- 1Liyuan Cardiovascular Center, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- 2Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- 3Department of Rehabilitation, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- 4Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
Macrophages are essential immune cells involved in pathogen clearance, initiation and resolution of inflammation, and tissue repair across multiple organ systems. They exhibit remarkable phenotypic diversity, encompassing classical M1 and M2 subtypes-further subdivided into M2a, M2b, M2c, and M2d-as well as newly identified subsets such as Mreg, M4, Mox, and Mhem, each with distinct functional roles. Emerging evidence highlights cellular metabolism as a central regulator of macrophage phenotype and function. Distinct metabolic programs underpin the polarization of M1 and M2 macrophages in response to environmental cues, thereby critically influencing disease progression and tissue outcomes. Cardiovascular disease remains a leading cause of morbidity and mortality worldwide. In the heart, macrophages represent a dominant immune cell population and play integral roles in both pathological injury and tissue regeneration. This review provides a comprehensive overview of macrophage ontogeny, phenotypic heterogeneity, and metabolic reprogramming, with a particular focus on their roles in inflammatory heart diseases. We synthesize current findings on how metabolic pathways shape macrophage behavior and function within the cardiac microenvironment and discuss the therapeutic potential of targeting macrophage metabolism to modulate inflammation, promote repair, and improve clinical outcomes in cardiovascular disease.
1 Introduction
Cardiovascular diseases remain a leading cause of morbidity and mortality worldwide, particularly in developed countries (1). Accumulating evidence indicates that immune cells-including macrophages, neutrophils, T Lymphocytes (T cells), and B Lymphocytes (B cells)-play critical roles in both the injury and repair phases of inflammatory heart diseases (2). Among these, macrophages represent the predominant immune cell population in the heart and are indispensable in shaping cardiac inflammation, remodeling, and regeneration (3–6). Notably, increasing attention has been directed toward the metabolic reprogramming of macrophages and its impact on cardiovascular pathophysiology (7–9). Macrophages are ubiquitously distributed throughout the body and serve as key mediators of innate immunity. Through phagocytosis, antigen presentation, and cytokine production, they orchestrate both proinflammatory and anti-inflammatory responses, while also facilitating tissue repair and homeostasis (10). Traditionally, macrophages have been broadly classified into two functional subsets: classically activated (M1) and alternatively activated (M2) macrophages (11). However, advances in immunology and transcriptomics have revealed a much more nuanced spectrum of macrophage phenotypes. M2 macrophages can be further subdivided into M2a, M2b, M2c, and M2d subtypes, each with distinct activation stimuli and functional roles. In addition, emerging subtypes such as regulatory macrophages (Mreg), M4, Mox, and Mhem have been identified, underscoring the complexity and plasticity of the macrophage population (12). Cellular metabolism is fundamental to maintaining survival, supporting biosynthetic demands, and driving immune cell function. In macrophages, metabolic pathways not only fulfill energy requirements but also shape immune phenotypes and functional responses. Recent studies have shown that macrophages undergo dynamic metabolic shifts in response to microenvironmental cues, and these shifts critically influence their role in disease progression and resolution (13).
In this review, we provide a comprehensive overview of macrophage ontogeny, phenotypic diversity, and functional specialization, with a particular focus on the metabolic programming that underlies these states. We further examine how macrophage metabolism contributes to the pathogenesis and resolution of inflammatory heart diseases, highlighting potential therapeutic targets to enhance cardiac repair and improve clinical outcomes.
2 Metabolism directs the function and polarization of macrophages
2.1 The functions of macrophages
2.1.1 Sources of macrophages
Macrophages are crucial components of the innate immune system. They can be classified into two categories: those derived from monocytes and those derived from tissue-resident macrophages (TRMs).
Generally, monocytes originate from the bone marrow. In the bone marrow, monocyte development includes the following progenitor cells: common myeloid progenitors (CMPs), granulocyte–macrophage progenitors (GMPs), and monocyte–macrophage and dendritic cell (DC) precursors (MDPs) (14). MDPs can differentiate into common monocyte progenitors (cMoPs), which may give rise to monocyte precursors. These precursors then develop into Ly6Chi monocytes, migrate from the bone marrow and circulate in the blood, and after a few days, they enter tissues and differentiate into macrophages (15). Monocyte-derived macrophages play a key role in clearing pathogens and cellular debris, antigen presentation, and initiating adaptive immune responses (16).
TRMs typically enter organs during embryonic or early postnatal development and exist largely independently of monocyte input throughout life (17). Many TRM populations (in the heart, lungs, liver, kidneys, and brain) originate from fetal liver or yolk sac progenitors (15, 18). There are also populations of TRMs (in the stomach, intestine, and colon) that originate from peripheral monocytes and are subsequently maintained through local proliferation (19, 20). TRMs are often replenished by monocytes, especially in infection or injury. During inflammatory disease, the initiation of inflammation in different tissues leads to a rapid decline in TRMs (21). At this time, inflammatory monocytes/macrophages are recruited to the site of inflammation, where they play an important role in immune defense but may also contribute to the pathogenesis of various inflammatory diseases and organ failure (22). In acute lung injury, peripheral blood monocytes can differentiate into interstitial macrophages and alveolar macrophages, and interstitial macrophages can further differentiate into alveolar macrophages (23). TRMs acquire unique functions on the basis of the organs in which they reside (24). In basal states, TRMs are considered functionally immunosuppressive; they play important roles in maintaining homeostasis, inhibiting T cell activation, and promoting the resolution of inflammation (25). However, in inflammatory or pathological conditions, the functions of TRMs can be altered significantly. For example, in the tumor microenvironment, TRMs can be reprogrammed to exhibit pro-tumorigenic effects by promoting angiogenesis and tumor invasion (26, 27). During inflammation, Alveolar macrophages have also been shown to initiate inflammation by secreting C-X-C Motif Chemokine Ligand 1 (CXCL1), which enhances the recruitment of neutrophils (21).
2.1.2 Polymorphism of macrophages
Monocyte-derived macrophages differentiate into different phenotypes on the basis of various environmental stimuli, with the most classic classification being M1 and M2 macrophages (28). In addition to the classic classification, Mreg macrophages are another newly identified type (29) (Table 1).
M1 macrophages arise in response to activation by pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and proinflammatory cytokines (such as interferon-γ, IFN-γ and tumor necrosis factor, TNF) (30). Following LPS activation, Fcγ receptor-mediated phagocytosis and Mitogen-Activated Protein Kinase (MAPK), Janus Kinase 1(JAK1), and JAK3 signaling are activated in M1 macrophages (31). These pathways control several inflammatory genes, endowing macrophages with proinflammatory characteristics (32, 33). M1 macrophages play crucial roles in defending against pathogens and initiating inflammatory responses. These cells express markers such as Cluster of Differentiation 86 (CD86), CD68, MHC class II molecules, and inducible nitric oxide synthase (iNOS). Moreover, they secrete high levels of proinflammatory cytokines, including interleukin-1β (IL-1β), IL-6, IL-12, IL-23, and TNF-α, thereby promoting inflammation and cytotoxicity (34). M1 macrophages can produce abundant reactive oxygen species (ROS) and reactive nitrogen species (RNS) to combat pathogen invasion. Concurrently, they secrete proinflammatory cytokines and stimulate the polarization of T helper 1 (Th1) cells, resulting in robust inflammation (35). However, the excessive proinflammatory and cytotoxic actions of these macrophages can lead to severe tissue damage (36).
In contrast to M1 macrophages, M2 macrophages play a central role in tissue repair, immune regulation, and fibrosis (37). These cells are typically activated by cytokines secreted by T helper 2 (Th2) cells and exhibit reduced antigen-presenting capabilities along with elevated levels of anti-inflammatory cytokines, such as IL-10 and transforming growth factor-β (TGF-β) (38). This anti-inflammatory effect helps to balance excessive inflammatory responses. However, the overactivation of M2-like macrophages can lead to tissue fibrosis, potentially resulting in organ dysfunction (35). M2 macrophages can be divided into four subtypes, M2a, M2b, M2c, and M2d macrophages, each with distinct functions (28, 39). M2a macrophages are induced by IL-4 and IL-13, M2b macrophages are activated by IL-1R or TLR agonists, M2c macrophages are induced by IL-10 and glucocorticoids, and M2d macrophages are induced by A2 adenosine receptors (A2Rs) and IL-6 (28).
M2a macrophages express high levels of the mannose receptor (CD206) and secrete profibrotic factors, such as TGF-β, which promote tissue repair (12). M2a macrophages can activate the STAT6 pathway through the coreceptor IL-4Rα, a pathway that primarily mediates tissue repair and antifungal responses, inducing the production of arginase 1 (Arg 1). Following this, arginine is degraded into polyamines and proline. These byproducts increase cell division and the accumulation of collagen fibers, which are vital processes for the repair and regeneration of tissues (35).
M2b macrophages are phenotypically and functionally similar to Mregs and are capable of producing both proinflammatory and anti-inflammatory cytokines, such as TNF-α, IL-1β, IL-6, and IL-10 (12, 40). In lupus nephritis, Granulin (GRN) enhances M2b polarization, leading to increased release of pro-inflammatory factors, which in turn worsens kidney inflammation and fibrosis (41). However, in the myocardial ischemia-reperfusion injury model, α-lipoic acid (ALA) induces M2b polarization, reducing inflammatory damage to cardiomyocytes and promoting IL-10-mediated immune tolerance. This will improve heart function (42).
M2c macrophages, also known as deactivated macrophages, possess anti-inflammatory functions. They secrete IL-10 and TGF-β and effectively phagocytose and eliminate apoptotic cells (28). Recent studies reveal that peroxisome proliferator-activated receptor γ (PPARγ)-dependent lipid synthesis is crucial for their repair functions—dephosphorylation of PPARγ T166 enhances fatty acid metabolism, fueling the production of PDGF, TGF-β, and VEGF to support tissue regeneration in skin and liver injuries (43).
M2d macrophages exhibit phenotypic and functional resemblance to tumor-associated macrophages (TAMs) (35). They secrete IL-10, TGF-β, and vascular endothelial growth factor (VEGF), promoting angiogenesis and tumor metastasis (44). For example, in gastric cancer, M2d macrophages can secrete these cytokines to promote tumor proliferation and metastasis (44, 45).
Mregs, a special subtype, are primarily formed through the differentiation of bone marrow precursors and peripheral blood mononuclear cells in in vivo microenvironments under special circumstances (35). Dehydrogenase/Reductase Member 9 (DHRS9) is a unique and stable marker of Mregs (46). Mregs possess distinct immunosuppressive functions (47) and play a key role in immune responses, especially during the process of tissue damage repair. A notable characteristic of Mregs is their high expression of MHC-II and CD80 molecules, which not only endows them with potent immunosuppressive capabilities but also enables them to effectively present antigens (46). Mregs can suppress immune responses both directly and indirectly. They can directly inhibit the proliferation of activated allogeneic and xenogeneic T lymphocytes and can also indirectly suppress immune responses by inducing regulatory T cells (Tregs) or by secreting molecules such as IL-10, TGF-β, NOS, and Indoleamine 2,3-Dioxygenase (IDO) (28, 35, 47).
Unlike monocyte-derived macrophages, TRMs present distinct phenotypes under homeostatic conditions. TRMs are characterized primarily by the expression of different markers according to specific phenotypes; monocyte-derived macrophages are characterized by the expression of CD11b, CD209, CD64, and c-mer tyrosine kinase (MerTK) (47). TRMs are divided into different subpopulations, such as Kupffer cells in the liver, alveolar macrophages in the lungs, microglia in the brain, and macrophages in the heart, on the basis of their anatomical location and function (37). TRM is a complex cell population. The current definition of TRM is not complete, and markers vary across different organs. For example, kupffer cells in the liver highly express inhibitor of DNA binding 3 (ID3) and Dectin1; alveolar macrophages express CD11c, sialic acid-binding Ig-like lectin F (Siglec-F) (in mice) or CD169 (in humans); and microglia express purinergic receptor P2Y, G-protein coupled, 12 (P2RY12) and transmembrane protein 119 (TMEM119) (48–50). Therefore, further researches are needed to provide accurate clues. Studies show that TRMs can also polarize according to their functions. For instance, alveolar macrophages can be categorized into two groups: M1 macrophages, which are characterized by pro-inflammatory and profibrotic activities and actively recruit neutrophils, and M2 macrophages, which exhibit anti-inflammatory and anti-fibrotic properties, promote asthma development, and facilitate tissue regeneration and resolution of inflammation (21). Microglia in the M1 polarization state exhibit strong pro-inflammatory and phagocytic functions, whereas in the M2 polarization state, they possess anti-inflammatory and tissue repair functions (51).
In human and murine atherosclerotic lesions, other types of macrophages have been identified, such as M4, Mox, Mhem, and M(Hb) macrophages (12). M4 macrophages, which are induced by the platelet chemokine CXCL-4, primarily express CD68, the calcium-binding protein S100A8, and matrix metalloproteinase (MMP) 7 in the adventitia and intima of arteries, and they are associated mainly with plaque instability (52). Mox macrophages have been identified in the atherosclerotic plaques of mice, where they are induced by oxidized phospholipids (oxPL). Mox macrophages account for up to 30 % of total plaque macrophages, and their specific functions remain to be further investigated (53). M(Hb) and Mhem macrophages are two subtypes of macrophages found in atherosclerotic plaques that coexist at sites of neovascular bleeding or unstable plaques (54). M(Hb) macrophages, which are stimulated by hemoglobin, typically express high levels of mannose and CD163 receptors and are involved in the clearance of hemoglobin/haptoglobin (Hb/Hp) complexes after plaque hemorrhage (55). After Hb/Hp complexes and erythrocytes are engulfed, the released heme can shift macrophages toward the Mhem phenotype (56). This particular subtype is capable of inhibiting the development of foam cells and offers specific protective benefits against atherosclerosis (57).
2.2 Metabolism directs the function and polarization of macrophages
Traditional cellular metabolism includes anabolism and catabolism, which produce ATP and biosynthetic precursors necessary for cell maintenance and growth. Like those of most cells, the metabolic pathways of resting macrophages are primarily based on the uptake of glucose and fatty acids. The aerobic oxidation of glucose and β-oxidation of fatty acids, accompanied by a robust tricarboxylic acid (TCA) cycle and mitochondrial oxidative phosphorylation (OXPHOS), continuously generate ATP to supply energy. An increasing number of studies indicate that metabolic processes and reactions play broader roles in immune effects (58). With the alteration of homeostasis in the body and the infiltration of related pathological products and exogenous substances, macrophages can activate their associated metabolic patterns to adapt to these changes (59). M1 and M2 macrophages have distinct metabolic characteristics depending on their degree of polarization. Typically, M1 macrophages express iNOS to produce nitric oxide (NO) from arginine. They exhibit enhanced glycolytic metabolism, an active pentose phosphate pathway (PPP), fatty acid synthesis, and impaired function of the TCA cycle and OXPHOS (60) (Figure 1). In contrast, M2 macrophages metabolize arginine through Arg-1, which is characterized by increased OXPHOS, fatty acid synthesis, and glutamine metabolism, as well as a suppressed PPP (60) (Figure 2).

Figure 1. Metabolism in M1 macrophages. M1 macrophages exhibit increased glycolysis; an increase in CARKL leads to increased flux through the PPP. Owing to the suppression of IDH and SDH production, the TCA cycle is interrupted, leading to the accumulation of citrate and succinate. Citrate is exported from the mitochondria via a citrate transporter and participates in lipid synthesis. The accumulation of succinate stabilizes HIF-1α, inducing the activation of NF-κB, which promotes inflammation. The increased expression of SREBP-1α leads to increased fatty acid synthesis. FAO is also increased, facilitating inflammasome activation. M1 macrophages highly express iNOS, promoting the synthesis of NO from arginine, which further enhances inflammation (figure was created with Biorender.com).

Figure 2. Metabolism in M2 macrophages. M2 macrophages exhibit reduced expression of CARKL, leading to the inhibition of the PPP. They have an intact TCA cycle. AMPK is involved in the activation of OXPHOS and FAO, which are their primary functional pathways. They also highly express arginase, which catalyzes the hydrolysis of arginine. Glutamine metabolism is increased, promoting the epigenetic activation of M2-type genes in macrophages (figure was created with Biorender.com).
2.2.1 Characteristics and metabolism of macrophages
2.2.1.1 Glucose metabolism
Various types of macrophages harness distinct glycolytic pathways to derive energy. M1 macrophages exhibit increased glycolytic activity, which is associated with their proinflammatory functions (60). This reliance on glycolysis, despite its lower ATP yield compared to oxidative phosphorylation, may prioritize speed over efficiency—a logical adaptation for rapidly responding to pathogens or tissue damage. LPS-induced bone marrow-derived macrophages (BMDMs) exhibit increased glycolytic metabolism (61). In M1 macrophages, the production of isocitrate dehydrogenase (IDH) and succinate dehydrogenase (SDH) is inhibited, leading to the disruption of the TCA cycle and the accumulation of citrate and succinate (59). The accumulated citrate is exported from the mitochondria via a citrate transporter involved in fatty acid production (62). This citrate shunt likely supports membrane biogenesis and prostaglandin synthesis, underscoring how metabolic intermediates directly influence inflammatory output. Activated M1 macrophages increase succinate levels, which stabilize hypoxia-inducible factor 1-alpha (HIF-1α), further promoting glycolysis (63, 64). HIF-1α is a transcription factor essential for the induction of multiple enzymes involved in glycolysis and may also be involved in the activation of NF-κB (63). High glucose-6-phosphate dehydrogenase (a key enzyme in the PPP) expression also activates the NF-κB signaling pathway, leading to intracellular oxidative stress and the induction of M1 macrophage polarization (62, 65). In contrast, M2 macrophages have an intact TCA cycle and can obtain the required energy through oxidative phosphorylation (66). The metabolic dichotomy between M1 and M2 macrophages mirrors their functional divergence: M1 cells sacrifice efficiency for rapid effector functions, while M2 cells prioritize energy conservation for tissue repair and homeostasis. M1 macrophages can produce nicotinamide adenine dinucleotide phosphate hydrogen (NADPH) through the PPP, which is involved in fatty acid synthesis for the production of inflammatory mediators and cell membrane remodeling (67). LPS-stimulated BMDMs exhibit increased PPP flux (68, 69). Carbohydrate kinase-like protein (CARKL), a protein resembling a carbohydrate kinase, restricts the PPP. Haschemi et al. found that CARKL expression decreases significantly during LPS-induced macrophage activation, triggering pro-inflammatory factor production. Conversely, when BMDMs are activated by IL-4, CARKL expression rises, helping to suppress the PPP (68). Studies have also found that macrophages induced by OxPL, known as Mox macrophages, exhibit unique metabolic features that are distinct from those of the traditional M1 and M2 macrophages. Mox macrophages rely on glucose metabolism and the PPP to support the production of glutathione (GSH) and the expression of Nrf2-dependent antioxidant genes (70).
2.2.1.2 Lipid metabolism
The activation of macrophages is closely related to changes in lipid metabolism. Lipidomic studies have elucidated the importance of lipid metabolism in the polarization of macrophages to an inflammatory phenotype. The synthesis of fatty acids in M1 macrophages is positively regulated to promote proinflammatory functions (71, 72). In LPS-activated BMDMs, the expression of sterol regulatory element-binding protein 1α (SREBP-1α) increases, activating not only fatty acid synthesis but also the Nlrp1α gene, a core component of the inflammasome, which promotes the secretion of proinflammatory cytokines (73). In M1 macrophages, fatty acid synthetase (FAS) is a key factor in regulating fatty acid synthesis, and its absence can prevent chronic inflammation in mice (74). Researchers believe that FAS is necessary for macrophage membrane remodeling; a deficiency in FAS leads to changes in cell membrane components and alterations in the transport of Rho GTPases, thereby reducing inflammatory signaling in macrophages (75). The preference of M1 macrophages for fatty acid synthesis may be related to their pro-inflammatory functions and the regulation of signaling pathways. However, M2 macrophages primarily utilize OXPHOS and fatty acid oxidation (FAO) as their main functional pathways, engaging in activities related to tissue damage repair (76). Adenosine monophosphate-activated protein kinase (AMPK) is a key energy-sensing enzyme, which can be regulated by multiple factors, including the ATP/AMP ratio, phosphorylation by upstream kinases (such as LKB1 and CaMKKβ), and metabolic stress signals (77). The AMPK signaling pathway is activated in the M2 subtype and is involved in increasing OXPHOS and FAO (78). AMPK activation is important for macrophage polarization. For instance, IL-25 induces mitophagy by activating AMPK and promoting the polarization of macrophages toward the M2 phenotype (79). Meanwhile, quercetin inhibits inflammation associated with M1 macrophage polarization and promotes M2 macrophage polarization by activating the AMPK and Akt signaling pathways (80). In hyperhomocysteinemia, intermedin can rescue the imbalance of the M1/M2 macrophage ratio by activating the AMPK pathway (81). Wang et al. (82) discovered that endosome-associated trafficking regulator 1 (ENTR1) can inhibit M1 macrophage polarization by enhancing AMPK phosphorylation. Interestingly, recent studies found that FAO may also contribute to inflammasome activation. Previous research revealed that NADPH oxidase 4 (NOX4), a source of cellular superoxide anions, induces FAO through carnitine palmitoyltransferase 1A (CPT1A) during NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation and helps in the development of the inflammatory macrophage phenotype (76). Through transcriptomics and extracellular flux analysis, it has been found that in an obese environment, adipose tissue macrophages (ATMs) show an increase in both glycolysis and OXPHOS (83). This metabolic feature is dependent on factors originating from adipose tissue. Meanwhile, human macrophages isolated from the adipose tissue of obese individuals also exhibit a similar metabolic profile, which is associated with a pro-inflammatory phenotype (83).
The specific mechanisms by which differences in fatty acid metabolism lead to opposing activation patterns and distinct functions in macrophages need to be investigated further. The proinflammatory and anti-inflammatory phenotypes may influence different metabolic patterns because of their differing dependencies on energy demands and conversion efficiency (66).
2.2.1.3 Amino acid metabolism
Amino acid synthesis, degradation, and interconversion play significant roles in the polarization and function of macrophages. They can be transformed into various physiologically active substances or generate specific chemical groups through appropriate degradation. Arginine metabolism is a key metabolic pathway that regulates macrophage polarization and inflammatory responses (84). In macrophages, the arginine metabolic pathways mediated by iNOS and arginase play important roles in the M1 and M2 processes. iNOS catalyzes the conversion of arginine to NO. M1 macrophages highly express iNOS, which is essential for their inflammatory functions. In contrast, the arginase pathway, which breaks down arginine and thus reduces NO production, is highly expressed in M2 macrophages. This pathway regulates macrophage proliferation and collagen synthesis (66). Glutamine metabolism also plays a crucial role in regulating immune cell functions. Glutamine metabolism is also a key input for the synthesis of NO through arginine, demonstrating the role of glutamine in macrophage cytotoxicity and antibacterial functions (66). Another characteristic of M2 macrophages compared with M1 macrophages is increased glutamine metabolism. Alpha-ketoglutarate produced by glutamine catabolism promotes the epigenetic activation of M2-type genes in macrophages, whereas glutamine is not required for the development of LPS-stimulated M1-type macrophages (69). Recent studies have shown that the inhibition of serine metabolism can enhance the polarization of M1 while inhibiting the polarization of M2 (85). Additionally, the depletion of intracellular citrulline is crucial for the polarization of proinflammatory macrophages and immune responses (86). Researchers have revealed that impaired glutamine catabolism in macrophages exacerbates atherosclerosis. High-throughput transcriptional and metabolic analyses have shown that the phagocytic ability of macrophages depends on the atypical transaminase pathway rather than traditional glutamate dehydrogenase 1 (GLUD1) to support alpha-ketoglutarate-dependent immune metabolism (87).
2.2.1.4 Trace element metabolism
In addition to the aforementioned metabolic processes, the metabolism of certain trace elements also plays a crucial role in the function of macrophages. Macrophages handle most of the iron released from aging and damaged red blood cells, returning it to the circulation or storing it in ferritin within the cytoplasm to maintain iron homeostasis (88). Iron significantly affects macrophage polarization. Generally, with high levels of intracellular iron activating proinflammatory M1 macrophages while reducing the numbers of anti-inflammatory M2 macrophages (89). Over 60 % of iron-related genes are differentially expressed between M1 and M2 macrophages, which may explain why iron levels in M2 macrophages are typically lower than those in M1 macrophages (90). In the non-alcoholic steatohepatitis (NASH) model, Kupffer cells with iron accumulation exhibit a pro-inflammatory and profibrotic phenotype. Iron accumulation activates the MiT/TFE transcription factors, which in turn upregulate the expression of pro-inflammatory and profibrotic genes (91). However, recent studies found that in some cases, iron-rich macrophages also have anti-inflammatory effects. In macrophages infected with Salmonella, restricting intracellular iron appears to change immune function; for example, IFN-γ can increase the expression of the iron exporter ferroportin (Fpn), increasing iron efflux while stimulating the production of TNF-α (88). Conversely, the absence of nitric oxide synthase 2 (Nos2) reduces Fpn, leading to increased iron accumulation in macrophages and the reduced production of cytokines such as TNF-α, IL-12, and IFN-γ (92). The reduction in iron in macrophages diminishes the signaling of TLR4/TRIF-associated adaptor molecules, leading to decreased production of TNF-α, IL-6, and IFN-β in response to LPS stimulation or Salmonella infection (93). MFehi macrophages are a subset of adipose tissue-resident macrophages characterized by high intracellular iron stores (94). MFehi macrophages exhibit gene expression profiles associated with anti-inflammatory functions and iron recycling (95). In the obese state, the iron-handling capacity of MFehi macrophages is impaired, they become more pro-inflammatory. These changes occur concurrently with adipocyte iron overload and reduced adipocyte adiponectin expression (95). The impact of iron metabolism and other trace element metabolisms on macrophage function still requires further investigation.
2.2.2 Mitochondrial function and biogenesis on macrophages
Mitochondria are crucial for the viability of eukaryotic cells, as they play important roles in bioenergetics, metabolism, and signaling (96). Many studies have focused on the impact of mitochondrial function and biogenesis on macrophages.
Mitochondrial fusion and fission can regulate macrophage polarization and function. Ganglioside-induced differentiation-associated protein 1 (GDAP1L1) phosphorylates dynamin-related protein 1 (Drp1), promoting its translocation from the cytosol to mitochondria, leading to mitochondrial fission. This will promote the M1 macrophage polarization. Consequently, the production of cytokines and chemokines is augmented via the MAPK and NF-κB pathways, leading to psoriasis inflammation (97). In contrast, MFN-mediated normal mitochondrial fusion could promote the M2 macrophage phenotype and inhibit the M1 phenotype. According to the study by Li et al. (98), Xuanfei Baidu Formula (XBF) alleviates casp11-dependent NLRP3 inflammasome activation through promoting MFN1-induced mitochondrial fusion, which prevent macrophage polarization to the M1 pro-inflammatory phenotype.
Mitochondrial biogenesis can also regulate macrophage polarization and function. Research indicates that γ-amino butyric acid (GABA) suppresses α-ketoglutarate dehydrogenase (OGDH) activity, lowering succinyl-CoA (SCOA) levels. This reduction diminishes mitochondrial protein succinylation, thereby boosting oxidative phosphorylation efficiency and promoting M2 macrophage polarization (99). Olfactomedin-like protein 3 (OLFML3) protein enhances M2 polarization by promoting immune-responsive gene 1 (IRG1) mitochondrial localization, catalyzing itaconate production, which maintaining mitochondrial membrane potential and mitochondrial reactive oxygen species (mtROS) homeostasis (100). During chronic kidney disease (CKD), the AMPK signaling pathway is inhibited, which result in mitochondrial biogenesis (nuclear respiratory factor 1 and mitochondrial transcription factor A) as well as mitochondrial-specific proteins (cytochrome C and mitochondrial complex IV) downregulated, leading to M1 polarization of macrophages (101).
2.3 Effects of metabolism on macrophages
Some cytokines, metabolic intermediates, and metabolites also have interactive effects on the metabolism of macrophages. They may originate from the macrophages themselves, whereas others may originate from the circulation or other cells (Table 2).
2.3.1 Cytokines
2.3.1.1 IFN-γ
The activation of macrophages to the M1 state through inflammatory stimuli such as IFN-γ is associated with increased glycolytic metabolism (102). The enhancement of glycolytic metabolism is associated with the activation of 6-phosphofructo-2-kinase (PFK 2). Phosphofructokinase-1 (PFK1) is a key enzyme that regulates the flux of glycolysis, and PFK2, the most effective allosteric activator of PFK1, catalyzes the formation of fructose 2,6-bisphosphate. Under stimulation by IFN-γ, the expression of PFK 2 in the liver shifts to a more active, ubiquitously expressed isoform (uPFK2), which helps maintain higher concentrations of fructose 2,6-bisphosphate, thereby enhancing the glycolytic process (66). The increased glycolytic rate allows M1 macrophages to rapidly produce enough energy and biosynthetic intermediates to respond quickly to hypoxic microenvironments (69).
2.3.1.2 IL-4
Unlike those stimulated with IFN-γ, macrophages stimulated with IL-4 primarily undergo glucose metabolism and an energy supply through the TCA cycle and OXPHOS. The TCA cycle in M2 macrophages is involved in the production of uridine diphospho-N-acetylglucosamine (UDP-GlcNAc) intermediates, which are associated with the clearance function of M2 macrophages (66). IL-4 promotes mitochondrial respiration and FAO in M2 macrophages by activating PPARγ coactivator 1β (PGC-1β) (66).
2.3.1.3 IL-10
IL-10 can inhibit LPS-induced glucose uptake and glycolysis in macrophages and promote OXPHOS (103). Furthermore, IL-10 suppresses the activity of the mammalian target of rapamycin (mTOR) by inducing the mTOR inhibitor DDIT4. Consequently, IL-10 promotes mitophagy, which eliminates dysfunctional mitochondria characterized by low membrane potential and high levels of ROS (103). In the absence of IL-10 signaling, macrophages in mouse models of colitis and inflammatory bowel disease accumulate damaged mitochondria, leading to the dysregulation of NLRP3 inflammasome activation and IL-1β production (103).
2.3.1.4 TNF-α
NAD+ is an important intracellular redox coenzyme. Studies have shown that TNF-α is related to NAD+ levels in macrophages. M1 macrophages stimulated with LPS exhibit biphasic and transient increases in NAD+ levels associated with the release of TNF-α (104). TNF-α can regulate the expression of several enzymes involved in NAD+ homeostasis (104). TNF-α can regulate the expression of nicotinamide phosphoribosyl transferase (NAMPT), which is a key enzyme in the NAD+ salvage pathway (104). By affecting the expression of NAMPT, TNF-α indirectly influences the synthesis of NAD+.
2.3.2 Metabolic intermediates
2.3.2.1 Lactate
Lactate, the end product of glycolysis, has long been considered a metabolic waste product (105). However, an increasing body of evidence suggests that lactate has multiple biological functions; it acts as a signaling molecule, regulating various processes such as metabolism, immune responses, and cell–cell communication (106). As a central molecule, lactate plays a crucial role in modulating macrophage metabolism. Studies have shown that lactate can inhibit the polarization of M1 proinflammatory macrophages while promoting the polarization of M2 macrophages toward an anti-inflammatory and proangiogenic phenotype, with these effects being mediated through various mechanisms, including histone posttranslational modifications and signaling pathway regulation (107, 108). Lactate increases phosphorylation activation of the signal-regulated kinase (ERK)/ signal transducer and activator of transcription 3 (STAT3) pathway in macrophages and promotes M2 polarization (109). In 2019, Zhang et al. (110) reported that M1 macrophages possess an endogenous “lactate clock,” which regulates the expression of M2-type characteristics in M1 macrophages during the later stages of polarization through histone lactylation. Some studies have pointed out that, under specific circumstances, lactylation can alter the activity of glycolytic enzymes, thereby inhibiting glycolysis, reducing lactate production, and promoting the shift from M1 phenotype to M2 phenotype (109). The impact of lactate on macrophage polarization remains somewhat controversial and requires further investigation.
2.3.2.2 Citrate
The production and conversion of citrate link mitochondrial and cytoplasmic metabolism. Citrate is generated in the TCA cycle through the condensation of oxaloacetate and acetyl-CoA, with the latter derived from pyruvate produced by glycolysis or the breakdown of fatty acids. Citrate is converted to isocitrate and then transformed into α-ketoglutarate (αKG) by IDH (111). Citrate entering the cytoplasm can indirectly inhibit glycolysis by acting on pyruvate kinase (PK), stimulate fatty acid synthesis by activating acetyl-CoA carboxylase (ACC), and promote gluconeogenesis by activating fructose-1,6-bisphosphatase (111). As previously introduced, the disruption of the TCA cycle in M1 macrophages leads to the accumulation of citrate. Owing to the downregulation of IDH expression and the upregulation of the expression of the mitochondrial citrate carrier (CIC), M1 macrophages characteristically accumulate citrate, which is then transported out of the mitochondria (112). This accumulation and transfer of citrate are crucial for the functions of M1 macrophages, affecting the production of NO, ROS, and prostaglandin E2 (PGE2) in M1 macrophages (112, 113).
2.3.2.3 Itaconate
Itaconate is produced from cis-aconitate in the TCA cycle by the enzyme itaconate decarboxylase 1 (ACOD1). Itaconate has antibacterial properties and may also play a role in immune modulation, the suppression of inflammation, and the promotion of tolerance (114, 115). ACOD1 expression is upregulated in M1 macrophages (111). Reduced levels of itaconate favor M2-like polarization in macrophages, possibly due to the inhibition of SDH, which increases OXPHOS flux (116).
2.3.2.4 Succinate
Succinate is an intermediate product of the TCA cycle, is produced from succinyl-CoA and is a substrate for succinate dehydrogenase (SDH). As previously noted, the disruption of the TCA cycle in M1 macrophages also results in succinate accumulation. Succinate affects the stability of HIF-1α by inhibiting prolyl hydroxylase domain enzymes (PHDs), which are a class of αKG-dependent dioxygenases that can regulate HIF-1α stability in an oxygen-dependent manner, preventing its degradation (63). In the absence of ATP production, the substantial oxidation of succinate can lead to reverse electron transport (RET) to complex I, which is associated with a significant release of ROS and can activate HIF1-α in M1 macrophages (117, 118). HIF-1α can then interact with coactivators to induce the glycolytic metabolic program, and drive inflammation by increasing transcription of IL-1β (119). Although succinate is primarily associated with the pro-inflammatory responses of M1 macrophages, studies have also shown that it can promote the polarization of M2 macrophages through the succinate receptor-1(SUCNR-1) mediated signaling pathway (120). The impact of succinate on macrophage polarization and function is worth further investigation.
2.3.2.5 ROS
Elevated levels of ROS and hypoxia promote metabolic reprogramming in macrophages, making the cells more reliant on glycolytic processes (66). M1 macrophages generate ROS through the NADPH oxidase complex (such as NOX2) by catalyzing the one-electron reduction of oxygen, which is crucial for macrophage phagocytosis and antimicrobial defense (121). The formation of a high-concentration ROS microenvironment is accompanied by the upregulation of the expression of inflammatory markers such as iNOS, leading to increased glucose uptake and enhanced glycolytic metabolism (122). NO is produced by iNOS, which catalyzes the conversion of arginine and is involved in the proinflammatory response of macrophages (123).
2.3.3 Sirtuins
Sirtuins (SIRT), initially identified in yeast as the first nicotinamide adenine dinucleotide (NAD)+-dependent epigenetic and metabolic regulators, belong to the class III histone deacetylases. Their activity relies on the presence of NAD+ (124). The sirtuins protein family primarily comprises seven members. SIRT1, SIRT6, and SIRT7 are typically localized in the nucleus, where they are involved in chromosomal stability and transcriptional regulation; SIRT2 is mainly found in the cytoplasm but can also be present in the nucleus; SIRT3, SIRT4, and SIRT5 are usually located in the mitochondria, where they regulate metabolic enzymes and stress response mechanisms (125). The substrates of nuclear sirtuins include histones and non-histone proteins, such as nuclear transcription factors and cofactors. In contrast, cytoplasmic and mitochondrial sirtuins play crucial roles in key enzymes involved in oxidative and metabolic pathways, including glycolysis, fatty acid oxidation, and the TCA cycle (126).
Sirtuins influence macrophage polarization through multiple pathways. SIRT1/2 can directly inhibit the NF-κB signaling pathway by deacetylating the p65 subunit of NF-κB, reducing the expression of pro-inflammatory cytokines (such as IL-6, IL-1β, and TNF-α) and suppressing M1 polarization (127–129). SIRT1 can also upregulate the expression of STAT6 to promote M2 polarization (130). SIRT3 promotes M2 polarization by activating mitochondrial autophagy, which reduces the release of inflammatory cytokines like IL-1β and TNF-α (131, 132). SIRT4 decreases M1 polarization by inhibiting STAT3 and suppressing inflammatory factors such as ROS, MMP-13, IL-6, and TNF-α. Meanwhile, SIRT4 promotes M2 polarization, increases the mRNA levels of Arg-1 and CD206 and enhances the expression of the IL-10 (133, 134). SIRT5 promotes the acetylation of p65 and activates the NF-κB signaling pathway to augment IL-1β production during inflammation (135, 136). SIRT7 activates the NF-κB signaling pathway, increases the expression of IL-1β, and promotes inflammatory responses (137). In contrast, SIRT6 upregulates the expression of anti-inflammatory cytokines IL-4 and IL-10, increases M2 polarization, and attenuates inflammatory responses (138).
The pre-activation state of macrophages also affects the function and expression of Sirtuins. NAD+ is an important cofactor regulating metabolic homeostasis and a rate-limiting substrate for sirtuin deacylases (139). Studies have shown that supplementation with NMN (a precursor of NAD+) increases NAD+ levels and activate SIRT3 (140). Under pro-inflammatory conditions, M1 macrophages produce large amounts of ROS through glycolysis, which may lead to the depletion of NAD+ and indirectly inhibit the enzymatic activity of SIRT1 and SIRT3 (59, 141, 142). In contrast, M2 macrophages rely on oxidative phosphorylation and may maintain higher NAD+ levels through fatty acid oxidation and OXPHOS, thereby activating SIRT3 and promoting anti-inflammatory and tissue repair functions (59, 141, 143, 144).
2.3.4 NLRP3 inflammasome
The NLRP3 inflammasome is a high-molecular-weight multiprotein complex composed of NLR family members that assembles in response to various stimuli in the cytoplasm (145); it plays a key role in host defense, inflammation, autoimmunity, and the development of metabolic diseases (146). Macrophages undergo profound metabolic reprogramming when they are sensing infections and sterile stimuli, and this metabolic shift supports and regulates the activation of the NLRP3 inflammasome (147).
The role of glycolysis in inflammasome activation is complex and involves enzymes involved in the glycolytic process and changes in glycolytic flux. The blockade of pyruvate kinase M2 (PKM2) limits the classical activation of NLRP3 in mouse BMDMs (148). In contrast, the inhibition of hexokinase 2 (HK2) by N-acetylglucosamine (NAG) leads to the detachment of HK2 from the mitochondrial outer membrane, thereby increasing inflammasome activation in mouse BMDMs (149). Effective glycolysis results in the conversion of NAD+ to NADH, the secretion of lactate, and the production of ATP. The inhibition of GAPDH and α-enolase, leading to the interruption of glycolytic flux, triggers the NLRP3 inflammasome in mouse BMDMs in a K+-independent manner (150).
Intermediates in the TCA cycle play a significant role in cellular signaling, including the regulation of NLRP3 inflammasome activation. The accumulation of itaconate is associated with the inhibition of the NLRP3 inflammasome. Irg1−/− mouse BMDMs, which cannot synthesize itaconate, exhibit increased NLRP3 activation (114). Itaconate inhibits the activation of the NLRP3 inflammasome through various mechanisms, including posttranslational modifications of kelch-like ECH-associated protein 1 (KEAP1), NIMA-related kinase 7 (NEK7), and gasdermin-D (GSDMD) (151–155). Succinate plays a significant role in regulating NLRP3 inflammasome activation by succinylating the Cys192 site of GSDMD, inhibiting GSDMD oligomerization and pyroptosis (156).
Lipid metabolism is also crucial for NLRP3 inflammasome production. Saturated fatty acids (SFAs) can activate the NLRP3 inflammasome, whereas unsaturated fatty acids, such as oleic acid (OA), can counteract SFA-mediated NLRP3 activation or directly suppress NLRP3 activation (155, 157–159). Polyunsaturated fatty acids (PUFAs), such as docosahexaenoic acid (DHA), can sense and inhibit LPS-preactivated human macrophage NLRP3 activation (151, 152).
2.3.5 Metabolites
2.3.5.1 Blood glucose
Chronic hyperglycemia enhances the proinflammatory response in lipopolysaccharide-stimulated macrophages, including the production of TNF-α, IL-1β, and IL-6 (160). Studies have also shown that under high-glucose culture conditions, BMDMs and monocytes exhibit increased glycolysis, altered metabolic profiles, and a shift toward proinflammatory subtype (M1 type) polarization, promoting the expression of proinflammatory genes in macrophages. This may explain how hyperglycemia induces trained immunity in macrophages and their precursor cells (161).
2.3.5.2 Insulin
Insulin also exerts an impact on the metabolism and function of macrophages. Studies have shown that in patients with type 2 diabetes, the number of macrophages in adipose tissue and the circulating levels of TNF-α and IL-1β increase following the initiation of insulin therapy (162). Hyperinsulinemia can exacerbate the infiltration of macrophages into adipose tissue and promote the development of insulin resistance (163). A previous study revealed that insulin enhances the phagocytic capacity of macrophages and the production of hydrogen peroxide (H2O2) by promoting glucose metabolism and inhibiting glutamine metabolism, thereby strengthening their immune function (164).
2.3.5.3 Ketone bodies
Ketone bodies (KBs), which include β-hydroxybutyric acid (β-HB) and acetoacetic acid (AcAc), play critical roles in organismal energy homeostasis (165). Studies have shown that in atherosclerosis, β-HB inhibits Ox-LDL-induced macrophage proliferation (166). AcAc inhibits proliferation but exacerbates lipid accumulation (166). These two may exert opposite effects on atherosclerotic progression. β-HB could promote M2 macrophage polarization through the STAT6-dependent signaling pathway, and suppress M1 macrophage polarization through β-hydroxybutyrylation of the STAT1 protein (167, 168). When rats were treated with a ketone diet, it was found that ketogenic metabolism could promote the polarization of macrophages toward the M2 phenotype and inhibit inflammatory responses (169).
2.3.5.4 Heme
Heme is an early important substance in the activation of innate immunity and has been shown to directly act on macrophages, neutrophils, epithelial cells, and endothelial cells (170). As a typical proinflammatory response molecule, heme can directly induce macrophages to produce proinflammatory lipid mediators such as leukotriene B4 (LTB4) and release proinflammatory cytokines, leading to local to systemic inflammatory responses (171). Studies have also indicated that heme can stimulate NLRP3 inflammasome activation in endothelial cells, inducing macrophages to produce large amounts of IL-1β, which often leads to inflammatory responses in hemolytic diseases (172).
2.3.5.5 Blood lipids
The impact of blood lipids on macrophages is crucial in various types of inflammation. Mice fed a high-fat diet (Western diet) present increased levels of cholesterol and triglycerides, with the activation of endoplasmic reticulum stress pathways in cardiac macrophages leading to the activation of inflammatory genes and affecting myocardial inflammation in heart failure with preserved ejection fraction (173). Monocyte-derived macrophages can take up oxidized low-density lipoprotein in the subendothelial space of blood vessels, leading to the accumulation of cholesterol and the formation of foam cells, which is a hallmark of atherosclerotic disease progression (174). In contrast to LDL, high-density lipoprotein (HDL) inhibits the expression of surface markers and inflammation-related genes such as TNF-α, IL-6, and monocyte chemotactic protein-1 (MCP-1) in macrophages, reducing the production of ROS and RNS and blocking the polarization of macrophages toward the M1 type (175).
2.3.5.6 Bile acids
Bile acids (BAs) are steroid substances synthesized from cholesterol in the liver that are capable of binding to plasma proteins and entering the systemic circulation, where they are distributed to various organs throughout the body (such as the intestines, brain, and heart). BAs affect the polarization state of macrophages by activating different receptors, such as the farnesoid X receptor (FXR) and the G protein-coupled bile acid receptor 1 (TGR5) (176). The activation of TGR5 can reduce the secretion of TNF, IFN-γ, IL-6, and IL-1β in proinflammatory M1 macrophages and promote the polarization of anti-inflammatory M2 macrophages (176).
2.3.5.7 Creatinine
Creatinine is a slightly toxic metabolic product produced by muscle metabolism and is primarily excreted by the glomerular filtration of the kidneys, and its levels significantly increase after extensive exercise or the excessive consumption of meat. Macrophages mainly take up creatinine through solute carrier family 6 member 8 (SLC6A8), and subsequently, creatinine inhibits the IFN-γ/JAK/STAT1 signaling pathway, reducing the expression of iNOS, IL-12, and TNF-α and blocking the polarization of macrophages toward the M1 type. On the other hand, creatinine can promote chromatin remodeling and activate IL-4/STAT6 signaling, promoting the expression of M2 markers such as Arg-1 (177).
2.3.5.8 Uric acid
Uric acid is the end product of purine metabolism, and disturbances in purine metabolism often lead to abnormally high levels of blood uric acid, causing hyperuricemia, which is closely related to the occurrence of gouty arthritis, joint deformity, and gouty nephropathy. Studies have shown that uric acid significantly increases the expression of TNF-α and TLR4 in macrophages and enhances their phagocytic activity, while the expression levels of CD206, C-X3-C motif chemokine receptor 1 (CX3CR1), and C-C chemokine receptor 2 (CCR2) are significantly reduced, leading to M1-type polarization and the downregulation of urate transporter 1 (URAT1) expression (178). Probenecid treatment significantly inhibits UA-induced M1 macrophage polarization, reducing the production of proinflammatory cytokines and weakening their phagocytic activity (178).
3 The role of macrophages in inflammatory heart disease
3.1 Macrophages in the heart
3.1.1 Classification and function of macrophages in the heart
In the cardiac tissue microenvironment, macrophages constitute the largest group of immune cells. Cardiac macrophages can be divided into two major categories: infiltrating macrophages and resident macrophages.
Infiltrating cardiac macrophages are mainly CCR2+ macrophages derived from recruited monocytes, which play a major role in the inflammatory response (179). After myocardial infarction, macrophages and other inflammatory cells infiltrate the infarct area, leading to the production of pro-inflammatory cytokines, exacerbating inflammation, and clearing necrotic tissue (180). In ischemic myocardial injury, CCR2+ macrophages infiltrated via MCP-1 dominate the inflammatory and fibrotic responses in the early stage of injury (181). Additionally, in dilated cardiomyopathy, NLRP3 inflammasomes synthesized by infiltrating macrophages can promote the cleavage of apoptosis-associated speck-like protein (ASC), caspase-1, IL-1β, IL-18, and GSDMD into their active forms, which will exacerbate inflammation, induce cardiomyocyte pyroptosis and promote myocardial fibrosis (182, 183).
Resident cardiac macrophages originate from different macrophage precursor cells and possess distinct functions and behaviors (184). Resident cardiac macrophages in the heart can be distinguished on the basis of their expression of CCR2; CCR2− macrophages are derived from the primitive yolk sac and fetal monocyte precursors, whereas CCR2+ macrophages originate from hematopoietic progenitors in the blood (185, 186).
Under homeostatic conditions, resident cardiac macrophages are involved in a range of functions that are crucial for maintaining cardiac homeostasis. Studies have shown that CCR2− macrophages, which originate from the yolk sac, are necessary for the proper remodeling of the coronary arterial plexus and the functional maturation of coronary vessels (187). These findings also suggest that macrophage-derived insulin-like growth factor 1 (IGF-1) is a potential mediator of coronary arteriogenesis (187). CCR2− macrophage depletion in embryos leads to abnormalities in developmental processes and vascular maturation (187).
Resident cardiac macrophages mediate immune surveillance, which maintains the homeostasis of CCR2− and CCR2+ macrophages that express MHC II and act as antigen-presenting cells; they function by processing and presenting antigens to circulating T cells (184). Moreover, CCR2− resident cardiac macrophages expressing MERTK have a greater capacity for phagocytosing cardiac myocyte debris and contents than CCR2+ macrophages do. Several studies have described the efferocytosis of apoptotic cells by MERTK-expressing cardiac macrophages as an important contributing factor to cardiac homeostasis (188, 189). Additionally, MERTK-expressing resident cardiac macrophages in the hearts of young mice can protect the heart from infection by Mycobacterium avium, which has been shown to cause cardiac arrhythmias and promote bacterial dissemination to the cardiac tissues of older mice (190).
Importantly, studies have shown that resident cardiac macrophages promote mitochondrial homeostasis in cardiomyocytes (191). Mertk deletion in resident cardiac macrophages limits their phagocytic capacity and prevents them from taking up dysfunctional mitochondria (191). The disruption of mitochondrial clearance in macrophages impairs autophagy, leading to the accumulation of cellular debris and metabolic dysfunction in cardiomyocytes, which may result in impaired ventricular function (191).
With the development of technologies such as single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics, further classification of cardiac macrophage subsets has been achieved. Using scRNA-seq, trypanosome lytic factor (TLF+) macrophages have been identified (192–194). The replenishment of TLF+ macrophages is independent of circulating monocytes (192), and their transcriptomic features are mainly enriched in homeostatic functions, such as endocytosis, cellular transport, and angiogenesis (192, 193). The application of scRNA-seq in acute myocardial infarction research has further subdivided distinct subsets of cardiac macrophages. It was found that macrophages with high expression of interferon-stimulated genes (ISG+) and MHC-II+ macrophages are key pro-inflammatory subsets during the inflammatory phase, whereas macrophages with high expression of triggering receptor expressed on myeloid cells 2 (Trem2+) are the main anti-inflammatory subsets during the repair phase (194–196). A study using spatial transcriptomics analyzed different anatomical regions of the heart and identified a novel population of tissue-resident macrophages (LYVE1+ macrophages), which are present in the atrioventricular node and sinoatrial node (197). Another study utilizing spatial transcriptomics discovered a subset of monocyte-derived macrophages expressing basic helix-loop-helix family member e41 (Bhlhe41+) in the infarct area during the early stage of myocardial infarction (198). By analyzing spatial ligand-receptor interactions and combining experiments in animal models, the results showed that Bhlhe41+ macrophages can upregulate the secretion of progranulin (GRN). GRN antagonizes the effect of TNF-α on TNF receptor 1 (TNFR1), which will inhibit myofibroblast activation and limit the expansion of the infarct area (198). In the future, with the continuous advancement of technologies and in-depth research, the classification of cardiac macrophages will be further refined.
3.1.2 Crosstalk between cardiac macrophages and other cells
Cardiac macrophages participate in cross talk with fibroblasts, lymphocytes, endothelial cells, cardiomyocytes, and other cell types in the heart (199). Following myocardial infarction, pro-inflammatory M1-type macrophages release exosomes rich in miR-155, which suppress the expression of key genes such as Rac family small GTPase 1 (RAC1), p21-activated kinase 2 (PAK2), SIRT1, and AMP-activated protein kinase α2 (AMPKα2) in endothelial cells. This inhibition of gene expression could impair angiogenesis and exacerbates myocardial injury (200). During the healing phase of myocardial infarction, infiltrating cardiac macrophages can secrete Angiotensin II (Ang II), which activates the angiotensin II type 1 (AT1) receptor on fibroblasts, upregulates TGF-β1, induces the transformation of fibroblasts into myofibroblasts and increases collagen deposition (201). Meanwhile, infiltrating cardiac macrophages are capable of secreting exosomes enriched with miR-155; after being internalized by fibroblasts, these exosomes downregulate the expression of son of sevenless homolog 1 (SOS1) and inhibit fibroblast proliferation (202). Interestingly, the expression of angiopoietin 2 (Angpt2) in endothelial cells is increased, and Angpt2 can promote the polarization of macrophages toward a pro-inflammatory phenotype via the α5β1/extracellular ERK pathway (203). Similarly, after myocardial infarction, neutrophil gelatinase-associated lipocalin (NGAL) in the secretome of neutrophils affects the proliferation of cardiac macrophages by modulating the expression of the macrophage receptor MertK on the cardiac macrophage membrane (204). In the absence of NGAL, the expression of MertK on the cardiac macrophage membrane is reduced, which impairs the phagocytic capacity of macrophages (204). Th1 cells promote the polarization of pro-inflammatory macrophages, whereas Th2 cells and Treg cells (which secrete IL-10 and TGF-β) facilitate the acquisition of an anti-inflammatory macrophage phenotype. These regulatory effects ultimately influence myocardial healing and scar formation (205). A pioneering study reported that resident cardiac macrophages promote electrical conduction in the heart by intermingling with distal atrioventricular nodal conduction cells through macrophages expressing connexin 43 (206). Both mouse and human hearts are rich in CCR2− and CCR2+ macrophages in the atrioventricular node (206). Resident cardiac macrophages in the atrioventricular node maintain direct contact with cardiomyocytes through gap junctions. The removal of resident cardiac macrophages or conditional knockout of the Gja1 gene, which encodes connexin 43, leads to impaired atrioventricular conduction, ultimately resulting in atrioventricular block, thereby confirming that resident cardiac macrophages are necessary for cardiac electrical conduction. Furthermore, compelling studies have indicated that sudden cardiac death due to arrhythmias caused by myocardial infarction is a result of an imbalance in the cardiac leukocyte population (207, 208). More specifically, researchers have described an “electrical storm,” during which rapidly infiltrating neutrophils contribute to ventricular tachycardia (207, 208). The depletion of CCR2− resident cardiac macrophages or complete depletion of macrophages leads to an increased incidence of arrhythmias, demonstrating that macrophages have a protective effect on electrical conduction after myocardial infarction.
3.2 Macrophages function in inflammatory heart disease
Macrophages not only are crucial for maintaining cardiac homeostasis but also play a significant role in heart diseases (Table 3). Research has shown that, in heart-related diseases such as myocardial infarction, myocarditis, valvular heart disease, and heart failure, macrophage polarization, metabolic changes and functions are important (Figure 3).

Figure 3. Macrophages in inflammatory heart disease. Macrophages play different roles in various heart diseases. In atherosclerosis, the recruitment, polarization, and foam cell formation of macrophages affect plaque formation. After MI, cardiac macrophages play different roles at various stages, including pro-inflammatory functions, clearance of necrotic cells, and tissue repair. In aortic aneurysms, M1 macrophages drive the inflammatory process, while M2 macrophages promote vascular remodeling and repair. In myocarditis, M1 macrophages exacerbate the condition, while M2 macrophages alleviate inflammation. CCR2+ macrophages promote inflammatory responses in transplant rejection, whereas CCR2− macrophages have anti-inflammatory effects and promote coronary artery formation and cardiomyocyte proliferation. In valvular heart disease, CCR2+ macrophages are associated with disease progression, while M2 macrophages can reduce inflammation and promote valve repair. The proliferation of CCR2+ macrophages is associated with the deterioration of cardiac function in heart failure. Myocardial macrophages are crucial for maintaining normal cardiac electrical conduction, and the inflammatory factors they secrete are closely related to arrhythmias (figure was created with Biorender.com).
3.2.1 Atherosclerosis
Atherosclerosis (AS) is a process characterized by the formation of fibrofatty lesions in the intima of arteries, a term derived from Greek, meaning “gruel-like,” which vividly describes the lipid core of typical atherosclerotic plaques (209, 210). Over time, atherosclerotic plaques become more fibrotic and accumulate calcium (209). Plaques can trigger thrombosis and cause tissue ischemia by obstructing blood flow through rupture, superficial erosion, or protrusion into the arterial lumen (211). Inflammation plays a crucial role in AS development. Atherosclerotic plaques harbor a variety of inflammatory cells, such as monocytes/macrophages and T cells. These cells are recruited to sites of arterial wall injury, where they release a range of inflammatory factors, including cytokines (IL-1, IL-6, TNF-α, etc.), which contribute to the inflammatory response (212, 213). After engulfing oxidized low-density lipoprotein (oxLDL), monocytes/macrophages transform into foam cells, and the accumulation of foam cells is one of the key features of atherosclerotic plaque formation (214).
Macrophages are involved in the occurrence, development, and regression of AS. The recruitment and polarization of macrophages and the formation of foam cells can affect the fate of plaques (66). When the endothelium is exposed to atherogenic risk factors, endothelial cells are disturbed. These risk factors include oxidized and other modified LDL particles, pro-inflammatory cytokines, local disturbances in the hemodynamic environment, and cardiovascular-related risk factors (215, 216). Activated endothelial cells express adhesion molecules and chemokines, which promote the rolling, adhesion of circulating monocytes to the endothelium, and the migration of bound monocytes into the arterial wall (217).
Histological analyses of human plaques have revealed varying numbers and degrees of macrophages. M1 macrophages are located primarily in lipid-rich areas away from M2 macrophages; these macrophages change their phenotypic expression over time, depending on their location and microenvironment, all of which regulate plaque progression and composition (66). Macrophages take up oxidized lipid particles and transform into cholesterol-filled foam cells, which is an early event in plaque formation. The formation of foam cells is associated with multiple transcription factors that play key roles in macrophage polarization and the development of atherosclerosis (218). The dynamic equilibrium of macrophages within atherosclerotic plaques is a complex process involving multiple aspects, including recruitment, retention, polarization, egression, and death. The interplay of these processes determines the inflammatory status and stability of the plaque (219). In the early stages of atherosclerosis, the dominant M2 macrophages help maintain the stability of the arterial wall (220). However, as the disease progresses, the proportion of M1 macrophages gradually increases, while the proportion of M2 macrophages relatively decreases. This imbalance exacerbates the inflammatory response and promotes the development of atherosclerosis (221).
3.2.2 Myocardial infarction
Myocardial infarction (MI) is a clinical condition characterized by cardiomyocyte death and tissue injury resulting from myocardial oxygen deprivation. The loss of cardiomyocytes is accompanied by alterations in cellular composition, which in turn elicits an inflammatory response and triggers remodeling of the ischemic myocardium (222). Various cells, including cardiomyocytes, endothelial cells, neutrophils, monocytes/macrophages, lymphocytes, and fibroblasts, are involved in the inflammatory response. These cells promote the infiltration of inflammatory cells and the clearance of tissue damage by releasing cytokines, chemokines, and cell adhesion molecules (180). MI is one of the leading cardiovascular diseases worldwide, and its high incidence and mortality rates pose a significant burden on society and healthcare systems (223). Immune inflammation plays a crucial role in MI and subsequent ventricular remodeling, significantly impacting the extent of tissue damage, cardiac function, and disease prognosis (224). As essential innate immune cells, macrophages play a vital role in immune responses, pathogen clearance, and tissue repair (225). After MI, there are significant changes in the subpopulation distribution, metabolic characteristics, and functional phenotypes of cardiac macrophages, which promote inflammation, dead cell debris clearance, inflammation/repair transition, tissue repair promotion, and other functions at different stages (184, 226).
The insufficient cardiac blood perfusion after MI leads to a lack of oxygen and nutrients, cell damage, and the release of DAMPs. These environmental changes trigger adaptive alterations in macrophage metabolic pathways, significantly affecting their movement, phagocytic functions, and cellular phenotypes, thereby influencing the role of macrophages in disease occurrence and development (111, 224). Transcriptome profiling has revealed differential gene enrichment in glycolytic pathways in macrophages on the first day post-MI; metabolic flux analysis has revealed that macrophage glycolysis increases on the Day 1 and Day 3 post-MI. Enhanced glycolysis in macrophages early after MI is conducive to their adaptation to hypoxic environments and activation (227). Mechanistically, macrophages post-MI recognize DAMPs released from necrotic cells through Toll-like receptors (TLRs), activating downstream signaling pathways such as the mTOR pathway and entering an activated state with increased energy demands (224, 226). Hypoxia-induced factor (HIF)-1α, a downstream molecule of the TLR–mTOR axis, increases in expression post-MI and enhances glycolysis by upregulating the expression of key enzymes in the glycolytic pathway, such as hexokinase 1, lactate dehydrogenase, and glucose transporter 1 (228). In a mouse MI model, the double knockout of HIF-1α and HIF-2α reduces macrophage glycolytic levels, leading to increased macrophage necrosis, impaired tissue fibrosis, and increased cardiac rupture in mice, suggesting that increased macrophage glycolysis post-MI is beneficial for their survival in hypoxic environments (229). Additionally, the enhancement of glycolysis in macrophages post-MI provides energy for cell movement, phagocytosis, and the secretion of inflammatory factors while reducing oxygen consumption, meeting their increased energy demands upon activation (119, 184, 226).
To date, there is a lack of research evidence on lipid synthesis metabolism in macrophages post-MI. Some studies suggest that macrophages enhance FAO when phagocytosing necrotic cells post-MI, activating NAD+-dependent signal transduction and promoting the SIRT1–Pbx1–IL-10 metabolic signaling pathway, which promotes macrophage polarization toward the M2 type and improves cardiac damage repair post-MI (230). The changes in amino acid metabolism in macrophages post-MI have also been less studied. Glutamine metabolism in macrophages increases post-MI in mice, and exogenous glutamine supplementation can improve left ventricular function on the first, third, and seventh days post-MI (231). Recent studies have found that during the subacute phase of myocardial infarction, TREM2+ macrophages are recruited to the infarcted myocardial tissue and show upregulation of anti-inflammatory genes (232). These cells, derived from myeloid cells, play an important role in myocardial infarction repair (233). After efferocytosis, TREM2+ macrophages inhibit the mitochondrial NAD+ transporter SLC25A53 via the SYK-SMAD4 signaling pathway. This affects mitochondrial NAD+ transport, interrupts the TCA cycle, and increases itaconate production. Itaconate, an anti-inflammatory metabolite, helps regulate macrophage function and cardiac repair after myocardial infarction (233).
3.2.3 Myocarditis
Myocarditis is an inflammatory heart disease that may be triggered by infections, exposure to toxic substances, and activation of the immune system. It is classified as a secondary cardiomyopathy in the 1996 World Health Organization classification (234). The most thoroughly studied to date is enterovirus infection, particularly myocarditis caused by Coxsackie virus B3 (CVB3) infection (235). The mechanisms by which this virus causes myocardial injury include both direct viral damage and immune-mediated damage. The innate immune response of the host is excessively activated (236, 237), leading to the release of a large number of inflammatory factors and the formation of a cytokine storm, which is the main mechanism causing severe myocardial injury and pump failure (238).
Research on the role of macrophages in myocarditis has focused primarily on the M1 and M2 polarization phenotypes, with a general consensus that M1 macrophages exacerbate myocarditis, whereas M2 macrophages alleviate myocardial inflammation. In viral myocarditis caused by CVB3, macrophages are enriched in cardiac tissue as early as the third day post infection (239), and they secrete high levels of IL-1β, which triggers the upregulation of NLRP3 in macrophages, potentially polarizing them to the M1 phenotype (240, 241). M1 macrophages are characterized by enhanced glycolysis and disrupted TCA cycle, leading to the accumulation of HIF-1α and the expression of pro-inflammatory factors (iNOS, IL-1β, IL-6) (242). This further exacerbates inflammation, creating a vicious cycle. The adoptive transfer of M1 macrophages significantly promotes myocarditis in mice. Transferring M2 macrophages into susceptible male mice significantly reduces myocardial inflammation by modulating local cytokines (243). Similarly, in myocarditis induced by CVB3, IL-13 deficiency increases the severity of CVB3-induced myocarditis by reducing the number of M2 macrophages but does not affect viral replication in the heart postinfection. The reduction in M2 macrophages leads to a significant increase in the levels of IL-1, IL-18, IFN-α, TGF-β, IL-4, and histamine, as well as an increase in CD4+ T cells and autoreactive antibodies (244). In the experimental autoimmune myocarditis (EAM) model, macrophages are one of the major cellular components of cardiac inflammation. Studies have shown that they are not only key players in the inflammatory response but also regulate disease progression and tissue repair through interactions with different immune cells (245). Granulocyte-macrophage colony-stimulating factor (GM-CSF) stimulates macrophages to secrete IL-6, which in turn promotes the development of Th17 cells and enhances autoimmune reactions. The IL-17 secreted by Th17 cells can further activate macrophages, forming a positive feedback loop (246). Prominin-1+/CD133+ cells (a type of hematopoietic cell with stem cell characteristics) can differentiate into macrophages with immune regulatory functions in EAM. These macrophages can secrete anti-inflammatory cytokines (such as IL-10) to suppress inflammatory responses, alleviating the severity of myocarditis (247). IL-13 alleviates the severity of myocarditis by regulating the polarization and function of monocytes/macrophages. Its absence may lead to enhanced T cell activation and altered macrophage activation states, resulting in exacerbated myocarditis, cardiac fibrosis, and heart failure (244). During the chronic phase of myocarditis, Ly6Chigh monocytes infiltrated the heart transition into Ly6Clow macrophages, while Ly6Clow monocytes continue to infiltrate the heart and differentiate into macrophages (248). These macrophages predominantly exhibit an M2 phenotype and secrete a variety of cytokines that mediate tissue repair: arginase 1, TGF-β, and IL-10 induce myofibroblasts to produce collagen, VEGF promotes angiogenesis, and MMPs along with tissue inhibitors of metalloproteinases (TIMPs) regulate the extracellular matrix network. In contrast, TNF-α and IL-1 secreted by M1 macrophages stimulate fibroblasts to secrete excessive extracellular matrix, leading to matrix deposition and maladaptive fibrosis (249).
3.2.4 Heart allograft rejection
Inflammation in heart allograft rejection is one of the important factors leading to transplant failure, including hyperacute rejection, acute rejection, and chronic rejection. Hyperacute rejection is an immune response that occurs rapidly, usually within minutes to hours after transplantation. The inflammatory manifestations include vascular congestion, thrombosis, hyaline microthrombi, and necrotizing vasculitis of small to medium-sized vessels (250). Acute rejection includes acute cellular rejection (ACR) and antibody-mediated rejection (AMR). ACR is mainly mediated by T cells and is the most common type of rejection after cardiac transplantation, usually occurring within 3 to 6 months after transplantation. The inflammatory manifestations include T cell infiltration and cytokine secretion like IL-2, TNF-α, leading to myocardial injury and necrosis (251). AMR is caused by B cells producing antibodies against donor antigens, leading to graft injury through complement activation and inflammatory cell infiltration (252). Chronic rejection, also known as cardiac allograft vasculopathy (CAV), is one of the long-term complications after cardiac transplantation. The inflammatory manifestations include intimal thickening of the coronary arteries, leading to luminal stenosis (253).
The accumulation of macrophages and rejection macrophages has long been considered a characteristic of allograft rejection, with the total number of graft-infiltrating macrophages being associated with poorer clinical outcomes (28, 254). The heart contains both CCR2+ and CCR2− macrophages, which exhibit different phenotypes under homeostatic conditions (17). After transplantation, the macrophage population in the heart includes donor-derived CCR2+ and CCR2− macrophages, as well as recipient-derived neutrophils, monocytes, monocyte-derived macrophages, and monocyte-derived dendritic cells (255–257). These cells play different roles in rejection and tolerance. CCR2+ macrophages are enriched in proinflammatory genes, and their activation represents a mechanism that drives inflammation; CCR2− macrophages promote coronary arteriogenesis and cardiomyocyte proliferation through the secretion of IL-10 and TGF-β and possess potential anti-inflammatory effects (17). Studies have found that MEK1/2 promotes the pro-inflammatory phenotype and glycolytic capacity of alloimmune infiltrating macrophages (AIMs) by regulating PKM2 expression and nuclear translocation, playing a key role in cardiac transplant rejection (258). Further research is needed to elucidate the roles of macrophage metabolism and polarization in cardiac transplant rejection, including their mechanisms in various rejection types and potential therapeutic targets.
3.2.5 Valvular heart disease
Valvular heart disease can be categorized into primary and secondary types. Primary (or degenerative) disease is caused by intrinsic dysfunction of the valve itself, whereas secondary (or functional) disease results from underlying myocardial pathology (259). Inflammatory responses in valvular heart disease involve the activation of endothelial cells and immune cells, which secrete inflammatory cytokines such as TGF-β1, TNF-α, IL-1β, and IL-2, and they promote disease progression (260).
Valvular heart disease is a leading cause of disability and diminished quality of life and is a major contributor to cardiovascular morbidity and mortality worldwide (261). In recent years, an increasing number of studies have highlighted the significant role of macrophages in the development of valvular heart disease (262). In myxomatous valve disease (MVD), there is an increase in the number of CCR2+ monocytes and CD206+ macrophages, and the accumulation of these cells in immune-active extracellular matrix remodeling areas of the valve leaflets may be associated with disease progression (263). In calcific aortic valve disease (CAVD), macrophages increase infiltration and maturation in response to inflammatory factors secreted by aortic valve cells, thereby promoting osteogenic calcification, affecting the splicing of STAT3 and reducing the expression of the STAT3β isoform, which may promote disease progression (264). Macrophages, particularly the M2 subtype, play a key role in calcific aortic stenosis, and their expression of the pattern recognition receptor Toll-like receptor 7 (TLR7) is associated with inflammation and calcification processes in the aortic valve (265). The activation of TLR7 leads to increased secretion of the immunomodulatory cytokine IL-10, which may help mitigate inflammatory responses and promote valve tissue repair (265). Few studies have explored the role of macrophage metabolism and polarization in valvular heart disease. More research should be devoted to understanding their specific mechanisms in disease progression and repair.
3.2.6 Heart failure
Heart failure (HF) is a multifactorial cardiovascular disease characterized by impaired cardiac pumping function, leading to inadequate organ perfusion and oxygenation (266). Classified by left ventricular ejection fraction (LVEF), HF is typically categorized into heart failure with reduced ejection fraction (HFrEF), heart failure with mildly reduced ejection fraction (HFmrEF), and heart failure with preserved ejection fraction (HFpEF) (266). Endothelial inflammation, activation of the innate immune system, and humoral immunity can all affect the structure and function of the heart, leading to heart failure (267). Heart failure itself can also cause sterile inflammation, with cardiomyocytes and cardiac fibroblasts releasing pro-inflammatory cytokines (such as TNF-α, IL-6, IL-1β) when subjected to mechanical stretching and increased wall tension, activating the NLRP3 inflammasome and further exacerbating inflammation (268). Acute myocardial infarction (AMI) is a major cause of heart failure (269). When AMI occurs, Ly6Chigh monocytes are the first monocytic population to arrive. They differentiate into M1 macrophages, produce proteases, and secrete MMPs that degrade dying cardiomyocytes and the extracellular matrix (ECM) (270). Hypoxia-inducible factors (HIFs) rapidly accumulate in the nuclei of macrophages in response to the hypoxic microenvironment (271). HIFs promote the transcription of the M1 macrophage gene profile and shift glycolysis to become the primary mode of metabolic energy production in M1 macrophages by driving the expression of the glycolytic gene Slc2al (272).
Clinically, in HF patients undergoing left ventricular assist device implantation, the proliferation of CCR2+ macrophages is correlated with the deterioration of left ventricular function and chamber dilation (255). Moreover, these CCR2+ macrophages enriched in the hearts of these patients also produce large amounts of IL-1β upon exposure to necrotic cardiomyocytes. IL-1β is known to predict adverse outcomes in HFrEF patients (273). In myocardial biopsies from HFpEF patients, the number of cardiac macrophages is doubled (274), and there is an increase in the gene expression of profibrotic TGF-β (275). These events seem to contribute to the activation of fibroblasts and excessive deposition of collagen (274, 275). Cardiac macrophages also secrete galectin-3, which promotes myocardial fibrosis by activating myofibroblasts (276), as well as phagocytosis-induced TGF-β expression (277). In HFpEF patients, plasma levels of galectin-3 are significantly increased, and elevated levels are associated with a worse prognosis (278).
3.2.7 Arrhythmias
Arrhythmias refer to abnormalities or perturbations in the normal activation or beating of the myocardium. The normal cardiac rhythm is initiated by the sinoatrial (SA) node and conducted through the atrioventricular (AV) node and the His-Purkinje system, resulting in systematic ventricular depolarization. There are numerous types of cardiac arrhythmias. Sinus rhythm, the normal cardiac rhythm, can be disrupted by sinoatrial node dysfunction or inappropriate sinus tachycardia. Additionally, premature atrial contractions and premature ventricular contractions may occur. The severity of arrhythmias is associated with the presence or absence of structural heart disease. Atrial fibrillation (AF) is generally considered benign; however, structural heart diseases such as coronary artery disease or left ventricular dysfunction superimposed with AF may lead to heart failure or sudden cardiac death (279). Inflammation induces atrial arrhythmias by promoting atrial fibrosis, abnormal regulation of gap junctions, and intracellular calcium regulation, causing atrial electrical and structural remodeling (280). Inflammatory markers such as CRP, hs-CRP, and IL-6 are associated with the incidence and duration of atrial fibrillation (280). Following myocardial infarction, macrophages shift toward the M1 phenotype, which impacts the electrical properties of cardiomyocytes via gap junctions and KCa3.1 channel. This leads to variability in cardiomyocyte action potential duration (APD) and contributes to the development of arrhythmias (281).
In both human and murine atrioventricular nodes, CCR2− and CCR2+ macrophages are present (282). Cardiac macrophages facilitate signal transmission between cardiomyocytes through connexin 43, thereby maintaining cardiac impulse conduction. Macrophage-produced dual-regulatory proteins are key mediators controlling the phosphorylation and translocation of connexin 43 (206, 283). In vivo, the depletion of cardiac resident macrophages or macrophage gap junction protein 43 leads to impaired cardiac conduction (282). Macrophages can promote sympathoexcitation, and proinflammatory factors produced by macrophages, such as IL-1β, IL-6, and TNF-α, can activate the sympathetic nervous system through the circulation or afferent fibers, forming a substrate for proarrhythmia and directly affecting cardiac electrophysiology (284). Recent studies have shown that in atrial fibrillation, the expansion of a subset of macrophages, particularly those positive for secreted phosphoprotein 1 (SPP1), plays a key role in the development of arrhythmias; they promote inflammation and fibrosis through interactions with local cardiac immune and stromal cells, providing a conducive environment for the occurrence and maintenance of atrial fibrillation (285). Currently, there are limited studies on the role of macrophage polarization and metabolic changes in arrhythmias, and more researches are needed to fill this gap.
3.2.8 Aortic aneurysm
Aortic aneurysm (AA) is a chronic aortic disease characterized by permanent, localized dilatation of the aorta resulting from pathological remodeling of the aortic wall. It can further progress to fatal aortic rupture, which is associated with a high mortality rate (286). Aortic rupture is not only associated with the continuous enlargement of the aneurysm diameter; multiple pathological processes also contribute to this progression, including ECM degradation, inflammation, phenotypic transformation of vascular smooth muscle cells (SMCs), oxidative stress, and neovascularization (287). Macrophages play a crucial role in all stages of aortic aneurysm development. M1-type macrophages promote the progression of AA by secreting inflammatory factors and MMPs, which facilitate extracellular matrix destruction and apoptosis of vascular smooth muscle cells (VSMCs). In contrast, M2-type macrophages are mainly involved in vascular repair by inhibiting inflammation (288, 289).
When arteries are damaged, monocytes are recruited to the injury site under the guidance of chemokines such as CCR2 and CCR1, and further differentiate into macrophages (290). Inflammation is one of the core characteristics of aortic aneurysms, and macrophages play a key regulatory role in this process (197). During the clearance of early cellular debris, M1 macrophages generate large amounts of ROS. These ROS act synergistically with ROS derived from endothelial cells, vascular smooth muscle cells, and other immune cells in the aortic wall to continuously activate macrophages, forming a vicious cycle that amplifies the inflammatory response (197, 291). Additionally, M1-type macrophages can secrete pro-inflammatory cytokines such as TNF-α and IL-1β, which further drive the inflammatory process (289, 292). In contrast to M1 macrophages, M2 macrophages secrete anti-inflammatory factors such as IL-10 and TGF-β to inhibit the production of inflammatory mediators and MMPs, and they can also clear free hemoglobin and regulate oxidative stress, which will promote vascular remodeling and repair (288, 293). This process may involve the upregulation of the transcription factor Krüppel-like factor 6 (KLF6) or the downregulation of PPARδ activity (294, 295).
3.3 Sex difference in macrophage polarization and inflammatory heart disease
Sex differences also have an impact on macrophage polarization and inflammatory heart diseases. In female mice infected with CVB3, the infiltrating macrophages in the myocardium predominantly exhibit M2 polarization, which alleviates the severity of myocarditis by modulating the local cytokine environment and promoting the differentiation of regulatory T cells (243). In atherosclerosis models, aged female mice have aortas enriched with pro-inflammatory Il1b+ M1-like macrophages, showing a stronger inflammatory phenotype, while aged male mice exhibit increased Trem2+ macrophages, including anti-inflammatory foam cells and M2-like macrophages (296). Interestingly, another study showed that in female patients, atherosclerotic plaques contain a relatively higher proportion of anti-inflammatory M2 macrophages, which promote inflammation resolution and plaque stability by phagocytosing apoptotic cells and secreting anti-inflammatory cytokines (297); in contrast, male plaques demonstrate more significant infiltration of pro-inflammatory M1 macrophages and inflammation-associated monocyte subsets, such as intermediate monocytes (298). Further research is needed to elucidate the impact of macrophages in different sex groups on inflammatory heart diseases.
3.4 New research hotspots: immunosenescence and trained immunity
3.4.1 Immunosenescence of macrophages in inflammatory heart disease
The immune system is essential for defending the body against external pathogens and maintaining homeostasis. However, as people age, the functionality of the immune system gradually declines, a process known as “immunosenescence.” This phenomenon describes the weakening of the immune system associated with aging, characterized by alterations in immune cell populations and functional impairments of both innate and adaptive immunity. As a result, immunosenescence leads to increased susceptibility to infections and various chronic inflammatory diseases (299). Moreover, the aging of the immune system is a cause of immune dysfunction, which can eliminate senescent cells and lead to acquired tissue damage (300). Inflammaging is a chronic, sterile, low-grade inflammatory state associated with aging and is an important hallmark of immunosenescence. This condition is characterized by persistent immune activation and elevated levels of pro-inflammatory cytokines in the circulation, such as TNF-α, IL-6, and IL-1β (301).
The role of immunosenescence in inflammation-related heart diseases is increasingly being investigated. During the process of inflammaging, macrophages accumulate in the arterial wall, where they take up lipids from the blood, particularly LDL, leading to the formation of foam cells. As foam cells accumulate within the arterial wall, they contribute to the formation of atherosclerotic plaques, resulting in vessel stenosis or occlusion (219). With advancing age, the production of NAD+ and the levels of NAD+-dependent enzymes, such as the SIRT family, decrease in macrophages. This reduction affects the regulation of NF-κB activity, leading to heightened inflammatory responses (302). In aged individuals, the prolonged course of wound repair is associated with prolonged inflammation, delayed neovascularization, and delayed restoration of the extracellular matrix (303). Studies have shown that in aged mice following myocardial infarction, there is impaired inflammation characterized by decreased and delayed neutrophil and macrophage infiltration, reduced cytokine and chemokine expression, and impaired phagocytosis of dead cardiomyocytes. These factors may lead to maladaptive vascular remodeling and tissue repair in the healing heart, accelerating the transition to heart failure (304).
3.4.2 Trained immunity of macrophages in inflammatory heart disease
Previous researchers believed that innate immune cells, such as macrophages, could only non-specifically eliminate pathogens through biological processes like phagocytosis. However, an increasing number of studies have shown that monocytes/macrophages may also develop memory capabilities similar to those of the adaptive immune system after exposure to pathogens (305). Myeloid cells of the innate immune system become more sensitive after activation with the same or different stimuli, resulting in a persistent inflammatory monocyte/macrophage phenotype. This phenomenon is known as “trained immunity” or “innate immune memory” (306). This persistent overactivation of the innate immune system may promote the progression of inflammatory cardiovascular diseases. Risk factors for cardiovascular diseases, such as diet, smoking, hypercholesterolemia, and diabetes, are often associated with low-grade inflammation, which can induce trained immunity and subsequently affect bone marrow hematopoietic progenitor cells through epigenetic modifications, leading to chronic metabolic disorders and vascular inflammation (307). Bekkering et al. (308) proposed that trained innate immunity is associated with the progression of atherosclerosis. Wang et al. (309) also found that trained monocytes/macrophages undergo metabolic and epigenetic changes that keep these cells in a state of chronic overactivation, thereby exacerbating atherosclerosis. Myocardial infarction reprograms bone marrow monocytes through epigenetic modifications, and transplantation of bone marrow from infarcted mice to other mice results in aggravated atherosclerosis and increased infiltration of myeloid cells into plaques (310).
4 Macrophage-based target therapy in inflammatory heart disease
As research on macrophages in heart diseases deepens, an increasing number of studies have focused on regulating macrophage polarization, inflammation, and metabolism as potential therapeutic approaches for inflammatory heart diseases (Table 4).
4.1 Treatment for atherosclerosis
In atherosclerosis, the infiltration of macrophages is closely related to plaque growth and instability. Therefore, depleting macrophages has emerged as a potential therapeutic strategy (311). Using clodronate liposomes (Clo-Lip) to deplete macrophages through inducing programmed cell death has been shown to improve systolic flow velocity in patients with atherosclerosis (312, 313). Some antihyperglycemic drugs, such as metformin, pioglitazone, and sitagliptin, have been found to induce macrophages to shift to the M2 phenotype (314, 315). These drugs exert their anti-atherosclerotic effects by influencing the phenotype of macrophages. Natural pharmaceutical compounds, like curcumin and polyphenols, can also induce macrophage polarization (316, 317). New drug delivery systems, including nanoparticles, stents, oligopeptide complexes, liposomes, and monoclonal antibodies, can selectively modify macrophages. Targeting macrophage surface markers (CD11b, CD163, CD206) in atherosclerotic plaques in vivo can achieve macrophage-targeted drug delivery, significantly enhancing drug bioavailability and specificity (318). In lipid metabolism, many microRNAs regulate cholesterol efflux by targeting ATP-binding cassette transporter (ABCA1/ABCG1). For example, miR-33 inhibits ABCA1 expression, it can reduce cholesterol efflux (319). Anti-miR-33 nano therapy can modulate the polarization of macrophages to the M2 phenotype. Targeting microRNAs in macrophages may be a promising therapeutic approach for atherosclerosis (320).
4.2 Treatment for myocardial infraction
Nucleophosmin1 (NPM1) knockout inhibits glycolysis in macrophages after myocardial infarction (MI) by suppressing mTOR signal transduction, enhances OXPHOS, transforms cardiac macrophages into a reparative phenotype, and alleviates myocardial ischemic injury and adverse ventricular remodeling (321). The application of antisense oligonucleotide drugs (ASO-NPM1) and polymerization inhibitors (NSC348884) targeting NPM1 can also significantly promote the transformation of reparative macrophages, thereby improving cardiac function and mitigating ventricular remodeling after MI (321). The protective role of TREM2+ macrophages in myocardial infarction has been discussed. Adenoviral-induced TREM2 overexpression and the exogenous administration of the itaconate derivative 4-octyl itaconate (4-OI) can enhance the anti-inflammatory function of macrophages and improve cardiac function and ventricular remodeling after MI (233). Dimethyl fumarate can promote the polarization of macrophages to the M2 phenotype by increasing mitochondrial OXPHOS levels and inhibiting the expression of HIF-1α, improving damage repair after MI (322).
4.3 Treatment for other inflammatory heart diseases
IL-6 has the capacity to regulate cardiomyocytes and macrophages and plays a crucial role in the pathogenesis of heart failure with HFpEF (323). Statins can reduce the expression levels of IL-6, contributing to the improved prognosis of patients with HFpEF (324). Inhibition of IL-6 in patients with HFpEF can effectively improve their clinical condition (325). Newly discovered FAP-targeted CAR-Ms can effectively alleviate myocardial fibrosis following myocardial ischemia-reperfusion injury, improve cardiac function, and demonstrate long-term cardioprotective effects (326). In Marfan syndrome (MFS) mice, depletion of CCR2+ macrophages can protect against the progression of myxomatous valve degeneration (327). Further investigation is required to develop more therapeutic approaches targeting macrophages.
4.4 Limitation of the treatment
The spatial structure of biomolecules (such as cytokines and chemokines) is affected in vivo by biological, physical, and chemical factors, including biological enzymes, temperature, and pH value. Additionally, these biomolecules face issues such as off-target effects and difficulty in crossing biological membrane barriers, which to a certain extent hinder the exertion of drug efficacy (328, 329). Nano delivery technologies can reduce off-target effects, help drugs cross barriers, and improve bioavailability; however, the problem of high manufacturing costs urgently needs to be addressed (197). Furthermore, it is difficult to determine the therapeutic time window for different diseases. For example, early inhibition of IL-1β secreted by macrophages in acute myocardial infarction leads to insufficient scar formation and cardiac rupture (330), whereas early inhibition of IL-1β in ischemia-reperfusion injury can reduce infarct size and improve ventricular remodeling (331). In the future, we need more research to solve these problems in practical applications.
5 Conclusions and perspectives
The occurrence, development, and prognosis of heart diseases are closely related to immune inflammation. Immune inflammation plays a significant role in the occurrence and development of various heart diseases, and anti-inflammatory therapy has become a research hotspot in the treatment of them. Identifying more anti-inflammatory treatment targets and drugs is highly important. Macrophages, as key immune cells, play a central role during inflammatory heart diseases. A more in-depth investigation into therapies targeting macrophages may bring new hope for inflammatory heart diseases.
Author contributions
AX: Validation, Visualization, Writing – review & editing. AC: Validation, Writing – original draft. JZ: Validation, Writing – original draft. JJ: Validation, Writing – original draft. XZ: Validation, Visualization, Writing – original draft, Writing – review & editing. QW: Visualization, Writing – original draft, Writing – review & editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This work was supported by the Hubei Provincial Natural Science Foundation General Project (2024AFB578, 2025AFB477), the Intramural Research Program of Liyuan hospital, Tongji Medical College, Huazhong University of Science and Technology (2023LYYYSZRP0001), the National Natural Science Foundation of China (82572044), the China Postdoctoral Science Foundation (2024T170310, 2023M741285), the Science foundation of union hospital (2024XHYN046), and the Research Grant of Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education Huazhong University of Science and Technology (2024SWBS013).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Gen AI was used in the creation of this manuscript.
Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.
Publisher's note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Benjamin EJ, Virani SS, Callaway CW, Chamberlain AM, Chang AR, Cheng S, et al. Heart Disease and stroke statistics-2018 update a report from the American Heart Association. Circulation. (2018) 137:E67–E492. doi: 10.1161/CIR.0000000000000573
2. Simoes FC, Riley PR. Immune cells in cardiac repair and regeneration. Development. (2022) 149:dev199906. doi: 10.1242/dev.199906
3. Peet C, Ivetic A, Bromage DI, Shah AM. Cardiac monocytes and macrophages after myocardial infarction. Cardiovasc Res. (2020) 116:1101–12. doi: 10.1093/cvr/cvz336
4. Barrett TJ. Macrophages in atherosclerosis regression. Arterioscler Thromb Vasc Biol. (2020) 40:20–33. doi: 10.1161/ATVBAHA.119.312802
5. Wang MR, Li CH, Liu YC, Jin YY, Yu Y, Tan XQ, et al. The effect of macrophages and their exosomes in ischemic heart disease. Front Immunol. (2024) 15:1402468. doi: 10.3389/fimmu.2024.1402468
6. Moskalik A, Niderla-Bielinska J, Ratajska A. Multiple roles of cardiac macrophages in heart homeostasis and failure. Heart Fail Rev. (2022) 27:1413–30. doi: 10.1007/s10741-021-10156-z
7. Cai SS, Zhao MY, Zhou B, Yoshii A, Bugg D, Villet O, et al. Mitochondrial dysfunction in macrophages promotes inflammation and suppresses repair after myocardial infarction. J Clin Invest. (2023) 133:e159498. doi: 10.1172/JCI159498
8. Fan AD, Lin HQ, Wang XZ, Yang K, Guo HX, Zhang HX, et al. Advances in macrophage metabolic reprogramming in myocardial ischemia-reperfusion. Cell Signal. (2024) 123:111370. doi: 10.1016/j.cellsig.2024.111370
9. Zhang CY, Shi YK, Liu CZ, Sudesh SM, Hu Z, Li PY, et al. Therapeutic strategies targeting mechanisms of macrophages in diabetic heart disease. Cardiovasc Diabetol. (2024) 23:169. doi: 10.1186/s12933-024-02300-4
10. Zhang CY, Yang M, Ericsson AC. Function of macrophages in disease: current understanding on molecular mechanisms. Front Immunol. (2021) 12:620510. doi: 10.3389/fimmu.2021.620510
11. Atri C, Guerfali FZ, Laouini D. Role of human macrophage polarization in inflammation during infectious diseases. Int J Mol Sci. (2018) 19:1801. doi: 10.3390/ijms19061801
12. Jinnouchi H, Guo L, Sakamoto A, Torii S, Sato Y, Cornelissen A, et al. Diversity of macrophage phenotypes and responses in atherosclerosis. Cell Mol Life Sci. (2020) 77:1919–32. doi: 10.1007/s00018-019-03371-3
13. Saha S, Shalova IN, Biswas SK. Metabolic regulation of macrophage phenotype and function. Immunol Rev. (2017) 280:102–11. doi: 10.1111/imr.12603
14. Geissmann F, Manz MG, Jung S, Sieweke MH, Merad M, Ley K. Development of monocytes, macrophages, and dendritic cells. Science. (2010) 327:656–61. doi: 10.1126/science.1178331
15. Zhao Y, Zou WL, Du JF, Zhao Y. The origins and homeostasis of monocytes and tissue-resident macrophages in physiological situation. J Cell Physiol. (2018) 233:6425–39. doi: 10.1002/jcp.26461
16. Jakubzick CV, Randolph GJ, Henson PM. Monocyte differentiation and antigen-presenting functions. Nat Rev Immunol. (2017) 17:349–62. doi: 10.1038/nri.2017.28
17. Kopecky BJ, Frye C, Terada Y, Balsara KR, Kreisel D, Lavine KJ. Role of donor macrophages after heart and lung transplantation. Am J Transplant. (2020) 20:1225–35. doi: 10.1111/ajt.15751
18. Bleriot C, Chakarov S, Ginhoux F. Determinants of resident tissue macrophage identity and function. Immunity. (2020) 52:957–70. doi: 10.1016/j.immuni.2020.05.014
19. Bain CC, Bravo-Blas A, Scott CL, Perdiguero EG, Geissmann F, Henri S, et al. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat Immunol. (2014) 15:929–U236. doi: 10.1038/ni.2967
20. Bain CC, Scott CL, Uronen-Hansson H, Gudjonsson S, Jansson O, Grip O, et al. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. (2013) 6:498–510. doi: 10.1038/mi.2012.89
21. Mu XJ Li YT, Fan GC. Tissue-Resident Macrophages in the control of infection and resolution of inflammation. Shock. (2021) 55:14–23. doi: 10.1097/SHK.0000000000001601
22. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol. (2011) 11:762–74. doi: 10.1038/nri3070
23. Dang WP, Tao YM, Xu XX, Zhao H, Zou LJ Li YS. The role of lung macrophages in acute respiratory distress syndrome. Inflamm Res. (2022) 71:1417–32. doi: 10.1007/s00011-022-01645-4
24. Davies LC, Jenkins SJ, Allen JE, Taylor PR. Tissue-resident macrophages. Nat Immunol. (2013) 14:986–95. doi: 10.1038/ni.2705
25. Ginhoux F, Schultze JL, Murray PJ, Ochando J, Biswas SK. New insights into the multidimensional concept of macrophage ontogeny, activation and function. Nat Immunol. (2016) 17:34–40. doi: 10.1038/ni.3324
26. Imani S, Farghadani R, Roozitalab G, Maghsoudloo M, Emadi M, Moradi A, et al. Reprogramming the breast tumor immune microenvironment: cold-to-hot transition for enhanced immunotherapy. J Exp Clin Cancer Res. (2025) 44:131. doi: 10.1186/s13046-025-03394-8
27. Sheban F, Phan TS, Xie K, Ingelfinger F, Gur C, Shapir Itai Y, et al. ZEB2 is a master switch controlling the tumor-associated macrophage program. Cancer Cell. (2025) 43:1227–41.e11. doi: 10.1016/j.ccell.2025.03.021
28. Ordikhani F, Pothula V, Sanchez-Tarjuelo R, Jordan S, Ochando J. Macrophages in organ transplantation. Front Immunol. (2020) 11:582939. doi: 10.3389/fimmu.2020.582939
29. Hutchinson JA, Riquelme P, Geissler EK, Fändrich F. Human regulatory macrophages. Methods Mol Biol. (2011) 677:181–92. doi: 10.1007/978-1-60761-869-0_13
30. Panzer SE. Macrophages in transplantation: a matter of plasticity, polarization, and diversity. Transplantation. (2022) 106:257–67. doi: 10.1097/TP.0000000000003804
31. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. Macrophage polarization: different gene signatures in M1(LPS+) vs. classically and M2(LPS-) vs. alternatively activated macrophages. Front Immunol. (2019) 10:1084. doi: 10.3389/fimmu.2019.01084
32. Müller E, Christopoulos PF, Halder S, Lunde A, Beraki K, Speth M, et al. Toll-like receptor ligands and interferon-γ synergize for induction of antitumor M1 macrophages. Front Immunol. (2017) 8:1383. doi: 10.3389/fimmu.2017.01383
33. Müller E, Speth M, Christopoulos PF, Lunde A, Avdagic A, Oynebråten I, et al. Both type I and type II interferons can activate antitumor M1 macrophages when combined with TLR stimulation. Front Immunol. (2018) 9:2520. doi: 10.3389/fimmu.2018.02520
34. Duque GA, Descoteaux A. Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol. (2014) 5:1–12. doi: 10.3389/fimmu.2014.00491
35. Chen Y, Wang J, An C, Bao S, Zhang C. The role and research progress of macrophages after heart transplantation. Heliyon. (2024) 10:e33844. doi: 10.1016/j.heliyon.2024.e33844
36. Nathan C, Ding AH. Nonresolving inflammation. Cell. (2010) 140:871–82. doi: 10.1016/j.cell.2010.02.029
37. Lackner K, Ebner S, Watschinger K, Maglione M. Multiple shades of gray-macrophages in acute allograft rejection. Int J Mol Sci. (2023) 24:8257. doi: 10.3390/ijms24098257
38. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. (2013) 496:445–55. doi: 10.1038/nature12034
39. Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. (2018) 233:6425–40. doi: 10.1002/jcp.26429
40. Yue Y, Yang X, Feng KN, Wang LX, Hou J, Mei B, et al. M2b macrophages reduce early reperfusion injury after myocardial ischemia in mice: a predominant role of inhibiting apoptosis A20. Int J Cardiol. (2017) 245:228–35. doi: 10.1016/j.ijcard.2017.07.085
41. Chen X, Wen ZK, Xu W, Xiong SD. Granulin exacerbates lupus nephritis via enhancing macrophage M2b polarization. PLoS ONE. (2013) 8:e65542. doi: 10.1371/journal.pone.0065542
42. Wang YC, Zheng Y, Qi BC, Liu YW, Cheng X, Feng JY, et al. α-Lipoic acid alleviates myocardial injury and induces M2b macrophage polarization after myocardial infarction via HMGB1/NF-kB signaling pathway. Int Immunopharmacol. (2023) 121:110435. doi: 10.1016/j.intimp.2023.110435
43. Zuo SM, Wang YX, Bao HJ, Zhang ZH, Yang NF, Jia M, et al. Lipid synthesis, triggered by PPARγ T166 dephosphorylation, sustains reparative function of macrophages during tissue repair. Nat Commun. (2024) 15:7269. doi: 10.1038/s41467-024-51736-5
44. Zhang QD, Sioud M. Tumor-associated macrophage subsets: shaping polarization and targeting. Int J Mol Sci. (2023) 24:7493. doi: 10.3390/ijms24087493
45. Ma YY, He XJ, Wang HJ, Xia YJ, Wang SL, Ye ZY, et al. Interaction of coagulation factors and tumor-associated macrophages mediates migration and invasion of gastric cancer. Cancer Sci. (2011) 102:336–42. doi: 10.1111/j.1349-7006.2010.01795.x
46. Riquelme P, Amodio G, Macedo C, Moreau A, Obermajer N, Brochhausen C, et al. DHRS9 Is a stable marker of human regulatory macrophages. Transplantation. (2017) 101:2731–8. doi: 10.1097/TP.0000000000001814
47. Li J, Li C, Zhuang Q, Peng B, Zhu Y, Ye Q, et al. The evolving roles of macrophages in organ transplantation. J Immunol Res. (2019) 2019:5763430. doi: 10.1155/2019/5763430
48. Deng ZH, Loyher PL, Lazarov T, Li L, Shen ZY, Bhinder B, et al. The nuclear factor ID3 endows macrophages with a potent anti-tumour activity. Nature. (2024) 626:864–73. doi: 10.1038/s41586-023-06950-4
49. Joshi N, Walter JM, Misharin AV. Alveolar macrophages. Cell Immunol. (2018) 330:86–90. doi: 10.1016/j.cellimm.2018.01.005
50. Prinz M, Jung S, Priller J. Microglia biology: one century of evolving concepts. Cell. (2019) 179:292–311. doi: 10.1016/j.cell.2019.08.053
51. Orihuela R, McPherson CA, Harry GJ. Microglial M1/M2 polarization and metabolic states. Br J Pharmacol. (2016) 173:649–65. doi: 10.1111/bph.13139
52. Erbel C, Tyka M, Helmes CM, Akhavanpoor M, Rupp G, Domschke G, et al. CXCL4-induced plaque macrophages can be specifically identified by co-expression of MMP7+S100A8+ in vitro and in vivo. Innate Immun. (2015) 21:255–65. doi: 10.1177/1753425914526461
53. Kadl A, Meher AK, Sharma PR, Lee MY, Doran AC, Johnstone SR, et al. Identification of a novel macrophage phenotype that develops in response to atherogenic phospholipids via Nrf2. Circ Res. (2010) 107:737–U155. doi: 10.1161/CIRCRESAHA.109.215715
54. Mushenkova NV, Nikiforov NG, Melnichenko AA, Kalmykov V, Shakhpazyan NK, Orekhova VA, et al. Functional phenotypes of intraplaque macrophages and their distinct roles in atherosclerosis development and atheroinflammation. Biomedicines. (2022) 10:452. doi: 10.3390/biomedicines10020452
55. Finn AV, Nakano M, Polavarapu R, Karmali V, Saeed O, Zhao XQ, et al. Hemoglobin directs macrophage differentiation and prevents foam cell formation in human atherosclerotic plaques. J Am Coll Cardiol. (2012) 59:166–77. doi: 10.1016/j.jacc.2011.10.852
56. Boyle JJ, Johns M, Kampfer T, Nguyen AT, Game L, Schaer DJ, et al. Activating transcription factor 1 directs mhem atheroprotective macrophages through coordinated iron handling and foam cell protection. Circ Res. (2012) 110:20–33. doi: 10.1161/CIRCRESAHA.111.247577
57. Wu JY, He SP, Song ZK, Chen SK, Lin XF, Sun HM, et al. Macrophage polarization states in atherosclerosis. Front Immunol. (2023) 14:1185587. doi: 10.3389/fimmu.2023.1185587
58. Diskin C, Pålsson-McDermott EM. Metabolic modulation in macrophage effector function. Front Immunol. (2018) 9:270. doi: 10.3389/fimmu.2018.00270
59. Sun X, Li Y, Deng Q, Hu Y, Dong J, Wang W, et al. Macrophage polarization, metabolic reprogramming, and inflammatory effects in ischemic heart disease. Front Immunol. (2022) 13:934040. doi: 10.3389/fimmu.2022.934040
60. Liu Y, Xu R, Gu H, Zhang E, Qu J, Cao W, et al. Metabolic reprogramming in macrophage responses. Biomarker Res. (2021) 9:1. doi: 10.1186/s40364-020-00251-y
61. Van den Bossche J, Baardman J, de Winther MPJ. Metabolic characterization of polarized M1 and M2 bone marrow-derived macrophages using real-time extracellular flux analysis. Jove J Vis Exp. (2015). doi: 10.3791/53424-v
62. O'Neill LAJ. A broken krebs cycle in macrophages. Immunity. (2015) 42:393–4. doi: 10.1016/j.immuni.2015.02.017
63. Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. (2013) 496:238–42. doi: 10.1038/nature11986
64. Semba H, Takeda N, Isagawa T, Sugiura Y, Honda K, Wake M, et al. HIF-1α-PDK1 axis-induced active glycolysis plays an essential role in macrophage migratory capacity. Nat Commun. (2016) 7:11635. doi: 10.1038/ncomms11635
65. O'Neill LAJ, Kishton RJ, Rathmell J, A. guide to immunometabolism for immunologists. Nat Rev Immunol. (2016) 16:553–65. doi: 10.1038/nri.2016.70
66. Xiao Q, Hou R, Xie L, Niu M, Pan X, Zhu X. Macrophage metabolic reprogramming and atherosclerotic plaque microenvironment: Fostering each other? Clin Transl Med. (2023) 13:e1257. doi: 10.1002/ctm2.1257
67. Forrester JV, Kuffova L, Delibegovic M. The role of inflammation in diabetic retinopathy. Front Immunol. (2020) 11:583687. doi: 10.3389/fimmu.2020.583687
68. Haschemi A, Kosma P, Gille L, Evans CR, Burant CF, Starkl P, et al. The sedoheptulose kinase CARKL directs macrophage polarization through control of glucose metabolism. Cell Metab. (2012) 15:813–26. doi: 10.1016/j.cmet.2012.04.023
69. Jha AK, Huang SCC, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. (2015) 42:419–30. doi: 10.1016/j.immuni.2015.02.005
70. Serbulea V, Upchurch CM, Ahern KW, Bories G, Voigt P, DeWeese DE, et al. Macrophages sensing oxidized DAMPs reprogram their metabolism to support redox homeostasis and inflammation through a TLR2-Syk-ceramide dependent mechanism. Mol Metab. (2018) 7:23–34. doi: 10.1016/j.molmet.2017.11.002
71. Yan JW, Horng T. Lipid metabolism in regulation of macrophage functions. Trends Cell Biol. (2020) 30:979–89. doi: 10.1016/j.tcb.2020.09.006
72. Batista-Gonzalez A, Vidal R, Criollo A, Carreño LJ. New insights on the role of lipid metabolism in the metabolic reprogramming of macrophages. Front Immunol. (2020) 10:2993. doi: 10.3389/fimmu.2019.02993
73. Im SS, Yousef L, Blaschitz C, Liu JZ, Edwards RA, Young SG, et al. Linking lipid metabolism to the innate immune response in macrophages through sterol regulatory element binding protein-1a. Cell Metab. (2011) 13:540–9. doi: 10.1016/j.cmet.2011.04.001
74. Maier T, Leibundgut M, Ban N. The crystal structure of a mammalian fatty acid synthase. Science. (2008) 321:1315–22. doi: 10.1126/science.1161269
75. Wei XC, Song HW, Yin L, Rizzo MG, Sidhu R, Covey DF, et al. Fatty acid synthesis configures the plasma membrane for inflammation in diabetes. Nature. (2016) 539:294–8. doi: 10.1038/nature20117
76. Van den Bossche J, O'Neill LA, Menon D. Macrophage Immunometabolism: where are we (Going)? Trends Immunol. (2017) 38:395–406. doi: 10.1016/j.it.2017.03.001
77. Tarasiuk O, Miceli M, Di Domizio A, Nicolini G. AMPK and diseases: state of the art regulation by AMPK-targeting molecules. Biology. (2022) 11:1041. doi: 10.3390/biology11071041
78. Wang F, Zhang S, Vuckovic I, Jeon R, Lerman A, Folmes CD, et al. Glycolytic stimulation is not a requirement for M2 macrophage differentiation. Cell Metab. (2018) 28:463–75.e4. doi: 10.1016/j.cmet.2018.08.012
79. Tsai ML, Tsai YG, Lin YC, Hsu YL, Chen YT, Tsai MK, et al. IL-25 induced ROS-mediated M2 macrophage polarization via AMPK-associated mitophagy. Int J Mol Sci. (2022) 23:3. doi: 10.3390/ijms23010003
80. Tsai CF, Chen GW, Chen YC, Shen CK, Lu DY, Yang LY, et al. Regulatory effects of quercetin on M1/M2 macrophage polarization and oxidative/antioxidative balance. Nutrients. (2022) 14:67. doi: 10.3390/nu14010067
81. Zhou DX, Huang C, Lin Z, Zhan SX, Kong LN, Fang CB, et al. Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal. (2014) 26:192–7. doi: 10.1016/j.cellsig.2013.11.004
82. Wang X, Gui HD, Liu CH, Huo FL, Lan WP, Zhu XY, et al. ENTR1 regulates periodontitis by modulating macrophage M1 polarization via AMPK activation. Life Sci. (2025) 369:123525. doi: 10.1016/j.lfs.2025.123525
83. Kolliniati O, Ieronymaki E, Vergadi E, Tsatsanis C. Metabolic regulation of macrophage activation. J Innate Immun. (2022) 14:51–67. doi: 10.1159/000516780
84. Rath M, Müller I, Kropf P, Closs EI, Munder M. Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Front Immunol. (2014) 5:532. doi: 10.3389/fimmu.2014.00532
85. Shan X, Hu PH Ni LN, Shen L, Zhang YA Ji ZM, et al. Serine metabolism orchestrates macrophage polarization by regulating the IGF1-p38 axis. Cell Mol Immunol. (2022) 19:1263–78. doi: 10.1038/s41423-022-00925-7
86. Mao YX, Shi D, Li G, Jiang P. Citrulline depletion by ASS1 is required for proinflammatory macrophage activation and immune responses. Mol Cell. (2022) 82:527–41.e7. doi: 10.1016/j.molcel.2021.12.006
87. Merlin J, Ivanov S, Dumont A, Sergushichev A, Gall J, Stunault M, et al. Non-canonical glutamine transamination sustains efferocytosis by coupling redox buffering to oxidative phosphorylation. Nat Metab. (2021) 3:1313–26. doi: 10.1038/s42255-021-00471-y
88. Mu QD, Chen LY, Gao XT, Shen SY, Sheng WJ, Min JX, et al. The role of iron homeostasis in remodeling immune function and regulating inflammatory disease. Sci Bull. (2021) 66:1806–16. doi: 10.1016/j.scib.2021.02.010
89. Recalcati S, Locati M, Gammella E, Invernizzi P, Cairo G. Iron levels in polarized macrophages: regulation of immunity and autoimmunity. Autoimmun Rev. (2012) 11:883–9. doi: 10.1016/j.autrev.2012.03.003
90. Recalcati S, Locati M, Marini A, Santambrogio P, Zaninotto F, De Pizzol M, et al. Differential regulation of iron homeostasis during human macrophage polarized activation. Eur J Immunol. (2010) 40:824–35. doi: 10.1002/eji.200939889
91. Kanamori Y, Tanaka M, Itoh M, Ochi K, Ito A, Hidaka I, et al. Iron-rich Kupffer cells exhibit phenotypic changes during the development of liver fibrosis in NASH. Iscience. (2021) 24:102032. doi: 10.1016/j.isci.2020.102032
92. Nairz M, Schleicher U, Schroll A, Sonnweber T, Theurl I, Ludwiczek S, et al. Nitric oxide-mediated regulation of ferroportin-1 controls macrophage iron homeostasis and immune function in infection. J Exp Med. (2013) 210:855–73. doi: 10.1084/jem.20121946
93. Wang LJ, Harrington L, Trebicka E, Shi HN, Kagan JC, Hong CC, et al. Selective modulation of TLR4-activated inflammatory responses by altered iron homeostasis in mice. J Clin Invest. (2009) 119:3322–8. doi: 10.1172/JCI39939
94. Hubler MJ, Peterson KR, Hasty AH. Iron homeostasis: a new job for macrophages in adipose tissue? Trends Endocrinol Metab. (2015) 26:101–9. doi: 10.1016/j.tem.2014.12.005
95. Li YJ, Yun K, Mu RQ. A review on the biology and properties of adipose tissue macrophages involved in adipose tissue physiological and pathophysiological processes. Lipids Health Dis. (2020) 19:164. doi: 10.1186/s12944-020-01342-3
96. Pfanner N, Warscheid B, Wiedemann N. Mitochondrial proteins: from biogenesis to functional networks. Nat Rev Mol Cell Biol. (2019) 20:267–84. doi: 10.1038/s41580-018-0092-0
97. Alalaiwe A, Chen CY, Chang ZY, Sung JT, Chuang SY, Fang JY. Psoriasiform inflammation is associated with mitochondrial fission/GDAP1L1 signaling in macrophages. Int J Mol Sci. (2021) 22:10410. doi: 10.3390/ijms221910410
98. Li ZH, Pan HT, Yang JH, Chen DJ, Wang Y, Zhang H, et al. Xuanfei Baidu formula alleviates impaired mitochondrial dynamics and activated NLRP3 inflammasome by repressing NF-κB and MAPK pathways in LPS-induced ALI and inflammation models. Phytomedicine. (2023) 108:154545. doi: 10.1016/j.phymed.2022.154545
99. Fu J, Han ZY, Wu ZB, Xia YY, Yang G, Yin YL, et al. GABA regulates IL-1b production in macrophages. Cell Rep. (2022) 41:111770. doi: 10.1016/j.celrep.2022.111770
100. Yu QJ, Mei H, Gu Q, Zeng R, Li YA, Zhang JJ, et al. OLFML3 promotes IRG1 mitochondrial localization and modulates mitochondrial function in macrophages. Int J Biol Sci. (2025) 21:2275–95. doi: 10.7150/ijbs.103859
101. Li C, Ding XY, Xiang DM, Xu J, Huang XL, Hou FF, et al. Enhanced M1 and impaired M2 macrophage polarization and reduced mitochondria! Biogenesis via inhibition of AMP kinase in chronic kidney disease. Cell Physiol Biochem. (2015) 36:358–72. doi: 10.1159/000430106
102. Rodríguez-Prados JC, Través PG, Cuenca J, Rico D, Aragonés J, Martín-Sanz P, et al. Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol. (2010) 185:605–14. doi: 10.4049/jimmunol.0901698
103. Ip WKE, Hoshi N, Shouval DS, Snapper S, Medzhitov R. Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages. Science. (2017) 356:513–9. doi: 10.1126/science.aal3535
104. Al-Shabany AJ, Moody AJ, Foey AD, Billington RA. Intracellular NAD+ levels are associated with LPS-induced TNF-α release in pro-inflammatory macrophages. Biosci Rep. (2016) 36:e00301. doi: 10.1042/BSR20150247
105. Heiden MGV, Cantley LC, Thompson CB. Understanding the warburg effect: the metabolic requirements of cell proliferation. Science. (2009) 324:1029–33. doi: 10.1126/science.1160809
106. Ippolito L, Morandi A, Giannoni E, Chiarugi P. Lactate: a metabolic driver in the tumour landscape. Trends Biochem Sci. (2019) 44:153–66. doi: 10.1016/j.tibs.2018.10.011
107. Ivashkiv LB. The hypoxia-lactate axis tempers inflammation. Nat Rev Immunol. (2020) 20:85–6. doi: 10.1038/s41577-019-0259-8
108. Kes MMG, Van den Bossche J, Griffioen AW, Huijbers EJM. Oncometabolites lactate and succinate drive pro-angiogenic macrophage response in tumors. Biochim Biophys Acta Rev Cancer. (2020) 1874:188427. doi: 10.1016/j.bbcan.2020.188427
109. Xu BJ, Liu Y, Li N, Geng Q. Lactate and lactylation in macrophage metabolic reprogramming: current progress and outstanding issues. Front Immunol. (2024) 15:1395786. doi: 10.3389/fimmu.2024.1395786
110. Zhang D, Tang ZY, Huang H, Zhou GL, Cui C, Weng YJ, et al. Metabolic regulation of gene expression by histone lactylation. Nature. (2019) 574:575–80. doi: 10.1038/s41586-019-1678-1
111. Viola A, Munari F, Sánchez-Rodríguez R, Scolaro T, Castegna A. The metabolic signature of macrophage responses. Front Immunol. (2019) 10:1462. doi: 10.3389/fimmu.2019.01462
112. Infantino V, Iacobazzi V, Menga A, Avantaggiati ML, Palmieri F, A. key role of the mitochondrial citrate carrier (SLC25A1) in TNFα- and IFNγ-triggered inflammation. Biochim Biophysi Acta Gene Regulatory Mech. (2014) 1839:1217–25. doi: 10.1016/j.bbagrm.2014.07.013
113. Infantino V, Iacobazzi V, Palmieri F, Menga A. ATP-citrate lyase is essential for macrophage inflammatory response. Biochem Biophys Res Commun. (2013) 440:105–11. doi: 10.1016/j.bbrc.2013.09.037
114. Lampropoulou V, Sergushichev A, Bambouskova M, Nair S, Vincent EE, Loginicheva E, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. (2016) 24:158–66. doi: 10.1016/j.cmet.2016.06.004
115. Naujoks J, Tabeling C, Dill BD, Hoffmann C, Brown AS, Kunze M, et al. IFNs modify the proteome of containing vacuoles and restrict infection via IRG1-derived itaconic acid. PLoS Pathog. (2016) 12:1005408. doi: 10.1371/journal.ppat.1005408
116. Puchalska P, Huang XJ, Martin SE, Han XL, Patti GJ, Crawford PA. Isotope tracing untargeted metabolomics reveals macrophage polarization-state-specific metabolic coordination across intracellular compartments. Iscience. (2018) 9:298–313. doi: 10.1016/j.isci.2018.10.029
117. Benmoussa K, Garaude J, Acín-Pérez R. How mitochondrial metabolism contributes to macrophage phenotype and functions. J Mol Biol. (2018) 430:3906–21. doi: 10.1016/j.jmb.2018.07.003
118. Mills EL, Kelly B, Logan A, Costa ASH, Varma M, Bryant CE, et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell. (2016) 167:457–70.e13. doi: 10.1016/j.cell.2016.08.064
119. Kelly B, O'Neill LAJ. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. (2015) 25:771–84. doi: 10.1038/cr.2015.68
120. Wu KK. Extracellular succinate: a physiological messenger and a pathological trigger. Int J Mol Sci. (2023) 24:11165. doi: 10.3390/ijms241311165
121. Tan HY, Wang N, Li S, Hong M, Wang XB, Feng YB. The reactive oxygen species in macrophage polarization: reflecting its dual role in progression and treatment of human diseases. Oxid Med Cell Longev. (2016) 2016:2795090. doi: 10.1155/2016/2795090
122. Förstermann U, Xia N, Li HG. Roles of vascular oxidative stress and nitric oxide in the pathogenesis of atherosclerosis. Circ Res. (2017) 120:713–35. doi: 10.1161/CIRCRESAHA.116.309326
123. Aarup A, Pedersen TX, Junker N, Christoffersen C, Bartels ED, Madsen M, et al. Hypoxia-inducible factor-1 expression in macrophages promotes development of atherosclerosis. Arterioscler Thromb Vasc Biol. (2016) 36:1782–90. doi: 10.1161/ATVBAHA.116.307830
124. Almeida M, Porter RM. Sirtuins and FoxOs in osteoporosis and osteoarthritis. Bone. (2019) 121:284–92. doi: 10.1016/j.bone.2019.01.018
125. Xu CQ, Li J, Liang ZQ, Zhong YL, Zhang ZH, Hu XQ, et al. Sirtuins in macrophage immune metabolism: a novel target for cardiovascular disorders. Int J Biol Macromol. (2024) 256:128270. doi: 10.1016/j.ijbiomac.2023.128270
126. Hardeland R. Aging, melatonin, and the pro- and anti-inflammatory networks. Int J Mol Sci. (2019) 20:1223. doi: 10.3390/ijms20051223
127. Wan XX, Chowdhury IH, Jie ZL, Choudhuri S, Garg NJ. Origin of monocytes/macrophages contributing to chronic inflammation in chagas disease: SIRT1 inhibition of FAK-NFκB-dependent proliferation and proinflammatory activation of macrophages. Cells. (2020) 9:80. doi: 10.3390/cells9010080
128. Wang XF, Buechler NL, Martin A, Wells J, Yoza B, McCall CE, et al. Sirtuin-2 Regulates sepsis inflammation in ob/ob mice. PLoS ONE. (2016) 11:162560. doi: 10.1371/journal.pone.0162560
129. Zhang BC, Ma YF, Xiang CH. SIRT2 decreases atherosclerotic plaque formation in low-density lipoprotein receptor-deficient mice by modulating macrophage polarization. Biomed Pharmacother. (2018) 97:1238–42. doi: 10.1016/j.biopha.2017.11.061
130. Zhou B, Yang Y, Li CP. SIRT1 inhibits hepatocellular carcinoma metastasis by promoting M1 macrophage polarization via NF-κB pathway. Onco Targets Ther. (2019) 12:2519–29. doi: 10.2147/OTT.S195234
131. Dai SH, Wei JL, Zhang HC, Luo P, Yang YF, Jiang XF, et al. Intermittent fasting reduces neuroinflammation in intracerebral hemorrhage through the Sirt3/Nrf2/HO-1 pathway. J Neuroinflammation. (2022) 19:122. doi: 10.1186/s12974-022-02474-2
132. Zhou W, Hu GL, He JL, Wang TS, Zuo Y, Cao Y, et al. SENP1-Sirt3 signaling promotes α-ketoglutarate production during M2 macrophage polarization. Cell Rep. (2022) 39:110660. doi: 10.1016/j.celrep.2022.110660
133. Li Z, Li H, Zhao ZB, Zhu W, Feng PP, Zhu XW, et al. SIRT4 silencing in tumor-associated macrophages promotes HCC development via PPARδ signalling-mediated alternative activation of macrophages. J Exp Clin Cancer Res. 2019 38. doi: 10.1186/s13046-019-1456-9
134. Tomaselli D, Steegborn C, Mai AT, Rotili D. Sirt4: A Multifaceted Enzyme at the Crossroads of Mitochondrial Metabolism and Cancer. Front Oncol. 2020 10. doi: 10.3389/fonc.2020.00474
135. Polletta L, Vernucci E, Carnevale I, Arcangeli T, Rotili D, Palmerio S, et al. SIRT5 regulation of ammonia-induced autophagy and mitophagy. Autophagy. (2015) 11:253–70. doi: 10.1080/15548627.2015.1009778
136. Qin KW, Han CF, Zhang H, Li TL Li N, Cao XT. 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: 10.1016/j.jaut.2017.04.006
137. Yoshizawa T, Sato Y, Sobuz SU, Mizumoto T, Tsuyama T, Karim MF, et al. SIRT7 suppresses energy expenditure and thermogenesis by regulating brown adipose tissue functions in mice. Nat Commun. (2022) 13:7439. doi: 10.1038/s41467-022-35219-z
138. Ji LQ, Chen YF, Wang HQ, Zhang W, He LX, Wu JM, et al. Overexpression of Sirt6 promotes M2 macrophage transformation, alleviating renal injury in diabetic nephropathy. Int J Oncol. (2019) 55:103–15. doi: 10.3892/ijo.2019.4800
139. Mouchiroud L, Houtkooper RH, Moullan N, Katsyuba E, Ryu D, Cantó C, et al. The NAD/Sirtuin pathway modulates longevity through activation of mitochondrial UPR and FOXO signaling. Cell. (2013) 154:430–41. doi: 10.1016/j.cell.2013.06.016
140. Wang H, Sun YA, Pi CC, Yu X, Gao XY, Zhang C, et al. Nicotinamide mononucleotide supplementation improves mitochondrial dysfunction and rescues cellular senescence by NAD/Sirt3 pathway in mesenchymal stem cells. Int J Mol Sci. (2022) 23:14739. doi: 10.3390/ijms232314739
141. Hershberger KA, Martin AS, Hirschey MD. Role of NAD+ and mitochondrial sirtuins in cardiac and renal diseases. Nat Rev Nephrol. (2017) 13:213–25. doi: 10.1038/nrneph.2017.5
142. Liu Q, Zhang LQ, Chen Z, He YH, Huang YH, Qiu C, et al. Metabolic profiling of cochlear organoids identifies α-ketoglutarate and NAD as limiting factors for hair cell reprogramming. Adv Sci. (2024) 11:32. doi: 10.1002/advs.202308032
143. Artyomov MN, Van den Bossche J. Immunometabolism in the single-cell era. Cell Metab. (2020) 32:710–25. doi: 10.1016/j.cmet.2020.09.013
144. Teng YJ, Xu LC, Li WJ, Liu PY, Tian LL, Liu M. Targeting reactive oxygen species and fat acid oxidation for the modulation of tumor-associated macrophages: a narrative review. Front Immunol. (2023) 14:122443. doi: 10.3389/fimmu.2023.1224443
145. Lupfer C, Anand PK. Integrating inflammasome signaling in sexually transmitted infections. Trends Immunol. (2016) 37:703–14. doi: 10.1016/j.it.2016.08.004
146. Anand PK. Lipids, inflammasomes, metabolism, and disease. Immunol Rev. (2020) 297:108–22. doi: 10.1111/imr.12891
147. Olona A, Leishman S, Anand PK. The NLRP3 inflammasome: regulation by metabolic signals. Trends Immunol. (2022) 43:978–89. doi: 10.1016/j.it.2022.10.003
148. Xie M, Yu Y, Kang R, Zhu S, Yang LC, Zeng L, et al. PKM2-dependent glycolysis promotes NLRP3 and AIM2 inflammasome activation. Nat Commun. (2016) 7:13280. doi: 10.1038/ncomms13280
149. Wolf AJ, Reyes CN, Liang WB, Becker C, Shimada K, Wheeler ML, et al. Hexokinase is an innate immune receptor for the detection of bacterial peptidoglycan. Cell. (2016) 166:624–36. doi: 10.1016/j.cell.2016.05.076
150. Sanman LE, Qian Y, Eisle NA, Ng TM, Van der Linden WA, Monack DM, et al. Disruption of glycolytic flux is a signal for inflammasome signaling and pyroptotic cell death. Elife. (2016) 5:e13663. doi: 10.7554/eLife.13663
151. Martínez-Micaelo N, González-Abuin N, Pinent M, Ardévol A, Blay M. Dietary fatty acid composition is sensed by the NLRP3 inflammasome: omega-3 fatty acid (DHA) prevents NLRP3 activation in human macrophages. Food Funct. (2016) 7:3480–7. doi: 10.1039/C6FO00477F
152. Shen LL, Yang Y, Ou TT, Key CCC, Tong SH, Sequeira RC, et al. Dietary PUFAs attenuate NLRP3 inflammasome activation via enhancing macrophage autophagy. J Lipid Res. (2017) 58:1808–21. doi: 10.1194/jlr.M075879
153. Hooftman A, Angiari S, Hester S, Corcoran SE, Runtsch MC, Ling C, et al. The immunomodulatory metabolite itaconate modifies NLRP3 and inhibits inflammasome activation. Cell Metab. (2020) 32:468–78.e7. doi: 10.1016/j.cmet.2020.07.016
154. Bambouskova M, Potuckova L, Paulenda T, Kerndl M, Mogilenko DA, Lizotte K, et al. Itaconate confers tolerance to late NLRP3 inflammasome activation. Cell Rep. (2021) 34:108756. doi: 10.1016/j.celrep.2021.108756
155. Huang YM, Yong P, Dickey D, Vora SM, Wu H, Bernlohr DA. Inflammasome activation and pyroptosis via a lipid-regulated SIRT1-p53-ASC axis in macrophages from male mice and humans. Endocrinology. (2022) 163:bqac014. doi: 10.1210/endocr/bqac014
156. Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, Li S, Wang BW, Nemmara VV, et al. Succination inactivates gasdermin D and blocks pyroptosis. Science. (2020) 369:1633–7. doi: 10.1126/science.abb9818
157. Wen HT, Gris D, Lei Y, Jha S, Zhang L, Huang MTH, et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat Immunol. (2011) 12:408–U61. doi: 10.1038/ni.2022
158. L'homme L, Esser N, Riva L, Scheen A, Paquot N, Piette J, et al. Unsaturated fatty acids prevent activation of NLRP3 inflammasome in human monocytes/macrophages. J Lipid Res. (2013) 54:2998–3008. doi: 10.1194/jlr.M037861
159. Finucane OM, Lyons CL, Murphy AM, Reynolds CM, Klinger R, Healy NP, et al. Monounsaturated fatty acid-enriched high-fat diets impede adipose NLRP3 inflammasome-mediated IL-1β secretion and insulin resistance despite obesity. Diabetes. (2015) 64:2116–28. doi: 10.2337/db14-1098
160. Suzuki T, Yamashita S, Hattori K, Matsuda N, Hattori Y. Impact of a long-term high-glucose environment on pro-inflammatory responses in macrophages stimulated with lipopolysaccharide. Naunyn Schmiedebergs Arch Pharmacol. (2021) 394:2129–39. doi: 10.1007/s00210-021-02137-8
161. Edgar L, Akbar N, Braithwaite AT, Krausgruber T, Gallart-Ayala H, Bailey J, et al. Hyperglycemia induces trained immunity in macrophages and their precursors and promotes atherosclerosis. Circulation. (2021) 144:961–82. doi: 10.1161/CIRCULATIONAHA.120.046464
162. Jansen HJ, Stienstra R, van Diepen JA, Hijmans A, van der Laak JA, Vervoort GM, et al. Start of insulin therapy in patients with type 2 diabetes mellitus promotes the influx of macrophages into subcutaneous adipose tissue. Diabetologia. (2013) 56:2573–81. doi: 10.1007/s00125-013-3018-6
163. Puschel GP, Klauder J, Henkel J. Macrophages, low-grade inflammation, insulin resistance and hyperinsulinemia: a mutual ambiguous relationship in the development of metabolic diseases. J Clin Med. (2022) 11:4358. doi: 10.3390/jcm11154358
164. Costa Rosa LF, Safi DA, Cury Y, Curi R. The effect of insulin on macrophage metabolism and function. Cell Biochem Funct. (1996) 14:33–42. doi: 10.1002/cbf.637
165. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. (2017) 25:262–84. doi: 10.1016/j.cmet.2016.12.022
166. Sato A, Nemoto H, Yabuki A, Sato G, Ogawa Y, Ohira M. The role of ketone bodies in oxidized LDL-induced cell proliferation and lipid accumulation of macrophages. Int J Exp Pathol. (2025) 106:e70002. doi: 10.1111/iep.70002
167. Bai YP, Xing YJ, Ma T, Li K, Zhang T, Wang DG, et al. beta-Hydroxybutyrate suppresses M1 macrophage polarization through beta-hydroxybutyrylation of the STAT1 protein. Cell Death Dis. (2024) 15:874. doi: 10.1038/s41419-024-07268-3
168. Huang C, Wang J, Liu H, Huang R, Yan X, Song M, et al. Ketone body beta-hydroxybutyrate ameliorates colitis by promoting M2 macrophage polarization through the STAT6-dependent signaling pathway. BMC Med. (2022) 20:148. doi: 10.1186/s12916-022-02352-x
169. Lin J, Huang Z, Liu J, Huang Z, Liu Y, Liu Q, et al. Neuroprotective effect of ketone metabolism on inhibiting inflammatory response by regulating macrophage polarization after acute cervical spinal cord injury in rats. Front Neurosci. (2020) 14:583611. doi: 10.3389/fnins.2020.583611
170. Zhong WW Di CX, Lv JJ, Zhang YJ, Lin XL, Yuan YF, et al. Heme oxygenase-1 inhibits basophil maturation and activation but promotes its apoptosis in T helper type 2-mediated allergic airway inflammation. Immunology. (2016) 147:321–37. doi: 10.1111/imm.12564
171. Cosgrove S, Chotirmall SH, Greene CM, McElvaney NG. Pulmonary proteases in the cystic fibrosis lung induce interleukin 8 expression from bronchial epithelial cells via a heme/meprin/epidermal growth factor receptor/toll-like receptor pathway. J Biol Chem. (2011) 286:7692–704. doi: 10.1074/jbc.M110.183863
172. Erdei J, Tóth A, Balogh E, Nyakundi BB, Bányai E, Ryffel B, et al. Induction of NLRP3 inflammasome activation by heme in human endothelial cells. Oxid Med Cell Longev. (2018) 2018:4310816. doi: 10.1155/2018/4310816
173. Panico C, Felicetta A, Kunderfranco P, Cremonesi M, Salvarani N, Carullo P, et al. Single-Cell RNA sequencing reveals metabolic stress-dependent activation of cardiac macrophages in a model of dyslipidemia-induced diastolic dysfunction. Circulation. (2024) 150:1517–32. doi: 10.1161/CIRCULATIONAHA.122.062984
174. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. (2011) 145:341–55. doi: 10.1016/j.cell.2011.04.005
175. Lee MK, Moore XL, Fu Y, Al-Sharea A, Dragoljevic D, Fernandez-Rojo MA, et al. High-density lipoprotein inhibits human M1 macrophage polarization through redistribution of caveolin-1. Br J Pharmacol. (2016) 173:741–51. doi: 10.1111/bph.13319
176. Jia W, Li YT, Cheung KCP, Zheng XJ. Bile acid signaling in the regulation of whole body metabolic and immunological homeostasis. Sci China Life Sci. (2024) 67:865–78. doi: 10.1007/s11427-023-2353-0
177. Ji LL, Zhao XB, Zhang B, Kang L, Song WX, Zhao BH, et al. Slc6a8-mediated creatine uptake and accumulation reprogram macrophage polarization via regulating cytokine responses. Immunity. (2019) 51:272–84.e7. doi: 10.1016/j.immuni.2019.06.007
178. Martínez-Reyes CP, Manjarrez-Reyna AN, Méndez-García LA, Aguayo-Guerrero JA, Aguirre-Sierra B, Villalobos-Molina R, et al. Uric acid has direct proinflammatory effects on human macrophages by increasing proinflammatory mediators and bacterial phagocytosis probably via URAT1. Biomolecules. (2020) 10:576. doi: 10.3390/biom10040576
179. Lafuse WP, Wozniak DJ, Rajaram MVS. Role of cardiac macrophages on cardiac inflammation, fibrosis and tissue repair. Cells. (2020) 10:51. doi: 10.3390/cells10010051
180. Prabhu SD, Frangogiannis NG. The biological basis for cardiac repair after myocardial infarction: from inflammation to fibrosis. Circ Res. (2016) 119:91–112. doi: 10.1161/CIRCRESAHA.116.303577
181. Hishikari K, Suzuki J, Ogawa M, Isobe K, Takahashi T, Onishi M, et al. Pharmacological activation of the prostaglandin E2 receptor EP4 improves cardiac function after myocardial ischaemia/reperfusion injury. Cardiovasc Res. (2009) 81:123–32. doi: 10.1093/cvr/cvn254
182. Marchetti C, Toldo S, Chojnacki J, Mezzaroma E, Liu K, Salloum FN, et al. Pharmacologic inhibition of the NLRP3 inflammasome preserves cardiac function after ischemic and nonischemic injury in the mouse. J Cardiovasc Pharmacol. (2015) 66:1–8. doi: 10.1097/FJC.0000000000000247
183. Zhang L, Jiang YH, Fan C, Zhang Q, Jiang YH Li Y, et al. MCC950 attenuates doxorubicin-induced myocardial injury in vivo and in vitro by inhibiting NLRP3-mediated pyroptosis. Biomed Pharmacother. (2021) 143:112133. doi: 10.1016/j.biopha.2021.112133
184. Yap J, Irei J, Lozano-Gerona J, Vanapruks S, Bishop T, Boisvert WA. Macrophages in cardiac remodelling after myocardial infarction. Nat Rev Cardiol. (2023) 20:373–85. doi: 10.1038/s41569-022-00823-5
185. Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. (2014) 40:91–104. doi: 10.1016/j.immuni.2013.11.019
186. Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci USA. (2014) 111:16029–34. doi: 10.1073/pnas.1406508111
187. Leid J, Carrelha J, Boukarabila H, Epelman S, Jacobsen SEW, Lavine KJ. Primitive embryonic macrophages are required for coronary development and maturation. Circ Res. (2016) 118:1498–U127. doi: 10.1161/CIRCRESAHA.115.308270
188. Howangyin KY, Zlatanova I, Pinto C, Ngkelo A, Cochain C, Rouanet M, et al. Myeloid- epithelial-reproductive receptor tyrosine kinase and milk fat globule epidermal growth factor 8 coordinately improve remodeling after myocardial infarction via local delivery of vascular endothelial growth factor. Circulation. (2016) 133:826–39. doi: 10.1161/CIRCULATIONAHA.115.020857
189. Penberthy KK, Ravichandran KS. Apoptotic cell recognition receptors and scavenger receptors. Immunol Rev. (2016) 269:44–59. doi: 10.1111/imr.12376
190. Saljoughian N, Wu Q, Kumar N, Ganesan LP, Lafuse WP, Rajaram M. Role of cardiac macrophages in controlling the infection and homeostasis of steady state heart function in aging. J Immunol. (2020) 204:75.1-.1. doi: 10.4049/jimmunol.204.Supp.75.1
191. Nicolás-Avila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. (2020) 183:94–109.e23. doi: 10.1016/j.cell.2020.08.031
192. Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci Immunol. (2022) 7:eabf7777. doi: 10.1126/sciimmunol.abf7777
193. Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. (2019) 20:29–39. doi: 10.1038/s41590-018-0272-2
194. Rizzo G, Gropper J, Piollet M, Vafadarnejad E, Rizakou A, Bandi SR, et al. Dynamics of monocyte-derived macrophage diversity in experimental myocardial infarction. Cardiovasc Res. (2023) 119:772–85. doi: 10.1093/cvr/cvac113
195. King KR, Aguirre AD, Ye YX, Sun Y, Roh JD, Ng RP Jr., et al. IRF3 and type I interferons fuel a fatal response to myocardial infarction. Nat Med. (2017) 23:1481–7. doi: 10.1038/nm.4428
196. Jung SH, Hwang BH, Shin S, Park EH, Park SH, Kim CW, et al. Spatiotemporal dynamics of macrophage heterogeneity and a potential function of Trem2(hi) macrophages in infarcted hearts. Nat Commun. (2022) 13:4580. doi: 10.1038/s41467-022-32284-2
197. Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, et al. Macrophages in cardiovascular diseases: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. (2024) 9:130. doi: 10.1038/s41392-024-01840-1
198. Xu Y, Jiang K, Su F, Deng R, Cheng Z, Wang D, et al. A transient wave of Bhlhe41(+) resident macrophages enables remodeling of the developing infarcted myocardium. Cell Rep. (2023) 42:113174. doi: 10.1016/j.celrep.2023.113174
199. Jian Y, Zhou X, Shan W, Chen C, Ge W, Cui J, et al. Crosstalk between macrophages and cardiac cells after myocardial infarction. Cell Commun Signal. (2023) 21:109. doi: 10.1186/s12964-023-01105-4
200. Liu S, Chen J, Shi J, Zhou W, Wang L, Fang W, et al. M1-like macrophage-derived exosomes suppress angiogenesis and exacerbate cardiac dysfunction in a myocardial infarction microenvironment. Basic Res Cardiol. (2020) 115:22. doi: 10.1007/s00395-020-0781-7
201. Weber KT, Sun Y, Bhattacharya SK, Ahokas RA, Gerling IC. Myofibroblast-mediated mechanisms of pathological remodelling of the heart. Nat Rev Cardiol. (2013) 10:15–26. doi: 10.1038/nrcardio.2012.158
202. Wang C, Zhang C, Liu L, Xi A, Chen B, Li Y, et al. Macrophage-derived mir-155-containing exosomes suppress fibroblast proliferation and promote fibroblast inflammation during cardiac injury. Mol Ther. (2017) 25:192–204. doi: 10.1016/j.ymthe.2016.09.001
203. Lee SJ, Lee CK, Kang S, Park I, Kim YH, Kim SK, et al. Angiopoietin-2 exacerbates cardiac hypoxia and inflammation after myocardial infarction. J Clin Invest. (2018) 128:5018–33. doi: 10.1172/JCI99659
204. Horckmans M, Ring L, Duchene J, Santovito D, Schloss MJ, Drechsler M, et al. Neutrophils orchestrate post-myocardial infarction healing by polarizing macrophages towards a reparative phenotype. Eur Heart J. (2017) 38:187–97. doi: 10.1093/eurheartj/ehw002
205. Ramos G, Hofmann U, Frantz S. Myocardial fibrosis seen through the lenses of T-cell biology. J Mol Cell Cardiol. (2016) 92:41–5. doi: 10.1016/j.yjmcc.2016.01.018
206. Hulsmans M, Clauss S, Xiao L, Aguirre AD, King KR, Hanley A, et al. Macrophages facilitate electrical conduction in the heart. Cell. (2017) 169:510–22.e20. doi: 10.1016/j.cell.2017.03.050
207. Grune J, Lewis AJM, Yamazoe M, Hulsmans M, Rohde D, Xiao L, et al. Neutrophils incite and macrophages avert electrical storm after myocardial infarction. Nat Cardiovasc Res. (2022) 1:649–64. doi: 10.1038/s44161-022-00094-w
208. Lim GB. Macrophages and neutrophils modulate arrhythmia risk after myocardial infarction. Nat Rev Cardiol. (2022) 19:573. doi: 10.1038/s41569-022-00758-x
209. Libby P, Buring JE, Badimon L, Hansson CK, Deanfield J, Bittencourt MS, et al. Atherosclerosis. Nat Rev Dis Primers. (2019) 5:56. doi: 10.1038/s41572-019-0106-z
210. Libby P. The changing landscape of atherosclerosis. Nature. (2021) 592:524–33. doi: 10.1038/s41586-021-03392-8
211. Bentzon JF, Otsuka F, Virmani R, Falk E. Mechanisms of plaque formation and rupture. Circ Res. (2014) 114:1852–66. doi: 10.1161/CIRCRESAHA.114.302721
212. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease - reply. N Engl J Med. (2005) 353:429–30. doi: 10.1056/NEJM200507283530425
213. Tedgui A, Mallat Z. Cytokines in atherosclerosis: pathogenic and regulatory pathways. Physiol Rev. (2006) 86:515–81. doi: 10.1152/physrev.00024.2005
214. Taleb S. Inflammation in atherosclerosis. Arch Cardiovasc Dis. (2016) 109:708–15. doi: 10.1016/j.acvd.2016.04.002
215. Levitan I, Volkov S, Subbaiah PV. Oxidized LDL: diversity, patterns of recognition, and pathophysiology. Antioxid Redox Signaling. (2010) 13:39–75. doi: 10.1089/ars.2009.2733
216. Miller YI, Choi SH, Wiesner P, Fang LH, Harkewicz R, Hartvigsen K, et al. Oxidation-specific epitopes are danger-associated molecular patterns recognized by pattern recognition receptors of innate immunity. Circ Res. (2011) 108:235–48. doi: 10.1161/CIRCRESAHA.110.223875
217. Munro JM. Endothelial-leukocyte adhesive interactions in inflammatory diseases. Eur Heart J. (1993) 14 Suppl K:72-7.
218. Kuznetsova T, Prange KHM, Glass CK, de Winther MPJ. Transcriptional and epigenetic regulation of macrophages in atherosclerosis. Nat Rev Cardiol. (2020) 17:216–28. doi: 10.1038/s41569-019-0265-3
219. Moore KJ, Sheedy FJ, Fisher EA. Macrophages in atherosclerosis: a dynamic balance. Nat Rev Immunol. (2013) 13:709–21. doi: 10.1038/nri3520
220. de Gaetano M, Crean D, Barry M, Belton O. M1-and M2-type macrophage responses are predictive of adverse outcomes in human atherosclerosis. Front Immunol. (2016) 7:275. doi: 10.3389/fimmu.2016.00275
221. Hou P, Fang J, Liu Z, Shi Y, Agostini M, Bernassola F, et al. Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis. (2023) 14:691. doi: 10.1038/s41419-023-06206-z
222. Frangogiannis NG. The inflammatory response in myocardial injury, repair, and remodelling. Nat Rev Cardiol. (2014) 11:255–65. doi: 10.1038/nrcardio.2014.28
223. Vaduganathan M, Mensah GA, Turco JV, Fuster V, Roth GA. The global burden of cardiovascular diseases and risk a compass for future health. J Am Coll Cardiol. (2022) 80:2361–71. doi: 10.1016/j.jacc.2022.11.005
224. Ong SB, Hernández-Reséndiz S, Crespo-Avilan GE, Mukhametshina RT, Kwek XY, Cabrera-Fuentes HA, et al. Inflammation following acute myocardial infarction: multiple players, dynamic roles, and novel therapeutic opportunities. Pharmacol Ther. (2018) 186:73–87. doi: 10.1016/j.pharmthera.2018.01.001
225. Steffens S, Nahrendorf M, Madonna R. Immune cells in cardiac homeostasis and disease: emerging insights from novel technologies. Eur Heart J. (2022) 43:1533–41. doi: 10.1093/eurheartj/ehab842
226. Mouton AJ, Aitken NM, Moak SP, do Carmo JM, da Silva AA, Omoto ACM, et al. Temporal changes in glucose metabolism reflect polarization in resident and monocyte-derived macrophages after myocardial infarction. Front Cardiovasc Med. (2023) 10:1136252. doi: 10.3389/fcvm.2023.1136252
227. Mouton AJ, DeLeon-Pennell KY, Gonzalez OJR, Flynn ER, Freeman TC, Saucerman JJ, et al. Mapping macrophage polarization over the myocardial infarction time continuum. Basic Res Cardiol. (2018) 113:26. doi: 10.1007/s00395-018-0686-x
228. Pan JY, Zhang L, Li DX, Li Y, Lu MK, Hu YL, et al. Hypoxia-inducible factor-1: Regulatory mechanisms and drug therapy in myocardial infarction. Eur J Pharmacol. (2024) 963:176277. doi: 10.1016/j.ejphar.2023.176277
229. DeBerge M, Lantz C, Dehn S, Sullivan DP, van der Laan AM, Niessen HWM, et al. Hypoxia-inducible factors individually facilitate inflammatory myeloid metabolism and inefficient cardiac. J Exp Med. (2021) 218:667. doi: 10.1084/jem.20200667
230. Zhang S, Weinberg S, DeBerge M, Gainullina A, Schipma M, Kinchen JM, et al. Efferocytosis fuels requirements of fatty acid oxidation and the electron transport chain to polarize macrophages for tissue repair. Cell Metab. (2019) 29:443–56.e5. doi: 10.1016/j.cmet.2018.12.004
231. Mouton AJ, Aitken NM, Morato JG, O'Quinn KR, do Carmo JM, da Silva AA, et al. Glutamine metabolism improves left ventricular function but not macrophage-mediated inflammation following myocardial infarction. Am J Physiol Cell Physiol. (2024) 327:C571–C86. doi: 10.1152/ajpcell.00272.2024
232. Kim SH, Lee KY, Chang KY. The protective role of TREM2 in the heterogenous population of macrophages during post-myocardial infarction inflammation. Int J Mol Sci. (2023) 24:65556. doi: 10.3390/ijms24065556
233. Gong SY, Zhai M, Shi JY Yu GY, Lei ZJ, Shi YF, et al. TREM2 macrophage promotes cardiac repair in myocardial infarction by reprogramming metabolism via SLC25A53. Cell Death Differ. (2024) 31:239–53. doi: 10.1038/s41418-023-01252-8
234. Ammirati E, Frigerio M, Adler ED, Basso C, Birnie DH, Brambatti M, et al. Management of acute myocarditis and chronic inflammatory cardiomyopathy an expert consensus document. Circ Heart Fail. (2020) 13:7405. doi: 10.1161/CIRCHEARTFAILURE.120.007405
235. Klingel K, Sauter M, Bock CT, Szalay G, Schnorr JJ, Kandolf R. Molecular pathology of inflammatory cardiomyopathy. Med Microbiol Immunol. (2004) 193:101–7. doi: 10.1007/s00430-003-0190-1
236. Hang WJ, Chen C, Seubert JM, Wang DW. Fulminant myocarditis: a comprehensive review from etiology to treatments and outcomes. Signal Transduct Targeted Ther. (2020) 5:287. doi: 10.1038/s41392-020-00360-y
237. Li HH, Zhang MZ, Zhao QY, Zhao WQ, Zhuang Y, Wang J, et al. Self-recruited neutrophils trigger over-activated innate immune response and phenotypic change of cardiomyocytes in fulminant viral myocarditis. Cell Discov. (2023) 9:103. doi: 10.1038/s41421-023-00593-5
238. Wang J, He MY, Li HH, Chen YH, Nie X, Cai YY, et al. Soluble ST2 is a sensitive and specific biomarker for fulminant myocarditis. J Am Heart Assoc. (2022) 11:24417. doi: 10.1161/JAHA.121.024417
239. Fairweather D, Rose NR. Coxsackievirus-induced myocarditis in mice: a model of autoimmune disease for studying immunotoxicity. Methods. (2007) 41:118–22. doi: 10.1016/j.ymeth.2006.07.009
240. Bao J, Sun T, Yue Y, Xiong S. Macrophage NLRP3 inflammasome activated by CVB3 capsid proteins contributes to the development of viral myocarditis. Mol Immunol. (2019) 114:41–8. doi: 10.1016/j.molimm.2019.07.012
241. Fei X, Chen SH, Li LY, Xu XB, Wang H, Ke HJ, et al. Helicobacter pylori infection promotes M1 macrophage polarization and gastric inflammation by activation of NLRP3 inflammasome via TNF/TNFR1 axis. Cell Commun Signaling. (2025) 23:6. doi: 10.1186/s12964-024-02017-7
242. Wu KKL, Xu XF, Wu MY, Li XM, Hoque M, Li GHY, et al. MDM2 induces pro-inflammatory and glycolytic responses in M1 macrophages by integrating iNOS-nitric oxide and HIF-1α pathways in mice. Nat Commun. (2024) 15:8624. doi: 10.1038/s41467-024-53006-w
243. Li K, Xu W, Guo Q, Jiang Z, Wang P, Yue Y, et al. Differential macrophage polarization in male and female BALB/c mice infected with coxsackievirus B3 defines susceptibility to viral myocarditis. Circ Res. (2009) 105:353–64. doi: 10.1161/CIRCRESAHA.109.195230
244. Cihakova D, Barin JG, Afanasyeva M, Kimura M, Fairweather D, Berg M, et al. Interleukin-13 protects against experimental autoimmune myocarditis by regulating macrophage differentiation. Am J Pathol. (2008) 172:1195–208. doi: 10.2353/ajpath.2008.070207
245. Barin JG, Rose NR, Ciháková D. Macrophage diversity in cardiac inflammation: a review. Immunobiology. (2012) 217:468–75. doi: 10.1016/j.imbio.2011.06.009
246. Sonderegger I, Iezzi G, Maier R, Schmitz N, Kurrer M, Kopf M, et al. mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J Exp Med. (2008) 205:2281–94. doi: 10.1084/jem.20071119
247. Kania G, Blyszczuk P, Valaperti A, Dieterle T, Leimenstoll B, Dirnhofer S, et al. Prominin-1+/CD133+ bone marrow-derived heart-resident cells suppress experimental autoimmune myocarditis. Cardiovasc Res. (2008) 80:236–45. doi: 10.1093/cvr/cvn190
248. Hilgendorf I, Gerhardt LMS, Tan TC, Winter C, Holderried TAW, Chousterman BG, et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. (2014) 114:1611–22. doi: 10.1161/CIRCRESAHA.114.303204
249. Duncan SE, Gao S, Sarhene M, Coffie JW, Deng LH, Bao XR, et al. Macrophage activities in myocardial infarction and heart failure. Cardiol Res Pract. (2020) 2020:4375127. doi: 10.1155/2020/4375127
250. Kobashigawa J, Mehra M, West L, Kerman R, George J, Rose M, et al. Report from a consensus conference on the sensitized patient awaiting heart transplantation. J Heart Lung Transplant. (2009) 28:213–25. doi: 10.1016/j.healun.2008.12.017
251. Farcas AO, Stoica MC, Maier IM, Maier AC, Sin AI. Heart transplant rejection: from the endomyocardial biopsy to gene expression profiling. Biomedicines. (2024) 12:1926. doi: 10.3390/biomedicines12081926
252. De Angelis D, Chinali M, Franceschini A, Esposito C, Calcagni G, Parisi F, et al. Advanced parameters of cardiac function out perform traditional echocardiographic indices in the identification of pediatric patients with acute antibody-mediated cardiac transplant rejection. J Heart Lung Transplant. (2018) 37:S394-S. doi: 10.1016/j.healun.2018.01.1015
253. Loupy A, Toquet C, Rouvier P, Beuscart T, Bories MC, Varnous S, et al. Late failing heart allografts: pathology of cardiac allograft vasculopathy and association with antibody-mediated rejection. Am J Transplant. (2016) 16:111–20. doi: 10.1111/ajt.13529
254. Zhang X, Xu H, Yu JZ, Cui JK, Chen Z, Li Y, et al. Immune regulation of the liver through the PCSK9/CD36 pathway during heart transplant rejection. Circulation. (2023) 148:336–53. doi: 10.1161/CIRCULATIONAHA.123.062788
255. Bajpai G, Schneider C, Wong N, Bredemeyer A, Hulsmans M, Nahrendorf M, et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat Med. (2018) 24:1234–45. doi: 10.1038/s41591-018-0059-x
256. Ochando J, Kwan WH, Ginhoux F, Hutchinson JA, Hashimoto D, Collin M. The mononuclear phagocyte system in organ transplantation. Am J Transplant. (2016) 16:1053–69. doi: 10.1111/ajt.13627
257. Spahn JH Li WJ, Kreisel D. Innate immune cells in transplantation. Curr Opin Organ Transplant. (2014) 19:14–9. doi: 10.1097/MOT.0000000000000041
258. Chen Z, Li Y, Niu YQ, Zhang X, Yu JZ, Cui JK, et al. MEK1/2-PKM2 pathway modulates the immunometabolic reprogramming of proinflammatory allograft-infiltrating macrophages during heart transplant rejection. Transplantation. (2024) 108:1127–41. doi: 10.1097/TP.0000000000004899
259. Praz F, Beyersdorf F, Haugaa K, Prendergast B. Valvular heart disease: from mechanisms to management. Lancet. (2024) 403:1576–89. doi: 10.1016/S0140-6736(23)02755-1
260. Ferrari S, Pesce M. The complex interplay of inflammation, metabolism, epigenetics, and sex in calcific disease of the aortic valve. Front Cardiovasc Med. (2022) 8:791646. doi: 10.3389/fcvm.2021.791646
261. Vahanian A, Beyersdorf F, Praz F, Milojevic M, Baldus S, Bauersachs J, et al. 2021 ESC/EACTS guidelines for the management of valvular heart disease. Eur Heart J. (2022) 43:561–632. doi: 10.1093/ejcts/ezac209
262. Kim AJ, Xu N, Yutzey KE. Macrophage lineages in heart valve development and disease. Cardiovasc Res. (2021) 117:663–73. doi: 10.1093/cvr/cvaa062
263. Hulin A, Anstine LJ, Kim AJ, Potter SJ, DeFalco T, Lincoln J, et al. Macrophage transitions in heart valve development and myxomatous valve disease. Arterioscler Thromb Vasc Biol. (2018) 38:636–44. doi: 10.1161/ATVBAHA.117.310667
264. Raddatz MA, Huffstater T, Bersi MR, Reinfeld B, Madden MZ, Booton SE, et al. Macrophages promote aortic valve cell calcification and alter STAT3 splicing. Arterioscler Thromb Vasc Biol. (2020) 40:E153–E65. doi: 10.1161/ATVBAHA.120.314360
265. Karadimou G, Plunde O, Pawelzik SC, Carracedo M, Eriksson P, Franco-Cereceda A, et al. TLR7 expression is associated with M2 macrophage subset in calcific aortic valve stenosis. Cells. (2020) 9:1710. doi: 10.3390/cells9071710
266. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm M, et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur Heart J. (2021) 42:3599–726. doi: 10.1002/ejhf.2333
267. Murphy SP, Kakkar R, McCarthy CP, Januzzi JL Jr. Inflammation in heart failure: JACC state-of-the-art review. J Am Coll Cardiol. (2020) 75:1324–40. doi: 10.1016/j.jacc.2020.01.014
268. Nakayama H, Otsu K. Translation of hemodynamic stress to sterile inflammation in the heart. Trends Endocrinol Metab. (2013) 24:546–53. doi: 10.1016/j.tem.2013.06.004
269. Jenca D, Melenovsky V, Stehlik J, Stanek V, Kettner J, Kautzner J, et al. Heart failure after myocardial infarction: incidence and predictors. Esc Heart Failure. (2021) 8:222–37. doi: 10.1002/ehf2.13144
270. Zhu ZQ, Wang M, Lu SH, Dai SS, Liu JH. Role of macrophage polarization in heart failure and traditional Chinese medicine treatment. Front Pharmacol. (2024) 15:1434654. doi: 10.3389/fphar.2024.1434654
271. Zeng H, Vaka VR, He XC, Booz GW, Chen JX. High-fat diet induces cardiac remodelling and dysfunction: assessment of the role played by SIRT3 loss. J Cell Mol Med. (2015) 19:1847–56. doi: 10.1111/jcmm.12556
272. Fujisaka S, Usui I, Ikutani M, Aminuddin A, Takikawa A, Tsuneyama K, et al. Adipose tissue hypoxia induces inflammatory M1 polarity of macrophages in an HIF-1α-dependent and HIF-1α-independent manner in obese mice. Diabetologia. (2013) 56:1403–12. doi: 10.1007/s00125-013-2885-1
273. Deswal A, Petersen NJ, Feldman AM, Young JB, White BG, Mann DL. Cytokines and cytokine receptors in advanced heart failure - An analysis of the cytokine database from the vesnarinone trial (VEST). Circulation. (2001) 103:2055–9. doi: 10.1161/01.CIR.103.16.2055
274. Hulsmans M, Sager HB, Roh JD, Valero-Muñoz M, Houstis NE, Iwamoto Y, et al. Cardiac macrophages promote diastolic dysfunction. J Exp Med. (2018) 215:423–40. doi: 10.1084/jem.20171274
275. Westermann D, Lindner D, Kasner M, Zietsch C, Savvatis K, Escher F, et al. Cardiac Inflammation contributes to changes in the extracellular matrix in patients with heart failure and normal ejection fraction. Circ Heart Fail. (2011) 4:44–52. doi: 10.1161/CIRCHEARTFAILURE.109.931451
276. González GE, Rhaleb NE, D'Ambrosio MA, Nakagawa P, Liao TD, Peterson EL, et al. Cardiac-deleterious role of galectin-3 in chronic angiotensin II-induced hypertension. Am J Physiol Heart Circ Physiol. (2016) 311:H1287–H96. doi: 10.1152/ajpheart.00096.2016
277. Mukaro VR, Bylund J, Hodge G, Holmes M, Jersmann H, Reynolds PN, et al. Lectins offer new perspectives in the development of macrophage-targeted therapies for COPD/emphysema. PLoS ONE. (2013) 8:e56147. doi: 10.1371/journal.pone.0056147
278. Edelmann F, Holzendorf V, Wachter R, Nolte K, Schmidt AG, Kraigher-Krainer E, et al. Galectin-3 in patients with heart failure with preserved ejection fraction: results from the Aldo-DHF trial. Eur J Heart Fail. (2015) 17:214–23. doi: 10.1002/ejhf.203
279. Fu DG. Cardiac arrhythmias: diagnosis, symptoms, and treatments. Cell Biochem Biophys. (2015) 73:291–6. doi: 10.1007/s12013-015-0626-4
280. Armbruster AL, Campbell KB, Kahanda MG, Cuculich PS. The role of inflammation in the pathogenesis and treatment of arrhythmias. Pharmacotherapy. (2022) 42:250–62. doi: 10.1002/phar.2663
281. Fei YD, Wang Q, Hou JW Li W, Cai XX, Yang YL, et al. Macrophages facilitate post myocardial infarction arrhythmias: roles of gap junction and KCa31. Theranostics. (2019) 9:6396–411. doi: 10.7150/thno.34801
282. Lavine KJ, Pinto AR, Epelman S, Kopecky BJ, Clemente-Casares X, Godwin J, et al. The macrophage in cardiac homeostasis and disease: JACC macrophage in CVD series (Part 4). J Am Coll Cardiol. (2018) 72:2213–30. doi: 10.1016/j.jacc.2018.08.2149
283. Sugita J, Fujiu K, Nakayama Y, Matsubara T, Matsuda J, Oshima T, et al. Cardiac macrophages prevent sudden death during heart stress. Nat Commun. (2021) 12:1910. doi: 10.1038/s41467-021-22178-0
284. Chen MX, Li XP, Wang SY, Yu LL, Tang JJ, Zhou SH. The role of cardiac macrophage and cytokines on ventricular arrhythmias. Front Physiol. (2020) 11:1113. doi: 10.3389/fphys.2020.01113
285. Hulsmans M, Schloss MJ, Lee IH, Bapat A, Iwamoto Y, Vinegoni C, et al. Recruited macrophages elicit atrial fibrillation. Science. (2023) 381:231–8. doi: 10.1126/science.abq3061
286. Golledge J. Abdominal aortic aneurysm: update on pathogenesis and medical treatments. Nat Rev Cardiol. (2019) 16:225–42. doi: 10.1038/s41569-018-0114-9
287. Cho MJ, Lee MR, Park JG. Aortic aneurysms: current pathogenesis and therapeutic targets. Exp Mol Med. (2023) 55:2519–30. doi: 10.1038/s12276-023-01130-w
288. Cheng Z, Zhou YZ, Wu Y, Wu QY, Liao XB, Fu XM, et al. Diverse roles of macrophage polarization in aortic aneurysm: destruction and repair. J Transl Med. (2018) 16:354. doi: 10.1186/s12967-018-1731-0
289. Yuan Z, Lu Y, Wei J, Wu J, Yang J, Cai Z. Abdominal aortic aneurysm: roles of inflammatory cells. Front Immunol. (2020) 11:609161. doi: 10.3389/fimmu.2020.609161
290. Mellak S, Ait-Oufella H, Esposito B, Loyer X, Poirier M, Tedder TF, et al. Angiotensin II mobilizes spleen monocytes to promote the development of abdominal aortic aneurysm in Apoe-/- mice. Arterioscler Thromb Vasc Biol. (2015) 35:378–88. doi: 10.1161/ATVBAHA.114.304389
291. Raffort J, Lareyre F, Clement M, Hassen-Khodja R, Chinetti G, Mallat Z. Monocytes and macrophages in abdominal aortic aneurysm. Nat Rev Cardiol. (2017) 14:457–71. doi: 10.1038/nrcardio.2017.52
292. Batra R, Suh MK, Carson JS, Dale MA, Meisinger TM, Fitzgerald M, et al. IL-1beta (Interleukin-1beta) and TNF-alpha (Tumor Necrosis Factor-alpha) impact abdominal aortic aneurysm formation by differential effects on macrophage polarization. Arterioscler Thromb Vasc Biol. (2018) 38:457–63. doi: 10.1161/ATVBAHA.117.310333
293. Gao JP, Guo W. Mechanisms of abdominal aortic aneurysm progression: a review. Vasc Med. (2022) 27:88–96. doi: 10.1177/1358863X211021170
294. Hwang JS, Kim HJ, Kim G, Kang ES, Ham SA, Yoo T, et al. PPARdelta reduces abdominal aortic aneurysm formation in angiotensin II-infused apolipoprotein E-deficient mice by regulating extracellular matrix homeostasis and inflammatory responses. Int J Cardiol. (2014) 174:43–50. doi: 10.1016/j.ijcard.2014.03.138
295. Son BK, Sawaki D, Tomida S, Fujita D, Aizawa K, Aoki H, et al. Granulocyte macrophage colony-stimulating factor is required for aortic dissection/intramural haematoma. Nat Commun. (2015) 6:6994. doi: 10.1038/ncomms7994
296. Smit V, de Mol J, Kleijn MNAB, Depuydt MAC, de Winther MPJ, Bot I, et al. Sexual dimorphism in atherosclerotic plaques of aged mice. Immun Ageing. (2024) 21:27. doi: 10.1186/s12979-024-00434-3
297. van Dam-Nolen DHK, van Egmond NCM, Koudstaal PJ, van der Lugt A, Bos D. Sex differences in carotid atherosclerosis: a systematic review and meta-analysis. Stroke. (2023) 54:315–26. doi: 10.1161/STROKEAHA.122.041046
298. Yuan XM, Ward LJ, Forssell C, Siraj N, Li W. Carotid atheroma from men has significantly higher levels of inflammation and iron metabolism enabled by macrophages. Stroke. (2018) 49:419–25. doi: 10.1161/STROKEAHA.117.018724
299. Kim NH, Sim SJ, Han HG, Yoon JH, Han YH. Immunosenescence and age-related immune cells: causes of age-related diseases. Arch Pharm Res. (2025) 48:132–49. doi: 10.1007/s12272-024-01529-7
300. Yousefzadeh MJ, Flores RR, Zhu Y, Schmiechen ZC, Brooks RW, Trussoni CE, et al. An aged immune system drives senescence and ageing of solid organs. Nature. (2021) 594:100–5. doi: 10.1038/s41586-021-03547-7
301. Fulop T, Larbi A, Dupuis G, Le Page A, Frost EH, Cohen AA, et al. Immunosenescence and inflamm-aging as two sides of the same coin: friends or foes? Front Immunol. (2018) 8:1960. doi: 10.3389/fimmu.2017.01960
302. Yarbro JR, Emmons RS, Pence BD. Macrophage immunometabolism and inflammaging: roles of mitochondrial dysfunction, cellular senescence, CD38, and NAD. Immunometabolism. (2020) 2:e200026. doi: 10.20900/immunometab20200026
303. Chen W, Frangogiannis NG. The role of inflammatory and fibrogenic pathways in heart failure associated with aging. Heart Fail Rev. (2010) 15:415–22. doi: 10.1007/s10741-010-9161-y
304. Linton PJ, Thoman ML. Immunosenescence in monocytes, macrophages, and dendritic cells: lessons learned from the lung and heart. Immunol Lett. (2014) 162:290–7. doi: 10.1016/j.imlet.2014.06.017
305. Arts RJW, Moorlag SJCFM, Novakovic B, Li Y, Wang SY, Oosting M, et al. BCG vaccination protects against experimental viral infection in humans through the induction of cytokines associated with trained immunity. Cell Host Microbe. (2018) 23:89–100.e5. doi: 10.1016/j.chom.2017.12.010
306. Netea MG, Domínguez-Andrés J, Barreiro LB, Chavakis T, Divangahi M, Fuchs E, et al. Defining trained immunity and its role in health and disease. Nat Rev Immunol. (2020) 20:375–88. doi: 10.1038/s41577-020-0285-6
307. Devesa A, Lobo-González M, Martínez-Milla J, Oliva B, García-Lunar I, Mastrangelo A, et al. Bone marrow activation in response to metabolic syndrome and early atherosclerosis. Eur Heart J. (2022) 43:1809–28. doi: 10.1093/eurheartj/ehac102
308. Bekkering S, Joosten LAB, Netea MG, Riksen NP. Trained innate immunity as a mechanistic link between sepsis and atherosclerosis. Critical Care. (2014) 18:645. doi: 10.1186/s13054-014-0645-3
309. Wang J, Liu YM, Hu J, Chen C. Trained immunity in monocyte/macrophage: novel mechanism of phytochemicals in the treatment of atherosclerotic cardiovascular disease. Front Pharmacol. (2023) 14:1109576. doi: 10.3389/fphar.2023.1109576
310. Dutta P, Courties G, Wei Y, Leuschner F, Gorbatov R, Robbins CS, et al. Myocardial infarction accelerates atherosclerosis. Nature. (2012) 487:325–9. doi: 10.1038/nature11260
311. De Meyer I, Martinet W, De Meyer GRY. Therapeutic strategies to deplete macrophages in atherosclerotic plaques. Br J Clin Pharmacol. (2012) 74:246–63. doi: 10.1111/j.1365-2125.2012.04211.x
312. Han XN Li Q, Lan X, EL-Mufti L, Ren HL, Wang J. Microglial depletion with clodronate liposomes increases proinflammatory cytokine levels, induces astrocyte activation, and damages blood vessel integrity. Mol Neurobiol. (2019) 56:6184–96. doi: 10.1007/s12035-019-1502-9
313. Shoulders H, Garner KH, Singla DK. Macrophage depletion by clodronate attenuates bone morphogenetic protein-7 induced M2 macrophage differentiation and improved systolic blood velocity in atherosclerosis. Transl Res. (2019) 203:1–14. doi: 10.1016/j.trsl.2018.07.006
314. Brenner C, Franz WM, Kühlenthal S, Kuschnerus K, Remma F, Gross L, et al. DPP-4 inhibition ameliorates atherosclerosis by priming monocytes into M2 macrophages. Int J Cardiol. (2015) 199:163–9. doi: 10.1016/j.ijcard.2015.07.044
315. Tokutome M, Matoba T, Nakano Y, Okahara A, Fujiwara M, Koga JI, et al. Peroxisome proliferator-activated receptor-gamma targeting nanomedicine promotes cardiac healing after acute myocardial infarction by skewing monocyte/macrophage polarization in preclinical animal models. Cardiovasc Res. (2019) 115:419–31. doi: 10.1093/cvr/cvy200
316. Wang DD, Yang Y, Lei YN, Tzvetkov NT, Liu XD, Yeung AWK, et al. Targeting foam cell formation in atherosclerosis: therapeutic potential of natural products. Pharmacol Rev. (2019) 71:596–670. doi: 10.1124/pr.118.017178
317. Zhao SJ, Li JK, Wang LF, Wu XX. Pomegranate peel polyphenols inhibit lipid accumulation and enhance cholesterol efflux in raw2647 macrophages. Food Funct. (2016) 7:3201–10. doi: 10.1039/C6FO00347H
318. Poznyak AV, Nikiforov NG, Starodubova AV, Popkova TV, Orekhov AN. Macrophages and foam cells: brief overview of their role, linkage, and targeting potential in atherosclerosis. Biomedicines. (2021) 9:1221. doi: 10.3390/biomedicines9091221
319. Rayner KJ, Suárez Y, Dávalos A, Parathath S, Fitzgerald ML, Tamehiro N, et al. MiR-33 contributes to the regulation of cholesterol homeostasis. Science. (2010) 328:1570–3. doi: 10.1126/science.1189862
320. Li CW, Dou Y, Chen YD, Qi YT, Li LL, Han SL, et al. Site-specific MicroRNA-33 antagonism by pH-responsive nanotherapies for treatment of atherosclerosis via regulating cholesterol efflux and adaptive immunity. Adv Funct Mater. (2020) 30:2131. doi: 10.1002/adfm.202002131
321. Zhang S, Zhang Y, Duan X, Wang B, Zhan Z. Targeting NPM1 epigenetically promotes postinfarction cardiac repair by reprogramming reparative macrophage metabolism. Circulation. (2024) 149:1982–2001. doi: 10.1161/CIRCULATIONAHA.123.065506
322. Mouton AJ, Flynn ER, Moak SP, Aitken NM, Omoto ACM Li X, et al. Dimethyl fumarate preserves left ventricular infarct integrity following myocardial infarction via modulation of cardiac macrophage and fibroblast oxidative metabolism. J Mol Cell Cardiol. (2021) 158:38–48. doi: 10.1016/j.yjmcc.2021.05.008
323. Mauer J, Chaurasia B, Goldau J, Vogt MC, Ruud J, Nguyen KD, et al. Signaling by IL-6 promotes alternative activation of macrophages to limit endotoxemia and obesity-associated resistance to insulin. Nat Immunol. (2014) 15:423–30. doi: 10.1038/ni.2865
324. Alehagen U, Benson L, Edner M, Dahlström U, Lund LH. Association between use of statins and mortality in patients with heart failure and ejection fraction of ≥50%. Circ Heart Fail. (2015) 8:862–70. doi: 10.1161/CIRCHEARTFAILURE.115.002143
325. Alogna A, Koepp KE, Sabbah M, Netto JME, Jensen MD, Kirkland JL, et al. Interleukin-6 in patients with heart failure and preserved ejection fraction. Jacc Heart Fail. (2023) 11:1549–61. doi: 10.1016/j.jchf.2023.06.031
326. Wang JW, Du H, Xie WR Bi JM, Zhang H, Liu X, et al. CAR-macrophage therapy alleviates myocardial ischemia-reperfusion injury. Circ Res. (2024) 135:1161–74. doi: 10.1161/CIRCRESAHA.124.325212
327. Kim AJ, Xu N, Umeyama K, Hulin A, Ponny SR, Vagnozzi RJ, et al. Deficiency of circulating monocytes ameliorates the progression of myxomatous valve degeneration in marfan syndrome. Circulation. (2020) 141:132–46. doi: 10.1161/CIRCULATIONAHA.119.042391
328. Machtakova M, Therien-Aubin H, Landfester K. Polymer nano-systems for the encapsulation and delivery of active biomacromolecular therapeutic agents. Chem Soc Rev. (2022) 51:128–52. doi: 10.1039/D1CS00686J
329. Zhao L, Ren TH, Wang DD. Clinical pharmacology considerations in biologics development. Acta Pharmacol Sin. (2012) 33:1339–47. doi: 10.1038/aps.2012.51
330. Hwang MW, Matsumori A, Furukawa Y, Ono K, Okada M, Iwasaki A, et al. Neutralization of interleukin-1beta in the acute phase of myocardial infarction promotes the progression of left ventricular remodeling. J Am Coll Cardiol. (2001) 38:1546–53. doi: 10.1016/S0735-1097(01)01591-1
331. Bujak M, Dobaczewski M, Chatila K, Mendoza LH Li N, Reddy A, et al. Interleukin-1 receptor type I signaling critically regulates infarct healing and cardiac remodeling. Am J Pathol. (2008) 173:57–67. doi: 10.2353/ajpath.2008.070974
332. Zhang FL, Zhang JD, Cao P, Sun ZJ, Wang W. The characteristics of regulatory macrophages and their roles in transplantation. Int Immunopharmacol. (2021) 91:107322. doi: 10.1016/j.intimp.2020.107322
333. Zheng WX, Yang LX, Jiang SM, Chen MX, Li JZ, Liu ZJ, et al. Role of Kupffer cells in tolerance induction after liver transplantation. Front Cell Dev Biol. (2023) 11:1179077. doi: 10.3389/fcell.2023.1179077
Keywords: inflammatory, heart, disease, macrophage, metabolic, reprogramming, immunometabolism, therapeutic targeting
Citation: Xiang A, Chang A, Zhou J, Jin J, Zhang X and Wang Q (2025) Macrophage metabolism in inflammatory heart disease: new insights and therapeutic implications. Front. Med. 12:1664538. doi: 10.3389/fmed.2025.1664538
Received: 12 July 2025; Accepted: 18 September 2025;
Published: 08 October 2025.
Edited by:
Dongze Qin, Albert Einstein College of Medicine, United StatesReviewed by:
Harikrishnan Venugopal, Albert Einstein College of Medicine, United StatesIzabela Tuleta, Albert Einstein College of Medicine, United States
Copyright © 2025 Xiang, Chang, Zhou, Jin, Zhang and Wang. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Qianyan Wang, MjAxNWx5MDkxMUBodXN0LmVkdS5jbg==; Xi Zhang, emhhbmd4aTc5OEBzaW5hLmNvbQ==
†These authors have contributed equally to this work