MAIT cells promote inflammatory monocyte differentiation into dendritic cells during pulmonary intracellular infection

Anda I Meierovics, Siobhán C Cowley, Anda I Meierovics, Siobhán C Cowley

Abstract

Mucosa-associated invariant T (MAIT) cells are a unique innate T cell subset that is necessary for rapid recruitment of activated CD4+ T cells to the lungs after pulmonary F. tularensis LVS infection. Here, we investigated the mechanisms behind this effect. We provide evidence to show that MAIT cells promote early differentiation of CCR2-dependent monocytes into monocyte-derived DCs (Mo-DCs) in the lungs after F. tularensis LVS pulmonary infection. Adoptive transfer of Mo-DCs to MAIT cell-deficient mice (MR1-/- mice) rescued their defect in the recruitment of activated CD4+ T cells to the lungs. We further demonstrate that MAIT cell-dependent GM-CSF production stimulated monocyte differentiation in vitro, and that in vivo production of GM-CSF was delayed in the lungs of MR1-/- mice. Finally, GM-CSF-deficient mice exhibited a defect in monocyte differentiation into Mo-DCs that was phenotypically similar to MR1-/- mice. Overall, our data demonstrate that MAIT cells promote early pulmonary GM-CSF production, which drives the differentiation of inflammatory monocytes into Mo-DCs. Further, this delayed differentiation of Mo-DCs in MR1-/- mice was responsible for the delayed recruitment of activated CD4+ T cells to the lungs. These findings establish a novel mechanism by which MAIT cells function to promote both innate and adaptive immune responses.

Figures

Figure 1.
Figure 1.
Accumulation of CCR2-dependent CD11b+ DCs in the lungs of MR1−/− mice is impaired after F. tularensis i.n. LVS infection. Wild-type C57BL/6, MR1−/−, and CCR2−/− mice were i.n. infected with a sublethal dose of 2 × 102 CFU F. tularensis LVS, and total lung cells were harvested at the indicated time points after infection to evaluate the presence of myeloid cells. (A) Representative flow cytometry dot plots of lung cells on day 4 after infection, demonstrating the gating scheme used to identify CD103+ DCs (gate R1), macrophages (gate R2), and CD11b+ DCs (gate R3). (B) Accumulation of CD103+ DCs, macrophages, and CD11b+ DCs in the lungs of WT mice after i.n. LVS infection. (C) Total numbers of CD11b+ DCs in the lungs of WT, MR1−/−, and CCR2−/− mice on day 4 after i.n. LVS infection. All data are representative of three independent experiments (n = 5 mice per group) and are the mean ± SEM. *, P < 0.01, compared with LVS-infected WT mice at the same time point. Data were analyzed via one-way ANOVA, followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 2.
Figure 2.
Characterization of CD11b+ DCs in the lungs of mice after i.n. F. tularensis LVS infection. Wild-type C57BL/6 mice were infected with a sublethal dose of 2 × 102 CFU F. tularensis i.n. LVS, and total lung cells were harvested on day 4 after infection. (A) Expression of myeloid cell markers by CD11b+ DCs. In the flow cytometry histograms shown, CD11b+ DCs were first gated as indicated in Fig. 1 A, gate R3. Gray histogram shows fluorescence minus one control, and white histogram shows staining by the indicated antibody. Data are representative of two independent experiments (n = 3 mice per group). (B) Quantitation of myeloid marker expression by lung CD11b+ DCs (based on percentage of positive cells). Data are representative of two independent experiments (n = 3 mice per group). (C) Cytospin preparations of CD11b+ DCs purified from the lungs of 10 LVS-infected WT mice on day 4 after infection by fluorescence-activated cell sorting and examined by light microscopy. Bars, 10 µm. Data are representative of two independent experiments. (D) Lung Mo-DCs and CD11b+ DCs are the same population by flow cytometry analysis. Wild-type C57BL/6 mice were infected with a sublethal dose of 2 × 102 CFU F. tularensis i.n. LVS, and total lung cells were harvested on day 4 after infection to evaluate Mo-DCs and CD11b+ DCs. Representative dot plots of lung cells showing the gating scheme for Mo-DCs (gate R1) and CD11b+ DCs (gate R2). Overlay of the Mo-DC gating scheme is shown in red, and the CD11b+ DC gating scheme shown in blue. Data are representative of three independent experiments (n = 5 mice).
Figure 3.
Figure 3.
Inflammatory monocyte recruitment to the lungs of MR1−/− mice is not impaired after i.n. F. tularensis LVS infection. Wild-type C57BL/6, MR1−/−, and CCR2−/− mice were i.n. infected with a sublethal dose of 2 × 102 CFU F. tularensis LVS, and (A) total lung cells were harvested on day 4 after infection to evaluate the presence of Ly6Chi CD11b+ inflammatory monocytes. Representative flow cytometry dot plots are shown, as well as enumeration of the total number of Ly6Chi CD11b+ inflammatory monocytes in the lungs of mice. Data are representative of four pooled experiments (n = 3–5 mice per group) and are the mean ± SEM. *, P < 0.01, compared with LVS-infected WT mice at the same time point. (B) Total bone marrow cells were harvested on day 4 after infection to evaluate the presence of Ly6Chi CD11b+ monocytes. Representative flow cytometry dot plots are shown, as well as the percentage of Ly6Chi CD11b+ monocytes in the bone marrow of mice. Data are representative of three independent experiments (n = 5 mice per group) and are the mean ± SEM. *, P < 0.01, compared with LVS-infected WT mice at the same time point. Data were analyzed via one-way ANOVA, followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 4.
Figure 4.
Differentiation of inflammatory monocytes into Mo-DCs is impaired in the lungs of MR1−/− mice after i.n. F. tularensis LVS infection. Wild-type C57BL/6 and MR1−/− mice were infected with a sublethal dose of 2 × 102 CFU F. tularensis i.n. LVS, and (A) total lung cells were harvested at the indicated time points to evaluate the presence of Mo-DCs (Ly6Chi CD11b+ MHCII+ CD11c+ cells). Data are representative of three independent experiments (n = 5 mice per group), and are the mean ± SEM. *, P < 0.05, compared with LVS-infected WT mice at the same time point. (B) Flow cytometry analyses of Mo-DCs in the lungs of WT and MR1−/− mice on day 4 after infection. Representative dot plots are shown. (C) Analysis of CD11c, CD86, and MHCII expression by Ly6Chi CD11b+ cells in the lungs of mice on day 4 after i.n. LVS infection. Data are representative of three independent experiments (n = 5 mice per group), and are the mean ± SEM. *, P < 0.05, compared with LVS-infected WT mice at the same time point. (D) Cytospin preparations of CD11c− and CD11c+ inflammatory monocytes (Ly6Chi CD11b+ cells) purified from the lungs of LVS-infected mice on day 4 after infection by fluorescence-activated cell sorting and examined by light microscopy (top) or confocal microscopy (bottom) using wheat germ agglutinin-Alexa Fluor 594–conjugated membrane stain (red) and Hoechst nuclear stain (blue). Bars, 10 µm. Lung cells were pooled from 10 mice for flow sorting, and data are representative of three independent experiments. (E) WT mice were infected i.n. with LVS-GFP or LVS, and GFP+ cells were evaluated in CD11c− and CD11c+ inflammatory monocytes (Ly6Chi CD11b+ cells) by flow cytometry on day 4 after infection. Representative dot plots are shown. (F) Quantification of the percentage of GFP+ CD11c− and CD11c+ inflammatory monocytes (Ly6Chi CD11b+ cells) in the lungs of mice on day 4 after infection with either LVS-GFP or LVS (negative GFP control). Data are representative of three independent experiments (n = 5 mice per group) and are the mean ± SEM. *, P < 0.05, compared with LVS-infected WT mice at the same time point. Data were analyzed via one-way ANOVA followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 5.
Figure 5.
Adoptive transfer of Mo-DCs to MR1−/− mice rescues their impairment in the accumulation of activated CD4+ T cells in the lungs after i.n. F. tularensis LVS infection. Wild-type C57BL/6 mice were infected with a sublethal dose of 2 × 102 CFU F. tularensis i.n. LVS, and Mo-DCs were purified from the lungs on day 4 after infection by fluorescence-activated cell sorting using the gating scheme shown in Fig. 1 A, gate R3. 106 Mo-DCs were transferred i.v. to MR1−/− mice on day 3 after i.n. LVS infection. Lungs were harvested from WT mice, MR1−/− mice, and MR1−/− adoptive transfer recipients and the total numbers of CD4+ T cells and activated (CD69+ CD44+) CD4+ T cells were quantified by flow cytometry on day 8 after i.n. LVS infection. Data are representative of three independent experiments, and are the mean ± SEM. *, P < 0.05, compared with LVS-infected WT mice. ^, P < 0.05, compared with LVS-infected MR1−/− mice. Data were analyzed via one-way ANOVA, followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 6.
Figure 6.
Bone marrow monocytes from MR1−/− mice do not exhibit a defect in differentiation into MHCII+CD11c+ cells in response to recombinant cytokines in vitro. Monocytes (Ly6Chi CD11b+ cells) were purified from the bone marrow of naive WT and MR1−/− mice by fluorescence-activated cell sorting and cultured for 3 d in the presence of recombinant IFN-γ or GM-CSF (100 ng/ml), then assessed for coexpression of MHCII and CD11c by flow cytometry. Representative flow cytometry dot plots are shown (A). The percentage (B) and total number (C) of MHCII+ CD11c+ monocytes is shown. Bone marrow cells were pooled and sorted from 50 mice; data includes values ± SEM from three replicates, and is representative of three independent experiments. *, P < 0.01, compared with IFN-γ–treated cells. Data were analyzed via one-way ANOVA, followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 7.
Figure 7.
Vα19iTg T cells promote the differentiation of bone marrow monocytes into MHCII+CD11c+ cells in a GM-CSF–dependent manner in vitro. Monocytes (Ly6Chi CD11b+ cells) were purified from the bone marrow of naive WT and MR1−/− mice by fluorescence-activated cell sorting and cultured for 3 d in the presence of MAIT cells and LVS-infected macrophages, then assessed for coexpression of MHCII and CD11c by flow cytometry. Thy1.2+ cells purified from transgenic Vα19iTgCα−/−MR1+/+ mice were the source of MAIT cells. In some cases, as indicated, neutralizing antibodies or isotype control antibodies were added to the cultures at the time of addition of purified MAIT cells. In A, representative flow cytometry dot plots and the number of MHCII+ CD11c+ monocytes are shown for cultures containing WT bone marrow monocytes and LVS-infected WT macrophages in the presence or absence of Vα19iTg Thy1.2+ cells. In B, the number of MHCII+ CD11c+ monocytes are shown for cultures containing WT bone marrow monocytes, LVS-infected WT macrophages, and either Vα19iTg Thy1.2+ cells or naive WT C57BL/6 Thy1.2+ cells. In C, representative flow cytometry dot plots of MHCII+ CD11c+ monocytes are shown for cultures containing MR1−/− bone marrow monocytes, LVS-infected MR1−/− macrophages, and Vα19iTg Thy1.2+ cells. Graph of the numbers of MHCII+ CD11c+ monocytes obtained from MR1−/− cultures as compared with WT cultures (note that A and B are all from the same representative experiment). In D, IFN-γ and GM-CSF in the supernatants of cultures containing WT bone marrow monocytes, LVS-infected WT macrophages, and Vα19iTg Thy1.2+ cells after 3 d are shown. Bone marrow cells were pooled from 50 mice per experiment; data includes values ± SEM from two pooled experiments, each containing two replicate samples, and is representative of five independent experiments. *, P < 0.01, compared with No T cells. ^, P < 0.01, compared with +Vα19i Thy1.2 cells + control Ab. #, P < 0.01, compared with +Vα19i Thy1.2 cells. Data were analyzed via one-way ANOVA followed by the Student-Newman-Keuls multiple stepwise comparison.
Figure 8.
Figure 8.
MAIT cells are required for early GM-CSF production in the lungs after i.n. F. tularensis LVS infection. Wild-type C57BL/6 and MR1−/− mice were infected with a sublethal dose of 2 × 102 CFU F. tularensis i.n. LVS, and (A) the levels of GM-CSF and M-CSF were determined in lung homogenates during the first 5 d of infection (n = 5 mice per group). (B) Expression of CD115 and CSF2Rα by inflammatory monocytes (Ly6Chi CD11b+ cells) in the bone marrow and lungs on day 4 after i.n. LVS infection. Red line, fluorescence minus one control; blue line, staining by the indicated antibody. (C) Enumeration of the total number of Ly6Chi CD11b+ inflammatory monocytes in the lungs of WT, MR1−/−, and GM-CSF–depleted WT mice on day 4 after i.n. LVS infection. (D) Representative flow cytometry dot plots of Mo-DC accumulation in the lungs of WT, MR1−/−, and GM-CSF–depleted WT mice on day 4 after i.n. LVS infection. (E) Enumeration of the total number of Mo-DCs in the lungs of WT, MR1−/−, and GM-CSF–depleted WT mice on day 4 after i.n. LVS infection. Data are representative of four independent experiments (n = 5 mice per group), and are the mean ± SEM *, P < 0.01, compared with WT mice. Data were analyzed via one-way ANOVA followed by the Student-Newman-Keuls multiple stepwise comparison.

References

    1. Bar-Haim E., Gat O., Markel G., Cohen H., Shafferman A., and Velan B.. 2008. Interrelationship between dendritic cell trafficking and Francisella tularensis dissemination following airway infection. PLoS Pathog. 4:e1000211 10.1371/journal.ppat.1000211
    1. Barouch D.H., Santra S., Tenner-Racz K., Racz P., Kuroda M.J., Schmitz J.E., Jackson S.S., Lifton M.A., Freed D.C., Perry H.C., et al. . 2002. Potent CD4+ T cell responses elicited by a bicistronic HIV-1 DNA vaccine expressing gp120 and GM-CSF. J. Immunol. 168:562–568. 10.4049/jimmunol.168.2.562
    1. Bosschaerts T., Guilliams M., Stijlemans B., Morias Y., Engel D., Tacke F., Hérin M., De Baetselier P., and Beschin A.. 2010. Tip-DC development during parasitic infection is regulated by IL-10 and requires CCL2/CCR2, IFN-gamma and MyD88 signaling. PLoS Pathog. 6:e1001045 10.1371/journal.ppat.1001045
    1. Campbell I.K., van Nieuwenhuijze A., Segura E., O’Donnell K., Coghill E., Hommel M., Gerondakis S., Villadangos J.A., and Wicks I.P.. 2011. Differentiation of inflammatory dendritic cells is mediated by NF-κB1-dependent GM-CSF production in CD4 T cells. J. Immunol. 186:5468–5477. 10.4049/jimmunol.1002923
    1. Chen G.H., Teitz-Tennenbaum S., Neal L.M., Murdock B.J., Malachowski A.N., Dils A.J., Olszewski M.A., and Osterholzer J.J.. 2016. Local GM-CSF-dependent differentiation and activation of pulmonary dendritic cells and macrophages protect against progressive cryptococcal lung infection in mice. J. Immunol. 196:1810–1821. 10.4049/jimmunol.1501512
    1. Chong S.Z., Wong K.L., Lin G., Yang C.M., Wong S.C., Angeli V., Macary P.A., and Kemeny D.M.. 2011. Human CD8+ T cells drive Th1 responses through the differentiation of TNF/iNOS-producing dendritic cells. Eur. J. Immunol. 41:1639–1651. 10.1002/eji.201041022
    1. Chua W.J., Truscott S.M., Eickhoff C.S., Blazevic A., Hoft D.F., and Hansen T.H.. 2012. Polyclonal mucosa-associated invariant T cells have unique innate functions in bacterial infection. Infect. Immun. 80:3256–3267. 10.1128/IAI.00279-12
    1. Codarri L., Gyülvészi G., Tosevski V., Hesske L., Fontana A., Magnenat L., Suter T., and Becher B.. 2011. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 12:560–567. 10.1038/ni.2027
    1. Cowley S.C. 2014. MAIT cells and pathogen defense. Cell. Mol. Life Sci. 71:4831–4840. 10.1007/s00018-014-1708-y
    1. Cowley S.C., and Elkins K.L.. 2003. Multiple T cell subsets control Francisella tularensis LVS intracellular growth without stimulation through macrophage interferon gamma receptors. J. Exp. Med. 198:379–389. 10.1084/jem.20030687
    1. Croxford A.L., Lanzinger M., Hartmann F.J., Schreiner B., Mair F., Pelczar P., Clausen B.E., Jung S., Greter M., and Becher B.. 2015. The Cytokine GM-CSF drives the inflammatory signature of CCR2+ monocytes and licenses autoimmunity. Immunity. 43:502–514. 10.1016/j.immuni.2015.08.010
    1. Cui Y., Franciszkiewicz K., Mburu Y.K., Mondot S., Le Bourhis L., Premel V., Martin E., Kachaner A., Duban L., Ingersoll M.A., et al. . 2015. Mucosal-associated invariant T cell-rich congenic mouse strain allows functional evaluation. J. Clin. Invest. 125:4171–4185. 10.1172/JCI82424
    1. Dennis D.T., Inglesby T.V., Henderson D.A., Bartlett J.G., Ascher M.S., Eitzen E., Fine A.D., Friedlander A.M., Hauer J., Layton M., et al. Working Group on Civilian Biodefense . 2001. Tularemia as a biological weapon: medical and public health management. JAMA. 285:2763–2773. 10.1001/jama.285.21.2763
    1. Dusseaux M., Martin E., Serriari N., Péguillet I., Premel V., Louis D., Milder M., Le Bourhis L., Soudais C., Treiner E., and Lantz O.. 2011. Human MAIT cells are xenobiotic-resistant, tissue-targeted, CD161hi IL-17-secreting T cells. Blood. 117:1250–1259. 10.1182/blood-2010-08-303339
    1. Elkins K.L., Cowley S.C., and Bosio C.M.. 2003. Innate and adaptive immune responses to an intracellular bacterium, Francisella tularensis live vaccine strain. Microbes Infect. 5:135–142. 10.1016/S1286-4579(02)00084-9
    1. Ellis J., Oyston P.C., Green M., and Titball R.W.. 2002. Tularemia. Clin. Microbiol. Rev. 15:631–646. 10.1128/CMR.15.4.631-646.2002
    1. Espinosa V., Jhingran A., Dutta O., Kasahara S., Donnelly R., Du P., Rosenfeld J., Leiner I., Chen C.C., Ron Y., et al. . 2014. Inflammatory monocytes orchestrate innate antifungal immunity in the lung. PLoS Pathog. 10:e1003940 10.1371/journal.ppat.1003940
    1. Förster R., Schubel A., Breitfeld D., Kremmer E., Renner-Müller I., Wolf E., and Lipp M.. 1999. CCR7 coordinates the primary immune response by establishing functional microenvironments in secondary lymphoid organs. Cell. 99:23–33. 10.1016/S0092-8674(00)80059-8
    1. Fortier A.H., Slayter M.V., Ziemba R., Meltzer M.S., and Nacy C.A.. 1991. Live vaccine strain of Francisella tularensis: infection and immunity in mice. Infect. Immun. 59:2922–2928.
    1. Geissler M., Gesien A., Tokushige K., and Wands J.R.. 1997. Enhancement of cellular and humoral immune responses to hepatitis C virus core protein using DNA-based vaccines augmented with cytokine-expressing plasmids. J. Immunol. 158:1231–1237.
    1. Georgel P., Radosavljevic M., Macquin C., and Bahram S.. 2011. The non-conventional MHC class I MR1 molecule controls infection by Klebsiella pneumoniae in mice. Mol. Immunol. 48:769–775. 10.1016/j.molimm.2010.12.002
    1. Gold M.C., and Lewinsohn D.M.. 2013. Co-dependents: MR1-restricted MAIT cells and their antimicrobial function. Nat. Rev. Microbiol. 11:14–19. 10.1038/nrmicro2918
    1. Gold M.C., Cerri S., Smyk-Pearson S., Cansler M.E., Vogt T.M., Delepine J., Winata E., Swarbrick G.M., Chua W.J., Yu Y.Y., et al. . 2010. Human mucosal associated invariant T cells detect bacterially infected cells. PLoS Biol. 8:e1000407 10.1371/journal.pbio.1000407
    1. Greter M., Helft J., Chow A., Hashimoto D., Mortha A., Agudo-Cantero J., Bogunovic M., Gautier E.L., Miller J., Leboeuf M., et al. . 2012. GM-CSF controls nonlymphoid tissue dendritic cell homeostasis but is dispensable for the differentiation of inflammatory dendritic cells. Immunity. 36:1031–1046. 10.1016/j.immuni.2012.03.027
    1. Guilliams M., Movahedi K., Bosschaerts T., VandenDriessche T., Chuah M.K., Hérin M., Acosta-Sanchez A., Ma L., Moser M., Van Ginderachter J.A., et al. . 2009. IL-10 dampens TNF/inducible nitric oxide synthase-producing dendritic cell-mediated pathogenicity during parasitic infection. J. Immunol. 182:1107–1118. 10.4049/jimmunol.182.2.1107
    1. Guilliams M., Lambrecht B.N., and Hammad H.. 2013. Division of labor between lung dendritic cells and macrophages in the defense against pulmonary infections. Mucosal Immunol. 6:464–473. 10.1038/mi.2013.14
    1. Hegde S., Chen X., Keaton J.M., Reddington F., Besra G.S., and Gumperz J.E.. 2007. NKT cells direct monocytes into a DC differentiation pathway. J. Leukoc. Biol. 81:1224–1235. 10.1189/jlb.1206718
    1. Helft J., Böttcher J., Chakravarty P., Zelenay S., Huotari J., Schraml B.U., Goubau D., and Reis e Sousa C.. 2015. GM-CSF Mouse Bone Marrow Cultures Comprise a Heterogeneous Population of CD11c(+)MHCII(+) Macrophages and Dendritic Cells. Immunity. 42:1197–1211. 10.1016/j.immuni.2015.05.018
    1. Hohl T.M., Rivera A., Lipuma L., Gallegos A., Shi C., Mack M., and Pamer E.G.. 2009. Inflammatory monocytes facilitate adaptive CD4 T cell responses during respiratory fungal infection. Cell Host Microbe. 6:470–481. 10.1016/j.chom.2009.10.007
    1. Huang S., Gilfillan S., Cella M., Miley M.J., Lantz O., Lybarger L., Fremont D.H., and Hansen T.H.. 2005. Evidence for MR1 antigen presentation to mucosal-associated invariant T cells. J. Biol. Chem. 280:21183–21193. 10.1074/jbc.M501087200
    1. Huang S., Martin E., Kim S., Yu L., Soudais C., Fremont D.H., Lantz O., and Hansen T.H.. 2009. MR1 antigen presentation to mucosal-associated invariant T cells was highly conserved in evolution. Proc. Natl. Acad. Sci. USA. 106:8290–8295. 10.1073/pnas.0903196106
    1. Kang S.J., Liang H.E., Reizis B., and Locksley R.M.. 2008. Regulation of hierarchical clustering and activation of innate immune cells by dendritic cells. Immunity. 29:819–833. 10.1016/j.immuni.2008.09.017
    1. Kawachi I., Maldonado J., Strader C., and Gilfillan S.. 2006. MR1-restricted V alpha 19i mucosal-associated invariant T cells are innate T cells in the gut lamina propria that provide a rapid and diverse cytokine response. J. Immunol. 176:1618–1627. 10.4049/jimmunol.176.3.1618
    1. Kjer-Nielsen L., Patel O., Corbett A.J., Le Nours J., Meehan B., Liu L., Bhati M., Chen Z., Kostenko L., Reantragoon R., et al. . 2012. MR1 presents microbial vitamin B metabolites to MAIT cells. Nature. 491:717–723.
    1. Ko H.J., Brady J.L., Ryg-Cornejo V., Hansen D.S., Vremec D., Shortman K., Zhan Y., and Lew A.M.. 2014. GM-CSF-responsive monocyte-derived dendritic cells are pivotal in Th17 pathogenesis. J. Immunol. 192:2202–2209. 10.4049/jimmunol.1302040
    1. Kurioka A., Ussher J.E., Cosgrove C., Clough C., Fergusson J.R., Smith K., Kang Y.H., Walker L.J., Hansen T.H., Willberg C.B., and Klenerman P.. 2015. MAIT cells are licensed through granzyme exchange to kill bacterially sensitized targets. Mucosal Immunol. 8:429–440. 10.1038/mi.2014.81
    1. Le Bourhis L., Dusseaux M., Bohineust A., Bessoles S., Martin E., Premel V., Coré M., Sleurs D., Serriari N.E., Treiner E., et al. . 2013. MAIT cells detect and efficiently lyse bacterially-infected epithelial cells. PLoS Pathog. 9:e1003681 10.1371/journal.ppat.1003681
    1. León B., López-Bravo M., and Ardavín C.. 2007. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity. 26:519–531. 10.1016/j.immuni.2007.01.017
    1. Lepore M., Kalinichenko A., Colone A., Paleja B., Singhal A., Tschumi A., Lee B., Poidinger M., Zolezzi F., Quagliata L., et al. . 2014. Parallel T-cell cloning and deep sequencing of human MAIT cells reveal stable oligoclonal TCRβ repertoire. Nat. Commun. 5:3866.
    1. Lin K.L., Suzuki Y., Nakano H., Ramsburg E., and Gunn M.D.. 2008. CCR2+ monocyte-derived dendritic cells and exudate macrophages produce influenza-induced pulmonary immune pathology and mortality. J. Immunol. 180:2562–2572. 10.4049/jimmunol.180.4.2562
    1. Louis C., Cook A.D., Lacey D., Fleetwood A.J., Vlahos R., Anderson G.P., and Hamilton J.A.. 2015. Specific contributions of CSF-1 and GM-CSF to the dynamics of the mononuclear phagocyte system. J. Immunol. 195:134–144. 10.4049/jimmunol.1500369
    1. Meierovics A., Yankelevich W.J., and Cowley S.C.. 2013. MAIT cells are critical for optimal mucosal immune responses during in vivo pulmonary bacterial infection. Proc. Natl. Acad. Sci. USA. 110:E3119–E3128. 10.1073/pnas.1302799110
    1. Nakano H., Lin K.L., Yanagita M., Charbonneau C., Cook D.N., Kakiuchi T., and Gunn M.D.. 2009. Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses. Nat. Immunol. 10:394–402. 10.1038/ni.1707
    1. Nambiar J.K., Ryan A.A., Kong C.U., Britton W.J., and Triccas J.A.. 2010. Modulation of pulmonary DC function by vaccine-encoded GM-CSF enhances protective immunity against Mycobacterium tuberculosis infection. Eur. J. Immunol. 40:153–161. 10.1002/eji.200939665
    1. Osterholzer J.J., Chen G.H., Olszewski M.A., Curtis J.L., Huffnagle G.B., and Toews G.B.. 2009. Accumulation of CD11b+ lung dendritic cells in response to fungal infection results from the CCR2-mediated recruitment and differentiation of Ly-6Chigh monocytes. J. Immunol. 183:8044–8053. 10.4049/jimmunol.0902823
    1. Peters W., Scott H.M., Chambers H.F., Flynn J.L., Charo I.F., and Ernst J.D.. 2001. Chemokine receptor 2 serves an early and essential role in resistance to Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. USA. 98:7958–7963. 10.1073/pnas.131207398
    1. Peters W., Cyster J.G., Mack M., Schlöndorff D., Wolf A.J., Ernst J.D., and Charo I.F.. 2004. CCR2-dependent trafficking of F4/80dim macrophages and CD11cdim/intermediate dendritic cells is crucial for T cell recruitment to lungs infected with Mycobacterium tuberculosis. J. Immunol. 172:7647–7653. 10.4049/jimmunol.172.12.7647
    1. Rahimpour A., Koay H.F., Enders A., Clanchy R., Eckle S.B., Meehan B., Chen Z., Whittle B., Liu L., Fairlie D.P., et al. . 2015. Identification of phenotypically and functionally heterogeneous mouse mucosal-associated invariant T cells using MR1 tetramers. J. Exp. Med. 212:1095–1108. 10.1084/jem.20142110
    1. Sakala I.G., Kjer-Nielsen L., Eickhoff C.S., Wang X., Blazevic A., Liu L., Fairlie D.P., Rossjohn J., McCluskey J., Fremont D.H., et al. . 2015. Functional heterogeneity and antimycobacterial effects of mouse mucosal-associated invariant T cells specific for riboflavin metabolites. J. Immunol. 195:587–601. 10.4049/jimmunol.1402545
    1. Scheicher C., Mehlig M., Zecher R., and Reske K.. 1992. Dendritic cells from mouse bone marrow: in vitro differentiation using low doses of recombinant granulocyte-macrophage colony-stimulating factor. J. Immunol. Methods. 154:253–264. 10.1016/0022-1759(92)90199-4
    1. Serbina N.V., and Pamer E.G.. 2006. Monocyte emigration from bone marrow during bacterial infection requires signals mediated by chemokine receptor CCR2. Nat. Immunol. 7:311–317. 10.1038/ni1309
    1. Serbina N.V., Salazar-Mather T.P., Biron C.A., Kuziel W.A., and Pamer E.G.. 2003. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity. 19:59–70. 10.1016/S1074-7613(03)00171-7
    1. Serbina N.V., Jia T., Hohl T.M., and Pamer E.G.. 2008. Monocyte-mediated defense against microbial pathogens. Annu. Rev. Immunol. 26:421–452. 10.1146/annurev.immunol.26.021607.090326
    1. Shi C., and Pamer E.G.. 2011. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 11:762–774. 10.1038/nri3070
    1. Sköld M., and Behar S.M.. 2008. Tuberculosis triggers a tissue-dependent program of differentiation and acquisition of effector functions by circulating monocytes. J. Immunol. 181:6349–6360. 10.4049/jimmunol.181.9.6349
    1. Sung S.S., Fu S.M., Rose C.E. Jr., Gaskin F., Ju S.T., and Beaty S.R.. 2006. A major lung CD103 (alphaE)-beta7 integrin-positive epithelial dendritic cell population expressing Langerin and tight junction proteins. J. Immunol. 176:2161–2172. 10.4049/jimmunol.176.4.2161
    1. Szeliga J., Daniel D.S., Yang C.H., Sever-Chroneos Z., Jagannath C., and Chroneos Z.C.. 2008. Granulocyte-macrophage colony stimulating factor-mediated innate responses in tuberculosis. Tuberculosis (Edinb.). 88:7–20. 10.1016/j.tube.2007.08.009
    1. Tezuka H., Abe Y., Iwata M., Takeuchi H., Ishikawa H., Matsushita M., Shiohara T., Akira S., and Ohteki T.. 2007. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature. 448:929–933. 10.1038/nature06033
    1. Trapnell B.C., Whitsett J.A., and Nakata K.. 2003. Pulmonary alveolar proteinosis. N. Engl. J. Med. 349:2527–2539. 10.1056/NEJMra023226
    1. Ussher J.E., Bilton M., Attwod E., Shadwell J., Richardson R., de Lara C., Mettke E., Kurioka A., Hansen T.H., Klenerman P., and Willberg C.B.. 2014. CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner. Eur. J. Immunol. 44:195–203. 10.1002/eji.201343509
    1. Wüthrich M., Ersland K., Sullivan T., Galles K., and Klein B.S.. 2012. Fungi subvert vaccine T cell priming at the respiratory mucosa by preventing chemokine-induced influx of inflammatory monocytes. Immunity. 36:680–692. 10.1016/j.immuni.2012.02.015
    1. Yee D., Rhinehart-Jones T.R., and Elkins K.L.. 1996. Loss of either CD4+ or CD8+ T cells does not affect the magnitude of protective immunity to an intracellular pathogen, Francisella tularensis strain LVS. J. Immunol. 157:5042–5048.
    1. Yoon H.A., Aleyas A.G., George J.A., Park S.O., Han Y.W., Lee J.H., Cho J.G., and Eo S.K.. 2006. Cytokine GM-CSF genetic adjuvant facilitates prophylactic DNA vaccine against pseudorabies virus through enhanced immune responses. Microbiol. Immunol. 50:83–92. 10.1111/j.1348-0421.2006.tb03773.x
    1. Zhan Y., Xu Y., Seah S., Brady J.L., Carrington E.M., Cheers C., Croker B.A., Wu L., Villadangos J.A., and Lew A.M.. 2010. Resident and monocyte-derived dendritic cells become dominant IL-12 producers under different conditions and signaling pathways. J. Immunol. 185:2125–2133. 10.4049/jimmunol.0903793

Source: PubMed

3
S'abonner