Skip to main content
Top
Published in: Arthritis Research & Therapy 3/2012

01-06-2012 | Review

Energy metabolism and rheumatic diseases: from cell to organism

Authors: Cornelia M Spies, Rainer H Straub, Frank Buttgereit

Published in: Arthritis Research & Therapy | Issue 3/2012

Login to get access

Abstract

In rheumatic and other chronic inflammatory diseases, high amounts of energy for the activated immune system have to be provided and allocated by energy metabolism. In recent time many new insights have been gained into the control of the immune response through metabolic signals. Activation of immune cells as well as reduced nutrient supply and hypoxia in inflamed tissues cause stimulation of glycolysis and other cellular metabolic pathways. However, persistent cellular metabolic signals can promote ongoing chronic inflammation and loss of immune tolerance. On the organism level, the neuroendocrine immune response of the hypothalamic-pituitary adrenal axis and sympathetic nervous system, which is meant to overcome a transient inflammatory episode, can lead to metabolic disease sequelae if chronically activated. We conclude that, on cellular and organism levels, a prolonged energy appeal reaction is an important factor of chronic inflammatory disease etiology.
Appendix
Available only for authorised users
Literature
1.
go back to reference Buttgereit F, Burmester GR, Brand MD: Bioenergetics of immune functions: fundamental and therapeutic aspects. Immunol Today. 2000, 21: 192-199.CrossRefPubMed Buttgereit F, Burmester GR, Brand MD: Bioenergetics of immune functions: fundamental and therapeutic aspects. Immunol Today. 2000, 21: 192-199.CrossRefPubMed
2.
go back to reference Straub RH, Cutolo M, Buttgereit F, Pongratz G: Energy regulation and neuroendocrine-immune control in chronic inflammatory diseases. J Intern Med. 2010, 267: 543-560. 10.1111/j.1365-2796.2010.02218.x.CrossRefPubMed Straub RH, Cutolo M, Buttgereit F, Pongratz G: Energy regulation and neuroendocrine-immune control in chronic inflammatory diseases. J Intern Med. 2010, 267: 543-560. 10.1111/j.1365-2796.2010.02218.x.CrossRefPubMed
4.
go back to reference Fox CJ, Hammerman PS, Thompson CB: Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol. 2005, 5: 844-852. 10.1038/nri1710.CrossRefPubMed Fox CJ, Hammerman PS, Thompson CB: Fuel feeds function: energy metabolism and the T-cell response. Nat Rev Immunol. 2005, 5: 844-852. 10.1038/nri1710.CrossRefPubMed
5.
go back to reference Mathis D, Shoelson SE: Immunometabolism: an emerging frontier. Nat Rev Immunol. 2011, 11: 81-83. 10.1038/nri2922.CrossRefPubMed Mathis D, Shoelson SE: Immunometabolism: an emerging frontier. Nat Rev Immunol. 2011, 11: 81-83. 10.1038/nri2922.CrossRefPubMed
7.
go back to reference Tannahill GM, O'Neill LA: The emerging role of metabolic regulation in the functioning of Toll-like receptors and the NOD-like receptor Nlrp3. FEBS Lett. 2011, 585: 1568-1572. 10.1016/j.febslet.2011.05.008.CrossRefPubMed Tannahill GM, O'Neill LA: The emerging role of metabolic regulation in the functioning of Toll-like receptors and the NOD-like receptor Nlrp3. FEBS Lett. 2011, 585: 1568-1572. 10.1016/j.febslet.2011.05.008.CrossRefPubMed
8.
go back to reference Inoki K, Kim J, Guan KL: AMPK and mTOR in cellular energy homeostasis and drug targets. Annu Rev Pharmacol Toxicol. 2012, 52: 381-400. 10.1146/annurev-pharmtox-010611-134537.CrossRefPubMed Inoki K, Kim J, Guan KL: AMPK and mTOR in cellular energy homeostasis and drug targets. Annu Rev Pharmacol Toxicol. 2012, 52: 381-400. 10.1146/annurev-pharmtox-010611-134537.CrossRefPubMed
9.
go back to reference Nutsch K, Hsieh C: When T cells run out of breath: the HIF-1α story. Cell. 2011, 146: 673-674. 10.1016/j.cell.2011.08.018.CrossRefPubMed Nutsch K, Hsieh C: When T cells run out of breath: the HIF-1α story. Cell. 2011, 146: 673-674. 10.1016/j.cell.2011.08.018.CrossRefPubMed
11.
go back to reference Procaccini C, Galgani M, De Rosa V, Matarese G: Intracellular metabolic pathways control immune tolerance. Trends Immunol. 2012, 33: 1-7. 10.1016/j.it.2011.09.002.CrossRefPubMed Procaccini C, Galgani M, De Rosa V, Matarese G: Intracellular metabolic pathways control immune tolerance. Trends Immunol. 2012, 33: 1-7. 10.1016/j.it.2011.09.002.CrossRefPubMed
12.
go back to reference Gatza E, Wahl DR, Opipari AW, Sundberg TB, Reddy P, Liu C, Glick GD, Ferrara JL: Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci Transl Med. 2011, 3: 67ra8-PubMedCentralCrossRefPubMed Gatza E, Wahl DR, Opipari AW, Sundberg TB, Reddy P, Liu C, Glick GD, Ferrara JL: Manipulating the bioenergetics of alloreactive T cells causes their selective apoptosis and arrests graft-versus-host disease. Sci Transl Med. 2011, 3: 67ra8-PubMedCentralCrossRefPubMed
13.
go back to reference Jones RG, Thompson CB: Revving the engine: signal transduction fuels T cell activation. Immunity. 2007, 27: 173-178. 10.1016/j.immuni.2007.07.008.CrossRefPubMed Jones RG, Thompson CB: Revving the engine: signal transduction fuels T cell activation. Immunity. 2007, 27: 173-178. 10.1016/j.immuni.2007.07.008.CrossRefPubMed
14.
go back to reference Vander Heiden MG, Cantley LC, Thompson CB: Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009, 324: 1029-1033. 10.1126/science.1160809.PubMedCentralCrossRefPubMed Vander Heiden MG, Cantley LC, Thompson CB: Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009, 324: 1029-1033. 10.1126/science.1160809.PubMedCentralCrossRefPubMed
15.
go back to reference Summers SA, Yin VP, Whiteman EL, Garza LA, Cho H, Tuttle RL, Birnbaum MJ: Signaling pathways mediating insulin-stimulated glucose transport. Ann N Y Acad Sci. 1999, 892: 169-186. 10.1111/j.1749-6632.1999.tb07795.x.CrossRefPubMed Summers SA, Yin VP, Whiteman EL, Garza LA, Cho H, Tuttle RL, Birnbaum MJ: Signaling pathways mediating insulin-stimulated glucose transport. Ann N Y Acad Sci. 1999, 892: 169-186. 10.1111/j.1749-6632.1999.tb07795.x.CrossRefPubMed
16.
go back to reference Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB: The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002, 16: 769-777. 10.1016/S1074-7613(02)00323-0.CrossRefPubMed Frauwirth KA, Riley JL, Harris MH, Parry RV, Rathmell JC, Plas DR, Elstrom RL, June CH, Thompson CB: The CD28 signaling pathway regulates glucose metabolism. Immunity. 2002, 16: 769-777. 10.1016/S1074-7613(02)00323-0.CrossRefPubMed
17.
go back to reference Wofford JA, Wieman HL, Jacobs SR, Zhao Y, Rathmell JC: IL-7 promotes Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support T-cell survival. Blood. 2008, 111: 2101-2111. 10.1182/blood-2007-06-096297.PubMedCentralCrossRefPubMed Wofford JA, Wieman HL, Jacobs SR, Zhao Y, Rathmell JC: IL-7 promotes Glut1 trafficking and glucose uptake via STAT5-mediated activation of Akt to support T-cell survival. Blood. 2008, 111: 2101-2111. 10.1182/blood-2007-06-096297.PubMedCentralCrossRefPubMed
18.
19.
go back to reference Gingras AC, Raught B, Sonenberg N: Regulation of translation initiation by FRAP/mTOR. Genes Dev. 2001, 15: 807-826. 10.1101/gad.887201.CrossRefPubMed Gingras AC, Raught B, Sonenberg N: Regulation of translation initiation by FRAP/mTOR. Genes Dev. 2001, 15: 807-826. 10.1101/gad.887201.CrossRefPubMed
20.
go back to reference Hay N, Sonenberg N: Upstream and downstream of mTOR. Genes Dev. 2004, 18: 1926-1945. 10.1101/gad.1212704.CrossRefPubMed Hay N, Sonenberg N: Upstream and downstream of mTOR. Genes Dev. 2004, 18: 1926-1945. 10.1101/gad.1212704.CrossRefPubMed
21.
go back to reference Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD: Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell. 2010, 39: 171-183. 10.1016/j.molcel.2010.06.022.PubMedCentralCrossRefPubMed Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL, Triantafellow E, Ma Q, Gorski R, Cleaver S, Vander Heiden MG, MacKeigan JP, Finan PM, Clish CB, Murphy LO, Manning BD: Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell. 2010, 39: 171-183. 10.1016/j.molcel.2010.06.022.PubMedCentralCrossRefPubMed
22.
go back to reference Ghillebert R, Swinnen E, Wen J, Vandesteene L, Ramon M, Norga K, Rolland F, Winderickx J: The AMPK/SNF1/SnRK1 fuel gauge and energy regulator: structure, function and regulation. FEBS J. 2011, 278: 3978-3990. 10.1111/j.1742-4658.2011.08315.x.CrossRefPubMed Ghillebert R, Swinnen E, Wen J, Vandesteene L, Ramon M, Norga K, Rolland F, Winderickx J: The AMPK/SNF1/SnRK1 fuel gauge and energy regulator: structure, function and regulation. FEBS J. 2011, 278: 3978-3990. 10.1111/j.1742-4658.2011.08315.x.CrossRefPubMed
23.
go back to reference Hardie DG: AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011, 25: 1895-1908. 10.1101/gad.17420111.PubMedCentralCrossRefPubMed Hardie DG: AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev. 2011, 25: 1895-1908. 10.1101/gad.17420111.PubMedCentralCrossRefPubMed
24.
go back to reference Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ: AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008, 30: 214-226. 10.1016/j.molcel.2008.03.003.PubMedCentralCrossRefPubMed Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ: AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell. 2008, 30: 214-226. 10.1016/j.molcel.2008.03.003.PubMedCentralCrossRefPubMed
26.
go back to reference Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, Thompson CB, Evans RM: AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science. 2009, 326: 437-440. 10.1126/science.1172156.PubMedCentralCrossRefPubMed Lamia KA, Sachdeva UM, DiTacchio L, Williams EC, Alvarez JG, Egan DF, Vasquez DS, Juguilon H, Panda S, Shaw RJ, Thompson CB, Evans RM: AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science. 2009, 326: 437-440. 10.1126/science.1172156.PubMedCentralCrossRefPubMed
27.
go back to reference Dziurla R, Gaber T, Fangradt M, Hahne M, Tripmacher R, Kolar P, Spies CM, Burmester GR, Buttgereit F: Effects of hypoxia and/or lack of glucose on cellular energy metabolism and cytokine production in stimulated human CD4+ T lymphocytes. Immunol Lett. 2010, 131: 97-105. 10.1016/j.imlet.2010.02.008.CrossRefPubMed Dziurla R, Gaber T, Fangradt M, Hahne M, Tripmacher R, Kolar P, Spies CM, Burmester GR, Buttgereit F: Effects of hypoxia and/or lack of glucose on cellular energy metabolism and cytokine production in stimulated human CD4+ T lymphocytes. Immunol Lett. 2010, 131: 97-105. 10.1016/j.imlet.2010.02.008.CrossRefPubMed
28.
go back to reference Gaber T, Dziurla R, Tripmacher R, Burmester GR, Buttgereit F: Hypoxia inducible factor (HIF) in rheumatology: low O2! See what HIF can do!. Ann Rheum Dis. 2005, 64: 971-980. 10.1136/ard.2004.031641.PubMedCentralCrossRefPubMed Gaber T, Dziurla R, Tripmacher R, Burmester GR, Buttgereit F: Hypoxia inducible factor (HIF) in rheumatology: low O2! See what HIF can do!. Ann Rheum Dis. 2005, 64: 971-980. 10.1136/ard.2004.031641.PubMedCentralCrossRefPubMed
29.
go back to reference Tripmacher R, Gaber T, Dziurla R, Haupl T, Erekul K, Grutzkau A, Tschirschmann M, Scheffold A, Radbruch A, Burmester GR, Buttgereit F: Human CD4+ T cells maintain specific functions even under conditions of extremely restricted ATP production. Eur J Immunol. 2008, 38: 1631-1642. 10.1002/eji.200738047.CrossRefPubMed Tripmacher R, Gaber T, Dziurla R, Haupl T, Erekul K, Grutzkau A, Tschirschmann M, Scheffold A, Radbruch A, Burmester GR, Buttgereit F: Human CD4+ T cells maintain specific functions even under conditions of extremely restricted ATP production. Eur J Immunol. 2008, 38: 1631-1642. 10.1002/eji.200738047.CrossRefPubMed
30.
go back to reference Falchuk KH, Goetzl EJ, Kulka JP: Respiratory gases of synovial fluids. An approach to synovial tissue circulatory-metabolic imbalance in rheumatoid arthritis. Am J Med. 1970, 49: 223-231. 10.1016/S0002-9343(70)80078-X.CrossRefPubMed Falchuk KH, Goetzl EJ, Kulka JP: Respiratory gases of synovial fluids. An approach to synovial tissue circulatory-metabolic imbalance in rheumatoid arthritis. Am J Med. 1970, 49: 223-231. 10.1016/S0002-9343(70)80078-X.CrossRefPubMed
31.
go back to reference Lund-Olesen K: Oxygen tension in synovial fluids. Arthritis Rheum. 1970, 13: 769-776. 10.1002/art.1780130606.CrossRefPubMed Lund-Olesen K: Oxygen tension in synovial fluids. Arthritis Rheum. 1970, 13: 769-776. 10.1002/art.1780130606.CrossRefPubMed
32.
go back to reference Sivakumar B, Akhavani MA, Winlove CP, Taylor PC, Paleolog EM, Kang N: Synovial hypoxia as a cause of tendon rupture in rheumatoid arthritis. J Hand Surg Am. 2008, 33: 49-58. 10.1016/j.jhsa.2007.09.002.CrossRefPubMed Sivakumar B, Akhavani MA, Winlove CP, Taylor PC, Paleolog EM, Kang N: Synovial hypoxia as a cause of tendon rupture in rheumatoid arthritis. J Hand Surg Am. 2008, 33: 49-58. 10.1016/j.jhsa.2007.09.002.CrossRefPubMed
33.
go back to reference Ng CT, Biniecka M, Kennedy A, McCormick J, Fitzgerald O, Bresnihan B, Buggy D, Taylor CT, O'Sullivan J, Fearon U, Veale DJ: Synovial tissue hypoxia and inflammation in vivo. Ann Rheum Dis. 2010, 69: 1389-1395. 10.1136/ard.2009.119776.PubMedCentralCrossRefPubMed Ng CT, Biniecka M, Kennedy A, McCormick J, Fitzgerald O, Bresnihan B, Buggy D, Taylor CT, O'Sullivan J, Fearon U, Veale DJ: Synovial tissue hypoxia and inflammation in vivo. Ann Rheum Dis. 2010, 69: 1389-1395. 10.1136/ard.2009.119776.PubMedCentralCrossRefPubMed
34.
go back to reference Kennedy A, Ng CT, Chang TC, Biniecka M, O'Sullivan JN, Heffernan E, Fearon U, Veale DJ: Tumor necrosis factor blocking therapy alters joint inflammation and hypoxia. Arthritis Rheum. 2011, 63: 923-932. 10.1002/art.30221.CrossRefPubMed Kennedy A, Ng CT, Chang TC, Biniecka M, O'Sullivan JN, Heffernan E, Fearon U, Veale DJ: Tumor necrosis factor blocking therapy alters joint inflammation and hypoxia. Arthritis Rheum. 2011, 63: 923-932. 10.1002/art.30221.CrossRefPubMed
35.
go back to reference Wang GL, Semenza GL: Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J Biol Chem. 1993, 268: 21513-21518.PubMed Wang GL, Semenza GL: Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J Biol Chem. 1993, 268: 21513-21518.PubMed
36.
go back to reference Gaber T, Schellmann S, Erekul KB, Fangradt M, Tykwinska K, Hahne M, Maschmeyer P, Wagegg M, Stahn C, Kolar P, Dziurla R, Lohning M, Burmester GR, Buttgereit F: Macrophage migration inhibitory factor counterregulates dexamethasone-mediated suppression of hypoxia-inducible factor-1 alpha function and differentially influences human CD4+ T cell proliferation under hypoxia. J Immunol. 2011, 186: 764-774. 10.4049/jimmunol.0903421.CrossRefPubMed Gaber T, Schellmann S, Erekul KB, Fangradt M, Tykwinska K, Hahne M, Maschmeyer P, Wagegg M, Stahn C, Kolar P, Dziurla R, Lohning M, Burmester GR, Buttgereit F: Macrophage migration inhibitory factor counterregulates dexamethasone-mediated suppression of hypoxia-inducible factor-1 alpha function and differentially influences human CD4+ T cell proliferation under hypoxia. J Immunol. 2011, 186: 764-774. 10.4049/jimmunol.0903421.CrossRefPubMed
37.
go back to reference Nakamura H, Makino Y, Okamoto K, Poellinger L, Ohnuma K, Morimoto C, Tanaka H: TCR engagement increases hypoxia-inducible factor-1 alpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J Immunol. 2005, 174: 7592-7599.CrossRefPubMed Nakamura H, Makino Y, Okamoto K, Poellinger L, Ohnuma K, Morimoto C, Tanaka H: TCR engagement increases hypoxia-inducible factor-1 alpha protein synthesis via rapamycin-sensitive pathway under hypoxic conditions in human peripheral T cells. J Immunol. 2005, 174: 7592-7599.CrossRefPubMed
38.
go back to reference Hollander AP, Corke KP, Freemont AJ, Lewis CE: Expression of hypoxia-inducible factor 1α by macrophages in the rheumatoid synovium: implications for targeting of therapeutic genes to the inflamed joint. Arthritis Rheum. 2001, 44: 1540-1544. 10.1002/1529-0131(200107)44:7<1540::AID-ART277>3.0.CO;2-7.CrossRefPubMed Hollander AP, Corke KP, Freemont AJ, Lewis CE: Expression of hypoxia-inducible factor 1α by macrophages in the rheumatoid synovium: implications for targeting of therapeutic genes to the inflamed joint. Arthritis Rheum. 2001, 44: 1540-1544. 10.1002/1529-0131(200107)44:7<1540::AID-ART277>3.0.CO;2-7.CrossRefPubMed
39.
go back to reference Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, Haase VH, Jaenisch R, Corr M, Nizet V, Firestein GS, Gerber HP, Ferrara N, Johnson RS: HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 2003, 112: 645-657. 10.1016/S0092-8674(03)00154-5.PubMedCentralCrossRefPubMed Cramer T, Yamanishi Y, Clausen BE, Forster I, Pawlinski R, Mackman N, Haase VH, Jaenisch R, Corr M, Nizet V, Firestein GS, Gerber HP, Ferrara N, Johnson RS: HIF-1alpha is essential for myeloid cell-mediated inflammation. Cell. 2003, 112: 645-657. 10.1016/S0092-8674(03)00154-5.PubMedCentralCrossRefPubMed
40.
go back to reference van Hal TW, van Bon L, Radstake TR: A system out of breath: how hypoxia possibly contributes to the pathogenesis of systemic sclerosis. Int J Rheumatol. 2011, 2011: 824972-PubMedCentralCrossRefPubMed van Hal TW, van Bon L, Radstake TR: A system out of breath: how hypoxia possibly contributes to the pathogenesis of systemic sclerosis. Int J Rheumatol. 2011, 2011: 824972-PubMedCentralCrossRefPubMed
41.
go back to reference Beyer C, Schett G, Gay S, Distler O, Distler JH: Hypoxia. Hypoxia in the pathogenesis of systemic sclerosis. Arthritis Res Ther. 2009, 11: 220-10.1186/ar2598.PubMedCentralCrossRefPubMed Beyer C, Schett G, Gay S, Distler O, Distler JH: Hypoxia. Hypoxia in the pathogenesis of systemic sclerosis. Arthritis Res Ther. 2009, 11: 220-10.1186/ar2598.PubMedCentralCrossRefPubMed
42.
go back to reference Distler O, Distler JH, Scheid A, Acker T, Hirth A, Rethage J, Michel BA, Gay RE, Muller-Ladner U, Matucci-Cerinic M, Plate KH, Gassmann M, Gay S: Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res. 2004, 95: 109-116. 10.1161/01.RES.0000134644.89917.96.CrossRefPubMed Distler O, Distler JH, Scheid A, Acker T, Hirth A, Rethage J, Michel BA, Gay RE, Muller-Ladner U, Matucci-Cerinic M, Plate KH, Gassmann M, Gay S: Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res. 2004, 95: 109-116. 10.1161/01.RES.0000134644.89917.96.CrossRefPubMed
43.
go back to reference Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD: The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol. 2011, 12: 295-303.PubMedCentralCrossRefPubMed Delgoffe GM, Pollizzi KN, Waickman AT, Heikamp E, Meyers DJ, Horton MR, Xiao B, Worley PF, Powell JD: The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat Immunol. 2011, 12: 295-303.PubMedCentralCrossRefPubMed
44.
go back to reference Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG, Rathmell JC: Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011, 186: 3299-3303. 10.4049/jimmunol.1003613.PubMedCentralCrossRefPubMed Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG, Rathmell JC: Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol. 2011, 186: 3299-3303. 10.4049/jimmunol.1003613.PubMedCentralCrossRefPubMed
45.
go back to reference Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z, Fu J, Kim Y, Yen HR, Luo W, Zeller K, Shimoda L, Topalian SL, Semenza GL, Dang CV, Pardoll DM, Pan F: Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell. 2011, 146: 772-784. 10.1016/j.cell.2011.07.033.PubMedCentralCrossRefPubMed Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z, Fu J, Kim Y, Yen HR, Luo W, Zeller K, Shimoda L, Topalian SL, Semenza GL, Dang CV, Pardoll DM, Pan F: Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell. 2011, 146: 772-784. 10.1016/j.cell.2011.07.033.PubMedCentralCrossRefPubMed
46.
go back to reference Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, Chi H: HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011, 208: 1367-1376. 10.1084/jem.20110278.PubMedCentralCrossRefPubMed Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, Chi H: HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J Exp Med. 2011, 208: 1367-1376. 10.1084/jem.20110278.PubMedCentralCrossRefPubMed
47.
go back to reference Moran EM, Heydrich R, Ng CT, Saber TP, McCormick J, Sieper J, Appel H, Fearon U, Veale DJ: IL-17A expression is localised to both mononuclear and polymorphonuclear synovial cell infiltrates. PLoS One. 2011, 6: e24048-10.1371/journal.pone.0024048.PubMedCentralCrossRefPubMed Moran EM, Heydrich R, Ng CT, Saber TP, McCormick J, Sieper J, Appel H, Fearon U, Veale DJ: IL-17A expression is localised to both mononuclear and polymorphonuclear synovial cell infiltrates. PLoS One. 2011, 6: e24048-10.1371/journal.pone.0024048.PubMedCentralCrossRefPubMed
48.
go back to reference Sitkovsky MV, Kjaergaard J, Lukashev D, Ohta A: Hypoxia-adenosinergic immunosuppression: tumor protection by T regulatory cells and cancerous tissue hypoxia. Clin Cancer Res. 2008, 14: 5947-5952. 10.1158/1078-0432.CCR-08-0229.CrossRefPubMed Sitkovsky MV, Kjaergaard J, Lukashev D, Ohta A: Hypoxia-adenosinergic immunosuppression: tumor protection by T regulatory cells and cancerous tissue hypoxia. Clin Cancer Res. 2008, 14: 5947-5952. 10.1158/1078-0432.CCR-08-0229.CrossRefPubMed
49.
go back to reference Chang X, Wei C: Glycolysis and rheumatoid arthritis. Int J Rheum Dis. 2011, 14: 217-222. 10.1111/j.1756-185X.2011.01598.x.CrossRefPubMed Chang X, Wei C: Glycolysis and rheumatoid arthritis. Int J Rheum Dis. 2011, 14: 217-222. 10.1111/j.1756-185X.2011.01598.x.CrossRefPubMed
50.
go back to reference Henderson B, Bitensky L, Chayen J: Glycolytic activity in human synovial lining cells in rheumatoid arthritis. Ann Rheum Dis. 1979, 38: 63-67. 10.1136/ard.38.1.63.PubMedCentralCrossRefPubMed Henderson B, Bitensky L, Chayen J: Glycolytic activity in human synovial lining cells in rheumatoid arthritis. Ann Rheum Dis. 1979, 38: 63-67. 10.1136/ard.38.1.63.PubMedCentralCrossRefPubMed
51.
go back to reference Matsumoto I, Lee DM, Goldbach-Mansky R, Sumida T, Hitchon CA, Schur PH, Anderson RJ, Coblyn JS, Weinblatt ME, Brenner M, Duclos B, Pasquali JL, El-Gabalawy H, Mathis D, Benoist C: Low prevalence of antibodies to glucose-6-phosphate isomerase in patients with rheumatoid arthritis and a spectrum of other chronic autoimmune disorders. Arthritis Rheum. 2003, 48: 944-954. 10.1002/art.10898.CrossRefPubMed Matsumoto I, Lee DM, Goldbach-Mansky R, Sumida T, Hitchon CA, Schur PH, Anderson RJ, Coblyn JS, Weinblatt ME, Brenner M, Duclos B, Pasquali JL, El-Gabalawy H, Mathis D, Benoist C: Low prevalence of antibodies to glucose-6-phosphate isomerase in patients with rheumatoid arthritis and a spectrum of other chronic autoimmune disorders. Arthritis Rheum. 2003, 48: 944-954. 10.1002/art.10898.CrossRefPubMed
52.
go back to reference Matsumoto I, Staub A, Benoist C, Mathis D: Arthritis provoked by linked T and B cell recognition of a glycolytic enzyme. Science. 1999, 286: 1732-1735. 10.1126/science.286.5445.1732.CrossRefPubMed Matsumoto I, Staub A, Benoist C, Mathis D: Arthritis provoked by linked T and B cell recognition of a glycolytic enzyme. Science. 1999, 286: 1732-1735. 10.1126/science.286.5445.1732.CrossRefPubMed
53.
go back to reference Fernandez D, Bonilla E, Mirza N, Niland B, Perl A: Rapamycin reduces disease activity and normalizes T cell activation-induced calcium fluxing in patients with systemic lupus erythematosus. Arthritis Rheum. 2006, 54: 2983-2988. 10.1002/art.22085.PubMedCentralCrossRefPubMed Fernandez D, Bonilla E, Mirza N, Niland B, Perl A: Rapamycin reduces disease activity and normalizes T cell activation-induced calcium fluxing in patients with systemic lupus erythematosus. Arthritis Rheum. 2006, 54: 2983-2988. 10.1002/art.22085.PubMedCentralCrossRefPubMed
54.
go back to reference Warner LM, Adams LM, Sehgal SN: Rapamycin prolongs survival and arrests pathophysiologic changes in murine systemic lupus erythematosus. Arthritis Rheum. 1994, 37: 289-297. 10.1002/art.1780370219.CrossRefPubMed Warner LM, Adams LM, Sehgal SN: Rapamycin prolongs survival and arrests pathophysiologic changes in murine systemic lupus erythematosus. Arthritis Rheum. 1994, 37: 289-297. 10.1002/art.1780370219.CrossRefPubMed
55.
go back to reference Fernandez D, Perl A: mTOR signaling: a central pathway to pathogenesis in systemic lupus erythematosus?. Discov Med. 2010, 9: 173-178.PubMedCentralPubMed Fernandez D, Perl A: mTOR signaling: a central pathway to pathogenesis in systemic lupus erythematosus?. Discov Med. 2010, 9: 173-178.PubMedCentralPubMed
56.
go back to reference Yoshizaki A, Yanaba K, Iwata Y, Komura K, Ogawa F, Takenaka M, Shimizu K, Asano Y, Hasegawa M, Fujimoto M, Sato S: Treatment with rapamycin prevents fibrosis in tight-skin and bleomycin-induced mouse models of systemic sclerosis. Arthritis Rheum. 2010, 62: 2476-2487. 10.1002/art.27498.CrossRefPubMed Yoshizaki A, Yanaba K, Iwata Y, Komura K, Ogawa F, Takenaka M, Shimizu K, Asano Y, Hasegawa M, Fujimoto M, Sato S: Treatment with rapamycin prevents fibrosis in tight-skin and bleomycin-induced mouse models of systemic sclerosis. Arthritis Rheum. 2010, 62: 2476-2487. 10.1002/art.27498.CrossRefPubMed
57.
go back to reference Su TI, Khanna D, Furst DE, Danovitch G, Burger C, Maranian P, Clements PJ: Rapamycin versus methotrexate in early diffuse systemic sclerosis: results from a randomized, single-blind pilot study. Arthritis Rheum. 2009, 60: 3821-3830. 10.1002/art.24986.PubMedCentralCrossRefPubMed Su TI, Khanna D, Furst DE, Danovitch G, Burger C, Maranian P, Clements PJ: Rapamycin versus methotrexate in early diffuse systemic sclerosis: results from a randomized, single-blind pilot study. Arthritis Rheum. 2009, 60: 3821-3830. 10.1002/art.24986.PubMedCentralCrossRefPubMed
58.
go back to reference Qu Y, Zhang B, Zhao L, Liu G, Ma H, Rao E, Zeng C, Zhao Y: The effect of immunosuppressive drug rapamycin on regulatory CD4+CD25+Foxp3+ T cells in mice. Transpl Immunol. 2007, 17: 153-161. 10.1016/j.trim.2007.01.002.CrossRefPubMed Qu Y, Zhang B, Zhao L, Liu G, Ma H, Rao E, Zeng C, Zhao Y: The effect of immunosuppressive drug rapamycin on regulatory CD4+CD25+Foxp3+ T cells in mice. Transpl Immunol. 2007, 17: 153-161. 10.1016/j.trim.2007.01.002.CrossRefPubMed
59.
go back to reference Ogino H, Nakamura K, Iwasa T, Ihara E, Akiho H, Motomura Y, Akahoshi K, Igarashi H, Kato M, Kotoh K, Ito T, Takayanagi R: Regulatory T cells expanded by rapamycin in vitro suppress colitis in an experimental mouse model. J Gastroenterol. 2011, 47: 366-376.CrossRefPubMed Ogino H, Nakamura K, Iwasa T, Ihara E, Akiho H, Motomura Y, Akahoshi K, Igarashi H, Kato M, Kotoh K, Ito T, Takayanagi R: Regulatory T cells expanded by rapamycin in vitro suppress colitis in an experimental mouse model. J Gastroenterol. 2011, 47: 366-376.CrossRefPubMed
60.
go back to reference Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R: mTOR regulates memory CD8 T-cell differentiation. Nature. 2009, 460: 108-112. 10.1038/nature08155.PubMedCentralCrossRefPubMed Araki K, Turner AP, Shaffer VO, Gangappa S, Keller SA, Bachmann MF, Larsen CP, Ahmed R: mTOR regulates memory CD8 T-cell differentiation. Nature. 2009, 460: 108-112. 10.1038/nature08155.PubMedCentralCrossRefPubMed
61.
go back to reference Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y: Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009, 460: 103-107. 10.1038/nature08097.PubMedCentralCrossRefPubMed Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang LS, Jones RG, Choi Y: Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009, 460: 103-107. 10.1038/nature08097.PubMedCentralCrossRefPubMed
62.
go back to reference Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, Cross JR, Jung E, Thompson CB, Jones RG, Pearce EJ: Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010, 115: 4742-4749. 10.1182/blood-2009-10-249540.PubMedCentralCrossRefPubMed Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, Cross JR, Jung E, Thompson CB, Jones RG, Pearce EJ: Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010, 115: 4742-4749. 10.1182/blood-2009-10-249540.PubMedCentralCrossRefPubMed
63.
go back to reference Kim SY, Choi YJ, Joung SM, Lee BH, Jung YS, Lee JY: Hypoxic stress up-regulates the expression of Toll-like receptor 4 in macrophages via hypoxia-inducible factor. Immunology. 2010, 129: 516-524. 10.1111/j.1365-2567.2009.03203.x.PubMedCentralCrossRefPubMed Kim SY, Choi YJ, Joung SM, Lee BH, Jung YS, Lee JY: Hypoxic stress up-regulates the expression of Toll-like receptor 4 in macrophages via hypoxia-inducible factor. Immunology. 2010, 129: 516-524. 10.1111/j.1365-2567.2009.03203.x.PubMedCentralCrossRefPubMed
64.
go back to reference Rodriguez-Prados JC, Traves PG, Cuenca J, Rico D, Aragones J, Martin-Sanz P, Cascante M, Bosca L: Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol. 2010, 185: 605-614. 10.4049/jimmunol.0901698.CrossRefPubMed Rodriguez-Prados JC, Traves PG, Cuenca J, Rico D, Aragones J, Martin-Sanz P, Cascante M, Bosca L: Substrate fate in activated macrophages: a comparison between innate, classic, and alternative activation. J Immunol. 2010, 185: 605-614. 10.4049/jimmunol.0901698.CrossRefPubMed
65.
go back to reference Garedew A, Moncada S: Mitochondrial dysfunction and HIF1alpha stabilization in inflammation. J Cell Sci. 2008, 121: 3468-3475. 10.1242/jcs.034660.CrossRefPubMed Garedew A, Moncada S: Mitochondrial dysfunction and HIF1alpha stabilization in inflammation. J Cell Sci. 2008, 121: 3468-3475. 10.1242/jcs.034660.CrossRefPubMed
66.
go back to reference Roiniotis J, Dinh H, Masendycz P, Turner A, Elsegood CL, Scholz GM, Hamilton JA: Hypoxia prolongs monocyte/macrophage survival and enhanced glycolysis is associated with their maturation under aerobic conditions. J Immunol. 2009, 182: 7974-7981. 10.4049/jimmunol.0804216.CrossRefPubMed Roiniotis J, Dinh H, Masendycz P, Turner A, Elsegood CL, Scholz GM, Hamilton JA: Hypoxia prolongs monocyte/macrophage survival and enhanced glycolysis is associated with their maturation under aerobic conditions. J Immunol. 2009, 182: 7974-7981. 10.4049/jimmunol.0804216.CrossRefPubMed
67.
go back to reference Hannah S, Mecklenburgh K, Rahman I, Bellingan GJ, Greening A, Haslett C, Chilvers ER: Hypoxia prolongs neutrophil survival in vitro. FEBS Lett. 1995, 372: 233-237. 10.1016/0014-5793(95)00986-J.CrossRefPubMed Hannah S, Mecklenburgh K, Rahman I, Bellingan GJ, Greening A, Haslett C, Chilvers ER: Hypoxia prolongs neutrophil survival in vitro. FEBS Lett. 1995, 372: 233-237. 10.1016/0014-5793(95)00986-J.CrossRefPubMed
68.
go back to reference Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, Sobolewski A, Condliffe AM, Cowburn AS, Johnson N, Chilvers ER: Hypoxia-induced neutrophil survival is mediated by HIF-1α-dependent NF-κB activity. J Exp Med. 2005, 201: 105-115. 10.1084/jem.20040624.PubMedCentralCrossRefPubMed Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, Sobolewski A, Condliffe AM, Cowburn AS, Johnson N, Chilvers ER: Hypoxia-induced neutrophil survival is mediated by HIF-1α-dependent NF-κB activity. J Exp Med. 2005, 201: 105-115. 10.1084/jem.20040624.PubMedCentralCrossRefPubMed
69.
go back to reference Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM, Dixit VD: The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011, 17: 179-188. 10.1038/nm.2279.PubMedCentralCrossRefPubMed Vandanmagsar B, Youm YH, Ravussin A, Galgani JE, Stadler K, Mynatt RL, Ravussin E, Stephens JM, Dixit VD: The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat Med. 2011, 17: 179-188. 10.1038/nm.2279.PubMedCentralCrossRefPubMed
70.
go back to reference Gergely P, Grossman C, Niland B, Puskas F, Neupane H, Allam F, Banki K, Phillips PE, Perl A: Mitochondrial hyperpolarization and ATP depletion in patients with systemic lupus erythematosus. Arthritis Rheum. 2002, 46: 175-190. 10.1002/1529-0131(200201)46:1<175::AID-ART10015>3.0.CO;2-H.PubMedCentralCrossRefPubMed Gergely P, Grossman C, Niland B, Puskas F, Neupane H, Allam F, Banki K, Phillips PE, Perl A: Mitochondrial hyperpolarization and ATP depletion in patients with systemic lupus erythematosus. Arthritis Rheum. 2002, 46: 175-190. 10.1002/1529-0131(200201)46:1<175::AID-ART10015>3.0.CO;2-H.PubMedCentralCrossRefPubMed
71.
go back to reference Pedersen-Lane JH, Zurier RB, Lawrence DA: Analysis of the thiol status of peripheral blood leukocytes in rheumatoid arthritis patients. J Leukoc Biol. 2007, 81: 934-941. 10.1189/jlb.0806533.CrossRefPubMed Pedersen-Lane JH, Zurier RB, Lawrence DA: Analysis of the thiol status of peripheral blood leukocytes in rheumatoid arthritis patients. J Leukoc Biol. 2007, 81: 934-941. 10.1189/jlb.0806533.CrossRefPubMed
72.
go back to reference Shah D, Wanchu A, Bhatnagar A: Interaction between oxidative stress and chemokines: possible pathogenic role in systemic lupus erythematosus and rheumatoid arthritis. Immunobiology. 2011, 216: 1010-1017. 10.1016/j.imbio.2011.04.001.CrossRefPubMed Shah D, Wanchu A, Bhatnagar A: Interaction between oxidative stress and chemokines: possible pathogenic role in systemic lupus erythematosus and rheumatoid arthritis. Immunobiology. 2011, 216: 1010-1017. 10.1016/j.imbio.2011.04.001.CrossRefPubMed
73.
go back to reference Pedersen BK: Exercise-induced myokines and their role in chronic diseases. Brain Behav Immun. 2011, 25: 811-816. 10.1016/j.bbi.2011.02.010.CrossRefPubMed Pedersen BK: Exercise-induced myokines and their role in chronic diseases. Brain Behav Immun. 2011, 25: 811-816. 10.1016/j.bbi.2011.02.010.CrossRefPubMed
74.
go back to reference Straub RH, Besedovsky HO: Integrated evolutionary, immunological, and neuroendocrine framework for the pathogenesis of chronic disabling inflammatory diseases. FASEB J. 2003, 17: 2176-2183. 10.1096/fj.03-0433hyp.CrossRefPubMed Straub RH, Besedovsky HO: Integrated evolutionary, immunological, and neuroendocrine framework for the pathogenesis of chronic disabling inflammatory diseases. FASEB J. 2003, 17: 2176-2183. 10.1096/fj.03-0433hyp.CrossRefPubMed
75.
go back to reference Williams GC: Pleiotropy, natural selection, and the evolution of senescence. Evolution. 1957, 11: 398-411. 10.2307/2406060.CrossRef Williams GC: Pleiotropy, natural selection, and the evolution of senescence. Evolution. 1957, 11: 398-411. 10.2307/2406060.CrossRef
76.
go back to reference Straub RH: [Neuroendocrine immunology: new pathogenetic aspects and clinical application]. Z Rheumatol. 2011, 70: 767-774. 10.1007/s00393-011-0784-8.CrossRefPubMed Straub RH: [Neuroendocrine immunology: new pathogenetic aspects and clinical application]. Z Rheumatol. 2011, 70: 767-774. 10.1007/s00393-011-0784-8.CrossRefPubMed
77.
go back to reference Cooper MS, Stewart PM: Corticosteroid insufficiency in acutely ill patients. N Engl J Med. 2003, 348: 727-734. 10.1056/NEJMra020529.CrossRefPubMed Cooper MS, Stewart PM: Corticosteroid insufficiency in acutely ill patients. N Engl J Med. 2003, 348: 727-734. 10.1056/NEJMra020529.CrossRefPubMed
78.
go back to reference Straub RH: Concepts of evolutionary medicine and energy regulation contribute to the etiology of systemic chronic inflammatory diseases. Brain Behav Immun. 2011, 25: 1-5. 10.1016/j.bbi.2010.08.002.CrossRefPubMed Straub RH: Concepts of evolutionary medicine and energy regulation contribute to the etiology of systemic chronic inflammatory diseases. Brain Behav Immun. 2011, 25: 1-5. 10.1016/j.bbi.2010.08.002.CrossRefPubMed
79.
go back to reference Buttgereit F, Brand MD, Muller M: ConA induced changes in energy metabolism of rat thymocytes. Biosci Rep. 1992, 12: 381-386. 10.1007/BF01121501.CrossRefPubMed Buttgereit F, Brand MD, Muller M: ConA induced changes in energy metabolism of rat thymocytes. Biosci Rep. 1992, 12: 381-386. 10.1007/BF01121501.CrossRefPubMed
80.
go back to reference Princiotta MF, Finzi D, Qian SB, Gibbs J, Schuchmann S, Buttgereit F, Bennink JR, Yewdell JW: Quantitating protein synthesis, degradation, and endogenous antigen processing. Immunity. 2003, 18: 343-354. 10.1016/S1074-7613(03)00051-7.CrossRefPubMed Princiotta MF, Finzi D, Qian SB, Gibbs J, Schuchmann S, Buttgereit F, Bennink JR, Yewdell JW: Quantitating protein synthesis, degradation, and endogenous antigen processing. Immunity. 2003, 18: 343-354. 10.1016/S1074-7613(03)00051-7.CrossRefPubMed
81.
go back to reference Maravillas-Montero JL, Santos-Argumedo L: The myosin family: unconventional roles of actin-dependent molecular motors in immune cells. J Leukoc Biol. 2011, 91: 35-46.CrossRefPubMed Maravillas-Montero JL, Santos-Argumedo L: The myosin family: unconventional roles of actin-dependent molecular motors in immune cells. J Leukoc Biol. 2011, 91: 35-46.CrossRefPubMed
82.
83.
go back to reference Sauer RT, Baker TA: AAA+ proteases: ATP-fueled machines of protein destruction. Annu Rev Biochem. 2011, 80: 587-612. 10.1146/annurev-biochem-060408-172623.CrossRefPubMed Sauer RT, Baker TA: AAA+ proteases: ATP-fueled machines of protein destruction. Annu Rev Biochem. 2011, 80: 587-612. 10.1146/annurev-biochem-060408-172623.CrossRefPubMed
84.
go back to reference Procko E, Gaudet R: Antigen processing and presentation: TAPping into ABC transporters. Curr Opin Immunol. 2009, 21: 84-91. 10.1016/j.coi.2009.02.003.CrossRefPubMed Procko E, Gaudet R: Antigen processing and presentation: TAPping into ABC transporters. Curr Opin Immunol. 2009, 21: 84-91. 10.1016/j.coi.2009.02.003.CrossRefPubMed
85.
go back to reference Authier F, Posner BI, Bergeron JJ: Endosomal proteolysis of internalized proteins. FEBS Lett. 1996, 389: 55-60. 10.1016/0014-5793(96)00368-7.CrossRefPubMed Authier F, Posner BI, Bergeron JJ: Endosomal proteolysis of internalized proteins. FEBS Lett. 1996, 389: 55-60. 10.1016/0014-5793(96)00368-7.CrossRefPubMed
86.
go back to reference Togashi K, Kataoka T, Nagai K: Characterization of a series of vacuolar type H+-ATPase inhibitors on CTL-mediated cytotoxicity. Immunol Lett. 1997, 55: 139-144. 10.1016/S0165-2478(97)02698-9.CrossRefPubMed Togashi K, Kataoka T, Nagai K: Characterization of a series of vacuolar type H+-ATPase inhibitors on CTL-mediated cytotoxicity. Immunol Lett. 1997, 55: 139-144. 10.1016/S0165-2478(97)02698-9.CrossRefPubMed
87.
go back to reference White C, Lee J, Kambe T, Fritsche K, Petris MJ: A role for the ATP7A copper-transporting ATPase in macrophage bactericidal activity. J Biol Chem. 2009, 284: 33949-33956. 10.1074/jbc.M109.070201.PubMedCentralCrossRefPubMed White C, Lee J, Kambe T, Fritsche K, Petris MJ: A role for the ATP7A copper-transporting ATPase in macrophage bactericidal activity. J Biol Chem. 2009, 284: 33949-33956. 10.1074/jbc.M109.070201.PubMedCentralCrossRefPubMed
88.
go back to reference Marques-da-Silva C, Chaves MM, Rodrigues JC, Corte-Real S, Coutinho-Silva R, Persechini PM: Differential modulation of ATP-induced P2X7-associated permeabilities to cations and anions of macrophages by infection with Leishmania amazonensis. PLoS One. 2011, 6: e25356-10.1371/journal.pone.0025356.PubMedCentralCrossRefPubMed Marques-da-Silva C, Chaves MM, Rodrigues JC, Corte-Real S, Coutinho-Silva R, Persechini PM: Differential modulation of ATP-induced P2X7-associated permeabilities to cations and anions of macrophages by infection with Leishmania amazonensis. PLoS One. 2011, 6: e25356-10.1371/journal.pone.0025356.PubMedCentralCrossRefPubMed
89.
go back to reference Schenk U, Frascoli M, Proietti M, Geffers R, Traggiai E, Buer J, Ricordi C, Westendorf AM, Grassi F: ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci Signal. 2011, 4: ra12-10.1126/scisignal.2001270.CrossRefPubMed Schenk U, Frascoli M, Proietti M, Geffers R, Traggiai E, Buer J, Ricordi C, Westendorf AM, Grassi F: ATP inhibits the generation and function of regulatory T cells through the activation of purinergic P2X receptors. Sci Signal. 2011, 4: ra12-10.1126/scisignal.2001270.CrossRefPubMed
90.
go back to reference Straub RH: Evolutionary medicine and chronic inflammatory state-known and new concepts in pathophysiology. J Mol Med (Berl). 2012, 90: 523-534. 10.1007/s00109-012-0861-8.CrossRef Straub RH: Evolutionary medicine and chronic inflammatory state-known and new concepts in pathophysiology. J Mol Med (Berl). 2012, 90: 523-534. 10.1007/s00109-012-0861-8.CrossRef
Metadata
Title
Energy metabolism and rheumatic diseases: from cell to organism
Authors
Cornelia M Spies
Rainer H Straub
Frank Buttgereit
Publication date
01-06-2012
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 3/2012
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/ar3885

Other articles of this Issue 3/2012

Arthritis Research & Therapy 3/2012 Go to the issue