Back to Journals » ImmunoTargets and Therapy » Volume 14
The Regulatory Network of Transcription Factors in Macrophage Polarization
Authors Liu J, Wang M , Zhao Y
Received 10 October 2024
Accepted for publication 13 March 2025
Published 6 June 2025 Volume 2025:14 Pages 555—575
DOI https://doi.org/10.2147/ITT.S494550
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 4
Editor who approved publication: Dr Sarah Wheeler
Jie Liu,1,2 Mengran Wang,1,2 Yong Zhao1,2
1State Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, People’s Republic of China; 2Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen, People’s Republic of China
Correspondence: Yong Zhao, Faculty of Synthetic Biology, Shenzhen University of Advanced Technology, Shenzhen, 518055, People’s Republic of China, Email [email protected]
Abstract: Macrophage polarization, a dynamic process crucial for immune responses and tissue homeostasis, is tightly regulated by transcription factors. Understanding the transcriptional regulation of macrophage polarization holds significant therapeutic implications for various diseases, including cancer, autoimmune disorders, and metabolic syndromes. Studies have shown that transcription factors, including signal transducer and activator of transcription (STAT), nuclear transcription factor-κB (NF-κB), peroxisome proliferator-activated receptors (PPARs), interferon regulatory factors (IRFs), BTB and CNC homology (BACH), CCAAT-enhancer binding proteins (C/EBPs), kruppel-like factors (KLFs), Cellular Myc (c-Myc), the SNAIL family, v-Maf Musculoaponeurotic Fibrosarcoma Oncogene Homolog (Maf), and hypoxia-inducible factor alpha (HIFα), are highly involved in shaping macrophage polarization. Targeting transcription factors involved in macrophage polarization may provide promising avenues for immunomodulatory therapies aimed at restoring immune homeostasis and combating pathological conditions characterized by dysregulated macrophage activation. Here, we review the intricate transcriptional networks that govern macrophage polarization, highlighting the pivotal role of transcription factors in orchestrating these processes.
Keywords: macrophages, transcription factor, macrophage polarization, inflammation
Introduction
Macrophages are vital components of the immune system, playing key roles in development, scavenging, inflammation, and pathogen defense by directly removing foreign agents and coordinating the stages of inflammation and tissue repair.1–3 In response to various environmental cues (eg, microbial products, harmful organisms, apoptotic cells, insult-related debris, and activated lymphocytes) or under different pathophysiological conditions, macrophages can adopt a variety of functional phenotypes through tightly regulated phenotypic polarization.4,5 Two well-established polarized phenotypes are commonly known as classically activated macrophages (M1 macrophages) and alternatively activated macrophages (M2 macrophages). Bacterial lipopolysaccharide (LPS), tumor necrosis factor-α (TNF-α), and interferon-gamma (IFN-γ)-induced M1 macrophage activation is characterized by a robust capacity for antigen presentation, elevated production of interleukin-12 (IL-12), IL-23, and toxic intermediates such as nitric oxide and reactive oxygen species (ROS), which together drive a polarized type I immune response. Thus, M1 macrophages are widely regarded as powerful effector cells that effectively kill microorganisms and tumor cells while producing substantial amounts of proinflammatory cytokines. In contrast, various signals (eg, IL-4, IL-10, IL-13, glucocorticoids, and immunoglobulin complexes/Toll-like receptor (TLR) ligands) induce distinct M2 functional polarization, which can attenuate inflammatory responses, induce adaptive T-helper 2 immunity, scavenge debris, and promote angiogenesis, tissue repair and remodeling. Macrophage polarization significantly influences disease outcomes. In infectious diseases, M1 polarization enhances pathogen clearance, but excessive activation can lead to chronic inflammation. Conversely, M2 macrophages resolve inflammation and promote tissue repair, but their functions can also support tumor progression and metastasis in cancer. The polarization state of macrophages directly affects their ability to recognize, phagocytose and kill pathogens in infectious diseases, while also influencing the degree and duration of inflammation in rheumatoid arthritis, inflammatory bowel disease, and other diseases. The polarization status of macrophages is closely associated with tumor growth, metastasis, and treatment response, impacting the prognosis and treatment response of many diseases and serving as a key factor in prognosis and therapy.6–8 As transcription factors play pivotal roles in orchestrating the fate and biological functions of all cells, many transcription factors are closely involved in mastering the dynamic polarization of macrophages in response to environmental cues, which has been widely studied (Figure 1). An in-depth understanding of the regulatory network of transcription factors underlying macrophage polarization would greatly help us to develop novel therapeutic approaches for inflammation-related diseases. Here, we review the current understanding of how transcription factor-mediated regulation of macrophage polarization occurs in physiological and pathological contexts. The transcription factors discussed in the present review include STAT, NF-κB, PPARs, IRFs, BACH, C/EBPs, KLFs, c-Myc, the SNAIL family, Maf, HIFα, and others.
The STAT Family
STAT family members are pivotal transcription factors that regulate macrophage polarization and key macrophage functions such as inflammation, tissue repair, and tumor progression (Figure 2). STATs shape macrophage phenotypes in response to cytokines and growth factors by activating diverse signaling pathways, critically impacting the functional programming of tumor-associated macrophages (TAMs).9 STAT1 is a critical mediator of M1 macrophage polarization driven by IFN-γ, forming homodimers that bind to cis elements known as IFN-γ-activated sequences in the promoters of genes such as nitric oxide synthase 2 (NOS2), the major histocompatibility complex (MHC) class II transactivator and IL-12, crucial for M1’s proinflammatory responses and pathogen defense. In vivo, this can be driven by IFN-γ derived from T cells or innate lymphocyte-like natural killer cells, playing a critical role in defending against intracellular pathogens, such as viruses, Listeria monocytogenes, and Mycobacterium tuberculosis.10 Conversely, STAT6 serves as the key transcription factor in the polarization of M2 macrophages mediated by IL-4 or IL-13. STAT6 initiates the transcription of genes characteristic of M2 polarization, such as chitinase-like molecule 3(YM1/Chi3l3), resistance-like α (Retnlα/Fizz1) and mannose receptor C type 1(Mrc1), which support tissue repair, immune regulation, and macrophage polarization toward the TAM phenotype, contributing to tumor progression and tissue remodeling. Besides, IL-4-activated STAT6 mediates transcriptional repression in alternative macrophage polarization by inhibiting p300 and RNA polymerase II binding, reducing enhancer RNA expression, and overlapping with the NF-κB p65 cistrome, leading to reduced inflammatory responses.11,12 STAT3 is activated by various cytokines, including IL-6 and IL-10, and has a context-dependent role in macrophage polarization, supporting both proinflammatory M1 and anti-inflammatory M2 responses. In the presence of IL-6, STAT3 activation can lead to the expression of genes involved in inflammation, while IL-10-induced STAT3 activation promotes angiogenesis, tumor progression and tissue repair functions. This dual role allows STAT3 to finely tune the immune response according to the specific needs of the tissue environment, making it a promising target for altering the M1/M2 ratio and effectively controlling disease progression.13,14 STAT2 primarily mediates IFN-α/β signaling, which is essential for antiviral defenses. STAT2 activation contributes to the antiviral state and supports the expression of certain M1-related genes, but its direct role in macrophage polarization is less prominent.15 STAT4, which is activated by IL-12 and IL-23, plays a significant role in driving Th1 responses and promoting M1 macrophage polarization.16 STAT5a and STAT5b play a role in macrophage survival and proliferation, but few studies have investigated macrophage polarization.17 Overall, STAT family members play distinct and overlapping roles in regulating macrophage plasticity, each contributing to the complexity and specificity of the immune response.
NF-κB
NF-κB plays a crucial role as a transcriptional regulator in the M1 macrophage polarization induced by TLR4. The activation of NF-κB begins with the phosphorylation of inhibitor of kappa B (IκB) in response to microenvironmental stimuli such as LPS. Following IκB phosphorylation and subsequent degradation, NF-κB translocates to the nucleus, where it binds to specific DNA sequences and regulates the expression of proinflammatory cytokines.18,19 NF-κB has two subunits: p65/p50, which promotes a proinflammatory M1 response, and p50/p50, which is associated with an anti-inflammatory M2 response (Figure 3). When NF-κB is activated in the p65/p50 form, the production of proinflammatory cytokines significantly increases, resulting in an M1 phenotype. However, p50 lacks a transactivation domain. When the p50/p50 homodimer binds to specific DNA sequences in various promoters, it does not activate transcription and blocks the access of heterodimers such as p65/p50, thereby inhibiting the production of proinflammatory cytokines and resolving inflammation.20–22 Subsequent studies have shown that p50 also plays a crucial role in directly promoting IL-10 transcription. Upon LPS stimulation, the NF-κB binding site on the IL-10 proximal promoter facilitates p50 homodimerization and interaction with the transcriptional coactivator cAMP responsive element-binding protein (CREB), resulting in transcriptional activation.23 In addition, upon activation of IL-10R, IL-10 subsequently induces STAT3 activation, p50/p50 dimer formation and M2 polarization.22,24 Consistently, p50-deficient mice also exhibit heightened M1-driven inflammation and impaired ability to initiate M2-polarized inflammatory responses in allergic and helminth-driven conditions. Importantly, when macrophages are exposed long-term to microbial components, such as bacterial LPS, they can also induce a shift in the macrophage phenotype and produce M2-type cytokines via p50/p50, highlighting a mechanism that regulates macrophage plasticity and facilitates rapid responses to infections and changes in the microenvironment.21,24,25 In the tumor microenvironment, upon activation by IL-1, IL-1R recruits the adaptor protein myeloid differentiation primary response gene 88 (MyD88), which in turn activates the IκB kinase complex, leading to NF-κB activation. This pathway is essential for sustaining the immunosuppressive phenotype of TAMs, which is marked by elevated levels of IL-10, TNF-α, and arginase 1 (Arg1), along with reduced levels of IL-12, MHC II, and NOS2 in ovarian cancer.26 Conversely, IL-10 from TAMs inhibits IL-12 production by preventing NF-κB activation, thereby promoting tumor survival, but this effect is reversed by blocking IL-10, which restores IL-12 production in a fibrosarcoma mouse model.27 In liver cancer, NF-κB inhibitor JSH-23 regulates macrophage polarization by inhibiting NF-κB in M1-like TAMs, reducing cell proliferation and promoting apoptosis.28 Similarly, BAY-11-7082 in Guillain-Barré syndrome reduces M1 macrophages and pro-inflammatory cytokines, alleviating clinical symptoms.29 Therefore, NF-κB activation may have differential effects on these distinct populations of macrophage and play complex roles in macrophage polarization, which provide a therapeutic strategy for cancer and immune-mediated diseases.
The Nuclear Receptor PPARs
PPARs are structurally conserved members of the ligand-activated nuclear hormone receptor superfamily and include PPARα, PPARδ, and PPARγ. Research has demonstrated that PPARs transcriptionally regulate macrophage activation in various health and disease states, including obesity, insulin resistance, cardiovascular disease, liver fibrosis, nonalcoholic fatty liver disease and Chagas disease30–41 (Table 1). PPARγ, which is highly expressed in adipose tissue and macrophages, is a key regulator of lipid uptake and adipogenesis. PPARγ activation suppresses M1 markers such as NOS2, IL-1β, TNFα, IL-6, and monocyte chemotactic protein 1 through ligand-responsive interference with the STAT-1, AP-1 and NF-κB pathways.42,43 PPARγ expression levels are positively associated with M2 marker expression (IL-10, Mrc1, and alternative macrophage activation-associated CC chemokine 1) in human carotid atherosclerotic lesions.44 PPARγ transcriptional activity is induced by IL-4, IL-13, insulin and the C peptide through PI3-kinase and primes macrophages for M2-induced tissue repair. Additionally, PPARγ also binds to DNA and recruits P300 and RAD21, creating a permissive chromatin environment that promotes STAT6 and RNA polymerase II binding. This enables transcriptional memory and robust gene expression upon IL-4 re-stimulation, regulating extracellular matrix remodeling during muscle regeneration in a mouse injury model.44–47 Macrophages lacking PPARγ are resistant to M2 polarization, which promotes insulin insensitivity and anti-inflammatory effects and leads to increased pulmonary collagen deposition following influenza infection.48–51 PPARα plays a key role in fatty acid oxidation and energy metabolism. Upon activation, PPARα enhances transcriptional activity, promoting M2 macrophage polarization and increasing the expression of M2 marker genes such as Arg1, YM1, Mrc1, and transforming growth factor-β (TGF-β), while simultaneously inhibiting the expression of NOS2 and proinflammatory cytokines in murine macrophages. After stroke, microglia/macrophages shift from the neuroprotective M2 phenotype to the neurotoxic M1 phenotype, influencing brain injury and repair. The protective role of PPARα in acute ischemic stroke was confirmed in PPARα-deficient mice, where PPARα activation promoted M2 polarization of microglia/macrophages, thereby enhancing neuronal survival and improving recovery.31,52,53 PPARδ is expressed ubiquitously and is involved in lipid metabolism, insulin resistance, wound healing, and inflammation.54–56 PPARδ expression in macrophages is driven by STAT6 binding to its promoter, induced by Th2 cytokines IL-13 and IL-4 produced by adipocytes, facilitating alternative activation. The ablation of PPARδ prevents macrophages from transitioning to the M2 phenotype, leading to inflammation and metabolic dysregulation in adipocytes reducing the expression of glucose transporter 4 and hindering insulin-stimulated glucose uptake. Myeloid-specific PPARδ-deficient mice on a high-fat diet also showed elevated M1 markers and reduced M2 markers in white adipose tissue and liver, leading to a decrease in insulin sensitivity. Additionally, transferring PPARδ-deficient bone marrow into wild-type mice inhibited the alternative activation of macrophages, causing hepatic dysfunction and systemic insulin resistance.57,58 Overall, PPARs are crucial regulators of macrophage polarization, influencing the immune response and inflammation through their transcriptional activities. Modulating PPAR function offers promising therapeutic potential for managing conditions such as cardiovascular diseases, insulin resistance, obesity, and infectious diseases by restoring the balance between proinflammatory and anti-inflammatory macrophage activities.
![]() |
Table 1 Summary of PPAR’s Roles in Macrophage Polarization |
IRFs
IRFs, originally identified as regulators of type I interferon expression and signaling, are also recognized as crucial mediators involved in macrophage polarization. The IRF family comprises nine members (IRF1-9) that regulate both the development and activation of immune cells.59 Among the IRF family members, only seven (IRF1-5 and IRF-8,9) are involved in the differentiation and polarization of macrophages. Notably, IRF1, IRF5, IRF8 and IRF9 play crucial roles in the proinflammatory polarization of macrophages, whereas IRF3 and IRF4 are key contributors to M2 macrophage polarization60–66 (Figure 4). IRF4, which is induced by IL-4 via jumonji domain-containing protein 3(JMJD3), is crucial for the M2 macrophage response to helminth infection. Both IRF4 and the histone demethylase JMJD3 play crucial roles in IL-4-induced M2 macrophage polarization by binding to specific promoter regions of M2-specific genes, including CD206, Arg1, Fizz1, and YM1.67 Furthermore, IL-4 modulates a subset of M2 phenotype-associated genes, including MHC-II genes, Class II transactivator genes, IL-1R antagonist gene, and cytochrome P450 1B1 genes, which are disrupted in IRF4-deficient macrophages.68 Additionally, the protein levels of IRF4, together with those of STAT3 and phosphorylated STAT3, were found to be elevated in IL-6-induced monocyte-derived M2 macrophages in vitro.69 Interestingly, the competition between IRF4 and IRF5 for binding to the middle region of MyD88 plays a crucial role in determining macrophage polarization toward either the M1 or M2 phenotype.70 IRF5 is central to macrophage polarization toward the proinflammatory M1 phenotype, mediating TLR-dependent induction of key inflammatory cytokines, including TNF, IL-6, and IL-12.71 When IRF5 is co-expressed with IκB kinase β, a kinase responsible for phosphorylating and activating IRF5, it drives the polarization of TAMs toward the M1 phenotype, effectively suppressing tumor development in models of advanced-stage ovarian cancer, metastatic melanoma, and glioblastoma.72 IRF9 lacks known transcriptional activity independently and is recognized primarily as a subunit of interferon-stimulated gene factor 3 or as a complex with STAT1 or STAT2.73 Studies have demonstrated that IRF9 deficiency provides significant protection against colon inflammation induced by dextran sodium sulfate. Besides, IRF9 promotes the progression of rheumatoid arthritis by regulating macrophage polarization through the proteasome 20S alpha 5 signaling pathway. IRF9 and proteasome subunit alpha 5 were significantly elevated in RA patients, M1/M2 ratio was also increased. Knockdown of IRF9 in RAW264.7 cells suppresses proteasome 20S alpha 5 expression, reduces the M1/M2 ratio, and decreases the secretion of pro-inflammatory factors. IRF9 is also involved in regulating type I interferon signaling and contributes to the promotion of M1 macrophage polarization.74–76 Overall, IRFs represent critical regulators of macrophage polarization, influencing the balance between proinflammatory and anti-inflammatory responses in health and diseases. Further elucidating the specific roles and mechanisms of IRFs in macrophage polarization will pave the way for novel therapeutic strategies aimed at restoring immune homeostasis.
BACH Proteins
BACH proteins, including BACH1 and BACH2, are transcriptional repressors belonging to the basic region leucine zipper (bZIP) transcription factor family and play widespread roles in governing the development and function of both the innate and adaptive immune systems.77 BACH1 has an important function in inflammatory macrophage differentiation.78 BACH1-deficient mice have a lifespan comparable to that of wild-type mice but they show resistance to inflammation by upregulating the expression of heme oxygenase-1 across multiple scenarios, including spinal cord injury, ischemia/reperfusion myocardial injury, hypoxic lung injury, LPS-induced hepatic injury, atherosclerosis and colitis triggered by 2,4,6-trinitrobenzene sulfonic acid.79–86 Peritoneal macrophages derived from BACH1‑deficient animals exhibited elevated levels of genes linked to M2 macrophage differentiation, such as Arg1, CD206, Fizz1, and YM1, due to the release of transcriptional activity inhibition. Additionally, BACH1-deficient mice show partial resistance to the onset of experimental autoimmune encephalomyelitis due to a reduced antigen-presentation capacity, which results from decreased proportions of macrophages and dendritic cells that express MHC-II.86 Similarly, BACH2-deficient alveolar macrophages display defects in phagocytosis and cholesterol handling, and further studies have identified altered gene expression related to chemotaxis, lipid metabolism, and alternative M2 macrophage activation, including increased levels of YM1, Arg1, and the M2 regulator IRF4. These changes lead to impaired lipid processing and the accumulation of surfactant proteins, contributing to the development of pulmonary alveolar proteinosis. In addition, BACH2-deficient peritoneal macrophages also exhibit increased YM1 expression upon stimulation with IL-4.87 Overall, accumulating evidence suggests that BACH factors are involved in macrophage polarization. Further work is needed to understand the precise functions of BACH factors in human immunity and the modification of immune function in patients with allergies, chronic infections, autoimmune diseases, and cancer.
C/EBPs
C/EBPs are a family of bZIP transcription factors that play vital roles in myeloid development and macrophage activation.88 C/EBPα is known primarily for its role in the differentiation of myeloid cells. It facilitates the development of monocytes and macrophages from hematopoietic progenitors. In macrophages, C/EBPα is associated with the promotion of anti-inflammatory and homeostatic functions. Macrophage-specific deficiency of C/EBPα protects against high-fat-induced inflammation in skeletal muscle. Moreover, C/EBPα-deficient macrophages exhibit a blunted response to cytokine-induced expression of both M1 and M2 macrophage markers, indicating that C/EBPα regulates both M1 and M2 polarization.89 Previous studies have indicated that the transcription factors C/EBPβ and C/EBPδ are involved in TLR-induced M1 activation through MyD88 and L-1R-associated kinase 4. In macrophages deficient in MyD88 or IL-1R-associated kinase 4, the expression of C/EBPβ and C/EBPδ is diminished following LPS treatment. Furthermore, the absence of both C/EBPβ and C/EBPδ leads to impaired induction of proinflammatory cytokines in response to several TLR ligands.90 Conversely, another study demonstrated that C/EBPβ interacts with STAT factors specifically regulates M2-associated genes involved in tissue repair, such as Arg1, IL-10, and Msr1, when its expression is transcriptionally activated by the CREB binding protein, another transcription factor from the bZIP family.91,92 Importantly, although CREB is essential for LPS-induced expression of C/EBPβ, only M2-associated genes were suppressed in the mutant mice, while M1-associated genes, such as NOS2 and IL-12, remained unaffected, suggesting that the M2 program seems to be specifically sensitive to C/EBPβ levels.92,93 Additionally, C/EBPβ also regulates arginine metabolism by activating Arg1 expression in macrophages via STAT6 and C/EBPβ binding sites in response to IL-4, which may provide a foundation for developing strategies to modulate arginase expression in Th2 cytokine-predominant diseases.94 However, the role of C/EBPs in regulating macrophage polarization and the molecular mechanisms involved in atherosclerosis, autoimmune diseases, and cancer remains unclear. Therefore, more studies, such as the development of relevant in vivo and in vitro pathological models, single-cell transcriptomic analysis of clinical samples, and gene editing, are needed to reveal the roles of C/EBPs in the cross-linking networks that modulate macrophage function in various disease contexts.
KLFs
KLFs are a subfamily of zinc-finger transcription factors involved in macrophage polarization in response to various stimuli.95,96 KLF4 plays a crucial role in M2 macrophage polarization and enhances the expression of genes associated with tissue repair, immune regulation, and resolution of inflammation. KLF4 cross-talk with STAT6 signaling impairs NF-κB activity by sequestering essential coactivators, p300 and p300/CBP-associated factors, leading to the induction of M2-related genetic reprogramming and effectively preventing M1 polarization. KLF4-deficient macrophages exhibit impaired expression of typical M2 markers after IL-4 or IL-13 stimulation in vitro. Additionally, these macrophages exhibit increased proinflammatory gene expression, enhanced bactericidal activity, and altered metabolic responses.97 IL-4 induces STAT6 phosphorylation, which promotes KLF4 gene expression. In turn, KLF4 cooperates with STAT6 to enhance the M2 gene profile through the activation of monocyte chemotactic protein-induced protein expression and the upregulation of Arg1 and Fizz1 expression.98 Furthermore, KLF4 deficiency in macrophages infiltrating the kidney enhances M1 polarization, exacerbating glomerular matrix deposition and tubular epithelial damage in a murine model of chronic kidney disease.99 However, other studies have shown that KLF4 is significantly induced in macrophages by pro-inflammatory stimuli such as IFN-γ, LPS, or TNF-α, while its expression is decreased by TGF-β. Overexpression of KLF4 enhances M1 polarization by activating STAT1 and inducing iNOS, which leads to increased production of inflammatory cytokines and tissue damage. This effect is further amplified when KLF4 is overexpressed in the presence of IFN-γ and LPS.100,101 A potential explanation for KLF4’s dual role in promoting both M1 and M2 polarization may lie in the use of different cell types across studies. The studies showing KLF4 promoting M1 polarization often utilized immortalized cell lines (eg, J774a, THP-1, RAW264.7), while those demonstrating its role in M2 polarization typically used primary macrophage cell lines. Additionally, variations in LPS concentration (ranging from 10 ng/mL to 1000 ng/mL) and its source (eg, E. coli, Porphyromonas gingivalis, and Salmonella enterica) could also account for the divergent results observed. Recent research has shown that KLF14 also regulates glycolysis and immune function in macrophages. KLF14 expression is elevated in septic patients, and its deletion results in significantly higher mortality in lethal models of murine endotoxemia and sepsis via the transcription of hexokinase 2(HK2) to promote glycolysis and the release of inflammatory cytokines.102 KLF6 is another crucial member of the KLF family that significantly influences macrophage polarization. KLF6 expression is strongly induced by LPS and IFN-γ and is essential for LPS- and IFN-γ-induced macrophage polarization to the M1-like phenotype, working in concert with NF-κB signaling. It suppresses anti-inflammatory gene expression by downregulating PPARγ in macrophages, both in vitro and in KLF6-deficient mice.103 KLF6 overexpression elevates inducible HIF-1α expression in macrophages, whereas KLF6 deficiency reduces it.104 Notably, overexpressing oxygen-stable HIF-1α in KLF6-deficient macrophages restores proinflammatory and glycolytic gene expression, enabling a coordinated inflammatory and hypoxic gene program for an effective immune response104 (Figure 5).
c-Myc
The c-Myc transcription factor is crucial for regulating M2 and TAM activation.105 c-Myc expression is primarily limited to the M2 phenotype and is nearly undetectable in M0 and M1 macrophages in human cells. The study demonstrated that c-Myc expression is induced in human M2-like macrophages in response to various stimuli, including IL-4, IL-10, IL-13, and TGF-β. However, murine M0 macrophages express small but detectable levels of c-Myc.105,106 c-Myc expression has been observed in certain types of human TAMs in vivo, which exhibit an M2-like macrophage activation status. Studies with mouse bone marrow-derived macrophages (BMDMs) cultured in conditioned medium from Hepa1-6 cell have indicated that Wnt/β-catenin signaling mediates M2 macrophage polarization through c-Myc-mediated expression of mannose receptor (MR), Arg1, and YM1, which supports the progression of hepatocellular carcinoma.107 In addition, in a coculture model of human monocytes and hepatocellular carcinoma cells, IL-12 inhibited the transcriptional activity of STAT3 and the expression of c-Myc in monocytes, promoting M1 polarization, affecting T cell infiltration and suppressing hepatocellular carcinoma growth.108 Furthermore, the inhibition or deletion of c-Myc reduces the expression of pro-angiogenic molecules (eg, vascular endothelial growth factor (VEGF), matrix metalloproteinase 9, and HIF-1α) and diminishes tumor growth 109 Besides, c-Myc serves as a transcriptional activator by binding to enhancer box sequences in the promoter regions of its target genes. It directly induces the expression of key M2-associated genes, including arachidonate 15-lipoxygenase, Mrc1, scavenger receptor class B member 1, STAT6, and PPARγ, thereby driving M2 macrophage polarization. In response to IL-4, STAT6 and PPARγ further enhance M2 activation by directly binding to the promoters of M2 target genes, such as CD209 and CD36.105,109 Thus, c-Myc is a critical regulator of macrophage polarization, particularly in promoting the M2 phenotype and supporting tumor progression (Figure 6). Its inhibition or deletion in macrophages suppresses protumor gene expression and reduces tumor growth, making it a potential therapeutic target for modulating macrophage activity in cancer. In addition, future research could explore the potential of overexpressing c-Myc in M0 or M1 macrophages to induce M2 polarization, offering a promising strategy for the treatment of autoimmune and inflammatory diseases.
SNAIL Family
The transcription factor SNAIL may play a regulatory role in macrophage polarization. In human THP-1 macrophages, SNAIL expression induced by TGF-β, through the transcriptional activation of PI3K/AKT and Smad2/3 signaling pathways, drives M2-like macrophage polarization. SNAIL overexpression promotes M2-like differentiation by reducing the expression of proinflammatory cytokines and enhancing the expression of M2-specific markers. Conversely, SNAIL knockdown favors M1 polarization by increasing proinflammatory cytokine production and blocking TGF-β-induced M2 polarization.110 Moreover, Jagged1-mediated myeloid Notch1 signaling regulates NOD-like receptor family pyrin domain containing 3(NLRP3) function and promotes the activation of heat shock transcription factor 1, which subsequently induces the expression of SNAIL. Furthermore, SNAIL increases thioredoxin-1 expression and reduces thioredoxin-interacting protein, NLRP3/caspase-1, and ROS production, which in turn suppresses NLRP3 function and hepatocellular apoptosis, resulting in the reduction of ischemia/reperfusion-induced liver injury. Ablation of myeloid SNAIL expression significantly increased apoptotic signals regulating kinase 1 activation by transcriptionally regulates the thioredoxin 1/ thioredoxin-interacting protein and thioredoxin 1/ apoptosis signal-regulating kinase 1 complexes, leading to enhanced NLRP3 inflammasome activation and ROS-induced hepatocellular apoptosis in a mouse model of ischemia/reperfusion-induced liver injury.111,112 Studies have shown that SNAIL also regulates M1 polarization. In the cecal ligation and puncture-induced sepsis models, high glucose and LPS stimulation promoted M1 macrophage polarization and reduced miR-3061 levels, which were associated with increased SNAIL expression in RAW264.7 cells. Furthermore, overexpression of miR-3061 inhibited SNAIL expression, thereby suppressing M1 macrophage polarization and the production of inflammatory cytokines, which ultimately exacerbated sepsis-induced intestinal injury.113 However, there are few reports on the direct involvement of SNAIL in regulating macrophage polarization in TAMs. Instead, SNAIL influences macrophage polarization indirectly through its abnormal expression in tumor cells.114–118 Overall, additional research is required to fully understand the mechanisms through which SNAIL directly regulates macrophage polarization in TAMs. Furthermore, there is a need to develop effective strategies, such as small-molecule inhibitors, cell therapies, or gene editing, to modulate its activity in different disease contexts.
Maf
The Maf transcription factor family consists of several members, including MafA, MafB, c-Maf, Neuroretina Leucine zipper protein 11, MafF12, MafG13, and MafK. Among them, MafB and c-Maf are recognized as significant factors that regulate macrophage differentiation and polarization in both mouse and human models.119–121 In BMDMs from adult wild-type mice, MafB expression is stimulated by IL-10 or IL-4/IL-13, while it is suppressed by LPS or granulocyte-macrophage colony-stimulating factor (GM-CSF).120 Immunostaining analysis revealed strong MafB expression in CD204+ and CD68+ TAMs at stage 3 of human lung cancer. Furthermore, in MafB-GFP knock-in heterozygous mice with Lewis lung carcinoma, MafB+ macrophages significantly expressed protumor factors such as IL-10, Arg1 and TNFα.119 Additionally, increased MafB expression in TAMs was noted in a murine model of breast cancer.122 These findings suggest that MafB expression can be a potential marker of TAMs in malignant tumor. In human primary macrophages, IL-10-induced MafB activated the STAT3 signaling pathway, increasing the levels of the matrix metalloproteinase 9 and IL-7R genes, which help resolve inflammation and restore tissue integrity.123 Consistently, wound healing was significantly delayed in MafB-deficient mice, as MafB deficiency downregulated the expression of C-C motif chemokine ligand 12, C-C motif chemokine ligand 2, and Arg1, leading to reduced macrophage recruitment and impaired tissue repair.124 Conversely, c-Maf expression is stimulated by IL-10 and suppressed by IL-4 in combination with IL-13 or GM-CSF in BMDMs.120 However, recent research has shown that M2 macrophages induced by IL-4 and IL-13 also express high levels of c-Maf, which directly transcriptionally induces the expression of colony-stimulating factor 1 receptor, thereby regulating the expression of M2-related genes (IL-12, IL-6, IL-10, IL-1β, Arg1, TGF-β, VEGF, IRF4, and the chemokine C-C-motif receptor 2).125 In human non-small cell lung carcinoma, TAMs express c-Maf, which facilitates M2-mediated T-cell suppression and tumor progression by regulating M2-related genes in vivo. Knockout of c-Maf in macrophages diminished the suppression of effector T cells, decreased the expression of CD206, and increased MHC-II levels. Consequently, the absence of c-Maf in myeloid cells results in delayed tumor growth compared with that in mice with c-Maf–competent myeloid cells.125,126 Additionally, c-Maf acts as a key regulator of the transcriptional program in perivascular macrophages. Compared to wild-type mice, high-fat diet-fed mice with c-Maf-deficient macrophages demonstrated improved metabolic outcomes, including reduced weight gain, enhanced glucose tolerance, and a decreased inflammatory cell profile in white adipose tissue.127 Overall, MafB and c-Maf are pivotal transcription factors, with both loss-of-function and gain-of-function studies demonstrating their roles in promoting M2 macrophage polarization and regulating TAMs. These findings highlight their potential as therapeutic targets, with approaches such as small molecule inhibitors, gene editing, and cell therapy showing promise in cancer and metabolic diseases. However, the widespread expression of MafB and c-Maf in various cell types, as well as the unique immunogenicity associated with macrophages, must also be carefully considered to ensure the safety and efficacy in therapeutic applications.
HIFα
HIFα, which includes the isoforms HIF-1α and HIF-2α, is a master transcriptional regulator of the cellular response to hypoxia and plays a crucial role in macrophage polarization128,129 (Figure 7). Notably, HIFα stabilization in macrophages can occur in an oxygen-independent manner. In the presence of different pathogens, the activation of HIF-1α expression has been observed in macrophages cultured under normoxic conditions.130 LPS/IFNγ rapidly induces HIF-1α expression in macrophages, thereby promoting NOS production independently of hypoxia. In contrast, HIF-2α mRNA responds more slowly to IL-4/IL-13, which induces Arg1 expression via the JAK/STAT6 pathway. However, IFNγ and LPS can suppress IL-4/STAT6 signaling by inducing suppressor of cytokine signaling proteins, which ultimately inhibits HIF-2α synthesis. Under low IFNγ conditions, HIF-2α promotes the expression of arginase 1, thereby reducing NO production. In contrast, under high IFNγ conditions, HIF-2α levels decrease, leading to the activation of iNOS, which produces NO.131,132 Furthermore, the overexpression of HIF-1α in macrophages increases the expression of M1 markers, whereas HIF-2α promotes M2 polarization through the expression of markers such as Arg1.132,133 Despite these roles, deletion of myeloid HIF-1α or HIF-2α does not affect macrophage polarization or function during skeletal muscle regeneration induced by sterile tissue damage.134 However, HIF-1α deficiency attenuates pro-inflammatory pathways and impairs M1 macrophage polarization in stenotic artery macrophages. Interestingly, myeloid cell-specific knockout of HIF-1α and HIF-2α has been shown to alleviates sepsis, reducing proinflammatory cytokine production and improving survival in LPS-induced endotoxemia models.135–137 These observations underscore the plasticity of macrophages and their context-dependent roles in inflammation and repair. Loss of HIF-1α in myeloid cells downregulates iNOS and Arg1 expression, relieving T-cell proliferation suppression, which significantly reduces tumor mass and inhibits tumor progression.138 Similarly, in a murine colitis-associated colorectal cancer model, the loss of HIF-2α also reduced tumor burden and progression by impairing macrophage migration and invasion through the downregulation of macrophage colony-stimulating factor receptor and C-X-C chemokine receptor type 4 expression.139 LPS-induced HIF-1α stabilization leads to M1 polarization, highlighting the role of HIF-1α in inflammatory responses to bacteria and viruses. HIF-1α increases following Mycobacterium tuberculosis infection, increasing IL-1β production and reducing bacillary survival. In addition, HIF-1α is an essential mediator of IFN-γ-dependent control of Mycobacterium tuberculosis infection, and RNA sequencing revealed that almost half of all genes inducible by IFN-γ are regulated in HIF-1α–deficient macrophages during infection.140,141 In H1N1 virus-infected macrophages, the levels of HIF-1α mRNA and protein actually remain constant, yet its transcriptional activity increases, resulting in elevated TNF-α and IL-6 production, reduced IL-10, and heightened inflammation.142 HIF-1α promotes VEGF induction in macrophages in response to GM-CSF and low oxygen levels, whereas HIF-2α, dependent on JAK2/STAT5 signaling, induces soluble VEGF receptor 1 to neutralize VEGF as oxygen levels decrease. This indicates that HIF-1α and HIF-2α may have opposing roles in regulating VEGF signaling across different oxygen concentrations.143–145 In general, these studies indicate that the functions of HIF-1α and HIF-2α in macrophage polarization are complex, sometimes overlapping and dependent on the pathophysiological context. Although many small molecule inhibitors targeting HIF-1α and HIF-2α, such as PX-478 and PT2385, are currently used in cancer therapy, the widespread expression of these factors across various cell types makes it important to focus on identifying macrophage-specific inhibitors and drug strategies as a key direction for future research. Furthermore, gene editing-based cell therapy represents a promising targeted strategy to precisely modulate HIF-1α and HIF-2α activity in macrophages, potentially enhancing targeting specificity while minimizing side effects.
Other Transcription Factors
In addition, an increasing number of studies have revealed a broader correlation between transcription factors and macrophage polarization. Zinc fingers and homeobox 2 (zhx2) play important roles in B-cell differentiation, NK cell maturation and macrophage survival.146–148 It is also highly expressed in LPS-stimulated macrophages, where it promotes glycolysis and inflammatory responses during sepsis by binding to the promoter and enhancing the transcription of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3, a rate-limiting enzyme in glycolysis.149 Furthermore, zhx2 regulates macrophage polarization in inflammatory environments and tumor microenvironments by associating with NF-κB (p65) and binding to the IRF1 promoter, which leads to the induction of IRF1 transcription in macrophages. Deletion of zhx2 in myeloid cells suppresses LPS-driven proinflammatory polarization while promoting anti-inflammatory and protumor phenotypes induced by IL-4 and the tumor microenvironment in liver tumor models.150 Myocyte enhancer factor 2C (MEF2C) is essential for bone, neuronal, cardiac, and skeletal muscle development.151 MEF2C binding sites were the most significantly enriched motifs identified in a ChIP-seq analysis in M. tuberculosis-infected macrophages, suggesting their role in macrophage responses to infection. Indeed, MEF2C promotes M1 macrophage polarization by controlling the expression of cytokines characteristic of macrophage lineages, and myeloid-specific Mef2c-knockout mice display diminished IL-12 production and weakened Th1 responses, resulting in increased susceptibility to Listeria monocytogenes infection but protection against dextran sulfate sodium salt-induced inflammatory bowel disease.152,153 Activating transcription factor 3 (ATF3) belongs to the mammalian ATF/CREB family of transcription factors and responds to various physiological and pathological processes.154 In macrophages, ATF3 acts as a significant negative regulator of proinflammatory cytokines. Upon the activation by TLRs, ATF3 expression is rapidly induced, which subsequently recruits histone Deacetylase 1 to the ATF3/p65 complex, facilitating the deacetylation of p65 and thereby suppressing the inflammatory gene expression triggered by TLR signaling.155,156 Moreover, ATF3 is induced in both mouse and human immune cells in response to IFN-α and IFN-β. It acts as a transcriptional repressor by directly binding to the Ifnb1 promoter, playing a critical role in a negative feedback loop that regulates IFN-β expression. This regulation is achieved through modulation of both basal and inducible IFN-β levels, as well as influencing the expression of IFN-γ and downstream genes regulated by IFN signaling pathways 157 Furthermore, ATF3 overexpression enhances macrophage migration and promotes the expression of markers associated with the M2 phenotype while inhibiting markers of the M1 phenotype by upregulating tenascin through the Wnt/β-catenin signaling pathway.158,159 Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor pivotal in the cellular defense against oxidative stress.160 Increasing evidence underscores the significant role of Nrf2 in shaping the distinct metabolic and inflammatory profiles of M1 and M2 macrophages.161 Nrf2 has been shown to negatively regulate M1 macrophage polarization while promoting M2 macrophage polarization through redox control via the upregulation of heme oxygenase-1 and glutathione S-transferase expression.162,163 In response to immunological stresses, such as those induced by bacterial products and tumor growth, the NF-κB(p50)–CSF-R1–C3aR axis activates Nrf2, leading to the upregulation of heme oxygenase-1 expression in TAMs. This process promotes immunosuppression, angiogenesis, and epithelial-mesenchymal transition, which together enhance metastasis formation. Accordingly, in vivo treatment of mice bearing fibrosarcoma with brusatol, a selective Nrf2 inhibitor, significantly reduced lung metastasis formation 164 Impaired wound healing is one of the major complications of diabetes, treatment with the Nrf2 activator dimethyl fumarate markedly improved wound healing in streptozocin-induced diabetic rats by boosting antioxidant enzyme expression and reducing pro-inflammatory cytokines, while the Nrf2 inhibitor ML385 replicated diabetic effects, highlighting Nrf2 activation as a potential therapeutic strategy for diabetic wound healing.165 Moreover, Nrf2 activation by diethyl maleate suppresses inflammation by preventing RNA Pol II recruitment to the IL-6 and IL-1β gene loci without affecting p65 recruitment. This anti-inflammatory mechanism is independent of the Nrf2-binding motif and ROS levels. Additionally, Nrf2 activation plays a pivotal role in regulating macrophage polarization by suppressing M1 polarization while simultaneously enhancing the expression of M2 markers, through its interaction with several key signaling pathways, such as TGF-β/SMAD, TLR/NF-κB, JAK/STAT, Notch, PI3K/AKT, NLRP3, and MAPK. Together, these pathways create a complex network that not only drives macrophage polarization but also highlights potential therapeutic targets for addressing osteoarthritis.166,167
Conclusions and Future Directions
Transcription factors play pivotal roles in regulating macrophage polarization, which determines the functional phenotypes of immune responses. To achieve a comprehensive understanding of how macrophage polarization is regulated by transcription factors, we summarize the properties and roles of these transcription factors in determining macrophage phenotypes, as shown in Figure 8. Under physiological or pathological conditions, LPS, IFNα/β/γ, or TNFα activate STAT1/2/4, NF-κB (p65/50), IRF1/5/8/9, CEBPδ, KLF6, HIF-1α, Zhx2 and MEF2C to promote M1 macrophage polarization. Conversely, IL-4, IL-10, and IL-13, or tumor-conditioned medium, mediate STAT6, PPARα/γ/δ, IRF4, HIF-2α, NF-κB (p50/50), MafB, c-Maf, c-Myc, ATF3 and Nrf2 transcriptional activities, promoting macrophage M2 polarization. Notably, STAT3, CEBPα/β, KLF4, and SNAIL induce macrophage polarization toward the M1 or M2 phenotype in various settings, underscoring the complexity, plasticity and context dependence of macrophage polarization. Furthermore, PPARα/γ/δ, KLF4/14, NF-κB (p50/50), IRF3/4, STAT6, ATF3 and Nrf2 also inhibited M1 phenotype differentiation in the presence of IL-4, IL-6 or IL-10, whereas BACH1/2, KLF6 and NF-κB (p65/50) inhibited M2 polarization under the stimulation of LPS, IFNβ or TNFα via autocrine and paracrine mechanisms. Besides, the interactions between transcription factors and their cascading effects responding to different stimuli, such as direct interactions (eg, NF-κB with KLF4/6 and STAT1/3), competitive binding (eg, IRF4 and IRF5 competing for the middle region of MyD88), epigenetic inhibition (eg, STAT6 or Nrf2 binding to RNA polymerase II to suppress NF-κB and RNA polymerase II initiation), and cascading effects (eg, NF-κB inducing KLF4/6/14 expression, which then binds to induce M1 polarization), further highlighting the intricate regulation, cross-linking and environmental sensitivity involved in macrophage polarization. Understanding the intricate transcriptional regulatory networks governing macrophage polarization holds great promise for therapeutic interventions in inflammatory diseases. Recent advancements in single-cell RNA sequencing and genome editing technologies have offered unprecedented insights into the transcriptional landscape of macrophage polarization. For instance, a single-cell RNA sequencing map of human macrophage specification dynamics from postconceptional weeks 4–26 across 19 tissues identified a population of microglia-like cells in the skin, testis, and heart, which resemble central nervous system microglia in transcriptome, protein expression, and morphology. Additionally, a pan-cancer analysis of single myeloid cells from 210 patients across 15 cancer types reveals tissue-specific gene expression profiles in pro-angiogenic TAMs, such as secreted phosphoprotein 1(SPP1)+ in breast cancer and VCAN+ in melanoma.168,169 Integrating multi-omics data and computational modeling enables the identification of key transcription factors and their target genes involved in polarization dynamics. For example, integrating time-course proteomics, phosphoproteomics, and transcriptomics revealed metabolic shifts in M1/M2 polarization, highlighting PPARγ-induced retinoic acid and mitogen-activated protein kinase signaling as key regulators of M2 polarization. Additionally, single-cell RNA sequencing and spatial imaging identified prognostically relevant macrophage subpopulations in CRC, such as IL4I1+ macrophages in high-cell-turnover regions (favorable prognosis) and SPP1+ macrophages in hypoxic tumor areas (poor prognosis), providing potential therapeutic insights.170–173 By deciphering the molecular mechanisms that govern macrophage polarization, researchers aim to manipulate transcription factor activities to direct macrophages toward desirable phenotypes. This could be achieved through emerging immunotherapeutic approaches, such as small molecules, nucleic acid-based products, gene editing, and cell therapy, ultimately leading to novel therapeutic strategies for inflammatory disorders, infectious diseases, and cancers.174,175 In the future, exploiting transcriptional regulation to fine-tune macrophage polarization holds immense potential for personalized medicine and targeted immunotherapy, particularly in enhancing both efficacy and safety. However, further research is certainly needed to elucidate the context-specific roles of transcription factors in macrophage polarization and their immunotherapy implications for pathogenesis, which is essential to translate these findings into clinical applications, ultimately improving patient outcomes.
Abbreviations
ALOX15, Arachidonate 15-Lipoxygenase; ATF3, Activating transcription factor 3; Arg1, Arginase 1; BACH, BTB and CNC homology; BMDM, Bone marrow derived macrophage; bZIP, belonging to the basic region leucine zipper; C/EBP, CCAAT-enhancer binding protein; Chi3l3, Chitinase-like molecule 3; c-Myc, Cellular Myc; CREB, cAMP responsive element-binding protein; GM-CSF, Granulocyte-Macrophage Colony-Stimulating Factor; HK2, Hexokinase 2; HIF-1α, Hypoxia-inducible factor alpha; IκB, inhibitor of kappa B; IFN, Interferon-gamma; IL-12, Interleukin-12; IRF, Interferon regulatory factor; JMJD3, Jumonji domain-containing protein 3; KLF, Kruppel-like factor; LPS, Lipopolysaccharide; Maf, v-Maf Musculoaponeurotic Fibrosarcoma Oncogene Homolog; MEF2C, Myocyte enhancer factor 2C; MHC, Major histocompatibility complex; Mrc1, Mannose receptor C type 1; MR, Mannose receptor; MyD88, myeloid differentiation primary response gene 88; NF-Κb, Nuclear transcription factor-κB; NOS2, Nitric oxide synthase 2; Nrf2, Nuclear factor erythroid 2-related factor 2; PPAR, Peroxisome proliferator-activated receptor; PCAF, p300/CBP-associated factor; RBP-J, Recombination Signal Binding Protein for Immunoglobulin kappa J region; Retnlα, Resistance-like α; SCARB1, Scavenger Receptor Class B Member 1; SPP1, secreted phosphoprotein 1; STAT, Signal transducer and activator of transcription; TAM, Tumor-associated macrophages; TBK1, TANK-binding kinase 1; TGF-β, Transforming growth factor-β; TLR, Toll-like receptors; TNF-α, Tumor necrosis factor-α; VEGF, Vascular endothelial growth factor; zhx2, Zinc-fingers and homeoboxes 2.
Acknowledgments
We would like to thank Drs. Jianing Zhang and Na Liang for their careful review of our manuscript. This work was supported by grants from the National Natural Science Foundation of China for Key Program (32330037, Y.Z.), the National Key Research and Development Program of China (2023YFA0915000, Y.Z.), Shenzhen Medical Research Fund (B2302030, Y.Z.).
Disclosure
The authors declare no competing interests.
References
1. Watanabe S, Alexander M, Misharin AV, Budinger GRS. The role of macrophages in the resolution of inflammation. J Clin Invest. 2019;129(7):2619–2628. doi:10.1172/JCI124615
2. Christofides A, Strauss L, Yeo A, Cao C, Charest A, Boussiotis VA. The complex role of tumor-infiltrating macrophages. Nat Immunol. 2022;23(8):1148–1156. doi:10.1038/s41590-022-01267-2
3. Mantovani A, Allavena P, Marchesi F, Garlanda C. Macrophages as tools and targets in cancer therapy. Nat Rev Drug Discov. 2022;21(11):799–820. doi:10.1038/s41573-022-00520-5
4. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol. 2017;14(7):399–416. doi:10.1038/nrclinonc.2016.217
5. DeNardo DG, Ruffell B. Macrophages as regulators of tumour immunity and immunotherapy. Nat Rev Immunol. 2019;19(6):369–382. doi:10.1038/s41577-019-0127-6
6. Murray PJ. Macrophage Polarization. Annu Rev Physiol. 2017;79:541–566. doi:10.1146/annurev-physiol-022516-034339
7. Cassetta L, Cassol E, Poli G. Macrophage polarization in health and disease. ScientificWorldJournal. 2011;11:2391–2402. doi:10.1100/2011/213962
8. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. doi:10.1016/j.ejphar.2020.173090
9. Ma B, Yang Y, Li Z, et al. Modular bioinformatics analysis demonstrates that a Toll‑like receptor signaling pathway is involved in the regulation of macrophage polarization. Mol Med Rep. 2018;18(5):4313–4320. doi:10.3892/mmr.2018.9486
10. Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J Immunol. 2005;174(8):4880–4891. doi:10.4049/jimmunol.174.8.4880
11. Goenka S, Kaplan MH. Transcriptional regulation by STAT6. Immunol Res. 2011;50(1):87–96. doi:10.1007/s12026-011-8205-2
12. Czimmerer Z, Daniel B, Horvath A, et al. The transcription factor STAT6 mediates direct repression of inflammatory enhancers and limits activation of alternatively polarized macrophages. Immunity. 2018;48(1):75–90.e76. doi:10.1016/j.immuni.2017.12.010
13. Solís-Martínez R, Cancino-Marentes M, Hernández-Flores G, et al. Regulation of immunophenotype modulation of monocytes-macrophages from M1 into M2 by prostate cancer cell-culture supernatant via transcription factor STAT3. Immunol Letters. 2018;196:S016524781730367X.
14. Xia T, Zhang M, Lei W, et al. Advances in the role of STAT3 in macrophage polarization. Front Immunol. 2023;14.
15. Gopal R, Lee B, Mchugh KJ, et al. STAT2 signaling regulates macrophage phenotype during influenza and bacterial super-infection. Front Immunol. 2018;9. doi:10.3389/fimmu.2018.02151
16. Blum A, Setiawan T, Hang L, Stoyanoff K, Weinstock JV. Interleukin-12 (IL-12) and IL-23 induction of substance p synthesis in murine T cells and macrophages is subject to IL-10 and transforming growth factor beta regulation. Infection Immun. 2008;76(8):3651–3656. doi:10.1128/IAI.00358-08
17. Feldman GM, Rosenthal LA, Liu X, et al. STAT5A-deficient mice demonstrate a defect in granulocyte-macrophage colony-stimulating factor-induced proliferation and gene expression. Blood. 1997;90(5):1768–1776. doi:10.1182/blood.V90.5.1768
18. Baker RG, Hayden MS, Ghosh S. NF-κB, inflammation, and metabolic disease. Cell Metab. 2011;13(1):11–22. doi:10.1016/j.cmet.2010.12.008
19. Zhu HT, Bian C, Yuan JC, et al. Curcumin attenuates acute inflammatory injury by inhibiting the TLR4/MyD88/NF-κB signaling pathway in experimental traumatic brain injury. J Neuroinfl. 2014;11:59. doi:10.1186/1742-2094-11-59
20. Ziegler-Heitbrock L. The p50-homodimer mechanism in tolerance to LPS. J Endotoxin Res. 2001;7(3):219–222. doi:10.1177/09680519010070030401
21. Porta C, Rimoldi M, Raes G, et al. Tolerance and M2 (alternative) macrophage polarization are related processes orchestrated by p50 nuclear factor κB. Proc Nat Acad Sci. 2009;106(35):14978–14983. doi:10.1073/pnas.0809784106
22. Chen S, Saeed AF, Liu Q, et al. Macrophages in immunoregulation and therapeutics. Signal Transduction Targeted Ther. 2023;8(1):207. doi:10.1038/s41392-023-01452-1
23. Cao S, Zhang X, Edwards JP, Mosser DM. NF-κB1 (p50) homodimers differentially regulate pro-and anti-inflammatory cytokines in macrophages. J Biol Chem. 2006;281(36):26041–26050. doi:10.1074/jbc.M602222200
24. Miao X, Leng X, Zhang Q. The current state of nanoparticle-induced macrophage polarization and reprogramming research. Int J Mol Sci. 2017;18(2):336. doi:10.3390/ijms18020336
25. Igor M, Yuri M. current concept and update of the macrophage plasticity concept: intracellular mechanisms of reprogramming and M3 macrophage “Switch” phenotype. BioMed Res Int. 2015;2015:341308. doi:10.1155/2015/341308
26. Hagemann T, Lawrence T, McNeish I, et al. “Re-educating” tumor-associated macrophages by targeting NF-kappaB. J Exp Med. 2008;205(6):1261–1268. doi:10.1084/jem.20080108
27. Sica A, Saccani A, Bottazzi B, et al. Autocrine production of IL-10 mediates defective IL-12 production and NF-kappa B activation in tumor-associated macrophages. J Immunol. 2000;164(2):762–767. doi:10.4049/jimmunol.164.2.762
28. Sharen G, Cheng H, Hu X, Miao J, Zhao D. M1‑like tumor‑associated macrophages enhance proliferation and anti‑apoptotic ability of liver cancer cells via activating the NF‑κB signaling pathway. Mol Med Rep. 2022;26(5):1–10. doi:10.3892/mmr.2022.12847
29. Shen D, Chu F, Lang Y, et al. Nuclear factor kappa B inhibitor suppresses experimental autoimmune neuritis in mice via declining macrophages polarization to M1 type. Clin Exp Immunol. 2021;206(1):110–117. doi:10.1111/cei.13637
30. Chawla A. Control of macrophage activation and function by PPARs. Circ Res. 2010;106(10):1559–1569. doi:10.1161/CIRCRESAHA.110.216523
31. Penas F, Mirkin GA, Vera M, et al. Treatment in vitro with PPARα and PPARγ ligands drives M1-to-M2 polarization of macrophages from T. cruzi-infected mice. Biochim Biophys Acta. 2015;1852(5):893–904. doi:10.1016/j.bbadis.2014.12.019
32. Luo W, Xu Q, Wang Q, Wu H, Hua J. Effect of modulation of PPAR-γ activity on Kupffer cells M1/M2 polarization in the development of non-alcoholic fatty liver disease. Scientific Rep. 2017;7(1):44612. doi:10.1038/srep44612
33. Chen Q, Bao L, Lv L, et al. Schisandrin B regulates macrophage polarization and alleviates liver fibrosis via activation of PPARγ. Annals Transl Med. 2021;9(19):1500. doi:10.21037/atm-21-4602
34. Chen M, Lin W, Ye R, Yi J, Zhao Z. PPARβ/δ agonist alleviates diabetic osteoporosis via regulating M1/M2 macrophage polarization. Front Cell Dev Biol. 2021;9:753194. doi:10.3389/fcell.2021.753194
35. Li X-Y, Ji P-X, Ni -X-X, et al. Regulation of PPAR-γ activity in lipid-laden hepatocytes affects macrophage polarization and inflammation in nonalcoholic fatty liver disease. World J Hepatol. 2022;14(7):1365. doi:10.4254/wjh.v14.i7.1365
36. Chang HY, Lee H-N, Kim W, Surh Y-J. Docosahexaenoic acid induces M2 macrophage polarization through peroxisome proliferator-activated receptor γ activation. Life Sci. 2015;120:39–47. doi:10.1016/j.lfs.2014.10.014
37. Nelson VL, Nguyen HC, Garcìa-Cañaveras JC, et al. PPARγ is a nexus controlling alternative activation of macrophages via glutamine metabolism. Genes Dev. 2018;32(15–16):1035–1044. doi:10.1101/gad.312355.118
38. Mirza RE, Fang MM, Novak ML, et al. Macrophage PPARγ and impaired wound healing in type 2 diabetes. J Pathol. 2015;236(4):433–444. doi:10.1002/path.4548
39. Hevener AL, Olefsky JM, Reichart D, et al. Macrophage PPARγ is required for normal skeletal muscle and hepatic insulin sensitivity and full antidiabetic effects of thiazolidinediones. J clin Investig. 2007;117(6):1658–1669. doi:10.1172/JCI31561
40. Chinetti G, Lestavel S, Fruchart J-C, Clavey V, Staels B. Peroxisome proliferator-activated receptor α reduces cholesterol esterification in macrophages. Circul Re. 2003;92(2):212–217. doi:10.1161/01.RES.0000053386.46813.E9
41. Nakamachi T, Nomiyama T, Gizard F, et al. PPARα agonists suppress osteopontin expression in macrophages and decrease plasma levels in patients with type 2 diabetes. Diabetes. 2007;56(6):1662–1670. doi:10.2337/db06-1177
42. Mingzhi S, Jiafu C, Jin H, et al. The in vitro and in vivo anti-inflammatory effects of a phthalimide PPAR-γ agonist. Marine Drugs. 2017;15(1):7.
43. Blanquart C, Barbier O, Fruchart JC, Staels B, Glineur C. Peroxisome proliferator-activated receptors: regulation of transcriptional activities and roles in inflammation. J Steroid Biochem Mol Biol. 2003;85(2–5):267–273. doi:10.1016/S0960-0760(03)00214-0
44. Bouhlel M, Derudas B, Rigamonti E, et al. PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab. 2007;6(2):137. doi:10.1016/j.cmet.2007.06.010
45. Daniel B, Nagy G, Czimmerer Z, et al. The nuclear receptor pparγ controls progressive macrophage polarization as a ligand-insensitive epigenomic ratchet of transcriptional memory. Immunity. 2018;49(4):615–626.e616. doi:10.1016/j.immuni.2018.09.005
46. Al-Rasheed NM, Chana RS, Baines RJ, Willars GB, Brunskill NJ. Ligand-independent activation of peroxisome proliferator-activated receptor-γ by insulin and C-peptide in kidney proximal tubular cells: dependent on phosphatidylinositol 3-kinase activity. J Biol Chem. 2004;279(48):49747–49754. doi:10.1074/jbc.M408268200
47. Varga T, Mounier R, Patsalos A, et al. Macrophage PPARγ, a lipid activated transcription factor controls the growth factor GDF3 and skeletal muscle regeneration. Immunity. 2016;45(5):1038–1051. doi:10.1016/j.immuni.2016.10.016
48. Huang S, Goplen NP, Zhu B, Cheon IS, Sun J. Macrophage PPAR-γ suppresses long-term lung fibrotic sequelae following acute influenza infection. PLoS One. 2019;14(10):e0223430. doi:10.1371/journal.pone.0223430
49. Kang S, Nakanishi Y, Kioi Y, et al. Semaphorin 6D reverse signaling controls macrophage lipid metabolism and anti-inflammatory polarization. Nat Immunol. 2018;19(6):561–570. doi:10.1038/s41590-018-0108-0
50. Ni XX, Ji PX, Chen YX, et al. Regulation of the macrophage‐hepatic stellate cell interaction by targeting macrophage peroxisome proliferator‐activated receptor gamma to prevent non‐alcoholic steatohepatitis progression in mice. Liver Int. 2022;42(12):2696–2712. doi:10.1111/liv.15441
51. Guirado E, Rajaram MV, Chawla A, et al. Deletion of PPARγ in lung macrophages provides an immunoprotective response against M. tuberculosis infection in mice. Tuberculosis. 2018;111:170–177. doi:10.1016/j.tube.2018.06.012
52. Feng X, Gao X, Wang S, et al. PPAR-α agonist fenofibrate prevented diabetic nephropathy by inhibiting M1 macrophages via improving endothelial cell function in db/db mice. Front Med. 2021;8:652558. doi:10.3389/fmed.2021.652558
53. Li Y, Zhang Y, Wang Q, Wu C, Du G, Yang L. Oleoylethanolamide protects against acute ischemic stroke by promoting PPARα-mediated microglia/macrophage M2 polarization. Pharmaceuticals. 2023;16(4):621. doi:10.3390/ph16040621
54. Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications-a review. Nutr J. 2014;13:1–10. doi:10.1186/1475-2891-13-17
55. Girroir EE, Hollingshead HE, He P, Zhu B, Perdew GH, Peters JM. Quantitative expression patterns of peroxisome proliferator-activated receptor-beta/delta (PPARbeta/delta) protein in mice. Biochem biophys Res Commun. 2008;371(3):456–461. doi:10.1016/j.bbrc.2008.04.086
56. Michalik L, Wahli W. Involvement of PPAR nuclear receptors in tissue injury and wound repair. J Clin Investig. 2006;116(3):598–606. doi:10.1172/JCI27958
57. Kang K, Reilly SM, Karabacak V, et al. Adipocyte-derived Th2 cytokines and myeloid PPARdelta regulate macrophage polarization and insulin sensitivity. Cell Metab. 2008;7(6):485–495. doi:10.1016/j.cmet.2008.04.002
58. Odegaard JI, Ricardo-Gonzalez RR, Eagle AR, et al. Alternative (M2) activation of Kupffer cells by PPARδ ameliorates obesity-induced insulin resistance. Cell Metab. 2008;7(6):496–507.
59. Mancino A, Natoli G. Specificity and function of IRF family transcription factors: insights from genomics. J Interferon Cytokine Res. 2016;36(7):462–469. doi:10.1089/jir.2016.0004
60. Jeyakumar T, Fodil N, Van Der Kraak L, et al. Inactivation of interferon regulatory factor 1 causes susceptibility to colitis-associated colorectal cancer. Sci Rep. 2019;9(1):18897. doi:10.1038/s41598-019-55378-2
61. Chistiakov DA, Myasoedova VA, Revin VV, Orekhov AN, Bobryshev YV. The impact of interferon-regulatory factors to macrophage differentiation and polarization into M1 and M2. Immunobiology. 2018;223(1):101–111. doi:10.1016/j.imbio.2017.10.005
62. Günthner R, Anders HJ. Interferon-regulatory factors determine macrophage phenotype polarization. Mediators Infl. 2013;2013:731023. doi:10.1155/2013/731023
63. Xu H, Zhu J, Smith S, et al. Notch–RBP-J signaling regulates the transcription factor IRF8 to promote inflammatory macrophage polarization. Nat Immunol. 2012;13(7):642–650. doi:10.1038/ni.2304
64. Tarassishin L, Suh H-S, Lee SC. Interferon regulatory factor 3 plays an anti-inflammatory role in microglia by activating the PI3K/Akt pathway. J Neuroinfl. 2011;8:1–18. doi:10.1186/1742-2094-8-187
65. Negishi H, Fujita Y, Yanai H, et al. Evidence for licensing of IFN-γ-induced IFN regulatory factor 1 transcription factor by MyD88 in Toll-like receptor-dependent gene induction program. Proc Nat Acad Sci. 2006;103(41):15136–15141. doi:10.1073/pnas.0607181103
66. Langlais D, Barreiro LB, Gros P. The macrophage IRF8/IRF1 regulome is required for protection against infections and is associated with chronic inflammation. J Exp Med. 2016;213(4):585–603. doi:10.1084/jem.20151764
67. Satoh T, Takeuchi O, Vandenbon A, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 2010;11(10):936. doi:10.1038/ni.1920
68. Chartouni CE, Schwarzfischer L, Rehli M. Interleukin-4 induced interferon regulatory factor (Irf) 4 participates in the regulation of alternative macrophage priming. Immunobiology. 2010;215(9–10):821–825. doi:10.1016/j.imbio.2010.05.031
69. Su CY, Fu XL, Duan W, Yu PW, Zhao YL. High density of CD68+ tumor-associated macrophages predicts a poor prognosis in gastric cancer mediated by IL-6 expression. Oncol Letters. 2018. doi:10.3892/ol.2018.8119
70. Negishi H, Ohba Y, Yanai H, et al. Negative regulation of Toll-like-receptor signaling by IRF-4. Proc Nat Acad Sci. 2005;102(44):15989–15994. doi:10.1073/pnas.0508327102
71. Takaoka A, Yanai H, Kondo S, et al. Integral role of IRF-5 in the gene induction programme activated by Toll-like receptors. Nature. 2005;434(7030):243–249. doi:10.1038/nature03308
72. Zhang F, Parayath NN, Ene CI, Stephan SB, Stephan MT. Genetic programming of macrophages to perform anti-tumor functions using targeted mRNA nanocarriers. Nat Commun. 2019;10(1):3974.
73. Platanitis E, Decker T. Regulatory networks involving STATs, IRFs, and NFκB in inflammation. Front Immunol. 2018;9. doi:10.3389/fimmu.2018.02542
74. Rauch I, Rosebrock F, Hainzl E, et al. Noncanonical effects of IRF9 in intestinal inflammation: more than type I and type III interferons. Mol Cell Biol. 2015;35(13):2332–2343. doi:10.1128/MCB.01498-14
75. Ganta VC, Choi MH, Kutateladze A, Fox TE, Farber CR, Annex BH. A MicroRNA93-interferon regulatory Factor-9-Immunoresponsive Gene-1-Itaconic acid pathway modulates M2-like macrophage polarization to revascularize ischemic muscle. Circulation. 2017;135(24):2403–2425. doi:10.1161/CIRCULATIONAHA.116.025490
76. Guan Y, Li X, Yang H, et al. Role and mechanism of IRF9 in promoting the progression of rheumatoid arthritis by regulating macrophage polarization via PSMA5. Heliyon. 2024;10(15):e35589. doi:10.1016/j.heliyon.2024.e35589
77. Igarashi K, Kurosaki T, Roychoudhuri R. BACH transcription factors in innate and adaptive immunity. Nat Rev Immunol. 2017;17(7):437–450. doi:10.1038/nri.2017.26
78. Haldar M, Kohyama M, So AY, et al. Heme-mediated SPI-C induction promotes monocyte differentiation into iron-recycling macrophages. Cell. 2014;156(6):1223–1234. doi:10.1016/j.cell.2014.01.069
79. Ota K, Brydun A, Itoh-Nakadai A, Sun J, Igarashi K. Bach1 deficiency and accompanying overexpression of heme oxygenase-1 do not influence aging or tumorigenesis in mice. Oxid Med Cell Longev. 2014;2014:757901. doi:10.1155/2014/757901
80. Yano Y, Ozono R, Oishi Y, et al. Genetic ablation of the transcription repressor Bach1 leads to myocardial protection against ischemia/reperfusion in mice. Genes Cells. 2006;11(7):791–803. doi:10.1111/j.1365-2443.2006.00979.x
81. Kanno H, Ozawa H, Dohi Y, Sekiguchi A, Igarashi K, Itoi E. Genetic ablation of transcription repressor Bach1 reduces neural tissue damage and improves locomotor function after spinal cord injury in mice. J Neurotrauma. 2009;26(1):31–39. doi:10.1089/neu.2008.0667
82. Yamada K, Tanaka N, Nakanishi K, et al. Modulation of the secondary injury process after spinal cord injury in Bach1-deficient mice by heme oxygenase-1. J Neurosurg Spine. 2008;9(6):611–620. doi:10.3171/SPI.2008.10.08488
83. Iida A, Inagaki K, Miyazaki A, Yonemori F, Ito E, Igarashi K. Bach1 deficiency ameliorates hepatic injury in a mouse model. Tohoku J Exp Med. 2009;217(3):223–229. doi:10.1620/tjem.217.223
84. Tanimoto T, Hattori N, Senoo T, et al. Genetic ablation of the Bach1 gene reduces hyperoxic lung injury in mice: role of IL-6. Free Radic Biol Med. 2009;46(8):1119–1126. doi:10.1016/j.freeradbiomed.2009.01.017
85. Watari Y, Yamamoto Y, Brydun A, et al. Ablation of the bach1 gene leads to the suppression of atherosclerosis in bach1 and apolipoprotein E double knockout mice. Hypertens Res. 2008;31(4):783–792. doi:10.1291/hypres.31.783
86. Harusato A, Naito Y, Takagi T, et al. BTB and CNC homolog 1 (Bach1) deficiency ameliorates TNBS colitis in mice: role of M2 macrophages and heme oxygenase-1. Inflamm Bowel Dis. 2013;19(4):740–753. doi:10.1097/MIB.0b013e3182802968
87. Nakamura A, Ebina-Shibuya R, Itoh-Nakadai A, et al. Transcription repressor Bach2 is required for pulmonary surfactant homeostasis and alveolar macrophage function. J Exp Med. 2013;210(11):2191–2204. doi:10.1084/jem.20130028
88. Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. 2011;11(11):750–761. doi:10.1038/nri3088
89. Lee B, Qiao L, Lu M, et al. C/EBPα regulates macrophage activation and systemic metabolism. Ame J Physiol-Endocrinol Metab. 2014;306(10):E1144–E1154. doi:10.1152/ajpendo.00002.2014
90. Lu YC, Kim I, Lye E, et al. Differential role for c-Rel and C/EBPβ/δ in TLR-mediated induction of proinflammatory cytokines. J Immunol. 2009;182(11):7212–7221. doi:10.4049/jimmunol.0802971
91. Ali AT, Hochfeld WE, Myburgh R, Pepper MS. Adipocyte and adipogenesis. Eur J Cell Biol. 2013;92(6–7):229–236. doi:10.1016/j.ejcb.2013.06.001
92. Ruffell D, Mourkioti F, Gambardella A, et al. A CREB-C/EBPβ cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair. Proc Nat Acad Sci. 2009;106(41):17475–17480. doi:10.1073/pnas.0908641106
93. Ramji DP, Foka P. CCAAT/enhancer-binding proteins: structure, function and regulation. Biochem J. 2002;365(Pt 3):561–575. doi:10.1042/bj20020508
94. Gray MJ, Poljakovic M, Kepka-Lenhart D, Morris SM Jr. Induction of arginase I transcription by IL-4 requires a composite DNA response element for STAT6 and C/EBPβ. Gene. 2005;353(1):98–106. doi:10.1016/j.gene.2005.04.004
95. van Vliet J, Crofts LA, Quinlan KG, Czolij R, Perkins AC, Crossley M. Human KLF17 is a new member of the Sp/KLF family of transcription factors. Genomics. 2006;87(4):474–482. doi:10.1016/j.ygeno.2005.12.011
96. Nayak L, Tugal D, Jain MK. Kruppel-like factors in monocyte–macrophage biology. InMacrophages. 2014;2014:487–495.
97. Liao X, Sharma N, Kapadia F, et al. Kruppel-like factor 4 regulates macrophage polarization. J Clin Investig. :2011;1217:121.
98. Kapoor N, Niu J, Saad Y, et al. Transcription factors STAT6 and KLF4 implement macrophage polarization via the dual catalytic powers of MCPIP. J Immunol. 2015;194(12):6011. doi:10.4049/jimmunol.1402797
99. Wen Y, Lu X, Ren J, et al. klf4 in macrophages attenuates tnfa-mediated kidney injury and fibrosis. J Ame Soc Nephrol. 2019; 30(10):1925–38.
100. Ye Q, Luo F, Yan T. Transcription factor KLF4 regulated STAT1 to promote M1 polarization of macrophages in rheumatoid arthritis. Aging. 2022;14(14):5669. doi:10.18632/aging.204128
101. Feinberg MW, Cao Z, Wara AK, Lebedeva MA, SenBanerjee S, Jain MK. Kruppel-like factor 4 is a mediator of proinflammatory signaling in macrophages. J Biol Chem. 2005;280(46):38247–38258. doi:10.1074/jbc.M509378200
102. Yuan Y, Fan G, Liu Y, et al. The transcription factor KLF14 regulates macrophage glycolysis and immune function by inhibiting HK2 in sepsis. Cell Mol Immunol. 2022;19:504–515.
103. Date D, Das R, Narla G, Simon DI, Jain MK, Mahabeleshwar GH. Kruppel-like transcription factor 6 regulates inflammatory macrophage polarization. J Biol Chem. 2014;289(15):10318–10329. doi:10.1074/jbc.M113.526749
104. Kim GD, Ng HP, Chan ER, Mahabeleshwar GH. Kruppel‐like factor 6 promotes macrophage inflammatory and hypoxia response. FASEB J. 2020;34(2): 3209–23.
105. Pello OM, Pizzol MD, Mirolo M, Soucek L, Locati M. Role of c-MYC in alternative activation of human macrophages and tumor-associated macrophage biology. Blood. 2011;119(2):411–421. doi:10.1182/blood-2011-02-339911
106. Jablonski KA, Amici SA, Webb LM, et al. Novel markers to delineate murine M1 and M2 macrophages. PLoS One. 2015;10(12):e0145342. doi:10.1371/journal.pone.0145342
107. Yang Y, Yu-Chen Y, Yan C, et al. Crosstalk between hepatic tumor cells and macrophages via Wnt/β-catenin signaling promotes M2-like macrophage polarization and reinforces tumor malignant behaviors. Cell Death Dis. 2018;9(8):793. doi:10.1038/s41419-018-0818-0
108. Wang Q, Cheng F, Ma TT, et al. Interleukin-12 inhibits the hepatocellular carcinoma growth by inducing macrophage polarization to the M1-like phenotype through downregulation of Stat-3. Mol Cell Biochem Int J Chem Biol. 2016; 415:157–68.
109. Pello OM, Chèvre R, Laoui D, et al. In vivo inhibition of c-MYC in myeloid cells impairs tumor-associated macrophage maturation and pro-tumoral activities. PLoS One. 2012;7(9):e45399. doi:10.1371/journal.pone.0045399
110. Zhang F, Wang H, Wang X, Jiang G, Du J. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. 2016;7(32):52294–52306. doi:10.18632/oncotarget.10561
111. Li C, Sheng M, Lin Y, et al. Functional crosstalk between myeloid Foxo1–β-catenin axis and Hedgehog/Gli1 signaling in oxidative stress response. Cell Death Differentiation. 2021;28(5):1705–1719. doi:10.1038/s41418-020-00695-7
112. Jin Y, Li C, Xu D, et al. Jagged1-mediated myeloid Notch1 signaling activates HSF1/Snail and controls NLRP3 inflammasome activation in liver inflammatory injury. Cell Mol Immunol. 2020;17(12):1245–1256. doi:10.1038/s41423-019-0318-x
113. Tan F, Cao Y, Zheng L, et al. Diabetes exacerbated sepsis-induced intestinal injury by promoting M1 macrophage polarization via miR-3061/Snail1 signaling. Front Immunol. 2022;13:922614. doi:10.3389/fimmu.2022.922614
114. Bao Z, Zeng W, Zhang D, et al. SNAIL induces EMT and lung metastasis of tumours secreting CXCL2 to promote the invasion of M2-type immunosuppressed macrophages in colorectal cancer. Int J Biol Sci. 2022;18(7):2867. doi:10.7150/ijbs.66854
115. Hsu D-S-S, Wang H-J, Tai S-K, et al. Acetylation of snail modulates the cytokinome of cancer cells to enhance the recruitment of macrophages. Cancer Cell. 2014;26(4):534–548. doi:10.1016/j.ccell.2014.09.002
116. Fu X-T, Dai Z, Song K, et al. Macrophage-secreted IL-8 induces epithelial-mesenchymal transition in hepatocellular carcinoma cells by activating the JAK2/STAT3/Snail pathway. Int J Oncol. 2015;46(2):587–596. doi:10.3892/ijo.2014.2761
117. Zhang Q, Mao Z, Sun J. NF-κB inhibitor, BAY11-7082, suppresses M2 tumor-associated macrophage induced EMT potential via miR-30a/NF-κB/Snail signaling in bladder cancer cells. Gene. 2019;710:91–97. doi:10.1016/j.gene.2019.04.039
118. Wu J, Wang Y, Yang Y, et al. TNFSF9 promotes metastasis of pancreatic cancer through Wnt/Snail signaling and M2 polarization of macrophages. Aging. 2021;13(17):21571. doi:10.18632/aging.203497
119. Yadav MK, Inoue Y, Nakane-Otani A, et al. Transcription factor MafB is a marker of tumor-associated macrophages in both mouse and humans. Biochem Biophys Res Commun. 2020;521(3):590–595. doi:10.1016/j.bbrc.2019.10.125
120. Daassi D, Hamada M, Jeon H, Imamura Y, Nhu Tran MT, Takahashi S. Differential expression patterns of MafB and c-Maf in macrophages in vivo and in vitro. Biochem Biophys Res Commun. 2016;473(1):118–124. doi:10.1016/j.bbrc.2016.03.063
121. Hasegawa H, Watanabe T, Kato S, et al. The role of macrophage transcription factor MafB in atherosclerotic plaque stability. Atherosclerosis. 2016;250:133–143. doi:10.1016/j.atherosclerosis.2016.05.021
122. Franklin RA, Liao W, Sarkar A, et al. The cellular and molecular origin of tumor-associated macrophages. Science. 2014;344(6186):921–925. doi:10.1126/science.1252510
123. Gemelli C, Zanocco Marani T, Bicciato S, et al. MafB is a downstream target of the IL-10/STAT3 signaling pathway, involved in the regulation of macrophage de-activation ☆. BBA - Mol Cell Res. 2014;1843(5):955–964. doi:10.1016/j.bbamcr.2014.01.021
124. Inoue Y, Liao C-W, Tsunakawa Y, Tsai I-L, Takahashi S, Hamada M. Macrophage-specific, mafb-deficient mice showed delayed skin wound healing. Int J Mol Sci. 2022;23(16):9346. doi:10.3390/ijms23169346
125. Liu M, Tong Z, Ding C, Luo F, Yan J. Transcription factor c-Maf is a checkpoint that programs macrophages in lung cancer. J Clin Investig. 2020;130(4):2081–2096. doi:10.1172/JCI131335
126. Larionova I, Kazakova E, Patysheva M, Kzhyshkowska J. Transcriptional, epigenetic and metabolic programming of tumor-associated macrophages. Cancers. 2020;12(6):1411. doi:10.3390/cancers12061411
127. Silva HM, Kitoko JZ, Queiroz CP, et al. c-MAF–dependent perivascular macrophages regulate diet-induced metabolic syndrome. Sci Immunol. 2021;6(64):eabg7506. doi:10.1126/sciimmunol.abg7506
128. Mcgettrick AF, LAJ O. The Role of HIF in Immunity and Inflammation. Cell Metabol. 2020;32(4):524–536. doi:10.1016/j.cmet.2020.08.002
129. Qiu B, Yuan P, Du X, Jin H, Du J, Huang Y. Hypoxia inducible factor-1α is an important regulator of macrophage biology. Heliyon. 2023;9(6):e17167. doi:10.1016/j.heliyon.2023.e17167
130. Peyssonnaux C, Datta V, Cramer T, et al. HIF-1alpha expression regulates the bactericidal capacity of phagocytes. J Clin Investig. 2005;115(7):115.
131. Blouin CC, Page EL, Soucy GM, Richard DE. Hypoxic gene activation by lipopolysaccharide in macrophages: implication of hypoxia-inducible factor 1α. Blood. 2004;103(3):1124–1130. doi:10.1182/blood-2003-07-2427
132. Takeda N, O’Dea EL, Doedens A, et al. Differential activation and antagonistic function of HIF-α isoforms in macrophages are essential for NO homeostasis. Genes Dev. 2010;24(5):491–501. doi:10.1101/gad.1881410
133. Wang T, Liu H, Lian G, Zhang SY, Jiang C. HIF1±-induced glycolysis metabolism is essential to the activation of inflammatory macrophages. Mediators Inflamm. 2017;2017(11):1–10. doi:10.1155/2017/3102737
134. Gondin J, Theret M, Duhamel G, et al. Myeloid HIFs are dispensable for resolution of inflammation during skeletal muscle regeneration. J Immunol. 2015;194(7):3389–3399. doi:10.4049/jimmunol.1401420
135. Amy IZHWPEHMMPASDC. Hypoxia-inducible factor 2α regulates macrophage function in mouse models of acute and tumor inflammation. J Clin Investig. 2010;120:1.
136. Peyssonnaux C, Cejudo-Martin P, Doedens A, Zinkernagel AS, Johnson RS, Nizet V. Cutting edge: essential role of hypoxia inducible factor-1α in development of lipopolysaccharide-induced sepsis. J Immunol. 2007;178(12):7516–7519. doi:10.4049/jimmunol.178.12.7516
137. Tannahill GM, Curtis AM, Adamik J, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238. doi:10.1038/nature11986
138. Doedens AL, Stockmann C, Rubinstein MP, et al. Macrophage expression of hypoxia-inducible factor-1α suppresses T-cell function and promotes tumor progression. Cancer Res. 2010;70(19):7465–7475. doi:10.1158/0008-5472.CAN-10-1439
139. Imtiyaz HZ, Williams EP, Hickey MM, et al. Hypoxia-inducible factor 2alpha regulates macrophage function in mouse models of acute and tumor inflammation. J Clin Investig. 2010;120(8):2699–2714. doi:10.1172/JCI39506
140. Flynn JL, Chan J, Triebold KJ, Dalton DK, Stewart TA, Bloom BR. An essential role for interferon gamma in resistance to Mycobacterium tuberculosis infection. J Exp Med. 1993;178(6):2249–2254. doi:10.1084/jem.178.6.2249
141. Braverman J, Sogi KM, Benjamin D, Nomura DK, Stanley SA. HIF-1 alpha is an essential mediator of IFN-gamma-dependent immunity to mycobacterium tuberculosis. J Immunol. 2016;197(4):1287–1297. doi:10.4049/jimmunol.1600266
142. Guo X, Zhu Z, Zhang W, et al. Nuclear translocation of HIF-1α induced by influenza A (H1N1) infection is critical to the production of proinflammatory cytokines. Emerg Microbes Infect. 2017;6(5):e39. doi:10.1038/emi.2017.21
143. Eubank TD, Roda JM, Liu H, O’Neil T, Marsh CB. Opposing roles for HIF-1alpha and HIF-2alpha in the regulation of angiogenesis by mononuclear phagocytes. Blood. 2011;117:320–32.
144. Roda JM, Wang Y, Sumner LA, Phillips GS, Marsh CB, Eubank TD. Stabilization of HIF-2α induces sVEGFR-1 production from tumor-associated macrophages and decreases tumor growth in a murine melanoma model. J Immunol. 2012;189(6):3168. doi:10.4049/jimmunol.1103817
145. Ahn G-O, Seita J, Hong B-J. Transcriptional activation of hypoxia-inducible factor-1 (HIF-1) in myeloid cells promotes angiogenesis through VEGF and S100A8. Proc Nat Acad Sci. 2014;111(7):2698–2703. doi:10.1073/pnas.1320243111
146. Nagel S, Ehrentraut S, Meyer C, Kaufmann M, Drexler HG, MacLeod RA. Aberrantly expressed OTX homeobox genes deregulate B-cell differentiation in Hodgkin lymphoma. PLoS One. 2015;10(9):e0138416. doi:10.1371/journal.pone.0138416
147. Tan S, Guo X, Li M, et al. Transcription factor Zhx2 restricts NK cell maturation and suppresses their antitumor immunity. J Exp Med. 2021;218(9):e20210009. doi:10.1084/jem.20210009
148. Erbilgin A, Seldin MM, Wu X, et al. Transcription factor Zhx2 deficiency reduces atherosclerosis and promotes macrophage apoptosis in mice. Arteriosclerosis Thrombosis Vasc Biol. 2018;38(9):2016–2027. doi:10.1161/ATVBAHA.118.311266
149. Wang Z, Kong L, Tan S, et al. Zhx2 accelerates sepsis by promoting macrophage glycolysis via Pfkfb3. J Immunol. 2020;204(8):2232–2241. doi:10.4049/jimmunol.1901246
150. Tan S, Wang Z, Li N, et al. Transcription factor Zhx2 is a checkpoint that programs macrophage polarization and antitumor response. Cell Death Differentiation. 2023;30(9):2104–2119. doi:10.1038/s41418-023-01202-4
151. Potthoff MJ, Olson EN. MEF2: a central regulator of diverse developmental programs. Development. 2007;134(23):4131–4140. doi:10.1242/dev.008367
152. Bouttier M, Laperriere D, Memari B, et al. Alu repeats as transcriptional regulatory platforms in macrophage responses to M. tuberculosis infection. Nucleic Acids Res. 2016;44(22):10571–10587. doi:10.1093/nar/gkw782
153. Zhao X, Di Q, Liu H, et al. MEF2C promotes M1 macrophage polarization and Th1 responses. Cell Mol Immunol. 2022;19(4):540–553. doi:10.1038/s41423-022-00841-w
154. Ku H-C, Cheng C-F. Master regulator activating transcription factor 3 (ATF3) in metabolic homeostasis and cancer. Front Endocrinol. 2020;11:556. doi:10.3389/fendo.2020.00556
155. Whitmore MM, Iparraguirre A, Kubelka L, Weninger W, Hai T, Williams BR. Negative regulation of TLR-signaling pathways by activating transcription factor-3. J Immunol. 2007;179(6):3622–3630. doi:10.4049/jimmunol.179.6.3622
156. Kwon J-W, Kwon H-K, Shin H-J, Choi Y-M, Anwar MA, Choi S. Activating transcription factor 3 represses inflammatory responses by binding to the p65 subunit of NF-κB. Scientific Rep. 2015;5(1):14470. doi:10.1038/srep14470
157. Labzin LI, Schmidt SV, Masters SL, et al. ATF3 is a key regulator of macrophage IFN responses. J Immunol. 2015;195(9):4446–4455. doi:10.4049/jimmunol.1500204
158. Sha H, Zhang D, Zhang Y, Wen Y, Wang Y. ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin‑C via Wnt/β‑catenin pathway. Mol Med Rep. 2017;16(3):3641–3647. doi:10.3892/mmr.2017.6992
159. Wang W, Xu R, He P, et al. Role of ATF3 triggering M2 macrophage polarization to protect against the inflammatory injury of sepsis through ILF3/NEAT1 axis. Mol Med. 2024;30(1):30. doi:10.1186/s10020-023-00711-9
160. Kensler TW, Wakabayashi N, Biswal S. Cell survival responses to environmental stresses via the Keap1-Nrf2-ARE pathway. Annu Rev Pharmacol Toxicol. 2007;47(1):89–116. doi:10.1146/annurev.pharmtox.46.120604.141046
161. Saha S, Buttari B, Panieri E, Profumo E, Saso L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules. 2020;25(22):5474. doi:10.3390/molecules25225474
162. Marchev AS, Dimitrova PA, Burns AJ, Kostov RV, Dinkova‐Kostova AT, Georgiev MI. Oxidative stress and chronic inflammation in osteoarthritis: can NRF2 counteract these partners in crime? Annals New York Acad Sci. 2017;1401(1):114–135. doi:10.1111/nyas.13407
163. Hayes JD, Dinkova-Kostova AT. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem Sci. 2014;39(4):199–218. doi:10.1016/j.tibs.2014.02.002
164. Consonni FM, Bleve A, Totaro MG, et al. Heme catabolism by tumor-associated macrophages controls metastasis formation. Nat Immunol. 2021;22(5):595–606. doi:10.1038/s41590-021-00921-5
165. Li M, Yu H, Pan H, et al. Nrf2 suppression delays diabetic wound healing through sustained oxidative stress and inflammation. Front Pharmacol. 2019;10:1099. doi:10.3389/fphar.2019.01099
166. Wang L, He C. Nrf2-mediated anti-inflammatory polarization of macrophages as therapeutic targets for osteoarthritis. Front Immunol. 2022;13:967193. doi:10.3389/fimmu.2022.967193
167. Kobayashi EH, Suzuki T, Funayama R, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat Commun. 2016;7. doi:10.1038/ncomms11624
168. Wang Z, Wu Z, Wang H, et al. An immune cell atlas reveals the dynamics of human macrophage specification during prenatal development. Cell. 2023;186(20):4454–4471.e4419. doi:10.1016/j.cell.2023.08.019
169. Cheng S, Li Z, Gao R, et al. A pan-cancer single-cell transcriptional atlas of tumor infiltrating myeloid cells. Cell. 2021;184(3):792–809.e723. doi:10.1016/j.cell.2021.01.010
170. Yan K, Da -T-T, Bian Z-H, et al. Multi-omics analysis identifies FoxO1 as a regulator of macrophage function through metabolic reprogramming. Cell Death Dis. 2020;11(9):800. doi:10.1038/s41419-020-02982-0
171. Yousuf S, Qiu M, von Voithenberg LV, et al. Spatially resolved multi-omics single-cell analyses inform mechanisms of immune dysfunction in pancreatic cancer. Gastroenterology. 2023;165(4):891–908.e814. doi:10.1053/j.gastro.2023.05.036
172. He L, Jhong J-H, Chen Q, et al. Global characterization of macrophage polarization mechanisms and identification of M2-type polarization inhibitors. Cell Rep. 2021;37(5). doi:10.1016/j.celrep.2021.109955.
173. Matusiak M, Hickey JW, van IJzendoorn DG, et al. Spatially segregated macrophage populations predict distinct outcomes in colon cancer. Cancer Discovery. 2024;14:OF1–OF22.
174. Lei A, Yu H, Lu S, et al. A second-generation M1-polarized CAR macrophage with antitumor efficacy. Nat Immunol. 2024;25(1):102–116. doi:10.1038/s41590-023-01687-8
175. Chen Y, Chen X, Zhang Y, et al. Macrophage-specific in vivo RNA editing promotes phagocytosis and antitumor immunity in mice. Sci Transl Med. 2025;17(781):eadl5800. doi:10.1126/scitranslmed.adl5800
© 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The
full terms of this license are available at https://www.dovepress.com/terms.php
and incorporate the Creative Commons Attribution
- Non Commercial (unported, 4.0) License.
By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted
without any further permission from Dove Medical Press Limited, provided the work is properly
attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.
Recommended articles

The Role of Podoplanin in the Immune System and Inflammation
Zhang Z, Zhang N, Yu J, Xu W, Gao J, Lv X, Wen Z
Journal of Inflammation Research 2022, 15:3561-3572
Published Date: 17 June 2022

A Tissue-Tended Mycophenolate-Modified Nanoparticle Alleviates Systemic Lupus Erythematosus in MRL/Lpr Mouse Model Mainly by Promoting Local M2-Like Macrophagocytes Polarization
Jiang B, Zhang Y, Li Y, Chen Y, Sha S, Zhao L, Li D, Wen J, Lan J, Lou Y, Su H, Zhang C, Zhu J, Tao J
International Journal of Nanomedicine 2022, 17:3251-3267
Published Date: 28 July 2022

P2X7R-NEK7-NLRP3 Inflammasome Activation: A Novel Therapeutic Pathway of Qishen Granule in the Treatment of Acute Myocardial Ischemia
Li Y, Sun X, Liu X, Li J, Li X, Wang G, Liu Y, Lu X, Cui L, Shao M, Wang Y, Wang W, Li C
Journal of Inflammation Research 2022, 15:5309-5326
Published Date: 13 September 2022

CCL20/CCR6 Mediated Macrophage Activation and Polarization Can Promote Adenoid Epithelial Inflammation in Adenoid Hypertrophy
Ye C, Guo X, Wu J, Wang M, Ding H, Ren X
Journal of Inflammation Research 2022, 15:6843-6855
Published Date: 23 December 2022

Theaflavin-3,3’-Digallate Ameliorates Collagen-Induced Arthritis Through Regulation of Autophagy and Macrophage Polarization
Zhang L, Li W, Hou Z, Wang Z, Zhang W, Liang X, Wu Z, Wang T, Liu X, Peng X, Yang X, Yang H, Geng D
Journal of Inflammation Research 2023, 16:109-126
Published Date: 10 January 2023