Cancer cell–intrinsic properties caused by oncogenic mutations have been well characterized; however, how specific oncogenes and tumor suppressors impact the tumor microenvironment (TME) is not well understood. Here, we present a novel non–cell-autonomous function of the retinoblastoma (RB) tumor suppressor in controlling the TME. RB inactivation stimulated tumor growth and neoangiogenesis in a syngeneic and orthotropic murine soft-tissue sarcoma model, which was associated with recruitment of tumor-associated macrophages (TAM) and immunosuppressive cells such as Gr1+CD11b+ myeloid-derived suppressor cells (MDSC) or Foxp3+ regulatory T cells (Treg). Gene expression profiling and analysis of genetically engineered mouse models revealed that RB inactivation increased secretion of the chemoattractant CCL2. Furthermore, activation of the CCL2–CCR2 axis in the TME promoted tumor angiogenesis and recruitment of TAMs and MDSCs into the TME in several tumor types including sarcoma and breast cancer. Loss of RB increased fatty acid oxidation (FAO) by activating AMP-activated protein kinase that led to inactivation of acetyl-CoA carboxylase, which suppresses FAO. This promoted mitochondrial superoxide production and JNK activation, which enhanced CCL2 expression. These findings indicate that the CCL2–CCR2 axis could be an effective therapeutic target in RB-deficient tumors.

Significance:

These findings demonstrate the cell-nonautonomous role of the tumor suppressor retinoblastoma in the tumor microenvironment, linking retinoblastoma loss to immunosuppression.

The tumor microenvironment (TME) in solid tumors consists of extracellular matrix as well as the associated stromal cells including immune cells, fibroblasts, and vascular networks. Bidirectional interactions between tumor cells and the TME enhance tumor progression at multiple levels: by supplying a variety of cytokines and growth factors, and nurturing cancer cells via the promotion of angiogenesis (1). Immune checkpoint blockade (ICB) accelerates the antitumor function of the TME through cytotoxic T-cell–mediated immunosurveillance, which is frequently attenuated by the aberrant expression of immune checkpoint molecules such as PD-L1 (2). Furthermore, the recruitment of immunosuppressive cells such as myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) into the TME also leads to a poorly immunogenic tumor; thus the ratio of tumor-infiltrating CD8+ cytotoxic T cells to immunosuppressive cells has been shown to predict patient outcomes for cancer immunotherapies using ICB (1).

Recent advances in cancer immunotherapies including ICB have been dramatic; however, they still provide limited benefits for the majority of patients. For example, according to an early-phase I trial in patients with breast cancer, the effectiveness of ICB has been recognized particularly in triple-negative breast cancer (TNBC) due to its lack of targetable molecules such as estrogen receptor (ER) and human EGFR-2 (HER2), with an approximately 20% overall response rate (3). In addition to the expression of immune checkpoint molecules or the frequencies of tumor-infiltrating CD8+ cytotoxic T cells in tumor tissues, several groups recently demonstrated that genetic aberrations in cancer cells such as loss-of-function mutations in JAK1/2, APLNR, PTPN2, and PBRM1 are significantly correlated with the efficacy of cancer immunotherapies (4–9). In spite of such extensive efforts, it remains highly challenging to effectively determine which patient will respond to current immunotherapies because of the complexity of tumor heterogeneity in terms of genetic, epigenetic, and/or microenvironment levels in aggressive tumors.

Although cancer cell–intrinsic properties resulting from oncogenic mutations have been well characterized, it is becoming increasingly clear that oncogenic mutations impact angiogenesis and/or the recruitment and phenotype of immune cells in the TME via increasing secretion of cytokines and chemokines by tumor cells (10). The BRAFV600E mutation in melanoma cells promotes the secretion of multiple cytokines including IL6 and VEGFα, contributing to the establishment of a protumoral microenvironment (11). The oncogenic KRAS mutation in pancreatic ductal adenocarcinoma induced GM-CSF secretion, which enhanced the recruitment of Gr1+CD11b+ MDSCs and subsequent suppression of CD8+ cytotoxic T-cell infiltration in the TME (12, 13). In addition, the loss of tumor suppressor genes such as STK11/LKB1 and PTEN affects T-cell infiltration into the TME by modulating the secretion of cytokines and chemokines (14, 15). We previously reported that retinoblastoma (RB) loss in Trp53-null sarcoma cells and ARF-deficient breast cancer cells increased the secretion of cytokines such as IL6 via enhanced mitochondrial superoxide (MS) production, which stimulates their self-renewal activity in a cell-autonomous manner (16, 17). However, the role of enhanced cytokine production following RB inactivation in the TME remains unclear.

In this study, to elucidate the significance of non–cell-autonomous RB function in tumor tissue, we employed a syngeneic and orthotopic murine soft-tissue sarcoma model and a mammary carcinogenesis model to analyze the TME of RB-deficient tumors. We found that enhanced CCL2 secretion following RB inactivation contributes to the establishment of a tumor-promoting microenvironment due to the recruitment of immunosuppressive cells such as Gr1+CD11b+ MDSCs and F4/80+ tumor-associated macrophages (TAM) into the TME.

Mice

Trp53 knockout mice (18) were obtained from RIKEN BioResource Center (#CDB0001K). Ccr2 knockout mice were obtained from Dr. W.A. Kuziel (PDL Bio Pharma Inc., Incline Village, NV; ref. 19). Ccl2 knockout mice (#004434) and MMTV-Cre mice (#003553) were purchased from the Jackson Laboratory. Rbflox/flox mice were gifted from Dr. A. Berns (Netherlands Cancer Institute, Amsterdam). Wild-type C57BL/6 mice were purchased from Japan SLC. Female of MMTV-Cre; Rbflox/flox mice for mammary carcinogenesis were never mated to male mice during analysis. Mouse experiments were conducted in accordance with a Kanazawa University Institutional Animal Care and Use Committee-approved protocol (AP-153426).

Cell line and primary cell culture

Minced pieces of soft-tissue sarcoma samples derived from p53-knockout mice were digested with 300 U/mL collagenase (#C-5138, Sigma-Aldrich), 100 U/mL hyaluronidase (#H3506-1G, Sigma-Aldrich) and 100 μg/mL DNase I (#DN25-100MG, Sigma-Aldrich) in α-modified Eagle's medium (αMEM) supplemented with 10% FBS (17). Primary cells from surgically removed human breast carcinomas were established in the laboratory of Dr. Noriko Gotoh (Kanazawa University; ref. 20), and maintained in HuMEC (#12752-010, Life Technologies). The institutional review boards of Kanazawa University (Ishikawa, Japan; #335) and the Institute of Medical Science, The University of Tokyo (Tokyo, Japan), approved this study. RAW 264.7 (RIKEN BRC, RCB0535), THP-1 (RIKEN BRC, RCB1189), MCF7 (RIKEN BRC, RCB1904), and MDA-MB-231 [ATCC (HTB-26)] were cultured in DMEM containing 10% FBS. Hs578t [ATCC (HTB-126)] were maintained in DMEM supplemented with 10% FBS and 10 μg/mL recombinant insulin. HCC1187 [ATCC (CRL-2322)] were maintained in RPMI1640 supplemented with 10% FBS. These cell lines were authenticated by RIKEN BRC and ATCC, and all experiments were performed before reaching 10 passages. Mycoplasma infection was regularly checked by PCR using the conditioned media derived from each cell line (#G238, abm).

Generation of lentivirus

MISSION TRC validated shRNA target sets for mouse Rb (TRCN0000042543 and TRCN0000042544), human RB (TRCN0000040163 and TRCN0000010419), and negative control (Scramble; SHC002) were purchased from Sigma-Aldrich. pQCXIH-PSM-RB7LP was purchased from Addgene (#37106), and RB7LP-lacking stop codon was amplified by PCR (16). PCR products were then cloned into pDONR223, and subcloned into pLX304. pCL20c-CMV-EGFP and pCL20c-CMV-EGFP-DN-c-Jun were obtained from Dr. Katsuji Yoshioka (Kanazawa University; ref. 21). Generation and infection of lentivirus were performed according to the manufacturer's instructions.

In vivo tumor formation assay

Cells suspended in 50 μL αMEM with 10% FBS were mixed with 50 μL Matrigel (#354234; Corning) and injected subcutaneously into wild-type or Ccr2 KO C57BL/6 male mice. Tumors were assessed 14–28 days after injection.

qRT-PCR

tRNA was isolated from cultured cells or tumor tissues by using TRIzol (#15596018, Life Technologies) according to the manufacturer's instructions. qPCR of tRNA was performed as described previously (17) using Taqman probes. Taqman probes: mouse Actb (Mm00607939_s1), mouse Rb (Mm00485586_m1), mouse Vegfα (Mm01281449_m1), mouse Perforin1 (Mm00812512_m1), mouse Ccl2 (Mm00441242_m1), mouse Ccr2 (Mm99999051_gH), mouse Il6 (Mm00446190_ m1), mouse Il1α (Mm00439620_m1), mouse Cxcl1 (Mm04207460_ m1), mouse Cxcl5 (Mm00436451_g1), mouse Ptgs2 (Mm00478374_m1), human ACTB (Hs99999903_m1), human RB (Hs01078066_m1), human CCL2 (Hs00234140_ m1), and human CCL5 (Hs00982282_m1). The relative level of gene expression was normalized using the level of Actb or ACTB.

IHC

IHC was performed on paraffin-embedded subcutaneous tumor sections. After deparaffinizing tissue blocks, antigen retrieval was performed by 0.01% trypsin at 37°C for 10 minutes (CD31, Gr-1, and F4/80), 10 mmol/L Tris-1 mmol/L EDTA buffer (pH 9.0) at 90°C for 10 minutes (CD3, CD4, CD8, and Foxp3), or 10 mmol/L citrate at 100°C for 10 minutes (PCNA and CCL2). To block nonspecific signal, tissue sections were incubated for 10 minutes at room temperature using PBS containing 5% goat serum, 1% BSA, and 0.1% TritonX-100. Serial tissue sections were stained with following antibodies: CD31 (#557355, BD Pharmingen), Gr-1 (#550291, BD Pharmingen), F4/80 (#MCA497G, Bio-Rad), CD3 (#MCA1477, Bio-Rad), CD4 (ab183685, Abcam), CD8a (#14-0195, Thermo Fisher Scientific), Foxp3 (#623801, BioLegend), PCNA (#13110, Cell Signaling Technology), and CCL2 (#ab25124, Abcam). Sections were visualized with ZEISS microscope equipped with ZEISS AxioCam HRc camera and Axio Vision 4.1. Obtained digital images were analyzed by Photoshop to determine the proportion of immune-positive cells.

Flow cytometry

Tumors were surgically removed from transplanted mice. The tumors were cut into smaller fragments, and digested in 5-mL DMEM medium containing 3,045 units collagenase, 1,050 units hyaluronidase, and 200–400 units deoxyribonuclease at 37°C for 1 hour. Then 1 × 106 cells were stained by CD11b-APC (#553312, BD Pharmingen), Gr-1-FITC (#11-5931, eBioscience), F4/80-PE (#12-4801, eBioscience), CD45-PerCP-Cy5.5 (#45-0451, eBioscience), and CCR2-PE (#150609, BioLegend) antibodies. MS level was determined by MitoSOX Red (#M36008, Life Technologies). A total of 1 × 106 cells suspended in 500 μL PBS containing 3% FBS were analyzed by FACSCanto ll (BD Biosciences).

RNA sequence

The tRNA was extracted using the TRIzol reagent (#15596018, Life Technologies). From 15 μg of tRNA, a RNA-sequence library was constructed using the mRNA-sequence Sample Preparation Kit, according to the manufacturer's instructions (Illumina). A total of 36 base-pair single-end-read RNA-sequence tags were generated using a HiSeq 2000 sequencer, according to the standard protocol. The RNA-sequence tags were mapped to the mouse genomic sequence (mm9 from the UCSC Genome Browser) using the ELAND program (Illumina). Unmapped or redundantly mapped sequences were removed from the dataset, and only uniquely mapped sequences without any mismatches were used for the analyses (22). The raw data of RNA sequence are available in DNA Data Bank of Japan (DDBJ; DRA002911).

ELISA assay

Blood samples were collected directly from the heart of anesthetized Ccl2 KO mice 17 days after subcutaneous tumor injection, incubated for 24 hours at 4°C, and then centrifuged at 1,200 rcf for 30 minutes at 4°C to separate the serum. Conditioned medium was collected from 53KOSTS and MCF7 cells transduced with shRNA. Mouse Ccl2 ELISA (#88-7391, eBioscience) and human CCL2 (#DCP00, R&D Systems) and human CCL5 ELISA (#DRN00B, R&D Systems) were performed according to the manufacturer's instructions.

Reagents

The following reagents were used: anti-mouse Ccl2 antibody (#554440, BD Biosciences), Armenian Hamster-IgG (#400916, BioLegend), carmine alum (#07070, STEMCELL Technologies), N-acetylcysteine (#A7250, Sigma-Aldrich), Trolox (#202-17891, Wako), mitoquinone (#10-1363, Focus Biomolecures), carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP; #C2920, Sigma-Aldrich), and SP600125 (#8177, Cell Signaling Technology).

CRISPR/Cas9 system

Target sequences for CRISPR interference were designed using the sgRNA designer (http://portals.broadinstitute.org/gpp/public/analysis-tools/sgrna-design), and then cloned into pCRISPRv2. A nontargeting sgRNA from the Gecko library v2 was used as a scramble sgRNA.

Scramble sgRNA: ACGTGTAAGGCGAACGCCTT

Mouse Ccl2 sgRNA: TGTCACCAAGCTCAAGAGAG

Microarray analysis

tRNA was extracted using the RNeasy Mini Kit (#74106, Qiagen) according to the manufacturer's instructions. The quality of the tRNA samples was assessed using the RNA 6000 Nano Lab Chip Kit (Bio analyzer 2100, Agilent Technologies). The microarray analysis was performed with a Human Gene Expression 4 × 44K v2 Microarray (#26652, Agilent Technologies). The fluorescence intensity was measured by the G2505C Microarray Scanner (Agilent Technologies). Three samples were analyzed per group. Data were analyzed by Gene Spring 12.6.1 - GX - PA (Agilent Technologies) and R version 3.1.1. The raw data of microarray analysis are available in Gene Expression Omnibus database (GSE64525).

Immunoblotting

Whole-cell lysates were prepared as described previously (23). Immunoblotting was performed as described previously (23) using following antibodies: Phospho-Rb (#9308, Cell Signaling Technology), total RB (#554136, BD Biosciences), Phospho-ACC (#3661, Cell Signaling Technology), total ACC (#3676, Cell Signaling Technology), Phospho-AMPK (#2535, Cell Signaling Technology), total AMPK (#5832, Cell Signaling Technology), α-tubulin (#CP06-100UG, EMD Millipore), and β-Actin (#3700, Cell Signaling Technology).

Chemotaxis assay

Conditioned medium was collected from MCF7 transduced with shRNA cultured in serum-free RPMI1640 for 48 hours. A total of 1 × 105 THP-1 cells suspended in serum-free RPMI1640 (WAKO) were loaded into the upper chamber of the 24-well–type Microchemotaxis Chamber (#CLS3421, Sigma-Aldrich), and 600 μL conditioned medium derived from MCF7 cells was added to the lower chamber. After 24-hour incubation at 37°C, the number of THP-1 cells migrated into the lower chamber was analyzed by BZ analysis software on BZ-9000 (Keyence) and Photoshop.

Immunofluorescence

Immunofluorescence was performed on paraffin-embedded subcutaneous tumor sections. After deparaffinizing tissue blocks, antigen retrieval was performed by boiling the sections in pH6 10 mmol/L citrate buffer for 10 minutes. To block nonspecific signal, tissue sections were incubated for 10 minutes at room temperature using PBS containing 5% goat serum, 1% BSA, and 0.1% TritonX-100. Serial tissue sections were stained with following antibodies: F4/80 (#MCA497G, Bio-Rad), and CK18 (#GTX105624, GeneTex). Put the sections in 4 °C overnight. The next day, we labeled the F4/80 antibody with Alexa Fluor 633 (#A-21094, Thermo Fisher Scientific), labeled the CK18 antibody with Alexa Fluor 488 (#A-11034, Thermo Fisher Scientific), and then we mount the sections with Antifade Mounting Medium with DAPI (#H-1200, VECTOR). Sections were visualized with Leica TCS SP8 microscope and LAS X 1.8. Obtained digital images were analyzed by photoshop to determine the proportion of immune-positive cells.

Statistical analysis

Statistical significance was assessed using unpaired two-tailed Student t test, or one-way ANOVA followed by Tukey post hoc test. P values less than 0.05 were considered significant. Asterisks used to indicate significance correspond with: *, P < 0.05; **, P < 0.01. Columns represent means ± SD. In one-way ANOVA followed by post hoc tests, we showed asterisks only in pairs of our interest. GraphPad Prism7 was used for all statistical analysis, data processing, and presentation.

Rb loss alters the TME

To assess whether RB status affects tumor progression in a non–cell-autonomous manner, we established a syngeneic and orthotropic murine soft-tissue sarcoma model to analyze the TME of RB-deficient tumors (Fig. 1A). First, we depleted Rb in a Trp53 knockout (KO) C57BL/6 mouse–derived sarcoma cell line named 53KOSTS (Trp53 knockout soft-tissue sarcoma) cells (Supplementary Fig. S1A), which we established previously (16, 17). As we expected, Rb depletion in 53KOSTS cells caused an acceleration of cell growth in vitro (Supplementary Fig. S1B) and an increase in tumor size following orthotopic engraftment in wild-type C57BL/6 mice. Interestingly, tumor tissues derived from Rb-depleted 53KOSTS cells appeared to be more vascularized than the control tissues (Fig. 1B). To validate this observation, we performed endothelial cell marker CD31 staining to confirm higher angiogenesis in Rb-depleted tumors (Fig. 1C). Consistent with higher CD31 expression in Rb-depleted tumors and a previous report describing induction of Vegfα expression following Rb-inactivation (24), Vegfα expression was 3-fold higher in tumor tissues derived from Rb-depleted 53KOSTS cells, even though Rb depletion only weakly induced Vegfα expression in 53KOSTS cells themselves in vitro (Fig. 1D and E). On the basis of these findings, we expected that stromal cells that formed the TME might also be stimulated to express Vegfα upon Rb depletion in 53KOSTS cells due to cell–cell interactions including humoral factors. Indeed, we observed that treatment with conditioned medium (CM) derived from Rb-depleted 53KOSTS cells significantly increased expression of Vegfα, as well as that of two other activation markers, IL6 and Il1α, in an RAW264.7 mouse macrophage cell line (Fig. 1F), implying that Rb depletion in tumor cells might affect the TME via secreted factors.

Figure 1.

Rb inactivation induces a protumoral microenvironment in the syngeneic and orthotopic murine soft-tissue sarcoma model. A, A schematic drawing of syngeneic and orthotopic murine soft-tissue sarcoma model. SC, subcutaneous. B, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA. A total of 1 × 105 cells were injected subcutaneously into syngeneic C57BL/6 mice (day 24). Scale bar, 10 mm. Tumors were weighed (right; n = 9). C, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. CD31 cells were quantified (right; n = 8). D, qRT-PCR of the indicated gene in 53KOSTS cells transduced with the indicated shRNA (n = 3). E, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 5). F, qRT-PCR of the indicated gene in RAW264.7 cells treated with the CM derived from 53KOSTS cells transduced with the indicated shRNA (n = 3). Growth medium was used as a negative control. G, J, and K, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. Gr-1+, CD3+, CD4+, Foxp3+, or CD8+ cells were quantified (n > 8). H and I, The profiles (% in CD45+ cells) of Gr-1/CD11b (H) or F4/80+ (I) cells in tumors derived from 53KOSTS cells transduced with the indicated shRNA (n > 4). The insets show Gr1/CD11b double-positive cells (H). L, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 8). *, P < 0.05; **, P < 0.01.

Figure 1.

Rb inactivation induces a protumoral microenvironment in the syngeneic and orthotopic murine soft-tissue sarcoma model. A, A schematic drawing of syngeneic and orthotopic murine soft-tissue sarcoma model. SC, subcutaneous. B, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA. A total of 1 × 105 cells were injected subcutaneously into syngeneic C57BL/6 mice (day 24). Scale bar, 10 mm. Tumors were weighed (right; n = 9). C, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. CD31 cells were quantified (right; n = 8). D, qRT-PCR of the indicated gene in 53KOSTS cells transduced with the indicated shRNA (n = 3). E, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 5). F, qRT-PCR of the indicated gene in RAW264.7 cells treated with the CM derived from 53KOSTS cells transduced with the indicated shRNA (n = 3). Growth medium was used as a negative control. G, J, and K, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. Gr-1+, CD3+, CD4+, Foxp3+, or CD8+ cells were quantified (n > 8). H and I, The profiles (% in CD45+ cells) of Gr-1/CD11b (H) or F4/80+ (I) cells in tumors derived from 53KOSTS cells transduced with the indicated shRNA (n > 4). The insets show Gr1/CD11b double-positive cells (H). L, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 8). *, P < 0.05; **, P < 0.01.

Close modal

Given these changes in the TME, we next characterized infiltration of immune cells into tumor tissues derived from Rb-depleted 53KOSTS cells. Interestingly, Rb depletion significantly promoted the infiltration of Gr1+CD11b+ MDSCs, which are known to be very potent suppressors of cytotoxic T-cell immunity (Fig. 1G and H). In addition, Rb depletion slightly enhanced the infiltration of F4/80+ TAMs in the TME (Fig. 1I). Moreover, consistent with a higher number of CD3+ pan-T cells in the TME (Fig. 1J), we observed a significantly higher number of CD4+ effector or Foxp3+ Tregs in the TME of Rb-depleted tumors, although the infiltration of CD8+ cytotoxic T cells was slightly lower in Rb-depleted tumors (Fig. 1K; Supplementary Fig. S1C). We confirmed lower expression of perforin, the cytolytic granule effector molecule of CD8+ T cells, in Rb-depleted tumors by qRT-PCR (Fig. 1L). Taken together, Rb depletion not only enhances angiogenesis, but also contributes to the establishment of a protumoral microenvironment by recruiting immunosuppressive cells such as MDSCs into the TME.

Rb depletion elevates Ccl2 expression

We next systematically assessed RB-regulated factors that could induce these changes in the TME by performing RNA sequencing of Rb-depleted 53KOSTS cells. This uncovered multiple chemokine genes including Ccl2 that were upregulated following Rb depletion in these cells (Fig. 2A). Furthermore, pathway analysis using Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that cytokine–cytokine receptor interactions and chemokine signaling pathways were significantly upregulated in Rb-depleted 53KOSTS cells (Table 1). We then validated the upregulation of Ccl2 and other chemokines such as Cxcl1 and Cxcl5 by qRT-PCR ((Fig. 2B; Supplementary Fig. S1D). In addition, we detected enhanced Ccl2 secretion following Rb depletion in 53KOSTS cells using ELISA (Fig. 2C). Despite higher secretion levels of Ccl2 from Rb-depleted 53KOSTS cells, they themselves seemed not to receive Ccl2 because of the lack of Ccr2 expression, which encodes the main receptor for Ccl2 (Fig. 2D). These data suggested that enhanced Ccl2 secretion from Rb-depleted 53KOSTS cells mainly involved stromal cells, but not tumor cells in vivo, and might contribute to the remodeling of the TME in a non–cell-autonomous manner. Consistent with this idea, in addition to Ccl2 expression, tumor tissues derived from Rb-depleted 53KOSTS cells showed significantly high Il1α expression (Fig. 2E), which was induced in the macrophage cell line by treatment with CM derived from Rb-depleted 53KOSTS cells (Fig. 1F).

Figure 2.

Ccl2 is upregulated following Rb depletion in mouse sarcoma cells. A, Heatmap indicating fold change of the top 50 genes upregulated by Rb depletion in 53KOSTS cells based on RNA-sequence analysis. Fifty genes were reranked by average RPKM value. B, qRT-PCR of the indicated gene in 53KOSTS cells transduced with the indicated shRNA (n = 3). #1 and #2, independent shRNA. C, ELISA of mouse Ccl2 levels in 53KOSTS cells transduced with the indicated shRNA following 10-hour culture (n = 3). D, qRT-PCR of the indicated gene in RAW264.7 and 53KOSTS cells (n = 3). E, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 3). *, P < 0.05; **, P < 0.01.

Figure 2.

Ccl2 is upregulated following Rb depletion in mouse sarcoma cells. A, Heatmap indicating fold change of the top 50 genes upregulated by Rb depletion in 53KOSTS cells based on RNA-sequence analysis. Fifty genes were reranked by average RPKM value. B, qRT-PCR of the indicated gene in 53KOSTS cells transduced with the indicated shRNA (n = 3). #1 and #2, independent shRNA. C, ELISA of mouse Ccl2 levels in 53KOSTS cells transduced with the indicated shRNA following 10-hour culture (n = 3). D, qRT-PCR of the indicated gene in RAW264.7 and 53KOSTS cells (n = 3). E, qRT-PCR of the indicated gene in tumor nodules derived from 53KOSTS cells transduced with the indicated shRNA (n = 3). *, P < 0.05; **, P < 0.01.

Close modal
Table 1.

Pathway analysis using KEGG of genes upregulated by Rb depletion in 53KOSTS cells

KEGG pathway termPBonferroniBenjamini
Steroid biosynthesis 2.32E-05 1.60E-03 1.60E-03 
Cytokine–cytokine receptor interaction 8.67E-04 5.81E-02 2.95E-02 
NOD-like receptor signaling pathway 3.84E-03 2.33E-01 8.46E-02 
Chemokine signaling pathway 4.07E-02 9.43E-01 5.12E-01 
KEGG pathway termPBonferroniBenjamini
Steroid biosynthesis 2.32E-05 1.60E-03 1.60E-03 
Cytokine–cytokine receptor interaction 8.67E-04 5.81E-02 2.95E-02 
NOD-like receptor signaling pathway 3.84E-03 2.33E-01 8.46E-02 
Chemokine signaling pathway 4.07E-02 9.43E-01 5.12E-01 

Rb depletion induces tumor progression, depending on Ccl2–Ccr2 axis

To determine the specific contribution of enhanced Ccl2 secretion to tumor progression in vivo, we next subcutaneously injected 53KOSTS cells into Ccl2 KO C57BL/6 mice to determine the concentration of tumor-derived Ccl2 in serum (Fig. 3A). Consistent with the results in vitro, Rb depletion elevated the concentration of tumor-derived Ccl2 in serum in vivo (Fig. 3B). Next, to elucidate the role of Ccl2 in vivo in tumor progression promoted by Rb depletion, we employed Ccr2 KO C57BL/6 mice to abolish the Ccl2–Ccr2 axis in the TME (Fig. 3C). Interestingly, Rb-depleted cells generated significantly smaller tumors in Ccr2 KO C57BL/6 mice than in wild-type C57BL/6 mice (Fig. 3D). To further study how the Ccl2–Ccr2 axis promoted tumor development, we then assessed angiogenesis and infiltration of immune cells such as MDSCs and T cells into the TME. Angiogenesis induced by Rb depletion in the TME was markedly suppressed in Ccr2 KO C57BL/6 mice. (Fig. 3E). In addition, infiltration of TAMs and MDSCs induced upon Rb depletion was clearly suppressed in Ccr2 KO C57BL/6 mice (Fig. 3E–G). Previously, several groups have shown that TAMs and MDSCs are recruited into the TME by CCL2 secreted from tumor cells and contribute to tumor angiogenesis by producing angiogenic factors such as VEGF (25, 26). Consistent with these reports, we observed that approximately 95% of Gr1+CD11b+ MDSCs in the TME express Ccr2 (Fig. 3H). However, the infiltration of T cells including Tregs did not decrease, suggesting that this was independent of the activation of the Ccl2–Ccr2 pathway (Supplementary Fig. S1E and S1F). Moreover, we demonstrated that CRISPR/Cas9–mediated depletion of Ccl2 significantly attenuated tumor growth, angiogenesis, and the infiltration of TAMs and MDSCs in Rb-depleted 53KOSTS cells in C57BL/6 mice (Fig. 3I–K; Supplementary Fig. S1G–S1I). Taken together, these findings suggest that tumor progression induced by Rb depletion, at least in part, depends on the elevated Ccl2 secretion and the subsequent activation of Ccr2-dependent angiogenesis or the recruitment of immunosuppressive cells into the TME.

Figure 3.

Tumor progression induced by Rb depletion depends on Ccl2–Ccr2 axis. A, A schematic drawing of ELISA for serum derived from Ccl2 KO C57BL/6 mouse. B, ELISA of mouse Ccl2 levels in serum derived from tumor-bearing Ccl2 KO C57BL/6 mouse. Ccl2 concentration in serum was normalized by tumor weight (n = 7). C, A schematic drawing of subcutaneous tumor implantation of 53KOSTS cells in Ccr2 WT or KO C57BL/6 mice. D, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA. A total of 3 × 105 cells were injected subcutaneously into Ccr2 WT or KO C57BL/6 mice (day 17). Scale bar, 10 mm. Tumors were weighed (right; n = 7). E, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. CD31 or Gr-1+ cells were quantified (right; n = 3). F and G, The profiles (% in CD45+ cells) of F4/80+ cells (F) or Gr-1+/CD11b+ cells (G) in tumors derived from 53KOSTS cells transduced with the indicated shRNA (n > 4). The insets show Gr1/CD11b double-positive cells (G). H, The 4 × 105 cells were injected subcutaneously into C57BL/6 mice. Tumors were analyzed at day 14. Ccr2 expression in CD45 or CD45+/Gr1+/CD11b+ cells in the TME. I, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA. Scale bar, 10 mm. Tumors were weighed (right; n = 5). J, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA. Gr-1+ cells were quantified (right; n = 5). K, The proportion of Gr-1+/CD11b+ cells in tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA (n = 10). n.s., not significant; *, P < 0.05; **, P < 0.01.

Figure 3.

Tumor progression induced by Rb depletion depends on Ccl2–Ccr2 axis. A, A schematic drawing of ELISA for serum derived from Ccl2 KO C57BL/6 mouse. B, ELISA of mouse Ccl2 levels in serum derived from tumor-bearing Ccl2 KO C57BL/6 mouse. Ccl2 concentration in serum was normalized by tumor weight (n = 7). C, A schematic drawing of subcutaneous tumor implantation of 53KOSTS cells in Ccr2 WT or KO C57BL/6 mice. D, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA. A total of 3 × 105 cells were injected subcutaneously into Ccr2 WT or KO C57BL/6 mice (day 17). Scale bar, 10 mm. Tumors were weighed (right; n = 7). E, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA. Scale bar, 100 μm. CD31 or Gr-1+ cells were quantified (right; n = 3). F and G, The profiles (% in CD45+ cells) of F4/80+ cells (F) or Gr-1+/CD11b+ cells (G) in tumors derived from 53KOSTS cells transduced with the indicated shRNA (n > 4). The insets show Gr1/CD11b double-positive cells (G). H, The 4 × 105 cells were injected subcutaneously into C57BL/6 mice. Tumors were analyzed at day 14. Ccr2 expression in CD45 or CD45+/Gr1+/CD11b+ cells in the TME. I, Representative pictures of tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA. Scale bar, 10 mm. Tumors were weighed (right; n = 5). J, IHC analysis of the indicated protein in tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA. Gr-1+ cells were quantified (right; n = 5). K, The proportion of Gr-1+/CD11b+ cells in tumors derived from 53KOSTS cells transduced with the indicated shRNA or sgRNA (n = 10). n.s., not significant; *, P < 0.05; **, P < 0.01.

Close modal

RB depletion upregulates CCL2 expression in human breast cancer cells

To validate the role of the CCL2–CCR2 axis and confirm its relevance to human cancer, we focused on human breast cancer because the RB gene shows genetic alterations in approximately 10% of patients with breast cancer (27). Furthermore, we previously reported enhanced cytokine secretion following RB inactivation in breast cancer cells (16, 17). According to gene expression profiling using DNA microarray and subsequent pathway analysis in RB-depleted MCF7 cells (RB intact and ARF deficient), we confirmed that cytokine signaling is highly upregulated following RB depletion in MCF7 cells as observed in 53KOSTS cells (Fig. 4A; Supplementary Fig. S2A). Furthermore, among CC chemokine family members, the expression levels of CCL2 and CCL5 were specifically upregulated, although we did not observe Ccl5 upregulation in Rb-depleted 53KOSTS cells (Supplementary Table S1). We next examined CCL2 and CCL5 expression across a panel of RB-positive breast cancer cell lines. In most cell lines, CCL2 and CCL5 expression was significantly higher following RB depletion (Fig. 4B; Supplementary Fig. S2B). We further examined patient-derived primary breast cancer cells, in which RB depletion also upregulated both CCL2 and CCL5 (Fig. 4C). Moreover, we confirmed enhanced CCL2 and CCL5 secretion following RB depletion by ELISA (Fig. 4D). In contrast to RB depletion, overexpression of the constitutively active (nonphosphorylatable) form of RB (RB7LP; ref. 28) clearly decreased both CCL2 and CCL5 expression (Fig. 4E). Consistent with these findings, CCL2 and CCL5 expression showed a weak inverse correlation with RB expression according to gene expression profiling data from breast cancer cell lines found in the Cancer Cell Line Encyclopedia (CCLE) database (Fig. 4F; Supplementary Table S2). To functionally validate the role of the RB–CCL2 axis, we next examined CCL2-dependent migration of THP-1 cells using a transwell migration assay. Importantly, compared with the CM derived from control MCF7 cells, CM derived from RB-depleted MCF7 cells exhibited stronger chemoattractant activity for THP-1 cells, which was significantly antagonized by treatment with an anti–CCL2-neutralizing antibody (Fig. 4G). Taken together, these data suggest that RB inactivation might influence the TME of breast cancer through enhanced chemokine secretion including CCL2.

Figure 4.

CCL2 is upregulated following Rb depletion in human breast cancer cells. A, Heatmap indicating fold change of CC chemokines following Rb depletion in MCF7 cells based on DNA microarray analysis. B, qRT-PCR of the indicated gene in RB-positive breast cancer cell lines transduced with the indicated shRNA (n > 3). #1 and #2, independent shRNA. C, qRT-PCR of the indicated gene in patient-derived primary breast cancer cells transduced with the indicated shRNA (n = 3). D, ELISA of human CCL2 and CCL5 levels in MCF7 cells transduced with the indicated shRNA following 48-hour culture (n = 3). N.D., not detected. E, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated vector (n = 3). Lac, LacZ. 7LP: RB7LP. F, Correlation between RB and CCL2 or CCL5 expression according to CCLE database. r and P values based on Pearson correlation. G, The number of THP1 cells migrated into the lower compartment of the microchemotaxis chamber containing conditioned medium derived from MCF7 cells transduced with the indicated shRNA with or without 20 μg/mL anti–CCL2-neutralizing antibody. n.s., not significant; *, P < 0.05; **, P < 0.01.

Figure 4.

CCL2 is upregulated following Rb depletion in human breast cancer cells. A, Heatmap indicating fold change of CC chemokines following Rb depletion in MCF7 cells based on DNA microarray analysis. B, qRT-PCR of the indicated gene in RB-positive breast cancer cell lines transduced with the indicated shRNA (n > 3). #1 and #2, independent shRNA. C, qRT-PCR of the indicated gene in patient-derived primary breast cancer cells transduced with the indicated shRNA (n = 3). D, ELISA of human CCL2 and CCL5 levels in MCF7 cells transduced with the indicated shRNA following 48-hour culture (n = 3). N.D., not detected. E, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated vector (n = 3). Lac, LacZ. 7LP: RB7LP. F, Correlation between RB and CCL2 or CCL5 expression according to CCLE database. r and P values based on Pearson correlation. G, The number of THP1 cells migrated into the lower compartment of the microchemotaxis chamber containing conditioned medium derived from MCF7 cells transduced with the indicated shRNA with or without 20 μg/mL anti–CCL2-neutralizing antibody. n.s., not significant; *, P < 0.05; **, P < 0.01.

Close modal

Enhanced FAO and MS production induces CCL2

Previously, we found that RB inactivation induced the enhanced secretion of several cytokines such as IL6 in breast cancer cell lines through enhanced MS production (17). In brief, the transcription of mitochondria-related genes especially related to fatty acid oxidation (FAO) such as CPT1 is upregulated due to deregulation of E2F activity following RB inactivation. CPT1 provides a rate-limiting step in long-chain fatty acid oxidation. CPT1 controls transportation of long-chain fatty acids into mitochondria. Long-chain fatty acids transported into mitochondria are used for β-oxidation. Consistent with higher expression of FAO-related genes, RB-depleted cells showed an elevated oxygen consumption rate upon palmitate stimulation and also an elevated MS production (17). In the same previous work (17), we demonstrated that RB inactivation significantly decrease malonyl-CoA level. This finding was highly consistent with elevated FAO because malonyl-CoA strongly suppresses FAO through inhibition of CPT1. However, why malonyl-CoA level drops following RB inactivation was not fully cleared in the previous study. In this work, we discovered that RB loss increases the phosphorylation of AMP-activated protein kinase (AMPK) and one of its substrates acetyl-CoA carboxylase (ACC; Fig. 5A). ACC plays a crucial role in regulating FAO. The malonyl-CoA, which is generated by ACC, specifically inhibits the CPT1 activation. The phosphorylated AMPK phosphorylates and thus inactivates ACC. Therefore, upon AMPK phosphorylation, the level of malonyl-CoA drops, leading to increased activity of CPT1. The RB–AMPK–ACC axis may provide a possible pathway whereby RB loss increases FAO.

Figure 5.

Enhanced MS production following RB depletion induces CCL2 but not CCL5. A, Immunoblot of the indicated proteins in MCF7 cells transduced with the indicated shRNA. B, Correlation between MS levels and CCL2 or CCL5 expression in breast cancer cell lines (MCF7, HCC1187, Hs578t, MDA-MB-231, MDA-MB-436, MDA-MB-468, and HCC70). r and P values based on Pearson correlations. C, MS levels in RB-positive breast cancer cell lines transduced with the indicated shRNA (n = 4). D, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA, treated with 5 mmol/L NAC, 100 μmol/L trolox, or 5 nmol/L mitoquinone for 24 hours (n = 3). E, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA, and treated with 1 μmol/L DMSO (DM) or 1 μmol/L FCCP (FC) for 4 hours (n = 3). F, qRT-PCR of the indicated gene in MCF7 transduced with the indicated shRNA and treated with or without 20 μmol/L SP600125 (SP) for 4 hours (n = 3). G, MCF7 cells transduced with the indicated lentiviral vectors observed under phase contrast or fluorescence microscope. Scale bar, 100 μm. H, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA and the indicated lentiviral expression vector (n = 3). DN, dominant negative c-Jun.

Figure 5.

Enhanced MS production following RB depletion induces CCL2 but not CCL5. A, Immunoblot of the indicated proteins in MCF7 cells transduced with the indicated shRNA. B, Correlation between MS levels and CCL2 or CCL5 expression in breast cancer cell lines (MCF7, HCC1187, Hs578t, MDA-MB-231, MDA-MB-436, MDA-MB-468, and HCC70). r and P values based on Pearson correlations. C, MS levels in RB-positive breast cancer cell lines transduced with the indicated shRNA (n = 4). D, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA, treated with 5 mmol/L NAC, 100 μmol/L trolox, or 5 nmol/L mitoquinone for 24 hours (n = 3). E, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA, and treated with 1 μmol/L DMSO (DM) or 1 μmol/L FCCP (FC) for 4 hours (n = 3). F, qRT-PCR of the indicated gene in MCF7 transduced with the indicated shRNA and treated with or without 20 μmol/L SP600125 (SP) for 4 hours (n = 3). G, MCF7 cells transduced with the indicated lentiviral vectors observed under phase contrast or fluorescence microscope. Scale bar, 100 μm. H, qRT-PCR of the indicated gene in MCF7 cells transduced with the indicated shRNA and the indicated lentiviral expression vector (n = 3). DN, dominant negative c-Jun.

Close modal

To further understand whether the induction of CCL2 and CCL5 following RB depletion depends on MS, we first analyzed the association between CCL2 or CCL5 expression and MS production in a variety of breast cancer cell lines, noting that CCL2, but not CCL5, showed a significant positive correlation with MS production (Fig. 5B). Moreover, consistent with our previous study (17), MS production was significantly upregulated following RB depletion in multiple breast cancer cell lines except MDA-MB-231 (Fig. 5C) in which CCL2 was not upregulated by RB depletion (Fig. 4B). To examine whether higher MS production in RB-depleted cells directly contributes to higher CCL2 production, we treated MCF7 cells with several antioxidants such as N-acetyl cysteine (NAC), Trolox, and mitochondria-targeted antioxidant MitoQ, and found that CCL2 production was, at least in part, dependent on MS (Fig. 5D). However, CCL5 induction following RB depletion was not antagonized upon treatment with antioxidants (Supplementary Fig. S2C). In addition, treatment with trifluoromethoxy carbonylcyanide phenylhydrazone (FCCP), a potent uncoupler of oxidative phosphorylation in mitochondria known to induce MS production, strongly induced CCL2, but not CCL5 expression, to the same degree as RB depletion in MCF7 cells (Fig. 5E; Supplementary Fig. S2D). Collectively, these data suggest that enhanced MS production following RB inactivation induces CCL2 expression. Finally, as we previously reported that enhanced cytokine secretion following RB inactivation was mediated by JNK activation, we verified the JNK dependency of CCL2 induction by treatment with JNK inhibitor SP600125 (Fig. 5F) or introduction of a dominant negative form of c-JUN (Fig. 5G and H).

Ccr2−/− background antagonizes in vivo mammary carcinogenesis induced by Rb deficiency

To examine whether the RB–CCL2 axis is involved in carcinogenesis induced by RB deficiency, we analyzed mouse mammary carcinogenesis in vivo using MMTV-Cre;Rbflox/flox mice with various Ccr2 genetic backgrounds, including Ccr2+/+, Ccr2+/−, and Ccr2−/−. Previously, several groups have reported that MMTV-Cre;Rbflox/flox female mice develop focal hyperplastic lesions in the mammary glands. (29, 30). As expected, the percentage of PCNA-positive cells in the mammary glands of MMTV-Cre;Rbflox/flox; Ccr2+/+ nulliparous mice (45.3 ± 13.3%) at an average age of examination of 429 ± 13.2 days was dramatically higher than that of wild-type C57BL/6 mice at a similar age (6.5 ± 3.0%). Importantly, the percentage of PCNA-positive cells in mammary glands was lower in Ccr2± (17.8 ± 2.0%) and Ccr2−/− (4.5 ± 5.0%) backgrounds at a similar age (Fig. 6A; Supplementary Fig. S2E). Overall survival did not show statistically significant differences among Ccr2+/+, Ccr2±, and Ccr2−/− backgrounds (Supplementary Fig. S2F). We did not observe palpable mammary tumors in mice with any background. Upon autopsy, we frequently observed lymphoma (Supplementary Fig. S2G), which may explain relatively shorter survival of MMTV-Cre;Rbflox/flox mice (around 16 months). Although not frequently, we observed hepatic and thyroid tumors (Supplementary Fig. S2H and SI). These findings are consistent with nonmammary tissue-specific activation of the MMTV promoter (31).

Figure 6.

Activation of Ccl2–Ccr2 pathway is required for mammary carcinogenesis induced by Rb deficiency. A, IHC analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. PCNA-positive cells in mammary glands were quantified (n > 3). Insets show higher magnification of mammary gland. B, Representative pictures of whole-mount carmine alum staining of MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (> 400 days). Scale bar, 5 mm (left), 500 μm (middle), and 100 μm (right). LN, lymph node. C, IHC analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. CCL2-positive area in mammary glands was quantified (n > 7). Insets show higher magnification of mammary gland. D, Immunofluorescence analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. F4/80+ cells in mammary glands were quantified (n > 3). Insets show higher magnification of mammary gland. n.s., not significant; **, P < 0.01.

Figure 6.

Activation of Ccl2–Ccr2 pathway is required for mammary carcinogenesis induced by Rb deficiency. A, IHC analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. PCNA-positive cells in mammary glands were quantified (n > 3). Insets show higher magnification of mammary gland. B, Representative pictures of whole-mount carmine alum staining of MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (> 400 days). Scale bar, 5 mm (left), 500 μm (middle), and 100 μm (right). LN, lymph node. C, IHC analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. CCL2-positive area in mammary glands was quantified (n > 7). Insets show higher magnification of mammary gland. D, Immunofluorescence analysis of the indicated protein in MMTV-Cre; Rbf/f; Ccr2 WT or KO, or C57BL/6 (WT) mammary glands (>400 days). Scale bar, 50 μm. F4/80+ cells in mammary glands were quantified (n > 3). Insets show higher magnification of mammary gland. n.s., not significant; **, P < 0.01.

Close modal

Consistent with higher PCNA signal, MMTV-Cre;Rbflox/flox;Ccr2+/+ nulliparous mice mammary glands examined at 436 ± 40.2 days frequently exhibited hyperplastic features upon whole-mount carmine alum staining and hematoxylin and eosin staining, whereas MMTV-Cre;Rbflox/flox;Ccr2−/− mammary glands at a similar age exhibited morphologies reminiscent of those of wild-type C57BL/6 mice at a similar age (Fig. 6B; Supplementary Fig. S2J). More accurately, we examined mammary gland of 14 MMTV-Cre;Rbflox/flox;Ccr2+/+ mice, and 11 of them (78.6%) showed hyperplastic phenotype. However, in 22 MMTV-Cre;Rbflox/flox;Ccr2−/− mice, we observed no (0%) hyperplastic phenotype. Furthermore, the infiltration of F4/80+ macrophages into mammary glands was significantly upregulated in MMTV-Cre;Rbflox/flox;Ccr2+/+ but not in the Ccr2−/− background, even though CCL2 expression in mammary glands was upregulated following RB deletion both in Ccr2+/+ and Ccr2−/− backgrounds (Fig. 6C and D). Taken together, these data suggest that activation of the Ccl2–Ccr2 pathway in mammary glands via enhanced Ccl2 secretion is required for carcinogenesis induced by Rb deficiency.

Here, we provide novel evidence indicating that RB inactivation in tumor cells results in the formation of a protumoral microenvironment by promoting angiogenesis and recruitment of TAMs or immunosuppressive cells such as MDSCs into the TME. By using a syngeneic and orthotropic murine soft-tissue sarcoma model, and confirming these findings in a second murine mammary carcinogenesis model, we demonstrated that CCL2 induction following RB inactivation in tumor cells and subsequent activation of the CCL2–CCR2 axis in the TME accelerates tumor progression. CCL2 has been shown to play a critical role in tumor progression in various cancer types including breast cancer via macrophage recruitment into the TME (32, 33). Macrophages in the TME support tumor growth via multiple mechanisms, including the secretion of growth factors and the promotion of angiogenesis (34). In addition to the proliferative advantage imparted to tumors, elevated CCL2 secretion contributes to the formation of an immunosuppressive TME via MDSCs and Tregs recruitment, and results in the evasion of cytotoxic T cells (35). Despite many studies that identify CCL2 as a protumorigenic chemokine, the therapeutic effect of blocking the CCL2–CCR2 axis via treatment with a neutralizing antibody, for example, has been disappointing in clinical trials (36–38). Thus, to achieve the therapeutic benefits of CCL2-CCR2 blockade, we must understand the regulatory mechanisms of CCL2 and determine which patient subtypes respond to this therapy (i.e., which tumor mutations and gene expression signatures are susceptible to CCL2 blockade).

According to our findings that demonstrate the functional relevance between the RB and CCL2–CCR2 axis, aberrant RB expression in certain tumor types might act as a possible marker for the development of an effective therapy by CCL2-CCR2 blockade. Currently, it is thought that the CCL2-CCR2 blockade exhibits tumor-suppressive function, at least in part, by enhancing the antitumoral function of the TME via inhibition of the infiltration of immunosuppressive cells (1). TNBC is a highly heterogeneous subtype compared with others such as ER-positive or HER2-positive breast cancer (39). RB inactivation by genetic and epigenetic factors and significantly higher CCL2 levels is frequently found in this aggressive subtype (40, 41). Although TNBC is sensitive to chemotherapy, the overall outcomes of TNBC are worse in patients with breast cancer because of the lack of targetable molecules such as ER and HER2 (39). Thus, immunotherapy including ICB treatment is emerging as a promising new option for patients with TNBC, but there are no established prognosis markers to estimate its efficacy. Our current study revealed that RB inactivation via genetic mutation or transcriptional suppression via DNA hypermethylation in the RB gene promoter in patients with TNBC could be a potential marker for both CCL2-CCR2 blockade efficacy and poor immunogenicity in TNBC, and anti-CCL2 treatment might enhance therapeutic effect of ICB treatment. However, it is now becoming clear that the accumulation of DNA damage in tumors resulting from defects in the DNA repair pathway, DNA-damaging chemotherapy, and/or radiotherapy is associated with immunogenic cell death and neoantigen production, promoting an antitumor immune response (42–44). It is possible that increased genomic instability and subsequent responses to DNA damage in RB-inactivated cancer cells might potentiate the efficacy of immunotherapy, but further research is needed to uncover how RB inactivation alters tumor immunogenicity in both cell-autonomous and non–cell-autonomous manners.

Tumor angiogenesis in the TME is crucial for tumor progression (26). By using a syngeneic and orthotropic murine soft-tissue sarcoma model with a Ccr2-null background, we demonstrated that Rb inactivation in cancer cells promotes aberrant angiogenesis through the activation of the Ccl2–Ccr2 axis in the TME. In particular, RB inactivation–dependent infiltration of TAMs into the TME is clearly suppressed in Ccr2 KO mice. TAMs secrete high levels of angiogenic factors including Vegfα, leading to neovascularization in the TME. Because Rb inactivation also induces Vegfα secretion from tumor cells themselves (24), RB might regulate tumor angiogenesis via both cell-intrinsic and cell-extrinsic mechanisms.

In our previous study (17), we partially clarified the mechanism whereby RB inactivation leads to increased production of IL6 via increased FAO activity and MS production. We disclosed that RB inactivation increased the expression of a number of genes involved in FAO in an E2F-dependent manner, therefore increased oxidative metabolism leads to increased MS production and JNK activation. JNK activation is critical for IL6 secretion. In addition, we demonstrated that malonyl-CoA level drops following RB inactivation. This finding is the most consistent with increased FAO, because malonyl-CoA is the strongest suppressor of FAO (17). However, in the previous work, we did not clarify why malonyl-CoA level drops following RB inactivation. In this work, we provided evidence that increased CCL2 secretion following RB inactivation is due to elevated FAO activity and MS production similar to in the case of IL6. Moreover, we linked RB inactivation to downregulation of malonyl-CoA by AMPK and ACC. RB inactivation dramatically increased AMPK phosphorylation (Fig. 5A). Phosphorylated AMPK phosphorylates ACC. Phosphorylated ACC loses its activity to synthesize malonyl-CoA from acetyl-CoA. Decrease in malonyl-CoA allows CPT1 to transport long-chain fatty acids into mitochondria for FAO. Why RB loss increases AMPK phosphorylation is currently under investigation. We are determining ATP/AMP and NAD+/NADH ratio in cells before and after RB depletion (45). In addition, RB has been suggested to be involved in various facets of cellular metabolism (46). Further study would be necessary to thoroughly determine the mechanism of RB–AMPK axis.

No potential conflicts of interest were disclosed.

Conception and design: F. Li, S. Kitajima, C. Takahashi

Development of methodology: F. Li, S. Kohno, N. Okada, C. Takahashi

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): F. Li, S. Kitajima, S. Kohno, A. Yoshida, S. Tange, S. Sasaki, Y. Nishimoto, H. Muranaka, N. Nagatani, M. Suzuki, T. Nishiuchi, T. Tanaka, N. Mukaida

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): F. Li, S. Kitajima, S. Kohno, S. Tange, T. Tanaka, C. Takahashi

Writing, review, and/or revision of the manuscript: F. Li, S. Kitajima, H. Muranaka, T. Tanaka, D.A. Barbie, N. Mukaida, C. Takahashi

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): S. Kitajima, N. Okada, H. Muranaka, S. Masuda, T.C. Thai, C. Takahashi

Study supervision: S. Kitajima, C. Takahashi

We thank Dr. T. Baba for technical instruction and useful discussion, Dr. N. Mahadevan for pathologic diagnosis, and Mr. S. Sundararaman for critical reading of the manuscript. This work was supported by Funding Program for Next Generation World-Leading Researchers LS049 (to C. Takahashi), Grant-in-Aid for Scientific Research on Innovative Areas 15H01487 and 17H05615 (to C. Takahashi), Grant-in-Aid for Scientific Research 17H03576 (to C. Takahashi) and 25830077 (to S. Kitajima), Hokuriku Bank Research Grant for Young Scientist (to S Kitajima), the Uehara Memorial Foundation Post-Doctoral Fellowship (to S. Kitajima), the Strategic Young Researcher Overseas Visit Program for Accelerating Brain Circulation (to S. Kitajima), and JSPS Postdoctoral Fellowship for Research Abroad (to S. Kitajima).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Binnewies
M
,
Roberts
EW
,
Kersten
K
,
Chan
V
,
Fearon
DF
,
Merad
M
, et al
Understanding the tumor immune microenvironment (TIME) for effective therapy
.
Nat Med
2018
;
24
:
541
50
.
2.
Wei
SC
,
Duffy
CR
,
Allison
JP
. 
Fundamental mechanisms of immune checkpoint blockade therapy
.
Cancer Discov
2018
;
8
:
1069
86
.
3.
Li
Z
,
Qiu
Y
,
Lu
W
,
Jiang
Y
,
Wang
J
. 
Immunotherapeutic interventions of triple negative breast cancer
.
J Transl Med
2018
;
16
:
147
.
4.
Manguso
RT
,
Pope
HW
,
Zimmer
MD
,
Brown
FD
,
Yates
KB
,
Miller
BC
, et al
In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target
.
Nature
2017
;
547
:
413
8
.
5.
Pan
D
,
Kobayashi
A
,
Jiang
P
,
Ferrari de Andrade
L
,
Tay
RE
,
Luoma
AM
, et al
A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing
.
Science
2018
;
359
:
770
5
.
6.
Miao
D
,
Margolis
CA
,
Gao
W
,
Voss
MH
,
Li
W
,
Martini
DJ
, et al
Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma
.
Science
2018
;
359
:
801
6
.
7.
Shin
DS
,
Zaretsky
JM
,
Escuin-Ordinas
H
,
Garcia-Diaz
A
,
Hu-Lieskovan
S
,
Kalbasi
A
, et al
Primary resistance to PD-1 blockade mediated by JAK1/2 mutations
.
Cancer Discov
2017
;
7
:
188
201
.
8.
Zaretsky
JM
,
Garcia-Diaz
A
,
Shin
DS
,
Escuin-Ordinas
H
,
Hugo
W
,
Hu-Lieskovan
S
, et al
Mutations associated with acquired resistance to PD-1 blockade in melanoma
.
N Engl J Med
2016
;
375
:
819
29
.
9.
Patel
SJ
,
Sanjana
NE
,
Kishton
RJ
,
Eidizadeh
A
,
Vodnala
SK
,
Cam
M
, et al
Identification of essential genes for cancer immunotherapy
.
Nature
2017
;
548
:
537
42
.
10.
Spranger
S
,
Gajewski
TF.
Impact of oncogenic pathways on evasion of antitumour immune responses
.
Nat Rev Cancer
2018
;
18
:
139
47
.
11.
Sumimoto
H
,
Imabayashi
F
,
Iwata
T
,
Kawakami
Y
. 
The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells
.
J Exp Med
2006
;
203
:
1651
6
.
12.
Pylayeva-Gupta
Y
,
Lee
KE
,
Hajdu
CH
,
Miller
G
,
Bar-Sagi
D
. 
Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia
.
Cancer Cell
2012
;
21
:
836
47
.
13.
Bayne
LJ
,
Beatty
GL
,
Jhala
N
,
Clark
CE
,
Rhim
AD
,
Stanger
BZ
, et al
Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer
.
Cancer Cell
2012
;
21
:
822
35
.
14.
Koyama
S
,
Akbay
EA
,
Li
YY
,
Aref
AR
,
Skoulidis
F
,
Herter-Sprie
GS
, et al
STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T-cell activity in the lung tumor microenvironment
.
Cancer Res
2016
;
76
:
999
1008
.
15.
Peng
W
,
Chen
JQ
,
Liu
C
,
Malu
S
,
Creasy
C
,
Tetzlaff
MT
, et al
Loss of PTEN promotes resistance to T cell-mediated immunotherapy
.
Cancer Discov
2016
;
6
:
202
16
.
16.
Yoshida
A
,
Kitajima
S
,
Li
F
,
Cheng
C
,
Takegami
Y
,
Kohno
S
, et al
MicroRNA-140 mediates RB tumor suppressor function to control stem cell-like activity through interleukin-6
.
Oncotarget
2017
;
8
:
13872
85
.
17.
Kitajima
S
,
Yoshida
A
,
Kohno
S
,
Li
F
,
Suzuki
S
,
Nagatani
N
, et al
The RB-IL-6 axis controls self-renewal and endocrine therapy resistance by fine-tuning mitochondrial activity
.
Oncogene
2017
;
36
:
5145
57
.
18.
Tsukada
T
,
Tomooka
Y
,
Takai
S
,
Ueda
Y
,
Nishikawa
S
,
Yagi
T
, et al
Enhanced proliferative potential in culture of cells from p53-deficient mice
.
Oncogene
1993
;
8
:
3313
22
.
19.
Kuziel
WA
,
Morgan
SJ
,
Dawson
TC
,
Griffin
S
,
Smithies
O
,
Ley
K
, et al
Severe reduction in leukocyte adhesion and monocyte extravasation in mice deficient in CC chemokine receptor 2
.
Proc Natl Acad Sci U S A
1997
;
94
:
12053
8
.
20.
Hinohara
K
,
Kobayashi
S
,
Kanauchi
H
,
Shimizu
S
,
Nishioka
K
,
Tsuji
E
, et al
ErbB receptor tyrosine kinase/NF-kappaB signaling controls mammosphere formation in human breast cancer
.
Proc Natl Acad Sci U S A
2012
;
109
:
6584
9
.
21.
Sato
T
,
Ishikawa
M
,
Mochizuki
M
,
Ohta
M
,
Ohkura
M
,
Nakai
J
, et al
JSAP1/JIP3 and JLP regulate kinesin-1-dependent axonal transport to prevent neuronal degeneration
.
Cell Death Differ
2015
;
22
:
1260
74
.
22.
Kanai
A
,
Suzuki
K
,
Tanimoto
K
,
Mizushima-Sugano
J
,
Suzuki
Y
,
Sugano
S
. 
Characterization of STAT6 target genes in human B cells and lung epithelial cells
.
DNA Res
2011
;
18
:
379
92
.
23.
Kitajima
S
,
Miki
T
,
Takegami
Y
,
Kido
Y
,
Noda
M
,
Hara
E
, et al
Reversion-inducing cysteine-rich protein with Kazal motifs interferes with epidermal growth factor receptor signaling
.
Oncogene
2011
;
30
:
737
50
.
24.
Lasorella
A
,
Rothschild
G
,
Yokota
Y
,
Russell
RG
,
Iavarone
A
. 
Id2 mediates tumor initiation, proliferation, and angiogenesis in Rb mutant mice
.
Mol Cell Biol
2005
;
25
:
3563
74
.
25.
Kumar
V
,
Patel
S
,
Tcyganov
E
,
Gabrilovich
DI
. 
The nature of myeloid-derived suppressor cells in the tumor microenvironment
.
Trends Immunol
2016
;
37
:
208
20
.
26.
De Palma
M
,
Biziato
D
,
Petrova
TV
. 
Microenvironmental regulation of tumour angiogenesis
.
Nat Rev Cancer
2017
;
17
:
457
74
.
27.
Ciriello
G
,
Gatza
ML
,
Beck
AH
,
Wilkerson
MD
,
Rhie
SK
,
Pastore
A
, et al
Comprehensive molecular portraits of invasive lobular breast cancer
.
Cell
2015
;
163
:
506
19
.
28.
Knudsen
ES
,
Wang
JY.
Dual mechanisms for the inhibition of E2F binding to RB by cyclin-dependent kinase-mediated RB phosphorylation
.
Mol Cell Biol
1997
;
17
:
5771
83
.
29.
Jiang
Z
,
Deng
T
,
Jones
R
,
Li
H
,
Herschkowitz
JI
,
Liu
JC
, et al
Rb deletion in mouse mammary progenitors induces luminal-B or basal-like/EMT tumor subtypes depending on p53 status
.
J Clin Invest
2010
;
120
:
3296
309
.
30.
Cheng
L
,
Zhou
Z
,
Flesken-Nikitin
A
,
Toshkov
IA
,
Wang
W
,
Camps
J
, et al
Rb inactivation accelerates neoplastic growth and substitutes for recurrent amplification of cIAP1, cIAP2 and Yap1 in sporadic mammary carcinoma associated with p53 deficiency
.
Oncogene
2010
;
29
:
5700
11
.
31.
Wagner
KU
,
McAllister
K
,
Ward
T
,
Davis
B
,
Wiseman
R
,
Hennighausen
L
. 
Spatial and temporal expression of the Cre gene under the control of the MMTV-LTR in different lines of transgenic mice
.
Transgenic Res
2001
;
10
:
545
53
.
32.
Qian
BZ
,
Li
J
,
Zhang
H
,
Kitamura
T
,
Zhang
J
,
Campion
LR
, et al
CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis
.
Nature
2011
;
475
:
222
5
.
33.
Lu
X
,
Kang
Y
. 
Chemokine (C-C motif) ligand 2 engages CCR2+ stromal cells of monocytic origin to promote breast cancer metastasis to lung and bone
.
J Biol Chem
2009
;
284
:
29087
96
.
34.
Mantovani
A
,
Marchesi
F
,
Malesci
A
,
Laghi
L
,
Allavena
P
. 
Tumour-associated macrophages as treatment targets in oncology
.
Nat Rev Clin Oncol
2017
;
14
:
399
416
.
35.
Chang
AL
,
Miska
J
,
Wainwright
DA
,
Dey
M
,
Rivetta
CV
,
Yu
D
, et al
CCL2 produced by the glioma microenvironment is essential for the recruitment of regulatory T cells and myeloid-derived suppressor cells
.
Cancer Res
2016
;
76
:
5671
82
.
36.
Sandhu
SK
,
Papadopoulos
K
,
Fong
PC
,
Patnaik
A
,
Messiou
C
,
Olmos
D
, et al
A first-in-human, first-in-class, phase I study of carlumab (CNTO 888), a human monoclonal antibody against CC-chemokine ligand 2 in patients with solid tumors
.
Cancer Chemother Pharmacol
2013
;
71
:
1041
50
.
37.
Vela
M
,
Aris
M
,
Llorente
M
,
Garcia-Sanz
JA
,
Kremer
L
. 
Chemokine receptor-specific antibodies in cancer immunotherapy: achievements and challenges
.
Front Immunol
2015
;
6
:
12
.
38.
Brana
I
,
Calles
A
,
LoRusso
PM
,
Yee
LK
,
Puchalski
TA
,
Seetharam
S
, et al
Carlumab, an anti-C-C chemokine ligand 2 monoclonal antibody, in combination with four chemotherapy regimens for the treatment of patients with solid tumors: an open-label, multicenter phase 1b study
.
Target Oncol
2015
;
10
:
111
23
.
39.
Bianchini
G
,
Balko
JM
,
Mayer
IA
,
Sanders
ME
,
Gianni
L
. 
Triple-negative breast cancer: challenges and opportunities of a heterogeneous disease
.
Nat Rev Clin Oncol
2016
;
13
:
674
90
.
40.
Cancer Genome Atlas
Network
. 
Comprehensive molecular portraits of human breast tumours
.
Nature
2012
;
490
:
61
70
.
41.
Dutta
P
,
Sarkissyan
M
,
Paico
K
,
Wu
Y
,
Vadgama
JV
. 
MCP-1 is overexpressed in triple-negative breast cancers and drives cancer invasiveness and metastasis
.
Breast Cancer Res Treat
2018
;
170
:
477
86
.
42.
Brown
JS
,
Sundar
R
,
Lopez
J
. 
Combining DNA damaging therapeutics with immunotherapy: more haste, less speed
.
Br J Cancer
2018
;
118
:
312
24
.
43.
Patel
SA
,
Minn
AJ.
Combination cancer therapy with immune checkpoint blockade: mechanisms and strategies
.
Immunity
2018
;
48
:
417
33
.
44.
Gotwals
P
,
Cameron
S
,
Cipolletta
D
,
Cremasco
V
,
Crystal
A
,
Hewes
B
, et al
Prospects for combining targeted and conventional cancer therapy with immunotherapy
.
Nat Rev Cancer
2017
;
17
:
286
301
.
45.
Lan
F
,
Cacicedo
JM
,
Ruderman
N
,
Ido
Y
. 
SIRT1 modulation of the acetylation status, cytosolic localization, and activity of LKB1. Possible role in AMP-activated protein kinase activation
.
J Biol Chem
2008
;
283
:
27628
35
.
46.
Kitajima
S
,
Takahashi
C.
Intersection of retinoblastoma tumor suppressor function, stem cells, metabolism, and inflammation
.
Cancer Sci
2017
;
108
:
1726
31
.