Skip to main content
Epigenetics & Chromatin logoLink to Epigenetics & Chromatin
. 2025 Mar 29;18:16. doi: 10.1186/s13072-025-00575-9

Crosstalk between metabolism and epigenetics during macrophage polarization

Kangling Zhang 1,, Chinnaswamy Jagannath 2,
PMCID: PMC11954343  PMID: 40156046

Abstract

Macrophage polarization is a dynamic process driven by a complex interplay of cytokine signaling, metabolism, and epigenetic modifications mediated by pathogens. Upon encountering specific environmental cues, monocytes differentiate into macrophages, adopting either a pro-inflammatory (M1) or anti-inflammatory (M2) phenotype, depending on the cytokines present. M1 macrophages are induced by interferon-gamma (IFN-γ) and are characterized by their reliance on glycolysis and their role in host defense. In contrast, M2 macrophages, stimulated by interleukin-4 (IL-4) and interleukin-13 (IL-13), favor oxidative phosphorylation and participate in tissue repair and anti-inflammatory responses. Metabolism is tightly linked to epigenetic regulation, because key metabolic intermediates such as acetyl-coenzyme A (CoA), α-ketoglutarate (α-KG), S-adenosylmethionine (SAM), and nicotinamide adenine dinucleotide (NAD+) serve as cofactors for chromatin-modifying enzymes, which in turn, directly influences histone acetylation, methylation, RNA/DNA methylation, and protein arginine methylation. These epigenetic modifications control gene expression by regulating chromatin accessibility, thereby modulating macrophage function and polarization. Histone acetylation generally promotes a more open chromatin structure conducive to gene activation, while histone methylation can either activate or repress gene expression depending on the specific residue and its methylation state. Crosstalk between histone modifications, such as acetylation and methylation, further fine-tunes macrophage phenotypes by regulating transcriptional networks in response to metabolic cues. While arginine methylation primarily functions in epigenetics by regulating gene expression through protein modifications, the degradation of methylated proteins releases arginine derivatives like asymmetric dimethylarginine (ADMA), which contribute directly to arginine metabolism—a key factor in macrophage polarization. This review explores the intricate relationships between metabolism and epigenetic regulation during macrophage polarization. A better understanding of this crosstalk will likely generate novel therapeutic insights for manipulating macrophage phenotypes during infections like tuberculosis and inflammatory diseases such as diabetes.

Keywords: Macrophages, M1, M2, Epigenetics, Sirtuins, IFN-γ, IL-4, IL10, IL-13, Glucose metabolism, Metabolism, Macrophage polarization, Histones, Methylation, Acetylation

Introduction

Macrophage differentiation, plasticity, and functional polarization

Typically, macrophages develop from monocytes which are produced in the bone marrow and circulate in the bloodstream before differentiating into macrophages when entering tissues to play critical roles in maintaining homeostasis and orchestrating the immune response [1]. An exception is that alveolar macrophages arise within the lungs. As key components of the innate immune system, macrophages are essential for pathogen recognition, phagocytosis, antigen presentation, and the regulation of inflammatory processes. Their ability to adapt to diverse environmental stimuli enables them to fulfill distinct functional roles, ranging from pro-inflammatory activities to anti-inflammatory and tissue-repair functions [2]. A defining feature of macrophages is their plasticity, which allows them to polarize into different phenotypes based on external cues. The two most widely studied phenotypes are classically activated (M1) macrophages and alternatively activated (M2) macrophages [3, 4]. These differentiation processes can be driven by growth factors and cytokines [5]. For instance, growth factors like macrophage colony-stimulating factor (M-CSF) and granulocyte-macrophage colony-stimulating factor (GM-CSF) promote the differentiation of monocytes into macrophages. Further, interferon-gamma (IFN-γ) and lipopolysaccharide (LPS), alone or together, promotes the formation of M1 macrophages, which are associated with pro-inflammatory cytokines including IL-1β, IL-6 and tumor necrosis factor α (TNFα), for pathogen elimination [610]. Conversely, cytokines like IL-4 and IL-13 induce M2a macrophages, which are involved in tissue repair, fibrosis, and inflammation resolution. Other M2 subtypes include M2b, which arises in response to immune complexes and IL-1R signaling and plays a regulatory role in immune responses and adaptive immunity, and M2c, which is driven by IL-10, TGF-β, and glucocorticoids, contributing to immune suppression and inflammation resolution [11]. It is noted that the M1/M2 macrophage polarization concept has been widely adopted in immunology as a broad model to understand macrophage responses to different stimuli. Therefore, throughout this review, we use the M1/M2 framework to describe macrophage functional states while acknowledging the complexity and plasticity of macrophage polarization.

The role of macrophages in tumors, metabolism, and epigenetic regulation

Besides their roles in infection and inflammation, macrophages are pivotal players in the tumor microenvironment (TME) [12]. Tumor-associated macrophages (TAMs), which often exhibit M2-like characteristics, promote tumor progression through several mechanisms. These include immune suppression, enhancement of angiogenesis, remodeling of the extracellular matrix, and facilitation of metastatic dissemination. TAMs can inhibit anti-tumor immune responses by producing immunosuppressive cytokines and modulating T-cell function, thereby contributing to immune evasion. Conversely, macrophages can exhibit anti-tumor properties under certain conditions, particularly when polarized to an M1-like phenotype [13]. This dual role underscores the complexity of macrophage function within the TME and highlights their potential as therapeutic targets [14].

Cytokines drive macrophage differentiation by binding to specific cell surface receptors, which activate intracellular signaling pathways such as JAK-STAT, NF-κB, and MAPK [15]. These pathways change gene expression and activate transcription factors (TFs) that dictate macrophage polarization. For example, IFN-γ activates TFs like IRF5 to drive M1 macrophages [16], whereas IL-4 and IL-13 activate STAT6 to promote M2 macrophages [17]. Interestingly, recent research has uncovered that macrophage function and polarization are tightly regulated by metabolic and epigenetic mechanisms [18, 19]. Metabolic pathways, such as glycolysis, the tricarboxylic acid (TCA) cycle, fatty acid oxidation, and the pentose phosphate pathway (PPP), dictate the energy requirements and biosynthetic precursors necessary for macrophage activation. For instance, M1 macrophages rely heavily on glycolysis to fuel their pro-inflammatory responses and reactive oxygen species (ROS) production, while exhibiting a disrupted TCA cycle with increased succinate accumulation. Succinate stabilizes hypoxia-inducible factor-1α (HIF-1α), promoting the production of inflammatory cytokines like interleukin-1β (IL-1β) [20]. In contrast, M2 macrophages depend on oxidative metabolism and fatty acid oxidation to sustain their anti-inflammatory functions and maintain mitochondrial integrity. The PPP supports NADPH production in both phenotypes but is differentially regulated to meet their specific metabolic demands.

Concurrently, epigenetic modifications, including histone acetylation, methylation, and succinylation, and DNA methylation, modulate gene expression programs that guide macrophage differentiation and stabilize macrophage phenotypes [21, 22]. For example, histone acetylation at promoter regions of inflammatory genes enhances their transcription in M1 macrophages, while M2 macrophages exhibit histone H3K27 demethylation that promotes anti-inflammatory gene expression [23]. DNA methylation and hydroxymethylation, regulated by factors such as ten-eleven translocation (TET) methylcytosine dioxygenases and methyltransferases, further influence macrophage polarization and function. Additionally, metabolites like α-ketoglutarate (α-KG), succinate, acetyl coenzyme A (acetyl-CoA), S-adenosylmethionine (SAM), and nicotinamide adenine dinucleotide (NAD+) serve as cofactors for these epigenetic regulators, linking metabolism directly to epigenetic control.

Understanding the metabolic-epigenetic axis in macrophages has significant implications for human health and disease. In the context of cancer, metabolic reprogramming and epigenetic alterations in TAMs can shift their phenotype toward tumor-promoting states, contributing to immune evasion and resistance to therapy [24]. Similarly, in infectious diseases such as tuberculosis, metabolic and epigenetic changes induced by pathogens can impair macrophage function, leading to immune dysregulation and chronic infections [25]. Emerging evidence also implicates macrophages in metabolic disorders, such as obesity and diabetes, where their polarization states influence systemic inflammation and insulin resistance [26]. Thus, targeting the metabolic and epigenetic networks that govern macrophage polarization offers promising therapeutic avenues for modulating immune responses in diverse pathological conditions.

Metabolic regulation of histone acetylation and methylation in gene expression and macrophage function

Histone methylation is a key epigenetic modification involving the addition of methyl groups to specific amino acid residues, predominantly lysine and arginine, on histone proteins. The patterns of histone methylation are intricate and highly specific, influencing gene expression by either activating or repressing transcription depending on the modified residue and the degree of methylation [27, 28]. For instance, tri-methylation of lysine 4 on histone H3 (H3K4me3) is commonly associated with active gene transcription, transcriptional pause-release, and elongation [2931], whereas tri-methylation of lysine 9 (H3K9me3) and lysine 27 (H3K27me3) on histone H3 are typically linked to gene repression [32, 33]. Histone H4 K20 mono-methylation facilitates chromatin openness, particularly during mitosis [3436], while di- and tri-methylation (H4K20me2 and H4K20me3), interacting with H3K9me3, increase quiescence and promote chromatin compaction [37, 38]. Additionally, methylation of histone H3 lysine 36 (H3K36me) plays a critical role in the regulation of alternative splicing of RNA and RNA elongation [39, 40], and methylation of histone H3 lysine 79 (H3K79me) is involved in the DNA damage response [41].

Histone acetylation also regulates gene expression and chromatin dynamics [42]. The acetylation process involves the addition of acetyl groups to lysine residues on histone proteins, which neutralizes their positive charge and reduces their affinity for the negatively charged DNA [43]. As a result, the chromatin structure becomes more relaxed and open, allowing greater ‘accessibility’ for the transcriptional machinery. This openness facilitates the binding of TFs and RNA polymerase to specific gene promoters, thereby enhancing gene activation and expression [44]. Moreover, acetylation of histones is instrumental in the recruitment of transcriptional coactivators and other regulatory proteins, which are essential for the initiation and maintenance of gene transcription [45]. This process is crucial for various cellular functions, including cell differentiation and development. In particular, histone acetylation influences the differentiation of macrophages in different functional states from homeostasis to host defense [21]. In addition, histone acetylation impacts DNA repair and replication [46]. The relaxed chromatin structure resulting from acetylation facilitates the accessibility of damaged DNA to repair enzymes, thereby supporting efficient DNA repair mechanisms. In addition, acetylation plays a role in DNA replication by regulating chromatin structure during the cell cycle, ensuring proper genome duplication [42]. It is noted here that histone acetylation and methylation are not isolated processes; rather, they often interact through complex crosstalk that influences chromatin structure and gene expression. These two types of histone modifications can affect each other’s actions and work in tandem to regulate cellular processes [47]. For instance, the presence of acetyl groups on histones can influence the binding of methyltransferases or demethylases, and conversely, specific methylation marks can affect the efficiency of acetylation processes [48]. Moreover, certain histone marks can create a “histone code” that guides the recruitment of other regulatory proteins, thereby integrating signals from both acetylation and methylation to fine tune gene expression and chromatin dynamics [49]. This interplay is crucial for maintaining the balance between gene activation and repression, influencing processes such as gene silencing, transcriptional activation, and cellular differentiation. Overall, the crosstalk between histone acetylation and methylation underscores the complexity of epigenetic regulation and its impact on cellular functions [47], including macrophage polarization [50].

Interestingly, histone acetylation and methylation are deeply intertwined with cellular metabolism, creating a dynamic relationship [51, 52]. Intriguingly, histone acetylation depends on acetyl-CoA, a metabolite derived from glycolysis, fatty acid oxidation, deacetylation of acetylated molecules, and ATP citrate lyase (ACLY)-mediated citrate cleavage. As metabolic activity changes, such as during increased glycolysis or fatty acid β-oxidation in stem cells or developing cells, the availability of acetyl-CoA can lead to altered levels of histone acetylation [5357]. This modification relaxes chromatin structure, facilitating gene activation. Histone acetylation can be removed by histone deacetylases (HDACs). Specifically, sirtuins, a family of NAD+ HDACs (Type III HDACs), play a crucial role in metabolism [58, 59]. In addition to NAD+, sirtuins bind to zinc and, therefore, sirtuin signaling is zinc dependent and requires an adequate level of zinc for proper enzymatic activity [60, 61]. Sirtuins adjust histone acetylation in response to the cellular energy status and nutrient availability, linking nutrient intake and metabolic states to transcriptional regulation [59]. By deacetylation of histones and other proteins, sirtuin consume NAD+ by breaking down the NAD+ molecule into nicotinamide and ADP-ribose.

On the other hand, histone methylation is regulated by SAM, which serves as the primary methyl donor for histone methyltransferases. The availability of SAM, which is influenced by one-carbon metabolism and dietary nutrients like folate and vitamin B12, directly impacts histone methylation patterns [62]. SAM levels, especially the SAM/SAH (S-adenosylhomocysteine) ratio, controls histone methylation, influencing gene expression and cellular functions [63, 64]. Moreover, cellular metabolic states such as hypoxia can affect histone methylation by altering the expression or activation of histone methyltransferases and demethylases involved during methylation processes [65]. Under hypoxic conditions, changes in metabolic pathways, including glycolysis and one-carbon metabolism, can lead to a shift in histone methylation pattern, linking environmental stress to epigenetic modifications [66].

Histone demethylation is also intricately connected to cellular metabolism, as metabolic states directly influence the activity of histone demethylases [67]. Key demethylases, such as those in the Jumonji C (JmjC) domain family, rely on alpha-ketoglutarate (α-KG) as a cofactor, a metabolite from the TCA cycle. Changes in α-KG levels can impact the efficiency of these demethylases, thereby altering histone methylation patterns and gene expression [68]. Additionally, the availability of other metabolites involved in one-carbon metabolism, such as folate, also affects demethylase activity [69]. Nutrient availability can therefore modulate these metabolites which in turn, influence histone demethylation. Conditions such as hypoxia, which alter metabolic pathways, further affect the activity of histone demethylases and the associated epigenetic modifications [65, 70, 71]. The interplay between metabolism and histone demethylation allows cells to adjust their epigenetic landscape in response to varying metabolic conditions, thereby integrating metabolic signals into gene regulation and cellular adaptation.

The metabolic and epigenetic landscapes in M1 and M2 macrophages

The metabolic and epigenetic landscapes of M1 and M2 macrophages are intricately linked to their contrasting functional roles within the immune system. M1 macrophages are equipped for aggressive immune responses and inflammation, characterized by glycolytic metabolism, a metabolic pathway that produces energy (ATP) rapidly and generates ROS. ROS drive inflammatory signaling by acting as redox-sensitive modulators of key cellular pathways: They activate TFs like NF-κB by oxidizing cysteine residues in regulatory proteins, leading to the expression of pro-inflammatory cytokines [72]. ROS also activate the NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome by destabilizing mitochondrial membranes and releasing activators such as thioredoxin-interacting protein (TXNIP), enhancing the maturation of IL-1β and IL-18 [73]. Additionally, ROS generate reactive aldehydes (e.g., 4-hydroxynonenal (4-HNE)) from membrane lipids, which function as signaling molecules to further promote inflammation [74]. They also stimulate MAPK pathways [75], driving inflammatory gene transcription and modulating epigenetic processes such as histone modifications and DNA methylation [76, 77]. Furthermore, ROS amplify their production through positive feedback loops involving nicotinamide adenine dinucleotide phosphate (NADPH) oxidases and mitochondrial dysfunction, intensifying cellular stress and immune cell recruitment [78, 79]. The shift toward glycolysis in M1 macrophages, even in the presence of oxygen, supports their role in pro-inflammatory responses and pathogen elimination [19]. Moreover, arginine is metabolized into citrulline releasing nitric oxide (NO) to form reactive nitrogen species (RNS), by activated iNOS enzyme; RNS are major antimicrobial agents. M1 macrophages also produce metabolites like succinate, which stabilizes hypoxia-inducible factor 1-alpha (HIF-1α), further enhancing their inflammatory responses [80]. The epigenetic landscape of M1 macrophages is characterized by specific histone modifications that facilitate the expression of pro-inflammatory genes. For instance, increased histone acetylation at the promoters of IL-6 and TNF-α enhances their accessibility of TFs and expression [81, 82]. Other key TFs, such as NF-κB/RelA and STAT1, are also activated in M1 macrophages to drive the expression of cytokines and other factors involved in inflammation and immune activation [83, 84]. In addition, DNA methylation patterns in M1 macrophages can also influence the accessibility and expression of inflammatory genes, contributing to their functional state [85, 86]. In contrast, M2 macrophages, also known as alternatively activated macrophages, rely predominantly on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to meet their energy demands and support their anti-inflammatory functions, such as tissue repair and wound healing [19]. Unlike the glycolytic metabolism of pro-inflammatory M1 macrophages, M2 macrophages utilize FAO to fuel the TCA cycle, providing substrates for OXPHOS [87]. This metabolic reprogramming is driven by cytokines like IL-4 and IL-13, which activate the STAT6 and PPARγ pathways, upregulating key enzymes involved in FAO [87, 88]. Mitochondrial biogenesis, regulated by PPAR Gamma Coactivator 1β (PGC-1β), is essential for maintaining OXPHOS, and mitochondrial integrity is critical for M2 polarization, as dysfunctional mitochondria impair their function [89]. Additionally, the metabolic shift towards FAO and OXPHOS is coupled with epigenetic modifications, such as histone acetylation, which promote the transcription of M2-specific genes [90]. The epigenetic landscape of M2 macrophages is distinct, with histone modifications that promote the expression of anti-inflammatory genes and support tissue repair [91]. TFs, such as STAT6 and PPAR-γ, play crucial roles in regulating genes, including Arg1, Dectin-1, Mrc1, Fizz1, and Ym1 [4]. The expression of these genes or proteins is increased by IL-4 stimulation but reduced by treatment with either LPS or IFN-γ [92]. Among these regulatory genes, Mrc1 (Mediator of Replication Checkpoint 1) is a replisome-associated histone chaperone essential for the symmetrical transfer of parental histones, particularly H3-H4 tetramers, to newly replicated DNA strands. Through distinct effects on the histone-binding domain (HBD) and other structural domains, Mrc1 coordinates with replisome components (FPC, Cdc45, MCM2, and POLE) to ensure balanced histone recycling and epigenetic inheritance during DNA replication [93]. Higher expression of Mrc1 is potentially correlated with increased expression of histone proteins and the MCM (minichromosome maintenance) complex, which together support elevated DNA replication activity. In M2 macrophages, arginine is metabolized by ARG1 into ornithine, which is used to synthesize polyamine including spermine, spermidine, and putrescine. Polyamine differentially regulates histone methylation: putrescine increases H3K9me3 and decreases H3K9ac, favoring M2 macrophages [94]. Spermine also favors M2 macrophages, while spermidine favors M1 macrophages [94]. In addition to histone modifications, DNA methylation patterns in M2 macrophages also help maintain the expression of genes that support tissue repair and the resolution of inflammation, reflecting their role during immune regulation and tissue homeostasis [50, 95]. Finally, M2 macrophages produce lower levels of ROS and RNS, aligning with their function in promoting healing rather than inflammation [96]. Conversely, this weakens their antimicrobial functions. Other metabolic pathways, including PPP, glutamine and arginine metabolism, tryptophan metabolism, and one-carbon metabolism, are also distinctly regulated between M1 and M2 macrophages. These pathways support the metabolic demands and functional specialization of macrophages during polarization. We have extensively studied and reviewed these metabolic alterations in prior publications, providing detailed insights into their mechanisms of macrophage function and polarization [9799].

Acetyl-CoA dependent histone acetylation during macrophage polarization

Acetyl-CoA, a critical metabolic intermediate, is generated through several key metabolic pathways. It can be produced through the oxidative decarboxylation of pyruvate derived from glycolysis, the oxidation of long-chain fatty acids in the mitochondria, the oxidative degradation of certain amino acids, and the deacetylation of acetylated biomolecules [97]. In addition, acetyl-CoA can be synthesized by ACLY, which converts citrate into acetyl-CoA and oxaloacetate, both of which play key roles in cellular metabolism and energy production. Furthermore, two acetyl-CoA synthetase enzymes, acetyl-CoA synthetase 1 and 2 (ACSS1 and ACSS2), also contribute to acetyl-CoA production by catalyzing the condensation of acetate with thiol groups, a reaction that occurs in both the cytoplasm and the mitochondria [99].

As the macrophage’s energy demands change, particularly during polarization, metabolic intermediates like acetyl-CoA serve as both a fuel source and a signaling molecule. This dual role of acetyl-CoA is essential for maintaining macrophage activation and ensuring proper immune responses. In the early phase of stimulation of macrophages (0.5–2 h post-stimulation with LPS), glycolysis is increased, driving pyruvate oxidation by the pyruvate dehydrogenase complex (PDC) to produce acetyl-CoA which either enters TCA or is used for fatty acid synthesis and protein acetylation. This also leads to enhanced citrate production and its cleavage by activated ACLY, resulting in elevated acetyl-CoA levels and increased global histone acetylation [100, 101].

In macrophages, the generation of acetyl-CoA and its subsequent use in histone acetylation is tightly regulated during polarization. During the early phase of macrophage stimulation (0.5–2 h post-stimulation with LPS), macrophages experience an upregulation of glycolysis, which is a hallmark of M1 polarization [19]. Increased glycolytic flux leads to an accumulation of pyruvate, which is shuttled into the mitochondria where it is oxidized by PDC to produce acetyl-CoA. This acetyl-CoA can either enter the TCA cycle, fueling oxidative phosphorylation for energy production, or it can be used for other cellular processes such as fatty acid synthesis or protein acetylation [98]. The increased acetyl-CoA levels, driven by both glycolysis and citrate cleavage, enable the acetylation of histones and non-histone proteins, which supports the transcription of inflammatory cytokines and other immune effector molecules that promote the pro-inflammatory M1 state [99]. Thus, the balance of acetyl-CoA production via glycolysis and mitochondrial activity directly influences the macrophage’s ability to respond to external stimuli and polarize toward M1 or M2 phenotypes. However, in the late phase (48–72 h post-stimulation with LPS and IFN-γ), NO inhibits PDC and the oxoglutarate/α-KG complex (OGDC), impairing glycolysis and the TCA cycle [102104]. This reduces acetyl-CoA production, along with histone deacetylation, leading to a decrease in global histone acetylation. Therefore, global histone acetylation in M1 macrophages follows a biphasic pattern upon stimulation.

Lactate-mediated regulation of macrophage polarization: a negative feedback mechanism

As glycolysis is upregulated in M1 macrophages, lactate production also increases, leading to enhanced histone lactylation [105]. Increased lactate production from pyruvate generates NAD+ to maintain NAD+ homeostasis, compensating for the consumption of NAD+ during glycolysis and the decreased de novo biosynthesis from tryptophan metabolism [98, 99]. However, accumulated lactate, in turn, has negative feedback effects on macrophage polarization. Lactate accumulation can induce the expression of immune suppressive markers and inhibit further M1 polarization by activating the lactate receptor, GPR81, which has been shown to inhibit NF-κB signaling and inflammatory cytokine production [106]. Additionally, histone lactylation activates genes linked to anti-inflammatory functions favoring an M2-like phenotype in tumor and chronic inflammatory environments [105]. Interestingly, lactate can enhance histone acetylation by inhibiting HDACs due to its structural similarity to pyruvate, resulting in a combined effect on lactylation and acetylation that stabilizes the M2 phenotype and suppresses pro-inflammatory pathways [107109]. In the tumor microenvironment, lactate drives M2 macrophage polarization via a TCA cycle-dependent mechanism that requires mitochondrial pyruvate uptake and ACLY activity for histone acetylation and immune-suppressive gene expression [110]. Disruption of this lactate-driven pathway impairs M2 polarization and tumor progression, highlighting a critical metabolic-epigenetic link during immune evasion [110, 111].

NAD+-dependent histone deacetylation by sirtuin proteins during macrophage polarization

Sirtuin proteins play a critical role in macrophage polarization, particularly in diseases like tuberculosis, as reviewed by us previously [99]. Among the seven members of the sirtuin protein family, SIRT2 is prominently expressed in M2 macrophages, supporting an anti-inflammatory phenotype, whereas SIRT5 is more prevalent in M1 macrophages associated with pro-inflammatory responses [8]. Moreover, SIRT2 deletion promotes inflammatory responses with enhanced expression of pro-inflammatory cytokines related to M1 macrophages by increasing NF-κB acetylation and reducing the M2-associated anti-inflammatory pathways [112]. This review will therefore focus on these two proteins, SIRT2 and SIRT5, and their distinct roles in macrophage polarization from a broad perspective.

SIRT2 in macrophage polarization: epigenetic, metabolic, and therapeutic implications

SIRT2 is the most expressed sirtuin in myeloid cells and macrophages. It plays a significant role in macrophage development and functional polarization [97, 113]. During macrophage development, SIRT2 governs key signal transduction pathways and transcriptional programs that determine macrophage polarization [112]. For instance, SIRT2 is highly expressed in M2 macrophages, where it promotes anti-inflammatory and tissue-reparative functions. Mechanistically, RelA/p65 forms a complex with SIRT2 and is deacetylated by SIRT2 at lysine 310 (K310) in the cytoplasm [112, 114]. Upon stimulation by TNFα, RelA translocates to the nucleus to function as a transcriptional activator. Inhibition of this deacetylation at K310 leads to increased expression of a subset of NF-κB-dependent genes [114]. These NF-κB-regulated genes include IL-1β, IL-6, IL-12, IL-23, TNFα, and iNOS, which are key markers of pro-inflammatory M1 macrophages [112, 115118]. SIRT2 depletion seems to have the same effect as its inhibition, which results in hyperacetylation of RelA at Lys310 in bone marrow-derived macrophages (BMDMs), promoting inflammatory responses characteristic of M1 macrophages while reducing the expression of anti-inflammatory genes associated with M2 macrophages [112]. However, one contradictory finding has also been reported: decrease of SIRT2 expression in BMDMs reduced LPS-induced pro-inflammatory or M1-related cytokine expression, iNOS/NO production, through inhibition of RelA activation due to SIRT2-induced reduction of RelA phosphorylation and translocation [119].

SIRT2 also intersects with metabolic pathways pivotal for macrophage function. By modulating glycolysis, mitochondrial respiration, and autophagy, SIRT2 ensures the metabolic flexibility of macrophages, enabling them to adapt to various stress conditions. Predominantly localized in the cytoplasm, SIRT2 deacetylates key enzymes in glycolysis and other metabolic pathways, thereby regulating their activity and influencing macrophage polarization and function. The role of SIRT2 (and other sirtuin proteins)-dependent metabolism, particularly in macrophages during mycobacterium tuberculosis (Mtb) infection, has been comprehensively reviewed by us elsewhere [99].

In addition to its metabolic functions, SIRT2 can be transported from the cytosol to the nucleus by importin-7 in response to bacterial infection [120]. In the nucleus, SIRT2 exerts epigenetic regulation by deacetylating histones. It modulates chromatin accessibility and transcriptional programs by targeting specific histone residues, such as H3K18ac and H4K16ac [120125]. These modifications are essential for defining the transcriptional landscapes that underpin M1 or M2 phenotypes. Therefore, SIRT2 interplays among epigenetics, metabolism, and the signaling pathways underscoring its multifaceted role in macrophage polarization.

Targeting SIRT2 offers significant therapeutic potential in modulating macrophage polarization for various diseases. Enhancing SIRT2 activity may promote M2 polarization, aiding in tissue repair and resolution of chronic inflammation. On the other hand, inhibiting SIRT2 could strengthen M1-mediated immune responses, providing benefits in combating infections and tumors. For example, inhibition of SIRT2 has been shown to significantly suppress the growth of Mtb [126129]. Individuals with diabetes are more susceptible to bacterial infections such as Mtb and often experience worse infection outcomes compared to non-diabetic individuals [130132]. This heightened susceptibility is linked to factors such as the availability of glucose as a carbon source for pathogen growth, increased gluconeogenesis and impaired glucose utilization, decreased ketoacidosis, and weakened GLUT1-facilitated phagocytosis [133], which reduces the production of ROS and RNS [134]. Diabetes is also associated with alterations in pro-inflammatory cytokine profiles during infection, though the literature presents conflicting findings. Some studies report elevated pro-inflammatory cytokine expression, while others show the opposite [135138]. This suggests that cytokine production in diabetic individuals may vary depending on the severity of the disease or blood glucose management. Inhibition of SIRT2 has the potential to enhance pro-inflammatory cytokine production and promote M1 macrophage polarization, potentially improving pathogenic bacterial clearance. However, this inhibition also carries the risk of exacerbating insulin resistance due to heightened pro-inflammatory cytokine production, despite the ability of SIRT2 to negatively regulate insulin resistance [139, 140]. Resolving this dilemma—enhancing pathogen killing while preventing insulin resistance—poses a critical research challenge.

SIRT5 in mitochondrial metabolism and macrophage polarization

SIRT5, a non-nuclear sirtuin protein localized exclusively in mitochondria, plays a crucial role as a desuccinylase, regulating key enzymes involved in mitochondrial metabolism. The substrates of SIRT5 have been extensively identified through affinity mass spectrometry in mouse liver tissue [141143]. SIRT5’s enzymatic activities include the deacetylation and desuccinylation of key metabolic enzymes, such as pyruvate kinase 1 and 2 (PKM1/2) in glycolysis [144], the pyruvate dehydrogenase complex (PDC), and succinate dehydrogenase (SDH), along with other dehydrogenases involved in the TCA cycle [141, 142]. Additionally, SIRT5 targets hydroxyacyl-CoA dehydrogenase (HADH) in fatty acid beta-oxidation, 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) in ketogenesis, and complexes I, II, and IV of the electron transport chain [141, 143]. SIRT5 also regulates crucial components of the urea cycle and extended arginine metabolism, including carbamoyl-phosphate synthase 1 (CPS1), argininosuccinate synthase 1 (ASS1), ornithine aminotransferase (OAT), glutamate-oxaloacetic transaminase (GOT1/2), and glutaminase (GLS) [98, 143, 145147].

While most of SIRT5’s substrates and desuccinylation sites have been identified in mouse liver tissue, as well as in yeast, Escherichia coli and various cell lines [141143, 148, 149], it remains unclear whether these sites overlap with those in human immune cells. It is particularly unknown if distinct modification sites exist in immune cells, especially during macrophage polarization. However, SIRT5 has been observed to desuccinylate and activate PKM2, which in turn blocks IL-1β production in macrophages [144]. In contrast, SIRT5 has been found to promote M2 macrophage polarization, contributing to mucosal edema and exacerbated Th2-type inflammation in chronic rhinosinusitis with nasal polyps (CRSwNP) [150]. Interestingly, while SIRT5 deficiency does not appear to compromise innate immune responses to bacterial infections such as endotoxemia, E.coli peritonitis, Streptococcus pneumoniae pneumonia, Klebsiella pneumoniae infection, or listeriosis (its impact on tuberculosis remains unknown) [151], inhibition of SIRT5 with the desuccinylation inhibitor MC3482 has been shown to decrease inflammation. This occurs by increasing M2 macrophage levels and reducing M1 macrophage levels in complete Freund’s adjuvant (CFA)-induced adjuvant arthritis models, via the SIRT5-GLUD1-α-KG axis [152]. Further research is necessary to fully understand SIRT5’s role in macrophage polarization and its responses to various microenvironments. For example, an area of research would be whether treatment with SIRT5 inhibitors or activators promotes M1 or M2 macrophage polarization.

Though SIRT5 is primarily recognized for its role in mitochondrial metabolism rather than direct involvement in epigenetic regulation, it competes with other sirtuin proteins for acetylation sites and NAD+, which is required for its enzymatic activity. This competition affects metabolic pathways, influencing the production of acetyl-CoA, methyl donors, and the consumption and regeneration of NAD+. Consequently, while SIRT5 does not directly impact epigenetic modifications, its influence on these metabolic processes may indirectly affect the epigenetic landscape. Lysine succinylation often overlaps with acetylation, frequently appearing at the same modification sites in mitochondrial proteins [148]. Given that SIRT2 is primarily a cytosolic protein and SIRT5 is specific to mitochondria, the distribution of lysine acetylation and succinylation varies across different cell compartments [148]. Acetyl-CoA is predominantly produced through glycolysis, while succinyl-CoA originates from the TCA cycle [148, 153]. Glycolysis is upregulated in M1 macrophages, whereas the TCA cycle is upregulated in M2 macrophages [19, 97]. Our observations indicate that SIRT5 is upregulated in M1 macrophages, while SIRT2 is upregulated in M2 macrophages [8]. Considering the differential distribution of enzymes and cofactors in M1 and M2 macrophages—specifically, high SIRT5 and low SIRT2 with high acetyl-CoA in M1 macrophages, versus low SIRT5 and high SIRT2 with high succinyl-CoA in M2 macrophages—we predict that lysine acetylation would be high and succinylation low in M1 macrophages, and the reverse would be true in M2 macrophages (Fig. 1). However, the stoichiometry of lysine acetylation and succinylation during macrophage polarization still requires experimental validation through mass spectrometry and immunological methods. Notably, SIRT5 and SIRT2 may compete to regulate the acetylation state of NF-κB p65/RelA, thereby influencing the activation of the NF-κB pathway and its downstream cytokines [154, 155].

Fig. 1.

Fig. 1

The stoichiometry of lysine acetylation and succinylation regulated by SIRT2 and SIRT5 determines the polarization states of macrophages. SIRT5 is highly expressed in M1 macrophages, it desuccinylates protein succinylation, resulting in hyperacetylation and hyposuccinylation; SIRT2 is highly expressed in M2 macrophages, it deacetylates protein acetylation resulting in hypersuccinylation and hypoacetylation. Modulation of the expression or activity of SIRT2 and SIRT5 drives macrophage polarization towards either M1 or M2. Activation of SIRT2 and SIRT5 is dependent of homeostatic level of NAD+, which is regulated by the NAD+de novo synthesis from tryptophan catabolism and the salvage pathway [98]. NAD+ is consumed by glycolysis and TCA, but can be regenerated through the Electron-Transporting-Complexes (ETC). Sirtinol: SIRT2 inhibitor; MC3482: SIRT5 inhibitor. Suc: succinylation; Ac: acetylation. Red color: up-regulated proteins or pathways; green color: down-regulated proteins or pathways

Regulation of Sirtuin activity by NAD+ and its impact on macrophage polarization

NAD+ is a vital coenzyme involved in redox reactions and serves as a substrate for various enzymes, including sirtuins and poly(ADP-ribose) polymerases (PARPs). Its subcellular distribution spans the cytoplasm, mitochondria, and nucleus, each maintaining distinct NAD⁺ pools that are interconnected and buffered and tightly regulated [156]. This compartmentalization is crucial for modulating metabolic pathways, DNA repair, chromatin remodeling, and other cellular processes. Recent research has illuminated the significant role of NAD⁺ metabolism in influencing macrophage function and polarization [157].

One pathway of interest is the de novo NAD⁺ synthesis via the kynurenine pathway (KP) in macrophages. Activation of the KP leads to the production of quinolinic acid (QA), which is subsequently converted to NAD⁺ by the enzyme quinolinate phosphoribosyltransferase (QPRT). Studies have shown that during aging, there is a suppression of QPRT expression, resulting in decreased NAD⁺ levels. This decline impairs mitochondrial function and skews macrophages toward a pro-inflammatory state or the M1-like state but with a declined phagocytosis activity [158]. Restoring QPRT expression in aged macrophages has been demonstrated to replenish NAD⁺ levels, enhance mitochondrial respiration, and promote a more balanced immune response [158]. Contradictory to the non-stimulated macrophages [97], stimulated macrophages with LPS increases the expression of IDO1 while reducing the expression of QPRT, resulting in an impeded conversion of QA to NAD+ and the accumulation of QA [158]. In addition to the de novo NAD+ synthesis, salvage pathways are crucial for macrophage activation as well. A decrease in de novo NAD+ synthesis coupled with an increase in the salvage pathway is normally associated with M1 macrophage polarization [98]. For instance, inhibiting IDO1 or QPRT, which are involved in tryptophan metabolism for NAD+ production, leads to increased production of pro-inflammatory cytokines, including TNF-α, IL-1β, LPS, and IFN-γ [99, 159, 160]. These cytokines activate nicotinamide phosphoribosyltransferase (NAMPT) expression, essential for NAD+ production and required for CD38 (a NAD+ hydrolase) consumption and ROS-induced DNA damage [161, 162]. CD38 is upregulated in both mouse and human M1 macrophages [163165]. CD38 activity contributes to increased autophagy, which mediates early control of Mtb proliferation in lungs [166]. Mtb infection depletes NAD+, which is critical for glycolysis and immune cell protection. Supplementation with NAM can limit Mtb proliferation by restoring NAD+ levels [167, 168]. However, there are contradictory findings regarding NAD+ supplementation. Increasing NAD+ production through niacin (NA) or its metabolite NAM via NAMPT can shift macrophage polarization towards the M2 phenotype, resulting in reduced iNOS and NO production [169171]. This discrepancy suggests that the host’s NAD+ homeostasis critically influences the balance between M1 and M2 macrophages that impacts the outcome of bacterial infection [98, 99]. One possible explanation is that during the early phase of Mtb infection, upregulated glycolysis, and reduced de novo NAD+ biosynthesis leads to NAD+ depletion, reduced sirtuin activity, and increased histone acetylation. Over time, NAD+ levels are rebalanced through the salvage pathway, with upregulated NAMPT in M1-macrophages restoring sirtuin activity [172], which in turn reduces global histone acetylation and acetylation of CPS1 [9799, 146]. Therefore, we propose that the balance between NAD+ biosynthesis and salvage pathways and other metabolic pathways that consume or produce NAD+ from NADH, together with the activation of SIRT2 and SIRT5, plays a crucial role macrophage polarization (Fig. 2).

Fig. 2.

Fig. 2

Regulation of NAD+level and SIRT2/5 expression in M1 and M2 Macrophages. M1 Macrophages (Left): The pro-inflammatory M1 phenotype exhibits enhanced glycolysis, as indicated by the upregulation of GLUT1 and key glycolytic intermediates (NAD+ consumption), leading to lactate production (NAD+ production). NAD+ biosynthesis is downregulated via the de novo (tryptophan-derived quinolinic acid pathway) due to reduced expression of QPRT, though it may be compensated by salvage pathways (mediated by increased NAMPT). SIRT5 supports ETC function and TCA cycle flux by modulating NAD+ levels. Epigenetically, TET2 (the most abundant in macrophages compared to TET1 and TET3) enhances transcription of TNF-α, IL-1β, and other pro-inflammatory cytokines through DNA demethylation, while the enhanced acetylation of histones and NF-κB/RelA (K18ac, K310ac) due to abolished NAD+-dependent deacetylase, SIRT2, promotes cytokine gene expression. M2 Macrophages (Right): The anti-inflammatory M2 phenotype relies on oxidative metabolism and fatty acid oxidation, driven by TCA cycle flux. Glycolysis proceeds minimally, with limited lactate production. NAD+ levels are increased by unregulated de novo synthesis and ETC complex I, and decreased consumption by glycolysis. Together with high expression of SIRT2, acetylation of histones and RelA is decreased resulting in suppression of pro-inflammatory cytokine expression. In contrast, histone H3K27me3 marks are removed by JMJD3, enabling transcription of STAT3, STAT6, PPARδ, and PPARγ, which is also facilitated by TET2 demethylation, contributing to the expression of M2-specific genes. Green color: Up-regulated; Red color: Down-regulated

In summary, sirtuin proteins, particularly SIRT2 and SIRT5, together with NAD+ metabolism, play critical roles in regulating macrophage polarization through metabolic, epigenetic, and immune pathways. SIRT2, highly expressed in M2 macrophages, promotes anti-inflammatory responses by deacetylating NF-κB/RelA and histones, enhancing tissue repair but suppressing pro-inflammatory cytokines. In contrast, SIRT5, predominantly found in M1 macrophages, regulates mitochondrial metabolism via desuccinylation of key metabolic enzymes, influencing inflammatory responses and macrophage function. SIRT2 inhibition fosters M1 polarization, enhancing glycolysis, histone acetylation, and immune responses against infections, such as Mtb, though it may exacerbate insulin resistance for diabetes patients who are more vulnerable to Mtb infection. SIRT5 modulates lysine acetylation and succinylation stoichiometry, indirectly shaping macrophage metabolism and polarization states (Fig. 1). Both sirtuins compete for NAD+ and acetylation sites, highlighting their interplay during macrophage polarization. However, the distribution of NAD+ and sirtuins (beyond SIRT2 and SIRT5) across different cellular compartments—such as the cytosol, mitochondria, and nucleus—and their roles in regulating metabolism, energy production, redox reactions, DNA repair, and epigenetics during macrophage development and polarization remain poorly understood. This compartmentalization appears crucial for the immune response to foreign invaders but poses significant challenges for investigation as techniques to measure sirtuin targets and compartment-specific NAD+ levels are limited and not yet widely accessible [156, 173].

SAM-dependent methylation during macrophage polarization

SAM serves as a critical methyl donor in cellular methylation reactions and consists of two primary components: an adenosyl group and methionine, the latter can donate a methyl group. The adenosyl group contains adenosine composed of a ribose sugar and a purine base. Ribose, an essential sugar component of adenosine, is derived from the PPP pathway and the degradation of nucleosides and nucleotides. Purine and methionine, the two essential precursors of SAM, can either be obtained directly from nutrients or synthesized internally. Purine synthesis involves a complex pathway relying on intermediates from the PPP and folate metabolism, while methionine synthesis depends on the one-carbon metabolism pathway, which provides the methyl groups necessary for methionine’s conversion to SAM.

SAM serves as a cofactor for numerous methyltransferases, including histone lysine methyltransferases (HMTs), protein arginine methyltransferases (PRMTs), DNA methyltransferases (DNMTs), RNA methyltransferases, and various metabolic methyltransferases. The synthesis of SAM through pathways such as the PPP, SSP, and one-carbon metabolism is synergistically regulated during LPS-induced inflammation and is essential for activating the pro-inflammatory gene IL-1β via H3K36 tri-methylation [174]. Interestingly, extracellular treatment with a high concentration of SAM (500 µM) has been shown to repress the expression of LPS-induced pro-inflammatory genes, such as TNF-α, while increasing the expression of the anti-inflammatory gene IL-10 and enhancing global DNA methylation [175]. While more studies on histone, DNA, and RNA methylation during macrophage polarization can be found elsewhere [22, 176], this review will focus specifically on protein arginine methylation, which not only depends on SAM as a methyl donor but also generates degradation products—monomethylarginine (MMA), symmetrical dimethylarginine (SDMA), and asymmetrical dimethylarginine (ADMA)—that are involved in arginine metabolism and NO production [98, 177].

Protein arginine methylation: a link between epigenetics, arginine metabolism, and host-pathogen interactions

Protein arginine methylation, regulated by a family of 11 identified protein arginine methyltransferases (PRMTs), plays a crucial role in both immune responses and host-pathogen interactions. Among the enzymes in the PRMT family, PRMT1, PRMT5, CARM1/PRMT4 were the predominant proteins identified in proteomic analyses of human monocyte-derived macrophages (Data are available via ProteomeXchange with identifier PXD058308, and our recently unpublished data) [178]. Extensive studies have highlighted their critical roles in macrophages, particularly in modulating immune responses to pathogens and in cancer invasion [179, 180]. Therefore, in this review, we will center our discussion exclusively on these three PRMTs. PRMT1, an asymmetrical dimethyltransferase, catalyzes the formation of ADMA during protein degradation. ADMA serves as an endogenous inhibitor of nitric oxide synthase (NOS) in both circulating blood and macrophages, thereby reducing NO production [181]. PRMT1 is closely linked to the regulation of inflammation and M2 macrophage polarization (Fig. 3A). For instance, PRMT1 knockout has been shown to increase the production of pro-inflammatory cytokines, a hallmark of M1 macrophages [182]. Mechanistically, PRMT1 regulates PPARγ gene expression by methylating histone H4 at arginine 3 (H4R3me2a) on the PPARγ promoter, thereby promoting M2 macrophage polarization [182]. Furthermore, PRMT1 acts as a negative regulator of anti-tumor immunity by suppressing MHC-I expression, a crucial molecule for presenting tumor antigens to immune cells [183]. Inhibition or knockout of PRMT1 enhances MHC-I expression and boosts the efficacy of anti-PD-1 immunotherapy, suggesting that targeting PRMT1 could improve the outcome of cancer immunotherapy [183].

Fig. 3.

Fig. 3

Regulation of Macrophage Polarization and Function by Protein/Histone Arginine Methylation. (A) PRMT1-mediated arginine methylation of histones and non-histone proteins promotes IL-4-induced M2 macrophage polarization via STAT6 and PPARγ activation. Inhibition or knockout of PRMT1 shifts macrophages from the anti-inflammatory M2 phenotype towards the pro-inflammatory M1 phenotype, characterized by increased TNF-α secretion and increased expression of IL-1β, iNOS, and IP-10. (B) PRMT5 plays a dual role in macrophage responses during Mtb infection. It enhances IFN-γ-induced MHC II transcription by symmetrically dimethylating histone H3R2, promoting antigen presentation to T cells. Simultaneously, PRMT5 represses ITCH expression via H4R3me2s methylation, stabilizing lipid accumulation proteins (ADRP, CD36) to support foam cell formation and Mtb survival. PRMT5 also suppresses autophagy by monomethylating ULK1 at R532. Inhibition of PRMT5 with EPZ015666 reduces lipid accumulation, restores autophagy, and shifts macrophages toward the pro-inflammatory M1 phenotype, enhancing Mtb clearance. (C) CARM1 methylates R35 of the transcription factor C/EBPα in B cells. Inhibition of CARM1, via pharmacological agents such as TP064, promotes the differentiation of B cells into macrophage-like cells through bone marrow transplantation (BMT), involving PU.1 and C/EBPα demethylation by TET2

PRMT5, another key enzyme, plays a significant role in immune regulation by symmetrically dimethylating histone H3 at arginine 2 (H3R2me2s) following IFN-γ stimulation, which in turn regulates CIITA-mediated MHC II transcription. This modification accumulates on the MHC II promoter, marking it as active for transcription, and facilitates the recruitment of CIITA, a critical transcriptional coactivator for MHC II gene expression. This synergy enhances MHC II transcription, which is vital for macrophage-mediated presentation of antigenic peptides to helper T cells and initiating an effective adaptive immune response [184]. Inhibition of PRMT5 with GSK3326595 has been shown to reduce MHC II expression in IFN-γ-stimulated peritoneal macrophages. However, PRMT5 inhibition also primes macrophages for IFN-γ-induced M1 polarization, leading to elevated mRNA expression levels of M1 markers such as TNF-α, IL-1β, iNOS, and IP-10 (Fig. 3B) [185].

Arginine methylation of histones has also emerged as a crucial determinant of Mtb survival within host cells. A significant example of this is the epigenetic regulation of the E3 ubiquitin ligase ITCH through arginine methylation of histone H4 by PRMT5, which profoundly impacts lipid accumulation during mycobacterial infection [186]. Lipid accumulation within the host is a pro-mycobacterial process that facilitates pathogen persistence and dormancy. Mtb infection represses the expression of ITCH via PRMT5, which induces repressive H4R3me2s methylation on the ITCH promoter, thereby inhibiting ITCH expression. This inhibition prevents the proteasomal degradation of key lipid accumulation molecules, such as Adipose Differentiation-Related Protein (ADRP) and CD36. Knockdown of PRMT5 via siRNA rescues ITCH expression, leading to reduced levels of Mtb-induced ADRP and CD36, thus limiting foam cell (FM) formation during Mtb infection. Furthermore, inhibition of PRMT5 enzyme activity with the PRMT5 inhibitor EPZ015666 results in reduced lipid levels and decreased mycobacterial survival in mouse peritoneal macrophages (ex vivo) and in a therapeutic mouse model of Mtb infection (in vivo) [186]. Additionally, this process may involve autophagy; genetic ablation or pharmacological inhibition of PRMT5 has been shown to trigger cytoprotective autophagy. Indeed, in breast cancer cells, PRMT5 catalyzes the monomethylation of ULK1 at R532, thereby suppressing ULK1 activation and attenuating autophagy (Fig. 3B) [187]. This underscores the complex role of PRMT5 in modulating autophagy and immune responses, with implications for various diseases [185].

CARM1, also known as PRMT4, is unique among PRMTs as it is the only one that methylates arginine residues in proteins with a nearby proline-rich sequence. Although its substrates may not be fully identified, CARM1 is known to target proteins involved in gene expression, chromatin organization and remodeling, mRNA metabolism, lipid metabolism via PPARα, and white adipocyte differentiation [188]. In addition to acting as a transcriptional coactivator, CARM1 plays a role in cell development and differentiation, is implicated in cancer progression and overexpressed in type 2 diabetes [189, 190]. Furthermore, CARM1 acts as a nutritional sensor by increasing in response to glucose and amino acid starvation. This increase is driven by the repression of its degradation via the SKP2-containing SCF E3 ubiquitin ligase complex in the nucleus. As a result, CARM1 binding to TFEB on autophagy gene promoters increases, where it catalyzes the asymmetric dimethylation of histone H3 at arginine 17 (H3R17me2a); this modification enhances transcriptional activation of autophagy genes [191]. Furthermore, CARM1 methylates the cofactor Pontin, which enhances Pontin’s interaction with FOXO3a and promotes the recruitment of the histone acetyltransferase Tip60 to the enhancer regions of autophagy genes [192]. This recruitment boosts histone H4 acetylation, creating a dual activation mechanism: CARM1 activates autophagy genes both at the promoter (through TFEB binding and H3R17me2a) and the enhancer (through FOXO3a-Pontin-Tip60 interaction). Together, these actions significantly upregulate autophagy under nutrient-deprived conditions, providing a comprehensive adaptive response to starvation [191, 192].

In summary, both PRMT1 and PRMT5 are seemingly associated with M2 macrophage polarization and host immune responses. Circulating monomethyl arginine (MMA) and ADMA inhibit NOS, reducing NO production [181, 193]. Inhibiting PRMT1 or PRMT5 alleviates the suppression of iNOS, elevating NO levels. This increase in NO production, combined with the upregulation of pro-inflammatory cytokines in M1 macrophages, enhances the immune response, aiding in the defense against infections and the destruction of cancer cells. Moreover, targeting PRMT5 may serve as a potential strategy for controlling mycobacterial infections by reducing lipid accumulation in macrophages and enhancing their autophagic activity [186].

Although CARM1 increase autophagy of cancer cells under nutrition stresses [191, 192], the inhibition of CARM1 enhances the efficacy of immunotherapy against resistant tumors by improving T-cell function, preserving memory T-cell populations, and amplifying tumor cell-intrinsic type 1 interferon responses [194]. Additionally, CARM1 methylates the transcription factor C/EBPα, which regulates B-cell trans-differentiation into macrophages (Fig. 3C) [195]. However, its role in macrophage polarization remains unknown, and further studies are needed to clarify the role of CARM1 in immune regulation, particularly during macrophage activation and polarization.

Linking α-Ketoglutarate (α-KG) to histone and DNA demethylation during macrophage polarization

α-KG, a rate-determining intermediate in the TCA cycle and a key metabolite produced through glutaminolysis, plays a crucial role in linking metabolism to epigenetic regulation, particularly in macrophage function. This anti-inflammatory metabolite not only enhances M2 macrophage activation but also regulates their metabolic reprogramming via mechanisms dependent on the demethylase Jumonji domain-containing 3 (JMJD3) [196]. The balance between α-KG and succinate levels is pivotal: a high α-KG/succinate ratio favors M2 macrophage polarization, while a low ratio promotes the pro-inflammatory M1 macrophage phenotype [196].

Role of α-KG in histone demethylation

α-KG serves as a cofactor for several histone demethylases, including JMJD3, which specifically demethylates trimethylated lysine 27 on histone H3 (H3K27me3), a repressive mark associated with gene silencing. IL-4 induces up-regulation of JMJD3 and JMJD3-mediated demethylation at the IRF4 locus activates this gene, promoting M2 macrophage polarization in response to stimuli such as helminth infection and chitin exposure [4, 23]. The JMJD3-IRF4 axis also plays a role in modulating type I interferon signaling in M1 macrophages, and this modulation influenced by the α-KG/succinate ratio, which is in turn regulated by IFN-β signaling [197]. Additionally, during Mtb infection, the RNA-binding protein MUSASHI (MSI) targets JMJD3 expression, facilitating the generation of lipid-laden foam macrophages and promoting M2 polarization through a TLR2-dependent mechanism [198].

Role of α-KG in DNA demethylation

In DNA, α-KG acts as a cofactor for TET enzymes including TET, TET2 and TET3, which are responsible for converting 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), a key step in DNA demethylation. This process is essential for regulating gene expression in macrophages. For example, TET1 has been shown to increase 5-hmC levels globally and at the TNF-α promoter during the differentiation of monocytes into macrophages, with further increases upon LPS stimulation. Knockout studies of TET1 reveal its role in promoting pro-inflammatory gene expression by reducing TNF-α levels [199]. In contrast, TET2 has been found to restrain pro-inflammatory gene expression, modulating the inflammatory response without significantly affecting IL-4-induced M2 macrophage polarization, and loss of TET2 leads to increased expression of inflammatory genes [200]. Therefore, TET1 promotes inflammatory gene expression and TET2 restrain it. The discrepancy between TET1 and TET2 may be due to their distinct biological roles, despite both being involved in DNA oxidation/demethylation. TET1 is primarily responsible for maintaining demethylation at promoters and CpG islands [201], helping to regulate gene expression, particularly in stem cell pluripotency [202] and in enhancing pro-inflammatory gene expression in macrophages [199]. On the other hand, TET2 has a broader scope, being active at enhancers and involved in both DNA and RNA methylation. TET2 regulates chromatin state and gene activation by mediating RNA m5C methylation, particularly in repetitive elements like LTR repeat RNAs, as its loss leads to global DNA hypomethylation resulting in an open chromatin state and transcription by antagonizing the m5C-MBD6-BAP1 gene activation pathway [203, 204]. TET3, like TET1, promotes the expression of pro-inflammatory genes in macrophages, playing a key role in regulating immune responses [205]. TET2 is significantly more expressed compared to TET1 and TET3 in mouse [200] and possibly human macrophages as suggested by our unpublished proteomics data, and its mRNA expression is notably induced during Mtb infection [206]. TET2 is essential for hematopoietic cell maturation, and its loss or mutation is associated with leukemogenesis [207]. It regulates immune tolerance in chronically activated mast cells [208]. It is important for immune cell differentiation via suppressing the production of MIF (macrophage migration inhibitory factor) through interaction with histone deacetylation and transcription factor EGR1 (early growth response 1) [209]. In addition, TET2 plays an important role in B cell development, T cell development and differentiation [210]. Interestingly, TET2 facilitates the de-repression of myeloid target genes during C/EBPα-induced transdifferentiation of pre-B cells [211]. This mechanism suggests a potential collaboration of TET2 with CARM1, as previous discussions have highlighted a similar role for CARM1 in the transdifferentiation of B cells to macrophages (Fig. 3C) [195]. This possibility is further supported by the fact that TET2 is one of the methylation substrates of CARM1 [188]. In macrophages, TET2 expression is induced by LPS but not IL-4 [200], and it interacts with RelA to activate TNF-α, a key component of M1 polarization [206]. Conversely, TET2 can modulate M2 polarization by recruiting HDAC2 to suppress IL-6 [200, 212] or regulating mRNA 5-methylcytosine methylation [213], in order to resolve inflammation [212].

Concluding remarks

Macrophage polarization is intricately regulated by both metabolism and epigenetic modifications. Pro-inflammatory M1 macrophages rely on glycolysis, while anti-inflammatory M2 macrophages favor oxidative phosphorylation and fatty acid oxidation, highlighting the metabolic shift essential for their functions [19]. Key metabolic pathways, such as glycolysis, TCA cycle, the PPP pathway, arginine metabolism, serine biosynthesis, and one-carbon metabolism for purine and methionine synthesis, are differentially activated in M1 and M2 macrophages, influencing their polarization states (Fig. 4). Epigenetic mechanisms, including histone acetylation, deacetylation, methylation, and demethylation, as well as DNA and RNA methylation, further shape macrophage function. Increased acetyl-CoA and histone acetylation enhances pro-inflammatory gene expression in M1 macrophages, while histone deacetylation represses inflammatory genes in M2 macrophages [98, 112]. Methylation patterns, such as DNA hypermethylation in the IL-12/IFN-γ signaling pathway, suppress pro-inflammatory responses [214], whereas demethylation by TET1 supports M1 polarization and demthylation by TET2 favors M2 macrophages [199]. RNA methylation, particularly N6-methyladenosine (m6A), also regulates inflammatory mRNA stability and translation [215]. Sirtuin proteins, like SIRT2 and SIRT5, integrate metabolic and epigenetic control in macrophages, with SIRT2 promoting M2 polarization and SIRT5 influencing arginine and glutamine metabolism with PRMT-catalyzed protein arginine methylation. Metabolites such as acetyl-CoA, SAM, α-KG, and NAD+ link cellular metabolism to epigenetic regulation, as they act as substrates or cofactors for enzymes driving acetylation, methylation, deacetylation/desuccinylation, and demethylation (Fig. 4). During early infection of pathogens or simulation with LPS, the balance between these epigenetic modifications defines the polarization state of macrophages, with M1 macrophages exhibiting active histone acetylation and reduced histone and DNA methylation while M2 macrophages showing increased histone deacetylation and histone/DNA methylation. The epigenetic landscape is dynamic, influenced by metabolites produced following stimulation, such as lactate and TCA cycle intermediates. Targeting these metabolic and epigenetic pathways offers therapeutic potential to modulate macrophage function in infectious and inflammatory diseases, such as tuberculosis and diabetes.

Fig. 4.

Fig. 4

Metabolic pathways that links epigenetics in macrophages. In M1 macrophages, SIRT5 is highly expressed and targets key enzymes involved in glycolysis, ETC, and glutamine/arginine metabolism. These targets include PKM in glycolysis, SDHA (succinate dehydrogenase) in the ETC, and GLS (glutaminase), CPS1 (carbamoyl-phosphate synthase 1), ASS1 (argininosuccinate synthase), and OAT (ornithine aminotransferase) in amino acid metabolism. In contrast, SIRT2 is highly expressed in M2 macrophages, where it regulates glycolytic enzymes such as PFKL (phosphofructokinase) and PKM, for acetyl-CoA synthesis, G6PD (glucose-6-phosphate dehydrogenase) in the pentose phosphate pathway, PHGDH (phosphoglycerate dehydrogenase) in serine biosynthesis, and CD38 and NAMPT in the NAD+ salvage pathway. SIRT2 activity is linked to epigenetic regulation, including the upregulation of JMJD3, a histone H3K27 demethylase that requires α-KG from the TCA cycle, IDO1 (indoleamine 2,3-dioxygenase) involved in tryptophan metabolism and NAD+ biosynthesis, and PRMT1 and PRMT5, which mediate arginine methylation. These metabolic pathways are closely tied to histone acetylation, succinylation, deacetylation, arginine methylation, RNA/DNA/histone methylation and demethylation, linking metabolic states with epigenetic modifications in macrophages. Enzymes targeted by SIRT5 are marked with a purple border, those targeted by SIRT2 with a blue border, while upregulated enzymes are filled in orange for M1 macrophages and blue for M2 macrophages, highlighting the metabolic-epigenetic interplay in macrophage polarization

Acknowledgements

The authors wish to acknowledge funding support from NIH RO1 AI161015 and 1R01AI184551-01.

Author contributions

KZ: Wrote. CJ: Edited.

Funding

The project is supported by NIH RO1 AI161015 and 1R01AI184551-01.

Data availability

No datasets were generated or analysed during the current study.

Declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

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.

Contributor Information

Kangling Zhang, Email: kazhang@utmb.edu.

Chinnaswamy Jagannath, Email: cjagannath@houstonmethodist.org.

References

  • 1.Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5:953–64. [DOI] [PubMed] [Google Scholar]
  • 2.Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Murray PJ. Macrophage polarization. Annu Rev Physiol. 2017;79:541–66. [DOI] [PubMed] [Google Scholar]
  • 4.Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. 2012;122:787–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol. 2014;5:514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Akira S, Taga T, Kishimoto T. Interleukin-6 in biology and medicine. Adv Immunol. 1993;54:1–78. [DOI] [PubMed] [Google Scholar]
  • 7.Dinarello CA. Biologic basis for interleukin-1 in disease. Blood. 1996;87:2095–147. [PubMed] [Google Scholar]
  • 8.Khan A, Zhang K, Singh VK, Mishra A, Kachroo P, Bing T, Won JH, Mani A, Papanna R, Mann LK, Ledezma-Campos E, Aguillon-Duran G, Canaday DH, David SA, Restrepo BI, Viet NN, Phan H, Graviss EA, Musser JM, Kaushal D, Gauduin MC, Jagannath C. Human M1 macrophages express unique innate immune response genes after mycobacterial infection to defend against tuberculosis. Commun Biol. 2022;5:480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tracey KJ, Cerami A. Tumor necrosis factor: a pleiotropic cytokine and therapeutic target. Annu Rev Med. 1994;45:491–503. [DOI] [PubMed] [Google Scholar]
  • 10.Held TK, Weihua X, Yuan L, Kalvakolanu DV, Cross AS. Gamma interferon augments macrophage activation by lipopolysaccharide by two distinct mechanisms, at the signal transduction level and via an autocrine mechanism involving tumor necrosis factor alpha and interleukin-1. Infect Immun. 1999;67:206–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lech M, Anders HJ. Macrophages and fibrosis: how resident and infiltrating mononuclear phagocytes orchestrate all phases of tissue injury and repair. Biochim Biophys Acta. 2013;1832:989–97. [DOI] [PubMed] [Google Scholar]
  • 12.Komohara Y, Jinushi M, Takeya M. Clinical significance of macrophage heterogeneity in human malignant tumors. Cancer Sci. 2014;105:1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.de Visser KE, Joyce JA. The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell. 2023;41:374–403. [DOI] [PubMed] [Google Scholar]
  • 14.Toledo B, Chen Z, Paniagua-Sancho L, Marchal M, Peran JA, M., and, Giovannetti E. Deciphering the performance of macrophages in tumour microenvironment: a call for precision immunotherapy. J Hematol Oncol. 2024;17:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Morris R, Kershaw NJ, Babon JJ. The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Sci. 2018;27:1984–2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, Hussell T, Feldmann M, Udalova IA. IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. 2011;12:231–8. [DOI] [PubMed] [Google Scholar]
  • 17.Waqas SFH, Ampem G, Roszer T. Analysis of IL-4/STAT6 signaling in macrophages. Methods Mol Biol. 2019;1966:211–24. [DOI] [PubMed] [Google Scholar]
  • 18.Xiong L, Darwanto A, Sharma S, Herring J, Hu S, Filippova M, Filippov V, Wang Y, Chen CS, Duerksen-Hughes PJ, Sowers LC, Zhang K. Mass spectrometric studies on epigenetic interaction networks in cell differentiation. J Biol Chem. 2011;286:13657–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Viola A, Munari F, Sanchez-Rodriguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol. 2019;10:1462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.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. and O’Neill, L. A. (2013) Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature 496:238–242. [DOI] [PMC free article] [PubMed]
  • 21.Chen S, Yang J, Wei Y, Wei X. Epigenetic regulation of macrophages: from homeostasis maintenance to host defense. Cell Mol Immunol. 2020;17:36–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ivashkiv LB. Epigenetic regulation of macrophage polarization and function. Trends Immunol. 2013;34:216–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, Miyake T, Matsushita K, Okazaki T, Saitoh T, Honma K, Matsuyama T, Yui K, Tsujimura T, Standley DM, Nakanishi K, Nakai K, Akira S. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 2010;11:936–44. [DOI] [PubMed] [Google Scholar]
  • 24.Puthenveetil A, Dubey S. Metabolic reprograming of tumor-associated macrophages. Ann Transl Med. 2020;8:1030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Llibre A, Dedicoat M, Burel JG, Demangel C, O’Shea MK, Mauro C. Host Immune-metabolic adaptations upon mycobacterial infections and Associated Co-morbidities. Front Immunol. 2021;12:747387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kraakman MJ, Murphy AJ, Jandeleit-Dahm K, Kammoun HL. Macrophage polarization in obesity and type 2 diabetes: weighing down our understanding of macrophage function? Front Immunol. 2014;5:470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Greer EL, Shi Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet. 2012;13:343–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Jambhekar A, Dhall A, Shi Y. Roles and regulation of histone methylation in animal development. Nat Rev Mol Cell Biol. 2019;20:625–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Lauberth SM, Nakayama T, Wu X, Ferris AL, Tang Z, Hughes SH, Roeder RG. H3K4me3 interactions with TAF3 regulate preinitiation complex assembly and selective gene activation. Cell. 2013;152:1021–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shilatifard A. The COMPASS family of histone H3K4 methylases: mechanisms of regulation in development and disease pathogenesis. Annu Rev Biochem. 2012;81:65–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang H, Fan Z, Shliaha PV, Miele M, Hendrickson RC, Jiang X, Helin K. H3K4me3 regulates RNA polymerase II promoter-proximal pause-release. Nature. 2023;615:339–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Bhaumik SR, Smith E, Shilatifard A. Covalent modifications of histones during development and disease pathogenesis. Nat Struct Mol Biol. 2007;14:1008–16. [DOI] [PubMed] [Google Scholar]
  • 33.McCarthy RL, Kaeding KE, Keller SH, Zhong Y, Xu L, Hsieh A, Hou Y, Donahue G, Becker JS, Alberto O, Lim B, Zaret KS. Diverse heterochromatin-associated proteins repress distinct classes of genes and repetitive elements. Nat Cell Biol. 2021;23:905–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Pesavento JJ, Yang H, Kelleher NL, Mizzen CA. Certain and progressive methylation of histone H4 at lysine 20 during the cell cycle. Mol Cell Biol. 2008;28:468–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Shoaib M, Chen Q, Shi X, Nair N, Prasanna C, Yang R, Walter D, Frederiksen KS, Einarsson H, Svensson JP, Liu CF, Ekwall K, Lerdrup M, Nordenskiold L, Sorensen CS. Histone H4 lysine 20 mono-methylation directly facilitates chromatin openness and promotes transcription of housekeeping genes. Nat Commun. 2021;12:4800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Yang H, Mizzen CA. The multiple facets of histone H4-lysine 20 methylation. Biochem Cell Biol. 2009;87:151–61. [DOI] [PubMed] [Google Scholar]
  • 37.Evertts AG, Manning AL, Wang X, Dyson NJ, Garcia BA, Coller HA. H4K20 methylation regulates quiescence and chromatin compaction. Mol Biol Cell. 2013;24:3025–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schotta G, Lachner M, Sarma K, Ebert A, Sengupta R, Reuter G, Reinberg D, Jenuwein T. A silencing pathway to induce H3-K9 and H4-K20 trimethylation at constitutive heterochromatin. Genes Dev. 2004;18:1251–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Schier AC, Taatjes DJ. Structure and mechanism of the RNA polymerase II transcription machinery. Genes Dev. 2020;34:465–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Wagner EJ, Carpenter PB. Understanding the language of Lys36 methylation at histone H3. Nat Rev Mol Cell Biol. 2012;13:115–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Huyen Y, Zgheib O, Ditullio RA Jr., Gorgoulis VG, Zacharatos P, Petty TJ, Sheston EA, Mellert HS, Stavridi ES, Halazonetis TD. Methylated lysine 79 of histone H3 targets 53BP1 to DNA double-strand breaks. Nature. 2004;432:406–11. [DOI] [PubMed] [Google Scholar]
  • 42.Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell Res. 2011;21:381–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Roth SY, Denu JM, Allis CD. Histone acetyltransferases. Annu Rev Biochem. 2001;70:81–120. [DOI] [PubMed] [Google Scholar]
  • 44.Tsompana M, Buck MJ. Chromatin accessibility: a window into the genome. Epigenetics Chromatin. 2014;7:33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Eberharter A, Becker PB. Histone acetylation: a switch between repressive and permissive chromatin. Second in review series on chromatin dynamics. EMBO Rep. 2002;3:224–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Aricthota S, Rana PP, Haldar D. Histone acetylation dynamics in repair of DNA double-strand breaks. Front Genet. 2022;13:926577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Suganuma T, Workman JL. Crosstalk among histone modifications. Cell. 2008;135:604–7. [DOI] [PubMed] [Google Scholar]
  • 48.Zhang T, Cooper S, Brockdorff N. The interplay of histone modifications - writers that read. EMBO Rep. 2015;16:1467–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Jenuwein T, Allis CD. Translating the histone code. Science. 2001;293:1074–80. [DOI] [PubMed] [Google Scholar]
  • 50.Kapellos TS, Iqbal AJ. (2016) Epigenetic Control of Macrophage Polarisation and Soluble Mediator Gene Expression during Inflammation. Mediators Inflamm 2016:6591703. [DOI] [PMC free article] [PubMed]
  • 51.Etchegaray JP, Mostoslavsky R. Interplay between metabolism and epigenetics: A Nuclear adaptation to environmental changes. Mol Cell. 2016;62:695–711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Reid MA, Dai Z, Locasale JW. The impact of cellular metabolism on chromatin dynamics and epigenetics. Nat Cell Biol. 2017;19:1298–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Willnow P, Teleman AA. Nuclear position and local acetyl-CoA production regulate chromatin state. Nature. 2024;630:466–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Yucel N, Wang YX, Mai T, Porpiglia E, Lund PJ, Markov G, et al. Glucose metabolism drives histone Acetylation Landscape transitions that dictate muscle stem cell function. Cell Rep. 2019;27:3939–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Mews P, Donahue G, Drake AM, Luczak V, Abel T, Berger SL. Acetyl-CoA synthetase regulates histone acetylation and hippocampal memory. Nature. 2017;546:381–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nagaraj R, Sharpley MS, Chi F, Braas D, Zhou Y, Kim R, et al. Nuclear localization of mitochondrial TCA cycle enzymes as a critical step in mammalian zygotic genome activation. Cell. 2017;168:210–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sutendra G, Kinnaird A, Dromparis P, Paulin R, Stenson TH, Haromy A, Hashimoto K, Zhang N, Flaim E, Michelakis ED. A nuclear pyruvate dehydrogenase complex is important for the generation of acetyl-CoA and histone acetylation. Cell. 2014;158:84–97. [DOI] [PubMed] [Google Scholar]
  • 58.Houtkooper RH, Canto C, Wanders RJ, Auwerx J. The secret life of NAD+: an old metabolite controlling new metabolic signaling pathways. Endocr Rev. 2010;31:194–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Houtkooper RH, Pirinen E, Auwerx J. Sirtuins as regulators of metabolism and healthspan. Nat Rev Mol Cell Biol. 2012;13:225–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chakrabarty SP, Balaram H. Reversible binding of zinc in Plasmodium Falciparum Sir2: structure and activity of the apoenzyme. Biochim Biophys Acta. 2010;1804:1743–50. [DOI] [PubMed] [Google Scholar]
  • 61.Feldman JL, Dittenhafer-Reed KE, Denu JM. Sirtuin catalysis and regulation. J Biol Chem. 2012;287:42419–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Serefidou M, Venkatasubramani AV, Imhof A. The impact of one Carbon metabolism on histone methylation. Front Genet. 2019;10:764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Fukumoto K, Ito K, Saer B, Taylor G, Ye S, Yamano M, Toriba Y, Hayes A, Okamura H, Fustin JM. Excess S-adenosylmethionine inhibits methylation via catabolism to adenine. Commun Biol. 2022;5:313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Karimian A, Vogelauer M, Kurdistani SK. Metabolic sinkholes: histones as methyl repositories. PLoS Biol. 2023;21:e3002371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kim J, Lee H, Yi SJ, Kim K. Gene regulation by histone-modifying enzymes under hypoxic conditions: a focus on histone methylation and acetylation. Exp Mol Med. 2022;54:878–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zhang K, Xu P, Sowers JL, Machuca DF, Mirfattah B, Herring J, Tang H, Chen Y, Tian B, Brasier AR, Sowers LC. Proteome Analysis of Hypoxic Glioblastoma cells reveals sequential metabolic adaptation of one-Carbon Metabolic pathways. Mol Cell Proteom. 2017;16:1906–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Teperino R, Schoonjans K, Auwerx J. Histone methyl transferases and demethylases; can they link metabolism and transcription? Cell Metab. 2010;12:321–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tran TQ, Lowman XH, Kong M. Molecular pathways: Metabolic Control of Histone Methylation and gene expression in Cancer. Clin Cancer Res. 2017;23:4004–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Luka Z, Moss F, Loukachevitch LV, Bornhop DJ, Wagner C. Histone demethylase LSD1 is a folate-binding protein. Biochemistry. 2011;50:4750–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chervona Y, Costa M. The control of histone methylation and gene expression by oxidative stress, hypoxia, and metals. Free Radic Biol Med. 2012;53:1041–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Salminen A, Kaarniranta K, Kauppinen A. Hypoxia-inducible histone lysine demethylases: impact on the aging process and age-related diseases. Aging Dis. 2016;7:180–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011;21:103–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tschopp J, Schroder K. NLRP3 inflammasome activation: the convergence of multiple signalling pathways on ROS production? Nat Rev Immunol. 2010;10:210–5. [DOI] [PubMed] [Google Scholar]
  • 74.Uchida K. 4-Hydroxy-2-nonenal: a product and mediator of oxidative stress. Prog Lipid Res. 2003;42:318–43. [DOI] [PubMed] [Google Scholar]
  • 75.Kamata H, Hirata H. Redox regulation of cellular signalling. Cell Signal. 1999;11:1–14. [DOI] [PubMed] [Google Scholar]
  • 76.Niu Y, DesMarais TL, Tong Z, Yao Y, Costa M. Oxidative stress alters global histone modification and DNA methylation. Free Radic Biol Med. 2015;82:22–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Chen Y, Shen YQ. Role of reactive oxygen species in regulating epigenetic modifications. Cell Signal. 2025;125:111502. [DOI] [PubMed] [Google Scholar]
  • 78.D’Autreaux B, Toledano MB. ROS as signalling molecules: mechanisms that generate specificity in ROS homeostasis. Nat Rev Mol Cell Biol. 2007;8:813–24. [DOI] [PubMed] [Google Scholar]
  • 79.Mittal M, Siddiqui MR, Tran K, Reddy SP, Malik AB. Reactive oxygen species in inflammation and tissue injury. Antioxid Redox Signal. 2014;20:1126–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Corcoran SE, O’Neill LA. HIF1alpha and metabolic reprogramming in inflammation. J Clin Invest. 2016;126:3699–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lee JY, Kim NA, Sanford A, Sullivan KE. Histone acetylation and chromatin conformation are regulated separately at the TNF-alpha promoter in monocytes and macrophages. J Leukoc Biol. 2003;73:862–71. [DOI] [PubMed] [Google Scholar]
  • 82.Vanden Berghe W, De Bosscher K, Boone E, Plaisance S, Haegeman G. The nuclear factor-kappab engages CBP/p300 and histone acetyltransferase activity for transcriptional activation of the interleukin-6 gene promoter. J Biol Chem. 1999;274:32091–8. [DOI] [PubMed] [Google Scholar]
  • 83.Chen S, Saeed A, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L, Duo Y. Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. 2023;8:207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang N, Liang H, Zen K. Molecular mechanisms that influence the macrophage m1-m2 polarization balance. Front Immunol. 2014;5:614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Migliaccio G, Morikka J, Del Giudice G, Vaani M, Mobus L, Serra A, Federico A, Greco D. Methylation and transcriptomic profiling reveals short term and long term regulatory responses in polarized macrophages. Comput Struct Biotechnol J. 2024;25:143–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Yang X, Wang X, Liu D, Yu L, Xue B, Shi H. Epigenetic regulation of macrophage polarization by DNA methyltransferase 3b. Mol Endocrinol. 2014;28:565–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Huang SC, Smith AM, Everts B, Colonna M, Pearce EL, Schilling JD, Pearce EJ. Metabolic reprogramming mediated by the mTORC2-IRF4 Signaling Axis is essential for Macrophage Alternative Activation. Immunity. 2016;45:817–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Huang SC, Everts B, Ivanova Y, O’Sullivan D, Nascimento M, Smith AM, Beatty W, Love-Gregory L, Lam WY, O’Neill CM, Yan C, Du H, Abumrad NA, Urban JF Jr., Artyomov MN, Pearce EL, Pearce EJ. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat Immunol. 2014;15:846–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Vats D, Mukundan L, Odegaard JI, Zhang L, Smith KL, Morel CR, Wagner RA, Greaves DR, Murray PJ, Chawla A. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 2006;4:13–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Covarrubias AJ, Aksoylar HI, Yu J, Snyder NW, Worth AJ, Iyer SS, et al. Akt-mTORC1 signaling regulates Acly to integrate metabolic input to control of macrophage activation. Elife. 2016;5:11612. [DOI] [PMC free article] [PubMed]
  • 91.Placek K, Schultze JL, Aschenbrenner AC. Epigenetic reprogramming of immune cells in injury, repair, and resolution. J Clin Invest. 2019;129:2994–3005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Menzies FM, Henriquez FL, Alexander J, Roberts CW. Sequential expression of macrophage anti-microbial/inflammatory and wound healing markers following innate, alternative and classical activation. Clin Exp Immunol. 2010;160:369–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Yu J, Zhang Y, Fang Y, Paulo JA, Yaghoubi D, Hua X, et al. A replisome-associated histone H3-H4 chaperone required for epigenetic inheritance. Cell. 2024;187:5010–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Latour YL, Gobert AP, Wilson KT. The role of polyamines in the regulation of macrophage polarization and function. Amino Acids. 2020;52:151–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Hey J, Paulsen M, Toth R, Weichenhan D, Butz S, Schatterny J, Liebers R, Lutsik P, Plass C, Mall MA. Epigenetic reprogramming of airway macrophages promotes polarization and inflammation in muco-obstructive lung disease. Nat Commun. 2021;12:6520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. The role of macrophages in Acute and Chronic Wound Healing and interventions to Promote Pro-wound Healing phenotypes. Front Physiol. 2018;9:419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Sowers ML, Tang H, Singh VK, Khan A, Mishra A, Restrepo BI, Jagannath C, Zhang K. Multi-OMICs analysis reveals metabolic and epigenetic changes associated with macrophage polarization. J Biol Chem. 2022;298:102418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zhang K, Mishra A, Jagannath C. New insight into arginine and tryptophan metabolism in macrophage activation during tuberculosis. Front Immunol. 2024;15:1363938. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Zhang K, Sowers ML, Cherryhomes EI, Singh VK, Mishra A, Restrepo BI, Khan A, Jagannath C. Sirtuin-dependent metabolic and epigenetic regulation of macrophages during tuberculosis. Front Immunol. 2023;14:1121495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lauterbach MA, Hanke JE, Serefidou M, Mangan MSJ, Kolbe CC, Hess T, et al. Toll-like receptor signaling rewires macrophage metabolism and promotes histone acetylation via ATP-Citrate lyase. Immunity. 2019;51:997–1011. [DOI] [PubMed] [Google Scholar]
  • 101.Moussaieff A, Rouleau M, Kitsberg D, Cohen M, Levy G, Barasch D, Nemirovski A, Shen-Orr S, Laevsky I, Amit M, Bomze D, Elena-Herrmann B, Scherf T, Nissim-Rafinia M, Kempa S, Itskovitz-Eldor J, Meshorer E, Aberdam D, Nahmias Y. Glycolysis-mediated changes in acetyl-CoA and histone acetylation control the early differentiation of embryonic stem cells. Cell Metab. 2015;21:392–402. [DOI] [PubMed] [Google Scholar]
  • 102.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. Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat Commun. 2020;11:698. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Palsson-McDermott EM, O’Neill LAJ. Targeting immunometabolism as an anti-inflammatory strategy. Cell Res. 2020;30:300–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Seim GL, Britt EC, John SV, Yeo FJ, Johnson AR, Eisenstein RS, Pagliarini DJ, Fan J. Two-stage metabolic remodelling in macrophages in response to lipopolysaccharide and interferon-gamma stimulation. Nat Metab. 2019;1:731–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.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. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Yang K, Xu J, Fan M, Tu F, Wang X, Ha T, Williams DL, Li C. Lactate suppresses macrophage pro-inflammatory response to LPS stimulation by inhibition of YAP and NF-kappaB activation via GPR81-Mediated signaling. Front Immunol. 2020;11:587913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Latham T, Mackay L, Sproul D, Karim M, Culley J, Harrison DJ, Hayward L, Langridge-Smith P, Gilbert N, Ramsahoye BH. Lactate, a product of glycolytic metabolism, inhibits histone deacetylase activity and promotes changes in gene expression. Nucleic Acids Res. 2012;40:4794–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Thangaraju M, Carswell KN, Prasad PD, Ganapathy V. Colon cancer cells maintain low levels of pyruvate to avoid cell death caused by inhibition of HDAC1/HDAC3. Biochem J. 2009;417:379–89. [DOI] [PubMed] [Google Scholar]
  • 109.Ohashi T, Aoki M, Tomita H, Akazawa T, Sato K, Kuze B, Mizuta K, Hara A, Nagaoka H, Inoue N, Ito Y. M2-like macrophage polarization in high lactic acid-producing head and neck cancer. Cancer Sci. 2017;108:1128–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Noe JT, Rendon BE, Geller AE, Conroy LR, Morrissey SM, Young LEA, Bruntz RC, Kim EJ, Wise-Mitchell A, Barbosa, de Souza Rizzo M, Relich ER, Baby BV, Johnson LA, Affronti HC, McMasters KM, Clem BF, Gentry MS, Yan J, Wellen KE, Sun RC, Mitchell RA. (2021) Lactate supports a metabolic-epigenetic link in macrophage polarization. Sci Adv. 7:eabi8602. [DOI] [PMC free article] [PubMed]
  • 111.Hayes C, Donohoe CL, Davern M, Donlon NE. The oncogenic and clinical implications of lactate induced immunosuppression in the tumour microenvironment. Cancer Lett. 2021;500:75–86. [DOI] [PubMed] [Google Scholar]
  • 112.Lo Sasso G, Menzies KJ, Mottis A, Piersigilli A, Perino A, Yamamoto H, Schoonjans K, Auwerx J. SIRT2 deficiency modulates macrophage polarization and susceptibility to experimental colitis. PLoS ONE. 2014;9:e103573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ciarlo E, Heinonen T, Theroude C, Herderschee J, Mombelli M, Lugrin J, Pfefferle M, Tyrrell B, Lensch S, Acha-Orbea H, Le Roy D, Auwerx J, Roger T. Sirtuin 2 Deficiency increases bacterial phagocytosis by macrophages and protects from chronic staphylococcal infection. Front Immunol. 2017;8:1037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Rothgiesser KM, Fey M, Hottiger MO. Acetylation of p65 at lysine 314 is important for late NF-kappaB-dependent gene expression. BMC Genomics. 2010;11:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Brasier AR. The nuclear factor-kappab-interleukin-6 signalling pathway mediating vascular inflammation. Cardiovasc Res. 2010;86:211–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Murphy TL, Cleveland MG, Kulesza P, Magram J, Murphy KM. Regulation of interleukin 12 p40 expression through an NF-kappa B half-site. Mol Cell Biol. 1995;15:5258–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Xie QW, Kashiwabara Y, Nathan C. Role of transcription factor NF-kappa B/Rel in induction of nitric oxide synthase. J Biol Chem. 1994;269:4705–8. [PubMed] [Google Scholar]
  • 119.Lee AS, Jung YJ, Kim D, Nguyen-Thanh T, Kang KP, Lee S, Park SK, Kim W. SIRT2 ameliorates lipopolysaccharide-induced inflammation in macrophages. Biochem Biophys Res Commun. 2014;450:1363–9. [DOI] [PubMed] [Google Scholar]
  • 120.Eldridge MJG, Pereira JM, Impens F, Hamon MA. (2020) Active nuclear import of the deacetylase Sirtuin-2 is controlled by its C-terminus and importins. Sci Rep. 10:2034. [DOI] [PMC free article] [PubMed]
  • 121.Eskandarian HA, Impens F, Nahori MA, Soubigou G, Coppee JY, Cossart P, Hamon MA. A role for SIRT2-dependent histone H3K18 deacetylation in bacterial infection. Science. 2013;341:1238858. [DOI] [PubMed] [Google Scholar]
  • 122.Inoue T, Hiratsuka M, Osaki M, Oshimura M. The molecular biology of mammalian SIRT proteins: SIRT2 in cell cycle regulation. Cell Cycle. 2007;6:1011–8. [DOI] [PubMed] [Google Scholar]
  • 123.Inoue T, Hiratsuka M, Osaki M, Yamada H, Kishimoto I, Yamaguchi S, Nakano S, Katoh M, Ito H, Oshimura M. SIRT2, a tubulin deacetylase, acts to block the entry to chromosome condensation in response to mitotic stress. Oncogene. 2007;26:945–57. [DOI] [PubMed] [Google Scholar]
  • 124.Vaquero A, Scher MB, Lee DH, Sutton A, Cheng HL, Alt FW, Serrano L, Sternglanz R, Reinberg D. SirT2 is a histone deacetylase with preference for histone H4 lys 16 during mitosis. Genes Dev. 2006;20:1256–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Zhang H, Park SH, Pantazides BG, Karpiuk O, Warren MD, Hardy CW, Duong DM, Park SJ, Kim HS, Vassilopoulos A, Seyfried NT, Johnsen SA, Gius D, Yu DS. SIRT2 directs the replication stress response through CDK9 deacetylation. Proc Natl Acad Sci U S A. 2013;110:13546–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Bhaskar A, Kumar S, Khan MZ, Singh A, Dwivedi VP, Nandicoori VK. Host sirtuin 2 as an immunotherapeutic target against tuberculosis. Elife. 2020;9:e55415. [DOI] [PMC free article] [PubMed]
  • 127.Bhaskar A, Pahuja I, Negi K, Verma A, Ghoshal A, Mathew B, Tripathi G, Maras JS, Chaturvedi S, Dwivedi VP. (2023) SIRT2 inhibition by AGK2 enhances mycobacteria-specific stem cell memory responses by modulating beta-catenin and glycolysis. iScience 26:106644. [DOI] [PMC free article] [PubMed]
  • 128.Kalsum S, Akber M, Loreti MG, Andersson B, Danielson E, Lerm M, Brighenti S. Sirtuin inhibitors reduce intracellular growth of M. Tuberculosis in human macrophages via modulation of host cell immunity. Sci Rep. 2024;14:28150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Roychowdhury S, Gandhirajan A, Kibler C, Wang X, Vachharajani V. Sirtuin 2 dysregulates autophagy in high-fat-exposed immune-tolerant macrophages. Cells. 2021;10:731. [DOI] [PMC free article] [PubMed]
  • 130.Dooley KE, Chaisson RE. Tuberculosis and diabetes mellitus: convergence of two epidemics. Lancet Infect Dis. 2009;9:737–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Dooley KE, Tang T, Golub JE, Dorman SE, Cronin W. Impact of diabetes mellitus on treatment outcomes of patients with active tuberculosis. Am J Trop Med Hyg. 2009;80:634–9. [PMC free article] [PubMed] [Google Scholar]
  • 132.Hodgson K, Morris J, Bridson T, Govan B, Rush C, Ketheesan N. Immunological mechanisms contributing to the double burden of diabetes and intracellular bacterial infections. Immunology. 2015;144:171–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Freemerman AJ, Johnson AR, Sacks GN, Milner JJ, Kirk EL, Troester MA, Macintyre AN, Goraksha-Hicks P, Rathmell JC, Makowski L. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J Biol Chem. 2014;289:7884–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Thurlow LR, Stephens AC, Hurley KE, Richardson AR. Lack of nutritional immunity in diabetic skin infections promotes Staphylococcus aureus virulence. Sci Adv. 2020;6:eabc5569. [DOI] [PMC free article] [PubMed]
  • 135.Geerlings SE, Brouwer EC, Van Kessel KC, Gaastra W, Stolk RP, Hoepelman AI. Cytokine secretion is impaired in women with diabetes mellitus. Eur J Clin Invest. 2000;30:995–1001. [DOI] [PubMed] [Google Scholar]
  • 136.Kumar S, Ingle H, Prasad DV, Kumar H. Recognition of bacterial infection by innate immune sensors. Crit Rev Microbiol. 2013;39:229–46. [DOI] [PubMed] [Google Scholar]
  • 137.Medzhitov R. Recognition of microorganisms and activation of the immune response. Nature. 2007;449:819–26. [DOI] [PubMed] [Google Scholar]
  • 138.Zykova SN, Jenssen TG, Berdal M, Olsen R, Myklebust R, Seljelid R. Altered cytokine and nitric oxide secretion in vitro by macrophages from diabetic type II-like db/db mice. Diabetes. 2000;49:1451–8. [DOI] [PubMed] [Google Scholar]
  • 139.Arora A, Dey CS. SIRT2 negatively regulates insulin resistance in C2C12 skeletal muscle cells. Biochim Biophys Acta. 2014;1842:1372–8. [DOI] [PubMed] [Google Scholar]
  • 140.Wortham M, Ramms B, Zeng C, Benthuysen JR, Sai S, Pollow DP, Liu F, Schlichting M, Harrington AR, Liu B, Prakash TP, Pirie EC, Zhu H, Baghdasarian S, Auwerx J, Shirihai OS, Sander M. (2024) Metabolic control of adaptive beta-cell proliferation by the protein deacetylase SIRT2. bioRxiv.
  • 141.Park J, Chen Y, Tishkoff DX, Peng C, Tan M, Dai L, Xie Z, Zhang Y, Zwaans BM, Skinner ME, Lombard DB, Zhao Y. SIRT5-mediated lysine desuccinylation impacts diverse metabolic pathways. Mol Cell. 2013;50:919–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rardin MJ, He W, Nishida Y, Newman JC, Carrico C, Danielson SR, Guo A, Gut P, Sahu AK, Li B, Uppala R, Fitch M, Riiff T, Zhu L, Zhou J, Mulhern D, Stevens RD, Ilkayeva OR, Newgard CB, Jacobson MP, Hellerstein M, Goetzman ES, Gibson BW, Verdin E. SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks. Cell Metab. 2013;18:920–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zhang R, Fang J, Xie X, Carrico C, Meyer JG, Wei L, Bons J, Rose J, Riley R, Kwok R, Kumaar A, Zhang PV, He Y, Nishida W, Liu Y, Locasale X, Schilling JW, B., and, Verdin E. Regulation of urea cycle by reversible high-stoichiometry lysine succinylation. Nat Metab. 2024;6:550–66. [DOI] [PubMed] [Google Scholar]
  • 144.Wang F, Wang K, Xu W, Zhao S, Ye D, Wang Y, Xu Y, Zhou L, Chu Y, Zhang C, Qin X, Yang P, Yu H. SIRT5 desuccinylates and activates pyruvate kinase M2 to Block Macrophage IL-1beta production and to prevent DSS-Induced Colitis in mice. Cell Rep. 2017;19:2331–44. [DOI] [PubMed] [Google Scholar]
  • 145.Nakagawa T, Guarente L. Urea cycle regulation by mitochondrial sirtuin, SIRT5. Aging. 2009;1:578–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Nakagawa T, Lomb DJ, Haigis MC, Guarente L. SIRT5 Deacetylates carbamoyl phosphate synthetase 1 and regulates the urea cycle. Cell. 2009;137:560–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Greene KS, Lukey MJ, Wang X, Blank B, Druso JE, Lin MJ, Stalnecker CA, Zhang C, Abril N, Erickson Y, Wilson JW, Lin KF, Weiss H, R. S., and, Cerione RA. SIRT5 stabilizes mitochondrial glutaminase and supports breast cancer tumorigenesis. Proc Natl Acad Sci U S A. 2019;116:26625–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Weinert BT, Scholz C, Wagner SA, Iesmantavicius V, Su D, Daniel JA, Choudhary C. Lysine succinylation is a frequently occurring modification in prokaryotes and eukaryotes and extensively overlaps with acetylation. Cell Rep. 2013;4:842–51. [DOI] [PubMed] [Google Scholar]
  • 149.Sadhukhan S, Liu X, Ryu D, Nelson OD, Stupinski JA, Li Z, Chen W, Zhang S, Weiss RS, Locasale JW, Auwerx J, Lin H. Metabolomics-assisted proteomics identifies succinylation and SIRT5 as important regulators of cardiac function. Proc Natl Acad Sci U S A. 2016;113:4320–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Cha XD, Zou QY, Li FZ, Wang TY, Wang SL, Cai BY, et al. SIRT5 exacerbates eosinophilic chronic rhinosinusitis by promoting polarization of M2 macrophage. J Allergy Clin Immunol. 2024;154:644–56. [DOI] [PubMed]
  • 151.Heinonen T, Ciarlo E, Theroude C, Pelekanou A, Herderschee J, Le Roy D, Roger T. Sirtuin 5 Deficiency does not compromise Innate Immune responses to bacterial infections. Front Immunol. 2018;9:2675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Wang YQ, Wang HL, Xu J, Tan J, Fu LN, Wang JL, Zou TH, Sun DF, Gao QY, Chen YX, Fang JY. Sirtuin5 contributes to colorectal carcinogenesis by enhancing glutaminolysis in a deglutarylation-dependent manner. Nat Commun. 2018;9:545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Wellen KE, Hatzivassiliou G, Sachdeva UM, Bui TV, Cross JR, Thompson CB. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. 2009;324:1076–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Hamaidi I, Zhang L, Kim N, Wang MH, Iclozan C, Fang B, et al. Sirt2 inhibition enhances metabolic fitness and effector functions of tumor-reactive T cells. Cell Metab. 2020;32:420–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Qin K, Han C, Zhang H, Li T, Li N, Cao X. NAD(+) dependent deacetylase sirtuin 5 rescues the innate inflammatory response of endotoxin tolerant macrophages by promoting acetylation of p65. J Autoimmun. 2017;81:120–9. [DOI] [PubMed] [Google Scholar]
  • 156.Hoyland LE, VanLinden MR, Niere M, Stromland O, Sharma S, Dietze J, Tolas I, Lucena E, Bifulco E, Sverkeli LJ, Cimadamore-Werthein C, Ashrafi H, Haukanes KF, van der Hoeven B, Dolle C, Davidsen C, Pettersen IKN, Tronstad KJ, Mjos SA, Hayat F, Makarov MV, Migaud ME, Heiland I, Ziegler M. Subcellular NAD(+) pools are interconnected and buffered by mitochondrial NAD(). Nat Metab. 2024;6:2319–37. [DOI] [PubMed] [Google Scholar]
  • 157.Fang J, Chen W, Hou P, Liu Z, Zuo M, Liu S, Feng C, Han Y, Li P, Shi Y, Shao C. NAD(+) metabolism-based immunoregulation and therapeutic potential. Cell Biosci. 2023;13:81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Minhas PS, Liu L, Moon PK, Joshi AU, Dove C, Mhatre S, Contrepois K, Wang Q, Lee BA, Coronado M, Bernstein D, Snyder MP, Migaud M, Majeti R, Mochly-Rosen D, Rabinowitz JD, Andreasson KI. Macrophage de novo NAD(+) synthesis specifies immune function in aging and inflammation. Nat Immunol. 2019;20:50–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Stone TW, Williams RO. Interactions of IDO and the kynurenine pathway with cell transduction systems and metabolism at the inflammation-cancer interface. Cancers (Basel). 2023;15:2895. [DOI] [PMC free article] [PubMed]
  • 160.Welz L, Harris DM, Kim NM, Alsaadi AI, Wu Q, Oumari M, Taubenheim J, Volk V, Credido G, Koncina E, Mukherjee PK, Tran F, Sievers LK, Pavlidis P, Powell N, Rieder F, Letellier E, Waschina S, Kaleta C, Feuerhake F, Verstockt B, McReynolds MR, Rosenstiel P, Schreiber S, Aden K. (2024) A metabolic constraint in the kynurenine pathway drives mucosal inflammation in IBD. medRxiv.
  • 161.Cameron AM, Castoldi A, Sanin DE, Flachsmann LJ, Field CS, Puleston DJ, Kyle RL, Patterson AE, Hassler F, Buescher JM, Kelly B, Pearce EL, Pearce EJ. Inflammatory macrophage dependence on NAD(+) salvage is a consequence of reactive oxygen species-mediated DNA damage. Nat Immunol. 2019;20:420–32. [DOI] [PubMed] [Google Scholar]
  • 162.Yarbro JR, Emmons RS, Pence BD. (2020) Macrophage Immunometabolism and Inflammaging: Roles of Mitochondrial Dysfunction, Cellular Senescence, CD38, and NAD. Immunometabolism. 2:e200026. [DOI] [PMC free article] [PubMed]
  • 163.Amici SA, Young NA, Narvaez-Miranda J, Jablonski KA, Arcos J, Rosas L, Papenfuss TL, Torrelles JB, Jarjour WN, Guerau-de-Arellano M. CD38 is robustly Induced in Human macrophages and monocytes in inflammatory conditions. Front Immunol. 2018;9:1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Jablonski KA, Amici SA, Webb LM, Ruiz-Rosado Jde D, Popovich PG, Partida-Sanchez S, Guerau-de-Arellano M. Novel markers to delineate murine M1 and M2 macrophages. PLoS ONE. 2015;10:e0145342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Li W, Li Y, Jin X, Liao Q, Chen Z, Peng H, Zhou Y. CD38: A Significant Regulator of macrophage function. Front Oncol. 2022;12:775649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Pisu D, Johnston L, Mattila JT, Russell D. The frequency of CD38+ alveolar macrophages correlates with mediate early control of M. Tuberculosis proliferation in the murine lung. Nat Commun. 2024;15:8522. [DOI] [PMC free article] [PubMed]
  • 167.Pacl HT, Chinta KC, Reddy VP, Nadeem S, Sevalkar RR, Nargan K, Lumamba K, Naidoo T, Glasgow JN, Agarwal A, Steyn AJC. NAD(H) homeostasis underlies host protection mediated by glycolytic myeloid cells in tuberculosis. Nat Commun. 2023;14:5472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Simmons JD, Peterson GJ, Campo M, Lohmiller J, Skerrett SJ, Tunaru S, Offermanns S, Sherman DR, Hawn TR. Nicotinamide limits replication of Mycobacterium tuberculosis and Bacille Calmette-Guerin within Macrophages. J Infect Dis. 2020;221:989–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bermudez S, Khayrullina G, Zhao Y, Byrnes KR. NADPH oxidase isoform expression is temporally regulated and may contribute to microglial/macrophage polarization after spinal cord injury. Mol Cell Neurosci. 2016;77:53–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Montserrat-de la Paz S, Naranjo MC, Lopez S, Abia R, Muriana FJ, Bermudez B. Niacin and olive oil promote skewing to the M2 phenotype in bone marrow-derived macrophages of mice with metabolic syndrome. Food Funct. 2016;7:2233–8. [DOI] [PubMed] [Google Scholar]
  • 171.Montserrat-de la Paz S, Naranjo MC, Lopez S, Abia R, Muriana FJG, Bermudez B. Niacin and its metabolites as master regulators of macrophage activation. J Nutr Biochem. 2017;39:40–7. [DOI] [PubMed] [Google Scholar]
  • 172.Halvorsen B, Espeland MZ, Andersen GO, Yndestad A, Sagen EL, Rashidi A, Knudsen EC, Skjelland M, Skagen KR, Krohg-Sorensen K, Holm S, Ritschel V, Holven KB, Biessen EA, Aukrust P, Dahl TB. Increased expression of NAMPT in PBMC from patients with acute coronary syndrome and in inflammatory M1 macrophages. Atherosclerosis. 2015;243:204–10. [DOI] [PubMed] [Google Scholar]
  • 173.Cambronne XA, Kraus WL. Location, location, location: compartmentalization of NAD(+) synthesis and functions in mammalian cells. Trends Biochem Sci. 2020;45:858–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Yu W, Wang Z, Zhang K, Chi Z, Xu T, Jiang D, Chen S, Li W, Yang X, Zhang X, Wu Y, Wang D. One-Carbon Metabolism supports S-Adenosylmethionine and histone methylation to Drive Inflammatory macrophages. Mol Cell. 2019;75:1147–60. e1145. [DOI] [PubMed] [Google Scholar]
  • 175.Pfalzer AC, Choi SW, Tammen SA, Park LK, Bottiglieri T, Parnell LD, Lamon-Fava S. S-adenosylmethionine mediates inhibition of inflammatory response and changes in DNA methylation in human macrophages. Physiol Genomics. 2014;46:617–23. [DOI] [PubMed] [Google Scholar]
  • 176.Pinello N, Song R, Lee Q, Calonne E, Duan KL, Wong E, Tieng J, Mehravar M, Rong B, Lan F, Roediger B, Ma CJ, Yuan BF, Rasko JEJ, Larance M, Ye D, Fuks F, Wong JJ. Dynamic changes in RNA m(6)A and 5 hmC influence gene expression programs during macrophage differentiation and polarisation. Cell Mol Life Sci. 2024;81:229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Rodriguez PC, Ochoa AC, Al-Khami AA. Arginine metabolism in myeloid cells shapes innate and adaptive immunity. Front Immunol. 2017;8:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Nicholas D, Tang H, Zhang Q, Rudra J, Xu F, Langridge W, Zhang K. Quantitative proteomics reveals a role for epigenetic reprogramming during human monocyte differentiation. Mol Cell Proteom. 2015;14:15–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Sauter C, Simonet J, Guidez F, Dumetier B, Pernon B, Callanan M, et al. Protein arginine methyltransferases as therapeutic targets in hematological malignancies. Cancers (Basel). 2022;14:5443. [DOI] [PMC free article] [PubMed]
  • 180.Wolf SS. The protein arginine methyltransferase family: an update about function, new perspectives and the physiological role in humans. Cell Mol Life Sci. 2009;66:2109–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Boger RH, Bode-Boger SM, Szuba A, Tsao PS, Chan JR, Tangphao O, Blaschke TF, Cooke JP. Asymmetric dimethylarginine (ADMA): a novel risk factor for endothelial dysfunction: its role in hypercholesterolemia. Circulation. 1998;98:1842–7. [DOI] [PubMed] [Google Scholar]
  • 182.Tikhanovich I, Zhao J, Olson J, Adams A, Taylor R, Bridges B, Marshall L, Roberts B, Weinman SA. Protein arginine methyltransferase 1 modulates innate immune responses through regulation of peroxisome proliferator-activated receptor gamma-dependent macrophage differentiation. J Biol Chem. 2017;292:6882–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Djajawi TM, Pijpers L, Srivaths A, Chisanga D, Chan KF, Hogg SJ, Neil L, Rivera SM, Bartonicek N, Ellis SL, Lim K, Sian TCC, Faridi P, Liao Y, Pal B, Behren A, Shi W, Vervoort SJ, Johnstone RW, Kearney CJ. PRMT1 acts as a suppressor of MHC-I and anti-tumor immunity. Cell Rep. 2024;43:113831. [DOI] [PubMed] [Google Scholar]
  • 184.Fan Z, Kong X, Xia J, Wu X, Li H, Xu H, Fang M, Xu Y. The arginine methyltransferase PRMT5 regulates CIITA-dependent MHC II transcription. Biochim Biophys Acta. 2016;1859:687–96. [DOI] [PubMed] [Google Scholar]
  • 185.Zhang Y, Verwilligen RAF, Van Eck M, Hoekstra M. PRMT5 inhibition induces pro-inflammatory macrophage polarization and increased hepatic triglyceride levels without affecting atherosclerosis in mice. J Cell Mol Med. 2023;27:1056–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Borbora SM, Rajmani RS, Balaji KN. PRMT5 epigenetically regulates the E3 ubiquitin ligase ITCH to influence lipid accumulation during mycobacterial infection. PLoS Pathog. 2022;18:e1010095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Brobbey C, Yin S, Liu L, Ball LE, Howe PH, Delaney JR, Gan W. Autophagy dictates sensitivity to PRMT5 inhibitor in breast cancer. Sci Rep. 2023;13:10752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Shishkova E, Zeng H, Liu F, Kwiecien NW, Hebert AS, Coon JJ, Xu W. Global mapping of CARM1 substrates defines enzyme specificity and substrate recognition. Nat Commun. 2017;8:15571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Porta M, Amione C, Barutta F, Fornengo P, Merlo S, Gruden G, Albano L, Ciccarelli M, Ungaro P, Durazzo M, Beguinot F, Berchialla P, Cavallo F, Trento M. The co-activator-associated arginine methyltransferase 1 (CARM1) gene is overexpressed in type 2 diabetes. Endocrine. 2019;63:284–92. [DOI] [PubMed] [Google Scholar]
  • 190.Suresh S, Huard S, Dubois T. CARM1/PRMT4: making its Mark beyond its function as a transcriptional Coactivator. Trends Cell Biol. 2021;31:402–17. [DOI] [PubMed] [Google Scholar]
  • 191.Shin HJ, Kim H, Oh S, Lee JG, Kee M, Ko HJ, Kweon MN, Won KJ, Baek SH. AMPK-SKP2-CARM1 signalling cascade in transcriptional regulation of autophagy. Nature. 2016;534:553–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Yu YS, Shin HR, Kim D, Baek SA, Choi SA, Ahn H, Shamim A, Kim J, Kim IS, Kim KK, Won KJ, Baek SH. Pontin arginine methylation by CARM1 is crucial for epigenetic regulation of autophagy. Nat Commun. 2020;11:6297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Fulton MD, Brown T, Zheng YG. The biological axis of protein arginine methylation and asymmetric dimethylarginine. Int J Mol Sci. 2019;20:3322. [DOI] [PMC free article] [PubMed]
  • 194.Kumar S, Zeng Z, Bagati A, Tay RE, Sanz LA, Hartono SR, Ito Y, Abderazzaq F, Hatchi E, Jiang P, Cartwright ANR, Olawoyin O, Mathewson ND, Pyrdol JW, Li MZ, Doench JG, Booker MA, Tolstorukov MY, Elledge SJ, Chedin F, Liu XS, Wucherpfennig KW. CARM1 inhibition enables immunotherapy of resistant tumors by Dual Action on Tumor Cells and T cells. Cancer Discov. 2021;11:2050–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Torcal Garcia G, Kowenz-Leutz E, Tian TV, Klonizakis A, Lerner J, De Andres-Aguayo L, et al. Carm1-arginine methylation of the transcription factor C/EBPalpha regulates transdifferentiation velocity. Elife. 2023;12:e83951. [DOI] [PMC free article] [PubMed]
  • 196.Liu PS, Wang H, Li X, Chao T, Teav T, Christen S, Di Conza G, Cheng WC, Chou CH, Vavakova M, Muret C, Debackere K, Mazzone M, Huang HD, Fendt SM, Ivanisevic J, Ho PC. Alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol. 2017;18:985–94. [DOI] [PubMed] [Google Scholar]
  • 197.Lee M-C, Achuthan K, De Souza AA, Lupancu DP, Binger TJ, Lee KJ, Xu MKS, McConville Y, de Weerd MJ, Dragoljevic NA, Hertzog D, Murphy PJ, Hamilton AJ, J. A., and, Fleetwood AJ. Type I interferon antagonism of the JMJD3-IRF4 pathway modulates macrophage activation and polarization. Cell Rep. 2022;39:110719. [DOI] [PubMed] [Google Scholar]
  • 198.Holla S, Prakhar P, Singh V, Karnam A, Mukherjee T, Mahadik K, Parikh P, Singh A, Rajmani RS, Ramachandra SG, Balaji KN. MUSASHI-Mediated expression of JMJD3, a H3K27me3 demethylase, is involved in Foamy Macrophage Generation during mycobacterial infection. PLoS Pathog. 2016;12:e1005814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Sun F, Abreu-Rodriguez I, Ye S, Gay S, Distler O, Neidhart M, Karouzakis E. TET1 is an important transcriptional activator of TNFalpha expression in macrophages. PLoS ONE. 2019;14:e0218551. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Cull AH, Snetsinger B, Buckstein R, Wells RA, Rauh MJ. Tet2 restrains inflammatory gene expression in macrophages. Exp Hematol. 2017;55:56–70. e13. [DOI] [PubMed] [Google Scholar]
  • 201.Williams K, Christensen J, Pedersen MT, Johansen JV, Cloos PA, Rappsilber J, Helin K. TET1 and hydroxymethylcytosine in transcription and DNA methylation fidelity. Nature. 2011;473:343–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Dawlaty MM, Ganz K, Powell BE, Hu YC, Markoulaki S, Cheng AW, Gao Q, Kim J, Choi SW, Page DC, Jaenisch R. Tet1 is dispensable for maintaining pluripotency and its loss is compatible with embryonic and postnatal development. Cell Stem Cell. 2011;9:166–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Figueroa ME, Abdel-Wahab O, Lu C, Ward PS, Patel J, Shih A, Li Y, Bhagwat N, Vasanthakumar A, Fernandez HF, Tallman MS, Sun Z, Wolniak K, Peeters JK, Liu W, Choe SE, Fantin VR, Paietta E, Lowenberg B, Licht JD, Godley LA, Delwel R, Valk PJ, Thompson CB, Levine RL, Melnick A. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell. 2010;18:553–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Zou Z, Dou X, Li Y, Zhang Z, Wang J, Gao B, et al. RNA m(5)C oxidation by TET2 regulates chromatin state and leukaemogenesis. Nature. 2024;634:986–94. [DOI] [PMC free article] [PubMed]
  • 205.Lv H, Liu B, Dai Y, Li F, Bellone S, Zhou Y, et al. TET3-overexpressing macrophages promote endometriosis. J Clin Invest. 2024;134:e181839. [DOI] [PMC free article] [PubMed]
  • 206.Zhu C, Cai Y, Mo S, Zhu J, Wang W, Peng B, Guo J, Zhang Z, Chen X. NF-kappaB-mediated TET2-dependent TNF promoter demethylation drives mtb-upregulation TNF expression in macrophages. Tuberculosis (Edinb). 2021;129:102108. [DOI] [PubMed] [Google Scholar]
  • 207.Rasmussen KD, Jia G, Johansen JV, Pedersen MT, Rapin N, Bagger FO, Porse BT, Bernard OA, Christensen J, Helin K. Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis. Genes Dev. 2015;29:910–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Rigo R, Chelbi R, Agopian J, Letard S, Griffon A, Ghamlouch H, et al. TET2 regulates immune tolerance in chronically activated mast cells. JCI Insight. 2022;7:e154191. [DOI] [PMC free article] [PubMed]
  • 209.Pronier E, Imanci A, Selimoglu-Buet D, Badaoui B, Itzykson R, Roger T, Jego C, Naimo A, Francillette M, Breckler M, Wagner-Ballon O, Figueroa ME, Aglave M, Gautheret D, Porteu F, Bernard OA, Vainchenker W, Delhommeau F, Solary E, Droin NM. Macrophage migration inhibitory factor is overproduced through EGR1 in TET2(low) resting monocytes. Commun Biol. 2022;5:110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Li J, Li L, Sun X, Deng T, Huang G, Li X, Xie Z, Zhou Z. Role of Tet2 in regulating adaptive and innate immunity. Front Cell Dev Biol. 2021;9:665897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Kallin EM, Rodriguez-Ubreva J, Christensen J, Cimmino L, Aifantis I, Helin K, Ballestar E, Graf T. Tet2 facilitates the derepression of myeloid target genes during CEBPalpha-induced transdifferentiation of pre-B cells. Mol Cell. 2012;48:266–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Zhang Q, Zhao K, Shen Q, Han Y, Gu Y, Li X, Zhao D, Liu Y, Wang C, Zhang X, Su X, Liu J, Ge W, Levine RL, Li N, Cao X. Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6. Nature. 2015;525:389–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Fan W, Liu P, Tan L, Lv H, Zhou H, Tao Z, Xu Y. Tet2 modulates M2 macrophage polarization via mRNA 5-methylcytosine in allergic rhinitis. Int Immunopharmacol. 2024;143:113495. [DOI] [PubMed] [Google Scholar]
  • 214.DiNardo AR, Rajapakshe K, Nishiguchi T, Grimm SL, Mtetwa G, Dlamini Q, Kahari J, Mahapatra S, Kay A, Maphalala G, Mace EM, Makedonas G, Cirillo JD, Netea MG, van Crevel R, Coarfa C, Mandalakas AM. DNA hypermethylation during tuberculosis dampens host immune responsiveness. J Clin Invest. 2020;130:3113–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Winkler R, Gillis E, Lasman L, Safra M, Geula S, Soyris C, Nachshon A, Tai-Schmiedel J, Friedman N, Le-Trilling VTK, Trilling M, Mandelboim M, Hanna JH, Schwartz S, Stern-Ginossar N. M(6)a modification controls the innate immune response to infection by targeting type I interferons. Nat Immunol. 2019;20:173–82. [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 datasets were generated or analysed during the current study.


Articles from Epigenetics & Chromatin are provided here courtesy of BMC

RESOURCES