Monocytes and macrophages express the transcription factor MAFB (V-maf musculoaponeurotic fibrosarcoma oncogene homolog B) and protect against ischemic acute kidney injury (AKI). However, the mechanism through which MAFB alleviates AKI in macrophages remains unclear. In this study, we induced AKI in macrophage lineage-specific Mafb-deficient mice (C57BL/6J) using the ischemia-reperfusion injury model to analyze these mechanisms. Our results showed that MAFB regulates the expression of Alox15 (arachidonate 15-lipoxygenase) in macrophages during ischemic AKI. The expression of ALOX15 was significantly decreased at the mRNA and protein levels in macrophages that infiltrated the kidneys of macrophage-specific Mafb-deficient mice at 24 h after ischemia-reperfusion injury. ALOX15 promotes the resolution of inflammation under acute conditions by producing specialized proresolving mediators by oxidizing essential fatty acids. Therefore, MAFB in macrophages promotes the resolution of inflammation in ischemic AKI by regulating the expression of Alox15. Moreover, MAFB expression in macrophages is upregulated via the COX-2/PGE2/EP4 pathway in ischemic AKI. Our in vitro assay showed that MAFB regulates the expression of Alox15 under the COX-2/PGE2/EP4 pathway in macrophages. PGE2 mediates the lipid mediator (LM) class switch from inflammatory LMs to specialized proresolving mediators. Therefore, MAFB plays a key role in the PGE2-mediated LM class switch by regulating the expression of Alox15. Our study identified a previously unknown mechanism by which MAFB in macrophages alleviates ischemic AKI and provides new insights into regulating the LM class switch in acute inflammatory conditions.

Ischemic acute kidney injury (AKI) is characterized by a rapid decline in renal function, occurring within a few hours to several days due to reduced blood flow into the kidneys, due to conditions such as sepsis or hypotension. The incidence of AKI has been increasing steadily annually. AKI is linked to severe outcomes: 10–15% of AKI patients succumb to the associated acute illness, 3–15% advance to end-stage renal disease, and 20–50% develop chronic kidney disease (CKD) (1, 2). Consequently, AKI significantly decreases the quality of life of patients. However, the only available treatment strategy for AKI focuses on restoring adequate blood flow to the kidneys. Moreover, no targeted therapies have been designed to suppress the underlying inflammatory processes specifically. Therefore, an in-depth understanding of the molecular mechanisms of AKI is essential for advancing AKI management.

Notably, the number of macrophages in the kidneys increases within 24 h of AKI onset. During the initial stage of AKI, macrophages promote tubular injury, whereas in later stages, they promote tubular proliferation, aiding the normal repair (3–5). Consequently, macrophages play a critical role in both the injury and recovery phases of AKI. Therefore, elucidating the specific functions of macrophages in promoting and resolving inflammation in AKI is crucial for understanding its pathology; however, the mechanisms remain unclear.

MAFB (V-maf musculoaponeurotic fibrosarcoma oncogene homolog B), a transcription factor belonging to the large Maf family, forms a dimer via a leucine zipper and binds to the Maf recognition element (MARE) in the DNA promoter region, thereby regulating the expression of downstream target genes (6, 7). MAFB plays an essential role in the differentiation and functional maintenance of various tissues and cells, including kidney glomerulus podocytes, pancreatic cells, and the parathyroid gland (8–10). In hematopoietic cell lines, MAFB is expressed specifically on monocytes and macrophages but not any other mature myeloid or lymphoid lineage cells (8, 11). Several studies have reported the function of MAFB in macrophages. MAFB promotes atherosclerosis development by regulating the expression of apoptotic inhibitors of macrophages (AIMs), in turn preventing foam cell apoptosis (12). MAFB in macrophages also plays a vital role in efferocytosis by regulating the expression of complement component C1q, as C1q binds to apoptotic cells to promote the recognition of apoptotic cells by macrophages (11). In addition, MAFB in macrophages promotes clearance of damage-associated molecular patterns (DAMPs) by regulating the expression of macrophage scavenger receptor 1 (MSR1) in ischemic stroke, and recently it has been reported that MAFB in macrophages promotes skin wound healing and cold-induced neuronal density in brown adipose tissue (13–15). Therefore, MAFB in macrophages is assumed to be crucial for homeostasis and inflammation suppression. Recently, it has been reported that MAFB expression is induced via the cyclooxygenase-2 (COX-2)/PGE2/PGE2 receptor 4 (EP4) pathway in macrophages infiltrating the ischemic AKI-induced kidney. The induced MAFB promotes recovery from ischemic AKI by polarizing renal myeloid cells into an anti-inflammatory and proresolving phenotype (16). However, the precise molecular mechanisms, such as target genes of MAFB, remain unknown.

Arachidonate 15-lypoxygenase (ALOX15) is a lipoxygenase that catalyzes the oxidation of essential fatty acids. ALOX15 in mice has been reported to play a role as a suppressor of inflammation and cancer (17). ALOX15 expressed in macrophages produces precursors of inflammation-suppressing specialized proresolving mediators (SPMs) by oxidizing essential fatty acids at the site of inflammation (17–19). SPMs have anti-inflammatory and proresolving functions in immune cells, and thereby they play a crucial role in orchestrating the resolution of tissue inflammation (20). PGE2, known as a proinflammatory lipid mediator (LM), exhibits diverse biological activities depending on the local inflammatory microenvironment and the receptor subtype on which it acts. Therefore, PGE2 also has immunosuppressive functions. For example, PGE2 inhibits inflammatory chemokine production by inhibiting NF-κB in a c-AMP–dependent manner via the EP4 receptor (21). In addition, PGE2 plays an important role in LM class switch from proinflammatory LMs to SPMs by inducing ALOX15 expression (22).

Mouse ALOX15 shares 74–81% amino acid identity with human ALOX15 and is considered an ortholog of human ALOX15. Usually, orthologs of enzymes perform similar functions between different species. Therefore, mouse ALOX15 is considered a functional homolog of human ALOX15, albeit with slightly different reaction specificity in the oxidation of arachidonic acid (23).

In the current study, the prognosis of ischemic AKI was observed to be worse in macrophage-specific Mafb-deficient mice. The finding was similar in the mice-transplanted fetal liver cells of mice carrying the human-type MAFB p.Leu239Pro mutant, which is a loss-of-function mutation of MAFB. Notably, ALOX15 expression decreased at both the mRNA and protein levels in Mafb-deficient macrophages infiltrating the kidney 24 h after ischemic AKI induction. Furthermore, our in vitro assay showed that MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway in macrophages. The results suggest that MAFB plays a crucial role for PGE2-mediated LM class switch by regulating the expression of ALOX15 under acute inflammatory conditions.

Macrophage lineage-specific Mafb-deficient mice (C57BL/6J) were generated by mating Mafbf/f mice with LysM-Cre mice (The Jackson Laboratory, Bar Harbor, ME) under the control of the endogenous Lys2 promoter. Mafbf/f and LysM-Cre mice were generated in our laboratory as described previously (24). Male 8- to 12-wk-old mice were used in the experiments.

Mafb GFP knock-in mice (Mafb heterozygous; Mafbgfp/−) with a C57BL/6J background were also used in this study (8).

The mice were maintained under specific pathogen-free conditions at the Laboratory Animal Resource Center of the University of Tsukuba. All experiments complied with relevant Japanese and institutional laws and guidelines, and were approved by the University of Tsukuba Animal Ethics Committee (23-041).

Renal ischemia-reperfusion injury (IRI) was performed as previously described (25). Briefly, a retroperitoneal incision was made under isoflurane anesthesia. The bilateral renal arteriovenous tissues were clamped for 60 (Fig. 1A) and 30 min (for other than Fig. 1A) using micro clamps (18055-02; Fine Science Tools). After the clamp was removed, reperfusion of both kidneys was visually confirmed. All experiments were performed on a 37°C warm plate to maintain body temperature.

FIGURE 1.

Loss of Mafb in macrophages worsens the prognosis of ischemic AKI. (A) Survival curve of Mafbf/f and Mafbf/f::LysM-Cre mice subjected to IRI with 60-min clamp (n = 6 for each group). p <0.01, by log-rank test. (B) Serum BUN and creatinine levels of Mafbf/f and Mafbf/f::LysM-Cre mice subjected to IRI with 30-min clamp (n = 6 for Mafbf/f, n = 5 for Mafbf/f::LysM-Cre). (C) Representative periodic acid–Schiff (PAS)–stained kidney from Mafbf/f and Mafbf/f::LysM-Cre mice at 7 d post-IRI. Scale bar, 100 µm. Arrowheads point to intraluminal debris. (D and E) Quantification of PAS+ intraluminal debris at the corticomedullary junction (D), as well as the acute tubular necrosis (ATN) scores (E) in the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice at 7 d post-IRI. Intraluminal debris (D) is presented as a percentage area per whole corticomedullary junction in a slide. (F) Fetal liver cells of WT or MAFB p.Leu239Pro (Mafbmt/mt) CD45.2 embryo were transplanted to lethally irradiated WT CD45.1 mice. After the 8-wk recovery period, mice were subjected to renal IRI. (G) Serum blood urea nitrogen (BUN) and creatinine levels of WT or Mafbmt/mt cell–transplanted mice subjected to IRI with a 30-min clamp. n = 5 for each group. (H) Representative PAS-stained kidney from WT or Mafbmt/mt cell–transplanted mice at 7 d post-IRI. Scale bar, 100 µm. Arrowheads point to intraluminal debris. (I) The ATN scores in the kidneys of WT or Mafbmt/mt cell–transplanted mice at 7 d post-IRI. All data are expressed as means ± SEM. *p <0.05, **p <0.01, ***p <0.001, by Welch t test.

FIGURE 1.

Loss of Mafb in macrophages worsens the prognosis of ischemic AKI. (A) Survival curve of Mafbf/f and Mafbf/f::LysM-Cre mice subjected to IRI with 60-min clamp (n = 6 for each group). p <0.01, by log-rank test. (B) Serum BUN and creatinine levels of Mafbf/f and Mafbf/f::LysM-Cre mice subjected to IRI with 30-min clamp (n = 6 for Mafbf/f, n = 5 for Mafbf/f::LysM-Cre). (C) Representative periodic acid–Schiff (PAS)–stained kidney from Mafbf/f and Mafbf/f::LysM-Cre mice at 7 d post-IRI. Scale bar, 100 µm. Arrowheads point to intraluminal debris. (D and E) Quantification of PAS+ intraluminal debris at the corticomedullary junction (D), as well as the acute tubular necrosis (ATN) scores (E) in the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice at 7 d post-IRI. Intraluminal debris (D) is presented as a percentage area per whole corticomedullary junction in a slide. (F) Fetal liver cells of WT or MAFB p.Leu239Pro (Mafbmt/mt) CD45.2 embryo were transplanted to lethally irradiated WT CD45.1 mice. After the 8-wk recovery period, mice were subjected to renal IRI. (G) Serum blood urea nitrogen (BUN) and creatinine levels of WT or Mafbmt/mt cell–transplanted mice subjected to IRI with a 30-min clamp. n = 5 for each group. (H) Representative PAS-stained kidney from WT or Mafbmt/mt cell–transplanted mice at 7 d post-IRI. Scale bar, 100 µm. Arrowheads point to intraluminal debris. (I) The ATN scores in the kidneys of WT or Mafbmt/mt cell–transplanted mice at 7 d post-IRI. All data are expressed as means ± SEM. *p <0.05, **p <0.01, ***p <0.001, by Welch t test.

Close modal

Blood samples were collected from the cheek veins of mice. Serum blood urea nitrogen (BUN) and creatinine concentrations were measured using DRI-CHEM (NX600; Fujifilm).

Kidney tissues were fixed in 4% paraformaldehyde and embedded in paraffin. Alternatively, for frozen sections, kidney tissues were fixed with 4% paraformaldehyde.

Periodic acid–Schiff (PAS) staining was performed on 3-µm sections of paraffin-embedded kidney blocks.

For the staining of MAFB, CD68, and F4/80, 5-µm sections of paraffin-embedded kidney blocks were incubated with a 1:100 dilution of rabbit monoclonal anti-MAFB Ab (E309X; Cell Signaling Technology), a 1:100 dilution of rabbit polyclonal anti-CD68 Ab (ab125212; Abcam), and a 1:200 dilution of rat monoclonal anti-F4/80 Ab (CL8940AP; Cedarlane). For staining of ALOX15 and GFP, 5-µm frozen sections were incubated with a 1:500 dilution of rabbit monoclonal anti–15-lipoxygenase 1 Ab (ab244205; Abcam) and a 1:500 dilution of chicken polyclonal anti-GFP Ab (Aves Lab). To stain MAFB and CD68, sections were incubated with Histofine simple mouse stain (rabbit; 414341; Nichirei). For the fluorescent staining of F4/80, ALOX15, and GFP, the sections were incubated with a 1:500 dilution of Alexa Fluor 647–conjugated donkey anti-rat IgG (ab150155; Abcam), a 1:500 dilution of Alexa Fluor 594–conjugated donkey anti-rabbit IgG (A21207; Invitrogen), and a 1:500 dilution of Alexa Fluor 488–conjugated goat anti-chicken IgG (A11039; Invitrogen). Nuclear staining was performed with Hoechst 33342 solution (346-07951; Dojindo), and sections were mounted using Fluoromount (K024; Diagnostic BioSystems). Specimens were analyzed using a Biolevo all-in-one fluorescence microscope (BZ-X810; Keyence).

The PAS-stained debris area at the corticomedullary junction was quantified using ImageJ software, and the percentage areas per whole section are presented. Six different fields (original magnification, ×100) were analyzed for each slide and the average value is displayed in the figure.

The acute tubular necrosis (ATN) score was based on the percentage of tubules that displayed cell necrosis, loss of brush border, cast formation, and tubule dilation. The scoring system ranged as follows: 0, none; 1, 10%; 2, 11–25%; 3, 26–45%; 4, 46–75%; and 5, >76% (26). Fifteen different fields (original magnification, ×200) were reviewed for each slide by a renal pathologist in a blinded manner.

Kidneys were digested with enzyme mixture (970 µl of HBSS, 20 µl of collagenase A [10103578001; Roche], 10 µl of DNase I). After digestion, the cells were incubated with anti-CD11b (clone M1/70; BioLegend), anti-Ly6C (clone HK1.4; BioLegend), and anti-Ly6G (clone RB6-8C5; GeneTex) Abs. Dead cells were stained by DAPI solution (340-07971; Cellstain). Flow cytometry analysis was performed using CytoFLEX (Beckman Coulter), and the results were analyzed using FlowJo software (Tree Star).

Kidneys were digested as described above and stained with Zombie Violet (BioLegend) at 100× dilution for 15 min at room temperature in the dark, followed by surface Abs for 25 min at 4°C in the dark. The cells were fixed with 1× fixation regent (10× fixation regent; 2.75 ml of formaldehyde in 47.25 ml of distilled water containing 0.32 g of NaH2PO4 and 0.2 g of Na2HPO4) for 30 min at room temperature in the dark. The cells were then permeabilized by washing with 0.1% saponin at 1000 rpm for 4 min at 25°C. The cells were incubated with anti–IL-6 Ab (BioLegend) for 30 min at room temperature in the dark, followed by washing with 0.1% saponin at 300 × g for 4 min at 25°C. Flow cytometry analysis was performed using CytoFLEX (Beckman Coulter), and the results were analyzed using FlowJo software (Tree Star). For intracellular staining of IL-6, mice were injected with 250 µg of brefeldin A (BioLegend) 6 h before the experiment, and all reagents for kidney digestion and staining contained brefeldin A 1× injection.

Kidneys were digested using the method described above and incubated with an anti-CD11b Ab conjugated to allophycocyanin. These cells were then incubated with anti-allophycocyanin microbeads (130-090-855; Miltenyi Biotec), applied to the mass spectrometry (MS) columns (130-042-201; Miltenyi Biotec), and passed through the magnetic column. The MS column was then removed from the magnet, and 1 ml of MACS buffer via the plunger was used for flushing out the microbead-labeled cells from the column, initially retained by the magnetic field. The microbead-labeled cells were incubated with anti-CD11b, anti-Ly6C, and anti-Ly6G Abs. Ly6GlowLy6C+CD11b+cells were sorted into Isogen (311-02501; Nippon Gene) using a MoFlo XDP cell sorter (Beckman Coulter).

Total RNA was extracted from Ly6GlowLy6C+CD11b+cells using TRIzol reagent (15596026; Thermo Fisher Scientific). The RNA sequencing (RNA-seq) library was prepared using the NEBNext Ultra directional RNA library prep kit (New England Biolabs, Ipswich, MA) after rRNA depletion (NEBNext rRNA depletion kit; New England Biolabs). Paired-end (2 × 36 bases) sequencing was performed using NextSeq 500 (Illumina). The FASTQ files were imported into the CLC Genomics Workbench (version 10.1.1; Qiagen). The sequence reads were mapped to the mouse reference genome (mm10). Gene expression levels were calculated as total read counts and normalized using the quantile method. Genes with zero counts in all samples were excluded, and differential expression was analyzed using the empirical analysis of DGE tool (edgeR test) within the CLC Main Workbench (version 7.7.3; Qiagen). Differentially expressed genes were extracted using false discovery rate–corrected p <0.05.

Total RNA was extracted using Isogen (311-02501; Nippon Gene). cDNA was synthesized using the QuantiTect reverse transcription kit (205313; Qiagen). The mRNA levels of the mouse genes of interest were measured using SYBR Green PCR master mix (QPS-201; Takara Bio). mRNA levels were normalized to mouse Hprt mRNA levels. The primer sequences for quantitative RT-PCR (qRT-PCR) were as follows: Hprt forward (5′-CAAACTTTGCTTTCCCTGGT-3′) and reverse (5′-CAAGGGCATATCCAACAACA-3′); Mafb forward (5′-TGAATTTGCTGGCACTGCTG-3′) and reverse (5′-AAGCACCATGCGGTTCATACA-3′); Alox15 forward (5′-CGTGGTTGAAGACTCTCAAGG-3′) and reverse (5′-CGAAATCGCTGGTCTACAGG-3′); C1qa forward (5′-GGATGGGGCTCCAGGAAATC-3′) and reverse (5′-CTGATATTGCCTGGATTGCC-3′); C1qb forward (5′-TGGCTCTGATGGCCAACCAG-3′) and reverse (5′-GACTTTCTGTGTAGCCCCGT-3′); C1qc forward (5′-AGGACGGGCATGATGGACTC-3′) and reverse (5′-TGAATACCGACTGGTCTTC-3′); Msr1 forward (5′-TGGAGGAGAGAATCGAAAGCA-3′) and reverse (5′-CTGGACTGACGAAATCAAGGAA-3′); and Ptgs2 forward (5′-GTATCAGAACCGCATTGCCTCTGA-3′) and reverse (5′-CGGCTTCCAGTATTGAGGAGAACAGAT-3′).

Liquid chromatography–tandem MS (LC-MS/MS)–based targeted lipidomics was performed as described previously (27). Briefly, frozen kidneys in ice-cold methanol (Wako) were homogenized by Zirconia beads of 3.0 and 5.0 mmφ (TOMY) using a Precellys 24 homogenizer (Bertin Technologies) and kept at −30°C overnight. The methanolic supernatant was applied to MonoSpin C18-AX cartridges (GL Science, Tokyo, Japan) to extract lipid metabolites in the presence of deuterated internal standards: 1 ng of arachidonic acid–d8, 15-hydroxyeicosatetraenoic acid–d8, LTB4-d4, LTD4-d5, and PGE2-d4 (Cayman Chemical, Ann Arbor, MI) for monitoring recovery rates during sample preparation. For LC-MS/MS analysis, a triple-quadrupole linear ion-trap mass spectrometer (QTRAP 4500, AB Sciex, Foster City, CA) equipped with an Acquity UPLC BEH C18 column (1.0 × 150 mm, 1.7-µm particle size; Waters, Milford, MA) was used. MS/MS analysis was performed in the negative ion mode, and metabolites were identified and quantified by multiple reaction monitoring. Calibration curves between 1 and 1000 pg and the LC retention times for each compound were established using synthetic standards. Raw data were analyzed using MultiQuant software (Sciex). PD1, PDX, and their stereoisomers were not separatable under the LC-MS/MS setting above.

The mouse macrophage RAW264.7 cell line was cultured in DMEM supplemented with 10% FBS and 1% l-glutamine.

To collect peritoneal macrophages, a thioglycolate medium was injected into the abdominal cavities of the mice. Four days after the injection, PBS was injected into the abdominal cavities of the mice. The PBS was then collected and centrifuged at 1000 rpm for 4 min at 4°C. The cell pellet was plated in macrophage serum-free medium (1265074; Life Technologies) supplemented with 1% penicillin/streptomycin for 3 h to remove neutrophils (nonadherent cells).

Macrophages were stimulated with LPS (500 ng/ml) for 3 d and PGE2 (Cayman Chemical) (1000 ng/ml) for 2 d.

PGE2 was from Cayman Chemical, and L161982 (EP4 antagonist) was from Tocris Bioscience for in vitro study.

The promoter region of the Alox15 gene was examined using the UCSC Genome Browser (http://genome.ucsc.edu/index.html). Fragments of 200 or 500 bp of the Alox15 promoter were extracted from C57BL/6 mouse genomic DNA and cloned into a pGL4.10 luciferase vector lacking a promoter (Promega). Site-specific mutations were introduced into the putative MARE sequence at site 2 using a QuickChange site-directed mutagenesis kit (Agilent Technologies) with the primer sequences as follows: Alox15 mut2-MARE forward (5′-GAGGCCCCGCCCCTTCGCGAGTCTAGTTTAGGGTTGAGC-3′) and reverse (5′-GCTCAACCCTAAACTAGACTCGCGAAGGGGCGGGGCCTC-3′). In addition, a mutated version of the putative MARE sequence at site 1 was synthesized by GenScript.

To investigate the potential activation of the Alox15 promoter by MAFB, vectors containing the Alox15 promoter were cotransfected with a Mafb expression vector into RAW264.7 cells. A pRL-TK vector (Promega) was also introduced into the RAW264.7 cells for normalization purposes. Transfection was carried out using FuGENE HD transfection reagent (Promega), followed by a 48-h cell culture. Luciferase activity was assessed using a Dual-Luciferase reporter assay system (Promega). Briefly, after transfection for 48 h, the cells were lysed, and 20 µl of cell lysate was transferred to 100 µl of luciferase assay reagent II. The samples were then measured using a luminometer. To halt the reaction, 100 µl of Stop&Glo reagent was added, and readings were taken again. A pRL-TK vector expressing Renilla reniformis luciferase was used to normalize the transfection efficiency.

Data are presented as mean ± SEM. Statistical significance for the difference in mean derived from several groups was determined using a Welch t test.

RNA-seq data are deposited in the DNA Databank of Japan (https://ddbj.nig.ac.jp/resource/sra-submission/DRA017447).

To elucidate the molecular mechanisms by which MAFB in myeloid cells alleviates AKI in detail, Mafbf/f::LysM-Cre mice, in which the Mafb gene was deleted specifically in the myeloid cell lineage, were used, as previously reported (11). AKI was induced in Mafbf/f and Mafbf/f::LysM-Cre mice using the IRI model and the prognosis was compared. Our results showed that with a 60-min clamp, Mafbf/f::LysM-Cre mice began to die on day 2, and the survival rate was ∼17% on day 6, although none of the Mafbf/f mice died after IRI (Fig. 1A). In addition, serum BUN and creatinine levels were higher in Mafbf/f::LysM-Cre mice (Supplemental Fig. 1A). Then, we shortened the clamping time to 30 min and conducted all subsequent experiments under these conditions. Although both Mafbf/f and Mafbf/f::LysM-Cre mice did not die by a 30-min clamp, the levels of serum BUN and creatinine were significantly higher in Mafbf/f::LysM-Cre mice (Fig. 1B). Because Mafbf/f::LysM-Cre mice started to die on day 2 after IRI with the 60-min clamp, and serum BUN and creatinine levels were significantly higher in Mafbf/f::LysM-Cre mice on day 1 after IRI in the 30-min clamp, MAFB in macrophages may play a significant role in suppressing inflammation from a very early stage. PAS staining of kidney sections revealed significantly higher amounts of debris in the kidneys of Mafbf/f::LysM-Cre mice, as well as impaired tissue repair, as indicated by the ATN score at 7 d post-IRI (Fig. 1C–E). It has been reported that the corticomedullary junction of the kidney is mainly injured in ischemic AKI (28). Consistently, we observed debris and injury mainly in the corticomedullary junction (Fig. 1C). These results indicated a worse prognosis for AKI in Mafbf/f::LysM-Cre mice than in Mafbf/f mice.

Whether similar results could be obtained in other MAFB-dysfunctional mouse models was tested. Previously, we have reported that human MAFB p.Leu239Pro mutant mice exhibit a phenotype similar to Mafb-deficient mice (29). To generate mice with this mutation only in myeloid cells, we transplanted fetal liver cells from MAFB p.Leu239Pro mutant (Mafbmt/mt) mice into 10 Gy–irradiated CD45.1 wild-type (WT) recipient mice (Fig. 1F). Chimerism in recipient mice was confirmed by FACS analysis, and mice with >95% chimerism were used for further experiments (Supplemental Fig. 1B). Ischemic AKI was induced in fetal liver cell–transplanted mice. Serum BUN and creatinine levels were significantly higher in Mafbmt/mt cell–transplanted mice than those in WT cell–transplanted mice (Fig. 1G). PAS staining of kidney sections revealed impaired tissue repair in Mafbmt/mt cell–transplanted mice, as indicated by the ATN score at 7 d post-IRI (Fig. 1H, 1I).

These results suggest that the loss of MAFB in myeloid cells leads to impaired kidney tissue repair after IRI, highlighting the pivotal role of MAFB in alleviating AKI in macrophages. After the onset of ischemic AKI, macrophages infiltrate damaged sites in the kidney (3). Therefore, we next examined MAFB expression in the macrophages, using Mafbgfp/+ mice in which the Mafb gene locus is heterozygously replaced by the gfp gene (8). Mafbgfp/+ mice were subjected to IRI and dissected 5, 12, or 24 h after IRI, and whole kidneys were extracted for FACS analysis. The gating strategy is presented in Supplemental Fig. 1C. We defined Ly6GlowLy6C+CD11b+cells as macrophages, and consistent with a previous study (3), their proportion among all live cells was significantly increased until at least 24 h after IRI (Fig. 2A, 2B). To examine whether these macrophages express MAFB, we measured the percentage of GFP+ macrophages among all macrophages. Before IRI, the percentage of GFP+ macrophages was ∼50%. However, following IRI, the percentage of GFP+ macrophages significantly increased at 5 h and was maintained at a high level for at least 24 h post-IRI (Fig. 2A, 2C). These results indicated that the number of MAFB+ macrophages increased in the kidney after AKI induction.

FIGURE 2.

MAFB deficiency alters leukocyte infiltration and activation patterns during AKI. (A) Kidney cells of WT or MafbGFP/+ mice at the indicated time points were stained with CD11b, Ly6G, and Ly6C Abs. Ly6GlowLy6C+CD11b+ cells were gated as macrophages, and the percentages of GFP+ macrophages among all macrophages were measured. (B and C) The percentage of all macrophages in total live cells (B) and the percentage of GFP+ macrophages in all macrophages (C) at the indicated time points are plotted with SEM (n = 3 for each group). (D) Immunostaining of IRI (−) or 5 h post-IRI kidney of Mafbf/f mice with anti-MAFB Ab. The image represents a corticomedullary junction of the kidney. Arrowheads point to the MAFB-positive areas. Scale bar, 100 µm. (E) CD11b+ cells were isolated from kidneys of Mafbf/f or Mafbf/f::LysM-Cre mice 24 h after IRI by MACS. Flow cytometric analysis was performed on these CD11b+ cells with anti-CD11b, anti-Ly6G, and anti-Ly6C Abs. Ly6GhighCD11b+cells were gated as neutrophils, and Ly6C+Ly6GlowCD11b+ cells were gated as macrophages. (F) The absolute number of cells in each cell population was calculated from the overall number of CD11b+ cells taken from the kidney tissues and the percent of each cell population (n = 9 for the Mafbf/f group and n = 8 for the Mafbf/f::LysM-Cre group). (G) Kidney cells of Mafbf/f or Mafbf/f::LysM-Cre mice at 24 h post-IRI were stained with CD11b, Ly6G, and Ly6C Abs. Ly6GlowLy6C+CD11b+ cells were gated as macrophages. Numbers indicate the percentage of Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in all macrophages. (H) Percentage of Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in all macrophages (n = 6 for each group). (I) IL-6 expression levels in Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in the kidney of Mafbf/f mice 24 h after IRI were measured by FACS analysis. (J) Percentages of Ly6Clow, Ly6Cint, and Ly6Chigh IL-6+ macrophages in all macrophages (n = 3 for each group). All data are expressed as means ± SEM. *p <0.05, **p <0.01, *** p <0.001, by Welch t test.

FIGURE 2.

MAFB deficiency alters leukocyte infiltration and activation patterns during AKI. (A) Kidney cells of WT or MafbGFP/+ mice at the indicated time points were stained with CD11b, Ly6G, and Ly6C Abs. Ly6GlowLy6C+CD11b+ cells were gated as macrophages, and the percentages of GFP+ macrophages among all macrophages were measured. (B and C) The percentage of all macrophages in total live cells (B) and the percentage of GFP+ macrophages in all macrophages (C) at the indicated time points are plotted with SEM (n = 3 for each group). (D) Immunostaining of IRI (−) or 5 h post-IRI kidney of Mafbf/f mice with anti-MAFB Ab. The image represents a corticomedullary junction of the kidney. Arrowheads point to the MAFB-positive areas. Scale bar, 100 µm. (E) CD11b+ cells were isolated from kidneys of Mafbf/f or Mafbf/f::LysM-Cre mice 24 h after IRI by MACS. Flow cytometric analysis was performed on these CD11b+ cells with anti-CD11b, anti-Ly6G, and anti-Ly6C Abs. Ly6GhighCD11b+cells were gated as neutrophils, and Ly6C+Ly6GlowCD11b+ cells were gated as macrophages. (F) The absolute number of cells in each cell population was calculated from the overall number of CD11b+ cells taken from the kidney tissues and the percent of each cell population (n = 9 for the Mafbf/f group and n = 8 for the Mafbf/f::LysM-Cre group). (G) Kidney cells of Mafbf/f or Mafbf/f::LysM-Cre mice at 24 h post-IRI were stained with CD11b, Ly6G, and Ly6C Abs. Ly6GlowLy6C+CD11b+ cells were gated as macrophages. Numbers indicate the percentage of Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in all macrophages. (H) Percentage of Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in all macrophages (n = 6 for each group). (I) IL-6 expression levels in Ly6Clow, Ly6Cint, and Ly6Chigh macrophages in the kidney of Mafbf/f mice 24 h after IRI were measured by FACS analysis. (J) Percentages of Ly6Clow, Ly6Cint, and Ly6Chigh IL-6+ macrophages in all macrophages (n = 3 for each group). All data are expressed as means ± SEM. *p <0.05, **p <0.01, *** p <0.001, by Welch t test.

Close modal

Next, to directly examine the expression of MAFB protein, we performed immunostaining of WT kidneys with an anti-MAFB Ab. In the kidney without IRI, there were only slight MAFB+ signals; however, 5 h after IRI, the MAFB+ signals increased, mainly at the corticomedullary junction of the kidney (Fig. 2D). The results suggest that the MAFB+ macrophages are recruited to the damaged site in the kidney after AKI onset.

Several research groups have suggested that myeloid cell infiltration affects the prognosis of ischemic AKI significantly (30, 31). Therefore, we examined the degree of myeloid cell infiltration into the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice 24 h after IRI using FACS analysis. CD11b+ cells were isolated from whole kidneys using MACS, and FACS analysis was performed. The absolute numbers of CD11b+ cells, neutrophils (Ly6GhighCD11b+ cells), and macrophages (Ly6C+Ly6GlowCD11b+ cells) were significantly higher in Mafbf/f::LysM-Cre mice (Fig. 2E, 2F), suggesting severe inflammation in Mafbf/f::LysM-Cre mice.

Proper migration to the injury site is essential for macrophages to perform their functions (3). To examine whether MAFB is involved in the proper migration of macrophages to the site of injury, immunostaining for macrophage markers CD68 and F4/80 was performed on the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice at 24 h post-IRI. According to PAS staining results, the corticomedullary junction of the kidney was primarily damaged by IRI (Fig. 1C, 1H). Consistently, CD68+ and F4/80+ cells were observed at the damaged corticomedullary junctions in the kidneys of both Mafbf/f and Mafbf/f::LysM-Cre mice (Supplemental Fig. 1E, 1F). Our results suggest that the infiltration ability of macrophages is not impaired by Mafb deficiency.

Mafb deficiency does not affect macrophage infiltrative capacity, suggesting that MAFB is involved in other functions of macrophages. Clements et al. (32) reported that macrophages infiltrating the kidney after IRI are divided into three populations according to the expression level of Ly6C: Ly6ChiCD11b+ proinflammatory population, Ly6CintCD11b+ wound healing population, and Ly6ClowCD11b+ profibrotic population. We examined whether the deficiency of Mafb altered the composition of these macrophage populations. We performed FACS analysis of kidneys from Mafbf/f and Mafbf/f::LysM-Cre mice after IRI. The gating strategy is shown in Supplemental Fig. 1D. The results showed that, without IRI, there was no difference in the composition of Ly6C populations between Mafbf/f and Mafbf/f::LysM-Cre mice (Supplemental Fig. 1G, 1H); however, at 24 h after AKI induction, the Ly6Cint macrophage population was increased in the kidney of Mafbf/f mice (Fig. 2G, 2H). The results suggested that MAFB shifts the macrophage phenotype toward an anti-inflammatory state in an IRI-dependent manner. To further analyze the feature of these Ly6C macrophage populations in detail, IL-6 expression in each population was measured by FACS. Ly6C exacerbates inflammation in the acute phase of AKI (33). Moreover, patients with higher IL-6 expression have a worse prognosis for AKI (34). FACS analysis showed significantly higher IL-6 expression in the Ly6Chigh population compared with in the Ly6Cint and Ly6Clow populations (Fig. 2I, 2J). The results reinforce previous reports that Ly6Chigh is an inflammatory population.

To investigate how Mafb deficiency causes a shift in the macrophage phenotype toward a proinflammatory state, we isolated macrophages from the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice at 24 h post-IRI and compared gene expression by RNA-seq analysis. For the isolation of macrophages, CD11b+ cells were isolated from the kidneys by MACS, and Ly6GlowLy6C+CD11b+ cells were sorted from these CD11b+ cells as macrophages (Supplemental Fig. 2A). RNA-seq analysis showed that gene expressions were altered in macrophages by the deficiency of Mafb, as shown by a heatmap (Supplemental Fig. 2B). Pathway analysis of these differentially expressed genes showed that the deficiency of Mafb caused abnormalities in the inflammatory response of macrophages (Supplemental Fig. 2C). These results suggest that the deficiency of Mafb in macrophages shifts the macrophage phenotype toward a proinflammatory state.

The volcano plot of the RNA-seq results showed that the expression of Alox15 and Syne1 was extremely downregulated in Mafb-deficient macrophages (Fig. 3A, 3B). Because ALOX15 is known to play a significant role in suppressing acute inflammation (17, 18), we hypothesized that MAFB promotes inflammation resolution in ischemic AKI by regulating the expression of Alox15 in macrophages. ALOX15 is a lipoxygenase that catalyzes the oxidation of essential fatty acids. ALOX15 expressed in macrophages produces precursors of inflammation-suppressing SPMs by oxidizing essential fatty acids at the site of inflammation (18, 19). SPMs have anti-inflammatory and proresolving functions in immune cells; therefore, they play essential roles in orchestrating the resolution of tissue inflammation (20).

FIGURE 3.

MAFB regulates the expression of Alox15 in macrophages infiltrated into the kidney of ischemic AKI. (A) Volcano plot of overall gene expression in macrophages sorted from the kidney of Mafbf/f and Mafbf/f::LysM-Cre mice at 24 h post-IRI. The blue and red dots indicate the downregulated and upregulated genes in the Mafbf/f::LysM-Cre group, respectively. (B) Normalized count of Alox15 mRNA from RNA-seq data (n = 3 for each group). ***False discovery rate<0.001, edgeR. (C) Immunostaining of kidney from Mafbf/f and Mafbf/f::LysM-Cre mice without IRI at 12 and 24 h post-IRI with anti-ALOX15 and anti-F4/80 Ab. Scale bars, 100 µm. (D) The number of ALOX15+F4/80+ cells per field is presented. Seven different fields (original magnification, ×200) were analyzed for each slide. All data are expressed as means ± SEM. **p <0.01, by Welch’s t test. (E) Immunostaining of kidney from WT and MafbGFP/+ mice at 24 h post-IRI with anti-ALOX15 and anti-GFP Ab. Arrowheads point to the ALOX15+GFP+ area. Scale bar, 50 µm.

FIGURE 3.

MAFB regulates the expression of Alox15 in macrophages infiltrated into the kidney of ischemic AKI. (A) Volcano plot of overall gene expression in macrophages sorted from the kidney of Mafbf/f and Mafbf/f::LysM-Cre mice at 24 h post-IRI. The blue and red dots indicate the downregulated and upregulated genes in the Mafbf/f::LysM-Cre group, respectively. (B) Normalized count of Alox15 mRNA from RNA-seq data (n = 3 for each group). ***False discovery rate<0.001, edgeR. (C) Immunostaining of kidney from Mafbf/f and Mafbf/f::LysM-Cre mice without IRI at 12 and 24 h post-IRI with anti-ALOX15 and anti-F4/80 Ab. Scale bars, 100 µm. (D) The number of ALOX15+F4/80+ cells per field is presented. Seven different fields (original magnification, ×200) were analyzed for each slide. All data are expressed as means ± SEM. **p <0.01, by Welch’s t test. (E) Immunostaining of kidney from WT and MafbGFP/+ mice at 24 h post-IRI with anti-ALOX15 and anti-GFP Ab. Arrowheads point to the ALOX15+GFP+ area. Scale bar, 50 µm.

Close modal

To confirm the reduced ALOX15 expression in macrophages in Mafbf/f::LysM-Cre mice at the protein level, immunostaining of ALOX15 and the macrophage marker F4/80 was performed on the kidneys of Mafbf/f and Mafbf/f::LysM-Cre mice before AKI induction and at 12 and 24 h post-AKI. The results showed no expression of ALOX15 in the kidney before IRI in both Mafbf/f and Mafbf/f::LysM-Cre mice (Fig. 3C, 3D, top). ALOX15+F4/80+ cells were already present in the kidneys of Mafbf/f mice at 12 h post-IRI and their number increased further at 24 h; however, the number of ALOX15+F4/80+ cells was significantly lower in the kidneys of Mafbf/f::LysM-Cre mice than in those of Mafbf/f mice at both time points (Fig. 3D, 3D, middle, bottom). The results suggest that MAFB induces ALOX15 expression in macrophages in an AKI-dependent manner. Subsequently, LC-MS analysis for LMs was performed on the kidneys of Mafbf/f mice before and at 12 and 24 h after AKI induction to verify whether the production of SPMs by ALOX15 actually takes place in the AKI-induced kidneys (Supplemental Fig. 3A). The results showed that 17-HDoHE, a precursor of the resolvin D series of SPMs, increased after 12 h. ALOX15 is essential for the production of 17-HDoHE from docosahexaenoic acid (DHA). After 24 h, 17-HDoHE was reduced, possibly due to conversion of 17-HDoHE to the resolvin D series. Previous studies have shown that the resolvin D series is protective against ischemic AKI (35, 36). Resolvins D1 and D2 were not detected in the current study; however, according to a previous study, the amount of the resolvin D series is not largely altered unless DHA is administrated before AKI induction (35). In the current study, the analysis was performed under natural conditions without DHA administration; therefore, resolvins would have been below detection sensitivity. Conversely, not much change was observed in the mediators produced from arachidonic acid by ALOX15. Indeed, there are reports of improved AKI prognosis in dogs and mice fed DHA rather thanarachidonic acid (37–39). PGE2, PGF2a, and TxB2, a metabolite of TxA2, was reduced at 24 h, suggesting that LM class switch from inflammatory LMs to SPMs would have occurred between 12 and 24 h after AKI induction.

To confirm that MAFB+ cells express ALOX15, immunostaining was performed on the kidneys of Mafbgfp/+ mice 24 h after IRI. The results showed that ALOX15 colocalized with GFP (Fig. 3E). However, not all GFP+ cells colocalized with ALOX15, suggesting that a limited macrophage population expresses ALOX15.

Several downstream target genes of MAFB in macrophages have been identified, such as C1q genes, AIM, and Msr1 (11) (12, 13). We measured the expression of these MAFB target genes in macrophages infiltrating the kidney at 24 h after IRI, using qRT-PCR. The expression of Msr1 and C1q genes was decreased significantly; however, the expression of AIM did not change in macrophages from Mafbf/f::LysM-Cre mice (Supplemental Fig. 3B). Because MSR1 and C1q are involved in the clearance of DAMPs and efferocytosis, respectively (11, 13), MAFB in macrophages may promote the suppression of inflammation by regulating the expression of the genes in ischemic AKI. Further studies are required to elucidate the involvement of MSR1 and C1q.

The Alox15 gene is expressed in M2-type macrophages following treatment with IL-4, IL-13, and RXR/LXR agonists (19). However, its expression is also induced in M1-type macrophages activated by LPS (40).

Therefore, we investigated whether MAFB regulates the expression of Alox15 in M1- or M2-type macrophages using in vitro–cultured macrophages. For this purpose, we first treated bone marrow–derived macrophages from Mafbf/f and Mafbf/f::LysM-Cre mice with the Th2 cytokines IL-4, IL-13, and 9cRA/T0901317 (an RXR/LXR agonist) and compared the expression of Alox15 by qRT-PCR. The expression of Alox15 was also measured in thioglycolate-elicited peritoneal macrophages from Mafbf/f and Mafbf/f::LysM-Cre mice because such macrophages are polarized to the M2 type (41). The results showed that in bone marrow–derived macrophages treated with Th2 cytokines, RXR/LXR agonists, and thioglycollate-elicited peritoneal macrophages, the expression of Alox15 was increased significantly in macrophages from Mafbf/f::LysM-Cre mice (Supplemental Fig. 3C, 3D). The results suggest that, in M2-type macrophages, MAFB negatively regulates Alox15 expression. M2 macrophages have an important profibrotic role in chronic inflammatory conditions (42). Under chronic inflammatory conditions, such as CKD, ALOX15 has been reported to worsen prognosis by promoting fibrosis (43–45). Therefore, in M2 macrophages, MAFB would downregulate ALOX15 expression to suppress fibrosis. This regulation differs from the response observed during acute inflammation, such as AKI. Further analyses are required to clarify the molecular mechanism by which MAFB suppresses ALOX15 expression in M2 macrophages.

Next, we examined the regulation of Alox15 expression by MAFB in M1-type macrophages. For this purpose, thioglycollate-elicited peritoneal macrophages from Mafbf/f and Mafbf/f::LysM-Cre mice were primed with LPS, and Alox15 expression was compared using qRT-PCR. In contrast to in the M2-type macrophages, Alox15 expression was decreased significantly in Mafb-deficient macrophages 3 d after LPS treatment (Fig. 4A). The trend was consistent with the in vivo results (Fig. 3B, 3D). Because MAFB expression is regulated by the COX-2/PGE2/EP4 pathway in macrophages infiltrating the kidney after IRI, and LPS induces COX-2 expression (16, 46), we hypothesized that MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway in macrophages. EP4 induced MAFB expression via the cAMP/PKA/CREB pathway (16). The expression of Ptgs2, a gene encoding COX-2, was induced after LPS treatment in both Mafbf/f and Mafbf/f::LysM-Cre mice, and induction of Mafb expression was observed in Mafbf/f-derived macrophages after 2 d of treatment (Fig. 4B). The results suggest that MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway. To further confirm that MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway, we primed peritoneal macrophages from Mafbf/f and Mafbf/f::LysM-Cre mice with PGE2 and performed qRT-PCR. The results showed that Mafb expression increased in Mafbf/f-derived macrophages significantly from day 1 after treatment (Fig. 4C). In addition, Alox15 expression was significantly lower in Mafb-deficient macrophages on day 2 after PGE2 treatment (Fig. 4D). As PGE2 exerts its effects at the picomolar to nanomolar levels in vivo, we investigated the PGE2 concentration required for MAFB-mediated Alox15 induction in macrophages. The result showed that Mafb expression was induced in Mafbf/f-derived macrophages from a PGE2 concentration of 10 nM (Fig. 4E, left). Moreover, Mafb induction led to Alox15 induction in Mafbf/f-derived macrophages starting at a PGE2 concentration of 10 nM, whereas no Alox15 induction was observed in Mafbf/f::LysM-Cre-derived macrophages (Fig. 4R, right). Furthermore, we examined whether administration of an EP4 antagonist (L161982) abolishes the expression of Mafb and Alox15 downstream of COX-2/PGE2/EP4. As expected, LPS-induced Mafb and Alox15 expression was suppressed by inhibition of EP4 receptors (Fig. 4F). With regard to Alox15 mRNA, two out of five L161982-administrated samples were excluded because their dissociation curves showed abnormal shapes. The findings suggest that Alox15 was not amplified properly in these two samples. The results indicate that MAFB regulates the expression of Alox15 under the COX-2/PGE2/EP4 pathway in macrophages.

FIGURE 4.

MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway in macrophages. (A and B) Thioglycollate-elicited PMs were primed with LPS, and Alox15 mRNA levels were measured 3 d after treatment using qRT-PCR (A). Ptgs2 mRNA in the Mafbf/f or Mafbf/f::LysM-Cre group (left) and Mafb mRNA in the Mafbf/f group (right) were measured using qRT-PCR (B). (CE) Thioglycollate-elicited peritoneal macrophages were primed with PGE2. Mafb mRNA in the Mafbf/f group was measured by qRT-PCR (C). The Alox15 mRNA was measured 2 d after treatment (D). In (A)–(D), n = 3 for each group. Thioglycollate-elicited PMs were primed with 10 nM, 100 nM, and 1 µM PGE2 and Mafb mRNA in the Mafbf/f group (left), and Alox15 mRNA levels in each group (right) were measured by RT-qPCR. Data are shown with the expression level at PGE2 (−) as 1. n = 8 for Mafbf/f, n = 5 for Mafbf/f::LysM-Cre (E). (F) LPS-induced Mafb and Alox15 expression was prevented using an EP4 antagonist (L161982) at day 3. All data are expressed as means ± SEM. *p <0.05, **p <0.01, ***p <0.001, by Welch t test. For (F), the p value was adjusted using the Holm method.

FIGURE 4.

MAFB regulates Alox15 expression under the COX-2/PGE2/EP4 pathway in macrophages. (A and B) Thioglycollate-elicited PMs were primed with LPS, and Alox15 mRNA levels were measured 3 d after treatment using qRT-PCR (A). Ptgs2 mRNA in the Mafbf/f or Mafbf/f::LysM-Cre group (left) and Mafb mRNA in the Mafbf/f group (right) were measured using qRT-PCR (B). (CE) Thioglycollate-elicited peritoneal macrophages were primed with PGE2. Mafb mRNA in the Mafbf/f group was measured by qRT-PCR (C). The Alox15 mRNA was measured 2 d after treatment (D). In (A)–(D), n = 3 for each group. Thioglycollate-elicited PMs were primed with 10 nM, 100 nM, and 1 µM PGE2 and Mafb mRNA in the Mafbf/f group (left), and Alox15 mRNA levels in each group (right) were measured by RT-qPCR. Data are shown with the expression level at PGE2 (−) as 1. n = 8 for Mafbf/f, n = 5 for Mafbf/f::LysM-Cre (E). (F) LPS-induced Mafb and Alox15 expression was prevented using an EP4 antagonist (L161982) at day 3. All data are expressed as means ± SEM. *p <0.05, **p <0.01, ***p <0.001, by Welch t test. For (F), the p value was adjusted using the Holm method.

Close modal

Next, to determine MAFB-responsive regions in the Alox15 promoter, we searched for putative transcription factor–binding sites in the promoter of the Alox15 gene using the UCSC Genome Browser (http://genome.ucsc.edu/index.html). Of note, a potential half of the MARE (half-MARE) sites were identified as −267 bp (site 1) and −58 bp (site 2) upstream of the transcription initiation site of Alox15. The half-MARE sequence in site 1 is relatively conserved among multiple mammalian species but not in site 2 (Fig. 5A). To evaluate the potential of MAFB to interact with these half-MAREs, we constructed luciferase reporter genes linked to the 500 or 200 bp of the Alox15 promoter. These constructs were cotransfected with an MAFB-expressing vector into RAW264.7 cell line. The results showed that the MAFB-expressing vector activated only 500 bp of the Alox15 promoter but not 200 bp (Fig. 5B), suggesting that MAFB regulates the expression of Alox15 through the half-MARE at site 1. To confirm this result, a mutation was inserted at the site 1 and site 2 half-MARE site (Fig. 5B). As expected, the Alox15 promoter was deactivated when the mutation was inserted at site 1 but not at site 2 (Fig. 5B). These results suggest that MAFB regulates the Alox15 gene through a half-MARE site −267 bp upstream of the transcriptional initiation site of Alox15.

FIGURE 5.

MAFB regulates the Alox15 gene through the half-MARE site at −267 bp upstream of the transcriptional initiation site of Alox15. (A) Half of the Maf recognition element (half-MARE) sites were identified in Alox15 gene promoters using the UCSC Genome Browser. The half-MARE site (site 1) in the Alox15 gene promoter (bold) was highly conserved among mammalian species. (B) An MAFB-expressing vector was cotransfected with luciferase reporter constructs of Alox15 promoter containing half-MARE sites or mutant half-MARE sites into RAW264.7 cells. The luciferase activity was analyzed 48 h after transfection. The term “mut” indicates three base mutations in the half-MARE sites. Data are presented as the means ± SEM. n = 3 for each group.

FIGURE 5.

MAFB regulates the Alox15 gene through the half-MARE site at −267 bp upstream of the transcriptional initiation site of Alox15. (A) Half of the Maf recognition element (half-MARE) sites were identified in Alox15 gene promoters using the UCSC Genome Browser. The half-MARE site (site 1) in the Alox15 gene promoter (bold) was highly conserved among mammalian species. (B) An MAFB-expressing vector was cotransfected with luciferase reporter constructs of Alox15 promoter containing half-MARE sites or mutant half-MARE sites into RAW264.7 cells. The luciferase activity was analyzed 48 h after transfection. The term “mut” indicates three base mutations in the half-MARE sites. Data are presented as the means ± SEM. n = 3 for each group.

Close modal

In conclusion, MAFB expressed on macrophages would promote the resolution of inflammation in ischemic AKI by inducing ALOX15 expression to produce SPMs under the COX-2/PGE2/EP4 pathway (Fig. 6).

FIGURE 6.

Mechanistic scenario for induction of ALOX15 by MAFB in macrophages. In macrophages infiltrating the kidney after IRI, COX-2–derived PGE2 activates the EP4 receptor, and MAFB expression is induced. Under the COX-2/PGE2/EP4 pathway, MAFB induces the expression of ALOX15. ALOX15 would promote the resolution of inflammation in ischemic AKI by producing specialized proresolving mediators (SPMs) from DHA.

FIGURE 6.

Mechanistic scenario for induction of ALOX15 by MAFB in macrophages. In macrophages infiltrating the kidney after IRI, COX-2–derived PGE2 activates the EP4 receptor, and MAFB expression is induced. Under the COX-2/PGE2/EP4 pathway, MAFB induces the expression of ALOX15. ALOX15 would promote the resolution of inflammation in ischemic AKI by producing specialized proresolving mediators (SPMs) from DHA.

Close modal

A previous study has reported participation of MAFB downstream of the COX-2/PGE2/EP4 pathway in ischemic AKI (16). In macrophage-specific Mafb-deficient mice, renal function deteriorates during AKI onset, and the absence of MAFB leads to a decline in anti-inflammatory gene expression (16). However, the target genes of MAFB in the pathway remain unknown. According to the findings of the current study, MAFB regulates Alox15 expression in macrophages under the COX-2/PGE2/EP4 pathway. ALOX15 is essential for production of SPMs. SPMs play a crucial role in the resolution of inflammation in ischemic AKI within 24 h by diminishing leukocyte infiltration and other associated inflammatory responses (35, 47). In addition, administration of DHA, the source of SPMs, reportedly improved the prognosis of AKI in mice and dogs within 24 h after the onset of AKI (37–39). Our data clearly demonstrated that MAFB regulates ALOX15 expression in macrophages infiltrated into the kidney 24 h after IRI (Fig. 3). Therefore, MAFB expressed on macrophages would promote the resolution of inflammation in ischemic AKI by inducing ALOX15 expression to produce SPMs (Fig. 6). PGE2 plays a pivotal role in the LM class switch from inflammatory LMs to SPMs; therefore, MAFB in macrophages would play a crucial role in the PGE2-mediated LM class switch.

A key limitation of the current study is that there is no direct evidence that ALOX15 promotes SPM production and improves AKI prognosis in vivo, which is a subject for future work. In addition, our luciferase assay clearly demonstrated that the half-MARE site at −267 bp upstream of the transcriptional initiation site of Alox15 is important for the MAFB-mediated regulation of Alox15 expression. However, further analyses such as chromatin immunoprecipitation assays are required to provide direct evidence for the direct regulation of Alox15 by MAFB.

We also observed that MAFB in macrophages regulates C1q and Msr1 expression in ischemic AKI. C1q induces efferocytosis, and MSR1 accelerates the clearance of DAMPs (11, 13). Therefore, MAFB may also promote AKI alleviation by regulating these genes. Further analyses are required to elucidate the involvement of C1q and MSR1 in the MAFB-mediated alleviation of ischemic AKI.

Recently, the mechanism by which PGE2 induces ALOX15 expression in human amniotic fibroblasts has been reported (48, 49). In human amniotic fibroblasts, PGE2 activates the cAMP/PKA pathway by binding to EP2 receptors, followed by the phosphorylation of CREB and STAT3. Phosphorylated CREB and STAT3 interact with p300 and glucocorticoid receptor (GR) at the ALOX15 gene promoter to induce ALOX15 expression. In ischemic AKI, PGE2 activates the cAMP/PKA/CREB pathway by binding to the EP4 receptor, increasing Mafb expression in macrophages (16). Our results showed that MAFB regulates Alox15 expression under this condition. Therefore, phosphorylated CREB, STAT3, p300, and GR may regulate Alox15 expression in macrophages by interacting with MAFB. In fact, MAFB has been reported to interact with GR and p300 (50, 51). Although there is no direct evidence for a relationship between MAFB and CREB or STAT3, they may interact to regulate Alox15 expression.

Previous studies have reported that MAFB in macrophages is activated in response to body abnormalities, such as autoimmune diseases, ischemic stroke, wound injury, obesity, and cold exposure; therefore, MAFB in macrophages is essential for homeostasis in the body (11, 13–15, 52). MAFB is upregulated by various nuclear receptor agonists, including PPARs, LXRs, RARs, and GRs (10, 11). Such agonists are potential candidates for therapeutic agents targeting MAFB under various pathological conditions. However, in the presence of an LXR/RXR agonist, MAFB downregulated Alox15 expression (Supplemental Fig. 3B); this is because in macrophages infiltrated into AKI-induced kidney, MAFB expression is regulated by the COX-2/PGE2/EP4 pathway, which is quite different from the regulation by nuclear receptors. Conversely, under chronic inflammatory conditions, such as CKD induced by unilateral ureteral obstruction or the 5/6 nephrectomized model, ALOX15 promotes fibrosis and worsens prognosis (43–45). In such CKD and in the chronic phase of AKI, MAFB would suppress ALOX15 expression, and LXR/RXR agonists could be therapeutic agents. Further analyses are required to clarify how MAFB regulation is altered by the surrounding environment.

In conclusion, the current study identified Alox15 as a target gene of MAFB in macrophages under the COX-2/PGE2/EP4 pathway. MAFB would play a crucial role in the PGE2-mediated LM class switch by inducing the expression of ALOX15, which catalyzes SPM production. Our findings provide new insights into the molecular mechanisms of LM regulation under acute inflammatory conditions and pave the way for the development of novel therapies for acute inflammatory diseases.

The authors have no financial conflicts of interest.

We acknowledge the School of Integrative and Global Majors (SIGMA) Ph.D. program in Human Biolog, University of Tsukuba, for student support. We thank Masami Ojima for providing technical support. We also thank Yukiyo Ida for assisting in purchasing the materials required for the experiments. We also acknowledge Yuri Inoue, Omar Samir, and Hibiki Ueno for supporting Mafbf/f::LysM-Cre mice colony management.

This work was supported by Ministry of Education, Culture, Sports, Science and Technology (MEXT) KAKENHI Grants 19K07499 and 23K05586, Japan Science and Technology Agency (JST) Grant JPMJPF2017 (to S.T.), and by Japan Science and Technology Agency (JST) SPRING Grant JPMJSP2124.

The online version of this article contains supplemental material.

The RNA-seq data presented in this article have been submitted to the DNA Databank of Japan (https://ddbj.nig.ac.jp/resource/sra-submission/) under accession number DRA017447.

AIM

apoptotic inhibitor of macrophages

AKI

acute kidney injury

ALOX15

arachidonate 15-lipoxygenase

ATN

acute tubular necrosis

BUN

blood urea nitrogen

CKD

chronic kidney disease

COX-2

cyclooxygenase-2

DAMP

damage-associated molecular pattern

DHA

docosahexaenoic acid

EP4

PGE2 receptor 4

GR

glucocorticoid receptor

half-MARE

half of the MARE

IRI

ischemia-reperfusion injury

LC-MS/MS

liquid chromatography–tandem MS

LM

lipid mediator

MAFB

V-maf musculoaponeurotic fibrosarcoma oncogene homolog B

MARE

Maf recognition element

MS

mass spectrometry

MSR1

macrophage scavenger receptor 1

PAS

periodic acid–Schiff

qRT-PCR

quantitative RT-PCR

RNA-seq

RNA sequencing

SPM

specialized proresolving mediator

WT

wild-type

1
Ronco
,
C.
,
R.
Bellomo
,
J. A.
Kellum
.
2019
.
Acute kidney injury
.
Lancet
394
:
1949
1964
.
2
Goldstein
,
S. L.
,
B. L.
Jaber
,
S.
Faubel
,
L. S.
Chawla
;
Acute Kidney Injury Advisory Group of American Society of Nephrology
.
2013
.
AKI transition of care: a potential opportunity to detect and prevent CKD
.
Clin. J. Am. Soc. Nephrol.
8
:
476
483
.
3
Huen
,
S. C.
,
L. G.
Cantley
.
2017
.
Macrophages in renal injury and repair
.
Annu. Rev. Physiol.
79
:
449
469
.
4
Cao
,
Q.
,
D. C. H.
Harris
,
Y.
Wang
.
2015
.
Macrophages in kidney injury, inflammation, and fibrosis
.
Physiology (Bethesda)
30
:
183
194
.
5
Kinsey
,
G. R.
,
L.
Li
,
M. D.
Okusa
.
2008
.
Inflammation in acute kidney injury
.
Nephron Exp. Nephrol.
109
:
e102
e107
.
6
Kataoka
,
K.
,
M.
Noda
,
M.
Nishizawa
.
1994
.
Maf nuclear oncoprotein recognizes sequences related to an AP-1 site and forms heterodimers with both Fos and Jun
.
Mol. Cell. Biol.
14
:
700
712
.
7
Yoshida
,
T.
,
T.
Ohkumo
,
S.
Ishibashi
,
K.
Yasuda
.
2005
.
The 5′-AT-rich half-site of Maf recognition element: a functional target for bZIP transcription factor Maf
.
Nucleic Acids Res.
33
:
3465
3478
.
8
Moriguchi
,
T.
,
M.
Hamada
,
N.
Morito
,
T.
Terunuma
,
K.
Hasegawa
,
C.
Zhang
,
T.
Yokomizo
,
R.
Esaki
,
E.
Kuroda
,
K.
Yoh
, et al
.
2006
.
MafB is essential for renal development and F4/80 expression in macrophages
.
Mol. Cell. Biol.
26
:
5715
5727
.
9
Katoh
,
M. C.
,
Y.
Jung
,
C. M.
Ugboma
,
M.
Shimbo
,
A.
Kuno
,
W. A.
Basha
,
T.
Kudo
,
H.
Oishi
,
S.
Takahashi
.
2018
.
MafB is critical for glucagon production and secretion in mouse pancreatic α cells in vivo
.
Mol. Cell. Biol.
38
:
e00504-17
.
10
Morito
,
N.
,
K.
Yoh
,
T.
Usui
,
H.
Oishi
,
M.
Ojima
,
A.
Fujita
,
R.
Koshida
,
H. H.
Shawki
,
M.
Hamada
,
M.
Muratani
, et al
.
2018
.
Transcription factor MafB may play an important role in secondary hyperparathyroidism
.
Kidney Int.
93
:
54
68
.
11
Tran
,
M. T. N.
,
M.
Hamada
,
H.
Jeon
,
R.
Shiraishi
,
K.
Asano
,
M.
Hattori
,
M.
Nakamura
,
Y.
Imamura
,
Y.
Tsunakawa
,
R.
Fujii
, et al
.
2017
.
MafB is a critical regulator of complement component C1q
.
Nat. Commun.
8
:
1700
.
12
Hamada
,
M.
,
M.
Nakamura
,
M. T. N.
Tran
,
T.
Moriguchi
,
C.
Hong
,
T.
Ohsumi
,
T. T. H.
Dinh
,
M.
Kusakabe
,
M.
Hattori
,
T.
Katsumata
, et al
.
2014
.
MafB promotes atherosclerosis by inhibiting foam-cell apoptosis
.
Nat. Commun.
5
:
3147
.
13
Shichita
,
T.
,
M.
Ito
,
R.
Morita
,
K.
Komai
,
Y.
Noguchi
,
H.
Ooboshi
,
R.
Koshida
,
S.
Takahashi
,
T.
Kodama
,
A.
Yoshimura
.
2017
.
MAFB prevents excess inflammation after ischemic stroke by accelerating clearance of damage signals through MSR1
.
Nat. Med.
23
:
723
732
.
14
Inoue
,
Y.
,
C.-W.
Liao
,
Y.
Tsunakawa
,
I.-L.
Tsai
,
S.
Takahashi
,
M.
Hamada
.
2022
.
Macrophage-specific, Mafb-deficient mice showed delayed skin wound healing
.
IJMS
23
:
9346
.
15
Yadav
,
M. K.
,
M.
Ishida
,
N.
Gogoleva
,
C.-W.
Liao
,
F. N.
Salim
,
M.
Kanai
,
A.
Kuno
,
T.
Hayashi
,
Z. J.
Shahri
,
K.
Kulathunga
, et al
.
2024
.
MAFB in macrophages regulates cold-induced neuronal density in brown adipose tissue
.
Cell Rep.
43
:
113978
.
16
Pan
,
Y.
,
S.
Cao
,
A. S.
Terker
,
J.
Tang
,
K.
Sasaki
,
Y.
Wang
,
A.
Niu
,
W.
Luo
,
D.
Daassi
,
X.
Fan
, et al
.
2022
.
Myeloid cyclooxygenase-2/prostaglandin E2/E-type prostanoid receptor 4 promotes transcription factor MafB-dependent inflammatory resolution in acute kidney injury
.
Kidney Int.
101
:
79
91
.
17
Tian
,
R.
,
X.
Zuo
,
J.
Jaoude
,
F.
Mao
,
J.
Colby
,
I.
Shureiqi
.
2017
.
ALOX15 as a suppressor of inflammation and cancer: lost in the link
.
Prostaglandins Other Lipid Mediat.
132
:
77
83
.
18
Lim
,
C. S.
,
D. W.
Porter
,
M. S.
Orandle
,
B. J.
Green
,
M. A.
Barnes
,
T. L.
Croston
,
M. G.
Wolfarth
,
L. A.
Battelli
,
M. E.
Andrew
,
D. H.
Beezhold
, et al
.
2020
.
Resolution of pulmonary inflammation induced by carbon nanotubes and fullerenes in mice: role of macrophage polarization
.
Front. Immunol.
11
:
1186
.
19
Snodgrass
,
R. G.
,
Y.
Benatzy
,
T.
Schmid
,
D.
Namgaladze
,
M.
Mainka
,
N. H.
Schebb
,
D.
Lütjohann
,
B.
Brüne
.
2021
.
Efferocytosis potentiates the expression of arachidonate 15-lipoxygenase (ALOX15) in alternatively activated human macrophages through LXR activation
.
Cell Death Differ.
28
:
1301
1316
.
20
Basil
,
M. C.
,
B. D.
Levy
.
2016
.
Specialized pro-resolving mediators: endogenous regulators of infection and inflammation
.
Nat. Rev. Immunol.
16
:
51
67
.
21
Takayama
,
K.
,
G.
García-Cardena
,
G. K.
Sukhova
,
J.
Comander
,
M. A.
Gimbrone
,
P.
Libby
.
2002
.
Prostaglandin E2 suppresses chemokine production in human macrophages through the EP4 receptor
.
J. Biol. Chem.
277
:
44147
44154
.
22
Serhan
,
C. N.
,
N.
Chiang
,
T. E.
Van Dyke
.
2008
.
Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators
.
Nat. Rev. Immunol.
8
:
349
361
.
23
Ivanov
,
I.
,
H.
Kuhn
,
D.
Heydeck
.
2015
.
Structural and functional biology of arachidonic acid 15-lipoxygenase-1 (ALOX15)
.
Gene
573
:
1
32
.
24
Suzuki
,
T.
,
V. P.
Kelly
,
H.
Motohashi
,
O.
Nakajima
,
S.
Takahashi
,
S.
Nishimura
,
M.
Yamamoto
.
2008
.
Deletion of the selenocysteine tRNA gene in macrophages and liver results in compensatory gene induction of cytoprotective enzymes by Nrf2
.
J. Biol. Chem.
283
:
2021
2030
.
25
Furuichi
,
K.
,
J.-L.
Gao
,
R.
Horuk
,
T.
Wada
,
S.
Kaneko
,
P. M.
Murphy
.
2008
.
Chemokine receptor CCR1 regulates inflammatory cell infiltration after renal ischemia-reperfusion injury
.
J. Immunol.
181
:
8670
8676
.
26
Yoshida
,
T.
,
H.
Sugiura
,
M.
Mitobe
,
K.
Tsuchiya
,
S.
Shirota
,
S.
Nishimura
,
S.
Shiohira
,
H.
Ito
,
K.
Nobori
,
S. R.
Gullans
, et al
.
2008
.
ATF3 protects against renal ischemia-reperfusion injury
.
J. Am. Soc. Nephrol.
19
:
217
224
.
27
Miyata
,
J.
,
Y.
Yokokura
,
K.
Moro
,
H.
Arai
,
K.
Fukunaga
,
M.
Arita
.
2021
.
12/15-lipoxygenase regulates IL-33-induced eosinophilic airway inflammation in mice
.
Front. Immunol.
12
:
687192
.
28
Bonventre
,
J. V.
,
L.
Yang
.
2011
.
Cellular pathophysiology of ischemic acute kidney injury
.
J. Clin. Invest.
121
:
4210
4221
.
29
Kanai
,
M.
,
H.
Jeon
,
M.
Ojima
,
T.
Nishino
,
T.
Usui
,
M. K.
Yadav
,
K.
Kulathunga
,
N.
Morito
,
S.
Takahashi
,
M.
Hamada
.
2020
.
Phenotypic analysis of mice carrying human-type MAFB p.Leu239Pro mutation
.
Biochem. Biophys. Res. Commun.
523
:
452
457
.
30
Kelly
,
K. J.
,
W. W.
Williams
,
R. B.
Colvin
,
S. M.
Meehan
,
T. A.
Springer
,
J. C.
Gutierrez-Ramos
,
J. V.
Bonventre
.
1996
.
Intercellular adhesion molecule-1-deficient mice are protected against ischemic renal injury
.
J. Clin. Invest.
97
:
1056
1063
.
31
Jang
,
H. R.
,
H.
Rabb
.
2015
.
Immune cells in experimental acute kidney injury
.
Nat. Rev. Nephrol.
11
:
88
101
.
32
Clements
,
M.
,
M.
Gershenovich
,
C.
Chaber
,
J.
Campos-Rivera
,
P.
Du
,
M.
Zhang
,
S.
Ledbetter
,
A.
Zuk
.
2016
.
Differential Ly6C expression after renal ischemia-reperfusion identifies unique macrophage populations
.
J. Am. Soc. Nephrol.
27
:
159
170
.
33
Nechemia-Arbely
,
Y.
,
D.
Barkan
,
G.
Pizov
,
A.
Shriki
,
S.
Rose-John
,
E.
Galun
,
J. H.
Axelrod
.
2008
.
IL-6/IL-6R axis plays a critical role in acute kidney injury
.
J. Am. Soc. Nephrol.
19
:
1106
1115
.
34
Shimazui
,
T.
,
T.
Nakada
,
Y.
Tateishi
,
T.
Oshima
,
T.
Aizimu
,
S.
Oda
.
2019
.
Association between serum levels of interleukin-6 on ICU admission and subsequent outcomes in critically ill patients with acute kidney injury
.
BMC Nephrol.
20
:
74
.
35
Duffield
,
J. S.
,
S.
Hong
,
V. S.
Vaidya
,
Y.
Lu
,
G.
Fredman
,
C. N.
Serhan
,
J. V.
Bonventre
.
2006
.
Resolvin D series and protectin D1 mitigate acute kidney injury
.
J. Immunol.
177
:
5902
5911
.
36
Hong
,
S.
,
Y.
Lu
.
2013
.
Omega-3 fatty acid-derived resolvins and protectins in inflammation resolution and leukocyte functions: targeting novel lipid mediator pathways in mitigation of acute kidney injury
.
Front. Immunol.
4
:
13
.
37
Hassan
,
I. R.
,
K.
Gronert
.
2009
.
Acute changes in dietary ω-3 and ω-6 Polyunsaturated fatty acids have a pronounced impact on survival following ischemic renal injury and formation of renoprotective docosahexaenoic acid-derived protectin D1
.
J. Immunol.
182
:
3223
3232
.
38
Neumayer
,
H. H.
,
M.
Heinrich
,
M.
Schmissas
,
H.
Haller
,
K.
Wagner
,
F. C.
Luft
.
1992
.
Amelioration of ischemic acute renal failure by dietary fish oil administration in conscious dogs
.
J. Am. Soc. Nephrol.
3
:
1312
1320
.
39
Kielar
,
M. L.
,
D. R.
Jeyarajah
,
X. J.
Zhou
,
C. Y.
Lu
.
2003
.
Docosahexaenoic acid ameliorates murine ischemic acute renal failure and prevents increases in mRNA abundance for both TNF-α and inducible nitric oxide synthase
.
J. Am. Soc. Nephrol.
14
:
389
396
.
40
Talamonti
,
E.
,
A. M.
Pauter
,
A.
Asadi
,
A. W.
Fischer
,
V.
Chiurchiù
,
A.
Jacobsson
.
2017
.
Impairment of systemic DHA synthesis affects macrophage plasticity and polarization: implications for DHA supplementation during inflammation
.
Cell. Mol. Life Sci.
74
:
2815
2826
.
41
Zubkova
,
E. S.
,
K. V.
Dergilev
,
I. B.
Beloglazova
,
Y. D.
Molokotina
,
M. A.
Boldyreva
,
Z. I.
Tsokolaeva
,
I. S.
Stafeev
,
M. Y.
Menshikov
,
Y. V.
Parfyonova
.
2021
.
Features of the population of mouse peritoneal macrophages isolated after stimulation with concanavalin A and thioglycolate
.
Bull. Exp. Biol. Med.
171
:
532
540
.
42
Braga
,
T. T.
,
J. S. H.
Agudelo
,
N. O. S.
Camara
.
2015
.
Macrophages during the fibrotic process: M2 as friend and foe
.
Front. Immunol.
6
:
602
.
43
Shinoda
,
Y.
,
H.
Tatsukawa
,
A.
Yonaga
,
R.
Wakita
,
T.
Takeuchi
,
T.
Tsuji
,
M.
Tanaka
,
T.
Suganami
,
K.
Hitomi
.
2023
.
Tissue transglutaminase exacerbates renal fibrosis via alternative activation of monocyte-derived macrophages
.
Cell Death Dis.
14
:
136
.
44
Montford
,
J. R.
,
C.
Bauer
,
J.
Rahkola
,
J. A.
Reisz
,
D.
Floyd
,
K.
Hopp
,
D. E.
Soranno
,
J.
Klawitter
,
M. C. M.
Weiser-Evans
,
R.
Nemenoff
, et al
.
2022
.
15-Lipoxygenase worsens renal fibrosis, inflammation, and metabolism in a murine model of ureteral obstruction
.
Am. J. Physiol. Renal Physiol.
322
:
F105
F119
.
45
Takahashi
,
N.
,
H.
Kikuchi
,
A.
Usui
,
T.
Furusho
,
T.
Fujimaru
,
T.
Fujiki
,
T.
Yanagi
,
Y.
Matsuura
,
K.
Asano
,
K.
Yamamoto
, et al
.
2021
.
Deletion of Alox15 improves kidney dysfunction and inhibits fibrosis by increased PGD2 in the kidney
.
Clin. Exp. Nephrol.
25
:
445
455
.
46
Eliopoulos
,
A. G.
,
C. D.
Dumitru
,
C.-C.
Wang
,
J.
Cho
,
P. N.
Tsichlis
.
2002
.
Induction of COX-2 by LPS in macrophages is regulated by Tpl2-dependent CREB activation signals
.
EMBO J.
21
:
4831
4840
.
47
Luan
,
H.
,
C.
Wang
,
J.
Sun
,
L.
Zhao
,
L.
Li
,
B.
Zhou
,
S.
Shao
,
X.
Shen
,
Y.
Xu
.
2020
.
Resolvin D1 protects against ischemia/reperfusion-induced acute kidney injury by increasing Treg percentages via the ALX/FPR2 pathway
.
Front. Physiol.
11
:
285
.
48
Zhang
,
F.
,
K.
Sun
,
W.-S.
Wang
.
2022
.
Identification of a feed-forward loop between 15(S)-HETE and PGE2 in human amnion at parturition
.
J. Lipid. Res.
63
:
100294
.
49
Zhang
,
F.
,
J.-W.
Lu
,
W.-J.
Lei
,
M.-D.
Li
,
F.
Pan
,
Y.-K.
Lin
,
W.-S.
Wang
,
K.
Sun
.
2023
.
Paradoxical induction of ALOX15/15B by cortisol in human amnion fibroblasts: implications for inflammatory responses of the fetal membranes at parturition
.
IJMS
24
:
10881
.
50
Morante-Palacios
,
O.
,
L.
Ciudad
,
R.
Micheroli
,
C.
de la Calle-Fabregat
,
T.
Li
,
G.
Barbisan
,
M.
Houtman
,
S. G.
Edalat
,
M.
Frank-Bertoncelj
,
C.
Ospelt
,
E.
Ballestar
.
2022
.
Coordinated glucocorticoid receptor and MAFB action induces tolerogenesis and epigenome remodeling in dendritic cells
.
Nucleic Acids Res.
50
:
108
126
.
51
Pajcini
,
K. V.
,
L.
Xu
,
L.
Shao
,
J.
Petrovic
,
K.
Palasiewicz
,
Y.
Ohtani
,
W.
Bailis
,
C.
Lee
,
G. B.
Wertheim
,
R.
Mani
, et al
.
2017
.
MAFB enhances oncogenic Notch signaling in T cell acute lymphoblastic leukemia
.
Sci. Signal.
10
:
eaam6846
.
52
Tran
,
M. T. N.
,
M.
Hamada
,
M.
Nakamura
,
H.
Jeon
,
R.
Kamei
,
Y.
Tsunakawa
,
K.
Kulathunga
,
Y.
Lin
,
K.
Fujisawa
,
T.
Kudo
,
S.
Takahashi
.
2016
.
MafB deficiency accelerates the development of obesity in mice
.
FEBS Open Bio.
6
:
540
547
.
This article is distributed under The American Association of Immunologists, Inc., Reuse Terms and Conditions for Author Choice articles.

Supplementary data