全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...

Heat Shock Proteins and Regulatory T Cells

DOI: 10.1155/2013/813256

Full-Text   Cite this paper   Add to My Lib

Abstract:

Heat shock proteins (HSPs) are important molecules required for ideal protein function. Extensive research on the functional properties of HSPs indicates that HSPs may be implicated in a wide range of physiological functions including immune function. In the immune system, HSPs are involved in cell proliferation, differentiation, cytokine release, and apoptosis. Therefore, the ability of the immune system, in particular immune cells, to function optimally and in unison with other physiological systems is in part dependent on signaling transduction processes, including bidirectional communication with HSPs. Regulatory T cells (Tregs) are important T cells with suppressive functions and impairments in their function have been associated with a number of autoimmune disorders. The purpose of this paper is to examine the relationship between HSPs and Tregs. The interrelationship between cells and proteins may be important in cellular functions necessary for cell survival and expansion during diseased state. 1. Introduction Optimal cellular function is regulated by several molecules including heat shock proteins (HSPs). These proteins have chaperone properties and are important in both stressed and unstressed cells. HSPs can be categorized into six diverse highly or less-conserved families. These include HSP10, HSP40, HSP60, HSP70, HSP90, and HSP100 [1–4]. HSP60 is found in the mitochondria [5]. HSP70 is implicated in protein transport assembly and synthesis. It has anti-apoptotic properties that are implicated in intrinsic and extrinsic apoptotic pathways. HSP70 interacts with the mitochondria through death receptor signaling where it binds to death receptors DR4 and DR5 impeding TNF-related apoptosis inducing ligand (TRAIL) [6]. Importantly, HSP70 can bind to either the unphosphorylated C terminus of protein kinase C or Akt prompting rephosphorylation and kinase stabilization [7]. ATP-dependent HSP90 regulates cell survival by stabilizing kinases such as Akt and suppressing apoptosis by inhibiting caspases [8, 9]. The exact structural domains for all human HSPs remains to be determined; however, HSP70 and HSP90 have been well characterized. HSP70 is comprised of an N-terminal nucleotide-binding domain with ATPase activity and a C terminal containing a substrate-binding domain [10–12]. HSP90 on the other hand has three characterized structural domains including an N-terminal nucleotide-binding domain, a middle segment and a C terminus [13–16]. Inter-domain interactions occur by a conserved linker [17]. HSPs are found in intracellular and extracellular spaces

References

[1]  B. Henderson, “Integrating the cell stress response: a new view of molecular chaperones as immunological and physiological homeostatic regulators,” Cell Biochemistry and Function, vol. 28, no. 1, pp. 1–14, 2010.
[2]  H. H. Kampinga, J. Hageman, M. J. Vos et al., “Guidelines for the nomenclature of the human heat shock proteins,” Cell Stress and Chaperones, vol. 14, no. 1, pp. 105–111, 2009.
[3]  C. Sarto, C. Valsecchi, F. Magni et al., “Expression of heat shock protein 27 in human renal cell carcinoma,” Proteomics, vol. 4, no. 8, pp. 2252–2260, 2004.
[4]  G. Tomasello, C. Sciume, F. Rappa et al., “Hsp10, Hsp70, and Hsp90 immunohistochemical levels change in ulcerative colitis after therapy,” European Journal of Histochemistry, vol. 55, no. 4, article e38, 2011.
[5]  R. Rajaiah and K. D. Moudgil, “Heat-shock proteins can promote as well as regulate autoimmunity,” Autoimmunity Reviews, vol. 8, no. 5, pp. 388–393, 2009.
[6]  Q. Pang, T. A. Christianson, W. Keeble, T. Koretsky, and G. C. Bagby, “The anti-apoptotic function of Hsp70 in the interferon-inducible double-stranded RNA-dependent protein kinase-mediated death signaling pathway requires the Fanconi anemia protein,” FANCC, vol. 277, pp. 49638–49643, 2002.
[7]  T. Gao and A. C. Newton, “The turn motif is a phosphorylation switch that regulates the binding of Hsp70 to protein kinase C,” Journal of Biological Chemistry, vol. 277, no. 35, pp. 31585–31592, 2002.
[8]  A. L. Joly, G. Wettstein, G. Mignot, F. Ghiringhelli, and C. Garrido, “Dual role of heat shock proteins as regulators of apoptosis and innate immunity,” Journal of Innate Immunity, vol. 2, no. 3, pp. 238–247, 2010.
[9]  S. K. Calderwood, S. S. Mambula, and P. J. Gray Jr., “Extracellular heat shock proteins in cell signaling and immunity,” Annals of the New York Academy of Sciences, vol. 1113, pp. 28–39, 2007.
[10]  R. Schlecht, A. H. Erbse, B. Bukau, and M. P. Mayer, “Mechanics of Hsp70 chaperones enables differential interaction with client proteins,” Nature Structural and Molecular Biology, vol. 18, no. 3, pp. 345–351, 2011.
[11]  S. Rüdiger, A. Buchberger, and B. Bukau, “Interaction of Hsp70 chaperones with substrates,” Nature Structural Biology, vol. 4, no. 5, pp. 342–349, 1997.
[12]  A. Cegielska and C. Georgopoulos, “Functional domains of the Escherichia coli dnaK heat shock protein as revealed by mutational analysis,” Journal of Biological Chemistry, vol. 264, no. 35, pp. 21122–21130, 1989.
[13]  C. Prodromou, S. M. Roe, R. O'Brien, J. E. Ladbury, P. W. Piper, and L. H. Pearl, “Identification and structural characterization of the ATP/ADP-binding site in the Hsp90 molecular chaperone,” Cell, vol. 90, no. 1, pp. 65–75, 1997.
[14]  T. Gidalevitz, C. Biswas, H. Ding et al., “Identification of the N-terminal peptide binding site of glucose-regulated protein 94,” The Journal of Biological Chemistry, vol. 279, pp. 16543–16552, 2004.
[15]  S. Vogen, T. Gidalevitz, C. Biswas et al., “Radicicol-sensitive peptide binding to the N-terminal portion of GRP94,” Journal of Biological Chemistry, vol. 277, no. 43, pp. 40742–40750, 2002.
[16]  S. F. Harris, A. K. Shiau, and D. A. Agard, “The crystal structure of the carboxy-terminal dimerization domain of htpG, the Escherichia coli Hsp90, reveals a potential substrate binding site,” Structure, vol. 12, no. 6, pp. 1087–1097, 2004.
[17]  J. Jiang, K. Prasad, E. M. Lafer, and R. Sousa, “Structural basis of interdomain communication in the Hsc70 chaperone,” Molecular Cell, vol. 20, no. 4, pp. 513–524, 2005.
[18]  E. Schmitt, M. Gehrmann, M. Brunet, G. Multhoff, and C. Garrido, “Intracellular and extracellular functions of heat shock proteins: repercussions in cancer therapy,” Journal of Leukocyte Biology, vol. 81, no. 1, pp. 15–27, 2007.
[19]  U. Banerji, M. Walton, F. Raynaud et al., “Pharmacokinetic-pharmacodynamic relationships for the heat shock protein 90 molecular chaperone inhibitor 17-allylamino, 17-demethoxygeldanamycin in human ovarian cancer xenograft models,” Clinical Cancer Research, vol. 11, no. 19, pp. 7023–7032, 2005.
[20]  K. Nanbu, I. Konishi, M. Mandai et al., “Prognostic significance of heat shock proteins HSP70 and HSP90 in endometrial carcinomas,” Cancer Detection and Prevention, vol. 22, no. 6, pp. 549–555, 1998.
[21]  P. Workman, “Combinatorial attack on multistep oncogenesis by inhibiting the Hsp90 molecular chaperone,” Cancer Letters, vol. 206, no. 2, pp. 149–157, 2004.
[22]  A. Clayton, A. Turkes, H. Navabi, M. D. Mason, and Z. Tabi, “Induction of heat shock proteins in B-cell exosomes,” Journal of Cell Science, vol. 118, no. 16, pp. 3631–3638, 2005.
[23]  I. Guzhova, K. Kislyakova, O. Moskaliova et al., “In vitro studies show that Hsp70 can be released by glia and that exogenous Hsp70 can enhance neuronal stress tolerance,” Brain Research, vol. 914, no. 1-2, pp. 66–73, 2001.
[24]  C. Hunter-Lavin, E. L. Davies, M. M. F. V. G. Bacelar, M. J. Marshall, S. M. Andrew, and J. H. H. Williams, “Hsp70 release from peripheral blood mononuclear cells,” Biochemical and Biophysical Research Communications, vol. 324, no. 2, pp. 511–517, 2004.
[25]  G. I. Lancaster and M. A. Febbraio, “Mechanisms of stress-induced cellular HSP72 release: implications for exercise-induced increases in extracellular HSP72,” Exercise immunology review., vol. 11, pp. 46–52, 2005.
[26]  M. A. Bausero, R. Gastpar, G. Multhoff, and A. Asea, “Alternative mechanism by which IFN-γ enhances tumor recognition: active release of heat shock protein 72,” Journal of Immunology, vol. 175, no. 5, pp. 2900–2912, 2005.
[27]  B. Dybdahl, S. A. Sl?rdahl, A. Waage, P. Kierulf, T. Espevik, and A. Sundan, “Myocardial ischaemia and the inflammatory response: release of heat shock protein 70 after myocardial infarction,” Heart, vol. 91, no. 3, pp. 299–304, 2005.
[28]  F. U. Hartl and M. Hayer-Hartl, “Protein folding. Molecular chaperones in the cytosol: from nascent chain to folded protein,” Science, vol. 295, no. 5561, pp. 1852–1858, 2002.
[29]  E. A. Craig, B. D. Gambill, and R. J. Nelson, “Heat shock proteins: molecular chaperones of protein biogenesis,” Microbiological Reviews, vol. 57, no. 2, pp. 402–414, 1993.
[30]  S. Lindquist, “The heat-shock response,” Annual Review of Biochemistry, vol. 55, pp. 1151–1191, 1986.
[31]  C. S?ti, E. Nagy, Z. Giricz, L. Vígh, P. Csermely, and P. Ferdinandy, “Heat shock proteins as emerging therapeutic targets,” British Journal of Pharmacology, vol. 146, no. 6, pp. 769–780, 2005.
[32]  K. D. Sarge, S. P. Murphy, and R. I. Morimoto, “Activation of heat shock gene transcription by heat shock factor 1 involves oligomerization, acquisition of DNA-binding activity, and nuclear localization and can occur in the absence of stress,” Molecular and Cellular Biology, vol. 13, no. 3, pp. 1392–1407, 1993.
[33]  A. Gragerov and M. E. Gottesman, “Different peptide binding specificities of hsp70 family members,” Journal of Molecular Biology, vol. 241, no. 2, pp. 133–135, 1994.
[34]  J. G. Facciponte, I. J. MacDonald, X. Y. Wang, H. Kim, M. H. Manjili, and J. R. Subjeck, “Heat shock proteins and scavenger receptors: role in adaptive immune responses,” Immunological Investigations, vol. 34, no. 3, pp. 325–342, 2005.
[35]  U. Kumaraguru, C. D. Pack, and B. T. Rouse, “Toll-like receptor ligand links innate and adaptive immune responses by the production of heat-shock proteins,” Journal of Leukocyte Biology, vol. 73, no. 5, pp. 574–583, 2003.
[36]  G. Multhoff, “Activation of natural killer cells by heat shock protein 70,” International Journal of Hyperthermia, vol. 25, no. 3, pp. 169–175, 2009.
[37]  C. Gross, I. G. Schmidt-Wolf, S. Nagaraj et al., “Heat shock protein 70-reactivity is associated with increased cell surface density of CD94/CD56 on primary natural killer cells,” Cell Stress & Chaperones, vol. 8, pp. 348–360, 2003.
[38]  S. Stangl, C. Gross, A. G. Pockley, A. A. Asea, and G. Multhoff, “Influence of Hsp70 and HLA-E on the killing of leukemic blasts by cytokine/Hsp70 peptide-activated human natural killer (NK) cells,” Cell Stress and Chaperones, vol. 13, no. 2, pp. 221–230, 2008.
[39]  M. F. Tsan and B. Gao, “Heat shock proteins and immune system,” Journal of Leukocyte Biology, vol. 85, no. 6, pp. 905–910, 2009.
[40]  W. Van Eden, R. Van Der Zee, and B. Prakken, “Heat-shock proteins induce T-cell regulation of chronic inflammation,” Nature Reviews Immunology, vol. 5, no. 4, pp. 318–330, 2005.
[41]  W. Van Eden, G. Wick, S. Albani, and I. Cohen, “Stress, heat shock proteins, and autoimmunity: how immune responses to heat shock proteins are to be used for the control of chronic inflammatory diseases,” Annals of the New York Academy of Sciences, vol. 1113, pp. 217–237, 2007.
[42]  J. M. van Noort, M. Bsibsi, P. Nacken, W. H. Gerritsen, and S. Amor, “The link between small heat shock proteins and the immune system,” The International Journal of Biochemistry & Cell Biology, vol. 44, no. 10, pp. 1670–1679, 2012.
[43]  Y. Bulut, K. S. Michelsen, L. Hayrapetian et al., “Mycobacterium tuberculosis heat shock proteins use diverse toll-like receptor pathways to activate pro-inflammatory signals,” Journal of Biological Chemistry, vol. 280, no. 22, pp. 20961–20967, 2005.
[44]  J. S. H. Gaston, “Heat shock proteins as potential targets in the therapy of inflammatory arthritis,” Biotherapy, vol. 10, no. 3, pp. 197–203, 1998.
[45]  P. A. MacAry, B. Javid, R. A. Floto et al., “HSP70 peptide binding mutants separate antigen delivery from dendritic cell stimulation,” Immunity, vol. 20, no. 1, pp. 95–106, 2004.
[46]  S. Pawaria, M. N. Messmer, Y. J. Zhou, and R. J. Binder, “A role for the heat shock protein-CD91 axis in the initiation of immune responses to tumors,” Immunologic Research, vol. 50, pp. 255–260, 2011.
[47]  F. Stelter, “Structure/function relationships of CD14,” Chemical Immunology, vol. 74, pp. 25–41, 2000.
[48]  A. Asea, S. K. Kraeft, E. A. Kurt-Jones et al., “HSP70 stimulates cytokine production through a CD 14-dependant pathway, demonstrating its dual role as a chaperone and cytokine,” Nature Medicine, vol. 6, no. 4, pp. 435–442, 2000.
[49]  T. Kawai and S. Akira, “TLR signaling,” Cell Death & Differentiation, vol. 13, pp. 816–825, 2006.
[50]  T. S. Blackwell and J. W. Christman, “The role of nuclear factor-kappa B in cytokine gene regulation,” American Journal of Respiratory Cell and Molecular Biology, vol. 17, pp. 3–9, 1997.
[51]  A. R. Brasier, “The NF-kappaB regulatory network,” Cardiovascular Toxicology, vol. 6, pp. 111–130, 2006.
[52]  D. Sun, D. Chen, B. Du, and J. Pan, “Heat shock response inhibits NF-kappaB activation and cytokine production in murine Kupffer cells,” The Journal of Surgical Research, vol. 129, pp. 114–121, 2005.
[53]  Y. Wang, C. Li, X. Wang, J. Zhang, and Z. Chang, “Heat shock response inhibits IL-18 expression through the JNK pathway in murine peritoneal macrophages,” Biochemical and Biophysical Research Communications, vol. 296, no. 3, pp. 742–748, 2002.
[54]  S. K. Calderwood, S. S. Mambula, P. J. Gray Jr., and J. R. Theriault, “Extracellular heat shock proteins in cell signaling,” FEBS Letters, vol. 581, no. 19, pp. 3689–3694, 2007.
[55]  M. Cohen-Sfady, G. Nussbaum, M. Pevsner-Fischer et al., “Heat shock protein 60 activates B cells via the TLR4-MyD88 pathway,” Journal of Immunology, vol. 175, pp. 3594–3602, 2005.
[56]  A. Zanin-Zhorov, G. Nussbaum, S. Franitza, I. R. Cohen, and O. Lider, “T cells respond to heat shock protein 60 via TLR2: activation of adhesion and inhibition of chemokine receptors,” The FASEB Journal, vol. 17, no. 11, pp. 1567–1569, 2003.
[57]  P. Matzinger, “The danger model: a renewed sense of self,” Science, vol. 296, no. 5566, pp. 301–305, 2002.
[58]  W. van Eden, R. Spiering, F. Broere, and R. van der Zee, “A case of mistaken identity: HSPs are no DAMPs but DAMPERs,” Cell Stress & Chaperones, vol. 17, pp. 281–292, 2012.
[59]  H. Kono and K. L. Rock, “How dying cells alert the immune system to danger,” Nature Reviews Immunology, vol. 8, no. 4, pp. 279–289, 2008.
[60]  D. Tang, Y. Shi, L. Jang, K. Wang, W. Xiao, and X. Xiao, “Heat shock response inhibits release of high mobility group box 1 protein induced by endotoxin in murine macrophages,” Shock, vol. 23, no. 5, pp. 434–440, 2005.
[61]  A. Hasegawa, H. Iwasaka, S. Hagiwara, and T. Noguchi, “Relationship between HMGB1 and tissue protective effects of HSP72 in a LPS-induced systemic inflammation model,” Journal of Surgical Research, vol. 169, no. 1, pp. 85–91, 2011.
[62]  I. S. Singh, J. R. He, S. Calderwood, and J. D. Hasday, “A high affinity HSF-1 binding site in the 5′-untranslated region of the murine tumor necrosis factor-alpha gene is a transcriptional repressor,” The Journal of Biological Chemistry, vol. 277, pp. 4981–4988, 2002.
[63]  W. van Eden, “Immunoregulation of autoimmune diseases,” Human Immunology, vol. 67, no. 6, pp. 446–453, 2006.
[64]  W. Van Eden, F. Hauet-Broere, S. Berlo et al., “Stress proteins as inducers and targets of regulatory T cells in arthritis,” International Reviews of Immunology, vol. 24, no. 3-4, pp. 181–197, 2005.
[65]  W. van Eden, U. Wendling, L. Paul, B. Prakken, P. van Kooten, and R. van der Zee, “Arthritis protective regulatory potential of self-heat shock protein cross-reactive T cells,” Cell Stress & Chaperones, vol. 5, pp. 452–457, 2000.
[66]  Y. M. Snyder, L. Guthrie, G. F. Evans, and S. H. Zuckerman, “Transcriptional inhibition of endotoxin-induced monokine synthesis following heat shock in murine peritoneal macrophages,” Journal of Leukocyte Biology, vol. 51, no. 2, pp. 181–187, 1992.
[67]  X. Z. Ding, C. M. Fernandez-Prada, A. K. Bhattacharjee, and D. L. Hoover, “Over-expression of hsp-70 inhibits bacterial lipopolysaccharide-induced production of cytokines in human monocyte-derived macrophages,” Cytokine, vol. 16, pp. 210–219, 2001.
[68]  B. Gao and M. F. Tsan, “Induction of cytokines by heat shock proteins and endotoxin in murine macrophages,” Biochemical and Biophysical Research Communications, vol. 317, no. 4, pp. 1149–1154, 2004.
[69]  C. J. Workman, A. L. Szymczak-Workman, L. W. Collison, M. R. Pillai, and D. A. Vignali, “The development and function of regulatory T cells,” Cellular and Molecular Life Sciences, vol. 66, pp. 2603–2622, 2009.
[70]  E. M. Shevach, “CD4+CD25+ suppressor T cells: more questions than answers,” Nature Reviews Immunology, vol. 2, no. 6, pp. 389–400, 2002.
[71]  S. Sakaguchi, “Naturally arising Foxp3-expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self,” Nature Immunology, vol. 6, pp. 345–352, 2005.
[72]  S. Sakaguchi, “Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses,” Annual Review of Immunology, vol. 22, pp. 531–562, 2004.
[73]  M. Yadav, C. Louvet, D. Davini et al., “Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo,” The Journal of Experimental Medicine, vol. 209, no. 10, pp. 1713–1722, 2012.
[74]  S. Huang, L. Li, S. Liang, and W. Wang, “Conversion of peripheral CD4+CD25- T cells to CD4+CD25+ regulatory T cells by IFN-γ in patients with Guillain-Barré syndrome,” Journal of Neuroimmunology, vol. 217, no. 1-2, pp. 80–84, 2009.
[75]  W. Chen, W. Jin, N. Hardegen et al., “Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-β induction of transcription factor Foxp3,” Journal of Experimental Medicine, vol. 198, no. 12, pp. 1875–1886, 2003.
[76]  K. Kretschmer, I. Apostolou, D. Hawiger, K. Khazaie, M. C. Nussenzweig, and H. von Boehmer, “Inducing and expanding regulatory T cell populations by foreign antigen,” Nature Immunology, vol. 6, no. 12, pp. 1219–1227, 2005.
[77]  M. H. Kaplan, Y. L. Sun, T. Hoey, and M. J. Grusby, “Impaired IL-12 responses and enhanced development of Th2 cells in Stat4-deficient mice,” Nature, vol. 382, pp. 174–177, 1996.
[78]  R. A. Seder, R. Gazzinelli, A. Sher, and W. E. Paul, “Interleukin 12 acts directly on CD4+ T cells to enhance priming for interferon γ production and diminishes interleukin 4 inhibition of such priming,” Proceedings of the National Academy of Sciences of the United States of America, vol. 90, no. 21, pp. 10188–10192, 1993.
[79]  F. Cottrez and H. Groux, “Specialization in tolerance: innate CD4+CD25+ versus acquired Tr1 and Th3 regulatory T cells,” Transplantation, vol. 77, no. 1, Supplement, pp. S12–S15, 2004.
[80]  A. Skapenko, J. R. Kalden, P. E. Lipsky, and H. Schulze-Koops, “The IL-4 receptor alpha-chain-binding cytokines, IL-4 and IL-13, induce forkhead box P3-expressing CD25+CD4+ regulatory T cells from CD25-CD4+ precursors,” Journal of Immunology, vol. 175, pp. 6107–6116, 2005.
[81]  Y. Carrier, J. Yuan, V. K. Kuchroo, and H. L. Weiner, “Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-beta T cell-transgenic mice,” Journal of Immunology, vol. 178, pp. 179–185, 2007.
[82]  E. Maggi, L. Cosmi, F. Liotta, P. Romagnani, S. Romagnani, and F. Annunziato, “Thymic regulatory T cells,” Autoimmunity Reviews, vol. 4, no. 8, pp. 579–586, 2005.
[83]  L. S. Taams and A. N. Akbar, “Peripheral generation and function of CD4+CD25+ regulatory T cells,” Current Topics in Microbiology and Immunology, vol. 293, pp. 115–131, 2005.
[84]  E. M. Shevach, “Mechanisms of foxp3+ T regulatory cell-mediated suppression,” Immunity, vol. 30, pp. 636–645, 2009.
[85]  D. K. Sojka, Y. H. Huang, and D. J. Fowell, “Mechanisms of regulatory T-cell suppression—a diverse arsenal for a moving target,” Immunology, vol. 124, no. 1, pp. 13–22, 2008.
[86]  G. S. Whitehead, R. H. Wilson, K. Nakano, L. H. Burch, H. Nakano, and D. N. Cook, “IL-35 production by inducible costimulator (ICOS)-positive regulatory T cells reverses established IL-17-dependent allergic airways disease,” Journal of Allergy and Clinical Immunology, vol. 129, pp. 207–215, 2012.
[87]  L. W. Collison, C. J. Workman, T. T. Kuo et al., “The inhibitory cytokine IL-35 contributes to regulatory T-cell function,” Nature, vol. 450, pp. 566–569, 2007.
[88]  S. Yamagiwa, J. D. Gray, S. Hashimoto, and D. A. Horwitz, “A role for TGF-β in the generation and expansion of CD4+CD25+ regulatory t cells from human peripheral blood,” Journal of Immunology, vol. 166, no. 12, pp. 7282–7289, 2001.
[89]  M. Czystowska, L. Strauss, C. Bergmann, M. Szajnik, H. Rabinowich, and T. L. Whiteside, “Reciprocal granzyme/perforin-mediated death of human regulatory and responder T cells is regulated by interleukin-2 (IL-2),” Journal of Molecular Medicine, vol. 88, no. 6, pp. 577–588, 2010.
[90]  X. Cao, S. F. Cai, T. A. Fehniger et al., “Granzyme B and perforin are important for regulatory T cell-mediated suppression of tumor clearance,” Immunity, vol. 27, no. 4, pp. 635–646, 2007.
[91]  M. Vaeth, T. Gogishvili, T. Bopp et al., “Regulatory T cells facilitate the nuclear accumulation of inducible cAMP early repressor (ICER) and suppress nuclear factor of activated T cell c1 (NFATc1),” Proceedings of the National Academy of Sciences of the United States of America, vol. 108, no. 6, pp. 2480–2485, 2011.
[92]  X. Zhu, P. Yang, H. Zhou et al., “CD4+CD25+Tregs express an increased LAG-3 and CTLA-4 in anterior chamber-associated immune deviation,” Graefe's Archive for Clinical and Experimental Ophthalmology, vol. 245, pp. 1549–1557, 2007.
[93]  H. Liu, M. Komai-Koma, D. Xu, and F. Y. Liew, “Toll-like receptor 2 signaling modulates the functions of CD4+ CD25+ regulatory T cells,” Proceedings of the National Academy of Sciences of the United States of America, vol. 103, pp. 7048–7053, 2006.
[94]  A. Zanin-Zhorov, L. Cahalon, G. Tal, R. Margalit, O. Lider, and I. R. Cohen, “Heat shock protein 60 enhances CD4+CD25+ regulatory T cell function via innate TLR2 signaling,” Journal of Clinical Investigation, vol. 116, no. 7, pp. 2022–2032, 2006.
[95]  M. Sakaguchi, H. Kitahara, T. Seto et al., “Surgery for acute type A aortic dissection in pregnant patients with Marfan syndrome,” European Journal of Cardio-thoracic Surgery, vol. 28, no. 2, pp. 280–283, 2005.
[96]  J. A. Aalberse, B. Kapitein, S. de Roock et al., “Cord blood CD4+ T cells respond to self heat shock protein 60 (HSP60),” PloS One, vol. 6, article e24119, 2011.
[97]  G. H. M. Van Puijvelde, T. Van Es, E. J. A. Van Wanrooij et al., “Induction of oral tolerance to HSP60 or an HSP60-peptide activates t cell regulation and reduces atherosclerosis,” Arteriosclerosis, Thrombosis, and Vascular Biology, vol. 27, no. 12, pp. 2677–2683, 2007.
[98]  H. Li, Y. Ding, G. Yi, Q. Zeng, and W. Yang, “Establishment of nasal tolerance to heat shock protein-60 alleviates atherosclerosis by inducing TGF-beta-dependent regulatory T cells,” Journal of Huazhong University of Science and Technology, vol. 32, pp. 24–30, 2012.
[99]  R. Sutmuller, A. Garritsen, and G. J. Adema, “Regulatory T cells and toll–like receptors: regulating the regulators,” Annals of the Rheumatic Diseases, vol. 66, Supplement 3, pp. iii91–iii95, 2007.
[100]  E. F. de Zoeten, L. Wang, K. Butler et al., “Histone deacetylase 6 and heat shock protein 90 control the functions of Foxp3(+) T-regulatory cells,” Molecular and Cellular Biology, vol. 31, pp. 2066–2078, 2011.
[101]  J. Dai, B. Liu, S. M. Ngoi, S. Sun, A. T. Vella, and Z. Li, “TLR4 hyperresponsiveness via cell surface expression of heat shock protein gp96 potentiates suppressive function of regulatory T cells,” Journal of Immunology, vol. 178, no. 5, pp. 3219–3225, 2007.
[102]  I. Caramalho, T. Lopes-Carvalho, D. Ostler, S. Zelenay, M. Haury, and J. Demengeot, “Regulatory T cells selectively express toll-like receptors and are activated by lipopolysaccharide,” Journal of Experimental Medicine, vol. 197, no. 4, pp. 403–411, 2003.
[103]  M. J. van Herwijnen, L. Wieten, R. van der Zee et al., “Regulatory T cells that recognize a ubiquitous stress-inducible self-antigen are long-lived suppressors of autoimmune arthritis,” Proceedings of the National Academy of Sciences of the United States of America, vol. 109, pp. 14134–14139, 2012.
[104]  L. Wieten, S. E. Berlo, C. B. T. Brink et al., “IL-10 is critically involved in mycobacterial HSP70 induced suppression of proteoglycan-induced arthritis,” PLoS ONE, vol. 4, no. 1, article e4186, Article ID e4186, 2009.
[105]  T. J. Borges, B. N. Porto, C. A. Teixeira et al., “Prolonged survival of allografts induced by mycobacterial Hsp70 is dependent on CD4+CD25+ regulatory T cells,” PLoS ONE, vol. 5, no. 12, article e14264, Article ID e14264, 2010.
[106]  M. Jaattela, D. Wissing, K. Kokholm, T. Kallunki, and M. Egeblad, “Hsp70 exerts its anti-apoptotic function downstream of caspase-3-like proteases,” The EMBO Journal, vol. 17, pp. 6124–6134, 1998.
[107]  J. C. Young, I. Moarefi, and F. U. Hartl, “Hsp90: a specialized but essential protein-folding tool,” Journal of Cell Biology, vol. 154, no. 2, pp. 267–273, 2001.
[108]  S. Aoyagi and T. K. Archer, “Modulating molecular chaperone Hsp90 functions through reversible acetylation,” Trends in Cell Biology, vol. 15, no. 11, pp. 565–567, 2005.
[109]  H. Zhang, Y. Xiao, Z. Zhu, B. Li, and M. I. Greene, “Immune regulation by histone deacetylases: a focus on the alteration of FOXP3 activity,” Immunology & Cell Biology, vol. 90, pp. 95–100, 2012.
[110]  R. Tao, E. F. De Zoeten, E. Ozkaynak et al., “Deacetylase inhibition promotes the generation and function of regulatory T cells,” Nature Medicine, vol. 13, no. 11, pp. 1299–1307, 2007.
[111]  E. F. de Zoeten, L. Wang, H. Sai, W. H. Dillmann, and W. W. Hancock, “Inhibition of HDAC9 Increases T Regulatory Cell Function and Prevents Colitis in Mice,” Gastroenterology, vol. 138, no. 2, pp. 583–594, 2010.
[112]  Y. Xiao, B. Li, Z. Zhou, W. W. Hancock, H. Zhang, and M. I. Greene, “Histone acetyltransferase mediated regulation of FOXP3 acetylation and Treg function,” Current Opinion in Immunology, vol. 22, no. 5, pp. 583–591, 2010.
[113]  U. H. Beier, T. Akimova, Y. Liu, L. Wang, and W. W. Hancock, “Histone/protein deacetylases control Foxp3 expression and the heat shock response of T-regulatory cells,” Current Opinion in Immunology, vol. 23, pp. 670–678, 2011.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413