Differential Accumulation and Activation of Monocyte and Dendritic Cell Subsets in Inflamed Synovial Fluid Discriminates Between Juvenile Idiopathic Arthritis and Septic Arthritis

Maïlys Cren, Nadège Nziza, Aurélia Carbasse, Perrine Mahe, Emilie Dufourcq-Lopez, Marion Delpont, Hugues Chevassus, Mirna Khalil, Thibault Mura, Isabelle Duroux-Richard, Florence Apparailly, Eric Jeziorski, Pascale Louis-Plence, Maïlys Cren, Nadège Nziza, Aurélia Carbasse, Perrine Mahe, Emilie Dufourcq-Lopez, Marion Delpont, Hugues Chevassus, Mirna Khalil, Thibault Mura, Isabelle Duroux-Richard, Florence Apparailly, Eric Jeziorski, Pascale Louis-Plence

Abstract

Despite their distinct etiology, several lines of evidence suggest that innate immunity plays a pivotal role in both juvenile idiopathic arthritis (JIA) and septic arthritis (SA) pathophysiology. Indeed, monocytes and dendritic cells (DC) are involved in the first line of defense against pathogens and play a critical role in initiating and orchestrating the immune response. The aim of this study was to compare the number and phenotype of monocytes and DCs in peripheral blood (PB) and synovial fluid (SF) from patients with JIA and SA to identify specific cell subsets and activation markers associated with pathophysiological mechanisms and that could be used as biomarkers to discriminate both diseases. The proportion of intermediate and non-classical monocytes in the SF and PB, respectively, were significantly higher in JIA than in SA patients. In contrast the proportion of classical monocytes and their absolute numbers were higher in the SF from SA compared with JIA patients. Higher expression of CD64 on non-classical monocyte was observed in PB from SA compared with JIA patients. In SF, higher expression of CD64 on classical and intermediate monocyte as well as higher CD163 expression on intermediate monocytes was observed in SA compared with JIA patients. Moreover, whereas the number of conventional (cDC), plasmacytoid (pDC) and inflammatory (infDC) DCs was comparable between groups in PB, the number of CD141+ cDCs and CD123+ pDCs in the SF was significantly higher in JIA than in SA patients. CD14+ infDCs represented the major DC subset in the SF of both groups with potent activation assessed by high expression of HLA-DR and CD86 and significant up-regulation of HLA-DR expression in SA compared with JIA patients. Finally, higher activation of SF DC subsets was monitored in SA compared with JIA with significant up-regulation of CD86 and PDL2 expression on several DC subsets. Our results show the differential accumulation and activation of innate immune cells between septic and inflammatory arthritis. They strongly indicate that the relative high numbers of CD141+ cDC and CD123+ pDCs in SF are specific for JIA while the over-activation of DC and monocyte subsets is specific for SA.

Keywords: CD123 pDC; CD141 cDC; dendritic cells; juvenile idiopathic arthritis; monocytes; multiparametric flow cytometry; septic arthritis.

Copyright © 2020 Cren, Nziza, Carbasse, Mahe, Dufourcq-Lopez, Delpont, Chevassus, Khalil, Mura, Duroux-Richard, Apparailly, Jeziorski and Louis-Plence.

Figures

Figure 1
Figure 1
Representative staining of monocyte subsets. Representative dot plots of the classical (CD14++CD16−, red), intermediate (CD14++CD16+, blue) and non-classical (CD14+CD16++, green) monocytes and representative histograms for the various markers (CCR2, CD163, CD64, HLA-DR, and SLAN) on each subset. (A) in peripheral blood (B) in synovial fluid of patients with JIA (left) or SA (right). Black histograms correspond to negative control MFI.
Figure 2
Figure 2
Representative staining of DC subsets. Representative dot plot of the CD1c+ cDC (1, red), CD14+ InfDCs (2, blue), CD141+ cDC (3, orange) and CD123+ pDC(4, green) and representative histograms for the various markers (PDL2, CD86, HLA-DR) on each subset (A) in peripheral blood and (B) in synovial fluid of patients with JIA (left) or SA (right). Black histograms correspond to negative control MFI.
Figure 3
Figure 3
Stratification of patients with JIA and SA based on monocyte proportion and all cell counts. (A) Heatmap of monocyte proportion and all cell counts in peripheral blood (PB) and synovial fluid (SF) by multi-parametric flow cytometry. In the color scale, yellow indicates relative higher and blue relative lower cell numbers/proportion. Gray data are undetermined. The dendrogram shows the results of the non-supervised hierarchical clustering analysis; in the heatmap: rows, cell subsets; and columns, individual patients with JIA (gray, n = 15) and SA (black, n = 14). (B) Heatmap showing markers that were significantly differentially expressed between patients with SA and JIA. The dendrogram shows the results of the non-supervised hierarchical clustering analysis using an FDR cut-off of 5%. (C) Direct comparison of the 5 differentially expressed cells counts in synovial fluid (SF) and non-classical monocyte proportion in peripheral blood (PB) from JIA (white, n = 15) and SA (gray, n = 14) patients. Data are represented for all patients and horizontal bars are the mean ± SEM. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001 (two-sided t or Mann–Whitney test, according to data distribution after FDR adjustment).
Figure 4
Figure 4
Stratification of patients with JIA and SA based on DC and monocyte activation markers. (A) Heatmap showing the mean fluorescence intensities (MFI, rows) of activation markers that were differentially expressed in JIA (gray, n = 15) and SA (black, n = 14) patients (column). The dendrogram shows the results of the non-supervised hierarchical clustering analysis using an FDR cut-off of 5%. Yellow indicates relative higher expression, blue relative lower expression of the markers and gray undetermined data. Direct comparison of the expression of (B) CD64 and (C) CD163 in monocyte subsets and (D) CD86, (E) PDL2, and (F) HLA-DR in DC subsets in blood (PB) and synovial fluid (SF) samples of patients with JIA (white, n = 15) and SA (gray, n = 14); Data are represented for all patients and horizontal bars are mean ± SEM. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.001 (two-sided t or Mann–Whitney test, according to data distribution after FDR adjustment).
Figure 5
Figure 5
Stratification of patients with JIA and SA based on all parameters. (A) Heatmap showing the markers (markers, rows) that were differentially represented in JIA (gray, n = 15) and SA (black, n = 14) patients (column). The dendrogram shows the results of the non-supervised hierarchical clustering analysis using an FDR cut-off of 5%. Yellow indicates relative higher expression, blue relative lower expression of the markers. (B) Receiver operating curves (ROC) for markers with AUC values > 0.94.

References

    1. Aupiais C, Ilharreborde B, Doit C, Blachier A, Desmarest M, Job-Deslandre C, et al. . Aetiology of arthritis in hospitalised children: an observational study. Arch Dis Child. (2015) 100:742–7. 10.1136/archdischild-2014-307490
    1. Wall C, Donnan L. Septic arthritis in children. Aust Fam Phys. (2015) 44:213–5.
    1. Al-Qwbani M, Jiang N, Yu B. Kingella kingae-associated pediatric osteoarticular infections: an overview of 566 reported cases. Clin Pediatr (Phila). (2016) 55:1328–37. 10.1177/0009922816629620
    1. Petty RE. Growing pains: the ILAR classification of juvenile idiopathic arthritis. J Rheumatol. (2001) 28:927–8.
    1. Prakken B, Albani S, Martini A. Juvenile idiopathic arthritis. Lancet. (2011) 377:2138–49. 10.1016/S0140-6736(11)60244-4
    1. Macaubas C, Nguyen K, Deshpande C, Phillips C, Peck A, Lee T, et al. . Distribution of circulating cells in systemic juvenile idiopathic arthritis across disease activity states. Clin Immunol. (2010) 134:206–16. 10.1016/j.clim.2009.09.010
    1. Smolewska E, Stanczyk J, Brózik H, Biernacka-Zielinska M, Cebula B, Robak T, et al. . Distribution and clinical significance of blood dendritic cells in children with juvenile idiopathic arthritis. Ann Rheum Dis. (2008) 67:762–8. 10.1136/ard.2007.077669
    1. Segura E, Touzot M, Bohineust A, Cappuccio A, Chiocchia G, Hosmalin A, et al. . Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity. (2013) 38:336–48. 10.1016/j.immuni.2012.10.018
    1. Reynolds G, Gibbon JR, Pratt AG, Wood MJ, Coady D, Raftery G, et al. . Synovial CD4+ T-cell-derived GM-CSF supports the differentiation of an inflammatory dendritic cell population in rheumatoid arthritis. Ann Rheum Dis. (2016) 75:899–907. 10.1136/annrheumdis-2014-206578
    1. Wong KL, Jing-Yi Tai J, Wong W-C, Han H, Sem X, Yeap W-H, et al. . Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood. (2011) 118:e16–31. 10.1182/blood-2010-12-326355
    1. Carlin LM, Stamatiades EG, Cedric A, Hanna RN, Glover L, Vizcay-Barrena G, et al. . Nr4a1-dependent Ly6C(low) monocytes monitor endothelial cells and orchestrate their disposal. Cell. (2013) 153:362–75. 10.1016/j.cell.2013.03.010
    1. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, Kayal S, et al. . Monitoring of blood vessels and tissues by a population of monocytes with patrolling behavior. Science. (2007) 317:666–70. 10.1126/science.1142883
    1. Geissmann F, Jung S, Littman DR. Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity. (2003) 19:71–82. 10.1016/S1074-7613(03)00174-2
    1. Cros J, Cagnard N, Woollard K, Patey N, Zhang SY, Senechal B, et al. . Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity. (2010) 33:375–86. 10.1016/j.immuni.2010.08.012
    1. Wong KL, Yeap WH, Tai JJ, Ong SM, Dang TM, Wong SC. The three human monocyte subsets: implications for health and disease. Immunol Res. (2012) 53:41–57. 10.1007/s12026-012-8297-3
    1. Skrzeczyñska J, Kobylarz K, Hartwich Z, Zembala M, Pryjma J. CD14+CD16+ monocytes in the course of sepsis in neonates and small children: monitoring and functional studies. Scand J Immunol. (2002) 55:629–38. 10.1046/j.1365-3083.2002.01092.x
    1. Smiljanovic B, Radzikowska A, Kuca-Warnawin E, Kurowska W, Grün JR, Stuhlmüller B, et al. . Monocyte alterations in rheumatoid arthritis are dominated by preterm release from bone marrow and prominent triggering in the joint. Ann Rheum Dis. (2018) 77:300–8. 10.1136/annrheumdis-2017-211649
    1. Gaur P, Myles A, Misra R, Aggarwal A. Intermediate monocytes are increased in enthesitis-related arthritis, a category of juvenile idiopathic arthritis. Clin Exp Immunol. (2017) 187:234–41. 10.1111/cei.12880
    1. Yoon BR, Yoo SJ, Choi Yh, Chung YH, Kim J, Yoo IS, et al. . Functional phenotype of synovial monocytes modulating inflammatory T-cell responses in rheumatoid arthritis (RA). PLoS ONE. (2014) 9:e109775. 10.1371/journal.pone.0109775
    1. Rossol M, Kraus S, Pierer M, Baerwald C, Wagner U. The CD14(bright) CD16+ monocyte subset is expanded in rheumatoid arthritis and promotes expansion of the Th17 cell population. Arthritis Rheum. (2012) 64:671–7. 10.1002/art.33418
    1. de Jager W, Hoppenreijs EP, Wulffraat NM, Wedderburn LR, Kuis W, Prakken BJ. Blood and synovial fluid cytokine signatures in patients with juvenile idiopathic arthritis: a cross-sectional study. Ann Rheum Dis. (2007) 66:589–98. 10.1136/ard.2006.061853
    1. Aupiais C, Basmaci R, Ilharreborde B, Blachier A, Desmarest M, Job-Deslandre C, et al. . Arthritis in children: comparison of clinical and biological characteristics of septic arthritis and juvenile idiopathic arthritis. Arch Dis Child. (2017) 102:316–22. 10.1136/archdischild-2016-310594
    1. Hanley JA, McNeil BJ. The meaning and use of the area under a receiver operating characteristic (ROC) curve. Radiology. (1982) 143:29–36. 10.1148/radiology.143.1.7063747
    1. Hochberg Y, Benjamini Y. More powerful procedures for multiple significance testing. Stat Med. (1990) 9:811–8. 10.1002/sim.4780090710
    1. Boltjes A, van Wijk F. Human dendritic cell functional specialization in steady-state and inflammation. Front Immunol. (2014) 5:131. 10.3389/fimmu.2014.00131
    1. Dutertre CA, Becht E, Irac SE, Khalilnezhad A, Narang V, Khalilnezha S, et al. . Single-cell analysis of human mononuclear phagocytes reveals subset-defining markers and identifies circulating inflammatory dendritic cells. Immunity. (2019) 51:573–89. e578. 10.1016/j.immuni.2019.08.008
    1. Gregorio A, Gambini C, Gerloni V, Parafioriti A, Sormani MP, Gregorio S et al. Lymphoid neogenesis in juvenile idiopathic arthritis correlates with ANA positivity and plasma cells infiltration. Rheumatology. (2007) 46:308–13. 10.1093/rheumatology/kel225
    1. Wedderburn LR, Robinson N, Patel A, Varsani H, Woo P. Selective recruitment of polarized T cells expressing CCR5 and CXCR3 to the inflamed joints of children with juvenile idiopathic arthritis. Arthritis Rheum. (2000) 43:765–74. 10.1002/1529-0131(200004)43:4<765::AID-ANR7>;2-B
    1. Mukherjee R, Barman PK, Thatoi PK, Tripathy R, Das BK, Ravindran B. Non-Classical monocytes display inflammatory features: validation in sepsis and systemic lupus Erythematous. Sci Rep. (2015) 5:13886. 10.1038/srep13886
    1. Kim OY, Monsel A, Bertrand M, Coriat P, Cavaillon JM, Adib-Conquy M. Differential down-regulation of HLA-DR on monocyte subpopulations during systemic inflammation. Crit Care. (2010) 14:R61. 10.1186/cc8959
    1. Li Y, Lee PY, Kellner ES, Paulus M, Switanek J, Xu Y, et al. . Monocyte surface expression of Fcgamma receptor RI (CD64), a biomarker reflecting type-I interferon levels in systemic lupus erythematosus. Arthritis Res Ther. (2010) 12:R90. 10.1186/ar3017
    1. Tanaka S, Nishino J, Matsui T, Komiya A, Nishimura K, Tohma S. Neutrophil CD64 expression in the diagnosis of local musculoskeletal infection and the impact of antibiotics. J Bone Joint Surg Br. (2009) 91:1237–42. 10.1302/0301-620X.91B9.22051
    1. Li S, Yu Y, Yue Y, Zhang Z, Su K. Microbial infection and rheumatoid arthritis. J Clin Cell Immunol. (2013) 4:174. 10.4172/2155-9899.1000174
    1. Oppegaard O, Skodvin B, Halse AK, Langeland N. CD64 as a potential biomarker in septic arthritis. BMC Infect Dis. (2013) 13:278. 10.1186/1471-2334-13-278
    1. Fjaertoft G, Håkansson L, Ewald U, Foucard T, Venge P. Neutrophils from term and preterm newborn infants express the high affinity Fcgamma-receptor I (CD64) during bacterial infections. Pediatr Res. (1999) 45:871–6. 10.1203/00006450-199906000-00016
    1. Fjaertoft G, Håkansson LD, Pauksens K, Sisask G, Venge P. Neutrophil CD64 (FcgammaRI) expression is a specific marker of bacterial infection: a study on the kinetics and the impact of major surgery. Scand J Infect Dis. (2007) 39:525–35. 10.1080/00365540601113693
    1. Groselj-Grenc M, Ihan A, Pavcnik-Arnol M, Kopitar AN, Gmeiner-Stopar T, Derganc M. Neutrophil and monocyte CD64 indexes, lipopolysaccharide-binding protein, procalcitonin and C-reactive protein in sepsis of critically ill neonates and children. Intensive Care Med. (2009) 35:1950–8. 10.1007/s00134-009-1637-7
    1. Hofer TP, Zawada AM, Frankenberger M, Skokann K, Satzl AA, Gesierich W, et al. . Slan-defined subsets of CD16-positive monocytes: impact of granulomatous inflammation and M-CSF receptor mutation. Blood. (2015) 126:2601–10. 10.1182/blood-2015-06-651331
    1. Throm AA, Moncrieffe H, Orandi AB, Pingel JT, Geurs TL, Miller HL, et al. . Identification of enhanced IFN-γ signaling in polyarticular juvenile idiopathic arthritis with mass cytometry. JCI Insight. (2018) 3:e121544. 10.1172/jci.insight.121544
    1. Canavan M, Walsh AM, Bhargava V, Wade SM, McGarry T, Marzaioli V, et al. . Enriched Cd141+ DCs in the joint are transcriptionally distinct, activated, and contribute to joint pathogenesis. JCI Insight. (2018) 3:e95228. 10.1172/jci.insight.95228
    1. Villadangos JA, Young L. Antigen-presentation properties of plasmacytoid dendritic cells. Immunity. (2008) 29:352–61. 10.1016/j.immuni.2008.09.002
    1. Swiecki M, Colonna M. The multifaceted biology of plasmacytoid dendritic cells. Nat Rev Immunol. (2015) 15:471–85. 10.1038/nri3865
    1. Hanabuchi S, Ito T, Rark W-P, Watanabe N, Shaw JL, Roman E, et al. . Thymic stromal lymphopoietin-activated plasmacytoid dendritic cells induce the generation of FOXP3+ regulatory T cells in human thymus. J Immunol. (2010) 184:2999–3007. 10.4049/jimmunol.0804106
    1. Martín-Gayo E, Sierra-Filardi E, Corbí AL, Toribio ML. Plasmacytoid dendritic cells resident in human thymus drive natural Treg cell development. Blood. (2010) 115:5366–75. 10.1182/blood-2009-10-248260
    1. Moseman EA, Liang X, Dawson AJ, Panoskaltsis-Mortari A, Krieg AM, Liu YJ, et al. . Human plasmacytoid dendritic cells activated by CpG oligodeoxynucleotides induce the generation of CD4+CD25+ regulatory T cells. J Immunol. (2004) 173:4433–42. 10.4049/jimmunol.173.7.4433
    1. Kavousanaki M, Makrigiannakis A, Boumpas D, Verginis P. Novel role of plasmacytoid dendritic cells in humans: induction of interleukin-10-producing Treg cells by plasmacytoid dendritic cells in patients with rheumatoid arthritis responding to therapy. Arthritis Rheum. (2010) 62:53–63. 10.1002/art.25037
    1. van Bon L, Affandi AJ, Broen J, Christmann RB, Marijnissen RJ, Stawski L, et al. . Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J Med. (2014) 370:433–43. 10.1056/NEJMc1402401
    1. Gilliet M, Cao W, Liu YJ. Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nat Rev Immunol. (2008) 8:594–606. 10.1038/nri2358
    1. Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, et al. . Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjögren's syndrome. Proc Natl Acad Sci USA. (2006) 103:2770–5. 10.1073/pnas.0510837103
    1. López de Padilla CM, Vallejo AN, McNallan KT, Vehe R, Smith SA, Dietz AB, et al. . Plasmacytoid dendritic cells in inflamed muscle of patients with juvenile dermatomyositis. Arthritis Rheum. (2007) 56:1658–68. 10.1002/art.22558
    1. Ganguly D, Haak S, Sisirak V, Reizis B. The role of dendritic cells in autoimmunity. Nat Rev Immunol. (2013) 13:566–77. 10.1038/nri3477
    1. Rönnblom LE, Alm GV, Oberg KE. Autoimmunity after alpha-interferon therapy for malignant carcinoid tumors. Ann Intern Med. (1991) 115:178–83. 10.7326/0003-4819-115-3-178
    1. van Krinks CH, Matyszak MK, Gaston JS. Characterization of plasmacytoid dendritic cells in inflammatory arthritis synovial fluid. Rheumatology. (2004) 43:453–60. 10.1093/rheumatology/keh115
    1. Cavanagh LL, Boyce A, Smith L, Padmanabha J, Filgueira L, Pietschmann P, et al. . Rheumatoid arthritis synovium contains plasmacytoid dendritic cells. Arthritis Res Ther. (2005) 7:R230–40. 10.1186/ar1467
    1. Villani AC, Satija R, Reynolds G, Sarkizova S, Shekhar K, Fletcher J, et al. . Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. (2017) 356:eaah4573. 10.1126/science.aah4573
    1. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. . PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol. (2001) 2:261–8. 10.1038/85330
    1. Lesterhuis WJ, Steer H, Lake RA. PD-L2 is predominantly expressed by Th2 cells. Mol Immunol. (2011) 49:1–3. 10.1016/j.molimm.2011.09.014
    1. Messal N, Serriari NE, Pastor S, Nunès JA. Olive D, PD-L2 is expressed on activated human T cells and regulates their function. Mol Immunol. (2011) 48:2214–19. 10.1016/j.molimm.2011.06.436
    1. Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, O'Shea MA, et al. . The common gamma-chain cytokines IL-2, IL-7, IL-15, and IL-21 induce the expression of programmed death-1 and its ligands. J Immunol. (2008) 181:6738–46. 10.4049/jimmunol.181.10.6738
    1. Zhong X, Tumang JR, Gao W, Bai C, Rothstein TL. PD-L2 expression extends beyond dendritic cells/macrophages to B1 cells enriched for V(H)11/V(H)12 and phosphatidylcholine binding. Eur J Immunol. (2007) 37:2405–10. 10.1002/eji.200737461
    1. Guignant C, Lepape A, Huang X, Kherouf H, Denis L, Poitevin F, et al. . Programmed death-1 levels correlate with increased mortality, nosocomial infection and immune dysfunctions in septic shock patients. Crit Care. (2011) 15:R99. 10.1186/cc10112
    1. Loke P, Allison JP. PD-L1 and PD-L2 are differentially regulated by Th1 and Th2 cells. Proc Natl Acad Sci USA. (2003) 100:5336–41. 10.1073/pnas.0931259100
    1. Nziza N, Jeziorski E, Delpont M, Cren M, Chevassus H, Carbasse A, et al. . Synovial-fluid miRNA signature for diagnosis of juvenile idiopathic arthritis. Cells. (2019) 8:1521. 10.3390/cells8121521

Source: PubMed

3
Subskrybuj