CD16 (FcRgammaIII) as a potential marker of osteoclast precursors in psoriatic arthritis

Yahui Grace Chiu, Tianmeng Shao, Changyong Feng, Kofi A Mensah, Michael Thullen, Edward M Schwarz, Christopher T Ritchlin, Yahui Grace Chiu, Tianmeng Shao, Changyong Feng, Kofi A Mensah, Michael Thullen, Edward M Schwarz, Christopher T Ritchlin

Abstract

Introduction: Psoriatic arthritis (PsA) is a chronic inflammatory arthritis characterized by bone erosion mediated by osteoclasts (OC). Our previous studies showed an elevated frequency of OC precursors (OCP) in PsA patients. Here, we examined if OC arise from CD16-positive monocytes in PsA.

Methods: Peripheral blood mononuclear cells (PBMC) or monocytes were isolated from human peripheral blood and sorted based on CD16 expression. Sorted cells were cultured alone or with bone wafers in the presence of receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). Enumeration and bone erosion activity of OC were examined after culture. The effects of tumor necrosis factor-alpha (TNFalpha), OC-promoting (M-CSF plus RANKL), and dendritic cell (DC)-promoting (GM-CSF plus interleukin (IL)-4) cytokines on CD16 surface expression were examined by flow cytometry.

Results: PsA and psoriasis (Ps) subjects had a higher percentage of circulating inflammatory CD14+CD16+ cells than healthy controls (HC). Exposure of cells to OC-promoting, but not DC-promoting media, was associated with CD16 up-regulation. PBMC of Ps and PsA had a higher frequency of cells expressing intermediate levels of CD16. OC were mainly derived from CD16+ cells in PsA. Increased CD16 expression was associated with a higher bone erosion activity in PsA.

Conclusions: An increased frequency of circulating CD14+CD16+ cells was noted in PsA compared to controls, and intermediate levels of CD16 may suggest a transitional state of OCP during osteoclastogenesis. Intriguingly, TNFalpha blocked CD16 expression on a subset of CD14+ monocytes. Collectively, our data suggest that CD16 has the potential to serve as an OCP marker in inflammatory arthritis.

Figures

Figure 1
Figure 1
Ps and PsA patients have a higher percentage of CD14+CD16+ cells. Human peripheral blood mononuclear cells (PBMC) were isolated from peripheral blood, stained with an antibody cocktail composed of CD14-APC, CD16-PE, 7AAD, and analyzed by flow cytometry. Dead cells were excluded by 7AAD+. (a) Classical (CD14+CD16-, red line) and non-classical (CD14+CD16+, blue line) monocytes were labeled based on the classification by Strauss-Ayali and colleagues [5]. (b) The percentage of CD14+CD16+ cells in the PBMC of 16 healthy controls (HC), and 29 psoriasis (Ps), 28 psoriatic arthritis (PsA), and 8 rheumatoid arthritis (RA) patients. (c) The percentage of CD14+CD16+ cells in enriched human monocytes from HC and PsA. Monocytes were enriched by the Human Monocyte Enrichment Cocktail. The percentage of CD14+CD16+ cells in enriched monocytes from HC (2.6 ± 1.6%) and PsA patients (10.3 ± 9.5%) are shown in [a] and [b], respectively. The data are representative of 10 independent experiments.
Figure 2
Figure 2
Cytokines alter the cell surface expression of CD16 and human CD14+ cells undergo a transitional stage of CD16 up-regulation in OC-promoting culture conditions. (a) Enriched human monocytes were cultured in osteoclast (OC)-promoting media (receptor activator of nuclear factor kappa-B ligand (RANKL) + macrophage colony-stimulating factor (M-CSF), pink line) or dendritic cell (DC)-promoting media (IL-4 + granulocyte-macrophage colony-stimulating factor (GM-CSF), green line). Freshly isolated monocytes (blue line) and the isotype control (black line) are also shown. Surface expression of CD16 was monitored by FACS analysis on [a] day 0, [b] day 1, and [c] day 3, respectively. (b) Human peripheral blood mononuclear cells (PBMC) were cultured in OC-promoting media (RANKL + M-CSF) and the cell surface expression of CD14 and CD16 was monitored on [a] day 0, [b] day 3, and [c] day 5. Data shown here are live cells after forward scatter/side scatter (FSC/SSC) gating followed by dead cell exclusion using 7-amino-actinomycin D (AAD). Numbers shown in each quadrant are the percentage of total gated cells.
Figure 3
Figure 3
Cells that express intermediate levels of CD16 (CD16int) are found in vivo in human PBMC. (a) CD16 expression on human peripheral blood mononuclear cells (PBMC) is heterogeneous. The data shown here are the representatives of CD16 expression patterns on fresh human PBMC from 82 subjects. [a] PBMC that expressed high and low levels of CD16 without the CD16int population. [b to d] PBMC that have CD16int populations with different CD16 expression levels. (b) Ex vivo analysis of CD14 and CD16 expression. Representative examples of individuals [a] without a CD16int population, [b] CD14- CD16int (arrow) cells and [c] CD14+ CD16int (arrow) cells are shown. In total, 16 healthy control (HC), 29 psoriasis (Ps), 29 psoriatic arthritis (PsA), and 8 rheumatoid arthritis (RA)patients were included in this analysis. See Table 1 for the summary of CD16int percentage in HC, Ps, PsA and RA groups.
Figure 4
Figure 4
CD16- and CD16+ cells are the major reservoirs of OC precursors in HC and PsA patients, respectively. (a) 30 to 50% of MHCII+CD16+ cells are CD14+. Human peripheral blood mononuclear cells (PBMC) were isolated from the peripheral blood and stained with an antibody cocktail composed of MHCII-FITC, CD14-APC, CD16-PE, 7AAD. The gating strategy is sequentially illustrated from a to d. [a] PBMC were gated by FSC/SSC; [b] live cells were gated by 7AAD; [c] dot plot analysis of MHCII and CD16 expression on live cells; [d] the expression of CD14 on MHCII+CD16+ cells gated from c. Red line is the isotype control and blue line is staining from CD14-APC. The number shown in the histogram represents the frequency of CD14+ cells per total MHCII+CD16+ cells. [e] The relative percentage of CD14+CD16+ to MHCII+CD16+ cells. Sample shown here is the representative of 3 healthy controls (HC). (b) Human PBMC from HC and psoriatic arthritis (PsA) patients were isolated, stained with antibodies, and sterile sorted into three populations, MHCII+CD16-, MHCII+CD16int, and MHCII+CD16+. Osteoclastogenesis was examined by osteoclast (OC) count after eight-day culture in the presence of receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) (MHCII+CD16-: purple, MHCII+CD16int: turquoise, MHCII+CD16+: orange). Subject numbers 1 to 3 are HC and numbers 4 to 8 are PsA patients.
Figure 5
Figure 5
OC derived from MHCII+CD16+ and MHCII+CD16- cells of HC and PsA patients have distinct phenotypes. Peripheral blood mononuclear cells (PBMC) isolated from (a and b) psoriatic arthritis (PsA) and (c and d) healthy controls (HC) and were sterile sorted into (a and c) MHCII+CD16+ and (b and d) MHCII+CD16- populations. Sorted cells were cultured in the presence of receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) for eight days, and tartrate-resistant acid phosphatase stained for osteoclast (OC) quantification. Arrows indicate mature OC. The details of differences in OC generation potential and numbers of nuclei from these sorted subsets are summarized in Table 4.
Figure 6
Figure 6
CD14+cells from HC and PsA respond differently to TNFα in regards to CD16 cell surface expression. The expression of CD16 on freshly isolated peripheral blood mononuclear cells (PBMC) from healthy control (HC) and psoriatic arthritis (PsA) was shown in (a) and (b), respectively. After PBMC were cultured in the (c and e) absence or (d and f) presence of TNFα for three days, cell surface expression of CD16 was examined again using the same antibody cocktail composed of anti-CD16 (PE), anti-CD14 (APC), anti-CD3 (Pacific Blue), and anti-CD19 (APC-Cy7) antibodies with 7AAD. The anti-CD3, anti-CD19 antibodies and 7AAD were included to exclude T cells, B cells, and dead cells, respectively. Numbers shown in each quadrant (c to f) represent the percentage of cells per total gated live cells. Data shown here are the representative of two HC and three PsA samples.
Figure 7
Figure 7
Elevated CD16 surface expression was associated with increased bone erosion activity in PsA monocytes. Sterile sorted MHCII+CD16+ and MHCII+CD16int cells were co-cultured with bone slices in the presence of receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) for 14 days. (a and b) Osteoclast (OC) were identified by tartrate-resistant acid phosphatase (TRAP) staining, and (c and d) bone erosion pits were visualized after toluidine blue staining.
Figure 8
Figure 8
CD16+ cells have a higher bone resorption activity than CD16int cells. Sterile sorted MHCII+CD16+ and MHCII+CD16int cells were co-cultured with bone slices in the presence of receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) for 14 days. (a) Quantification of bone pits eroded by MHCII+CD16int and MHCII+CD16+ cells. (b) Reconstructed three-dimensional micro-CT images of the bone wafer incubated with MHCII+CD16int [a] and MHCII+CD16+ [b] sorted cells. The data are representative of three independent sorting experiments.

References

    1. Kane D, Pathare S. Early psoriatic arthritis. Rheum Dis Clin North Am. 2005;31:641–657. doi: 10.1016/j.rdc.2005.07.009.
    1. Teitelbaum SL, Ross FP. Genetic regulation of osteoclast development and function. Nat Rev Genet. 2003;4:638–649. doi: 10.1038/nrg1122.
    1. Ritchlin CT, Haas-Smith SA, Li P, Hicks DG, Schwarz EM. Mechanisms of TNF-alpha- and RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis. J Clin Invest. 2003;111:821–831.
    1. Ziegler-Heitbrock HW. Heterogeneity of human blood monocytes: the CD14+ CD16+ subpopulation. Immunol Today. 1996;17:424–428. doi: 10.1016/0167-5699(96)10029-3.
    1. Strauss-Ayali D, Conrad SM, Mosser DM. Monocyte subpopulations and their differentiation patterns during infection. J Leukoc Biol. 2007;82:244–252. doi: 10.1189/jlb.0307191.
    1. Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leukoc Biol. 2007;81:584–592. doi: 10.1189/jlb.0806510.
    1. Fingerle G, Pforte A, Passlick B, Blumenstein M, Strobel M, Ziegler-Heitbrock HW. The novel subset of CD14+/CD16+ blood monocytes is expanded in sepsis patients. Blood. 1993;82:3170–3176.
    1. Todd I, Radford PM, Ziegler-Heitbrock L, Ghaemmaghami AM, Powell RJ, Tighe PJ. Elevated CD16 expression by monocytes from patients with tumor necrosis factor receptor-associated periodic syndrome. Arthritis Rheum. 2007;56:4182–4188. doi: 10.1002/art.23133.
    1. Takeyama N, Yabuki T, Kumagai T, Takagi S, Takamoto S, Noguchi H. Selective expansion of the CD14(+)/CD16(bright) subpopulation of circulating monocytes in patients with hemophagocytic syndrome. Ann Hematol. 2007;86:787–792. doi: 10.1007/s00277-007-0332-4.
    1. Nockher WA, Scherberich JE. Expanded CD14+ CD16+ monocyte subpopulation in patients with acute and chronic infections undergoing hemodialysis. Infect Immun. 1998;66:2782–2790.
    1. Baeten D, Boots AM, Steenbakkers PG, Elewaut D, Bos E, Verheijden GF, Berheijden G, Miltenburg AM, Rijnders AW, Veys EM, De Keyser F. Human cartilage gp-39+,CD16+ monocytes in peripheral blood and synovium: correlation with joint destruction in rheumatoid arthritis. Arthritis Rheum. 2000;43:1233–1243. doi: 10.1002/1529-0131(200006)43:6<1233::AID-ANR6>;2-9.
    1. Wu W, Zhang X, Zhang C, Tang T, Ren W, Dai K. Expansion of CD14+CD16+ peripheral monocytes among patients with aseptic loosening. Inflamm Res. 2009;58:561–570. doi: 10.1007/s00011-009-0020-z.
    1. Belge KU, Dayyani F, Horelt A, Siedlar M, Frankenberger M, Frankenberger B, Espevik T, Ziegler-Heitbrock L. The proinflammatory CD14+CD16+DR++ monocytes are a major source of TNF. J Immunol. 2002;168:3536–3542.
    1. Perussia B. Fc receptors on natural killer cells. Curr Top Microbiol Immunol. 1998;230:63–88.
    1. Langlet C, Bernard AM, Drevot P, He HT. Membrane rafts and signaling by the multichain immune recognition receptors. Curr Opin Immunol. 2000;12:250–255. doi: 10.1016/S0952-7915(00)00084-4.
    1. Pinheiro da Silva F, Aloulou M, Skurnik D, Benhamou M, Andremont A, Velasco IT, Chiamolera M, Verbeek JS, Launay P, Monteiro RC. CD16 promotes Escherichia coli sepsis through an FcR gamma inhibitory pathway that prevents phagocytosis and facilitates inflammation. Nat Med. 2007;13:1368–1374. doi: 10.1038/nm1665.
    1. da Silva FP, Aloulou M, Benhamou M, Monteiro RC. Inhibitory ITAMs: a matter of life and death. Trends Immunol. 2008;29:366–373. doi: 10.1016/j.it.2008.05.001.
    1. Moll JM, Wright V. Psoriatic arthritis. Semin Arthritis Rheum. 1973;3:55–78. doi: 10.1016/0049-0172(73)90035-8.
    1. Yano R, Yamamura M, Sunahori K, Takasugi K, Yamana J, Kawashima M, Makino H. Recruitment of CD16+ monocytes into synovial tissues is mediated by fractalkine and CX3CR1 in rheumatoid arthritis patients. Acta Med Okayama. 2007;61(2):89–98.
    1. Ancuta P, Rao R, Moses A, Mehle A, Shaw SK, Luscinskas FW, Gabuzda D. Fractalkine preferentially mediates arrest and migration of CD16+ monocytes. J Exp Med. 2003;197:1701–1707. doi: 10.1084/jem.20022156.
    1. Gordon S, Taylor PR. Monocyte and macrophage heterogeneity. Nat Rev Immunol. 2005;5(12):953–964. doi: 10.1038/nri1733.
    1. Schwarz EM, Looney RJ, Drissi MH, O'Keefe RJ, Boyce BF, Xing L, Ritchlin CT. Autoimmunity and bone. Ann N Y Acad Sci. 2006;1068:275–283. doi: 10.1196/annals.1346.031.
    1. Moreno JL, Mikhailenko I, Tondravi MM, Keegan AD. IL-4 promotes the formation of multinucleated giant cells from macrophage precursors by a STAT6-dependent, homotypic mechanism: contribution of E-cadherin. J Leukoc Biol. 2007;82:1542–1553. doi: 10.1189/jlb.0107058.
    1. Komano Y, Nanki T, Hayashida K, Taniguchi K, Miyasaka N. Identification of a human peripheral blood monocyte subset that differentiates into osteoclasts. Arthritis Res Ther. 2006;8:R152. doi: 10.1186/ar2046.
    1. Skinner NA, MacIsaac CM, Hamilton JA, Visvanathan K. Regulation of Toll-like receptor (TLR)2 and TLR4 on CD14dimCD16+ monocytes in response to sepsis-related antigens. Clin Exp Immunol. 2005;141:270–278. doi: 10.1111/j.1365-2249.2005.02839.x.
    1. Zhang Q, Guo R, Lu Y, Zhao L, Zhou Q, Schwarz EM, Huang J, Chen D, Jin ZG, Boyce BF, Xing L. VEGF-C, a lymphatic growth factor, is a RANKL target gene in osteoclasts that enhances osteoclastic bone resorption through an autocrine mechanism. J Biol Chem. 2008;283:13491–13499. doi: 10.1074/jbc.M708055200.
    1. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol. 2008;8:958–969. doi: 10.1038/nri2448.
    1. Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schakel K. The CD16(+) (FcgammaRIII(+)) subset of human monocytes preferentially becomes migratory dendritic cells in a model tissue setting. J Exp Med. 2002;196:517–527. doi: 10.1084/jem.20011608.
    1. Lari R, Kitchener PD, Hamilton JA. The proliferative human monocyte subpopulation contains osteoclast precursors. Arthritis Res Ther. 2009;11:R23. doi: 10.1186/ar2616.
    1. Pinheiro da Silva F, Aloulou M, Benhamou M, Monteiro RC. Inhibitory ITAMs: a matter of life and death. Trends Immunol. 2008;29:366–373. doi: 10.1016/j.it.2008.05.001.
    1. Ivashkiv LB. Cross-regulation of signaling by ITAM-associated receptors. Nat Immunol. 2009;10:340–347. doi: 10.1038/ni.1706.
    1. Humphrey MB, Daws MR, Spusta SC, Niemi EC, Torchia JA, Lanier LL, Seaman WE, Nakamura MC. TREM2, a DAP12-associated receptor, regulates osteoclast differentiation and function. J Bone Miner Res. 2006;21:237–245. doi: 10.1359/JBMR.051016.
    1. Humphrey MB, Lanier LL, Nakamura MC. Role of ITAM-containing adapter proteins and their receptors in the immune system and bone. Immunol Rev. 2005;208:50–65. doi: 10.1111/j.0105-2896.2005.00325.x.
    1. Koga T, Inui M, Inoue K, Kim S, Suematsu A, Kobayashi E, Iwata T, Ohnishi H, Matozaki T, Kodama T, Taniguchi T, Takayanagi H, Takai T. Costimulatory signals mediated by the ITAM motif cooperate with RANKL for bone homeostasis. Nature. 2004;428:758–763. doi: 10.1038/nature02444.
    1. Helming L, Tomasello E, Kyriakides TR, Martinez FO, Takai T, Gordon S, Vivier E. Essential role of DAP12 signaling in macrophage programming into a fusion-competent state. Sci Signal. 2008;1:ra11. doi: 10.1126/scisignal.1159665.
    1. Humphrey MB, Ogasawara K, Yao W, Spusta SC, Daws MR, Lane NE, Lanier LL, Nakamura MC. The signaling adapter protein DAP12 regulates multinucleation during osteoclast development. J Bone Miner Res. 2004;19:224–234. doi: 10.1359/JBMR.0301234.
    1. Barber DF, Bartolomé A, Hernandez C, Flores JM, Redondo C, Fernandez-Arias C, Camps M, Rückle T, Schwarz MK, Rodríguez S, Martinez-A C, Balomenos D, Rommel C, Carrera AC. PI3Kgamma inhibition blocks glomerulonephritis and extends lifespan in a mouse model of systemic lupus. Nat Med. 2005;11:933–935.
    1. Kramer PR, Winger V, Reuben J. PI3K limits TNF-alpha production in CD16-activated monocytes. Eur J Immunol. 2009;39:561–570. doi: 10.1002/eji.200838801.
    1. Ancuta P, Wang J, Gabuzda D. CD16+ monocytes produce IL-6, CCL2, and matrix metalloproteinase-9 upon interaction with CX3CL1-expressing endothelial cells. J Leukoc Biol. 2006;80(5):1156–1164. doi: 10.1189/jlb.0206125.

Source: PubMed

3
Subscribe