Volume 21, Issue 2, Pages (August 2004)

Slides:



Advertisements
Similar presentations
Shamim A. Mollah, Joseph S. Dobrin, Rachel E
Advertisements

Volume 28, Issue 2, Pages (February 2008)
Volume 19, Issue 1, Pages (July 2003)
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,
Skin-draining lymph nodes contain dermis-derived CD103− dendritic cells that constitutively produce retinoic acid and induce Foxp3+ regulatory T cells.
Volume 25, Issue 1, Pages (July 2006)
Mouse Lymphoid Tissue Contains Distinct Subsets of Langerin/CD207+ Dendritic Cells, Only One of Which Represents Epidermal-Derived Langerhans Cells  Patrice.
Following the Development of a CD4 T Cell Response In Vivo
Wei Hu, Ty Dale Troutman, Ramakrishna Edukulla, Chandrashekhar Pasare 
EpCAM Expressed by Murine Epidermal Langerhans Cells Modulates Immunization to an Epicutaneously Applied Protein Antigen  Takeshi Ouchi, Gaku Nakato,
Volume 31, Issue 2, Pages (August 2009)
Volume 31, Issue 5, Pages (November 2009)
Accelerated Migration of Respiratory Dendritic Cells to the Regional Lymph Nodes Is Limited to the Early Phase of Pulmonary Infection  Kevin L Legge,
by Dior Kingston, Michael A
Volume 34, Issue 1, Pages (January 2011)
Volume 16, Issue 2, Pages (February 2002)
Frederic Geissmann, Steffen Jung, Dan R. Littman  Immunity 
Volume 11, Issue 12, Pages (June 2015)
The Late Endosomal Adaptor Molecule p14 (LAMTOR2) Regulates TGFβ1-Mediated Homeostasis of Langerhans Cells  Florian Sparber, Christoph H. Tripp, Kerstin.
Volume 31, Issue 5, Pages (November 2009)
Mouse Lymphoid Tissue Contains Distinct Subsets of Langerin/CD207+ Dendritic Cells, Only One of Which Represents Epidermal-Derived Langerhans Cells  Patrice.
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 
Γδ T Cells Augment Rejection of Skin Grafts by Enhancing Cross-Priming of CD8 T Cells to Skin-Derived Antigen  Azad Rahimpour, Stephen R. Mattarollo,
Volume 18, Issue 5, Pages (May 2003)
Volume 13, Issue 3, Pages (September 2000)
Dynamic Interplay among Monocyte-Derived, Dermal, and Resident Lymph Node Dendritic Cells during the Generation of Vaccine Immunity to Fungi  Karen Ersland,
Sandra Holzmann, BSc, Christoph H
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 23, Issue 6, Pages (December 2005)
B Cells Acquire Particulate Antigen in a Macrophage-Rich Area at the Boundary between the Follicle and the Subcapsular Sinus of the Lymph Node  Yolanda.
NKT Cells Inhibit the Onset of Diabetes by Impairing the Development of Pathogenic T Cells Specific for Pancreatic β Cells  Lucie Beaudoin, Véronique.
Volume 21, Issue 4, Pages (October 2004)
Volume 25, Issue 3, Pages (September 2006)
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 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.
Volume 14, Issue 1, Pages (January 2001)
CD44-Deficient Mice Do Not Exhibit Impairment of Epidermal Langerhans Cell Migration to Lymph Nodes after Epicutaneous Sensitization with Protein  Shi-Chuen.
Volume 44, Issue 3, Pages (March 2016)
Volume 31, Issue 5, Pages (November 2009)
CD24a Expression Levels Discriminate Langerhans Cells from Dermal Dendritic Cells in Murine Skin and Lymph Nodes  Susanne Stutte, Bettina Jux, Charlotte.
Dynamics of Blood-Borne CD8 Memory T Cell Migration In Vivo
Volume 29, Issue 6, Pages (December 2008)
Elicited and steady-state transport of skin self-antigens in PYNOD-KO mice. Elicited and steady-state transport of skin self-antigens in PYNOD-KO mice.
Impaired Initiation of Contact Hypersensitivity by FTY720
Volume 11, Issue 2, Pages (August 1999)
Epidermal Langerhans Cells Rapidly Capture and Present Antigens from C-Type Lectin- Targeting Antibodies Deposited in the Dermis  Vincent Flacher, Christoph.
Volume 36, Issue 6, Pages (June 2012)
Oral Tolerance Can Be Established via Gap Junction Transfer of Fed Antigens from CX3CR1+ Macrophages to CD103+ Dendritic Cells  Elisa Mazzini, Lucia Massimiliano,
T Cell–Mediated Elimination of B7.2 Transgenic B Cells
Volume 33, Issue 3, Pages (September 2010)
Keratinocytes Function as Accessory Cells for Presentation of Endogenous Antigen Expressed in the Epidermis  Brian S. Kim, Fumi Miyagawa, Young-Hun Cho,
CD301b+ Dermal Dendritic Cells Drive T Helper 2 Cell-Mediated Immunity
Engagement of CD47 Inhibits the Contact Hypersensitivity Response Via the Suppression of Motility and B7 Expression by Langerhans Cells  Xijun Yu, Atsushi.
Volume 29, Issue 5, Pages (November 2008)
Sandra Holzmann, BSc, Christoph H
CD44 Regulates Survival and Memory Development in Th1 Cells
Volume 35, Issue 6, Pages (December 2011)
Volume 28, Issue 5, Pages (May 2008)
Volume 31, Issue 5, Pages (November 2009)
SOCS1 Prevents Potentially Skin-Reactive Cytotoxic T Lymphocytes from Gaining the Ability to Cause Inflammatory Lesions  Galaxia Maria Rodriguez, Dante.
Toll-Dependent Control Mechanisms of CD4 T Cell Activation
Volume 38, Issue 3, Pages (March 2013)
Cell-Autonomous Defects in Dendritic Cell Populations of Ikaros Mutant Mice Point to a Developmental Relationship with the Lymphoid Lineage  Li Wu, Aliki.
Patrizia Stoitzner, Christoph H
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 20, Issue 6, Pages (June 2004)
Presentation transcript:

Volume 21, Issue 2, Pages 279-288 (August 2004) CCR7 Governs Skin Dendritic Cell Migration under Inflammatory and Steady-State Conditions  Lars Ohl, Mariette Mohaupt, Niklas Czeloth, Gabriele Hintzen, Ziba Kiafard, Jörg Zwirner, Thomas Blankenstein, Golo Henning, Reinhold Förster  Immunity  Volume 21, Issue 2, Pages 279-288 (August 2004) DOI: 10.1016/j.immuni.2004.06.014

Figure 1 Effects of CCR7 Deficiency on Distribution and Mobilization of Langerhans Cells and Skin Dendritic Cells (A–F) To analyze LC distribution in the untreated skin, epidermal sheets were prepared from the ear of wild-type (A) or CCR7−/− (B) mice and stained with rat anti-Langerin mAb (clone 929F3) and mouse anti-rat-Cy3. In the explant model, skin flaps were isolated from wild-type (C and E) and CCR7−/− (D and F) ear and floated on tissue culture medium for 48 hr before epidermis (C and D) was separated from dermis (E and F) followed by staining with a rat anti-Langerin (C–F) or FITC-conjugated anti-MHCII mAb (E and F). (G) The number of CD86+MHCII+ cells emigrated into the tissue culture medium 24 hr and 48 hr after the onset of culture. (H) Ears of C57BL6 and CCR7−/− mice were treated with 0.1% FITC in acetone/dibutylphtalat or carrier alone. Mice were sacrificed 18 hr later and epidermal sheets were stained with rat anti-Langerin mAb (clone 929F3) and mouse anti-rat-Cy3, and the number of Langerin-positive cells was determined by epi-fluorescent microscopy. Data are derived from three images per epidermal sheet (10× original magnification, 0.8 mm2/image) of at least six ears of each genotype and treatment (**p < 0.01, unpaired t test). Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 2 CCR7-Deficient DC Fail to Migrate to Skin-Draining Lymph Nodes (A) In vitro differentiated, bone marrow-derived DC from wild-type and CCR7-deficient mice were labeled with CFSE and TAMRA, respectively, mixed at equal numbers, and injected i.c. into the right foot pad. After 36 hr, the popliteal LN was removed and analyzed for the presence of immigrated DC. (B) Same as (A) with the exception that wild-type DC were labeled with TAMRA and CCR7-deficient DC with CFSE and cells were injected into the left food pad of the same mice used in (A). (C) Mean ± SEM of the data derived from 8 popliteal LN of the experiments shown in (A) and (B). Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 3 Reduced Numbers of Distinct DC Subsets in CCR7-Deficient Mice The total number of different DC subtypes from wild-type (filled columns) and CCR7-deficient (open columns) mice was determined per LN by flow cytometry using the markers as indicated (pools of 10 skin-draining LN per animal; data from three C57BL6 and three CCR7-deficient C57BL6 mice). Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 4 Lack of Skin-Derived DC and Langerhans Cells in Skin-Draining LN of CCR7-Deficient Mice (A and B) LN cells derived from 10 skin-draining LN (inguinal, axillary, brachial, popliteal, and facial) of C57BL6 (A) and CCR7-deficient C57BL6 (B) mice were pooled and analyzed for the expression of CD11c and MHCII. (C) Number of cells falling within the upper right gate shown in (A) and (B) (data derived from three wild-type and three mutant mice of one experiment; similar data were derived from at least six additional mice of each genotype). (D) Langerin+ cells were found only in the CD11c+MHCIIhigh DC population (solid line, upper right gate shown in [A] and [B]) present in wild-type mice, whereas MHCIIinter DC (dotted line, middle right gate shown in [A] and [B]) do not express Langerin. (E) In CCR7-deficient mice, neither the residual MHCIIhigh nor the MHCIIinter DC population contained any Langerin+ cells. (F and G) Skin-draining lymph nodes of CCR7-deficient mice lack CD40high DC populations containing skin-derived DC. Cells from skin-draining LN of four wild-type mice (F) and four CCR7−/− mice were enriched for DC by density gradient centrifugation. DC were stained with anti-CD11c and anti-CD40. Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 5 Expression of CCR7 on LN Dendritic Cells and Freshly Isolated Epidermal Dendritic Cells Lymph node cells (upper panel) or cells isolated from ear epidermis (lower panel) of C57BL6 mice were stained with anti-MHCII, anti-CD11c, and rat anti-mCCR7 (solid line) or isotype control (shaded area) and mouse anti-rat-Cy5. Expression of CCR7 on CD11c+ cells expressing intermediate levels of MHCII (left column) and high levels of MHCII (right column) is shown. Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 6 Steady-State CD11c+MHCIIhigh DC in Wild-Type Mice Display a “Semimature” Phenotype Cells from skin-draining LN of naive wild-type mice (A) or of mice 24 hr after FITC skin painting (B) were analyzed for expression of MHCII and CD11c. The numbers shown represent the percentage of cells within the CD11c+MHCIIhigh population derived from six draining LN each. (B) The CD11c+MHCIIhigh subpopulation was further analyzed for expression of costimulatory molecules CD40 (C), CD80 (D), and CD86 (E) as well as for Langerin (F). Shaded area, isotype control; dotted line, gate on CD11c+MHCIIhigh cells of untreated mice; solid line, gate on CD11c+MHCIIhigh FITC+ cells 18 hr after FITC skin painting. Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)

Figure 7 CCR7-Deficient Mice Show Impaired T Cell Response in a Model of T Cell Triggering by s.c. Applied Antigen under Inflammatory and Steady-State Conditions (A–D) 24 hr after adoptive transfer of CFSE-labeled OT-II cells into C57BL6 and CCR7−/− mice, OVA (0.1–10 μg as indicated) was injected s.c. into recipient's left ear (A and C) while PBS was injected into the same recipient's right ear (B and D) together with 10 μg CpG1668 (A and B) or without adjuvant (C and D). Proliferation of the OT-II cells in the draining LN was analyzed 72 hr after antigen application. Data shown for each experimental setup are representative for at least six LN. For PBS injection, data are shown for right facial LN of mice that received 10 μg OVA into their left ear. (E) Instead of soluble OVA, ovalbumin chemically coupled to NLDC145 mAb (1 μg) was used for s.c. application in the same adoptive transfer model. (F) OT-II cells, adoptively transferred to C57BL6 wild-type mice, strongly upregulate the activation marker CD69 36 hr after s.c. application of 1 μg LPS-free OVA, while OT-II cells transferred to CCR7-deficient mice show weak upregulation on few TCR transgenic cells; shaded area, isotype control. (G) Intravenous application of 1 mg OVA induces comparable proliferation of adoptively transferred OT-II cells in peripheral LN of wild-type and CCR7-deficient mice. Immunity 2004 21, 279-288DOI: (10.1016/j.immuni.2004.06.014)