Go to JCI Insight
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
  • Clinical Research and Public Health
  • Current issue
  • Past issues
  • By specialty
    • COVID-19
    • Cardiology
    • Gastroenterology
    • Immunology
    • Metabolism
    • Nephrology
    • Neuroscience
    • Oncology
    • Pulmonology
    • Vascular biology
    • All ...
  • Videos
    • Conversations with Giants in Medicine
    • Video Abstracts
  • Reviews
    • View all reviews ...
    • Complement Biology and Therapeutics (May 2025)
    • Evolving insights into MASLD and MASH pathogenesis and treatment (Apr 2025)
    • Microbiome in Health and Disease (Feb 2025)
    • Substance Use Disorders (Oct 2024)
    • Clonal Hematopoiesis (Oct 2024)
    • Sex Differences in Medicine (Sep 2024)
    • Vascular Malformations (Apr 2024)
    • View all review series ...
  • Viewpoint
  • Collections
    • In-Press Preview
    • Clinical Research and Public Health
    • Research Letters
    • Letters to the Editor
    • Editorials
    • Commentaries
    • Editor's notes
    • Reviews
    • Viewpoints
    • 100th anniversary
    • Top read articles

  • Current issue
  • Past issues
  • Specialties
  • Reviews
  • Review series
  • Conversations with Giants in Medicine
  • Video Abstracts
  • In-Press Preview
  • Clinical Research and Public Health
  • Research Letters
  • Letters to the Editor
  • Editorials
  • Commentaries
  • Editor's notes
  • Reviews
  • Viewpoints
  • 100th anniversary
  • Top read articles
  • About
  • Editors
  • Consulting Editors
  • For authors
  • Publication ethics
  • Publication alerts by email
  • Advertising
  • Job board
  • Contact
Top
  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal
  • Top
  • Abstract
  • Introduction to the cardiac macrophage
  • Cardiac macrophages in the acute response to myocardial infarction
  • Relevance of macrophage metabolic signaling
  • Inflammatory metabolism of monocytes and macrophages after acute MI
  • Reparative cardiac macrophages
  • Macrophage metabolic switches implicated in cardiac repair
  • Future research directions and therapeutic implications
  • Acknowledgments
  • Footnotes
  • References
  • Version history
  • Article usage
  • Citations to this article

Advertisement

Review Open Access | 10.1172/JCI171953

Cardiac macrophages and emerging roles for their metabolism after myocardial infarction

Edward B. Thorp

Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, USA.

Address correspondence to: Edward B. Thorp, Feinberg School of Medicine, Northwestern University, Department of Pathology, 303 E. Chicago Avenue Ward 4-116, Chicago, Illinois 60611, USA. Phone: 312.503.4309; Email: ebthorp@northwestern.edu.

Find articles by Thorp, E. in: JCI | PubMed | Google Scholar |

Published September 15, 2023 - More info

Published in Volume 133, Issue 18 on September 15, 2023
J Clin Invest. 2023;133(18):e171953. https://doi.org/10.1172/JCI171953.
© 2023 Thorp This work is licensed under the Creative Commons Attribution 4.0 International License. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
Published September 15, 2023 - Version history
View PDF
Abstract

Interest in cardioimmunology has reached new heights as the experimental cardiology field works to tap the unrealized potential of immunotherapy for clinical care. Within this space is the cardiac macrophage, a key modulator of cardiac function in health and disease. After a myocardial infarction, myeloid macrophages both protect and harm the heart. To varying degrees, such outcomes are a function of myeloid ontogeny and heterogeneity, as well as functional cellular plasticity. Diversity is further shaped by the extracellular milieu, which fluctuates considerably after coronary occlusion. Ischemic limitation of nutrients constrains the metabolic potential of immune cells, and accumulating evidence supports a paradigm whereby macrophage metabolism is coupled to divergent inflammatory consequences, although experimental evidence for this in the heart is just emerging. Herein we examine the heterogeneous cardiac macrophage response following ischemic injury, with a focus on integrating putative contributions of immunometabolism and implications for therapeutically relevant cardiac injury versus cardiac repair.

Introduction to the cardiac macrophage

In the adult, macrophages are found in most organs (1). Defined by its “professional” phagocytic prowess (2), the macrophage’s specific cellular identity is also imprinted by its organ of residence (3). In the heart, macrophage phagocytes are interwoven between the cardiac parenchyma, and account for approximately 7% of the non-cardiomyocyte cellular population (4). Thanks to single-cell technologies and elegant genetic lineage traces, we now recognize that cardiac macrophages are composed of multiple subpopulations with distinct ontological origins. These subtypes include long-lived yolk sac–derived macrophages as well as adult monocyte-derived macrophages (5, 6). Cardiac macrophages are also broadly distinguished by expression of chemokine (C-C motif) receptor 2 (CCR2), with recruited CCR2+ cells arising recently from circulating monocytes or adult hematopoietic lineages, and resident macrophages lacking CCR2 (CCR2–). In experimental rodents, CCR2– macrophages often arise from embryonic seeding and harbor the potential for self-renewal (5). It is important to note that this is an oversimplification (7). For example, CCR2– macrophages are further divided by expression of major histocompatibility complex (MHC) class II, as MHCIIhi and MHCIIlo populations, the latter of which are characterized as T cell immunoglobulin and mucin domain containing 4 (TimD4+), lymphatic vessel endothelial hyaluronan receptor 1 (Lyve1+), and folate receptor 2 (FolR2+) (7). CCR2 is also expressed in human macrophages, including monocyte-derived macrophages (8), and most human CCR2– macrophages are predominantly characterized by high expression of HLA-DRh (9).

Once thought to primarily act as sentinels, functioning as innate responders to blood-borne pathogens or tissue injury, cardiac macrophages are now appreciated as having more diverse functions. Various roles include those associated with critical developmental, homeostatic, and disease-associated functions. For example, during heart development, embryonic macrophages participate in coronary maturation (10) and lymphatic vessel growth, which is necessary for proper patterning of coronary blood vessels (11). In the adult, macrophages also facilitate electrical conductance by regulating the membrane potential of cardiomyocytes, and deficiency of adult macrophages induces progressive atrioventricular block (12). With aging, an increase in the contribution of blood-borne monocyte-derived macrophages to the resident pool correlates with increased inflammation (13).

Cardiac macrophages in the acute response to myocardial infarction

Though clinical advances have reduced acute mortality after myocardial infarction (MI), ischemic heart disease remains a substantial cause of chronic morbidity and progression to heart failure (14). After infarction, macrophages are cardioprotective (15), yet also contribute to cardiac injury (16). This latter outcome is particularly relevant in the setting of reperfusion injury, where excessive inflammation portends poor clinical outcome (17). Also, blockade of CCR2-mediated recruitment of monocyte-derived macrophages after experimental MI has been shown to attenuate adverse cardiac remodeling (18). This type of injury is correlated with excess production of reactive oxygen species (ROS) (19). Moreover, aging-related defects in macrophage phagocytosis are implicated in poor cardiac outcomes (20). Detrimental functions of cardiac macrophages may be a consequence of an unevolved response to a disease that occurs in post-reproductive years.

Following MI, peripheral CCR2+ monocytes are recruited to the heart from the circulation and differentiate into macrophages. Monocytic accumulation follows infiltration by blood neutrophils (21), which help debride the cardiac wound but can also contribute to tissue injury. Additional signals from cardiomyocytes and myocardial B cells contribute to monocyte recruitment (22). Blood-borne macrophages serve multiple functions that protect the heart from even more devastating outcomes, such as cardiac rupture. This includes the clearance of dead parenchymal and stromal cells, which is a prerequisite for replacement fibrosis. The physical removal of actively dying cells also prevents secondary necrosis, limiting infarct expansion (23). Moreover, CCR2+ macrophages perpetuate inflammation by releasing monocyte chemoattractant proteins, and this fuels further monocyte recruitment (24). This positive-feedback loop is particularly important given that resident cardiac macrophage survival is markedly reduced soon after cardiac ischemia (6). Inflammatory macrophages also produce interleukin-1β (IL-1β), the blockade of which reduced clinical events in MI patients (25).

As introduced above, cardiac macrophages are not entirely derived from monocytes, as the resident cardiac macrophage pool is prenatally seeded and maintained by self-renewal. Resident macrophages sense injury, which also triggers macrophage proliferation (26). As introduced above, and in both humans and mice, resident macrophages are classified by lack of CCR2 expression (9). In mice, CCR2– macrophages regulate monocyte fate specification, and their depletion alters the ratio of macrophage subsets following cardiac injury (24). These tissue-resident CCR2– macrophages also counterbalance inflammation through the inhibition of monocyte recruitment (24). Both mouse and human CCR2– macrophage subsets express higher levels of tissue-resident macrophage markers such as the mannose receptor CD206, the scavenger receptor CD163, the sialic acid–binding lectin SIGLEC1, and LYVE1, as well as growth factors IGF-1 and PDGF-C, relative to their CCR2+ counterparts (9). Thus, a dichotomy emerges whereby CCR2– cardiac macrophages are involved in orchestrating inflammation resolution and cardiac repair, in contrast to their CCR2+ counterparts, which act to promote inflammation (Figure 1).

Heterogeneous cardiac macrophages and interacting cell types respond to MI.Figure 1

Heterogeneous cardiac macrophages and interacting cell types respond to MI. At steady state, resident CCR2– macrophages, recruited CCR2+ cardiac macrophages, and fibroblasts exhibit non-inflammatory activity. Within 1 week after MI, CCR2– and CCR2+ macrophages show increased macrophage functions related to repair and inflammation. Cardiac macrophages interact with various cell types, including cardiomyocytes, neutrophils (PMNs), fibroblasts, monocytes, B cells, apoptotic cells, regulatory T cells (Tregs), endothelial cells, lymphatic endothelial cells, and myofibroblasts. Related processes include macrophage phagocytic clearance of apoptotic cells (efferocytosis) and released cardiomyocyte-derived exophers. Efferocytosis in the heart may contribute to activation of myofibroblasts and resultant scar formation. Cardiomyocytes may secrete factors that activate endothelial adhesion molecules. Macrophage receptors that interact with the cardiac milieu include the chemokine receptor CCR2, the pattern recognition receptor TLR4, and the phagocytic molecule CD36. Key effector cytokines produced after MI include IL-1β (produced from the inflammasome) and TGF-β. Reparative gene activation involves the vascular endothelial growth factors Vegfa and Vegfc.

Relevance of macrophage metabolic signaling

In addition to ontogenetic factors, cell-intrinsic transcriptional and functional plasticity is a hallmark of macrophages (27). Inflammatory activation is particularly sensitive to and facilitated by triggering of macrophage Toll-like receptors (TLRs) such as TLR4 (28), the ligands of which are generated after MI (29). On the other hand, cytokines secreted from T helper 2 (Th2) lymphocytes, such as IL-4 and IL-13, polarize transcriptomes toward an antiinflammatory state (30) that includes increased expression of IL-10. In the heart, such alternatively activated macrophages appear to be critical to cardiac repair (31). Thus, macrophage polarization as a function of exposure to TLR versus IL-4 receptor ligands has been widely studied in an effort to elucidate fundamental mechanisms of functional cellular polarization. This model framework has also been exploited in vitro to investigate metabolic contributions to macrophage polarization. While this model has been tremendously insightful at the fundamental level, it is now time to prioritize a focus on in vivo mechanisms and translational potential. Our grasp of this dichotomy is particularly critical in the heart, where excessive inflammation contributes to collateral loss of non-regenerative cardiomyocytes.

Cell metabolism may conventionally be described as a balance between biosynthetic and catabolic reactions, summarized as the totality of chemical processes that are necessary to power cells via the biosynthesis of ATP. Yet a more evolved view is that metabolites also contribute to select signaling pathways that specifically regulate functional cellular plasticity. As such, it stands to reason that metabolite signaling may be sensitive to metabolite availability, which varies considerably after MI. Acute MI substantially reduces blood flow and so limits oxygen and nutrient availability within the myocardial perfusion bed that is distal to the occluded coronary artery. Access to extracellular glucose, glutamine, and lipids may also be limited, presenting a challenge to metabolically demanding macrophages.

Inflammatory metabolism of monocytes and macrophages after acute MI

Glucose fuels the mobilization of cardiac monocytes after MI. Multiple lines of evidence support a paradigm wherein acute inflammatory mobilization of myeloid cells is fueled by glycolysis. Glycolysis provides a rapid pathway to generate the necessary building blocks for cell proliferation and the energy that is needed to secrete inflammatory cytokines. CCR2+ tissue macrophages differentiate from blood-borne monocytes, both of which increase their glucose appetite during inflammation. This thirst for sugar begins early in myeloid differentiation, external to the myocardial infarct, as disruption of glucose transporters (such as Glut1) in hematopoietic stem cells impairs both intracellular glucose flux and inflammatory myelopoiesis (32). In patient-derived monocytes and macrophages, increased glucose uptake fuels the generation of mitochondrial ROS (mROS). This activates the glycolytic enzyme pyruvate kinase M2 (PKM2), which in turn signals to boost IL-6 and IL-1β production. Thus, PKM2 integrates glycolysis, oxidative stress, and inflammation (33). Glycolysis is also required for the adhesion of human CD14+CD16− monocytes to endothelial layers (34). This binding is necessary for the transmigration of blood monocytes into the heart. Many of these immunometabolic features can be recapitulated in isolated cells. For example, healthy human CD14+ monocytes activated with a TLR4 agonist increase aerobic Warburg (35) glycolysis and suppress mitochondrial oxidative phosphorylation (OXPHOS) (36). Glucose can also be sourced from glycogen stores to fuel inflammation (37). In patients after MI, peripheral monocytes are enriched for both glycolytic enzyme and inflammatory transcripts, and these patterns appear to be conserved between humans and experimental rodents (38). In experimental animals, monocytes just 1 day after MI exhibited higher basal extracellular acidification rates and elevated glycolytic reserve, consistent with a high glycolytic capacity. Additionally, oxygen consumption rates revealed decreased basal OXPHOS and spare respiratory capacity, and maximal respiration was also reduced, consistent with a higher dependence on glycolysis relative to oxidative metabolism (39). Glycolytic affinity in monocytes has been exploited clinically to track monocytes by fluorodeoxyglucose positron emission tomography (FDG-PET). FDG uptake is increased within the infarct concomitant with monocyte infiltration (40). In acute MI patients, peripheral monocyte abundance is also associated with elevated myocardial FDG uptake, larger infarcts, and worsened systolic function (41).

Inflammatory and metabolic adaptations to hypoxia. The appetite for glycolysis continues after monocytes differentiate into CCR2+ cardiac macrophages. Tracking macrophage polarity over the span of a week after experimental ligation of coronary arteries revealed a notable parallel induction of glycolysis and inflammatory cytokine gene transcripts (42). This response likely reflected the activity of recruited, monocyte-derived CCR2+ macrophages (26), which also exhibit glycolytic transcriptional signatures.

The cardiac microenvironment instructs divergent functional fates in myeloid cells during inflammation (43). Coronary obstruction reduces nutrient and oxygen (44) availability, and experimental permanent coronary occlusion leads to substantial loss of resident cardiac macrophages (6). This depleted state contrasts with that which occurs after coronary reperfusion, which salvages resident macrophages. Given that the majority of patients undergo a form of clinical reperfusion, this point stresses the importance of inducing reperfusion in studies that examine therapeutic relevance. In those cardiac macrophages that survive acute ischemia, or alternatively are recruited from the circulation, hypoxia triggers the stabilization of hypoxia-inducible transcription factors (HIFs) (45). HIF-1α promotes metabolic adaptations in a cell-autonomous manner (46), particularly by increasing mRNAs of glucose transporters and glycolytic enzymes (45). HIF-1α also polarizes toward glycolysis by suppressing metabolism through the mitochondrial tricarboxylic acid (TCA) cycle. Glycolysis is favored because HIF-1α directly transactivates the gene encoding pyruvate dehydrogenase kinase 1 (PDK1). PDK1 in turn inactivates the TCA cycle enzyme pyruvate dehydrogenase, which converts pyruvate to acetyl-CoA (47). Increased glycolytic intermediates also feed the pentose phosphate pathway (PPP), which generates NADPH-dependent ROS. Importantly, reperfusion injury has been associated with activation of NADPH oxidase in macrophages (19).

HIF-1α–dependent metabolism is further coupled to induction of inflammatory cytokines, notably IL-1β (48). Though HIFs contribute to bactericidal capacity in macrophages (49), tempering of HIF-1α activity in myeloid (50) and hematopoietic stem cells (51) has been found to attenuate inflammation and improve cardiac function after experimental MI. One immunometabolic explanation for this maladaptation is provided by shedding of cell-surface phagocytic receptors through the action of hypoxia-inducible proteases. For example, inhibiting HIF-1α and glycolysis reduced soluble levels of the phagocytic receptor MerTK, which is required for cardiac repair (52), maintenance of cardiomyocyte metabolism through removal of spent cardiomyocyte mitochondria (also known as exophers) (53), and cardiac angiogenesis (54). In addition to HIF-1α, the structurally related HIF-2α accumulates in cardiac macrophages after MI (50), and separately has been implicated in inflammatory macrophage functions (55). Like HIF-1α, HIF-2α fuels maladaptive glycolytic pathways within macrophages, albeit indirectly. That is, HIF-2α contributes to the cellular glycolytic shift by sequestering fatty acids in lipid droplets (56), in essence starving macrophage mitochondrial metabolism (50). Despite this evidence for myeloid HIF-mediated cardiopathogenic roles, others have reported that chronic hypoxia has the capacity to promote reparative macrophage functions. For example, prolonged hypoxia enhances macrophage-mediated phagocytic processes (57) through the PPP, and hypoxia preconditioning of macrophages has been shown to reduce scar size after experimental MI (58). Given the substantial interest in targeting HIFs in the setting of inflammatory ischemia/reperfusion injury (59), it will be of further interest to parse out more specific temporal and spatial contributions of hypoxic metabolism in the heterogeneous macrophage response to cardiac inflammation.

Mitochondrial metabolism and macrophage and cardiac inflammation. Flux of metabolites does not occur in isolation, and alterations in one pathway have consequences in another. Metabolites also enter divergent catabolic fates, depending on the activities or localization of metabolic enzymes and metabolite carriers. For example, glycolysis-derived pyruvate can be reduced to lactate in the cytosol or transported into the mitochondrion via the mitochondrial pyruvate carrier to feed oxidative metabolism. Prior studies have implicated mitochondrial pyruvate import in the inflammatory polarization of macrophages (60), although further experimentation is needed to test this concept in cardiac macrophages. Though often associated with antiinflammatory macrophage polarization, mitochondrial metabolism can also promote macrophage inflammation. For example, the mitochondrial electron transport chain is necessary for the activation of the macrophage inflammasome (61).

Within the mitochondrion of inflammatory macrophages, a so-called “broken,” or disrupted, TCA cycle explains reductions in coupled OXPHOS. This is characterized by an accumulation of TCA intermediates, including succinate, citrate, fumarate, and malate (62). In the case of succinate, its accumulation is a broad feature of ischemia and ischemic hearts (63). Upon reperfusion, accumulated succinate is oxidized by succinate dehydrogenase (SDH; complex II of the electron transport chain), leading to mROS accumulation at mitochondrial complex I (63) and in macrophages (64). mROS activates HIF-1α (65), and succinate further stabilizes HIF-1α by leading to the inhibition of prolyl hydroxylase domain (PHD) enzymes (66). Consistent with these signaling axes, inhibition of SDH reduces mROS and myocardial reperfusion-associated injury (63). For example, malonate is a competitive inhibitor of SDH and is cardioprotective when administered during cardiac ischemia (63), including by reducing infarct size (67). Succinate can further be excreted through plasma membrane transporters of the SLC13 family (68) and is recognized by neighboring cells through G protein–coupled receptor 91 (GPR91), thereby acting as a cell crosstalk signaling molecule (69). GPR91 activation in cardiomyocytes leads to pathological hypertrophy (70). Interestingly, activation of succinate receptor 1 (SUNCR1) can promote an antiinflammatory macrophage phenotype (71). This response is not completely surprising, as inflammation often programs its own resolution (72) through negative-feedback circuits.

The TCA metabolite citrate also accumulates in ischemic hearts (73). In some cases, citrate leaves the TCA cycle and mitochondrion via the citrate carrier (74). Once in the cytosol, cytosolic ATP citrate synthase, also known as ATP citrate lyase (ACLY), converts citrate to acetyl-CoA. Since this reaction increases the abundance of acetyl-CoA, which serves as a substrate for histone acetylation, it promotes alteration of the chromatin structure. Epigenetic mechanisms of inflammatory gene expression are triggered in inflammatory macrophages, and these responses correlate with increased ACLY and histone acetyltransferase activity (75). In macrophages, metabolic tracing studies revealed that TLR signaling redirects metabolic flux and generates acetyl-CoA from glucose to increase inflammatory gene expression (60). ACLY has been shown to be activated in cardiac vasculature macrophages during atherosclerosis (76), yet its role in myocardial tissue macrophages remains unknown and worth testing. The TCA intermediate fumarate is also implicated in inflammatory macrophages. Accumulation of fumarate induces monocytic epigenetic reprogramming to increase the inflammatory cytokines TNF-α and IL-6 (77). Moreover, inhibition of fumarate hydratase in macrophages leads to impaired mitochondrial respiration, the release of inflammatory mitochondrial RNA, and increased TNF production (78). Separately, knockout of fumarate hydratase in cardiomyocytes is cardioprotective after myocardial ischemia/reperfusion (79). If similar results are found after inhibition of cardiac macrophage fumarate hydratase, this approach could simplify potential therapeutic strategies; otherwise, cell-specific targeting would be necessary for optimal cardioprotection.

Inflammatory amino acid metabolism. Amino acids are also an important contributor to macrophage activation. For example, serine deprivation diminishes TLR4-triggered IL1B mRNA (80). Also, the amino acid arginine can be metabolized for either inflammatory or antiinflammatory macrophage actions. In the case of inflammation, the enzyme nitric oxide synthase (NOS) converts arginine to citrulline and nitric oxide (NO). Increased levels of inducible NOS (iNOS) are a signature feature of TLR-activated macrophages (81), and NO can antagonize mitochondrial metabolism by interfering with the activity of the TCA cycle enzyme aconitase 2 and pyruvate dehydrogenase (82). In patients, l-arginine, when added to standard postinfarction therapies, did not improve ejection fraction and may in fact may be associated with higher postinfarction mortality (83).

Taken together, accumulating evidence supports a concept wherein the metabolic bias of cardiac macrophages can substantially alter the amplitude of the inflammatory response to cardiac injury. From these studies, an integrated working model emerges that illustrates potential contributions of cardiac macrophage metabolites to acute cardiac inflammation. This model is depicted in Figure 2. It is important to note that not all outcomes of these inflammatory circuits are detrimental, as inflammation per se is necessary to trigger protective inflammation-resolution and tissue repair programs. Furthermore, functions of metabolic pathways are certainly context dependent, as metabolites that act to inflame macrophages in one scenario can alternatively act in an antiinflammatory or cardiac repair capacity, as discussed below.

Working model of inflammatory cardiac macrophage metabolism after MI.Figure 2

Working model of inflammatory cardiac macrophage metabolism after MI. Inflammatory immunometabolic reactions of cardiac macrophages are implicated in the response after MI. In CCR2+ macrophages, glucose utilization (detected by 18FDG-PET) through GLUT1 fuels glycolysis and inflammatory pathways including ROS, mROS, and other intermediates that promote inflammation. Metabolic HIF-1α activation may induce a disintegrin and metalloproteinase (ADAM) proteases and lead to proteolysis of phagocytic receptors such as MerTK to suppress phagocytosis of exophers and dying cells. Altered mitochondrial metabolism is also associated with the accumulation of TCA intermediates (such as succinate [SUC]) that may signal intercellular crosstalk and have the capacity to alter epigenetic regulation of proinflammatory cytokine genes. ACLY, ATP citrate lyase; CCL2, also known as monocyte chemoattractant protein-1 (MCP-1); CIT, citrate; DAMP, damage-associated molecular pattern; EPI, epigenetic; FDG, fluorodeoxyglucose; GLUT1, glucose transporter protein type 1, also known as SLC2A1; HIF-1α, hypoxia-inducible factor 1α; MPC, mitochondrial pyruvate carrier; mROS, mitochondrial reactive oxygen species; OXPHOS, oxidative phosphorylation; PDH, pyruvate dehydrogenase; PKM2, pyruvate kinase M2; PPP, pentose phosphate pathway; SDH, succinate dehydrogenase; TCA, tricarboxylic acid; TLR, Toll-like receptor.

Reparative cardiac macrophages

The response to cardiac injury follows a series of ordered and coordinated steps, initially relying on inflammatory macrophages examined above, followed by a macrophage functional polarization that actively dampens inflammation and orchestrates crosstalk with fibroblasts and other cell types, as examined below. As such, macrophages are dynamic and encoded with the capacity to divergently polarize over time and space (27). Tissue remodeling by macrophages requires cooperative signaling that is triggered through recognition of dying cells and the cytokine IL-4 (84). IL-4 facilitates antiinflammatory macrophage programming, which assists in preventing cardiac rupture (31). Repair macrophages also secrete TGF-β, which activates cardiac fibroblasts (85) and promotes replacement fibrosis (86). Cardiac macrophage function is also coupled to ontogeny. Embryo-derived resident cardiac macrophages exhibit enhanced tissue repair activity relative to their adult monocyte-derived counterparts (87). Unfortunately, the tissue-resident subset succumbs to cell death after prolonged ischemia (6), soon to be replenished by more inflammatory monocyte-derived macrophages. Reperfusion of coronary arteries spares the death of resident cardiac macrophages (88); however, it also amplifies inflammation by recruiting excess neutrophils and monocytes. Therefore, research into mechanisms or immunometabolites that polarize the macrophage repair response, particularly in the setting of reperfusion, is of considerable interest.

Macrophage metabolic switches implicated in cardiac repair

Antiinflammatory macrophage polarization is often correlated with oxidative mitochondrial metabolism that is distinct from glycolysis (89). This metabolic stereotype, however, is not absolute given the integrated nature of metabolic circuitry. For example, in IL-4–treated macrophages, glutamine and glucose carbons can be traced as sources of nitrogen and carbon, respectively, into the nucleotide sugar uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) (90), which is necessary for protein glycosylation of the prototypical antiinflammatory macrophage marker CD206. CD206, or mannose receptor, is a C-type lectin that functions in homeostatic phagocytic processes (91) and has been tested as a nuclear imaging marker for repair-phase macrophages after experimental MI (92). Glycolysis also generates lactic acid, which serves as a substrate for histone lactylation (93). Lactylation of histones can boost reparative gene activation in bone marrow and circulating monocytes, before recruitment of monocytes to infarcted hearts (39). Specifically, lactylation was enriched in the gene promoter of angiogenic Vegfa. Consistent with the therapeutic potential of this pathway, treatment with sodium lactate after experimental MI improved cardiac function, and this improvement was coupled to the angiogenic macrophage signature.

Contributions of macrophage mitochondrial metabolism to repair. After MI, macrophages increase the expression of genes that contribute to mitochondrial metabolism and OXPHOS (42). A notable mitochondrial metabolite that is induced after cardiac injury and in activated macrophages (94) is itaconate. Itaconate is generated from aconitate of the TCA cycle, and through increased expression of aconitate decarboxylase 1 (encoded by immune-responsive gene 1 [Irg1]), which can also be induced by TLR ligands. Irg1 exhibits selective expression in activated immune cells (95) and inflammatory macrophages (96). Buildup of itaconate is also facilitated by reductions of isocitrate dehydrogenase 1 (Idh1), which in resting macrophages normally acts on aconitate in the TCA cycle to generate α-ketoglutarate (α-KG), as discussed below. Thus, itaconate is a consequence of diverting aconitate away from the TCA cycle. Itaconate has notable immunosuppressive activity (97) and can act in a negative-feedback loop during inflammation. Itaconate suppresses inflammation in part by antagonizing SDH of the TCA cycle (64). This in turn lowers mROS generation from complex I of the electron transport chain. Itaconate can also suppress inflammation by alkylating an inhibitor of the antioxidant transcription factor nuclear factor erythroid 2–related factor 2 (NRF2) (97). Moreover, itaconate has been reported to inhibit components of the inflammasome and IL-1β biosynthesis (98). These activities are not all cell intrinsic, as itaconate can also be secreted and act as an agonist on neighboring cells through targeting the G protein–coupled receptor 2-oxogluarate receptor 1 (OXGR1) (99). Thus, the therapeutic potential of itaconate has been pursued by multiple groups. Dimethyl itaconate when infused intravenously can reduce myocardial infarct size (100). Also, dimethyl itaconate delivered via subcutaneous nanofiber patches attenuated adverse remodeling after experimental MI. This effect was associated with increased antiinflammatory macrophage polarization (101). It remains unknown the degree to which itaconate is produced specifically in cardiac macrophages, and whether macrophage polarization in this setting could enhance itaconate production for therapeutic effect.

Roles for additional TCA intermediates have emerged in antiinflammatory macrophages and cardioprotection. For example, α-KG from the TCA cycle is a substrate of 2-oxoglutarate–dependent dioxygenases (2-OGDDs). A notable 2-OGDD is the HIF PHD enzyme, which acts to suppress HIF levels (102). TCA metabolites are also substrates of chromatin-modifying enzymes. For example, α-KG, fueled by glutaminolysis, contributes to alternative activation of macrophages. High ratios of α-KG to succinate favor epigenetic activation of IL-4–induced macrophage genes by histone demethylases such as Jumonji domain–containing proteins (103). Low cardiac α-KG has been implicated in myocardial ischemic injury (104), but the connection between α-KG and cardiac macrophages has not yet been made. 2-Hydroxyglutarate (2-HG) is structurally similar to α-KG, and is a metabolite that can be derived from α-KG (105). 2-HG can also rewire the epigenetic landscape by inhibition of histone and DNA demethylases (106). Exposure to TLR4 ligands leads to accumulation of 2-HG in macrophages yet appears to act in an antiinflammatory capacity. Exogenous administration of the d-2-HG enantiomer of 2-HG attenuates both macrophage TNF-α production and systemic inflammation during experimental sepsis. 2-HG protects against cardiac ischemia/reperfusion injury, yet the role of cardiac macrophages was not investigated (107).

NAD+, macrophage function, and cardiac inflammation. A key metabolic function of the TCA cycle is to generate the reducing equivalents NADH2 and FADH2. NADH is oxidized by electron transport chain complex I for OXPHOS, and this generates NAD+. Mitochondrial ratios of NAD+ to NADH are reflected to an extent in the cytosol, and this distribution occurs by transfer of reducing equivalents through mitochondrial matrix shuttles, such as the malate aspartate shuttle. Increases in cytosolic NAD+ cofactors can in turn activate NAD+-dependent signaling proteins and therefore influence cell-fate decisions. Importantly, declining NAD+ levels are correlated with many diseases of aging. Macrophages from aged subjects showed increased activities of the NAD+-consuming enzyme CD38, which is linked to macrophage inflammatory polarization (108). Reduced NAD+ levels are also found in experimental models of heart failure (109). Administration of NAD+ precursors, such as nicotinamide riboside, boost low mitochondrial respiration capacity found in peripheral blood mononuclear cells from patients with systolic heart failure (110). Moreover, NAD+ has been shown to restore α-KG levels, favoring HIF-1α degradation (111) and promotion of antiinflammatory macrophage polarization (103). NAD+ is also increased after macrophages engulf apoptotic cells (discussed more below) and is required for the biosynthesis of antiinflammatory IL-10 (112). Interestingly, exogenous supplemental NAD+ was found to protect myocardium against myocardial ischemic/reperfusion injury in an animal model (113).

Amino acids, macrophage repair functions, and the heart. Amino acid availability is critical to macrophages, which encode sensors such as mTORC1 for amino acid sensing (114). Glutamine is the most abundant amino acid in plasma and is internalized in macrophages via the solute carrier family 1 member 5 (SLC1A5) transporter. Catabolism of glutamine by glutaminolysis can contribute to inflammatory macrophage activation (such as through anaplerosis), but glutamine can also support IL-4–induced macrophage polarization (90). Glutamine is uniquely metabolized during macrophage phagocytosis of apoptotic cells (efferocytosis). This buffers the mitochondrial oxidative stress that is generated during efferocytosis (115). Also, environments enriched in glutamate fuel glutaminolysis to maintain phagocytic respiratory bursts (116). Compellingly, glutamine has been reported to improve cardiac function following experimental myocardial ischemia/reperfusion (117).

Another amino acid implicated in both reparative macrophages and the heart is arginine, the catabolism of which is intricately linked to divergent macrophage polarization phenotypes. Arginase, which hydrolyzes arginine to ornithine and urea, is induced in macrophages exposed to IL-4. During efferocytosis, metabolism of apoptotic cell–derived arginine to putrescine facilitates actin-signaling pathways that enhance multiple rounds of efferocytic engulfment (118). More recent data suggest that the import of arginine-derived polyamines (such as spermidine and spermine), also from engulfed apoptotic cells, can suppress IL-1β or IL-6 (119). Despite these inflammation resolution phenotypes, it is important to remember that in macrophages arginine can also be metabolized to inflammatory NO, as discussed above, and treatment with l-arginine is not recommended following MI (83). Thus, enhancing specific downstream metabolites of arginine may more specifically promote pathways that resolve excess inflammation.

Metabolism of dying cells: contributions to cardiac repair. Resolution of inflammation acts in part through the programmed cell death of accumulated immune cells. A feature of this program is to recruit macrophages, which engulf dying cells through the process of efferocytosis. If this process is inefficient, secondary necrosis can ensue and expose self-antigen to conditions that facilitate autoimmunity. For example, MI triggers chronic cardiac autoimmunity in type 1 diabetic patients (120). Separately, diabetes is a risk factor for defective efferocytosis (121), and blockade of CCR2+ macrophages after MI in experimental nonobese diabetic animals reduced the formation of autoimmune markers (122). In addition to physically removing dead cells, efferocytosis is also a cue to activate repair signaling from within cardiac macrophages (52). After MI, this repair process can include the expression of angiogenic (54) and lymphangiogenic factors (123). Thus, efferocytosis and dying cell metabolism by cardiac macrophages is at the interface of cardiac inflammation and cardiac repair.

After MI, a sizable mass of injured tissue must be cleared and metabolized. This includes necrotic cardiomyocytes, apoptotic bodies from apoptotic cardiomyocytes (124), and apoptotic neutrophils, which turn over rapidly at injured sites. Engulfment of dying cells is a metabolic strain for macrophages, leading to oxidative stress (125). In response, phagocytes induce antioxidant mRNAs (126). Exposure to high amounts of dying cells can lead to additional compensatory mechanisms, including the activation of both mitochondrial uncoupling proteins (127) and mitochondrial fission machinery (128). Efferocytosis also triggers increased glycolysis and generation of lactate. Secreted lactate in this context is recognized by receptors on neighboring macrophages and can promote antiinflammatory macrophage polarization (129). After MI, lactate has been shown to preserve ventricular function (130).

What is the fate of metabolites sourced from dying cells in the heart? If metabolites are limiting in ischemic tissue, it could be advantageous for cardiac macrophages to recycle or metabolize substrates from dying cells, for the purpose of tissue repair. Alternatively, engulfed metabolites could serve as signaling cues to modulate the macrophage response. Indeed, there is experimental evidence supporting these concepts, such as with arginine, as aforementioned. Moreover, apoptotic cell–derived nucleotides trigger intracellular signaling that promotes macrophage proliferation (131). This finding is particularly intriguing in light of cardiac macrophage populations that expand through proliferation (132). Thus, specific apoptotic cell–derived factors may be utilized to potentially expand selective cardioprotective macrophage populations.

Dying cells also deliver a large payload of lipids and membrane-derived cholesterol to phagocytes. While macrophage lipid accumulation is linked to inflammatory responses, such as during atherosclerosis, the process of efferocytosis triggers compensatory reactions that not only mitigate lipid-induced cellular stress (133) but also trigger tissue-reparative signaling. For example, lysosomal acid lipase hydrolyzes cholesterol esters in endocytic compartments, and this response can lead to antiinflammatory oxysterol production (134). Also, increased fatty acids in apoptotic cells contribute to antiinflammatory macrophage responses (e.g., IL-10) (112), and separately, fatty acid oxidation and activation of PPARγ coactivator-1β (PGC-1β) have been shown to polarize macrophages to an antiinflammatory phenotype (135). Furthermore, genetic silencing of mitochondrial lipid importers and components of the electron transport chain inhibit IL-10 production during efferocytosis, suggesting a role for fatty acid oxidation (112). Finally, disruption of electron transport chain complexes I (136) and III (112) in myeloid cells leads to poor outcomes after experimental MI.

Collectively, the metabolic signatures of antiinflammatory and pro-repair macrophages discussed above are consistent with the concept that specific metabolic pathways and metabolites have the capacity to enhance tissue repair through cardiac macrophages. A working model emerges from these data that outlines potential contributions of cardiac macrophage metabolism to cardiac healing (Figure 3).

Working model of the contributions of cardiac macrophage metabolism to inflFigure 3

Working model of the contributions of cardiac macrophage metabolism to inflammation resolution and cardiac repair after MI. Repair macrophage metabolism is associated with both glycolytic and mitochondrial pathways of cellular metabolism. Glycolysis-derived lactate has the capacity to polarize macrophages to an antiinflammatory state. Fatty acids, including fatty acids derived from apoptotic cells, may enter the mitochondrion of the phagocyte (such as through MerTK-dependent efferocytosis) to fuel increases in mitochondrial respiration and the generation of NAD+. NAD+ can facilitate cellular signaling pathways that lead to induction of pro-repair cytokines. ARG, arginine; CPT, carnitine palmitoyltransferase; EPI, epigenetic; FA, fatty acid; FAO, fatty acid oxidation; GLN, glutamine; HIF, hypoxia-inducible factor; ITA, itaconate; α-KG, α-ketoglutarate; LA, lactylation; LIPA, lysosomal acid lipase; NAD+, nicotinamide adenine dinucleotide; NMN, nicotinamide mononucleotide; NFR2, nuclear factor erythroid 2–related factor 2; NR, nicotinamide riboside; PYR, pyruvate; PUT, putrescine; TCA, tricarboxylic acid.

Future research directions and therapeutic implications

Like the overgeneralized dichotomy between M1 and M2 macrophages (137), current dogmas that divide inflammatory versus repair programming into glycolytic versus oxidative metabolism in the macrophage are also an oversimplification. Moreover, a profound limitation to our working models is that most of our immunometabolic insight originates from studies performed on cells cultured in vitro. New technical approaches are needed that preserve in vivo metabolic phenotypes. Another cautionary tale is that we too often make metabolic inferences from static quantifications of metabolite levels that can be deceiving. For example, higher metabolite concentrations can be a function of slower metabolite consumption or accelerated production. It is even formally possible that macrophages could undergo higher metabolite flux without any change in detectable levels. Looking ahead, experimental approaches that incorporate multiple orthogonal perspectives, including real-time in vivo metabolic tracing, will be key in understanding the true evolution of cardiac macrophage metabolism, as an element of macrophage function, after MI.

There is increasing evidence that mitochondrial dysfunction is associated with disease. Mitochondrial OXPHOS is a key signature of tissue macrophage homeostasis (138). Important questions remain unanswered with respect to the extent to which natural age-associated declines in macrophage metabolism may contribute to the efficiency of recovery after MI. Future studies are also needed to determine the extent to which metabolic MI risk factors contribute to cardiac macrophage responsiveness. For example, hyperlipidemic individuals are at increased risk for heart failure after MI (14), yet our understanding of how excess lipid load likely affects cardiac macrophage functions remains vague and incomplete. Also, advancements in the imaging of in vivo mitochondrial dynamics may provide further clues to the relationship between cardiac macrophage metabolism and disease progression (139).

Beyond the satisfaction of discovering fundamental new mechanisms linking metabolism to immune function, inflammation, and cardiac repair, this space also harbors powerful potential for therapeutic intervention. Macrophages have been implicated in the benefits associated with cell therapy after MI (140, 141). As such, optimizing cardiac macrophage metabolism to achieve optimal cardioprotection is a logical next step. Effective cardioprotection may be achieved through the administration of select bioactive metabolites such as itaconate or NAD+ (111), or alternatively by targeting of broader metabolic pathways, such as OXPHOS (142). Whatever the case may be, the field is moving fast, and new discoveries and therapeutic approaches are on the horizon.

Acknowledgments

Thank you to members of the laboratory for their dedication to new discoveries and ideas in the field of innate immunology, inflammation, and cardiovascular disease, particularly to Mallory Filipp for critical comments on the manuscript. Special thanks also to key collaborators and colleagues in this space, including Laurent Yvan-Charvet, Nav Chandel, and Sam Weinberg. This Review was supported by NIH National Heart, Lung, and Blood Institute grant R01HL122309.

Address correspondence to: Edward B. Thorp, Feinberg School of Medicine, Northwestern University, Department of Pathology, 303 E. Chicago Avenue Ward 4-116, Chicago, Illinois 60611, USA. Phone: 312.503.4309; Email: ebthorp@northwestern.edu.

Footnotes

Conflict of interest: The author has declared that no conflict of interest exists.

Copyright: © 2023, Thorp et al. This is an open access article published under the terms of the Creative Commons Attribution 4.0 International License.

Reference information: J Clin Invest. 2023;133(18):e171953. https://doi.org/10.1172/JCI171953.

References
  1. Ginhoux F, Guilliams M. Tissue-resident macrophage ontogeny and homeostasis. Immunity. 2016;44(3):439–449.
    View this article via: CrossRef PubMed Google Scholar
  2. Gordon S. The macrophage: past, present and future. Eur J Immunol. 2007;37(1):9–17.
    View this article via: CrossRef PubMed Google Scholar
  3. Guilliams M, et al. Establishment and maintenance of the macrophage niche. Immunity. 2020;52(3):434–451.
    View this article via: CrossRef PubMed Google Scholar
  4. Pinto AR, et al. Revisiting cardiac cellular composition. Circ Res. 2016;118(3):400–409.
    View this article via: CrossRef PubMed Google Scholar
  5. Epelman S, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. 2014;40(1):91–104.
    View this article via: CrossRef PubMed Google Scholar
  6. Heidt T, et al. Differential contribution of monocytes to heart macrophages in steady-state and after myocardial infarction. Circ Res. 2014;115(2):284–295.
    View this article via: CrossRef PubMed Google Scholar
  7. Dick SA, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Sci Immunol. 2022;7(67):eabf7777.
    View this article via: CrossRef PubMed Google Scholar
  8. Kaufmann A, et al. Increase of CCR1 and CCR5 expression and enhanced functional response to MIP-1 alpha during differentiation of human monocytes to macrophages. J Leukoc Biol. 2001;69(2):248–252.
    View this article via: CrossRef PubMed Google Scholar
  9. Bajpai G, et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat Med. 2018;24(8):1234–1245.
    View this article via: CrossRef PubMed Google Scholar
  10. Leid J, et al. Primitive embryonic macrophages are required for coronary development and maturation. Circ Res. 2016;118(10):1498–1511.
    View this article via: CrossRef PubMed Google Scholar
  11. Cahill TJ, et al. Tissue-resident macrophages regulate lymphatic vessel growth and patterning in the developing heart. Development. 2021;148(3):dev194563.
    View this article via: CrossRef PubMed Google Scholar
  12. Hulsmans M, et al. Macrophages facilitate electrical conduction in the heart. Cell. 2017;169(3):510–522.
    View this article via: CrossRef PubMed Google Scholar
  13. Molawi K, et al. Progressive replacement of embryo-derived cardiac macrophages with age. J Exp Med. 2014;211(11):2151–2158.
    View this article via: CrossRef PubMed Google Scholar
  14. Tsao CW, et al. Heart disease and stroke statistics—2023 update: a report from the American Heart Association. Circulation. 2023;147(8):93–621.
    View this article via: CrossRef PubMed Google Scholar
  15. van Amerongen MJ, et al. Macrophage depletion impairs wound healing and increases left ventricular remodeling after myocardial injury in mice. Am J Pathol. 2007;170(3):818–829.
    View this article via: CrossRef PubMed Google Scholar
  16. Fan Q, et al. Dectin-1 contributes to myocardial ischemia/reperfusion injury by regulating macrophage polarization and neutrophil infiltration. Circulation. 2019;139(5):663–678.
    View this article via: CrossRef PubMed Google Scholar
  17. Davidson SM, et al. Multitarget strategies to reduce myocardial ischemia/reperfusion injury: JACC review topic of the week. J Am Coll Cardiol. 2019;73(1):89–99.
    View this article via: CrossRef PubMed Google Scholar
  18. Kaikita K, et al. Targeted deletion of CC chemokine receptor 2 attenuates left ventricular remodeling after experimental myocardial infarction. Am J Pathol. 2004;165(2):439–447.
    View this article via: CrossRef PubMed Google Scholar
  19. Yu L, et al. Megakaryocytic leukemia 1 bridges epigenetic activation of NADPH oxidase in macrophages to cardiac ischemia-reperfusion injury. Circulation. 2018;138(24):2820–2836.
    View this article via: CrossRef PubMed Google Scholar
  20. Bujak M, et al. Aging-related defects are associated with adverse cardiac remodeling in a mouse model of reperfused myocardial infarction. J Am Coll Cardiol. 2008;51(14):1384–1392.
    View this article via: CrossRef PubMed Google Scholar
  21. Soehnlein O, et al. Neutrophil secretion products pave the way for inflammatory monocytes. Blood. 2008;112(4):1461–1471.
    View this article via: CrossRef PubMed Google Scholar
  22. Zouggari Y, et al. B lymphocytes trigger monocyte mobilization and impair heart function after acute myocardial infarction. Nat Med. 2013;19(10):1273–1280.
    View this article via: CrossRef PubMed Google Scholar
  23. Dehn S, Thorp EB. Myeloid receptor CD36 is required for early phagocytosis of myocardial infarcts and induction of Nr4a1-dependent mechanisms of cardiac repair. FASEB J. 2018;32(1):254–264.
    View this article via: CrossRef PubMed Google Scholar
  24. Bajpai G, et al. Tissue resident CCR2- and CCR2+ cardiac macrophages differentially orchestrate monocyte recruitment and fate specification following myocardial injury. Circ Res. 2019;124(2):263–278.
    View this article via: CrossRef PubMed Google Scholar
  25. Ridker PM, et al. Antiinflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119–1131.
    View this article via: CrossRef PubMed Google Scholar
  26. Dick SA, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. 2019;20(1):29–39.
    View this article via: CrossRef PubMed Google Scholar
  27. Murray PJ, et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 2014;41(1):14–20.
    View this article via: CrossRef PubMed Google Scholar
  28. West AP, et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472(7344):476–480.
    View this article via: CrossRef PubMed Google Scholar
  29. Zhang W, et al. Necrotic myocardial cells release damage-associated molecular patterns that provoke fibroblast activation in vitro and trigger myocardial inflammation and fibrosis in vivo. J Am Heart Assoc. 2015;4(6):e001993.
    View this article via: CrossRef PubMed Google Scholar
  30. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 2014;6:13.
    View this article via: CrossRef PubMed Google Scholar
  31. Shiraishi M, et al. Alternatively activated macrophages determine repair of the infarcted adult murine heart. J Clin Invest. 2016;126(6):2151–2166.
    View this article via: JCI CrossRef PubMed Google Scholar
  32. Sarrazy V, et al. Disruption of Glut1 in hematopoietic stem cells prevents myelopoiesis and enhanced glucose flux in atheromatous plaques of ApoE(-/-) mice. Circ Res. 2016;118(7):1062–1077.
    View this article via: CrossRef PubMed Google Scholar
  33. Shirai T, et al. The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. J Exp Med. 2016;213(3):337–354.
    View this article via: CrossRef PubMed Google Scholar
  34. Lee MKS, et al. Glycolysis is required for LPS-induced activation and adhesion of human CD14(+)CD16(-) monocytes. Front Immunol. 2019;10:2054.
    View this article via: CrossRef PubMed Google Scholar
  35. Warburg O, et al. The metabolism of tumors in the body. J Gen Physiol. 1927;8(6):519–530.
    View this article via: CrossRef PubMed Google Scholar
  36. Lachmandas E, et al. Microbial stimulation of different Toll-like receptor signalling pathways induces diverse metabolic programmes in human monocytes. Nat Microbiol. 2016;2(3):16246.
    View this article via: CrossRef PubMed Google Scholar
  37. Ma J, et al. Glycogen metabolism regulates macrophage-mediated acute inflammatory responses. Nat Commun. 2020;11(1):1769.
    View this article via: CrossRef PubMed Google Scholar
  38. Ruparelia N, et al. Acute myocardial infarction activates distinct inflammation and proliferation pathways in circulating monocytes, prior to recruitment, and identified through conserved transcriptional responses in mice and humans. Eur Heart J. 2015;36(29):1923–1934.
    View this article via: CrossRef PubMed Google Scholar
  39. Wang N, et al. Histone lactylation boosts reparative gene activation post-myocardial infarction. Circ Res. 2022;131(11):893–908.
    View this article via: CrossRef PubMed Google Scholar
  40. Lee WW, et al. PET/MRI of inflammation in myocardial infarction. J Am Coll Cardiol. 2012;59(2):153–163.
    View this article via: CrossRef PubMed Google Scholar
  41. Rischpler C, et al. Prospective evaluation of 18F-fluorodeoxyglucose uptake in postischemic myocardium by simultaneous positron emission tomography/magnetic resonance imaging as a prognostic marker of functional outcome. Circ Cardiovasc Imaging. 2016;9(4):e004316.
    View this article via: CrossRef PubMed Google Scholar
  42. Mouton AJ, et al. Mapping macrophage polarization over the myocardial infarction time continuum. Basic Res Cardiol. 2018;113(4):26.
    View this article via: CrossRef PubMed Google Scholar
  43. Hou X, et al. The cardiac microenvironment instructs divergent monocyte fates and functions in myocarditis. Cell Rep. 2019;28(1):172–189.
    View this article via: CrossRef PubMed Google Scholar
  44. Handley MG, et al. PET imaging of cardiac hypoxia: opportunities and challenges. J Mol Cell Cardiol. 2011;51(5):640–650.
    View this article via: CrossRef PubMed Google Scholar
  45. Iyer NV, et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1 alpha. Genes Dev. 1998;12(2):149–162.
    View this article via: CrossRef PubMed Google Scholar
  46. Seagroves TN, et al. Transcription factor HIF-1 is a necessary mediator of the Pasteur effect in mammalian cells. Mol Cell Biol. 2001;21(10):3436–3444.
    View this article via: CrossRef PubMed Google Scholar
  47. Kim JW, et al. HIF-1-mediated expression of pyruvate dehydrogenase kinase: a metabolic switch required for cellular adaptation to hypoxia. Cell Metab. 2006;3(3):177–185.
    View this article via: CrossRef PubMed Google Scholar
  48. Tannahill GM, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–242.
    View this article via: CrossRef PubMed Google Scholar
  49. Peyssonnaux C, et al. HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Invest. 2005;115(7):1806–1815.
    View this article via: JCI CrossRef PubMed Google Scholar
  50. DeBerge M, et al. Hypoxia-inducible factors individually facilitate inflammatory myeloid metabolism and inefficient cardiac repair. J Exp Med. 2021;218(9):e20200667.
    View this article via: CrossRef PubMed Google Scholar
  51. Dong F, et al. Critical role for leukocyte hypoxia inducible factor-1alpha expression in post-myocardial infarction left ventricular remodeling. Circ Res. 2010;106(3):601–610.
    View this article via: CrossRef PubMed Google Scholar
  52. Wan E, et al. Enhanced efferocytosis of apoptotic cardiomyocytes through myeloid-epithelial-reproductive tyrosine kinase links acute inflammation resolution to cardiac repair after infarction. Circ Res. 2013;113(8):1004–1012.
    View this article via: CrossRef PubMed Google Scholar
  53. Nicolás-Ávila JA, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. 2020;183(1):94–109.
    View this article via: CrossRef PubMed Google Scholar
  54. Howangyin KY, 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(9):826–839.
    View this article via: CrossRef PubMed Google Scholar
  55. Imtiyaz HZ, et al. Hypoxia-inducible factor 2alpha regulates macrophage function in mouse models of acute and tumor inflammation. J Clin Invest. 2010;120(8):2699–2714.
    View this article via: JCI CrossRef PubMed Google Scholar
  56. Rankin EB, et al. Hypoxia-inducible factor 2 regulates hepatic lipid metabolism. Mol Cell Biol. 2009;29(16):4527–4538.
    View this article via: CrossRef PubMed Google Scholar
  57. Wang YT, et al. Metabolic adaptation supports enhanced macrophage efferocytosis in limited-oxygen environments. Cell Metab. 2023;35(2):316–331.
    View this article via: CrossRef PubMed Google Scholar
  58. Zhu Y, et al. Hypoxia-primed monocytes/macrophages enhance postinfarction myocardial repair. Theranostics. 2022;12(1):307–323.
    View this article via: CrossRef PubMed Google Scholar
  59. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med. 2011;364(7):656–665.
    View this article via: CrossRef PubMed Google Scholar
  60. Lauterbach MA, et al. Toll-like receptor signaling rewires macrophage metabolism and promotes histone acetylation via ATP-citrate lyase. Immunity. 2019;51(6):997–1011.
    View this article via: CrossRef PubMed Google Scholar
  61. Billingham LK, et al. Mitochondrial electron transport chain is necessary for NLRP3 inflammasome activation. Nat Immunol. 2022;23(5):692–704.
    View this article via: CrossRef PubMed Google Scholar
  62. O’Neill LA. A broken Krebs cycle in macrophages. Immunity. 2015;42(3):393–394.
    View this article via: CrossRef PubMed Google Scholar
  63. Chouchani ET, et al. Ischaemic accumulation of succinate controls reperfusion injury through mitochondrial ROS. Nature. 2014;515(7527):431–435.
    View this article via: CrossRef PubMed Google Scholar
  64. Mills EL, et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell. 2016;167(2):457–470.
    View this article via: CrossRef PubMed Google Scholar
  65. Chandel NS, et al. Reactive oxygen species generated at mitochondrial complex III stabilize hypoxia-inducible factor-1alpha during hypoxia: a mechanism of O2 sensing. J Biol Chem. 2000;275(33):25130–25138.
    View this article via: CrossRef PubMed Google Scholar
  66. MacKenzie ED, et al. Cell-permeating alpha-ketoglutarate derivatives alleviate pseudohypoxia in succinate dehydrogenase-deficient cells. Mol Cell Biol. 2007;27(9):3282–3289.
    View this article via: CrossRef PubMed Google Scholar
  67. Valls-Lacalle L, et al. Selective inhibition of succinate dehydrogenase in reperfused myocardium with intracoronary malonate reduces infarct size. Sci Rep. 2018;8(1):2442.
    View this article via: CrossRef PubMed Google Scholar
  68. Khamaysi A, et al. A dynamic anchor domain in slc13 transporters controls metabolite transport. J Biol Chem. 2020;295(24):8155–8163.
    View this article via: CrossRef PubMed Google Scholar
  69. Littlewood-Evans A, et al. GPR91 senses extracellular succinate released from inflammatory macrophages and exacerbates rheumatoid arthritis. J Exp Med. 2016;213(9):1655–1662.
    View this article via: CrossRef PubMed Google Scholar
  70. Aguiar CJ, et al. Succinate causes pathological cardiomyocyte hypertrophy through GPR91 activation. Cell Commun Signal. 2014;12:78.
    View this article via: CrossRef PubMed Google Scholar
  71. Keiran N, et al. SUCNR1 controls an anti-inflammatory program in macrophages to regulate the metabolic response to obesity. Nat Immunol. 2019;20(5):581–592.
    View this article via: CrossRef PubMed Google Scholar
  72. Serhan CN, Savill J. Resolution of inflammation: the beginning programs the end. Nat Immunol. 2005;6(12):1191–1197.
    View this article via: CrossRef PubMed Google Scholar
  73. Hassel B, et al. Cardiac accumulation of citrate during brief myocardial ischaemia and reperfusion in the pig in vivo. Acta Physiol Scand. 1998;164(1):53–59.
    View this article via: CrossRef PubMed Google Scholar
  74. Tan M, et al. Inhibition of the mitochondrial citrate carrier, Slc25a1, reverts steatosis, glucose intolerance, and inflammation in preclinical models of NAFLD/NASH. Cell Death Differ. 2020;27(7):2143–2157.
    View this article via: CrossRef PubMed Google Scholar
  75. Wellen KE, et al. ATP-citrate lyase links cellular metabolism to histone acetylation. Science. 2009;324(5930):1076–1080.
    View this article via: CrossRef PubMed Google Scholar
  76. Baardman J, et al. Macrophage ATP citrate lyase deficiency stabilizes atherosclerotic plaques. Nat Commun. 2020;11(1):6296.
    View this article via: CrossRef PubMed Google Scholar
  77. Arts RJ, et al. Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity. Cell Metab. 2016;24(6):807–819.
    View this article via: CrossRef PubMed Google Scholar
  78. Hooftman A, et al. Macrophage fumarate hydratase restrains mtRNA-mediated interferon production. Nature. 2023;615(7952):490–498.
    View this article via: CrossRef PubMed Google Scholar
  79. Ashrafian H, et al. Fumarate is cardioprotective via activation of the Nrf2 antioxidant pathway. Cell Metab. 2012;15(3):361–371.
    View this article via: CrossRef PubMed Google Scholar
  80. Rodriguez AE, et al. Serine metabolism supports macrophage IL-1β production. Cell Metab. 2019;29(4):1003–1011.
    View this article via: CrossRef PubMed Google Scholar
  81. MacMicking J, et al. Nitric oxide and macrophage function. Annu Rev Immunol. 1997;15:323–350.
    View this article via: CrossRef PubMed Google Scholar
  82. Palmieri EM, et al. Nitric oxide orchestrates metabolic rewiring in M1 macrophages by targeting aconitase 2 and pyruvate dehydrogenase. Nat Commun. 2020;11(1):698.
    View this article via: CrossRef PubMed Google Scholar
  83. Schulman SP, et al. L-arginine therapy in acute myocardial infarction: the Vascular Interaction With Age in Myocardial Infarction (VINTAGE MI) randomized clinical trial. JAMA. 2006;295(1):58–64.
    View this article via: CrossRef PubMed Google Scholar
  84. Bosurgi L, et al. Macrophage function in tissue repair and remodeling requires IL-4 or IL-13 with apoptotic cells. Science. 2017;356(6342):1072–1076.
    View this article via: CrossRef PubMed Google Scholar
  85. Davis J, Molkentin JD. Myofibroblasts: trust your heart and let fate decide. J Mol Cell Cardiol. 2014;70:9–18.
    View this article via: CrossRef PubMed Google Scholar
  86. Frangogiannis N. Transforming growth factor-β in tissue fibrosis. J Exp Med. 2020;217(3):e20190103.
    View this article via: CrossRef PubMed Google Scholar
  87. Lavine KJ, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci U S A. 2014;111(45):16029–16034.
    View this article via: CrossRef PubMed Google Scholar
  88. Leuschner F, et al. Rapid monocyte kinetics in acute myocardial infarction are sustained by extramedullary monocytopoiesis. J Exp Med. 2012;209(1):123–137.
    View this article via: CrossRef PubMed Google Scholar
  89. Wang F, et al. Glycolytic stimulation is not a requirement for M2 macrophage differentiation. Cell Metab. 2018;28(3):463–475.e4.
    View this article via: CrossRef PubMed Google Scholar
  90. Jha AK, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419–430.
    View this article via: CrossRef PubMed Google Scholar
  91. Feinberg H, et al. Structural analysis of carbohydrate binding by the macrophage mannose receptor CD206. J Biol Chem. 2021;296:100368.
    View this article via: CrossRef PubMed Google Scholar
  92. Varasteh Z, et al. In vivo visualization of M2 macrophages in the myocardium after myocardial infarction (MI) using 68Ga-NOTA-Anti-MMR Nb: targeting mannose receptor (MR, CD206) on M2 macrophages. Front Cardiovasc Med. 2022;9:889963.
    View this article via: CrossRef PubMed Google Scholar
  93. Zhang D, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574(7779):575–580.
    View this article via: CrossRef PubMed Google Scholar
  94. O’Neill LAJ, Artyomov MN. Itaconate: the poster child of metabolic reprogramming in macrophage function. Nat Rev Immunol. 2019;19(5):273–281.
    View this article via: CrossRef PubMed Google Scholar
  95. Michelucci A, et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc Natl Acad Sci U S A. 2013;110(19):7820–7825.
    View this article via: CrossRef PubMed Google Scholar
  96. Strelko CL, et al. Itaconic acid is a mammalian metabolite induced during macrophage activation. J Am Chem Soc. 2011;133(41):16386–16389.
    View this article via: CrossRef PubMed Google Scholar
  97. Mills EL, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556(7699):113–117.
    View this article via: CrossRef PubMed Google Scholar
  98. Hooftman A, et al. The immunomodulatory metabolite itaconate modifies NLRP3 and inhibits inflammasome activation. Cell Metab. 2020;32(3):468–478.
    View this article via: CrossRef PubMed Google Scholar
  99. Zeng YR, et al. The immunometabolite itaconate stimulates OXGR1 to promote mucociliary clearance during the pulmonary innate immune response. J Clin Invest. 2023;133(6):e160463.
    View this article via: JCI CrossRef PubMed Google Scholar
  100. Lampropoulou V, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 2016;24(1):158–166.
    View this article via: CrossRef PubMed Google Scholar
  101. Nakkala JR, et al. Dimethyl itaconate-loaded nanofibers rewrite macrophage polarization, reduce inflammation, and enhance repair of myocardic infarction. Small. 2021;17(17):2006992.
    View this article via: CrossRef PubMed Google Scholar
  102. Bruick RK, McKnight SL. A conserved family of prolyl-4-hydroxylases that modify HIF. Science. 2001;294(5545):1337–1340.
    View this article via: CrossRef PubMed Google Scholar
  103. Liu PS, et al. α-Ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat Immunol. 2017;18(9):985–994.
    View this article via: CrossRef PubMed Google Scholar
  104. Kjellman U, et al. Alpha-ketoglutarate for myocardial protection in heart surgery. Lancet. 1995;345(8949):552–553.
    View this article via: CrossRef PubMed Google Scholar
  105. de Goede KE, et al. d-2-Hydroxyglutarate is an anti-inflammatory immunometabolite that accumulates in macrophages after TLR4 activation. Biochim Biophys Acta Mol Basis Dis. 2022;1868(9):166427.
    View this article via: CrossRef PubMed Google Scholar
  106. Martínez-Reyes I, Chandel NS. Mitochondrial TCA cycle metabolites control physiology and disease. Nat Commun. 2020;11(1):102.
    View this article via: CrossRef PubMed Google Scholar
  107. He H, et al. L-2-Hydroxyglutarate protects against cardiac injury via metabolic remodeling. Circ Res. 2022;131(7):562–579.
    View this article via: CrossRef PubMed Google Scholar
  108. Covarrubias AJ, et al. Senescent cells promote tissue NAD+ decline during ageing via the activation of CD38+ macrophages. Nat Metab. 2020;2(11):1265–1283.
    View this article via: CrossRef PubMed Google Scholar
  109. Diguet N, et al. Nicotinamide riboside preserves cardiac function in a mouse model of dilated cardiomyopathy. Circulation. 2018;137(21):2256–2273.
    View this article via: CrossRef PubMed Google Scholar
  110. Zhou B, et al. Boosting NAD level suppresses inflammatory activation of PBMCs in heart failure. J Clin Invest. 2020;130(11):6054–6063.
    View this article via: JCI CrossRef PubMed Google Scholar
  111. Lee CF, et al. Targeting NAD+ metabolism as interventions for mitochondrial disease. Sci Rep. 2019;9(1):3073.
    View this article via: CrossRef PubMed Google Scholar
  112. Zhang S, et al. Efferocytosis fuels requirements of fatty acid oxidation and the electron transport chain to polarize macrophages for tissue repair. Cell Metab. 2019;29(2):443–456.
    View this article via: CrossRef PubMed Google Scholar
  113. Zhai X, et al. Exogenous supplemental NAD+ protect myocardium against myocardial ischemic/reperfusion injury in swine model. Am J Transl Res. 2019;11(9):6066–6074.
    View this article via: PubMed Google Scholar
  114. Zoncu R, et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H(+)-ATPase. Science. 2011;334(6056):678–683.
    View this article via: CrossRef PubMed Google Scholar
  115. Merlin J, et al. Non-canonical glutamine transamination sustains efferocytosis by coupling redox buffering to oxidative phosphorylation. Nat Metab. 2021;3(10):1313–1326.
    View this article via: CrossRef PubMed Google Scholar
  116. Davies LC, et al. Peritoneal tissue-resident macrophages are metabolically poised to engage microbes using tissue-niche fuels. Nat Commun. 2017;8(1):2074.
    View this article via: CrossRef PubMed Google Scholar
  117. Bolotin G, et al. Glutamine improves myocardial function following ischemia-reperfusion injury. Asian Cardiovasc Thorac Ann. 2007;15(6):463–467.
    View this article via: CrossRef PubMed Google Scholar
  118. Yurdagul A Jr. , et al. Macrophage metabolism of apoptotic cell-derived arginine promotes continual efferocytosis and resolution of injury. Cell Metab. 2020;31(3):518–533.
    View this article via: CrossRef PubMed Google Scholar
  119. McCubbrey AL, et al. Polyamine import and accumulation causes immunomodulation in macrophages engulfing apoptotic cells. Cell Rep. 2022;38(2):110222.
    View this article via: CrossRef PubMed Google Scholar
  120. Gottumukkala RV, et al. Myocardial infarction triggers chronic cardiac autoimmunity in type 1 diabetes. Sci Transl Med. 2012;4(138):138ra80.
    View this article via: CrossRef PubMed Google Scholar
  121. Maschalidi S, et al. Targeting SLC7A11 improves efferocytosis by dendritic cells and wound healing in diabetes. Nature. 2022;606(7915):776–784.
    View this article via: CrossRef PubMed Google Scholar
  122. DeBerge M, et al. Monocytes prime autoreactive T cells after myocardial infarction. Am J Physiol Heart Circ Physiol. 2020;318(1):H116–H123.
    View this article via: CrossRef PubMed Google Scholar
  123. Glinton KE, et al. Macrophage-produced VEGFC is induced by efferocytosis to ameliorate cardiac injury and inflammation. J Clin Invest. 2022;132(9):e140685.
    View this article via: JCI CrossRef PubMed Google Scholar
  124. Zhang S, et al. Cardiomyocytes induce macrophage receptor shedding to suppress phagocytosis. J Mol Cell Cardiol. 2015;87:171–179.
    View this article via: CrossRef PubMed Google Scholar
  125. Yvan-Charvet L, et al. ABCA1 and ABCG1 protect against oxidative stress-induced macrophage apoptosis during efferocytosis. Circ Res. 2013;106(12):1861–1869.
    View this article via: CrossRef PubMed Google Scholar
  126. Lantz C, et al. Single-cell RNA sequencing uncovers heterogenous transcriptional signatures in macrophages during efferocytosis. Sci Rep. 2020;10(1):14333.
    View this article via: CrossRef PubMed Google Scholar
  127. Park D, et al. Continued clearance of apoptotic cells critically depends on the phagocyte Ucp2 protein. Nature. 2011;477(7363):220–224.
    View this article via: CrossRef PubMed Google Scholar
  128. Wang Y, et al. Mitochondrial fission promotes the continued clearance of apoptotic cells by macrophages. Cell. 2017;171(2):331–345.
    View this article via: CrossRef PubMed Google Scholar
  129. Morioka S, et al. Efferocytosis induces a novel SLC program to promote glucose uptake and lactate release. Nature. 2018;563(7733):714–718.
    View this article via: CrossRef PubMed Google Scholar
  130. Chen Y, et al. LDHA-mediated metabolic reprogramming promoted cardiomyocyte proliferation by alleviating ROS and inducing M2 macrophage polarization. Redox Biol. 2022;56:102446.
    View this article via: CrossRef PubMed Google Scholar
  131. Gerlach BD, et al. Efferocytosis induces macrophage proliferation to help resolve tissue injury. Cell Metab. 2021;33(12):2445–2463.
    View this article via: CrossRef PubMed Google Scholar
  132. Hilgendorf I, et al. Ly-6Chigh monocytes depend on Nr4a1 to balance both inflammatory and reparative phases in the infarcted myocardium. Circ Res. 2014;114(10):1611–1622.
    View this article via: CrossRef PubMed Google Scholar
  133. Cui D, et al. Pivotal advance: macrophages become resistant to cholesterol-induced death after phagocytosis of apoptotic cells. J Leukoc Biol. 2007;82(5):1040–1050.
    View this article via: CrossRef PubMed Google Scholar
  134. Viaud M, et al. Lysosomal cholesterol hydrolysis couples efferocytosis to anti-inflammatory oxysterol production. Circ Res. 2018;122(10):1369–1384.
    View this article via: CrossRef PubMed Google Scholar
  135. Vats D, et al. Oxidative metabolism and PGC-1beta attenuate macrophage-mediated inflammation. Cell Metab. 2006;4(1):13–24.
    View this article via: CrossRef PubMed Google Scholar
  136. Cai S, et al. Mitochondrial dysfunction in macrophages promotes inflammation and suppresses repair after myocardial infarction. J Clin Invest. 2023;133(4):e159498.
    View this article via: JCI CrossRef PubMed Google Scholar
  137. Nahrendorf M, Swirski FK. Abandoning M1/M2 for a network model of macrophage function. Circ Res. 2016;119(3):414–417.
    View this article via: CrossRef PubMed Google Scholar
  138. Wculek SK, et al. Oxidative phosphorylation selectively orchestrates tissue macrophage homeostasis. Immunity. 2023;56(3):516–530.e9.
    View this article via: CrossRef PubMed Google Scholar
  139. Li Y, et al. Imaging of macrophage mitochondria dynamics in vivo reveals cellular activation phenotype for diagnosis. Theranostics. 2020;10(7):2897–2917.
    View this article via: CrossRef PubMed Google Scholar
  140. de Couto G, et al. Mechanism of enhanced MerTK-dependent macrophage efferocytosis by extracellular vesicles. Arterioscler Thromb Vasc Biol. 2019;39(10):2082–2096.
    View this article via: CrossRef PubMed Google Scholar
  141. Vagnozzi RJ, et al. An acute immune response underlies the benefit of cardiac stem cell therapy. Nature. 2020;577(7790):405–409.
    View this article via: CrossRef PubMed Google Scholar
  142. Singh A, et al. Pharmacological advances in mitochondrial therapy. EBioMedicine. 2021;65:103244.
    View this article via: CrossRef PubMed Google Scholar
Version history
  • Version 1 (September 15, 2023): Electronic publication

Article tools

  • View PDF
  • Download citation information
  • Send a comment
  • Terms of use
  • Standard abbreviations
  • Need help? Email the journal

Metrics

  • Article usage
  • Citations to this article

Go to

  • Top
  • Abstract
  • Introduction to the cardiac macrophage
  • Cardiac macrophages in the acute response to myocardial infarction
  • Relevance of macrophage metabolic signaling
  • Inflammatory metabolism of monocytes and macrophages after acute MI
  • Reparative cardiac macrophages
  • Macrophage metabolic switches implicated in cardiac repair
  • Future research directions and therapeutic implications
  • Acknowledgments
  • Footnotes
  • References
  • Version history
Advertisement
Advertisement

Copyright © 2025 American Society for Clinical Investigation
ISSN: 0021-9738 (print), 1558-8238 (online)

Sign up for email alerts