Heat shock proteins and chronic fatigue in primary Sjögren's syndrome

Kjetil Bårdsen, Mari Mæland Nilsen, Jan Terje Kvaløy, Katrine Brække Norheim, Grete Jonsson, Roald Omdal, Kjetil Bårdsen, Mari Mæland Nilsen, Jan Terje Kvaløy, Katrine Brække Norheim, Grete Jonsson, Roald Omdal

Abstract

Fatigue occurs frequently in patients with cancer, neurological diseases and chronic inflammatory diseases, but the biological mechanisms that lead to and regulate fatigue are largely unknown. When the innate immune system is activated, heat shock proteins (HSPs) are produced to protect cells. Some extracellular HSPs appear to recognize cellular targets in the brain, and we hypothesize that fatigue may be generated by specific HSPs signalling through neuronal or glial cells in the central nervous system. From a cohort of patients with primary Sjögren's syndrome, 20 patients with high and 20 patients with low fatigue were selected. Fatigue was evaluated with a fatigue visual analogue scale. Plasma concentrations of HSP32, HSP60, HSP72 and HSP90α were measured and analysed to determine if there were associations with the level of fatigue. Plasma concentrations of HSP90α were significantly higher in patients with high fatigue compared with those with low fatigue, and there was a tendency to higher concentrations of HSP72 in patients with high fatigue compared with patients with low fatigue. There were no differences in concentrations of HSP32 and HSP60 between the high- and low-fatigue groups. Thus, extracellular HSPs, particularly HSP90α, may signal fatigue in chronic inflammation. This supports the hypothesis that fatigue is generated by cellular defence mechanisms.

Keywords: Chronic fatigue; Sjögren’s syndrome; autoimmune diseases; cellular stress; heat shock proteins; innate immunity.

© The Author(s) 2016.

Figures

Figure 1.
Figure 1.
HSP plasma concentrations compared with levels of fatigue. HSP90α concentration differed between high- and low-fatigue groups (P = 0.02). The difference in HSP72 concentrations between high- and low-fatigue groups was close to significant (P = 0.06). There were no associations between fatigue level and plasma concentrations for HSP32 and HSP60. For ease of presentation, one data point for HSP60 (210 ng/ml) in a low-fatigue patient was not included in the Figure.
Figure 2.
Figure 2.
HSP plasma concentrations vs. BDI score. Patients were dichotomized into one group with no depression (BDI 

References

    1. Krupp LB, Pollina DA. Mechanisms and management of fatigue in progressive neurological disorders. Curr Opin Neurol 1996; 9: 456–460.
    1. Norheim KB, Jonsson G, Omdal R. Biological mechanisms of chronic fatigue. Rheumatology (Oxford) 2011; 50: 1009–1018.
    1. Morris G, Berk M, Walder K, et al. Central pathways causing fatigue in neuro-inflammatory and autoimmune illnesses. BMC Med 2015; 13: 28–28.
    1. Maes M. Inflammatory and oxidative and nitrosative stress pathways underpinning chronic fatigue, somatization and psychosomatic symptoms. Curr Opin Psychiatry 2009; 22: 75–83.
    1. Hart BL. Biological basis of the behavior of sick animals. Neurosci Biobehav Rev 1988; 12: 123–37.
    1. Dantzer R, Heijnen CJ, Kavelaars A, et al. The neuroimmune basis of fatigue. Trends Neurosci 2014; 37: 39–46.
    1. Smith DE, Lipsky BP, Russell C, et al. A central nervous system-restricted isoform of the interleukin-1 receptor accessory protein modulates neuronal responses to interleukin-1. Immunity 2009; 30: 817–31.
    1. Harboe E, Tjensvoll AB, Vefring HK, et al. Fatigue in primary Sjögren’s syndrome—a link to sickness behaviour in animals? Brain Behav Immun 2009; 23: 1104–8.
    1. Norheim KB, Harboe E, Gøransson LG, et al. Interleukin-1 inhibition and fatigue in primary Sjögren’s syndrome—a double blind, randomised clinical trial. PLoS One 2012; 7: e30123–e30123.
    1. Omdal R, Gunnarsson R. The effect of interleukin-1 blockade on fatigue in rheumatoid arthritis—a pilot study. Rheumatol Int 2005; 25: 481–484.
    1. Avalos I, Chung CP, Oeser A, et al. Oxidative stress in systemic lupus erythematosus: relationship to disease activity and symptoms. Lupus 2007; 16: 195–200.
    1. Kennedy G, Spence Va, McLaren M, et al. Oxidative stress levels are raised in chronic fatigue syndrome and are associated with clinical symptoms. Free Radic Biol Med 2005; 39: 584–589.
    1. Brkic S, Tomic S, Maric D, et al. Lipid peroxidation is elevated in female patients with chronic fatigue syndrome. Med Sci Monit 2010; 16: CR628–R632.
    1. Segal B, Thomas W, Zhu X, et al. Oxidative stress and fatigue in systemic lupus erythematosus. Lupus 2012; 21: 984–992.
    1. Chung CP, Titova D, Oeser A, et al. Oxidative stress in fibromyalgia and its relationship to symptoms. Clin Rheumatol 2009; 28: 435–8.
    1. Stetler RA, Gan Y, Zhang W, et al. Heat shock proteins: cellular and molecular mechanisms in the central nervous system. Prog Neurobiol 2010; 92: 184–211.
    1. Basu S, Binder RJ, Ramalingam T, et al. CD91 is a common receptor for heat shock proteins gp96, hsp90, hsp70, and calreticulin. Immunity 2001; 14: 303–313.
    1. Triantafilou M, Triantafilou K. Heat-shock protein 70 and heat-shock protein 90 associate with Toll-like receptor 4 in response to bacterial lipopolysaccharide. Biochem Soc Trans 2004; 32: 636–639.
    1. Jonsson R, Vogelsang P, Volchenkov R, et al. The complexity of Sjögren’s syndrome: novel aspects on pathogenesis. Immunol Lett 2011; 141: 1–9.
    1. Karageorgas T, Fragioudaki S, Nezos A, et al. Fatigue in primary Sjogren’s syndrome: clinical, laboratory, psychometric and biological associations. Arthritis Care Res (Hoboken) 2016; 68: 123–131.
    1. Segal B, Thomas W, Rogers T, et al. Prevalence, severity, and predictors of fatigue in subjects with primary Sjögren’s syndrome. Arthritis Rheum 2008; 59: 1780–1787.
    1. Mengshoel AM, Norheim KB, Omdal R. Primary Sjögren’s syndrome: fatigue is an ever-present, fluctuating, and uncontrollable lack of energy. Arthritis Care Res (Hoboken) 2014; 66: 1227–1232.
    1. Vitali C, Bombardieri S, Jonsson R, et al. Classification criteria for Sjögren’s syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis 2002; 61: 554–558.
    1. Wolfe F. Fatigue assessments in rheumatoid arthritis: comparative performance of visual analog scales and longer fatigue questionnaires in 7760 patients. J Rheumatol 2004; 31: 1896–1902.
    1. Arnold LM. Understanding fatigue in major depressive disorder and other medical disorders. Psychosomatics 2008; 49: 185–190.
    1. Omdal R, Waterloo K, Koldingsnes W, et al. Fatigue in patients with systemic lupus erythematosus: the psychosocial aspects. J Rheumatol 2003; 30: 283–287.
    1. Goshen I, Kreisel T, Ben-Menachem-Zidon O, et al. Brain interleukin-1 mediates chronic stress-induced depression in mice via adrenocortical activation and hippocampal neurogenesis suppression. Mol Psychiatry 2008; 13: 717–728.
    1. Lawson MA, McCusker RH, Kelley KW. Interleukin-1 beta converting enzyme is necessary for development of depression-like behavior following intracerebroventricular administration of lipopolysaccharide to mice. J Neuroinflammation 2013; 10: 54–54.
    1. Shevtsov MA, Nikolaev BP, Yakovleva LY, et al. Neurotherapeutic activity of the recombinant heat shock protein Hsp70 in a model of focal cerebral ischemia in rats. Drug Des Devel Ther 2014; 8: 639–650.
    1. Benarroch EE. Circumventricular organs: receptive and homeostatic functions and clinical implications. Neurology 2011; 77: 1198–1204.
    1. Svensson KJ, Christianson HC, Wittrup A, et al. Exosome uptake depends on ERK1/2-heat shock protein 27 signaling and lipid raft-mediated endocytosis negatively regulated by caveolin-1. J Biol Chem 2013; 288: 17713–17724.
    1. Antonucci F, Turola E, Riganti L, et al. Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J 2012; 31: 1231–1240.
    1. Chivet M, Javalet C, Laulagnier K, et al. Exosomes secreted by cortical neurons upon glutamatergic synapse activation specifically interact with neurons. J Extracell Vesicles 2014; 3: 24722–24722.
    1. Hegmans JPJJ, Bard MPL, Hemmes A, et al. Proteomic analysis of exosomes secreted by human mesothelioma cells. Am J Pathol 2004; 164: 1807–1815.
    1. Lancaster GI, Febbraio MA. Exosome-dependent trafficking of HSP70: a novel secretory pathway for cellular stress proteins. J Biol Chem 2005; 280: 23349–23355.
    1. Merendino AM, Bucchieri F, Campanella C, et al. Hsp60 is actively secreted by human tumor cells. PLoS One 2010; 5: e9247–e9247.
    1. Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by systemic injection of targeted exosomes. Nat Biotechnol 2011; 29: 341–345.
    1. Lucas K, Morris G, Anderson G, et al. The Toll-like receptor radical cycle pathway: a new drug target in immune-related chronic fatigue. CNS Neurol Disord Drug Targets 2015; 14: 838–854.
    1. Hines DJ, Choi HB, Hines RM, et al. Prevention of LPS-induced microglia activation, cytokine production and sickness behavior with TLR4 receptor interfering peptides. PLoS One 2013; 8: e60388–e60388.
    1. Asea A, Rehli M, Kabingu E, et al. Novel signal transduction pathway utilized by extracellular HSP70. Role of toll-like receptor (TLR) 2 and TLR4. J Biol Chem 2002; 277: 15028–15034.
    1. Kakimura J-I, Kitamura Y, Takata K, et al. Microglial activation and amyloid-beta clearance induced by exogenous heat-shock proteins. FASEB J 16: 601–603.
    1. Ohara K, Shimizu K, Matsuura S, et al. Toll-like receptor 4 signaling in trigeminal ganglion neurons contributes tongue-referred pain associated with tooth pulp inflammation. J Neuroinflammation 2013; 10: 139–139.
    1. Elfaitouri A, Herrmann B, Bölin-Wiener A, et al. Epitopes of microbial and human heat shock protein 60 and their recognition in myalgic encephalomyelitis. PLoS One 2013; 8: e81155–e81155.

Source: PubMed

3
Subscribe