DOI QR코드

DOI QR Code

GM-CSF Grown Bone Marrow Derived Cells Are Composed of Phenotypically Different Dendritic Cells and Macrophages

  • Na, Yi Rang (Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine) ;
  • Jung, Daun (Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine) ;
  • Gu, Gyo Jeong (Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine) ;
  • Seok, Seung Hyeok (Department of Microbiology and Immunology, and Institute of Endemic Disease, Seoul National University College of Medicine)
  • Received : 2016.06.28
  • Accepted : 2016.09.19
  • Published : 2016.10.31

Abstract

Granulocyte-macrophage colony stimulating factor (GM-CSF) has a role in inducing emergency hematopoiesis upon exposure to inflammatory stimuli. Although GM-CSF generated murine bone marrow derived cells have been widely used as macrophages or dendritic cells in research, the exact characteristics of each cell population have not yet been defined. Here we discriminated GM-CSF grown bone marrow derived macrophages (GM-BMMs) from dendritic cells (GM-BMDCs) in several criteria. After C57BL/6J mice bone marrow cell culture for 7 days with GM-CSF supplementation, two main populations were observed in the attached cells based on MHCII and F4/80 marker expressions. GM-BMMs had $MHCII^{low}F4/80^{high}$ as well as $CD11c^+CD11b^{high}CD80^-CD64^+MerTK^+$ phenotypes. In contrast, GM-BMDCs had $MHCII^{high}F4/80^{low}$ and $CD11c^{high}CD8{\alpha}^-CD11b^+CD80^+CD64^-MerTK^{low}$ phenotypes. Interestingly, the GM-BMM population increased but GM-BMDCs decreased in a GM-CSF dose-dependent manner. Functionally, GM-BMMs showed extremely high phagocytic abilities and produced higher IL-10 upon LPS stimulation. GM-BMDCs, however, could not phagocytose as well, but were efficient at producing $TNF{\alpha}$, $IL-1{\beta}$, IL-12p70 and IL-6 as well as inducing T cell proliferation. Finally, whole transcriptome analysis revealed that GM-BMMs and GM-BMDCs are overlap with in vivo resident macrophages and dendritic cells, respectively. Taken together, our study shows the heterogeneicity of GM-CSF derived cell populations, and specifically characterizes GM-CSF derived macrophages compared to dendritic cells.

Keywords

References

  1. Banchereau, J., and Steinman, R.M. (1998). Dendritic cells and the control of immunity. Nature 392, 245-252. https://doi.org/10.1038/32588
  2. Becker, L., Liu, N.C., Averill, M.M., Yuan, W., Pamir, N., Peng, Y., Irwin, A.D., Fu, X., Bornfeldt, K.E., and Heinecke, J.W. (2012). Unique proteomic signatures distinguish macrophages and dendritic cells. PLoS One 7, e33297. https://doi.org/10.1371/journal.pone.0033297
  3. Bhattacharya, P., Gopisetty, A., Ganesh, B.B., Sheng, J.R., and Prabhakar, B.S. (2011). GM-CSF-induced, bone-marrow-derived dendritic cells can expand natural Tregs and induce adaptive Tregs by different mechanisms. J. Leukocyte Biol. 89, 235-249. https://doi.org/10.1189/jlb.0310154
  4. Cebon, J., Layton, J.E., Maher, D., and Morstyn, G. (1994). Endogenous haemopoietic growth factors in neutropenia and infection. Br. J. Haematol. 86, 265-274. https://doi.org/10.1111/j.1365-2141.1994.tb04725.x
  5. Cheers, C., Haigh, A.M., Kelso, A., Metcalf, D., Stanley, E.R., and Young, A.M. (1988). Production of colony-stimulating factors (CSFs) during infection: separate determinations of macrophage-, granulocyte-, granulocyte-macrophage-, and multi-CSFs. Infect. Immun. 56, 247-251.
  6. Chung, S., Ranjan, R., Lee, Y.G., Park, G.Y., Karpurapu, M., Deng, J., Xiao, L., Kim, J.Y., Unterman, T.G., and Christman, J.W. (2015). Distinct role of FoxO1 in M-CSF- and GM-CSF-differentiated macrophages contributes LPS-mediated IL-10: implication in hyperglycemia. J. Leukocyte Biol. 97, 327-339. https://doi.org/10.1189/jlb.3A0514-251R
  7. Crozat, K., Guiton, R., Guilliams, M., Henri, S., Baranek, T., Schwartz-Cornil, I., Malissen, B., and Dalod, M. (2010). Comparative genomics as a tool to reveal functional equivalences between human and mouse dendritic cell subsets. Immunol. Rev. 234, 177-198. https://doi.org/10.1111/j.0105-2896.2009.00868.x
  8. Egawa, M., Mukai, K., Yoshikawa, S., Iki, M., Mukaida, N., Kawano, Y., Minegishi, Y., and Karasuyama, H. (2013). Inflammatory monocytes recruited to allergic skin acquire an anti-inflammatory M2 phenotype via basophil-derived interleukin-4. Immunity 38, 570-580. https://doi.org/10.1016/j.immuni.2012.11.014
  9. Fleetwood, A.J., Lawrence, T., Hamilton, J.A., and Cook, A.D. (2007). Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J. Immunol. 178, 5245-5252. https://doi.org/10.4049/jimmunol.178.8.5245
  10. Ganesh, B.B., Cheatem, D.M., Sheng, J.R., Vasu, C., and Prabhakar, B.S. (2009). GM-CSF-induced CD11c+CD8a--dendritic cells facilitate Foxp3+ and IL-10+ regulatory T cell expansion resulting in suppression of autoimmune thyroiditis. Int. Immunol. 21, 269-282. https://doi.org/10.1093/intimm/dxn147
  11. Gautier, E.L., Shay, T., Miller, J., Greter, M., Jakubzick, C., Ivanov, S., Helft, J., Chow, A., Elpek, K.G., Gordonov, S., et al. (2012). Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13, 1118-1128. https://doi.org/10.1038/ni.2419
  12. Hashimoto, D., Miller, J., and Merad, M. (2011). Dendritic cell and macrophage heterogeneity in vivo. Immunity 35, 323-335. https://doi.org/10.1016/j.immuni.2011.09.007
  13. Helft, J., Bottcher, J., Chakravarty, P., Zelenay, S., Huotari, J., Schraml, B.U., Goubau, D., and Reis e Sousa, C. (2015). GMCSF mouse bone marrow cultures comprise a heterogeneous population of CD11c(+)MHCII(+) macrophages and dendritic cells. Immunity 42, 1197-1211. https://doi.org/10.1016/j.immuni.2015.05.018
  14. Heng, T.S., Painter, M.W., and Immunological Genome Project, C. (2008). The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091-1094. https://doi.org/10.1038/ni1008-1091
  15. Hercus, T.R., Thomas, D., Guthridge, M.A., Ekert, P.G., King-Scott, J., Parker, M.W., and Lopez, A.F. (2009). The granulocytemacrophage colony-stimulating factor receptor: linking its structure to cell signaling and its role in disease. Blood 114, 1289-1298. https://doi.org/10.1182/blood-2008-12-164004
  16. Inaba, K., Inaba, M., Deguchi, M., Hagi, K., Yasumizu, R., Ikehara, S., Muramatsu, S., and Steinman, R.M. (1993). Granulocytes, macrophages, and dendritic cells arise from a common major histocompatibility complex class II-negative progenitor in mouse bone marrow. Proc. Natl. Acad. Sci. USA 90, 3038-3042. https://doi.org/10.1073/pnas.90.7.3038
  17. Mellman, I., and Steinman, R.M. (2001). Dendritic cells: specialized and regulated antigen processing machines. Cell 106, 255-258. https://doi.org/10.1016/S0092-8674(01)00449-4
  18. Miller, J.C., Brown, B.D., Shay, T., Gautier, E.L., Jojic, V., Cohain, A., Pandey, G., Leboeuf, M., Elpek, K.G., Helft, J., et al. (2012). Deciphering the transcriptional network of the dendritic cell lineage. Nat. Immunol. 13, 888-899. https://doi.org/10.1038/ni.2370
  19. Murray, P.J., Allen, J.E., Biswas, S.K., Fisher, E.A., Gilroy, D.W., Goerdt, S., Gordon, S., Hamilton, J.A., Ivashkiv, L.B., Lawrence, T., et al. (2014). Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14-20. https://doi.org/10.1016/j.immuni.2014.06.008
  20. Nikolic, T., de Bruijn, M.F., Lutz, M.B., and Leenen, P.J. (2003). Developmental stages of myeloid dendritic cells in mouse bone marrow. Int. Immunol. 15, 515-524. https://doi.org/10.1093/intimm/dxg050
  21. Paine, R., 3rd, Morris, S.B., Jin, H., Wilcoxen, S.E., Phare, S.M., Moore, B.B., Coffey, M.J., and Toews, G.B. (2001). Impaired functional activity of alveolar macrophages from GM-CSF-deficient mice. Am. J. Physiol. Lung Cell. Mol. Physiol. 281, L1210-1218. https://doi.org/10.1152/ajplung.2001.281.5.L1210
  22. Robbins, S.H., Walzer, T., Dembele, D., Thibault, C., Defays, A., Bessou, G., Xu, H., Vivier, E., Sellars, M., Pierre, P., et al. (2008). Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol. 9, R17. https://doi.org/10.1186/gb-2008-9-1-r17
  23. Saraiva, M., and O'Garra, A. (2010). The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 10, 170-181. https://doi.org/10.1038/nri2711
  24. Satpathy, A.T., Wu, X., Albring, J.C., and Murphy, K.M. (2012). Re(de)fining the dendritic cell lineage. Nat. Immunol. 13, 1145-1154. https://doi.org/10.1038/ni.2467
  25. Seok, S.H., Heo, J.I., Hwang, J.H., Na, Y.R., Yun, J.H., Lee, E.H., Park, J.W., and Cho, C.H. (2013). Angiopoietin-1 elicits pro-inflammatory responses in monocytes and differentiating macrophages. Mol. Cells 35, 550-556. https://doi.org/10.1007/s10059-013-0088-8
  26. Xu, Y., Zhan, Y., Lew, A.M., Naik, S.H., and Kershaw, M.H. (2007). Differential development of murine dendritic cells by GM-CSF versus Flt3 ligand has implications for inflammation and trafficking. J. Immunol. 179, 7577-7584. https://doi.org/10.4049/jimmunol.179.11.7577
  27. Zhang, Y., Harada, A., Wang, J.B., Zhang, Y.Y., Hashimoto, S., Naito, M., and Matsushima, K. (1998). Bifurcated dendritic cell differentiation in vitro from murine lineage phenotype-negative c-kit+ bone marrow hematopoietic progenitor cells. Blood 92, 118-128.

Cited by

  1. Loss of lipid phosphatase SHIP1 promotes macrophage differentiation through suppression of dendritic cell differentiation pp.1555-8576, 2018, https://doi.org/10.1080/15384047.2018.1523846
  2. GM-CSF Quantity Has a Selective Effect on Granulocytic vs. Monocytic Myeloid Development and Function vol.9, pp.1664-3224, 2018, https://doi.org/10.3389/fimmu.2018.01922
  3. Integrated Network Pharmacology and Metabolomics Analysis of the Therapeutic Effects of Zi Dian Fang on Immune Thrombocytopenic Purpura vol.9, pp.1663-9812, 2018, https://doi.org/10.3389/fphar.2018.00597
  4. Increased B7-H4 expression during esophageal squamous cell carcinogenesis is associated with IL-6/STAT3 signaling pathway activation in mice vol.13, pp.4, 2016, https://doi.org/10.3892/ol.2017.5688
  5. Serum amyloid A inhibits dendritic cell differentiation by suppressing GM-CSF receptor expression and signaling vol.49, pp.8, 2017, https://doi.org/10.1038/emm.2017.120
  6. Listeria monocytogenes Replicate in Bone Marrow–Derived CD11c+ Cells but Not in Dendritic Cells Isolated from the Murine Gastrointestinal Tract vol.199, pp.11, 2016, https://doi.org/10.4049/jimmunol.1700970
  7. Tolerogenic bone marrow-derived dendritic cells induce neuroprotective regulatory T cells in a model of Parkinson’s disease vol.13, pp.None, 2016, https://doi.org/10.1186/s13024-018-0255-7
  8. Stable incorporation of GM-CSF into dissolvable microneedle patch improves skin vaccination against influenza vol.276, pp.None, 2018, https://doi.org/10.1016/j.jconrel.2018.02.033
  9. Development of endocytosis, degradative activity, and antigen processing capacity during GM-CSF driven differentiation of murine bone marrow vol.13, pp.5, 2018, https://doi.org/10.1371/journal.pone.0196591
  10. Androgen and Androgen Receptor as Enhancers of M2 Macrophage Polarization in Allergic Lung Inflammation vol.201, pp.10, 2018, https://doi.org/10.4049/jimmunol.1800352
  11. GM-CSF-Dependent Inflammatory Pathways vol.10, pp.None, 2016, https://doi.org/10.3389/fimmu.2019.02055
  12. The Pleiotropic Effects of the GM-CSF Rheostat on Myeloid Cell Differentiation and Function: More Than a Numbers Game vol.10, pp.None, 2016, https://doi.org/10.3389/fimmu.2019.02679
  13. MSU Crystals Enhance TDB-Mediated Inflammatory Macrophage IL-1β Secretion vol.42, pp.3, 2019, https://doi.org/10.1007/s10753-019-00976-5
  14. In vitro cellular responses to Neospora caninum glycosylphosphatidylinositols depend on the host origin of antigen presenting cells vol.119, pp.None, 2016, https://doi.org/10.1016/j.cyto.2019.03.014
  15. Downregulation of MHC Class II by Ubiquitination Is Required for the Migration of CD206+ Dendritic Cells to Skin-Draining Lymph Nodes vol.203, pp.11, 2019, https://doi.org/10.4049/jimmunol.1900593
  16. Quantitative MRI cell tracking of immune cell recruitment to tumors and draining lymph nodes in response to anti-PD-1 and a DPX-based immunotherapy vol.9, pp.1, 2016, https://doi.org/10.1080/2162402x.2020.1851539
  17. GM-CSF in inflammation vol.217, pp.1, 2020, https://doi.org/10.1084/jem.20190945
  18. Mouse T cell priming is enhanced by maturation-dependent stiffening of the dendritic cell cortex vol.9, pp.None, 2016, https://doi.org/10.7554/elife.55995
  19. In vitro Chicken Bone Marrow-Derived Dendritic Cells Comprise Subsets at Different States of Maturation vol.11, pp.None, 2020, https://doi.org/10.3389/fimmu.2020.00141
  20. TLR2 and Dectin-1 Signaling in Mouse Hematopoietic Stem and Progenitor Cells Impacts the Ability of the Antigen Presenting Cells They Produce to Activate CD4 T Cells vol.9, pp.5, 2016, https://doi.org/10.3390/cells9051317
  21. Allosteric Inhibition of SHP2 Stimulates Antitumor Immunity by Transforming the Immunosuppressive Environment vol.80, pp.13, 2016, https://doi.org/10.1158/0008-5472.can-19-3038
  22. Granulocyte/Macrophage Colony-Stimulating Factor-Derived Macrophages Exhibit Distinctive Early Immune Response to Lymphocytic Choriomeningitis Virus Infection vol.33, pp.6, 2020, https://doi.org/10.1089/vim.2019.0178
  23. Control of GM-CSF–Dependent Dendritic Cell Differentiation and Maturation by DEF6 and SWAP-70 vol.205, pp.5, 2020, https://doi.org/10.4049/jimmunol.2000020
  24. Toll-Like Receptor 2-Tpl2-Dependent ERK Signaling Drives Inverse Interleukin 12 Regulation in Dendritic Cells and Macrophages vol.89, pp.1, 2016, https://doi.org/10.1128/iai.00323-20
  25. Integrin Alpha E (CD103) Limits Virus-Induced IFN-I Production in Conventional Dendritic Cells vol.11, pp.None, 2016, https://doi.org/10.3389/fimmu.2020.607889
  26. Carbomer-based adjuvant elicits CD8 T-cell immunity by inducing a distinct metabolic state in cross-presenting dendritic cells vol.17, pp.1, 2016, https://doi.org/10.1371/journal.ppat.1009168
  27. Mutation in Irf8 Gene (Irf8R294C) Impairs Type I IFN-Mediated Antiviral Immune Response by Murine pDCs vol.12, pp.None, 2016, https://doi.org/10.3389/fimmu.2021.758190
  28. MRP8/14 mediates macrophage efferocytosis through RAGE and Gas6/MFG‐E8, and induces polarization via TLR4‐dependent pathway vol.236, pp.2, 2016, https://doi.org/10.1002/jcp.29944
  29. Complement-Opsonized Nano-Carriers Are Bound by Dendritic Cells (DC) via Complement Receptor (CR)3, and by B Cell Subpopulations via CR-1/2, and Affect the Activation of DC and B-1 Cells vol.22, pp.6, 2016, https://doi.org/10.3390/ijms22062869
  30. Differential TLR7-mediated cytokine expression by R848 in M-CSF- versus GM-CSF-derived macrophages after LCMV infection vol.102, pp.3, 2016, https://doi.org/10.1099/jgv.0.001541
  31. Combination Adjuvants Affect the Magnitude of Effector-Like Memory CD8 T Cells and Protection against Listeriosis vol.89, pp.7, 2016, https://doi.org/10.1128/iai.00768-20
  32. Crosstalk between Metabolic Disorders and Immune Cells vol.22, pp.18, 2016, https://doi.org/10.3390/ijms221810017
  33. Serum amyloid A delivers retinol to intestinal myeloid cells to promote adaptive immunity vol.373, pp.6561, 2016, https://doi.org/10.1126/science.abf9232
  34. The Sympathetic Nervous System Modulates Cancer Vaccine Activity through Monocyte-Derived Cells vol.207, pp.12, 2021, https://doi.org/10.4049/jimmunol.2100719
  35. IRF8 and BATF3 interaction enhances the cDC1 specific Pfkfb3 gene expression vol.371, pp.None, 2016, https://doi.org/10.1016/j.cellimm.2021.104468