International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
As a sentinel of the immune system, macrophages are characterized by the robust phagocytic capacity to play a pivotal role in pathogen clearance and tissue homeostasis (1). Based on their developmental origins, macrophages are categorized into hematopoietic monocyte-derived macrophages and embryonic-derived tissue-resident macrophages (2). Hematopoietic-derived macrophages originate from CD34+ hematopoietic stem cells (HSCs) in the bone marrow, progressing through granulocyte-monocyte progenitors and monocytic stages, before entering systemic circulation (3). Under specific microenvironmental signals, such as inflammatory mediators and chemokines, circulating monocytes are recruited to tissues and subsequently differentiate into functionally specialized macrophages. By contrast, embryonic-derived tissue-resident macrophages originate from yolk sac precursors during organogenesis (4-9), including microglia in the central nervous system (10), Kupffer cells in the liver (11) and Langerhans cells in the skin (12). These developmentally distinct populations differ fundamentally from their hematopoietic counterparts in both ontogeny and functional specialization (2,6,8,13-16).
Macrophages exhibit extraordinary plasticity and functional heterogeneity, dynamically adapting their polarization states in response to microenvironmental stimuli (17). The classical paradigm classifies macrophages into pro-inflammatory M1 and anti-inflammatory/reparative M2 phenotypes (18-20). M1 macrophages, activated by interferon (IFN)-γ and lipopolysaccharide (LPS) (21,22), amplify inflammatory responses and resist pathogenic infection through the production of IL-1β, IL-6, IL-12, TNF-α and chemokines (23-26). While being critical for pathogen containment, sustained M1 activation leads to chronic inflammation and tissue destruction. Conversely, M2 macrophages polarized by IL-4/IL-13 secrete immunosuppressive cytokines (such as IL-10 and TGF-β) to alleviate inflammation and promote tissue regeneration through growth factor-mediated extracellular matrix remodeling and angiogenesis (27). However, their immunosuppressive function may facilitate tumor progression by establishing pro-tumorigenic microenvironments. Importantly, the M1/M2 classification is considered an oversimplification of macrophage functional spectrum. Under pathophysiological conditions, macrophages exhibit a continuous spectrum of polarization from M1 to M2 phenotypes, rather than discrete binary states (28-31). Tissue macrophages frequently exhibit hybrid phenotypes with overlapping M1-M2 characteristics, reflecting their capacity to adapt to dynamic microenvironmental changes. To further clarify this conceptual framework, it is essential to distinguish between the processes of macrophage activation and polarization. Macrophage activation refers to the early process by which macrophages sense external stimuli through receptor-mediated signaling pathways, initiating intracellular cascades that endow them with enhanced responsiveness and effector potential. Macrophage activation represents a transition from resting state to functionally poised state. By contrast, macrophage polarization, dictated by specific cytokine milieu and environmental cues, denotes a more specialized process in which activated macrophages differentiate along a continuous spectrum into distinct functional phenotypes (32,33). Therefore, activation represents the initiation of macrophage responsiveness, whereas polarization describes the directional specification that follows.
Through coordinated phagocytosis, antigen presentation, cytokine secretion and crosstalk with immune and non-immune cells, macrophages play a central role in regulating inflammatory responses, host defense and tissue homeostasis. Dysregulated macrophage function underlies numerous pathologies. For instance, macrophages take up lipids from the blood to transform into foam cells, thereby promoting plaque formation and atherosclerotic progression (34-39). Obesity-associated metabolic disorders are featured by the macrophage-mediated chronic low-grade inflammation, which triggers the development of insulin resistance and type 2 diabetes (40-47). In certain cases, M2 macrophages may promote tumor cell growth, metastasis, invasion and drug resistance by secreting immunosuppressive factors and enhancing tumor angiogenesis through cytokines (such as VEGF) (48-52). As highly plastic innate immune cells derived from myeloid precursors, the differentiation of macrophage is orchestrated by the macrophage colony-stimulating factor (M-CSF, also known as CSF1) (53-55). This process is fundamentally governed by the transcription factor purine-rich U-box binding protein 1, which drives macrophage lineage differentiation through transcriptional activation of the M-CSF receptor CSF1R (56-58). NF-κB and STAT1 pathways drive classical M1 polarization (59,60), while alternative M2 polarization is mediated by JAK1/3-STAT6 signaling and nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) (61-65). Collectively, these observations underscore that macrophage differentiation, activation and polarization are governed by intricate transcriptional networks. Among these, the activating transcription factors (ATFs) have emerged as pivotal integrators of immune and metabolic cues, orchestrating macrophage responses under diverse physiological and pathological conditions. The current review sought to summarize the recent findings on how activated ATFs regulate macrophage development, survival, migration, phagocytosis, activation, cytokine secretion, polarization and their involvement in immune, metabolic, cardiovascular, neurological disorders and cancer, with a particular emphasis on their therapeutic potential (Fig. 1).
The ATF family, comprising seven members (ATF1-7), was initially characterized in 1987, as a group of transcriptional factors involved in the regulation of gene expression across diverse biological contexts (66). All ATF members share a highly conserved basic leucine zipper (bZIP) domain comprising a basic region responsible for DNA binding and a leucine zipper structure that mediates dimerization (67,68). ATFs are activated by upstream signals and exert transcriptional activities by forming either homodimers or heterodimers with other bZIP transcription factors such as AP-1 or C/EBP family members. ATF1 commonly forms heterodimers with cAMP response element-binding protein (CREB) or cAMP response element modulator (CREM) family members to bind to DNA and regulate target gene transcription (69-72). ATF2 dimerizes with c-Jun to form a canonical AP-1 complex (73,74), while its structurally related homolog ATF7 could interact with ATF2 to generate functional heterodimers (75-77). ATF3 cooperates with bZIP proteins including c-Jun, JunB, or C/EBP to regulate responsive genes (78,79). ATF4 integrates stress and metabolic signals through heterodimerization with members of the C/EBP family or with nuclear factor erythroid 2-related factor 2 (NRF2) to coordinate adaptive transcriptional programs (80-82). Similarly, ATF5 forms functional heterodimers with C/EBPγ (83). ATF6, however, undergoes a unique activation process. At steady state, ATF6 resides in the endoplasmic reticulum (ER) membrane as a monomer, dimer, or oligomer. Upon the challenge of ER stress, ATF6 translocates into the Golgi apparatus, where it is cleaved by S1P/S2P proteases to release its N-terminal fragment (50 kDa) that enters the nucleus. The nuclear p50 ATF6 forms homodimers or heterodimers with its homolog ATF6β (84) and both of them can associate with nuclear transcription factor Y (NF-Y) to form transcriptional complexes (85). In addition, ATF6 has been shown to heterodimerize with XBP1s, thereby broadening its target gene repertoire (86).
The specific dimerization partners of each ATF member largely determine its DNA-binding specificity and transcriptional outcomes. Accordingly, ATF1 and ATF5 predominantly recognize canonical CRE sequences (70,87), whereas the binding preferences of ATF2, ATF3 and ATF7 are largely determined by their dimerization partners (88-91). Once they form homodimers or heterodimers with the CREB family members, they preferentially bind to the CRE consensus sequence. By contrast, heterodimerization with AP-1 family proteins, such as c-Jun, redirects their binding toward AP-1 sites (74,78,92-94). Moreover, ATF3 and ATF4 can recognize C/EBP-ATF response elements (CARE), to composite motifs containing both CCAAT-box and CRE characteristics (95). Under stress conditions, the C/EBPγ-ATF4 heterodimer constitutes the predominant form to bind to the CARE motifs (81), whereas ATF4 can also form a complex with C/EBPβ to regulate differentiation-related gene expression (80). When ATF4 forms heterodimers with NRF2, the complex can recognize both antioxidant response elements (AREs) and ATF/CRE motifs, thereby synergistically activating antioxidant and detoxifying gene transcription (96). Likewise, ATF6α/ATF6β heterodimers, as well as ATF6α homodimers, associate with NF-Y to recognize ER stress response elements, which comprise a 5'-CCAAT-3' core, a GC-rich spacer and a terminal 5'-CCACG-3' motif (97-101). Within this composite motif, the CCAAT box serves as the NF-Y binding site, whereas ATF6 recognizes and binds to the terminal CCACG half-site to initiate transcription of target genes (85,102). Notably, CRE sequences vary among genes, often exhibiting one or two base substitutions and even half-CRE sites (such as TGACG or CGTCA) can be recognized by ATFs, suggesting that while the core CRE motif is evolutionarily conserved, a certain degree of sequence flexibility is tolerated (103).
The activation and function of the ATF family are governed by multi-layered regulatory mechanisms operating across epigenetic, transcriptional and post-translational dimensions. Epigenetic control involves DNA methylation (such as ATF3 silencing through promoter hypermethylation) (104), dynamic histone modifications and mRNA m6A modifications. Transcriptionally, factors, such as SP1/MYC and p53, modulate ATF activity via promoter binding (104). Post-translationally, numerous ATFs are activated through phosphorylation (PKA or MAPK) (105), while others undergo regulated proteolysis (such as cleavage of ATF6) or ubiquitin-mediated turnover (106), all of which influence their stability, localization and activity. Specifically, under stress conditions, JNK/p38 MAPK phosphorylates ATF2 to enhance its transcriptional activity (107), while ER stress activates ATF4 and ATF6 via the unfolded protein response (UPR) to restore proteostasis (106,108). Moreover, ATFs could also be regulated by signals such as IL-6, LPS or growth factors (109-111), thereby integrating immune and metabolic cues into coordinated transcriptional responses. Once activated, ATFs execute their transcriptional regulatory functions primarily through the recruitment of a wide range of transcriptional cofactors and chromatin-modifying complexes that sculpt gene expression programs. Among them, ATF family members recruit diverse histone-modifying enzymes and chromatin-remodeling complexes to mediate epigenetic regulation. For example, phosphorylated ATF1 recruits the histone acetyltransferase CBP/p300 to enhance local H3/H4 acetylation and chromatin accessibility (112,113). Similarly, following LPS stimulation, ATF2 recruits histone deacetylase 1 (HDAC1) to remove repressive acetylation marks, thereby facilitating target gene transcription (114). However, activated ATF3 recruits HDAC1 onto promoters of proinflammatory genes to erase permissive acetylation marks and repress gene transcription, implying the complex nature of ATF-related epigenetic regulation (115,116). Upon the insult of ER stress, ATF6α recruits the mediator complex together with multiple histone acetyltransferase assemblies, including SAGA and ATAC, to promoters such as HSPA5, to increase local histone acetylation level (117,118). ATF7 persistently recruits the histone H3K9 methyltransferase G9a in resting macrophages to deposit the repressive H3K9me2 mark, which silences the transcription of innate immune genes and maintain the quiescent state (119). Moreover, ATF6α recruits arginine methyltransferases such as PRMT1, to methylate histone arginine residues at target genes, by which it remodels chromatin architecture and regulates stress-responsive gene expression (120). Moreover, some ATF members can directly or indirectly recruit ATP-dependent chromatin-remodeling complexes, altering nucleosome positioning and conformation to reorganize the chromatin landscape (112).
ATFs play a crucial role in various biological processes. For instance, ATF family members regulate antioxidant gene expression during ER stress and oxidative stress responses, serving as a critical regulator in stress adaptation (121-123). In glucose and lipid metabolism, ATF proteins modulate the expression of multiple metabolic genes to maintain glucose and lipid homeostasis (111,124). Furthermore, ATF members participate in immune responses by regulating inflammatory cascades and immune evasion (125-128), the balance between autophagy and apoptosis (129) and the process of cell senescence (126). Notably, perturbation of ATF signaling networks demonstrates strong association with tumor (130), metabolic disorders (121), neurodegenerative diseases (121) and immune-related diseases (124,131). Such dynamic nature warrants ATFs as a crucial hub for developing targeted therapeutic strategies.
ATFs exert multidimensional regulatory effect on macrophage development and functional homeostasis through regulating the proliferation, differentiation and metabolic pathways. During macrophage development, ATF4 serves as a master regulator to govern monocytes differentiation to gut-resident macrophages (gMac). In septic models, ATF4 depletion in Ly6C+ monocyte precursors (P1 cells) disrupts gMac (P4 cell) differentiation coupled with aberrant proliferation and increased apoptosis of P1 cells, leading to exacerbated intestinal barrier dysfunction (132). ATF2 orchestrates monocyte differentiation through modulating the expression of phosphatase PPM1A by binding to its promoter (133). In atherosclerosis, the accumulation of macrophages is markedly reduced in plaques of mice overexpressing ATF3. ATF3 overexpression suppresses the PI3K/AKT signaling pathway, by which it downregulates the expression of matrix metalloproteinases (MMP-2/MMP-9) and reduces macrophage-mediated extracellular matrix degradation, coupled with plaque stabilization (134). Notably, the anti-proliferative effect of ATF3 persists throughout the macrophage lifespan by suppressing cell cycle genes (Mem2 and Cdk2) and inhibiting the Clec4e-Csf1 axis (135,136).
Regarding macrophage senescence and cell death, ATF4 is identified as the core responder to hypoxic stress and it rapidly accumulates in macrophage nuclei within 1-h of hypoxic challenge to activate adaptive gene networks to sustain cell viability (137). ATF3 exhibits anti-senescence properties in Pseudomonas aeruginosa infection models. Macrophages deficient in ATF3 exhibit elevated ROS levels and accelerated senescence, while ectopic ATF3 overexpression partially reverses these phenotypes (138). Additionally, in LPS-stimulated microglia, ATF2 exacerbates inflammatory pyroptosis by upregulating IL-1β, NLRP3 and key pyroptotic effector molecules such as GSDMDC1 and Caspase-1 (139).
ATF family members essentially regulate macrophage chemotaxis and phagocytosis by modulating chemokine expression and cytoskeletal remodeling. LPS induces phosphorylation of ATF2 in alveolar epithelial cells, which subsequently upregulates macrophage inflammatory protein-2 (MIP-2) to promote macrophage recruitment (140). Additionally, LPS enhances macrophage chemotactic capacity via ATF3, which binds to the CRE/AP-1 motif within the promoter of Regulated upon Activation, Normal T-cell Expressed and Secreted (RANTES) (141). Notably, ATF3 also regulates macrophage migration through other mechanisms. Transwell assays suggest that ATF3-overexpressing macrophages exhibit markedly enhanced migration under MCP-1 treatment (142,143). Mechanistically, ATF3 suppresses gelsolin expression and F-actin de-polymerization to promote cytoskeleton reorganization (144). On the other hand, ATF3 activates the Wnt/β-catenin signaling pathway to upregulate extracellular matrix protein tenascin C, which subsequently reinforces macrophage migration (142). These findings elucidate the molecular basis by which ATF family members, ATF3 in particular, integrate chemokine networks and cytoskeletal dynamics to precisely orchestrate macrophage migration.
Phagocytosis is another biological process relevant to cytoskeleton remodeling (145). ATF3 directly binds to the AP-1 motif within the promoter of Slc11a1, a gene that is critical for phagolysosomal iron transport and function, to downregulate SLC11A1 expression (146). Such inhibitory effect markedly impairs macrophage phagocytotic capacity, as evidenced by the enhanced uptake of fluorescent microspheres in ATF3-knockout models (138). In cyclophosphamide-induced immunosuppressive model, ATF3 further reduces macrophage phagocytosis of tumor cells by repressing antimicrobial peptide genes, such as Camp, Lcn2 and Ltf (147). Notably, ATF3 also influences lipid metabolism by upregulating the expression of scavenger receptor class B type 1 to promote macrophage reversing cholesterol transport (RCT), thereby preventing foam cell formation (148). By contrast, ATF2 enhances oxidized low-density lipoprotein (oxLDL) uptake and lipid accumulation by directly binding to the Cd36 promoter (-100 nt site) to drive its transcription (149). During Mycobacterium tuberculosis (Mtb) infection, overexpression of ATF2 enhances macrophage phagocytic capacity for pathogen clearance (133). Additionally, ATF1 coordinates the separation of iron and lipid metabolism during the response to heme-derived products, by regulating the expression of heme oxygenase-1 (HO-1), liver X receptor (LXR) and Spi-C transcription factor (SPIC). ATF1 deficiency disrupts phagolysosomal homeostasis, which impairs post-phagocytic processing (150). Further studies revealed that ATF1 and ATF2 exert synergistic effects on pathogen recognition. The two factors are involved in the O-acetylation of the cell wall component glycolipid in Mtb and Mycobacterium abscessus (M. abscessus), altering the molecular patterns on the pathogen surface and affecting macrophage phagocytotic efficiency (151). In fungal infection models, upon macrophage uptake of Mucor circinelloides, ATF1 and ATF2 are activated to regulate a series of gene networks, including Chi1 and Aqp1, thus enhancing fungal survival within the phagosome. This process supports fungal persistence and germination during phagocytosis (152).
Macrophage activation and cytokine secretion are orchestrated by ATFs that integrate microbial, metabolic and stress-derived signals, into coordinated transcriptional programs. ATF1, ATF5 and ATF6 predominantly act as pro-inflammatory drivers, while ATF3 and ATF7 serve as negative feedback regulators. Intriguingly, ATF2 and ATF4 exhibit context-dependent dual functions, capable of both amplifying and restraining inflammatory responses depending on the microenvironmental cues. The upstream stimuli, signaling connections and differential impacts on cytokine expression are summarized in Fig. 2 and Table I, which together outline the overall regulatory network and highlight the synergistic and antagonistic interactions among ATF members. The dynamic equilibrium among these ATF subsets ultimately determines the intensity and persistence of macrophage activation and cytokine production.
Table IDistinct regulatory roles of ATF family members in macrophage activation and cytokine secretion. |
ATF family members regulate macrophage pro-inflammatory activation through multiple pathways. ATF1, activated via sensing bacterial peptidoglycan (PGN), drives pro-inflammatory cytokine transcription either in its phosphorylated form or as a dimer with CREB (153). ATF5 directly amplifies inflammatory responses by upregulating TNF-α, IL-1β and IL-6 expression (154). ATF6, a key regulatory factor in ER stress (155), is essential for initiating the UPR triggered by pattern recognition receptors such as nucleotide-binding oligomerization domain-containing protein 2 (NOD2) (106). Upon the challenge of oxLDL, ATF6 dissociates from the ER membrane, moves into the Golgi apparatus, where it is cleaved into its active form (p50-ATF6), which subsequently translocates into the nucleus to regulate target genes, such as GRP78 and XBP-1 (156). Activated ATF6 stimulates the expression of genes involved in cholesterol biosynthesis, but represses the genes associated with cholesterol efflux (Abca1, Abcg1, Lxrα), to drive cholesterol accumulation (157). ATF6 directly binds to the Tnf-α promoter to enhance its transcriptional activity, thereby activating the NF-κB and TNF signaling pathways and driving macrophages to secrete pro-inflammatory cytokines, such as TNFα, IL-1β and IL-6 (156). In liver Kupffer cells, ATF6 also mediates the pro-inflammatory enhancement of TLR4 response and cytokine production (158). Markedly, ubiquitination at K152 residue of ATF6 is essential for its antimicrobial function. Mutations at this site (ATF6-K152A) exhibit defective bacterial uptake, coupled with reduced ROS generation, diminished LC3II/ATG5 expression and impaired intracellular pathogen clearance (159). Together, these findings establish ATF1, ATF5 and ATF6 as central mediators that translate microbial and metabolic stress into potent inflammatory cytokine programs.
In sharp contrast, ATF3 and ATF7 largely hinder the activation of macrophages. Under physiological condition, ATF7 acts as an epigenetic repressor by recruiting histone H3K9 dimethyltransferase G9a to the promoters of activation-related genes (such as Cxcl2, Ccl3, Stat1, Myo10, Nfkb2 and Tap1), thereby maintaining macrophage at the resting state. However, LPS stimulation induces p38-dependent phosphorylation of ATF7, prompting ATF7 dissociation from chromatin, which leads to a significant reduction of H3K9me2 levels at target genes and the unleashing of transcriptional repression. Such epigenetic change persists for weeks, which sustains low H3K9me2 levels and high transcriptional activity to confer long-term immune memory (119). Similarly, ATF3 functions as an essential effector in curbing excessive macrophage activation. First, ATF3 inhibits the phosphorylation of JNK, ERK and p38 MAPK, thereby reducing the production of TNF-α, IL-6, IL-1β and IL-12β induced by LPS, ROS, or Mycoplasma pneumoniae infection (125,138,160-162). Second, NRF2 binds to the antioxidant response elements (ARE1) within the Atf3 promoter to initiate Atf3 transcription (161). Activated ATF3 then recruits HDAC1 to pro-inflammatory gene promoters (143,163), thereby reducing H3/H4 histone acetylation (138,164) and counteracting Rel/NF-κB transcriptional activity (115,164,165). This epigenetic pathway is further reinforced in Clec4e-mediated response, in which ATF3 inhibits the downstream NF-κB/JNK pathway and reduces TNF-α and CCL2 secretion (136). Moreover, ATF3 also promotes GDF15 expression to foster the adaptation of macrophages to metabolic stress (125). Collectively, by counteracting the effects of ATF1, 2, 4, 5 and 6, ATF3 and ATF7 serve as critical rheostats in maintaining macrophage homeostasis.
ATF2 and ATF4 display dual regulatory activities that depend on the nature, strength and duration of environmental stimuli. As a key regulator in Toll-like receptor (TLR) signaling, ATF2 undergoes p38 MAPK-dependent phosphorylation at Thr71 (107,166), through which it promotes the expression of pro-inflammatory mediators, such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), TNF-α, IL-6 and IL-1β (167-172). This process is further amplified by the formation of AP-1 complex with c-JUN, which enhances the production of nitric oxide (NO) and prostaglandin E2 (PGE2) (173). Notably, ATF2 is highly expressed in M1 macrophages within the white adipose tissue from obese mice, where its phosphorylation is induced by ROS and LPS (139) and sustains the pro-inflammatory phenotype by suppressing the expression of anti-inflammatory ATF3 (174). Furthermore, ATF2 forms heterodimers with HDAC1 to induce Socs-3 transcription to negatively regulate TLR4-mediated inflammation (118), demonstrating its dual regulatory nature. Under metabolic stress, such as cholesterol accumulation, ATF4 is activated via the protein kinase R-like ER kinase (PERK)-eukaryotic translation initiation factor 2 alpha (eIF2α) pathway. During transient stress, ATF4 promotes macrophage survival, whereas persistent stress induces the expression of C/EBP homologous protein (CHOP), triggering macrophage apoptosis along with the release of inflammatory mediators (175,176). In obese microenvironments, ATF4 acts as a metabolic stress sensor, transcriptionally upregulating protein disulfide isomerase a3 (PDIA3) to drive TNF-α, IL-6 and chemokine (C-C motif) ligand 2 (CCL2) production, thereby intensifying adipose tissue inflammation (177). The dual regulatory role of ATF4 is further reflected in its ability to influence antioxidant defenses. Knockdown of ATF4 reduces NRF2 activity and HO-1 expression, leading to attenuated antioxidant defenses and increased NO production (82). Overall, ATF2 and ATF4 act as context-dependent signal integrators that fine-tune macrophage activation, coordinating inflammatory, oxidative and metabolic pathways to define the amplitude and persistence of cytokine responses.
ATFs exert multidimensional roles in macrophage polarization via metabolic reprogramming and signal transduction. ATF1 drives macrophage polarization toward the antioxidant 'Mhem' phenotype through coordinated regulation of iron and lipid metabolism. This involves the induction of HO-1, which promotes the degradation of heme into iron and antioxidant metabolites (biliverdin and bilirubin) to help mitigate oxidative stress. Furthermore, ATF1 activates liver X receptor-β (LXR-β) to augment the expression of cholesterol efflux proteins ABCA1 and ApoE, by which it reduces lipid accumulation and prevents foam cell formation (178). Phosphorylated ATF1 further improves macrophage adaptation to intraplaque hemorrhage, thus maintaining the homeostasis under dural iron-lipid stress (178). ATF2 deficiency (THP-ΔATF2) induces abnormal macrophage morphology, whereas ATF2 overexpression (THP-ATF2) triggers a rounded, flattened morphology resembling M1 macrophages, along with elevated expression of MHC class II, IL-1β and interferon-γ-induced protein 10 (133). Mechanistically, ATF2 drives M1 polarization by enhancing glycolytic flux. Metabolomic profiling confirmed that those ATF2-overexpressing macrophages recapitulate the classical M1 metabolic signatures (139).
ATF3 also plays a pivotal role in macrophage polarization. Overexpression of ATF3 suppresses the differentiation of M1 (pro-inflammatory) macrophage, as evidenced by the decreased expression of M1 markers such as TNF-α and CD11c in adipose tissues, thereby attenuating local inflammation (179). In atherosclerotic lesions, ATF3 mediated inhibition of M1 polarization reduces inflammatory responses and lowers the risk of plaque rupture. In tumor microenvironments, cyclophosphamide (CTX)-induced pro-tumor macrophage traits are reversed in ATF3-knockout mice, shifting macrophages toward anti-tumor phenotypes (147). ATF3 also enhances M2 (anti-inflammatory) macrophage program through indirect pathways. ATF3 suppresses the Nrf2/ARE signaling pathway, leading to the death of renal tubular epithelial cells (RTECs). The resulting apoptotic RTECs release miR-1306-5p-containing exosomes, which in turn inhibit M1 activation while enhance M2 polarization (180). In placental accreta spectrum (PAS) lesions, upregulated ATF3 facilitates M2 macrophage polarization by enhancing the expression of PD-L1, which interacts with PD-1 to strengthen the immunosuppressive microenvironment (181). Conversely, during receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis, ATF3 overexpression promotes osteoclast formation and activity. ATF3 directly binds to the promoters of key transcription factors c-Fos and NFATc1, to drive macrophage-to-osteoclast differentiation. However, as a pro-inflammatory ATF member, ATF4 could otherwise promote M2 program (122,182). Upon hypoxic insult, ATF4 upregulates the expression of M-CSF in hemangioma stem cells (HemSCs), to facilitate M2 macrophage polarization (183). ATF4 also activates RANKL signaling by promoting NFATc1 expression, synergistically regulating the differentiation of osteoclasts (184,185). Furthermore, ATF4 drives the transition of vascular smooth muscle cells (SMCs) into foam cells, as evidenced by the upregulation of macrophage markers such as Cd68 and Lgals3 (186).
Under various pathological conditions, macrophages exhibit disease-specific functional plasticity. Emerging evidence indicates that members of the ATF family in macrophages display distinct and highly disease-specific activation patterns rather than a uniform stress response. Overall, ATF1, ATF2, ATF4, ATF5 and ATF6 are persistently upregulated in most inflammatory, metabolic and stress-related diseases, acting primarily as transcriptional activators that drive pro-inflammatory programs or adaptive stress responses. By contrast, ATF3 and ATF7 are predominantly induced as negative feedback regulators to restrain excessive inflammation or mediate innate immune memory. Importantly, the direction and magnitude of these expression changes are strongly context-dependent. For instance, ATF1 is markedly upregulated during sepsis and atherosclerosis but suppressed in tissue hemorrhage (150,153,178). In addition, ATF3 is rapidly induced during the early hyperinflammatory phase of sepsis to attenuate cytokine storm, whereas its sustained overexpression at later stage contributes to immunosuppression (142,161,162,187). Furthermore, ATF4 is downregulated during early acute sepsis, but is elevated under chronic metabolic stress (132,177,182,186). These observations suggest that ATF activation is finely orchestrated by disease-specific microenvironments through the selective engagement of distinct upstream signaling pathways. A detailed summary of ATF expression dynamics in various diseases and their pathophysiological consequences, along with related signaling pathways, is presented in Table II.
In infectious and immune-related diseases, macrophages situate at the first line of defense (1,188). During the early inflammatory phase, they rapidly recognize and phagocytose pathogens, release pro-inflammatory cytokines and recruit other immune cells to the infection sites, thereby amplifying host defense (189-191). However, excessive or prolonged activation can lead to systemic inflammatory syndromes, such as sepsis and cause secondary tissue injury (191,192). As inflammation resolves, macrophages adopt a reparative phenotype that clears apoptotic cells and produces anti-inflammatory mediators to promote tissue repair (193). Moreover, macrophages can acquire innate immune memory, known as trained immunity, which enables them to respond more effectively to secondary challenges through enhanced cytokine production and antimicrobial activity (194). By contrast, excessive suppression of macrophage activation impairs pathogen clearance and increases vulnerability to secondary infections, thereby disturbing immune homeostasis (195,196). In chronic inflammatory conditions, persistent low-grade activation or failure to transition into a reparative state drive sustained cytokine production, tissue remodeling and fibrosis, all of which predispose to disease progression (197). During bacterial infection, ATF3 exhibits pathogen-specific regulatory effect. Specifically, ATF3 enhances macrophage clearance of Staphylococcus aureus by directly binding to the promoters of antimicrobial peptide genes, such as Reg3β and S100A8/9, to bolster host defense (198). Conversely, during Pseudomonas aeruginosa or uropathogenic E. coli infection, ATF3 attenuates host resistance by downregulating pro-inflammatory factors (TNF-α and IL-6) and upregulating IL-10 (138). Following Leishmania infection, ATF3 expression reaches the highest after 1-h of infection, which establishes an anti-inflammatory environment in favor of parasite survival (163). ATF4 enhances macrophage antioxidant capacity by promoting HO-1 expression to limit NO production, thus creating an environment conducive to Leishmania amazonensis survival (82). By contrast, ATF7 contributes to innate immune memory by modulating STAT1/NF-κB-dependent inflammatory networks (such as CXCL9/10), thus enhancing macrophage-mediated responses against bacteria and fungi (119). In antiviral immunity, ATF2 activates the p38 MAPK/ATF2/AP-1 axis to promote the secretion of interferon-stimulated genes and IFN-β, thereby enhancing the resistance to vesicular stomatitis virus, Newcastle disease virus and herpes simplex virus (105). However, ATF3 may compromise murine norovirus clearance by suppressing type I interferon expression or impairing Mx1 activity (125). During Aspergillus fumigatus infection, ATF4 is upregulated via the TLR4/LOX-1/MAPK pathway, which modulates corneal macrophage barrier function and disease severity (199).
In acute inflammatory diseases, ATFs exhibit a dynamic and balanced role. Hyperactivation of ATF1 induces excessive pro-inflammatory cytokine release from macrophages, contributing to systemic inflammatory response syndrome (SIRS) or septic shock (153). ATF3 plays a complex part at different septic stages. In LPS-induced septic models, ATF3 reduces TNF-α, IL-6 and IL-1β levels in plasma, potentially mitigating early-stage hyperinflammation (162,187). Additionally, ATF3 promotes macrophage polarization towards the M2 phenotype, enhances the expression of anti-inflammatory factors such as Arg-1 and PPARγ, but suppresses M1 markers (such as iNOS and TNF-α), thereby alleviating inflammatory responses and facilitating tissue repair (142). However, during the sepsis-associated immunosuppression phase, persistent ATF3 overexpression impairs immune responses and increases susceptibility to secondary infections (161). During septic progression, reduced ATF4 expression in P1 cells leads to diminished gMacs, which promotes bacterial translocation across the epithelial barrier and worsens systemic inflammation (132). ATF5 contributes to sepsis-associated liver injury by promoting the accumulation of inflammatory macrophages (CD45+ CD11b+ Ly6C+) through an NF-κB-dependent pathway, leading to the release of inflammatory cytokines (154). Excessive activation of ATF1 is also associated with the progression of SIRS and sepsis by enhancing macrophage cytokine secretion (153).
In chronic inflammatory diseases, ATF family members influence macrophage polarization and amplify inflammatory signaling, contributing to disease pathology. In LPS-induced acute lung injury, ATF2 exacerbates pulmonary inflammation by upregulating MIP-2 to further recruit macrophages (140), while ATF6 intensifies macrophage activation and cytokine release via ER stress signaling (200). ATF2 activation also amplifies TLR signaling cascades and exacerbating chronic inflammation (107,170). Moreover, upon LPS stimulation, ATF2 promotes IL-23 secretion in macrophages, which then induces pathogenic Th17 cell expansion in autoimmune disorders, such as multiple sclerosis, inflammatory bowel disease and rheumatoid arthritis (201). In organ-specific inflammatory injuries, ATF3 exhibits bidirectional regulatory properties. ATF3 attenuates renal ischemia-reperfusion injury (IRI) by inhibiting TLR4/NF-κB pathway (165). However, IRI-induced ATF3 overexpression in RTECs exacerbates ferroptosis and promotes the release of exosomes carrying miR-1306-5p. These exosomes, upon uptake by macrophages, drive M2 polarization and accelerate interstitial fibrosis (180). In alcohol-induced immunosuppression, ATF3 upregulation leads to enhanced Kupffer cell tolerance to LPS and reduced TNF-α production, impairing pathogen clearance and exacerbating alcoholic liver disease (143). In dextran sulfate sodium (DSS)-induced colitis, activation of the ATF4-CHOP pathway drives excessive release of inflammatory cytokines, intensifying intestinal inflammation and epithelial damage (176). It was also noted that ATF6 deficiency impairs macrophage antibacterial responses, worsening intestinal inflammation (106). During hepatic ischemia-reperfusion injury, ATF6 aggravates tissue damage by enhancing NF-κB activation and suppressing the anti-inflammatory Akt-GSK3β signaling pathway in Kupffer cells (158). However, during early acute liver injury, ATF6 activation via ER stress promotes IL-1α production, activating HSCs and driving liver fibrosis (159). ATF6 deficiency in microglia attenuates inflammation in autoimmune encephalomyelitis by promoting NF-κB p65 degradation (202).
In metabolic disorders, adipose tissue macrophages produce pro-inflammatory cytokines including TNF-α, IL-6 and IL-1β to impair insulin signaling and disrupt adipocyte homeostasis, thereby contributing to insulin resistance and metabolic syndrome (203-207). ATF4 exacerbates chronic inflammation in adipose tissue, driving the progression of metabolic syndrome and type 2 diabetes mellitus (177,182). By contrast, ATF3 mitigates obesity-associated inflammation by suppressing TLR4-mediated macrophage activation. Therefore, mice deficient in ATF3 exhibit aggravated obesity-related inflammation and insulin resistance, underscoring its protective role in maintaining metabolic homeostasis (162). Conversely, ATF2 activation intensifies macrophage M1 program within adipose tissue by perpetuating an 'inflammation-hypoxia-ROS' vicious cycle, worsening insulin resistance and metabolic syndrome (173). Additionally, ATF2 aggravates inflammation and metabolic disturbances in white adipose tissue by suppressing ATF3 expression, further enhancing M1 program (139).
In atherosclerosis, macrophages take up OxLDL to form foam cells and orchestrate inflammatory and reparative responses within vascular lesions, thereby playing a pivotal role in plaque formation and progression (28,34,36). ATFs influence disease progression by regulating macrophage lipid metabolism and plaque stability. ATF3 exerts an atheroprotective effect through multiple mechanisms. ATF3 suppresses cholesterol 25-hydroxylase to reduce oxidized cholesterol accumulation and promotes macrophage RCT, thereby limiting foam cell formation (148,208). Furthermore, ATF3 stabilizes plaque structure by inhibiting macrophage apoptosis and the release of inflammatory mediators, lowering the risk of myocardial infarction and stroke (134). ATF3 deficiency impairs ABCA1-mediated cholesterol efflux, aggravating oxLDL-induced lipid deposition (136). Moreover, ATF1 induces Mhem macrophage polarization and activates the HO-1/LXR-β pathway to enhance cholesterol efflux and iron chelation, thereby delaying plaque progression and rupture (150,178). By contrast, ATF6 activation positively correlates with the severity of atherosclerotic lesion and accelerates plaque development by facilitating cholesterol accumulation (157). ATF4 drives necrotic core expansion and plaque destabilization via switching an SMC phenotype ('transdifferentiated SMCs' which express both macrophage and fibroblast markers) and inducing CHOP-dependent cell apoptosis (186). As a downstream effector of PKCθ signaling, ATF2 upregulates CD36 to accelerate foam cell formation and plaque progression. PKCθ-deficient mice exhibit blunted ATF2 activation, along with reduced CD36 levels and markedly smaller plaque areas (149). Moreover, ATF1 deficiency impairs the segregation of iron and lipid within macrophages, resulting in their colocalization, which disrupts iron metabolic homeostasis, delays hematoma clearance following tissue hemorrhage and exacerbates oxidative stress-related damage (150). Collectively, ATFs spatiotemporally modulate macrophage inflammatory state, thus influencing the pathological progression of metabolic and cardiovascular diseases.
Macrophages are major immune infiltrates that regulate angiogenesis, immune suppression, antigen presentation and metastatic dissemination in the tumor microenvironment (1,209). In bone lesions, macrophages further contribute to osteoclast differentiation and tumor-induced bone destruction (210-212). ATF4 regulates tumor-associated macrophage polarization and promotes the differentiation of monocyte-macrophage precursors into osteoclasts to enhance bone resorption. In parallel, ATF4 exacerbates inflammation through the NF-κB signaling axis, further accelerating cancer-associated bone destruction (184). Unlike ATF4, ATF3 exhibits functional heterogeneity in tumor immunomodulation. Tumor-derived IL-1β upregulates ATF3 in bone marrow hematopoietic stem cells, driving myeloid precursor expansion and increasing peripheral CD11b+ myeloid cells, including TAMs, tumor-associated macrophages. In a breast cancer model, ATF3 deficiency inhibits monocyte-macrophage differentiation, while its overexpression serves as an early biomarker distinguishing malignant lesions (213). Furthermore, ATF3 activates immune checkpoint pathways to amplify CD14+ immunosuppressive macrophages in PAS lesions, exacerbating pathological progression (181).
In neurological diseases, macrophages and microglia not only phagocytose neuronal debris but also release neuroregulatory substances and inflammatory mediators, thereby modulating neuronal survival and degeneration (214,215). ATF2 promotes neuroinflammatory cascades in multiple sclerosis, Alzheimer's disease (AD) and Parkinson's disease by promoting pyroptosis in macrophages and microglia (139). Additionally, ATF4 is the core transcription factor for AD microglial cells to integrate stress response, by inducing them to enter the neurotoxic state and mediating neurodegeneration through lipid secretion (216). Furthermore, ATF4 synergizes with HIF-1α under hypoxic conditions in infantile hemangioma (IH) proliferative phases, driving M2 macrophage infiltration to fuel lesion expansion (183).
Targeting ATFs in macrophages offers a promising therapeutic strategy, as these key transcription factors orchestrate diverse macrophage functional states, thereby enabling precise modulation of disease processes (Table III). Currently, therapeutic strategies targeting ATFs can be broadly divided into three categories: Direct intervention, which employs small-molecule agonists or inhibitors and gene-editing approaches to directly modulate ATF expression or activity; indirect modulation, which targets upstream signaling pathways or downstream effector molecules to influence ATF-dependent regulatory networks; and emerging innovative techniques, including proteolysis-targeting chimera (PROTAC)-mediated protein degradation, peptide or peptidomimetic modulators and molecular glue technologies, which offer more efficient and specific manipulation of ATF functions.
Blocking the function of ATFs through small inhibitors, short interference (si)RNA, or gene-editing technologies offers a strategic approach to precisely control disease states driven by macrophages. Targeted inhibition of ATF1 reduces osteoclastogenesis by suppressing the miR-214-5p/ITGA7 axis, suggesting a novel direction for osteoporosis treatment (217). The traditional herbal medicine, HangAmDan-B (HAD-B), reduces LPS-induced NO and PGE2 production in macrophages by blocking ATF2 phosphorylation, consequently limiting tumor-promoting activity of TAMs in gastric and colorectal cancers (166). Ginsenoside Rc markedly reduces LPS-induced TNF-α release by suppressing the p38/ATF2 pathway, presenting a candidate strategy for rheumatoid arthritis and other inflammatory diseases (168). Moreover, the traditional compound formula Qingfei Paidu Decoction and its active component wogonoside, inhibit ATF2 phosphorylation and enhance its ubiquitin-mediated degradation, curbing macrophage-mediated inflammation and showing potential as adjunct therapies for coronavirus pneumonia and colitis (169). Moreover, extracts from Vaccinium oldhamii stems reduce ATF2 nuclear accumulation, which blocks MAPK signaling pathway activation and limits macrophage-driven osteoclast differentiation to mitigate bone resorption (218). Exosomes derived from adipose-derived stem cells (ADSCs) or mesenchymal stem cells attenuate ATF2 expression, resulting in reduced NF-κB activation, decreased ROS generation and diminished macrophage infiltration, thereby mitigating vascular dysfunction (219). Similarly, perfluorocarbon (PFC) could reduce inflammatory cell infiltration and alleviate acute lung injury by decreasing ATF2 activity in macrophages (140). The dynamic modulation of ATF3 requires consideration of disease progression. During the early hyperinflammatory phase of sepsis, induction of ATF3 expression in macrophages may counteract cytokine storms, while its inhibition during the immunosuppressive phase may help mitigate secondary infection risk (138). ATF4 suppression also requires meticulous evaluation due to its dualistic effect. The application of small-molecule inhibitors or siRNA-mediated knockdown of ATF4 has been shown to attenuate M-CSF secretion and limit M2 macrophage infiltration (183), which in turn impedes the progression of infantile hemangioma while simultaneously counteracts the pro-tumorigenic properties of TAMs (122). Inhibiting ATF4 can also mitigate Leishmania infection (82), though excessive inhibition might compromise the antifungal defense. Therefore, therapies targeting ATF4 must balance the anti-inflammatory outcomes with host immune protection (199). Naringenin, a flavanone compound found in grapefruits and other citrus fruits, could reduce ATF6 nuclear translocation and activity, by which it decreases the expression of ER stress markers while enhances the expression of cholesterol efflux genes (ABCA1, ABCG1 and LXRα). This action promotes macrophage cholesterol efflux, reducing atherosclerotic plaque formation (157). Furthermore, siRNA-mediated ATF6 knockdown alleviates macrophage-driven inflammation and fibrosis, highlighting its potential for treating chronic inflammatory disorders (159).
By contrast, activating or overexpressing ATFs through gene-editing technologies (such as adenoviral vectors) and small-molecule agonists offer an approach to enhance anti-inflammatory, anti-infectious and tissue-repairing functions of macrophages. Developing small-molecule drugs or gene therapies to enhance ATF1 activity presents a potential intervention for atherosclerosis by inducing the protective macrophage subset (178). ATF2 activation inhibits viral replication, with its agonists serving as potential therapeutics for chronic viral infections (167). High-salt diets have also been proposed as a strategy to enhance antiviral immunity via ATF2-mediated pathways in macrophages (105). Targeted activation or overexpression of ATF3 exhibited a promising therapeutic potential in inflammatory diseases, infections and metabolic disorders. Gene-editing technologies, such as lentiviral and adenoviral deliveries (Ad/ATF3) of ATF3, alleviate excessive inflammation following Mycoplasma pneumoniae infection (160), protect renal tubular cells from inflammatory damage (165) and reduce the risk of atherosclerosis development (134). ATF3 overexpression further diminishes LPS-induced pro-inflammatory cytokine release, thus improving the outcomes in chronic inflammatory diseases (138,164). In bacterial infections, enhancing ATF3 activity optimizes immune responses and promotes macrophage activation, migration and bactericidal activity, coupled with increased host clearance of Staphylococcus aureus (144). Across multiple disease models, berberine and metformin have been demonstrated to enhance ATF3 expression (162,220) and attenuates LPS-induced production of pro-inflammatory cytokines from macrophages (164). In particular, metformin exerts its anti-inflammatory effects by engaging the AMPK/ATF3 signaling axis, through which it potently manages inflammation associated with type 2 diabetes and obesity (187). ATF4 may represent a novel strategy for treating sepsis-related intestinal injury (142). Enhancing ATF4 expression through pharmacological or gene therapies help the restoration of monocyte differentiation into gMacs, thereby strengthening intestinal barrier function, reducing bacterial translocation and mitigating sepsis-induced inflammatory damage (132). In type 2 diabetes, upregulating ATF4 can improve insulin resistance by promoting M2 macrophage polarization (182). Targeting ATF7-mediated epigenetic modifications offers a strategy to refine vaccine adjuvant efficacy, by promoting macrophage activation and memory formation, ultimately advancing the development of more durable and effective vaccine-induced protection (119).
However, the clinical translation of strategies that directly targeting ATFs faces multiple inherent challenges. The first obstacle lies in the intrinsic 'undruggability' of these transcription factors. The interaction surfaces of ATFs with DNA or partner proteins are typically broad and shallow, lacking well-defined pockets that allow high-affinity binding of small molecules, thereby rendering the development of conventional inhibitors or agonists extremely difficult. Second, substantial structural similarity exists among ATF family members, particularly within their DNA-binding domains. As a result, small molecules targeting ATFs may suffer from poor subtype selectivity, potentially leading to off-target toxicity. Therefore, developing compounds capable of precisely distinguishing individual ATF isoforms or specific ATF homo- and heterodimers remains a formidable challenge, which warrants further intensive investigations (221).
Given the challenges associated with direct targeting, indirect modulation of upstream signaling pathways or downstream effector molecules have emerged as an attractive alternative strategy. Studies show that PERK inhibitors (such as integrated stress response inhibitor (ISRIB)) suppress the PERK-eIF2α-ATF4 pathway, thereby reducing foam cell formation and alleviating atherosclerotic plaque burden (186). The small molecule Limonin blocks the PERK-ATF4-CHOP axis to attenuate inflammatory cytokine release (186). Moreover, drugs, such as FTY720, modulate ATF4 expression via histone deacetylase 4 (HDAC4), thus reducing osteoclastic activity in inflammatory bone diseases (184). For ATF2, multiple upstream intervention strategies have been explored, including p38 MAPK inhibitors (SB203580) and JNK inhibitors (SP600125), which reduce inflammation by suppressing ATF2 phosphorylation (166). Additionally, PAR1 antagonists (such as SCH79797) or PKCθ inhibitors decrease ATF2 activation to improve insulin sensitivity (149). Novel compounds, such as bis(5-methyl)2-furylmethane and Javamide-II, reduce inflammatory mediator production by suppressing the p38 MAPK/ATF2 pathway. Particularly, Javamide-II manifests lower systemic toxicity compared with BFNM due to its selective inhibition of specific inflammatory cytokines without affecting TNF-α or IL-1β (107,170). For ATF1, anti-CD14 monoclonal antibodies (MY4) showed potential as an adjunctive therapy for sepsis by inhibiting PGN-induced ATF1 activation (153). Similarly, AMPK agonists (such as AICAR) induce ATF3 expression and offer therapeutic benefits for metabolic disorders (162). Conversely, the NRF2 inhibitor, trigonelline hydrochloride, markedly reduces parasitic loads in the liver and spleen by diminishing ATF3 expression (163). Additionally, NF-κB inhibitors [such as dehydroxy-methylepoxy-quinomicin (DHMEQ)] alleviate sepsis-associated liver damage by inhibiting ATF5-mediated pro-inflammatory macrophage differentiation (154).
Nevertheless, indirect modulation strategies also present substantial limitations. The fundamental drawback of such approaches lies in their lack of specificity. Upstream kinases or signaling pathways targeted by these interventions are often ubiquitously expressed across multiple cell types, leading to severe off-target effects and systemic toxicity. Second, the high redundancy of intracellular signaling networks frequently triggers compensatory activation of alternative pathways, thereby diminishing therapeutic efficacy or even resulting in treatment failure. Moreover, the regulatory effects of pathway-level interventions tend to attenuate along the signaling cascade and are highly dependent on the disease microenvironment, rendering the ultimate modulation of specific ATFs indirect and unpredictable. Finally, this relatively 'coarse-grained' mode of regulation is insufficient to precisely govern the bidirectional functions of ATF members or their heterodimeric interactions, limiting its ability to achieve fine-tuned control over their specific physiological and pathological activities.
To overcome the aforementioned obstacles, researchers have explored intervention strategies such as PROTACs and molecular glues, aiming to target traditionally 'undruggable' molecules through novel mechanisms of action. The advent of PROTAC technology has provided a new avenue for addressing such targets (222-225). A PROTAC molecule contains two functional domains, one binds to the target protein and the other recruits an E3 ubiquitin ligase, thereby tagging the target for selective proteasomal degradation (226,227). Although no highly potent small-molecule ligands have yet been reported for ATF proteins, researchers are creatively adapting the PROTAC concept for transcription factor degradation. One proposed generalizable strategy involves using DNA fragments recognized by transcription factors as targeting elements, which are linked to E3 ligase ligands to induce ubiquitination and subsequent degradation of transcription factor-DNA complexes (228). For example, the antioxidant response factor NRF2, a bZIP transcription factor that heterodimerizes with small Maf proteins to bind to the AREs, can be targeted by a chimeric molecule termed 'ARE-PROTAC', which successfully induced simultaneous degradation of the NRF2-MafG complex and synergistically suppressed Nrf2 signaling (229). This design suggests that DNA-binding factors, such as ATF/CREB, DNA mimetics or DNA-derived small molecules, could serve as decoy binding modules to achieve targeted degradation via the PROTAC mechanism. Unlike conventional inhibitors that merely block protein activity, this approach eliminates the target protein itself, offering more complete and durable inhibition while potentially alleviating feedback activation caused by persistent protein accumulation. Nevertheless, applying this technology to ATF targeting remains challenging. It requires the discovery of small, high-affinity ligands specific for ATF proteins and the relatively large molecular weight of PROTACs limits their intracellular delivery, especially into macrophages. To address this issue, recent studies have explored macrophage-targeted nanocarrier systems utilizing surface receptors such as citrate or scavenger receptors, enabling PROTAC accumulation and selective degradation at the inflammatory or tumor sites (230,231).
Building on this concept, molecular glues have emerged as another promising and simplified strategy for targeted protein degradation. Molecular glues are typically single small molecules that 'glue' the target protein and an E3 ubiquitin ligase, thereby promoting their interaction and inducing target degradation (232). Unlike PROTACs, molecular glues do not require a linker and can directly induce contact between transcription factors and E3 ligases, offering a simpler architecture and easier optimization. The classic example is lenalidomide, which remodels the substrate-binding surface of the CRBN E3 ligase, enabling selective recognition and degradation of the transcription factors IKZF1 and IKZF3 (233). This mechanism holds particular interest for targeting ATF family members. First, molecular glues generally have much smaller molecular weights than PROTACs, conferring superior membrane permeability and drug-like properties, thereby facilitating macrophage penetration. Second, their activity does not depend on identifying traditional ligand-binding pockets, which is advantageous for transcription factors lacking well-defined active sites. Nevertheless, challenges remain, including the difficulty of identifying suitable adaptor molecules and the dependency on specific E3 ligases, which may restrict the target spectrum.
In addition, peptide mimetics and interfering peptides offer new avenues for disrupting transcription factor activity. Researchers have developed a dominant-negative form of ATF5 (dn-ATF5) by deleting its N-terminal activation domain while retaining the bZIP dimerization and DNA-binding regions and further conjugated it to an HIV-TAT sequence to generate a cell-permeable synthetic peptide (CP-dn-ATF5) (234,235). This peptide can get into the nucleus and compete with endogenous ATF5 for partner proteins or DNA-binding sites, thereby blocking its transcriptional activity. The advantages of peptide-based therapies lie in their high specificity and flexible design. With advances in peptide drug modification techniques, such as cyclization, D-amino acid substitution and nanocarrier encapsulation, peptide-based ATF inhibitors are expected to become an important complement to small-molecule strategies.
Of note, enhancing the spatiotemporal specificity of ATF-targeting interventions is critical for improving therapeutic efficacy and safety. Advanced delivery systems are under development to achieve tissue- or cell-specific targeting, ensuring that drugs could be selectively delivered into macrophages in affected tissues (236). Encouragingly, advances in nanocarrier and bioengineering technologies now enable the precise delivery of therapeutic agents to macrophages, allowing for selective modulation of ATF-associated pathways without affecting other cell types. For instance, in atherosclerosis models, macrophage-targeted nanoparticles have been utilized to deliver drugs directly to macrophages within the plaques (237). These nanoparticles substantially improved the pharmacokinetic profiles of the encapsulated drugs, enhancing their accumulation at the lesion site and promoting the therapeutic efficacy. Similarly, in viral pneumonia models, lipid nanoparticles conjugated with macrophage-specific antibodies (such as anti-F4/80) have been used to deliver siRNA to lung macrophages, effectively silenced upstream inflammatory molecules such as TAK1 (238). These findings underscore the feasibility of macrophage-targeted delivery for therapeutic interventions and highlight the substantial potential of integrating delivery technologies into ATF-targeted therapies. Therefore, innovative targeted technologies coupled with advanced delivery systems, might overcome the barrier of ATFs-targeted therapies and accelerate their clinical translation.
By integrating mechanistic insights and disease-specific evidence, this review delineated how ATFs orchestrate macrophage development, survival, migration, phagocytosis, activation, cytokine secretion and polarization and how these regulatory axes converge to shape macrophage-driven pathology in infectious, inflammatory, metabolic, oncologic and neurodegenerative settings. Through transcriptional control of macrophage-expressed genes across diverse tissues and organs, ATFs are not only implicated in the initiation and progression of multiple disorders but also emerged as nodal regulators integrating pathophysiological signals with immune and metabolic reprogramming (Fig. 3). Of note, ATFs exhibit complex context-dependent versatility in macrophage-mediated immune responses. Mechanistically, the effector outcomes of ATFs largely hinge on the variety of stimuli and upstream signaling pathways in macrophages. For instance, TLR2 activation by Gram-positive bacteria induces moderate inflammation, during which ATF3 promotes antimicrobial gene expression, whereas LPS-TLR4 signaling from Gram-negative bacteria elicits strong NF-κB and IRF3 activation that rapidly induces ATF3 as a negative feedback regulator to suppress excessive cytokine production (115,144,161,239-242). In addition, the intensity and duration of stimulation further shape ATF activity. During early sepsis, ATF3 alleviates hyperinflammation and promotes tissue recovery, whereas its sustained overexpression contributes to late-phase immunosuppression and secondary infections (142,161,162,187). Transient stress activates ATF4 via the PERK-eIF2α pathway, upregulating adaptive and survival-related genes to enhance macrophage resilience; however, prolonged stress leads to persistent ATF4-CHOP signaling, promoting apoptosis and pro-inflammatory mediator release (175,176). Similarly, ATF6 activation under acute ER stress (such as triggered by pattern-recognition receptors) induces chaperone and autophagy programs that facilitate pathogen clearance (106,155,159). By contrast, during chronic metabolic stress, such as obesity or hyperlipidemia, sustained ATF6 activation increases cholesterol biosynthesis, suppresses cholesterol efflux, promotes foam cell formation and amplifies inflammatory responses (156,157). In addition, as bZIP transcription factors, ATF family members exert highly context-dependent effects determined by their interacting partners and chromatin accessibility. Microenvironmental factors, such as oxidative stress, metabolic stress and ER stress, not only regulate ATF activation levels but also influence their combination with c-Jun, CHOP, C/EBP, or HDAC1, thereby switching their transcriptional output between activation and repression. Upon inflammatory stimulation, ATF2 forms an AP-1 complex with c-Jun to transcribe Inos and Cox-2 expression, promoting NO and PGE2 production (156,168-173), while during inflammatory resolution stage, ATF2 recruits HDAC1 to transcribe Socs3 and suppresses excessive TLR4 signaling (162). ATF3 similarly shifts its binding preference. Under mild stimulation, ATF3 partners with c-Jun or CHOP to activate antimicrobial and metabolic genes, but under strong LPS-TLR4 signaling, ATF3 recruits HDAC1 to repress NF-κB target genes (115,138,143,163-165). The functional direction of ATF4 also depends on its binding partners. ATF4-NRF2 heterodimers drive adaptive metabolism and cell survival (82), while heterodimers formed with CHOP mediate inflammation and cell apoptosis (175,176). Likewise, ATF6 enhances cytoprotective UPR genes such as Grp78 and Xbp1 during acute stress, but under chronic lipotoxicity, it preferentially targets inflammatory and lipid metabolic genes to amplify macrophage inflammation (156). Moreover, changes in chromatin accessibility under infection or metabolic stress might reshape cis-element exposure, determining the preferential binding sites of ATFs and their downstream transcriptional outcomes. Collectively, these mechanisms highlight ATFs as crucial 'rheostats' that balance inflammatory signaling, metabolic adaptation and stress responses in macrophages. However, this functional duality also underscores the caution in therapeutic targeting: suppressing ATFs in pathogenic conditions must not compromise their protective roles and activating them to resolve inflammation should avoid inducing immunosuppression. A deeper understanding of the context-dependent mechanisms of ATF signaling is be essentially required.
It is noteworthy that alternative splicing may represent another critical but insufficiently investigated layer of ATF modulation. Except for ATF4, most ATF family members possess alternatively spliced isoforms, although their roles in macrophages remain largely unknown. Distinct ATF1 isoforms differ in DNA-binding affinity and subnuclear localization (243), whereas multiple ATF2 variants, including the human isoforms ATF2-sm and ATF2 (SV5) as well as the murine variant Atf2(Δ8,9) (244-246), exhibit reduced transcriptional activity and may alter gene expression by modifying nuclear-cytoplasmic trafficking or by interfering with canonical ATF2/AP-1 dimerization. Moreover, a truncated ATF3 isoform (ΔZip) lacking the leucine-zipper domain antagonizes the transcriptional repressive function mediated by full-length ATF3 (247), while cytoplasmic ATF7-4 acts as a molecular 'decoy' to sequester upstream kinases responsible for ATF7/ATF2 phosphorylation (77). These findings suggest that alternative splicing may establish negative-feedback circuits and intermolecular counter-regulatory balances responsible for the precise modulation of ATF function. Although their implications in macrophages remain to be defined, exploring ATF splicing isoforms represents an important direction for elucidating the dynamic regulatory mechanisms by which ATFs regulate immune homeostasis and stress responses.
The central involvement of ATFs in macrophage-driven pathologies positions them as promising therapeutic targets and biomarkers. On the one hand, ATF expression dynamics serve as diagnostic or prognostic indicators. Rapid ATF3 upregulation (8-fold within 24 h) in silicosis macrophages is an early diagnostic biomarker for silicosis detection (248). Additionally, due to its plaque-stabilizing effects on atherosclerosis, ATF3 expression aids in the identification of high-risk patients and guides personalized treatments (134). On the other hand, single-target interventions may be limited by compensatory mechanisms, necessitating the need for multi-target synergistic approaches for the regulation of macrophage functionality. Lastly, ATF-targeting strategies such as siRNA interference, gene editing, or more advanced protein degradation technologies such as PROTACs, all face common challenges related to in vivo delivery efficiency, tissue and cell specificity, immunogenicity and the durability of their effects. Although these interventions have shown remarkable efficacy in preclinical models, extensive studies are still required to validate their safety, efficacy and controllability in complex physiological settings and to further optimize targeted delivery systems to facilitate clinical translation.
Not applicable.
YCL conceived the study, wrote the manuscript and prepared the figures. JWZ, QJC and XTL curated data and collected references. SJR and FS organized the tables. SWL, CLY and CYW reviewed and revised the manuscript. Data authentication is not applicable. All authors approved the final manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
ATFs |
activating transcription factors |
|
ATM |
adipose tissue macrophage |
|
AD |
Alzheimer's disease |
|
bZIP |
basic leucine zipper |
|
CRE |
cAMP response element |
|
C/EBP |
CCAAT/enhancer-binding protein |
|
ER |
endoplasmic reticulum |
|
gMac |
gut-resident macrophages |
|
HSCs |
hematopoietic stem cells |
|
HO-1 |
heme oxygenase-1 |
|
IRI |
ischemia-reperfusion injury |
|
LPS |
lipopolysaccharide |
|
LXR |
liver X receptor |
|
M-CSF |
macrophage colony-stimulating factor |
|
NO |
nitric oxide |
|
oxLDL |
oxidized low-density lipoprotein |
|
PGN |
peptidoglycan |
|
PAS |
placental accreta spectrum |
|
PROTAC |
proteolysis-targeting chimera |
|
RTECs |
renal tubular epithelial cells |
|
SMCs |
smooth muscle cells |
|
SIRS |
systemic inflammatory response syndrome |
|
TLR |
Toll-like receptor |
|
UPR |
unfolded protein response |
Not applicable.
The present study was supported by the Noncommunicable Chronic Diseases-National Science and Technology Major Project (2024ZD0531400,2023ZD0507302), the National Key R&D Program of China (2022YFA0806101), the National Natural Science Foundation of China (81920108009, 82130023, 82570968, 82200923), the Research and Innovative Team Project for Scientific Breakthroughs at Shanxi Bethune Hospital (2024AOXIANG03), the Continuous Funding Program for High-Level Research Achievements at Shanxi Bethune Hospital (2024GSPYJ10 and 2024GSPYJ13) and the IGP Funding from QBRI, Hamad Bin Khalifa University.
|
Chen S, Saeed AFUH, Liu Q, Jiang Q, Xu H, Xiao GG, Rao L and Duo Y: Macrophages in immunoregulation and therapeutics. Signal Transduct Target Ther. 8:2072023. View Article : Google Scholar : PubMed/NCBI | |
|
Mass E, Ballesteros I, Farlik M, Halbritter F, Günther P, Crozet L, Jacome-Galarza CE, Händler K, Klughammer J, Kobayashi Y, et al: Specification of tissue-resident macrophages during organogenesis. Science. 353:aaf42382016. View Article : Google Scholar : PubMed/NCBI | |
|
Kuznetsova T, Prange KHM, Glass CK and de Winther MPJ: Transcriptional and epigenetic regulation of macrophages in atherosclerosis. Nat Rev Cardiol. 17:216–228. 2020. View Article : Google Scholar : | |
|
Ajami B, Bennett JL, Krieger C, Tetzlaff W and Rossi FM: Local self-renewal can sustain CNS microglia maintenance and function throughout adult life. Nat Neurosci. 10:1538–1543. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Bian Z, Gong Y, Huang T, Lee CZW, Bian L, Bai Z, Shi H, Zeng Y, Liu C, He J, et al: Deciphering human macrophage development at single-cell resolution. Nature. 582:571–576. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L, Garner H, Trouillet C, de Bruijn MF, Geissmann F and Rodewald HR: Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature. 518:547–551. 2015. View Article : Google Scholar | |
|
Guan F, Wang R, Yi Z, Luo P, Liu W, Xie Y, Liu Z, Xia Z, Zhang H and Cheng Q: Tissue macrophages: Origin, heterogenity, biological functions, diseases and therapeutic targets. Signal Transduct Target Ther. 10:932025. View Article : Google Scholar : PubMed/NCBI | |
|
Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, Becker CD, See P, Price J, Lucas D, et al: Tissue-resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. 38:792–804. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Rosmus DD, Koch J, Hausmann A, Chiot A, Arnhold F, Masuda T, Kierdorf K, Hansen SM, Kuhrt H, Fröba J, et al: Redefining the ontogeny of hyalocytes as yolk sac-derived tissue-resident macrophages of the vitreous body. J Neuroinflammation. 21:1682024. View Article : Google Scholar : PubMed/NCBI | |
|
Masuda T, Amann L, Monaco G, Sankowski R, Staszewski O, Krueger M, Del Gaudio F, He L, Paterson N, Nent E, et al: Specification of CNS macrophage subsets occurs postnatally in defined niches. Nature. 604:740–748. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kazankov K, Jorgensen SMD, Thomsen KL, Møller HJ, Vilstrup H, George J, Schuppan D and Grønbæk H: The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat Rev Gastroenterol Hepatol. 16:145–159. 2019. View Article : Google Scholar | |
|
Appios A, Davies J, Sirvent S, Henderson S, Trzebanski S, Schroth J, Law ML, Carvalho IB, Pinto MM, Carvalho C, et al: Convergent evolution of monocyte differentiation in adult skin instructs Langerhans cell identity. Sci Immunol. 9:eadp03442024. View Article : Google Scholar : PubMed/NCBI | |
|
Hassnain Waqas SF, Noble A, Hoang AC, Ampem G, Popp M, Strauß S, Guille M and Röszer T: Adipose tissue macrophages develop from bone marrow-independent progenitors in Xenopus laevis and mouse. J Leukoc Biol. 102:845–855. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Perdiguero EG and Geissmann F: The development and maintenance of resident macrophages. Nat Immunol. 17:2–8. 2016. View Article : Google Scholar : | |
|
Sakai M, Troutman TD, Seidman JS, Ouyang Z, Spann NJ, Abe Y, Ego KM, Bruni CM, Deng Z, Schlachetzki JCM, et al: Liver-derived signals sequentially reprogram myeloid enhancers to initiate and maintain kupffer cell identity. Immunity. 51:655–670 e8. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Yona S, Kim KW, Wolf Y, Mildner A, Varol D, Breker M, Strauss-Ayali D, Viukov S, Guilliams M, Misharin A, et al: Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity. 38:79–91. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Murray PJ and Wynn TA: Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 11:723–737. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Martinez FO and Gordon S: The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. 6:132014. View Article : Google Scholar : PubMed/NCBI | |
|
Mills CD: Anatomy of a discovery: m1 and m2 macrophages. Front Immunol. 6:2122015. View Article : Google Scholar : | |
|
Mills CD, Kincaid K, Alt JM, Heilman MJ and Hill AM: M-1/M-2 macrophages and the Th1/Th2 paradigm. J Immunol. 164:6166–6173. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Bosco MC: Macrophage polarization: Reaching across the aisle? J Allergy Clin Immunol. 143:1348–1350. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Locati M, Curtale G and Mantovani A: Diversity, Mechanisms, and Significance of Macrophage Plasticity. Annu Rev Pathol. 15:123–147. 2020. View Article : Google Scholar | |
|
Ivashkiv LB: Epigenetic regulation of macrophage polarization and function. Trends Immunol. 34:216–223. 2013. View Article : Google Scholar : | |
|
Mosser DM and Edwards JP: Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 8:958–969. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT and Sahebkar A: Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 233:6425–6440. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Dong T, Chen X, Xu H, Song Y, Wang H, Gao Y, Wang J, Du R, Lou H and Dong T: Mitochondrial metabolism mediated macrophage polarization in chronic lung diseases. Pharmacol Ther. 239:1082082022. View Article : Google Scholar : PubMed/NCBI | |
|
Gordon S and Martinez FO: Alternative activation of macrophages: Mechanism and functions. Immunity. 32:593–604. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Colin S, Chinetti-Gbaguidi G and Staels B: Macrophage phenotypes in atherosclerosis. Immunol Rev. 262:153–166. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ruytinx P, Proost P, Van Damme J and Struyf S: Chemokine-induced macrophage polarization in inflammatory conditions. Front Immunol. 9:19302018. View Article : Google Scholar : PubMed/NCBI | |
|
Xue J, Schmidt SV, Sander J, Draffehn A, Krebs W, Quester I, De Nardo D, Gohel TD, Emde M, Schmidleithner L, et al: Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity. 40:274–288. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Yan L, Wang J, Cai X, Liou YC, Shen HM, Hao J, Huang C, Luo G and He W: Macrophage plasticity: Signaling pathways, tissue repair, and regeneration. MedComm (2020). 5:e6582024. View Article : Google Scholar : PubMed/NCBI | |
|
Murray PJ, Allen JE, Biswas SK, Fisher EA, Gilroy DW, Goerdt S, Gordon S, Hamilton JA, Ivashkiv LB, Lawrence T, et al: Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity. 41:14–20. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Chen X, Tang J, Shuai W, Meng J, Feng J and Han Z: Macrophage polarization and its role in the pathogenesis of acute lung injury/acute respiratory distress syndrome. Inflamm Res. 69:883–895. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen R, Zhang H, Tang B, Luo Y, Yang Y, Zhong X, Chen S, Xu X, Huang S and Liu C: Macrophages in cardiovascular diseases: Molecular mechanisms and therapeutic targets. Signal Transduct Target Ther. 9:1302024. View Article : Google Scholar : PubMed/NCBI | |
|
Frangogiannis NG: Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med. 65:70–99. 2019. View Article : Google Scholar | |
|
Hou P, Fang J, Liu Z, Shi Y, Agostini M, Bernassola F, Bove P, Candi E, Rovella V, Sica G, et al: Macrophage polarization and metabolism in atherosclerosis. Cell Death Dis. 14:6912023. View Article : Google Scholar : PubMed/NCBI | |
|
Lopez B, Ravassa S, Moreno MU, José GS, Beaumont J, González A and Díez J: Diffuse myocardial fibrosis: Mechanisms, diagnosis and therapeutic approaches. Nat Rev Cardiol. 18:479–498. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Moore KJ and Tabas I: Macrophages in the pathogenesis of atherosclerosis. Cell. 145:341–355. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang Y, Xin W, Hu X, Wang H, Ye X, Xu C, Nan Y, Wu Z, Ju D and Fan J: Inhibition of Hedgehog signaling ameliorates foam cell formation by promoting autophagy in early atherosclerosis. Cell Death Dis. 14:7402023. View Article : Google Scholar : PubMed/NCBI | |
|
Byles V, Covarrubias AJ, Ben-Sahra I, Lamming DW, Sabatini DM, Manning BD and Horng T: The TSC-mTOR pathway regulates macrophage polarization. Nat Commun. 4:28342013. View Article : Google Scholar : PubMed/NCBI | |
|
Castoldi A, Naffah de Souza C, Camara NO and Moraes-Vieira PM: The macrophage switch in obesity development. Front Immunol. 6:6372016. View Article : Google Scholar : PubMed/NCBI | |
|
Chawla A, Nguyen KD and Goh YP: Macrophage-mediated inflammation in metabolic disease. Nat Rev Immunol. 11:738–749. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Duan H, Jing L, Xiang J, Ju C, Wu Z, Liu J, Ma X, Chen X, Liu Z, Feng J and Yan X: CD146 Associates with Gp130 to Control a macrophage pro-inflammatory program that regulates the metabolic response to obesity. Adv Sci (Weinh). 9:e21037192022. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang H, Westerterp M, Wang C, Zhu Y and Ai D: Macrophage mTORC1 disruption reduces inflammation and insulin resistance in obese mice. Diabetologia. 57:2393–2404. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Lu X, Kong X, Wu H, Hao J, Li S, Gu Z, Zeng X, Shen Y, Wang S, Chen J, et al: UBE2M-mediated neddylation of TRIM21 regulates obesity-induced inflammation and metabolic disorders. Cell Metab. 35:1390–1405 e8. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Lumeng CN, Bodzin JL and Saltiel AR: Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 117:175–184. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Lumeng CN, Deyoung SM, Bodzin JL and Saltiel AR: Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes. 56:16–23. 2007. View Article : Google Scholar | |
|
Cassetta L and Pollard JW: Targeting macrophages: Therapeutic approaches in cancer. Nat Rev Drug Discov. 17:887–904. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
De Palma M, Biziato D and Petrova TV: Microenvironmental regulation of tumour angiogenesis. Nat Rev Cancer. 17:457–474. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Orecchioni M, Ghosheh Y, Pramod AB and Ley K: Macrophage polarization: Different gene signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. alternatively activated macrophages. Front Immunol. 10:10842019. View Article : Google Scholar : PubMed/NCBI | |
|
Pombo Antunes AR, Scheyltjens I, Lodi F, Messiaen J, Antoranz A, Duerinck J, Kancheva D, Martens L, De Vlaminck K, Van Hove H, et al: Single-cell profiling of myeloid cells in glioblastoma across species and disease stage reveals macrophage competition and specialization. Nat Neurosci. 24:595–610. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ruffell B, Chang-Strachan D, Chan V, Rosenbusch A, Ho CM, Pryer N, Daniel D, Hwang ES, Rugo HS and Coussens LM: Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell. 26:623–637. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Inoue K, Qin Y, Xia Y, Han J, Yuan R, Sun J, Xu R, Jiang JX, Greenblatt MB and Zhao B: Bone marrow Adipoq-lineage progenitors are a major cellular source of M-CSF that dominates bone marrow macrophage development, osteoclastogenesis, and bone mass. Elife. 12:e821182023. View Article : Google Scholar : PubMed/NCBI | |
|
Kulkarni A, Chandrasekar V, Natarajan SK, Ramesh A, Pandey P, Nirgud J, Bhatnagar H, Ashok D, Ajay AK and Sengupta S: A designer self-assembled supramolecule amplifies macrophage immune responses against aggressive cancer. Nat Biomed Eng. 2:589–599. 2018. View Article : Google Scholar | |
|
Sathi GA, Farahat M, Hara ES, Taketa H, Nagatsuka H, Kuboki T and Matsumoto T: MCSF orchestrates branching morphogenesis in developing submandibular gland tissue. J Cell Sci. 130:1559–1569. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Deng Y, Wang H, Liu X, Yuan H, Xu J, de Thé H, Zhou J and Zhu J: Zbtb14 regulates monocyte and macrophage development through inhibiting pu.1 expression in zebrafish. Elife. 11:e807602022. View Article : Google Scholar : PubMed/NCBI | |
|
Solomon LA, Podder S, He J, Jackson-Chornenki NL, Gibson K, Ziliotto RG, Rhee J and DeKoter RP: Coordination of myeloid differentiation with reduced cell cycle progression by PU.1 induction of MicroRNAs targeting cell cycle regulators and lipid anabolism. Mol Cell Biol. 37:e00013–17. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Tu J, Chen W, Fang Y, Han D, Chen Y, Jiang H, Tan X, Xu Z, Wu X, Wang H, et al: PU.1 promotes development of rheumatoid arthritis via repressing FLT3 in macrophages and fibroblast-like synoviocytes. Ann Rheum Dis. 82:198–211. 2023. View Article : Google Scholar | |
|
Xiao X, Li JX, Li HH and Teng F: ACE2 alleviates sepsis-induced cardiomyopathy through inhibiting M1 macrophage via NF-ĸB/STAT1 signals. Cell Biol Toxicol. 40:822024. View Article : Google Scholar | |
|
Jin C, Zhang F, Luo H, Li B, Jiang X, Pirozzi CJ, Liang C and Zhang M: The CCL5/CCR5/SHP2 axis sustains Stat1 phosphorylation and activates NF-ĸB signaling promoting M1 macrophage polarization and exacerbating chronic prostatic inflammation. Cell Commun Signal. 22:5842024. View Article : Google Scholar | |
|
Wu Q, Li B, Li J and Sun S, Yuan J and Sun S: Cancer-associated adipocytes as immunomodulators in cancer. Biomark Res. 9:22021. View Article : Google Scholar : PubMed/NCBI | |
|
Gu L, Larson Casey JL, Andrabi SA, Lee JH, Meza-Perez S, Randall TD and Carter AB: Mitochondrial calcium uniporter regulates PGC-1α expression to mediate metabolic reprogramming in pulmonary fibrosis. Redox Biol. 26:1013072019. View Article : Google Scholar | |
|
Yu T, Gan S, Zhu Q, Dai D, Li N, Wang H, Chen X, Hou D, Wang Y, Pan Q, et al: Modulation of M2 macrophage polarization by the crosstalk between Stat6 and Trim24. Nat Commun. 10:43532019. View Article : Google Scholar : PubMed/NCBI | |
|
Deng C, Huo M, Chu H, Zhuang X, Deng G, Li W, Wei H, Zeng L, He Y, Liu H, et al: Exosome circATP8A1 induces macrophage M2 polarization by regulating the miR-1-3p/STAT6 axis to promote gastric cancer progression. Mol Cancer. 23:492024. View Article : Google Scholar : PubMed/NCBI | |
|
Choi JY, Seok HJ, Lee DH, Lee E, Kim TJ, Bae S, Shin I and Bae IH: Tumor-derived miR-6794-5p enhances cancer growth by promoting M2 macrophage polarization. Cell Commun Signal. 22:1902024. View Article : Google Scholar : PubMed/NCBI | |
|
Lee KA, Hai TY, SivaRaman L, Thimmappaya B, Hurst HC, Jones NC and Green MR: A cellular protein, activating transcription factor, activates transcription of multiple E1A-inducible adenovirus early promoters. Proc Natl Acad Sci USA. 84:8355–8359. 1987. View Article : Google Scholar : PubMed/NCBI | |
|
Hai T and Hartman MG: The molecular biology and nomenclature of the activating transcription factor/cAMP responsive element binding family of transcription factors: Activating transcription factor proteins and homeostasis. Gene. 273:1–11. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Vallejo M, Ron D, Miller CP and Habener JF: C/ATF, a member of the activating transcription factor family of DNA-binding proteins, dimerizes with CAAT/enhancer-binding proteins and directs their binding to cAMP response elements. Proc Natl Acad Sci USA. 90:4679–4683. 1993. View Article : Google Scholar : PubMed/NCBI | |
|
Montminy MR and Bilezikjian LM: Binding of a nuclear protein to the cyclic-AMP response element of the somatostatin gene. Nature. 328:175–178. 1987. View Article : Google Scholar : PubMed/NCBI | |
|
Moller E, Praz V, Rajendran S, Dong R, Cauderay A, Xing YH, Lee L, Fusco C, Broye LC, Cironi L, et al: EWSR1-ATF1 dependent 3D connectivity regulates oncogenic and differentiation programs in clear cell sarcoma. Nat Commun. 13:22672022. View Article : Google Scholar : PubMed/NCBI | |
|
Hai TW, Liu F, Allegretto EA, Karin M and Green MR: A family of immunologically related transcription factors that includes multiple forms of ATF and AP-1. Genes Dev. 2:1216–1226. 1988. View Article : Google Scholar : PubMed/NCBI | |
|
Foulkes NS, Borrelli E and Sassone-Corsi P: CREM gene: Use of alternative DNA-binding domains generates multiple antagonists of cAMP-induced transcription. Cell. 64:739–749. 1991. View Article : Google Scholar : PubMed/NCBI | |
|
Liu H, Deng X, Shyu YJ, Li JJ, Taparowsky EJ and Hu CD: Mutual regulation of c-Jun and ATF2 by transcriptional activation and subcellular localization. EMBO J. 25:1058–1069. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
De Cesare D, Vallone D, Caracciolo A, Sassone-Corsi P, Nerlov C and Verde P: Heterodimerization of c-Jun with ATF-2 and c-Fos is required for positive and negative regulation of the human urokinase enhancer. Oncogene. 11:365–376. 1995.PubMed/NCBI | |
|
Sun MH, Jiang WJ, Li XH, Lee SH, Heo G, Zhou D, Choi JS, Kim KS, Lv W and Cui XS: ATF7-dependent epigenetic changes induced by high temperature during early porcine embryonic development. Cell Prolif. 56:e133522023. View Article : Google Scholar | |
|
Liu Y, Maekawa T, Yoshida K, Muratani M, Chatton B and Ishii S: The transcription factor ATF7 controls adipocyte differentiation and thermogenic gene programming. iScience. 13:98–112. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Diring J, Camuzeaux B, Donzeau M, Vigneron M, Rosa-Calatrava M, Kedinger C and Chatton B: A cytoplasmic negative regulator isoform of ATF7 impairs ATF7 and ATF2 phosphorylation and transcriptional activity. PLoS One. 6:e233512011. View Article : Google Scholar : PubMed/NCBI | |
|
Tsujino H, Kondo E, Fukuoka T, Dai Y, Tokunaga A, Miki K, Yonenobu K, Ochi T and Noguchi K: Activating transcription factor 3 (ATF3) induction by axotomy in sensory and moto-neurons: A novel neuronal marker of nerve injury. Mol Cell Neurosci. 15:170–182. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Rodriguez-Martinez JA, Reinke AW, Bhimsaria D, Keating AE and Ansari AZ: Combinatorial bZIP dimers display complex DNA-binding specificity landscapes. Elife. 6:e192722017. View Article : Google Scholar : PubMed/NCBI | |
|
Tsushima H, Okazaki K, Ishihara K, Ushijima T and Iwamoto Y: CCAAT/enhancer-binding protein β promotes receptor activator of nuclear factor-kappa-B ligand (RANKL) expression and osteoclast formation in the synovium in rheumatoid arthritis. Arthritis Res Ther. 17:312015. View Article : Google Scholar | |
|
Huggins CJ, Mayekar MK, Martin N, Saylor KL, Gonit M, Jailwala P, Kasoji M, Haines DC, Quiñones OA and Johnson PF: C/EBPү is a critical regulator of cellular stress response networks through heterodimerization with ATF4. Mol Cell Biol. 36:693–713. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Dias-Teixeira KL, Calegari-Silva TC, Medina JM, Vivarini ÁC, Cavalcanti Á, Teteo N, Santana AKM, Real F, Gomes CM, Pereira RMS, et al: Emerging role for the PERK/eIF2alpha/ATF4 in human cutaneous leishmaniasis. Sci Rep. 7:170742017. View Article : Google Scholar | |
|
Newman JR and Keating AE: Comprehensive identification of human bZIP interactions with coiled-coil arrays. Science. 300:2097–2101. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Glembotski CC, Arrieta A, Blackwood EA and Stauffer WT: ATF6 as a nodal regulator of proteostasis in the heart. Front Physiol. 11:2672020. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M and Mori K: Endoplasmic reticulum stress-induced formation of transcription factor complex ERSF including NF-Y (CBF) and activating transcription factors 6alpha and 6beta that activates the mammalian unfolded protein response. Mol Cell Biol. 21:1239–1248. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Yamamoto K, Sato T, Matsui T, Sato M, Okada T, Yoshida H, Harada A and Mori K: Transcriptional induction of mammalian ER quality control proteins is mediated by single or combined action of ATF6alpha and XBP1. Dev Cell. 13:365–376. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Hao Q, Zhao X, Zhang Y, Dong Z, Hu T and Chen P: Targeting overexpressed activating transcription factor 1 (ATF1) inhibits proliferation and migration and enhances sensitivity to paclitaxel in esophageal cancer cells. Med Sci Monit Basic Res. 23:304–312. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Meijer BJ, Giugliano FP, Baan B, van der Meer JHM, Meisner S, van Roest M, Koelink PJ, de Boer RJ, Jones N, Breitwieser W, et al: ATF2 and ATF7 are critical mediators of intestinal epithelial repair. Cell Mol Gastroenterol Hepatol. 10:23–42. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Maekawa T, Kim S, Nakai D, Takagi T, Ogura H, Yamada K, Chatton B and Ishii S: Social isolation stress induces ATF-7 phosphorylation and impairs silencing of the 5-HT 5B receptor gene. EMBO J. 29:196–208. 2010. View Article : Google Scholar : | |
|
Gozdecka M and Breitwieser W: The roles of ATF2 (activating transcription factor 2) in tumorigenesis. Biochem Soc Trans. 40:230–234. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen M, Liu Y, Yang Y, Qiu Y, Wang Z, Li X and Zhang W: Emerging roles of activating transcription factor (ATF) family members in tumourigenesis and immunity: Implications in cancer immunotherapy. Genes Dis. 9:981–999. 2021. View Article : Google Scholar | |
|
Yan C and Boyd DD: ATF3 regulates the stability of p53: A link to cancer. Cell Cycle. 5:926–929. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Hai T, Wolfgang CD, Marsee DK, Allen AE and Sivaprasad U: ATF3 and stress responses. Gene Expr. 7:321–335. 1999.PubMed/NCBI | |
|
Giannoudis A, Malki MI, Rudraraju B, Mohhamed H, Menon S, Liloglou T, Ali S, Carroll JS and Palmieri C: Activating transcription factor-2 (ATF2) is a key determinant of resistance to endocrine treatment in an in vitro model of breast cancer. Breast Cancer Res. 22:1262020. View Article : Google Scholar : PubMed/NCBI | |
|
Fawcett TW, Martindale JL, Guyton KZ, Hai T and Holbrook NJ: Complexes containing activating transcription factor (ATF)/cAMP-responsive-element-binding protein (CREB) interact with the CCAAT/enhancer-binding protein (C/EBP)-ATF composite site to regulate Gadd153 expression during the stress response. Biochem J. 339:135–141. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Shahriari-Felordi M, Alikhani HK, Hashemian SR, Hassan M and Vosough M: Mini review ATF4 and GRP78 as novel molecular targets in ER-Stress modulation for critical COVID-19 patients. Mol Biol Rep. 49:1545–1549. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshida H, Okada T, Haze K, Yanagi H, Yura T, Negishi M and Mori K: ATF6 activated by proteolysis binds in the presence of NF-Y (CBF) directly to the cis-acting element responsible for the mammalian unfolded protein response. Mol Cell Biol. 20:6755–6767. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Thuerauf DJ, Morrison LE, Hoover H and Glembotski CC: Coordination of ATF6-mediated transcription and ATF6 degradation by a domain that is shared with the viral transcription factor, VP16. J Biol Chem. 277:20734–20739. 2002. View Article : Google Scholar : PubMed/NCBI | |
|
Thuerauf DJ, Arnold ND, Zechner D, Hanford DS, DeMartin KM, McDonough PM, Prywes R and Glembotski CC: p38 Mitogen-activated protein kinase mediates the transcriptional induction of the atrial natriuretic factor gene through a serum response element. A potential role for the transcription factor ATF6. J Biol Chem. 273:20636–20643. 1998. View Article : Google Scholar : PubMed/NCBI | |
|
Luo R, Lu JF, Hu Q and Maity SN: CBF/NF-Y controls endoplasmic reticulum stress induced transcription through recruitment of both ATF6(N) and TBP. J Cell Biochem. 104:1708–1723. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Kokame K, Kato H and Miyata T: Identification of ERSE-II, a new cis-acting element responsible for the ATF6-dependent mammalian unfolded protein response. J Biol Chem. 276:9199–9205. 2001. View Article : Google Scholar | |
|
Haze K, Yoshida H, Yanagi H, Yura T and Mori K: Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell. 10:3787–3799. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Niwano K, Arai M, Tomaru K, Uchiyama T, Ohyama Y and Kurabayashi M: Transcriptional stimulation of the eNOS gene by the stable prostacyclin analogue beraprost is mediated through cAMP-responsive element in vascular endothelial cells: Close link between PGI2 signal and NO pathways. Circ Res. 93:523–530. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Xiao X, Chen M, Sang Y, Xue J, Jiang K, Chen Y, Zhang L, Yu S, Lv W, Li Y, et al: Methylation-mediated silencing of ATF3 promotes thyroid cancer progression by regulating prognostic genes in the MAPK and PI3K/AKT pathways. Thyroid. 33:1441–1454. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang WC, Du LJ, Zheng XJ, Chen XQ, Shi C, Chen BY, Sun XN, Li C, Zhang YY, Liu Y, et al: Elevated sodium chloride drives type I interferon signaling in macrophages and increases antiviral resistance. J Biol Chem. 293:1030–1039. 2018. View Article : Google Scholar : | |
|
Ranjan K, Hedl M, Sinha S, Zhang X and Abraham C: Ubiquitination of ATF6 by disease-associated RNF186 promotes the innate receptor-induced unfolded protein response. J Clin Invest. 131:e1454722021. View Article : Google Scholar : PubMed/NCBI | |
|
Udompong S, Mankhong S, Jaratjaroonphong J and Srisook K: Involvement of p38 MAPK and ATF-2 signaling pathway in anti-inflammatory effect of a novel compound bis[(5-methyl)2-furyl] (4-nitrophenyl)methane on lipopolysaccharide-stimulated macrophages. Int Immunopharmacol. 50:6–13. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Herrema H, Guan D, Choi JW, Feng X, Salazar Hernandez MA, Faruk F, Auen T, Boudett E, Tao R, Chun H and Ozcan U: FKBP11 rewires UPR signaling to promote glucose homeostasis in type 2 diabetes and obesity. Cell Metab. 34:1004–1022 e8. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou S, Zhong Z, Huang P, Xiang B, Li X, Dong H, Zhang G, Wu Y and Li P: IL-6/STAT3 induced neuron apoptosis in hypoxia by downregulating ATF6 expression. Front Physiol. 12:7299252021. View Article : Google Scholar : PubMed/NCBI | |
|
Kwon JW, Kwon HK, Shin HJ, Choi YM, Anwar MA and Choi S: Activating transcription factor 3 represses inflammatory responses by binding to the p65 subunit of NF-ĸB. Sci Rep. 5:144702015. View Article : Google Scholar | |
|
Cui A, Ding D and Li Y: Regulation of hepatic metabolism and cell growth by the ATF/CREB family of transcription factors. Diabetes. 70:653–664. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Takii R, Fujimoto M, Tan K, Takaki E, Hayashida N, Nakato R, Shirahige K and akai A: ATF1 modulates the heat shock response by regulating the stress-inducible heat shock factor 1 transcription complex. Mol Cell Biol. 35:11–25. 2015. View Article : Google Scholar : | |
|
Karanam B, Wang L, Wang D, Liu X, Marmorstein R, Cotter R and Cole PA: Multiple roles for acetylation in the interaction of p300 HAT with ATF-2. Biochemistry. 46:8207–8216. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Nguyen HCB, Adlanmerini M, Hauck AK and Lazar MA: Dichotomous engagement of HDAC3 activity governs inflammatory responses. Nature. 584:286–290. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Gilchrist M, Thorsson V, Li B, Rust AG, Korb M, Roach JC, Kennedy K, Hai T, Bolouri H and Aderem A: Systems biology approaches identify ATF3 as a negative regulator of Toll-like receptor 4. Nature. 441:173–178. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Chen BP, Liang G, Whelan J and Hai T: ATF3 and ATF3 delta Zip. Transcriptional repression versus activation by alternatively spliced isoforms. J Biol Chem. 269:15819–15826. 1994. View Article : Google Scholar : PubMed/NCBI | |
|
Sela D, Chen L, Martin-Brown S, Washburn MP, Florens L, Conaway JW and Conaway RC: Endoplasmic reticulum stress-responsive transcription factor ATF6alpha directs recruitment of the mediator of RNA polymerase II transcription and multiple histone acetyltransferase complexes. J Biol Chem. 287:23035–23045. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Hirose N, Maekawa T, Shinagawa T and Ishii S: ATF-2 regulates lipopolysaccharide-induced transcription in macrophage cells. Biochem Biophys Res Commun. 385:72–77. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Yoshida K, Maekawa T, Zhu Y, Renard-Guillet C, Chatton B, Inoue K, Uchiyama T, Ishibashi K, Yamada T, Ohno N, et al: The transcription factor ATF7 mediates lipopolysaccharide-induced epigenetic changes in macrophages involved in innate immunological memory. Nat Immunol. 16:1034–1043. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Baumeister P, Luo S, Skarnes WC, Sui G, Seto E, Shi Y and Lee AS: Endoplasmic reticulum stress induction of the Grp78/BiP promoter: Activating mechanisms mediated by YY1 and its interactive chromatin modifiers. Mol Cell Biol. 25:4529–4540. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Ma J, Liu Y, Valladolid-Acebes I, Recio-López P, Peng G, Li J, Berggren PO, Juntti-Berggren L and Tong N: ATF5 is a regulator of ER stress and β-cell apoptosis in different mouse models of genetic- and diet-induced obesity and diabetes mellitus. Cell Signal. 102:1105352023. View Article : Google Scholar | |
|
Raines LN, Zhao H, Wang Y, Chen HY, Gallart-Ayala H, Hsueh PC, Cao W, Koh Y, Alamonte-Loya A, Liu PS, et al: PERK is a critical metabolic hub for immunosuppressive function in macrophages. Nat Immunol. 23:431–445. 2022. View Article : Google Scholar : PubMed/NCBI Wang Y, Zhang X, Fu Y, Fu D, Zhen D, Xing A, Chen Y, Gong G and Wei C: 1, 8-cineole protects against ISO-induced heart failure by inhibiting oxidative stress and ER stress in vitro and in vivo. Eur J Pharmacol. 910:1744722021. View Article : Google Scholar : PubMed/NCBI | |
|
Hu S, Li R, Gong D, Hu P, Xu J, Ai Y, Zhao X, Hu C, Xu M, Liu C, et al: Atf3-mediated metabolic reprogramming in hepatic macrophage orchestrates metabolic dysfunction-associated steatohepatitis. Sci Adv. 10:eado31412024. View Article : Google Scholar : PubMed/NCBI | |
|
Brocard M, Lu J, Hall B, Borah K, Moller-Levet C, Georgana I, Sorgeloos F, Beste DJV, Goodfellow IG and Locker N: Murine norovirus infection results in anti-inflammatory response downstream of amino acid depletion in macrophages. J Virol. 95:e01134212021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Ge MX, Li YH, Li JL, Yu Q, Xiao FH, Ao HS, Yang LQ, Li J, He Y and Kong QP: Longevity-associated transcription factor ATF7 promotes healthspan by suppressing cellular senescence and systematic inflammation. Aging Dis. 14:1374–1389. 2023.PubMed/NCBI | |
|
Liu H, Kuang X, Zhang Y, Ye Y, Li J, Liang L, Xie Z, Weng L, Guo J, Li H, et al: ADORA1 inhibition promotes tumor immune evasion by regulating the ATF3-PD-L1 axis. Cancer Cell. 37:324–339 e8. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Liu T, Wen Z, Shao L, Cui Y, Tang X, Miao H, Shi J, Jiang L, Feng S, Zhao Y, et al: ATF4 knockdown in macrophage impairs glycolysis and mediates immune tolerance by targeting HK2 and HIF-1alpha ubiquitination in sepsis. Clin Immunol. 254:1096982023. View Article : Google Scholar | |
|
Zhang Q, Liu G, Liu R, Liu J, Zeng X, Ren D, Yan X and Yuan X: Dual role of endoplasmic reticulum stress-ATF-6 activation in autophagy and apoptosis induced by cyclic stretch in myoblast. Apoptosis. 28:796–809. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dai W, Hong L, Xiao W, Zhang L, Sha W, Yu Z, Liu X, Liu S, Xiao Y, Yang P, et al: The ATF2/miR-3913-5p/CREB5 axis is involved in the cell proliferation and metastasis of colorectal cancer. Commun Biol. 6:10262023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang B, Zhang J, Liu X, Chai Q, Lu X, Yao X, Yang Z, Sun L, Johnson SF, Schwartz RC and Zheng YH: Protein disulfide isomerases (PDIs) negatively regulate ebolavirus structural glycoprotein expression in the endoplasmic reticulum (ER) via the autophagy-lysosomal pathway. Autophagy. 18:2350–2367. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wen Z, Xiong X, Chen D, Shao L, Tang X, Shen X, Zhang S, Huang S, Zhang L, Chen Y, et al: Activating transcription factor 4 protects mice against sepsis-induced intestinal injury by regulating gut-resident macrophages differentiation. Chin Med J (Engl). 135:2585–2595. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Rajabalee N, Siushansian H, Weerapura M, Berton S, Berbatovci F, Hooks B, Geoffrion M, Yang D, Harper ME, Rayner K, et al: ATF2 orchestrates macrophage differentiation and activation to promote antibacterial responses. J Leukoc Biol. 114:280–298. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Qin W, Yang H, Liu G, Bai R, Bian Y, Yang Z and Xiao C: Activating transcription factor 3 is a potential target and a new biomarker for the prognosis of atherosclerosis. Hum Cell. 34:49–59. 2021. View Article : Google Scholar | |
|
Nilsson R, Bajic VB, Suzuki H, di Bernardo D, Björkegren J, Katayama S, Reid JF, Sweet MJ, Gariboldi M, Carninci P, et al: Transcriptional network dynamics in macrophage activation. Genomics. 88:133–142. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Clement M, Basatemur G, Masters L, Baker L, Bruneval P, Iwawaki T, Kneilling M, Yamasaki S, Goodall J and Mallat Z: Necrotic cell sensor clec4e promotes a proatherogenic macrophage phenotype through activation of the unfolded protein response. Circulation. 134:1039–1051. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Elbarghati L, Murdoch C and Lewis CE: Effects of hypoxia on transcription factor expression in human monocytes and macrophages. Immunobiology. 213:899–908. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao Q, Luo YF, Tian M, Xiao YL, Cai HR and Li H: Activating transcription factor 3 involved in Pseudomonas aeruginosa PAO1-induced macrophage senescence. Mol Immunol. 133:122–127. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Li M, Lu H, Wang X, Duan C, Zhu X, Zhang Y, Ge X, Ji F, Wang X, Su J and Zhang D: Pyruvate kinase M2 (PKM2) interacts with activating transcription factor 2 (ATF2) to bridge glycolysis and pyroptosis in microglia. Mol Immunol. 140:250–266. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Y, Li CS, Li YQ, Liang Y, Cao L and Chen LA: Perfluorocarbon inhibits lipopolysaccharide-induced macrophage inflammatory protein-2 expression and activation of ATF-2 and c-Jun in A549 pulmonary epithelial cells. Cell Mol Biol (Noisy-le-grand). 62:18–24. 2016.PubMed/NCBI | |
|
Boehlk S, Fessele S, Mojaat A, Miyamoto NG, Werner T, Nelson EL, Schlöndorff D and Nelson PJ: ATF and Jun transcription factors, acting through an Ets/CRE promoter module, mediate lipopolysaccharide inducibility of the chemokine RANTES in monocytic Mono Mac 6 cells. Eur J Immunol. 30:1102–1112. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Sha H, Zhang D, Zhang Y, Wen Y and Wang Y: ATF3 promotes migration and M1/M2 polarization of macrophages by activating tenascin-C via Wnt/β-catenin pathway. Mol Med Rep. 16:3641–3647. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hu C, Meng X, Huang C, Shen C and Li J: Frontline Science: ATF3 is responsible for the inhibition of TNF-α release and the impaired migration of acute ethanol-exposed monocytes and macrophages. J Leukoc Biol. 101:633–642. 2017. View Article : Google Scholar | |
|
Du Y, Ma Z, Zheng J, Huang S, Yang X, Song Y, Dong D, Shi L and Xu D: ATF3 positively regulates antibacterial immunity by modulating macrophage killing and migration functions. Front Immunol. 13:8395022022. View Article : Google Scholar : PubMed/NCBI | |
|
Mylvaganam S, Freeman SA and Grinstein S: The cytoskeleton in phagocytosis and macropinocytosis. Curr Biol. 31:R619–R632. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Taka S, Gazouli M, Politis PK, Pappa KI and Anagnou NP: Transcription factor ATF-3 regulates allele variation phenotypes of the human SLC11A1 gene. Mol Biol Rep. 40:2263–2271. 2013. View Article : Google Scholar | |
|
Middleton JD, Fehlman J, Sivakumar S, Stover DG and Hai T: Stress-inducible gene Atf3 dictates a dichotomous macrophage activity in chemotherapy-enhanced lung colonization. Int J Mol Sci. 22:73562021. View Article : Google Scholar : PubMed/NCBI | |
|
Xu Y, Li Y, Jadhav K, Pan X, Zhu Y, Hu S, Chen S, Chen L, Tang Y, Wang HH, et al: Hepatocyte ATF3 protects against atherosclerosis by regulating HDL and bile acid metabolism. Nat Metab. 3:59–74. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Raghavan S, Singh NK, Gali S, Mani AM and Rao GN: Protein Kinase ctheta via activating transcription factor 2-mediated CD36 expression and foam cell formation of Ly6C(hi) cells contributes to atherosclerosis. Circulation. 138:2395–2412. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Seneviratne A, Han Y, Wong E, Walter ERH, Jiang L, Cave L, Long NJ, Carling D, Mason JC, Haskard DO and Boyle JJ: Hematoma resolution in vivo is directed by activating transcription factor 1. Circ Res. 127:928–944. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Illouz M, Leclercq LD, Dessenne C, Hatfull G, Daher W and Kremer L and Guérardel Y: Multiple Mycobacterium abscessus O-acetyltransferases influence glycopeptidolipid structure and colony morphotype. J Biol Chem. 299:1049792023. View Article : Google Scholar : PubMed/NCBI | |
|
Perez-Arques C, Navarro-Mendoza MI, Murcia L, Lax C, Martínez-García P, Heitman J, Nicolás FE and Garre V: Mucor circinelloides Thrives inside the phagosome through an Atf-Mediated germination pathway. mBio. 10:e02765–18. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Gupta D, Wang Q, Vinson C and Dziarski R: Bacterial peptidoglycan induces CD14-dependent activation of transcription factors CREB/ATF and AP-1. J Biol Chem. 274:14012–14020. 1999. View Article : Google Scholar : PubMed/NCBI | |
|
Shi R, Wang J, Zhang Z, Leng Y and Chen AF: ASGR1 promotes liver injury in sepsis by modulating monocyte-to-macrophage differentiation via NF-ĸB/ATF5 pathway. Life Sci. 315:1213392023. View Article : Google Scholar | |
|
Abe JI, Ko KA, Kotla S, Wang Y, Paez-Mayorga J, Shin IJ, Imanishi M, Vu HT, Tao Y, Leiva-Juarez MM, et al: MAGI1 as a link between endothelial activation and ER stress drives atherosclerosis. JCI Insight. 4:e1255702019. View Article : Google Scholar : PubMed/NCBI | |
|
Jin Z, Xu H, Zhao W, Zhang K, Wu S, Shu C, Zhu L, Wang Y, Wang L, Zhang H and Yan B: Macrophage ATF6 accelerates corticotomy-assisted orthodontic tooth movement through promoting Tnfα transcription. Int J Oral Sci. 17:282025. View Article : Google Scholar | |
|
Xu X, Lei T, Li W and Ou H: Enhanced cellular cholesterol efflux by naringenin is mediated through inhibiting endoplasmic reticulum stress - ATF6 activity in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids. 1864:1472–1482. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Rao J, Yue S, Fu Y, Zhu J, Wang X, Busuttil RW, Kupiec-Weglinski JW, Lu L and Zhai Y: ATF6 mediates a pro-inflammatory synergy between ER stress and TLR activation in the pathogenesis of liver ischemia-reperfusion injury. Am J Transplant. 14:1552–1561. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Wang Q, Zhu X, Li Z, Feng M and Liu X: ATF6 promotes liver fibrogenesis by regulating macrophage-derived interleukin-1alpha expression. Cell Immunol. 367:1044012021. View Article : Google Scholar | |
|
Wang J, Cheng W, Wang Z, Xin L and Zhang W: ATF3 inhibits the inflammation induced by Mycoplasma pneumonia in vitro and in vivo. Pediatr Pulmonol. 52:1163–1170. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Hoetzenecker W, Echtenacher B, Guenova E, Hoetzenecker K, Woelbing F, Brück J, Teske A, Valtcheva N, Fuchs K, Kneilling M, et al: ROS-induced ATF3 causes susceptibility to secondary infections during sepsis-associated immunosuppression. Nat Med. 18:128–134. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Bae YA and Cheon HG: Activating transcription factor-3 induction is involved in the anti-inflammatory action of berberine in RAW264.7 murine macrophages. Korean J Physiol Pharmacol. 20:415–424. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Saha S, Roy S, Dutta A, Jana K and Ukil A: Leishmania donovani targets host transcription factor NRF2 to activate antioxidant enzyme HO-1 and transcriptional repressor ATF3 for establishing infection. Infect Immun. 89:e00764202021. View Article : Google Scholar : PubMed/NCBI | |
|
Qian L, Zhao Y, Guo L, Li S and Wu X: Activating transcription factor 3 (ATF3) protects against lipopolysaccharide-induced acute lung injury via inhibiting the expression of TL1A. J Cell Physiol. 232:3727–3734. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng CF and Lin H: Acute kidney injury and the potential for ATF3-regulated epigenetic therapy. Toxicol Mech Methods. 21:362–366. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Yu T, Moh SH, Kim SB, Yang Y, Kim E, Lee YW, Cho CK, Kim KH, Yoo BC, Cho JY and Yoo HS: HangAmDan-B, an ethnomedicinal herbal mixture, suppresses inflammatory responses by inhibiting Syk/NF-ĸB and JNK/ATF-2 pathways. J Med Food. 16:56–65. 2013. View Article : Google Scholar : | |
|
Zhang C, He H, Wang L, Zhang N, Huang H, Xiong Q, Yan Y, Wu N, Ren H, Han H, et al: Virus-triggered ATP release limits viral replication through facilitating IFN-β Production in a P2X7-Dependent manner. J Immunol. 199:1372–1381. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Yu T, Yang Y, Kwak YS, Song GG, Kim MY, Rhee MH and Cho JY: Ginsenoside Rc from Panax ginseng exerts anti-inflammatory activity by targeting TANK-binding kinase 1/interferon regulatory factor-3 and p38/ATF-2. J Ginseng Res. 41:127–133. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Xu X, Xia J, Zhao S, Wang Q, Ge G, Xu F, Liu X, Zhang W and Yang Y: Qing-Fei-Pai-Du decoction and wogonoside exert anti-inflammatory action through down-regulating USP14 to promote the degradation of activating transcription factor 2. FASEB J. 35:e218702021. View Article : Google Scholar : PubMed/NCBI | |
|
Park JB, Peters R, Pham Q and Wang TTY: Javamide-II Inhibits IL-6 without Significant Impact on TNF-alpha and IL-1beta in macrophage-like cells. Biomedicines. 8:1382020. View Article : Google Scholar : PubMed/NCBI | |
|
Kim SM, Park EJ and Lee HJ: Nuciferine attenuates lipopolysaccharide-stimulated inflammatory responses by inhibiting p38 MAPK/ATF2 signaling pathways. Inflammopharmacology. 30:2373–2383. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Endale M, Kim TH, Kwak YS, Kim NM, Kim SH, Cho JY, Yun BS and Rhee MH: Torilin inhibits inflammation by limiting TAK1-mediated MAP kinase and NF-ĸB activation. Mediators Inflamm. 2017:72509682017. View Article : Google Scholar | |
|
Miyata Y, Fukuhara A, Otsuki M and Shimomura I: Expression of activating transcription factor 2 in inflammatory macrophages in obese adipose tissue. Obesity (Silver Spring). 21:731–736. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Y, Gu J, Wang Y, Lin J, Yu H, Yang F, Wu S, Yin J, Lv H, Ji X and Wang S: Promotion effect of EGCG on the raised expression of IL-23 through the signaling of STAT3-BATF2-c-JUN/ATF2. J Agric Food Chem. 69:7898–7909. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Sukhorukov VN, Khotina VA, Bagheri Ekta M, Ivanova EA, Sobenin IA and Orekhov AN: Endoplasmic reticulum stress in macrophages: The vicious circle of lipid accumulation and pro-inflammatory response. Biomedicines. 8:2102020. View Article : Google Scholar : PubMed/NCBI | |
|
Song C, Chen J, Li X, Yang R, Cao X, Zhou L, Zhou Y, Ying H, Zhang Q and Sun Y: Limonin ameliorates dextran sulfate sodium-induced chronic colitis in mice by inhibiting PERK-ATF4-CHOP pathway of ER stress and NF-kappaB signaling. Int Immunopharmacol. 90:1071612021. View Article : Google Scholar | |
|
Luo JH, Wang FX, Zhao JW, Yang CL, Rong SJ, Lu WY, Chen QJ, Zhou Q, Xiao J, Wang YN, et al: PDIA3 defines a novel subset of adipose macrophages to exacerbate the development of obesity and metabolic disorders. Cell Metab. 36:2262–2280 e5. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Boyle JJ, Johns M, Kampfer T, Nguyen AT, Game L, Schaer DJ, Mason JC and Haskard DO: Activating transcription factor 1 directs Mhem atheroprotective macrophages through coordinated iron handling and foam cell protection. Circ Res. 110:20–33. 2012. View Article : Google Scholar | |
|
Suganami T, Yuan X, Shimoda Y, Uchio-Yamada K, Nakagawa N, Shirakawa I, Usami T, Tsukahara T, Nakayama K, Miyamoto Y, et al: Activating transcription factor 3 constitutes a negative feedback mechanism that attenuates saturated Fatty acid/toll-like receptor 4 signaling and macrophage activation in obese adipose tissue. Circ Res. 105:25–32. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Tang Q, Xie J, Wang Y, Dong C and Sun Q: Exosomes secreted by ATF3/Nrf2-mediated ferroptotic renal tubular epithelial cells promote M1/M2 ratio imbalance inducing renal interstitial fibrosis following ischemia and reperfusion injury. Front Immunol. 16:15105002025. View Article : Google Scholar : PubMed/NCBI | |
|
Bartels HC, Hameed S, Young C, Nabhan M, Downey P, Curran KM, McCormack J, Fabre A, Kolch W, Zhernovkov V and Brennan DJ: Spatial proteomics and transcriptomics of the maternal-fetal interface in placenta accreta spectrum. Transl Res. 274:67–80. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Iwasaki Y, Suganami T, Hachiya R, Shirakawa I, Kim-Saijo M, Tanaka M, Hamaguchi M, Takai-Igarashi T, Nakai M, Miyamoto Y and Ogawa Y: Activating transcription factor 4 links metabolic stress to interleukin-6 expression in macrophages. Diabetes. 63:152–161. 2014. View Article : Google Scholar | |
|
Xia HF, Zhu JY, Wang JN, Ren JG, Cai Y, Wang FQ, Zhang W, Chen G, Zhao YF and Zhao JH: Association of ATF4 expression with tissue hypoxia and M2 macrophage infiltration in infantile hemangioma. J Histochem Cytochem. 65:285–294. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Zong S, Wang Z, Yang R, Guo Y, Wang Y, Chen X, Li Y and Wang S: FTY720 Attenuates LPS-Induced inflammatory bone loss by inhibiting osteoclastogenesis via the NF-kappaB and HDAC4/ATF pathways. J Immunol Res. 2023:85716492023. View Article : Google Scholar | |
|
Baek K, Park HJ, Baek JH and Kim HR: Isoproterenol increases RANKL expression in a ATF4/NFATc1-dependent manner in mouse osteoblastic cells. Int J Mol Sci. 18:22042017. View Article : Google Scholar : PubMed/NCBI | |
|
Chattopadhyay A, Kwartler CS, Kaw K, Li Y, Kaw A, Chen J, LeMaire SA, Shen YH and Milewicz DM: Cholesterol-induced phenotypic modulation of smooth muscle cells to macrophage/fibroblast-like cells is driven by an unfolded protein response. Arterioscler Thromb Vasc Biol. 41:302–316. 2021. View Article : Google Scholar | |
|
Kim J, Kwak HJ, Cha JY, Jeong YS, Rhee SD, Kim KR and Cheon HG: Metformin suppresses lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages via activating transcription factor-3 (ATF-3) induction. J Biol Chem. 289:23246–23255. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Luo M, Zhao F, Cheng H, Su M and Wang Y: Macrophage polarization: An important role in inflammatory diseases. Front Immunol. 15:13529462024. View Article : Google Scholar : PubMed/NCBI | |
|
Xiang M, Li P, Yue X, Liu L, Wang L, Sun N, Wang K, Zhang Y and Wang H: Dysregulated macrophage immunity in Helicobacter pylori infection: unveiling mechanistic insights and therapeutic implications. Front Immunol. 16:16367682025. View Article : Google Scholar : PubMed/NCBI | |
|
Fu YL and Harrison RE: Microbial phagocytic receptors and their potential involvement in cytokine induction in macrophages. Front Immunol. 12:6620632021. View Article : Google Scholar : PubMed/NCBI | |
|
Arango Duque G and Descoteaux A: Macrophage cytokines: Involvement in immunity and infectious diseases. Front Immunol. 5:4912014. View Article : Google Scholar : PubMed/NCBI | |
|
Lv K and Liang Q: Macrophages in sepsis-induced acute lung injury: exosomal modulation and therapeutic potential. Front Immunol. 15:15180082025. View Article : Google Scholar : PubMed/NCBI | |
|
A-Gonzalez N, Quintana JA, Garcia-Silva S, Mazariegos M, González de la Aleja A, Nicolás-Ávila JA, Walter W, Adrover JM, Crainiciuc G, Kuchroo VK, et al: Phagocytosis imprints heterogeneity in tissue-resident macrophages. J Exp Med. 214:1281–1296. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Ochando J, Mulder WJM, Madsen JC, Netea MG and Duivenvoorden R: Trained immunity-basic concepts and contributions to immunopathology. Nat Rev Nephrol. 19:23–37. 2023. View Article : Google Scholar | |
|
Nathan C and Ding A: Nonresolving inflammation. Cell. 140:871–882. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Cheng Y, Marion TN, Cao X, Wang W and Cao Y: Park 7: A novel therapeutic target for macrophages in sepsis-induced immunosuppression. Front Immunol. 9:26322018. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y, Cai R, Huang Y, Zhu L, Xiao L, Wang C and Wang L: Macrophages in organ fibrosis: From pathogenesis to therapeutic targets. Cell Death Discov. 10:4872024. View Article : Google Scholar : PubMed/NCBI | |
|
Ahmad S, Zaki A, Manda K, Mohan A and Syed MA: Vitamin-D ameliorates sepsis-induced acute lung injury via augmenting miR-149-5p and downregulating ER stress. J Nutr Biochem. 110:1091302022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang S, Meng P, Liu G, Liu K and Che C: ATF4 involvement in TLR4 and LOX-1-induced host inflammatory response to aspergillus fumigatus keratitis. J Ophthalmol. 2018:58302022018. | |
|
Li R, Ren T, Zeng J and Xu H: ALCAM deficiency alleviates LPS-Induced acute lung injury by inhibiting inflammatory response. Inflammation. 46:688–699. 2023. View Article : Google Scholar | |
|
Al-Salleeh F and Petro TM: Promoter analysis reveals critical roles for SMAD-3 and ATF-2 in expression of IL-23 p19 in macrophages. J Immunol. 181:4523–4533. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ta HM, Le TM, Ishii H, Takarada-Iemata M, Hattori T, Hashida K, Yamamoto Y, Mori K, Takahashi R, Kitao Y and Hori O: Atf6α deficiency suppresses microglial activation and ameliorates pathology of experimental autoimmune encephalomyelitis. J Neurochem. 139:1124–1137. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Wellen KE and Hotamisligil GS: Obesity-induced inflammatory changes in adipose tissue. J Clin Invest. 112:1785–1788. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Olefsky JM and Glass CK: Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 72:219–246. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Liang W, Qi Y, Yi H, Mao C, Meng Q, Wang H and Zheng C: The roles of adipose tissue macrophages in human disease. Front Immunol. 13:9087492022. View Article : Google Scholar : PubMed/NCBI | |
|
Guria S, Hoory A, Das S, Chattopadhyay D and Mukherjee S: Adipose tissue macrophages and their role in obesity-associated insulin resistance: An overview of the complex dynamics at play. Biosci Rep. 43:BSR202202002023. View Article : Google Scholar : PubMed/NCBI | |
|
Bai Y and Sun Q: Macrophage recruitment in obese adipose tissue. Obes Rev. 16:127–136. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
De Nardo D, Labzin LI, Kono H, Seki R, Schmidt SV, Beyer M, Xu D, Zimmer S, Lahrmann C, Schildberg FA, et al: High-density lipoprotein mediates anti-inflammatory reprogramming of macrophages via the transcriptional regulator ATF3. Nat Immunol. 15:152–160. 2014. View Article : Google Scholar | |
|
Xu J, Ding L, Mei J, Hu Y, Kong X, Dai S, Bu T, Xiao Q and Ding K: Dual roles and therapeutic targeting of tumor-associated macrophages in tumor microenvironments. Signal Transduct Target Ther. 10:2682025. View Article : Google Scholar : PubMed/NCBI | |
|
Takayanagi H: Osteoimmunology: Shared mechanisms and crosstalk between the immune and bone systems. Nat Rev Immunol. 7:292–304. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Li J, Xie X, Gu F, Sui Z, Zhang K and Yu T: Macrophage-osteoclast associations: Origin, polarization, and subgroups. Front Immunol. 12:7780782021. View Article : Google Scholar : PubMed/NCBI | |
|
Nakashima T, Hayashi M and Takayanagi H: New insights into osteoclastogenic signaling mechanisms. Trends Endocrinol Metab. 23:582–590. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Perrone M, Chiodoni C, Lecchi M, Botti L, Bassani B, Piva A, Jachetti E, Milani M, Lecis D, Tagliabue E, et al: ATF3 reprograms the bone marrow niche in response to early breast cancer transformation. Cancer Res. 83:117–129. 2023. View Article : Google Scholar : | |
|
Gao C, Jiang J, Tan Y and Chen S: Microglia in neurodegenerative diseases: Mechanism and potential therapeutic targets. Signal Transduct Target Ther. 8:3592023. View Article : Google Scholar : PubMed/NCBI | |
|
Fleiss B, Van Steenwinckel J, Bokobza C, I KS, Ross-Munro E and Gressens P: Microglia-mediated neurodegeneration in perinatal brain injuries. Biomolecules. 11:992021. View Article : Google Scholar : PubMed/NCBI | |
|
Flury A, Aljayousi L, Park HJ, Khakpour M, Mechler J, Aziz S, McGrath JD, Deme P, Sandberg C, González Ibáñez F, et al: A neurodegenerative cellular stress response linked to dark microglia and toxic lipid secretion. Neuron. 113:554–571 e14. 2025. View Article : Google Scholar : | |
|
Liu LL, Xiao YS, Huang WM, Liu S, Huang LX, Zhong JH, Jia P and Liu WY: ATF1/miR-214-5p/ITGA7 axis promotes osteoclastogenesis to alter OVX-induced bone absorption. Mol Med. 28:562022. View Article : Google Scholar : PubMed/NCBI | |
|
Kim HN, Baek JK, Park SB, Kim JD, Son HJ, Park GH, Eo HJ, Park JH, Jung HS and Jeong JB: Anti-inflammatory effect of Vaccinium oldhamii stems through inhibition of NF-kappaB and MAPK/ATF2 signaling activation in LPS-stimulated RAW264.7 cells. BMC Complement Altern Med. 19:2912019. View Article : Google Scholar | |
|
Comarita IK, Vilcu A, Constantin A, Procopciuc A, Safciuc F, Alexandru N, Dragan E, Nemecz M, Filippi A, Chiţoiu L, et al: Therapeutic potential of stem cell-derived extracellular vesicles on atherosclerosis-induced vascular dysfunction and its key molecular players. Front Cell Dev Biol. 10:8171802022. View Article : Google Scholar : PubMed/NCBI | |
|
Ho HH, Antoniv TT, Ji JD and Ivashkiv LB: Lipopolysaccharide-induced expression of matrix metalloproteinases in human monocytes is suppressed by IFN-gamma via superinduction of ATF-3 and suppression of AP-1. J Immunol. 181:5089–5097. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Ishida M, Ueki M, Morishita J, Ueno M, Shiozawa S and Maekawa N: T-5224, a selective inhibitor of c-Fos/activator protein-1, improves survival by inhibiting serum high mobility group box-1 in lethal lipopolysaccharide-induced acute kidney injury model. J Intensive Care. 3:492015. View Article : Google Scholar : PubMed/NCBI | |
|
Kubryn N, Fijalkowski L, Nowaczyk J, Jamil A and Nowaczyk A: PROTAC technology as a new tool for modern pharmacotherapy. Molecules. 30:21232025. View Article : Google Scholar : PubMed/NCBI | |
|
Hu Z and Crews CM: Recent developments in PROTAC-mediated protein degradation: From bench to clinic. Chembiochem. 23:e2021002702022. View Article : Google Scholar : | |
|
Bekes M, Langley DR and Crews CM: PROTAC targeted protein degraders: The past is prologue. Nat Rev Drug Discov. 21:181–200. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Ai M, Ma H, He J, Xu F, Ming Y, Ye Z, Zheng Q, Luo D, Yang K, Li J, et al: Targeting oncogenic transcriptional factor c-myc by oligonucleotide PROTAC for the treatment of hepatocellular carcinoma. Eur J Med Chem. 280:1169782024. View Article : Google Scholar : PubMed/NCBI | |
|
Simpson LM, Glennie L, Brewer A, Zhao JF, Crooks J, Shpiro N and Sapkota GP: Target protein localization and its impact on PROTAC-mediated degradation. Cell Chem Biol. 29:1482–1504 e7. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu Z, Hu M, Yang Y, Du C, Zhou H, Liu C, Chen Y, Fan L, Ma H, Gong Y and Xie Y: An overview of PROTACs: A promising drug discovery paradigm. Mol Biomed. 3:462022. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J, Chen H, Kaniskan HU, Xie L, Chen X, Jin J and Wei W: TF-PROTACs enable targeted degradation of transcription factors. J Am Chem Soc. 143:8902–8910. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Ji J, Ma S, Zhu Y, Zhao J, Tong Y, You Q and Jiang Z: ARE-PROTACs enable co-degradation of an Nrf2-MafG heterodimer. J Med Chem. 66:6070–6081. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang D, Liu Y, Chen Y, Dai C, Hu W, Han J, Li Z, Yin F, Zhang Y and Shi C: EGFR targeted liposomal PROTAC assisted with epigenetic regulation as an efficient strategy for osimertinib-resistant lung cancer therapy. Adv Sci (Weinh). 12:e101972025. View Article : Google Scholar : PubMed/NCBI | |
|
Chen S, Chen E, Su J, Gong Y, Tang S, Qin A, Shen A, Tang S and Zhang L: Magnetically navigated nano-PROTAC ameliorates acute lung injury. J Nanobiotechnology. 23:6222025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao L, Zhao J, Zhong K, Tong A and Jia D: Targeted protein degradation: Mechanisms, strategies and application. Signal Transduct Target Ther. 7:1132022. View Article : Google Scholar : PubMed/NCBI | |
|
Kronke J, Udeshi ND, Narla A, Grauman P, Hurst SN, McConkey M, Svinkina T, Heckl D, Comer E, Li X, et al: Lenalidomide causes selective degradation of IKZF1 and IKZF3 in multiple myeloma cells. Science. 343:301–305. 2014. View Article : Google Scholar : | |
|
Greene LA, Zhou Q, Siegelin MD and Angelastro JM: Targeting transcription factors ATF5, CEBPB and CEBPD with cell-penetrating peptides to treat brain and other cancers. Cells. 12:5812023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Angelastro JM, Merino D, Zhou Q, Siegelin MD and Greene LA: Dominant-negative ATF5 rapidly depletes survivin in tumor cells. Cell Death Dis. 10:7092019. View Article : Google Scholar : PubMed/NCBI | |
|
Rui Y, Eppler HB, Yanes AA and Jewell CM: Tissue-targeted drug delivery strategies to promote antigen-specific immune tolerance. Adv Healthc Mater. 12:e22022382023. View Article : Google Scholar | |
|
Chen W, Schilperoort M, Cao Y, Shi J, Tabas I and Tao W: Macrophage-targeted nanomedicine for the diagnosis and treatment of atherosclerosis. Nat Rev Cardiol. 19:228–249. 2022. View Article : Google Scholar | |
|
Zhao G, Xue L, Geisler HC, Xu J, Li X, Mitchell MJ and Vaughan AE: Precision treatment of viral pneumonia through macrophage-targeted lipid nanoparticle delivery. Proc Natl Acad Sci USA. 121:e23147471212024. View Article : Google Scholar : PubMed/NCBI | |
|
Hai T, Wolford CC and Chang YS: ATF3, a hub of the cellular adaptive-response network, in the pathogenesis of diseases: Is modulation of inflammation a unifying component? Gene Expr. 15:1–11. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Nguyen CT, Kim EH, Luong TT, Pyo S and Rhee DK: ATF3 confers resistance to pneumococcal infection through positive regulation of cytokine production. J Infect Dis. 210:1745–1754. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Bekeredjian-Ding I, Stein C and Uebele J: The innate immune response against staphylococcus aureus. Curr Top Microbiol Immunol. 409:385–418. 2017. | |
|
Nguyen CT, Kim EH, Luong TT, Pyo S and Rhee DK: TLR4 mediates pneumolysin-induced ATF3 expression through the JNK/p38 pathway in Streptococcus pneumoniae-infected RAW 264.7 cells. Mol Cells. 38:58–64. 2015. View Article : Google Scholar : | |
|
Guo B, Stein JL, van Wijnen AJ and Stein GS: ATF1 and CREB trans-activate a cell cycle regulated histone H4 gene at a distal nuclear matrix associated promoter element. Biochemistry. 36:14447–14455. 1997. View Article : Google Scholar : PubMed/NCBI | |
|
Watson G, Ronai ZA and Lau E: ATF2, a paradigm of the multifaceted regulation of transcription factors in biology and disease. Pharmacol Res. 119:347–357. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Lau E and Ronai ZA: ATF2 - at the crossroad of nuclear and cytosolic functions. J Cell Sci. 125:2815–2824. 2012.PubMed/NCBI | |
|
Claps G, Cheli Y, Zhang T, Scortegagna M, Lau E, Kim H, Qi J, Li JL, James B, Dzung A, et al: A transcriptionally inactive ATF2 variant drives melanomagenesis. Cell Rep. 15:1884–1892. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Hashimoto Y, Zhang C, Kawauchi J, Imoto I, Adachi MT, Inazawa J, Amagasa T, Hai T and Kitajima S: An alternatively spliced isoform of transcriptional repressor ATF3 and its induction by stress stimuli. Nucleic Acids Res. 0:2398–2406. 2002. View Article : Google Scholar | |
|
Chan JYW, Tsui JCC, Law PTW, So WKW, Leung DYP, Sham MMK, Tsui SKW and Chan CWH: RNA-Seq revealed ATF3-regulated inflammation induced by silica. Toxicology. 393:34–41. 2018. View Article : Google Scholar | |
|
Jeong BC, Kim JH, Kim K, Kim I, Seong S and Kim N: ATF3 modulates calcium signaling in osteoclast differentiation and activity by associating with c-Fos and NFATc1 proteins. Bone. 95:33–40. 2017. View Article : Google Scholar |