Saponin-based adjuvants induce cross-presentation in dendritic cells by intracellular lipid body formation

Martijn H den Brok, Christian Büll, Melissa Wassink, Annemarie M de Graaf, Jori A Wagenaars, Marthe Minderman, Mayank Thakur, Sebastian Amigorena, Eric O Rijke, Carla C Schrier, Gosse J Adema, Martijn H den Brok, Christian Büll, Melissa Wassink, Annemarie M de Graaf, Jori A Wagenaars, Marthe Minderman, Mayank Thakur, Sebastian Amigorena, Eric O Rijke, Carla C Schrier, Gosse J Adema

Abstract

Saponin-based adjuvants (SBAs) are being used in animal and human (cancer) vaccines, as they induce protective cellular immunity. Their adjuvant potency is a factor of inflammasome activation and enhanced antigen cross-presentation by dendritic cells (DCs), but how antigen cross-presentation is induced is not clear. Here we show that SBAs uniquely induce intracellular lipid bodies (LBs) in the CD11b+ DC subset in vitro and in vivo. Using genetic and pharmacological interference in models for vaccination and in situ tumour ablation, we demonstrate that LB induction is causally related to the saponin-dependent increase in cross-presentation and T-cell activation. These findings link adjuvant activity to LB formation, aid the application of SBAs as a cancer vaccine component, and will stimulate development of new adjuvants enhancing T-cell-mediated immunity.

Figures

Figure 1. Saponin-based adjuvants induce cross-presentation in…
Figure 1. Saponin-based adjuvants induce cross-presentation in dendritic cells.
(a,b) Tetramer staining of OVA specific T cells in draining lymph nodes, 10 days following s.c. injection of 20 μg OVA and/or 30 μg ISCOMs (a, vaccination), or 7 days following co-treatment of established B16OVA melanomas (5–8 mm diameter) with in situ tumour ablation and 30 μg peritumourally injected ISCOMs (b, Cryo-ablation). (c,d) In vitro B3Z cross-presentation assays demonstrating a concentration-dependent increase in cross-presentation of OVA, only when Matrix C ISCOMs was added. GM-CSF-cultured BMDCs were exposed to the indicated compounds and 80 μg ml−1 OVA protein for 5 h, washed and co-cultured o/n with B3Z cells, which produce β-galactosidase upon recognition of the OVA peptide in Kb. Data in (d) are excerpts from larger titrations. For the non-reactive adjuvants the maximum concentration used is shown (1.5 μl ml−1), for the ISCOMs the concentration shown is 400 ng ml−1. (e) In vitro cross-presentation after 5 h exposure to 400 ng ml−1 of various SBAs or controls. This contains a crude saponin mixture not purified for immunoactive fractions, commercially available immunoactive saponin fractions (VX, SS and FC) and ISCOM structures made with these saponins according to an ‘open access' protocol. Passive peptide loading with OVA Kb peptides was used to demonstrate viability and/or equal MHC-I levels. Similar results were obtained in two to three independent experiments. Statistical analysis was done using a one or two-way ANOVA with post hoc Bonferroni or Tukey tests.
Figure 2. SBA-induced cross-presentation is independent of…
Figure 2. SBA-induced cross-presentation is independent of co-stimulation or ROS.
(a) FACS analysis of surface markers CD206 (mannose receptor), MHC-I, and MHC-II. GM-CSF BMDCs were exposed for 5 h to ISCOMs or medium, and processed for FACS staining. Open black lines represent the corresponding isotype controls. Filled grey lines show the medium-treated cells, while the open red lines are the ISCOM treated samples. (b) FACS analysis of DC maturation markers CD80 and CD86 after 18 h stimulation with ISCOMs or 1 μg ml−1 CpG. (c) Bone-marrow of indicated knockout mice was used to generate GM-CSF BMDCs. Cells were exposed for 5 h to medium or ISCOMs, washed and co-cultured o/n with B3Z cells. External peptide pulsing (5 pg ml−1, 30 min) was used to control for viability and/or MHC-I levels. (d) Antigen uptake and processing as analysed by FACS. BMDCs were given 0.25 mg ml−1 OVA coupled to the fluorophore Alexa647, or 1 μg ml−1 DQ-OVA, during the 5 h exposure time to medium or ISCOMs. DQ-OVA will start to fluoresce once degraded. (e) Total cellular reactive oxygen species (ROS) were measured using the DHR123 probe. GM-CSF cultured BMDCs were plated in serum-free medium and pretreated for 10 min with 0.5 or 50 μM dihydrorhodamine 123. Next, ISCOMs or 1 μg ml−1 PMA was added and cells were incubated for 45 min before analysis by FACS. (f) In vitro cross-presentation in the presence of NADPH oxidase inhibitors. Indicated compounds and concentrations were added during the 5 h exposure period to ISCOMs. (g) In vitro cross-presentation using Gp91phox−/− bonemarrow DCs. Data in (e) represent single values in titration, whereas the other data show means from triplicates with s.e.m. Data are representative figures from two to three individual experiments. Statistical analyses were performed using two way ANOVA with post hoc Bonferroni tests.
Figure 3. SBAs induce in vitro and…
Figure 3. SBAs induce in vitro and in vivo lipid bodies in correlation with cross-presentation.
(ac) GM-CSF-cultured BMDCs following 5 h exposure to the indicated adjuvants were fixed and stained with the LB probe Bodipy493/503. Nuclei were counterstained with DAPI, and cells were analysed by confocal microscopy. (a) CLSM image showing SBA-induced LBs. (b) Quantification of lipid bodies following exposure to OVA alone, AlPO4, non-immunoactive saponin, FC saponin, or ISCOMs. Numbers of lipid bodies per cell were obtained from 90–150 cells found in 15 randomly taken CLSM images, and divided over three strata. (c) LB staining with counterstaining for ADRP (df) 32 h time-course showing concomitant increases in LB numbers (d,e) and cross-presentation (f). (g,h) In vitro cross-presentation assay on isolated lymph node CD11c+ DCs using OT-I cells (left) or B3Z cells (right) as a readout. (g) Three hundred microgram endotoxin-free OVA protein was injected s.c. on the femur of WT C57Bl/6 mice, with or without 30 μg ISCOMs. Twelve hours later, draining lymph nodes were harvested from which CD11c+ cells were isolated using MACS beads. Next, these cells were co-cultured with the reporter cells. CD69 levels on CD8+CD90.1+ OT-I cells were measured after 1 day, while B3Z cells were analysed after 2 days. (h) Established B16OVA melanomas (5–8 mm diameter) were injected i.t. with a booster shot of OVA (20 μg), after which they were treated with in situ tumour ablation and 30 μg peritumourally injected ISCOMs. 12 h later draining lymph nodes were harvested from which CD11c+ cells were isolated using MACS beads. Next, these cells were co-cultured with the reporter cells like in g. (i) Quantification of LBs per cell using CLSM (left) or FACS (right), on ex vivo CD11c+ cells as treated and isolated under (g). All data were reproduced in two to three independent experiments. Statistical analyses were performed using two-way ANOVA with post hoc Bonferroni tests, or Student's t-test for i.
Figure 4. Responsiveness to SBAs in vitro…
Figure 4. Responsiveness to SBAs in vitro is unique to CD11b+ DCs with monocytic origin.
(a) In vitro cross-presentation assay using GM-CSF- or Flt3-L-propagated DCs. (b) Confocal images with LB quantifications from Flt3-L-generated BMDCs exposed for 5 h to medium or ISCOMs. (c) Gating strategy of Flt3-L DCs. Sorted populations are gate 1 (pDCs), gate 3 (CD8α+ DCs) and gate 4 (CD11b+ DCs). (d) In vitro cross-presentation assay using the populations as sorted under c. (e) Gating strategy of GM-CSF DCs. Sorted populations are gate 2 (MHCIIlo CD11bhi), gate 3 (MHCIIint CD11bint) and gate 4 (MHCIIhi CD11bint). (f) In vitro cross-presentation assay using the populations as sorted under e. (g) CLSM image and LB quantification of populations as sorted under e, after 5 h exposure to ISCOMs or medium. (h) In vitro cross-presentation assay on Flt3-L- or GM-CSF-cultured bonemarrow DCs. In the 10 day Flt3-L cultures, medium was supplemented for the last 48 h with 20 ng ml−1 GM-CSF or medium. (i) In vitro cross-presentation assays on Flt3-L-cultured DCs, receiving GM-CSF for 72 h after the end of culture. In the left panel the entire day 10 cultures were supplemented with GM-CSF for another 72 h, after which the resulting populations were sorted (Supplementary Fig. 8). In the right panel the day 10 cultures were sorted, after which the individual populations were treated with fresh Flt3-L or GM-CSF. All data were reproduced in two to three independent experiments. Statistical analyses were performed using two-way ANOVA with post hoc Bonferroni tests.
Figure 5. Lymph node CD11b+ DCs uniquely…
Figure 5. Lymph node CD11b+ DCs uniquely possess the ability to respond to SBAs in vivo.
(a) Gating strategy for naive splenocytes. Sorted populations are gate 2 (CD8α+ DCs) and gate 3 (CD11b+ DCs). (b) In vitro cross-presentation assay using the populations as sorted under a. (c) Gating strategy for naïve lymph nodes. Lin- was defined as B220- and Ly6G-. Sorted populations are gate 4 (LN resident CD8α+ DCs), gate 5 (LN resident CD11b+ DCs), gate 6 (LN migratory CD103 DCs), and gate 7 (LN migratory CD11b+ DCs). (d) In vitro cross-presentation assay using the populations as sorted under c. (eg) Mice were injected with Flt3-L excreting B16 melanomas to expand endogenous DC pools. After 12 days, pooled lymph nodes were harvested and subjected to FACS sorting. (e) Gating strategy, in which sorted populations are gates 1 to 4. Right panels show additional stainings on populations 1 to 3. (f) In vitro cross-presentation assay using populations as sorted under e. Sorted cells were exposed for 5 h to the indicated compounds (ISCOMs 800 ng ml−1, OVA 300 μg ml−1, PLGA nano-particles corresponding to 30 μg ml−1 OVA). (g) LB quantification in populations as sorted under e. All data were reproduced in two independent experiments. Statistical analyses were performed using two-way ANOVA with post hoc Bonferroni tests.
Figure 6. SBA-induced cross-presentation is dependent on…
Figure 6. SBA-induced cross-presentation is dependent on LB formation.
(a) LB quantification as measured by CLSM in cells treated for 5 h with the indicated LB inhibitors: (Xanthohumol: 85 μM, A922500: 40 μM, Triacsin C: 10 μM, TOFA: 120 μM). (b) In vitro cross-presentation assays in the presence of the indicated LB inhibitors (inhibited enzymes in brackets). (c,d) Flowcytometry-based analysis of LB content. Wild-type (c), or Igtp+/+ and Igtp−/− (d) BMDCs were exposed to 800 ng ml−1 non-immunoactive saponin, FC saponin, ISCOMs or 250 ng ml−1 IFNγ for 5 h, washed, and stained with Bodipy493-503. (e) In vitro cross-presentation assay using IGTP knockout DCs. (f) Metabolic activity/viability (MTT) assay used to demonstrate cytosolic translocation of indicated toxins. (g) MTT assay showing endosomal acidification is needed for cytosolic translocation. Bafilomycin A1, (at 100 nM) is an inhibitor of endosomal v-ATPase, and thus endosomal acidification. (h) Quantification of LB induction in the presence of ISCOMs and Bafilomycin A1. (i) MTT assay showing that inhibition of LB induction does not abrogate cytosolic translocation. During the exposure to ISCOMs/Cyt C, the LB inhibitors TOFA (120 μM) and A922500 (60 μM) were added. (j) In vitro OT-I cross-presentation assay on isolated LN DCs from OVA protein or peptide-vaccinated wild-type, Igtp−/−, or Adrp−/− mice. (k) B16-OVA tumour bearing mice were treated with cryo-ablation, directly followed by peritumoural injection of 30 μg ISCOMs. A mixture of xanthohumol (500 μg) and A922500 (150 μg) was injected peritumourally, four hrs before ablation and directly after the ablation. 12 h later, draining LN CD11c+ cells were isolated that entered the cross-presentations assays. LacZ content in B3Z cells was analysed after 2 days. All data represent means with s.e.m., except for d where data represent mean MFI values normalized to medium values. Similar data were obtained in two to three independent experiments. Statistical analyses were done using ANOVA with post hoc Bonferroni tests.

References

    1. Kensil C. R., Patel U., Lennick M. & Marciani D. Separation and characterization of saponins with adjuvant activity from Quillaja saponaria Molina cortex. J. Immunol. 146, 431–437 (1991).
    1. Lorent J. H., Quetin-Leclercq J. & Mingeot-Leclercq M. P. The amphiphilic nature of saponins and their effects on artificial and biological membranes and potential consequences for red blood and cancer cells. Org. Biomol. Chem. 12, 8803–8822 (2014).
    1. Morein B., Sundquist B., Hoglund S., Dalsgaard K. & Osterhaus A. Iscom, a novel structure for antigenic presentation of membrane proteins from enveloped viruses. Nature 308, 457–460 (1984).
    1. Lovgren Bengtsson K., Morein B. & Osterhaus A. D. ISCOM technology-based Matrix M adjuvant: success in future vaccines relies on formulation. Expert Rev. Vaccines 10, 401–403 (2011).
    1. Maraskovsky E. et al.. Development of prophylactic and therapeutic vaccines using the ISCOMATRIX adjuvant. Immunol. Cell Biol. 87, 371–376 (2009).
    1. Pearse M. J. & Drane D. ISCOMATRIX adjuvant: a potent inducer of humoral and cellular immune responses. Vaccine 22, 2391–2395 (2004).
    1. Holzman D. C. New data cheer champions of the danger theory. J. Natl Cancer Inst. 102, 76–78 (2010).
    1. Drane D., Gittleson C., Boyle J. & Maraskovsky E. ISCOMATRIX adjuvant for prophylactic and therapeutic vaccines. Expert Rev. Vaccines 6, 761–772 (2007).
    1. McKenzie A., Watt M. & Gittleson C. ISCOMATRIX() vaccines: safety in human clinical studies. Hum. Vaccin. 6, 237–246 (2010).
    1. Maraskovsky E. et al.. NY-ESO-1 protein formulated in ISCOMATRIX adjuvant is a potent anticancer vaccine inducing both humoral and CD8+ t-cell-mediated immunity and protection against NY-ESO-1+ tumors. Clin. Cancer Res. 10, 2879–2890 (2004).
    1. den Brok M. H. et al.. Saponin-based adjuvants create a highly effective anti-tumor vaccine when combined with in situ tumor destruction. Vaccine 30, 737–744 (2012).
    1. Nicholaou T. et al.. Regulatory T-cell-mediated attenuation of T-cell responses to the NY-ESO-1 ISCOMATRIX vaccine in patients with advanced malignant melanoma. Clin. Cancer Res. 15, 2166–2173 (2009).
    1. Vantomme V. et al.. Immunologic analysis of a phase I/II study of vaccination with MAGE-3 protein combined with the AS02B adjuvant in patients with MAGE-3-positive tumors. J. Immunother. 27, 124–135 (2004).
    1. Klein O. et al.. Low-dose cyclophosphamide enhances antigen-specific CD4(+) T cell responses to NY-ESO-1/ISCOMATRIX vaccine in patients with advanced melanoma. Cancer Immunol. Immunother. 64, 507–518 (2015).
    1. Kim Y. J. et al.. Synthetic studies of complex immunostimulants from Quillaja saponaria: synthesis of the potent clinical immunoadjuvant QS-21Aapi. J. Am. Chem. Soc. 128, 11906–11915 (2006).
    1. Wilson N. S. et al.. Inflammasome-dependent and -independent IL-18 production mediates immunity to the ISCOMATRIX adjuvant. J. Immunol. 192, 3259–3268 (2014).
    1. Wilson N. S. et al.. ISCOMATRIX vaccines mediate CD8(+) T-cell cross-priming by a MyD88-dependent signaling pathway. Immunol. Cell Biol. 90, 540–552 (2011).
    1. Joffre O. P., Segura E., Savina A. & Amigorena S. Cross-presentation by dendritic cells. Nat. Rev. Immunol. 12, 557–569 (2012).
    1. Heath W. R. & Carbone F. R. Cross-presentation in viral immunity and self-tolerance. Nat. Rev. Immunol. 1, 126–134 (2001).
    1. Merad M., Sathe P., Helft J., Miller J. & Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 31, 563–604 (2013).
    1. Poulin L. F. et al.. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood 119, 6052–6062 (2012).
    1. Tamura T. et al.. IFN regulatory factor-4 and -8 govern dendritic cell subset development and their functional diversity. J. Immunol. 174, 2573–2581 (2005).
    1. den Haan J. M., Lehar S. M. & Bevan M. J. CD8(+) but not CD8(−) dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1696 (2000).
    1. Allan R. S. et al.. Epidermal viral immunity induced by CD8alpha+ dendritic cells but not by Langerhans cells. Science 301, 1925–1928 (2003).
    1. Nierkens S., Tel J., Janssen E. & Adema G. J. Antigen cross-presentation by dendritic cell subsets: one general or all sergeants? Trends Immunol. 34, 361–370 (2013).
    1. Cheong C. et al.. Microbial stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for immune T cell areas. Cell 143, 416–429 (2010).
    1. Farese R. V. Jr & Walther T. C. Lipid droplets finally get a little R-E-S-P-E-C-T. Cell 139, 855–860 (2009).
    1. Miura S. et al.. Functional conservation for lipid storage droplet association among Perilipin, ADRP, and TIP47 (PAT)-related proteins in mammals, Drosophila, and Dictyostelium. J. Biol. Chem. 277, 32253–32257 (2002).
    1. Brasaemle D. L., Dolios G., Shapiro L. & Wang R. Proteomic analysis of proteins associated with lipid droplets of basal and lipolytically stimulated 3T3-L1 adipocytes. J. Biol. Chem. 279, 46835–46842 (2004).
    1. Wan H. C., Melo R. C., Jin Z., Dvorak A. M. & Weller P. F. Roles and origins of leukocyte lipid bodies: proteomic and ultrastructural studies. FASEB J. 21, 167–178 (2007).
    1. Bougneres L. et al.. A role for lipid bodies in the cross-presentation of phagocytosed antigens by MHC class I in dendritic cells. Immunity 31, 232–244 (2009).
    1. 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).
    1. den Brok M. H. et al.. Efficient loading of dendritic cells following cryo and radiofrequency ablation in combination with immune modulation induces anti-tumour immunity. Br. J. Cancer 95, 896–905 (2006).
    1. Nierkens S. et al.. Immune adjuvant efficacy of CpG oligonucleotide in cancer treatment is founded specifically upon TLR9 function in plasmacytoid dendritic cells. Cancer Res. 71, 6428–6437 (2011).
    1. Schnurr M. et al.. ISCOMATRIX adjuvant induces efficient cross-presentation of tumor antigen by dendritic cells via rapid cytosolic antigen delivery and processing via tripeptidyl peptidase II. J. Immunol. 182, 1253–1259 (2009).
    1. Lewis K. L. et al.. Notch2 receptor signaling controls functional differentiation of dendritic cells in the spleen and intestine. Immunity 35, 780–791 (2011).
    1. Duewell P. et al.. ISCOMATRIX adjuvant combines immune activation with antigen delivery to dendritic cells in vivo leading to effective cross-priming of CD8+ T cells. J. Immunol. 187, 55–63 (2011).
    1. Schiott A. et al.. Posintro-HBsAg, a modified ISCOM including HBsAg, induces strong cellular and humoral responses. Int. J. Pharm. 414, 312–320 (2011).
    1. Cox R. J. et al.. Evaluation of a virosomal H5N1 vaccine formulated with Matrix M adjuvant in a phase I clinical trial. Vaccine 29, 8049–8059 (2011).
    1. Lövgren-Bengtsson K. & Morein B. in Methods in Molecular Medicine Vol. 42 (ed. O'Hagan, D.T.) 239–258 (2000).
    1. Kotsias F., Hoffmann E., Amigorena S. & Savina A. Reactive oxygen species production in the phagosome: impact on antigen presentation in dendritic cells. Antioxid. Redox Signal. 18, 714–729 (2013).
    1. Helft J. et al.. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c(+)MHCII(+) macrophages and dendritic cells. Immunity 42, 1197–1211 (2015).
    1. Naik S. H. et al.. Cutting edge: generation of splenic CD8+ and CD8− dendritic cell equivalents in Fms-like tyrosine kinase 3 ligand bone marrow cultures. J. Immunol. 174, 6592–6597 (2005).
    1. Mayer C. T. et al.. Selective and efficient generation of functional Batf3-dependent CD103+ dendritic cells from mouse bone marrow. Blood 124, 3081–3091 (2014).
    1. Mach N. et al.. Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or Flt3-ligand. Cancer Res. 60, 3239–3246 (2000).
    1. Mollah S. A. et al.. Flt3L dependence helps define an uncharacterized subset of murine cutaneous dendritic cells. J. Invest. Dermatol. 134, 1265–1275 (2014).
    1. Krahmer N., Guo Y., Farese R. V. Jr & Walther T. C. SnapShot: lipid droplets. Cell 139, 1024–1024 e1021 (2009).
    1. Herber D. L. et al.. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 16, 880–886 (2010).
    1. Paul A., Chang B. H., Li L., Yechoor V. K. & Chan L. Deficiency of adipose differentiation-related protein impairs foam cell formation and protects against atherosclerosis. Circ. Res. 102, 1492–1501 (2008).
    1. Weng A. et al.. Saponins modulate the intracellular trafficking of protein toxins. J. Control. Release 164, 74–86 (2012).
    1. Schnurr M. et al.. Tumor antigen processing and presentation depend critically on dendritic cell type and the mode of antigen delivery. Blood 105, 2465–2472 (2005).
    1. Shen L. & Rock K. L. Priming of T cells by exogenous antigen cross-presented on MHC class I molecules. Curr. Opin. Immunol. 18, 85–91 (2006).
    1. Everts B. et al.. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKvarepsilon supports the anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332 (2014).
    1. Ibrahim J. et al.. Dendritic cell populations with different concentrations of lipid regulate tolerance and immunity in mouse and human liver. Gastroenterology 143, 1061–1072 (2012).
    1. Cao W. et al.. Oxidized lipids block antigen cross-presentation by dendritic cells in cancer. J. Immunol. 192, 2920–2931 (2014).
    1. Eggermont A. M. et al.. Adjuvant ganglioside GM2-KLH/QS-21 vaccination versus observation after resection of primary tumor >1.5 mm in patients with stage II melanoma: results of the EORTC 18961 randomized phase III trial. J. Clin. Oncol. 31, 3831–3837 (2013).
    1. Silva A. et al.. The combination of ISCOMATRIX adjuvant and TLR agonists induces regression of established solid tumors in vivo. J. Immunol. 194, 2199–2207 (2015).
    1. McCluskie M. J. et al.. CpG ODN and ISCOMATRIX adjuvant: a synergistic adjuvant combination inducing strong T-Cell IFN-gamma responses. BioMed Res. Int. 2013, 636847 (2013).
    1. Chen J. L. et al.. NY-ESO-1 specific antibody and cellular responses in melanoma patients primed with NY-ESO-1 protein in ISCOMATRIX and boosted with recombinant NY-ESO-1 fowlpox virus. Int. J. Cancer 136, E590–E601 (2015).
    1. van den Bijgaart R. J. et al.. Thermal and mechanical high-intensity focused ultrasound: perspectives on tumor ablation, immune effects and combination strategies. Cancer Immunol. Immunother doi:10.1007/s00262-016-1891-9 (2016).
    1. Chang B. H. et al.. Protection against fatty liver but normal adipogenesis in mice lacking adipose differentiation-related protein. Mol. Cell Biol. 26, 1063–1076 (2006).
    1. Taylor G. A. et al.. Pathogen-specific loss of host resistance in mice lacking the IFN-gamma-inducible gene IGTP. Proc. Natl Acad. Sci. USA 97, 751–755 (2000).
    1. Lutz M. B. et al.. An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J. Immunol. Methods 223, 77–92 (1999).
    1. 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).
    1. Karttunen J., Sanderson S. & Shastri N. Detection of rare antigen-presenting cells by the lacZ T-cell activation assay suggests an expression cloning strategy for T-cell antigens. Proc. Natl Acad. Sci. USA 89, 6020–6024 (1992).
    1. Gilabert-Oriol R. et al.. Reporter assay for endo/lysosomal escape of toxin-based therapeutics. Toxins 6, 1644–1666 (2014).
    1. Nierkens S. et al.. In vivo colocalization of antigen and CpG [corrected] within dendritic cells is associated with the efficacy of cancer immunotherapy. Cancer Res. 68, 5390–5396 (2008).
    1. Cruz L. J. et al.. Targeted PLGA nano- but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. J. Control. Release 144, 118–126 (2010).
    1. Gocze P. M. & Freeman D. A. Factors underlying the variability of lipid droplet fluorescence in MA-10 Leydig tumor cells. Cytometry 17, 151–158 (1994).

Source: PubMed

3
Abonnere