The ability of the immune system to deliver the appropriate effector T cell to the anatomically correct place at the right time is essential for successful host defense. A widely held paradigm states that after initial exposure to antigen in lymph nodes draining a particular organ (e.g., skin), effector memory T cells are generated that express cell surface markers and chemokine receptors that facilitate their entry selectively into that organ (skin), but not into others (e.g., lung, gut). These markers include Cutaneous Lymphocyte antigen (CLA, a ligand for E and P selectin which we characterized extensively in the first cycle of funding), the chemokine receptor CCR4, and LFA-1. In parallel, a population of central memory T cells that express L selectin (ligand for PNAd expressed in lymph node postcapillary venules) and the chemokine receptor CCR& (ligand for Secondary Lymphoid Chemokine or SLC) are generated in the skin draining lymph node; these cell surface markers permit central memory cells to traffic like na?ve I cells and enter lymph node from blood. It was previously thought that the above CLA+ population of T cells lost either L selectin, CCR7, or both, and thus could not access the lymph node from blood like central memory T cells. We have demonstrated, however, that a large population of T cells in peripheral blood bears markers of both central memory T cells (L selectin, CCR7) and effector memory T cells (CLA, CCR4). We predict that these cells have the capacity to not only enter skin (particularly inflamed skin), but also lymph nodes throughout the body. In this proposal, we will analyze the phenotype and function of each of these populations of T cell, derived from blood and skin, with regards to their potential to extravasate at different anatomic sites. Moreover, we will examine T cells from skin and blood of patients with Cutaneous I Cell Lymphoma at different stages of disease, as we hypothesize that CTCL is a malignancy of the skin homing memory T cell. These experiments should help address questions about the plasticity of the organ specific homing response. Finally, we will use newly developed transgenic models and newly developed reagents to rigorously test the hypothesis that where a naive T cell encounters antigen determines its subsequent homing potential. This hypothesis is supported by substantial evidence; however, most of it is circumstantial. Taken together, these studies will allow us to further advance the paradigm of organ specific homing of effector and memory T cells. This should add to our fundamental understanding of both normal immunosurveillance as well as T cell mediated organ specific inflammatory disease.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
2R01AI041707-06
Application #
6545305
Study Section
General Medicine A Subcommittee 2 (GMA)
Program Officer
Plaut, Marshall
Project Start
1997-09-08
Project End
2007-02-28
Budget Start
2002-09-01
Budget End
2003-02-28
Support Year
6
Fiscal Year
2002
Total Cost
$190,796
Indirect Cost
Name
Brigham and Women's Hospital
Department
Type
DUNS #
071723621
City
Boston
State
MA
Country
United States
Zip Code
02115
Park, Chang Ook; Fu, Xiujun; Jiang, Xiaodong et al. (2018) Staged development of long-lived T-cell receptor ?? TH17 resident memory T-cell population to Candida albicans after skin infection. J Allergy Clin Immunol 142:647-662
Jiang, Xiaodong; Park, Chang Ook; Geddes Sweeney, Jenna et al. (2017) Dermal ?? T Cells Do Not Freely Re-Circulate Out of Skin and Produce IL-17 to Promote Neutrophil Infiltration during Primary Contact Hypersensitivity. PLoS One 12:e0169397
Pan, Youdong; Tian, Tian; Park, Chang Ook et al. (2017) Survival of tissue-resident memory T cells requires exogenous lipid uptake and metabolism. Nature 543:252-256
Collins, Nicholas; Jiang, Xiaodong; Zaid, Ali et al. (2016) Skin CD4(+) memory T cells exhibit combined cluster-mediated retention and equilibration with the circulation. Nat Commun 7:11514
Seneschal, Julien; Jiang, Xiaodong; Kupper, Thomas S (2014) Langerin+ dermal DC, but not Langerhans cells, are required for effective CD8-mediated immune responses after skin scarification with vaccinia virus. J Invest Dermatol 134:686-694
Hijnen, Dirkjan; Knol, Edward F; Gent, Yoony Y et al. (2013) CD8(+) T cells in the lesional skin of atopic dermatitis and psoriasis patients are an important source of IFN-?, IL-13, IL-17, and IL-22. J Invest Dermatol 133:973-9
Seneschal, Julien; Clark, Rachael A; Gehad, Ahmed et al. (2012) Human epidermal Langerhans cells maintain immune homeostasis in skin by activating skin resident regulatory T cells. Immunity 36:873-84
Jiang, Xiaodong; Clark, Rachael A; Liu, Luzheng et al. (2012) Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature 483:227-31
Kupper, Thomas S (2012) Old and new: recent innovations in vaccine biology and skin T cells. J Invest Dermatol 132:829-34
Purwar, Rahul; Schlapbach, Christoph; Xiao, Sheng et al. (2012) Robust tumor immunity to melanoma mediated by interleukin-9-producing T cells. Nat Med 18:1248-53

Showing the most recent 10 out of 27 publications