Summary

Economical and Efficient Protocol for Isolating and Culturing Bone Marrow-derived Dendritic Cells from Mice

Published: July 01, 2022
doi:

Summary

Here, we present an economical and efficient method to isolate and generate high-purity bone marrow-derived dendritic cells from mice after 7 days of culture with 10 ng/mL GM-CSF/IL-4.

Abstract

The demand for dendritic cells (DCs) is gradually increasing as immunology research advances. However, DCs are rare in all tissues. The traditional method for isolating DCs primarily involves inducing bone marrow (BM) differentiation into DCs by injecting large doses (>10 ng/mL) of granulocyte-macrophage colony-stimulating factor/interleukin-4 (GM-CSF/IL-4), making the procedure complex and expensive. In this protocol, using all BM cells cultured in 10 ng/mL GM-CSF/IL-4 medium, after 3-4 half-culture exchanges, up to 2.7 x 107 CD11c+ cells (DCs) per mouse (two femurs) were harvested with a purity of 80%-95%. After 10 days in culture, the expression of CD11c, CD80, and MHC II increased, whereas the number of cells decreased. The number of cells peaked after 7 days of culture. Moreover, this method only took 10 min to harvest all bone marrow cells, and a high number of DCs were obtained after 1 week of culture.

Introduction

Dendritic cells (DCs) are the most powerful antigen-presenting cells (APCs) for activating naïve T cells and inducing specific cytotoxic T lymphocyte (CTL) responses against infectious diseases, allergy diseases, and tumor cells1,2,3. DCs are the primary link between innate immunity and adaptive immunity and play an essential role in immunological defense and the maintenance of immune tolerance. In the last 40 years, many researchers have sought to define the subsets of DCs and their functions in inflammation and immunity. As per those studies, DCs develop along the myeloid and lymphoid lineages from bone marrow cells. Tumor vaccines have gained significant milestones in recent years and have a promising future. Mechanically, tumor vaccines modulate the immune response and prevent tumor growth by activating cytotoxic T lymphocytes using tumor antigens. The vaccine based on DCs plays an important role in tumor immunotherapy and has been identified as one of the most promising anti-tumor therapies1,4. In addition, DCs have been widely used in the testing of new molecular-targeted drugs and immune checkpoint inhibitors5.

Researchers urgently need a high number of high-purity DCs to further study the role of DCs. However, DCs are rare in various tissues and blood, accounting for only 1% of blood cells in humans and animals. In vitro culture of bone marrow dendritic cells (BMDC) is an important method for obtaining large amounts of DC cells. Meanwhile, The Lutz protocol for generating DCs from bone marrow has been widely used by researchers6. Although the protocol is effective in obtaining DC cells, it is complex and expensive, involving the addition of high concentrations of cytokines and the lysis of red blood cells.

In this study, we report a method for isolating almost all bone marrow cells from mouse bone marrow (BM) and inducing differentiation into BMDC after 7-9 days of incubation in vitro, with a lower concentration of GM-CSF and IL-4. This procedure only takes 10 min to harvest almost all bone marrow cells and to suspend them in a complete medium. In brief, we provide an efficient and cost-effective culturing method for BMDC in this research.

Protocol

All procedures were approved by the Nanjing Medical University Animal Care and Use Committee. 1. Isolation of bone marrow and preparation of BM cells Sacrifice C57BL/6 mice (18-22 g, 6-8 weeks old) via CO2 asphyxiation. Fix the mouse on the mouse operating table. Disinfect the surfaces with 70% ethanol. Cut the skin of the leg to expose the muscles and femoral artery. Clamp and tear off the femoral artery using two forceps, then pull the proxim…

Representative Results

The 1 x 107-1.7 x 107 cells were extracted from two femurs and were re-suspended in 24 mL of medium before being planted in a 6-well plate (Figure 1A). After 2 days, non-adherent cells were removed by completely changing the culture medium. Before changing the medium, a significant number of suspended cells were observed (Figure 1B). After 3 days of culture, small cell colonies began to form. On the sixth day, the size and number of colonie…

Discussion

Humans and mice have different DC subsets, including classical DCs (cDCs, including cDC1s and cDC2s) plasmacytoid DCs (pDCs), and monocyte-derived DCs (MoDCs)9,10,11. It is generally accepted that cDC1s regulate cytotoxic T lymphocyte (CTL) responses to intracellular pathogens and cancer, and cDC2s regulate immune responses to extracellular pathogens, parasites, and allergens12. A significant number of DC…

Divulgaciones

The authors have nothing to disclose.

Acknowledgements

This work was supported by Program of Tianjin Science and Technology Plan (20JCQNJC00550), Tianjin Health Science and Technology Project (TJWJ202021QN033 and TJWJ202021QN034).

Materials

β-Mercaptoethanol Solarbio M8211
6-well plate Corning 3516
APC-MHC II Biolegend 116417
FBS Gibco 10100
PE-CD80 Biolegend 104707
Penicillin-Streptomycin Solarbio P1400
Percp/cy5.5-CD11c Biolegend 117327
PRMI-1640 Thermo 11875093
Recombinant Mouse GM-CSF Solarbio P00184
Recombinant Mouse IL-4 Solarbio P00196
TruStain Fc PLUS (anti-mouse CD16/32) Antibody Biolegend 156603

Referencias

  1. Huang, M. N., et al. Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses. Journal of Clinical Investigation. 130 (2), 774-788 (2020).
  2. Wang, P., Dong, S., Zhao, P., He, X., Chen, M. Direct loading of CTL epitopes onto MHC class I complexes on dendritic cell surface in vivo. Biomaterials. 182, 92-103 (2018).
  3. Banchereau, J., Steinman, R. M. Dendritic cells and the control of immunity. Nature. 392 (6673), 245-252 (1998).
  4. Jiang, P. L., et al. Galactosylated liposome as a dendritic cell-targeted mucosal vaccine for inducing protective anti-tumor immunity. Acta Biomaterialia. 11, 356-367 (2015).
  5. Shi, Y., et al. Next-generation immunotherapies to improve anticancer immunity. Frontiers in Pharmacology. 11, 566401 (2020).
  6. Lutz, M. B., et al. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. Journal of Immunological Methods. 223 (1), 77-92 (1999).
  7. Son, Y. I., et al. A novel bulk-culture method for generating mature dendritic cells from mouse bone marrow cells. Journal of Immunological Methods. 262 (1-2), 145-157 (2002).
  8. Guo, L., et al. Fusion protein vaccine based on Ag85B and STEAP1 induces a protective immune response against prostate cancer. Vaccines. 9 (7), 786 (2021).
  9. Olweus, J., et al. Dendritic cell ontogeny: A human dendritic cell lineage of myeloid origin. Proceedings of the National Academy of Sciences of the United States of America. 94 (23), 12551-12556 (1997).
  10. Martin, P., et al. Concept of lymphoid versus myeloid dendritic cell lineages revisited: both CD8alpha(-) and CD8alpha(+) dendritic cells are generated from CD4(low) lymphoid-committed precursors. Blood. 96 (-), 2511-2519 (2000).
  11. Anderson, D. A., Dutertre, C. A., Ginhoux, F., Murphy, K. M. Genetic models of human and mouse dendritic cell development and function. Nature Reviews: Immunology. 21 (2), 101-115 (2021).
  12. Vu Manh, T. P., Bertho, N., Hosmalin, A., Schwartz-Cornil, I., Dalod, M. Investigating evolutionary conservation of dendritic cell subset identity and functions. Frontiers in Immunology. 6, 260 (2015).
  13. Scheicher, C., Mehlig, M., Zecher, R., Reske, K. Dendritic cells from mouse bone marrow: in vitro differentiation using low doses of recombinant granulocyte-macrophage colony-stimulating factor. Journal of Immunological Methods. 154 (2), 253-264 (1992).
  14. Brasel, K., De Smedt, T., Smith, J. L., Maliszewski, C. R. Generation of murine dendritic cells from flt3-ligand-supplemented bone marrow cultures. Blood. 96 (9), 3029-3039 (2000).
  15. Mayordomo, J. I., et al. marrow-derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumour immunity. Nature Medicine. 1 (12), 1297-1302 (1995).
  16. Condon, C., Watkins, S. C., Celluzzi, C. M., Thompson, K., Falo, L. D. DNA-based immunization by in vivo transfection of dendritic cells. Nature Medicine. 2 (10), 1122-1128 (1996).
  17. Brunner, G. A., et al. Post-prandial administration of the insulin analogue insulin aspart in patients with type 1 diabetes mellitus. Diabetic Medicine. 17 (5), 371-375 (2000).
  18. Koido, S., et al. Induction of antitumor immunity by vaccination of dendritic cells transfected with MUC1 RNA. Journal of Immunology. 165 (10), 5713-5719 (2000).
  19. Jonasson, P. S., et al. Strength of the porcine proximal femoral epiphyseal plate: The effect of different loading directions and the role of the perichondrial fibrocartilaginous complex and epiphyseal tubercle – An experimental biomechanical study. Journal of Experimental Orthopaedics. 1 (1), 4 (2014).
  20. Labeur, M. S., et al. Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage. Journal of Immunology. 162 (1), 168-175 (1999).
  21. Hinkel, A., et al. Immunomodulatory dendritic cells generated from nonfractionated bulk peripheral blood mononuclear cell cultures induce growth of cytotoxic T cells against renal cell carcinoma. Journal of Immunotherapy. 23 (1), 83-93 (2000).
check_url/es/63125?article_type=t

Play Video

Citar este artículo
Tang, H., Xie, H., Wang, Z., Peng, S., Ni, W., Guo, L. Economical and Efficient Protocol for Isolating and Culturing Bone Marrow-derived Dendritic Cells from Mice. J. Vis. Exp. (185), e63125, doi:10.3791/63125 (2022).

View Video