Open access peer-reviewed chapter

Immunometabolic Processes of Macrophages in Disease States

Written By

Filex Otieno

Submitted: 07 January 2023 Reviewed: 10 January 2023 Published: 02 March 2023

DOI: 10.5772/intechopen.109936

From the Edited Volume

Phagocytosis - Main Key of Immune System

Edited by Seyyed Shamsadin Athari and Entezar Mehrabi Nasab

Chapter metrics overview

76 Chapter Downloads

View Full Metrics

Abstract

Macrophages are immune cells functioning primarily as antigen-presenting cells. They are professional phagocytes and patrol tissues within the body contributing to immunological surveillance. The majority of circulating macrophages and to some extend tissue-resident macrophages differentiate from monocytes. A few of resident macrophages do however originate from embryo during fetal development and remain capable of self-renewal even in adulthood. Macrophages are highly plastic seeing that they play a dual function in inflammatory conditions: either pro-inflammatory or anti-inflammatory. Depending on state of the body, whether disease, healing or homeostatic state, macrophages can be polarized to either one of two phenotypes-M1 macrophages or M2 macrophages. The former phenotype is associated with pro-inflammatory processes, while the latter mediates anti-inflammatory process. Metabolic process and intermediate substrates influence macrophage activation, polarization and functioning within the body. Moreover, within macrophages themselves, the metabolic pathways activated also influences their polarization. As such inflammatory conditions from either infectious agents or metabolic diseases are a major drive for macrophage activation that determines disease severity and prognosis seemingly because macrophages also activate other immune cells. This interplay between immune system and metabolism is of interest especially in development newer treatment strategies for metabolic diseases and infectious agents.

Keywords

  • immune response
  • infections
  • metabolic diseases
  • metabolism
  • polarization

1. Introduction

Macrophages are innate immune cells derived either from blood-circulating monocytes or tissue-resident persistent embryonic stem cells [1]. Monocytes are hematopoietic stem cells formed in adult bone marrow and once mature are released into systemic circulation from where they go into different body tissues [2]. Once in the tissue, monocytes differentiate into and become macrophages [2]. Depending on the body tissue invaded, monocyte-derived macrophages function primarily to add onto the pool of total tissue macrophages during an immune response [3]. Contrary, during embryogenesis, macrophage stem cells and monocyte progenitor cells remain localized within body tissues permanently till adulthood. These cells are capable of self-renewal and in adults result in formation of resident macrophages i.e. the progenitor cells of embryonic origin directly differentiate into tissue-specific resident macrophage with signature molecules explicit for a given tissue [4]. Primarily, they serve homeostatic functions within tissues such as iron metabolism, removal of dead apoptotic cellular debris, synthesis of surfactant etc. [5]. They, however, are also involved in immune response against foreign particles in conjunction with monocyte-derived macrophages. In fact, during an immune response in a particular tissue, tissue-specific macrophages are the first type of macrophages to respond. As their population diminishes within the tissue, monocyte-derived macrophages migrate into the tissue to add to the pool and also fight the invading foreign particle.

In general, macrophages are phagocytic cells that function as antigen processing cells during a foreign invasion [6]. They use pattern recognition receptors (PRRs) to detect pathogen associated molecular patterns (PAMPs) in pathogens and damage associated molecular patterns (DAMPs) in damaged cells [7]. These patterns constitute molecules express on cellular surface that denote cellular damage or presence of a pathogenic microbe etc. Once detected, macrophages engulf the supposed recognized particle through a cell eating process termed phagocytosis [8]. The foreign materials are then digested in a sac-like organelle called the phagolysosomes into smaller particles [9]. These breakdown particles are then expressed on surface of macrophage membrane together with major histocompatibility molecules (MHC) for lymphocytes to recognized and trigger cytotoxic and humoral effects [10].

Although the functioning of macrophages seems straightforward, it is rather a complex process of active immune response. Presence of foreign particles within tissue surroundings alters the tissue microenvironment [11]. This microenvironment acts as a signaling pathway for activating of naïve macrophages [11]. Macrophages can either be polarized to be pro-inflammatory or anti-inflammatory in nature. Classically activated macrophages are pro-inflammatory in nature and function to eradicate foreign particle [12]. They are also known as M1 macrophages. Alternatively activated macrophages or M2 macrophages are anti-inflammatory in nature and are recruited during resolution of inflammation when invader has been eradicated to promote tissue healing and removal of debris [12].

Altogether, the entire process from activation, polarization, functioning and return to homeostatic steady-state, the macrophage cell is regulated by the microenvironment surrounding it. This consist of signaling molecules and metabolites, either intermediate or finished, that influence the working of the cell. While immune signaling molecules such as cytokines is discussed in detailed in the field of immunology, the latter has been recently recognized and its crucial role in chronic inflammation associated with specific chronic diseases; especially those inflammatory in nature. Noteworthy, it has led to emergence of the field of immunometabolism which focuses on how metabolism affects functioning of immune components. This chapter reviews current knowledge of how metabolic processes and metabolites influences functioning of macrophages.

Advertisement

2. Immunometabolic processes of macrophages

The term metabolism sums up chemical processes that occur within the body of a living organism to maintain life. The basic unit of life, which is a cell, utilizes cellular processes to break down large organic compounds into energy depending on their biogenetics. Metabolism can be viewed as either cellular or systemic. Cellular metabolism occurs within a cell while the latter depicts metabolism in which specific organs/tissues are regarded as producers of metabolites which are then utilized by consumer organs/tissues. For instance, the liver metabolizes much of carbohydrate and iron to form metabolites e.g. glucose that other organs such as the brain and bone tissue utilize for their own cellular processes. However, the general outlook of metabolism predominates around cellular metabolism and for this review; five metabolic pathways will be discussed in relation to influence on macrophage function. These are: glycolysis, pentose phosphate pathway (PPP), Tri-carboxylic acid cycle (TCA), amino acid (AA) and fatty acid (FA) metabolism.

In terms of macrophage activation, hypoxia and danger signals induce HIF1-α which that stimulates the glycolytic pathway [13]. This leads to rapid generation of ATP and macrophage polarization to M1 phenotype. Contrary, helminths stimulate interleukin 4 release which in addition to normoxia activates the TCA and electron transport chain pathways typical of M2 macrophages [14].

2.1 Glycolysis

Glycolysis defines the breakdown of glucose into pyruvate. The process occurs within the cytosol. Glucose is a monosaccharide which represents the unit molecule for carbohydrate compounds. Glycolysis can occur under either aerobic or anaerobic conditions. In aerobic conditions, pyruvate is converted to acetyl CoA which enters the TCA cycle or FA synthesis [15]. In anaerobic conditions, pyruvate is converted to lactate which is removed from cells and taken to the liver to be converted back to pyruvate for gluconeogenesis. Glycolysis is a 10-step process that results in a net amount of 2-ATP molecules being formed per unit of glucose [15]. Additionally, NAD+ is reduced to NADH which is used as a cofactor in various anabolic conditions [16].

During an immune response, naïve macrophages are polarized to the M1 subset due to changes in the microenvironment. In particular, when macrophages are stimulated by lipopolysaccharide (LPS) and interferon γ (IFN-γ), expression of glucose transporter 1 (GLUT1) increases significantly unlike that of succinate dehydrogenase A (SDHA), which is a component of electron transport chain [17]. High levels of intracellular glucose activates the glycolytic process which has been seen to be significantly ongoing during infectious conditions and carcinogenesis [18]. Inhibition of hexokinase using 2-deoxy-D-glucose, the first glycolytic enzyme that phosphorylates glucose leads to a reduction in the amount of TNF-α and interleukin-12 (IL-12) produced [19]. Notably, the cytokines stated previous mediate pro-inflammatory reaction. In addition, hexokinase inhibition also reduces expression of CD80, CD86 and inducible nitric oxide syntheses (iNOS) which are co-stimulatory molecules in macrophages and marker of M1 differentiation respectively [20, 21]. Hexokinase 1 also functions as a regulator of NOD-LRR and pyrin domain-containing 3 (NLRP3) found in the outer membrane of mitochondria [22]. NLRP3 functions to regulate activity of caspase 1 which is involved in generation of active and mature IL-18 and -1β respectively. Increased glycolytic activity activates hexokinase 1 which in turn activates NLRP3 to induce cell death via pyroptosis in macrophages [22].

Noteworthy is the effect of isomeric forms of pyruvate kinase on macrophage polarization. Pyruvate kinase mediates last step of glycolysis by converting intermediate metabolite- phosphoenolpyruvate to pyruvate with concomitant generation of ATP. The enzyme exists as two isoforms either as pyruvate kinase M1 (PKM1) or M2 (PKM2) [23]. Macrophage stimulation by LPS induces expression of PKM2 which is associated with slowing the rate of pyruvate formation unlike PKM1 [24]. This allows glycolytic intermediate metabolites to accumulate and divert to other pathways. For instance, glucose-6-phosphate is diverted to the PPP pathway to generate ribose for nucleotide synthesis while 3-phosphoglycerate is shunted to serine synthesis process. In addition, the PKM2 enzyme translocate into the nucleus where it interacts with HIF1-α and induce expression of HIF1-α genes [24]. These genes are responsible for expression of inflammatory proteins such as IL-1β and glycolytic enzymes [23]. At times however, the PKM2 enzyme is stabilized in the tetrameric form rather than dimeric. This inhibits its translocation into the nucleus [24]. The enzyme then becomes more concentrated in cytosol and predominantly participate in glycolysis reducing its immune-related function.

Ultimately, interference with glycolytic process in macrophages during an immune response reduces their pro-inflammatory activity. This is because, M1 macrophages depend largely on glycolysis for energy production and subsequent use of such energy for survival and synthesis of mediator molecules [25]. Furthermore, it would seem that in organisms with dysfunctional glycolytic process, a chronic inflammatory process is likely to ensue with fibrotic resolution and formation of granulomas; as the pathogen is not sufficiently eradicated.

2.2 Pentose phosphate pathway

The pentose phosphate pathway is a shunt from the glycolytic process occurring in the cytosol and leads to formation of pentoses and a major contributor of total NADPH produced in the human body. The pathway is branched into oxidative branch leading to generation of NADPH and non-oxidative branch leading to formation of pentoses [26]. Pentose sugars are ultimately utilized in the synthesis of nucleotides and amino acids. Pentose phosphate pathway starts with glucose-6-phosphate branching off from glycolytic process to form 6-phosphogluconolactone that is oxidized via intermediates to form ribulose-5-phosphate and NADPH molecules. Ribulose-5-phosphate in turn is converted to ribose- and xylulose-5-phosphate. These metabolites can be further metabolizes into intermediate molecules that enter the glycolytic pathway.

Macrophages and neutrophils are phagocytic in nature and during phagocytosis induce a respiratory burst that synthesizes reactive oxygen species (ROS) which oxidizes and interfere with integrity of biological structures of a pathogen. Macrophages utilize NADPH oxidase to synthesize ROS from NADPH [27]. Additionally, NADPH is used to generate glutathione which is an antioxidant that minimizes oxidative stress subjected to tissues [28]. Macrophage activation via LPS increases activity of the PPP shunt [29]. Noteworthy, the enzyme carbohydrate kinase-like protein (CARKL) which is a sedoheptulose kinase is critical in macrophage polarization. The enzyme phosphorylates sedoheptulose to sedoheptulose-7-phosphate which is an intermediate metabolite in the PPP pathway. This reaction is coupled to conversion of glyceraldehyde-3-phosphate into the non-oxidative branch of PPP. LPS activation of macrophages represses expression of CARKL genes leading to the intermediate product being used to synthesized pentose phosphates [30]. It remains unclear why such a regulatory activity occurs in M1 macrophages which for the larger part are not proliferative in nature and would not require much of the nucleotides. On the other hand, IL-4 activated macrophages (M2 polarization), show enhanced expression of the CARKL genes thus the enzyme’s catalyzed reactions increase in M2 macrophages [29].

2.3 Tri-carboxylic cycle

The TCA cycle or commonly known as Krebs cycle is an energy efficient mode of energy generation in form of ATP. Comparatively, from a single glucose molecule, glycolysis yields two ATP molecules while Krebs cycle yields a total of 36 ATP molecules. The process occurs within the mitochondrion and is commonly utilized by non-proliferative cells for energy generation. Krebs cycle has multiple input points notably via acetyl CoA and α-ketoglutarate (α-KG). Acetyl CoA together with oxaloacetate undergoes aldol condensation to form citrate while glutamate is converted to α-KG and altogether join the TCA. Ultimately, the cycle is meant to generate NADH and FADH2 by sequentially reducing the carbon atoms from acetyl CoA. Formed NADH and FADH2 enters ETC to generate ATP molecules.

M2 macrophages utilizes more or less the intact TCA cycle for energy production and further allows intermediates of UDP-GlcNAc to be generated which are utilized for glycosylation of M2-associated proteins e.g. mannose receptors [31]. Contrary, polarization of macrophages to the M1-phenotype is associated with break points in the intracellular TCA cycle [32]. First, conversion of citrate to isocitrate and secondly succinate to fumarate. This results in accumulation of citrate and succinate [32]. Citrate molecules are then directed to synthesis of fatty acid and itaconate and nitric oxide formation while the latter activates HIF1-α and production of IL-1β [33].

The first breakpoint occurs during the third reaction of Krebs cycle where isocitrate is converted to α-KG by the enzyme isocitrate dehydrogenase (IDH). M1 polarized macrophages transcriptional represses levels of IDH mRNA responsible for synthesis of IDH1 [34]. This leads to accumulation of isocitrate which isomerizes back to citrate via a two-step reaction process: isocitrate is first dehydrated to cis-aconitate which is then rehydrated to citrate. Ultimately, it is citrate and cis-aconitate that accumulates within mitochondrial matrix. As a result, cis-aconitate is diverted to synthesis of itaconate through decarboxylation reaction catalyzed by the immune-responsive gene 1 (IRG1). Itaconate is bactericidal in nature especially against Salmonella typhimurium and Mycobacterium tuberculosis [35]. Additionally, itaconate inhibits succinyl dehydrogenase (SDH) [36] which converts succinate to fumarate, the second break point in TCA cycle. Itaconate also alkylates the Kelch-like ECH-associated protein 1 which subsequently activates nuclear factor erythroid 2-related factor 2 (NRF2) that has anti-inflammatory activity [37].

Accumulation of citrate in M1 macrophages inhibits pyruvate dehydrogenase (PDH) and SDH ultimately decreasing formation of Acetyl CoA and FADH2 respectively. This additionally promotes consumption of ATP [38]. Accumulated citrate is transported from mitochondria matrix to cytosol via citrate transporter. Expression of citrate transporter is increased in proinflammatory macrophages in a NF-KB-dependent fashion [29]. Exported mitochondrial citrate is catabolized into products utilized in synthesis of inflammatory mediators e.g. nitric oxide and prostaglandins (PGE2) [29]. In M2 macrophages, since the TCA cycle is intact, much of α-KG is accumulated. α-KG has been shown to be immunosuppressive in nature by: preventing expression of pro-inflammatory IL-1β, inhibiting stabilization of HIF-1α and inactivating NF-Kβ signaling pathways [39]. Glutamate and glutamine provide precursors for formation of α-KG once deaminated via anaplerosis and will be discussed further under AA metabolism.

Conversion of α-KG to succinyl-CoA is a rate limiting step in the TCA cycle as it is one of the oxidoreductase reactions that result in formation of NADH. The reaction is catalyzed by the enzyme α-KG dehydrogenase (α-KGDH) which is highly sensitive to levels of ROS [40]. LPS activation and cytosolic accumulation of calcium within macrophages enhances activity of α-KGDH and limit production of anti-inflammatory IL-10 [29]. The intermediate product, succinyl-CoA is used to succinylate lysine residues within proteins such as SDH, PDH, Acyl-CoA and carbamoyl phosphate synthase 1 [32]. Notably, in LPS-activated macrophages, a lot of succinyl-CoA is produced which leads to succinylation of lysine 311 present on PKM2 [41, 42, 43]. As a result, the enzyme acquires a dimeric form which as earlier discussed facilitates entry of the enzyme into nucleus and consequent association with HIF-1α [41]. However, not all proteins succinylated activates pro-inflammatory activity as some have been shown to be immunosuppressive when succinylated. Formed succinyl-CoA is hydrolyzed by succinyl-CoA synthetase to succinate. Alternatively, succinate can also be produced via the gamma-aminobutyric acid shunt through which glutamine is deaminated to glutamate which is further catabolized to succinic semialdehyde and eventually succinate [17]. The latter product is of special importance in situations of inflammation and metabolic stress where it has been shown to regulate tumorigenesis, cellular inflammatory activity, signal transduction and epigenetics.

The second break occurs during conversion of succinate to fumarate by the enzyme SDH. Notably, the increased accumulation of succinate is not largely depended on SDH inhibition rather on glutamine anaplerosis as discussed above. Increased succinate is exported outside the mitochondria where it stabilizes HIF-1α and activate inflammatory genes leading to sustained production of IL-1β [32]. Within the mitochondrion, oxidation of succinate by SDH drives formation of ROS required during an immune response [44]. Although conversion of succinate to fumarate may be disrupted in M1 macrophages, LPS-activation has been shown to induce the aspartate-arginosuccinate shunt which feeds fumarate precursor molecules and leads to formation of fumarate [45]. Moreover, activation of the above stated shunt has been associated with upregulation of synthesis of NO and IL-6 [46] which are pro-inflammatory. At the same time, excess fumarate levels being formed inhibit pyroptosis increasing formation of gasdermin D [47]. Most likely, intermediates of aspartate-arginosuccinate shunt drives pro-inflammatory activities while the final product fumarate is anti-inflammatory in nature. Notably, might be another reason behind the second break that prevents conversion of succinate to fumarate in M1 macrophages.

Overall, metabolites of the TCA cycle on their own and in homeostatic concentrations seems to be immunosuppressive but activity of such metabolites on other pathways within and out of mitochondria most likely activate inflammatory pathways.

2.4 Lipid metabolism

Lipid metabolism functions to deliver lipid compounds to peripheral tissues and at the same time recycle lipids from peripheral tissues within the liver. It entails three pathways: exogenous, reverse cholesterol transport and endogenous pathways [48]. Dietary lipids are metabolized via exogenous pathway while endogenous pathways metabolize lipids synthesized in the liver. Reverse cholesterol transport pathway describes how cholesterol is removed from body tissues and transported to the liver for recycling since most peripheral tissue are not able to metabolize cholesterol.

Broadly, lipids entails triglycerides (TG), cholesterol and transporting molecules. Dietary lipids absorbed from intestinal lumen are package into chylomicrons and transported via lymphatic system where they become associated with apolipoproteins (ApoB, ApoC-II, -III, ApoE etc.) and enter systemic circulation to be delivered to various body tissues [49]. Apolipoproteins enable different body tissues to identify and uptake lipids; lipids bound to ApoC-II are recognized by adipose tissue which upon cellular entry are hydrolyzed to free fatty acids (FFA) and chylomicron remnants by lipoprotein lipase (LPL). Similarly, endogenously synthesized TGs and cholesterol get packaged into very low-density lipoprotein (VLDL) and ApoB respectively and then transported to body tissues. Adipocyte LDL hydrolyzes VLDL-bound TGs to FFAs and IDLs (VLDL remnants) [48]. The lipid transporting remnants are transported to the liver where hepatic lipases convert IDLs to low density lipoproteins (LDLs) which transport cholesterol across body tissues. In conditions such as chronic high fat intake where LDL is secreted in high amounts, excess LDL binds to free ApoA and resulting compound binds extracellular matrix within walls of vessels [50]. This has been implicated in the pathogenesis of atherosclerosis.

Macrophages are well fashioned to metabolize lipids especially liver, lung and adipose macrophages. They readily absorb and release lipoproteins and cholesterol respectively from dying cells. Lipoproteins, LDL and VLDL are absorbed either via phagocytosis, micropinocytosis or scavenger receptors such as CD36 and digested in the lysosomes by action of lysosomal acid lipase to form free FAs or cholesterol molecules [48]. Cytosolic esterification of free cholesterol to esters results in formation of lipid droplets that makes macrophages look like foam cells [51]. Additionally, free cytosolic cholesterol activates transcription factors such as peroxisome proliferator-activated receptor (PPAR), liver X receptor (LXR) and retinoid X receptor (RXR) [52]. The latter two receptors regulate lipogenesis in a more complex manner and in terms of macrophage function, their deficiency increases susceptibility to infections by Listeria monocytogenes [53] and mycobacterium tuberculosis [54] but at the same time confers protection from leishmania infections [55]. Nevertheless, binding of LXR by agonists inhibits expression of inflammatory genes that lead to synthesis of NO, PG and IL-6 [56].

Formed FFA on the other hand, enters mitochondria where it is oxidized to generate acetyl CoA and the reducing agents FADH2 and NADH. This latter process is largely seen in M2 polarized macrophages where lipolysis is registered but also occurs in M1 macrophages [57]. Blocking of the fatty acid oxidation pathway, as seen by use of etomoxir which inhibits carnitine palmitoyl-transferase 1, leads to inhibition of M2 macrophages polarization [58]. Additionally, formation of reducing agents FADH2 and NADH enables metabolism within macrophages to take an oxidative shift and by so doing activates PPAR-γ which activates expression of M2 signature genes. Activated PPAR-γ also enhances oxidation of glutamine within via anaplerosis which activates the TCA cycle [57]. In M1 macrophages on the other hand, fatty acid synthesis (FAS) is a more predominantly seen feature when macrophages are activated by TLR, LPS or IFN-γ [59]. Fatty acid synthesis ultimately leads to lipid formation which are necessary for macrophage cell membrane expansion during remodeling. Additionally, activation of FAS pathway induces the NLRP3 inflammasome with subsequent secretion of IL-1β [60].

2.5 Amino acid metabolism

Amino acids are essential building blocks for proteins that constitutively have an amino and carboxyl group attached to a central carbon. In mammals, 20 amino acids are utilized in protein synthesis out of which 9 are essential while the rest are non-essential. Noteworthy, there exist more than 20 amino acids but consensually, the 20 essential and non-essential amino acids are mostly described in biochemistry for cell function. The human body cannot synthesize essential amino acids and have to be supplied by dietary intake unlike non-essential amino acids [61]. They include valine, phenylalanine, lysine, methionine, tryptophan, isoleucine, leucine and histidine. Contrary, non-essential amino acids include arginine, tyrosine, alanine, serine, aspartate, glycine, asparagine, proline, glutamine and glutamate [61]. The role of amino acids and their metabolites during immune response is quite complex, varied and still an enigma. Of importance in macrophage functioning is tryptophan, arginine, serine, methionine and glutamine metabolism.

Tryptophan can either be metabolized via kynurenine or serotonin pathway [62]. Kynurenine pathway is key for de novo synthesis of NAD+ which is essential during redox reactions in glycolytic, TCA, fatty acid and electron transport chain pathways [63]. Contrary, serotonin pathway leads to formation of serotonin, a neurotransmitter, via action of tryptophan hydroxylase and aromatic amino acid decarboxylase [62]. Tryptophan can also be metabolized to indole-pyruvate that is skewed into TCA via anaplerosis. Kynurenine pathway starts with conversion of tryptophan to N-formyl-kynurenine mediated by indoleamine-2,3-dioxigenases (IDO) especially IDO1. Presences of infectious agents, TNF-α and IFN-γ stimulates expression of IDO genes which upregulates enzyme expression and tipping of tryptophan metabolism to the kynurenine pathway [64]. This additionally denies pathogens growth substrates within body tissues. In macrophages, serotonin formed from tryptophan is used to synthesize melatonin by action of N-acetyltransferase and O-methyltransferase. The product, melatonin has regulatory functions in terms of cytokine production by macrophages [65].

Serine and methionine are important during one-carbon metabolism in which methylation reactions occurs [62]. Serine, obtained exogenously or endogenously from 3-phosphoglycerate, donates one carbon atom to the folate and methionine cycles whereby in the latter, methionine acts as an intermediate donor in form of S-adenosylmethionine (SAM) thus not consumed. Macrophages utilize SAM to methylate histone molecules and synthesized IL-1beta in M1 phenotypes [66]. Additionally, LPS-activated macrophages utilize serine as a precursor molecule for synthesis of glycine which is subsequently used for glutathione synthesis. This enables rapid provision of the antioxidant as the NRF2-driven pathways generates glutathione molecules slowly [67].

Arginine can be catabolized either via arginase pathway or the nitric oxide synthesis pathway [62]. TNF-α and IFN-γ upregulate expression of inducible NO synthase (iNOS) in M1 macrophages to catabolize arginine into NO [68]. The enzyme iNOS mediates conversion of arginine to citrulline with concomitant production of NO which is an inflammatory mediator [68]. Upregulation of arginase enzyme, especially arginase 1, has been noted in macrophages stimulated by IL-4, -5 and -13 which are signature activators for naïve macrophages to M2 phenotype [69]. The enzyme converts arginine to ornithine limiting available substrate for iNOS within the cell. Ornithine is an important source of polyamines such as spermine which inhibits mitochondrial respiration and synthesis of pro-inflammatory cytokines [10].

In macrophages, glutamine is an important amino acid especially in M2 macrophages where it serves as a fuel source via anaplerotic processes [62]. In response to stimulation by IL-4, macrophages upregulate glutaminolysis to produce α-KG which mediates epigenetic reprogramming. Additionally, glutamine together with glycine and cysteine are used to synthesize glutathione which is a potent intracellular antioxidant. Glutamine is also a substrate for arginine biosynthesis [67] which is important for generation of NO in M1 macrophages.

Branched chain amino acids (leucine, isoleucine and valine) are major sources of carbon, and generation of glutamine, acetyl- and succinyl-CoA [67]. Increased uptake of leucine by macrophages activates the mTORC1 pathway leading to increased production of TNF-α and IL-1β in M1 macrophages [67].

Advertisement

3. Metabolic modulation of functions of tissue-specific resident macrophages

Resident macrophages especially in adults are formed through self-renewal of progenitor cells i.e. progenitor stem cells formed during embryogenesis that were responsible for primitive hematopoiesis persist in adulthood in various tissues though in smaller proportions [4]. These stem cells under the influence of factors such as colony stimulating factor-1 and IL-34 mediate activation of proliferation of stem cells to tissue macrophages dictated by transcription factors e.g. PU.1. Stem cell derived macrophages have differing transcriptional and gene expression profiles when compared to monocyte-derived macrophages [70]. They however, all perform similar functions depending on resident tissue and to which polarization end they are activated. Below is a discussion of some of tissue specific macrophages extensively studied and how they are metabolically programmed to function.

3.1 Alveolar macrophages

Alveolar macrophages are derived from fetal liver monocytes which during birth colonized the lungs and maintained self-perpetuation to adulthood. Primarily, their main function is to clear pulmonary surfactant that constantly being secreted into the alveolar space to maintain lung compliance [71]. Additionally, they also carry out immune surveillance and phagocytosis of foreign particles that have been inhaled [71]. Surfactants are predominately made up of lipids and as such, alveolar macrophages are metabolically equipped to handle lipid metabolism. During development, alveolar macrophages, under the influence of TGF-β and GM-CSF, activate the transcription factor PPARγ which regulates metabolism of fatty acids [72]. PPARγ activate genes responsible for increased fatty acid oxidation, esterification and efflux of cholesterol from cells [72]. Inability of alveolar macrophages to metabolize lipids leads to an accumulation of lung surfactant; a disease termed alveolar proteinosis [73]. Metabolically, alveolar macrophages conduct oxidation-phosphorylation reaction, fatty acid metabolism and cholesterol homeostasis.

3.2 Interstitial macrophages

Interstitial macrophages take residence in the space between epithelium and capillaries. They are derived from circulating blood monocytes and though are present in smaller numbers, their concentration increases in cases of immune response. Interstitial macrophages majorly junction as immune sentinels and once activated by a foreign particle, they differentiate to M1 phenotype [25]. Thus metabolically, interstitial cells conduct predominantly glycolysis and induction of nitric oxide synthase resulting in inhibition of mitochondrial oxidation-phosphorylation reaction [25].

3.3 Liver macrophages

Two types of liver macrophages have been documented: Kupffer cells and liver capsular macrophages [25]. Kupffer cells, located in the sinusoidal lumen, are derived from precursors of fetal liver monocytes and are capable of renewal. They carry out three major functions: clearance of damaged erythrocytes, immunological tolerance, clearance of blood-borne antigens [74]. In the presence of an antigen, Kupffer cells shift cellular metabolism towards glycolysis. This leads to increased glucose uptake and subsequently secretion of interleukin 10 [75]. Liver capsular macrophages on the other hand apart from performing immune surveillance, they also participate in neutrophil recruitment during an inflammatory episode. Less predominantly, liver macrophages conduct iron metabolism which is a major function of splenic macrophages [25]. During differentiation of Kupffer cells, notch ligands from endothelial cells of liver sinusoids induces expression of Spi-C. the latter is involved in activating genes that are responsible for iron metabolism 74].

3.4 Microglia

Microglia are CNS macrophages derived from embryonic yolk sac. They function to surveil the brain for pathogens, regulate neurogenesis and synaptic activity and have a role in clearance of apoptotic cells. Notably, microglia are active conductors of oxidative phosphorylation in inactive states to meet their energy requirement [76]. However, upon activation, they shift to a glycolytic model similar to that of blood monocyte derived macrophages. The metabolic profile of microglia is highly dynamic in nature and largely being influenced by the environment [76]. In steady state homeostatic conditions, microglia utilize oxidative phosphorylation for energy production; however, in hypoglycemic conditions, they switch to glutamine metabolism to support energy production [25].

3.5 Osteoclasts

Osteoclasts are multinucleated terminally differentiated monocyte-derived macrophages that are majorly found in the bone marrow. Predominantly, their function is bone resorption which they conduct via dissolution of collagen and mineral it the bone matrix [25]. This process is highly energy deficient thus osteoclasts have mitochondria not only in great numbers but size and complexity [25]. Formation of new osteoclasts is dependent on the factors RANK and osteoprotegerin. Activation of these systems is highly dependent on oxidative phosphorylation and mitochondrial biogenesis thus hypoxic conditions limit osteoclastogenesis process. Additionally, the metabolic profile of osteoclasts consists of elevate fatty acid oxidation, glutaminolysis, decreased glycolysis and activity of pentose phosphate pathway [77]. The former two serve to fuel oxidative phosphorylation processes which is required to produce energy for the energy driven process of bone resorption. Lactate, the end product of glycolysis has been shown to inhibit the osteoclastogenesis process [77]. During bone absorption by osteoclast, the metabolically switch to glycolysis.

3.6 Peritoneal macrophages

Two types of macrophages populate the peritoneum: large peritoneal macrophage (LPM) and small peritoneal macrophage. The former is derived from yolk sac progenitors with self-renewal capability thus forming the resident macrophages [25]. They function to phagocytose dead cells and bacteria. Small peritoneal macrophages are derived from circulating blood monocytes and predominantly function as immune sentinels, regulate immune response. In normal health conditions large peritoneal macrophages are more than small peritoneal macrophages but status changes often during immune stimulation or an ongoing inflammatory condition. Metabolically, Large peritoneal macrophages upon inflammatory stimulation exhibit increased activity in the mitochondria and electron transport chain which is linked to production of mtROS [78]. This high oxidative metabolism is fueled by fatty acids and glutamine whereby stimulated LPMs incorporate mitochondria into the phagosome and ensuing glutaminolysis induces complex II of the electron transport chain [78]. Notably, genes involved in lipid metabolism such as PPARγare downregulated in LPMS. SPMs on the other hand have higher glycolytic activity with reduced fatty acid oxidation and oxidative phosphorylation [78]. Stimulation of SPMs activates NF-kB which is associated with production of inflammatory cytokines.

3.7 Splenic macrophages

Currently, four different types of macrophages have been known to colonize the spleen: marginal zone macrophages, marginal metalophilic macrophages, tangible body macrophages and red pulp macrophages [25]. All except red pulp macrophages are derived from circulating blood monocytes. Red pulp macrophages differentiated from yolk sac and fetal liver progenitors that took residence in the spleen during embryogenesis. Red pulp macrophages act immune sentinels and in healthy conditions they primarily phagocytose platelets and red blood cells for iron metabolism [25]. Tangible body macrophages on the other hand phagocytose B cells that have undergone apoptosis. Marginal zone and marginal metallophillic macrophages function to blood-borne pathogens and clear them from circulation.

Advertisement

4. Conclusions

Macrophages, derived from either bone marrow monocytes or embryonic stem cells have crucial functions during immune response and homeostasis. Stem cell derived tissue specific macrophages are metabolically and functionally specialized to enable them play their role within specific tissues. In terms of immune response, macrophages are highly dynamic largely due to influence of low weight molecules such as intermediate metabolites within the tissue matrix. In turn, metabolic process occurring within macrophages and by extend extracellularly tend to modify the functioning of activated macrophages. As described, classically activated macrophages perform much of glycolysis and fatty acid synthesis to rapidly produce energy and remodel cell membrane. They also have breakpoints in the TCA cycle which allows intermediate products to accumulate and activate proinflammatory pathways. Contrary, alternatively activated macrophages predominantly utilize oxidative phosphorylation reactions and fatty acid oxidation to fuel their cellular activities and have an intact TCA cycle. However, to be noted, is that both M1 and M2 macrophages have been shown to depict a mixed metabolic picture. Additionally, the complexity of how metabolic processes are woven within cells makes it difficult to pin point a single pathway as either specialized in M1 or M2 macrophages. As such, we cannot conclusively state that above metabolic processes are delineated to specific macrophage polarization; rather it suggests that some metabolic pathways predominate either during inflammatory or anti-inflammatory events.

Advertisement

Conflict of interest

The author declares no conflict of interest.

References

  1. 1. Epelman S, Lavine K, Randolph G. Origin and functions of tissue macrophages. Immunity. 2014;41(1):21-35. DOI: 10.1016/j.immuni.2014.06.013
  2. 2. Yang J, Zhang L, Yu C, Yang X, Wang H. Monocyte and macrophage differentiation: Circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomarker Research. 2014;2(1):1-9. DOI: 10.1186/2050-7771-2-1
  3. 3. Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: Phenotypical vs. functional differentiation. Frontiers in Immunology. 2014;5(514):1-22. DOI: 10.3389/fimmu.2014.00514
  4. 4. Röszer T. Understanding the biology of self-renewing macrophages. Cell. 2018;7(8):103. DOI: 10.3390/cells7080103
  5. 5. Caputa G, Castoldi A, Pearce EJ. Metabolic adaptations of tissue-resident immune cells. Nature Immunology. 2019;20(7):793-801. DOI: 10.1038/s41590-019-0407-0
  6. 6. Pollard JW. Trophic macrophages in development and disease. Nature Reviews Immunology. 2009;9(4):259-270. DOI: 10.1038/nri2528
  7. 7. Plüddemann A, Mukhopadhyay S, Gordon S. Innate immunity to intracellular pathogens: Macrophage receptors and responses to microbial entry. Immunological Reviews. 2011;240(1):11-24. DOI: 10.1111/j.1600-065x.2010.00989.x
  8. 8. Rosales C, Uribe-Querol E. Phagocytosis: A fundamental process in immunity. BioMed Research International. 2017;2017:1-18. DOI: 10.1155/2017/9042851
  9. 9. Lee H, Woo Y, Hahn T, Jung YM, Jung Y. Formation and maturation of the phagosome: A key mechanism in innate immunity against intracellular bacterial infection. Microorganisms. 2020;8(9):1298. DOI: 10.3390/microorganisms8091298
  10. 10. Hesterberg R, Cleveland J, Epling-Burnette P. Role of polyamines in immune cell functions. Medical Science. 2018;6(1):22. DOI: 10.3390/medsci6010022
  11. 11. Ivashkiv LB. Epigenetic regulation of macrophage polarization and function. Trends in Immunology. 2013;34(5):216-223. DOI: 10.1016/j.it.2012.11.001
  12. 12. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. European Journal of Pharmacology. 2020;877:173090. DOI: 10.1016/j.ejphar.2020.173090
  13. 13. Corrado C, Fontana S. Hypoxia and HIF signaling: One axis with divergent effects. International Journal of Molecular Sciences. 2020;21(16):5611. DOI: 10.3390/ijms21165611
  14. 14. Gauthier T, Chen W. Modulation of macrophage Immunometabolism: A new approach to fight infections. Frontiers in Immunology. 2022;13(780839):1-21. DOI: 10.3389/fimmu.2022.780839
  15. 15. Harris RA, Harper ED. Glycolytic pathway. In: Encyclopedia of Life Sciences. 3rd ed. Chichester: John Wiley & Sons, Ltd; 2015. pp. 1-8
  16. 16. Brito-Arias M. Glycolysis. Enzymes Involved in Glycolysis, Fatty Acid and Amino Acid Biosynthesis: Active Site Mechanism and Inhibition. Sharjah, United Arab Emirates: Bentham Science Publishers; 2020. ISBN: 978-981-14-6088-3
  17. 17. Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Frontiers in Immunology. 2019;10(1462):1-16. DOI: 10.3389/fimmu.2019.01462
  18. 18. Rodríguez-Prados J, Través PG, Cuenca J, Rico D, Aragonés J, Martín-Sanz P, et al. Substrate fate in activated macrophages: A comparison between innate, classic, and alternative activation. The Journal of Immunology. 2010;185(1):605-614. DOI: 10.4049/jimmunol.0901698
  19. 19. Michl J, Ohlbaum DJ, Silverstein SC. 2-Deoxyglucose selectively inhibits Fc and complement receptor-mediated phagocytosis in mouse peritoneal macrophages. I. Description of the inhibitory effect. Journal of Experimental Medicine. 1976;144(6):1465-1483. DOI: 10.1084/jem.144.6.1465
  20. 20. Koo S, Garg NJ. Metabolic programming of macrophage functions and pathogens control. Redox Biology. 2019;24:101198. DOI: 10.1016/j.redox.2019.101198
  21. 21. Yu Q , Wang Y, Dong L, He Y, Liu R, Yang Q , et al. Regulations of glycolytic activities on macrophages functions in tumor and infectious inflammation. Frontiers in cellular and infection. Microbiology. 2020;10(287):1-14. DOI: 10.3389/fcimb.2020.00287
  22. 22. Wolf AJ, Reyes CN, Liang W, Becker C, Shimada K, Wheeler ML, et al. Hexokinase is an innate immune receptor for the detection of bacterial peptidoglycan. Cell. 2016;166(3):624-636. DOI: 10.1016/j.cell.2016.05.076
  23. 23. Min B, Park S, Kang H, Kim DW, Ham HJ, Ha C, et al. Pyruvate dehydrogenase kinase is a metabolic checkpoint for polarization of macrophages to the M1 phenotype. Frontiers in Immunology. 2019;10(944):1-14. DOI: 10.3389/fimmu.2019.00944
  24. 24. Palsson-McDermott E, Curtis A, Goel G, Lauterbach M, Sheedy F, Gleeson L, et al. Pyruvate kinase M2 regulates hif-1α activity and IL-1β induction and is a critical determinant of the Warburg effect in LPS-activated macrophages. Cell Metabolism. 2015;21(1):65-80. DOI: 10.1016/j.cmet.2014.12.005
  25. 25. Wculek SK, Dunphy G, Heras-Murillo I, Mastrangelo A, Sancho D. Metabolism of tissue macrophages in homeostasis and pathology. Cellular & Molecular Immunology. 2021;19(3):384-408. DOI: 10.1038/s41423-021-00791-9
  26. 26. Stincone A, Prigione A, Cramer T, Wamelink MM, Campbell K, Cheung E, et al. The return of metabolism: Biochemistry and physiology of the pentose phosphate pathway. Biological Reviews. 2014;90(3):927-963. DOI: 10.1111/brv.12140
  27. 27. Panday A, Sahoo MK, Osorio D, Batra S. NADPH oxidases: An overview from structure to innate immunity-associated pathologies. Cellular & Molecular Immunology. 2014;12(1):5-23. DOI: 10.1038/cmi.2014.89
  28. 28. Aranda-Rivera AK, Cruz-Gregorio A, Arancibia-Hernández YL, Hernández-Cruz EY, Pedraza-Chaverri J. RONS and oxidative stress: An overview of basic concepts. Oxygen. 2022;2(4):437-478. DOI: 10.3390/oxygen2040030
  29. 29. O'Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nature Reviews Immunology. 2016;16(9):553-565. DOI: 10.1038/nri.2016.70
  30. 30. Haschemi A, Kosma P, Gille L, Evans C, Burant C, Starkl P, et al. The Sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metabolism. 2012;15(6):813-826. DOI: 10.1016/j.cmet.2012.04.023
  31. 31. Jha A, Huang S, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419-430. DOI: 10.1016/j.immuni.2015.02.005
  32. 32. Ryan DG, O'Neill LA. Krebs cycle reborn in macrophage Immunometabolism. Annual Review of Immunology. 2020;38(1):289-313. DOI: 10.1146/annurev-immunol-081619-104850
  33. 33. Infantino V, Iacobazzi V, Menga A, Avantaggiati ML, Palmieri F. A key role of the mitochondrial citrate carrier (SLC25A1) in tnfα- and ifnγ-triggered inflammation. Biochimica et Biophysica Acta (BBA) - Gene Regulatory Mechanisms. 2014;1839(11):1217-1225. DOI: 10.1016/j.bbagrm.2014.07.013
  34. 34. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496:238-242. DOI: 10.1038/nature11986
  35. 35. Michelucci A, Cordes T, Ghelfi J, Pailot A, Reiling N, Goldmann O, et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proceedings of the National Academy of Sciences. 2013;110(19):7820-7825. DOI: 10.1073/pnas.1218599110
  36. 36. Lampropoulou V, Sergushichev A, Bambouskova M, Nair S, Vincent E, Loginicheva E, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metabolism. 2016;24(1):158-166. DOI: 10.1016/j.cmet.2016.06.004
  37. 37. Cheng C, Ku H, Shen T. The potential of using itaconate as treatment for inflammation-related heart diseases. Tzu Chi Medical Journal. 2022;34(2):113. DOI: 10.4103/tcmj.tcmj_83_21
  38. 38. Williams NC, O’Neill LA. A role for the Krebs cycle intermediate citrate in metabolic reprogramming in innate immunity and inflammation. Frontiers in Immunology. 2018;9(141):1-11. DOI: 10.3389/fimmu.2018.00141
  39. 39. Liu S, Yang J, Wu Z. The regulatory role of α-ketoglutarate metabolism in macrophages. Mediators of Inflammation. 2021;2021(11):1-7. DOI: 10.1155/2021/5577577
  40. 40. Tretter L, Adam-Vizi V. Alpha-ketoglutarate dehydrogenase: A target and generator of oxidative stress. Philosophical Transactions of the Royal Society B: Biological Sciences. 2005;360(1464):2335-2345. DOI: 10.1098/rstb.2005.1764
  41. 41. Harber KJ, De Goede KE, Verberk SG, Meinster E, De Vries HE, Van Weeghel M, et al. Succinate is an inflammation-induced immunoregulatory metabolite in macrophages. Metabolites. 2020;10(9):372. DOI: 10.3390/metabo10090372
  42. 42. Wang F, Wang K, Xu W, Zhao S, Ye D, Wang Y, et al. SIRT5 Desuccinylates and activates pyruvate kinase M2 to block macrophage IL-1β production and to prevent DSS-induced colitis in mice. Cell Reports. 2017;19(11):2331-2344. DOI: 10.1016/j.celrep.2017.05.065
  43. 43. Park J, Chen Y, Tishkoff D, Peng C, Tan M, Dai L, et al. SIRT5-mediated lysine Desuccinylation impacts diverse metabolic pathways. Molecular Cell. 2013;50(6):919-930. DOI: 10.1016/j.molcel.2013.06.001
  44. 44. Mills E, O’Neill LA. Succinate: A metabolic signal in inflammation. Trends in Cell Biology. 2014;24(5):313-320. DOI: 10.1016/j.tcb.2013.11.008
  45. 45. Fall F, Lamy E, Brollo M, Naline E, Lenuzza N, Thévenot E, et al. Metabolic reprograming of LPS-stimulated human lung macrophages involves tryptophan metabolism and the aspartate-arginosuccinate shunt. PLoS One. 2020;15(4):e0230813. DOI: 10.1371/journal.pone.0230813
  46. 46. Nassef MZ, Hanke JE, Hiller K. Mitochondrial metabolism in macrophages. American Journal of Physiology-Cell Physiology. 2021;321(6):C1070-C1081. DOI: 10.1152/ajpcell.00126.2021
  47. 47. Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, Li S, Wang B, Nemmara VV, et al. Succination inactivates gasdermin D and blocks pyroptosis. Science. 2020;369(6511):1633-1637. DOI: 10.1126/science.abb9818
  48. 48. Remmerie A, Scott CL. Macrophages and lipid metabolism. Cellular Immunology. 2018;330:27-42. DOI: 10.1016/j.cellimm.2018.01.020
  49. 49. Gaudet D, Drouin-Chartier J, Couture P. Lipid metabolism and emerging targets for lipid-lowering therapy. Canadian Journal of Cardiology. 2017;33(7):872-882. DOI: 10.1016/j.cjca.2016.12.019
  50. 50. Nordestgaard BG, Chapman MJ, Ray K, Borén J, Andreotti F, Watts GF, et al. Lipoprotein(a) as a cardiovascular risk factor: Current status. European Heart Journal. 2010;31(23):2844-2853. DOI: 10.1093/eurheartj/ehq386
  51. 51. Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: A dynamic balance. Nature Reviews Immunology. 2013;13(10):709-721. DOI: 10.1038/nri3520
  52. 52. Wang B, Tontonoz P. Liver X receptors in lipid signalling and membrane homeostasis. Nature Reviews Endocrinology. 2018;14(8):452-463. DOI: 10.1038/s41574-018-0037-x
  53. 53. Matalonga J, Glaria E, Bresque M, Escande C, Carbó JM, Kiefer K, et al. The nuclear receptor LXR limits bacterial infection of host macrophages through a mechanism that impacts cellular NAD metabolism. Cell Reports. 2017;18(5):1241-1255. DOI: 10.1016/j.celrep.2017.01.007
  54. 54. Korf H, Vander Beken S, Romano M, Steffensen KR, Stijlemans B, Gustafsson J, et al. Liver X receptors contribute to the protective immune response against mycobacterium tuberculosis in mice. Journal of Clinical Investigation. 2009;119(6):1626-1637. DOI: 10.1172/jci35288
  55. 55. Bruhn KW, Marathe C, Maretti-Mira AC, Nguyen H, Haskell J, Tran TA, et al. LXR deficiency confers increased protection against visceral leishmania infection in mice. PLoS Neglected Tropical Diseases. 2010;4(11):e886. DOI: 10.1371/journal.pntd.0000886
  56. 56. Ito A, Hong C, Rong X, Zhu X, Tarling EJ, Hedde PN, et al. LXRs link metabolism to inflammation through abca1-dependent regulation of membrane composition and TLR signaling. eLife. 2015;4(e08009):1-23. DOI: 10.7554/elife.08009
  57. 57. Batista-Gonzalez A, Vidal R, Criollo A, Carreño LJ. New insights on the role of lipid metabolism in the metabolic reprogramming of macrophages. Frontiers in Immunology. 2020;10(2993):1-7. DOI: 10.3389/fimmu.2019.02993
  58. 58. Divakaruni AS, Hsieh WY, Minarrieta L, Duong TN, Kim KK, Desousa BR, et al. Etomoxir inhibits macrophage polarization by disrupting CoA homeostasis. Cell Metabolism. 2018;28(3):490-503.e7. DOI: 10.1016/j.cmet.2018.06.001
  59. 59. Posokhova EN, Khoshchenko OM, Chasovskikh MI, Pivovarova EN, Dushkin MI. Lipid synthesis in macrophages during inflammation in vivo: Effect of agonists of peroxisome proliferator activated receptors α and γ and of retinoid X receptors. Biochemistry (Moscow). 2008;73(3):296-304. DOI: 10.1134/s0006297908030097
  60. 60. Anand PK. Lipids, inflammasomes, metabolism, and disease. Immunological Reviews. 2020;297(1):108-122. DOI: 10.1111/imr.12891
  61. 61. Kelly B, Pearce EL. Amino assets: How amino acids support immunity. Cell Metabolism. 2020;32(2):154-175. DOI: 10.1016/j.cmet.2020.06.010
  62. 62. Miyajima M. Amino acids: Key sources for immunometabolites and immunotransmitters. International Immunology. 2020;32(7):435-446. DOI: 10.1093/intimm/dxaa019
  63. 63. Badawy AA. Kynurenine pathway of tryptophan metabolism: Regulatory and functional aspects. International Journal of Tryptophan Research. 2017;10(1178646917691938):2-20. DOI: 10.1177/1178646917691938
  64. 64. Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: Tryptophan’s metabolites in exercise, inflammation, and mental health. Science. 2017;357(6349):eaaf9794. DOI: 10.1126/science.aaf9794
  65. 65. Xia Y, Chen S, Zeng S, Zhao Y, Zhu C, Deng B, et al. Melatonin in macrophage biology: Current understanding and future perspectives. Journal of Pineal Research. 2019;66(2):1-21. DOI: 10.1111/jpi.12547
  66. 66. Yu W, Wang Z, Zhang K, Chi Z, Xu T, Jiang D, et al. One-carbon metabolism supports S-adenosylmethionine and histone methylation to drive inflammatory macrophages. Molecular Cell. 2019;75(6):1147-1160.e5. DOI: 10.1016/j.molcel.2019.06.039
  67. 67. Halaby MJ, McGaha TL. Amino acid transport and metabolism in myeloid function. Frontiers in Immunology. 2021;12(695238):1-9. DOI: 10.3389/fimmu.2021.695238
  68. 68. Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via arginase or nitric oxide synthase: Two competing arginine pathways in macrophages. Frontiers in Immunology. 2014;5(532):1-10. DOI: 10.3389/fimmu.2014.00532
  69. 69. Yang Z, Ming X. Functions of arginase isoforms in macrophage inflammatory responses: Impact on cardiovascular diseases and metabolic disorders. Frontiers in Immunology. 2014;5(533):1-10. DOI: 10.3389/fimmu.2014.00533
  70. 70. Yadav S, Priya A, Borade DR, Agrawal-Rajput R. Macrophage subsets and their role: Co-relation with colony-stimulating factor-1 receptor and clinical relevance. Immunologic Research. 2022:1-23. Advance online publication. DOI: 10.1007/s12026-022-09330-8. Available from: https://link.springer.com/content/pdf/10.1007/s12026-022-09330-8.pdf?pdf=button%20sticky
  71. 71. Mass E. Delineating the origins, developmental programs and homeostatic functions of tissue-resident macrophages. International Immunology. 2018;30(11):493-501. DOI: 10.1093/intimm/dxy044
  72. 72. Schneider C, Nobs SP, Kurrer M, Rehrauer H, Thiele C, Kopf M. Induction of the nuclear receptor PPAR-γ by the cytokine GM-CSF is critical for the differentiation of fetal monocytes into alveolar macrophages. Nature Immunology. 2014;15(11):1026-1037. DOI: 10.1038/ni.3005
  73. 73. Kumar A, Abdelmalak B, Inoue Y, Culver DA. Pulmonary alveolar proteinosis in adults: Pathophysiology and clinical approach. The Lancet Respiratory Medicine. 2018;6(7):554-565. DOI: 10.1016/s2213-2600(18)30043-2
  74. 74. Nguyen-Lefebvre AT, Horuzsko A. Kupffer cell metabolism and function. Journal of Enzymology and Metabolism. 2015;1(1):101
  75. 75. Meszaros K, Bojta J, Bautista AP, Lang CH, Spitzer JJ. Glucose utilization by Kupffer cells, endothelial cells, and granulocytes in endotoxemic rat liver. American Journal of Physiology-Gastrointestinal and Liver Physiology. 1991;260(1):G7-G12. DOI: 10.1152/ajpgi.1991.260.1.g7
  76. 76. Lauro C, Limatola C. Metabolic reprograming of microglia in the regulation of the innate inflammatory response. Frontiers in Immunology. 2020;11(493):1-8. DOI: 10.3389/fimmu.2020.00493
  77. 77. Arnett TR, Orriss IR. Metabolic properties of the osteoclast. Bone. 2018;115:25-30. DOI: 10.1016/j.bone.2017.12.021
  78. 78. Pavlou S, Wang L, Xu H, Chen M. Higher phagocytic activity of thioglycollate-elicited peritoneal macrophages is related to metabolic status of the cells. Journal of Inflammation. 2017;14(4):1-6. DOI: 10.1186/s12950-017-0151-x

Written By

Filex Otieno

Submitted: 07 January 2023 Reviewed: 10 January 2023 Published: 02 March 2023