Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-wzw2p Total loading time: 0 Render date: 2024-06-02T01:32:31.614Z Has data issue: false hasContentIssue false

12 - Heat Shock Protein Release and Naturally Occurring Exogenous Heat Shock Proteins

Published online by Cambridge University Press:  10 August 2009

Johan Frostegård
Affiliation:
Unit of Rheumatology, Department of Medicine and Center for Molecular Medicine, Karolinska Hospital, Stockholm, Sweden
A. Graham Pockley
Affiliation:
Immunobiology Research Unit, Division of Clinical Sciences (North), (University of Sheffield), Northern General Hospital, Sheffield, United Kingdom
Brian Henderson
Affiliation:
University College London
A. Graham Pockley
Affiliation:
University of Sheffield
Get access

Summary

Introduction

Although for many years the perception has been that mammalian heat shock proteins are intracellular molecules that are only released into the extracellular environment in pathological situations such as necrotic cell death, it is now known that these molecules can be released from a variety of viable (non-necrotic) cell types [1–4]. Moreover, we and a number of others have reported Hsp60 and/or Hsp70 to be present in the peripheral circulation of normal individuals [5–12]. These observations have profound implications for the perceived role of these proteins as pro-inflammatory intercellular ‘danger’ signalling molecules and have prompted a re-evaluation of the functional significance and role(s) of these ubiquitously expressed and highly conserved families of molecules. The reader should refer to Chapter 2, which discusses the intracellular dispositions of molecular chaperones and also touches on the release of heat shock proteins, and Chapter 3, in which novel pathways of protein release are described.

The mechanism(s) leading to the release of heat shock proteins are unknown, as is the source of circulating heat shock proteins in the peripheral circulation and their physiological and pathophysiological role(s). The inverse relationship between levels of circulating Hsp70 and the progression of carotid atherosclerosis [13], or the presence of coronary artery disease (CAD) [14], appears to be inconsistent with the concept that this molecule is a danger signal and an in vitro activator of innate and pro-inflammatory immunity [15].

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

Hightower, L E and Guidon, P T. Selective release from cultured mammalian cells of heat-shock (stress) proteins that resemble glia-axon transfer proteins. J Cell Physiol 1989, 138: 257–266CrossRefGoogle ScholarPubMed
Child, D F, Williams, C P, Jones, R P, Hudson, P R, Jones, M and Smith, C J. Heat shock protein studies in type 1 and type 2 diabetes and human islet cell culture. Diabetic Med 1995, 12: 595–599CrossRefGoogle ScholarPubMed
Bassan, M, Zamostiano, R, Giladi, E, Davidson, A, Wollman, Y, Pitman, J, Hauser, J, Brenneman, D E and Gozes, I. The identification of secreted heat shock 60-like protein from rat glial cells and a human neuroblastoma cell line. Neurosci Letters 1998, 250: 37–40CrossRefGoogle Scholar
Liao, D-F, Jin, Z-G, Baas, A S, Daum, G, Gygi, S P, Aebersold, R and Berk, B C. Purification and identification of secreted oxidative stress-induced factors from vascular smooth muscle cells. J Biol Chem 2000, 275: 189–196CrossRefGoogle ScholarPubMed
Pockley, A G, Wu, R, Lemne, C, Kiessling, R, Faire, U and Frostegård, J. Circulating heat shock protein 60 is associated with early cardiovascular disease. Hypertension 2000, 36: 303–307CrossRefGoogle ScholarPubMed
Pockley, A G, Bulmer, J, Hanks, B M and Wright, B H. Identification of human heat shock protein 60 (Hsp60) and anti-Hsp60 antibodies in the peripheral circulation of normal individuals. Cell Stress Chaperon 1999, 4: 29–352.3.CO;2>CrossRefGoogle ScholarPubMed
Pockley, A G, Shepherd, J and Corton, J. Detection of heat shock protein 70 (Hsp70) and anti-Hsp70 antibodies in the serum of normal individuals. Immunol Invest 1998, 27: 367–377CrossRefGoogle ScholarPubMed
Xu, Q, Schett, G, Perschinka, H, Mayr, M, Egger, G, Oberhollenzer, F, Willeit, J, Kiechl, S and Wick, G. Serum soluble heat shock protein 60 is elevated in subjects with atherosclerosis in a general population. Circulation 2000, 102: 14–20CrossRefGoogle Scholar
Rea, I M, McNerlan, S and Pockley, A G. Serum heat shock protein and anti-heat shock protein antibody levels in aging. Exp Gerontol 2001, 36: 341–352CrossRefGoogle Scholar
Lewthwaite, J, Owen, N, Coates, A, Henderson, B and Steptoe, A. Circulating human heat shock protein 60 in the plasma of British civil servants. Circulation 2002, 106: 196–201CrossRefGoogle ScholarPubMed
Pockley, A G, Faire, U, Kiessling, R, Lemne, C, Thulin, T and Frostegård, J. Circulating heat shock protein and heat shock protein antibody levels in established hypertension. J Hypertension 2002, 20: 1815–1820CrossRefGoogle ScholarPubMed
Njemini, R, Lambert, M, Demanet, C and Mets, T. Elevated serum heat-shock protein 70 levels in patients with acute infection: use of an optimized enzyme-linked immunosorbent assay. Scand J Immunol 2003, 58: 664–669CrossRefGoogle ScholarPubMed
Pockley, A G, Georgiades, A, Thulin, T, Faire, U and Frostegård, J. Serum heat shock protein 70 levels predict the development of atherosclerosis in subjects with established hypertension. Hypertension 2003, 42: 235–238CrossRefGoogle ScholarPubMed
Zhu, J, Quyyumi, A A, Wu, H, Csako, G, Rott, D, Zalles-Ganley, A, Ogunmakinwa, J, Halcox, J and Epstein, S E. Increased serum levels of heat shock protein 70 are associated with low risk of coronary artery disease. Arterioscler Thromb Vasc Biol 2003, 23: 1055–1059CrossRefGoogle ScholarPubMed
Todryk, S M, Gough, M J and Pockley, A G. Facets of heat shock protein 70 show immunotherapeutic potential. Immunology 2003, 110: 1–9CrossRefGoogle ScholarPubMed
Bulmer, J, Bolton, A E and Pockley, A G. Effect of combined heat, ozonation and ultraviolet light (VasoCare™) on heat shock protein expression by peripheral blood leukocyte populations. J Biol Reg Homeostatic Agents 1997, 11: 104–110Google Scholar
Tytell, M, Greenberg, S G and Lasek, R J. Heat shock-like protein is transferred from glia to axon. Brain Res 1986, 363: 161–164CrossRefGoogle ScholarPubMed
Guzhova, I, Kislyakova, K, Moskaliova, O, Fridlanskaya, I, Tytell, M, Cheetham, M and Margulis, B. In vitro studies show that Hsp70 can be released by glia and that exogenous Hsp70 can enhance neuronal stress tolerance. Brain Res 2001, 914: 66–73CrossRefGoogle ScholarPubMed
Brudzynski, K and Martinez, V. Synaptophysin-containing microvesicles transport heat-shock protein Hsp60 in insulin-secreting β cells. Cytotechnology 1993, 11: 23–33CrossRefGoogle ScholarPubMed
Wang, M H, Grossmann, M E and Young, C Y. Forced expression of heat-shock protein 70 increases the secretion of Hsp70 and provides protection against tumour growth. Brit J Cancer 2004, 90: 926–931CrossRefGoogle ScholarPubMed
Barreto, A, Gonzalez, J M, Kabingu, E, Asea, A and Fiorentino, S. Stress-induced release of HSC70 from human tumors. Cell Immunol 2003, 222: 97–104CrossRefGoogle ScholarPubMed
Ménoret, A, Patry, Y, Burg, C and Pendu, J. Co-segregation of tumor immunogenicity with expression of inducible but not constitutive Hsp70 in rat colon carcinomas. J Immunol 1995, 155: 740–747Google Scholar
Martin, C A, Carsons, S E, Kowalewski, R, Bernstein, D, Valentino, M and Santiago-Schwarz, F. Aberrant extracellular and dendritic cell (DC) surface expression of heat shock protein (Hsp)70 in the rheumatoid joint: possible mechanisms of Hsp/DC-mediated cross-priming. J Immunol 2003, 171: 5736–5742CrossRefGoogle ScholarPubMed
Cleves, A E. Protein transports: the nonclassical ins and outs. Curr Biol 1997, 7: R318–R320CrossRefGoogle ScholarPubMed
Evdonin, A L, Guzhova, I V, Margulis, B A and Medvedeva, N D. Phospholipase C inhibitor, U73122, stimulates release of hsp-70 stress protein from A431 human carcinoma cells. Cancer Cell Int 2004, 4: 2CrossRefGoogle ScholarPubMed
Strous, G J and Gent, J. Dimerization, ubiquitylation and endocytosis go together in growth hormone receptor function. FEBS Letters 2002, 529: 102–109CrossRefGoogle ScholarPubMed
Katzmann, D J, Odorizzi, G and Emr, S D. Receptor downregulation and multivesicular-body sorting. Nat Rev Mol Cell Biol 2002, 3: 893–905CrossRefGoogle ScholarPubMed
Théry, C, Boussac, M, Véron, P, Ricciardi-Castagnoli, P, Raposo, G, Garin, G and Amigorena, S. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J Immunol 2001, 166: 7309–7318CrossRefGoogle ScholarPubMed
Denzer, K, Kleijmeer, M J, Heijnen, H F, Stoorvogel, W and Geuze, H J. Exosome: from internal vesicle of the multivesicular body to intercellular signalling device. J Cell Sci 2000, 113 Pt 19: 3365–3374Google Scholar
Théry, C, Zitvogel, L and Amigorena, S. Exosomes: composition, biogenesis and function. Nat Rev Immunol 2002, 2: 569–579CrossRefGoogle ScholarPubMed
Chaput, N, Taïeb, J, Schartz, N E, Andre, F, Angevin, E and Zitvogel, L. Exosome-based immunotherapy. Cancer Immunol Immunother 2004, 53: 234–239CrossRefGoogle ScholarPubMed
Mathew, A, Bell, A and Johnstone, R M. Hsp-70 is closely associated with the transferrin receptor in exosomes from maturing reticulocytes. Biochem J 1995, 308: 823–830CrossRefGoogle ScholarPubMed
Broquet, A H, Thomas, G, Masliah, J, Trugnan, G and Bachelet, M. Expression of the molecular chaperone Hsp70 in detergent-resistant microdomains correlates with its membrane delivery and release. J Biol Chem 2003, 278: 21601–21606CrossRefGoogle ScholarPubMed
Pralle, A, Keller, P, Florin, E L, Simons, K and Horber, J K. Sphingolipid-cholesterol rafts diffuse as small entities in the plasma membrane of mammalian cells. J Cell Biol 2000, 148: 997–1008CrossRefGoogle ScholarPubMed
Vereb, G, Matko, J, Vamosi, G, Ibrahim, S M, Magyar, E, Varga, S, Szollosi, J, Jenei, A, Gaspar, R J, Waldmann, T A and Damjanovich, S. Cholesterol-dependent clustering of IL-2Rα and its colocalization with HLA and CD48 on T lymphoma cells suggest their functional association with lipid rafts. Proc Natl Acad Sci USA 2000, 97: 6013–3018CrossRefGoogle Scholar
Horejsi, V, Cebecauer, M, Cerny, J, Brdicka, T, Angelisova, P and Drbal, K. Signal transduction in leucocytes via GPI-anchored proteins: an experimental artefact or an aspect of immunoreceptor function?Immunol Lett 1998, 63: 63–73CrossRefGoogle ScholarPubMed
Triantafilou, M, Miyake, K, Golenbock, D T and Triantafilou, K. Mediators of innate immune recognition of bacteria concentrate in lipid rafts and facilitate lipopolysaccharide-induced cell activation. J Cell Sci 2002, 115: 2603–2611Google ScholarPubMed
Walsh, R C, Koukoulas, I, Garnham, A, Moseley, P L, Hargreaves, M and Febbraio, M A. Exercise increases serum Hsp72 in humans. Cell Stress Chaperon 2001, 6: 386–3932.0.CO;2>CrossRefGoogle ScholarPubMed
Febbraio, M A, Ott, P, Nielsen, H B, Steensberg, A, Keller, C, Krustrup, P, Secher, N H and Pedersen, B K. Exercise induces hepatosplanchnic release of heat shock protein 72 in humans. J Physiol 2002, 544: 957–962CrossRefGoogle ScholarPubMed
Richardson A and Holbrook N J. Aging and the cellular response to stress: reduction in the heat shock response. In Holbrook, N. J., Martin, G. R. and Lockshin, R. A. (Eds.) Cellular Aging and Cell Death. Wiley-Liss, New York 1996, pp 67–79Google Scholar
Shelton, D N, Chang, E, Whittier, P S, Choi, D and Funk, W D. Microarray analysis of replicative senescence. Curr Biol 1999, 9: 939–945CrossRefGoogle ScholarPubMed
Fargnoli, J, Kunisada, T, Fornace, A J J, Schneider, E L and Holbrook, N J. Decreased expression of heat shock protein 70 mRNA and protein after heat treatment in cells of aged rats. Proc Natl Acad Sci USA 1990, 87: 846–850CrossRefGoogle ScholarPubMed
Heydari, A R, Conrad, C C and Richardson, A. Expression of heat shock genes in hepatocytes is affected by age and food restriction in rats. J Nutrition 1995, 125: 410–418Google ScholarPubMed
Hall, D, Xu, L, Drake, V J, Oberley, L W, Oberley, T D, Museley, P L and Kregel, K C. Aging reduces adaptive capacity and stress protein expression in the liver after heat stress. J Appl Physiol 2000, 2: 749–759CrossRefGoogle Scholar
Pahlavani, M A, Denny, M, Moore, S A, Weindruch, R and Richardson, A. The expression of heat shock protein 70 decreases with age in lymphocytes from rats and rhesus monkeys. Expl Cell Res 1995, 218: 310–318CrossRefGoogle ScholarPubMed
Gray, C C, Amrani, M, Smolenski, R T, Taylor, G I and Yacoub, M H. Age dependence of heat stress mediated cardioprotection. Ann Thoracic Surg 2000, 2: 621–626CrossRefGoogle Scholar
Nitta, Y, Abe, K, Aoki, M, Ohno, I and Isoyama, S. Diminished heat shock protein 70 mRNA induction in aged rats after ischemia. Am J Physiol 1994, 267: H1795–1803Google ScholarPubMed
Faassen, A E, O'Leary, J J, Rodysill, K J, Bergh, N and Hallgren, H M. Diminished heat-shock protein synthesis following mitogen stimulation of lymphocytes from aged donors. Exp Cell Res 1989, 183: 326–334CrossRefGoogle ScholarPubMed
Bernstein, S L, Liu, A M, Hansen, B C and Somiari, R I. Heat shock cognate-70 gene expression declines during normal aging of the primate retina. Invest Ophthalmol Vis Sci 2000, 10: 2857–2862Google Scholar
Liu, A Y, Lin, Z, Choi, H, Sorhage, F and Li, B. Attenuated induction of heat shock gene expression in aging diploid fibroblasts. J Biol Chem 1989, 264: 12037–12045Google ScholarPubMed
Luce, M C and Cristofalo, V J. Reduction in heat shock gene expression correlates with increased thermosensitivity in senescent human fibroblasts. Exp Cell Res 1992, 202: 9–16CrossRefGoogle ScholarPubMed
Effros, R B, Zhu, X and Walford, R L. Stress response of senescent T lymphocytes: reduced hsp70 is independent of the proliferative block. J Gerontol 1994, 49: B65–70CrossRefGoogle ScholarPubMed
Jin, X, Wang, R, Xiao, C, Cheng, L, Wang, F, Yang, L, Feng, T, Chen, M, Chen, S, Fu, X, Deng, J, Wang, R, Tang, F, Wei, Q, Tanguay, R M and Wu, T. Serum and lymphocyte levels of heat shock protein 70 in aging: a study in the normal Chinese population. Cell Stress Chaperon 2004, 9: 69–752.0.CO;2>CrossRefGoogle ScholarPubMed
Wick, G, Kleindienst, R, Schett, G, Amberger, A and Xu, Q. Role of heat shock protein 65/60 in the pathogenesis of atherosclerosis. Int Arch Allergy Immunol 1995, 107: 130–131CrossRefGoogle ScholarPubMed
Pockley, A G. Heat shock proteins, inflammation and cardiovascular disease. Circulation 2002, 105: 1012–1017CrossRefGoogle ScholarPubMed
Zal, B, Kaski, J C, Arno, G, Akiyu, J P, Xu, Q, Cole, D, Whelan, M, Russell, N, Madrigal, J A, Dodi, I A and Baboonian, C. Heat-shock protein 60-reactive CD4+CD28null T cells in patients with acute coronary syndromes. Circulation 2004, 109: 1230–1235CrossRefGoogle ScholarPubMed
Wright, B H, Corton, J, El-Nahas, A M, Wood, R F M and Pockley, A G. Elevated levels of circulating heat shock protein 70 (Hsp70) in peripheral and renal vascular disease. Heart Vessels 2000, 15: 18–22CrossRefGoogle ScholarPubMed
Kingston, A E, Hicks, C A, Colston, M J and Billingham, M E J. A 71-kD heat shock protein (hsp) from Mycobacterium tuberculosis has modulatory effects on experimental rat arthritis. Clin Exp Immunol 1996, 103: 77–82CrossRefGoogle ScholarPubMed
Tanaka, S, Kimura, Y, Mitani, A, Yamamoto, G, Nishimura, H, Spallek, R, Singh, M, Noguchi, T and Yoshikai, Y. Activation of T cells recognizing an epitope of heat-shock protein 70 can protect against rat adjuvant arthritis. J Immunol 1999, 163: 5560–5565Google ScholarPubMed
Wendling, U, Paul, L, Zee, R, Prakken, B, Singh, M and Eden, W. A conserved mycobacterial heat shock protein (hsp) 70 sequence prevents adjuvant arthritis upon nasal administration and induces IL-10-producing T cells that cross-react with the mammalian self-hsp70 homologue. J Immunol 2000, 164: 2711–2717CrossRefGoogle ScholarPubMed
Detanico, T, Rodrigues, L, Sabritto, A C, Keisermann, M, Bauer, M E, Zwickey, H and Bonorino, C. Mycobacterial heat shock protein 70 induced interleukin-10 production: immunomodulation of synovial cell cytokine profile and dendritic cell maturation. Clin Exp Immunol 2004, 135: 336–342CrossRefGoogle ScholarPubMed
Liuzzo, G, Kopecky, S L, Frye, R L, O' Fallon, W M, Maseri, A, Goronzy, J J and Weyand, C M. Perturbation of the T-cell repertoire in patients with unstable angina. Circulation 1999, 100: 2135–2139CrossRefGoogle ScholarPubMed
Nakajima, T, Schulte, S, Warrington, K J, Kopecky, S L, Frye, R L, Goronzy, J J and Weyand, C M. T-cell-mediated lysis of endothelial cells in acute coronary syndromes. Circulation 2002, 105: 570–575CrossRefGoogle ScholarPubMed
Pagetta, A, Folda, A, Brunati, A M and Finotti, P. Identification and purification from the plasma of type 1 diabetic subjects of a proteolytically active Grp94. Evidence that Grp94 is entirely responsible for plasma proteolytic activity. Diabetologia 2003, 46: 996–1006CrossRefGoogle Scholar
Finotti, P and Pagetta, A. A heat shock protein 70 fusion protein with α1-antitrypsin in plasma of type 1 diabetic subjects. Biochem Biophys Res Comm 2004, 315: 297–305CrossRefGoogle ScholarPubMed
Finotti, P, Carraro, P and Calderan, A. Purification of proteinase-like and Na+/K+-ATPase stimulating substance from plasma of insulin-dependent diabetics and its identification as α1-antitrypsin. Biochim Biophys Acta 1992, 1139: 122–132CrossRefGoogle Scholar
Hunter-Lavin, C, Hudson, P R, Mukherjee, S, Davies, G K, Williams, C P, Harvey, J N, Child, D F and Williams, J H H. Folate supplementation reduces serum Hsp70 levels in patients with type 2 diabetes. Cell Stress Chaperone, 2004, 9: 344–349CrossRefGoogle ScholarPubMed
Sampson, M J, Gopaul, N, Davies, I R, Hughes, D A and Carrier, M J. Plasma F2 isoprostanes: direct evidence of increased free radical damage during acute hyperglycemia in type 2 diabetes. Diabetes Care 2002, 25: 537–541CrossRefGoogle ScholarPubMed
Spanheimer, G R. Reducing cardiovascular risk in diabetes. Which factors to modify first?Postgrad Med 2001, 109: 33–36CrossRefGoogle ScholarPubMed
Stehouwer, C D, Gall, M A, Hougaard, P, Jakobs, C and Parving, H H. Plasma homocysteine concentration predicts mortality in non-insulin-dependent diabetic patients with and without microalbuminuria. Kidney Int 1999, 55: 308–314CrossRefGoogle Scholar
Marmot, M G, Smith, G D, Stansfeld, S, Patel, C, North, F, Head, J, White, I, Brunner, E and Feeney, A. Health inequalities among British civil servants: the Whitehall II study. Lancet 1991, 337: 1387–1393CrossRefGoogle ScholarPubMed
Fleshner, M, Campisi, J, Amiri, L and Diamond, D M. Cat exposure induces both intra- and extracellular Hsp72: the role of adrenal hormones. Psychoneuroendocrinology 2004, 29: 1142–1152CrossRefGoogle ScholarPubMed
Kimura, F, Itoh, H, Ambiru, S, Shimizu, H, Togawa, A, Yoshidome, H, Ohtsuka, M, Shimamura, F, Kato, A, Nukui, Y and Miyazaki, M. Circulating heat-shock protein 70 is associated with postoperative infection and organ dysfunction after liver resection. Am J Surg 2004, 187: 777–784CrossRefGoogle ScholarPubMed
Pittet, J F, Lee, H, Morabito, D, Howard, M B, Welch, W J and Mackersie, R C. Serum levels of Hsp 72 measured early after trauma correlate with survival. J Trauma 2002, 52: 611–617Google ScholarPubMed
Dybdahl, B, Wahba, A, Lien, E, Flo, T H, Waage, A, Qureshi, N, Sellevold, O F, Espevik, T and Sundan, A. Inflammatory response after open heart surgery: release of heat-shock protein 70 and signaling through Toll-like receptor-4. Circulation 2002, 105: 685–690CrossRefGoogle ScholarPubMed
Dybdahl, B, Wahba, A, Haaverstad, R, Kirkeby-Garstad, I, Kierulf, P, Espevik, T and Sundan, A. On-pump versus off-pump coronary artery bypass grafting: more heat-shock protein 70 is released after on-pump surgery. Eur J Cardiothorac Surg 2004, 25: 985–992CrossRefGoogle ScholarPubMed
Lewthwaite, J C, Coates, A R M, Tormay, P, Singh, M, Mascagni, P, Poole, S, Roberts, M, Sharp, L and Henderson, B. Mycobacterium tuberculosis chaperonin 60.1 is a more potent cytokine stimulator than chaperonin 60.2 (Hsp 65) and contains a CD14-binding domain. Infect Immun 2001, 69: 7349–7355CrossRefGoogle ScholarPubMed
Lewthwaite, J, George, R, Lund, P A, Poole, S, Tormay, P, Sharp, L, Coates, A R M and Henderson, B. Rhizobium leguminosarum chaperonin 60.3, but not chaperonin 60.1, induces cytokine production by human monocytes: activity is dependent on interaction with cell surface CD14. Cell Stress Chaperon 2002, 7: 130–1362.0.CO;2>CrossRefGoogle Scholar
Kirby, A C, Meghji, S, Nair, S P, White, P, Reddi, K, Nishihara, T, Nakashima, K, Willis, A C, Sim, R, Wilson, M and Henderson, B. The potent bone-resorbing mediator of Actinobacillus actinomycetemcomitans is homologous to the molecular chaperone GroEL. J Clin Invest 1995, 96: 1185–1194CrossRefGoogle ScholarPubMed
Reddi, K, Meghji, S, Nair, S P, Arnett, T R, Miller, A D, Preuss, M, Wilson, M, Henderson, B and Hill, P. The Escherichia coli chaperonin 60 (groEL) is a potent stimulator of osteoclast formation. J Bone Miner Res 1998, 13: 1260–1266CrossRefGoogle ScholarPubMed
Meghji, S, White, P, Nair, S P, Reddi, K, Heron, K, Henderson, B, Zaliani, A, Fossati, G, Mascagni, P, Hunt, J F, Roberts, M M and Coates, A R. Mycobacterium tuberculosis chaperonin 10 stimulates bone resorption: a potential contributory factor in Pott's disease. J Exp Med 1997, 186: 1241–1246CrossRefGoogle ScholarPubMed
Habich, C, Kempe, K, Zee, R, Burkart, V and Kolb, H. Different heat shock protein 60 species share pro-inflammatory activity but not binding sites on macrophages. FEBS Letters 2003, 533: 105–109CrossRefGoogle 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: 1277–1285CrossRefGoogle ScholarPubMed
Ramage, J M, Young, J L, Goodall, J C and Hill Gaston, J S. T cell responses to heat shock protein 60: differential responses by CD4+ T cell subsets according to their expression of CD45 isotypes. J Immunol 1999, 162: 704–710Google ScholarPubMed
Roon, J A G, Eden, W, Roy, J L A M, Lafeber, F J P G and Bijlsma, J W J. Stimulation of suppressive T cell responses by human but not bacterial 60-kD heat shock protein in synovial fluid of patients with rheumatoid arthritis. J Clin Invest 1997, 100: 459–463CrossRefGoogle Scholar
Res, P C, Schaar, C G, Breedveld, F C, Eden, W, Embden, J D S, Cohen, I R and Vries, R R P. Synovial fluid T cell reactivity against 65 kDa heat shock protein of mycobacteria in early chronic arthritis. Lancet 1988, : 478–480CrossRefGoogle Scholar
Gaston, J S H, Life, P F, Jenner, P J, Colston, M J and Bacon, P A. Recognition of a mycobacteria-specific epitope in the 65kD heat shock protein by synovial fluid derived T cell clones. J Exp Med 1990, 171: 831–841CrossRefGoogle Scholar
Graeff-Meeder, E R, Zee, R, Rijkers, G T, Schuurman, H J, Kuis, W, Bijlsma, J W J, Zegers, B J M and Eden, W. Recognition of human 60 kD heat shock protein by mononuclear cells from patients with juvenile chronic arthritis. Lancet 1991, 337: 1368–1372CrossRefGoogle ScholarPubMed
Wucherpfennig, K, Newcombe, J, Li, H, Keddy, C and Cuzner, M L. γδ T cell receptor repertoire in acute multiple sclerosis lesions. Proc Natl Acad Sci USA 1992, 89: 4588–4592CrossRefGoogle Scholar
Georgopoulos, C and McFarland, H. Heat shock proteins in multiple sclerosis and other autoimmune diseases. Immunology Today 1993, 14: 373–375CrossRefGoogle ScholarPubMed
Stinissen, P, Vandevyver, C, Medaer, R, Vandegaar, L, Nies, J, Tuyls, L, Hafler, D A, Raus, J and Zhang, J. Increased frequency of γδ T cells in cerebrospinal fluid and peripheral blood of patients with multiple sclerosis: reactivity, cytotoxicity, and T cell receptor V gene rearrangements. J Immunol 1995, 154: 4883–4894Google Scholar
Elias, D, Markovits, D, Reshef, T, Zee, R and Cohen, I R. Induction and therapy of autoimmune diabetes in the non-obese diabetic mouse by a 65-kDa heat shock protein. Proc Natl Acad Sci USA 1990, 87: 1576–1580CrossRefGoogle ScholarPubMed
Child, D, Smith, C and Williams, C. Heat shock protein and the double insult theory for the development of insulin-dependent diabetes. J Royal Soc Med (Eng) 1993, 86: 217–219Google ScholarPubMed
Tun, R Y M, Smith, M D, Lo, S S M, Rook, G A W, Lydyard, P and Leslie, R D G. Antibodies to heat shock protein 65 kD in type 1 diabetes mellitus. Diabetic Medicine 1994, 11: 66–70CrossRefGoogle ScholarPubMed
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: 1469–1476CrossRefGoogle ScholarPubMed
Broek, M F, Hogervorst, E J M, Bruggen, M C J, Eden, W, Zee, R and Berg, W. Protection against streptococcal cell wall induced arthritis by pretreatment with the 65kD heat shock protein. J Exp Med 1989, 170: 449–466CrossRefGoogle Scholar
Thompson, S J, Rook, G A W, Brealey, R J, Zee, R and Elson, C J. Autoimmune reactions to heat shock proteins in pristane induced arthritis. Eur J Immunol 1990, 20: 2479–2484CrossRefGoogle ScholarPubMed
Anderton, S M, Zee, R, Prakken, B, Noordzij, A and Eden, W. Activation of T cells recognizing self 60-kD heat shock protein can protect against experimental arthritis. J Exp Med 1995, 181: 943–952CrossRefGoogle ScholarPubMed
Anderton S M and van Eden W. T lymphocyte recognition of Hsp60 in experimental arthritis. In Eden, W. and Young, D. (Eds.) Stress Proteins in Medicine. Marcel Dekker, New York 1996, pp 73–91Google Scholar
Paul, A G A, Kooten, P J S, Eden, W and Zee, R. Highly autoproliferative T cells specific for 60-kDa heat shock protein produce IL-4/IL-10 and IFN-γ and are protective in adjuvant arthritis. J Immunol 2000, 165: 7270–7277CrossRefGoogle ScholarPubMed
Graeff-Meeder, E R, Eden, W, Rijkers, G T, Prakken, B J, Kuis, W, Voorhorst Ogink, M M, Zee, R, Schuurman, H J, Helders, P J and Zegers, B J. Juvenile chronic arthritis: T cell reactivity to human HSP60 in patients with a favorable course of arthritis. J Clin Invest 1995, 95: 934–940CrossRefGoogle ScholarPubMed
Roon, J, Eden, W, Gmelig-Meylig, E, Lafeber, F and Bijlsma, J. Reactivity of T cells from patients with rheumatoid arthritis towards human and mycobacterial hsp60. FASEB 1996, 10: A1312Google Scholar
Macht, L M, Elson, C J, Kirwan, J R, Gaston, J S H, Lamont, A G, Thompson, J M and Thompson, S J. Relationship between disease severity and responses by blood mononuclear cells from patients with rheumatoid arthritis to human heat-shock protein 60. Immunology 2000, 99: 208–214CrossRefGoogle ScholarPubMed
Pockley, A G. Heat shock proteins, heat shock protein reactivity and allograft rejection. Transplantation 2001, 71: 1503–1507CrossRefGoogle ScholarPubMed
Birk, O S, Gur, S L, Elias, D, Margalit, R, Mor, F, Carmi, P, Bockova, J, Altmann, D M and Cohen, I R. The 60-kDa heat shock protein modulates allograft rejection. Proc Nat Acad Sci USA 1999, 96: 5159–5163CrossRefGoogle ScholarPubMed
Caldas, C, Spadafora-Ferreira, M, Fonseca, J A, Luna, E, Iwai, L K, Kalil, J and Coelho, V. T-cell response to self HSP60 peptides in renal transplant recipients: a regulatory role?Transplant Proc 2004, 36: 833–835CrossRefGoogle ScholarPubMed
Cavanagh, A C. Identification of early pregnancy factor as chaperonin 10: implications for understanding its role. Rev Reprod 1996, 1: 28–32CrossRefGoogle ScholarPubMed
Rolfe, B, Cavanagh, A, Forde, C, Bastin, F, Chen, C and Morton, H. Modified rosette inhibition test with mouse lymphocytes for detection of early pregnancy factor in human pregnancy serum. J Immunol Methods 1984, 70: 1–11CrossRefGoogle ScholarPubMed
Zhang, B, Walsh, M D, Nguyen, K B, Hillyard, N C, Cavanagh, A C, McCombe, P A and Morton, H. Early pregnancy factor treatment suppresses the inflammatory response and adhesion molecule expression in the spinal cord of SJL/J mice with experimental autoimmune encephalomyelitis and the delayed-type hypersensitivity reaction to trinitrochlorobenzene in normal BALB/c mice. J Neurol Sci 2003, 212: 37–46CrossRefGoogle ScholarPubMed
Athanasas-Platsis, S, Zhang, B, Hillyard, N C, Cavanagh, A C, Csurhes, P A, Morton, H and McCombe, P A. Early pregnancy factor suppresses the infiltration of lymphocytes and macrophages in the spinal cord of rats during experimental autoimmune encephalomyelitis but has no effect on apoptosis. J Neurol Sci 2003, 214: 27–36CrossRefGoogle ScholarPubMed
De, A K, Kodys, K M, Yeh, B S and Miller-Graziano, C. Exaggerated human monocyte IL-10 concomitant to minimal TNF-α induction by heat-shock protein 27 (Hsp27) suggests Hsp27 is primarily an anti-inflammatory stimulus. J Immunol 2000, 165: 3951–3958CrossRefGoogle ScholarPubMed
Corrigall, V M, Bodman-Smith, M D, Fife, M S, Canas, B, Myers, L K, Wooley, P, Soh, C, Staines, N A, Pappin, D J, Berlo, S E, Eden, W, Zee, R, Lanchbury, J S and Panayi, G S. The human endoplasmic reticulum molecular chaperone BiP is an autoantigen for rheumatoid arthritis and prevents the induction of experimental arthritis. J Immunol 2001, 166: 1492–1498CrossRefGoogle ScholarPubMed
Kilmartin, B and Reen, D J. HSP60 induces self-tolerance to repeated HSP60 stimulation and cross-tolerance to other pro-inflammatory stimuli. Eur J Immunol 2004, 34: 2041–2051CrossRefGoogle ScholarPubMed
Guzhova, I V, Arnholdt, A C, Darieva, Z A, Kinev, A V, Lasunskaia, E B, Nilsson, K, Bozhkov, V M, Voronin, A P and Margulis, B A. Effects of exogenous stress protein 70 on the functional properties of human promonocytes through binding to cell surface and internalization. Cell Stress Chaperon 1998, 3: 67–772.3.CO;2>CrossRefGoogle ScholarPubMed
Sheth, K, De, A, Nolan, B, Friel, J, Duffy, A, Ricciardi, R, Miller-Graziano, C and Bankey, P. Heat shock protein 27 inhibits apoptosis in human neutrophils. J Surg Res 2001, 99: 129–133CrossRefGoogle ScholarPubMed
Hightower, L E. Heat shock, stress proteins, chaperones and proteotoxicity. Cell 1991, 66: 191–197CrossRefGoogle ScholarPubMed
Houenou, L J, Li, L, Lei, M, Kent, C R and Tytell, M. Exogenous heat shock cognate protein Hsc70 prevents axonomy-induced death of spinal sensory neurons. Cell Stress Chaperon 1996, 1: 161–1662.3.CO;2>CrossRefGoogle ScholarPubMed
Johnson, A D, Berberian, P A and Bond, M G. Effect of heat shock proteins on survival of isolated aortic cells from normal and atherosclerotic cynomolgus macaques. Atherosclerosis 1990, 84: 111–119CrossRefGoogle ScholarPubMed
Johnson, A D and Tytell, M. Exogenous Hsp70 becomes cell associated, but not internalised by stressed arterial smooth muscle cells. In Vitro Cell Dev Biol 1993, 29A: 807–812CrossRefGoogle ScholarPubMed
Berberian, P, Johnson, A and Bond, M. Exogenous 70kD heat shock protein increases survival of normal and atheromatous arterial cells. FASEB J 1990, 4: A1031Google Scholar
Jäättelä, M, Wissing, D, Bauer, P A and Li, G C. Major heat shock protein Hsp70 protects tumor cells from tumor necrosis factor cytotoxicity. EMBO J 1992, 11: 3507–3512Google ScholarPubMed
Simon, M M, Reikerstorfer, A, Schwarz, A, Krone, C, Luger, T A, Jäättelä, M and Schwarz, T. Heat shock protein 70 overexpression affects the response to ultraviolet light in murine fibroblasts. Evidence for increased cell viability and suppression of cytokine release. J Clin Invest 1995, 95: 926–933CrossRefGoogle ScholarPubMed
Samali, A and Cotter, T G. Heat shock proteins increase resistance to apoptosis. Exp Cell Res 1996, 223: 163–170CrossRefGoogle ScholarPubMed
Lasunskaia, E B, Fridlianskaia, I I, Guzhova, I V, Bozhkov, V M and Margulis, B A. Accumulation of major stress protein 70kDa protects myeloid and lymphoid cells from death by apoptosis. Apoptosis 1997, 2: 156–163CrossRefGoogle ScholarPubMed
Mosser, D D, Caron, A W, Bourget, L, Denis-Larose, C and Massie, B. Role of the human heat shock protein Hsp70 in protection against stress-induced apoptosis. Mol Cell Biol 1997, 17: 5317–5327CrossRefGoogle ScholarPubMed
Li, C-Y, Lee, J-S, Ko, Y-G, Kim, K-I and Seo, J-S. Heat shock protein 70 inhibits apoptosis downstream of cytochrome c release and upstream of caspase-3 activation. J Biol Chem 2000, 275: 25665–25671CrossRefGoogle ScholarPubMed
Alder, G M, Austen, B M, Bashford, C L, Mehlert, A and Pasternak, C A. Heat shock proteins induce pores in membranes. Biosci Rep 1990, 10: 509–518CrossRefGoogle ScholarPubMed
Yoshida, N, Oeda, K, Watanabe, E, Mikami, T, Fukita, Y, Nishimura, K, Komai, K and Matsuda, K. Protein function. Chaperonin turned insect toxin. Nature 2001, 411: 44CrossRefGoogle ScholarPubMed

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
×