Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Origin, precursors and differentiation of mouse dendritic cells

Key Points

  • Definition of the developmental origin and precursors of dendritic cells (DCs) is essential to assess whether DC functional diversity results from the existence of developmentally independent DC subpopulations or from environmentally regulated acquisition of specific functions by DC subsets that share a common differentiation origin.

  • Defining the origin of DCs will help us to understand the induction and control of immunity by DCs, as well as the use of DCs for immunotherapy.

  • New data on the origin and differentiation of DCs depend on a large diversity of experimental approaches including in vivo DC reconstitution experiments, analysis of genetically deficient mice, in vitro DC differentiation assays and studies of the recruitment of DC precursors and DC differentiation during microbial infections.

  • The concept that mouse CD8 and CD8+ DCs correspond to myeloid and lymphoid DCs, respectively, has been proven to be incorrect, as both subsets can be generated by either lymphoid- or myeloid-committed progenitors.

  • However, neither the proposed lymphoid origin for plasmacytoid DCs nor the existence of a developmentally restricted lymphoid DC subset have been formally shown, so far.

  • The description of a population of DC precursors in mice that can generate CD8 and CD8+ DCs, as well as B220+ plasmacytoid CDs, supports the hypothesis that all DC subsets are derived from a single DC common progenitor.

  • New data support the concept that induction of T-cell immunity against microbial infections involves the recruitment of DC precursors to lymphoid tissues and their local differentiation into DCs.

Abstract

Functional specialization allows defined dendritic-cell (DC) subsets to induce efficient defence mechanisms against pathogens and tumour cells, and maintain T-cell tolerance by inducing the inactivation of autoreactive T cells. A crucial question, which has important implications for both our understanding of the induction and control of immunity by DCs, as well as the use of DCs for immunotherapy, is whether the functional diversity of DCs results from the existence of developmentally independent DC subpopulations, or whether DC subsets that share a common differentiation origin acquire specific functions in response to environmental signals. This review discusses recent findings on mouse DC development.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Theoretical model of the developmental origin of mouse dendritic cells.
Figure 2: Theoretical model of CD8 and CD8+ dendritic-cell differentiation, activation and function.

Similar content being viewed by others

References

  1. Banchereau, B. et al. Immunobiology of dendritic cells. Annu. Rev. Immunol. 18, 767–811 (2000).

    Article  CAS  PubMed  Google Scholar 

  2. Fong, L. & Engleman, E. G. Dendritic cells in cancer immunotherapy. Annu. Rev. Immunol. 18, 245–273 (2000).

    Article  CAS  PubMed  Google Scholar 

  3. Jonuleit, H., Schmitt, E., Steinbrink, K. & Enk, A. H. Dendritic cells as a tool to induce anergic and regulatory T cells. Trends Immunol. 22, 394–400 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Shortman, K. & Liu, Y. -J. Mouse and human dendritic cell subtypes. Nature Rev. Immunol. 2, 151–161 (2002).

    Article  CAS  Google Scholar 

  5. Martín, P. et al. Concept of lymphoid versus myeloid dendritic cell lineages revisited: both CD8α and CD8α+ dendritic cells are generated from CD4low lymphoid-committed precursors. Blood 96, 2511–2519 (2000). A report showing that CD4low early thymic precursors can generate both CD8 and CD8+ DCs, invalidating the hypothesis that CD8 and CD8+ DCs are myeloid and lymphoid DCs, respectively.

    PubMed  Google Scholar 

  6. Hochrein, H. et al. Differential production of IL-12, IFN-α, and IFN-γ by mouse dendritic cell subsets. J. Immunol. 166, 5448–5455 (2001).

    Article  CAS  PubMed  Google Scholar 

  7. Kamath, A. T., Henri, S., Battye, F., Tough, D. F. & Shortman, K. Developmental kinetics and lifespan of dendritic cells in mouse lymphoid organs. Blood 100, 1734–1741 (2002).

    CAS  PubMed  Google Scholar 

  8. Anjuère, F. et al. Definition of dendritic cell subpopulations present in the spleen, Peyer's patches, lymph nodes and skin of the mouse. Blood 93, 590–598 (1999).

    PubMed  Google Scholar 

  9. Asselin-Paturel, C. et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nature Immunol. 2, 1144–1150 (2001). This paper defined the phenotypic and functional features of the mouse equivalent of human plasmacytoid DCs, which are characterized by their capacity to secrete large amounts of type-1 interferon (IFN) after stimulation with virus.

    Article  CAS  Google Scholar 

  10. Martín, P. et al. Characterization of a new subpopulation of mouse CD8α+ B220+ dendritic cells endowed with type 1 interferon production capacity and tolerogenic potential. Blood 100, 383–390 (2002). This report shows the capacity of mouse B220+ DCs, which are claimed to be the mouse equivalent of human plasmacytoid DCs, to induce the differentiation of T regulatory cells, indicating a role for these cells in the induction of T-cell tolerance.

    Article  PubMed  Google Scholar 

  11. Bilsborough, J., George, T. C., Norment, A. M. & Viney, J. L. Mucosal CD8α+ DC, with a plasmacytoid phenotype, induce differentiation and support function of T cells with regulatory properties. Immunology 108, 481–492 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Julia, V. et al. A restricted subset of dendritic cells captures airborne antigens and remains able to activate specific T cells long after antigen exposure. Immunity 16, 271–283 (2002).

    Article  CAS  PubMed  Google Scholar 

  13. Akbari, O., Dekruyff, R. H. & Umetsu, D. T. Pulmonary dendritic cells producing IL-10 mediate toerance induced by respiratory exposure to antigen. Nature Immunol. 2, 725–731 (2001).

    Article  CAS  Google Scholar 

  14. Hart, D. N. J. Dendritic cells: unique leukocyte populations which control the primary immune response. Blood 90, 3245–3287 (1997).

    CAS  PubMed  Google Scholar 

  15. van Furth, R. & Cohn, Z. A. The origin and kinetics of mononuclear phagocytes. J. Exp. Med. 128, 415 (1968).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Inaba, K. et al. 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 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Sallusto, F. & Lanzavecchia, A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor-α. J. Exp. Med. 179, 1109–1118 (1994). The first report to describe the generation of DCs from human monocytes in vitro in the presence of granulocyte–macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL–4).

    Article  CAS  PubMed  Google Scholar 

  18. Randolph, G. J., Inaba, K., Robbiani, D. F., Steinman, R. M. & Muller, W. A. Differentiation of phagocytic monocytes into lymph node dendritic cells in vivo. Immunity 11, 753–761 (1999).

    Article  CAS  PubMed  Google Scholar 

  19. Ardavín, C., Wu, L., Li, C. L. & Shortman, K. Thymic dendritic cells and T cells develop simultaneously in the thymus from a common precursor population. Nature 362, 761–763 (1993). A description of thymic DCs that are derived from CD4low early thymic precursors and are devoid of myeloid-reconstitution potential. This led to the concept that some DCs could be of lymphoid origin.

    Article  PubMed  Google Scholar 

  20. Wu, L., Li, C. L. & Shortman, K. Thymic dendritic cell precursors: relationship to the T lymphocyte lineage and phenotype of the dendritic cell progeny. J. Exp. Med. 184, 903–911 (1996).

    Article  CAS  PubMed  Google Scholar 

  21. Wu, L., Nichogiannopoulou, A., Shortman, K. & Georgopoulos, K. Cell-autonomous defects in dendritic cell populations of Ikaros mutant mice point to a developmental relationship with the lymphoid lineage. Immunity 7, 483–492 (1997).

    Article  CAS  PubMed  Google Scholar 

  22. Wu, L. et al. RelB is essential for the development of myeloid-related CD8α dendritic cells but not of lymphoid-related CD8α+ dendritic cells. Immunity 9, 839–847 (1998).

    Article  CAS  PubMed  Google Scholar 

  23. Traver, D. et al. Development of CD8α-positive dendritic cells from a common myeloid progenitor. Science 290, 2152–2154 (2000). This report shows that both bone-marrow common lymphoid progenitors and common myeloid progenitors have the capacity to generate CD8 and CD8+ DCs.

    Article  CAS  PubMed  Google Scholar 

  24. Manz, M. G., Traver, D., Miyamoto, T., Weissman, I. L. & Akashi, K. Dendritic cell potentials of early lymphoid and myeloid progenitors. Blood 97, 3333–3341 (2001).

    Article  CAS  PubMed  Google Scholar 

  25. Wu, L. et al. Development of thymic and splenic dendritic cell populations from different hemopoietic precursors. Blood 98, 3376–3382 (2001).

    Article  CAS  PubMed  Google Scholar 

  26. D'Amico, A. & Wu, L. The early progenitors of mouse dendritic cells and plasmacytoid pre-dendritic cells are within the bone marrow hemopoietic precursors expressing FLT3. J. Exp. Med. (in the press).

  27. Corcoran, L. et al. The lymphoid past of mouse plasmacytoid cells and thymic dendritic cells. J. Immunol. (in the press).

  28. Hacker, C. et al. Transcriptional profiling identifies Id2 function in dendritic cell develoment. Nature Immunol. 4, 380–386 (2003). References 28, 34 and 35 describe mice that are deficient in inhibitor of DNA binding 2 (Id2) or IFN consensus sequence binding protein (ICSBP), which both have a marked defect in the differentiation of CD8+ DCs, as well as a defective DC maturation process, supporting the concept that CD8+ DCs result from the differentiation and/or activation of CD8 DCs.

    Article  CAS  Google Scholar 

  29. Georgopoulos, K. et al. The Ikaros gene is required for the development of all lymphoid lineages. Cell 79, 143–156 (1994).

    Article  CAS  PubMed  Google Scholar 

  30. Rodewald, H. R., Brocker, T. & Haller, C. Developmental dissociation of thymic dendritic cell and thymocyte lineages revealed in growth factor receptor mutant mice. Proc. Natl Acad. Sci. USA 96, 15068–15073 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Radtke, F. et al. Notch1 deficiency dissociates the intrathymic development of dendritic cells and T cells. J. Exp. Med. 191, 1085–1093 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Guerriero, A., Langmuir, P. B., Spain, L. M. & Scott, E. W. PU.1 is required for myeloid-derived but not lymphoid-derived dendritic cells. Blood 95, 879–885 (2000).

    CAS  PubMed  Google Scholar 

  33. Anderson, K. L. et al. Transcription factor PU.1 is necessary for development of thymic and myeloid progenitor-derived dendritic cells. J. Immunol. 164, 1855–1861 (2000).

    Article  CAS  PubMed  Google Scholar 

  34. Aliberti, J. et al. Essential role for ICSBP in the in vivo development of murine CD8α+ dendritic cells. Blood 101, 305–310 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Schiavoni, G. et al. ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8α+ dendritic dells. J. Exp. Med. 196, 1415–1425 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zhang, Y. et al. Bifurcated dendritic cell differentiation in vitro from murine lineage phenotype-negative c-kit+ bone marrow hematopoietic progenitor cells. Blood 92, 118–128 (1998).

    CAS  PubMed  Google Scholar 

  37. Zhang, Y. et al. Transforming growth factor-β1 polarizes murine hematopoietic progenitor cells to generate Langerhans cell-like dendritic cells through a monocyte/macrophage differentiation pathway. Blood 93, 1208–1220 (1999).

    CAS  PubMed  Google Scholar 

  38. Merad, M., Fong, L., Bogenberger, J. & Engleman, E. G. Differentiation of myeloid dendritic cells into CD8α+ dendritic cells in vivo. Blood 96, 1865–1872 (2000).

    CAS  PubMed  Google Scholar 

  39. 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, 3029–3039 (2000). References 39 and 40 describe the differentiation of DCs from bone-marrow cultures in the presence of fms-related tyrosine kinase 3 ligand (FLT3L), representing an alternative culture method for the generation of DCs that are similar to those found in vivo , including plasmacytoid-like DCs.

    CAS  PubMed  Google Scholar 

  40. Brawand, P. et al. Murine plasmacytoid pre-dendritic cells generated from FLT3 ligand-supplemented bone marrow cultures are immature APCs. J. Immunol. 169, 6711–6719 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. Gilliet, M. et al. The development of murine plasmacytoid dendritic cell precursors is differentially regulated by FLT3-ligand and granulocyte/macrophage colony-stimulating factor. J. Exp. Med. 195, 953–958 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Boonstra, A. et al. Flexibility of mouse classical and plasmacytoid-derived dendritic cells in directing T helper type 1 and 2 cell development: dependency on antigen dose and differential Toll-like receptor ligation. J. Exp. Med. 197, 101–109 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ardavín, C. et al. Origin and differentiation of dendritic cells. Trends Immunol. 22, 691–700 (2001).

    Article  PubMed  Google Scholar 

  44. McKenna, H. J. et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood 95, 3489–3497 (2000).

    CAS  PubMed  Google Scholar 

  45. Borkowski, T. A., Letterio, J. J., Farr, A. G. & Udey, M. C. A role for endogenous transforming growth factor β1 in Langerhans cell biology: the skin of transforming growth factor β1 null mice is devoid of epidermal Langerhans cells. J. Exp. Med. 184, 2417–2422 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Merad, M. et al. Langerhans cells renew in the skin throughout life under steady-state conditions. Nature Immunol. 3, 1135–1141 (2002).

    Article  CAS  Google Scholar 

  47. Grouard, G. et al. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 6, 1101–1111 (1997).

    Article  Google Scholar 

  48. Res, P. C. M., Couwenberg, F., Vyth-Dreese, F. A. & Spits, H. Expression of pTα mRNA in a committed dendritic cell precursor in the human thymus. Blood 94, 2647–2657 (1999).

    CAS  PubMed  Google Scholar 

  49. Spits, H., Couwenberg, F., Bakker, A. Q., Weijer, K. & Uttenbogaart, C. H. Id2 and id3 inhibit development of CD34+ stem cells into predendritic cell (pre-DC)2 but not into pre-DC1: evidence for a lymphoid origin of pre-DC2. J. Exp. Med. 192, 1775–1783 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Schotte, R. et al. The transcription factor Spi-B is expressed in plasmacytoid DC precursors and inhibits T-, B-, and NK-cell development. Blood 101, 1015–1023 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Martínez del Hoyo, G. et al. Characterization of a common precursor population for dendritic cells. Nature 415, 1043–1047 (2002). The first description of a DC-restricted precursor population present in mouse blood that can fully reconstitute splenic CD8, CD8+ and plasmacytoid B220+ DC subpopulations, and is devoid of lymphoid- or myeloid-differentiation potential.

    Article  Google Scholar 

  52. Yoneyama, H. et al. Regulation by chemokines of circulating dendritic cell precursors, and the formation of portal tract-associated lymphoid tissue, in a granulomatous liver disease. J. Exp. Med. 193, 35–49 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Balázs, M., Martin, F., Zhou, T. & Kearney, J. F. Blood dendritic cells interact with splenic marginal zone B cells to initiate T-independent immune responses. Immunity 17, 341–352 (2002).

    Article  PubMed  Google Scholar 

  54. Leenen, P. J. M. et al. Heterogeneity of mouse spleen dendritic cells: in vivo phagocytic activity, expression of macrophage markers, and subpopulation turnover. J. Immunol. 160, 2166–2173 (1998).

    CAS  PubMed  Google Scholar 

  55. Iwasaki, A. & Kelsall, B. L. Localization of distinct Peyer's patch dendritic cell subsets and their recruitment by chemokines macrophage inflammatory protein (MIP)-3α, MIP-3β, and secondary lymphoid organ chemokine. J. Exp. Med. 191, 1381–1393 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. De Vinuesa, C. G. et al. Dendritic cells associated with plasmablast survival. Eur. J. Immunol. 29, 3712–3721 (1999).

    Article  Google Scholar 

  57. Pulendran, B., Banchereau, J., Maraskovsky, E. & Maliszewski, C. Modulating the immune response with dendritic cells and their growth factors. Trends Immunol. 22, 41–47 (2001).

    Article  CAS  PubMed  Google Scholar 

  58. Iyoda, T. et al. The CD8+ dendritic cell subset selectively endocytoses dying cells in culture and in vivo. J. Exp. Med. 195, 1289–1302 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Maldonado-Lopez, R. & Moser, M. Dendritic cell subsets and the regulation of TH1/TH2 responses. Semin. Immunol. 13, 275–282 (2001).

    Article  CAS  PubMed  Google Scholar 

  60. Manickasingham, S., Edwards, A. D., Schulz, O. & Reise Sousa, C. The ability of murine dendritic cell subsets to direct T helper cell differentiation is dependent on microbial signals. Eur. J. Immunol. 33, 101–107 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. Maldonado-López, R., Maliszewski, C., Urbain, J. & Moser, M. Cytokines regulate the capacity of CD8α+ and CD8α dendritic cells to prime TH1/TH2 cells in vivo. J. Immunol. 167, 4345–4350 (2001).

    Article  PubMed  Google Scholar 

  62. Fallarino, F. et al. CD40 ligand and CTLA-4 are reciprocally regulated in the TH1 cell proliferative response sustained by CD8+ dendritic cells. J. Immunol. 169, 1182–1188 (2002).

    Article  CAS  PubMed  Google Scholar 

  63. Ohteki, T. et al. Interleukin 12-dependent interferon-γ production by CD8α+ lymphoid dendritic cells. J. Exp. Med. 189, 1981–1986 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Maldonado-López, R. et al. CD8α+ and CD8α subclasses of dendritic cells direct the development of distinct T helper cells in vivo. J. Exp. Med. 189, 587–592 (1999).

    Article  PubMed  PubMed Central  Google Scholar 

  65. Den Haan, J. M. M., Lehar, S. M. & Bevan, M. J. CD8+ but not CD8 dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1695 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Den Haan, J. M. M. & Bevan, M. J. Constitutive versus activation-dependent cross-presentation of immune complexes by CD8+ and CD8 dendritic cells in vivo. J. Exp. Med. 196, 817–827 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Martínez del Hoyo, G., Martín, P., Fernández Arias, C., Rodríguez Marín, A. & Ardavín, C. CD8α+ dendritic cells originate from the CD8α dendritic cell subset by a maturation process involving CD8α, DEC-205 and CD24 upregulation. Blood 99, 999–1004 (2002).

    Article  PubMed  Google Scholar 

  68. Naik, S., Vremec, D., Wu, L., O'Keeffe, M. & Shortman, K. CD8α+ mouse spleen dendritic cells do not originate from the CD8 dendritic cell subset. Blood (in the press).

  69. Morón, G., Rueda, P., Casal, I. & Leclerc, C. CD8α CD11b+ dendritic cells present exogenous virus-like particles to CD8+ T cells and subsequently express CD8α and CD205 molecules. J. Exp. Med. 195, 1233–1245 (2002).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Morelli, A. E. et al. Internalization of circulating apoptotic cells by splenic marginal zone dendritic cells: dependence on complement receptors and effect on cytokine production. Blood 101, 611–620 (2003).

    Article  CAS  PubMed  Google Scholar 

  71. Aliberti, J. et al. CCR5 provides a signal for microbial induced production of IL-12 by CD8α dendritic cells. Nature Immunol. 1, 83–87 (2000).

    Article  CAS  Google Scholar 

  72. Steinman, R. M. & Cohn, Z. A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 137, 1142–1162 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Vremec, D., Pooley, J., Hochrein, H., Wu, L. & Shortman, K. CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J. Immunol. 164, 2978–2986 (2000).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

I would like to thank G. Martínez del Hoyo, V. Parrillas, M. López-Bravo and B. León for critical reading of this manuscript and help in its preparation, and K. Shortman, M. Zenke, M. Luisa Toribio and A. Corbí for helpful discussions. Our studies were funded by the European Commission, the Community of Madrid, Spain and the Ministry of Science and Technology of Spain.

Author information

Authors and Affiliations

Authors

Related links

Related links

DATABASES

LocusLink

B220

CD11c

CD4

CD8

c-kit

E2A

FLT3

FLT3L

GM-CSF

ICSBP

Id2

IFN-γ

Ikaros

IL-3

IL-4

IL-10

IL-12

LFA1

Notch1

PU.1

Relb

SCF

Spi-B

TGF-β

TNF

Glossary

DC RECONSTITUTION ASSAYS

Assays that allow the in vivo analysis of the differentiation potential of different precursor populations after transfer into irradiated recipients; the precursor progeny is subsequently studied by using a genetic marker that allows the detection of cells from donor or recipient origin.

BROMODEOXYURIDINE INCORPORATION ASSAY

(BrdU). An assay in which BrdU, a thymidine analogue, is incorporated into DNA during DNA replication. Cells that have incorporated BrdU, and presumably have divided, are then visualized using fluorescence-labelled BrdU-specific antibodies followed by flow cytometry.

PLASMACYTOID DCS

A subset of immature dendritic cells (DCs), originally described in humans, that are characterized by their capacity to produce high levels of type I interferon after stimulation with virus. They can differentiate into potent antigen-presenting cells after activation.

COLONY-FORMING UNIT EXPERIMENTS

These experiments address the differentiation capacity of haematopoietic precursors, by analysing the cell colonies that result from the proliferation and differentiation of single precursor cells, using in vivo transfer techniques or cell-culture systems.

COMMON LYMPHOID PROGENITORS

Bone-marrow clonogenic precursors that are committed to the lymphoid lineage, generating T cells, B cells and natural killer cells, but are devoid of myeloid differentiation potential.

COMMON MYELOID PROGENITORS

Bone-marrow clonogenic precursors that are committed to the myeloid lineage and give rise to megakaryocyte–erythrocyte or granulocyte–macrophage progenitors, but are devoid of lymphoid differentiation potential.

DC MATURATION

A process resulting from the engagement of activation receptors on dendritic cells (DCs), such as Fc receptors, Toll-like receptors, cytokine receptors or CD40, which involves the upregulation of MHC molecules and co-stimulatory molecules and the acquisition of specific functions that enable DCs to activate T cells efficiently.

DC PRECURSORS

(Pre-DCs). A population of dendritic cell (DC)-committed precursors present in mouse blood that can fully reconstitute the CD8, CD8+ and plasmacytoid B220+ DC subpopulations after transfer into irradiated recipients51. Precursor DCs are CD11c+ B220+CD11b+ CD43+CD44+ CD62L+ FcRγ+MHC class IICD19CD40CD86c-kit IL-7RαIL-3RαDEC205 F4/80Gr1.

CROSS-PRIMING

A mechanism by which an antigen-presenting cell processes exogenous cell-associated antigens and presents them in the context of MHC class I molecules, leading to the activation of antigen-specific CD8+ T cells.

DANGER SIGNALS

Cell-wall components and other products of pathogens that alert the innate immune system to the presence of potentially harmful invaders, usually by interacting with Toll-like receptors and other pattern recognition receptors expressed by tissue cells and dendritic cells, for example.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ardavín, C. Origin, precursors and differentiation of mouse dendritic cells. Nat Rev Immunol 3, 582–591 (2003). https://doi.org/10.1038/nri1127

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri1127

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing