Next Article in Journal
S1P Stimulates Erythropoietin Production in Mouse Renal Interstitial Fibroblasts by S1P1 and S1P3 Receptor Activation and HIF-2α Stabilization
Next Article in Special Issue
Pleiotropic Roles of NOTCH1 Signaling in the Loss of Maturational Arrest of Human Osteoarthritic Chondrocytes
Previous Article in Journal
Chemotherapy-Induced Intestinal Microbiota Dysbiosis Impairs Mucosal Homeostasis by Modulating Toll-like Receptor Signaling Pathways
Previous Article in Special Issue
Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Chondrocytes from Osteoarthritis Patients Adopt Distinct Phenotypes in Response to Central TH1/TH2/TH17 Cytokines

1
The Immunopharmacology Research Group, Faculty of Medicine and Health Technology, University of Tampere and Tampere University Hospital, 33100 Tampere, Finland
2
Coxa Hospital for Joint Replacement, 33520 Tampere, Finland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(17), 9463; https://doi.org/10.3390/ijms22179463
Submission received: 30 June 2021 / Revised: 30 July 2021 / Accepted: 6 August 2021 / Published: 31 August 2021
(This article belongs to the Special Issue Osteoarthritis: From Molecular Pathways to Therapeutic Advances)

Abstract

:
Chronic low-grade inflammation plays a central role in the pathogenesis of osteoarthritis (OA), and several pro- and anti-inflammatory cytokines have been implicated to mediate and regulate this process. Out of these cytokines, particularly IFNγ, IL-1β, IL-4 and IL-17 are associated with different phenotypes of T helper (TH) cells and macrophages, both examples of cells known for great phenotypic and functional heterogeneity. Chondrocytes also display various phenotypic changes during the course of arthritis. We set out to study the hypothesis of whether chondrocytes might adopt polarized phenotypes analogous to TH cells and macrophages. We studied the effects of IFNγ, IL-1β, IL-4 and IL-17 on gene expression in OA chondrocytes with RNA-Seq. Chondrocytes were harvested from the cartilage of OA patients undergoing knee replacement surgery and then cultured with or without the cytokines for 24 h. Total RNA was isolated and sequenced, and GO (Gene Ontology) functional analysis was performed. We also separately investigated genes linked to OA in recent genome wide expression analysis (GWEA) studies. The expression of more than 2800 genes was significantly altered in chondrocytes treated with IL-1β [in the C(IL-1β) phenotype] with a fold change (FC) > 2.5 in either direction. These included a large number of genes associated with inflammation, cartilage degradation and attenuation of metabolic signaling. The profile of genes differentially affected by IFNγ (the C(IFNγ) phenotype) was relatively distinct from that of the C(IL-1β) phenotype and included several genes associated with antigen processing and presentation. The IL-17-induced C(IL-17) phenotype was characterized by the induction of a more limited set of proinflammatory factors compared to C(IL-1β) cells. The C(IL-4) phenotype induced by IL-4 displayed a differential expression of a rather small set of genes compared with control, primarily those associated with TGFβ signaling and the regulation of inflammation. In conclusion, our results show that OA chondrocytes can adopt diverse phenotypes partly analogously to TH cells and macrophages. This phenotypic plasticity may play a role in the pathogenesis of arthritis and open new therapeutic avenues for the development of disease-modifying treatments for (osteo)arthritis.

1. Introduction

Osteoarthritis (OA) is the most common form of arthritis. It has been estimated to affect up to a half of the elderly population, and therefore causes widespread disability and human suffering as well as an immense burden to healthcare systems [1]. Once thought as a mostly mechanical “wear and tear” disease, the chronic inflammatory component of osteoarthritis has been increasingly recognized during recent decades [2]. Constant low-grade inflammation in the joint contributes to pain, oxidative stress, increased catabolism, and the eventual breakdown of articular cartilage [3,4]. Despite intense research, no disease-modifying pharmacological treatments are currently available for OA [5], demonstrating that our understanding of the pathogenesis of the disease remains limited.
When comparing chondrocytes from OA patients with healthy cells, several changes in gene expression can be observed [6,7]. The potential causal roles of these changes in the pathogenesis of OA are currently largely unknown. However, some of them can be considered harmful (such as secretion of catabolic enzymes and proinflammatory cytokines) and others protective (e.g., the production of extracellular matrix [ECM] components) [8,9]. The changes in OA chondrocyte phenotype are thought to be caused by several physical and chemical factors, among them local proinflammatory cytokines [10].
The T helper (TH) cell is probably the most well-known example of a cell capable of adopting distinct phenotypes in response to environmental factors. The different TH phenotypes, in turn, are associated with different cytokines. The TH1 phenotype drives inflammation and defense against intracellular pathogens. These cells are induced by interleukin 12 (IL-12) and produce mainly interferon gamma (IFNγ) as an effector cytokine [11]. In addition, they induce macrophages to produce IL-1β, which in turn promotes the proinflammatory effects of TH1 cells [12]. TH2 cells are induced by interleukins 2 and 4. They secrete various factors that promote humoral immunity and regulate inflammation, of which IL-4 is regarded as the central cytokine [11]. TH17 cells are most closely associated with autoimmunity; they are induced by transforming growth factor beta (TGFβ) along with several proinflammatory cytokines, such as interleukins 6, 21 and 23, and they produce IL-17 as the central effector [13].
The macrophage is another cell type with well-defined differential phenotypes. The so-called “macrophage polarization” has two main phenotypes analogous to TH1 and TH2. The proinflammatory or “classically activated” M1 phenotype is associated with proinflammatory cytokines such as IL-1β and IFNγ, while the healing-promoting “alternatively activated” M2 phenotype is mainly linked to IL-4 [14]. The effects of IL-17 on macrophage phenotype have also attracted considerable interest. The M17 phenotype is not as well-defined as the M1 and M2 phenotypes; however, macrophages stimulated by IL-17 are characterized by the increased production of chemotactic and proinflammatory factors in the initial stages of the inflammatory response [15] and by the clearance of apoptotic cells and resolution of inflammation in the later phase [16].
Some authors have noted similarities between the variable functions and gene expression profiles of macrophages and chondrocytes in the setting of arthritis [17]. As another intriguing observation, major TH1/2/17 cytokines have been shown to play roles in the development of different forms of arthritis. Of the cytokines that have been implicated in the development of OA, IL-1β is probably the most prominent. It has been shown to decrease the anabolic activity in chondrocytes and promote their apoptosis [18]. It also induces the expression of the proteolytic enzymes of the matrix metalloproteinase (MMP) and a disintegrin-like and metalloproteinase with trombospondin motifs (ADAMTS) families [19]. OA chondrocytes have been shown to upregulate the expression of IL-1 receptor (IL-1R) increasing their sensitivity to this cytokine [20]. Despite this, systemic treatment strategies specifically targeting IL-1β seem to have rather limited efficacy in OA [21], and none have reached clinical use.
Another major proinflammatory cytokine playing a role in the pathogenesis of arthritis is interleukin 17A (IL-17A) [22]. It promotes inflammation in concert with other proinflammatory cytokines [23], and its concentration in the synovial fluid correlates with radiographic severity of joint destruction [24]. In chondrocytes, it induces proinflammatory and catabolic factors and reduces proteoglycan synthesis [25,26,27]. Along with other proinflammatory cytokines, it also increases bone degradation by activating RANK ligand (RANKL) in osteoclasts [28]. In a murine model of collagen-induced arthritis, IL-17 deficiency has been shown to protect joints from the disease and IL-17 overexpression to exacerbate it [29,30]. Some functional gene expression analyses have actually implicated IL-17 signaling as a pathophysiological factor over IL-1β, the cytokine long known to drive OA [31].
In contrast to IL-1β and IL-17, the potential role of IFNγ as a causative factor in OA has attracted less interest. However, it has been found to be upregulated in chondrocytes by proinflammatory cytokines [32] as well as to be present in OA synovial fluid [33]. Some gene variants that affect the development of OA, particularly those of T-cell immunoglobulin and mucin-domain containing-3 (TIM-3), exert their effects via the modulation of IFNγ expression [34].
In the light of the above connections between the cytokines linked to major T helper cell/macrophage phenotypes and OA, it can be hypothesized that chondrocytes might also adopt phenotypes analogous to TH1/2/17 or M1/2/17 cells, and that these phenotypes might play a role in the development of OA. In the present study, we investigated the effects of the central TH1/2/17 cytokines on gene expression in OA chondrocytes. We sought to identify significantly differentially expressed genes and modulated pathways. The results were also compared to those of a recent genome-wide association study comparing degraded OA cartilage to preserved cartilage [35]. To our knowledge, this is the first study comparing the effects of the central TH1/2/17 cytokines on OA chondrocytes and to characterize the resulting phenotypes.

2. Results

2.1. Effects of IL-1β on Chondrocyte Phenotype

After normalization and correction for multiple testing, a total of 2822 genes were found to be differentially expressed in IL-1β-treated chondrocytes [in the C(IL-1β) phenotype] versus controls in a statistically significant manner (FDR-corrected p-value < 0.05) and with a fold change (FC) 2.5 or more in either direction. Of these, 1092 were up- and 1730 downregulated. The list of the 20 most strongly upregulated genes contains several proinflammatory cyto- and chemokines, while the most strongly downregulated ones include several factors associated with regulation of gene expression, such as histone proteins (Table 1).

2.2. Effects of IL-17 on Chondrocyte Phenotype

Three hundred and eighty genes were differentially expressed in IL-17-treated chondrocytes [in the C(IL-17) phenotype] versus controls with FC > 2.5 in either direction, 314 of which were up- and 66 downregulated. Among the 20 most strongly upregulated genes were several associated with inflammation and chemotaxis, while the most strongly downregulated include genes involved in connective tissue development (Table 2).

2.3. Effects of IFNγ on Chondrocyte Phenotype

After normalization and correction for multiple testing, a total of 548 genes were found to be differentially expressed in IFNγ-treated chondrocytes [in the C(IFNγ) phenotype] versus controls in a statistically significant manner and FC 2.5 or more in either direction. Of these, 462 were up- and 86 downregulated. The 20 genes most strongly upregulated in C(IFNγ) cells included many associated with inflammation, antigen processing and presentation, and the regulation of proliferation. The most strongly downregulated genes included those involved in cell adhesion, proliferation and migration, and in Wnt signaling (Table 3).

2.4. Effects of IL-4 on Chondrocyte Phenotype

Twenty-six genes were upregulated by IL-4 with FC > 2.5 (Table S1). No genes were downregulated by IL-4 to a similar extent, but 10 genes were downregulated with FC < −1.5 (Table S2). In the C(IL-4) phenotype, the upregulated genes included those associated with the regulation of inflammation and TGFβ signaling as well as metabolism and cell adhesion, while several genes linked to cell proliferation were among the downregulated ones.

2.5. Functional Gene Categories in Different Chondrocyte Phenotypes

Table 4 shows the Gene Ontology (GO) terms affected with a high significance (FDR-corrected p-value < 0.01) by at least one studied proinflammatory cytokine (IL-1β, IFNγ or IL-17). The C(IL-1β) phenotype was involved in the activation of a wide range of inflammatory terms and pathways, along with those related to cell adhesion as well as extracellular matrix production and degradation. The TH17-associated cytokine IL-17 affected a partly overlapping, but smaller, set of inflammatory cytokines compared to IL-1β. The C(IFNγ) phenotype was quite distinct compared to the C(IL-1β) and C(IL-17) phenotypes; several terms related to antigen processing and presentation were affected by this cytokine alone. Nitric oxide synthase biosynthetic process and chemotaxis were among the functions involved solely in the C(IL-17) phenotype. In addition, many high-level GO terms related to inflammation were affected by all of the three proinflammatory cytokines.
In C(IL-4) cells, no significantly affected GO terms were detected when analyzing the genes with FC > 2.5 in either direction. When the FC threshold was lowered to 1.5, GO terms associated with cell division were among the significant ones (Table S3).

2.6. Comparing the Effects of Different Proinflammatory Cytokines

Next, we cross-compared the genes markedly upregulated (FC > 2.5) in the C(IL-1β), C(IFNγ) and C(IL-17) phenotypes to further characterize the differences and similarities between the resulting phenotypes. As shown in Figure 1A, a large portion (nearly 85%) of genes markedly upregulated in C(IL-17) cells were included in the large set of those similarly affected by IL-1β, but 45 genes were solely affected by IL-17, and the overlap of C(IL-17) and C(IFNγ) phenotypes was considerable smaller than that of C(IL-17) and C(IL-1β). The intersection of genes upregulated by both IL-17 and IFNγ was nearly completely contained in those upregulated by IL-1β (Figure 1A). Many central regulators of inflammation such as IL6, PTGS2 (cyclo-oxygenase 2 or COX-2) and NOS2 (inducible nitric oxide synthase or iNOS) were markedly upregulated by all the three TH1/TH17 cytokines, in line with the widespread activation of inflammatory pathways observed in the GO analysis (Table 5).
When comparing genes markedly downregulated (FC < −2.5) by the three proinflammatory cytokines, the large (>1000 genes) list of genes downregulated by IL-1β again contained a large proportion (85%) of those downregulated by IL-17 and a smaller amount (48%) of genes similarly affected by IFNγ (Figure 1B). Genes downregulated by all of the three cytokines are presented in Table 6 and include, for example, those associated with cell proliferation and skeletal system development.

2.7. Effects of the Cytokines on Genes Differentially Expressed in Degraded and Preserved OA Cartilage

Some previous studies have investigated the differences in gene expression between degraded and preserved OA cartilage. Of these, the study by Almeida et al. [35] is probably the most comprehensive. To see whether the studied cytokines shift chondrocyte phenotype towards either degraded or preserved cartilage, we compared the differentially expressed genes in the phenotypes observed in the present study to those differentially expressed in the study by Almeida et al. [35] As a very large number (over 2300) of significantly differentially expressed genes were identified in that study, we focused on those 84 genes which were most strongly upregulated (FC > 2.5 and FDR-corrected p-value < 0.01) in the degraded cartilage. Of those 84 genes, 38 were significantly affected by at least one of the proinflammatory cytokines (IL-1, IL-17 or IFNγ) in our data. A large majority (30) of these 38 genes were also upregulated by IL-1β, showing that the cytokine shifts chondrocyte phenotype towards the one observed in the degraded cartilage. Several mediators of inflammation, such as LIF, CCL20 and TREM1, were especially strongly upregulated. Only four of the 84 genes (namely CLIC3, ERFE, SLC27A2 and ANK3) were downregulated by IL-1β.
In the C(IFNγ) phenotype, 13 of the 84 genes associated with degraded cartilage (including LIF and NGF) were upregulated compared with control, but nearly as many (nine) were downregulated, including TREM1. This shows that the effects of IFNγ on chondrocyte phenotype in relation to the degraded/preserved cartilage are more ambiguous than those of IL-1β.
In C(IL-17) chondrocytes, 25 of the 84 genes associated with degraded cartilage were upregulated compared to naïve chondrocytes (including CCL20 and IL11), and none were significantly downregulated. Nine genes, including IGFBP1, LIF and GPR158, were upregulated in all three inflammatory phenotypes C(IL-1β), C(IFNγ) and C(IL-17) and one (ANK3) was downregulated in all of them. (Figure 2 and Table S4).
In the study by Almeida et al. [35], 52 genes were associated with preserved rather than degraded cartilage (i.e., significantly downregulated in degraded cartilage with FC < −2.5). Of these, 19 were significantly affected by at least one of the proinflammatory cytokines in our data. In C(IL-1β) cells, 13 of these 19 genes were significantly downregulated with GDF10 displaying especially strong downregulation. In contrast, five of these genes were upregulated compared to control (including the especially strongly upregulated C3 and RSPO3). This again shows that the net effect of IL-1β is to shift chondrocyte phenotype towards degraded cartilage. IFNγ showed a directionally similar, but less pronounced effect: seven of the genes associated with preserved cartilage were significantly downregulated and three upregulated in the C(IFNγ) phenotype. In C(IL-17) cells, eight genes associated with preserved cartilage were down- and four upregulated; C3 once again displayed especially strong upregulation. Five genes, including PTGER3 and GDF10, were downregulated in all of the three chondrocyte phenotypes. On the other hand, RSPO3 and PRLR, both downregulated in degraded compared with preserved cartilage, were upregulated by all of the three cytokines. These data indicate that the C(IL-1β) and C(IL-17) phenotypes at least partly resemble the transcriptomic profile associated with degraded OA cartilage as identified by Almeida et al. [35]. In contrast, IFNγ seems to have a smaller effect on the genes directly linked to cartilage degradation in OA being instead characterized by the upregulation of genes associated with antigen processing and presentation. (Figure 3 and Table S5).
Relatively few genes were significantly affected by IL-4 in our data, and none of them were markedly (with FC > 2.5) associated with either degraded or preserved cartilage in the data of Almeida et al. [35]. However, looking at genes with a smaller proportional difference between degraded and preserved cartilage (FC > 1.5 in either direction) produced several genes that were significantly affected by IL-4. Ten genes (including DUSP5 and COL7A1) were upregulated in degraded cartilage and also upregulated in C(IL-4) cells. In contrast, one gene associated with degraded cartilage (HMMR) was downregulated by IL-4, and seven genes (including COL14A1) associated with preserved cartilage were upregulated by IL-4. (Table S6)
To demonstrate that naïve chondrocytes can be affected by the cytokines studied, we separately studied the expression of their receptors. As shown in Table S7, receptors for all studied cytokines were expressed in unstimulated OA chondrocytes at meaningful levels.

3. Discussion

Chondrocytes from OA patients were found to adopt distinct phenotypes in response to the central TH1/TH2/TH17 cytokines. The phenotype induced by the TH1 cytokine interleukin 1 (IL-1β), the C(IL-1β) phenotype, can be characterized by widespread, strong upregulation of inflammation and catabolism as well as downregulation of metabolic signaling. The effects of the TH17 cytokine IL-17 appear to be somewhat less widespread and partly overlapping those of IL-1β, with induction of inflammatory and chemotactic factors. The phenotype induced by the second TH1 cytokine interferon gamma (IFNγ) seems to be distinct from both C(IL-1β) and C(IL-17) phenotypes, with a significant theme of antigen processing and presentation. The effects of the TH2 cytokine IL-4 were much more modest; some factors involved in the regulation of inflammation and TGFβ signaling were upregulated, while the downregulated genes were mostly associated with cell proliferation and migration.
In T cells, the TH1 phenotype drives inflammation and defense against intracellular pathogens (cell-mediated immunity) and is associated with the production of proinflammatory cytokines such as IFNγ and IL-1β [36]. Conversely, TH2 cells promote humoral immunity, regulate inflammation and direct resolving and injury-healing responses [11]. Central TH2 cytokines are IL-4 and IL-13. A third relatively well-established population of TH cells is the TH17 phenotype. These cells produce IL-17, drive autoimmune reactions and activate neutrophils. This contrasts with TH1 cells that preferentially affect monocytes/macrophages, as well as TH2 cells that are associated with eosinophils, basophils and mast cells [37].
The central TH1/TH2/TH17 cytokines also induce loosely analogous macrophage phenotypes. Like TH1 cells, M1 or “classically activated” macrophages are induced by proinflammatory cytokines such as IL-1β and IFNγ and promote inflammation by secreting further proinflammatory factors. M2 or “alternatively activated” macrophages are induced canonically by IL-4. In addition to functioning as antiparasite effectors, they attenuate inflammation, direct wound-healing processes and promote the resolution of inflammation. [38] IL-17 induces a less-studied macrophage phenotype characterized by increased chemotaxis and the production of proinflammatory factors such as cyclo-oxygenase 2 (COX-2), IL-6 and tumor necrosis factor alpha (TNFα) [15,39] as well as resolution-promoting effects in the later phases of inflammation [16].
The chondrocyte phenotypes induced by different cytokines in our study can be considered analogous to TH cell and particularly macrophage phenotypes. IL-1β affects a very large number of genes and induces a phenotype characterized by the expression of inflammatory and matrix-degrading genes. The C(IL-17) phenotype appears likewise proinflammatory, but with a somewhat more limited repertoire of inflammatory genes. C(IFNγ) also appears to be a phenotype that is inflammatory, but is also characterized by genes linked to antigen presentation. The C(IL-4) phenotype is characterized by the expression of genes linked to TGFβ signaling and the regulation of inflammation.
The chondrocyte phenotypes induced by the TH1/TH2/TH17 cytokines appeared to be quite distinct as only 45 genes were markedly (FC > 2.5) upregulated and eight markedly downregulated (FC < −2.5) by all three proinflammatory cytokines, considering that hundreds of genes were up- and dozens downregulated to a similar extent by each of the three cytokines. The factors upregulated by all of the three proinflammatory cytokines (IL-1β, IFNy and IL-17) include the well-known inflammatory mediators IL6, nitric oxide synthase 2/inducible nitric oxide synthase (NOS2/iNOS) and prostaglandin-endoperoxide synthase 2/cyclooxygenase 2 (PTGS2/COX-2). On this list were also included, for example, pentraxin 3 (PTX3), toll-like receptor 2 (TLR2), chemokine (C-C motif) ligand 2 (CCL2), interferon regulatory factor 4 (IRF4) and prolactin receptor (PRLR). Pentraxin 3 (PTX) promotes inflammation by activating the classical complement pathway and by facilitating antigen recognition by mononuclear phagocytes [40], and it has been shown to be elevated in the serum and synovial fluid of patients with rheumatoid arthritis [41]. TLR2 is a pattern recognition receptor mediating innate immune activation by microbial particles. In osteoarthritis, it is activated by hyaluronan and aggrecan fragments leading to the activation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, which may contribute to OA progression and pain [42,43]. CCL2 is a monocyte-attracting chemokine that has been linked to OA development and pain [44,45]. IRF4 has recently been associated with cartilage destruction and pain in OA via the induction of CCL17 [46]. Prolactin has been implicated to promote chondrocyte differentiation and attenuate apoptosis, and thus the upregulation of its receptor might promote cartilage survival [47,48].
Factors downregulated by all of the three proinflammatory cytokines include asporin (ASPN) and prostaglandin EP3 receptor (PTGER3). Asporin belongs to the family of leucine-rich repeat proteins and is associated with cartilage matrix, also bearing a similarity to decorin [49]. The potential role of asporin in OA appears to be unclear; several studies have linked the protein to the development of the disease, where it might impair chondrogenesis by inhibiting TGF-β signaling [50]. Polymorphisms of the asporin gene have also been linked to OA risk [51], even though the most recent meta-analysis failed to find evidence for this [52]. Prostaglandin E2 (PGE2)-induced PTGER3 downregulation may contribute to cartilage inflammation and damage via NF-κB activation and IL-6 synthesis [53].
When the Gene Ontology (GO) terms significantly affected by the three different proinflammatory cytokines were studied, all three were found to affect those associated with inflammation. IL-1β was alone in significantly affecting several terms, such as cell adhesion, extracellular matrix metabolism and collagen catabolism, linking the chondrocyte phenotype induced by this cytokine to these functions. IL-17 solely affected nitric oxide synthase biosynthesis. This is intriguing, as the nitric oxide production is an important part of inflammatory response in chondrocytes [54]. The C(IFNγ) phenotype seems to be differentiated from others by activation of pathways related to antigen processing and presentation. Chondrocytes are not considered “professional” antigen-presenting cells, but they have, interestingly, been shown to present cartilage proteoglycans as antigens to CD8+ T cells, potentially contributing to local joint inflammation [55,56].
Previously published genome-wide expression analyses (GWEAs) have identified a number of differentially expressed genes between either damaged and intact OA cartilage or healthy and OA cartilage. These include genes involved in inflammation, skeletal system development, cell adhesion and monosaccharide metabolism [35,57,58,59]. When comparing our results to those of the comprehensive study by Almeida et al. [35], the C(IL-1β) phenotype most closely resembled degraded OA cartilage, while IL-17 upregulated a smaller number of proinflammatory factors associated with degraded cartilage in that study. Accordingly, some genes associated with preserved as opposed to degraded cartilage were also downregulated by these proinflammatory cytokines. Most of these genes are linked to cartilage anabolism. The effects of IFNγ and (especially) IL-4 on the genes identified by Almeida et al. [35] were more modest. It is important to note that the receptors for all cytokines studied were expressed at marked levels in our samples, which lends further validity to our results.
A potential limitation of the study is that whole thickness pieces of cartilage obtained from joint replacement surgery were used for chondrocyte isolation. Thus, the cells obtained are likely a mixture of chondrocytes from different layers of cartilage, and there might be some differences in the effects of cytokines between these groups. However, all chondrocytes can be expected to be exposed to cytokines diffused from the synovial fluid and/or produced by chondrocytes (in autocrine or paracrine manner). Thus, we think that the observed clear differences in the chondrocyte phenotypes in response to the major TH1/TH2/TH17 cytokines are relevant for further understanding of chondrocyte biology and OA pathophysiology. In future studies, cartilage layer-specific cell isolation methods or single-cell RNA-Seq could be considered to unravel possible zone-specific responses.
Another limitation of the study is that the chondrocytes used were obtained from OA joints; therefore, some of the detected effects of the cytokines might differ from those observed in healthy chondrocytes. Studying the effects of the cytokines on healthy chondrocytes would be an interesting avenue of future study; however, obtaining healthy primary human chondrocytes presents a practical challenge (compared to OA chondrocytes which can be obtained from joint replacement surgery). In the present study, we observed similarities between the C(IL-1β) and C(Il-17) phenotypes and the gene expression profile of chondrocytes from degraded OA cartilage published by Almeida et al. [35]; C(IFNγ) and especially C(IL-4) bore less resemblance to that phenotype. This suggests that the cytokine-induced phenotypes observed in our data have relevance regarding OA pathogenesis.
In conclusion, OA chondrocytes, analogously to macrophages, can assume distinct phenotypes in response to the cytokines associated with the TH1/TH2/TH17 phenotypes of T helper cells. These results provide novel information on chondrocyte biology and the pathogenesis of OA with further insights into the development of disease-modifying drugs for (osteo)arthritis.

4. Materials and Methods

4.1. Cartilage and Cell Culture

Leftover cartilage pieces were collected from nine patients undergoing total knee replacement surgery in Coxa Hospital for Joint Replacement, Tampere, Finland. All patients fulfilled the American College of Rheumatology classification criteria for knee OA [60]. Patients with diabetes mellitus were excluded from the study to avoid potential confounding effects on chondrocyte metabolism [61]. The study was approved by the Ethics Committee of Tampere University Hospital, Finland, and carried out in accordance with the Declaration of Helsinki. Written informed consent was obtained from the patients. Chondrocyte isolation and culture was carried out as previously described [62]. To ensure an adequate yield of chondrocytes, all available cartilage was removed aseptically using a scalpel from the bony parts received from joint replacement surgery and cut into small pieces. The pieces were first washed with phosphate buffered saline (PBS). After that, they were incubated for 24 h in the presence of Liberase enzyme (Roche, Mannheim, Germany) 0.25 mg/mL, diluted in serumless Dulbecco’s modified Eagle’s medium (DMEM, Sigma-Aldrich, St Louis, MO, USA) with glutamax-I containing penicillin (100 units/mL), streptomycin (100 μg/mL), and amphotericin B (250 ng/mL) (all three from Invitrogen, Carlsbad, CA, USA) at 37 °C. The resulting cell suspension was poured through a 70 μm nylon mesh and centrifuged for five minutes at 200 g. Cells were then washed twice and seeded on 24-well plates (0.2 million cells/mL) in DMEM supplemented with 10% heat-inactivated fetal bovine serum (Lonza) together with the aforementioned compounds. Confluent cultures were exposed to fresh culture medium alone, with 10 ng/mL IFNγ, with 100 pg/mL IL-1β, with 50 ng/mL IL-17 or with 10 ng/mL IL-4, for 24 h. The concentrations used were chosen based on our preliminary experiments with cultured chondrocytes.

4.2. RNA Isolation and Sample Preparation

Culture medium was removed at the indicated time points and total RNA of the chondrocytes was extracted with GenElute Mammalian Total RNA Miniprep kit (Sigma-Aldrich). The sample was treated with DNAse I (Fermentas UAB, Vilnius, Lithuania). RNA concentration and integrity were confirmed with the 2100 Bioanalyzer (Agilent Technologies, Santa Clara, CA, USA).

4.3. Next Generation Sequencing and Data Analysis

Sequencing of samples was performed in the Finnish Institute of Molecular Medicine (FIMM) sequencing core, Helsinki, Finland, using the Illumina HiSeq 2500 sequencing platform. Sequencing depth was 20 million paired-end reads 100 bp in length. Read quality was first assessed using FastQC [63], and the reads were trimmed using Trimmomatic [64]. Trimmed reads were aligned to reference human genome with STAR [65]. Count matrices were prepared with the featureCounts program [66]. Differential expression was assessed with DESeq2 [67]. Gene expression levels were given as DeSeq2-normalized counts, and genes with an average normalized count 10 or less across all samples were excluded from further analysis. For the purposes of further analysis, genes with a minimum of 2.5 fold change (FC) in abundance and FDR-corrected p-value < 0.05 were deemed biologically and statistically significant (unless otherwise indicated). Functional analysis was performed against the Gene Ontology (GO) database [68,69] using the DAVID tool [70], and REVIGO was used to reduce the resulting list [71].

4.4. Statistics

For NGS data analysis, normalization was performed and differential expression studied using a negative binomial model implemented in DESeq2.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/ijms22179463/s1.

Author Contributions

Conceptualization, A.P., T.L., M.H., T.M. and E.M.; funding acquisition, A.P. and E.M.; investigation, A.P., T.L., M.H. and T.M.; methodology, A.P., T.L., M.H., T.M. and E.M.; project administration, T.M. and E.M.; resources, T.M. and E.M.; supervision, E.M.; validation, A.P., T.L. and M.H.; visualization, A.P.; writing—original draft, A.P.; writing—review and editing, A.P., T.L., M.H., T.M. and E.M. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by grants from the Finnish Society of Rheumatology, Tampere Rheumatism Foundation, the competitive research funding of Pirkanmaa Hospital District and the Scandinavian Rheumatology Research Foundation. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Institutional Review Board Statement

This study was approved by the Ethics Committee of Tampere University Hospital, Finland (ref# ETL R09116).

Informed Consent Statement

This study was approved by the Ethics Committee of Tampere University Hospital, Finland. Written informed consent was obtained from all subjects involved in the study.

Data Availability Statement

Complete gene expression data for all samples are available from the corresponding author upon reasonable request.

Acknowledgments

We wish to thank research coordinator Heli Kupari for her skillful assistance with the cartilage samples. We are also thankful to Meiju Kukkonen and Salla Hietakangas for their excellent technical assistance in the laboratory, as well as Heli Määttä for great secretarial help.

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
  2. Scanzello, C.R. Chemokines and inflammation in osteoarthritis: Insights from patients and animal models. J. Orthop. Res. 2017, 35, 735–739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Greene, M.A.; Loeser, R.F. Aging-related inflammation in osteoarthritis. Osteoarth. Cartil. 2015, 23, 1966–1971. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-grade inflammation as a key mediator of the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592. [Google Scholar] [CrossRef]
  5. Karsdal, M.A.; Michaelis, M.; Ladel, C.; Siebuhr, A.S.; Bihlet, A.R.; Andersen, J.R.; Guehring, H.; Christiansen, C.; Bay-Jensen, A.C.; Kraus, V.B. Disease-modifying treatments for osteoarthritis (DMOADs) of the knee and hip: Lessons learned from failures and opportunities for the future. Osteoarthr. Cartil. 2016, 24, 2013–2021. [Google Scholar] [CrossRef] [PubMed]
  6. Aigner, T.; Fundel, K.; Saas, J.; Gebhard, P.M.; Haag, J.; Weiss, T.; Zien, A.; Obermayr, F.; Zimmer, R.; Bartnik, E. Large-scale gene expression profiling reveals major pathogenetic pathways of cartilage degeneration in osteoarthritis. Arthritis Rheum. 2006, 54, 3533–3544. [Google Scholar] [CrossRef] [PubMed]
  7. Sandy, J.D.; Chan, D.D.; Trevino, R.L.; Wimmer, M.A.; Plaas, A. Human genome-wide expression analysis reorients the study of inflammatory mediators and biomechanics in osteoarthritis. Osteoarthr. Cartil. 2015, 23, 1939–1945. [Google Scholar] [CrossRef] [Green Version]
  8. Hardingham, T. Extracellular matrix and pathogenic mechanisms in osteoarthritis. Curr. Rheumatol. Rep. 2008, 10, 30–36. [Google Scholar] [CrossRef] [PubMed]
  9. Maldonado, M.; Nam, J. The Role of Changes in Extracellular Matrix of Cartilage in the Presence of Inflammation on the Pathology of Osteoarthritis. BioMed Res. Int. 2013, 2013, 284873. [Google Scholar] [CrossRef] [Green Version]
  10. Loeser, R.F.; Collins, J.A.; Diekman, B.O. Ageing and the pathogenesis of osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 412–420. [Google Scholar] [CrossRef] [Green Version]
  11. Zhu, J.; Paul, W.E. CD4 T cells: Fates, Functions, and Faults. Blood, J. Am. Soc. Hematol. 2008, 112, 1557–1569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Dinarello, C.A. Immunological and Inflammatory Functions of the Interleukin-1 Family [Internet]. Annu. Rev. Immunol. 2009, 27, 519–550. [Google Scholar] [CrossRef]
  13. Harrington, L.E.; Hatton, R.D.; Mangan, P.R.; Turner, H.; Murphy, T.L.; Murphy, K.M.; Weaver, C.T. Interleukin 17-Producing CD4+ Effector T Cells Develop via a Lineage Distinct from the T Helper Type 1 and 2 Lineages. Nat. Immunol. 2005, 6, 1123–1132. [Google Scholar] [CrossRef] [PubMed]
  14. Kang, S.; Kumanogoh, A. The Spectrum of Macrophage Activation by Immunometabolism [Internet]. Int. Immunol. 2020, 32, 467–473. [Google Scholar] [CrossRef]
  15. Raucci, F.; Saviano, A.; Casillo, G.M.; Guerra-Rodriguez, M.; Mansour, A.A.; Piccolo, M.; Ferraro, M.G.; Panza, E.; Vellecco, V.; Irace, C.; et al. IL-17-Induced Inflammation Modulates the mPGES-1/PPAR-γ Pathway in Monocytes/Macrophages. Br. J. Pharmacol. 2021. [Google Scholar] [CrossRef]
  16. Zizzo, G.; Cohen, P.L. IL-17 Stimulates Differentiation of Human Anti-Inflammatory Macrophages and Phagocytosis of Apoptotic Neutrophils in Response to IL-10 and Glucocorticoids. J. Immunol. 2013, 190, 5237–5246. [Google Scholar] [CrossRef] [Green Version]
  17. Minguzzi, M.; Cetrullo, S.; D’Adamo, S.; Silvestri, Y.; Flamigni, F.; Borzì, R.M. Emerging Players at the Intersection of Chondrocyte Loss of Maturational Arrest, Oxidative Stress, Senescence and Low-Grade Inflammation in Osteoarthritis. Oxid. Med. Cell. Longev. 2018, 2018, 3075293. [Google Scholar] [CrossRef] [Green Version]
  18. López-Armada, M.J.; Caramés, B.; Lires-Deán, M.; Cillero-Pastor, B.; Ruiz-Romero, C.; Galdo, F.; Blanco, F.J. Cytokines, tumor necrosis factor-alpha and interleukin-1beta, differentially regulate apoptosis in osteoarthritis cultured human chondrocytes. Osteoarthr. Cartil. 2006, 14, 660–669. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Daheshia, M.; Yao, J.Q. The interleukin 1beta pathway in the pathogenesis of osteoarthritis. J. Rheumatol. 2008, 35, 2306–2312. [Google Scholar] [CrossRef]
  20. Martel-Pelletier, J.; McCollum, R.; DiBattista, J.; Faure, M.P.; Chin, J.A.; Fournier, S.; Sarfati, M.; Pelletier, J.P. The interleukin-1 receptor in normal and osteoarthritic human articular chondrocytes. Identification as the type I receptor and analysis of binding kinetics and biologic function. Arthritis Rheum. 1992, 35, 530–540. [Google Scholar] [CrossRef]
  21. Jotanovic, Z.; Mihelic, R.; Sestan, B.; Dembic, Z. Role of Interleukin-1 Inhibitors in Osteoarthritis. Drugs Aging 2012, 29, 343–358. [Google Scholar] [CrossRef]
  22. Yao, Z.; Painter, S.L.; Fanslow, W.C.; Ulrich, D.; Macduff, B.M.; Spriggs, M.K.; Armitage, R.J. Human IL-17: A novel cytokine derived from T cells. J. Immunol. 1995, 155, 5483–5486. [Google Scholar]
  23. Miossec, P.; Korn, T.; Kuchroo, V.K. Interleukin-17 and Type 17 Helper T Cells. N. Engl. J. Med. 2009, 361, 888–898. [Google Scholar] [CrossRef] [Green Version]
  24. Chen, B.; Deng, Y.; Tan, Y.; Qin, J.; Chen, L.-B. Association between severity of knee osteoarthritis and serum and synovial fluid interleukin 17 concentrations. J. Int. Med. Res. 2014, 42, 138–144. [Google Scholar] [CrossRef] [Green Version]
  25. Martel-Pelletier, J.; Mineau, F.; Jovanovic, D.; Di Battista, J.A.; Pelletier, J.-P. Mitogen-activated protein kinase and nuclear factor κB together regulate interleukin-17-induced nitric oxide production in human osteoarthritic chondrocytes: Possible role of transactivating factor mitogen-activated protein kinase-activated protein kinase. Arthritis Rheum. 1999, 42, 2399–2409. [Google Scholar] [CrossRef]
  26. Pacquelet, S.; Presle, N.; Boileau, C.; Dumond, H.; Netter, P.; Martel-Pelletier, J.; Pelletier, J.-P.; Terlain, B.; Jouzeau, J.-Y. Interleukin 17, a nitric oxide-producing cytokine with a peroxynitrite-independent inhibitory effect on proteoglycan synthesis. J. Rheumatol. 2002, 29, 2602–2610. [Google Scholar]
  27. Benderdour, M.; Tardif, G.; Pelletier, J.-P.; Di Battista, J.A.; Reboul, P.; Ranger, P.; Martel-Pelletier, J. Interleukin 17 (IL-17) induces collagenase-3 production in human osteoarthritic chondrocytes via AP-1 dependent activation: Differential activation of AP-1 members by IL-17 and IL-1beta. J. Rheumatol. 2002, 29, 1262–1272. [Google Scholar] [PubMed]
  28. Nakashima, T.; Kobayashi, Y.; Yamasaki, S.; Kawakami, A.; Eguchi, K.; Sasaki, H.; Sakai, H. Protein Expression and Functional Difference of Membrane-Bound and Soluble Receptor Activator of NF-κB Ligand: Modulation of the Expression by Osteotropic Factors and Cytokines. Biochem. Biophys. Res. Commun. 2000, 275, 768–775. [Google Scholar] [CrossRef] [PubMed]
  29. Nakae, S.; Nambu, A.; Sudo, K.; Iwakura, Y. Suppression of immune induction of collagen-induced arthritis in IL-17-deficient mice. J. Immunol. 2003, 171, 6173–6177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Chabaud, M.; Lubberts, E.; Joosten, L.; van den Berg, W.; Miossec, P. IL-17 derived from juxta-articular bone and synovium contributes to joint degradation in rheumatoid arthritis. Arthritis Res. 2001, 3, 168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Xu, Y.; Barter, M.J.; Swan, D.C.; Rankin, K.S.; Rowan, A.D.; Santibanez-Koref, M.; Loughlin, J.; Young, D.A. Identification of the pathogenic pathways in osteoarthritic hip cartilage: Commonality and discord between hip and knee OA. Osteoarthr. Cartil. 2012, 20, 1029–1038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Santangelo, K.S.; Nuovo, G.J.; Bertone, A.L. In vivo reduction or blockade of interleukin-1β in primary osteoarthritis influences expression of mediators implicated in pathogenesis. Osteoarthr. Cartil. 2012, 20, 1610–1618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Hoff, P.; Buttgereit, F.; Burmester, G.-R.; Jakstadt, M.; Gaber, T.; Andreas, K.; Matziolis, G.; Perka, C.; Röhner, E. Osteoarthritis synovial fluid activates pro-inflammatory cytokines in primary human chondrocytes. Int. Orthop. 2013, 37, 145–151. [Google Scholar] [CrossRef] [Green Version]
  34. Li, S.; Ren, Y.; Peng, D.; Yuan, Z.; Shan, S.; Sun, H.; Yan, X.; Xiao, H.; Li, G.; Song, H. TIM-3 Genetic Variations Affect Susceptibility to Osteoarthritis by Interfering with Interferon Gamma in CD4+ T Cells. Inflammation 2015, 38, 1857–1863. [Google Scholar] [CrossRef] [PubMed]
  35. de Almeida, R.C.; Ramos, Y.F.; Mahfouz, A.; den Hollander, W.; Lakenberg, N.; Houtman, E.; van Hoolwerff, M.; Suchiman, H.E.D.; Rodríguez Ruiz, A.; Slagboom, P.E.; et al. RNA sequencing data integration reveals an miRNA interactome of osteoarthritis cartilage. Ann. Rheum Dis. 2019, 78, 270–277. [Google Scholar] [CrossRef]
  36. Chizzolini, C.; Chicheportiche, R.; Burger, D.; Dayer, J.M. Human Th1 Cells Preferentially Induce Interleukin (IL)-1β While Th2 Cells Induce IL-1 Receptor Antagonist Production upon Cell/Cell Contact with Monocytes. Eur. J. Immunol. 1997, 27, 171–177. [Google Scholar] [CrossRef]
  37. Weaver, C.T.; Elson, C.O.; Fouser, L.A.; Kolls, J.K. The Th17 Pathway and Inflammatory Diseases of the Intestines, Lungs and Skin. Annu. Rev. Pathol. 2013, 8, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Hume, D.A. The Many Alternative Faces of Macrophage Activation. Front Immunol. 2015, 6, 370. [Google Scholar] [CrossRef] [Green Version]
  39. Alonso, M.N.; Wong, M.T.; Zhang, A.L.; Winer, D.; Suhoski, M.M.; Tolentino, L.L.; Gaitan, J.; Davidson, M.G.; Kung, T.H.; Galel, D.M.; et al. TH1, TH2, and TH17 Cells Instruct Monocytes to Differentiate into Specialized Dendritic Cell Subsets. Blood J. Am. Soc. Hematol. 2011, 118, 3311–3320. [Google Scholar] [CrossRef] [Green Version]
  40. Diniz, S.N.; Nomizo, R.; Cisalpino, P.S.; Teixeira, M.M.; Brown, G.D.; Mantovani, A.; Gordon, S.; Reis, L.F.L.; Dias, A.A.M. PTX3 function as an opsonin for the dectin-1-dependent internalization of zymosan by macrophages. J. Leukoc. Biol. 2004, 75, 649–656. [Google Scholar] [CrossRef] [Green Version]
  41. Sharma, A.; Khan, R.; Gupta, N.; Sharma, A.; Zaheer, M.S.; Abbas, M.; Khan, S.A. Acute phase reactant, Pentraxin 3, as a novel marker for the diagnosis of rheumatoid arthritis. Clin. Chim. Acta 2018, 480, 65–70. [Google Scholar] [CrossRef] [PubMed]
  42. Liu, Y.-X.; Wang, G.-D.; Wang, X.; Zhang, Y.-L.; Zhang, T.-L. Effects of TLR-2/NF-κB signaling pathway on the occurrence of degenerative knee osteoarthritis: An in vivo and in vitro study. Oncotarget 2017, 8, 38602–38617. [Google Scholar] [CrossRef] [PubMed]
  43. Miller, R.E.; Ishihara, S.; Tran, P.B.; Golub, S.B.; Last, K.; Miller, R.J.; Fosang, A.J.; Malfait, A.-M. An aggrecan fragment drives osteoarthritis pain through Toll-like receptor 2. JCI Insight 2018, 3, e95704. [Google Scholar] [CrossRef]
  44. Raghu, H.; Lepus, C.M.; Wang, Q.; Wong, H.H.; Lingampalli, N.; Oliviero, F.; Punzi, L.; Giori, N.J.; Goodman, S.B.; Chu, C.R.; et al. CCL2/CCR2, but not CCL5/CCR5, mediates monocyte recruitment, inflammation and cartilage destruction in osteoarthritis. Ann. Rheum. Dis. 2017, 76, 914–922. [Google Scholar] [CrossRef] [Green Version]
  45. Miller, R.J.; Malfait, A.M.; Miller, R.E. The Innate Immune Response as a Mediator of Osteoarthritis Pain [Internet]. Osteoarthr. Cartil. 2020, 28, 562–571. [Google Scholar] [CrossRef]
  46. Lee, M.-C.; Saleh, R.; Achuthan, A.; Fleetwood, A.J.; Förster, I.; Hamilton, J.A.; Cook, A.D. CCL17 blockade as a therapy for osteoarthritis pain and disease. Arthritis Res. Ther. 2018, 20, 62. [Google Scholar] [CrossRef] [Green Version]
  47. Adań, N.; Guzmán-Morales, J.; Ledesma-Colunga, M.G.; Perales-Canales, S.I.; Quintanar-Stéphano, A.; López-Barrera, F.; Méndez, I.; Moreno-Carranza, B.; Triebel, J.; Binart, N.; et al. Prolactin promotes cartilage survival and attenuates inflammation in inflammatory arthritis. J. Clin. Investig. 2013, 123, 3902–3913. [Google Scholar] [CrossRef]
  48. Ogueta, S.; Muñoz, J.; Obregon, E.; Delgado-Baeza, E.; García-Ruiz, J.P. Prolactin Is a Component of the Human Synovial Liquid and Modulates the Growth and Chondrogenic Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells. Mol. Cell. Endocrinol. 2002, 190, 51–63. [Google Scholar] [CrossRef]
  49. Lorenzo, P.; Aspberg, A.; Onnerfjord, P.; Bayliss, M.T.; Neame, P.J.; Heinegard, D. Identification and characterization of asporin. a novel member of the leucine-rich repeat protein family closely related to decorin and biglycan. J. Biol. Chem. 2001, 276, 12201–12211. [Google Scholar] [CrossRef] [Green Version]
  50. Xu, L.; Li, Z.; Liu, S.-Y.; Xu, S.-Y.; Ni, G.-X. Asporin and osteoarthritis. Osteoarthr. Cartil. 2015, 23, 933–939. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Zhu, X.; Jiang, L.; Lu, Y.; Wang, C.; Zhou, S.; Wang, H.; Tian, T. Association of aspartic acid repeat polymorphism in the asporin gene with osteoarthritis of knee, hip, and hand. Medicine (Baltimore) 2018, 97, e0200. [Google Scholar] [CrossRef]
  52. Wang, J.; Yang, A.; Zhang, J.; Sun, N.; Li, X.; Li, X.; Liu, Q.; Li, J.; Ren, X.; Ke, Z.; et al. Genetic polymorphism in the asporin gene is not a key risk factor for osteoarthritis: Evidence based on an updated cumulative meta-analysis. Exp. Ther. Med. 2018, 15, 3952–3966. [Google Scholar] [CrossRef] [Green Version]
  53. Wang, P.; Zhu, F.; Lee, N.H.; Konstantopoulos, K. Shear-induced interleukin-6 synthesis in chondrocytes: Roles of E prostanoid (EP) 2 and EP3 in cAMP/protein kinase A- and PI3-K/Akt-dependent NF-kappaB activation. J. Biol. Chem. 2010, 285, 24793–24804. [Google Scholar] [CrossRef] [Green Version]
  54. Eitner, A.; Müller, S.; König, C.; Wilharm, A.; Raab, R.; Hofmann, G.O.; Kamradt, T.; Schaible, H.G. Inhibition of Inducible Nitric Oxide Synthase Prevents il-1β-Induced Mitochondrial Dysfunction in Human Chondrocytes. Int. J. Mol. Sci. 2021, 22, 2477. [Google Scholar] [CrossRef]
  55. Kuhne, M.; Erben, U.; Schulze-Tanzil, G.; Köhler, D.; Wu, P.; Richter, F.J.; John, T.; Radbruch, A.; Sieper, J.; Appel, H. HLA-B27-Restricted Antigen Presentation by Human Chondrocytes to CD8+ T Cells: Potential Contribution to Local Immunopathologic Processes in Ankylosing Spondylitis. Arthritis Rheum. 2009, 60, 1635–1646. [Google Scholar] [CrossRef] [PubMed]
  56. Brennan, F.R.; Mikecz, K.; Buzás, E.I.; Glant, T.T. Interferon-Gamma but not Granulocyte/Macrophage Colony-Stimulating Factor Augments Proteoglycan Presentation by Synovial Cells and Chondrocytes to an Autopathogenic T Cell Hybridoma. Immunol. Lett. 1995, 45, 87–91. [Google Scholar] [CrossRef]
  57. Lewallen, E.A.; Bonin, C.A.; Li, X.; Smith, J.; Karperien, M.; Larson, A.N.; Lewallen, D.G.; Cool, S.M.; Westendorf, J.J.; Krych, A.J.; et al. The synovial microenvironment of osteoarthritic joints alters RNA-seq expression profiles of human primary articular chondrocytes. Gene 2016, 591, 456–464. [Google Scholar] [CrossRef]
  58. Dunn, S.L.; Soul, J.; Anand, S.; Schwartz, J.-M.; Boot-Handford, R.P.; Hardingham, T.E. Gene expression changes in damaged osteoarthritic cartilage identify a signature of non-chondrogenic and mechanical responses. Osteoarthr. Cartil. 2016, 24, 1431–1440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Ramos, Y.F.M.; den Hollander, W.; Bovée, J.V.M.G.; Bomer, N.; Breggen, R.; Lakenberg, N.; Keurentjes, J.C.; Goeman, J.J.; Slagboom, P.E.; Nelissen, R.G.H.H.; et al. Genes Involved in the Osteoarthritis Process Identified through Genome Wide Expression Analysis in Articular Cartilage; the RAAK Study. PLoS ONE 2014, 9, e103056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Altman, R.; Asch, E.; Bloch, D.; Bole, G.; Borenstein, D.; Brandt, K.; Christy, W.; Cooke, T.D.; Greenwald, R.; Hochberg, M.; et al. Development of criteria for the classification and reporting of osteoarthritis: Classification of osteoarthritis of the knee. Arthritis Rheum. 1986, 29, 1039–1049. [Google Scholar] [CrossRef]
  61. Mobasheri, A.; Vannucci, S.J.; Bondy, C.A.; Carter, S.D.; Innes, J.F.; Arteaga, M.F.; Trujillo, E.; Ferraz, I.; Shakibaei, M.; Martín-Vasallo, P. Glucose transport and metabolism in chondrocytes: A key to understanding chondrogenesis, skeletal development and cartilage degradation in osteoarthritis. Histol. Histopathol. 2002, 17, 1239–1267. [Google Scholar] [PubMed]
  62. Koskinen, A.; Juslin, S.; Nieminen, R.; Moilanen, T.; Vuolteenaho, K.; Moilanen, E. Adiponectin associates with markers of cartilage degradation in osteoarthritis and induces production of proinflammatory and catabolic factors through mitogen-activated protein kinase pathways. Arthritis Res. Ther. 2011, 13, R184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Andrews, S. FastQC: A Quality Control Tool for High Throughput Sequence Data. 2010. Available online: https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ (accessed on 19 April 2021).
  64. Bolger, A.M.; Lohse, M.; Usadel, B. Trimmomatic: A flexible trimmer for Illumina sequence data. Bioinformatics 2014, 30, 2114–2120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Dobin, A.; Davis, C.A.; Schlesinger, F.; Drenkow, J.; Zaleski, C.; Jha, S.; Batut, P.; Chaisson, M.; Gingeras, T.R. STAR: Ultrafast universal RNA-seq aligner. Bioinformatics 2013, 29, 15–21. [Google Scholar] [CrossRef]
  66. Liao, Y.; Smyth, G.K.; Shi, W. featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 2014, 30, 923–930. [Google Scholar] [CrossRef] [Green Version]
  67. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [Green Version]
  68. Ashburner, M.; Ball, C.A.; Blake, J.A.; Botstein, D.; Butler, H.; Cherry, J.M.; Davis, A.P.; Dolinski, K.; Dwight, S.S.; Eppig, J.T.; et al. Gene Ontology: Tool for the unification of biology. Nat. Genet. 2000, 25, 25–29. [Google Scholar] [CrossRef] [Green Version]
  69. The Gene Ontology Consortium. Expansion of the Gene Ontology knowledgebase and resources. Nucleic Acids Res. 2017, 45, D331–D338. [Google Scholar] [CrossRef] [Green Version]
  70. Huang, D.W.; Sherman, B.T.; Lempicki, R.A. Systematic and Integrative Analysis of Large Gene Lists Using DAVID Bioinformatics Resources. Nat. Protoc. 2009, 4, 44–57. [Google Scholar] [CrossRef]
  71. Supek, F.; Bošnjak, M.; Škunca, N.; Šmuc, T. REVIGO Summarizes and Visualizes Long Lists of Gene Ontology Terms. PLoS ONE 2011, 6, e21800. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Venn diagrams of genes markedly upregulated (FC > 2.5) (A) or markedly downregulated (FC < 2.5) (B) by IL-1β, IL-17 and IFNγ. Red denotes up- and blue downregulated genes.
Figure 1. Venn diagrams of genes markedly upregulated (FC > 2.5) (A) or markedly downregulated (FC < 2.5) (B) by IL-1β, IL-17 and IFNγ. Red denotes up- and blue downregulated genes.
Ijms 22 09463 g001
Figure 2. Heatmap of genes markedly upregulated (FC > 2.5) in degraded cartilage in the study by Almeida et al. [35] and significantly affected by at least one studied proinflammatory cytokine. Upregulated genes are marked with red, downregulated with blue, and genes with no significant fold change with white.
Figure 2. Heatmap of genes markedly upregulated (FC > 2.5) in degraded cartilage in the study by Almeida et al. [35] and significantly affected by at least one studied proinflammatory cytokine. Upregulated genes are marked with red, downregulated with blue, and genes with no significant fold change with white.
Ijms 22 09463 g002
Figure 3. Heatmap of genes markedly downregulated (FC < −2.5) in degraded cartilage in the study by Almeida et al. [35] and significantly affected by at least one studied proinflammatory cytokine. Upregulated genes are marked with red, downregulated with blue, and genes with no significant fold change with white.
Figure 3. Heatmap of genes markedly downregulated (FC < −2.5) in degraded cartilage in the study by Almeida et al. [35] and significantly affected by at least one studied proinflammatory cytokine. Upregulated genes are marked with red, downregulated with blue, and genes with no significant fold change with white.
Ijms 22 09463 g003
Table 1. Twenty most strongly up- and downregulated genes in interleukin 1-treated OA chondrocytes (IL1) relative to control (Co).
Table 1. Twenty most strongly up- and downregulated genes in interleukin 1-treated OA chondrocytes (IL1) relative to control (Co).
GeneNameFunctionMean (Co)Mean (IL1)Fold Changeadj. p
IL6Interleukin 6Inflammation12.418,406.93685.72<1.0 × 10−4
CXCL1C-X-C motif chemokine ligand 1Inflammation, chemotaxis13.823,793.73457.68<1.0 × 10−4
IL1BInterleukin 1 betaInflammation2.89575.73332.44<1.0 × 10−4
CXCL8C-X-C motif chemokine ligand 8Inflammation, chemotaxis329.5855,146.32968.9<1.0 × 10−4
CXCL6C-X-C motif chemokine ligand 6Inflammation, chemotaxis2.84951.82352.02<1.0 × 10−4
CXCL5C-X-C motif chemokine ligand 5Inflammation, chemotaxis7.47352.41239.8<1.0 × 10−4
CXCL2C-X-C motif chemokine ligand 2Inflammation, chemotaxis3.94798.21198.05<1.0 × 10−4
CXCL3C-X-C motif chemokine ligand 3Inflammation, chemotaxis3.13154.61130.76<1.0 × 10−4
CCL20C-C motif chemokine ligand 20Inflammation, chemotaxis418381,100.81128.35<1.0 × 10−4
IL36RNInterleukin 36 receptor antagonistRegulation of inflammation8.65863.8914.19<1.0 × 10−4
ADORA2AAdenosine A2a receptorRegulation of inflammation5.51550.7641.44<1.0 × 10−4
IL36GInterleukin 36 gammaInflammation1.81065.5562.03<1.0 × 10−4
EREGEpiregulinRegulation of proliferation31.913,697.7506.87<1.0 × 10−4
CSF3Colony stimulating factor 3Granulocyte-mediated inflammation0.163.9300.02<1.0 × 10−4
VNN1Vanin 1T cell migration9.22467.2273.35<1.0 × 10−4
CCL5C-C motif chemokine ligand 5Inflammation, chemotaxis4.11134.2271.85<1.0 × 10−4
C15orf48Chromosome 15 open reading frame 48?27.24669.1253.13<1.0 × 10−4
CCL3C-C motif chemokine ligand 3Inflammation, granulocyte activation0.5166.3242.88<1.0 × 10−4
FCAMRFc fragment of IgA and IgM receptorAdaptive immunity, leukocyte migration2.6492213.45<1.0 × 10−4
SERPINB7Serpin family B member 7Endoproteinase inhibition22.13747.9205.63<1.0 × 10−4
HRCT1Histidine rich carboxyl terminus 1?105.84.1−38.85<1.0 × 10−4
LSP1Lymphocyte specific protein 1Regulation of neutrophil mobility1749.658.1−31.39<1.0 × 10−4
HIST1H3GHistone cluster 1 H3 family member gRegulation of transcription183.49.6−28.26<1.0 × 10−4
ACTC1Actin, alpha, cardiac muscle 1Heart muscle constituent195.210.5−24.79<1.0 × 10−4
NXPH3Neurexophilin 3?39.22.4−23.89<1.0 × 10−4
SCN2BSodium voltage-gated channel beta subunit 2Cell adhesion and migration1678.7−22.19<1.0 × 10−4
HIST1H1AHistone cluster 1 H1 family member a?908.547.2−21.2<1.0 × 10−4
GDF10Growth differentiation factor 10Skeletal system development813.645.7−20.57<1.0 × 10−4
LINC02593Long intergenic non-protein coding RNA 2593?68.33.4−20.53<1.0 × 10−4
HIST1H3BHistone cluster 1 H3 family member bRegulation of transcription990.659.2−20.46<1.0 × 10−4
TMEM26Transmembrane protein 26?403.721.4−19.3<1.0 × 10−4
PHYHIPLPhytanoyl-CoA 2-hydroxylase interacting protein like?221.6−19.19<1.0 × 10−4
SARDHSarcosine dehydrogenaseMitochondrial metabolism25.82.4−19.08<1.0 × 10−4
HIST1H2BOHistone cluster 1 H2B family member oRegulation of transcription?234.412.7−18.99<1.0 × 10−4
ID3Inhibitor of DNA binding 3, HLH proteinRegulation of transcription676.545.8−18.32<1.0 × 10−4
HIST1H2AJHistone cluster 1 H2A family member jRegulation of transcription?85747.1−18.12<1.0 × 10−4
HIST1H1BHistone cluster 1 H1 family member bRegulation of transcription?73650.6−17.69<1.0 × 10−4
MFAP2Microfibril associated protein 2ECM organization333.2−17.52<1.0 × 10−4
TNNT3Troponin T3, fast skeletal typeMuscle constituent95.66.4−17.51<1.0 × 10−4
HIST1H2ALHistone cluster 1 H2A family member lRegulation of transcription?321.421.2−17.32<1.0 × 10−4
Red = upregulated genes; blue = downregulated genes.
Table 2. Twenty most strongly up- and downregulated genes in interleukin 17-treated OA chondrocytes (IL17) relative to control (Co).
Table 2. Twenty most strongly up- and downregulated genes in interleukin 17-treated OA chondrocytes (IL17) relative to control (Co).
GeneNameFunctionMean (Co)Mean (IL17)Fold Changeadj. p
SAA2Serum amyloid A2Chemotaxis5.5659.2319.99<1.0 × 10−4
IL6Interleukin 6Inflammation12.21431.4250.15<1.0 × 10−4
SAA1Serum amyloid A1Inflammation, chemotaxis63.73520.0183.26<1.0 × 10−4
SAA2-SAA4SAA2-SAA4 readthroughChemotaxis?2.9216.7156.18<1.0 × 10−4
CXCL6C-X-C motif chemokine ligand 6Inflammation, chemotaxis2.8276.4141.01<1.0 × 10−4
CXCL1C-X-C motif chemokine ligand 1Inflammation, chemotaxis13.61170.5136.48<1.0 × 10−4
VNN1Vanin 1T cell migration9.1820.584.13<1.0 × 10−4
CCL20C-C motif chemokine ligand 20Chemotaxis412.826,508.973.49<1.0 × 10−4
TNFSF18TNF superfamily member 18T cell survival4.2470.373.05<1.0 × 10−4
IL36RNInterleukin 36 receptor antagonistRegulation of inflammation8.5468.069.09<1.0 × 10−4
VNN3Vanin 3?1.8130.366.35<1.0 × 10−4
ADORA2AAdenosine A2a receptorInflammation, phagocytosis5.4105.964.74<1.0 × 10−4
CXCL2C-X-C motif chemokine ligand 2Inflammation, chemotaxis3.9220.355.90<1.0 × 10−4
CXCL8C-X-C motif chemokine ligand 8Inflammation, chemotaxis324.814,116.548.18<1.0 × 10−4
C15orf48Chromosome 15 open reading frame 48Mitochondrial respiration?26.9820.346.34<1.0 × 10−4
PDZK1IP1PDZK1 interacting protein 1Regulation of apoptosis5.2206.941.18<1.0 × 10−4
NOS2Nitric oxide synthase 2Inflammation137.93370.240.02<1.0 × 10−4
ODAPHOdontogenesis associated phosphoproteinEnamel production1.441.937.29<1.0 × 10−4
SLC28A3Solute carrier family 28 member 3Nucleoside transport4.3150.435.34<1.0 × 10−4
CXCL5C-X-C motif chemokine ligand 5Inflammation, chemotaxis7.3207.534.25<1.0 × 10−4
ACTC1Actin, alpha, cardiac muscle 1Cardiac muscle component191.726.7−8.14<1.0 × 10−4
TOXThymocyte selection associated high mobility group boxT cell development14.63.9−5.660.0010
TMEM26Transmembrane protein 26?396.369.8−5.47<1.0 × 10−4
TNNT3Troponin T3, fast skeletal typeMuscle component93.917.9−5.28<1.0 × 10−4
TENT5BTerminal nucleotidyltransferase 5BRegulation of cell proliferation152.539.7−4.81<1.0 × 10−4
TMEM26-AS1TMEM26 antisense RNA 1?32.014.4−4.773.8 × 10−4
RCAN2Regulator of calcineurin 2Regulation of transcription326.574.6−4.74<1.0 × 10−4
OPRL1Opioid related nociceptin receptor 1?11.83.0−4.510.0068
CSRNP3Cysteine and serine rich nuclear protein 3Regulation of apoptosis59.719.7−4.01<1.0 × 10−4
ASPNAsporinCartilage constituent2011.2505.2−3.92<1.0 × 10−4
HRCT1Histidine rich carboxyl terminus 1?104.125.8−3.85<1.0 × 10−4
AQP1Aquaporin 1 (Colton blood group)Regulation of osmotic pressure, angiogenesis, apoptosis42.913.4−3.69<1.0 × 10−4
YWHAZP5YWHAZ pseudogene 5?10.23.2−3.680.013
MRAP2Melanocortin 2 receptor accessory protein 2cAMP signaling1295.9376.5−3.62<1.0 × 10−4
C1QTNF7C1q and TNF related 7?63.420.1−3.54<1.0 × 10−4
MFAP2Microfibril associated protein 2Connective tissue organization32.48.7−3.47<1.0 × 10−4
CLEC3AC-type lectin domain family 3 member ASkeletal system development847.3264.6−3.46<1.0 × 10−4
GREM1Gremlin 1, DAN family BMP antagonistRegulation of connective tissue development5141.61566.4−3.41<1.0 × 10−4
CRISPLD1Cysteine rich secretory protein LCCL domain containing 1Morphogenesis946.1280.2−3.39<1.0 × 10−4
HRASLS5 (=PLAAT5)HRAS like suppressor family member 5Glycerophospholipid metabolism12.83.6−3.370.019
Red = upregulated genes; blue = downregulated genes.
Table 3. Twenty most strongly up- and downregulated genes in interferon gamma -treated OA chondrocytes (IFNγ) relative to control (Co).
Table 3. Twenty most strongly up- and downregulated genes in interferon gamma -treated OA chondrocytes (IFNγ) relative to control (Co).
GeneNameFunctionMean (Co)Mean (IFNγ)Fold changeadj. p
IDO1Indoleamine 2,3-dioxygenase 1Regulation of T cell -mediated immunity17.542,320.04643.74<1.0 × 10−4
LGALS17AGalectin 14 pseudogene?0.41065.11750.58<1.0 × 10−4
GBP1P1Guanylate binding protein 1 pseudogene 1?2.62838.81245.34<1.0 × 10−4
CXCL10C-X-C motif chemokine ligand 10Chemotaxis2.22065.21117.91<1.0 × 10−4
GBP5Guanylate binding protein 5Inflammasome activation1.41518.31112.44<1.0 × 10−4
CXCL9C-X-C motif chemokine ligand 9T cell chemotaxis1.11069.91033.80<1.0 × 10−4
GBP4Guanylate binding protein 4Inflammation?30.927,565.6955.57<1.0 × 10−4
IFI44LInterferon induced protein 44 like?9.76185.8694.66<1.0 × 10−4
GBP1Guanylate binding protein 1Negative regulation of inflammation124.354,562.1454.62<1.0 × 10−4
HLA-DRAMajor histocompatibility complex, class II, DR alphaAntigen presentation5.62338.3408.93<1.0 × 10−4
HLA-DRB1Major histocompatibility complex, class II, DR beta 1Antigen presentation10.72430.7383.18<1.0 × 10−4
CD74CD74 moleculeAntigen presentation31.911,211.5353.35<1.0 × 10−4
RSAD2Radical S-adenosyl methionine domain containing 2Antiviral action44.515,365.2338.82<1.0 × 10−4
RARRES3Retinoic acid receptor responder 3Phospholipid catabolism33.18271.1286.40<1.0 × 10−4
BST2Bone marrow stromal cell antigen 2Antiviral action10.12908.5285.04<1.0 × 10−4
GBP6Guanylate binding protein family member 6Inflammation1.0193.3273.26<1.0 × 10−4
HLA-DRB5Major histocompatibility complex, class II, DR beta 5Antigen presentation4.4825.4253.47<1.0 × 10−4
HLA-DRB6Major histocompatibility complex, class II, DR beta 6 (pseudogene)Antigen presentation?0.3125.7226.68<1.0 × 10−4
APOL4Apolipoprotein L4Lipid metabolism2.6500.8225.95<1.0 × 10−4
IFIT2Interferon induced protein with tetratricopeptide repeats 2Regulation of proliferation96.220,648.8225.79<1.0 × 10−4
TNFRSF10DTNF receptor superfamily member 10dInhibition of apoptosis4135.1501.9−7.65<1.0 × 10−4
ARHGAP9Rho gtpase activating protein 9?10.72.4−5.270.0028
NANOS1Nanos C2HC-type zinc finger 1Regulation of translation and cell migration83.416.9−4.94<1.0 × 10−4
SNORD108Small nucleolar RNA, C/D box 108?66.613.8−4.81<1.0 × 10−4
FAM189A2Family with sequence similarity 189 member A2?13.64.3−4.390.0033
PWAR6Prader Willi/Angelman region RNA 6?34.07.9−4.32<1.0 × 10−4
GABRA4Gamma-aminobutyric acid type A receptor alpha4 subunitSynaptic transmission2346.1549.2−4.28<1.0 × 10−4
CORO2ACoronin 2A?13.53.7−4.110.020
WFDC1WAP four-disulfide core domain 1Regulation of proliferation65.118.0−4.06<1.0 × 10−4
PRSS35Serine protease 35?51.413.5−4.01<1.0 × 10−4
SLC16A14Solute carrier family 16 member 14Organic acid transport40.213.3−3.98<1.0 × 10−4
PWAR5Prader Willi/Angelman region RNA 5?359.791.4−3.93<1.0 × 10−4
MTURNMaturin, neural progenitor differentiation regulator homolog?1857.1519.7−3.63<1.0 × 10−4
C1QTNF5C1q and TNF related 5Cell adhesion152.446.1−3.47<1.0 × 10−4
LONRF2LON peptidase N-terminal domain and ring finger 2?206.859.5−3.46<1.0 × 10−4
FGFR4Fibroblast growth factor receptor 4Cell proliferation and migration11.15.1−3.310.045
TRABD2BTrab domain containing 2BWnt signaling, proteolysis14.25.5−3.290.0014
TNNT3Troponin T3, fast skeletal typeMuscle contraction106.031.6−3.26<1.0 × 10−4
NCALDNeurocalcin deltaEndocytosis17.36.6−3.240.029
CDH2Cadherin 2Cell adhesion12.04.1−3.230.0012
Red = upregulated genes; blue = downregulated genes.
Table 4. GO terms affected by different proinflammatory cytokines. Genes with FC > 2.5 in either direction were analyzed with DAVID, and the resulting lists were reduced with REVIGO. GO terms significantly affected (with FDR-corrected p-value < 0.05) by a cytokine are marked with an X.
Table 4. GO terms affected by different proinflammatory cytokines. Genes with FC > 2.5 in either direction were analyzed with DAVID, and the resulting lists were reduced with REVIGO. GO terms significantly affected (with FDR-corrected p-value < 0.05) by a cytokine are marked with an X.
TermIL1IL17IFNγTermIL1IL17IFNγ
Inflammatory responseXXXNucleosome assemblyX
Immune responseXXXChromosome segregationX
Response to lipopolysaccharideXXXProtein heterotetramerizationX
ChemotaxisXXXWound healingX
Negative regulation of viral entryXXXRegulation of cell proliferationX
into host cell Cell migrationX
Negative regulation of type IXXXRegulation of gene silencingX
interferon production Positive regulation of interleukin-12 productionX
Response to progesteroneXX OdontogenesisX
Cell-cell signalingXX Cellular response to mechanical stimulusX
AngiogenesisXX Peptidyl-tyrosine phosphorylationX
Negative regulation of growthXX Collagen catabolic processX
Positive regulation of mitoticXX Positive regulation of cell divisionX
nuclear division Positive chemotaxis X
Negative regulation of cellXX Positive regulation of nitric-oxide synthase biosynthetic X
proliferation process
Signal transductionX XAcute-phase response X
Response to virusX XPositive regulation of cytosolic calcium ion concentration X
Positive regulation of interleukin-6X XPositive regulation of gtpase activity X
production Response to glucocorticoid X
Response to hydrogen peroxideX XResponse to wounding X
Positive regulation of I-kappabX XPositive regulation of NF-kappab transcription factor X
kinase/NF-kappab signaling activity
Response to drugX XNegative regulation of tumor necrosis factor production X
Cellular response to zinc ion XXCellular response to organic cyclic compound X
Response to toxic substance XXAntigen processing and presentation X
Tumor necrosis factor-mediated XXAntigen processing and presentation of peptide or X
signaling pathway polysaccharide antigen via MHC class II
Cell divisionX Antigen processing and presentation of exogenous peptide X
DNA replicationX antigen via MHC class I, TAP-independent
Telomere organizationX Response to interferon-beta X
Positive regulation of geneX Response to interferon-alpha X
expression T cell costimulation X
Cell adhesionX Positive regulation of T cell mediated cytotoxicity X
Extracellular matrix organizationX Defense response X
Skeletal system developmentX Protein trimerization X
Sister chromatid cohesionX Proteolysis X
DNA replication initiationX Defense response to protozoan X
Cellular protein metabolic processX Positive regulation of peptidyl-tyrosine phosphorylation X
Cell proliferationX Protein polyubiquitination X
Negative regulation of geneX
expression, epigenetic
Table 5. Genes upregulated by all studied proinflammatory cytokines with FC > 2.5. Shown are mean normalized expression levels in control (Co) and in C(IL1), C(IL17) and C(IFNγ) phenotypes, fold changes (FCs) for all comparisons vs. control and false discovery rate (FDR)-adjusted p values for them.
Table 5. Genes upregulated by all studied proinflammatory cytokines with FC > 2.5. Shown are mean normalized expression levels in control (Co) and in C(IL1), C(IL17) and C(IFNγ) phenotypes, fold changes (FCs) for all comparisons vs. control and false discovery rate (FDR)-adjusted p values for them.
GeneNameMean exp. (Co)Mean exp. (IL1)Mean exp. (IL17)Mean exp. (IFNγ)FC (IL1 vs. Co)adj. p (IL1 vs. Co)FC (IL17 vs. Co)adj. p (IL17 vs. Co)FC (IFNγ vs. Co)adj. p (IFNγ vs. Co)
IL6Interleukin 612.818,406.91431.494.23685.72<1.0 × 10−4250.15<1.0 × 10−412.34<1.0 × 10−4
IL36RNInterleukin 36 receptor antagonist8.95863.8468.036.7914.19<1.0 × 10−469.09<1.0 × 10−44.59<1.0 × 10−4
ESM1Endothelial cell specific molecule 1276.737,984.11373.51449.2157.25<1.0 × 10−45.09<1.0 × 10−44.70<1.0 × 10−4
SAA2Serum amyloid A25.8371.4659.227.1149.11<1.0 × 10−4319.99<1.0 × 10−48.73<1.0 × 10−4
iNOS/NOS2Inducible nitric oxide synthase/Nitric oxide synthase 2144.212,704.93370.23046.1131.22<1.0 × 10−440.02<1.0 × 10−430.16<1.0 × 10−4
NOD2Nucleotide binding oligomerization domain containing 27.6919.496.743.9116.73<1.0 × 10−413.67<1.0 × 10−45.61<1.0 × 10−4
PTX3Pentraxin 3184.418,888.74615.3479.6113.19<1.0 × 10−427.47<1.0 × 10−42.60<1.0 × 10−4
SAA1Serum amyloid A166.62188.73520.0227.694.66<1.0 × 10−4183.26<1.0 × 10−46.46<1.0 × 10−4
CD300ECD300e molecule3.6316.932.771.672.79<1.0 × 10−47.91<1.0 × 10−417.15<1.0 × 10−4
IL36BInterleukin 36 beta11.3466.380.139.167.27<1.0 × 10−49.65<1.0 × 10−43.60<1.0 × 10−4
TNFRSF1BTNF receptor superfamily member 1B40.02370.7525.8118.962.58<1.0 × 10−414.66<1.0 × 10−43.02<1.0 × 10−4
TNFAIP6TNF alpha induced protein 61176.442,950.35512.44561.236.87<1.0 × 10−44.59<1.0 × 10−43.59<1.0 × 10−4
TMEM132ATransmembrane protein 132A10.3328.1165.032.633.90<1.0 × 10−416.64<1.0 × 10−43.18<1.0 × 10−4
ICAM1Intercellular adhesion molecule 11415.242,657.24388.38524.531.66<1.0 × 10−43.15<1.0 × 10−45.54<1.0 × 10−4
C3AR1Complement C3a receptor 12.266.211.411.228.15<1.0 × 10−46.361.5 × 10−45.324.9 × 10−4
CLEC2BC-type lectin domain family 2 member B5.3145.048.520.627.53<1.0 × 10−49.35<1.0 × 10−43.85<1.0 × 10−4
COX-2/PTGS2Cyclooxygenase-2/Prostaglandin-endoperoxide synthase 21310.737,281.54678.65349.226.96<1.0 × 10−43.28<1.0 × 10−43.57<1.0 × 10−4
TLR2Toll like receptor 2134.93348.9782.0371.422.64<1.0 × 10−45.02<1.0 × 10−42.54<1.0 × 10−4
CCL7C-C motif chemokine ligand 72.136.720.624.420.66<1.0 × 10−412.14<1.0 × 10−410.56<1.0 × 10−4
CCL2C-C motif chemokine ligand 2150.42475.0815.0430.619.42<1.0 × 10−45.85<1.0 × 10−42.61<1.0 × 10−4
IRF4Interferon regulatory factor 423.5400.194.9114.218.20<1.0 × 10−44.62<1.0 × 10−44.69<1.0 × 10−4
CD274CD274 molecule61.81048.8350.13845.717.56<1.0 × 10−46.18<1.0 × 10−460.08<1.0 × 10−4
RBM47RNA binding motif protein 478.8122.330.622.814.96<1.0 × 10−43.38<1.0 × 10−42.670.040
CD38CD38 molecule9.8133.874.3211.414.81<1.0 × 10−47.67<1.0 × 10−420.76<1.0 × 10−4
BDKRB1Bradykinin receptor B129.0401.5129.6105.013.95<1.0 × 10−44.88<1.0 × 10−43.19<1.0 × 10−4
GCH1GTP cyclohydrolase 1591.77968.72212.73584.213.38<1.0 × 10−43.90<1.0 × 10−45.63<1.0 × 10−4
LRRC38Leucine rich repeat containing 3811.2132.144.435.811.59<1.0 × 10−43.79<1.0 × 10−42.98<1.0 × 10−4
KIAA1217KIAA121715.3157.855.1109.110.61<1.0 × 10−43.80<1.0 × 10−46.39<1.0 × 10−4
SSTR2Somatostatin receptor 290.0971.21549.7340.110.56<1.0 × 10−416.11<1.0 × 10−43.36<1.0 × 10−4
DUSP5Dual specificity phosphatase 577.3746.8302.4236.110.54<1.0 × 10−44.02<1.0 × 10−42.90<1.0 × 10−4
TYMPThymidine phosphorylase311.33020.11275.19324.010.15<1.0 × 10−44.24<1.0 × 10−428.71<1.0 × 10−4
GPR158G protein-coupled receptor 1586.938.022.021.59.98<1.0 × 10−46.770.00185.557.6 × 10−4
PRLRProlactin receptor8.378.829.733.09.93<1.0 × 10−43.050.00343.92<1.0 × 10−4
GSAPGamma-secretase activating protein122.21109.8378.0509.39.18<1.0 × 10−43.26<1.0 × 10−43.74<1.0 × 10−4
GPR39G protein-coupled receptor 3915.4110.639.141.49.17<1.0 × 10−43.241.7 × 10−42.71<1.0 × 10−4
LYPD1LY6/PLAUR domain containing 110.571.528.727.78.44<1.0 × 10−43.315.6 × 10−42.620.0023
ODF3BOuter dense fiber of sperm tails 3B34.6261.0106.0773.87.98<1.0 × 10−43.28<1.0 × 10−421.57<1.0 × 10−4
SLC15A3Solute carrier family 15 member 316.3119.454.7607.47.63<1.0 × 10−43.45<1.0 × 10−435.59<1.0 × 10−4
HALHistidine ammonia-lyase6.244.128.747.47.57<1.0 × 10−44.71<1.0 × 10−46.97<1.0 × 10−4
DOCK4Dedicator of cytokinesis 444.0306.8144.9139.26.94<1.0 × 10−43.21<1.0 × 10−42.91<1.0 × 10−4
RAB27BRAB27B, member RAS oncogene family16.577.260.584.55.98<1.0 × 10−43.85<1.0 × 10−45.62<1.0 × 10−4
CH25HCholesterol 25-hydroxylase7.436.525.841.84.41<1.0 × 10−43.270.0226.32<1.0 × 10−4
USP43Ubiquitin specific peptidase 434.412.813.616.13.940.0203.410.0134.500.0091
AC104966.1Ceruloplasmin (ferroxidase) (CP) pseudogene16.547.657.353.73.39<1.0 × 10−43.79<1.0 × 10−43.36<1.0 × 10−4
KLK10Kallikrein related peptidase 1014.037.133.043.13.110.0223.290.00672.650.0028
Red = upregulated genes.
Table 6. Genes downregulated by all studied proinflammatory cytokines with FC < −2.5. Shown are mean normalized expression levels in control (Co), in C(IL1), C(IL17) and C(IFNγ) phenotypes, fold changes (FCs) for all comparisons vs. control and false discovery rate (FDR)-adjusted p values for them.
Table 6. Genes downregulated by all studied proinflammatory cytokines with FC < −2.5. Shown are mean normalized expression levels in control (Co), in C(IL1), C(IL17) and C(IFNγ) phenotypes, fold changes (FCs) for all comparisons vs. control and false discovery rate (FDR)-adjusted p values for them.
GeneNameFunctionMean exp. (Co)Mean exp. (IL1)Mean exp. (IL17)Mean exp. (IFNγ)FC (IL1 vs. Co)adj. p (IL1 vs. Co)FC (IL17 vs. Co)adj. p (IL17 vs. Co)FC (IFNγ vs. Co)adj. p (IFNγ vs. Co)
SCN2BSodium voltage-gated channel beta subunit 2Sodium ion transport170.88.765.963.7−22.19<1.0 × 10−4−2.59<1.0 × 10−4−2.90<1.0 × 10−4
TNNT3Troponin T3, fast skeletal typeSkeletal muscle constituent97.86.417.931.6−17.51<1.0 × 10−4−5.28<1.0 × 10−4−3.26<1.0 × 10−4
MRAP2Melanocortin 2 receptor accessory protein 2Metabolism?1348.791.1376.5572.0−15.12<1.0 × 10−4−3.62<1.0 × 10−4−2.85<1.0 × 10−4
WFDC1WAP four-disulfide core domain 1Negative regulation of cell growth60.16.134.918.0−12.06<1.0 × 10−4−2.680.0019−4.06<1.0 × 10−4
RANBP3LRAN binding protein 3 likeNuclear export654.874.6284.8280.0−9.40<1.0 × 10−4−2.54<1.0 × 10−4−2.60<1.0 × 10−4
ASPNAsporinSkeletal system development, negative regulation of TGFβ signaling2094.0206.3505.2837.5−8.28<1.0 × 10−4−3.92<1.0 × 10−4−2.77<1.0 × 10−4
FGFR4Fibroblast growth factor receptor 4Cell proliferation and migration10.32.33.15.1−5.595.2 × 10−4−3.120.036−3.310.045
PTGER3Prostaglandin E receptor 3Inflammation, cell death494.1173.6162.3188.8−2.69<1.0 × 10−4−3.03<1.0 × 10−4−2.82<1.0 × 10−4
blue = downregulated genes.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pemmari, A.; Leppänen, T.; Hämäläinen, M.; Moilanen, T.; Moilanen, E. Chondrocytes from Osteoarthritis Patients Adopt Distinct Phenotypes in Response to Central TH1/TH2/TH17 Cytokines. Int. J. Mol. Sci. 2021, 22, 9463. https://doi.org/10.3390/ijms22179463

AMA Style

Pemmari A, Leppänen T, Hämäläinen M, Moilanen T, Moilanen E. Chondrocytes from Osteoarthritis Patients Adopt Distinct Phenotypes in Response to Central TH1/TH2/TH17 Cytokines. International Journal of Molecular Sciences. 2021; 22(17):9463. https://doi.org/10.3390/ijms22179463

Chicago/Turabian Style

Pemmari, Antti, Tiina Leppänen, Mari Hämäläinen, Teemu Moilanen, and Eeva Moilanen. 2021. "Chondrocytes from Osteoarthritis Patients Adopt Distinct Phenotypes in Response to Central TH1/TH2/TH17 Cytokines" International Journal of Molecular Sciences 22, no. 17: 9463. https://doi.org/10.3390/ijms22179463

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop