Decitabine Promotes Modulation in Phenotype and Function of Monocytes and Macrophages That Drive Immune Response Regulation

Fabiana Albani Zambuzi, Priscilla Mariane Cardoso-Silva, Ricardo Cardoso Castro, Caroline Fontanari, Flavio da Silva Emery, Fabiani Gai Frantz, Fabiana Albani Zambuzi, Priscilla Mariane Cardoso-Silva, Ricardo Cardoso Castro, Caroline Fontanari, Flavio da Silva Emery, Fabiani Gai Frantz

Abstract

Decitabine is an approved hypomethylating agent used for treating hematological malignancies. Although decitabine targets altered cells, epidrugs can trigger immunomodulatory effects, reinforcing the hypothesis of immunoregulation in treated patients. We therefore aimed to evaluate the impact of decitabine treatment on the phenotype and functions of monocytes and macrophages, which are pivotal cells of the innate immunity system. In vitro decitabine administration increased bacterial phagocytosis and IL-8 release, but impaired microbicidal activity of monocytes. In addition, during monocyte-to-macrophage differentiation, treatment promoted the M2-like profile, with increased expression of CD206 and ALOX15. Macrophages also demonstrated reduced infection control when exposed to Mycobacterium tuberculosis in vitro. However, cytokine production remained unchanged, indicating an atypical M2 macrophage. Furthermore, when macrophages were cocultured with lymphocytes, decitabine induced a reduction in the release of inflammatory cytokines such as IL-1β, TNF-α, and IFN-γ, maintaining IL-10 production, suggesting that decitabine could potentialize M2 polarization and might be considered as a therapeutic against the exacerbated immune response.

Keywords: DNMT inhibitors; immune cell; monocyte/macrophages; phagocytosis.

Conflict of interest statement

The authors declare that they have no commercial or other associations that might pose a conflict of interest in the manuscript.

Figures

Figure 1
Figure 1
Decitabine treatment modulated the monocyte response to mycobacterial infection. Monocytes were isolated from healthy individuals and pretreated with decitabine (0.25–5 µM), DMSO, or untreated for 24 h. After this period, cell viability was assessed by detecting resazurin metabolism (A). In addition, cells were infected with Mtb (MOI 5) for different periods. For phagocytosis analysis, cells were infected for 2 h, followed by washes with PBS, saponin lyses, and exposure to internalized bacteria. The number of phagocytized bacteria was determined indirectly by detecting resazurin metabolism, expressed in (B) and phagocytic index, normalized in relation to untreated cells, considered as 100% (C). Immune mediator release was determined after 24 h of pretreated monocyte infection with Mtb and quantified using a Magnetic Luminex Assay kit, as demonstrated in (D) IL-1β, (E) TNF-α, and (F) IL-8. (G,H) Microbicide activity was evaluated after 24 h of infection and the number of bacteria recovered from cells was quantified indirectly by detecting resazurin metabolization, expressed in (G), and the killing index was determined by normalization of RFU quantitation, in relation to untreated cells, considered as 100% (H). Data are expressed as mean ± standard deviation and analyzed by paired ANOVA, followed by a Tukey’s test. Phagocytosis and killing index were analyzed by a Friedman test (non-parametric), followed by a Dunn’s test. All the analyses consider * p < 0.05 as statistically significant (I) A schematic summarizing the effects of decitabine on monocyte function during Mtb infection.
Figure 2
Figure 2
Decitabine modulated macrophage function by potentializing the M2 like profile. Monocytes from healthy subjects were isolated from peripheral blood and cultured for 6 days in the RPMI 1640 medium supplemented with 10% FBS and 50 ng/mL granulocyte-macrophage colony-stimulating factor (GM-CSF), and 5 μM decitabine or DMSO (0.2%) added for macrophage differentiation. On the 6th day, differentiated macrophages were stimulated with cytokines for macrophage polarization or infected with Mtb for functional analysis. The M1 profile was defined by the addition of IFN-γ (100 ng/mL) and M2, by IL-4 and IL-13 (50 ng/mL each). After 24 h in the presence of polarizing cytokines, the macrophage profile was determined by gene expression of ALOX15 (A) and CXCL10 (D). The expression was normalized using Ct of the endogenous gene (ACTB) and the relative expression calculated using 2−ΔΔCt in relation to resting macrophages differentiated with DMSO. In addition, surface markers CD206 ((B) frequency, and (C) Mean Fluorescence Intensity) and CD86 ((E), frequency and (F), Mean Fluorescent Intensity—MFI) (D,F) were analyzed by flow cytometry. For functional analysis, after the differentiation period in the presence of decitabine or DMSO, macrophages were infected with Mtb and 24 h post-infection, cells were lysed to bacterial load determination through colony forming units (CFU) count, represented in (G), and the supernatant was collected for IL-10 (H), IL-1β (I), TNF-α (J), and IL-8 (K) quantitation by ELISA. Data were analyzed by paired tests, according to the normality distribution and number of groups per experiment. Gene expression and surface markers were analyzed through the ANOVA test for repeated measurements, followed by a Tukey test to evaluate the effect of stimulation with polarizing conditions, with # p < 0.05 M0 vs. M1 and M2 vs. M1. In addition, the effect of decitabine treatment in each condition was evaluated by the paired t-test with * p < 0.05 and ** p < 0.01 in relation to DMSO. CFU analysis was performed by the paired t-test, and cytokine release was analyzed by the non-parametric Wilcoxon test comparing decitabine to DMSO in infected conditions (*). All analyses considered p ≤ 0.05 as statistically significant. (L) Figure summarizing the effects of decitabine on macrophage phenotypes of polarization and their functions during Mtb infection. The symbols within the bars of the graphs represent each evaluated samples.
Figure 3
Figure 3
Decitabine reduces inflammation during Mtb infection in granuloma-like models. Peripheral blood mononuclear cells (PBMCs) were isolated and distributed (1 × 106 cells/well) into 48-well plates coated with an agarose layer (230 μL/well). Cells were incubated in RPMI medium supplemented with human serum (10%) for 24 h in the presence of 5 μM decitabine, DMSO, or untreated. The next day, cells were infected with Mtb H37Rv (1 × 104 bacteria/well) and incubated at 37 °C and 5% CO2 for 7 days. After the infection period, 0.2% saponin was added to promote cell lysis and recovery of bacteria. The bacterial suspension was diluted in 1X PBS and distributed in 7H11 plates for CFU count after 21 days of incubation. Data are represented as absolute count (A) and index of recovered bacteria normalized for each individual, considering untreated cells as 100% (B). (C) Viability assessment was performed using a Live/DeadTM Fixable Violet Dead cells staining kit (Thermo Fisher Scientific) and detected using a FACS Canto II flow cytometer (BD Biosciences, San Diego, CA, USA) (n = 3). The production of IL-1β (D), TNF-α (E), IFN-γ (F), and IL-10 (G) were quantified in culture supernatants after 7 days of culture by ELISA. All data were analyzed by ANOVA for repeated samples, followed by Tukey’s post-hoc test, considering * p < 0.05 For cytokine analysis, ANOVA for repeated samples, followed by Tukey’s post-hoc test were performed comparing decitabine to controls in infected conditions, considering # p < 0.05. (H) Figure summarizing the immunomodulation exerted by the hypomethylating agents during coculture of monocytes/macrophages and lymphocytes. The symbols within the bars of the graphs represent evaluated samples.

References

    1. Haniffa M., Bigley V., Collin M. Human mononuclear phagocyte system reunited. Semin Cell Dev. Biol. 2015;41:59–69. doi: 10.1016/j.semcdb.2015.05.004.
    1. Murray P.J., Wynn T.A. Protective and pathogenic functions of macrophage subsets. Nat. Rev. Immunol. 2011;11:723–737. doi: 10.1038/nri3073.
    1. Murray P.J. Macrophage polarization. Annu. Rev. Physiol. 2017;79:541–566. doi: 10.1146/annurev-physiol-022516-034339.
    1. de Groot A.E., Pienta K.J. Epigenetic control of macrophage polarization: Implications for targeting tumor-associated macrophages. Oncotarget. 2018;9:20908–20927. doi: 10.18632/oncotarget.24556.
    1. Enderlin Vaz da Silva Z., Lehr H.A., Velin D. In vitro and in vivo repair activities of undifferentiated and classically and alternatively activated macrophages. Pathobiology. 2014;81:86–93. doi: 10.1159/000357306.
    1. Gordon S., Martinez F.O. Alternative activation of macrophages: Mechanism and functions. Immunity. 2010;32:593–604. doi: 10.1016/j.immuni.2010.05.007.
    1. Fleming B.D., Mosser D.M. Regulatory macrophages: Setting the threshold for therapy. Eur. J. Immunol. 2011;41:2498–2502. doi: 10.1002/eji.201141717.
    1. Rőszer T. Understanding the mysterious m2 macrophage through activation markers and effector mechanisms. Mediators Inflamm. 2015;2015:816460. doi: 10.1155/2015/816460.
    1. Wilson C.B., Rowell E., Sekimata M. Epigenetic control of t-helper-cell differentiation. Nat. Rev. Immunol. 2009;9:91–105. doi: 10.1038/nri2487.
    1. Yang B.H., Floess S., Hagemann S., Deyneko I.V., Groebe L., Pezoldt J., Sparwasser T., Lochner M., Huehn J. Development of a unique epigenetic signature during in vivo th17 differentiation. Nucleic Acids Res. 2015;43:1537–1548. doi: 10.1093/nar/gkv014.
    1. Yang X., Wang X., Liu D., Yu L., Xue B., Shi H. Epigenetic regulation of macrophage polarization by DNA methyltransferase 3b. Mol. Endocrinol. 2014;28:565–574. doi: 10.1210/me.2013-1293.
    1. Cheng C., Huang C., Ma T.T., Bian E.B., He Y., Zhang L., Li J. Socs1 hypermethylation mediated by dnmt1 is associated with lipopolysaccharide-induced inflammatory cytokines in macrophages. Toxicol. Lett. 2014;225:488–497. doi: 10.1016/j.toxlet.2013.12.023.
    1. Kantarjian H., Issa J.P., Rosenfeld C.S., Bennett J.M., Albitar M., DiPersio J., Klimek V., Slack J., de Castro C., Ravandi F., et al. Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a phase iii randomized study. Cancer. 2006;106:1794–1803. doi: 10.1002/cncr.21792.
    1. Saba H.I. Decitabine in the treatment of myelodysplastic syndromes. Ther. Clin. Risk Manag. 2007;3:807–817.
    1. Frikeche J., Clavert A., Delaunay J., Brissot E., Grégoire M., Gaugler B., Mohty M. Impact of the hypomethylating agent 5-azacytidine on dendritic cells function. Exp. Hematol. 2011;39:1056–1063. doi: 10.1016/j.exphem.2011.08.004.
    1. Schmiedel B.J., Arélin V., Gruenebach F., Krusch M., Schmidt S.M., Salih H.R. Azacytidine impairs nk cell reactivity while decitabine augments nk cell responsiveness toward stimulation. Int. J. Cancer. 2011;128:2911–2922. doi: 10.1002/ijc.25635.
    1. Marcucci G., Silverman L., Eller M., Lintz L., Beach C.L. Bioavailability of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J. Clin. Pharmacol. 2005;45:597–602. doi: 10.1177/0091270004271947.
    1. Momparler R.L., Bouffard D.Y., Momparler L.F., Dionne J., Belanger K., Ayoub J. Pilot phase i-ii study on 5-aza-2’-deoxycytidine (decitabine) in patients with metastatic lung cancer. Anticancer Drugs. 1997;8:358–368. doi: 10.1097/00001813-199704000-00008.
    1. Espíndola M.S., Soares L.S., Galvão-Lima L.J., Zambuzi F.A., Cacemiro M.C., Brauer V.S., Marzocchi-Machado C.M., de Souza Gomes M., Amaral L.R., Martins-Filho O.A., et al. Epigenetic alterations are associated with monocyte immune dysfunctions in hiv-1 infection. Sci. Rep. 2018;8:5505. doi: 10.1038/s41598-018-23841-1.
    1. Kittan N.A., Allen R.M., Dhaliwal A., Cavassani K.A., Schaller M., Gallagher K.A., Carson W.F., Mukherjee S., Grembecka J., Cierpicki T., et al. Cytokine induced phenotypic and epigenetic signatures are key to establishing specific macrophage phenotypes. PLoS ONE. 2013;8:e78045. doi: 10.1371/journal.pone.0078045.
    1. Birkness K.A., Guarner J., Sable S.B., Tripp R.A., Kellar K.L., Bartlett J., Quinn F.D. An in vitro model of the leukocyte interactions associated with granuloma formation in mycobacterium tuberculosis infection. Immunol. Cell Biol. 2007;85:160–168. doi: 10.1038/sj.icb.7100019.
    1. Je S., Quan H., Na Y., Cho S.N., Kim B.J., Seok S.H. An in vitro model of granuloma-like cell aggregates substantiates early host immune responses against mycobacterium massiliense infection. Biol. Open. 2016;5:1118–1127. doi: 10.1242/bio.019315.
    1. Yamashiro L.H., Eto C., Soncini M., Horewicz V., Garcia M., Schlindwein A.D., Grisard E.C., Rovaris D.B., Báfica A. Isoniazid-induced control of mycobacterium tuberculosis by primary human cells requires interleukin-1 receptor and tumor necrosis factor. Eur. J. Immunol. 2016;46:1936–1947. doi: 10.1002/eji.201646349.
    1. Marino S., Cilfone N.A., Mattila J.T., Linderman J.J., Flynn J.L., Kirschner D.E. Macrophage polarization drives granuloma outcome during mycobacterium tuberculosis infection. Infect. Immun. 2015;83:324–338. doi: 10.1128/IAI.02494-14.
    1. Huang L., Nazarova E.V., Tan S., Liu Y., Russell D.G. Growth of mycobacterium tuberculosis in vivo segregates with host macrophage metabolism and ontogeny. J. Exp. Med. 2018;215:1135–1152. doi: 10.1084/jem.20172020.
    1. Lindblad K.E., Goswami M., Hourigan C.S., Oetjen K.A. Immunological effects of hypomethylating agents. Expert Rev. Hematol. 2017;10:745–752. doi: 10.1080/17474086.2017.1346470.
    1. Oliveira L.D.C. Efeito de Drogas Moduladoras da Estrutura da Cromatina Sobre a interação entre Macrófagos Murinos e Paracoccidioides Brasiliensis. [(accessed on 7 April 2021)]; Available online: .
    1. Kleinnijenhuis J., Oosting M., Joosten L.A., Netea M.G., Van Crevel R. Innate immune recognition of mycobacterium tuberculosis. Clin. Dev. Immunol. 2011;2011:405310. doi: 10.1155/2011/405310.
    1. Benakanakere M., Abdolhosseini M., Hosur K., Finoti L.S., Kinane D.F. Tlr2 promoter hypermethylation creates innate immune dysbiosis. J. Dent. Res. 2015;94:183–191. doi: 10.1177/0022034514557545.
    1. Oliveira N.F., Damm G.R., Andia D.C., Salmon C., Nociti F.H., Line S.R., de Souza A.P. DNA methylation status of the il8 gene promoter in oral cells of smokers and non-smokers with chronic periodontitis. J. Clin. Periodontol. 2009;36:719–725. doi: 10.1111/j.1600-051X.2009.01446.x.
    1. Dimberg J., Ström K., Löfgren S., Zar N., Lindh M., Matussek A. DNA promoter methylation status and protein expression of interleukin-8 in human colorectal adenocarcinomas. Int. J. Colorectal Dis. 2012;27:709–714. doi: 10.1007/s00384-011-1367-5.
    1. Kurashima K., Mukaida N., Fujimura M., Yasui M., Nakazumi Y., Matsuda T., Matsushima K. Elevated chemokine levels in bronchoalveolar lavage fluid of tuberculosis patients. Am. J. Respir. Crit. Care Med. 1997;155:1474–1477. doi: 10.1164/ajrccm.155.4.9105097.
    1. Sadek M.I., Sada E., Toossi Z., Schwander S.K., Rich E.A. Chemokines induced by infection of mononuclear phagocytes with mycobacteria and present in lung alveoli during active pulmonary tuberculosis. Am. J. Respir. Cell Mol. Biol. 1998;19:513–521. doi: 10.1165/ajrcmb.19.3.2815.
    1. Krupa A., Fol M., Dziadek B.R., Kepka E., Wojciechowska D., Brzostek A., Torzewska A., Dziadek J., Baughman R.P., Griffith D., et al. Binding of cxcl8/il-8 to mycobacterium tuberculosis modulates the innate immune response. Mediators Inflamm. 2015;2015:124762. doi: 10.1155/2015/124762.
    1. Schmid M.C., Varner J.A. Myeloid cells in the tumor microenvironment: Modulation of tumor angiogenesis and tumor inflammation. J. Oncol. 2010;2010:201026. doi: 10.1155/2010/201026.
    1. Zheng T., Ma G., Tang M., Li Z., Xu R. Il-8 secreted from m2 macrophages promoted prostate tumorigenesis via stat3/malat1 pathway. Int. J. Mol. Sci. 2018;20:98. doi: 10.3390/ijms20010098.
    1. Zhou D., Yang K., Chen L., Zhang W., Xu Z., Zuo J., Jiang H., Luan J. Promising landscape for regulating macrophage polarization: Epigenetic viewpoint. Oncotarget. 2017;8:57693–57706. doi: 10.18632/oncotarget.17027.
    1. Wang X., Cao Q., Yu L., Shi H., Xue B. Epigenetic regulation of macrophage polarization and inflammation by DNA methylation in obesity. JCI Insight. 2016;1:e87748. doi: 10.1172/jci.insight.87748.
    1. Thangavel J., Samanta S., Rajasingh S., Barani B., Xuan Y.T., Dawn B., Rajasingh J. Epigenetic modifiers reduce inflammation and modulate macrophage phenotype during endotoxemia-induced acute lung injury. J. Cell Sci. 2015;128:3094–3105. doi: 10.1242/jcs.170258.
    1. Jeong H.Y., Kang W.S., Hong M.H., Jeong H.C., Shin M.G., Jeong M.H., Kim Y.S., Ahn Y. 5-azacytidine modulates interferon regulatory factor 1 in macrophages to exert a cardioprotective effect. Sci. Rep. 2015;5:15768. doi: 10.1038/srep15768.
    1. Patel U., Rajasingh S., Samanta S., Cao T., Dawn B., Rajasingh J. Macrophage polarization in response to epigenetic modifiers during infection and inflammation. Drug Discov. Today. 2017;22:186–193. doi: 10.1016/j.drudis.2016.08.006.
    1. Joshi A.D., Oak S.R., Hartigan A.J., Finn W.G., Kunkel S.L., Duffy K.E., Das A., Hogaboam C.M. Interleukin-33 contributes to both m1 and m2 chemokine marker expression in human macrophages. BMC Immunol. 2010;11:52. doi: 10.1186/1471-2172-11-52.
    1. Tatano Y., Shimizu T., Tomioka H. Unique macrophages different from m1/m2 macrophages inhibit t cell mitogenesis while upregulating th17 polarization. Sci. Rep. 2014;4:4146. doi: 10.1038/srep04146.
    1. Piñeros A.R., Campos L.W., Fonseca D.M., Bertolini T.B., Gembre A.F., Prado R.Q., Alves-Filho J.C., Ramos S.G., Russo M., Bonato V.L. M2 macrophages or il-33 treatment attenuate ongoing mycobacterium tuberculosis infection. Sci. Rep. 2017;7:41240. doi: 10.1038/srep41240.
    1. Wang X., Wang J., Yu Y., Ma T., Chen P., Zhou B., Tao R. Decitabine inhibits t cell proliferation via a novel tet2-dependent mechanism and exerts potent protective effect in mouse auto- and allo-immunity models. Oncotarget. 2017;8:56802–56815. doi: 10.18632/oncotarget.18063.
    1. Fagone P., Mazzon E., Chikovani T., Saraceno A., Mammana S., Colletti G., Mangano K., Bramanti P., Nicoletti F. Decitabine induces regulatory t cells, inhibits the production of ifn-gamma and il-17 and exerts preventive and therapeutic efficacy in rodent experimental autoimmune neuritis. J. Neuroimmunol. 2018;321:41–48. doi: 10.1016/j.jneuroim.2018.05.013.

Source: PubMed

3
订阅