Next Article in Journal
Pre- and Postnatal Vitamin A Deficiency Impairs Motor Skills without a Consistent Effect on Trace Mineral Status in Young Mice
Next Article in Special Issue
The Role of Adipokines between Genders in the Pathogenesis of Osteoarthritis
Previous Article in Journal
Adiponectin and Inflammatory Marker Levels in the Elderly Patients with Diabetes, Mild Cognitive Impairment and Depressive Symptoms
Previous Article in Special Issue
TNFα-Induced Inflammation Model—Evaluation of Concentration and Passage-Dependent Effects on Bovine Chondrocytes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TNFα-Related Chondrocyte Inflammation Models: A Systematic Review

Department of Orthopedics and Trauma Surgery, University Hospital Bonn, 53127 Bonn, Germany
*
Authors to whom correspondence should be addressed.
These authors share last authorship.
Int. J. Mol. Sci. 2024, 25(19), 10805; https://doi.org/10.3390/ijms251910805
Submission received: 5 August 2024 / Revised: 26 September 2024 / Accepted: 4 October 2024 / Published: 8 October 2024
(This article belongs to the Special Issue Osteoarthritis Biomarkers, Diagnosis and Treatments)

Abstract

:
Tumor necrosis factor alpha (TNFα), as a key pro-inflammatory cytokine, plays a central role in joint diseases. In recent years, numerous models of TNFα-induced cartilage inflammation have been developed. However, due to the significant differences between these models and the lack of consensus in their construction, it becomes difficult to compare the results of different studies. Therefore, we summarized and compared these models based on important parameters for model construction, such as cell source, cytokine concentration, stimulation time, mechanical stimulation, and more. We attempted to analyze the advantages and disadvantages of each model and provide a compilation of the analytical methods used in previous studies. Currently, TNFα chondrocyte inflammation models can be categorized into four main types: monolayer-based, construct-based, explant-based TNFα chondrocyte inflammation models, and miscellaneous TNFα chondrocyte inflammation models. The most commonly used models were the monolayer-based TNFα chondrocyte inflammation models (42.86% of cases), with 10 ng/mL TNFα being the most frequently used concentration. The most frequently used chondrocyte cell passage is passage 1 (50%). Human tissues were most frequently used in experiments (51.43%). Only five articles included models with mechanical stimulations. We observed variations in design conditions between different models. This systematic review provides the essential experimental characteristics of the available chondrocyte inflammation models with TNFα, and it provides a platform for better comparison between existing and new studies in this field. It is essential to perform further experiments to standardize each model and to find the most appropriate experimental parameters.

1. Introduction

Articular cartilage consists of chondrocytes surrounded by an extracellular matrix (ECM). This tissue has an optimized composition to withstand mechanical forces. However, articular cartilage is aneural and avascular, and chondrocytes are nourished by the diffusion of synovial fluid, resulting in a low healing potential of the tissue after injury. Osteoarthritis (OA) is a major disease characterized by cartilage loss, subchondral bone changes, osteophyte formation, and persistent inflammation. The inflammatory response affects all of the joint structures, including cartilage, bone, and synovial tissue. Tumor necrosis factor alpha (TNFα) is a potent pro-inflammatory cytokine that has been implicated in many diseases, and it plays a critical role in the immune system during inflammation [1]. TNFα is mainly produced by activated macrophages, T cells, and natural killer cells, but it can also be secreted by chondrocytes. It exists in two forms: a soluble form of 17 kDa and a transmembrane form of 26 kDa [2]. The precursor form, the transmembrane form (tm-TNFα), is converted to the soluble form (sTNFα) to enable biological activities through the action of the TNFα-converting enzyme (TACE) [3]. TNFα interacts with its two distinct receptors, designated as TNFR1 and TNFR2 (TNFR1, also known as TNFRSF1A, CD120a, and p55; TNFR2, also known as TNFRSF1B, CD120b, and p75), which initiate signaling pathways leading to various cellular responses, including cell survival, differentiation, and proliferation and migration [4,5,6,7]. TNFα can inhibit cartilage matrix synthesis and increase its degradation by promoting the release of matrix metalloproteinases (MMPs), and it inhibits chondrogenesis through the nuclear factor-κB (NF-κB) pathway by preventing the synthesis of SOX9, a transcription factor required for the chondrocyte phenotype [8,9,10,11,12,13]. Given that TNFα-neutralizing therapies are effective in the treatment of autoimmune and chronic inflammatory diseases, the importance of TNFα in inflammation has been highlighted [14].
The aim of this review was to provide an overall summary of experimental approaches that have investigated the pro-inflammatory effect of TNFα on cartilage/chondrocyte tissue in vitro. This will allow for better referencing when developing new models or tactics and may also improve the translation of research from bench to bedside.

2. Results

2.1. Description of Studies

A total of 4304 reports were identified and 75 were selected for further review after reading the full text (Figure 1). In total, 25 studies met the inclusion criteria and were included in this systemic review and 50 studies were excluded. Twenty-two studies were excluded because some key information on the TNFα inflammation model was missing (chondrocyte passage and/or stimulus duration) [15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36]. Nine studies did not use primary chondrocytes [37,38,39,40,41,42,43,44,45]. One study was not accessible as a full text. Ten studies used TNFα together with other cytokines, such as IL-1β, to create an inflammation model and were therefore excluded. Two studies were not related to TNFα inflammation models. Six articles were excluded due to genetic modification. More specific model details are provided in Table 1, Table 2, Table 3 and Table 4.

2.2. Classification of TNFα Chondrocyte Inflammation Models

TNFα-related chondrocyte/cartilage inflammation models can be divided into four broad categories according to the method of model construction (Figure 2): (1) monolayer-based TNFα chondrocyte inflammation models (45.16%), (2) explant-based TNFα chondrocyte inflammation models (35.49%), (3) construct-based TNFα chondrocyte inflammation models (12.90%), and (4) other inflammation models (6.45%).

2.3. Concentrations and Stimulation Durations of TNFα

The choice of cytokine concentration would depend on the choice of model. The most commonly used TNFα concentration in monolayer-based, construct-based TNFα chondrocyte inflammation models was 10 ng/mL, but in explant-based TNFα chondrocyte inflammation models, the concentration was 100 ng/mL (Figure 3).
The duration of TNFα stimulation also depended on the type of model. In general, the stimulation time for monolayer-based models was shorter than the others (less than one week in all reported studies). In more than half of these studies, the stimulation time did not exceed 48 h, with 24 h being the most commonly used stimulation time (Figure 4).
For construct-based TNFα chondrocyte inflammation models, the shortest stimulation time was 48 h, which was also the most common stimulation time, and the longest stimulation time was 14 days. For explant-based TNFα chondrocyte inflammation models, the shortest time was 2 h [57], and the longest was 20 days, with the largest range of stimulation times between the studies (Figure 4).

2.4. Passages Used in TNFα Chondrocyte Inflammation Models

The chondrocyte passages used in the reported studies varied from passage 0 to 3. Passage 1 (60.87%) and passage 0 (21.73%) were the most commonly used (Figure 5).

2.5. Species Distribution

The species used in TNFα chondrocyte inflammation models in all of the reviewed literature included human (51.43%), rodent (14.28%), goat (2.86%), bovine (22.86%), equine (2.86%), and porcine (5.71%) (Figure 6).

2.6. Mechanical Stimuli

Five studies dealt with mechanical stimulation [61,62,66,67,68]. We summarized the different mechanical stimuli on the basis of the type of load, the type and magnitude of the load, the frequency and duration of the load, the experimental setting, and the device used to deliver the mechanical stimuli. The parameters used varied between the studies and models. In summary, the experiments primarily utilized either a construct-based or an explant-based TNFα chondrocyte inflammation model to investigate cartilage/chondrocyte responses under various conditions and loading scenarios. See Supplementary Table S1 for further details.

2.7. Common Methods of Evaluating Experimental Results

We summarized the target genes identified by PCR from the included literature [46,50,51,53,58,61,64,65,66,70]. Monolayer-based models used a wider variety of genes (16 in total). The target genes selected in the three models overlapped somewhat, with collagen type 2, aggrecan, matrix metalloproteinases (MMP-3, MMP-13), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5), and TNFα appearing in almost all of the models (Supplementary Table S2).
For tissue staining techniques, four histological staining methods were used to access cartilage degradation [47,58,60,61,68]. These were toluidine blue staining, safranin O/fast green staining, alcin blue staining, and picrosirus red staining. Toluidine blue was the most commonly used method (Supplementary Table S3).
The target proteins in the immunohistochemical analysis varied from the structural proteins collagen type 1, collagen type 2, and aggrecan to cartilage matrix degrading enzymes, including MMP-3 and MMP-13, and signaling pathway proteins [47,50,51,58,59,61]. Among these proteins, collagen type 2 was the most abundant. More specific results are shown in Supplementary Table S4.
We also summarized the signaling pathways frequently addressed in the literature [50,56,57,69,71,72,73,74,75,76,77,78]. These studies encompassed several pathways, including the NF-κb, MAPK, Jak-STAT, PI3K-Akt, and HIF pathways. The NF-κb and MAPK pathways were the most frequently investigated pathways (Supplementary Table S5).

3. Discussion

In this review, we systematically analyzed TNFα-related models of inflammation in chondrocytes and cartilage tissue. These models were classified into four main categories: monolayer-based, construct-based, explant-based TNFα chondrocyte inflammation models, and miscellaneous models. Among these, monolayer-based TNFα chondrocyte inflammation models (45.16%) were the most prevalent. The human species (51.43%) was most frequently identified in the included studies. Furthermore, the concentration of added TNFα and the duration of cytokine stimulation were model-dependent. The most commonly used concentrations for the monolayer, construct, and explant-based TNFα chondrocyte inflammation models were 10 ng/mL, 10 ng/mL, and 100 ng/mL, respectively (66.67%, 27.27%, and 45.46%). In addition, the most commonly used stimulation durations for monolayer-based and construct-based TNFα chondrocyte inflammation models were 24 h, 48 h, and 3 days, respectively (39.99%, 50.00%, and 18.19%). The chondrocytes used to construct the models were typically derived from passages 0–3, with passage 1 accounting for the majority (60.87%). We also summarized the mechanical parameters used to construct TNFα chondrocyte inflammation models, PCR gene targets, cartilage histology staining methods, immunohistochemistry targets, and pathways involved in these studies.

3.1. TNFα Chondrocyte Inflammation—Differences in Available Models

Different models have different strengths and are suitable for studying different aspects of the disease. Monolayer-based TNFα chondrocyte inflammation models have a precise regulation of chondrocyte culture composition and concentration, as well as oxygen pressure. However, monolayer cultures are not physiological and affect the functional phenotype of the cells and the resulting tissue [79,80]. Chondrocytes lose their cartilage phenotype and express a fibroblast phenotype during monolayer cell culture. Roman-Blas et al. [70] and Shakibaei et al. [69] maintained the chondrocyte phenotype by suspension culture and by combining 2D and 3D cultures. The disadvantage of the inability of this model to receive mechanical stimuli can also be overcome by combining this model with a 3D model and a joint-specific bioreactor [81].
Construct-based TNFα inflammation models could provide a 3D environment for chondrocytes. Cells are seeded into the constructs, which promotes chondrogenic differentiation. Mechanical stimulation can be applied to the constructs to better mimic the living environment of cartilage in the body. The scaffold materials for this model are the most critical. The material should be biocompatible, not cause severe inflammatory reactions, and allow adequate diffusion of nutrients to the cells [82]. The materials typically used for constructs are agarose, alginate, and hyaluronic acid, and, for scaffolds, polyethylene glycol, acrylamide hydrogels, or other porous materials [83,84]. The most common types of cells seeded are primary chondrocytes or differentiated chondrocytes derived from mesenchymal stromal cells. Mohanraj et al. [60] used an agarose construct model and showed that chondrocytes derived from bone marrow stem cells (MSCs) were more easily influenced by the inflammatory environment compared to native chondrocytes.
Cartilage explant models mimic the cytokine response in a relatively natural environment, allowing the degradation of the extracellular matrix to be monitored [79]. And it is easier to accept mechanical stimulation that can induce chondrogenesis [85,86]. The disadvantage is that the volume of cartilage from small animals such as the mouse is limited, and the cartilage structure is different between small animals and humans. The cartilage structure, biochemical composition, and functional properties of large animals are closer to those of humans. These models include porcine or equine species, but these experiments are more time consuming, costly, and usually involve ethical issues [87]. Animal samples can be obtained from animal models with standardized induced disease simulation or from completely healthy animals. Human cartilage tissue is usually obtained from healthy patients as organ donors after an accident or from patients with late-stage OA who undergo an arthroplasty procedure. These differences in phenotype are important to consider when comparing studies. In addition, organ donors often do not provide completely healthy cartilage, which is more likely to be expected from juvenile donors. Again, the age of the donor is important. It is therefore difficult to obtain completely healthy or relatively healthy cartilage, and such human cartilage tissue is not suitable for modeling the early stages of inflammatory diseases. There is also the problem that the quality of the sample depends on the site from which it is taken. It has been suggested that cartilage tissue from the knee joint loses more GAGs than cartilage tissue from the ankle joint when subjected to the same mechanical stimuli [68]. And cells at the edge of the cartilage explant are more likely to die than those in the center, skewing the observations at these sites [88].
The lack of harmonization in the construction of TNFα inflammation models makes it difficult to compare the results of different studies. Based on the results of this review, the commonly used TNFα concentration (100 ng/mL) for explant-based models of these studies is higher than that (10 ng/mL) for monolayer and construct-based models. While the most commonly used concentration remains the same for monolayer and construct-based models, the duration of TNFα stimulation (48 h) for construct-based models is longer than that (24 h) for monolayer-based models.
In this review, five studies reported the type of recombinant TNFα used [46,47,59,66,68]. It is unclear whether the use of different species of recombinant TNFα in homologous models results in differences. The need to use recombinant TNFα that is homologous to the experimental cells/cartilage remains to be clarified. The lack of uniformity in model construction makes it difficult to compare the results between the studies.

3.2. High Diversity of Outcome Assessments in Chondrocyte Inflammation Models with TNFα

In general, the primary goals of TNFα chondrocyte inflammation models are either to study the pathology of cartilage inflammatory diseases, to investigate the underlying mechanisms, or to test or improve the efficacy of treatments. Techniques such as histological assessment, biomarker measurement, and molecular biology analysis have proven useful in achieving these goals. For some models, mechanical assessments can play an important role.
The common genes associated with TNFα chondrocyte inflammation models are summarized in Supplementary Table S2. Genes can be categorized into several classes: those related to cartilage matrix components and their synthesis, such as aggrecan, PRG4, collagen type 2, collagen type 1, collagen type X, and SOX9; genes associated with extracellular matrix remodeling and turnover, such as MMPs, ADAMTS5, and their tissue inhibitors of metalloproteinases (TIMPs); and genes associated with the inflammatory response, such as iNOS and cyclooxygenase-2 (COX-2), interleukin (IL)-6, and IL-8. The selection of gene targets varied between the models, but there is no doubt that the matrix-related genes collagen type 2, aggrecan, MMP-3, MMP-13, and ADAMTS 5 are critical genes in the study of cartilage inflammation and degeneration.
Proteoglycans and collagen are important components of healthy joint cartilage [89]. In addition to HE staining, histological staining techniques include picrosirius red staining to visualize the collagen network in the extracellular matrix, and alcian blue and toluidine blue staining for sulfated glycosaminoglycans (sGAG), a critical component of proteoglycans. A combination of safranin-O and fast green staining is also used to highlight the presence of glycosaminoglycans. These staining methods provide a qualitative visualization of the spatial distribution of collagen and proteoglycans within the tissue. Different models show variations in the choice of staining methods.
Immunohistochemistry (IHC) can also be used to understand the distribution and content of different collagen fibers using antibodies against different collagen fibers (e.g., collagen type 1 and collagen type 2). Of the three models, collagen type 2 was selected as the target protein in all cases. When performing IHC, a 3D model is usually chosen. This is because there is more matrix to stain. When chondrocytes are cultured in a monolayer, there is little matrix secreted. Interestingly, however, Chen et al. [56] performed IHC staining of collagen type 2 by culturing cells on coverslips to identify chondrocytes.
In addition to histology and IHC, biochemical assays such as the dimethylmethylene blue (DMMB) method can assess the degradation of sulfated glycosaminoglycans in the cartilage matrix. These methods evaluate changes in the cartilage matrix composition from both a quantitative and qualitative perspective.
In 3D models, mechanical assessments are essential to understand the mechanical response of cartilage tissue or constructs to various stimuli. Compression tests and the determination of Young’s modulus are commonly used to assess mechanical properties [83,90,91,92]. In addition, there are other ways to measure mechanical properties that are related to specific experimental models. In the study by Bevill et al. [66], creep consolidation and the cyclic strain of cartilage were tested to assess the mechanical properties of cartilage tissue after mechanical stimulation. Djouad et al. [58] used a push-out test to determine the interface strength between the chondrocyte-loaded construct and natural cartilage.

3.3. Mechanical Stimulation as Key for the Simulation of Physiologic Conditions

In the physiological context, articular cartilage is routinely subjected to a variety of mechanical forces, including compression, shear, hydrostatic pressure, and tensile stress; it is dynamic, intermittent, and multi-axial. An adequate mechanical force is required to maintain extracellular homeostasis for chondrocytes. However, it is important to avoid overloading, as excessive mechanical stress can trigger apoptosis or inflammatory cascade within the tissue [79,93,94,95].
Regarding the duration of mechanical loading, it is common for mechanical injury simulations to involve a single application at the beginning of the experiment, followed by continued experiments with the injured specimens. In contrast, the duration of regular mechanical stimuli may vary depending on the specific research objectives.
Dynamic, intermittent, and multi-axial mechanical loading is often used to mimic the mechanical stresses experienced by articular cartilage during daily activities. The choice of frequency in mechanical loading experiments should be based on the research objectives and the physiological conditions you are trying to mimic. Lower frequencies (e.g., 0.01–1 Hz) are closer to the frequency of activities such as walking, rather than higher applied frequencies (e.g., 1–10 Hz). However, researchers should also consider the variations between the different models and research objectives. As different joint cartilage tissues experience varying levels of mechanical pressure under physiological conditions, the level of mechanical stress should be determined based on the experimental objectives and sample types. For example, during activities such as walking and standing, the knee joint typically experiences pressures in the range of 2 to 12 MPa, while the hip joint encounters 1 to 4 MPa. Shoulder joints typically experience 0.5 to 2 MPa. There may be a threshold for enhancing cartilage repair effects, but this threshold varies between the different models [61,66,96,97,98].

3.4. Limitations

This systematic review focused on the inflammation model with an external stimulation by the recombinant protein TNFα. There are other inflammation models available, which were not included in this review. In these models, only the stimulus induced by TNFα is taken into account, but not when TNFα is produced in vitro on the basis of the stimulus (e.g., by stimulating chondrocytes with MSU crystals or CPPD). This criticism is part of the fact that the translational significance of using chondrocytes to study cartilage inflammation is not yet clear.

4. Materials and Methods

4.1. Exclusion and Inclusion Criteria and Study Selection

This systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [99]. A comprehensive search of the electronic medical database PubMed was performed by two independent authors (SK and SW) from 1 August 1986 to 20 July 2023 using the search terms “TNF alpha” OR “TNFα” AND “chondrocyte” OR “cartilage”. Only English titles were included.
Studies included in the systematic review were those that investigated the pro-inflammatory effect of TNFα on primary chondrocytes, engineered chondrocyte constructs, or cartilage explants. A primary screening of titles and abstracts was conducted by including studies of any level of evidence published in peer-reviewed journals reporting results in English. After screening, all the included literature was read in full for further screening against the exclusion criteria. Exclusion criteria included (1) a lack of information on chondrocyte passage, species, stimulus duration, and the dosage of cytokines, or other stimuli. (2) Articles that did not use primary chondrocytes, such as those employing chondrocyte cell lines or chondrocytes derived from mesenchymal stem cells (MSCs), or fibroblast cells, were also excluded. This exclusion was justified for better comparability. (3) Articles using cytokines other than TNFα to induce inflammation were also excluded. (4) Full-text articles that were not accessible were also excluded.
Two authors independently performed the study selection and any discrepancies in the selection process were resolved by discussion. A senior investigator (RO) was consulted to review the selection process.

4.2. Data Collection Process

All data were extracted from the article text, tables, and figures. Two investigators independently reviewed each article. Discrepancies between the two reviewers were resolved through discussion and consensus. The final results were then reviewed by the senior investigator (RO). Data were extracted from each included report, including information on (1) the species of chondrocyte or cartilage, (2) the passage number of chondrocytes, (3) concentration and type of TNFα used, (4) the duration of each stimulus, (5) the type of intervention (including the type of TNFα, concentration, duration, and mechanical stimuli), (6) the type of inflammation model used, (7) the main results reported, and (8) the PCR primers used in the experiment.

5. Conclusions

Currently, there is a wide variety of TNFα chondrocyte inflammation models. In this review, models are categorized into four main groups: monolayer-based, construct-based, explant-based TNFα chondrocyte inflammation models, and other types of models. Different models need to be selected depending on the objective of the research. Meanwhile, specific factors such as species selection, TNFα concentration, and stimulation period, whether mechanical stimulation is used, vary between the models. The lack of standardization in these TNFα chondrocyte inflammation models makes comparisons between the models difficult. More systematic studies are needed to standardize the specific models. This systematic review provides an overview of the existing studies and is a reference for a better comparison of TNFα-induced inflammatory models.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms251910805/s1.

Author Contributions

Conceptualization, F.A.S. and R.O.; methodology, S.W., S.K. and R.O.; investigation, S.W., S.K., C.B., D.C.W., F.A.S. and R.O.; writing—original draft preparation, S.W., F.A.S. and R.O.; and writing—review and editing, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This study was funded by the Deutsche Arthrosehilfe e.V., the BONFOR-Forschungskommission der Medizinischen Fakultät Bonn, and the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) under Germany’s Excellence Strategy—EXC2151—390873048 and the John Templeton Foundation (grant #62214).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The original contributions presented in this study are included in the article/Supplementary Material; further inquiries can be directed to the corresponding author.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

References

  1. Baud, V.; Karin, M. Signal transduction by tumor necrosis factor and its relatives. Trends Cell Biol. 2001, 11, 372–377. [Google Scholar] [CrossRef] [PubMed]
  2. Horiuchi, T.; Mitoma, H.; Harashima, S.-I.; Tsukamoto, H.; Shimoda, T. Transmembrane TNF-alpha: Structure, function and interaction with anti-TNF agents. Rheumatology 2010, 49, 1215–1228. [Google Scholar] [CrossRef] [PubMed]
  3. Jiang, Y.; Yu, M.; Hu, X.; Han, L.; Yang, K.; Ba, H.; Zhang, Z.; Yin, B.; Yang, X.-P.; Li, Z.; et al. STAT1 mediates transmembrane TNF-alpha-induced formation of death-inducing signaling complex and apoptotic signaling via TNFR1. Cell Death Differ. 2017, 24, 660–671. [Google Scholar] [CrossRef] [PubMed]
  4. Bradley, J.R. TNF-mediated inflammatory disease. J. Pathol. 2008, 214, 149–160. [Google Scholar] [CrossRef] [PubMed]
  5. Jang, D.-I.; Lee, A.-H.; Shin, H.-Y.; Song, H.-R.; Park, J.-H.; Kang, T.-B.; Lee, S.-R.; Yang, S.-H. The Role of Tumor Necrosis Factor Alpha (TNF-α) in Autoimmune Disease and Current TNF-α Inhibitors in Therapeutics. Int. J. Mol. Sci. 2021, 22, 2719. [Google Scholar] [CrossRef]
  6. Smith, C.A.; Davis, T.; Anderson, D.; Solam, L.; Beckmann, M.P.; Jerzy, R.; Dower, S.K.; Cosman, D.; Goodwin, R.G. A Receptor for Tumor Necrosis Factor Defines an Unusual Family of Cellular and Viral Proteins. Science 1990, 248, 1019–1023. [Google Scholar] [CrossRef]
  7. Vandenabeele, P.; Declercq, W.; Beyaert, R.; Fiers, W. Two tumour necrosis factor receptors: Structure and function. Trends Cell Biol. 1995, 5, 392–399. [Google Scholar] [CrossRef]
  8. Catrina, A.I.; Klint, E.A.; Ernestam, S.; Catrina, S.; Makrygiannakis, D.; Botusan, I.R.; Klareskog, L.; Ulfgren, A. Anti-tumor necrosis factor therapy increases synovial osteoprotegerin expression in rheumatoid arthritis. Arthritis Rheum. 2006, 54, 76–81. [Google Scholar] [CrossRef]
  9. Wehling, N.; Palmer, G.D.; Pilapil, C.; Liu, F.; Wells, J.W.; Müller, P.E.; Evans, C.H.; Porter, R.M. Interleukin-1beta and tumor necrosis factor alpha inhibit chondrogenesis by human mesenchymal stem cells through NF-kappaB-dependent pathways. Arthritis Rheum. 2009, 60, 801–812. [Google Scholar] [CrossRef]
  10. Murakami, S.; Lefebvre, V.; de Crombrugghe, B. Potent inhibition of the master chondrogenic factor Sox9 gene by interleukin-1 and tumor necrosis factor-alpha. J. Biol. Chem. 2000, 275, 3687–3692. [Google Scholar] [CrossRef]
  11. Duprez, L.; Takahashi, N.; Van Hauwermeiren, F.; Vandendriessche, B.; Goossens, V.; Vanden Berghe, T.; Declercq, W.; Libert, C.; Cauwels, A.; Vandenabeele, P. RIP kinase-dependent necrosis drives lethal systemic inflammatory response syndrome. Immunity 2011, 35, 908–918. [Google Scholar] [CrossRef] [PubMed]
  12. Wu, S.; Fadoju, D.; Rezvani, G.; De Luca, F. Stimulatory effects of insulin-like growth factor-I on growth plate chondrogenesis are mediated by nuclear factor-kappaB p65. J. Biol. Chem. 2008, 283, 34037–34044. [Google Scholar] [CrossRef] [PubMed]
  13. Oberst, A.; Dillon, C.P.; Weinlich, R.; McCormick, L.L.; Fitzgerald, P.; Pop, C.; Hakem, R.; Salvesen, G.S.; Green, D.R. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 2011, 471, 363–367. [Google Scholar] [CrossRef] [PubMed]
  14. van Loo, G.; Bertrand, M.J.M. Death by TNF: A road to inflammation. Nat. Rev. Immunol. 2023, 23, 289–303. [Google Scholar] [CrossRef]
  15. Ahmad, R.; Sylvester, J.; Ahmad, M.; Zafarullah, M. Involvement of H-Ras and reactive oxygen species in proinflammatory cytokine-induced matrix metalloproteinase-13 expression in human articular chondrocytes. Arch. Biochem. Biophys. 2011, 507, 350–355. [Google Scholar] [CrossRef]
  16. Ben-Aderet, L.; Merquiol, E.; Fahham, D.; Kumar, A.; Reich, E.; Ben-Nun, Y.; Kandel, L.; Haze, A.; Liebergall, M.; Kosińska, M.K.; et al. Detecting cathepsin activity in human osteoarthritis via activity-based probes. Arthritis Res. Ther. 2015, 17, 1–14. [Google Scholar] [CrossRef]
  17. Guo, F.J.; Xiong, Z.; Lu, X.; Ye, M.; Han, X.; Jiang, R. ATF6 upregulates XBP1S and inhibits ER stress-mediated apoptosis in osteoarthritis cartilage. Cell. Signal. 2014, 26, 332–342. [Google Scholar] [CrossRef]
  18. Heinecke, L.F.; Grzanna, M.W.; Au, A.Y.; Mochal, C.A.; Rashmir-Raven, A.; Frondoza, C.G. Inhibition of cyclooxygenase-2 expression and prostaglandin E2 production in chondrocytes by avocado soybean unsaponifiables and epigallocatechin gallate. Osteoarthr. Cartil. 2010, 18, 220–227. [Google Scholar] [CrossRef]
  19. Imamura, T.; Imamura, C.; McAlinden, A.; Davies, S.R.; Iwamoto, Y.; Sandell, L.J. A novel tumor necrosis factor alpha-responsive CCAAT/enhancer binding protein site regulates expression of the cartilage-derived retinoic acid-sensitive protein gene in cartilage. Arthritis Rheum. 2008, 58, 1366–1376. [Google Scholar] [CrossRef]
  20. Kunisch, E.; Kinne, R.W.; Alsalameh, R.J.; Alsalameh, S. Pro-inflammatory IL-1beta and/or TNF-alpha up-regulate matrix metalloproteases-1 and -3 mRNA in chondrocyte subpopulations potentially pathogenic in osteoarthritis: In situ hybridization studies on a single cell level. Int. J. Rheum. Dis. 2016, 19, 557–566. [Google Scholar] [CrossRef]
  21. Madhavan, S.; Anghelina, M.; Sjostrom, D.; Dossumbekova, A.; Guttridge, D.C.; Agarwal, S. Biomechanical signals suppress TAK1 activation to inhibit NF-kappaB transcriptional activation in fibrochondrocytes. J. Immunol. 2007, 179, 6246–6254. [Google Scholar] [CrossRef] [PubMed]
  22. Qu, R.; Chen, X.; Wang, W.; Qiu, C.; Ban, M.; Guo, L.; Vasilev, K.; Chen, J.; Li, W.; Zhao, Y. Ghrelin protects against osteoarthritis through interplay with Akt and NF–kB signaling pathways. FASEB J. 2018, 32, 1044–1058. [Google Scholar] [CrossRef] [PubMed]
  23. Qu, Y.; Liu, Y.; Li, R. FSTL1 promotes inflammatory reaction and cartilage catabolism through interplay with NFkB signaling pathways in an in vitro ONFH model. Inflammation 2019, 42, 1491–1503. [Google Scholar] [CrossRef] [PubMed]
  24. Sirikaew, N.; Chomdej, S.; Tangyuenyong, S.; Tangjitjaroen, W.; Somgird, C.; Thitaram, C.; Ongchai, S. Proinflammatory cytokines and lipopolysaccharides up regulate MMP-3 and MMP-13 production in Asian elephant (Elephas maximus) chondrocytes: Attenuation by anti-arthritic agents. BMC Vet. Res. 2019, 15, 419. [Google Scholar] [CrossRef]
  25. Sommaggio, R.; Máñez, R.; Costa, C. TNF, pig CD86, and VCAM-1 identified as potential targets for intervention in xenotransplantation of pig chondrocytes. Cell Transplant. 2009, 18, 1381–1393. [Google Scholar] [CrossRef] [PubMed]
  26. Vaamonde-García, C.; Riveiro-Naveira, R.R.; Valcárcel-Ares, M.N.; Hermida-Carballo, L.; Blanco, F.J.; López-Armada, M.J. Mitochondrial dysfunction increases inflammatory responsiveness to cytokines in normal human chondrocytes. Arthritis Rheum. 2012, 64, 2927–2936. [Google Scholar] [CrossRef]
  27. Wang, W.; Li, J.; Li, F.; Peng, J.; Xu, M.; Shangguan, Y.; Li, Y.; Zhao, Y.; Qiu, C.; Qu, R.; et al. Scutellarin suppresses cartilage destruction in osteoarthritis mouse model by inhibiting the NF-kB and PI3K/AKT signaling pathways. Int. Immunopharmacol. 2019, 77, 105928. [Google Scholar] [CrossRef]
  28. Wang, J.; Wang, X.; Cao, Y.; Huang, T.; Song, D.X.; Tao, H.R. Therapeutic potential of hyaluronic acid/chitosan nanoparticles for the delivery of curcuminoid in knee osteoarthritis and an invitro evaluation in chondrocytes. Int. J. Mol. Med. 2018, 42, 2604–2614. [Google Scholar] [CrossRef]
  29. Weinmann, D.; Mueller, M.; Walzer, S.M.; Hobusch, G.M.; Lass, R.; Gahleitner, C.; Viernstein, H.; Windhager, R.; Toegel, S. Brazilin blocks catabolic processes in human osteoarthritic chondrocytes via inhibition of NFKB1/p50. J. Orthop. Res. 2018, 36, 2431–2438. [Google Scholar] [CrossRef]
  30. Woodell-May, J.; Matuska, A.; Oyster, M.; Welch, Z.; O’Shaughnessey, K.; Hoeppner, J. Autologous protein solution inhibits MMP-13 production by IL-1b and TNFa-stimulated human articular chondrocytes. J. Orthop. Res. 2011, 29, 1320–1326. [Google Scholar] [CrossRef]
  31. Zhi, L.Q.; Yao, S.X.; Liu, H.L.; Li, M.; Duan, N.; Ma, J.B. Hydroxytyrosol inhibits the inflammatory response of osteoarthritis chondrocytes via SIRT6-mediated autophagy. Mol. Med. Rep. 2017, 17, 4035–4042. [Google Scholar] [CrossRef] [PubMed]
  32. Zhou, R.-P.; Dai, B.-B.; Xie, Y.-Y.; Wu, X.-S.; Li, Y.; Wang, Z.-Q.; Zu, S.-Q.; Ge, J.-F.; Chen, F.-H. Interleukin-1b and tumor necrosis factor-a augment acidosis-induced rat articular chondrocyte apoptosis via nuclear factor-kappaB-dependent upregulation of ASIC1a channel. Biochim. Et Biophys. Acta(BBA)-Mol. Basis Dis. 2018, 1864, 162–177. [Google Scholar] [CrossRef] [PubMed]
  33. Žigon-Branc, S.; Barlič, A.; Knežević, M.; Jeras, M.; Vunjak-Novakovic, G. Testing the potency of anti-TNF-a and anti-IL-1b drugs using spheroid cultures of human osteoarthritic chondrocytes and donor-matched chondrogenically differentiated mesenchymal stem cells. Biotechnol. Prog. 2018, 34, 1045–1058. [Google Scholar] [CrossRef]
  34. Xiang, X.; Zhou, Y.; Sun, H.; Tan, S.; Lu, Z.; Huang, L.; Wang, W. Ivabradine abrogates TNF-a-induced degradation of articular cartilage matrix. Int. Immunopharmacol. 2019, 66, 347–353. [Google Scholar] [CrossRef] [PubMed]
  35. Cillero-Pastor, B.; Ruiz-Romero, C.; Caramés, B.; López-Armada, M.J.; Blanco, F.J. Proteomic analysis by two-dimensional electrophoresis to identify the normal human chondrocyte proteome stimulated by tumor necrosis factor alpha and interleukin-1beta. Arthritis Rheum. 2010, 62, 802–814. [Google Scholar] [CrossRef] [PubMed]
  36. Wenger, R. Hydrostatic Pressure Increases Apoptosis in Cartilage-Constructs Produced From Human Osteoarthritic Chondrocytes. Front. Biosci. 2006, 11, 1690. [Google Scholar] [CrossRef] [PubMed]
  37. Olkkonen, J.; Kouri, V.P.; Hynninen, J.; Konttinen, Y.T.; Mandelin, J. Differentially expressed in chondrocytes 2 (DEC2) increases the expression of IL-1b and is abundantly present in synovial membrane in rheumatoid arthritis. PLoS ONE 2015, 10, e0145279. [Google Scholar] [CrossRef]
  38. Wang, L.; Yang, M.; Zhang, C.; Huang, F. The protective effects of dehydrocostus lactone against TNF-a-induced degeneration of extracellular matrix (ECM) in SW1353 cells. Aging 2020, 12, 17137–17149. [Google Scholar] [CrossRef]
  39. Huang, Y.; Jiang, T.; Chen, J.; Yin, G.Y.; Fan, J. Effects of kartogenin on the attenuated nucleus pulposus cell degeneration of intervertebral discs induced by interleukin-1b and tumor necrosis factor-a. Int. J. Mol. Med. 2017, 41, 749–756. [Google Scholar] [CrossRef]
  40. Jones, S.W.; Brockbank, S.M.V.; Clements, K.M.; Le Good, N.; Campbell, D.; Read, S.J.; Newham, P. Mitogen-activated protein kinase-activated protein kinase 2 (MK2) modulates key biological pathways associated with OA disease pathology. Osteoarthr. Cartil. 2009, 17, 124–131. [Google Scholar] [CrossRef]
  41. Rainbow, R.S.; Kwon, H.; Foote, A.T.; Preda, R.C.; Kaplan, D.L.; Zeng, L. Muscle cell-derived factors inhibit inflammatory stimuli-induced damage in hMSC-derived chondrocytes. Osteoarthr. Cartil. 2013, 21, 990–998. [Google Scholar] [CrossRef] [PubMed]
  42. Wan, Q.; Xu, W.; Yan, J.-L.; Yokota, H.; Na, S. Distinctive subcellular inhibition of cytokine-induced src by salubrinal and fluid flow. PLoS ONE 2014, 9, e105699. [Google Scholar] [CrossRef] [PubMed]
  43. Wang, C.; Qu, L. The anti-fibrotic agent nintedanib protects chondrocytes against tumor necrosis factor-a (TNF-a)-induced extracellular matrix degradation. Bioengineered 2022, 13, 5318–5329. [Google Scholar] [CrossRef] [PubMed]
  44. Xiong, X.; Liu, L.; Xu, F.; Wu, X.; Yin, Z.; Dong, Y.; Qian, P. Feprazone ameliorates TNF-a-Induced loss of aggrecan via inhibition of the SOX-4/ADAMTS-5 signaling pathway. ACS Omega 2021, 6, 7638–7645. [Google Scholar] [CrossRef] [PubMed]
  45. Zhang, X.; Dong, Y.; Dong, H.; Cui, Y.; Du, Q.; Wang, X.; Li, L.; Zhang, H. Telmisartan mitigates TNF-a-Induced type II collagen reduction by upregulating SOX-9. ACS Omega 2021, 6, 11756–11761. [Google Scholar] [CrossRef]
  46. Kim, H.A.; Yeo, Y.; Jung, H.A.; Jung, Y.O.; Park, S.J.; Kim, S.J. Phase 2 enzyme inducer sulphoraphane blocks prostaglandin and nitric oxide synthesis in human articular chondrocytes and inhibits cartilage matrix degradation. Rheumatology 2012, 51, 1006–1016. [Google Scholar] [CrossRef]
  47. Malemud, C.J.; Sun, Y.; Pearlman, E.; Ginley, N.M.; Awadallah, A.; Wisler, B.A.; Dennis, J.E. Monosodium urate and tumor necrosis factor-a increase apoptosis in human chondrocyte cultures. Rheumatology 2012, 2, 113. [Google Scholar] [CrossRef]
  48. López-Armada, M.J.; Caramés, B.; Martín, M.A.; Cillero-Pastor, B.; Lires-Dean, M.; Fuentes-Boquete, I.; Arenas, J.; Blanco, F.J. Mitochondrial activity is modulated by TNFalpha and IL-1beta in normal human chondrocyte cells. Osteoarthr. Cartil. 2006, 14, 1011–1022. [Google Scholar] [CrossRef]
  49. López-Armada, M.J.; Caramés, B.; Lires-Deán, M.; Cillero-Pastor, B.; Ruiz-Romero, C.; Galdo, F.; Blanco, F. 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]
  50. Boileau, C.; Amiable, N.; Martel-Pelletier, J.; Fahmi, H.; Duval, N.; Pelletier, J.P. Activation of proteinase-activated receptor 2 in human osteoarthritic cartilage upregulates catabolic and proinflammatory pathways capable of inducing cartilage degradation: A basic science study. Arthritis Res. Ther. 2007, 9, R121. [Google Scholar] [CrossRef]
  51. Tardif, G. Differential regulation of the bone morphogenic protein antagonist chordin in human normal and osteoarthritic chondrocytes. Ann. Rheum. Dis. 2006, 65, 261–264. [Google Scholar] [CrossRef] [PubMed]
  52. Caramés, B.; López-Armada, M.J.; Cillero-Pastor, B.; Lires-Dean, M.; Vaamonde, C.; Galdo, F.; Blanco, F. Differential effects of tumor necrosis factor-alpha and interleukin-1beta on cell death in human articular chondrocytes. Osteoarthr. Cartil. 2008, 16, 715–722. [Google Scholar] [CrossRef] [PubMed]
  53. Yik, J.H.N.; Hu, Z.; Kumari, R.; Christiansen, B.A.; Haudenschild, D.R. Cyclin-Dependent Kinase 9 Inhibition Protects Cartilage From the Catabolic Effects of Proinflammatory Cytokines. Arthritis Rheumatol. 2014, 66, 1537–1546. [Google Scholar] [CrossRef] [PubMed]
  54. Ding, L.; Buckwalter, J.A.; Martin, J.A. DAMPs synergize with cytokines or fibronectin fragment on inducing chondrolysis but lose effect when acting alone. Mediat. Inflamm. 2017, 2017, 2642549. [Google Scholar] [CrossRef] [PubMed]
  55. Terkeltaub, R.; Yang, B.; Lotz, M.; Liu-Bryan, R. Chondrocyte AMP-activated protein kinase activity suppresses matrix degradation responses to proinflammatory cytokines interleukin-1b and tumor necrosis factor a. Arthritis Rheum. 2011, 63, 1928–1937. [Google Scholar] [CrossRef]
  56. Chen, H.; Shao, X.; Li, L.; Zheng, C.; Xu, X.; Hong, X.; Li, X.; Wu, M. Electroacupuncture serum inhibits TNF-a-mediated chondrocyte inflammation via the Ras-Raf-MEK1/2-ERK1/2 signaling pathway. Mol. Med. Rep. 2017, 16, 5807–5814. [Google Scholar] [CrossRef]
  57. Cao, Y.; Zhang, X.; Shang, W.; Xu, J.; Wang, X.; Hu, X.; Ao, Y.; Cheng, H. Proinflammatory Cytokines Stimulate Mitochondrial Superoxide Flashes in Articular Chondrocytes In Vitro and In Situ. PLoS ONE 2013, 8, e66444. [Google Scholar] [CrossRef]
  58. Djouad, F.; Rackwitz, L.; Song, Y.; Janjanin, S.; Tuan, R.S. ERK1/2 activation induced by inflammatory cytokines compromises effective host tissue integration of engineered cartilage. Tissue Eng. Part A 2009, 15, 2825–2835. [Google Scholar] [CrossRef]
  59. Chen, C.; Xie, J.; Rajappa, R.; Deng, L.; Fredberg, J.; Yang, L. Interleukin-1b and tumor necrosis factor-a increase stiffness and impair contractile function of articular chondrocytes. Acta Biochim. Biophys. Sin. 2015, 47, 121–129. [Google Scholar] [CrossRef]
  60. Mohanraj, B.; Huang, A.H.; Yeger–McKeever, M.J.; Schmidt, M.J.; Dodge, G.R.; Mauck, R.L. Chondrocyte and mesenchymal stem cell derived engineered cartilage exhibits differential sensitivity to pro–inflammatory cytokines. J. Orthop. Res. 2018, 36, 2901–2910. [Google Scholar] [CrossRef]
  61. Ossendorff, R.; Grad, S.; Stoddart, M.J.; Alini, M.; Schmal, H.; Südkamp, N.; Salzmann, G.M. Autologous chondrocyte implantation in osteoarthritic surroundings: TNFa and its inhibition by adalimumab in a knee-specific bioreactor. Am. J. Sports Med. 2018, 46, 431–440. [Google Scholar] [CrossRef] [PubMed]
  62. Tilwani, R.K.; Vessillier, S.; Pingguan-Murphy, B.; Lee, D.A.; Bader, D.L.; Chowdhury, T.T. Oxygen tension modulates the effects of TNFa in compressed chondrocytes. Inflamm. Res. 2017, 66, 49–58. [Google Scholar] [CrossRef] [PubMed]
  63. Morris, K.J.; Cs-Szabo, G.; Cole, A.A. Characterization of TIMP-3 in human articular talar cartilage. Connect. Tissue Res. 2010, 51, 478–490. [Google Scholar] [CrossRef] [PubMed]
  64. Little, C.B.; Flannery, C.R.; Hughes, C.E.; Goodship, A.; Caterson, B. Cytokine induced metalloproteinase expression and activity does not correlate with focal susceptibility of articular cartilage to degeneration. Osteoarthr. Cartil. 2005, 13, 162–170. [Google Scholar] [CrossRef] [PubMed]
  65. Little, C.B.; Flannery, C.R.; Hughes, C.E.; Mort, J.S.; Roughley, P.J.; Dent, C.; Caterson, B. Aggrecanase versus matrix metalloproteinases in the catabolism of the interglobular domain of aggrecan in vitro. Biochem. J. 1999, 344 Pt 1, 61–68. [Google Scholar] [CrossRef] [PubMed]
  66. Bevill, S.L.; Boyer, K.A.; Andriacchi, T.P. The Regional Sensitivity of Chondrocyte Gene Expression to Coactive Mechanical Load and Exogenous TNF-α Stimuli. J. Biomech. Eng. 2014, 136, 0910051–0910057. [Google Scholar] [CrossRef]
  67. Stevens, A.L.; Wishnok, J.S.; Chai, D.H.; Grodzinsky, A.J.; Tannenbaum, S.R. A sodium dodecyl sulfate-polyacrylamide gel electrophoresis-liquid chromatography tandem mass spectrometry analysis of bovine cartilage tissue response to mechanical compression injury and the inflammatory cytokines tumor necrosis factor alpha and interleukin-1beta. Arthritis Rheum. 2008, 58, 489–500. [Google Scholar] [CrossRef]
  68. Sui, Y.; Lee, J.H.; DiMicco, M.A.; Vanderploeg, E.J.; Blake, S.M.; Hung, H.; Plaas, A.H.K.; James, I.E.; Song, X.; Lark, M.W.; et al. Mechanical injury potentiates proteoglycan catabolism induced by interleukin-6 with soluble interleukin-6 receptor and tumor necrosis factor alpha in immature bovine and adult human articular cartilage. Arthritis Rheum. 2009, 60, 2985–2996. [Google Scholar] [CrossRef]
  69. Shakibaei, M.; John, T.; Schulze-Tanzil, G.; Lehmann, I.; Mobasheri, A. Suppression of NF-kappaB activation by curcumin leads to inhibition of expression of cyclo-oxygenase-2 and matrix metalloproteinase-9 in human articular chondrocytes: Implications for the treatment of osteoarthritis. Biochem. Pharmacol. 2007, 73, 1434–1445. [Google Scholar] [CrossRef]
  70. Roman-Blas, J.A.; Stokes, D.G.; Jimenez, S.A. Modulation of TGF-beta signaling by proinflammatory cytokines in articular chondrocytes. Osteoarthr. Cartil. 2007, 15, 1367–1377. [Google Scholar] [CrossRef]
  71. Yun, K.; Choi, Y.D.; Nam, J.H.; Park, Z.; Im, S.-H. NF-kappaB regulates Lef1 gene expression in chondrocytes. Biochem. Biophys. Res. Commun. 2007, 357, 589–595. [Google Scholar] [CrossRef] [PubMed]
  72. Saklatvala, J. Inflammatory signaling in cartilage: MAPK and NF-kappaB pathways in chondrocytes and the use of inhibitors for research into pathogenesis and therapy of osteoarthritis. Curr. Drug Targets 2007, 8, 305–313. [Google Scholar] [CrossRef] [PubMed]
  73. Allen, K.D.; Thoma, L.M.; Golightly, Y.M. Epidemiology of osteoarthritis. Osteoarthr. Cartil. 2022, 30, 184–195. [Google Scholar] [CrossRef] [PubMed]
  74. Li, Z.; Dai, A.; Yang, M.; Chen, S.; Deng, Z.; Li, L. p38MAPK Signaling Pathway in Osteoarthritis: Pathological and Therapeutic Aspects. J. Inflamm. Res. 2022, 15, 723–734. [Google Scholar] [CrossRef]
  75. Zhou, Q.; Ren, Q.; Jiao, L.; Huang, J.; Yi, J.; Chen, J.; Lai, J.; Ji, G.; Zheng, T. The potential roles of JAK/STAT signaling in the progression of osteoarthritis. Front. Endocrinol. 2022, 13, 1069057. [Google Scholar] [CrossRef]
  76. Saklatvala, J. Tumour necrosis factor alpha stimulates resorption and inhibits synthesis of proteoglycan in cartilage. Nature 1986, 322, 547–549. [Google Scholar] [CrossRef]
  77. Sun, K.; Luo, J.; Guo, J.; Yao, X.; Jing, X.; Guo, F. The PI3K/AKT/mTOR signaling pathway in osteoarthritis: A narrative review. Osteoarthr. Cartil. 2020, 28, 400–409. [Google Scholar] [CrossRef]
  78. Fu, L.; Zhang, L.; Zhang, X.; Chen, L.; Cai, Q.; Yang, X. Roles of oxygen level and hypoxia-inducible factor signaling pathway in cartilage, bone and osteochondral tissue engineering. Biomed. Mater. 2021, 16, 022006. [Google Scholar] [CrossRef]
  79. Johnson, C.I.; Argyle, D.J.; Clements, D.N. In vitro models for the study of osteoarthritis. Vet. J. 2016, 209, 40–49. [Google Scholar] [CrossRef]
  80. Cope, P.J.; Ourradi, K.; Li, Y.; Sharif, M. Models of osteoarthritis: The good, the bad and the promising. Osteoarthr. Cartil. 2019, 27, 230. [Google Scholar] [CrossRef]
  81. Grad, S.; Eglin, D.; Alini, M.; Stoddart, M.J. Physical stimulation of chondrogenic cells in vitro: A review. Clin. Orthop. Relat. Res. 2011, 469, 2764–2772. [Google Scholar] [CrossRef] [PubMed]
  82. O’Brien, F.J. Biomaterials & scaffolds for tissue engineering. Mater. Today 2011, 14, 88–95. [Google Scholar] [CrossRef]
  83. Patel, J.M.; Wise, B.C.; Bonnevie, E.D.; Mauck, R.L. A Systematic Review and Guide to Mechanical Testing for Articular Cartilage Tissue Engineering. Tissue Eng. Part C Methods 2019, 25, 593–608. [Google Scholar] [CrossRef] [PubMed]
  84. Reddi, A.H.; Becerra, J.; Andrades, J.A. Nanomaterials and hydrogel scaffolds for articular cartilage regeneration. Tissue Eng. Part B Rev. 2011, 17, 301–305. [Google Scholar] [CrossRef]
  85. van Haaften, E.E.; Ito, K.; van Donkelaar, C.C. The initial repair response of articular cartilage after mechanically induced damage. J. Orthop. Res. 2017, 35, 1265–1273. [Google Scholar] [CrossRef]
  86. Juhász, T.; Matta, C.; Somogyi, C.; Katona, E.; Takács, R.; Soha, R.F.; Szabó, I.A.; Cserháti, C.; Sződy, R.; Karácsonyi, Z.; et al. Mechanical loading stimulates chondrogenesis via the PKA/CREB-Sox9 and PP2A pathways in chicken micromass cultures. Cell. Signal. 2014, 26, 468–482. [Google Scholar] [CrossRef]
  87. McCoy, A.M. Animal Models of Osteoarthritis: Comparisons and Key Considerations. Vet. Pathol. 2015, 52, 803–818. [Google Scholar] [CrossRef]
  88. Huntley, J.S.; Simpson, A.H.; Hall, A.C. Use of non-degenerate human osteochondral tissue and confocal laser scanning microscopy for the study of chondrocyte death at cartilage surgery. Eur. Cells Mater. 2005, 9, 13–22. [Google Scholar] [CrossRef]
  89. Buckwalter, J.A.; Mankin, H.J.; Grodzinsky, A.J. Articular cartilage and osteoarthritis. Lect.-Am. Acad. Orthop. Surg. 2005, 54, 465–480. [Google Scholar]
  90. Athanasiou, K.A.; Darling, E.M.; Hu, J.C.; DuRaine, G.D.; Reddi, A.H. Articular Cartilage, 2nd ed.; Taylor & Francis: Abingdon, UK, 2016. [Google Scholar]
  91. Elahi, S.A.; Castro-Viñuelas, R.; Govaerts, A.; Lories, R.; Famaey, N.; Jonkers, I. Unconfined Compression Experimental Protocol for Cartilage Explants and Hydrogel Constructs: From Sample Preparation to Mechanical Characterization. Methods Mol. Biol. 2023, 2598, 271–287. [Google Scholar] [CrossRef]
  92. Olvera, D.; Daly, A.; Kelly, D.J. Mechanical Testing of Cartilage Constructs. Methods Mol. Biol. 2015, 1340, 279–287. [Google Scholar] [CrossRef] [PubMed]
  93. Henrotin, Y.; Pesesse, L.; Sanchez, C. Identification of Mechanosensitive Genes in Chondrocytes and Osteoblasts and Their Role in OA Pathogenesis. In Mechanical Stretch and Cytokines; Kamkin, A., Kiseleva, I., Eds.; Mechanosensitivity in Cells and Tissues; Springer: Dordrecht, The Netherlands, 2012; pp. 223–233. [Google Scholar] [CrossRef]
  94. Fang, T.; Zhou, X.; Jin, M.; Nie, J.; Li, X. Molecular mechanisms of mechanical load-induced osteoarthritis. Int. Orthop. 2021, 45, 1125–1136. [Google Scholar] [CrossRef] [PubMed]
  95. Logerstedt, D.S.; Ebert, J.R.; MacLeod, T.D.; Heiderscheit, B.C.; Gabbett, T.J.; Eckenrode, B.J. Effects of and Response to Mechanical Loading on the Knee. Sports Med. 2022, 52, 201–235. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Y.; Frank, E.H.; Wang, Y.; Chubinskaya, S.; Huang, H.H.; Grodzinsky, A.J. Moderate dynamic compression inhibits pro-catabolic response of cartilage to mechanical injury, tumor necrosis factor-a and interleukin-6, but accentuates degradation above a strain threshold. Osteoarthr. Cartil. 2013, 21, 1933–1941. [Google Scholar] [CrossRef] [PubMed]
  97. Torzilli, P.A.; Bhargava, M.; Park, S.; Chen, C. Mechanical Load Inhibits Il-1 Induced Matrix Degradation in Articular Cartilage. Osteoarthr. Cartil. 2010, 18, 97. [Google Scholar] [CrossRef]
  98. Natenstedt, J.; Kok, A.C.; Dankelman, J.; Tuijthof, G.J. What quantitative mechanical loading stimulates in vitro cultivation best? J. Exp. Orthop. 2015, 2, 15. [Google Scholar] [CrossRef]
  99. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: Explanation and elaboration. BMJ. 2009, 339, b2700. [Google Scholar] [CrossRef]
Figure 1. Identification and selection of studies via databases and registers.
Figure 1. Identification and selection of studies via databases and registers.
Ijms 25 10805 g001
Figure 2. Summary of TNFα chondrocyte inflammation models.
Figure 2. Summary of TNFα chondrocyte inflammation models.
Ijms 25 10805 g002
Figure 3. Concentration of TNFα in ng/mL used in different models (a): monolayer-based TNFα chondrocyte inflammation models, (b): construct-based TNFα chondrocyte inflammation models, and (c) explant-based TNFα chondrocyte inflammation models.
Figure 3. Concentration of TNFα in ng/mL used in different models (a): monolayer-based TNFα chondrocyte inflammation models, (b): construct-based TNFα chondrocyte inflammation models, and (c) explant-based TNFα chondrocyte inflammation models.
Ijms 25 10805 g003
Figure 4. Summary of TNFα stimulation durations in hours and/or days (a): monolayer-based models, (b): construct-based models, and (c): explant-based models.
Figure 4. Summary of TNFα stimulation durations in hours and/or days (a): monolayer-based models, (b): construct-based models, and (c): explant-based models.
Ijms 25 10805 g004
Figure 5. Passages’ (P0–P3) distribution of chondrocytes TNFα inflammation models.
Figure 5. Passages’ (P0–P3) distribution of chondrocytes TNFα inflammation models.
Ijms 25 10805 g005
Figure 6. Species distribution of TNFα chondrocyte inflammation models.
Figure 6. Species distribution of TNFα chondrocyte inflammation models.
Ijms 25 10805 g006
Table 1. Two-dimensional models: Monolayer-based TNFα chondrocyte inflammatory models.
Table 1. Two-dimensional models: Monolayer-based TNFα chondrocyte inflammatory models.
SpeciesAuthorsOriginAge
(Mean)
Concentration (ng/mL)PassageDurationMain Findings
HumanKim et al. [46]Knee OA69.5 y10124 hSulforaphane (SFN) hindered the synthesis of prostaglandin and nitric oxide in human articular chondrocytes, thereby preventing the degradation of cartilage matrix.
HumanMalemud et al. [47] OA
Adult knee
Juvenile knee
NA
NA
12 y
1011 hTNFα could increase apoptosis in normal human chondrocytes, OA chondrocytes and human juvenile chondrocyte pellet cultures, but not in chondrocyte pellet cultures initiated from MSCs.
HumanLópez-Armada et al. [48]Healthy knee59 y1016 daysTNFα and IL1β impacted the mitochondrial function of human articular chondrocytes, and the inhibition of complex I may be a factor in the cartilage breakdown caused by these cytokines.
HumanLópez-Armada et al. [49]Hip OANA1017 daysTNFα and IL-1β exerted distinct influences on the apoptotic pathway in human chondrocytes, with this variation being contingent upon the levels of PGE2 and caspase-8.
HumanBoileau et al. [50] Healthy knee
Knee OA
52 y
76 y
5172 hActivation of PAR-2 in osteoarthritic cartilage influenced disease pathways, making PAR-2 antagonists promising for OA treatment.
HumanTardif et al. [51]Healthy cartilage
OA
66 y
71 y
5124 hChordin was regulated differently in normal and osteoarthritic human chondrocytes.
HumanCaramés et al. [52]Healthy cartilageNA10124 hTNFα and IL-1β regulated apoptosis differently in human chondrocytes.
HumanYik et al. [53]Knee OA44–80 y1005 hCDK-9 activity was required for the primary inflammatory response in chondrocytes, CDK-9 inhibition provides protection of cartilage against catabolism effects of proinflammatory cytokines.
HumanDing et al. [54]Healthy ankle45 y100224 hDAMPs induced chondrolysis with cytokines or fibronectin fragment, but lost their effects when acting alone.
HumanTerkeltaub et al. [55]Healthy knee
OA knee
NA
NA
10118 hMaintaining AMPK activation protected the cartilage matrix from deterioration caused by inflammation.
RatChen et al. [56]Wild type4 w10372 hElectroacupuncture serum inhibits TNFα mediated chondrocyte inflammation via the Ras-Raf-MEK1/2-ERK1/2 signaling pathway.
MouseCao et al. [57]Wild type
Transgenic
4 w
4 w
1011 hTNFα stimulated mitochondrial superoxide flash activity by 2-fold in vitro and 5-fold in situ. Mitochondria are a significant source of cellular oxidants.
BovineDing et al. [54]Healthy stifle jointNA100124 hDAMPs induced chondrolysis with cytokines or fibronectin fragment, but lost their effects when acting alone.
BovineDjouad et al. [58]Carpometacarpal joint (healthy)6 m10172 hProinflammation cytokines had devastating effects on the cartilage constructs and those effects could be inhibited by the blockade of ERK signaling pathway.
GoatChen et al. [59]Healthy kneeNA40124 hProinflammatory cytokine could change the mechanical properties of chondrocyte in vitro.
Table 2. Three-dimensional models: Construct-based TNFα chondrocyte inflammatory models.
Table 2. Three-dimensional models: Construct-based TNFα chondrocyte inflammatory models.
SpeciesAuthorsOriginAge
(Mean)
ModelConcentration (ng/mL)PassageDurationMain Findings
BovineMohanraj
et al. [60]
Healthy knee2–6 mAgarose construct + TNFα1, 5, 1006 daysMSC-derived constructs were more easily influenced by the inflammatory environment compared to chondrocyte-derived constructs.
BovineOssendorff et al. [61] Healthy fetlock joint5–7 mPolyurethanePU scaffolds + TNFα20314 daysTNFα had negative effects on chondrogenesis under simulated ACI conditions. Dynamic load and adalimumab inhibited those effects.
BovineTilwani
et al. [62]
Healthy cartilage<18 mAgarose construct + TNFα0.1, 10, 100048 hTNF enhanced NO, PGE2, and MMP activity at 5% oxygen tension, and dynamic compression might counteract this impact.
HumanMorris et al. [63] Healthy knee and ankle65 yalginate beads + TNFα1, 10, 50, 100048 hTIMP3 may serve a chondroprotective role and it may be necessary to bind to ECM for its full function.
Table 3. Three-dimensional models: Explant-based TNFα chondrocyte inflammatory models.
Table 3. Three-dimensional models: Explant-based TNFα chondrocyte inflammatory models.
SpeciesAuthorsOriginAgeConcentration (ng/mL)DurationMain Findings
HumanBoileau et al. [50]Healthy knee
OA knee
52 y
76 y
52 daysPAR-2 activation had been linked to catabolic and inflammatory processes associated with OA development.
HumanKim et al. [46]OA knee69.5 y109 daysSulphoraphane (SFN) inhibited multiple catabolic mechanisms in cartilage.
HumanTardif et al. [51]Healthy cartilage
OA Cartilage
66
71
53 daysChordin was regulated differently in normal and osteoarthritic human chondrocytes.
MouseCao et al. [57]Hip and Knee
wild type/transgenic
4 w102 hTNFα stimulated mitochondrial superoxide flash activity by 2-fold in vitro and 5-fold in situ. Mitochondria are a significant source of cellular oxidants.
MouseTerkeltaub et al. [55] Healthy Femoral Head, wild-type2 m103 daysMaintaining AMPK activation protected the cartilage matrix from deterioration caused by inflammation.
HorseLittle et al. [64]Healthy fetlock joint2–12 y1004 daysRegional difference in response to catabolic cytokines was unlikely to be responsible for the initiation of focal cartilage degeneration in osteoarthritis.
Bovine/PorcineLittle
et al. [65]
Healthy fetlock joints2 w
3–6 m
1004–20 days (Bovine, Porcine)In vitro, aggrecan produced from normal and OA cartilage in response to TNFα is cleaved by aggrecanase rather than MMPs.
PorcineBevill et al. [66]Healthy knee6–8 m1006 hThe core and peripheral areas of tibial cartilage have distinct gene expression responses to mechanical strain and TNFα.
BovineStevens et al. [67]Healthy knee2–3 w1005 daysOverload compression damaged the cartilage matrix and disrupted the cell membrane. IL-1 and TNFα stimulate chondrocytes to release proteins linked with innate immune and stress responses, which may aid in host defense against infections and protect cells from stress-induced damage.
Bovine
Human
Sui et al. [68]Healthy knee
Healty ankle
1–2 w
26–61 y
25 (Bovine)
100 (Human)
6 days (Bovine)
8 days (Human)
Mechanical stimulation may enhance the catabolic effects of proinflammatory cytokine. In addition, IL-6/sIL-6R works in tandem with TNFα to cause cartilage deterioration.
Table 4. Other TNFα chondrocyte inflammatory models.
Table 4. Other TNFα chondrocyte inflammatory models.
SpeciesAuthorsModelOriginAgeConcentration
(ng/mL)
PassageDurationMain Findings
HumanShakibaei et al. [69]3D to 2D: chondrocytes migrated from alginate bead construct to monolayer culture + TNFαHip joint (femoral neck fracture)NA10224 hCurcumin reduced TNF-induced COX-2 and MMP-9 expression in chondrocytes.
HumanRoman-Blas et al. [70] 2D: Suspension culture + TNFαOA kneeNA1002 hIL-1β or TNFα suppressed Smad3/4 DNA-binding activity. IL-1β or TNFα suppression of TGF-β signaling pathways may no related to Smad7.
HumanMalemud et al. [47] 3D: Chondrocytes pellet
+ recombinant human TNFα
OA knee
adult knee
juvenile knee
NA
NA
12 y
10, 20, 50148 hTNFα could increase apoptosis in normal human chondrocytes, OA chondrocytes and human juvenile chondrocyte pellet cultures, but not in chondrocyte pellet cultures initiated from MSCs.
BovineDjouad et al. [58]3D: Agarose construct + cartilage ringHealthy fetlock joint6 m1, 10128 daysProinflammation cytokines had devastating effects on the cartilage constructs and those effects could be inhibited by the blockade of ERK signaling pathway.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, S.; Kurth, S.; Burger, C.; Wirtz, D.C.; Schildberg, F.A.; Ossendorff, R. TNFα-Related Chondrocyte Inflammation Models: A Systematic Review. Int. J. Mol. Sci. 2024, 25, 10805. https://doi.org/10.3390/ijms251910805

AMA Style

Wang S, Kurth S, Burger C, Wirtz DC, Schildberg FA, Ossendorff R. TNFα-Related Chondrocyte Inflammation Models: A Systematic Review. International Journal of Molecular Sciences. 2024; 25(19):10805. https://doi.org/10.3390/ijms251910805

Chicago/Turabian Style

Wang, Su, Sarah Kurth, Christof Burger, Dieter C. Wirtz, Frank A. Schildberg, and Robert Ossendorff. 2024. "TNFα-Related Chondrocyte Inflammation Models: A Systematic Review" International Journal of Molecular Sciences 25, no. 19: 10805. https://doi.org/10.3390/ijms251910805

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