Lysine-Specific Histone Demethylase 1A Regulates Macrophage Polarization and Checkpoint Molecules in the Tumor Microenvironment of Triple-Negative Breast Cancer

Abel H Y Tan, WenJuan Tu, Robert McCuaig, Kristine Hardy, Thomasina Donovan, Sofiya Tsimbalyuk, Jade K Forwood, Sudha Rao, Abel H Y Tan, WenJuan Tu, Robert McCuaig, Kristine Hardy, Thomasina Donovan, Sofiya Tsimbalyuk, Jade K Forwood, Sudha Rao

Abstract

Macrophages play an important role in regulating the tumor microenvironment (TME). Here we show that classical (M1) macrophage polarization reduced expression of LSD1, nuclear REST corepressor 1 (CoREST), and the zinc finger protein SNAIL. The LSD1 inhibitor phenelzine targeted both the flavin adenine dinucleotide (FAD) and CoREST binding domains of LSD1, unlike the LSD1 inhibitor GSK2879552, which only targeted the FAD domain. Phenelzine treatment reduced nuclear demethylase activity and increased transcription and expression of M1-like signatures both in vitro and in a murine triple-negative breast cancer model. Overall, the LSD1 inhibitors phenelzine and GSK2879552 are useful tools for dissecting the contribution of LSD1 demethylase activity and the nuclear LSD1-CoREST complex to switching macrophage polarization programs. These findings suggest that inhibitors must have dual FAD and CoREST targeting abilities to successfully initiate or prime macrophages toward an anti-tumor M1-like phenotype in triple-negative breast cancer.

Keywords: CoREST; LSD1; breast cancer; epigenetics; macrophage polarization; tumor associated macrophages; tumor microenvironment.

Figures

Figure 1
Figure 1
Phenelzine targets the FAD domain of LSD1 and potentially disrupts the LSD1/CoREST axis resulting in destabilization of LSD1 and its nuclear activity. (A) LSD1 protein crystals (top panel) grown in the absence (left) and presence of phenelzine (middle) and GSK2879552 (right). In the absence of inhibitors, strong density (bottom panels) was observed consistent with the FAD cofactor (shown as sticks colored with carbons black, nitrogen blue, oxygen red, and phosphate orange). The map is a simulated annealed omit map for FAD contoured at 2.5 sigma. LSD1 is colored gray and shown in cartoon mode. The FAD modifications by phenelzine (middle) and GSK (right) are supported by strong density, with corresponding maps and colors as per LSD1:FAD. (B) Structural superposition of LSD1 in the absence and presence of phenelzine and GSK. The structures solved in this study (left panel), LSD1 alone (pink) (PDB 6NQM), LSD1:GSK (yellow) (PDB 6NQU), and LSD1:phenelzine (green) (PDB 6NR5) are represented in cartoon mode. These structures are superimposed in the left panel, showing a high degree of structural homology in the LSD1 catalytic domain for all three structures. LSD1 alone and LSD1:GSK also show high structural conservation in the alpha-helical tails; however, LSD1:phenelzine has a 5.4 Å displacement in this region. This region is important for CoREST binding, as shown in the middle panel (PDB 2UXX). Superposition of all structures in the left and middle panels is shown on the right, highlighting that CoREST binding is mediated by the correct position of these domains. All images were generated in Pymol. (C) The plot-profile feature of ImageJ was used to plot the fluorescence signal intensity along a single line spanning the nucleus (n = 5 lines per nucleus, 5 individual cells) using the average fluorescent signal intensity for the indicated pair of antibodies plotted for each point on the line with SE. (D) Pearson's correlation coefficient (PCC) indicating the colocalization of LSD1/CoREST and CoREST/SNAIL. *p < 0.05, **p< 0.005, ***p < 0.0005, ****p < 0.0001 Mann–Whitney t-test.
Figure 2
Figure 2
Phenelzine or M1 polarization upregulates M1 protein CD38 and targets the nuclear activity of LSD1. RAW264.7 cells were treated with LPS + IFN-γ, IL-4, or 500 μM phenelzine or GSK for 24 h. Protein targets (A) EGR2 and CD38 in F4/80+ cells were localized by confocal laser scanning microscopy. Single 0.5 μm optical sections were obtained using an Olympus-ASI automated microscope with 100x oil immersion lens running ASI software. The final image was obtained by employing a high-throughput automated stage with ASI spectral capture software. Digital images were analyzed using automated ASI software to determine the distribution and intensities automatically with automatic thresholding and background correction. Graphs represent either a dot plot of the individual cell intensities or the average TFI (n = 2,000 cells). (B) LSD1 activity assay on nuclear extracts of RAW264.7 cells either untreated, M1/M2 polarized, or treated with phenelzine, GSK, or LSD1 inhibitor tranylcypromine. (C) Images of RAW264.7 cells treated with vehicle control, LPS + IFN-γ (M1), and IL-4 (M2) for 24 h. Phenelzine and GSK treated cells did not show morphology changes in 24 h (data not shown). In comparison, cells were treated with Phenelzine or GSK for 7 days.
Figure 3
Figure 3
LSD1 can regulate genes associated with macrophage polarization toward an M1 phenotype and checkpoint molecules. RAW264.7 cells were untreated or treated with LPS + IFN-γ (M1), IL-4 (M2), or 500 μM of phenelzine or GSK for 24 h. Quantitative real-time PCR of genes associated with (A) M1-associated genes and (B) M2-associated genes were used to compare different treatment groups. Graphs are represented as mRNA levels normalized to GAPDH. Graphs show means ± SE (n = 3). *p < 0.05, **p < 0.005 Mann-Whitney t-test. (C) and (D) shows chromatin accessibility of genes in the promoter (C) and enchancer (D) regions of associated genes using quantitative real-time PCR using FAIRE samples. (E) 10nM LSD1 siRNA transfected cells and (F) checkpoint molecules and were used to compare different treatment groups.
Figure 4
Figure 4
Phenelzine treatment polarizes macrophages in the tumor microenvironment toward an M1 phenotype. (A) Treatment regime using the BALB/c 4T1 breast cancer model. (B) Tumor volumes of mice treated with vehicle control, Abraxane, Phenelzine or PD1 (n = 4/5). (C) Flow cytometry for total macrophages, inflammatory macrophages, and M2-like macrophages in the TME. *p < 0.05, Mann–Whitney t-test (n = 4/5). Representative images of (D) M1 and (E) M2 staining of FFPE tumor tissues in 4T1 mouse model. (F) Sections of primary 4T1 tumors were fixed and IF microscopy performed probing with M1 focused primary antibodies to F4/80, iNOS, CD86, and PDL1 with DAPI (green = F4/80 red = iNOS, yellow = CD86, cyan = PDL1, blue = DAPI). The population % of F4/80 cells positive for iNOS, CD86 and PDL1 was measured using ASI's mIF system. Representative images for each dataset are shown. Graphs plots represent the % population (n ≥ 500 cells profiled per a group, n = 4 mice). (G) Section of primary 4T1 tumors were fixed and IF microscopy performed probing with M2 focused primary antibodies to F4/80, EGR2, CD206, and PDL2 with DAPI (green = F4/80 red = EGR2, yellow = CD206, cyan = PDL2, blue = DAPI). The population % of F4/80 cells positive for EGR2, CD206, and PDL2 was measured using ASI's mIF system. Representative images for each dataset are shown. Graphs plots represent the % population (n ≥ 500 cells profiled per a group, n = 4 mice).
Figure 5
Figure 5
Nanostring counts (log2 fold-change) from RNA isolated from macrophages in the TME for (A) M1 phenotypic signatures and pathways, (B) M1 signaling molecules and transcription factors, (C) T cell activation gene signatures, and (D) CD169+ macrophage gene signatures. *Indicates Benjamini–Yekutieli false discovery rate value < 0.05.
Figure 6
Figure 6
CpG content, GC content, histone marks, and accessibility of genes in the NanoString panel. (A) CpG and GC content, (B,D) histone levels and (C) accessibility of genes ± 1 kb from transcription start site (TSS) of NanoString genes up-regulated, downregulated (FDR < 0.15), or unchanged (FDR > 0.15) and genes from refSeq in RAW264.7 cells (47). (C) shows accessibility of Nanostring genes against gene sets from bone marrow derived macrophages (BMDMs) with or without 6 h of LPS stimulation (48). (E) Levels of up/down regulated NanoString genes that had enhancers within 10 kb of their TSS from BMDMs with or without 24 h of LPS stimulation (31). A t-test with unequal variance (Welch two sample t-test) was used with *p < 0.05, **p < 0.01 and ***p < 0.001.
Figure 7
Figure 7
Phenelzine affects Hippo, Wnt, and Ras signaling pathways and genes associated with ECM remodeling and metabolism. (A) Heatmap displaying the undirected and directed global significance score statistics using the default and (B) KEGG pathway annotations. Undirected scores measure the extent of differential expression of a geneset's genes against control ignoring whether each gene within the set is up- or downregulated. Orange denotes genesets whose genes exhibit extensive differential expression against control, and blue denotes genesets with less differential expression. Directed scores measure the extent to which a geneset is up- or downregulated compared to control. Red denotes genesets that show extensive overexpression and blue denotes genesets with extensive underexpression. (C) Venn diagram indicating genes that are differentially expressed with a false discovery rate of < 0.05 (Benjamini-Yekutieli) and top 6 pathways that those genes are fall under. (D) Table showing the upregulated and downregulated genes that are specific to Abraxane and phenelzine.
Figure 8
Figure 8
Putative model of how LSD1 can reprogram macrophage polarization. (A) When macrophages are stimulated with LPS and IFN-γ (classical activation; M1), disruption of CoREST destabilizes LSD1, which leads to LSD1 losing its repressive role in regulating M1-associated genes. It also increases LSD1 demethylase activity. (B) When macrophages are stimulated with IL-4 (alternative activation; M2), CoREST is not affected, resulting in stable expression of LSD1p and LSD1 maintaining its repressive role in regulating M1-associated genes. It also decreases LSD1 demethylase activity. (C) LSD1 inhibition using phenelzine can target both LSD1p and CoREST, mimicking a similar response to M1 polarization while (D) GSK was not able to achieve the same result because it did not disrupt the LSD1/CoREST complex.

References

    1. Torre LA, Siegel RL, Ward EM, Jemal A. Global cancer incidence and mortality rates and trends–an update. Cancer Epidemiol Biomarkers Prev. (2016) 25:16–27. 10.1158/1055-9965.EPI-15-0578
    1. Diana A, Franzese E, Centonze S, Carlino F, Della Corte CM, Ventriglia J, et al. . Triple-negative breast cancers: systematic review of the literature on molecular and clinical features with a focus on treatment with innovative drugs. Curr Oncol Rep. (2018) 20:76. 10.1007/s11912-018-0726-6
    1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. (2010) 363:1938–48. 10.1056/NEJMra1001389
    1. Dent R, Trudeau M, Pritchard KI, Hanna WM, Kahn HK, Sawka CA, et al. . Triple-negative breast cancer: clinical features and patterns of recurrence. Clin Cancer Res. (2007) 13(15 Pt 1):4429–34. 10.1158/1078-0432.CCR-06-3045
    1. Sharma P. Biology and management of patients with triple-negative breast cancer. Oncologist. (2016) 21:1050–62. 10.1634/theoncologist.2016-0067
    1. Abramson VG, Lehmann BD, Ballinger TJ, Pietenpol JA. Subtyping of triple-negative breast cancer: implications for therapy. Cancer. (2015) 121:8–16. 10.1002/cncr.28914
    1. Hudis CA, Gianni L. Triple-negative breast cancer: an unmet medical need. Oncologist. (2011) 16 (Suppl. 1):1–11. 10.1634/theoncologist.2011-S1-01
    1. Lee HJ, Lee JJ, Song IH, Park IA, Kang J, Yu JH, et al. . Prognostic and predictive value of NanoString-based immune-related gene signatures in a neoadjuvant setting of triple-negative breast cancer: relationship to tumor-infiltrating lymphocytes. Breast Cancer Res Treat. (2015) 151:619–27. 10.1007/s10549-015-3438-8
    1. Liu Z, Li M, Jiang Z, Wang X. A Comprehensive immunologic portrait of triple-negative breast cancer. Transl Oncol. (2018) 11:311–29. 10.1016/j.tranon.2018.01.011
    1. Chanmee T, Ontong P, Konno K, Itano N. Tumor-associated macrophages as major players in the tumor microenvironment. Cancers. (2014) 6:1670–90. 10.3390/cancers6031670
    1. Aras S, Zaidi MR. TAMeless traitors: macrophages in cancer progression and metastasis. Br J Cancer. (2017) 117:1583–91. 10.1038/bjc.2017.356
    1. Jablonski KA, Amici SA, Webb LM, Ruiz-Rosado Jde D, Popovich PG, Partida-Sanchez S, et al. . Novel Markers to Delineate Murine M1 and M2 Macrophages. PLoS ONE. (2015) 10:e0145342. 10.1371/journal.pone.0145342
    1. Jiang L, Li X, Zhang Y, Zhang M, Tang Z, Lv K. Microarray and bioinformatics analyses of gene expression profiles in BALB/c murine macrophage polarization. Mol Med Rep. (2017) 16:7382–90. 10.3892/mmr.2017.7511
    1. Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest. (2012) 122:787–95. 10.1172/JCI59643
    1. Aspeslagh S, Morel D, Soria JC, Postel-Vinay S. Epigenetic modifiers as new immunomodulatory therapies in solid tumours. Ann Oncol. (2018) 29:812–24. 10.1093/annonc/mdy050
    1. de Groot AE, Pienta KJ. Epigenetic control of macrophage polarization: implications for targeting tumor-associated macrophages. Oncotarget. (2018) 9:20908–27. 10.18632/oncotarget.24556
    1. Ismail T, Lee HK, Kim C, Kwon T, Park TJ, Lee HS. KDM1A microenvironment, its oncogenic potential, and therapeutic significance. Epigenetics Chromatin. (2018) 11:33. 10.1186/s13072-018-0203-3
    1. Ivashkiv LB. Epigenetic regulation of macrophage polarization and function. Trends Immunol. (2013) 34:216–23. 10.1016/j.it.2012.11.001
    1. Kerenyi MA, Shao Z, Hsu YJ, Guo G, Luc S, O'Brien K, et al. . Histone demethylase Lsd1 represses hematopoietic stem and progenitor cell signatures during blood cell maturation. Elife. (2013) 2:e00633. 10.7554/eLife.00633
    1. Qin Y, Vasilatos SN, Chen L, Wu H, Cao Z, Fu Y, et al. . Inhibition of histone lysine-specific demethylase 1 elicits breast tumor immunity and enhances antitumor efficacy of immune checkpoint blockade. Oncogene. (2018) 38:390–405. 10.1038/s41388-018-0451-5
    1. Foster SL, Hargreaves DC, Medzhitov R. Gene-specific control of inflammation by TLR-induced chromatin modifications. Nature. (2007) 447:972–8. 10.1038/nature05836
    1. Boulding T, McCuaig RD, Tan A, Hardy K, Wu F, Dunn J, et al. . LSD1 activation promotes inducible EMT programs and modulates the tumour microenvironment in breast cancer. Sci Rep. (2018) 8:73. 10.1038/s41598-017-17913-x
    1. Simon JM, Giresi PG, Davis IJ, Lieb JD. A detailed protocol for formaldehyde-assisted isolation of regulatory elements (FAIRE). Curr Protoc Mol Biol. (2013) Chapter 21:Unit21.26. 10.1002/0471142727.mb2126s102
    1. Benjamini Y, Yekutieli D. The control of the false discovery rate in multiple testing under dependency. Ann Stat. (2001) 29:1165–88. 10.1214/aos/1013699998
    1. Battye TG, Kontogiannis L, Johnson O, Powell HR, Leslie AG. iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM. Acta Crystallogr D Biol Crystallogr. (2011) 67(Pt 4):271–81. 10.1107/S0907444910048675
    1. Evans PR, Murshudov GN. How good are my data and what is the resolution? Acta Crystallogr D Biol Crystallogr. (2013) 69(Pt 7):1204–14. 10.1107/S0907444913000061
    1. Kantardjieff KA, Rupp B. Matthews coefficient probabilities: Improved estimates for unit cell contents of proteins, DNA, and protein-nucleic acid complex crystals. Protein Sci. (2003) 12:1865–71. 10.1110/ps.0350503
    1. Matthews BW. Solvent content of protein crystals. J Mol Biol. (1968) 33:491–7. 10.1016/0022-2836(68)90205-2
    1. Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr. (2004) 60(Pt 12 Pt 1):2126–32. 10.1107/S0907444904019158
    1. Murshudov GN, Skubak P, Lebedev AA, Pannu NS, Steiner RA, Nicholls RA, et al. . REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr D Biol Crystallogr. (2011) 67(Pt 4):355–67. 10.1107/S0907444911001314
    1. Ostuni R, Piccolo V, Barozzi I, Polletti S, Termanini A, Bonifacio S, et al. . Latent enhancers activated by stimulation in differentiated cells. Cell. (2013) 152:157–71. 10.1016/j.cell.2012.12.018
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. (2014) 30:2114–20. 10.1093/bioinformatics/btu170
    1. Langmead B, Salzberg SL. Fast gapped-read alignment with Bowtie 2. Nat Methods. (2012) 9:357–9. 10.1038/nmeth.1923
    1. Heinz S, Benner C, Spann N, Bertolino E, Lin YC, Laslo P, et al. . Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol Cell. (2010) 38:576–89. 10.1016/j.molcel.2010.05.004
    1. Quinlan AR, Hall IM. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics. (2010) 26:841–2. 10.1093/bioinformatics/btq033
    1. Culhane JC, Wang D, Yen PM, Cole PA. Comparative analysis of small molecules and histone substrate analogues as LSD1 lysine demethylase inhibitors. J Am Chem Soc. (2010) 132:3164–76. 10.1021/ja909996p
    1. Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, et al. . A DNA hypomethylation signature predicts antitumor activity of LSD1 Inhibitors in SCLC. Cancer Cell. (2015) 28:57–69. 10.1016/j.ccell.2015.06.002
    1. Binda C, Valente S, Romanenghi M, Pilotto S, Cirilli R, Karytinos A, et al. . Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J Am Chem Soc. (2010) 132:6827–33. 10.1021/ja101557k
    1. Schmidt DM, McCafferty DG. trans-2-Phenylcyclopropylamine is a mechanism-based inactivator of the histone demethylase LSD1. Biochemistry. (2007) 46:4408–16. 10.1021/bi0618621
    1. Binda C, Wang J, Li M, Hubalek F, Mattevi A, Edmondson DE. Structural and mechanistic studies of arylalkylhydrazine inhibition of human monoamine oxidases A and B. Biochemistry. (2008) 47:5616–25. 10.1021/bi8002814
    1. Yang M, Culhane JC, Szewczuk LM, Jalili P, Ball HL, Machius M, et al. . Structural basis for the inhibition of the LSD1 histone demethylase by the antidepressant trans-2-phenylcyclopropylamine. Biochemistry. (2007) 46:8058–65. 10.1021/bi700664y
    1. Zhang F, Wang H, Wang X, Jiang G, Liu H, Zhang G, et al. . TGF-beta induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget. (2016) 7:52294–306. 10.18632/oncotarget.10561
    1. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and Anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. (2018) 8:86. 10.3389/fonc.2018.00086
    1. Zou W, Wolchok JD, Chen L. PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med. (2016) 8:328rv4. 10.1126/scitranslmed.aad7118
    1. Jakubzick CV, Randolph GJ, Henson PM. Monocyte differentiation and antigen-presenting functions. Nat Rev Immunol. (2017) 17:349–62. 10.1038/nri.2017.28
    1. Roche PA, Furuta K. The ins and outs of MHC class II-mediated antigen processing and presentation. Nat Rev Immunol. (2015) 15:203–16. 10.1038/nri3818
    1. Soldi M, Mari T, Nicosia L, Musiani D, Sigismondo G, Cuomo A, et al. . Chromatin proteomics reveals novel combinatorial histone modification signatures that mark distinct subpopulations of macrophage enhancers. Nucleic Acids Res. (2017) 45:12195–213. 10.1093/nar/gkx821
    1. Atianand MK, Hu W, Satpathy AT, Shen Y, Ricci EP, Alvarez-Dominguez JR, et al. . A Long noncoding RNA lincRNA-EPS acts as a transcriptional brake to restrain inflammation. Cell. (2016) 165:1672–85. 10.1016/j.cell.2016.05.075
    1. Shi YJ, Matson C, Lan F, Iwase S, Baba T, Shi Y. Regulation of LSD1 histone demethylase activity by its associated factors. Mol Cell. (2005) 19:857–64. 10.1016/j.molcel.2005.08.027
    1. Lee MG, Wynder C, Cooch N, Shiekhattar R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature. (2005) 437:432–5. 10.1038/nature04021
    1. Metzger E, Wissmann M, Yin N, Muller JM, Schneider R, Peters AH, et al. . LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature. (2005) 437:436–9. 10.1038/nature04020
    1. Brenot A, Knolhoff BL, DeNardo DG, Longmore GD. SNAIL1 action in tumor cells influences macrophage polarization and metastasis in breast cancer through altered GM-CSF secretion. Oncogenesis. (2018) 7:32. 10.1038/s41389-018-0042-x
    1. Zafar A, Wu F, Hardy K, Li J, Tu WJ, McCuaig R, et al. . Chromatinized protein kinase C-theta directly regulates inducible genes in epithelial to mesenchymal transition and breast cancer stem cells. Mol Cell Biol. (2014) 34:2961–80. 10.1128/MCB.01693-13
    1. Brezar V, Tu WJ, Seddiki N. PKC-theta in regulatory and effector T-cell functions. Front Immunol. (2015) 6:530. 10.3389/fimmu.2015.00530
    1. Pfeifhofer-Obermair C, Albrecht-Schgoer K, Peer S, Nairz M, Siegmund K, Klepsch V, et al. . Role of PKCtheta in macrophage-mediated immune response to Salmonella typhimurium infection in mice. Cell Commun Signal. (2016) 14:14. 10.1186/s12964-016-0137-y
    1. Sutcliffe EL, Li J, Zafar A, Hardy K, Ghildyal R, McCuaig R, et al. . Chromatinized protein kinase C-theta: can it escape the clutches of NF-kappaB? Front Immunol. (2012) 3:260. 10.3389/fimmu.2012.00260
    1. Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, et al. . Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. (2009) 114:3244–54. 10.1182/blood-2009-04-217620
    1. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol. (2010) 11:889–96. 10.1038/ni.1937
    1. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. (2004) 25:677–86. 10.1016/j.it.2004.09.015
    1. Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Rep. (2014) 6:13. 10.12703/P6-13
    1. Murray PJ. Macrophage polarization. Annu Rev Physiol. (2017) 79:541–66. 10.1146/annurev-physiol-022516-034339
    1. Wang N, Liang H, Zen K. Molecular mechanisms that influence the macrophage m1-m2 polarization balance. Front Immunol. (2014) 5:614. 10.3389/fimmu.2014.00614
    1. Lawrence T, Natoli G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol. (2011) 11:750–61. 10.1038/nri3088
    1. Leopold Wager CM, Hole CR, Wozniak KL, Olszewski MA, Mueller M, Wormley FL, Jr. STAT1 signaling within macrophages is required for antifungal activity against Cryptococcus neoformans. Infect Immun. (2015) 83:4513–27. 10.1128/IAI.00935-15
    1. Fong CH, Bebien M, Didierlaurent A, Nebauer R, Hussell T, Broide D, et al. . An antiinflammatory role for IKKbeta through the inhibition of “classical” macrophage activation. J Exp Med. (2008) 205:1269–76. 10.1084/jem.20080124
    1. Saccani A, Schioppa T, Porta C, Biswas SK, Nebuloni M, Vago L, et al. . p50 nuclear factor-kappaB overexpression in tumor-associated macrophages inhibits M1 inflammatory responses and antitumor resistance. Cancer Res. (2006) 66:11432–40. 10.1158/0008-5472.CAN-06-1867
    1. Liu CP, Zhang X, Tan QL, Xu WX, Zhou CY, Luo M, et al. . NF-kappaB pathways are involved in M1 polarization of RAW 264.7 macrophage by polyporus polysaccharide in the tumor microenvironment. PLoS ONE. (2017) 12:e0188317. 10.1371/journal.pone.0188317
    1. Liu X, Li J, Peng X, Lv B, Wang P, Zhao X, et al. . Geraniin Inhibits LPS-Induced THP-1 macrophages switching to M1 Phenotype via SOCS1/NF-kappaB pathway. Inflammation. (2016) 39:1421–33. 10.1007/s10753-016-0374-7
    1. Xu X, Yin P, Wan C, Chong X, Liu M, Cheng P, et al. . Punicalagin inhibits inflammation in LPS-induced RAW264.7 macrophages via the suppression of TLR4-mediated MAPKs and NF-kappaB activation. Inflammation. (2014) 37:956–65. 10.1007/s10753-014-9816-2
    1. Liao X, Sharma N, Kapadia F, Zhou G, Lu Y, Hong H, et al. . Kruppel-like factor 4 regulates macrophage polarization. J Clin Invest. (2011) 121:2736–49. 10.1172/JCI45444
    1. Han YH, Kim HJ, Na H, Nam MW, Kim JY, Kim JS, et al. . RORalpha Induces KLF4-Mediated M2 polarization in the liver macrophages that protect against nonalcoholic steatohepatitis. Cell Rep. (2017) 20:124–35. 10.1016/j.celrep.2017.06.017
    1. Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, et al. . The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. (2010) 11:936–44. 10.1038/ni.1920
    1. Gunthner R, Anders HJ. Interferon-regulatory factors determine macrophage phenotype polarization. Mediators Inflamm. (2013) 2013:731023. 10.1155/2013/731023
    1. Krausgruber T, Blazek K, Smallie T, Alzabin S, Lockstone H, Sahgal N, et al. . IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol. (2011) 12:231–8. 10.1038/ni.1990
    1. Colado A, Genoula M, Cougoule C, Marin Franco JL, Almejun MB, Risnik D, et al. . Effect of the BTK inhibitor ibrutinib on macrophage- and gammadelta T cell-mediated response against Mycobacterium tuberculosis. Blood Cancer J. (2018) 8:100. 10.1038/s41408-018-0136-x
    1. Ni Gabhann J, Hams E, Smith S, Wynne C, Byrne JC, Brennan K, et al. . Btk regulates macrophage polarization in response to lipopolysaccharide. PLoS ONE. (2014) 9:e85834. 10.1371/journal.pone.0085834
    1. Anand-Apte B, Pepper MS, Voest E, Montesano R, Olsen B, Murphy G, et al. . Inhibition of angiogenesis by tissue inhibitor of metalloproteinase-3. Invest Ophthalmol Vis Sci. (1997) 38:817–23.
    1. Baker AH, Edwards DR, Murphy G. Metalloproteinase inhibitors: biological actions and therapeutic opportunities. J Cell Sci. (2002) 115(Pt 19):3719–27. 10.1242/jcs.00063
    1. Qi JH, Ebrahem Q, Moore N, Murphy G, Claesson-Welsh L, Bond M, et al. . A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med. (2003) 9:407–15. 10.1038/nm846
    1. Asano K, Nabeyama A, Miyake Y, Qiu CH, Kurita A, Tomura M, et al. . CD169-positive macrophages dominate antitumor immunity by crosspresenting dead cell-associated antigens. Immunity. (2011) 34:85–95. 10.1016/j.immuni.2010.12.011
    1. Chavez-Galan L, Olleros ML, Vesin D, Garcia I. Much More than M1 and M2 Macrophages, there are also CD169(+) and TCR(+) Macrophages. Front Immunol. (2015) 6:263. 10.3389/fimmu.2015.00263
    1. Martinez-Pomares L, Gordon S. CD169+ macrophages at the crossroads of antigen presentation. Trends Immunol. (2012) 33:66–70. 10.1016/j.it.2011.11.001
    1. Shiota T, Miyasato Y, Ohnishi K, Yamamoto-Ibusuki M, Yamamoto Y, Iwase H, et al. . The clinical significance of CD169-positive lymph node macrophage in patients with breast cancer. PLoS ONE. (2016) 11:e0166680. 10.1371/journal.pone.0166680
    1. van Dinther D, Veninga H, Iborra S, Borg EGF, Hoogterp L, Olesek K, et al. . Functional CD169 on macrophages mediates interaction with dendritic cells for CD8(+) T cell cross-priming. Cell Rep. (2018) 22:1484–95. 10.1016/j.celrep.2018.01.021
    1. Hashimoto H, Vertino PM, Cheng X. Molecular coupling of DNA methylation and histone methylation. Epigenomics. (2010) 2:657–69. 10.2217/epi.10.44
    1. Hansen CG, Moroishi T, Guan KL. YAP and TAZ: a nexus for Hippo signaling and beyond. Trends Cell Biol. (2015) 25:499–513. 10.1016/j.tcb.2015.05.002
    1. Yang Y, Ye YC, Chen Y, Zhao JL, Gao CC, Han H, et al. . Crosstalk between hepatic tumor cells and macrophages via Wnt/beta-catenin signaling promotes M2-like macrophage polarization and reinforces tumor malignant behaviors. Cell Death Dis. (2018) 9:793. 10.1038/s41419-018-0818-0
    1. Newman AC, Hughes CC. Macrophages and angiogenesis: a role for Wnt signaling. Vasc Cell. (2012) 4:13. 10.1186/2045-824X-4-13
    1. Ojalvo LS, Whittaker CA, Condeelis JS, Pollard JW. Gene expression analysis of macrophages that facilitate tumor invasion supports a role for Wnt-signaling in mediating their activity in primary mammary tumors. J Immunol. (2010) 184:702–12. 10.4049/jimmunol.0902360
    1. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene. (2017) 36:1461–73. 10.1038/onc.2016.304
    1. Downward J. Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. (2003) 3:11–22. 10.1038/nrc969
    1. George AJ, Thomas WG, Hannan RD. The renin-angiotensin system and cancer: old dog, new tricks. Nat Rev Cancer. (2010) 10:745–59. 10.1038/nrc2945
    1. Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Brown Swigart L, et al. . Myc cooperates with ras by programming inflammation and immune suppression. Cell. (2017) 171:1301–15 e14. 10.1016/j.cell.2017.11.013
    1. Gouirand V, Guillaumond F, Vasseur S. Influence of the tumor microenvironment on cancer cells metabolic reprogramming. Front Oncol. (2018) 8:117. 10.3389/fonc.2018.00117
    1. Lyssiotis CA, Kimmelman AC. Metabolic interactions in the tumor microenvironment. Trends Cell Biol. (2017) 27:863–75. 10.1016/j.tcb.2017.06.003
    1. Ocana MC, Martinez-Poveda B, Quesada AR, Medina MA. Metabolism within the tumor microenvironment and its implication on cancer progression: an ongoing therapeutic target. Med Res Rev. (2019) 39:70–113. 10.1002/med.21511
    1. Sainio A, Jarvelainen H. Extracellular matrix macromolecules: potential tools and targets in cancer gene therapy. Mol Cell Ther. (2014) 2:14. 10.1186/2052-8426-2-14
    1. Diskin C, Palsson-McDermott EM. Metabolic Modulation in Macrophage Effector Function. Front Immunol. (2018) 9:270. 10.3389/fimmu.2018.00270
    1. Hamers AAJ, Pillai AB. A sweet alternative: maintaining M2 macrophage polarization. Sci Immunol. (2018) 3:eaav7759. 10.1126/sciimmunol.aav7759

Source: PubMed

3
Se inscrever