Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3473 2023-10-31 09:04:22 |
2 update references and layout -6 word(s) 3467 2023-10-31 09:08:26 | |
3 update description + 4 word(s) 3471 2023-11-24 07:29:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Ejma-Multański, A.; Wajda, A.; Paradowska-Gorycka, A. Cell Cultures in the Autoimmune Connective Tissue Diseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/50961 (accessed on 18 May 2024).
Ejma-Multański A, Wajda A, Paradowska-Gorycka A. Cell Cultures in the Autoimmune Connective Tissue Diseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/50961. Accessed May 18, 2024.
Ejma-Multański, Adam, Anna Wajda, Agnieszka Paradowska-Gorycka. "Cell Cultures in the Autoimmune Connective Tissue Diseases" Encyclopedia, https://encyclopedia.pub/entry/50961 (accessed May 18, 2024).
Ejma-Multański, A., Wajda, A., & Paradowska-Gorycka, A. (2023, October 31). Cell Cultures in the Autoimmune Connective Tissue Diseases. In Encyclopedia. https://encyclopedia.pub/entry/50961
Ejma-Multański, Adam, et al. "Cell Cultures in the Autoimmune Connective Tissue Diseases." Encyclopedia. Web. 31 October, 2023.
Cell Cultures in the Autoimmune Connective Tissue Diseases
Edit

Cell cultures are an important part of the research and treatment of autoimmune connective tissue diseases. By culturing the various cell types involved in autoimmune connective tissue diseases (ACTDs), researchers are able to broaden the knowledge about these diseases that, in the near future, may lead to finding cures. Fibroblast cultures and chondrocyte cultures allow scientists to study the behavior, physiology and intracellular interactions of these cells.

ACTD cell cultures systemic sclerosis lupus erythematosus

1. Introduction

Autoimmune connective tissue diseases (ACTDs) are a group of complicated multisystem disorders in which patient’s immune system cells attack tissues such as joints, tendons, skeletal muscles, neural connections, cartilage, bones and other connective tissues [1]. Even though these diseases differ from each other in their symptoms as well as in affected cells, they all share the same pathological mechanism, which manifests as a prolonged state of inflammation in the affected area or areas, leading to the degeneration and degradation of afflicted structures [2]. The etiology of ACTDs remains mostly undiscovered with suspected involvement of genetic, hormonal and environmental factors [3]. Because of the unknown etiology and ACTDs’ multisystemic nature, proper diagnosis and treatment is a major challenge for healthcare systems. Therefore, cell cultures are one of the tools used to understand the molecular basis/mechanisms of/in ACTDs. Due to the lack of uniformed diagnosis criteria and the amount of ACTDs, it would be impossible to describe the cell culture types used in the research on all of them in one paper. So the authors have focused on few selected ACTDs, and the culture types used in research on them. These diseases are presented in Table 1.
Table 1. List of described ACTDs, tissues they affect, type of cell cultures that might be used in research on particular diseases, immune elements involved in pathological processes and their immunopathogenesis.
Cell culture development began in the early 1900s; however, their true potential was discovered and advanced in late 1940s when they were used to grow polioviruses in the production of the polio vaccine [13]. Conducting an experiment with cell cultures allows scientists to study the in vitro morphology, physiology and genetics of a single or few types of cells of interest. While cell cultures are a powerful tool in research, it is important to remember their limitations and requirements. First of all, working with tissue cultures requires strict conditions, such as CO2 levels, temperature, etc. Secondly, in order to grow cells must be placed in dedicated media which maintain their physiological state and functionality [14]. The media which are used in the cell cultures are listed in Table 2. Depending on the type of cell culture, its development might be a long and laborious process, which is why researchers often buy commercially prepared cell lines to hasten the experiment.
Table 2. List of medias used in the described cell cultures with their composition and application.
Connective tissue is a type of tissue that is present in every physiological system in the human body. It acts as a scaffolding for other tissues and organs, supports their functions, protects them against harmful factors and helps to repair damaged tissues [19]. During the course of ACTD-afflicted connective tissue with ongoing inflammation and, very often, fibrosis, is unable to perform its functions, thus starting a domino effect leading to deterioration of other tissues and organs.

2. Fibroblast Cultures

Fibroblast cell cultures are the most basic and best established of all known culture types; therefore, they can be considered a model culture. These cells are found in almost all of the tissues of the body, including skin, bones, muscles and organs. Fibroblasts play a crucial role in maintaining the structural integrity of tissues and organs. [20] Fibroblasts produce and secrete the extracellular matrix (ECM), a complex mixture of collagen, elastin and other proteins and carbohydrates, that forms the structural framework of tissues, providing them with necessary strength and flexibility. Fibroblasts also play a key role in early stages of wound healing and immune response by inducing receptor-linked chemokine synthesis [20]. As a fundamental type of connective tissue cell, fibroblasts are more or less affected in most of the ACTDs; however, in the course of SSc [11], DM [8] and PM [8] they play major roles. Briefly, in SSc, fibroblasts are targeted by immune cells which secrete profibrotic cytokines. These cytokines disrupt the balance of ECM production and degradation, causing fibroblasts to become dysfunctional and overproduce collagen and other proteins that form scar tissue. This excess collagen causes thickening and hardening of the skin, as well as damaging the blood vessels, muscles and internal organs [21]. In DM and PM, the muscle tissue is invaded by T and B lymphocytes, similarly to SSc. But, instead of secreting profibrotic cytokines, they produce cytokines and antibodies that destroy myocytes and damage the tissue leading to muscle weakness and atrophy. Even though fibroblasts are not directly targeted in myositis, they may be damaged by infiltrating immune cells, releasing MDA-5 antigen and mobilizing anti-MDA-5 antibodies, causing an increase in inflammation and a characteristic skin rash [22].
Establishing a primary cell culture of fibroblasts involves isolating these cells from tissue of interest and culturing them in a suitable growth medium. These tissues can be easily obtained through a biopsy or discarded surgical material. After acquisition, the collected material must be dissociated into smaller pieces, whether by using a sterile scalpel and/or by enzymatic digestion to release individual cells [20]. Isolated cells should be rinsed with PBS, resuspended in DMEM with 10% fetal bovine serum (FBS) and 1% antibiotic-antimycotic addition, and maintained in 5% CO2 environment at 37 °C [23][24][25].
One use of fibroblast cultures in the aforementioned diseases is to study the underlying mechanisms of the pathological processes involved in their development and progression [4][26][27]. Chadli et al. conducted an experiment in which they used skin biopsies collected from patients diagnosed with early SSc to generate fibroblast cell culture. Biopsies were minced into small fragments, treated with dispase II to separate dermis from epidermis and then the dermis was treated with mixture of dispase II and collagenase II to free fibroblasts, which were cultured in standard conditions in DMEM [26]. Subsequently, RNA was extracted from the cultured cells and micro-array was performed to measure the expression levels. The researchers found that fibroblasts derived from SSc skin biopsies retained most of the diseases’ molecular phenotype, therefore highlighting the value of phenotyped fibroblast cultures as a viable platform for SSc drug research [26]. Another use of the fibroblast cell cultures in ACTD is in the development and testing of new therapies, such as Wu and colleagues’ research in which they used skin biopsies from patients with SSc in order to limit SSc pathogenesis through fibroblast store-operated Ca2+ entry (SOCE) induced dedifferentiation [28].

3. Synovial Fibroblast Cultures

Synovial fibroblasts (SF) are a type of cell found only in the stroma of the joint synovium and are its main component. Based on their localization in the synovium and their expression patterns, the cells can be distinguished into two groups, intimal and subintimal [29]. The term “synovial fibroblasts” is a broad description that refers to both subpopulations; therefore, the intimal cells are also called type B synoviocytes or fibroblast-like-synoviocytes (FLS) [30]. Both groups of cells, despite distinct differences between them, will be called synovial fibroblasts, due to the fact that both subtypes are involved in the course of RA. Apart from being the basis of the stroma’s structure, synoviocytes’ main functions are producing components of the synovial fluid and immunosurveillance [31]. Production of synovial fluid is crucial for the integrity of the cartilaginous tissue present in the joint, as well as its lubrication and mobility [32]. SF play a key role in the development and progression of RA [33] and osteoarthritis (OA) [34], but, similarly to the chondrocytes which are found in the vicinity of SF, they are also indirectly affected in the course of other ACTDs that affect the joints with their symptoms, such as JIA, spondyloarthropathies and MCTD [5][10][35]. At this point, it is important to note that OA is not considered an ACTD as it does not have an autoimmune or inflammatory background. However, OA is often investigated in ACTD research, whether as a control group in inflammatory joint damage research, in studies on joint tissue renewal therapies or as a starting point for research into the development of joint models essential for ACTDs with joint pathogenesis. In RA, due to chronic inflammation, synoviocytes undergo a series of morphological and physiological changes that result in acquiring a form of a constantly activated tumor-like RA-FLS. These cells, resistant to receptor mediated apoptosis and capable of invading other cells, are causing further inflammation and degradation of joints by the production of inflammatory factors, stimulation and the accumulation of immune cells [33]. In OA, SF, mostly FLS, begin to intensively proliferate and differentiate into myofibroblast-like cells which synthesize immense amounts of rich in fiber ECM. This process leads to accumulation of ECM and joints fibrosis causing stiffness and chronic pain [36].
The main sources of SF are aspirates of synovial fluid from joints [37], fragments of tissue collected during joint surgeries [38] and tissue harvested during arthroscopic synovectomy [39]. SF culture from synovial fluid is conducted under the standard conditions using DMEM [37]. Deriving a cell culture from tissue fragments, either harvested during surgeries or synovectomy, first requires a manual fragmentation and digestion with collagenase in DMEM for 2 h at 37 °C. When freed from the fibrous scaffold, tissues can be harvested by centrifuging [37] or filtering through a mesh [38] and cultured in DMEM with 10% FBS and 1% antibiotic–antimycotic addition [37][38][39]. Synovial fibroblasts play an important role in the pathogenesis of various ACTDs, such as RA; therefore, their cultures are a valuable tool for studying the pathogenesis of these diseases. Wei et al. performed an experiment in which they used SF collected from RA patients and revealed that the upregulation of the neurogenic locus notch homolog protein 3 (NOTCH3) signaling pathway in SF plays a significant role in inflammation and tissue degradation processes during the course of RA [40]. Moreover, SF cultures can aid in identifying novel therapy targets and testing new potential therapeutics. Three different research groups conducted experiments in which they used SF cultures derived from cells collected from RA patients has shown significant upregulation of fucosyltransferase 1 [41] and Yes-associated protein (YAP) [42] expression, as well as a shift in glucose metabolism towards glycolysis [43] suggesting them as novel targets in RA treatment strategies.

4. Chondrocyte Cultures

Chondrocytes are the only type of cell present in human cartilaginous tissue. They can be considered as a highly differentiated form of fibroblasts therefore their main function is synthesis of an ECM rich in proteoglycan and type II collagen [44]. Moreover, chondrocytes maintain homeostasis in cartilage by alternating between anabolic and catabolic processes of creation and degradation of ECM proteins [44]. The matrix’s functions vary based on its location within tissue or organ and the number of structural proteins it consists of. Nevertheless, its fundamental roles are supporting the growth of cells and tissues, modulation of intercellular interaction and separation of tissues. Additionally, ECM is able to alter cell behavior and growth by secretion of various factors which crucial in cellular growth, fibrosis and thereby wound healing. It has been proven that ECM plays an important role in cell differentiation, migration and even in gene expression [45][46][47].
Chondrocytes are primarily affected in OA, which causes both cells and ECM to degrade, leading to the breakdown of joint cartilage and bone structure underneath [48]. Along with the progression of the disease occurs chronic inflammation and tightening of space in the afflicted joint, due to the lack of cartilaginous tissue and growth of pathological bone structure, causing stiffness, swelling and pain [49]. Chondrocytes are also indirectly affected in the course of other ACTD such as RA, JIA, MCTD and systemic lupus erythematosus (SLE) due to the persistent, prolonged inflammation that causes joint stiffness, swelling and pain, which may lead to ECM disbalance and degradation of chondrocytes [50][51][52][53].
Chondrocyte cell cultures are widely used in OA research, both in its etiology and treatment. In the case of ACTD studies, OA is used as a control model due to the lack of an autoimmune/inflammatory background [54]. They can be also used in treatment research by testing cellular response to drugs [55][56][57] inhibiting the diseases progress using genetic engineering [58][59][60] or by coculturing chondrocytes with other cells to enhance therapy effectiveness in articular chondrocyte implantation [61].
The main source of human cartilage cells is total knee arthroplasties (TKAs Partial knee arthroplasty is an equally valid source of cells acquisition [62]. Chondrocytes can be isolated from the afore-mentioned harvested tissue and be used in setting up a primary chondrocyte cell culture in standard conditions using DMEM medium. In order to do so, cartilage must be minced or cut into very small pieces and treated with type II 0.02% collagenase at 37 °C for 4 h [63][64]. The concentration of collagenase may be lower, but the digestion time should be adequately prolonged [44][65]. However, while being cultured in 2D in vitro conditions, chondrocytes undergo a process of dedifferentiation altering their appearance into one resembling a fibroblast. Recently it has been claimed that this change also affects the expression levels of various genes and proteins, making chondrocyte cultures a less reliable tool for research [66]. Chondrocyte cultures can be used to study the etiology of RA, as described in an experiment by Andreas et al., in which they created a model consisting of postmortem-acquired human chondrocytes, supernatants from cultures of RA SF and alginate. By stimulating chondrocytes with RA SF culture supernatants, they mimicked the RA joint environment, which allowed them to identify the key regulatory molecules driving cartilage destruction [67]. Chondrocyte cultures also allow for the study of changes in cell physiology during the course of the disease. By constructing coculture models consisting of either three [68] or two [69] types of cells deriving from patients or animals, respectively, researchers were able to recreate the joint environment, which allowed the first group to establish a complete model for cartilage destruction [68] and the second group to create a perfusion culture system [69]. Chondrocyte cell cultures can also be used to observe the effects of illness on gene expression levels and gene involvement in RA pathogenesis, as described by Barksby et al. In their experiment, the researchers cultured two types of cells, chondrocytes acquired from patients and commercially acquired human chondrosarcoma cells; both were cultured in standard conditions with use of serum-free DMEM. These cells were stimulated with a combination of interleukin 1 (IL-1) and oncostatin M (OSM) to mimic cytokine environment of RA synovial fluid. Subsequently, researchers isolated RNA from the cells and performed micro-array analysis, as well as a real-time PCR, which showed an overexpression of multiple genes, such as matrix metalloproteinase 1 (MMP1), a disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) and pentraxin-related protein 3 (PTX3) [70].

References

  1. Jeganathan, N.; Sathananthan, M. Connective Tissue Disease-Related Interstitial Lung Disease: Prevalence, Patterns, Predictors, Prognosis, and Treatment. Lung 2020, 198, 735–759.
  2. Huang, S.-W.; Lin, C.-L.; Lin, L.-F.; Huang, C.-C.; Liou, T.-H.; Lin, H.-W. Autoimmune Connective Tissue Diseases and the Risk of Rotator Cuff Repair Surgery: A Population-Based Retrospective Cohort Study. BMJ Open 2019, 9, e023848.
  3. Lai, B.; Wu, C.-H.; Wu, C.-Y.; Luo, S.-F.; Lai, J.-H. Ferroptosis and Autoimmune Diseases. Front. Immunol. 2022, 13, 916664.
  4. Kamperman, R.G.; van der Kooi, A.J.; de Visser, M.; Aronica, E.; Raaphorst, J. Pathophysiological Mechanisms and Treatment of Dermatomyositis and Immune Mediated Necrotizing Myopathies: A Focused Review. Int. J. Mol. Sci. 2022, 23, 4301.
  5. Agarwal, S.; Misra, R.; Aggarwal, A. Interleukin 17 levels are increased in juvenile idiopathic arthritis synovial fluid and induce synovial fibroblasts to produce proinflammatory cytokines and matrix metalloproteinases. J. Rheumatol. 2008, 35, 515–519.
  6. Blake, S.C.; Daniel, B.S. Cutaneous lupus erythematosus: A review of the literature. Int. J. Women’s Dermatol. 2019, 5, 320–329.
  7. Felis-Giemza, A.; Ornowska, S.; Haładyj, E.; Czuszyńska, Z.; Olesińska, M. Relationship between type of skin lesions and nailfold capillaroscopy pattern in mixed connective tissue disease. Clin. Rheumatol. 2022, 41, 281–288.
  8. Yang, S.-H.; Chang, C.; Lian, Z.-X. Polymyositis and dermatomyositis—Challenges in diagnosis and management. J. Transl. Autoimmun. 2019, 2, 100018.
  9. Chu, C.-Q. Highlights of Strategies Targeting Fibroblasts for Novel Therapies for Rheumatoid Arthritis. Front. Med. 2022, 9, 846300.
  10. Fearon, U.; Veale, D.J. Pathways of cell activation in spondyloarthropathies. Curr. Rheumatol. Rep. 2001, 3, 435–442.
  11. Asano, Y. Systemic sclerosis. J. Dermatol. 2018, 45, 128–138.
  12. Hoang, M.P.; Park, J. Vasculitis. In Hospital-Based Dermatopathology; Springer International Publishing: Berlin/Heidelberg, Germany, 2020; pp. 245–296.
  13. Taylor, M.W. A History of Cell Culture. In Viruses and Man: A History of Interactions; Springer International Publishing: Berlin/Heidelberg, Germany, 2014; pp. 41–52.
  14. Verma, P. (Ed.) Industrial Microbiology and Biotechnology; Springer: Singapore, 2022.
  15. Arora, M. Cell Culture Media: A Review. Mater. Methods 2013, 2020, 3.
  16. Kannan, S.; Ghosh, J.; Dhara, S.K. Osteogenic differentiation potential of porcine bone marrow mesenchymal stem cell subpopulations selected in different basal media. Biol. Open 2020, 9, bio053280.
  17. Gorfien, S.; Spector, A.; DeLuca, D.; Weiss, S. Growth and Physiological Functions of Vascular Endothelial Cells in a New Serum-Free Medium (SFM). Exp. Cell Res. 1993, 206, 291–301.
  18. Sukho, P.; Kirpensteijn, J.; Hesselink, J.W.; van Osch, G.J.V.M.; Verseijden, F.; Bastiaansen-Jenniskens, Y.M. Effect of Cell Seeding Density and Inflammatory Cytokines on Adipose Tissue-Derived Stem Cells: An in Vitro Study. Stem Cell Rev. Rep. 2017, 13, 267–277.
  19. Culav, E.M.; Clark, C.H.; Merrilees, M.J. Connective Tissues: Matrix Composition and Its Relevance to Physical Therapy. Phys. Ther. 1999, 79, 308–319.
  20. Plikus, M.V.; Wang, X.; Sinha, S.; Forte, E.; Thompson, S.M.; Herzog, E.L.; Driskell, R.R.; Rosenthal, N.; Biernaskie, J.; Horsley, V. Fibroblasts: Origins, definitions, and functions in health and disease. Cell 2021, 184, 3852–3872.
  21. Pattanaik, D.; Brown, M.; Postlethwaite, B.C.; Postlethwaite, A.E. Pathogenesis of Systemic Sclerosis. Front. Immunol. 2015, 6, 272.
  22. Shen, N.; Zhou, X.; Jin, X.; Lu, C.; Hu, X.; Zhang, Y.; Jiang, Y.; Xu, Q.; Xu, X.; Liu, M.; et al. MDA5 expression is associated with TGF-β-induced fibrosis: Potential mechanism of interstitial lung disease in anti-MDA5 dermatomyositis. Rheumatology 2022, 62, 373–383.
  23. Benchaprathanphorn, K.; Sakulaue, P.; Siriwatwechakul, W.; Muangman, P.; Chinaroonchai, K.; Namviriyachote, N.; Viravaidya-Pasuwat, K. Expansion of fibroblast cell sheets using a modified MEEK micrografting technique for wound healing applications. Sci. Rep. 2022, 12, 18541.
  24. Mahmoudi, S.; Mancini, E.; Xu, L.; Moore, A.; Jahanbani, F.; Hebestreit, K.; Srinivasan, R.; Li, X.; Devarajan, K.; Prélot, L.; et al. Heterogeneity in old fibroblasts is linked to variability in reprogramming and wound healing. Nature 2019, 574, 553–558.
  25. Plaisancié, P.; Buisson, C.; Fouché, E.; Martin, P.; Noirot, C.; Maslo, C.; Dupuy, J.; Guéraud, F.; Pierre, F. Study of the colonic epithelial-mesenchymal dialogue through establishment of two activated or not mesenchymal cell lines: Activated and resting ones differentially modulate colonocytes in co-culture. PLoS ONE 2022, 17, e0273858.
  26. Chadli, L.; Sotthewes, B.; Li, K.; Andersen, S.N.; Cahir-McFarland, E.; Cheung, M.; Cullen, P.; Dorjée, A.; de Vries-Bouwstra, J.K.; Huizinga, T.W.J.; et al. Identification of regulators of the myofibroblast phenotype of primary dermal fibroblasts from early diffuse systemic sclerosis patients. Sci. Rep. 2019, 9, 4521.
  27. Zou, Y.-Q.; Jin, W.-D.; Li, Y.-S. Roles of macrophage migration inhibitory factor in polymyositis: Inflammation and regeneration. J. Int. Med. Res. 2018, 46, 732–738.
  28. Wu, C.-Y.; Hsu, W.-L.; Tsai, M.-H.; Chai, C.-Y.; Yen, C.-J.; Chen, C.-H.; Lu, J.-H.; Yu, H.-S.; Yoshioka, T. A potential new approach for treating systemic sclerosis: Dedifferentiation of SSc fibroblasts and change in the microenvironment by blocking store-operated Ca2+ entry. PLoS ONE 2019, 14, e0213400.
  29. Croft, A.P.; Campos, J.; Jansen, K.; Turner, J.D.; Marshall, J.; Attar, M.; Savary, L.; Wehmeyer, C.; Naylor, A.J.; Kemble, S.; et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature 2019, 570, 246–251.
  30. Li, F.; Tang, Y.; Song, B.; Yu, M.; Li, Q.; Zhang, C.; Hou, J.; Yang, R. Nomenclature clarification: Synovial fibroblasts and synovial mesenchymal stem cells. Stem Cell Res. Ther. 2019, 10, 260.
  31. Ospelt, C. Synovial fibroblasts in 2017. RMD Open 2017, 3, e000471.
  32. Németh, T.; Nagy, G.; Pap, T. Synovial fibroblasts as potential drug targets in rheumatoid arthritis, where do we stand and where shall we go? Ann. Rheum. Dis. 2022, 81, 1055–1064.
  33. Pap, T.; Dankbar, B.; Wehmeyer, C.; Korb-Pap, A.; Sherwood, J. Synovial fibroblasts and articular tissue remodelling: Role and mechanisms. Semin. Cell Dev. Biol. 2020, 101, 140–145.
  34. Hu, T.; Zhang, Z.; Deng, C.; Ma, X.; Liu, X. Effects of β2 Integrins on Osteoclasts, Macrophages, Chondrocytes, and Synovial Fibroblasts in Osteoarthritis. Biomolecules 2022, 12, 1653.
  35. Ding, Q.; Hu, W.; Wang, R.; Yang, Q.; Zhu, M.; Li, M.; Cai, J.; Rose, P.; Mao, J.; Zhu, Y.Z. Signaling pathways in rheumatoid arthritis: Implications for targeted therapy. Signal Transduct. Target. Ther. 2023, 8, 68.
  36. Maglaviceanu, A.; Wu, B.; Kapoor, M. Fibroblast-like synoviocytes: Role in synovial fibrosis associated with osteoarthritis. Wound Repair Regen. 2021, 29, 642–649.
  37. Zafari, P.; Rafiei, A.; Faramarzi, F.; Ghaffari, S.; Amiri, A.H.; Taghadosi, M. Human fibroblast-like synoviocyte isolation matter: A comparison between cell isolation from synovial tissue and synovial fluid from patients with rheumatoid arthritis. Rev. Assoc. Médica Bras. 2021, 67, 1654–1658.
  38. Zheng, L.; Hu, F.; Bian, W.; Li, Y.; Zhang, L.; Shi, L.; Ma, X.; Liu, Y.; Zhang, X.; Li, Z. Dickkopf-1 perpetuated synovial fibroblast activation and synovial angiogenesis in rheumatoid arthritis. Clin. Rheumatol. 2021, 40, 4279–4288.
  39. Kaul, N.-C.; Mohapatra, S.R.; Adam, I.; Tucher, C.; Tretter, T.; Opitz, C.A.; Lorenz, H.-M.; Tykocinski, L.-O. Hypoxia decreases the T helper cell-suppressive capacity of synovial fibroblasts by downregulating IDO1-mediated tryptophan metabolism. Rheumatology 2020, 59, 1148–1158.
  40. Wei, K.; Korsunsky, I.; Marshall, J.L.; Gao, A.; Watts GF, M.; Major, T.; Croft, A.P.; Watts, J.; Blazar, P.E.; Lange, J.K.; et al. Notch signalling drives synovial fibroblast identity and arthritis pathology. Nature 2020, 582, 259–264.
  41. Isozaki, T.; Ruth, J.H.; Amin, M.A.; Campbell, P.L.; Tsou, P.-S.; Ha, C.M.; Haines, G.K.; Edhayan, G.; Koch, A.E. Fucosyltransferase 1 mediates angiogenesis, cell adhesion and rheumatoid arthritis synovial tissue fibroblast proliferation. Arthritis Res. Ther. 2014, 16, R28.
  42. Symons, R.A.; Colella, F.; Collins, F.L.; Rafipay, A.J.; Kania, K.; McClure, J.J.; White, N.; Cunningham, I.; Ashraf, S.; Hay, E.; et al. Targeting the IL-6–Yap–Snail signalling axis in synovial fibroblasts ameliorates inflammatory arthritis. Ann. Rheum. Dis. 2022, 81, 214–224.
  43. Garcia-Carbonell, R.; Divakaruni, A.S.; Lodi, A.; Vicente-Suarez, I.; Saha, A.; Cheroutre, H.; Boss, G.R.; Tiziani, S.; Murphy, A.N.; Guma, M. Critical Role of Glucose Metabolism in Rheumatoid Arthritis Fibroblast-like Synoviocytes. Arthritis Rheumatol. 2016, 68, 1614–1626.
  44. Park, S.; Baek, I.-J.; Ryu, J.H.; Chun, C.-H.; Jin, E.-J. PPARα−ACOT12 axis is responsible for maintaining cartilage homeostasis through modulating de novo lipogenesis. Nat. Commun. 2022, 13, 3.
  45. Lo, C.-M.; Wang, H.-B.; Dembo, M.; Wang, Y. Cell Movement Is Guided by the Rigidity of the Substrate. Biophys. J. 2000, 79, 144–152.
  46. Engler, A.J.; Sen, S.; Sweeney, H.L.; Discher, D.E. Matrix Elasticity Directs Stem Cell Lineage Specification. Cell 2006, 126, 677–689.
  47. Provenzano, P.P.; Inman, D.R.; Eliceiri, K.W.; Keely, P.J. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK–ERK linkage. Oncogene 2009, 28, 4326–4343.
  48. Krishnan, Y.; Grodzinsky, A.J. Cartilage diseases. Matrix Biol. 2018, 71–72, 51–69.
  49. Junker, S.; Frommer, K.W.; Krumbholz, G.; Tsiklauri, L.; Gerstberger, R.; Rehart, S.; Steinmeyer, J.; Rickert, M.; Wenisch, S.; Schett, G.; et al. Expression of adipokines in osteoarthritis osteophytes and their effect on osteoblasts. Matrix Biol. 2017, 62, 75–91.
  50. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O.; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 Rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. Dis. 2010, 69, 1580–1588.
  51. Colebatch-Bourn, A.N.; Edwards, C.J.; Collado, P.; D’Agostino, M.-A.; Hemke, R.; Jousse-Joulin, S.; Maas, M.; Martini, A.; Naredo, E.; Østergaard, M.; et al. EULAR-PReS points to consider for the use of imaging in the diagnosis and management of juvenile idiopathic arthritis in clinical practice. Ann. Rheum. Dis. 2015, 74, 1946–1957.
  52. John, K.J.; Sadiq, M.; George, T.; Gunasekaran, K.; Francis, N.; Rajadurai, E.; Sudarsanam, T.D. Clinical and Immunological Profile of Mixed Connective Tissue Disease and a Comparison of Four Diagnostic Criteria. Int. J. Rheumatol. 2020, 2020, 9692030.
  53. Aringer, M.; Costenbader, K.; Daikh, D.; Brinks, R.; Mosca, M.; Ramsey-Goldman, R.; Smolen, J.S.; Wofsy, D.; Boumpas, D.T.; Kamen, D.L.; et al. 2019 European League Against Rheumatism/American College of Rheumatology classification criteria for systemic lupus erythematosus. Ann. Rheum. Dis. 2019, 78, 1151–1159.
  54. Neidel, J.; Blum, W.F.; Schaeffer, H.-J.; Schulze, M.; Schönau, E.; Lindschau, J.; Föll, J. Elevated levels of insulin-like growth factor (IGF) binding protein-3 in rheumatoid arthritis synovial fluid inhibit stimulation by IGF-I of articular chondrocyte proteoglycan synthesis. Rheumatol. Int. 1997, 17, 29–37.
  55. Buhrmann, C.; Brockmueller, A.; Mueller, A.-L.; Shayan, P.; Shakibaei, M. Curcumin Attenuates Environment-Derived Osteoarthritis by Sox9/NF-kB Signaling Axis. Int. J. Mol. Sci. 2021, 22, 7645.
  56. Wang, B.; Jiang, Y.; Yao, Z.; Chen, P.; Yu, B.; Wang, S. Aucubin Protects Chondrocytes Against IL-1β-Induced Apoptosis In Vitro And Inhibits Osteoarthritis In Mice Model. Drug Des. Dev. Ther. 2019, 13, 3529–3538.
  57. Sun, K.; Luo, J.; Jing, X.; Xiang, W.; Guo, J.; Yao, X.; Liang, S.; Guo, F.; Xu, T. Hyperoside ameliorates the progression of osteoarthritis: An in vitro and in vivo study. Phytomedicine 2021, 80, 153387.
  58. Liang, C.; Wu, S.; Xia, G.; Huang, J.; Wen, Z.; Zhang, W.; Cao, X. Engineered M2a macrophages for the treatment of osteoarthritis. Front. Immunol. 2022, 13, 1054938.
  59. Liang, Y.; Xu, X.; Li, X.; Xiong, J.; Li, B.; Duan, L.; Wang, D.; Xia, J. Chondrocyte-Targeted MicroRNA Delivery by Engineered Exosomes toward a Cell-Free Osteoarthritis Therapy. ACS Appl. Mater. Interfaces 2020, 12, 36938–36947.
  60. Wu, H.; Peng, Z.; Xu, Y.; Sheng, Z.; Liu, Y.; Liao, Y.; Wang, Y.; Wen, Y.; Yi, J.; Xie, C.; et al. Engineered adipose-derived stem cells with IGF-1-modified mRNA ameliorates osteoarthritis development. Stem Cell Res. Ther. 2022, 13, 19.
  61. Esmaeili, A.; Hosseini, S.; Kamali, A.; Hosseinzadeh, M.; Shekari, F.; Baghaban Eslaminejad, M. Co-aggregation of MSC/chondrocyte in a dynamic 3D culture elevates the therapeutic effect of secreted extracellular vesicles on osteoarthritis in a rat model. Sci. Rep. 2022, 12, 19827.
  62. Hsu, H.; Siwiec, R.M. Knee Arthroplasty; StatPearls: St. Petersburg, FL, USA, 2023.
  63. Guo, Q.; Chen, X.; Chen, J.; Zheng, G.; Xie, C.; Wu, H.; Miao, Z.; Lin, Y.; Wang, X.; Gao, W.; et al. STING promotes senescence, apoptosis, and extracellular matrix degradation in osteoarthritis via the NF-κB signaling pathway. Cell Death Dis. 2021, 12, 13.
  64. Shi, Y.; Hu, X.; Cheng, J.; Zhang, X.; Zhao, F.; Shi, W.; Ren, B.; Yu, H.; Yang, P.; Li, Z.; et al. A small molecule promotes cartilage extracellular matrix generation and inhibits osteoarthritis development. Nat. Commun. 2019, 10, 1914.
  65. Yang, Y.; Lin, H.; Shen, H.; Wang, B.; Lei, G.; Tuan, R.S. Mesenchymal stem cell-derived extracellular matrix enhances chondrogenic phenotype of and cartilage formation by encapsulated chondrocytes in vitro and in vivo. Acta Biomater. 2018, 69, 71–82.
  66. Ghosh, S.; Scott, A.K.; Seelbinder, B.; Barthold, J.E.; Martin, B.M.S.; Kaonis, S.; Schneider, S.E.; Henderson, J.T.; Neu, C.P. Dedifferentiation alters chondrocyte nuclear mechanics during in vitro culture and expansion. Biophys. J. 2022, 121, 131–141.
  67. Andreas, K.; Lübke, C.; Häupl, T.; Dehne, T.; Morawietz, L.; Ringe, J.; Kaps, C.; Sittinger, M. Key regulatory molecules of cartilage destruction in rheumatoid arthritis: An in vitro study. Arthritis Res. Ther. 2008, 10, R9.
  68. Peck, Y.; Leom, L.T.; Low PF, P.; Wang, D.-A. Establishment of an in vitro three-dimensional model for cartilage damage in rheumatoid arthritis. J. Tissue Eng. Regen. Med. 2018, 12, e237–e249.
  69. Steinhagen, J.; Bruns, J.; Niggemeyer, O.; Fuerst, M.; Rüther, W.; Schünke, M.; Kurz, B. Perfusion culture system: Synovial fibroblasts modulate articular chondrocyte matrix synthesis in vitro. Tissue Cell 2010, 42, 151–157.
  70. Barksby, H.E.; Hui, W.; Wappler, I.; Peters, H.H.; Milner, J.M.; Richards, C.D.; Cawston, T.E.; Rowan, A.D. Interleukin-1 in combination with oncostatin M up-regulates multiple genes in chondrocytes: Implications for cartilage destruction and repair. Arthritis Rheum. 2006, 54, 540–550.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 116
Revisions: 3 times (View History)
Update Date: 24 Nov 2023
1000/1000