Skip to main content
  • Research article
  • Open access
  • Published:

CD14+CD16 monocytes are the main precursors of osteoclasts in rheumatoid arthritis via expressing Tyro3TK

Abstract

Background

Monocytes as precursors of osteoclasts in rheumatoid arthritis (RA) are well demonstrated, while monocyte subsets in osteoclast formation are still controversial. Tyro3 tyrosine kinase (Tyro3TK) is a member of the receptor tyrosine kinase family involved in immune homeostasis, the role of which in osteoclast differentiation was reported recently. This study aimed to compare the osteoclastic capacity of CD14+CD16+ and CD14+CD16 monocytes in RA and determine the potential involvement of Tyro3TK in their osteoclastogenesis.

Methods

Osteoclasts were induced from CD14+CD16+ and CD14+CD16 monocyte subsets isolated from healthy control (HC) and RA patients in vitro and evaluated by tartrate-resistant acid phosphatase (TRAP) staining. Then, the expression of Tyro3TK on CD14+CD16+ and CD14+CD16 monocyte subsets in the peripheral blood of RA, osteoarthritis (OA) patients, and HC were evaluated by flow cytometry and qPCR, and their correlation with RA patient clinical and immunological features was analyzed. The role of Tyro3TK in CD14+CD16 monocyte-mediated osteoclastogenesis was further investigated by osteoclast differentiation assay with Tyro3TK blockade.

Results

The results revealed that CD14+CD16 monocytes were the primary source of osteoclasts. Compared with HC and OA patients, the expression of Tyro3TK on CD14+CD16 monocytes in RA patients was significantly upregulated and positively correlated with the disease manifestations, such as IgM level, tender joint count, and the disease activity score. Moreover, anti-Tyro3TK antibody could inhibit Gas6-mediated osteoclast differentiation from CD14+CD16 monocytes in a dose-dependent manner.

Conclusions

These findings indicate that elevated Tyro3TK on CD14+CD16 monocytes serves as a critical signal for osteoclast differentiation in RA.

Background

Rheumatoid arthritis (RA) is one of the most common chronic systemic inflammatory rheumatic disease hallmarked by synovitis, aggressive lesions of the articular cartilage and bone, which leads to irreversible joint deformity and loss of function [1,2,3]. Bone erosion is the main pathological change in RA, which can even be observed in more than 45% of RA patients at an early stage [4]. It has been proved that excessive activation of local osteoclasts is involved in focal bone erosion in RA [5]. Osteoclasts are multinucleated cells which derived from the monocyte/macrophage lineage, especially from CD14+ monocytes [6].

Monocytes are plastic cells that can differentiate into macrophages, dendritic cells, and osteoclasts, which can accumulate in the blood and continuously migrate to inflammatory joints. Expanded monocytes in RA patients can lead to chronic joint inflammation and bone destruction [7]. Recently, based on differential surface expression of CD14 and CD16, human monocytes could be subdivided into two major subsets: CD14+CD16+ and CD14+CD16 monocytes, accounting for 5–10% and 90–95% of monocytes in healthy individuals, respectively [8].

However, the role of CD14+CD16+ and CD14+CD16 monocytes in osteoclast formation is still controversial. Bolzoni et al. demonstrated that bone marrow CD14+CD16+ monocytes from patients with multiple myeloma tended to differentiate into osteoclasts more remarkably than CD14+CD16 monocytes [9]. Chiu et al. also suggested that CD16+ monocytes from psoriatic arthritis patients were more prone to differentiate to osteoclasts [10]. In contrast, several studies illustrated that the osteoclasts were mainly derived from the CD14+CD16 monocytes in healthy donors [10,11,12]. Komano et al. further demonstrated that CD14+CD16 monocytes rather than CD14+CD16+ monocytes were the circulating osteoclast precursors in RA recently [11]. The different microenvironments of diseases would shape the phenotype of monocyte subsets and influence their capacity of osteoclast differentiation. In particular, studies have shown that multiple myeloma cells could profoundly modify the immune functions of the bone marrow cells as well as the bone marrow microenvironment [13, 14]. All these suggest that peripheral blood monocyte subsets may be directly involved in exacerbated osteoclast formation in RA. However, which monocyte subsets are the major sources of osteoclasts remains elusive.

Tyro3 tyrosine kinase (Tyro3TK) is one of the family members of TAM (Tyro3TK, AxlTK, MerTK) receptor tyrosine kinases (RTKs) [15], which could be expressed on the plasma membrane of a variety of cells, such as monocytes/macrophages, dendritic cells, NK cells, and nerve cells [16]. Tyro3TK could regulate the clearance of apoptotic cells, cytokine production, cell proliferation, thrombus formation, and hematopoiesis by binding to its ligand growth arrest-specific protein 6 (Gas6) and protein S (ProS1) [17, 18]. It was reported that Gas6 is expressed in RA synovium tissue and fluid and plays a role in RA synovium endothelial cell survival [19]. Furthermore, the expression of Gas6 appears to be stimulated by an inflammatory response, since elevated serum Gas6 levels were shown in sepsis and other systemic inflammation [20].

In 1998, Nakamura et al. firstly identified that Tyro3TK could be expressed in multinucleated osteoclasts, and the bone resorption activity of mature osteoclasts can be enhanced when binding with the ligand Gas6. However, Tyro3TK did not affect the differentiation of osteoclasts from bone marrow cells [21]. Katagiri et al. also found that Tyro3TK can be detected in mature osteoclasts while they showed that Gas6 demonstrated no apparent effect on osteoclast formation in mouse osteoclast progenitor cells [22]. Kawaguchi et al. found that Tyro3TK can only be detected in mouse mature osteoclasts among bone cells, while Gas6 is widely expressed in bone cells, stimulating the function of osteoclasts [23]. Recently, Ruiz-Heiland et al. illustrated that Tyro3TK-deficient mice showed an increased bone mass and impaired osteoclast differentiation in the arthritis model, suggesting the involvement of Tyro3TK in the differentiation and functional maturation of osteoclasts [24]. All these indicated that Tyro3TK might play a critical role in bone destruction in inflammatory arthritis. Despite these findings, the expression and osteogenic function of Tyro3TK on monocyte subsets in RA remain largely unknown.

In this study, we compared the osteoclastic capacity of CD14+CD16+ and CD14+CD16 monocytes in RA and determined the expression levels as well as the potential involvement of Tyro3TK in their osteoclastogenesis, aiming to further understand the mechanism of RA bone destruction.

Methods

Patients and controls

Fifty-seven patients with RA (Table 1), 28 osteoarthritis (OA) patients, and 49 age- and sex-matched healthy controls (HC) were enrolled in this study. All the patients met the 2010 American College of Rheumatology (ACR) revised criteria for RA [25] and 1986 ACR criteria for OA [26]. The study was approved by the Institutional Medical Ethics Review Board of Peking University People’s Hospital. Moreover, all participants provided informed consent.

Table 1 Demographic and clinical characteristics of RA patients

Clinical and laboratory indices of RA

The following data of patients with RA were recorded: gender, age, duration, swollen joint count (SJC), tender joint count (TJC), and laboratory parameters including white blood cells (WBC), red blood cells (RBC), hemoglobin (Hb), platelets (PLT), immunoglobulin (Ig) A, IgG, IgM, anti-cyclic citrullinated peptide antibody (anti-CCP antibody), erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP). Disease activity scores were calculated using the 28-joint Disease Activity Score-erythrocyte sedimentation rate (DAS28-ESR) in patients with RA. DAS28-ESR > 5.1 was considered a high disease activity according to the recommendations from the European League Against Rheumatism (EULAR).

Antibodies and reagents

Recombinant human macrophage colony-stimulating factor (rhM-CSF) (Cat# 300-25) was obtained from PerproTech GmbH (Rocky Hill, CT). Recombinant human RANKL (rhRANKL) (Cat# 390-TN), recombinant human Gas6 (rhGas6) (Cat# 885-GSB), human anti-Tyro3TK antibody (Cat# MAB859, Clone# 96201) proved to demonstrate blocking activity [27], human Tyro3TK PE-conjugated antibody (Cat# FAB859P), and mouse IgG2b PE-conjugated antibody (Cat# IC0041P) were purchased from R&D Systems (Minneapolis, MN). Human TruStain FcX™ (Fc Receptor Blocking Solution) (Cat# 422302) was purchased from BioLegend (San Diego, CA). Human CD14 FITC-conjugated antibody (Cat# 11-0141-81) and human CD16 APC-conjugated antibody (Cat# 17-0168-42) were purchased from eBioscience (San Diego, CA). The Leukocyte Acid Phosphatase Kit (Cat# 387A) was purchased from Sigma-Aldrich (St. Louis, MO). α-Minimum Essential Medium (α-MEM) (Cat# C11965500BT), 1% penicillin/streptomycin, and fetal bovine serum were purchased from Invitrogen (Carlsbad, CA).

Flow cytometry analysis and sorting

Peripheral blood mononuclear cells (PBMCs) were isolated from fresh EDTA blood samples using Ficoll density gradient centrifugation. Before staining with antibodies, single-cell suspensions were incubated with human Fc Receptor Blocking Solution for 10 min at room temperature to block the FcR-involved unwanted staining without interfering with antibody-mediated specific staining.

To detect the expression of Tyro3TK on CD14+CD16+ and CD14+CD16 monocytes, cells were stained with CD14 FITC-conjugated antibody, CD16 APC-conjugated antibody, and Tyro3TK PE-conjugated antibody. Corresponding negative isotype and fluorochrome-matched control (FMO) staining were also performed. The cells were then analyzed on FACS Aria II.

For CD14+CD16+ and CD14+CD16 monocyte sorting, cells were stained with CD14 FITC-conjugated antibody and CD16 APC-conjugated antibody. Then, the stained cells were sorted with FACS Aria II. The purified CD14+CD16+ and CD14+CD16 monocytes were further analyzed after sorting, the purity of which used for experiments was ~ 90%.

qPCR analysis of Tyro3TK expression

Total RNA was isolated from purified CD14+CD16 monocytes using the RNeasy mini kit (Qiagen, Hilden) then reverse transcribed into the oligo (dT)-primed cDNA by Revert Aid First Strand kit (Fermentas, Glen Burnie, MD). Real-time quantitative PCR (qPCR) was performed to analyze the expression of Tyro3TK mRNA in CD14+CD16 monocytes from RA patients and HC according to the manufacturer’s instructions. The sequences of the primers used in this study were as follows: the forward GAPDH primer was 5′-AAGGTGAAGGTCGGAGTCAA-3′, the reverse GAPDH primer was 5′-AATGAAGGGGTCATTGATGG-3′, the forward Tyro3TK primer was 5′-CAGCCGGTGAAGCTCAACT-3′, and the reverse Tyro3TK primer was 5′-TGGCACACCTTCTACCGTGA-3′.

In vitro osteoclast differentiation

CD14+CD16+ and CD14+CD16 monocytes from freshly isolated PBMCs were purified by FACS sorting. Then, the cells were cultivated 17 days separately in 96-well plates (5 × 104 cells/200 μl per well) in α-MEM with 1% PenStrep, 10% heat-inactivated fetal bovine serum, 30 ng/ml rhM-CSF, and 50 ng/ml rhRANKL. Different concentrations of rhGas6 and/or human anti-Tyro3TK antibody were added as indicated. The medium was changed with fresh medium every 6 days. Osteoclast differentiation was evaluated by staining cells for TRAP using a Leukocyte Acid Phosphatase kit (Sigma-Aldrich) according to the manufacturer’s instructions. TRAP-positive multinucleated cells were counted by an inverted fluorescence microscope (Olympus IX71-141, Tokyo, Japan).

Statistical analysis

All data were analyzed on the statistical software program SPSS 24.0 for Windows (SPSS, Chicago, IL). Differences between the groups were evaluated by Student’s t test, non-parametric Mann-Whitney U test, one-way ANOVA test, Kruskal-Wallis H test, and Spearman’s correlation test. P value less than 0.05 was considered statistically significant (*P < 0.05, ** P < 0.01, *** P < 0.001; ns, not significant).

Results

CD14+CD16 monocytes are the main precursors of osteoclasts in RA

To reveal which monocyte subset plays a significant role in osteoclast formation in RA, we performed osteoclast differentiation assay with monocyte subpopulation in vitro. CD14+CD16+ and CD14+CD16 monocytes were isolated from 5 HC and 5 RA patients by FACS sorting, respectively, the purity of which was confirmed by FACS (Fig. 1). Then osteoclast differentiation and TRAP staining were performed. Interestingly, the results showed that the number of TRAP-positive osteoclasts differentiated from CD14+CD16 monocytes were much more than that from CD14+CD16+ monocytes in HC (Fig. 2a). Moreover, CD14+CD16 monocytes demonstrated upregulated capacity of osteoclast differentiation in RA patients (Fig. 2b). However, there was no distinct difference for CD14+CD16+ monocytes between RA patients and HC (Fig. 2c).

Fig. 1
figure 1

Gating strategy for flow cytometry sorting of human CD14+CD16+ and CD14+CD16 monocytes. Peripheral blood mononuclear cells from RA and HC were stained with FITC-conjugated anti-CD14 antibody and APC-conjugated anti-CD16 antibody. CD14+CD16+ and CD14+CD16 monocytes were analyzed and sorted by flow cytometric; the purity of sorted CD14+CD16+ and CD14+CD16 monocytes used for experiments was ~ 90%

Fig. 2
figure 2

CD14+CD16 monocytes are the main osteoclast precursors in RA. Purified CD14+CD16+ and CD14+CD16 monocytes from RA (n = 5) and HC (n = 5) were cultured with rhM-CSF (30 ng/ml) and rhRANKL (50 ng/ml) for osteoclast differentiation. The cells were detected for tartrate-resistant acid phosphatase (TRAP) staining on day 17, and the TRAP-positive multinuclear cells were osteoclasts. The representative charts and the statistical results were shown. a CD14+CD16+ versus CD14+CD16 monocytes in HC (*P = 0.026). b RA versus HC for CD14+CD16 monocytes (*P = 0.019). c RA versus HC for CD14+CD16+ monocytes. *P < 0.05; ns, not significant (Student’s t test, ac)

Expression of Tyro3TK is enriched on CD14+CD16 monocytes and upregulated in RA patients

Then, we tried to reveal the effects of Tyro3TK on monocyte subset-mediated osteoclast differentiation. The expression of Tyro3TK on monocyte subsets in RA patients, OA patients, and HC were first analyzed and presented as mean fluorescence intensity (MFI). The gating strategy was demonstrated in Fig. 3a. We identified that there was no apparent difference in the expression of Tyro3TK on CD14+CD16+ and CD14+CD16 monocytes in HC and OA (Fig. 3b, c). Interestingly, the expression of Tyro3TK on CD14+CD16 monocytes in patients with RA was significantly higher than that of CD14+CD16+ monocytes (Fig. 3d). Moreover, the expression of Tyro3TK on CD14+CD16 monocytes was significantly increased in RA patients as compared with OA patients and HC. However, no significant difference was found for Tyro3TK expression on CD14+CD16+ monocytes between RA patients, OA patients, and HC (Fig. 3e, f). To further confirm our findings, we also detected the mRNA expression of Tyro3TK by qPCR. As shown in Fig. 3g, the results revealed that compared with HC, RA patient CD14+CD16 monocytes expressed significantly higher levels of Tyro3TK transcripts.

Fig. 3
figure 3

The expression of Tyro3TK on CD14+CD16 monocytes is increased in RA. a Gating strategy for identifying the expression of Tyro3TK on CD14+CD16+ and CD14+CD16 monocytes. Accordingly, the expression of Tyro3TK on CD14+CD16+ and CD14+CD16 monocytes in HC (n = 40) (b), OA (n = 28) (c), and RA patients (n = 40, **P = 0.008) (d) were analyzed and presented as the mean fluorescence intensity (MFI). e The expression of Tyro3TK on CD14+CD16+ monocytes were compared between HC, OA, and RA patients. f The expression of Tyro3TK on CD14+CD16 monocytes were compared between HC, OA, and RA patients (**P = 0.004, ***P < 0.001). g Flow cytometry-sorted CD14+CD16 monocytes from RA (n = 4) and HC (n = 4) were set to detect the mRNA expression of Tyro3TK by qPCR (*P = 0.029). *P < 0.05, **P < 0.01, ***P < 0.001; ns, not significant (Mann-Whitney U test, b, d, and g; Student’s t test, c; Kruskal-Wallis test followed by Dunn’s post-test for multiple comparisons, ef)

Tyro3TK on CD14+CD16 monocytes are associated with RA patient clinical and immunological features

Then, we analyzed the correlation of Tyro3TK on CD14+CD16+ and CD14+CD16 monocytes with RA patient clinical and immunological features, respectively. The results revealed substantial associations (Table 2). Notably, the levels of Tyro3TK on CD14+CD16 monocytes were found to be positively correlated with DAS28-ESR, TJC, and serum IgM (Fig. 4a–c). Detailed analyses showed that RA patients with high disease activity (DAS28-ESR > 5.1) showed higher levels of Tyro3TK on CD14+CD16 monocytes (Fig. 4d). Similar results were also seen in RA patients with tender joints and RF positivity (Fig. 4e, f). However, no apparent association was found between the levels of Tyro3TK on CD14+CD16 monocytes and RA patient’s gender, anti-CCP, or swollen joints (Fig. 4g–i).

Table 2 Correlation of Tyro3TK expression on monocyte subsets with RA patient clinical and immunological features
Fig. 4
figure 4

Correlation analysis of Tyro3TK on CD14+CD16 monocytes with RA patient clinical manifestations. The associations of Tyro3TK on CD14+CD16 monocytes with RA patient DAS28-ESR (r = 0.323, *P = 0.042) (a), tender joint counts (TJC) (r = 0.514, **P = 0.001) (b), and IgM (r = 0.432, **P = 0.005) (c) were analyzed. The expression of Tyro3TK on CD14+CD16 monocytes were also compared between the different RA patient groups: d RA with high disease activity (DAS28-ESR > 5.1) and non-high disease activity (DAS28-ESR ≤ 5.1) (*P = 0.034), e RA with and without tender joints (*P = 0.031), f rheumatoid factor (RF) positive and negative RA (*P = 0.024), g anti-CCP antibody positive and negative RA, h male and female RA, and i RA with and without swollen joints. *P < 0.05, **P < 0.01; ns, not significant (Spearman’s rank correlation test, ac; Mann-Whitney U test, di)

Upregulated Tyro3TK on CD14+CD16 monocytes promotes their osteoclast differentiation in RA

To further illustrate the osteoclast-priming effects of Tyro3TK on CD14+CD16 monocytes in RA patients, we performed osteoclast differentiation assay with or without Tyro3TK blockade. As shown in Fig. 5a, the co-culture of CD14+CD16 monocytes isolated from RA patients with rhGas6 promoted TRAP-positive osteoclast formation, especially at the dose of 50 ng/ml. Strikingly, anti-Tyro3TK antibody significantly compromised this rhGas6-mediated exacerbation of osteoclast differentiation in a dose-dependent manner. At the dose of 200 ng/ml, anti-Tyro3TK antibody could almost abolish the formation of osteoclasts (Fig. 5b). Collectively, these results revealed the critical role of Tyro3TK in mediating CD14+CD16 monocyte differentiation into osteoclasts.

Fig. 5
figure 5

Tyro3TK promotes CD14+CD16 monocyte-mediated osteoclastogenesis in RA. Purified CD14+CD16 monocytes from RA patients (n = 8) were cultured with rhM-CSF (30 ng/ml) and rhRANKL (50 ng/ml) under different conditions for osteoclast differentiation. Seventeen days later, the cells were harvested for TRAP staining. The representative charts and the statistical results were shown. a Different concentrations of rhGas6 (0 ng/ml, 10 ng/ml, 50 ng/ml, and 100 ng/ml) were supplemented for osteoclast differentiation (n = 3 per group, **P = 0.006). b Different concentrations of anti-Tyro3TK antibody (0 ng/ml, 50 ng/ml, 100 ng/ml, and 200 ng/ml), and 50 ng/ml rhGas6 was supplemented for osteoclast differentiation (n = 5 per group, ***P < 0.001). **P < 0.01, ***P < 0.001 (one-way ANOVA test followed by Dunn’s post-test for multiple comparisons, a, b)

Discussion

In this study, we found that CD14+CD16 monocytes were more potent in osteoclast differentiation in HC, the capacity of which was more powerful in RA patients. The expression of Tyro3TK on CD14+CD16 monocytes were upregulated in RA, positively correlating with the clinical features of the patients. Moreover, upregulated Tyro3TK on CD14+CD16 monocytes promotes their osteoclast differentiation in RA.

Peripheral blood monocytes played an essential role in secreting inflammatory factors, regulating innate immunity, and inducing osteoclast formation [28]. Monocyte heterogeneity has been recognized in humans for a long time. Based on phenotypic characteristics, human monocytes can be divided into CD14+CD16+ and CD14+CD16 monocytes, and the CD14+CD16+ monocytes can be further divided into non-classical (CD14+CD16++) and intermediate (CD14++CD16+) monocytes [7]. In this study, we mainly focus on the role of CD14+CD16+ and CD14+CD16 monocytes in osteoclast formation with Tyro3TK expression. Nevertheless, the different roles of non-classical and intermediate monocytes in osteoclastogenesis, as well as the involvement of Tyro3TK are of significance, which will be revealed in our future study. In addition, it should be noticed that there is also a specific subset of DCs derived from monocytes known to express CD14 but not CD16 (named Mo-DC) [29]. Our result showed that the frequencies of Mo-DC in CD14+CD16 cells were ~ 1.54% (data not shown). This might induce minimal interference to the current results yet could not be excluded, and the role of Mo-DC in osteoclastogenesis deserves to be further studied.

CD14+CD16+ and CD14+CD16 monocyte subsets might possess different functions in RA. Our previous study showed that CD14+CD16+ monocytes in patients with systemic lupus erythematosus showed inflammatory phenotype, with increased CD80, CD86, HLA-DR, and CX3CR1, which could promote Th17 response [30]. IL-17 is a pro-inflammatory cytokine mainly produced by CD4+ T cells and plays a critical role in RA synovitis [31]. Kotake et al. illustrated that the level of cytokine IL-17 was significantly increased in RA synovial fluid, and IL-17 could promote osteoclast differentiation from CD14+ monocytes [32]. CD14+CD16+ monocytes can also migrate to RA synovium and produce high levels of TNF-α, IL-6, and IL-1β. These cytokines could promote the production of cytokine IL-17, thus playing a critical role in synovial inflammation and osteoclasts formation [33,34,35]. Here, we showed that CD14+CD16 monocytes were more prone to differentiate into osteoclasts than CD14+CD16+ monocytes in healthy controls. Moreover, the osteoclastic capacity of CD14+CD16 monocytes was significantly enhanced in RA patients. Although with controversial, these results were consistent with most previous studies [10,11,12]. Therefore, we speculate that CD14+CD16 monocytes are the main osteoclast precursors in RA, while CD14+CD16+ monocytes are more competent in producing pro-inflammatory cytokines. Detailed mechanistic studies are still needed to reveal the differential functions of these two monocyte subsets.

Tyro3TK was initially discovered as a therapeutic target in tumors [36]. Increasing studies have focused on their critical role in autoimmune diseases [37, 38]. Barth et al. demonstrated that Tyro3TK could express in monocytes [39]. As the ligand of Tyro3TK, Gas6 was evaluated in RA synovium tissue and fluid [19]. It can promote RA synovial hyperplasia, which is hallmarked by the abundant synovial fibroblasts and associated with bone destruction in RA [40]. Besides, Gas6-Tyro3TK interaction may play a critical osteoclast-priming role [21,22,23,24]. In this study, we showed that Tyro3TK on CD14+CD16 monocytes of RA patients was significantly upregulated, which was associated with clinical features and disease activity. Furthermore, Gas6 can promote the osteoclasts formation of CD14+CD16 monocytes, while disrupts Gas6-Tyro3TK interaction, the number of osteoclasts differentiated from CD14+CD16 monocytes decreased significantly with a dose-dependent anti-Tyro3TK antibody. The study also extends our findings, demonstrating that Tyro3TK has a distinct role in regulating CD14+CD16 monocyte osteoclastogenesis, suggesting that Tyro3TK might be a possible therapeutic target for RA bone destruction. Therefore, it is intriguing to propose that targeting Tyro3TK and CD14+CD16 monocytes simultaneously may have a more apparent inhibitory effect on bone destruction in RA. However, the detailed signal mechanisms of Tyro3TK on CD14+CD16 in RA need to be further studied.

Conclusion

In summary, this study reveals that CD14+CD16 monocytes are the main precursors of osteoclasts in RA. Moreover, upregulated Tyro3TK expression on these cells provides a pivotal role for osteoclastogenesis, which might serve as therapeutic targets for the persistent disease.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Abbreviations

RA:

Rheumatoid arthritis

OA:

Osteoarthritis

HC:

Healthy control

TRAP:

Tartrate-resistant acid phosphatase

Gas6:

Growth arrest-specific protein 6

ProS1:

Protein S

RTKs:

Receptor tyrosine kinases

ACR:

American College of Rheumatology

EULAR:

European League Against Rheumatism

SJC:

Swollen joint count

TJC:

Tender joint count

WBC:

White blood cells

RBC:

Red blood cells

Hb:

Hemoglobin

PLT:

Platelets

IgA/G/M:

Immunoglobulin

A/G/M:

Anti-CCP antibody, anti-cyclic citrullinated peptide antibody

ESR:

Erythrocyte sedimentation rate

CRP:

C-reactive protein

PBMCs:

Peripheral blood mononuclear cells

PBS:

Phosphate-buffered saline

FMO:

Fluorochrome-matched controls

MFI:

Mean fluorescence intensity

α-MEM:

α-Minimum Essential Medium

M-CSF:

Macrophage colony-stimulating factor

RANKL:

Nuclear factor-κB ligand

References

  1. Smolen JS, Aletaha D, Barton A, Burmester GR, Emery P, Firestein GS, Kavanaugh A, McInnes IB, Solomon DH, Strand V, et al. Rheumatoid arthritis. Nat Rev Dis Primers. 2018;4:18001.

    PubMed  Google Scholar 

  2. McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. N Engl J Med. 2011;365(23):2205–19.

    CAS  Google Scholar 

  3. Firestein GS. Evolving concepts of rheumatoid arthritis. Nature. 2003;423(6937):356–61.

    CAS  PubMed  Google Scholar 

  4. Adamopoulos IE, Mellins ED. Alternative pathways of osteoclastogenesis in inflammatory arthritis. Nat Rev Rheumatol. 2015;11(3):189–94.

    CAS  PubMed  Google Scholar 

  5. Okamoto K, Nakashima T, Shinohara M, Negishi-Koga T, Komatsu N, Terashima A, Sawa S, Nitta T, Takayanagi H. Osteoimmunology: the conceptual framework unifying the immune and skeletal systems. Physiol Rev. 2017;97(4):1295–349.

    CAS  PubMed  Google Scholar 

  6. Massey HM, Flanagan AM. Human osteoclasts derive from CD14-positive monocytes. Br J Haematol. 1999;106(1):167–70.

    CAS  PubMed  Google Scholar 

  7. Rana AK, Li Y, Dang Q, Yang F. Monocytes in rheumatoid arthritis: circulating precursors of macrophages and osteoclasts and, their heterogeneity and plasticity role in RA pathogenesis. Int Immunopharmacol. 2018;65:348–59.

    CAS  PubMed  Google Scholar 

  8. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, Leenen PJ, Liu YJ, MacPherson G, Randolph GJ, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116(16):e74–80.

    CAS  PubMed  Google Scholar 

  9. Bolzoni M, Ronchetti D, Storti P, Donofrio G, Marchica V, Costa F, Agnelli L, Toscani D, Vescovini R, Todoerti K, et al. IL21R expressing CD14+CD16+ monocytes expand in multiple myeloma patients leading to increased osteoclasts. Haematologica. 2017;102(4):773–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Chiu YG, Shao T, Feng C, Mensah KA, Thullen M, Schwarz EM, Ritchlin CT. CD16 (FcRgammaIII) as a potential marker of osteoclast precursors in psoriatic arthritis. Arthritis Res Ther. 2010;12(1):R14.

    PubMed  PubMed Central  Google Scholar 

  11. 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(5):R152.

    PubMed  PubMed Central  Google Scholar 

  12. Lari R, Kitchener PD, Hamilton JA. The proliferative human monocyte subpopulation contains osteoclast precursors. Arthritis Res Ther. 2009;11(1):R23.

    PubMed  PubMed Central  Google Scholar 

  13. Noll JE, Williams SA, Tong CM, Wang H, Quach JM, Purton LE, Pilkington K, To LB, Evdokiou A, Gronthos S, et al. Myeloma plasma cells alter the bone marrow microenvironment by stimulating the proliferation of mesenchymal stromal cells. Haematologica. 2014;99(1):163–71.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Terpos E, Ntanasis-Stathopoulos I, Gavriatopoulou M, Dimopoulos MA. Pathogenesis of bone disease in multiple myeloma: from bench to bedside. Blood Cancer J. 2018;8(1):7.

    PubMed  PubMed Central  Google Scholar 

  15. Lemke G. Phosphatidylserine is the signal for TAM receptors and their ligands. Trends Biochem Sci. 2017;42(9):738–48.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Rothlin CV, Carrera-Silva EA, Bosurgi L, Ghosh S. TAM receptor signaling in immune homeostasis. Annu Rev Immunol. 2015;33:355–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhou J, Yang A, Wang Y, Chen F, Zhao Z, Davra V, Suzuki-Inoue K, Ozaki Y, Birge RB, Lu Q, et al. Tyro3, Axl, and Mertk receptors differentially participate in platelet activation and thrombus formation. Cell Commun Signal. 2018;16(1):98.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Peeters MJW, Rahbech A, Thor Straten P. TAM-ing T cells in the tumor microenvironment: implications for TAM receptor targeting. Cancer Immunol Immunother. 2020;69(2):237–44.

    CAS  PubMed  Google Scholar 

  19. O’Donnell K, Harkes IC, Dougherty L, Wicks IP. Expression of receptor tyrosine kinase Axl and its ligand Gas6 in rheumatoid arthritis: evidence for a novel endothelial cell survival pathway. Am J Pathol. 1999;154(4):1171–80.

    PubMed  PubMed Central  Google Scholar 

  20. Hurtado B, de Frutos PG. GAS6 in systemic inflammatory diseases: with and without infection. Crit Care. 2010;14(5):1003.

    PubMed  PubMed Central  Google Scholar 

  21. Nakamura YS, Hakeda Y, Takakura N, Kameda T, Hamaguchi I, Miyamoto T, Kakudo S, Nakano T, Kumegawa M, Suda T. Tyro 3 receptor tyrosine kinase and its ligand, Gas6, stimulate the function of osteoclasts. Stem Cells. 1998;16(3):229–38.

    CAS  PubMed  Google Scholar 

  22. Katagiri M, Hakeda Y, Chikazu D, Ogasawara T, Takato T, Kumegawa M, Nakamura K, Kawaguchi H. Mechanism of stimulation of osteoclastic bone resorption through Gas6/Tyro 3, a receptor tyrosine kinase signaling, in mouse osteoclasts. J Biol Chem. 2001;276(10):7376–82.

    CAS  PubMed  Google Scholar 

  23. Kawaguchi H, Katagiri M, Chikazu D. Osteoclastic bone resorption through receptor tyrosine kinase and extracellular signal-regulated kinase signaling in mature osteoclasts. Mod Rheumatol. 2004;14(1):1–5.

    PubMed  Google Scholar 

  24. Ruiz-Heiland G, Zhao Y, Derer A, Braun T, Engelke K, Neumann E, Mueller-Ladner U, Liu Y, Zwerina J, Schett G. Deletion of the receptor tyrosine kinase Tyro3 inhibits synovial hyperplasia and bone damage in arthritis. Ann Rheum Dis. 2014;73(4):771–9.

    CAS  PubMed  Google Scholar 

  25. Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham CO 3rd, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD, et al. 2010 rheumatoid arthritis classification criteria: an American College of Rheumatology/European League Against Rheumatism collaborative initiative. Arthritis Rheum. 2010;62(9):2569–81.

    PubMed  Google Scholar 

  26. Altman R, Asch E, Bloch D, Bole G, Borenstein D, Brandt K, Christy W, Cooke TD, Greenwald R, Hochberg M, et al. Development of criteria for the classification and reporting of osteoarthritis. Classification of osteoarthritis of the knee. Diagnostic and Therapeutic Criteria Committee of the American Rheumatism Association. Arthritis Rheum. 1986;29(8):1039–49.

    CAS  PubMed  Google Scholar 

  27. Jiang L, Chen XQ, Gao MJ, Lee W, Zhou J, Zhao YF, Wang GD. The Pros1/Tyro3 axis protects against periodontitis by modulating STAT/SOCS signalling. J Cell Mol Med. 2019;23(4):2769–81.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Kikuta J, Ishii M. Osteoclast migration, differentiation and function: novel therapeutic targets for rheumatic diseases. Rheumatology (Oxford). 2013;52(2):226–34.

    CAS  Google Scholar 

  29. Tang-Huau TL, Segura E. Human in vivo-differentiated monocyte-derived dendritic cells. Semin Cell Dev Biol. 2019;86:44–9.

    CAS  PubMed  Google Scholar 

  30. Zhu H, Hu F, Sun X, Zhang X, Zhu L, Liu X, Li X, Xu L, Shi L, Gan Y, et al. CD16(+) monocyte subset was enriched and functionally exacerbated in driving T-cell activation and B-cell response in systemic lupus erythematosus. Front Immunol. 2016;7:512.

    PubMed  PubMed Central  Google Scholar 

  31. van Hamburg JP, Corneth OB, Paulissen SM, Davelaar N, Asmawidjaja PS, Mus AM, Lubberts E. IL-17/Th17 mediated synovial inflammation is IL-22 independent. Ann Rheum Dis. 2013;72(10):1700–7.

    PubMed  Google Scholar 

  32. Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh K, Ishiyama S, Saito S, Inoue K, Kamatani N, Gillespie MT, et al. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest. 1999;103(9):1345–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Amoruso A, Sola D, Rossi L, Obeng JA, Fresu LG, Sainaghi PP, Pirisi M, Brunelleschi S. Relation among anti-rheumatic drug therapy, CD14+CD16+ blood monocytes and disease activity markers (DAS28 and US7 scores) in rheumatoid arthritis: a pilot study. Pharmacol Res. 2016;107:308–14.

    CAS  PubMed  Google Scholar 

  34. 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(7):3536–42.

    CAS  PubMed  Google Scholar 

  35. Yoon BR, Yoo SJ, Choi Y, Chung YH, Kim J, Yoo IS, Kang SW, Lee WW. Functional phenotype of synovial monocytes modulating inflammatory T-cell responses in rheumatoid arthritis (RA). PLoS One. 2014;9(10):e109775.

    PubMed  PubMed Central  Google Scholar 

  36. Smart SK, Vasileiadi E, Wang X, DeRyckere D, Graham DK. The emerging role of TYRO3 as a therapeutic target in cancer. Cancers (Basel). 2018;10(12).

  37. Pagani S, Bellan M, Mauro D, Castello LM, Avanzi GC, Lewis MJ, Sainaghi PP, Pitzalis C, Nerviani A. New insights into the role of Tyro3, Axl, and Mer receptors in rheumatoid arthritis. Dis Markers. 2020;2020:1614627.

    PubMed  PubMed Central  Google Scholar 

  38. Rothlin CV, Lemke G. TAM receptor signaling and autoimmune disease. Curr Opin Immunol. 2010;22(6):740–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Barth ND, Marwick JA, Heeb MJ, Gale AJ, Rossi AG, Dransfield I. Augmentation of human monocyte responses to lipopolysaccharide by the protein S and Mer/Tyro3 receptor tyrosine kinase axis. J Immunol. 2018;201(9):2602–11.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Danks L, Komatsu N, Guerrini MM, Sawa S, Armaka M, Kollias G, Nakashima T, Takayanagi H. RANKL expressed on synovial fibroblasts is primarily responsible for bone erosions during joint inflammation. Ann Rheum Dis. 2016;75(6):1187–95.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the patients with RA and OA and healthy donors included in the study.

Funding

The study was supported by grants from the National Natural Science Foundation of China (81671609 and 81871290 to Dr. Y. Su; 81971523 and 81671604 to Dr. F. Hu), the Beijing Science and Technology Planning Project (Z191100006619111 to Dr. Y. Su), the Beijing Municipal Natural Science Foundation (7194329 to Dr. L. Xu), and the Beijing Nova Program (Z181100006218044 to Dr. F. Hu), as well as by the Peking University People’s Hospital Research and Development Funds (RDF2019-03 to Dr. F. Hu).

Author information

Authors and Affiliations

Authors

Contributions

Performed the experiments: JM.X. and LL.X. Analyzed the data: HQ.Z. and X.L. Contributed reagents/materials/analysis tools: MX.B., Z.Z., H.Z., G.C., and X.L. Wrote the manuscript: JM.X. and LL.X. Conceived the study, reviewed, and edited the manuscript: FL.H. and Y.S. The authors read and approved the final manuscript.

Corresponding authors

Correspondence to Fanlei Hu or Yin Su.

Ethics declarations

Ethics approval and consent to participate

All subjects gave written informed consent under the Declaration of Helsinki. The protocol was approved by the Institutional Medical Ethics Review Board of Peking University People’s Hospital.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xue, J., Xu, L., Zhu, H. et al. CD14+CD16 monocytes are the main precursors of osteoclasts in rheumatoid arthritis via expressing Tyro3TK. Arthritis Res Ther 22, 221 (2020). https://doi.org/10.1186/s13075-020-02308-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13075-020-02308-7

Keywords