Volume 21, Issue 4, Pages (October 2004)

Slides:



Advertisements
Similar presentations
Fig. 1. TLR4 expression is much greater on macrophages than on DCs
Advertisements

Volume 6, Issue 4, Pages (October 2009)
Volume 45, Issue 6, Pages (December 2016)
Shamim A. Mollah, Joseph S. Dobrin, Rachel E
Dynamic Changes in Resident and Infiltrating Epidermal Dendritic Cells in Active and Resolved Psoriasis  Elisa Martini, Maria Wikén, Stanley Cheuk, Irène.
Volume 25, Issue 1, Pages (July 2006)
Critical Role for Skin-Derived Migratory DCs and Langerhans Cells in TFH and GC Responses after Intradermal Immunization  Clément Levin, Olivia Bonduelle,
Tatsukuni Ohno, Yuta Kondo, Chenyang Zhang, Siwen Kang, Miyuki Azuma 
Novel function for interleukin-7 in dendritic cell development
Volume 42, Issue 3, Pages (March 2015)
EpCAM Expressed by Murine Epidermal Langerhans Cells Modulates Immunization to an Epicutaneously Applied Protein Antigen  Takeshi Ouchi, Gaku Nakato,
Volume 16, Issue 2, Pages (February 2002)
Langerhans Cells Facilitate UVB-Induced Epidermal Carcinogenesis
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 21, Issue 2, Pages (August 2004)
CD4+CD3− Cells Induce Peyer's Patch Development
The Late Endosomal Adaptor Molecule p14 (LAMTOR2) Regulates TGFβ1-Mediated Homeostasis of Langerhans Cells  Florian Sparber, Christoph H. Tripp, Kerstin.
CD69 Modulates Sphingosine-1-Phosphate-Induced Migration of Skin Dendritic Cells  Amalia Lamana, Pilar Martin, Hortensia de la Fuente, Laura Martinez-Muñoz,
Antigen-Specific Peripheral Tolerance Induced by Topical Application of NF-κB Decoy Oligodeoxynucleotide  Iwao Isomura, Kunio Tsujimura, Akimichi Morita 
Volume 24, Issue 2, Pages (February 2006)
Volume 40, Issue 3, Pages (March 2014)
IL-23 from Langerhans Cells Is Required for the Development of Imiquimod-Induced Psoriasis-Like Dermatitis by Induction of IL-17A-Producing γδ T Cells 
Sandra Holzmann, BSc, Christoph H
Langerhans Cells Are Required for UVR-Induced Immunosuppression
Depletion of Host CCR7+ Dendritic Cells Prevented Donor T Cell Tissue Tropism in Anti- CD3–Conditioned Recipients  Wei He, Jeremy J. Racine, Heather F.
Volume 29, Issue 2, Pages (August 2008)
Volume 23, Issue 6, Pages (December 2005)
Volume 42, Issue 5, Pages (May 2015)
IL-10-Producing Langerhans Cells and Regulatory T Cells Are Responsible for Depressed Contact Hypersensitivity in Grafted Skin  Ryutaro Yoshiki, Kenji.
Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity  Zhang-Xu Liu, Sugantha Govindarajan,
Volume 7, Issue 2, Pages (February 2010)
Volume 103, Issue 5, Pages (November 2000)
Volume 37, Issue 6, Pages (December 2012)
Volume 21, Issue 3, Pages (September 2004)
Interleukin-1β But Not Tumor Necrosis Factor is Involved in West Nile Virus-Induced Langerhans Cell Migration from the Skin in C57BL/6 Mice  Scott N.
Volume 45, Issue 6, Pages (December 2016)
Activation of the Arylhydrocarbon Receptor Causes Immunosuppression Primarily by Modulating Dendritic Cells  Anika Bruhs, Thomas Haarmann-Stemmann, Katrin.
Volume 37, Issue 5, Pages (November 2012)
Integrin αE(CD103) Is Involved in Regulatory T-Cell Function in Allergic Contact Hypersensitivity  Andrea Braun, Nadin Dewert, Fiona Brunnert, Viktor.
Differential Effects of Corticosteroids and Pimecrolimus on the Developing Skin Immune System in Humans and Mice  Simone Meindl, Christine Vaculik, Josef.
Volume 14, Issue 1, Pages (January 2001)
CD24a Expression Levels Discriminate Langerhans Cells from Dermal Dendritic Cells in Murine Skin and Lymph Nodes  Susanne Stutte, Bettina Jux, Charlotte.
Volume 24, Issue 2, Pages (February 2006)
Volume 17, Issue 5, Pages (May 2013)
Lymphatic Dysfunction Impairs Antigen-Specific Immunization, but Augments Tissue Swelling Following Contact with Allergens  Makoto Sugaya, Yoshihiro Kuwano,
Volume 29, Issue 6, Pages (December 2008)
Impaired Initiation of Contact Hypersensitivity by FTY720
Pivotal Role of Dermal IL-17-Producing γδ T Cells in Skin Inflammation
Volume 48, Issue 4, Pages e4 (April 2018)
Jeffery M. Cowden, Mai Zhang, Paul J. Dunford, Robin L. Thurmond 
Volume 38, Issue 3, Pages (March 2013)
Overexpression of IL-4 Alters the Homeostasis in the Skin
Volume 11, Issue 6, Pages (December 1999)
B Cells Activated in Lymph Nodes in Response to Ultraviolet Irradiation or by Interleukin-10 Inhibit Dendritic Cell Induction of Immunity  Scott N. Byrne,
Volume 119, Issue 1, Pages (July 2000)
Volume 6, Issue 4, Pages (October 2009)
Simone C. Zimmerli, Conrad Hauser  Journal of Investigative Dermatology 
Engagement of CD47 Inhibits the Contact Hypersensitivity Response Via the Suppression of Motility and B7 Expression by Langerhans Cells  Xijun Yu, Atsushi.
Sandra Holzmann, BSc, Christoph H
Linda J. Johnston, Nicholas J.C. King 
Karima R.R. Siddiqui, Sophie Laffont, Fiona Powrie  Immunity 
Volume 42, Issue 4, Pages (April 2015)
Volume 35, Issue 6, Pages (December 2011)
Topical Application of Sphingosine-1-Phosphate and FTY720 Attenuate Allergic Contact Dermatitis Reaction through Inhibition of Dendritic Cell Migration 
Volume 28, Issue 5, Pages (May 2008)
No defect in T-cell priming, secondary response, or tolerance induction in response to inhaled antigens in Fms-like tyrosine kinase 3 ligand–deficient.
Volume 25, Issue 1, Pages (July 2006)
Adenosine Slows Migration of Dendritic Cells but Does Not Affect Other Aspects of Dendritic Cell Maturation  Susanne Hofer, Lennart Ivarsson, Patrizia.
Dendritic Cells Require T Cells for Functional Maturation In Vivo
Volume 24, Issue 2, Pages (February 2006)
Presentation transcript:

Volume 21, Issue 4, Pages 561-574 (October 2004) Dyslipidemia Associated with Atherosclerotic Disease Systemically Alters Dendritic Cell Mobilization  Véronique Angeli, Jaime Llodrá, James X. Rong, Kei Satoh, Satoshi Ishii, Takao Shimizu, Edward A. Fisher, Gwendalyn J. Randolph  Immunity  Volume 21, Issue 4, Pages 561-574 (October 2004) DOI: 10.1016/j.immuni.2004.09.003

Figure 1 Perturbations in LNs and Skin of Apoe−/− Mice Wt and Apoe−/− mice were studied at 6, 16, and 34 weeks of age. Some were placed on a Western diet at 6 week of age. (A) Relative number of cells in skin-draining LNs was determined in order to combine values from the four experiments conducted (n = 4). By 16 weeks and thereafter, the values between wt and atherosclerotic mice were significantly different; p < 0.05 independent of diet type. (B) Epidermal sheets from wt and Apoe−/− mice were stained with anti-IAb mAb. Low power (top two rows) and high power (last two rows) magnifications are shown. For analysis of MHC class II distribution, epidermal sheets were stained with anti-IAb mAb (red) and DAPI (blue). (C) The number of LC/mm2 was evaluated (upper panel). Values between wt and atherosclerotic mice were significantly different, p < 1 × 10−5. The number of LCs per LN was calculated by multiplying the percentage of langerin-positive cells by the number of total LN cells (bottom). Data are combined from three experiments (n = 3). Significant differences from wt controls are designated by *p < 0.05. (D) Ear skin sections were stained for histologic analysis (top rows) or with anti-CD68 mAb (bottom row). All scale bars, 40 μm. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)

Figure 2 Analysis of DC Migration to Draining LNs in Apoe−/− Mice (A) Wt and Apoe−/−mice received epicutaneous FITC, and the accumulation of FITC+ cells (x axis) in the LNs was assessed. DCs were identified by using PE-conjugated anti-CD11c mAb (y axis). (B) Percentage of FITC+CD11c+ DCs in LNs from wt and Apoe−/− mice was determined (means of four experiments). (C) To combine values from four independent experiments, the relative number of FITC+CD11c+ DCs per LN for each data point in each experiment was calculated. (D) Epidermal sheets from wt and Apoe−/− mice fed a chow or a Western diet were stained with anti-IAb mAb after one ear was painted with FITC. Percentage of migrated LCs was calculated by comparing densities between painted and unpainted ears. Data are from four experiments. (E) CCR7 mRNA expression in emigrated LCs from wt and Apoe−/− epidermis was assessed in the steady state and 18 hr after FITC painting. (F) Epidermal suspension were prepared 18 hr after FITC application and stained with CD45. Gated CD45+ cells were analyzed for FITC content. Significant differences from wt controls are designated by *p < 0.05. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)

Figure 3 Effect of Elevating HDL on DC Migration in Apoe−/− Mice (A) The accumulation of FITC+ DCs in LNs after FITC painting, (B) the baseline LC density, and (C) LN cellularity were evaluated in wt, Apoe−/−, and Apoe−/−/hApoA-I+/+ mice fed a Western diet for 10 weeks. (D) Wt and Apoe−/− mice fed a Western diet for 10 weeks received i.v. 500 μg of human albumin (control) or HDL isolated from normal plasma (HDL) for 7 days before FITC application. One typical experiment is shown for each part of the figure. Each experiment was repeated three times with similar results. Significant differences from untreated Apoe−/− controls are shown; *p < 0.001. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)

Figure 4 Role of HDL-Associated PAFAH in DC Migration (A) PAFAH activity was measured in HDL isolated from normal plasma (normal), in HDL derived from individuals who are homozygous deficient for PAFAH (PAFAH−/−), or in HDL from normal plasma treated with the Pefabloc (Pefabloc). (B) The effect of normal HDL (Normal), PAFAH-deficient HDL (PAFAH−/−), or (C) Pefabloc-inactivated PAFAH HDL (Pefabloc) treatment on DC migration was analyzed in Apoe−/− mice fed a Western diet after FITC painting. The results are expressed as the percentage of migration restored, where 100% reversal is defined as the difference between the percentage of FITC+ DC in untreated wt and Apoe−/− mice. (D) Wt or Apoe−/− mice kept on the Western diet received 0.1 mg/kg i.v. of placebo (Placebo) or recombinant PAFAH (PAFAH) for 7 consecutive days before FITC painting. (E) Wt and Apoe−/− mice kept on the Western diet for 10 weeks were treated with normal HDL (Normal), Pefabloc-inactivated PAFAH HDL (Pefabloc), or recombinant PAFAH (PAFAH). Baseline LC density was determined by immunostaining with anti-IAb mAb. (F) Total cells numbers in skin draining LNs were evaluated. Data are combined from three experiments. Significant differences are designated by *p < 0.05. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)

Figure 5 Effect of PAFAH-Associated HDL on Immunologic Parameters in Apoe−/− Mice Wt and Apoe−/− mice kept on the Western diet for 10 weeks were treated with HDL, Pefabloc-inactivated HDL, or recombinant PAFAH for 7 consecutive days before sensitization and one day after sensitization. (A) CHS and (B) DTH responses were assessed and expressed as ear swelling. Baseline unchallenged ear thickness was 40 ± 2 μm in wt mice and 50 ± 5 μm in Apoe−/− mice. (C) Expression of costimulatory molecules on CD11c+ DCs (steady state) or on CD11c+FITC+ DCs (18h FITC painting) was assessed by flow cytometry in LNs from wt (unfilled histograms) and Apoe−/− mice (filled histograms) at 16 weeks of age and fed a Western diet. Results shown are from one experiment typical of three conducted. Significant differences from wt controls are designated by *p < 0.01. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)

Figure 6 PAFR Expression and Function during DC Migration (A) Expression of mRNA for PAFR by wt and Pafr−/− LCs emigrated from skin explants and by wt and Pafr−/− BMDCs was assessed. (B) After FITC application on skin of wt and Pafr−/− mice, FITC+ CD11c+ cells in LNs were analyzed. Percent and total number of FITC+CD11c+ DCs in LNs from wt and Pafr−/− mice were determined. Data points are pooled from two experiments. (C) Migration of DCs after FITC painting was assessed in wt mice treated with the PAFR antagonist CV3988 or ethanol (control solvent) 1 day before and the day of FITC painting. (D) Adoptive transfer of BMDCs from WT or Pafr−/− CD45.2 mice (donor) was performed in the back skin of wt CD45.1 recipients, or conversely, BMDCs from wt CD45.1 mice (donor) were transferred to wt and Pafr−/− CD45.2 recipients. 2 days after cell transfer, the number of DC that migrated to brachial LNs was determined. All results are representative of at least two experiments (n ≥ 4). Significant differences are designated by *p < 0.05. (E) Expression of mRNA for PAFR in human LCs was analyzed (left). Calcium flux in human LCs was measured in response to cPAF. Arrow indicates point of cPAF addition (middle). Culture medium of human skin explants was supplemented with IL-1β, CV3988, its control solvent (ethanol), or with no additives. Emigration was quantified after 24 hr (right). Data points represent normalized emigration from individual 4 cm2 explants. Immunity 2004 21, 561-574DOI: (10.1016/j.immuni.2004.09.003)