. Cell Rep 17:684–696 [DOI] [PubMed] [Google Scholar]
- 44.Palmieri EM, Gonzalez-Cotto M, Baseler WA, Davies LC, Ghesquiere B, Maio N, Rice CM, Rouault TA, Cassel T, Higashi RM, Lane AN, Fan TW, Wink DA, McVicar DW (2020) Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat Commun 11:698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Doulias PT, Tenopoulou M, Greene JL, Raju K, Ischiropoulos H (2013) Nitric oxide regulates mitochondrial fatty acid metabolism through reversible protein S-nitrosylation. Sci Signal 6:rs1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Everts B, Amiel E, van der Windt GJ, Freitas TC, Chott R, Yarasheski KE, Pearce EL, Pearce EJ (2012) Commitment to glycolysis sustains survival of NO-producing inflammatory dendritic cells. Blood 120:1422–1431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Mishra BB, Rathinam VA, Martens GW, Martinot AJ, Kornfeld H, Fitzgerald KA, Sassetti CM (2013) Nitric oxide controls the immunopathology of tuberculosis by inhibiting NLRP3 inflammasome-dependent processing of IL-1beta. Nat Immunol 14:52–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bambouskova M, Potuckova L, Paulenda T, Kerndl M, Mogilenko DA, Lizotte K, Swain A, Hayes S, Sheldon RD, Kim H, Kapadnis U, Ellis AE, Isaguirre C, Burdess S, Laha A, Amarasinghe GK, Chubukov V, Roddy TP, Diamond MS, Jones RG, Simons DM, Artyomov MN (2021) Itaconate confers tolerance to late NLRP3 inflammasome activation. Cell Rep 34:108756 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Braverman J, Stanley SA (2017) Nitric oxide modulates macrophage responses to mycobacterium tuberculosis infection through activation of HIF-1alpha and repression of NF-kappaB. J Immunol 199:1805–1816 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH, Zheng L, Gardet A, Tong Z, Jany SS, Corr SC, Haneklaus M, Caffrey BE, Pierce K, Walmsley S, Beasley FC, Cummins E, Nizet V, Whyte M, Taylor CT, Lin H, Masters SL, Gottlieb E, Kelly VP, Clish C, Auron PE, Xavier RJ, O’Neill LA (2013) Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature 496:238–242 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Vasquez-Vivar J, Kalyanaraman B, Martasek P, Hogg N, Masters BS, Karoui H, Tordo P, Pritchard KA Jr (1998) Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci U S A 95:9220–9225 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.El Kasmi KC, Qualls JE, Pesce JT, Smith AM, Thompson RW, Henao-Tamayo M, Basaraba RJ, Konig T, Schleicher U, Koo MS, Kaplan G, Fitzgerald KA, Tuomanen EI, Orme IM, Kanneganti TD, Bogdan C, Wynn TA, Murray PJ (2008) Toll-like receptor-induced arginase 1 in macrophages thwarts effective immunity against intracellular pathogens. Nat Immunol 9:1399–1406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Mattila JT, Ojo OO, Kepka-Lenhart D, Marino S, Kim JH, Eum SY, Via LE, Barry CE 3rd, Klein E, Kirschner DE, Morris SM Jr, Lin PL, Flynn JL (2013) Microenvironments in tuberculous granulomas are delineated by distinct populations of macrophage subsets and expression of nitric oxide synthase and arginase isoforms. J Immunol 191:773–784 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Pauleau AL, Rutschman R, Lang R, Pernis A, Watowich SS, Murray PJ (2004) Enhancer-mediated control of macrophage-specific arginase I expression. J Immunol 172:7565–7573 [DOI] [PubMed] [Google Scholar]
- 55.Gray MJ, Poljakovic M, Kepka-Lenhart D, Morris SM Jr (2005) Induction of arginase I transcription by IL-4 requires a composite DNA response element for STAT6 and C/EBPbeta. Gene 353:98–106 [DOI] [PubMed] [Google Scholar]
- 56.Zhang D, Tang Z, Huang H, Zhou G, Cui C, Weng Y, Liu W, Kim S, Lee S, Perez-Neut M, Ding J, Czyz D, Hu R, Ye Z, He M, Zheng YG, Shuman HA, Dai L, Ren B, Roeder RG, Becker L, Zhao Y (2019) Metabolic regulation of gene expression by histone lactylation. Nature 574:575–580 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wijnands KA, Hoeksema MA, Meesters DM, van den Akker NM, Molin DG, Briede JJ, Ghosh M, Kohler SE, van Zandvoort MA, de Winther MP, Buurman WA, Lamers WH, Poeze M (2014) Arginase-1 deficiency regulates arginine concentrations and NOS2-mediated NO production during endotoxemia. PLoS ONE 9:e86135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Qualls JE, Neale G, Smith AM, Koo MS, DeFreitas AA, Zhang H, Kaplan G, Watowich SS, Murray PJ (2010) Arginine usage in mycobacteria-infected macrophages depends on autocrine-paracrine cytokine signaling. Sci Signal 3:ra62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Dowling JK, Afzal R, Gearing LJ, Cervantes-Silva MP, Annett S, Davis GM, De Santi C, Assmann N, Dettmer K, Gough DJ, Bantug GR, Hamid FI, Nally FK, Duffy CP, Gorman AL, Liddicoat AM, Lavelle EC, Hess C, Oefner PJ, Finlay DK, Davey GP, Robson T, Curtis AM, Hertzog PJ, Williams BRG, McCoy CE (2021) Mitochondrial arginase-2 is essential for IL-10 metabolic reprogramming of inflammatory macrophages. Nat Commun 12:1460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Hannemann N, Cao S, Eriksson D, Schnelzer A, Jordan J, Eberhardt M, Schleicher U, Rech J, Ramming A, Uebe S, Ekici A, Canete JD, Chen X, Bauerle T, Vera J, Bogdan C, Schett G, Bozec A (2019) Transcription factor Fra-1 targets arginase-1 to enhance macrophage-mediated inflammation in arthritis. J Clin Invest 129:2669–2684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.El-Gayar S, Thuring-Nahler H, Pfeilschifter J, Rollinghoff M, Bogdan C (2003) Translational control of inducible nitric oxide synthase by IL-13 and arginine availability in inflammatory macrophages. J Immunol 171:4561–4568 [DOI] [PubMed] [Google Scholar]
- 62.Schleicher U, Paduch K, Debus A, Obermeyer S, Konig T, Kling JC, Ribechini E, Dudziak D, Mougiakakos D, Murray PJ, Ostuni R, Korner H, Bogdan C (2016) TNF-Mediated restriction of Arginase 1 expression in myeloid cells triggers type 2 NO synthase activity at the site of infection. Cell Rep 15:1062–1075 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Modolell M, Corraliza IM, Link F, Soler G, Eichmann K (1995) Reciprocal regulation of the nitric oxide synthase/arginase balance in mouse bone marrow-derived macrophages by TH1 and TH2 cytokines. Eur J Immunol 25:1101–1104 [DOI] [PubMed] [Google Scholar]
- 64.Rapovy SM, Zhao J, Bricker RL, Schmidt SM, Setchell KD, Qualls JE (2015) Differential requirements for L-Citrulline and L-Arginine during antimycobacterial macrophage activity. J Immunol 195:3293–3300 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pesce JT, Ramalingam TR, Mentink-Kane MM, Wilson MS, El Kasmi KC, Smith AM, Thompson RW, Cheever AW, Murray PJ, Wynn TA (2009) Arginase-1-expressing macrophages suppress Th2 cytokine-driven inflammation and fibrosis. PLoS Pathog 5:e1000371 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rutschman R, Lang R, Hesse M, Ihle JN, Wynn TA, Murray PJ (2001) Cutting edge: Stat6-dependent substrate depletion regulates nitric oxide production. J Immunol 166:2173–2177 [DOI] [PubMed] [Google Scholar]
- 67.Mondanelli G, Bianchi R, Pallotta MT, Orabona C, Albini E, Iacono A, Belladonna ML, Vacca C, Fallarino F, Macchiarulo A, Ugel S, Bronte V, Gevi F, Zolla L, Verhaar A, Peppelenbosch M, Mazza EMC, Bicciato S, Laouar Y, Santambrogio L, Puccetti P, Volpi C, Grohmann U (2017) A relay pathway between arginine and tryptophan metabolism confers immunosuppressive properties on dendritic cells. Immunity 46:233–244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Boutard V, Havouis R, Fouqueray B, Philippe C, Moulinoux JP, Baud L (1995) Transforming growth factor-beta stimulates arginase activity in macrophages. Implications for the regulation of macrophage cytotoxicity. J Immunol 155:2077–2084 [PubMed] [Google Scholar]
- 69.Lang R, Patel D, Morris JJ, Rutschman RL, Murray PJ (2002) Shaping gene expression in activated and resting primary macrophages by IL-10. J Immunol 169:2253–2263 [DOI] [PubMed] [Google Scholar]
- 70.Munder M, Eichmann K, Modolell M (1998) Alternative metabolic states in murine macrophages reflected by the nitric oxide synthase/arginase balance: competitive regulation by CD4+ T cells correlates with Th1/Th2 phenotype. J Immunol 160:5347–5354 [PubMed] [Google Scholar]
- 71.Chen F, El-Naccache DW, Ponessa JJ, Lemenze A, Espinosa V, Wu W, Lothstein K, Jin L, Antao O, Weinstein JS, Damani-Yokota P, Khanna K, Murray PJ, Rivera A, Siracusa MC, Gause WC (2022) Helminth resistance is mediated by differential activation of recruited monocyte-derived alveolar macrophages and arginine depletion. Cell Rep 38:110215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Esser-von Bieren J, Mosconi I, Guiet R, Piersgilli A, Volpe B, Chen F, Gause WC, Seitz A, Verbeek JS, Harris NL (2013) Antibodies trap tissue migrating helminth larvae and prevent tissue damage by driving IL-4Ralpha-independent alternative differentiation of macrophages. PLoS Pathog 9:e1003771 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hargrave KE, Woods S, Millington O, Chalmers S, Westrop GD, Roberts CW (2019) Multi-Omics studies demonstrate toxoplasma gondii-induced metabolic reprogramming of murine dendritic cells. Front Cell Infect Microbiol 9:309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Mandal A, Das S, Kumar A, Roy S, Verma S, Ghosh AK, Singh R, Abhishek K, Saini S, Sardar AH, Purkait B, Kumar A, Mandal C, Das P (2017) l-Arginine uptake by cationic amino acid transporter promotes intra-macrophage survival of leishmania donovani by enhancing arginase-mediated polyamine synthesis. Front Immunol 8:839 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 75.Hesse M, Modolell M, La Flamme AC, Schito M, Fuentes JM, Cheever AW, Pearce EJ, Wynn TA (2001) Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of L-arginine metabolism. J Immunol 167:6533–6544 [DOI] [PubMed] [Google Scholar]
- 76.Choi BS, Martinez-Falero IC, Corset C, Munder M, Modolell M, Muller I, Kropf P (2009) Differential impact of L-arginine deprivation on the activation and effector functions of T cells and macrophages. J Leukoc Biol 85:268–277 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.West EE, Merle NS, Kaminski MM, Palacios G, Kumar D, Wang L, Bibby JA, Overdahl K, Jarmusch AK, Freeley S, Lee DY, Thompson JW, Yu ZX, Taylor N, Sitbon M, Green DR, Bohrer A, Mayer-Barber KD, Afzali B, Kazemian M, Scholl-Buergi S, Karall D, Huemer M, Kemper C (2023) Loss of CD4(+) T cell-intrinsic arginase 1 accelerates Th1 response kinetics and reduces lung pathology during influenza infection. Immunity 56(2036–53):e12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Munder M, Schneider H, Luckner C, Giese T, Langhans CD, Fuentes JM, Kropf P, Mueller I, Kolb A, Modolell M, Ho AD (2006) Suppression of T-cell functions by human granulocyte arginase. Blood 108:1627–1634 [DOI] [PubMed] [Google Scholar]
- 79.Munder M, Mollinedo F, Calafat J, Canchado J, Gil-Lamaignere C, Fuentes JM, Luckner C, Doschko G, Soler G, Eichmann K, Muller FM, Ho AD, Goerner M, Modolell M (2005) Arginase I is constitutively expressed in human granulocytes and participates in fungicidal activity. Blood 105:2549–2556 [DOI] [PubMed] [Google Scholar]
- 80.Herbert DR, Orekov T, Roloson A, Ilies M, Perkins C, O’Brien W, Cederbaum S, Christianson DW, Zimmermann N, Rothenberg ME, Finkelman FD (2010) Arginase I suppresses IL-12/IL-23p40-driven intestinal inflammation during acute schistosomiasis. J Immunol 184:6438–6446 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Barron L, Smith AM, El Kasmi KC, Qualls JE, Huang X, Cheever A, Borthwick LA, Wilson MS, Murray PJ, Wynn TA (2013) Role of arginase 1 from myeloid cells in th2-dominated lung inflammation. PLoS ONE 8:e61961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Duque-Correa MA, Kuhl AA, Rodriguez PC, Zedler U, Schommer-Leitner S, Rao M, Weiner J 3rd, Hurwitz R, Qualls JE, Kosmiadi GA, Murray PJ, Kaufmann SH, Reece ST (2014) Macrophage arginase-1 controls bacterial growth and pathology in hypoxic tuberculosis granulomas. Proc Natl Acad Sci U S A 111:E4024–E4032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Monin L, Griffiths KL, Lam WY, Gopal R, Kang DD, Ahmed M, Rajamanickam A, Cruz-Lagunas A, Zuniga J, Babu S, Kolls JK, Mitreva M, Rosa BA, Ramos-Payan R, Morrison TE, Murray PJ, Rangel-Moreno J, Pearce EJ, Khader SA (2015) Helminth-induced arginase-1 exacerbates lung inflammation and disease severity in tuberculosis. J Clin Invest 125:4699–4713 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.De Santi C, Nally FK, Afzal R, Duffy CP, Fitzsimons S, Annett SL, Robson T, Dowling JK, Cryan SA, McCoy CE (2022) Enhancing arginase 2 expression using target site blockers as a strategy to modulate macrophage phenotype. Mol Ther Nucleic Acids 29:643–655 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Fitzsimons S, Munoz-San Martin M, Nally F, Dillon E, Fashina IA, Strowitzki MJ, Ramio-Torrenta L, Dowling JK, De Santi C, McCoy CE (2023) Inhibition of pro-inflammatory signaling in human primary macrophages by enhancing arginase-2 via target site blockers. Mol Ther Nucleic Acids 33:941–959 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ming XF, Rajapakse AG, Yepuri G, Xiong Y, Carvas JM, Ruffieux J, Scerri I, Wu Z, Popp K, Li J, Sartori C, Scherrer U, Kwak BR, Montani JP, Yang Z (2012) Arginase II promotes macrophage inflammatory responses through mitochondrial reactive oxygen species, contributing to insulin resistance and atherogenesis. J Am Heart Assoc 1:e000992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hardbower DM, Asim M, Luis PB, Singh K, Barry DP, Yang C, Steeves MA, Cleveland JL, Schneider C, Piazuelo MB, Gobert AP, Wilson KT (2017) Ornithine decarboxylase regulates M1 macrophage activation and mucosal inflammation via histone modifications. Proc Natl Acad Sci U S A 114:E751–E760 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chaturvedi R, Asim M, Hoge S, Lewis ND, Singh K, Barry DP, de Sablet T, Piazuelo MB, Sarvaria AR, Cheng Y, Closs EI, Casero RA Jr, Gobert AP, Wilson KT (2010) Polyamines impair immunity to helicobacter pylori by inhibiting L-Arginine uptake required for nitric oxide production. Gastroenterology 139:1686–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Nakamura A, Kurihara S, Takahashi D, Ohashi W, Nakamura Y, Kimura S, Onuki M, Kume A, Sasazawa Y, Furusawa Y, Obata Y, Fukuda S, Saiki S, Matsumoto M, Hase K (2021) Symbiotic polyamine metabolism regulates epithelial proliferation and macrophage differentiation in the colon. Nat Commun 12:2105 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Bussiere FI, Chaturvedi R, Cheng Y, Gobert AP, Asim M, Blumberg DR, Xu H, Kim PY, Hacker A, Casero RA Jr, Wilson KT (2005) Spermine causes loss of innate immune response to Helicobacter pylori by inhibition of inducible nitric-oxide synthase translation. J Biol Chem 280:2409–2412 [DOI] [PubMed] [Google Scholar]
- 91.Fritsch SD, Sukhbaatar N, Gonzales K, Sahu A, Tran L, Vogel A, Mazic M, Wilson JL, Forisch S, Mayr H, Oberle R, Weiszmann J, Brenner M, Vanhoutte R, Hofmann M, Pirnes-Karhu S, Magnes C, Kuhnast T, Weckwerth W, Bock C, Klavins K, Hengstschlager M, Moissl-Eichinger C, Schabbauer G, Egger G, Pirinen E, Verhelst SHL, Weichhart T (2023) Metabolic support by macrophages sustains colonic epithelial homeostasis. Cell Metab 35(1931–43):e8 [DOI] [PubMed] [Google Scholar]
- 92.Van den Bossche J, Lamers WH, Koehler ES, Geuns JM, Alhonen L, Uimari A, Pirnes-Karhu S, Van Overmeire E, Morias Y, Brys L, Vereecke L, De Baetselier P, Van Ginderachter JA (2012) Pivotal Advance: Arginase-1-independent polyamine production stimulates the expression of IL-4-induced alternatively activated macrophage markers while inhibiting LPS-induced expression of inflammatory genes. J Leukoc Biol 91:685–699 [DOI] [PubMed] [Google Scholar]
- 93.Van den Bossche J, Bogaert P, van Hengel J, Guerin CJ, Berx G, Movahedi K, Van den Bergh R, Pereira-Fernandes A, Geuns JM, Pircher H, Dorny P, Grooten J, De Baetselier P, Van Ginderachter JA (2009) Alternatively activated macrophages engage in homotypic and heterotypic interactions through IL-4 and polyamine-induced E-cadherin/catenin complexes. Blood 114:4664–4674 [DOI] [PubMed] [Google Scholar]
- 94.Puleston DJ, Buck MD, Klein Geltink RI, Kyle RL, Caputa G, O’Sullivan D, Cameron AM, Castoldi A, Musa Y, Kabat AM, Zhang Y, Flachsmann LJ, Field CS, Patterson AE, Scherer S, Alfei F, Baixauli F, Austin SK, Kelly B, Matsushita M, Curtis JD, Grzes KM, Villa M, Corrado M, Sanin DE, Qiu J, Pallman N, Paz K, Maccari ME, Blazar BR, Mittler G, Buescher JM, Zehn D, Rospert S, Pearce EJ, Balabanov S, Pearce EL (2019) Polyamines and eIF5A hypusination modulate mitochondrial respiration and macrophage activation. Cell Metab 30(352–63):e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Al-Habsi M, Chamoto K, Matsumoto K, Nomura N, Zhang B, Sugiura Y, Sonomura K, Maharani A, Nakajima Y, Wu Y, Nomura Y, Menzies R, Tajima M, Kitaoka K, Haku Y, Delghandi S, Yurimoto K, Matsuda F, Iwata S, Ogura T, Fagarasan S, Honjo T (2022) Spermidine activates mitochondrial trifunctional protein and improves antitumor immunity in mice. Science 378:eabj3510 [DOI] [PubMed] [Google Scholar]
- 96.Hofer SJ, Simon AK, Bergmann M, Eisenberg T, Kroemer G, Madeo F (2022) Mechanisms of spermidine-induced autophagy and geroprotection. Nat Aging 2:1112–1129 [DOI] [PubMed] [Google Scholar]
- 97.Eisenberg T, Knauer H, Schauer A, Buttner S, Ruckenstuhl C, Carmona-Gutierrez D, Ring J, Schroeder S, Magnes C, Antonacci L, Fussi H, Deszcz L, Hartl R, Schraml E, Criollo A, Megalou E, Weiskopf D, Laun P, Heeren G, Breitenbach M, Grubeck-Loebenstein B, Herker E, Fahrenkrog B, Frohlich KU, Sinner F, Tavernarakis N, Minois N, Kroemer G, Madeo F (2009) Induction of autophagy by spermidine promotes longevity. Nat Cell Biol 11:1305–1314 [DOI] [PubMed] [Google Scholar]
- 98.Germic N, Frangez Z, Yousefi S, Simon HU (2019) Regulation of the innate immune system by autophagy: monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ 26:715–727 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jiang J, Wang W, Sun F, Zhang Y, Liu Q, Yang D (2021) Bacterial infection reinforces host metabolic flux from arginine to spermine for NLRP3 inflammasome evasion. Cell Rep 34:108832 [DOI] [PubMed] [Google Scholar]
- 100.McCubbrey AL, McManus SA, McClendon JD, Thomas SM, Chatwin HB, Reisz JA, D’Alessandro A, Mould KJ, Bratton DL, Henson PM, Janssen WJ (2022) Polyamine import and accumulation causes immunomodulation in macrophages engulfing apoptotic cells. Cell Rep 38:110222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Yurdagul A Jr, Subramanian M, Wang X, Crown SB, Ilkayeva OR, Darville L, Kolluru GK, Rymond CC, Gerlach BD, Zheng Z, Kuriakose G, Kevil CG, Koomen JM, Cleveland JL, Muoio DM, Tabas I (2020) Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab 31(518–33):e10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Medina CB, Mehrotra P, Arandjelovic S, Perry JSA, Guo Y, Morioka S, Barron B, Walk SF, Ghesquiere B, Krupnick AS, Lorenz U, Ravichandran KS (2020) Metabolites released from apoptotic cells act as tissue messengers. Nature 580:130–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Campbell L, Saville CR, Murray PJ, Cruickshank SM, Hardman MJ (2013) Local arginase 1 activity is required for cutaneous wound healing. J Invest Dermatol 133:2461–2470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Yang Y, Ma L, Song M, Li X, He F, Wang C, Chen M, Zhou J, Mei C (2020) The role of the complement factor B-arginase-polyamine molecular axis in uremia-induced cardiac remodeling in mice. Eur J Immunol 50:220–233 [DOI] [PubMed] [Google Scholar]
- 105.Rahim AB, Lim HK, Tan CYR, Jia L, Leo VI, Uemura T, Hardman-Smart J, Common JEA, Lim TC, Bellanger S, Paus R, Igarashi K, Yang H, Vardy LA (2021) The polyamine regulator AMD1 upregulates spermine levels to drive epidermal differentiation. J Invest Dermatol 141(2178–88):e6 [DOI] [PubMed] [Google Scholar]
- 106.Heitmann L, Abad Dar M, Schreiber T, Erdmann H, Behrends J, McKenzie AN, Brombacher F, Ehlers S, Holscher C (2014) The IL-13/IL-4Ralpha axis is involved in tuberculosis-associated pathology. J Pathol 234:338–350 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Schworer S, Berisa M, Violante S, Qin W, Zhu J, Hendrickson RC, Cross JR, Thompson CB (2020) Proline biosynthesis is a vent for TGFbeta-induced mitochondrial redox stress. EMBO J 39:e103334 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Patriarca EJ, Cermola F, D’Aniello C, Fico A, Guardiola O, De Cesare D, Minchiotti G (2021) The multifaceted roles of proline in cell behavior. Front Cell Dev Biol 9:728576 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Pellegrinelli V, Rodriguez-Cuenca S, Rouault C, Figueroa-Juarez E, Schilbert H, Virtue S, Moreno-Navarrete JM, Bidault G, Vazquez-Borrego MC, Dias AR, Pucker B, Dale M, Campbell M, Carobbio S, Lin YH, Vacca M, Aron-Wisnewsky J, Mora S, Masiero MM, Emmanouilidou A, Mukhopadhyay S, Dougan G, den Hoed M, Loos RJF, Fernandez-Real JM, Chiarugi D, Clement K, Vidal-Puig A (2022) Dysregulation of macrophage PEPD in obesity determines adipose tissue fibro-inflammation and insulin resistance. Nat Metab 4:476–494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Li H, Ma L, Li W, Zheng B, Wang J, Chen S, Wang Y, Ge F, Qin B, Zheng X, Deng Y, Zeng R (2022) Proline metabolism reprogramming of trained macrophages induced by early respiratory infection combined with allergen sensitization contributes to development of allergic asthma in childhood of mice. Front Immunol 13:977235 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Silao FGS, Ward M, Ryman K, Wallstrom A, Brindefalk B, Udekwu K, Ljungdahl PO (2019) Mitochondrial proline catabolism activates Ras1/cAMP/PKA-induced filamentation in Candida albicans. PLoS Genet 15:e1007976 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Wyss M, Kaddurah-Daouk R (2000) Creatine and creatinine metabolism. Physiol Rev 80:1107–1213 [DOI] [PubMed] [Google Scholar]
- 113.Barcelos RP, Stefanello ST, Mauriz JL, Gonzalez-Gallego J, Soares FA (2016) Creatine and the liver: metabolism and possible interactions. Mini Rev Med Chem 16:12–18 [DOI] [PubMed] [Google Scholar]
- 114.Kazak L, Cohen P (2020) Creatine metabolism: energy homeostasis, immunity and cancer biology. Nat Rev Endocrinol 16:421–436 [DOI] [PubMed] [Google Scholar]
- 115.Ji L, Zhao X, Zhang B, Kang L, Song W, Zhao B, Xie W, Chen L, Hu X (2019) Slc6a8-Mediated creatine uptake and accumulation reprogram macrophage polarization via regulating cytokine responses. Immunity 51(272–84):e7 [DOI] [PubMed] [Google Scholar]
- 116.Yu L, Wang L, Hu G, Ren L, Qiu C, Li S, Zhou X, Chen S, Chen R (2022) Reprogramming alternative macrophage polarization by GATM-mediated endogenous creatine synthesis: a potential target for HDM-induced asthma treatment. Front Immunol 13:937331 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Ulland TK, Song WM, Huang SC, Ulrich JD, Sergushichev A, Beatty WL, Loboda AA, Zhou Y, Cairns NJ, Kambal A, Loginicheva E, Gilfillan S, Cella M, Virgin HW, Unanue ER, Wang Y, Artyomov MN, Holtzman DM, Colonna M (2017) TREM2 maintains microglial metabolic fitness in alzheimer’s disease. Cell 170(649–63):e13 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Kuiper JW, Pluk H, Oerlemans F, van Leeuwen FN, de Lange F, Fransen J, Wieringa B (2008) Creatine kinase-mediated ATP supply fuels actin-based events in phagocytosis. PLoS Biol 6:e51 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Hotchkiss RS, Moldawer LL, Opal SM, Reinhart K, Turnbull IR, Vincent JL (2016) Sepsis and septic shock. Nat Rev Dis Primers 2:16045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Luiking YC, Poeze M, Ramsay G, Deutz NE (2009) Reduced citrulline production in sepsis is related to diminished de novo arginine and nitric oxide production. Am J Clin Nutr 89:142–152 [DOI] [PubMed] [Google Scholar]
- 121.Wijnands KA, Castermans TM, Hommen MP, Meesters DM, Poeze M (2015) Arginine and citrulline and the immune response in sepsis. Nutrients 7:1426–1463 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Jensen IJ, Sjaastad FV, Griffith TS, Badovinac VP (2018) Sepsis-Induced T cell immunoparalysis: the ins and outs of impaired T cell immunity. J Immunol 200:1543–1553 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Reizine F, Gregoire M, Lesouhaitier M, Coirier V, Gauthier J, Delaloy C, Dessauge E, Creusat F, Uhel F, Gacouin A, Dessauge F, Le Naoures C, Moreau C, Bendavid C, Daniel Y, Petitjean K, Bordeau V, Lamaison C, Piau C, Cattoir V, Roussel M, Fromenty B, Michelet C, Le Tulzo Y, Zmijewski J, Thibault R, Cogne M, Tarte K, Tadie JM (2022) Beneficial effects of citrulline enteral administration on sepsis-induced T cell mitochondrial dysfunction. Proc Natl Acad Sci U S A 119:e2115139119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Rosenthal MD, Carrott PW, Patel J, Kiraly L, Martindale RG (2016) Parenteral or enteral arginine supplementation safety and efficacy. J Nutr 146:2594S-S2600 [DOI] [PubMed] [Google Scholar]
- 125.Colonna M, Butovsky O (2017) Microglia function in the central nervous system during health and neurodegeneration. Annu Rev Immunol 35:441–468 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Stratoulias V, Ruiz R, Kanatani S, Osman AM, Keane L, Armengol JA, Rodriguez-Moreno A, Murgoci AN, Garcia-Dominguez I, Alonso-Bellido I, Gonzalez Ibanez F, Picard K, Vazquez-Cabrera G, Posada-Perez M, Vernoux N, Tejera D, Grabert K, Cheray M, Gonzalez-Rodriguez P, Perez-Villegas EM, Martinez-Gallego I, Lastra-Romero A, Brodin D, Avila-Carino J, Cao Y, Airavaara M, Uhlen P, Heneka MT, Tremblay ME, Blomgren K, Venero JL, Joseph B (2023) ARG1-expressing microglia show a distinct molecular signature and modulate postnatal development and function of the mouse brain. Nat Neurosci 26:1008–1020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Cai W, Dai X, Chen J, Zhao J, Xu M, Zhang L, Yang B, Zhang W, Rocha M, Nakao T, Kofler J, Shi Y, Stetler RA, Hu X, Chen J (2019) STAT6/Arg1 promotes microglia/macrophage efferocytosis and inflammation resolution in stroke mice. JCI Insight 4:e131355 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Fenn AM, Hall JC, Gensel JC, Popovich PG, Godbout JP (2014) IL-4 signaling drives a unique arginase+/IL-1beta+ microglia phenotype and recruits macrophages to the inflammatory CNS: consequences of age-related deficits in IL-4Ralpha after traumatic spinal cord injury. J Neurosci 34:8904–8917 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Hunt JB Jr, Nash KR, Placides D, Moran P, Selenica ML, Abuqalbeen F, Ratnasamy K, Slouha N, Rodriguez-Ospina S, Savlia M, Ranaweera Y, Reid P, Dickey CA, Uricia R, Yang CG, Sandusky LA, Gordon MN, Morgan D, Lee DC (2015) Sustained arginase 1 expression modulates pathological tau deposits in a mouse model of tauopathy. J Neurosci 35:14842–14860 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Cherry JD, Olschowka JA, O’Banion MK (2015) Arginase 1+ microglia reduce Abeta plaque deposition during IL-1beta-dependent neuroinflammation. J Neuroinflammation 12:203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Kan MJ, Lee JE, Wilson JG, Everhart AL, Brown CM, Hoofnagle AN, Jansen M, Vitek MP, Gunn MD, Colton CA (2015) Arginine deprivation and immune suppression in a mouse model of Alzheimer’s disease. J Neurosci 35:5969–5982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, van Wijngaarden P, Wagers AJ, Williams A, Franklin RJM, Ffrench-Constant C (2013) M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 16:1211–1218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Standiford MM, Grund EM, Howe CL (2021) Citrullinated myelin induces microglial TNFalpha and inhibits endogenous repair in the cuprizone model of demyelination. J Neuroinflammation 18:305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Pegg AE (2016) Functions of Polyamines in Mammals. J Biol Chem 291:14904–14912 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Sandusky-Beltran LA, Kovalenko A, Placides DS, Ratnasamy K, Ma C, Hunt JB Jr, Liang H, Calahatian JIT, Michalski C, Fahnestock M, Blair LJ, Darling AL, Baker JD, Fontaine SN, Dickey CA, Gamsby JJ, Nash KR, Abner E, Selenica MB, Lee DC (2021) Aberrant AZIN2 and polyamine metabolism precipitates tau neuropathology. J Clin Invest 131(4):e126299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Lewandowski NM, Ju S, Verbitsky M, Ross B, Geddie ML, Rockenstein E, Adame A, Muhammad A, Vonsattel JP, Ringe D, Cote L, Lindquist S, Masliah E, Petsko GA, Marder K, Clark LN, Small SA (2010) Polyamine pathway contributes to the pathogenesis of Parkinson disease. Proc Natl Acad Sci U S A 107:16970–16975 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Soulet D, Rivest S (2003) Polyamines play a critical role in the control of the innate immune response in the mouse central nervous system. J Cell Biol 162:257–268 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Freitag K, Sterczyk N, Wendlinger S, Obermayer B, Schulz J, Farztdinov V, Mulleder M, Ralser M, Houtman J, Fleck L, Braeuning C, Sansevrino R, Hoffmann C, Milovanovic D, Sigrist SJ, Conrad T, Beule D, Heppner FL, Jendrach M (2022) Spermidine reduces neuroinflammation and soluble amyloid beta in an Alzheimer’s disease mouse model. J Neuroinflammation 19:172 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Yang Q, Zheng C, Cao J, Cao G, Shou P, Lin L, Velletri T, Jiang M, Chen Q, Han Y, Li F, Wang Y, Cao W, Shi Y (2016) Spermidine alleviates experimental autoimmune encephalomyelitis through inducing inhibitory macrophages. Cell Death Differ 23:1850–1861 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Veis DJ, O’Brien CA (2023) Osteoclasts, master sculptors of bone. Annu Rev Pathol 18:257–281 [DOI] [PubMed] [Google Scholar]
- 141.Cao S, Li Y, Song R, Meng X, Fuchs M, Liang C, Kachler K, Meng X, Wen J, Schlotzer-Schrehardt U, Taudte V, Gessner A, Kunz M, Schleicher U, Zaiss MM, Kastbom A, Chen X, Schett G, Bozec A (2024) L-arginine metabolism inhibits arthritis and inflammatory bone loss. Ann Rheum Dis 83:72–87 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Brunner JS, Vulliard L, Hofmann M, Kieler M, Lercher A, Vogel A, Russier M, Bruggenthies JB, Kerndl M, Saferding V, Niederreiter B, Junza A, Frauenstein A, Scholtysek C, Mikami Y, Klavins K, Kronke G, Bergthaler A, O’Shea JJ, Weichhart T, Meissner F, Smolen JS, Cheng P, Yanes O, Menche J, Murray PJ, Sharif O, Bluml S, Schabbauer G (2020) Environmental arginine controls multinuclear giant cell metabolism and formation. Nat Commun 11:431 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kondo T, Otsuka Y, Aoki H, Goto Y, Kawaguchi Y, Waguri-Nagaya Y, Miyazawa K, Goto S, Aoyama M (2021) The inducible nitric oxide synthase pathway promotes osteoclastogenesis under hypoxic culture conditions. Am J Pathol 191:2072–2079 [DOI] [PubMed] [Google Scholar]
- 144.Nilforoushan D, Gramoun A, Glogauer M, Manolson MF (2009) Nitric oxide enhances osteoclastogenesis possibly by mediating cell fusion. Nitric Oxide 21:27–36 [DOI] [PubMed] [Google Scholar]
- 145.Watanuki M, Sakai A, Sakata T, Tsurukami H, Miwa M, Uchida Y, Watanabe K, Ikeda K, Nakamura T (2002) Role of inducible nitric oxide synthase in skeletal adaptation to acute increases in mechanical loading. J Bone Miner Res 17:1015–1025 [DOI] [PubMed] [Google Scholar]
- 146.Gyurko R, Shoji H, Battaglino RA, Boustany G, Gibson FC 3rd, Genco CA, Stashenko P, Van Dyke TE (2005) Inducible nitric oxide synthase mediates bone development and P. gingivalis-induced alveolar bone loss. Bone 36:472–479 [DOI] [PubMed] [Google Scholar]
- 147.Jin Z, Kho J, Dawson B, Jiang MM, Chen Y, Ali S, Burrage LC, Grover M, Palmer DJ, Turner DL, Ng P, Nagamani SC, Lee B (2021) Nitric oxide modulates bone anabolism through regulation of osteoblast glycolysis and differentiation. J Clin Invest 131(5):e138935 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Krishnamurthy A, Joshua V, Haj Hensvold A, Jin T, Sun M, Vivar N, Ytterberg AJ, Engstrom M, Fernandes-Cerqueira C, Amara K, Magnusson M, Wigerblad G, Kato J, Jimenez-Andrade JM, Tyson K, Rapecki S, Lundberg K, Catrina SB, Jakobsson PJ, Svensson C, Malmstrom V, Klareskog L, Wahamaa H, Catrina AI (2016) Identification of a novel chemokine-dependent molecular mechanism underlying rheumatoid arthritis-associated autoantibody-mediated bone loss. Ann Rheum Dis 75:721–729 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Bronte V, Brandau S, Chen SH, Colombo MP, Frey AB, Greten TF, Mandruzzato S, Murray PJ, Ochoa A, Ostrand-Rosenberg S, Rodriguez PC, Sica A, Umansky V, Vonderheide RH, Gabrilovich DI (2016) Recommendations for myeloid-derived suppressor cell nomenclature and characterization standards. Nat Commun 7:12150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bied M, Ho WW, Ginhoux F, Bleriot C (2023) Roles of macrophages in tumor development: a spatiotemporal perspective. Cell Mol Immunol 20:983–992 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam V, Cyrus N, Brokowski CE, Eisenbarth SC, Phillips GM, Cline GW, Phillips AJ, Medzhitov R (2014) Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 513:559–563 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Geeraerts X, Fernandez-Garcia J, Hartmann FJ, de Goede KE, Martens L, Elkrim Y, Debraekeleer A, Stijlemans B, Vandekeere A, Rinaldi G, De Rycke R, Planque M, Broekaert D, Meinster E, Clappaert E, Bardet P, Murgaski A, Gysemans C, Nana FA, Saeys Y, Bendall SC, Laoui D, Van den Bossche J, Fendt SM, Van Ginderachter JA (2021) Macrophages are metabolically heterogeneous within the tumor microenvironment. Cell Rep 37:110171 [DOI] [PubMed] [Google Scholar]
- 153.LaRue MM, Parker S, Puccini J, Cammer M, Kimmelman AC, Bar-Sagi D (2022) Metabolic reprogramming of tumor-associated macrophages by collagen turnover promotes fibrosis in pancreatic cancer. Proc Natl Acad Sci U S A 119:e2119168119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Su X, Xu Y, Fox GC, Xiang J, Kwakwa KA, Davis JL, Belle JI, Lee WC, Wong WH, Fontana F, Hernandez-Aya LF, Kobayashi T, Tomasson HM, Su J, Bakewell SJ, Stewart SA, Egbulefu C, Karmakar P, Meyer MA, Veis DJ, DeNardo DG, Lanza GM, Achilefu S, Weilbaecher KN (2021) Breast cancer-derived GM-CSF regulates arginase 1 in myeloid cells to promote an immunosuppressive microenvironment. J Clin Invest 131(20):e145296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Zhan Y, Lew AM, Chopin M (2019) The pleiotropic effects of the GM-CSF rheostat on myeloid cell differentiation and function: more than a numbers game. Front Immunol 10:2679 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Hajji N, Garcia-Revilla J, Soto MS, Perryman R, Symington J, Quarles CC, Healey DR, Guo Y, Orta-Vazquez ML, Mateos-Cordero S, Shah K, Bomalaski J, Anichini G, Tzakos AG, Crook T, O’Neill K, Scheck AC, Venero JL, Syed N (2022) Arginine deprivation alters microglial polarity and synergizes with radiation to eradicate non-arginine-auxotrophic glioblastoma tumors. J Clin Invest 132(6):e142137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Dussold C, Zilinger K, Turunen J, Heimberger AB, Miska J (2024) Modulation of macrophage metabolism as an emerging immunotherapy strategy for cancer. J Clin Invest 134(2):e175445 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Dahal A, Hong Y, Mathew JS, Geber A, Eckl S, Renner S, Sailer CJ, Ryan AT, Mir S, Lim K, Linehan DC, Gerber SA, Kim M (2024) Platelet-activating factor (PAF) promotes immunosuppressive neutrophil differentiation within tumors. Proc Natl Acad Sci U S A 121:e2406748121 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Rodriguez PC, Zea AH, Culotta KS, Zabaleta J, Ochoa JB, Ochoa AC (2002) Regulation of T cell receptor CD3zeta chain expression by L-arginine. J Biol Chem 277:21123–21129 [DOI] [PubMed] [Google Scholar]
- 160.Rodriguez PC, Quiceno DG, Ochoa AC (2007) L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood 109:1568–1573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Rodriguez PC, Quiceno DG, Zabaleta J, Ortiz B, Zea AH, Piazuelo MB, Delgado A, Correa P, Brayer J, Sotomayor EM, Antonia S, Ochoa JB, Ochoa AC (2004) Arginase I production in the tumor microenvironment by mature myeloid cells inhibits T-cell receptor expression and antigen-specific T-cell responses. Cancer Res 64:5839–5849 [DOI] [PubMed] [Google Scholar]
- 162.Rodriguez PC, Zea AH, DeSalvo J, Culotta KS, Zabaleta J, Quiceno DG, Ochoa JB, Ochoa AC (2003) L-arginine consumption by macrophages modulates the expression of CD3 zeta chain in T lymphocytes. J Immunol 171:1232–1239 [DOI] [PubMed] [Google Scholar]
- 163.Steggerda SM, Bennett MK, Chen J, Emberley E, Huang T, Janes JR, Li W, MacKinnon AL, Makkouk A, Marguier G, Murray PJ, Neou S, Pan A, Parlati F, Rodriguez MLM, Van de Velde LA, Wang T, Works M, Zhang J, Zhang W, Gross MI (2017) Inhibition of arginase by CB-1158 blocks myeloid cell-mediated immune suppression in the tumor microenvironment. J Immunother Cancer 5:101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Sippel TR, White J, Nag K, Tsvankin V, Klaassen M, Kleinschmidt-DeMasters BK, Waziri A (2011) Neutrophil degranulation and immunosuppression in patients with GBM: restoration of cellular immune function by targeting arginase I. Clin Cancer Res 17:6992–7002 [DOI] [PubMed] [Google Scholar]
- 165.Oberlies J, Watzl C, Giese T, Luckner C, Kropf P, Muller I, Ho AD, Munder M (2009) Regulation of NK cell function by human granulocyte arginase. J Immunol 182:5259–5267 [DOI] [PubMed] [Google Scholar]
- 166.Zhang H, Zhu X, Friesen TJ, Kwak JW, Pisarenko T, Mekvanich S, Velasco MA, Randolph TW, Kargl J, Houghton AM (2022) Annexin A2/TLR2/MYD88 pathway induces arginase 1 expression in tumor-associated neutrophils. J Clin Invest 132(22):e153643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Nagaraj S, Schrum AG, Cho HI, Celis E, Gabrilovich DI (2010) Mechanism of T cell tolerance induced by myeloid-derived suppressor cells. J Immunol 184:3106–3116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Patil MD, Bhaumik J, Babykutty S, Banerjee UC, Fukumura D (2016) Arginine dependence of tumor cells: targeting a chink in cancer’s armor. Oncogene 35:4957–4972 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Granados-Principal S, Liu Y, Guevara ML, Blanco E, Choi DS, Qian W, Patel T, Rodriguez AA, Cusimano J, Weiss HL, Zhao H, Landis MD, Dave B, Gross SS, Chang JC (2015) Inhibition of iNOS as a novel effective targeted therapy against triple-negative breast cancer. Breast Cancer Res 17:25 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Eyler CE, Wu Q, Yan K, MacSwords JM, Chandler-Militello D, Misuraca KL, Lathia JD, Forrester MT, Lee J, Stamler JS, Goldman SA, Bredel M, McLendon RE, Sloan AE, Hjelmeland AB, Rich JN (2011) Glioma stem cell proliferation and tumor growth are promoted by nitric oxide synthase-2. Cell 146:53–66 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Reddy TP, Glynn SA, Billiar TR, Wink DA, Chang JC (2023) Targeting nitric oxide: say NO to metastasis. Clin Cancer Res 29:1855–1868 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Apiz Saab JJ, Dzierozynski LN, Jonker PB, AminiTabrizi R, Shah H, Menjivar RE, Scott AJ, Nwosu ZC, Zhu Z, Chen RN, Oh M, Sheehan C, Wahl DR, Pasca di Magliano M, Lyssiotis CA, Macleod KF, Weber CR, Muir A (2023) Pancreatic tumors exhibit myeloid-driven amino acid stress and upregulate arginine biosynthesis. Elife 12:e81289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Chia TY, Zolp A, Miska J (2022) Polyamine immunometabolism: central regulators of inflammation, cancer and autoimmunity. Cells 11:896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Miska J, Rashidi A, Lee-Chang C, Gao P, Lopez-Rosas A, Zhang P, Burga R, Castro B, Xiao T, Han Y, Hou D, Sampat S, Cordero A, Stoolman JS, Horbinski CM, Burns M, Reshetnyak YK, Chandel NS, Lesniak MS (2021) Polyamines drive myeloid cell survival by buffering intracellular pH to promote immunosuppression in glioblastoma. Sci Adv 7:eabc8929 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Ye C, Geng Z, Dominguez D, Chen S, Fan J, Qin L, Long A, Zhang Y, Kuzel TM, Zhang B (2016) Targeting ornithine decarboxylase by alpha-difluoromethylornithine inhibits tumor growth by impairing myeloid-derived suppressor cells. J Immunol 196:915–923 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Alexander ET, Minton A, Peters MC, Ot P, Gilmour SK (2017) A novel polyamine blockade therapy activates an anti-tumor immune response. Oncotarget 8:84140–84152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Rashidi A, Billingham LK, Zolp A, Chia TY, Silvers C, Katz JL, Park CH, Delay S, Boland L, Geng Y, Markwell SM, Dmello C, Arrieta VA, Zilinger K, Jacob IM, Lopez-Rosas A, Hou D, Castro B, Steffens AM, McCortney K, Walshon JP, Flowers MS, Lin H, Wang H, Zhao J, Sonabend A, Zhang P, Ahmed AU, Brat DJ, Heiland DH, Lee-Chang C, Lesniak MS, Chandel NS, Miska J (2024) Myeloid cell-derived creatine in the hypoxic niche promotes glioblastoma growth. Cell Metab 36(62–77):e8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Veglia F, Sanseviero E, Gabrilovich DI (2021) Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol 21:485–498 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Rodriguez PC, Ochoa AC (2008) Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev 222:180–191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Wu Y, Yi M, Niu M, Mei Q, Wu K (2022) Myeloid-derived suppressor cells: an emerging target for anticancer immunotherapy. Mol Cancer 21:184 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Movahedi K, Guilliams M, Van den Bossche J, Van den Bergh R, Gysemans C, Beschin A, De Baetselier P, Van Ginderachter JA (2008) Identification of discrete tumor-induced myeloid-derived suppressor cell subpopulations with distinct T cell-suppressive activity. Blood 111:4233–4244 [DOI] [PubMed] [Google Scholar]
- 182.Aarts CEM, Kuijpers TW (2018) Neutrophils as myeloid-derived suppressor cells. Eur J Clin Invest 48(Suppl 2):e12989 [DOI] [PubMed] [Google Scholar]
- 183.Miret JJ, Kirschmeier P, Koyama S, Zhu M, Li YY, Naito Y, Wu M, Malladi VS, Huang W, Walker W, Palakurthi S, Dranoff G, Hammerman PS, Pecot CV, Wong KK, Akbay EA (2019) Suppression of Myeloid Cell Arginase Activity leads to Therapeutic Response in a NSCLC Mouse Model by Activating Anti-Tumor Immunity. J Immunother Cancer 7:32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Vonwirth V, Bulbul Y, Werner A, Echchannaoui H, Windschmitt J, Habermeier A, Ioannidis S, Shin N, Conradi R, Bros M, Tenzer S, Theobald M, Closs EI, Munder M (2020) Inhibition of arginase 1 liberates potent T cell immunostimulatory activity of human neutrophil granulocytes. Front Immunol 11:617699 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Pallett LJ, Gill US, Quaglia A, Sinclair LV, Jover-Cobos M, Schurich A, Singh KP, Thomas N, Das A, Chen A, Fusai G, Bertoletti A, Cantrell DA, Kennedy PT, Davies NA, Haniffa M, Maini MK (2015) Metabolic regulation of hepatitis B immunopathology by myeloid-derived suppressor cells. Nat Med 21:591–600 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, Ochoa AC (2009) Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res 69:1553–1560 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Wang Y, Tian J, Tang X, Rui K, Tian X, Ma J, Ma B, Xu H, Lu L, Wang S (2016) Exosomes released by granulocytic myeloid-derived suppressor cells attenuate DSS-induced colitis in mice. Oncotarget 7:15356–15368 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Zea AH, Rodriguez PC, Atkins MB, Hernandez C, Signoretti S, Zabaleta J, McDermott D, Quiceno D, Youmans A, O’Neill A, Mier J, Ochoa AC (2005) Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res 65:3044–3048 [DOI] [PubMed] [Google Scholar]
- 189.Bian Z, Abdelaal AM, Shi L, Liang H, Xiong L, Kidder K, Venkataramani M, Culpepper C, Zen K, Liu Y (2018) Arginase-1 is neither constitutively expressed in nor required for myeloid-derived suppressor cell-mediated inhibition of T-cell proliferation. Eur J Immunol 48:1046–1058 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Rodriguez PC, Hernandez CP, Quiceno D, Dubinett SM, Zabaleta J, Ochoa JB, Gilbert J, Ochoa AC (2005) Arginase I in myeloid suppressor cells is induced by COX-2 in lung carcinoma. J Exp Med 202:931–939 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Vasquez-Dunddel D, Pan F, Zeng Q, Gorbounov M, Albesiano E, Fu J, Blosser RL, Tam AJ, Bruno T, Zhang H, Pardoll D, Kim Y (2013) STAT3 regulates arginase-I in myeloid-derived suppressor cells from cancer patients. J Clin Invest 123:1580–1589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Mohammadpour H, MacDonald CR, Qiao G, Chen M, Dong B, Hylander BL, McCarthy PL, Abrams SI, Repasky EA (2019) beta2 adrenergic receptor-mediated signaling regulates the immunosuppressive potential of myeloid-derived suppressor cells. J Clin Invest 129:5537–5552 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Fletcher M, Ramirez ME, Sierra RA, Raber P, Thevenot P, Al-Khami AA, Sanchez-Pino D, Hernandez C, Wyczechowska DD, Ochoa AC, Rodriguez PC (2015) l-Arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells. Cancer Res 75:275–283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Nagaraj S, Gupta K, Pisarev V, Kinarsky L, Sherman S, Kang L, Herber DL, Schneck J, Gabrilovich DI (2007) Altered recognition of antigen is a mechanism of CD8+ T cell tolerance in cancer. Nat Med 13:828–835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Niu F, Yu Y, Li Z, Ren Y, Li Z, Ye Q, Liu P, Ji C, Qian L, Xiong Y (2022) Arginase: an emerging and promising therapeutic target for cancer treatment. Biomed Pharmacother 149:112840 [DOI] [PubMed] [Google Scholar]
- 196.Iannone F, Porzia A, Peruzzi G, Birarelli P, Milana B, Sacco L, Dinatale G, Peparini N, Prezioso G, Battella S, Caronna R, Morrone S, Palmieri G, Mainiero F, Chirletti P (2015) Effect of surgery on pancreatic tumor-dependent lymphocyte asset: modulation of natural killer cell frequency and cytotoxic function. Pancreas 44:386–393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Reinhardt R, Pohlmann S, Kleinertz H, Hepner-Schefczyk M, Paul A, Flohe SB (2015) Invasive surgery impairs the regulatory function of human CD56 bright natural killer cells in response to staphylococcus aureus. Suppression of interferon-gamma synthesis. PLoS One 10:e0130155 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Tai LH, de Souza CT, Belanger S, Ly L, Alkayyal AA, Zhang J, Rintoul JL, Ananth AA, Lam T, Breitbach CJ, Falls TJ, Kirn DH, Bell JC, Makrigiannis AP, Auer RA (2013) Preventing postoperative metastatic disease by inhibiting surgery-induced dysfunction in natural killer cells. Cancer Res 73:97–107 [DOI] [PubMed] [Google Scholar]
- 199.Tai LH, Alkayyal AA, Leslie AL, Sahi S, Bennett S, Tanese de Souza C, Baxter K, Angka L, Xu R, Kennedy MA, Auer RC (2018) Phosphodiesterase-5 inhibition reduces postoperative metastatic disease by targeting surgery-induced myeloid derived suppressor cell-dependent inhibition of Natural Killer cell cytotoxicity. Oncoimmunology 7:e1431082 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Angka L, Tanese de Souza C, Baxter KE, Khan ST, Market M, Martel AB, Tai LH, Kennedy MA, Bell JC, Auer RC (2022) Perioperative arginine prevents metastases by accelerating natural killer cell recovery after surgery. Mol Ther 30:3270–3283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Hubner M, Tomasi R, Effinger D, Wu T, Klein G, Bender M, Kilger E, Juchem G, Schwedhelm E, Kreth S (2019) Myeloid-Derived suppressor cells mediate immunosuppression after cardiopulmonary bypass. Crit Care Med 47:e700–e709 [DOI] [PubMed] [Google Scholar]
- 202.Ochoa JB, Bernard AC, O’Brien WE, Griffen MM, Maley ME, Rockich AK, Tsuei BJ, Boulanger BR, Kearney PA, Morris SM Jr (2001) Arginase I expression and activity in human mononuclear cells after injury. Ann Surg 233:393–399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Lamas B, Vergnaud-Gauduchon J, Goncalves-Mendes N, Perche O, Rossary A, Vasson MP, Farges MC (2012) Altered functions of natural killer cells in response to L-Arginine availability. Cell Immunol 280:182–190 [DOI] [PubMed] [Google Scholar]
- 204.Adiamah A, Skorepa P, Weimann A, Lobo DN (2019) The impact of preoperative immune modulating nutrition on outcomes in patients undergoing surgery for gastrointestinal cancer: a systematic review and meta-analysis. Ann Surg 270:247–256 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No original data was generated for this review article.