Abstract Background The polarization status and function of tumor-associated macrophages (TAMs) influence tumor progression and patients’ prognosis. CCAAT/enhancer-binding proteins (CEBPs) family are important transcriptional factors in macrophages differentiation physically and pathologically. This study aims to explore the mechanism of CEBPs in TAMs polarization in clear cell renal cell carcinoma (ccRCC) immune microenvironment and its impact on immune checkpoint blockers (ICBs) therapy. Methods The expression of CEBPs in ccRCC was analyzed by single-cell transcriptome and western blot. Immunofluorescence and in-vitro polarization assay were used to evaluate the effect of CEBP delta (CEBPD) on TAMs. Chromatin immunoprecipitation sequencing was used to explore targets of CEBPD. Dual-luciferase reporter assay and electrophoretic mobility shift assay were performed to confirm the regulation of CEBPD to RGS2. Specimens of patients received ICB therapy were used to analyze the relationship between CEBPD and immunotherapy. Results The study identified CEBPD as a key transcriptional factor in ccRCC TAM polarization. Upregulation of CEBPD correlates to decreased M1/M2 ratio of TAMs and poorer clinical outcomes. CEBPD inhibited M1-like polarization in vitro and in vivo via the RGS2/PAR2 axis. Furthermore, CEBPD also affected the therapeutic efficacy of ICB. Conclusion This study revealed CEBPD regulated TAM polarization via the CEBPD/RGS2/PAR2 axis. Targeting CEBPD may be a potential approach and a complementary strategy to ICB therapies in ccRCC. Keywords: Kidney Cancer, Macrophage, Immunotherapy __________________________________________________________________ WHAT IS ALREADY KNOWN ON THIS TOPIC * Tumor-associated macrophages (TAMs) are highly plastic and their polarization status affects clinical outcomes of patients with clear cell renal cell carcinoma (ccRCC). Previous studies have shown that the CCAAT/enhancer-binding protein (CEBP) family is involved in the differentiation of macrophages but the specific roles of family members in TAM polarization are not thoroughly revealed. WHAT THIS STUDY ADDS * Our study showed for the first time that a CEBP family member, CEBP delta (CEBPD) regulated TAM polarization via the CEBPD/RGS2/PAR2 axis and promoted tumor progression. Furthermore, CEBPD was related to the efficacy of immune checkpoint blockers (ICBs) in ccRCC. HOW THIS STUDY MIGHT AFFECT RESEARCH, PRACTICE OR POLICY * Our findings revealed the role of CEBPD in regulating TAM polarization and targeting CEBPD may enhance the ICBs therapeutic efficacy in patients with ccRCC. Introduction Clear cell renal cell carcinoma (ccRCC), is the most common histologic subtype of kidney malignancies.[55]^1 Nearly 25% of patients with ccRCC are diagnosed at the metastatic stage, and a third of the patients who receive resection of localized tumors will have a recurrence.[56]^2 Tyrosine kinase inhibitors (TKIs) are applied to treat patients with advanced or metastatic renal cell carcinoma, but most patients develop resistance to TKI therapy.[57]^3 In recent years, treatment with immune checkpoint blockers (ICBs) in ccRCC patients have made remarkable success, which reveals enormous potential for immune therapy. However, low response rates and drug resistance still remain intractable problems. Thus, there is an urgent need to investigate the tumor immune microenvironment and explore new treatment targets. Due to the dysfunction of von Hippel-Lindau protein, ccRCC is characterized by a highly hypoxic tumor microenvironment (TME). Hypoxia-related signal pathways are hyperactivated, such as HIF, which lead to excessive infiltration of tumor-associated macrophages (TAMs) in the TME.[58]^4 5 TAMs are preferentially recruited in the hypoxic region and gradually polarize into immune-suppressive M2-like phenotype,[59]^6 7 whereas the proportion of proinflammatory M1-like TAMs decreases. This phenotype shift represented a risk factor for poor prognosis, shorter survival and worse therapeutic efficacy.[60]8,[61]10 Enhancing the transformation of TAMs into antitumor M1 phenotype could provide a therapeutic strategy.[62]^11 However, the molecular mechanism of influencing the TAMs polarization in the hypoxic TME remains to be further specified. CCAAT/enhancer-binding proteins (CEBPs), are a group of transcription factors significant in the development and differentiation of macrophages.[63]^12 13 Previous studies reported that CEBPs can be regulated by hypoxic stimulation.[64]14,[65]16 However, whether CEBPs play a significant role in TAM polarization in the hypoxic TME, still remains to be clarified. The CEBP family is comprised of six members: CEBPα, CEBPβ, CEBPγ, CEBPδ, CEBPε, CEBPζ. Family members have a high degree of homology structurally and share many common targets,[66]^17 which makes it complicated to explore their individual function. Therefore, it’s necessary to analyze the expression pattern of CEBPs in ccRCC transcriptional landscape by single-cell transcriptome and combined with large-scale specimen verification in order to clarify the role of CEBPs played in the TAMs polarization in hypoxia TME of ccRCC. In this study, we conducted a comprehensive analysis of the CEBP family in ccRCC TAMs and identified CEBP delta (CEBPD) as a pivotal transcriptional factor influencing TAM polarization. Combining patient specimens and in-vitro polarization assay, we confirmed that CEBPD reduces the M1/M2 ratio and leads to poorer survival outcomes. Furthermore, through DNA-protein interactive assay, we reported RGS2 was the target of CEPBD and elucidated the involvement of the RGS2/PAR2 axis in TAM polarization. Finally, we explored the impact of CEBPD on patients received ICB therapy. Materials and methods Processing of single cell RNA-seq data Process single cell RNA-seq (scRNA-seq) data using R (V.4.3.0). Use the Read10X function in Seurat (R package, V.4.3.0.1) to separately import unique molecular identifier (UMI) count matrices generated from 20 samples into R. To remove the low-expressed genes and low-quality cells, cells that had fewer than 200 genes, over 6,000, UMI over 10,000, or more than 25% mitochondrial reads were depleted, and the genes expressed in at least 3 cells were kept from the original data. Follow the DoubletFinder (R package, V.2.0.4) to computationally infer and remove doublets in each sample individually, with default parameters. All of this information was merged into one Seurat object using the merge function. UMI counts were normalized using the “NormalizeData” function. The 2000 highly variable genes were identified by the “FindVariableFeatures” function. Principal component analysis (PCA) was performed on the single-cell expression matrix using the ‘‘RunPCA’’ function and the top 30 principal components were used for clustering with the Louvain graph-clustering method. Then, we used the “Runharmony” to perform the batch-effect correction for each dataset. Subsequently, Uniform Manifold Approximation and Projection (UMAP) was established based on harmony dimensions for cell clustering analysis. As to subclusters, the same methods were used for recognizing the variable genes, reducing dimensions and clustering. Cell type annotation and cluster marker identification “FindAllMarkers” function was used to find markers for each of the identified clusters and annotated on the basis of the expression of canonical markers of particular cell types based on the published single-cell transcriptome data of kidney cancer. The gene set variation analysis (GSVA, R package, V.1.48.2) was applied to estimate pathway activity scores for single cells and visualize it through the heatmap using gene sets of C2 and C5 collection obtained from the molecular signature database. The differential activities of pathways were calculated using the limma R package. Gene set variation analysis Regulon activity analysis pySCENIC (V.0.12.1) was used to build the gene regulatory network in all cells combined with cis-Target human motif database (V.10). Raw expression data were extracted from the Seurat data. Enriched motifs were identified with default parameters of pySCENIC. AUCell function assigns specific values to each cell to represent their regulon activity based on Jensen-Shannon divergence. Cell culture and transfection THP-1 cells were obtained from the cell bank of the typical culture preservation committee of the Chinese Academy of Sciences. THP-1 cells were cultured in RPMI 1640 (Gibco) supplemented with 10% fetal bovine serum (Gibco). Human peripheral mononuclear cells were obtained from donors and were used for isolating CD14+ monocytes. 50 ng/mL Phorbol 12-myristate 13-acetate (PMA) (Selleck) were added for 48 hours to induce M0 macrophages. 100 ng/mL lipopolysaccharides (LPS) (Selleck) and interferon-gamma (IFN-γ) (20 ng/mL) (PeproTech) were added to induce M1-like macrophages and interleukin (IL)-4 (20 ng/mL) (PeproTech) was added to induce M2-like macrophages. THP-1 cells were transduced with short hairpin RNA lentivirus targeting CEBPD (CEBPD-kd1 and CEBPD-kd2) and RGS2 (RGS2-kd1 and RGS2-kd2), overexpressing lentiviral vector (RGS2-oe) or control lentivirus. Lentiviruses were purchased from OBIOSH. Cells infected with virus were selected by 2 µg/mL puromycin (Selleck). PAR1 inhibitor (HY-14993), PAR2 inhibitor (HY-P1260) and PAR2 agonist (HY-P4803) were purchased from MCE. Patient specimens Written informed consent was also obtained from all the patients. 196 patient specimens were derived from patients underwent radical or partial nephrectomy at Xinhua Hospital between 2016 and 2018. All tumor specimens were pathologically confirmed ccRCC. After resection, the samples were immediately frozen in liquid nitrogen and stored at −80℃ for further studies. Specimen collection for research purposes was approved by the Clinical Research Ethics Board of Xinhua Hospital. Western blotting assay Total protein was extracted from the cultured cells or specimens using lysis buffer according to manufacturer’s protocol, and resolved by sodium dodecyl sulfate (SDS) polyacrylamide gels, and then transferred to polyvinylidene fluoride (PVDF) membranes. The antibodies were provided in the [67]online supplemental materials. The immunoreactive bands were analyzed by chemiluminescence. Flow cytometry Cells (1×10^6) were incubated with Fc receptors blocker, then GhostDye staining of dead cells was incubated at 4°C in the dark for 30 min and centrifuged to remove the supernatant. Then, the cells were resuspended in fluorescence-activated cell sorting (FACS), after which extracellular staining was performed for the following proteins: CD86 and CD206. The cells were incubated in cell staining buffer at 4°C in the dark for 30 min. The cells were analyzed using a Beckman Analytical Flow Cytometer and analyzed using CytExpert software (Beckman Coulter). The following antibodies were used: anti-CD86, PE (Miltenyi Biotec, 130-116-160); anti-CD206, PE (Invitrogen, MA5-23594). Tissues and cells immunofluorescence assay Tissue sections were deparaffinized, rehydrated with graded ethanol, incubated with 0.3% hydrogen peroxide for 20 min, and blocked with 10% goat serum. THP-1 cells were fixed on slides with 4% paraformaldehyde, permeabilized with a buffer containing 0.1% Triton X-100 and 0.25% bovine serum albumin (BSA), and blocked with goat serum and BSA. The slides were incubated with primary antibody overnight at 4℃. Next, slides were labeled with secondary antibody for 1 hour at room temperature. After washing three times with phosphate-buffered saline, the slides were subsequently stained with 4',6-diamidino-2-phenylindole (DAPI) for visualization of the nucleus. Images were scanned and analyzed using a FLUOVIEW vs200 microscope (Olympus). In multiplex immunohistochemistry (mIHC), we used CD86 to label M1-like macrophages and CD163 to label M2-like macrophages. Tumors with a CEBPD positivity rate exceeding 50% in TAMs were classified into the CEBPD-high group, while those below 50% were low group. The antibodies were provided in the [68]online supplemental materials. RNA extraction and real-time quantitative PCR Total RNAs were isolated using TRIzol reagent (Invitrogen), and 2 µg of total RNA was subjected to reverse transcription using Superscript III transcriptase (Invitrogen). Real-time quantitative PCR (RT-qPCR) was conducted using a Bio-Rad CFX96 system with SYBR green to determine the messenger RNA expression level of a gene of interest. Expression levels were normalized to the ACTB level using the 2−∆∆Ct methods. The sequences of the primers were provided in [69]online supplemental materials. Chromatin immunoprecipitation sequencing (ChIP-seq) Cells were cross-linked with 1% formaldehyde solution, then lysed and sonicated. The sonicated chromatin was precleared with chromatin immunoprecipitation (ChIP) Grade protein A/G for 1 hour. Then the lysates were incubated at 4℃ overnight with the target-specific antibody CEBPD (Invitrogen, PA5-30262) or negative control anti-IgG antibody. DNA was eluted from immunoprecipitated complexes, reverse cross-linked, and purified. High-quality ChIP DNA was used for sequencing. The ChIP-seq results were visualized with Integrative Genomics Viewer. The number of CEBPD binding sites in the promoter region of interested genes was analyzed to find target genes that can be transcriptionally regulated by CEBPD. Dual-luciferase reporter assay The full-length segments and truncated segments of the RGS2 promoter were amplified by PCR from genome DNA of ccRCC tissues and were purified by Quick Gel Extraction Kit (TransDetect, China). The purified DNA segments were inserted upstream of the firefly luciferase complementary DNA (cDNA) in the pGL3-basic vector to construct pGL3-RGS2-Full-length, pGL3-RGS2-Truncated#1 and pGL3-RGS2-Truncated#2. The coding sequence of CEBPD was amplified by PCR from cDNA of ccRCC tissues and was also purified and inserted downstream of the promoter in the pcDNA3.1 vector to construct pcDNA3.1-CEBPD. Cells were transfected using Lipo3.0 Efficient Transfection Reagent (SuperKine, China). Dual-luciferase reporter assay was conducted using the Double-Luciferase Reporter Assay Kit (TransDetect, China) as per manufacturer’s protocol. All experiments were repeated at least three times. The primers used to construct pcDNA3.1-CEBPD and pGL3-RGS2-promoter were provided in [70]online supplemental materials. Electrophoretic mobility shift assay (EMSA) The full-length segments of RGS2 promoter were obtained as described above. Recombinant Human CEBPD protein was purchased from Cusabio (catalog#EP005182HU). The RGS2-promoter DNA and CEBPD proteins were mixed at indicated concentrations in binding buffer and incubated at room temperature for 30 min. A 1% agarose gel containing 0.5×Tris-Borate-EDTA (TBE) buffer was prepared. The incubated mixture was then subjected to the gel and run in 0.5×TBE buffer at 10 V/cm for 1 hour on ice. After electrophoresis, the gel was steeped in 0.5×TBE buffer containing 4×Ultra Gelred (Vazyme, China) to stain nucleic acid for ∼40 min and then scanned using an ultraviolet emitter. After that, the gel was steeped in FastBlue (Biosharp, China) to stain protein for 2 hour and then photographed. Equal doses of BSA were used as a negative control. Statistical analysis All data were analyzed by GraphPad Prism V.9.0 and presented as the mean±SD. Statistical tests for data analysis included Student’s t-test, Spearman correlation analysis. P values were determined by a two-tailed Student’s t-test for comparing two groups. P values were indicated with *p<0.05, **p<0.01, and ***p<0.001 on graphs. Graphs not labeled with an asterisk indicate that the differences between the test groups and the control groups were not statistically significant. All in vitro experiments were repeated with at least three replicates. Results CEBPD upregulates in ccRCC and correlates to suppressed immune response To provide a landscape of ccRCC TME, we performed scRNA-seq on 13 ccRCC samples and seven adjacent kidney samples. Following UMAP dimensionality reduction, these cells clustered into seven groups annotated with specific marker genes ([71]online supplemental figure S1A–C). Myeloid cells were further categorized into monocytes, macrophages (including TAM1, TAM2 and TAM3), and dendritic cells ([72]figure 1A,B, [73]online supplemental figure S2A,B and S3). Our clustering strategy of TAMs was adapted from the study of Mei et al.[74]^18 The TAM1 cluster was similar to tissue-resident macrophages. The TAM2 cluster was defined as an immune-suppressive subtype, transcriptionally expressing a high level of CD163. The TAM3 cluster was described as a proinflammatory subtype, with an increasing expression of M1-like markers like IL-1β. We noticed a significant decrease in TAM3 in ccRCC tissues compared with adjacent kidney tissues, indicating a decline in inflammation and antitumor immune response ([75]figure 1C). We hypothesized that CEBP-mediated macrophage differentiation contributes to this phenomenon. Analysis of CEBP family expression in myeloid cells from ccRCC and adjacent tissues revealed that the expression of CEBPB and CEBPD was the highest among all members ([76]figure 1D). Notably, CEBPD in TAM3 differed significantly between tumors and adjacent tissues. Further analysis of CEBP family transcriptional activity showed a marked increase in CEBPD activity in TAM3 in ccRCC compared with adjacent kidney tissues ([77]figure 1F), while CEBPB and other members showed no significant differences ([78]figure 1E and [79]online supplemental figure S2C,D). Moreover, CEBPD-high ccRCCs had a significantly lower proportion of TAM3 compared with CEBPD-low ccRCCs ([80]figure 1G). Pathway enrichment analysis showed CEBPD upregulation in ccRCCs was positively associated with negative regulation of IL-6 pathway, negative regulation of monocyte chemotaxis and macrophage fusion ([81]figure 1H), while it was negatively associated with positive regulation of T cell differentiation ([82]figure 1H). Similarly, CEBPD upregulation correlated to low expression of IL-1α, IL-1β, IL-6, suggesting impaired antitumor function ([83]figure 1I). These results indicated that the activation of CEBPD in ccRCC leads to a decrease and dysfunction of proinflammatory TAMs and suppression of immune response. To identify downstream of CEBPD, we analyzed differentially expressed genes ([84]figure 1J) and performed function enrichment ([85]online supplemental figure S4) in TAM3 between CEBPD-high and CEBPD-low groups, and RGS2 showed upregulation. Figure 1. CEBPD is upregulated in ccRCCs and correlated with impaired immune response. (A) The cell type clusters of myeloid cells. (B) The group clusters of myeloid cells. (C) The proportion of myeloid cells in ccRCCs and adjacent kidney tissues. (D) The expression of CEBPD family in cell type clusters, and the yellow dot represents high level of average expression, while the purple dot represents low level of average expression. Besides, the size of the dot represents the percentage of cells expressing the gene. Transcription factor activity of (E) CEBPB and (F) CEBPD in myeloid cells. (G) Proportion of 5 groups of myeloid cells in CEBPD-high and CEBPD-low ccRCCs. (H) Pathway enrichment analysis of 13 ccRCC samples. (I) Heatmap of cytokines expression of 13 ccRCC samples. (J) Volcano map of differentially expressed genes between CEBPD-high and low TAM3. Data were presented as mean±SD. A significant difference between the groups, **p<0.01, ***p<0.001, and ****p<0.0001. ccRCC, clear cell renal cell carcinoma; CEBP, CCAAT/enhancer-binding protein; CEBPD, CEBP delta; DC, dendritic cell; GSVA: gene set variation analysis; IL, interleukin; ns, not significant; RCC, renal cell carcinoma; TAM, tumor-associated macrophage. [86]Figure 1 [87]Open in a new tab CEBPD upregulation in ccRCC tissues correlates to low M1/M2 proportion and poorer clinical prognosis We analyzed the protein levels of CEBP family members in six matched patient samples. Compared with adjacent kidney tissues, ccRCC tissues showed elevated levels of CEBPD ([88]figure 2A), while other CEBP family members did not show significant differences. Elevated CEBPD expression in ccRCC was related to inferior overall survival probability in The Cancer Genome Atlas (TCGA) cohort ([89]figure 2B). To further investigate changes in the immune microenvironment in ccRCC tissues, we performed a multiplex immunofluorescence assay.[90]^19 We used CD86 to define M1-like antitumoral TAMs and CD163 to define M2-like protumorous TAMs. Results showed a higher infiltration of macrophages in ccRCC compared with adjacent kidney tissue ([91]figure 2D, [92]online supplemental figure S5). Besides, tumors with higher expression of TAMs CEBPD showed increased CD163, which was considered as M2-like TAMs, while M1 marker CD86 was reduced, compared with tumor with lower expression of TAMs CEBPD ([93]figure 2D,E). Furthermore, higher TAM-CEBPD patients showed worse overall survival probability than the low group ([94]figure 2C, [95]online supplemental table S1). These results further indicated that CEBPD upregulation would disturb the proportion of macrophage subtypes, which may lead to poor clinical outcomes. Figure 2. CEBPD upregulation influenced TAM proportion and was associated with poorer prognosis. (A) Western blot results showed the protein expression of CEBPA, CEBPB, CEBPD, CEBPG, CEBPZ in six patient specimens. (B) Kaplan-Meier curves of TCGA-KIRC cohort. (C) Kaplan-Meier curves of survival probability in Xinhua Hospital cohort. Patients were divided by IHC score. (D) Representative immunofluorescence images of adjacent kidney tissue (left), CEBPD-low tumor (middle) and CEBPD-high tumor (right), and (E) quantitative analysis of M1-like and M2-like TAMs.Data were presented as mean±SD. A significant difference between the groups, *p<0.05. ccRCC, clear cell renal cell carcinoma; CEBP, CCAAT/enhancer-binding protein; CEBPD, CEBP delta; IHC, immunohistochemistry; KIRC, kidney renal clear cell carcinoma; TAM, tumor-associated macrophage; TCGA, The Cancer Genome Atlas. [96]Figure 2 [97]Open in a new tab CEBPD attenuates the M1 polarization of macrophages in vitro To explore the role of CEBPD in macrophage polarization, we conducted experiments using human acute monocytic leukemia THP-1 cells, which were induced by PMA into the M0 stage. M0 THP-1 exhibited low levels of both M1 and M2 markers, and the knockdown or overexpression of CEBPD did not affect the basal expression of these markers ([98]online supplemental figure S6). We then induced M1 polarization using LPS and IFN-γ, and M2 polarization using IL-4. Notably, results of flow cytometry showed CEBPD knockdown resulted in an increasing proportion of cells expressing CD86 ([99]figure 3A). In addition, western blot and RT-qPCR showed CEBPD knockdown increased the expression of IL-1β and IL-6, indicating enhanced M1 polarization ([100]figure 3B,C). However, M2 polarization was not affected by CEBPD knockdown ([101]figure 3D–F). Similar results were shown in monocytes that CEBPD knocked down could enhance M1 polarization while not affecting M2 polarization ([102]online supplemental figure S7). Immunofluorescence showed that CEBPD knockdown enhanced the translocation of p65 from cytoplasm to the nucleus ([103]figure 3G). These findings supported that CEBPD impeded M1 polarization via the NF-κB pathway. Overexpression of CEBPD was found to reduce the production of proinflammation factors, potentially impairing the antitumor effect of macrophages within the TME. Figure 3. CEBPD knockdown enhanced M1 type polarization of THP-1 cells. (A) CD86 positive cells in CEBPD-kd1, CEBPD-kd2 and control group. (B–C) CEBPD knockdown enhanced the expression of IL-1β and IL-6. (D) CD206 positive cells in CEBPD-kd1, CEBPD-kd2 and control group. (E–F) CEBPD knockdown did not affect the expression of M2 markers. (G) CEBPD knockdown promoted the translocation of p65 from cytoplasm to nucleus. A significant difference between the groups, ***p<0.001. CEBPD, CCAAT/enhancer-binding protein delta; DAPI: 4',6-diamidino-2-phenylindole; IFN-γ, interferon-gamma; IL, interleukin; LPS, lipopolysaccharides; ns, not significant; UN, uninduced, [104]Figure 3 [105]Open in a new tab CEBPD binds to RGS2 promoter region and upregulates RGS2 expression To elucidate the underlying mechanism of CEBPD in TAM polarization, ChIP sequencing (ChIP-seq) was performed in THP-1 cells to identify downstream genes regulated by CEBPD. The analysis revealed that nearly 1/4 binding regions of CEBPD were located in promoter regions ([106]figure 4A), with the binding peaks almost concentrated in transcription start sites ([107]figure 4B). Differential fold change and peak score analysis of downstream genes regulated by CEBPD highlighted RGS2 as having the highest fold change and peak score ([108]figure 4C,D). Functional enrichment analysis of these genes indicated involvement in the production of molecular mediators of immune response ([109]figure 4E). Visualization with IGV software confirmed that the CEBPD binding peak was positioned upstream of the transcription start site of RGS2 ([110]figure 4F). Additionally, a positive correlation between RGS2 and CEBPD was validated in TAM3 using scRNA-seq ([111]figure 4G), and a similar result was observed in the TCGA-KIRC (kidney renal clear cell carcinoma) database ([112]figure 4H). Figure 4. Chromatin immunoprecipitation sequencing analysis of CEBPD. (A) Pie chart of binding region of CEBPD. (B) Genomic distribution of CEBPD binding peaks. (C) Fold changes and (D) peak score of genes regulated by CEBPD. (E) Function enrichment of genes regulated by CEBPD. (F) Visualization of binding peak of CEBPD on promoter region of RGS2. Correlation analysis between CEBPD and RGS2 in (G) TAM3 and (H) TCGA-KIRC database. ccRCC, clear cell renal cell carcinoma; CEBPD, CCAAT/enhancer-binding protein delta; ChIP, chromatin immunoprecipitation; FC, fold change; KIRC, kidney renal clear cell carcinoma. TAM, tumor-associated macrophage; TCGA, The Cancer Genome Atlas. [113]Figure 4 [114]Open in a new tab To confirm the direct transcriptional regulation of RGS2 by CEBPD, we performed dual-luciferase reporter assay first. We designed a 2000 bp promoter sequence containing CEBPD binding peak, and identified three potential binding sites by JASPR database ([115]figure 5A, top). Through sequential truncation of these binding sites, we determined that the first site was responsible for binding and transcriptional activity ([116]figure 5A, bottom). Next, the western blot assay showed RGS2 increased as CEBPD was overexpressed, while RGS2 decreased as CEBPD was knocked down ([117]figure 5B). To further confirm the direct binding between CEBPD protein and RGS2 promoter, we performed an electrophoretic mobility shift assay (EMSA). As shown in [118]figure 5C, a shifted band of the RGS2 promoter appeared as the concentration of CEBPD protein increased, indicating the DNA-protein complex formed by direct binding between the RGS2 promoter and CEBPD protein ([119]figure 5D, top). In contrast, no shift bind was observed when BSA was used instead of CEBPD protein ([120]figure 5D, bottom). These results established that CEBPD directly binds to the promoter region of RGS2, thereby promoting its transcription and expression. Figure 5. CEBPD upregulated RGS2 by binding to its promoter region. (A) Dual-luciferase reporter assay results showed CEBPD bound to the first site on RGS2 promoter region. Western blot assay showed RGS2 increased as CEBPD was overexpressed while RGS2 decreased as CEBPD was knocked down in (B) THP-1 cells and (C) human monocytes. (D) EMSA results confirmed the combination between CEBPD protein and RGS2 promoter region. Data were presented as mean±SD. A significant difference between the groups, **p<0.01, ***p<0.001, and ****p<0.0001. BS, binding site. BS, binding site; BSA, bovine serum albumin; CEBPD, CCAAT/enhancer-binding protein delta; EMSA, electrophoretic mobility shift assay. [121]Figure 5 [122]Open in a new tab RGS2 inhibits M1 polarization through deactivating the PAR2 pathway We further explored whether RGS2 was the target of CEBPD involved in regulating macrophage polarization. Western blot analysis showed that THP-1 cells and monocytes with CEBPD knockdown expressed more IL-1β and IL-6; however, the overexpression of RGS2 reversed this trend ([123]figure 6A, [124]online supplemental figure S8A). Additionally, CEBPD knockdown promoted the translocation of p65 to the nucleus, but this effect was diminished by RGS2 overexpression ([125]figure 6B). Figure 6. RGS2 inhibited M1 polarization through deactivating PAR2. (A) CEBPD knockdown increased IL-1β and IL-6, whereas RGS2 overexpression reversed this effect. (B) CEBPD knockdown promoted the translocation of p65 from cytoplasm to nucleus, whereas RGS2 overexpression reversed this effect. (C) PAR2 was upregulated during M1 polarization. (D) RGS2 knockdown activated PAR2-mediated MAPK signaling pathway. (E) PAR2 inhibitors reduced IL-1β and IL-6. (F) PAR2 inhibitors impeded the nuclear translocation of p65. CEBPD, CCAAT/enhancer-binding protein delta; DAPI: 4',6-diamidino-2-phenylindole; IFN-γ, interferon-gamma; IL, interleukin; LPS, lipopolysaccharides; PAR, proteinase-activated receptor; UN, uninduced. [126]Figure 6 [127]Open in a new tab RGS2, a member of the regulator of G protein signaling family, functions as GTPase activating proteins. RGS family inhibits GPCR-mediated signaling by accelerating the GTPase activity of G protein alpha subunits, thus driving them into their inactive GDP-bound form.[128]^20 21 Proteinase-activated receptors (PARs) are a subfamily of G protein-coupled receptors with four members, PAR1, PAR2, PAR3, PAR4. PARs play an important role in inflammation[129]22,[130]24 and are reported to interact with RGS family members.[131]25,[132]27 We observed an increase in PAR2 expression during the M1 polarization of macrophages, indicating the involvement of the PAR2-associated pathway in this process ([133]figure 6C). Further investigation showed PAR2 agonists increased phosphorylated ERK and NLRP3 levels, whereas PAR2 inhibitors had the opposite effect ([134]figure 6D, [135]online supplemental figure S8B). RGS2 knockdown also elevated phosphorylated ERK and NLRP3, which could be reversed by PAR2 inhibitors ([136]figure 6D, [137]online supplemental figure S8B). Additionally, PAR2 inhibitors reduced IL-1β and IL-6, effectively blocking M1 polarization ([138]figure 6E, [139]online supplemental figure S8C), while PAR1 inhibitor did not affect this outcome. Furthermore, PAR2 inhibitors impeded the nuclear translocation of p65 ([140]figure 6F). These results demonstrated that RGS2 mitigated IL-1β and IL-6 production by deactivating PAR2 via the MAPK/NF-κB signaling pathway ([141]figure 7). Figure 7. Schematic diagram of CEBPD affecting M1-type polarization of macrophages through the RGS2/PAR2 axis. CEBPD, CCAAT/enhancer-binding protein delta; IL, interleukin; PAR, proteinase-activated receptor. [142]Figure 7 [143]Open in a new tab CEBPD correlates with ICB therapeutic efficacy Given that ICBs are widely applied in patients with advanced and metastatic ccRCC, and that TAMs are also associated with the efficacy of ICBs,[144]^28 29 we further examined whether CEBPD influences the therapeutic effect of ICBs. We performed mIHC on ccRCC patients who received postoperative ICB therapy. The results showed that patients respond to ICB therapy had low expression of CEBPD, whereas patients who did not respond to ICB therapy had significantly elevated expression of CEBPD ([145]figure 8A,B). Meanwhile, the response group showed a higher level of CD86 and lower CD163 compared with non-response group ([146]figure 8A). Additionally, low-CEBPD patients had longer overall survival compared with high-CEBPD patients ([147]figure 8C). Furthermore, we investigated the association between CEBPD and the reported pathways through which TAMs influence ICB therapeutic efficacy by TCGA and scRNA-seq ([148]online supplemental figure S7). The result showed that CEBPD was not correlated with VEGF or TGF-β pathway, while it was negatively correlated with M1 macrophage gene set score. These results indicated that higher CEBPD expression in TAMs correlated with worse ICB efficacy, which may mainly mediate the polarization of TAMs. Additionally, in vivo experiments demonstrated enhanced efficacy of anti-PD1 therapy in CEBPD-knockout mice compared with wild type ([149]online supplemental figure S10), which indicated that targeting CEBPD may serve as a complement to existing immunotherapy. Figure 8. CEBPD correlated with ICB efficacy in patients with RCC. (A) Representative immunofluorescence images of patient respond to ICB therapy (left) and patient non-respond to ICB therapy (right), and (B) quantitative analysis of CEBPD intensity in two groups. (C) Kaplan-Meier curves of survival probability in CEBPD-high group and CEBPD-low group. CEBPD, CCAAT/enhancer-binding protein delta; ICB, immune checkpoint blocker; RCC, renal cell carcinoma. [150]Figure 8 [151]Open in a new tab Discussion Macrophages are an important part of the immune system to eliminate tumor cells. In ccRCC, though macrophages are abundant in the TME, it seems like macrophages are “hijacked” by tumor cells and lose antitumor function. The ratio of M1/M2 TAMs is a dynamic indicator influenced by tumor progression and therapy. Low M1/M2 ratio represents an immunosuppressive TME and leads to poor clinical outcomes. It is well established that TAMs are susceptible to polarization to the M2 phenotype and contribute to tumorigenesis, metastasis, tumor angiogenesis, immunosuppression and drug resistance.[152]30,[153]33 In recent years, more specific clusters of TAMs were identified, some of which expressed both traditional M1 and M2 markers. But we can still broadly categorize TAMs into proinflammatory type and immunosuppressive type. Therefore, future research should focus on reversing this trend to enhance the ratio of proinflammatory TAMs while reducing immunosuppressive types.[154]^34 35 In this study, bioinformatics analysis indicated that CEBPD negatively influences antitumor effects. In vitro experiments showed that the knockdown of CEBPD promoted the M1 type polarization of macrophages without affecting M2 type polarization. However, multiple immune-fluorescence analyses on patient specimens showed that tumors with high CEBPD expression had fewer M1-like TAMs and more M2-like TAMs compared with tumors with low CEBPD expression. This revealed a competitive dynamic between M1 and M2 polarization in the TME.[155]^36 Previous studies have demonstrated that TAMs impact the efficacy of ICBs.[156]37,[157]39 TAM-derived cytokines can influence the expression of PD-L1. For instance, in pancreatic ductal adenocarcinoma, TAM-derived TGF-β upregulates tumor PD-L1 expression via the AKT/NF-κB pathway.[158]^40 In bladder cancer, PEG2 can increase the expression of PD-L1 in tumor-infiltrated myeloid cells and exclude CD8^+ T cells.[159]^41 Besides, TAM-expressed ligands, such as V-domain Ig-containing suppressor of T-cell activation, can block anti-PD-1/PD-L1 immune efficacy.[160]42,[161]44 Moreover, TAMs can also express PD-1; Gordon et al found that PD-1^+ TAMs exhibited a reduced phagocytic capacity compared with PD-1^− TAMs,[162]^45 indicating impaired capacity to present tumor antigen and failure to activate T cells. In our study, patients with ccRCC who responded to ICB therapy showed lower TAM levels of CEBPD, alongside a higher proportion of M1 TAMs and a lower proportion of M2 TAMs. Prior research has established a correlation between macrophage polarization status and the therapeutic outcomes of ICBs,[163]46,[164]49 which is consistent with our findings. Although the precise mechanism by which CEBPD influences anti-PD-1 efficacy remains to be elucidated, our findings suggest that CEBPD could be a potential target for immunotherapy in ccRCC. We reported for the first time that RGS2 is involved in TAM polarization. To confirm that RGS2 is directly regulated by CEBPD, we performed ChIP-seq, dual-luciferase assay and EMSA. RGS2, acting as GTPase activating protein, modulates G protein alpha subunits and drives G proteins into inactive GDP-bound form.[165]^20 21 Prior research has indicated that RGS2 plays a suppressive role in proinflammatory responses of macrophages. Macrophages derived from RGS2-KO mice showed increased response to TLR2 agonist and tended to produce more iNOS.[166]^50 Lee et al revealed that RGS2 could repress STAT3-mediated transcriptional activation of Nox1, thereby attenuating innate immunity.[167]^51 In this study, we discovered that RGS2 deactivates PAR2 and inhibits the MAPK/NF-κB pathway, resulting in blocked M1 polarization and reduced production of IL-1β and IL-6. Targeting the RGS2/PAR2 axis holds promise for enhancing M1 TAM activation and promoting their proinflammatory functions against tumors. Conclusion Our study demonstrated that upregulation of CEBPD is associated with impaired M1 polarization in ccRCC. We identified the CEBPD/RGS2/PAR2 axis involved in this process. Furthermore, our findings suggested that targeting CEBPD could serve as a promising immunotherapy approach and a complementary strategy to ICB therapies. Supplementary material online supplemental file 1 [168]jitc-13-7-s001.docx^ (4.9MB, docx) DOI: 10.1136/jitc-2024-010898 Footnotes Funding: This work was sponsored by the National Natural Science Foundation of China (No. 82330094, 82072806, 82173265); Leading health talents of Shanghai Municipal Health Commission (2022LJ002); Shanghai Rising-Star Program (23QC1401400); the Natural Science Foundation of Shanghai (23ZR1441300); Hospital Funded Clinical Research, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (21XHDB06). Provenance and peer review: Not commissioned; externally peer reviewed. Patient consent for publication: Not applicable. Ethics approval: This study involves human participants and was approved by Ethics Committee of Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Approval No. XHEC-D-2024-140. Participants gave informed consent to participate in the study before taking part. Correction notice: This article has been corrected since it was first published. The corresponding author information has been updated. 7th August 2025. Data availability statement Data are available upon reasonable request. References