Enhanced Lipid Accumulation and Metabolism Are Required for the Differentiation and Activation of Tumor-Associated Macrophages

Pan Su, Qiang Wang, Enguang Bi, Xingzhe Ma, Lintao Liu, Maojie Yang, Jianfei Qian, Qing Yi, Pan Su, Qiang Wang, Enguang Bi, Xingzhe Ma, Lintao Liu, Maojie Yang, Jianfei Qian, Qing Yi

Abstract

Tumor-associated macrophages (TAM) are important tumor-promoting cells. However, the mechanisms underlying how the tumor and its microenvironment reprogram these cells remain elusive. Here we report that lipids play a crucial role in generating TAMs in the tumor microenvironment (TME). Macrophages from both human and murine tumor tissues were enriched with lipids due to increased lipid uptake by macrophages. TAMs expressed elevated levels of the scavenger receptor CD36, accumulated lipids, and used fatty acid oxidation (FAO) instead of glycolysis for energy. High levels of FAO promoted mitochondrial oxidative phosphorylation, production of reactive oxygen species, phosphorylation of JAK1, and dephosphorylation of SHP1, leading to STAT6 activation and transcription of genes that regulate TAM generation and function. These processes were critical for TAM polarization and activity, both in vitro and in vivo. In summary, we highlight the importance of lipid metabolism in the differentiation and function of protumor TAMs in the TME. SIGNIFICANCE: This study highlights the role of lipid metabolism in the differentiation and function of TAMs and suggests targeting TAM fatty acid oxidation as a potential therapeutic modality for human cancers.

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

©2020 American Association for Cancer Research.

Figures

Fig. 1.. Lipid accumulation in TAMs
Fig. 1.. Lipid accumulation in TAMs
A-B, Representative fluorescent confocal images showing the morphology and lipid levels of human MΦs sorted from BM samples of normal donors (A) and MM patients (B). TD represents contrast image by transmitted light differential interference. C-E, Representative images of lipid levels and CD68 expression in human frozen sections of breast, colon, and prostate cancer and normal tissues. F, Representative histogram showing the levels of lipids in CD11b+CD68+ MΦs from human colon normal and cancer tissues (left panel). Right panel showing the statistic result of the mean fluorescence intensity (MFI) of lipids in MΦs from normal and tumor tissues. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001. G, Representative histograms (left panel) and quantitative result (right panel) of the lipid levels in MΦs from human normal and melanoma tissues. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001. H, Bar graph representing the lipid levels in CD11b+CD68+ MΦs from normal, MGUS, and MM BM samples. Statistical significance was determined by two-tailed Student’s t test between normal and disease group. **, P < 0.01; ***, P < 0.001. I, Statistic results of lipid levels in F4/80+CD11b+ MΦs of tumor tissues and their corresponding normal tissues from indicated tumor-bearing (TB) or normal mice. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001. J-K, Bar graphs depicting the MFI of lipids in MΦs cultured with or without indicated tumor cells (J) or TCM (K). Statistical significance was determined by two-tailed Student’s t test between control MΦs and different TAMs. **, P < 0.01; ***, P < 0.001. L-M, Representative confocal images of lipid droplet in MΦs cultured with tumor cells (L) or TCM (M).
Fig. 2.. Blocking CD36 abolishes lipid accumulation…
Fig. 2.. Blocking CD36 abolishes lipid accumulation in TAMs
A, Representative fluorescent confocal images showing Fatty Acid-Red C12 uptake by human control MΦs and TAMs. B, Bar graph depicting ELISA results of fatty acid uptake by control MΦs and TAMs using a Free Fatty Acid Uptake Assay kit. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001. C, Heat map showing the relative expression of human scavenger receptors in TAMs compared with control MΦs. D, Representative histograms (left) and statistic results (right) showing lipids in MΦs and TAMs with indicated blocking antibody treatments. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. **, P < 0.01; ***, P < 0.001; n.s., not significant. E-F, Representative histograms (left) and statistic results (right) showing lipid levels in control MΦs and indicated TAMs cultured with or without SSO. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001. G-H, Representative confocal images of lipids of murine MΦs from CD36 WT and KO mice cultured with or without 5TGM1 (G) or EL4 (H) cells in vitro.
Fig. 3.. Culture with tumor cells increases…
Fig. 3.. Culture with tumor cells increases the fatty acid oxidation in TAMs
A-B, Microarray analysis of genes related to lipid β-oxidation in control MΦs and TAMs. Pie charts showing the percentage of total gene changes (A) in each indicated group of cells. Heat map showing the relative expression of lipid β-oxidation-associated genes (B) in control MΦs and TAMs. C, Relative mRNA expression of indicated genes related to lipid β-oxidation in control MΦs and TAMs. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001; n.s., not significant. D-E, Oxygen consumption rates (OCRs) of control MΦs, TAMs and etomoxir treated TAMs by Seahorse XF Analyzer (D). Statistic results depicting the basal and maximal respiration and spare respiratory capacity in control MΦs and TAMs by analyzing the OCRs (E). Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. *, P < 0.05; **, P < 0.01; ***, P < 0.001. F, Relative mRNA expression of indicated genes in control MΦs and TAMs from CD36 WT and KO mice using qPCR. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. **, P < 0.01; ***, P < 0.001; n.s., not significant. G-H, OCRs of control MΦs and TAMs from CD36 WT and KO mice (G) and statistic results (H) depicting the basal and maximal respiration and spare respiratory capacity. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. *, P < 0.05; **, P < 0.01; ***, P < 0.001, n.s., not significant.
Fig. 4.. Fatty acid oxidation promotes TAM…
Fig. 4.. Fatty acid oxidation promotes TAM differentiation
A, Heat map showing the relative expression of TAM signature genes in control MΦs and TAMs. B, Relative mRNA expression of TAM signature genes in control MΦs and TAMs by qPCR. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. *, P < 0.05; ***, P < 0.001, n.s., not significant. C, Relative mRNA expression of indicated genes in the MΦs with indicated treatment. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. **, P < 0.01; ***, P < 0.001, n.s., not significant. D, Venn diagram showing the changes in gene expression in control MΦs and TAMs under various conditions. Arg1 and Ccl2 were in the intersection between these three gene sets. E-F, Relative mRNA expression (e) and protein levels (f) of Ccl2 and Arg1 in control MΦs and TAMs from CD36 WT and KO mice. Statistical significance was determined by two-tailed Student’s t test between control MΦs and TAMs. ***, P < 0.001, n.s., not significant.
Fig. 5.. Inhibition of fatty acid oxidation…
Fig. 5.. Inhibition of fatty acid oxidation impairs protumor function of TAMs
A-D, Representative histograms (left) and statistic results (right) of the expression of Ki-67 in human U266 (A) and ARP1 (B) myeloma cells, LNCap-LN3 prostate tumor cells (C), and MDA-MB-468 breast tumor cells (D), cultured with control MΦs or TAMs in the presence or absence of etomoxir. Statistical significance was determined by two-tailed Student’s t test between indicated groups. **, P < 0.01; ***, P < 0.001. E, Representative dot plots showing cell cycle progression of tumor cells with indicated coculture conditions using APC-BrdU and 7-AAD. F, Bar graphs showing the percentage of human U266, ARP1, LNCaP-LN3, and MDA-MB-468 cells in the indicated cell cycle stages under indicated coculture conditions. Statistical significance was determined by two-tailed Student’s t test between indicated groups. *, P < 0.05; **, P < 0.01; ***, P < 0.001. G-H, Representative histograms (G) and statistic results (H) showing the expression of Ki-67 in 5TGM1 cells cultured with control MΦs or TAMs from CD36 WT and KO mice. Statistical significance was determined by two-tailed Student’s t test between indicated groups. ***, P < 0.001; n.s., not significant.
Fig. 6.. High level of fatty acid…
Fig. 6.. High level of fatty acid oxidation enhances the phosphorylation of STAT6
A, IPA analysis of canonical signaling pathways in control MΦs and TAMs. The circle surface area is proportional to −log (P-value) and the color intensity of circles indicates the Z score. B, Representative western blots (upper) of 3 independent experiments and statistic result (lower) showing STAT6 phosphorylation in control MΦs and TAMs. Statistical significance was determined by two-tailed Student’s t test between indicated groups. ***, P < 0.001. C, Representative histograms showing the levels of superoxide determined by Mitosox Red (upper) and cellular ROS by DCFDA (lower) in control MΦs and TAMs. D, Representative immunoblots of JAK1 and JAK3 phosphorylation in control MΦs and TAMs. E, Representative immunoblots (lower) and quantitative result (upper) of the SHP1 phosphorylation in control MΦs and TAMs. Statistical significance was determined by two-tailed Student’s t test between indicated groups. ***, P < 0.001. F, immunoblots of the phosphorylated and total JAK1, STAT6, and SHP1 in MΦs with indicated treatment. G, Western blots of STAT6 and SHP1 phosphorylation in control MΦs and TAMs from CD36 WT and KO mice. Bar graph showing means ± SEM of 3 independent experiments. Statistical significance was determined by two-tailed Student’s t test between indicated groups. ***, P < 0.001, n.s., not significant.
Fig. 7.. Effect of fatty acid oxidation…
Fig. 7.. Effect of fatty acid oxidation inhibition on protumor function of TAMs
A, SPEP was performed on a representative Vk*MYC-bearing CD36 WT and KO mice. Arrows indicate M protein in Vk*MYC-bearing mice. B, Survival curves of CD36 WT and KO mice bearing Vk*MYC myeloma cells. The results were analyzed by Log-rank (Mantel-Cox) test (**, P < 0.01). C, EL4 lymphoma cells were mixed with or without CD36 WT or KO TAMs at a ratio of 5:1, and then injected subcutaneously into C57BL/6J mice. Tumor growth was monitored for 7 days. Results of tumor volumes are shown as means ± SEM. (* P < 0.05) D, EL4-bearing NSG mice were treated with 50 mg/kg etomoxir intraperitoneally every two days after tumor injection. Tumor growth was followed for 9 days. Results of tumor volumes were shown as means ± SEM. (* P < 0.05) E, Bioluminescent images of 5TGM1-luc myeloma cell-bearing NSG mice treated with 50 mg/kg etomoxir or vehicle on day 21. F, Tumor burden measured as serum concentration of IgG2b in 5TGM1-bearing mice treated with etomoxir or vehicle. Results are shown as means ± SEM. **, P < 0.01. G-H, Bioluminescent images (G) and statistic results of bioluminescent signals (H) of human MM.1S-luc cell-bearing NSG mice on day 21. Results are presented as means ± SEM. **, P < 0.01.

References

    1. Gordon S, Pluddemann A, Martinez Estrada F. Macrophage heterogeneity in tissues: phenotypic diversity and functions. Immunological reviews 2014;262:36–55
    1. Ye H, Zhou Q, Zheng S, Li G, Lin Q, Wei L, et al. Tumor-associated macrophages promote progression and the Warburg effect via CCL18/NF-kB/VCAM-1 pathway in pancreatic ductal adenocarcinoma. Cell death & disease 2018;9:453.
    1. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 2014;6:1670–90
    1. Wang Q, Lu Y, Li R, Jiang Y, Zheng Y, Qian J, et al. Therapeutic effects of CSF1R-blocking antibodies in multiple myeloma. Leukemia 2018;32:176–83
    1. Yang C, He L, He P, Liu Y, Wang W, He Y, et al. Increased drug resistance in breast cancer by tumor-associated macrophages through IL-10/STAT3/bcl-2 signaling pathway. Med Oncol 2015;32:352.
    1. Zheng Y, Wang Q, Li T, Qian J, Lu Y, Li Y, et al. Role of Myeloma-Derived MIF in Myeloma Cell Adhesion to Bone Marrow and Chemotherapy Response. Journal of the National Cancer Institute 2016;108
    1. Georgoudaki AM, Prokopec KE, Boura VF, Hellqvist E, Sohn S, Ostling J, et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell reports 2016;15:2000–11
    1. Mantovani A, Locati M. Macrophage Metabolism Shapes Angiogenesis in Tumors. Cell metabolism 2016;24:653–4
    1. Osterborg A, Yi Q, Henriksson L, Fagerberg J, Bergenbrant S, Jeddi-Tehrani M, et al. Idiotype immunization combined with granulocyte-macrophage colony-stimulating factor in myeloma patients induced type I, major histocompatibility complex-restricted, CD8- and CD4-specific T-cell responses. Blood 1998;91:2459–66
    1. Yan H, Dong M, Liu X, Shen Q, He D, Huang X, et al. Multiple myeloma cell-derived IL-32gamma increases the immunosuppressive function of macrophages by promoting indoleamine 2,3-dioxygenase (IDO) expression. Cancer letters 2019;446:38–48
    1. Petty AJ, Yang Y. Tumor-associated macrophages: implications in cancer immunotherapy. Immunotherapy 2017;9:289–302
    1. Cha HR, Lee JH, Hensel JA, Sawant AB, Davis BH, Lee CM, et al. Prostate cancer-derived cathelicidin-related antimicrobial peptide facilitates macrophage differentiation and polarization of immature myeloid progenitors to protumorigenic macrophages. The Prostate 2016;76:624–36
    1. Henze AT, Mazzone M. The impact of hypoxia on tumor-associated macrophages. The Journal of clinical investigation 2016;126:3672–9
    1. Laoui D, Van Overmeire E, Di Conza G, Aldeni C, Keirsse J, Morias Y, et al. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer research 2014;74:24–30
    1. Osinsky S, Bubnovskaya L, Ganusevich I, Kovelskaya A, Gumenyuk L, Olijnichenko G, et al. Hypoxia, tumour-associated macrophages, microvessel density, VEGF and matrix metalloproteinases in human gastric cancer: interaction and impact on survival. Clinical & translational oncology : official publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico 2011;13:133–8
    1. Vatolin S, Phillips JG, Jha BK, Govindgari S, Hu J, Grabowski D, et al. Novel Protein Disulfide Isomerase Inhibitor with Anticancer Activity in Multiple Myeloma. Cancer research 2016;76:3340–50
    1. Cassol E, Cassetta L, Rizzi C, Alfano M, Poli G. M1 and M2a polarization of human monocyte-derived macrophages inhibits HIV-1 replication by distinct mechanisms. J Immunol 2009;182:6237–46
    1. Li Y, Zheng Y, Li T, Wang Q, Qian J, Lu Y, et al. Chemokines CCL2, 3, 14 stimulate macrophage bone marrow homing, proliferation, and polarization in multiple myeloma. Oncotarget 2015;6:24218–29
    1. Wang Q, Zhang S, Jiang H, Wang J, Weng L, Mao Y, et al. PA from an H5N1 highly pathogenic avian influenza virus activates viral transcription and replication and induces apoptosis and interferon expression at an early stage of infection. Virology journal 2012;9:106.
    1. Zheng Y, Yang J, Qian J, Qiu P, Hanabuchi S, Lu Y, et al. PSGL-1/selectin and ICAM-1/CD18 interactions are involved in macrophage-induced drug resistance in myeloma. Leukemia 2013;27:702–10
    1. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P. Tumour-associated macrophages as treatment targets in oncology. Nature reviews Clinical oncology 2017;14:399–416
    1. Wang N, Liang H, Zen K. Molecular mechanisms that influence the macrophage m1-m2 polarization balance. Frontiers in immunology 2014;5:614.
    1. Rahal OM, Wolfe AR, Mandal PK, Larson R, Tin S, Jimenez C, et al. Blocking Interleukin (IL)4- and IL13-Mediated Phosphorylation of STAT6 (Tyr641) Decreases M2 Polarization of Macrophages and Protects Against Macrophage-Mediated Radioresistance of Inflammatory Breast Cancer. International journal of radiation oncology, biology, physics 2018;100:1034–43
    1. Johnson DJ, Pao LI, Dhanji S, Murakami K, Ohashi PS, Neel BG. Shp1 regulates T cell homeostasis by limiting IL-4 signals. The Journal of experimental medicine 2013;210:1419–31
    1. Lambrechts D, Wauters E, Boeckx B, Aibar S, Nittner D, Burton O, et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nature medicine 2018;24:1277–89
    1. Azizi E, Carr AJ, Plitas G, Cornish AE, Konopacki C, Prabhakaran S, et al. Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment. Cell 2018;174:1293–308 e36
    1. Jones J, Otu H, Spentzos D, Kolia S, Inan M, Beecken WD, et al. Gene signatures of progression and metastasis in renal cell cancer. Clinical cancer research : an official journal of the American Association for Cancer Research 2005;11:5730–9
    1. Beroukhim R, Brunet JP, Di Napoli A, Mertz KD, Seeley A, Pires MM, et al. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer research 2009;69:4674–81
    1. Sun L, Hui AM, Su Q, Vortmeyer A, Kotliarov Y, Pastorino S, et al. Neuronal and glioma-derived stem cell factor induces angiogenesis within the brain. Cancer cell 2006;9:287–300
    1. Chesi M, Matthews GM, Garbitt VM, Palmer SE, Shortt J, Lefebure M, et al. Drug response in a genetically engineered mouse model of multiple myeloma is predictive of clinical efficacy. Blood 2012;120:376–85
    1. Bi E, Li R, Bover LC, Li H, Su P, Ma X, et al. E-cadherin expression on multiple myeloma cells activates tumor-promoting properties in plasmacytoid DCs. The Journal of clinical investigation 2018;128:4821–31
    1. Herber DL, Cao W, Nefedova Y, Novitskiy SV, Nagaraj S, Tyurin VA, et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nature medicine 2010;16:880–6
    1. Hossain F, Al-Khami AA, Wyczechowska D, Hernandez C, Zheng L, Reiss K, et al. Inhibition of Fatty Acid Oxidation Modulates Immunosuppressive Functions of Myeloid-Derived Suppressor Cells and Enhances Cancer Therapies. Cancer immunology research 2015;3:1236–47
    1. Freemerman AJ, Johnson AR, Sacks GN, Milner JJ, Kirk EL, Troester MA, et al. Metabolic reprogramming of macrophages: glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. The Journal of biological chemistry 2014;289:7884–96
    1. Huang SC, Everts B, Ivanova Y, O’Sullivan D, Nascimento M, Smith AM, et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nature immunology 2014;15:846–55
    1. Penny HL, Sieow JL, Adriani G, Yeap WH, See Chi Ee P, San Luis B, et al. Warburg metabolism in tumor-conditioned macrophages promotes metastasis in human pancreatic ductal adenocarcinoma. Oncoimmunology 2016;5:e1191731.
    1. Muller S, Kohanbash G, Liu SJ, Alvarado B, Carrera D, Bhaduri A, et al. Single-cell profiling of human gliomas reveals macrophage ontogeny as a basis for regional differences in macrophage activation in the tumor microenvironment. Genome biology 2017;18:234.
    1. Zhang Y, Sun Y, Rao E, Yan F, Li Q, Silverstein KA, et al. Fatty acid-binding protein E-FABP restricts tumor growth by promoting IFN-beta responses in tumor-associated macrophages. Cancer research 2014;74:2986–98
    1. Schlager SI, Madden LD, Meltzer MS, Bara S, Mamula MJ. Role of macrophage lipids in regulating tumoricidal activity. Cellular immunology 1983;77:52–68
    1. Zhao J, Zhi Z, Wang C, Xing H, Song G, Yu X, et al. Exogenous lipids promote the growth of breast cancer cells via CD36. Oncology reports 2017;38:2105–15
    1. Watt MJ, Clark AK, Selth LA, Haynes VR, Lister N, Rebello R, et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Science translational medicine 2019;11
    1. Ladanyi A, Mukherjee A, Kenny HA, Johnson A, Mitra AK, Sundaresan S, et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018;37:2285–301
    1. Enciu AM, Radu E, Popescu ID, Hinescu ME, Ceafalan LC. Targeting CD36 as Biomarker for Metastasis Prognostic: How Far from Translation into Clinical Practice? BioMed research international 2018;2018:7801202.
    1. Nath A, Li I, Roberts LR, Chan C. Elevated free fatty acid uptake via CD36 promotes epithelial-mesenchymal transition in hepatocellular carcinoma. Scientific reports 2015;5:14752.
    1. Hale JS, Otvos B, Sinyuk M, Alvarado AG, Hitomi M, Stoltz K, et al. Cancer stem cell-specific scavenger receptor CD36 drives glioblastoma progression. Stem Cells 2014;32:1746–58
    1. Al-Khami AA, Zheng L, Del Valle L, Hossain F, Wyczechowska D, Zabaleta J, et al. Exogenous lipid uptake induces metabolic and functional reprogramming of tumor-associated myeloid-derived suppressor cells. Oncoimmunology 2017;6:e1344804.
    1. Carracedo A, Cantley LC, Pandolfi PP. Cancer metabolism: fatty acid oxidation in the limelight. Nature reviews Cancer 2013;13:227–32
    1. Goossens P, Rodriguez-Vita J, Etzerodt A, Masse M, Rastoin O, Gouirand V, et al. Membrane Cholesterol Efflux Drives Tumor-Associated Macrophage Reprogramming and Tumor Progression. Cell metabolism 2019;29:1376–89 e4

Source: PubMed

3
Abonnere