Location, location, location: the impact of migratory heterogeneity on T cell function

Bas J G Baaten, Andrea M Cooper, Susan L Swain, Linda M Bradley, Bas J G Baaten, Andrea M Cooper, Susan L Swain, Linda M Bradley

Abstract

T cell migration is crucial for an effective adaptive immune response to invading pathogens. Naive and memory T cells encounter pathogen antigens, become activated, and differentiate into effector cells in secondary lymphoid tissues, and then migrate to the site(s) of infection where they exert effector activities that control and eliminate pathogens. To achieve activation, efficient effector function, and good memory formation, T cells must traffic between lymphoid and non-lymphoid tissues within the body. This complex process is facilitated by chemokine receptors, selectins, CD44, and integrins that mediate the interactions of T cells with the environment. The expression patterns of these migration receptors (MR) dictate the tissues into which the effector T cells migrate and enable them to occupy specific niches within the tissue. While MR have been considered primarily to facilitate cell movement, we highlight how the heterogeneity of signaling through these receptors influences the function and fate of T cells in situ. We explore what drives MR expression heterogeneity, how this affects migration, and how this impacts T cell effector function and memory formation.

Keywords: T cell; cellular; heterogeneity; immunity; memory; migration; motility; subset.

Figures

Figure 1
Figure 1
Induction of migratory heterogeneity during priming. MR phenotype is impacted by TCR engagement, the level of co-stimulation, and the cytokine milieu (left). MR play a direct role in the formation of the immunological synapse, but TCR signaling subsequently impacts MR expression [panel 1, adapted from Ref. (8)]. Cytoskeletal rearrangements that involve the actin-binding ezrin (ezr), radixin, and moesin (mo) proteins are necessary for TCR signaling complex polarization. The integrin LFA-1 forms a ring surrounding the cSMAC that supports prolonged T cell-DC engagement, while other MR become excluded from the cSMAC. This process is possibly due to differential polarization of ezrin and moesin. Co-stimulatory signaling through molecules also contributes to the migratory heterogeneity of T cells (panel 2). For example, CD28 controls migration through upregulation of OX40, which is instrumental for CXCR5 expression and T cell localization to germinal centers (GC). In addition CD28/TCR signaling activates the PI3K/AKT pathway, which inhibits Foxo1 leading to decreased KLF2 expression. Differential co-stimulation can impact the levels of CD62L, CCR7, and S1P1 and thereby regulate the egress of T cells into the circulation. Cytokines released by DC promote specific transcriptional profiles that introduce further MR heterogeneity (panel 3). DC-derived IL-12 induces expression of the transcription factor T-bet and determines a CD4+ Th1 or CD8+ effector transcriptional program that results in part in the expression of CXCR3 and PSGL-1, which contribute to homing to peripheral sites. Alternatively, induction of the Tfh-associated transcription factor Bcl6 by IL-6 and IL-21 results in the downregulation of PSGL-1 and increased expression of CXCR5, which allows these cells to migrate from the T cell zones in the paracortex into GC.
Figure 2
Figure 2
Induction of tissue-specific MR profile. Expression of distinct MR combinations results in tissue-specific migration. T cells that are primed in gut-associated lymphoid tissues preferentially express functional PSGL-1, α4β7, and CCR9, which support migration through the post capillary venules of the small intestine to enter the lamina propria and intraepithelial compartment (panel 1). CD103-expressing DC impart the intestinal homing signature on T cells during priming by expression of retinoic acid (RA) in response to microbiota-driven toll-like receptor (TLR) signaling. A different program of MR usage is induced during priming of naïve T cells in skin-draining LN (panel 2). T cells in the skin express the cutaneous lymphocyte antigen (CLA), an inducible carbohydrate modification of PSGL-1, CCR4, CCR8, CCR10, α4β1, and LFA-1, which mitigates their migration into the skin. Analogous to intestinal imprinting, CCR10, but not CCR4, expression is regulated by skin-draining DCs that synthesize the vitamin D3 metabolite, 1,25(OH)2D3. 1,25(OH)2D3 suppresses α4β7 and CCR9 expression and RA inhibits CCR4 and CCR10 expression. In addition, CCR8 expression is imprinted by epidermal keratinocytes, although the skin-specific factors that induce CCR8 remain unknown. Evidence for imprinting of T cells in the lung is limited, but recent evidence suggests that lung DC-activated T cell migrate more efficiently into the lung, which was attributed to CCR4, although other MR are likely to contribute (panel 3). Many lymphocytes in the lung express high levels of α4β1, α1β1, and LFA-1 and CD8+ T cells primed in the mediastinal LN are enriched for CCR5 and CXCR3 expression, suggesting a pulmonary profile driven by distinct molecular mechanisms.

References

    1. Koning JJ, Mebius RE. Interdependence of stromal and immune cells for lymph node function. Trends Immunol (2012) 33(6):264–7010.1016/j.it.2011.10.006
    1. Forster R, Braun A, Worbs T. Lymph node homing of T cells and dendritic cells via afferent lymphatics. Trends Immunol (2012) 33(6):271–8010.1016/j.it.2012.02.007
    1. Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol (2013) 31:563–60410.1146/annurev-immunol-020711-074950
    1. Tate MD, Schilter HC, Brooks AG, Reading PC. Responses of mouse airway epithelial cells and alveolar macrophages to virulent and avirulent strains of influenza A virus. Viral Immunol (2011) 24(2):77–8810.1089/vim.2010.0118
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S. Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol (2007) 7(9):678–8910.1038/nri2156
    1. Castellino F, Huang AY, Altan-Bonnet G, Stoll S, Scheinecker C, Germain RN. Chemokines enhance immunity by guiding naive CD8+ T cells to sites of CD4+ T cell-dendritic cell interaction. Nature (2006) 440(7086):890–510.1038/nature04651
    1. Cyster JG. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu Rev Immunol (2005) 23:127–5910.1146/annurev.immunol.23.021704.115628
    1. Gordon-Alonso M, Veiga E, Sanchez-Madrid F. Actin dynamics at the immunological synapse. Cell Health Cytoskelet (2010) 2:33–47
    1. Ilani T, Khanna C, Zhou M, Veenstra TD, Bretscher A. Immune synapse formation requires ZAP-70 recruitment by ezrin and CD43 removal by moesin. J Cell Biol (2007) 179(4):733–4610.1083/jcb.200707199
    1. Shaffer MH, Dupree RS, Zhu P, Saotome I, Schmidt RF, McClatchey AI, et al. Ezrin and moesin function together to promote T cell activation. J Immunol (2009) 182(2):1021–32
    1. Montoya MC, Sancho D, Vicente-Manzanares M, Sanchez-Madrid F. Cell adhesion and polarity during immune interactions. Immunol Rev (2002) 186:68–8210.1034/j.1600-065X.2002.18607.x
    1. Gerard A, Khan O, Beemiller P, Oswald E, Hu J, Matloubian M, et al. Secondary T cell-T cell synaptic interactions drive the differentiation of protective CD8+ T cells. Nat Immunol (2013) 14(4):356–6310.1038/ni.2547
    1. Guy CS, Vignali KM, Temirov J, Bettini ML, Overacre AE, Smeltzer M, et al. Distinct TCR signaling pathways drive proliferation and cytokine production in T cells. Nat Immunol (2013) 14(3):262–7010.1038/ni.2538
    1. Morris GP, Allen PM. How the TCR balances sensitivity and specificity for the recognition of self and pathogens. Nat Immunol (2012) 13(2):121–810.1038/ni.2190
    1. Tubo NJ, Pagan AJ, Taylor JJ, Nelson RW, Linehan JL, Ertelt JM, et al. Single naive CD4(+) T cells from a diverse repertoire produce different effector cell types during infection. Cell (2013) 153(4):785–9610.1016/j.cell.2013.04.007
    1. Gerlach C, Rohr JC, Perie L, van Rooij N, van Heijst JW, Velds A, et al. Heterogeneous differentiation patterns of individual CD8+ T cells. Science (2013) 340(6132):635–910.1126/science.1235487
    1. Stemberger C, Huster KM, Koffler M, Anderl F, Schiemann M, Wagner H, et al. A single naive CD8+ T cell precursor can develop into diverse effector and memory subsets. Immunity (2007) 27(6):985–9710.1016/j.immuni.2007.10.012
    1. Schneider H, Valk E, da Rocha Dias S, Wei B, Rudd CE. CTLA-4 up-regulation of lymphocyte function-associated antigen 1 adhesion and clustering as an alternate basis for coreceptor function. Proc Natl Acad Sci U S A (2005) 102(36):12861–610.1073/pnas.0505802102
    1. Shimizu Y, van Seventer GA, Ennis E, Newman W, Horgan KJ, Shaw S. Crosslinking of the T cell-specific accessory molecules CD7 and CD28 modulates T cell adhesion. J Exp Med (1992) 175(2):577–8210.1084/jem.175.2.577
    1. Mirenda V, Jarmin SJ, David R, Dyson J, Scott D, Gu Y, et al. Physiologic and aberrant regulation of memory T-cell trafficking by the costimulatory molecule CD28. Blood (2007) 109(7):2968–77
    1. Walker LS, Gulbranson-Judge A, Flynn S, Brocker T, Raykundalia C, Goodall M, et al. Compromised OX40 function in CD28-deficient mice is linked with failure to develop CXC chemokine receptor 5-positive CD4 cells and germinal centers. J Exp Med (1999) 190(8):1115–2210.1084/jem.190.8.1115
    1. Delgoffe GM, Kole TP, Zheng Y, Zarek PE, Matthews KL, Xiao B, et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity (2009) 30(6):832–4410.1016/j.immuni.2009.04.014
    1. Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, et al. mTOR regulates memory CD8 T-cell differentiation. Nature (2009) 460(7251):108–1210.1038/nature08155
    1. Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol (2011) 12(4):295–30310.1038/ni.2005
    1. Ouyang W, Beckett O, Flavell RA, Li MO. An essential role of the Forkhead-box transcription factor Foxo1 in control of T cell homeostasis and tolerance. Immunity (2009) 30(3):358–7110.1016/j.immuni.2009.02.003
    1. Kerdiles YM, Beisner DR, Tinoco R, Dejean AS, Castrillon DH, DePinho RA, et al. Foxo1 links homing and survival of naive T cells by regulating L-selectin, CCR7 and interleukin 7 receptor. Nat Immunol (2009) 10(2):176–8410.1038/ni.1689
    1. Carlson CM, Endrizzi BT, Wu J, Ding X, Weinreich MA, Walsh ER, et al. Kruppel-like factor 2 regulates thymocyte and T-cell migration. Nature (2006) 442(7100):299–30210.1038/nature04882
    1. Moore TV, Clay BS, Cannon JL, Histed A, Shilling RA, Sperling AI. Inducible costimulator controls migration of T cells to the lungs via down-regulation of CCR7 and CD62L. Am J Respir Cell Mol Biol (2011) 45(4):843–5010.1165/rcmb.2010-0466OC
    1. Pham TH, Okada T, Matloubian M, Lo CG, Cyster JG. S1P1 receptor signaling overrides retention mediated by G alpha i-coupled receptors to promote T cell egress. Immunity (2008) 28(1):122–3310.1016/j.immuni.2007.11.017
    1. Mittelbrunn M, Molina A, Escribese MM, Yanez-Mo M, Escudero E, Ursa A, et al. VLA-4 integrin concentrates at the peripheral supramolecular activation complex of the immune synapse and drives T helper 1 responses. Proc Natl Acad Sci U S A (2004) 101(30):11058–6310.1073/pnas.0307927101
    1. Ariel A, Lider O, Brill A, Cahalon L, Savion N, Varon D, et al. Induction of interactions between CD44 and hyaluronic acid by a short exposure of human T cells to diverse pro-inflammatory mediators. Immunology (2000) 100(3):345–5110.1046/j.1365-2567.2000.00059.x
    1. Mummert ME, Mummert D, Edelbaum D, Hui F, Matsue H, Takashima A. Synthesis and surface expression of hyaluronan by dendritic cells and its potential role in antigen presentation. J Immunol (2002) 169(8):4322–31
    1. Jiang S, Dong C. A complex issue on CD4(+) T-cell subsets. Immunol Rev (2013) 252(1):5–1110.1111/imr.12041
    1. Groom JR, Richmond J, Murooka TT, Sorensen EW, Sung JH, Bankert K, et al. CXCR3 chemokine receptor-ligand interactions in the lymph node optimize CD4+ T helper 1 cell differentiation. Immunity (2012) 37(6):1091–10310.1016/j.immuni.2012.08.016
    1. Chang JT, Ciocca ML, Kinjyo I, Palanivel VR, McClurkin CE, Dejong CS, et al. Asymmetric proteasome segregation as a mechanism for unequal partitioning of the transcription factor T-bet during T lymphocyte division. Immunity (2011) 34(4):492–50410.1016/j.immuni.2011.03.017
    1. Matheu MP, Teijaro JR, Walsh KB, Greenberg ML, Marsolais D, Parker I, et al. Three phases of CD8 T cell response in the lung following H1N1 influenza infection and sphingosine 1 phosphate agonist therapy. PLoS One (2013) 8(3):e58033.10.1371/journal.pone.0058033
    1. Johnston RJ, Poholek AC, DiToro D, Yusuf I, Eto D, Barnett B, et al. Bcl6 and Blimp-1 are reciprocal and antagonistic regulators of T follicular helper cell differentiation. Science (2009) 325(5943):1006–1010.1126/science.1175870
    1. Poholek AC, Hansen K, Hernandez SG, Eto D, Chandele A, Weinstein JS, et al. In vivo regulation of Bcl6 and T follicular helper cell development. J Immunol (2010) 185(1):313–2610.4049/jimmunol.0904023
    1. Crotty S. Follicular helper CD4 T cells (TFH). Annu Rev Immunol (2011) 29:621–6310.1146/annurev-immunol-031210-101400
    1. Haddad W, Cooper CJ, Zhang Z, Brown JB, Zhu Y, Issekutz A, et al. P-selectin and P-selectin glycoprotein ligand 1 are major determinants for Th1 cell recruitment to nonlymphoid effector sites in the intestinal lamina propria. J Exp Med (2003) 198(3):369–7710.1084/jem.20020691
    1. Mora JR, Bono MR, Manjunath N, Weninger W, Cavanagh LL, Rosemblatt M, et al. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature (2003) 424(6944):88–9310.1038/nature01726
    1. Annacker O, Coombes JL, Malmstrom V, Uhlig HH, Bourne T, Johansson-Lindbom B, et al. Essential role for CD103 in the T cell-mediated regulation of experimental colitis. J Exp Med (2005) 202(8):1051–6110.1084/jem.20040662
    1. Johansson-Lindbom B, Svensson M, Pabst O, Palmqvist C, Marquez G, Forster R, et al. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J Exp Med (2005) 202(8):1063–7310.1084/jem.20051100
    1. Iwata M, Hirakiyama A, Eshima Y, Kagechika H, Kato C, Song SY. Retinoic acid imprints gut-homing specificity on T cells. Immunity (2004) 21(4):527–3810.1016/j.immuni.2004.08.011
    1. Wang S, Villablanca EJ, De Calisto J, Gomes DC, Nguyen DD, Mizoguchi E, et al. MyD88-dependent TLR1/2 signals educate dendritic cells with gut-specific imprinting properties. J Immunol (2011) 187(1):141–5010.4049/jimmunol.1003740
    1. Hammerschmidt SI, Ahrendt M, Bode U, Wahl B, Kremmer E, Forster R, et al. Stromal mesenteric lymph node cells are essential for the generation of gut-homing T cells in vivo. J Exp Med (2008) 205(11):2483–9010.1084/jem.20080039
    1. Igyarto BZ, Kaplan DH. Antigen presentation by Langerhans cells. Curr Opin Immunol (2013) 25(1):115–910.1016/j.coi.2012.11.007
    1. Fuhlbrigge RC, Kieffer JD, Armerding D, Kupper TS. Cutaneous lymphocyte antigen is a specialized form of PSGL-1 expressed on skin-homing T cells. Nature (1997) 389(6654):978–8110.1038/40166
    1. Zarbock A, Ley K, McEver RP, Hidalgo A. Leukocyte ligands for endothelial selectins: specialized glycoconjugates that mediate rolling and signaling under flow. Blood (2011) 118(26):6743–5110.1182/blood-2011-07-343566
    1. McCully ML, Moser B. The human cutaneous chemokine system. Front Immunol (2011) 2:33.10.3389/fimmu.2011.00033
    1. McCully ML, Ladell K, Hakobyan S, Mansel RE, Price DA, Moser B. Epidermis instructs skin homing receptor expression in human T cells. Blood (2012) 120(23):4591–810.1182/blood-2012-05-433037
    1. Sigmundsdottir H, Pan J, Debes GF, Alt C, Habtezion A, Soler D, et al. DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat Immunol (2007) 8(3):285–9310.1038/ni1433
    1. Grabbe S, Varga G, Beissert S, Steinert M, Pendl G, Seeliger S, et al. Beta2 integrins are required for skin homing of primed T cells but not for priming naive T cells. J Clin Invest (2002) 109(2):183–9210.1172/JCI11703
    1. Mackay LK, Stock AT, Ma JZ, Jones CM, Kent SJ, Mueller SN, et al. Long-lived epithelial immunity by tissue-resident memory T (TRM) cells in the absence of persisting local antigen presentation. Proc Natl Acad Sci U S A (2012) 109(18):7037–4210.1073/pnas.1202288109
    1. Mikhak Z, Strassner JP, Luster AD. Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4. J Exp Med (2013) 210(9):1855–6910.1084/jem.20130091
    1. Strutt TM, McKinstry KK, Kuang Y, Bradley LM, Swain SL. Memory CD4+ T-cell-mediated protection depends on secondary effectors that are distinct from and superior to primary effectors. Proc Natl Acad Sci U S A (2012) 109(38):E2551–6010.1073/pnas.1205894109
    1. Campbell JJ, Brightling CE, Symon FA, Qin S, Murphy KE, Hodge M, et al. Expression of chemokine receptors by lung T cells from normal and asthmatic subjects. J Immunol (2001) 166(4):2842–8
    1. Clark JG, Mandac-Dy JB, Dixon AE, Madtes DK, Burkhart KM, Harlan JM, et al. Trafficking of Th1 cells to lung: a role for selectins and a P-selectin glycoprotein-1-independent ligand. Am J Respir Cell Mol Biol (2004) 30(2):220–710.1165/rcmb.2003-0208OC
    1. Ray SJ, Franki SN, Pierce RH, Dimitrova S, Koteliansky V, Sprague AG, et al. The collagen binding alpha1beta1 integrin VLA-1 regulates CD8 T cell-mediated immune protection against heterologous influenza infection. Immunity (2004) 20(2):167–7910.1016/S1074-7613(04)00021-4
    1. Thatte J, Dabak V, Williams MB, Braciale TJ, Ley K. LFA-1 is required for retention of effector CD8 T cells in mouse lungs. Blood (2003) 101(12):4916–2210.1182/blood-2002-10-3159
    1. Wolber FM, Curtis JL, Maly P, Kelly RJ, Smith P, Yednock TA, et al. Endothelial selectins and alpha4 integrins regulate independent pathways of T lymphocyte recruitment in the pulmonary immune response. J Immunol (1998) 161(8):4396–403
    1. Ferguson AR, Engelhard VH. CD8 T cells activated in distinct lymphoid organs differentially express adhesion proteins and coexpress multiple chemokine receptors. J Immunol (2010) 184(8):4079–8610.4049/jimmunol.0901903
    1. Kohlmeier JE, Reiley WW, Perona-Wright G, Freeman ML, Yager EJ, Connor LM, et al. Inflammatory chemokine receptors regulate CD8(+) T cell contraction and memory generation following infection. J Exp Med (2011) 208(8):1621–3410.1084/jem.20102110
    1. Brown DM, Lee S, Garcia-Hernandez Mde L, Swain SL. Multifunctional CD4 cells expressing gamma interferon and perforin mediate protection against lethal influenza virus infection. J Virol (2012) 86(12):6792–80310.1128/JVI.07172-11
    1. Strutt TM, McKinstry KK, Marshall NB, Vong AM, Dutton RW, Swain SL. Multipronged CD4(+) T-cell effector and memory responses cooperate to provide potent immunity against respiratory virus. Immunol Rev (2013) 255(1):149–6410.1111/imr.12088
    1. McGill J, Van Rooijen N, Legge KL. Protective influenza-specific CD8 T cell responses require interactions with dendritic cells in the lungs. J Exp Med (2008) 205(7):1635–4610.1084/jem.20080314
    1. Cooper AM. Cell-mediated immune responses in tuberculosis. Annu Rev Immunol (2009) 27:393–42210.1146/annurev.immunol.021908.132703
    1. Khader SA, Guglani L, Rangel-Moreno J, Gopal R, Junecko BA, Fountain JJ, et al. IL-23 is required for long-term control of Mycobacterium tuberculosis and B cell follicle formation in the infected lung. J Immunol (2011) 187(10):5402–710.4049/jimmunol.1101377
    1. Khader SA, Rangel-Moreno J, Fountain JJ, Martino CA, Reiley WW, Pearl JE, et al. In a murine tuberculosis model, the absence of homeostatic chemokines delays granuloma formation and protective immunity. J Immunol (2009) 183(12):8004–1410.4049/jimmunol.0901937
    1. Pearl JE, Torrado E, Tighe M, Fountain JJ, Solache A, Strutt T, et al. Nitric oxide inhibits the accumulation of CD4+CD44hiTbet+CD69lo T cells in mycobacterial infection. Eur J Immunol (2012) 42(12):3267–7910.1002/eji.201142158
    1. Calzascia T, Masson F, Di Berardino-Besson W, Contassot E, Wilmotte R, Aurrand-Lions M, et al. Homing phenotypes of tumor-specific CD8 T cells are predetermined at the tumor site by crosspresenting APCs. Immunity (2005) 22(2):175–8410.1016/j.immuni.2004.12.008
    1. Oyoshi MK, Elkhal A, Scott JE, Wurbel MA, Hornick JL, Campbell JJ, et al. Epicutaneous challenge of orally immunized mice redirects antigen-specific gut-homing T cells to the skin. J Clin Invest (2011) 121(6):2210–2010.1172/JCI43586
    1. Villablanca EJ, Russo V, Mora JR. Dendritic cell migration and lymphocyte homing imprinting. Histol Histopathol (2008) 23(7):897–910
    1. Baaten BJ, Tinoco R, Chen AT, Bradley LM. Regulation of antigen-experienced T cells: lessons from the quintessential memory marker CD44. Front Immunol (2012) 3:23.10.3389/fimmu.2012.00023
    1. Leppert D, Waubant E, Galardy R, Bunnett NW, Hauser SL. T cell gelatinases mediate basement membrane transmigration in vitro. J Immunol (1995) 154(9):4379–89
    1. Abraham M, Shapiro S, Karni A, Weiner HL, Miller A. Gelatinases (MMP-2 and MMP-9) are preferentially expressed by Th1 vs. Th2 cells. J Neuroimmunol (2005) 163(1–2):157–6410.1016/j.jneuroim.2005.02.001
    1. Oviedo-Orta E, Bermudez-Fajardo A, Karanam S, Benbow U, Newby AC. Comparison of MMP-2 and MMP-9 secretion from T helper 0, 1 and 2 lymphocytes alone and in coculture with macrophages. Immunology (2008) 124(1):42–5010.1111/j.1365-2567.2007.02728.x
    1. Aoudjit F, Potworowski EF, St-Pierre Y. Bi-directional induction of matrix metalloproteinase-9 and tissue inhibitor of matrix metalloproteinase-1 during T lymphoma/endothelial cell contact: implication of ICAM-1. J Immunol (1998) 160(6):2967–73
    1. Wu B, Crampton SP, Hughes CC. Wnt signaling induces matrix metalloproteinase expression and regulates T cell transmigration. Immunity (2007) 26(2):227–3910.1016/j.immuni.2006.12.007
    1. Yakubenko VP, Lobb RR, Plow EF, Ugarova TP. Differential induction of gelatinase B (MMP-9) and gelatinase A (MMP-2) in T lymphocytes upon alpha(4)beta(1)-mediated adhesion to VCAM-1 and the CS-1 peptide of fibronectin. Exp Cell Res (2000) 260(1):73–8410.1006/excr.2000.5002
    1. Moore C, Shen XD, Gao F, Busuttil RW, Coito AJ. Fibronectin-alpha4beta1 integrin interactions regulate metalloproteinase-9 expression in steatotic liver ischemia and reperfusion injury. Am J Pathol (2007) 170(2):567–7710.2353/ajpath.2007.060456
    1. Esparza J, Vilardell C, Calvo J, Juan M, Vives J, Urbano-Marquez A, et al. Fibronectin upregulates gelatinase B (MMP-9) and induces coordinated expression of gelatinase A (MMP-2) and its activator MT1-MMP (MMP-14) by human T lymphocyte cell lines. A process repressed through RAS/MAP kinase signaling pathways. Blood (1999) 94(8):2754–66
    1. Kambara C, Nakamura T, Furuya T, Migita K, Ida H, Kawakami A, et al. Vascular cell adhesion molecule-1-mediated matrix metalloproteinase-2 induction in peripheral blood T cells is up-regulated in patients with HTLV-I-associated myelopathy. J Neuroimmunol (1999) 99(2):242–710.1016/S0165-5728(99)00110-1
    1. Romanic AM, Madri JA. The induction of 72-kD gelatinase in T cells upon adhesion to endothelial cells is VCAM-1 dependent. J Cell Biol (1994) 125(5):1165–7810.1083/jcb.125.5.1165
    1. Xia M, Leppert D, Hauser SL, Sreedharan SP, Nelson PJ, Krensky AM, et al. Stimulus specificity of matrix metalloproteinase dependence of human T cell migration through a model basement membrane. J Immunol (1996) 156(1):160–7
    1. Xia M, Sreedharan SP, Dazin P, Damsky CH, Goetzl EJ. Integrin-dependent role of human T cell matrix metalloproteinase activity in chemotaxis through a model basement membrane. J Cell Biochem (1996) 61(3):452–810.1002/(SICI)1097-4644(19960601)61:3<452::AID-JCB12>;2-L
    1. Stefanidakis M, Koivunen E. Cell-surface association between matrix metalloproteinases and integrins: role of the complexes in leukocyte migration and cancer progression. Blood (2006) 108(5):1441–5010.1182/blood-2006-02-005363
    1. Brooks PC, Stromblad S, Sanders LC, von Schalscha TL, Aimes RT, Stetler-Stevenson WG, et al. Localization of matrix metalloproteinase MMP-2 to the surface of invasive cells by interaction with integrin alpha v beta 3. Cell (1996) 85(5):683–9310.1016/S0092-8674(00)81235-0
    1. Rowe RG, Weiss SJ. Breaching the basement membrane: who, when and how? Trends Cell Biol (2008) 18(11):560–7410.1016/j.tcb.2008.08.007
    1. Parks WC, Wilson CL, Lopez-Boado YS. Matrix metalloproteinases as modulators of inflammation and innate immunity. Nat Rev Immunol (2004) 4(8):617–2910.1038/nri1418
    1. Sanchez-Madrid F, Serrador JM. Bringing up the rear: defining the roles of the uropod. Nat Rev Mol Cell Biol (2009) 10(5):353–910.1038/nrm2680
    1. del Pozo MA, Sanchez-Mateos P, Nieto M, Sanchez-Madrid F. Chemokines regulate cellular polarization and adhesion receptor redistribution during lymphocyte interaction with endothelium and extracellular matrix. Involvement of cAMP signaling pathway. J Cell Biol (1995) 131(2):495–50810.1083/jcb.131.2.495
    1. de Curtis I, Meldolesi J. Cell surface dynamics – how Rho GTPases orchestrate the interplay between the plasma membrane and the cortical cytoskeleton. J Cell Sci (2012) 125(Pt 19):4435–4410.1242/jcs.108266
    1. Nieto M, Frade JM, Sancho D, Mellado M, Martinez AC, Sanchez-Madrid F. Polarization of chemokine receptors to the leading edge during lymphocyte chemotaxis. J Exp Med (1997) 186(1):153–810.1084/jem.186.1.153
    1. Urzainqui A, Serrador JM, Viedma F, Yanez-Mo M, Rodriguez A, Corbi AL, et al. ITAM-based interaction of ERM proteins with Syk mediates signaling by the leukocyte adhesion receptor PSGL-1. Immunity (2002) 17(4):401–1210.1016/S1074-7613(02)00420-X
    1. Rougerie P, Delon J. Rho GTPases: masters of T lymphocyte migration and activation. Immunol Lett (2012) 142(1–2):1–1310.1016/j.imlet.2011.12.003
    1. Martinelli S, Chen EJ, Clarke F, Lyck R, Affentranger S, Burkhardt JK, et al. Ezrin/Radixin/Moesin proteins and flotillins cooperate to promote uropod formation in T cells. Front Immunol (2013) 4:84.10.3389/fimmu.2013.00084
    1. Mrass P, Kinjyo I, Ng LG, Reiner SL, Pure E, Weninger W. CD44 mediates successful interstitial navigation by killer T cells and enables efficient antitumor immunity. Immunity (2008) 29(6):971–8510.1016/j.immuni.2008.10.015
    1. Overstreet MG, Gaylo A, Angermann BR, Hughson A, Hyun YM, Lambert K, et al. Inflammation-induced interstitial migration of effector CD4(+) T cells is dependent on integrin alphaV. Nat Immunol (2013) 14(9):949–5810.1038/ni.2682
    1. Hufford MM, Kim TS, Sun J, Braciale TJ. Antiviral CD8+ T cell effector activities in situ are regulated by target cell type. J Exp Med (2011) 208(1):167–8010.1084/jem.20101850
    1. Catron DM, Rusch LK, Hataye J, Itano AA, Jenkins MK. CD4+ T cells that enter the draining lymph nodes after antigen injection participate in the primary response and become central-memory cells. J Exp Med (2006) 203(4):1045–5410.1084/jem.20051954
    1. D’Souza WN, Hedrick SM. Cutting edge: latecomer CD8 T cells are imprinted with a unique differentiation program. J Immunol (2006) 177(2):777–81
    1. Darrah PA, Patel DT, De Luca PM, Lindsay RW, Davey DF, Flynn BJ, et al. Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nat Med (2007) 13(7):843–5010.1038/nm1592
    1. Dooms H, Wolslegel K, Lin P, Abbas AK. Interleukin-2 enhances CD4+ T cell memory by promoting the generation of IL-7R alpha-expressing cells. J Exp Med (2007) 204(3):547–5710.1084/jem.20062381
    1. Debes GF, Arnold CN, Young AJ, Krautwald S, Lipp M, Hay JB, et al. Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat Immunol (2005) 6(9):889–9410.1038/ni1238
    1. Jennrich S, Lee MH, Lynn RC, Dewberry K, Debes GF. Tissue exit: a novel control point in the accumulation of antigen-specific CD8 T cells in the influenza a virus-infected lung. J Virol (2012) 86(7):3436–4510.1128/JVI.07025-11
    1. Debes GF, Bonhagen K, Wolff T, Kretschmer U, Krautwald S, Kamradt T, et al. CC chemokine receptor 7 expression by effector/memory CD4+ T cells depends on antigen specificity and tissue localization during influenza A virus infection. J Virol (2004) 78(14):7528–3510.1128/JVI.78.14.7528-7535.2004
    1. Bromley SK, Thomas SY, Luster AD. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat Immunol (2005) 6(9):895–90110.1038/ni1240
    1. Ledgerwood LG, Lal G, Zhang N, Garin A, Esses SJ, Ginhoux F, et al. The sphingosine 1-phosphate receptor 1 causes tissue retention by inhibiting the entry of peripheral tissue T lymphocytes into afferent lymphatics. Nat Immunol (2008) 9(1):42–5310.1038/ni1534
    1. Carbone FR, Mackay LK, Heath WR, Gebhardt T. Distinct resident and recirculating memory T cell subsets in non-lymphoid tissues. Curr Opin Immunol (2013) 25(3):329–3310.1016/j.coi.2013.05.007
    1. Gebhardt T, Whitney PG, Zaid A, Mackay LK, Brooks AG, Heath WR, et al. Different patterns of peripheral migration by memory CD4+ and CD8+ T cells. Nature (2011) 477(7363):216–910.1038/nature10339
    1. Richter M, Ray SJ, Chapman TJ, Austin SJ, Rebhahn J, Mosmann TR, et al. Collagen distribution and expression of collagen-binding alpha1beta1 (VLA-1) and alpha2beta1 (VLA-2) integrins on CD4 and CD8 T cells during influenza infection. J Immunol (2007) 178(7):4506–16
    1. Chapman TJ, Topham DJ. Identification of a unique population of tissue-memory CD4+ T cells in the airways after influenza infection that is dependent on the integrin VLA-1. J Immunol (2010) 184(7):3841–910.4049/jimmunol.0902281
    1. Richter MV, Topham DJ. The {alpha}1beta1 integrin and TNF receptor II protect airway CD8+ effector T cells from apoptosis during influenza infection. J Immunol (2007) 179(8):5054–63
    1. Baaten BJ, Li CR, Deiro MF, Lin MM, Linton PJ, Bradley LM. CD44 regulates survival and memory development in Th1 cells. Immunity (2010) 32(1):104–1510.1016/j.immuni.2009.10.011
    1. Maeshima N, Poon GF, Dosanjh M, Felberg J, Lee SS, Cross JL, et al. Hyaluronan binding identifies the most proliferative activated and memory T cells. Eur J Immunol (2011) 41(4):1108–1910.1002/eji.201040870
    1. Gretz JE, Norbury CC, Anderson AO, Proudfoot AE, Shaw S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J Exp Med (2000) 192(10):1425–4010.1084/jem.192.10.1425
    1. Kaech SM, Wherry EJ. Heterogeneity and cell-fate decisions in effector and memory CD8+ T cell differentiation during viral infection. Immunity (2007) 27(3):393–40510.1016/j.immuni.2007.08.007
    1. Obar JJ, Jellison ER, Sheridan BS, Blair DA, Pham QM, Zickovich JM, et al. Pathogen-induced inflammatory environment controls effector and memory CD8+ T cell differentiation. J Immunol (2011) 187(10):4967–7810.4049/jimmunol.1102335
    1. Jung YW, Rutishauser RL, Joshi NS, Haberman AM, Kaech SM. Differential localization of effector and memory CD8 T cell subsets in lymphoid organs during acute viral infection. J Immunol (2010) 185(9):5315–2510.4049/jimmunol.1001948
    1. Zinselmeyer BH, Heydari S, Sacristán C, Nayak D, Cammer M, Herz J, et al. PD-1 promotes immune exhaustion by inducing antiviral T cell motility paralysis. J Exp Med (2013) 210(4):757–7410.1084/jem.20121416
    1. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity (2007) 27(1):111–2210.1016/j.immuni.2007.05.016

Source: PubMed

3
Předplatit