Fluorine-18 labeled mouse bone marrow-derived dendritic cells can be detected in vivo by high resolution projection imaging

https://doi.org/10.1016/S0022-1759(01)00528-2Get rights and content

Abstract

Immunization with ex vivo generated dendritic cells has become a focus for many clinical applications. The optimal site of injection and the migration pattern of these cells remain to be elucidated. We therefore developed a novel method for labeling mouse bone marrow-derived dendritic cells (BMDC) with the positron emitting radioisotope F-18 using N-succinimidyl-4-[F-18]fluorobenzoate, which covalently binds to the lysine residues of cell surface proteins. When we determined the stability of F-18 labeled BMDC, we found that at 4 h only 44±10% of the initial cell-bound activity was retained at 37 °C, whereas considerably more (91±3%) was retained at 4 °C. Labeled cells did not exhibit any significant alteration in cell viability or phenotype as determined by trypan blue exclusion and FACS analysis 24 h after radiolabeling. Furthermore, F-18-labeled BMDC stimulated allogeneic T cells in a mixed leukocyte reaction as potently as did sham-treated BMDC and migrated towards secondary lymphoid tissue chemokine (SLC) in a chemotaxis assay in vitro with the same efficiency as sham-treated BMDC. Migration of F-18-labeled BMDC was studied after footpad injection by (1) ex vivo counting of dissected tissues using a gamma counter and (2) in vivo by imaging mice with PiPET, a 2-mm resolution positron projection imager. After 4 h, the ratio between measured activity in draining vs. contralateral (D/C) lymph nodes (LN) was 166±96 (n=7) in the case of live cell injections, whereas if we injected heat-killed F-18-labeled BMDC or F-18-labeled macrophages the D/C ratios were 17±2 (n=2) and 14±4 (n=2), respectively. Injection of cell-free activity in the form of F-18-labeled 4-fluorobenzoic acid resulted in a D/C ratio of 7±2 (n=3), suggesting that the activity measured in the draining lymph node was associated with migrated F-18-labeled BMDC. When F-18-labeled live cells were injected into the footpad, 0.18±0.04% (n=7) of footpad activity was found in the draining LN within 4 h, whereas none was found in the contralateral LN. Quantitative assessment of cell migration by PET projection imaging of mice confirmed the ex-vivo counting results. These studies indicate that PET imaging offers a new approach for in vivo studies of dendritic cell biodistribution and migration.

Introduction

Active immunotherapy using tumor antigen loaded DC in mice have been extensively studied Celluzzi et al., 1996, Porgador et al., 1996, Zitvogel et al., 1996, Lotze et al., 1997, Lotze et al., 2000, Tuting et al., 1997, Fields et al., 1998a, Fields et al., 1998b, Nestle et al., 1998, Salgaller et al., 1998. Other studies demonstrated protection against viruses (De Bruijn et al., 1998) and bacteria (Mbow et al., 1997). Furthermore, human trials utilizing DC loaded with antigen are underway in several institutions. The induction of immunity depends on the interaction between DC and T cells. However, the best route of DC administration for ensuring migration to the T cell areas of lymph nodes, thereby ensuring optimal interactions, is uncertain, in part, because the sites where human DCs localize after injection are not fully known. Although bone marrow-derived DCs (BMDC) or their precursors circulate in the peripheral blood and subsequently reside in peripheral tissues, acquire antigen and migrate to regional lymph nodes in physiological circumstances [Steinman, 1991 #63; Banchereau, 1998 #62; Flores-Romo, 2001 #1], the migration pattern is still under intensive investigation. Migration of dendritic cells after intravenous, intradermal or subcutaneous injection has been assessed by using different tracing methods, such as membrane or cytosolic fluorescent dyes Austyn et al., 1988, Barratt-Boyes et al., 1997, Ingulli et al., 1997, Lappin et al., 1999 or cells labeled with radionuclides such as Cr-51 Cruz et al., 1990, Saeki et al., 1999 or Indium-111 Kupiec-Weglinski et al., 1988, Larsen et al., 1990, Morse et al., 1999, Thomas et al., 1999. In most of these studies the organs are removed at various times after injection of the label.

The aim of our study was to develop a method to label DC with the positron emitting radionuclide F-18, in order to detect DC migration in vivo by a high-resolution small animal PET camera. Here we describe our labeling method, the effects of labeling on the viability, phenotype and function of BMDC in vitro, and how F-18 labeled cells can be visualized and followed by a high resolution animal scanner.

Section snippets

Mice

Female BALB/c (I-Ad), C57BL/6 (I-Ab) and F1 (BALB/cxC57BL/6) mice (6–10 weeks old) were obtained from the National Institutes of Health animal facility. All animals were treated in accordance with institutional protocol.

Cells

BMDC were generated using the method of Fields et al. (1998a) with minor modifications. Briefly, bone marrow was flushed from tibias and femurs of female BALB/c or C57BL/6 mice and depleted of red blood cells with ammonium chloride. Cells were plated at 1×106 cell/ml in T162 T

Labeling efficiency and retention of radionuclide with BMDC

The F-18 labeling efficiency—percentage of original activity used for labeling remained with the cells after labeling and three washes—was 19±4% (n=18). Based on the specific activity of F-18, the cells were labeled with an average of 107 label/cell. Fig. 1 shows the distribution of F-18 in cells and supernatants as a function of time after labeling. At 37 °C considerable release occurred during the first hour and then remained essentially unchanged. The release of F-18 by BMDC may result from

Discussion

This is the first description of a method for labeling mouse BMDC with a positron emitting radionuclide F-18 in order to follow their migration with a high-resolution imager in vivo. F-18 is a radionuclide with a short (109.7 min) half life, and a high specific activity (>200 Ci/mmol at EOB). The short half life allows detection of the radionuclide for a maximum of 5–6 h, which in the case of BMDC migration is rather short, since the maximum number of injected BMDC can be found in the draining

Acknowledgements

The authors would like to thank to Elaine Jagoda, John L. Holt for helpful discussions and Jay Linton for providing expert technical help.

References (34)

  • J.M. Austyn et al.

    Migration patterns of dendritic cells in the mouse. Homing to T cell-dependent areas of spleen, and binding within marginal zone

    J. Exp. Med.

    (1988)
  • E.P. Balaban et al.

    Toxicity of indium-111 on the radiolabeled lymphocyte

    J. Nucl. Med.

    (1987)
  • S.M. Barratt-Boyes et al.

    In vivo migration of dendritic cells differentiated in vitro: a chimpanzee model

    J. Immunol.

    (1997)
  • Y. Belkaid et al.

    Analysis of cytokine production by inflammatory mouse macrophages at the single-cell level: selective impairment of IL-12 induction in Leishmania-infected cells

    Eur. J. Immunol.

    (1998)
  • C. Botti et al.

    Comparison of three different methods for radiolabelling human activated T lymphocytes

    Eur. J. Nucl. Med.

    (1997)
  • C.M. Celluzzi et al.

    Peptide-pulsed dendritic cells induce antigen-specific CTL-mediated protective tumor immunity [see comments]

    J. Exp. Med.

    (1996)
  • P.D. Cruz et al.

    Langerhans cells that migrate to skin after intravenous infusion regulate the induction of contact hypersensitivity

    J. Immunol.

    (1990)
  • C. Curiel-Lewandrowski et al.

    Transfection of immature murine bone marrow-derived dendritic cells with the granulocyte-macrophage colony-stimulating factor gene potently enhances their in vivo antigen-presenting capacity

    J. Immunol.

    (1999)
  • M.L. De Bruijn et al.

    Immunization with human papillomavirus type 16 (HPV16) oncoprotein-loaded dendritic cells as well as protein in adjuvant induces MHC class I-restricted protection to HPV16-induced tumor cells

    Cancer Res.

    (1998)
  • M.C. Dieu et al.

    Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites

    J. Exp. Med.

    (1998)
  • R.C. Fields et al.

    Comparative analysis of murine dendritic cells derived from spleen and bone marrow

    J. Immunother.

    (1998)
  • R.C. Fields et al.

    Murine dendritic cells pulsed with whole tumor lysates mediate potent antitumor immune responses in vitro and in vivo

    Proc. Natl. Acad. Sci. U.S.A.

    (1998)
  • E. Ingulli et al.

    In vivo detection of dendritic cell antigen presentation to CD4(+) T cells

    J. Exp. Med.

    (1997)
  • A.I. Kassis et al.

    Chemotoxicity of indium-111 oxine in mammalian cells

    J. Nucl. Med.

    (1985)
  • J.W. Kupiec-Weglinski et al.

    Migration patterns of dendritic cells in the mouse. Traffic from the blood, and T cell-dependent and -independent entry to lymphoid tissues

    J. Exp. Med.

    (1988)
  • J. Kuyama et al.

    Indium-111 labelled lymphocytes: isotope distribution and cell division

    Eur. J. Nucl. Med.

    (1997)
  • M.B. Lappin et al.

    Analysis of mouse dendritic cell migration in vivo upon subcutaneous and intravenous injection

    Immunology

    (1999)
  • Cited by (64)

    • Molecular imaging of innate immunity and immunotherapy

      2023, Advanced Drug Delivery Reviews
    • Engineering antigen as photosensitiser nanocarrier to facilitate ROS triggered immune cascade for photodynamic immunotherapy

      2020, Biomaterials
      Citation Excerpt :

      Meanwhile, comparing with controls, the obviously elevated red fluorescence intensity of MON can also be directly visualized in confocal laser scanning microscopy (CLSM) image (Fig. 3e), which further confirmed the presence of the homophilic targeting effect. BMDCs were isolated from femurs and tibias of 3–4 weeks old C57BL/6 mice according to the previous report with minor modification [45]. DC activation and antigen cross-presentation are key steps in the generation of potent immune responses and the schematic illustration of operation steps were showed in Fig. S5.

    • Nanoparticles for dendritic cell-based immunotherapy

      2018, International Journal of Pharmaceutics
    • Prolongation of the rat composite tissue allograft survival by the combination of tolerogenic immature dendritic cells and short-term treatment with FK506

      2013, Transplantation Proceedings
      Citation Excerpt :

      In contrast, subcutaneously or intradermally injected DCs preferentially home to the T-cell areas of draining lymph nodes. These results demonstrate that DC distribution to lymphoid tissue sites is dramatically affected by the mode of administration.25,26 In conclusion, we have demonstrated that alloantigen-pulsed recipient BLDCs in conjunction with transient immunosuppression have potential therapeutic application to induce TH2 immune responses and T-regulatory cell infiltration into composite tissue grafts.

    View all citing articles on Scopus
    View full text