Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-2pzkn Total loading time: 0 Render date: 2024-05-16T02:30:40.331Z Has data issue: false hasContentIssue false

10 - Heat Shock Proteins, Their Cell Surface Receptors and Effect on the Immune System

Published online by Cambridge University Press:  10 August 2009

Thomas Lehner
Affiliation:
Peter Gorer Department of Immunobiology, Guy's, King's and St. Thomas Hospital Medical School London, United Kingdom
Yufei Wang
Affiliation:
Peter Gorer Department of Immunobiology, Guy's, King's and St. Thomas' Hospital Medical School, London, United Kingdom
Trevor Whittall
Affiliation:
Peter Gorer Department of Immunobiology, Guy's, King's and St. Thomas' Hospital Medical School, London, United Kingdom
Lesley A. Bergmeier
Affiliation:
Peter Gorer Department of Immunobiology, Guy's, King's and St. Thomas Hospital Medical School, London, United Kingdom
Brian Henderson
Affiliation:
University College London
A. Graham Pockley
Affiliation:
University of Sheffield
Get access

Summary

Introduction

Heat shock or stress proteins are important intracellular protein chaperones that control their trafficking. The function of heat shock proteins in the immunopathology of infections, tumours and autoimmune diseases has been the subject of numerous experimental and clinical investigations over the past few decades and some of their properties are summarised in Table 10.1. Because there is extensive homology between mammalian and microbial heat shock proteins, immunological cross-reactions were considered to account for a number of autoimmune diseases. However, although the biological significance of lipopolysaccharide (LPS) found in Gram-negative bacteria has been well appreciated, heat shock proteins, which are found more widely in Gram-negative and -positive bacteria, especially those in the gut, have received more limited attention. A relatively new phase in this area of biology was initiated by the discovery of specific heat shock protein receptors and by rapid advances in our understanding of the signalling pathways. This chapter will deal with the receptors used by heat shock proteins, with particular reference to Hsp70, involvement of these proteins in the stimulation of chemokine production, maturation of dendritic cells (DCs), their intrinsic adjuvanticity and capacity to enhance immunogenicity.

Structural features of Hsp70

Although the overall three-dimensional structure of Hsp70 is not known, the structures of the two domains from various members of the family have been solved separately. The crystal structures of the ATPase domains of bovine Hsc70 (heat shock constitutive protein) and human Hsp70 have been determined [1, 2].

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2005

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Flaherty, K M, Deluca-Flaherty, C and McKay, D B.Three-dimensional structure of the ATPse fragment of a 70K heat-shock cognate protein. Nature 1990, 346: 623–628Google Scholar
Osipiuk, J, Walsh, M A, Freeman, B C, Morimoto, R I and Joachimiak, A. Structure of a new crystal form of human hsp70 ATPse domain. D. Biol. Crystallogr. 1999, D55: 1105–1107Google Scholar
Zhu, X, Zhao, X, Burkholder, W F, Gragerov, A, Ogata, C M, Gottesman, M E and Hendrickson, W A. Structural analysis of substrate binding by the molecular chaperone DnaK. Science 1996, 272: 1606–1614Google Scholar
Kol, A, Lichtman, A H, Finberg, R W, Libby, P and Kurt-Jones, E A. Heat shock protein (HSP) 60 activates the innate immune response: CD14 is an essential receptor for HSP60 activation of mononuclear cells. J Immunol 2000, 164: 13–17Google Scholar
Asea, A, Kraeft, S-K, Kurt-Jones, E A, Stevenson, M A, Chen, L B, Finberg, R W, Koo, G C and Calderwood, S K. Hsp70 stimulates cytokine production through a CD14-dependent pathway, demonstrating its dual role as a chaperone and cytokine. Nat Med 2000, 6: 435–442Google Scholar
Wang, Y, Kelly, C G, Karttunen, T, Whittall, T, Lehner, P J, Duncan, L, MacAry, P, Younson, J S, Singh, M, Oehlmann, W, Cheng, G, Bergmeier, L and Lehner, T. CD40 is a cellular receptor mediating mycobacterial heat shock protein 70 stimulation of CC-chemokines. Immunity 2001, 15: 971–983Google Scholar
Wang, Y, Kelly, C G, Singh, M, McGowan, E G, Carrara, A S, Bergmeier, L A and Lehner, T. Stimulation of Th1-polarizing cytokines, C-C chemokines, maturation of dendritic cells, and adjuvant function by the peptide binding fragment of heat shock protein 70. J Immunol 2002, 169: 2422–2429Google Scholar
Wang, Y, Whittal, T, McGowan, E, Younson, J, Kelly, C, Bergmeier, L A, Singh, M, Lehner, T.Identification of stimulating and inhibitory epitopes within the Hsp70 molecule which modulate cytokine production and maturation of dendritic cells. J Immunology 2005, 174: 3306–3316Google Scholar
Becker, T, Hartl, F U and Wieland, F. CD40, an extracellular receptor for binding and uptake of Hsp70-peptide complexes. J Cell Biol 2002, 158: 1277–1285Google Scholar
Millar, D G, Garza, K M, Odermatt, B, Elford, A R, Ono, N, Li, Z and Ohashi, P S. Hsp70 promotes antigen-presenting cell function and converts T-cell tolerance to autoimmunity in vivo. Nat Med 2003, 9: 1469–1476Google Scholar
Lazarevic, V, Myers, A J, Scanga, C A and Flynn, J L. CD40, but not CD40L, is required for the optimal priming of T cells and control of aerosol M. tuberculosis infection. Immunity 2003, 19: 823–835Google Scholar
Basu, S, Binder, R J, Suto, R, Anderson, K M and Srivastava, P K. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activates the NF-κB pathway. Int Immunol 2000, 12: 1539–1546Google Scholar
Binder, R J, Han, D K and Srivastava, P K. CD91: a receptor for heat shock protein gp96. Nat Immunol 2000, 1: 151–155Google Scholar
Ohashi, K, Burkart, V, Flohé, S and Kolb, H. Heat shock protein 60 is a putative endogenous ligand of the Toll-like receptor-4 complex. J Immunol 2000, 164: 558–561Google Scholar
Vabulas, R M, Ahmad-Nejad, P, da Costa, C, Miethke, T, Kirschning, C J, Hacker, H and Wagner, H. Endocytosed HSP60s use toll-like receptor 2 (TLR2) and TLR4 to activate the toll/interleukin-1 receptor signaling pathway in innate immune cells. J Biol Chem 2001, 276: 31332–31339Google Scholar
Vabulas, R M, Ahmad-Nejad, P, Ghose, S, Kirschning, C J, Issels, R D and Wagner, H. HSP70 as endogenous stimulus of the Toll/interleukin-1 receptor signal pathway. J Biol Chem 2002, 277: 15107–15112Google Scholar
Vabulas, R M, Braedel, S, Hilf, N, Singh-Jasuja, H, Herter, S, Ahmad-Nejad, P, Kirschning, C J, Da Costa, C, Rammensee, H G, Wagner, H and Schild, H. The endoplasmic reticulum-resident heat shock protein Gp96 activates dendritic cells via the Toll-like receptor 2/4 pathway. J Biol Chem 2002, 277: 20847–20853Google Scholar
Draude, G, Hrboticky, N and Lorenz, R L. The expression of the lectin-like oxidized low-density lipoprotein receptor (LOX-1) on human vascular smooth muscle cells and monocytes and its down-regulation by lovastatin. Biochem Pharmacol 1999, 57: 383–386Google Scholar
Delneste, Y, Magistrelli, G, Gauchat, J, Haeuw, J, Aubry, J, Nakamura, K, Kawakami-Honda, N, Goetsch, L, Sawamura, T, Bonnefoy, J and Jeannin, P. Involvement of LOX-1 in dendritic cell-mediated antigen cross-presentation. Immunity 2002, 17: 353–362Google Scholar
Lehner, T, Bergmeier, L A, Wang, Y, Tao, L, Singh, M, Spallek, R and Zee, R. Heat shock proteins generate β-chemokines which function as innate adjuvants enhancing adaptive immunity. Eur J Immunol 2000, 30: 594–603Google Scholar
Retzlaff, C, Yamamoto, Y, Hoffman, P S, Friedman, H and Klein, T W. Bacterial heat shock proteins directly induce cytokine mRNA and interleukin-1 secretion in macrophage cultures. Infect Immun 1994, 62: 5689–5693Google Scholar
Castellino, F, Boucher, P E, Eichelberg, K, Mayhew, M, Rothman, J E, Houghton, A N and Germain, R N. Receptor-mediated uptake of antigen/heat shock protein complexes results in major histocompatibility complex class I antigen presentation via two distinct pathways. J Exp Med 2000, 191: 1957–1964Google Scholar
Fujihara, S M and Nadler, S G. Intranuclear targeted delivery of functional NF-κB by 70kDa heat shock protein. EMBO J 1999, 18: 411–419Google Scholar
Wright, S D, Ramos, R A, Tobias, P S, Ulevitch, R J and Mathison, J C. CD14, a receptor for complexes of lipopolysaccharide (LPS) and LPS binding protein. Science 1990, 249: 1431–1433Google Scholar
Yang, R B, Mark, M R, Gurney, A L and Godwski, P J. Signalling events induced by lipopolysaccharide-activated toll-like receptor 2. J Immunol 1999, 163: 639–643Google Scholar
da Silva Correia, J, Soldau, K, Christen, U, Tobias, P S and Ulevitch, R J. Lipopolysaccharide is in close proximity to each of the proteins in its membrane receptor complex. Transfer from CD14 to TLR4 and MD-2. J Biol Chem 2001, 276: 21129–21135Google Scholar
Triantafilou, K, Triantafilou, M and Dedrick, R L. A CD14-independent LPS receptor cluster. Nat Immunol 2001, 2: 338–344Google Scholar
Asea, A, Rehli, M, Kabingu, E, Boch, J A, Baré, O, Auron, P E, Stevenson, M A and Calderwood, S K. Novel signal transduction pathway utilized by extracellular HSP70. Role of Toll-like receptor (TLR) 2 and TLR4. J Biol Chem 2002, 277: 15028–15034Google Scholar
Lipsker, D, Ziylan, U, Spehner, D, Proamer, F, Bausinger, H, Jeannin, P, Salamero, J, Bohbot, A, Cazenave, J P, Drillien, R, Delneste, Y, Hanau, D and Salle, H. Heat shock proteins 70 and 60 share common receptors which are expressed on human monocyte-derived but not epidermal dendritic cells. Eur J Immunol 2002, 32: 322–332Google Scholar
Kooten, C and Banchereau, J. Functions of CD40 on B cells, dendritic cells and other cells. Cur Opin Immunol 1997, 9: 330–337Google Scholar
Young, L S, Eliopoulos, A G, Gallagher, N J and Dawson, C W. CD40 and epithelial cells: across the great divide. Immunol Today 1998, 19: 502–506Google Scholar
Bourgeois, C, Rocha, B and Tanchot, C. A role for CD40 expression on CD8+ T cells in the generation of CD8+ T cell memory. Science 2002, 297: 2060–2063Google Scholar
Bennett, S R, Carbone, F R, Karamalis, F, Flavell, R A, Miller, J F and Heath, W R. Help for cytotoxic-T cell responses is mediated by CD40 signalling. Nature 1998, 393: 478–480Google Scholar
Schoenberger, S P, Toes, R E, Voort, E I, Offringa, R and Melief, C J. T cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature 1998, 393: 480–483Google Scholar
Herold, K C, Lu, J, Rulifson, I, Vezys, V, Taub, D, Grusby, M J and Bluestone, J A. Regulation of C-C chemokine production by murine T cells by CD28/B7 costimulation. J Immunol 1997, 159: 4150–4153Google Scholar
Kornbluth, R S, Kee, K and Richman, D D. CD40 ligand (CD154) stimulation of macrophages to produce HIV-1 suppressive chemokines. Proc Natl Acad Sci USA 1998, 99: 5205–5210Google Scholar
McDyer, J F, Dybul, M, Goletz, T J, Kinter, A L, Thomas, E K, Berzofsky, J A, Fauci, A S and Seder, R A. Differential effects of CD40 ligand/trimer stimulation on the ability of dendritic cells to replicate and transmit HIV infection: evidence for CC-chemokine-dependent and -independent mechanisms. J Immunol 1999, 162: 3711–3717Google Scholar
Yasui, T, Muraoka, M, Takaoka-Shichijo, Y, Ishida, I, Takegahara, N, Uchida, J, Kumanogoh, A, Suematsu, S, Suzuki, M and Kikutani, H. Dissection of B cell differentiation during primary immune responses in mice with altered CD40 signals. Int Immunol 2002, 14: 319–329Google Scholar
Janeway, C A J and Medzhitov, R. Innate immune recognition. Ann Rev Immunol 2002, 20: 197–216Google Scholar
Medzhitov, R. Toll-like receptors and innate immunity. Nat Rev Immunol 2001, 1: 135–145Google Scholar
Uronen-Hansson, H, Allen, J, Osman, M, SquiresG, Klein N G, Klein N and Callard, R E. Toll-like receptor 2 (TLR2) and TLR4 are present inside human dendritic cells, associated with microtubules and the Golgi apparatus but are not detectable on the cell surface: integrity of microtubules is required for interleukin-12 production in response to internalized bacteria. Immunology 2004, 111: 173–178Google Scholar
Armstrong, P B and Quigley, J P. α2 Macroglobulin: an evolutionary conserved arm of the innate immune system. Dev Comp Immunol 1999, 23: 375–390Google Scholar
Basu, S, Binder, R J, Ramalingam, T and Srivastava, P K. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70 and calreticulin. Immunity 2001, 14: 303–313Google Scholar
Srivastava, P. Roles of heat-shock proteins in innate and adaptive immunity. Nat Rev Immunol 2002, 2: 185–194Google Scholar
Krieger, M. The other side of scavenger receptors: pattern recognition for host defense. Cur Opin Lipidol 1997, 8: 275–280Google Scholar
Gough, P J and Gordon, S. The role of scavenger receptors in the innate immune system. Microbes Infect 2000, 2: 305–311Google Scholar
Panjwani, N N, Popova, L and Srivastava, P K. Heat shock proteins gp96 and hsp70 activate the release of nitric oxide by APCs. J Immunol 2002, 168: 2997–3003Google Scholar
MacAry, P A, Javid, B, Floto, R A, Smith, K G C, Singh, M and Lehner, P J. HSP70 peptide binding mutants separate antigen delivery from dendritic cell stimulation. Immunity 2004, 20: 95–106Google Scholar
Coutinho, A and Meo, T. Genetic basis for unresponsiveness to lipopolysaccharide in C57BL/10Cr mice. Immunogenetics 1978, 7: 17–24Google Scholar
Quershi, S T, Larivière, L, Leveque, G, Clermont, S, Moore, K J, Gros, P and Malo, D. Endotoxin-tolerant mice have mutations in Toll-like receptor 4 (TLR4). J Exp Med 1999, 189: 615–625Google Scholar
Randow, F and Seed, B. Endoplasmic reticulum chaperone gp96 is required for innate immunity but not cell viability. Nat Cell Biol 2001, 3: 891–896Google Scholar
Kadowaki, N, Ho, S, Antonenko, S, Malefyt, R W, Kastelein, R A and Bazan, F. Subsets of human dendritic cell precursors express different toll-like receptor and respond to different microbial antigens. J Exp Med 2001, 163: 5786–5795Google Scholar
Lee, H H, Dempsey, P W, Parks, T P, Zhu, X, Baltimore, D and Cheng, G. Specificities of CD40 signaling: involvement of TRAF2 in CD40-induced NF-κB activation and intercellular adhesion molecule-1 up-regulation. Proc Natl Acad Sci USA 1999, 96: 1421–1426Google Scholar
Quezada, S A, Jarvinen, L Z, Lind, E F and Noelle, R J. CD40/CD154 interactions at the interface of tolerance and immunity. Ann Rev Immunol 2004, 22: 307–328Google Scholar
Vabulas, R M, Wagner, H and Schild, H. Heat shock proteins as ligands of Toll-like receptors. Cur Topics Microbiol Immunol 2002, 270: 169–184Google Scholar
Markees, T G, Phillips, N E, Noelle, R J, Shultz, L D, Mordes, J P, Greiner, D L and Rossini, A A. Prolonged survival of mouse skin allografts in recipients treated with donor splenocytes and antibody to CD40 ligand. Transplantation 1997, 64: 329–335Google Scholar
Gallucci, S, Lolkema, M and Matzinger, P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med 1999, 11: 1249–1255Google Scholar
Sauter, B, Albert, M L, Francisco, L, Larsson, M, Somersan, S and Bhardwaj, N. Consequences of cell death: exposure to necrotic tumour cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med 2000, 191: 423–433Google Scholar
Shi, Y and Rock, K L. Cell death releases endogenous adjuvants that selectively enhance immune surveillance of particulate antigens. Eur J Immunol 2002, 32: 155–162Google Scholar
Shi, Y, Zhang, W and Rock, K L. Cell injury releases endogenous adjuvants that stimulate cytotoxic T cell responses. Proc Natl Acad Sci USA 2000, 97: 14590–14595Google Scholar
Steinman, R M, Turley, S, Mellman, I and Inaba, K. The induction of tolerance by dendritic cells that have captured apoptotic cells. J Exp Med 2000, 191: 411–416Google Scholar
Steinman, R M, Hawiger, D and Nussenzweig, M C. Tolerogenic dendritic cells. Ann Rev Immunol 2003, 21: 685–711Google Scholar
Lutz, M B and Schuler, G. Immature, semi-mature and fully mature dendritic cells: which signals induce tolerance or immunity. Trends Immunol 2002, 23: 445–449Google Scholar
Menges, M, Rößner, S, Voigtländer, C, Schindler, H, Kukutsch, N A, Bogdan, C, Erb, K, Schuler, G and Lutz, M B. Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce antigen-specific protection of mice from autoimmunity. J Exp Med 2002, 195: 15–21Google Scholar
Dhodapkar, M V, Steinman, R M, Krasovsky, J, Munz, C and Bhardwaj, N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med 2001, 193: 233–238Google Scholar
Legge, K L, Gregg, R K, Maldonado-Lopez, R, Li, L, Caprio, J C, Moser, M and Zaghouani, H. On the role of dendritic cells in peripheral T cell tolerance and modulation of autoimmunity. J Exp Med 2002, 196: 217–227Google Scholar
Ferguson, T A, Herndon, J, Elzey, B, Griffith, T S, Schoenberger, S and Green, D R. Uptake of apoptotic cells by lymphoid dendritic cells and cross-priming of CD8+ T cells produce active immune unresponsiveness. J Immunol 2002, 168: 5589–5595Google Scholar
Miga, A J, Masters, S R, Durell, B G, Gonzalez, M, Jenkins, M K, Maliszewski, C, Kikutani, H, Wade, W F and Noelle, R J. Dendritic cell longevity and T cell persistence is controlled by CD154-CD40 interactions. Eur J Immunol 2001, 31: 959–965Google Scholar
Grohmann, U, Fallarino, F, Silla, S, Biachi, R, Belladonna, M L, Vacca, C, Micheletti, A, Fioretti, M C and Puccetti, P. CD40 ligation abrogates the tolerogenic potential of lymphoid dendritic cells. J Immunol 2001, 166: 277–283Google Scholar
Mycko, M P, Cwiklinska, H, Szymanski, J, Szymanska, B, Kudla, G, Kilianek, L, Odyniec, A, Brosnan, C F and Selmaj, K W. Inducible heat shock protein 70 promotes myelin autoantigen presentation by the HLA class II. J Immunol 2004, 172: 202–213Google Scholar
Steinman, R M and Nussenzweig, M C. Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci USA 2002, 99: 351–358Google Scholar
Bretscher, P A. A two-step, two-signal model for the primary activation of precursor helper T cells. Proc Natl Acad Sci USA 1999, 96: 185–190Google Scholar
Roth, S, Willcox, N, Rzepka, R, Mayer, M P and Melchers, I. Major differences in antigen-processing correlate with a single Arg71↔Lys substitution in HLA-DR molecules predisposing to rheumatoid arthritis and with their selective interactions with 70-kDa heat shock protein chaperones. J Immunol 2002, 169: 3015–3020Google Scholar
Lussow, A R, Barrios, C, Embden, J, Zee, R, Verdini, A S, Pessi, A, Louis, J A, Lambert, P-H and Del Giudice, G. Mycobacterial heat-shock proteins as carrier molecules. Eur J Immunol 1991, 21: 2297–2302Google Scholar
Barrios, C, Lussow, J A, Embden, J, Zee, R, Rappouli, R, Costantino, P, Louis, J A, Lambert, P-H and Del Giudice, G. Mycobacterial heat-shock proteins as carrier molecules. II. The use of the 70kDa mycobacterial carrier for conjugated vaccines can circumvent the need for adjuvants and Bacillus Calmette Guerin priming. Eur J Immunol 1992, 22: 1365–1372Google Scholar
Perraut, R, Lussow, A R, Gavoille, S, Garraud, O, Matile, H, Tougne, C, Embden, J, Zee, R, Lambert, P-H, Gysin, J and Del Giudice, G. Successful primate immunization with peptide conjugated to purified protein derrivative or mycobacterial heat shock proteins in the absence of adjuvants. Clin Exp Immunol 1993, 93: 382–386Google Scholar
Suzue, K and Young, R A. Adjuvant-free hsp70 fusion protein system elicits humoral and cellular immune responses to HIV-1 p24. J Immunol 1996, 156: 873–879Google Scholar
Udono, H and Srivastava, P K. Heat shock protein 70-associated peptides elicit specific cancer immunity. J Exp Med 1993, 178: 1391–1396Google Scholar
Nieland, T J F, Tan, M C A A, Monee-van, Muijen M, Koning, F, Kruisbeek, A M and Bleek, G M. Isolation of an immunodominant viral peptide that is endogenously bound to the stress protein GP96/GRP94. Proc Natl Acad Sci USA 1996, 93: 6135–6139Google Scholar
Ciupitu, A T, Petersson, M, O'Donnell, C L, Williams, K, Jindal, S, Kiessling, R and Welsh, R M. Immunization with a lymphocytic choriomeningitis virus peptide mixed with heat shock protein 70 results in protective antiviral immunity and specific cytotoxic T lymphocytes. J Exp Med 1998, 187: 685–691Google Scholar
Schall, T J, Bacon, K, Toy, K J and Goedell, D V. Selective attraction of monocytes and T lymphocytes of the memory phenotype by cytokine RANTES. Nature 1990, 347: 669–671Google Scholar
Murphy, W J, Taub, D D, Anver, M, Conlon, K, Oppenheim, J J, Kelvin, D J and Longo, D L. Human RANTES induces the migration of human T lymphocytes into the peripheral tissues of mice with severe combined immune deficiency. Eur J Immunol 1994, 24: 1823–1827Google Scholar
Meurer, R, Riper, G, Feeney, W, Cunningham, P, Hora, D J, Springer, M S, MacIntyre, D E and Rosen, H. Formation of eosinophilic and monocytic intradermal inflammatory sites in the dog by injection of human RANTES but not human monocyte chemoattractant protein 1, human macrophage inflammatory protein 1 α, or human interleukin 8. J Exp Med 1993, 178: 1913–1921Google Scholar
Kim, J J, Nottingham, L K, Sin, J I, Tsai, A, Morrison, L, Oh, J, Dang, K, Hu, Y, Kazahaya, K, Bennett, M, Dentchev, T, Wilson, D M, Chalian, A A, Boyer, J D, Agadjanyan, M G and Weiner, D B. CD8 positive influence antigen-specific immune responses through the expression of chemokines. J Clin Invest 1998, 102: 1112–1124Google Scholar
Schall, T J, Bacon, K, Camp, R D, Kaspari, J W and Goeddel, D V. Human macrophage inflammatory protein alpha (MIP-1α) and MIP-1β chemokines attract distinct populations of lymphocytes. J Exp Med 1993, 177: 1821–1826Google Scholar
Dieu, M C, Vanbervliet, B, Vicari, A, Bridon, J M, Oldham, E, Ait-Yahia, S, Briere, F, Zlotnik, A, Lebecque, S and Caux, C. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J Exp Med 1998, 188: 373–386Google Scholar
Trinchieri, G. Interleukin-12: a cytokine produced by antigen presenting cells with immunoregulatory functions in the generation of T-helper cells type 1 and cytotoxic lymphocytes. Blood 1994, 84: 4008–4027Google Scholar
Thole, J E, Schooten, W C, Keulen, W J, Hermans, P W, Janson, A A, Vries, R R, Kolk, A H and Embden, J D. Use of recombinant antigens expressed in Escherichia coli K-12 to map B-cell and T-cell epitopes on the immunodominant 65-kilodalton protein of Mycobacterium bovis BCG. Infect Immun 1988, 56: 1633–1640Google Scholar
Ivanyi, J, Sharp, K, Jackett, P and Bothamley, G. Immunological study of defined constituents of mycobacteria. Springer Semin Immunopathol 1988, 10: 279–300Google Scholar
Medzhitov, R M and Janeway, C A J. Innate immunity: impact on the adaptive immune response. Cur Opin Immunol 1997, 9: 4–9Google Scholar
Fearon, D T and Locksley, R M. The instructive role of innate immunity in the acquired immune response. Science 1996, 272: 50–53Google Scholar
Matzinger, P. Tolerance, danger, and the extended family. Ann Rev Immunol 1994, 12: 991–1045Google Scholar
Stewart, G R, Snewin, V A, Walzl, G, Hussell, T, Tormay, P, O'Gaora, P, Goyal, M, Betts, J, Brown, I N and Young, D B. Overexpression of heat-shock proteins reduces survival of Mycobacterium tuberculosis in the chronic phase of infection. Nat Med 2001, 7: 732–737Google Scholar
Bogers, W M, Bergmeier, L A, Ma, J, Oostermeijer, H, Wang, Y, Kelly, C G, Ten Haaft, P, Singh, M, Heeney, J L and Lehner, T. A novel HIV-CCR5 receptor vaccine strategy in the control of mucosal SIV/HIV infection. AIDS 2004, 18: 25–36Google Scholar
Multhoff, G, Mizzen, L, Winchester, C C, Milner, C M, Wenk, S, Eissner, G, Kampinga, H H, Laumbacher, B and Johnson, J. Heat shock protein 70 (Hsp70) stimulates proliferation and cytolytic activity of natural killer cells. Exp Hematol 1999, 27: 1627–1636Google Scholar
Gastpar, R, Gross, C, Rossbacher, L, Ellwart, J, Riegger, J and Multhoff, G. The cell surface-localized heat shock protein 70 epitope TKD induces migration and cytolytic activity selectively in human NK cells. J Immunol 2004, 172: 972–980Google Scholar
Michaëlsson, J, Teixeira Matos, C T, Achour, A, Lanier, L L, Kärre, K and Söderström, K. A signal peptide derived from hsp60 binds HLA-E and interferes with CD94/NKG2A recognition. J Exp Med 2002, 196: 1403–1414Google Scholar
Lehner, T, Mitchell, E, Bergmeier, L, Singh, M, Spallek, R, Cranage, M, Hall, G, Dennis, M, Villinger, F and Wang, Y.The role of γδ T cells in generating antiviral factors and β-chemokines in protection against mucosal simian immunodeficiency virus infection. Eur J Immunol 2000, 30: 2245–2256Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×