Circulating precursors of human CD1c+ and CD141+ dendritic cells

Gaëlle Breton, Jaeyop Lee, Yu Jerry Zhou, Joseph J Schreiber, Tibor Keler, Sarah Puhr, Niroshana Anandasabapathy, Sarah Schlesinger, Marina Caskey, Kang Liu, Michel C Nussenzweig, Gaëlle Breton, Jaeyop Lee, Yu Jerry Zhou, Joseph J Schreiber, Tibor Keler, Sarah Puhr, Niroshana Anandasabapathy, Sarah Schlesinger, Marina Caskey, Kang Liu, Michel C Nussenzweig

Abstract

Two subsets of conventional dendritic cells (cDCs) with distinct cell surface markers and functions exist in mouse and human. The two subsets of cDCs are specialized antigen-presenting cells that initiate T cell immunity and tolerance. In the mouse, a migratory cDC precursor (pre-CDC) originates from defined progenitors in the bone marrow (BM). Small numbers of short-lived pre-CDCs travel through the blood and replace cDCs in the peripheral organs, maintaining homeostasis of the highly dynamic cDC pool. However, the identity and distribution of the immediate precursor to human cDCs has not been defined. Using a tissue culture system that supports the development of human DCs, we identify a migratory precursor (hpre-CDC) that exists in human cord blood, BM, blood, and peripheral lymphoid organs. hpre-CDCs differ from premonocytes that are restricted to the BM. In contrast to earlier progenitors with greater developmental potential, the hpre-CDC is restricted to producing CD1c(+) and CD141(+) Clec9a(+) cDCs. Studies in human volunteers demonstrate that hpre-CDCs are a dynamic population that increases in response to levels of circulating Flt3L.

© 2015 Breton et al.

Figures

Figure 1.
Figure 1.
Expression of cytokine receptors on DCs and monocytes. Flow cytometry plots show expression of CD117, CD135, CD115, CD123, CD45RA, CD34, and CD116 on gated CD3−CD19−CD56− cells (gray), differentiated monocytes (CD14+ orange), pDCs (CD303+ green), CD1c+ cDCs (blue), and CD141+ cDCs (red) from peripheral blood. Table summarizes results of flow cytometry.
Figure 2.
Figure 2.
Screening of populations in the cord blood for committed cDC lineage potential. (a) Flow cytometry plots show gating of CD45+CD3−CD19−CD56−CD14−CD66b−CD1c−CD141−CD303− cells in human cord blood can be divided into four populations based on CD117, CD34, and CD135: CD34+CD117+ (CD34+), CD34−CD117− (CD117−), CD34−CD117+CD135− (CD135−), and CD34−CD117+CD135+ (CD135+). Numbers indicate the frequency of respective gates. (b) Differentiation potential of 1,000 purified cells from each of 4 populations indicated in (a) in MS5+FSG cultures for 7 d. Flow cytometry plots show gated live human CD45+ cells. (c) Flow cytometry plots show CD135+ cells indicated as in (a) can be further separated into 4 populations based on CD116, CD115, and CD45RA: CD135+CD116− (CD116−), CD135+CD116+CD45RA−(CD45RA−), CD135+CD116+CD45RA+CD115+ (CD115+), and CD135+CD116+CD45RA+ CD115− (CD115−). (d and e) Differentiation potential of 100 purified cells from each of 4 populations indicated in (c) in MS5+FSG cultures for 7 d. (d) Flow cytometry plots of CD45+ cells gated as in (c), showing expression of CD141, CLEC9a, CD1c, and CD19. (e) Graphs show mean output of pDC, CD1c+ cDC, CD141+ cDC and monocytes from each population from three independent experiments. (f) Histograms show expression of CD11c, HLA-DR, CD123, CD135, CD117, CD45RA, CD116, and CD115 on indicated cell-type. (g) Morphology of purified cord blood hpre-CDCs by Giemsa staining of cytospin preparations (100×). Dotted lines indicate cropping out of white space between cells. Bar, 10 µm.
Figure 3.
Figure 3.
hpre-CDCs in human lymphoid organs and blood. (a) Representative flow cytometry plots of gated Lin(CD3/19/56/14/66b)−DC(CD1c/141/303)−CD34− cells show gating strategy and composition of hpre-CDCs (SSCloCD117+CD116+CD135+ CD45RA+CD115−, red) and premonocytes (SSCloCD117+CD116+CD135+ CD45RA+CD115+, green) in human cord blood (CB), BM, peripheral blood (PB), and tonsil. Numbers indicate frequency of cells of parent gate (CB, n = 5; BM, n = 4, PB, n = 5; Tonsil, n = 3; n, number of donors). (b) Differentiation potential of purified hpre-CDCs indicated in (a) in MS5+FSG cultures for 5 d. Flow cytometry plots of gated live human CD45+ cells show culture output of CD141+ cDC (red) and CD1c+ cDC (blue). Representative results of four (CB), three (BM), three (PB), and two (tonsil) independent experiments are shown. (c) Differentiation potential of purified premonocytes from cord blood (CB) and BM in MS5+FSG cultures for 5 d. FACs plots show phenotype of gated live human CD45+ culture-derived cells including monocytes (orange) and CD1c+ cDCs (blue). Representative of three independent experiments are shown.
Figure 4.
Figure 4.
Proliferative capacity of hpre-CDCs. (a) Expansion of purified hpre-CDCs or CD34+ HSPCs from peripheral blood (PB) or cord blood (CB) in MS5+FSG cultures for 7 d. Graph shows the mean fold change of live human CD45+ cells from 100 input cells from three independent experiments. *, P < 0.005, unpaired two-tailed Student’s t test. (b and c) CD34+ HSPCs and hpre-CDCs were purified from CB, labeled with CFSE, and cultured in MS5+FSG for 2 or 7 d; proliferation was assessed by flow cytometry. FACs plots show (b) gated CD45+ culture-derived cells, including CD34+ cells (purple), CD34−CD1c−CD141− cells (orange), CD141+ cDCs (red), and CD1c+ cDCs (blue) and (c) their CFSE dilution. Dotted lines mark last division by hpre-CDCs. Plots are representative of three independent experiments.
Figure 5.
Figure 5.
Clonal efficiency and potential of hpre-CDC. (a) Limiting dilution outgrowth assay show clonal efficiency of CD34+ HSPCs (n = 3) or hpre-CDCs from peripheral (PB, n = 4) and cord blood (CB, n = 3) in MS5+FSG cultures for 5 d (for hpre-CDCs) or 14 d (for CD34+ HSPCs; Materials and methods). **, P < 0.05, pairwise test by ELDA. (b) Representative flow cytometry plots of gated CD45+ cells derived from single hpre-CDC clones indicated in (a) show output of CD1c+ cDCs (blue) and CD141+ cDCs (red). (c) Graphs summarize the lineage output of single clones from CD34+ HSPCS, and hpre-CDCs from cord (CB) or peripheral blood (PB). G, granulocyte; M, monocyte; L, lymphocytes.
Figure 6.
Figure 6.
hpre-CDCs descend from hCDPs. (a) Flow cytometry plots show comparison of hCDP (purple) and hpre-CDC (orange) in cord blood. (b) Flow cytometry plots of gated CD45+Lin(CD3/19/56/14)−CD34+ show phenotype and purity of magnetically enriched cord blood (CB) CD34+ cells (presorting, upper) and sorted cells (post-sorting, bottom). hCDPs were gated as CD34+CD38hiCD45RA+CD10−CD123hi. (c) Differentiation kinetics of hCDPs purified from CB in MS5+FSG cultures for 1, 2, or 4 d. FACs plots show culture output of live human CD45+ cells, including CD34+ cells (purple), pDC (green), CD1c+ cDC (blue), CD141+ cDCs (red), and hpre-CDCs (orange). Representative of four independent experiments are shown. Graphs summarize composition of indicated populations among total hCD45+ cells. Bars are mean values from four independent experiments, and error bars are SEM.
Figure 7.
Figure 7.
Flt3L administration increases circulating DC subsets and hpre-CDCs in humans. PBMCs from Flt3L-treated volunteers (n = 3; 25 µg/kg for 10 consecutive days) were analyzed by flow cytometry over a 21-d period to assess the expansion of DC subsets (CD1c+ cDCs [blue], CD141+ cDCs [red], and pDCs [green]; hpre-CDCs (gray); monocytes (orange); and granulocytes (brown). (a) Representative flow cytometry dot plots show DC subsets and hpre-CDCs in blood (gating strategy in Fig. S4). (b) Graphs show the kinetics of cell number of cDC subsets, pDCs, and hpre-CDCs over 21 d in 3 Flt3L-treated volunteers. The absolute numbers per milliliter of blood were obtained by multiplying the number of cells (obtained by flow cytometry) by the total number of PBMCs per milliliter of blood. (c) Representative histograms show CD135 expression on CD141+ cDCs, CD1c+ cDCs, pDCs and hpre-CDCs over 21 d in one Flt3L-treated volunteer. (d) Differentiation potential of purified hpre-CDCs from blood of Flt3L-injected volunteers in MS5+FSG cultures after 7 d. Flow cytometry plots of gated CD45+ cells from culture show output of CD141+ cDCs (red), CD1c+ cDCs (blue), and lack of pDCs (green gate). (e) Representative flow cytometry dot plots show CD14+ monocytes and CD66b+ granulocytes in blood (gating strategy in Fig. S4). (f) Graphs show the kinetics of cell number of monocytes and granulocytes over 21 d in 3 Flt3L-treated volunteers. The absolute numbers per milliliter of blood were obtained by multiplying the number of cells (obtained by flow cytometry) by the total number of PBMCs per milliliter of blood. (g) Graph showing the mean fold change increase from the three patients (d1 vs. d14) of hpre-CDCs (gray), CD1c+ cDCs (blue), CD141+ cDCs (red), pDCs (green), monocytes (orange), and granulocytes (brown) per milliliter of blood. Error bars are SDs.

References

    1. Bachem A., Güttler S., Hartung E., Ebstein F., Schaefer M., Tannert A., Salama A., Movassaghi K., Opitz C., Mages H.W., et al. . 2010. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. Med. 207:1273–1281 10.1084/jem.20100348
    1. Banchereau J., and Steinman R.M.. 1998. Dendritic cells and the control of immunity. Nature. 392:245–252 10.1038/32588
    1. Bogunovic M., Ginhoux F., Helft J., Shang L., Hashimoto D., Greter M., Liu K., Jakubzick C., Ingersoll M.A., Leboeuf M., et al. . 2009. Origin of the lamina propria dendritic cell network. Immunity. 31:513–525 10.1016/j.immuni.2009.08.010
    1. Chicha L., Jarrossay D., and Manz M.G.. 2004. Clonal type I interferon-producing and dendritic cell precursors are contained in both human lymphoid and myeloid progenitor populations. J. Exp. Med. 200:1519–1524 10.1084/jem.20040809
    1. Cohn L., Chatterjee B., Esselborn F., Smed-Sörensen A., Nakamura N., Chalouni C., Lee B.C., Vandlen R., Keler T., Lauer P., et al. . 2013. Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation. J. Exp. Med. 210:1049–1063 10.1084/jem.20121251
    1. Collin M., Bigley V., Haniffa M., and Hambleton S.. 2011. Human dendritic cell deficiency: the missing ID? Nat. Rev. Immunol. 11:575–583 10.1038/nri3046
    1. Crozat K., Guiton R., Contreras V., Feuillet V., Dutertre C.A., Ventre E., Vu Manh T.P., Baranek T., Storset A.K., Marvel J., et al. . 2010. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J. Exp. Med. 207:1283–1292 10.1084/jem.20100223
    1. Dickinson R.E., Milne P., Jardine L., Zandi S., Swierczek S.I., McGovern N., Cookson S., Ferozepurwalla Z., Langridge A., Pagan S., et al. . 2014. The evolution of cellular deficiency in GATA2 mutation. Blood. 123:863–874 10.1182/blood-2013-07-517151
    1. Doulatov S., Notta F., Eppert K., Nguyen L.T., Ohashi P.S., and Dick J.E.. 2010. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat. Immunol. 11:585–593 10.1038/ni.1889
    1. Dudziak D., Kamphorst A.O., Heidkamp G.F., Buchholz V.R., Trumpfheller C., Yamazaki S., Cheong C., Liu K., Lee H.W., Park C.G., et al. . 2007. Differential antigen processing by dendritic cell subsets in vivo. Science. 315:107–111 10.1126/science.1136080
    1. Eidenschenk C., Crozat K., Krebs P., Arens R., Popkin D., Arnold C.N., Blasius A.L., Benedict C.A., Moresco E.M., Xia Y., and Beutler B.. 2010. Flt3 permits survival during infection by rendering dendritic cells competent to activate NK cells. Proc. Natl. Acad. Sci. USA. 107:9759–9764 10.1073/pnas.1005186107
    1. Fogg D.K., Sibon C., Miled C., Jung S., Aucouturier P., Littman D.R., Cumano A., and Geissmann F.. 2006. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science. 311:83–87 10.1126/science.1117729
    1. Ginhoux F., and Schlitzer A.. 2014. CD11b+ DCs rediscovered: implications for vaccination. Expert Rev. Vaccines. 13:445–447 10.1586/14760584.2014.893196
    1. Ginhoux F., Liu K., Helft J., Bogunovic M., Greter M., Hashimoto D., Price J., Yin N., Bromberg J., Lira S.A., et al. . 2009. The origin and development of nonlymphoid tissue CD103+ DCs. J. Exp. Med. 206:3115–3130 10.1084/jem.20091756
    1. Granelli-Piperno A., Shimeliovich I., Pack M., Trumpfheller C., and Steinman R.M.. 2006. HIV-1 selectively infects a subset of nonmaturing BDCA1-positive dendritic cells in human blood. J. Immunol. 176:991–998 10.4049/jimmunol.176.2.991
    1. Grouard G., Rissoan M.C., Filgueira L., Durand I., Banchereau J., and Liu Y.J.. 1997. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 185:1101–1111 10.1084/jem.185.6.1101
    1. Guermonprez P., Helft J., Claser C., Deroubaix S., Karanje H., Gazumyan A., Darasse-Jèze G., Telerman S.B., Breton G., Schreiber H.A., et al. . 2013. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection. Nat. Med. 19:730–738 10.1038/nm.3197
    1. Haniffa M., Shin A., Bigley V., McGovern N., Teo P., See P., Wasan P.S., Wang X.N., Malinarich F., Malleret B., et al. . 2012. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity. 37:60–73 10.1016/j.immuni.2012.04.012
    1. Haniffa M., Collin M., and Ginhoux F.. 2013. Ontogeny and functional specialization of dendritic cells in human and mouse. Adv. Immunol. 120:1–49 10.1016/B978-0-12-417028-5.00001-6
    1. Hettinger J., Richards D.M., Hansson J., Barra M.M., Joschko A.C., Krijgsveld J., and Feuerer M.. 2013. Origin of monocytes and macrophages in a committed progenitor. Nat. Immunol. 14:821–830 10.1038/ni.2638
    1. Jongbloed S.L., Kassianos A.J., McDonald K.J., Clark G.J., Ju X., Angel C.E., Chen C.J., Dunbar P.R., Wadley R.B., Jeet V., et al. . 2010. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J. Exp. Med. 207:1247–1260 10.1084/jem.20092140
    1. Kabashima K., Banks T.A., Ansel K.M., Lu T.T., Ware C.F., and Cyster J.G.. 2005. Intrinsic lymphotoxin-beta receptor requirement for homeostasis of lymphoid tissue dendritic cells. Immunity. 22:439–450 10.1016/j.immuni.2005.02.007
    1. Kamphorst A.O., Guermonprez P., Dudziak D., and Nussenzweig M.C.. 2010. Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J. Immunol. 185:3426–3435 10.4049/jimmunol.1001205
    1. Karsunky H., Merad M., Cozzio A., Weissman I.L., and Manz M.G.. 2003. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. J. Exp. Med. 198:305–313 10.1084/jem.20030323
    1. KC W., Satpathy A.T., Rapaport A.S., Briseño C.G., Wu X., Albring J.C., Russler-Germain E.V., Kretzer N.M., Durai V., Persaud S.P., et al. . 2014. L-Myc expression by dendritic cells is required for optimal T-cell priming. Nature. 507:243–247 10.1038/nature12967
    1. Lee J., Breton G., Oliveira T.Y.K., Zhou Y.J., Aljoufi A., Puhr S., Cameron M.J., Sékaly R.-P., Nussenzweig M.C., and Liu K.. 2015. Restricted dendritic cell and monocyte progenitors in human cord blood and bone marrow. J. Exp. Med. 212:xxx–xxx 10.1084/jem.20141441
    1. Liu Y.J.2005. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu. Rev. Immunol. 23:275–306 10.1146/annurev.immunol.23.021704.115633
    1. Liu K., and Nussenzweig M.C.. 2010. Development and homeostasis of dendritic cells. Eur. J. Immunol. 40:2099–2102 10.1002/eji.201040501
    1. Liu K., Waskow C., Liu X., Yao K., Hoh J., and Nussenzweig M.. 2007. Origin of dendritic cells in peripheral lymphoid organs of mice. Nat. Immunol. 8:578–583 10.1038/ni1462
    1. Liu K., Victora G.D., Schwickert T.A., Guermonprez P., Meredith M.M., Yao K., Chu F.F., Randolph G.J., Rudensky A.Y., and Nussenzweig M.. 2009. In vivo analysis of dendritic cell development and homeostasis. Science. 324:392–397 10.1126/science.1170540
    1. Maraskovsky E., Pulendran B., Brasel K., Teepe M., Roux E.R., Shortman K., Lyman S.D., and McKenna H.J.. 1997. Dramatic numerical increase of functionally mature dendritic cells in FLT3 ligand-treated mice. Adv. Exp. Med. Biol. 417:33–40 10.1007/978-1-4757-9966-8_6
    1. Maraskovsky E., Daro E., Roux E., Teepe M., Maliszewski C.R., Hoek J., Caron D., Lebsack M.E., and McKenna H.J.. 2000. In vivo generation of human dendritic cell subsets by Flt3 ligand. Blood. 96:878–884.
    1. McKenna H.J., Stocking K.L., Miller R.E., Brasel K., De Smedt T., Maraskovsky E., Maliszewski C.R., Lynch D.H., Smith J., Pulendran B., et al. . 2000. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood. 95:3489–3497.
    1. Merad M., Sathe P., Helft J., Miller J., and Mortha A.. 2013. 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 10.1146/annurev-immunol-020711-074950
    1. Murphy K.M.2013. Transcriptional control of dendritic cell development. Adv. Immunol. 120:239–267 10.1016/B978-0-12-417028-5.00009-0
    1. Naik S.H., Metcalf D., van Nieuwenhuijze A., Wicks I., Wu L., O’Keeffe M., and Shortman K.. 2006. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat. Immunol. 7:663–671 10.1038/ni1340
    1. Naik S.H., Sathe P., Park H.Y., Metcalf D., Proietto A.I., Dakic A., Carotta S., O’Keeffe M., Bahlo M., Papenfuss A., et al. . 2007. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat. Immunol. 8:1217–1226 10.1038/ni1522
    1. Nizzoli G., Krietsch J., Weick A., Steinfelder S., Facciotti F., Gruarin P., Bianco A., Steckel B., Moro M., Crosti M., et al. . 2013. Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T-cell responses. Blood. 122:932–942 10.1182/blood-2013-04-495424
    1. O’Doherty U., Peng M., Gezelter S., Swiggard W.J., Betjes M., Bhardwaj N., and Steinman R.M.. 1994. Human blood contains two subsets of dendritic cells, one immunologically mature and the other immature. Immunology. 82:487–493.
    1. Onai N., Obata-Onai A., Schmid M.A., Ohteki T., Jarrossay D., and Manz M.G.. 2007. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8:1207–1216 10.1038/ni1518
    1. Patterson S., Donaghy H., Amjadi P., Gazzard B., Gotch F., and Kelleher P.. 2005. Human BDCA-1-positive blood dendritic cells differentiate into phenotypically distinct immature and mature populations in the absence of exogenous maturational stimuli: differentiation failure in HIV infection. J. Immunol. 174:8200–8209 10.4049/jimmunol.174.12.8200
    1. Poulin L.F., Salio M., Griessinger E., Anjos-Afonso F., Craciun L., Chen J.L., Keller A.M., Joffre O., Zelenay S., Nye E., et al. . 2010. Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J. Exp. Med. 207:1261–1271 10.1084/jem.20092618
    1. Pulendran B., Banchereau J., Burkeholder S., Kraus E., Guinet E., Chalouni C., Caron D., Maliszewski C., Davoust J., Fay J., and Palucka K.. 2000. Flt3-ligand and granulocyte colony-stimulating factor mobilize distinct human dendritic cell subsets in vivo. J. Immunol. 165:566–572 10.4049/jimmunol.165.1.566
    1. Robbins S.H., Walzer T., Dembélé D., Thibault C., Defays A., Bessou G., Xu H., Vivier E., Sellars M., Pierre P., et al. . 2008. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol. 9:R17 10.1186/gb-2008-9-1-r17
    1. Schlitzer A., McGovern N., Teo P., Zelante T., Atarashi K., Low D., Ho A.W., See P., Shin A., Wasan P.S., et al. . 2013. IRF4 transcription factor-dependent CD11b+ dendritic cells in human and mouse control mucosal IL-17 cytokine responses. Immunity. 38:970–983 10.1016/j.immuni.2013.04.011
    1. Segura E., Valladeau-Guilemond J., Donnadieu M.H., Sastre-Garau X., Soumelis V., and Amigorena S.. 2012. Characterization of resident and migratory dendritic cells in human lymph nodes. J. Exp. Med. 209:653–660 10.1084/jem.20111457
    1. Siegal F.P., Kadowaki N., Shodell M., Fitzgerald-Bocarsly P.A., Shah K., Ho S., Antonenko S., and Liu Y.J.. 1999. The nature of the principal type 1 interferon-producing cells in human blood. Science. 284:1835–1837 10.1126/science.284.5421.1835
    1. Varol C., Vallon-Eberhard A., Elinav E., Aychek T., Shapira Y., Luche H., Fehling H.J., Hardt W.D., Shakhar G., and Jung S.. 2009. Intestinal lamina propria dendritic cell subsets have different origin and functions. Immunity. 31:502–512 10.1016/j.immuni.2009.06.025
    1. Waskow C., Liu K., Darrasse-Jèze G., Guermonprez P., Ginhoux F., Merad M., Shengelia T., Yao K., and Nussenzweig M.. 2008. The receptor tyrosine kinase Flt3 is required for dendritic cell development in peripheral lymphoid tissues. Nat. Immunol. 9:676–683 10.1038/ni.1615
    1. Wright D.E., Wagers A.J., Gulati A.P., Johnson F.L., and Weissman I.L.. 2001. Physiological migration of hematopoietic stem and progenitor cells. Science. 294:1933–1936 10.1126/science.1064081
    1. Yu C.I., Becker C., Wang Y., Marches F., Helft J., Leboeuf M., Anguiano E., Pourpe S., Goller K., Pascual V., et al. . 2013. Human CD1c+ dendritic cells drive the differentiation of CD103+ CD8+ mucosal effector T cells via the cytokine TGF-β. Immunity. 38:818–830 10.1016/j.immuni.2013.03.004

Source: PubMed

3
订阅