Subsets of activated monocytes and markers of inflammation in incipient and progressed multiple sclerosis

Mikkel Carstensen Gjelstrup, Morten Stilund, Thor Petersen, Holger Jon Møller, Eva Lykke Petersen, Tove Christensen, Mikkel Carstensen Gjelstrup, Morten Stilund, Thor Petersen, Holger Jon Møller, Eva Lykke Petersen, Tove Christensen

Abstract

Multiple sclerosis (MS) is an immune mediated, inflammatory and demyelinating disease of the central nervous system (CNS). Substantial evidence points toward monocytes and macrophages playing prominent roles early in disease, mediating both pro- and anti-inflammatory responses. Monocytes are subdivided into three subsets depending on the expression of CD14 and CD16, representing different stages of inflammatory activation. To investigate their involvement in MS, peripheral blood mononuclear cells from 40 patients with incipient or progressed MS and 20 healthy controls were characterized ex vivo. In MS samples, we demonstrate a highly significant increase in nonclassical monocytes (CD14+CD16++), with a concomitant significant reduction in classical monocytes (CD14++CD16-) compared with healthy controls. Also, a significant reduction in the surface expression of CD40, CD163, and CD192 was found, attributable to the upregulation of the nonclassical monocytes. In addition, significantly increased levels of human endogenous retrovirus (HERV) envelope (Env) epitopes, encoded by both HERV-H/F and HERV-W, were specifically found on nonclassical monocytes from patients with MS; emphasizing their involvement in MS disease. In parallel, serum and cerebrospinal fluid (CSF) samples were analyzed for soluble biomarkers of inflammation and neurodegeneration. For sCD163 versus CD163, no significant correlations were found, whereas highly significant correlations between levels of soluble neopterine and the intermediate monocyte (CD14++CD16+) population was found, as were correlations between levels of soluble osteopontin and the HERV Env expression on nonclassical monocytes. The results from this study emphasize the relevance of further focus on monocyte subsets, particularly the nonclassical monocytes in monitoring of inflammatory diseases.

Keywords: ELISA; MS; Biomarkers; FLOW Cytometry; HERVs; monocyte subsets; monocytes; multiple sclerosis.

© 2017 The Authors Immunology and Cell Biology published by John Wiley & Sons Australia, Ltd on behalf of Australasian Society for Immunology Inc.

Figures

Figure 1
Figure 1
Gating strategy used in the flow cytometric analysis of patient and healthy control samples. A sample from a representative patient with RRMS was used for this figure. (a) From left to right: Total monocytes (> 20,000 events) were gated according to their size and granularity in forward scatter‐height (FSC‐H)/side scatter‐height (SSC‐H), aggregated cells were removed according to forward scatter‐area (FSC‐A)/FSC‐H and side scatter‐area (SSC‐A)/SSC‐H, and finally dead cells were removed according to staining with a LIVE/DEAD ® cell stain. (b) From left to right: The tree monocyte subsets (classical, intermediate, nonclassical) were gated on the “Live Cells” gate, as were the CD40+, CD163+, and CD192+ cells (blue). Appropriate isotype controls (red) were used to determine the unspecific antibody binding. (c, d) From left to right: Human endogenous retrovirus (HERV) expression was determined on the total monocyte population (Live cells) and the three monocyte subsets (classical, intermediate, nonclassical) by incubation with sera from rabbits immunized with HERV H3 Env (c) or HERV W3 Env (d) peptide antigens (blue) as described previously36 and with the appropriate control (pre‐immune sera) (red) to determine the median fluorescence index (MFI).
Figure 2
Figure 2
Differences in the expression of CD40, CD163, CD192, and of the three monocyte subsets in the patient group normalised to the median of the healthy controls. The differences in expression of CD40, CD163, and CD192 on the total monocyte population (Live cells, Figure 1) (a) and the three monocyte sub‐populations; classical, intermediate, and nonclassical (b) were determined as % positive cells of the monocytes, divided by the respective median levels of the healthy control samples to give the fold change. Bars represent the median of the populations and braces indicate a significant difference (Mann–Whitney U‐test) between the median of the patient group (n = 40) and the median of the healthy control group (n = 20). P‐values are listed. Pt, patients; HC, healthy controls.
Figure 3
Figure 3
Differences in the expression of HERV H3 Env and HERV W3 Env on the three monocyte subsets in the patient group and the HC group. The differences in expression of HERV H3 Env (a) and HERV W3 Env (b) on the three monocyte subsets; classical, intermediate, and nonclassical were determined as the median fluorescence index by calculating the median fluorescence for each sample and dividing by the median fluorescence of the appropriate control (pre‐immune sera). Bars represent the median of the subsets and braces indicate a significant difference (Mann–Whitney U‐test) between the median of the patient group (n = 40) and the median of the HC group (n = 20). P‐values are listed. Pt, patients, HC, healthy controls.
Figure 4
Figure 4
Logistic regression analyses with ROC curve output of patients with CIS or MS plotted against the HC group. AUC, with 95% CI, is given for each parameter. The surface expression of each parameter for patients with CIS or MS (n = 40) are combined as true positives and plotted against HC as true negatives (n = 20). The diagonal dividing the ROC space represents the random event. A logistic regression analysis with combined parameter results has been performed for “all parameters”, parameters with AUC > 0.70, and AUC > 0.75. ROC, receiver operating characteristic; AUC, Area under the curve; RRMS, relapsing‐remitting MS; PPMS, primary‐progressive MS; CIS, clinically isolated syndrome; HCs,healthy controls.

References

    1. Pan G, Simpson S Jr, van der Mei I, et al Role of genetic susceptibility variants in predicting clinical course in multiple sclerosis: a cohort study. J Neurol Neurosurg Psychiatry 2016; 87: 1204–1211.
    1. Dendrou CA, Fugger L, Friese MA. Immunopathology of multiple sclerosis. Nat Rev Immunol 2015; 15: 545–558.
    1. Compston A, Coles A. Multiple sclerosis. Lancet 2008; 372: 1502–1517.
    1. Miller D, Barkhof F, Montalban X, et al Clinically isolated syndromes suggestive of multiple sclerosis, part 1: natural history, pathogenesis, diagnosis, and prognosis. Lancet Neurol 2005; 4: 281–288.
    1. Harris VK, Sadiq SA. Disease biomarkers in multiple sclerosis potential for use in therapeutic decision making. Mol Diagn Ther 2009; 13: 225–244.
    1. Nielsen HH, Beck HC, Kristensen LP, et al The urine proteome profile is different in neuromyelitis optica compared to multiple sclerosis: a clinical proteome study. PLoS ONE 2015; 10: e0139659.
    1. Housley WJ, Pitt D, Hafler DA. Biomarkers in multiple sclerosis. Clin Immunol 2015; 161: 51–58.
    1. Polman CH, Reingold SC, Banwell B, et al Diagnostic criteria for multiple sclerosis: 2010 revisions to the McDonald criteria. Ann Neurol 2011; 69: 292–302.
    1. Stilund M, Gjelstrup MC, Petersen T, et al Biomarkers of inflammation and axonal degeneration/damage in patients with newly diagnosed multiple sclerosis: contributions of the soluble CD163 CSF/serum ratio to a biomarker panel. PLoS ONE 2015; 10: e0119681.
    1. Xia J, Broadhurst DI, Wilson M, et al Translational biomarker discovery in clinical metabolomics: an introductory tutorial. Metabolomics 2013; 9: 280–299.
    1. Fabriek BO, Moller HJ, Vloet RP, et al Proteolytic shedding of the macrophage scavenger receptor CD163 in multiple sclerosis. J Neuroimmunol 2007; 187: 179–186.
    1. Bar‐Or A, Nuttall RK, Duddy M, et al Analyses of all matrix metalloproteinase members in leukocytes emphasize monocytes as major inflammatory mediators in multiple sclerosis. Brain 2003; 126: 2738–2749.
    1. Etzerodt A, Rasmussen MR, Svendsen P, et al Structural basis for inflammation‐driven shedding of CD163 ectodomain and tumor necrosis factor‐alpha in macrophages. J Biol Chem 2014; 289: 778–788.
    1. Moller HJ. Soluble CD163. Scand J Clin Lab Invest 2012; 72: 1–13.
    1. Kjaergaard AG, Rodgaard‐Hansen S, Dige A, et al Monocyte expression and soluble levels of the haemoglobin receptor (CD163/sCD163) and the mannose receptor (MR/sMR) in septic and critically ill non‐septic ICU patients. PLoS ONE 2014; 9: e92331.
    1. Chitadze G, Lettau M, Bhat J, et al Shedding of endogenous MHC class I‐related chain molecules A and B from different human tumor entities: heterogeneous involvement of the “a disintegrin and metalloproteases” 10 and 17. Int J Cancer 2013; 133: 1557–1566.
    1. Wong KL, Yeap WH, Tai JJY, et al The three human monocyte subsets: implications for health and disease. Immunol Res 2012; 53: 41–57.
    1. Miron VE, Boyd A, Zhao JW, et al M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 2013; 16: U1211–U1275.
    1. Vogel DY, Vereyken EJ, Glim JE, et al Macrophages in inflammatory multiple sclerosis lesions have an intermediate activation status. J Neuroinflammation 2013; 10: 35.
    1. Ziegler‐Heitbrock L. Blood monocytes and their subsets: established features and open questions. Front Immunol 2015; 6: 423.
    1. Kouris A, Pistiki A, Katoulis A, et al Proinflammatory cytokine responses in patients with psoriasis. Eur Cytokine Netw 2014; 25: 63–68.
    1. Kawanaka N, Yamamura M, Aita T, et al CD14+, CD16+ blood monocytes and joint inflammation in rheumatoid arthritis. Arthritis Rheum 2002; 46: 2578–2586.
    1. Burdo TH, Soulas C, Orzechowski K, et al Increased monocyte turnover from bone marrow correlates with severity of SIV encephalitis and CD163 levels in plasma. PLoS Pathog 2010; 6: e1000842.
    1. Waschbisch A, Schroder S, Schraudner D, et al Pivotal role for CD16+ monocytes in immune surveillance of the central nervous system. J Immunol 2016; 196:1558–1567.
    1. Ashhurst TM, Vreden CV, Niewold P, et al The plasticity of inflammatory monocyte responses to the inflamed central nervous system. Cell Immunol 2014; 291: 49–57.
    1. Chitnis T, Khoury SJ. Role of costimulatory pathways in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis. J Allergy Clin Immunol 2003; 112: 837–849.
    1. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 2011; 11: 762–774.
    1. Stilund M, Reuschlein AK, Christensen T, et al Soluble CD163 as a marker of macrophage activity in newly diagnosed patients with multiple sclerosis. PLoS ONE 2014; 9: e98588.
    1. Katsavos S, Anagnostouli M. Biomarkers in multiple sclerosis: an up‐to‐date overview. Mult Scler Int 2013; 2013: 340508.
    1. Stilund M, Gjelstrup MC, Christensen T, et al A multi‐biomarker follow‐up study of patients with multiple sclerosis. Brain Behav 2016; 6: e00509.
    1. Brettschneider J, Czerwoniak A, Senel M, et al The chemokine CXCL13 is a prognostic marker in clinically isolated syndrome (CIS). PLoS ONE 2010; 5: e11986.
    1. Murr C, Widner B, Wirleitner B, et al Neopterin as a marker for immune system activation. Curr Drug Metab 2002; 3: 175–187.
    1. Teunissen CE, Khalil M. Neurofilaments as biomarkers in multiple sclerosis. Mult Scler 2012; 18: 552–556.
    1. Kivisakk P, Healy BC, Francois K, et al Evaluation of circulating osteopontin levels in an unselected cohort of patients with multiple sclerosis: relevance for biomarker development. Mult Scler 2014; 20: 438–444.
    1. Christensen T. HERVs in neuropathogenesis. J Neuroimmune Pharmacol 2010; 5: 326–335.
    1. Brudek T, Christensen T, Aagaard L, et al B cells and monocytes from patients with active multiple sclerosis exhibit increased surface expression of both HERV‐H Env and HERV‐W Env, accompanied by increased seroreactivity. Retrovirology 2009; 6: 104.
    1. Laska MJ, Brudek T, Nissen KK, et al Expression of HERV‐Fc1, a human endogenous retrovirus, is increased in patients with active multiple sclerosis. J Virol 2012; 86: 3713–3722.
    1. Christensen T, Petersen T, Thiel S, et al Gene‐environment interactions in multiple sclerosis: Innate and adaptive immune responses to human endogenous retrovirus and herpesvirus antigens and the lectin complement activation pathway. J Neuroimmunol 2007; 183: 175–188.
    1. Petersen T, Moller‐Larsen A, Ellermann‐Eriksen S, et al Effects of interferon‐beta therapy on elements in the antiviral immune response towards the human herpesviruses EBV, HSV, and VZV, and to the human endogenous retroviruses HERV‐H and HERV‐W in multiple sclerosis. J Neuroimmunol 2012; 249: 105–108.
    1. Christensen T, Sorensen PD, Hansen HJ, et al Antibodies against a human endogenous retrovirus and the preponderance of env splice variants in multiple sclerosis patients. Mult Scler 2003; 9: 6–15.
    1. Hauser SL, Waubant E, Arnold DL, et al B‐cell depletion with rituximab in relapsing‐remitting multiple sclerosis. N Engl J Med 2008; 358: 676–688.
    1. Kappos L, Li D, Calabresi PA, et al Ocrelizumab in relapsing‐remitting multiple sclerosis: a phase 2, randomised, placebo‐controlled, multicentre trial. Lancet 2011; 378: 1779–1787.
    1. Polman CH, O'Connor PW, Havrdova E, et al A randomized, placebo‐controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med 2006; 354: 899–910.
    1. Yokoyama Y, Watanabe K, Ito H, et al Factors associated with treatment outcome, and long‐term prognosis of patients with ulcerative colitis undergoing selective depletion of myeloid lineage leucocytes: a prospective multicenter study. Cytotherapy 2015; 17: 680–688.
    1. Ziegler‐Heitbrock L, Ancuta P, Crowe S, et al Nomenclature of monocytes and dendritic cells in blood. Blood 2010; 116: e74–e80.
    1. Fischer‐Smith T, Croul S, Sverstiuk AE, et al CNS invasion by CD14+/CD16+ peripheral blood‐derived monocytes in HIV dementia: perivascular accumulation and reservoir of HIV infection. J Neurovirol 2001; 7: 528–541.
    1. Chuluundorj D, Harding SA, Abernethy D, et al Expansion and preferential activation of the CD14(+)CD16(+) monocyte subset during multiple sclerosis. Immunol Cell Biol 2014; 92: 509–517.
    1. Wiesemann E, Deb M, Trebst C, et al Effects of interferon‐beta on co‐signaling molecules: upregulation of CD40, CD86 and PD‐L2 on monocytes in relation to clinical response to interferon‐beta treatment in patients with multiple sclerosis. Mult Scler 2008; 14: 166–176.
    1. Moller HJ, Aerts H, Gronbaek H, et al Soluble CD163: a marker molecule for monocyte/macrophage activity in disease. Scand J Clin Lab Invest 2002; 62: 29–33.
    1. Contin C, Pitard V, Itai T, et al Membrane‐anchored CD40 is processed by the tumor necrosis factor‐alpha‐converting enzyme ‐ Implications for CD40 signaling. J Biol Chem 2003; 278: 32801–32809.
    1. Chen P, Su B, Zhang T, et al Perturbations of monocyte subsets and their association with T helper cell differentiation in acute and chronic hiV‐1‐infected patients. Front Immunol 2017; 8:272.
    1. Davis BH, Zarev PV. Human monocyte CD163 expression inversely correlates with soluble CD163 plasma levels. Cytometry B Clin Cytom 2005; 63: 16–22.
    1. Christensen T. Human endogenous retroviruses in neurologic disease. Apmis. 2016; 124: 116–126.
    1. Antony JM, Deslauriers AM, Bhat RK, et al Human endogenous retroviruses and multiple sclerosis: innocent bystanders or disease determinants? Biochim Biophys Acta 2011; 1812:162–176.
    1. Nexo BA, Christensen T, Frederiksen J, et al The etiology of multiple sclerosis: genetic evidence for the involvement of the human endogenous retrovirus HERV‐Fc1. PLoS ONE 2011; 6: e16652.
    1. Mameli G, Poddighe L, Mei A, et al Expression and activation by epstein barr virus of human endogenous retroviruses‐Win blood cells and astrocytes: inference for multiple sclerosis. PLoS ONE 2012; 7: e44991.
    1. Antony JM, Ellestad KK, Hammond R, et al The human endogenous retrovirus envelope glycoprotein, syncytin‐1, regulates neuroinflammation and its receptor expression in multiple sclerosis: a role for endoplasmic reticulum chaperones in astrocytes. J Immunol 2007; 179: 1210–1224.
    1. Perron H, Lazarini F, Ruprecht K, et al Human endogenous retrovirus (HERV)‐W ENV and GAG proteins: physiological expression in human brain and pathophysiological modulation in multiple sclerosis lesions. J Neurovirol 2005; 11: 23–33.
    1. Petersen T, Moller‐Larsen A, Thiel S, et al Effects of interferon‐beta therapy on innate and adaptive immune responses to the human endogenous retroviruses HERV‐H and HERV‐W, cytokine production, and the lectin complement activation pathway in multiple sclerosis. J Neuroimmunol 2009; 215: 108–116.
    1. Johnston JB, Silva C, Holden J, et al Monocyte activation and differentiation augment human endogenous retrovirus expression: implications for inflammatory brain diseases. Ann Neurol 2001; 50: 434–442.
    1. Christensen T. Association of human endogenous retroviruses with multiple sclerosis and possible interactions with herpes viruses. Rev Med Virol 2005; 15: 179–211.
    1. Hansen DT, Petersen T, Christensen T. Retroviral envelope proteins: Involvement in neuropathogenesis. J Neurol Sci 2017; 380: 151–163.
    1. Sharma P, Azebi S, England P, et al Citrullination of histone H3 Interferes with HP1‐mediated transcriptional repression. PLoS Genet 2012; 8: e1002934.
    1. Kurtzke JF. Rating neurologic impairment in multiple sclerosis: an expanded disability status scale (EDSS). Neurology 1983; 33: 1444–1452.
    1. Teunissen CE, Petzold A, Bennett JL, et al A consensus protocol for the standardization of cerebrospinal fluid collection and biobanking. Neurology 2009; 73: 1914–1922.
    1. Moller HJ, Hald K, Moestrup SK. Characterization of an enzyme‐linked immunosorbent assay for soluble CD163. Scand J Clin Lab Invest 2002; 62: 293–299.

Source: PubMed

Подписаться