Suk-Jo Kang, Hong-Erh Liang, Boris Reizis, Richard M. Locksley 

Slides:



Advertisements
Similar presentations
The Humoral Immune Response Is Initiated in Lymph Nodes by B Cells that Acquire Soluble Antigen Directly in the Follicles  Kathryn A. Pape, Drew M. Catron,
Advertisements

Volume 42, Issue 1, Pages (January 2015)
Volume 40, Issue 1, Pages (January 2014)
Volume 6, Issue 5, Pages (November 2009)
Volume 31, Issue 1, Pages (July 2009)
Volume 29, Issue 3, Pages (September 2008)
Volume 42, Issue 3, Pages (March 2015)
Volume 27, Issue 4, Pages (October 2007)
Volume 34, Issue 3, Pages (March 2011)
Volume 40, Issue 6, Pages (June 2014)
Hans-Peter Raué, Carol Beadling, Jennifer Haun, Mark K. Slifka 
Volume 31, Issue 5, Pages (November 2009)
Lung Natural Helper Cells Are a Critical Source of Th2 Cell-Type Cytokines in Protease Allergen-Induced Airway Inflammation  Timotheus Y.F. Halim, Ramona H.
Volume 45, Issue 1, Pages (July 2016)
Frederic Geissmann, Steffen Jung, Dan R. Littman  Immunity 
Volume 31, Issue 5, Pages (November 2009)
Cellular Mechanisms of Fatal Early-Onset Autoimmunity in Mice with the T Cell-Specific Targeting of Transforming Growth Factor-β Receptor  Julien C. Marie,
Volume 40, Issue 3, Pages (March 2014)
Inflammatory Monocytes Activate Memory CD8+ T and Innate NK Lymphocytes Independent of Cognate Antigen during Microbial Pathogen Invasion  Saïdi M'Homa.
Volume 165, Issue 3, Pages (April 2016)
Volume 29, Issue 2, Pages (August 2008)
Volume 29, Issue 2, Pages (August 2008)
Volume 36, Issue 1, Pages (January 2012)
Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity  Zhang-Xu Liu, Sugantha Govindarajan,
Volume 43, Issue 4, Pages (October 2015)
Volume 7, Issue 2, Pages (February 2010)
Volume 33, Issue 4, Pages (October 2010)
B-1a and B-1b Cells Exhibit Distinct Developmental Requirements and Have Unique Functional Roles in Innate and Adaptive Immunity to S. pneumoniae  Karen.
Volume 34, Issue 3, Pages (March 2011)
Volume 34, Issue 5, Pages (May 2011)
Volume 37, Issue 5, Pages (November 2012)
Volume 33, Issue 6, Pages (December 2010)
Volume 31, Issue 5, Pages (November 2009)
Volume 27, Issue 2, Pages (August 2007)
Volume 6, Issue 5, Pages (November 2009)
Volume 22, Issue 3, Pages (March 2005)
A Mutation in the Nlrp3 Gene Causing Inflammasome Hyperactivation Potentiates Th17 Cell-Dominant Immune Responses  Guangxun Meng, Fuping Zhang, Ivan Fuss,
Volume 40, Issue 1, Pages (January 2014)
Volume 36, Issue 6, Pages (June 2012)
Blimp-1 Transcription Factor Is Required for the Differentiation of Effector CD8+ T Cells and Memory Responses  Axel Kallies, Annie Xin, Gabrielle T.
Volume 22, Issue 2, Pages (February 2005)
Francis Coffey, Boris Alabyev, Tim Manser  Immunity 
Oral Tolerance Can Be Established via Gap Junction Transfer of Fed Antigens from CX3CR1+ Macrophages to CD103+ Dendritic Cells  Elisa Mazzini, Lucia Massimiliano,
Volume 30, Issue 2, Pages (February 2009)
Volume 14, Issue 2, Pages (August 2013)
Volume 38, Issue 6, Pages (June 2013)
Volume 38, Issue 3, Pages (March 2013)
Volume 34, Issue 3, Pages (March 2011)
Volume 22, Issue 5, Pages e5 (November 2017)
Volume 38, Issue 3, Pages (March 2013)
Volume 22, Issue 4, Pages (April 2005)
Opposing Effects of TGF-β and IL-15 Cytokines Control the Number of Short-Lived Effector CD8+ T Cells  Shomyseh Sanjabi, Munir M. Mosaheb, Richard A.
Volume 29, Issue 5, Pages (November 2008)
Volume 22, Issue 5, Pages e5 (November 2017)
Volume 19, Issue 9, Pages (May 2017)
Volume 71, Issue 7, Pages (April 2007)
Karima R.R. Siddiqui, Sophie Laffont, Fiona Powrie  Immunity 
Volume 34, Issue 5, Pages (May 2011)
Volume 26, Issue 4, Pages (April 2007)
Volume 19, Issue 1, Pages (July 2003)
Volume 32, Issue 1, Pages (January 2010)
Volume 38, Issue 2, Pages (February 2013)
Volume 31, Issue 5, Pages (November 2009)
Volume 35, Issue 4, Pages (October 2011)
Volume 33, Issue 5, Pages (November 2010)
Volume 30, Issue 5, Pages (May 2009)
Volume 3, Issue 5, Pages (May 2001)
Volume 37, Issue 4, Pages (October 2012)
Volume 33, Issue 2, Pages (August 2010)
Volume 25, Issue 4, Pages (October 2006)
Presentation transcript:

Regulation of Hierarchical Clustering and Activation of Innate Immune Cells by Dendritic Cells  Suk-Jo Kang, Hong-Erh Liang, Boris Reizis, Richard M. Locksley  Immunity  Volume 29, Issue 5, Pages 819-833 (November 2008) DOI: 10.1016/j.immuni.2008.09.017 Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 1 Innate Cell Clustering and IFN-γ Production in Response to Listeria (A) Positioning of myeloid (CD11b+), NK cells (NK1.1+), and B cells (B220+) in the spleens from mice either uninfected or infected with Listeria 24 hr previously. T and B areas are labeled; RP denotes red pulp; arrows indicate the central arteriole; 40×. (B) Positioning of IFN-γ-producing cells. Conditions are as in (A). (C) Gating strategy for NK cells, NKT cells, and T cells in spleen before (−) and 24 hr after (+) Listeria infection. Intracellular IFN-γ shown in right panels for cell types designated. (D) Percentages of IFN-γ-positive cells among designated cell types before (−) and 24 hr after (+) infection with Listeria. n = 3 for (−), 4 for (+); red bars represent means. (E) Total numbers of IFN-γ-positive cells in the spleen before (−) and 24 hr after (+) infection with Listeria. n = 3 for (−), 4 for (+); red bars represent means. In (A) and (B), data are representative from over four independent experiments. In (C)–(E), data are representative from two independent experiments. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 2 Time Course of Innate Immune Response after Listeria Infection in Spleen Spleens from the IFN-γ reporter mice infected with Listeria were harvested at the indicated times. Molecules stained for are color matched and shown to the left of each row: eCFP, enhanced cyan fluorescent protein as a marker for IFN-γ, green; cytokines (IFN-γ, IL-12), red; Thy1.2 staining marks T cells, blue; NK1.1, NK cells, red; CD11b, CD11b+ myeloid cells, green in the third and fourth rows, blue in the bottom row; Listeria, red; iNOS, red; HKLM, heat-killed Listeria; LLO-, listeriolysin O-deficient Listeria; RP, red pulp; WP, white pulp; arrowheads, terminal arteriole; arrow, central arteriole; 40× except the penultimate row (100×). Approximately 300 WPs, as based on morphology and staining, were counted in multiple tissue sections at indicated times and related to the numbers of CD11b+ clusters visible in the same fields. Data are representative of two independent experiments. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 3 Characterization of CD11b+ Myeloid Cells in Spleen (A) Gating scheme and markers for the delineation by flow cytometry of pDCs (orange), conventional cDCs (green), PMNs (neutrophils, blue), and monocytes (mono, red) from spleens of uninfected mice. Expression of Ly6G on various myeloid populations, which are color coded as in the third panel and graphed at top right. (B) Simplified scheme to separate PMNs and monocytes in uninfected and infected mouse spleen and differentiation of monocytes to TipDCs. Mice were infected 48 hr previously with Listeria. Expression of iNOS and MHC class II from cells gated is depicted, with percentage of monocytes expressing iNOS shown. Gray histogram shows isotype control; solid line indicates iNOS or class II. (C) Distinguishing monocytes and neutrophils by using 7/4, Ly6C, and Ly6G. Mice were infected 48 hr previously with Listeria. Percentages of gated cell types among live cells are shown. Expression of Ly6G (G1, red line; G2, blue line) and iNOS on gated populations is graphed at right. Gray histogram shows isotype control; solid line indicates iNOS. (D) Positioning of monocytes and neutrophils in the spleen of mice before and 24 hr after Listeria infection. The inset areas in the left-most panels are shown at right. WP, white pulp; RP, red pulp; T, T cell area; B, B cell area; arrows, monocytes; arrowheads, neutrophils; 200×. Data are representative from at least two independent experiments. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 4 IFN-γ from NK Cells Mediates the Maturation of Monocytes to TipDCs (A) Quantitation of numbers of respective cells after depletion and infection with 2000 Listeria is shown. Graph shows mean ± SD; n = 6 for each group; ∗∗, p < 0.01. (B) Immunohistochemistry of mice treated with anti-Gr1 or isotype control antibody and infected 48 hr later with Listeria. The spleens were examined 24 hr later. Myeloid cell depletion had no effect on NK cell clustering. 40×. (C) Spleen immunohistochemistry of positioning of NK cells, myeloid cells, and cells producing IFN-γ 24 hr after Listeria infection in wild-type or CCR2-deficient (Ccr2−/−) mice. 40×. (D) Mice were treated with anti-asialoGM1 (anti-aGM1) or control Ab (rabbit IgG), and spleens were analyzed 48 hr later for proportions of NK cells (uppermost gates) and NKT cells (right gates). Distribution of asialoGM1 staining on NK and NKT cells from spleen denoted in the right panel, with percentage of NKT cells that were stained with anti-aGM1 indicated. (E) NK cell-independent clustering of CD11b+ myeloid cells. Mice were treated with control rabbit antibody (control) or anti-asialoGM1(aGM1) and infected 48 hr later with Listeria. The next day, spleens were harvested and imaged with immunohistochemistry. 40×. (F) Monocytes in the spleen were gated as in Figure 3B and analyzed for expression of iNOS and MHC class II from the wild-type (WT), IFN-γ-deficient (Ifng−/−), and RAG1-deficient (Rag1−/−) mice infected 48 hr earlier with Listeria. Top: representative flow-cytometry plots. Bottom: summary of results. Mean of WT arbitrarily set as 100%. MFI denotes mean fluorescence intensity; n = 4 for each group; ∗∗, p < 0.01. (G) Analysis of TipDC-like monocytes in spleen from WT and γc-deficient (Il2rg−/−) mice infected 48 hr earlier with Listeria. Summary with quantitation is shown. Mean of WT arbitrarily set as 100%. n = 4 for WT and 3 for Il2rg−/− group; ∗∗, p < 0.01. (H) Analysis of TipDC-like monocytes in spleen from Rag1−/− mice treated with control rabbit antibody (control) or anti-asialoGM1(anti-aGM1) and infected with Listeria 48 hr before. Summary with quantitation is shown. Mean of control Ab-treated mice arbitrarily set as 100%. n = 3 for control and 4 for anti-aGM1 group; ∗, p < 0.05. (I) Analysis of TipDC-like monocytes in spleen from Rag1−/− mice and Rag1−/−Ifng−/− mice infected with Listeria 48 hr before. Summary with quantitation is shown. Mean of Rag1−/− mice arbitrarily set as 100%. n = 3 for each group; ∗∗, p < 0.01. Data are representative from at least two independent experiments. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 5 CD11c+ Cells Are Required for Innate Cell Clustering and Activation in Response to Listeria (A) Spleen immunohistochemistry for indicated markers 24 hr after (upper panels) and before (bottom panels) infection with Listeria. DTA denotes diphtheria toxin A; Cre denotes CD11c-cre recombinase; CD169 (sialoadhesin, MOMA-1) is a marker for metallophilic macrophages; CD209b (SIGNR1, ER-TR9) is a marker for marginal-zone macrophages; 40×. Data are representative of four independent experiments. (B) Representative flow-cytometric analysis of NK cells, NKT cells, and T cells that express intracellular IFN-γ when isolated from the indicated mice 24 hr after infection with Listeria. Percentages of IFN-γ-positive cells among designated cell types are shown. Data are representative of two independent experiments. (C) Quantitation of flow-cytometric analysis from two combined experiments from (B). Graphs show mean ± SD; ∗, p < 0.05; ∗∗, p < 0.01. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 6 NK Cell Clustering and Activation Require a Pertussis-Toxin-Sensitive Receptor (A) Spleen immunohistochemistry for indicated markers 24 hr after Listeria infection of mice pretreated with PBS or PTX. 40×. (B) Representative (upper panels, percentages of IFN-γ-positive cells among designated cell types shown) and quantitative (lower panels) summary of intracellular IFN-γ detection from the indicated spleen cells 24 hr after Listeria infection. Graphs show mean ± SD; n = 4 for each group; ∗∗, p < 0.01. (C) NK cells purified from Rag1−/− mice were labeled with CFSE and treated with PBS (vehicle control) or PTX before transfer to recipient WT mice. Spleen sections were stained for CFSE and IFN-γ 24 hr after infection with Listeria. Quantitation of movement of donor (CFSE+), as compared to host (CFSE−), NK cells into clusters, as defined by association with an IFN-γ-producing cluster, from multiple experiments is shown below. n = 3 for PBS and 4 for PTX group; ∗∗, p < 0.01. (D) NK cells prepared as in (C) were isolated from infected mice 24 hr after Listeria infection and stained for IFN-γ. Loss of IFN-γ production in PTX-treated NK cells is shown in graph. Mean of IFN-γ-producing host cells was set arbitrarily as 100%. n = 4 for each group; ∗∗, p < 0.01. (E) Spleen immunohistochemistry of positioning of NK cells, myeloid cells, and cells producing IFN-γ 24 hr after Listeria infection in wild-type or Ccr5−/− mice. 40×. (F) Quantitation of flow-cytometric analysis from multiple experiments of spleen cells staining for intracellular IFN-γ 24 hr after Listeria infection from the designated mice. Mean of WT was set arbitrarily as 100%; graph shows mean ± SD; n = 9 for WT and 8 for Ccr5−/− group; ∗, p < 0.05; ∗∗, p < 0.01. In (A), (B), (E), and (F), data are representative of three independent experiments. In (C) and (D), data are representative of two independent experiments. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions

Figure 7 Signaling Pathways Involved in Innate Immune Cell Clustering and IFN-γ Production (A–F) Immunohistochemistry of coinfected matched WT or mice with specific gene deletion (MyD88, Myd88; IL-12p40, Il12b; IL-8, Il18; ASC, Pycard; NOD1, Nod1; NOD2, Nod2; IRF3, Irf3) to assess NK cell clustering (upper panels) and IFN-γ production (lower panels) 24 hr after Listeria infection. Data are representative of at least two independent experiments with two or more mice per group for each experiment. Immunity 2008 29, 819-833DOI: (10.1016/j.immuni.2008.09.017) Copyright © 2008 Elsevier Inc. Terms and Conditions