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 -- 3227 2023-10-27 19:19:47 |
2 Reference format revised. Meta information modification 3227 2023-10-30 01:52:33 |

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.
Oberemok, V.V.; Andreeva, O.; Laikova, K.; Alieva, E.; Temirova, Z. Treatment of  Rheumatoid Arthritis. Encyclopedia. Available online: https://encyclopedia.pub/entry/50887 (accessed on 30 June 2024).
Oberemok VV, Andreeva O, Laikova K, Alieva E, Temirova Z. Treatment of  Rheumatoid Arthritis. Encyclopedia. Available at: https://encyclopedia.pub/entry/50887. Accessed June 30, 2024.
Oberemok, Volodymyr V., Oksana Andreeva, Kateryna Laikova, Edie Alieva, Zenure Temirova. "Treatment of  Rheumatoid Arthritis" Encyclopedia, https://encyclopedia.pub/entry/50887 (accessed June 30, 2024).
Oberemok, V.V., Andreeva, O., Laikova, K., Alieva, E., & Temirova, Z. (2023, October 27). Treatment of  Rheumatoid Arthritis. In Encyclopedia. https://encyclopedia.pub/entry/50887
Oberemok, Volodymyr V., et al. "Treatment of  Rheumatoid Arthritis." Encyclopedia. Web. 27 October, 2023.
Treatment of  Rheumatoid Arthritis
Edit

Rheumatoid arthritis refers to joint diseases of unclear etiology whose final stages can lead to unbearable pain and complete immobility of the affected joints. Modern treatment with anti-inflammatory drugs, including janus kinase (JAK) inhibitors, monoclonal antibodies, and botanicals (polyphenols, glycosides, alkaloids, etc.) has achieved some success and hope for improving the course of the disease. 

:rheumatoid arthritis joints inflammation

1. Introduction

Rheumatoid arthritis (RA) is a difficult to treat disease. This disease is characterized by synovial inflammation (synovitis), which affects bone turnover and the ability of bone to adapt to bone tissue when replacing the cartilaginous matrix with mineralized bone [1]; it also leads to degeneration of bone tissue [2]. Patients with this disease experience pain in their joints, accompanied by stiffness, which limits their mobility. In addition, over time, this disease can destroy both cartilage and bone; limited mobility becomes active disability and may lead to deformed joints. Along with deformity, extra-articular manifestations such as vasculitis and scleritis can also be observed [3][4].

Among common risk factors, cigarette smoking has the strongest association with RA [5][6][7][8]. Tobacco smoking, does not cause rheumatoid arthritis but leads to a worsening of the disease by means of citrullination of tissue proteins [9][10]. Interestingly, Jiang et al. reported that the impact of cigarette uses on the development of RA increased only when smoke was inhaled from cigarettes, but not when tobacco products containing nicotine were chewed, which suggests that nicotine is not significantly involved in the pathogenesis of RA [8][9].
As mentioned above, RA is the most common autoimmune inflammatory arthritis, with incidence of 0.5–1.0% in the northern hemisphere population [11][12]; annually, it occurs in 24–45 people per 100,000 [13]. The disability that attends rheumatoid arthritis is serious and debilitating. An extensive review showed that within 2–3 years of onset, approximately one-third of sick people with rheumatoid arthritis stopped working because of the disease; by 10–15 years after onset, almost two-thirds (50–60%) may be unable to work [14][15]. In addition to deterioration of patients’ health, this leads to adverse effects in the global economy [16][17].
There is no single cause of rheumatoid arthritis, and its prognosis remains uncertain [18]. But microbiological infections, genetic and environmental factors play a special role in the development of RA. Meta-estimates of the regional prevalence of rheumatoid arthritis in low- or middle-income countries were 0.4% (Southeast Asia), 0.37% (Eastern Mediterranean), 0.62% (Europe), 1.25% (North, Central, and South America), and 0.42% (Western Pacific). A formal meta-analysis cannot be performed for sub-Saharan Africa due to limited data. No significant difference in the prevalence of RA was found between urban and rural areas. While the prevalence of the disease in low- and middle-income men was 0.16%, it climbed to 0.75% in women, a statistically significant difference [19]. Hormones and genetic (X-linked) factors may explain the more frequent occurrence of rheumatoid arthritis among women [20]. For example, estrogens, which control the body’s immune response, can lead to autoimmune diseases [21]. To explain gender bias, scientists have shown that women with RA exhibit non-random X chromosome inactivation (XCI), which can cause autoimmunity. It is believed that this error correlates with the presence of a shared epitope and the duration of the disease. Premature immuno aging, characterized by shorter telomere length, is also associated with the presence of SE [22].

2. A Mystery That Still Needs to Be Deciphered

Although assimilation of knowledge in the study of pathogenesis of RA is still incomplete, this disease is widely accepted as an immune-mediated disorder. That immune cells play an important role in the pathogenesis of RA has been shown by studies in which treatment with anticytokine agents, for example a neutralizing Abs against TNF-α, soluble TNF receptor fusion proteins, or rIL-1β receptor antagonists resulted in the successful suppression of joint inflammation [23]. Agents that target cytokine-driven immune processes are among the most important clinical treatments used to manage disease in RA patients [24]. Disease-modifying antirheumatic drugs (DMARDs), which are divided into synthetic (sDMARDs) and biological (bDMARDs), are widely used [25].

There are three pronounced determinants that can describe the clinical picture of RA: the inflammatory process (swelling, pain, stiffness during movement); the proliferative-destructive process (destruction of joints); and the enzymatic collagenolytic process (primary necrotism) [26]. The intensity of pain is directly related to the activity of the disease as a whole [27]. The pathogenesis of RA has different etiologies of origin. Since the early 1980s, it has been assumed that the spread has a genetic link [28], including exposure to various chemicals on the respiratory tract [29]. There are a variety of ways to counteract RA, and one of the first measures is dietary nutrition, since there are already a number of scientific justifications that show an advantage in combating the disease and lead to a reduction in symptoms [30].
The pathogenesis of the studied disease involves a complex interconnection between B cells, CD4+ and CD8+ T cells, and dendritic cells [31]. Because B cells undergo isotype switching, they are more capable of supporting the inflammatory cascade. In addition, the rheumatoid factor (RF), a group of autoantibodies with the ability to respond to Fc of human IgG, behaves like heterophilic antibodies and cross-reacts with other types of antibodies [32][33]. The participation of the RF in the formation of the immune complex can lead to further fixation of the complement and the involvement of cells that cause inflammation such as macrophages, neutrophils, and lymphocytes. This leads to tissue damage and provides positive feedback for the production of even more autoantibodies. At the moment, a long list of causes for rheumatoid arthritis has been identified, including epigenetic, genetic, hormonal, reproductive, neuroendocrine and comorbid host factors [34].
It appears that macrophage-derived cytokines, for example, tumor necrosis factor alpha (TNF-α) and IL-1β, are critical to the mediation of inflammatory synovitis; however, it has also been suggested that synovial T cells participate both in triggering the disease and in contributing to the development of the disease in RA. Significant infiltration of the synovial tissue by T lymphocytes has been observed frequently in RA [35]. However, little is known about the direct action of T cells in the development of pathogenesis. While the synovial T cells observed in RA are mainly classified by type of memory type: CD4+ CD45 RO [36] with a pro-inflammatory Th1 phenotype. Compared to TNF-α and IL-1β, the classical Th1 T-cell-derived cytokine IFN-γ is also rarely seen in rheumatoid joints [37].
Scientists believe that T cells-produced cytokine IL-17 participates in the RA development. IL-17 is frequently produced by T cells clones taken from patients with RA, and it was shown that IL-17 is found in abundance in the synovial fluids of RA patients [38]. Therefore, it comes as no surprise that T cells specifically predisposed to antigen have been shown to be efficacious in the medication of rheumatoid arthritis. Generally, these data suggest that T cells are quite significant albeit their still incompletely understood role in the pathogenesis of RA.

3. Achieving Movement without Pain

In addition to adalimumab, drugs such as methotrexate [39], etanercept [40][41], prednisone [42], and leflunomide [43] quite often used in the treatment of RA. These drugs have been in use longer, and for a variety of conditions. Their side effects are also serious and must be considered.
According to EULAR (European alliance of associations for rheumatology) recommendations the biologic and targeting synthetic drugs should be used only after treatment with methotrexate [44]. Methotrexate is the first-choice treatment among most patients [45]. Treatment with methotrexate reduces inflammation but also degrades cartilage. It is unknown whether these clinical responses to methotrexate are evidence of a specific mechanism of action or merely a common final pathway. While it is possible that changes in the production of PGE2 and levels of TIMP 1 may represent the downstream effects of methotrexate on the formation of IL-1 and IL-6, respectively, no conclusive evidence exists to support this. Some of these changes are also observed following treatment with nonsteroidal anti-inflammatory drugs, and the methotrexate-specific pathways have yet to be elucidated [46]. In addition, the number of side effects—some severe—associated with methotrexate are many: nausea, headaches, fatigue, mucositis and hair loss, cytopenia, interstitial lung disease (ILD pneumonitis), and drug-related liver diseases (fibrosis and cirrhosis of the liver) [47]. Unfortunately, the exact mechanisms of methotrexate toxicity remain unclear [48].
Prednisone is another drug used to treat RA. It is a corticosteroid with anti-inflammatory properties and immunosuppressive activity [49][50]. The mechanism of action is the binding of prednisone to the glucocorticoid receptor; it promotes conformational changes in the DNA-binding domain, which leads to a displacement of the receptor into the nucleus. In the latter, various genes are activated, including anti-inflammatory genes [49]. Among these are the genes encoding annexin-1 (formerly known as lipocortin 1), IαB (NFαB inhibitor), IL-10, and the anti-inflammatory protein MAPK-phosphatase-1. This process, called transactivation, is responsible in part for the anti-inflammatory action of glucocorticoids.
The undesirable effects of corticosteroids are doses and time dependent and vary depending on the drug administered [51]. Some adverse effects follow a linear dose-response pattern, where the incidence increases together with dose increases (ecchymosis, cushingoid features, parchment-like skin and sleep disturbance). 
Patients with RA are often prescribed anti-rheumatic drug leflunomide whose primary aim is to reduce swelling and inflammation in the affected joints [50]. The active metabolite of leflunomide (teriflunomidum, or A77 1726) in reversible manner inhibits dehydroorotate dehydrogenase, a step limiting the rate of de novo pyrimidine synthesis [52]. This results in a reduction in the level of circulating pyrimidines and affects their availability for DNA and RNA synthesis, which in turn has an effect on the spreading of immune cells as well as the expression of inflammatory cytokines.

4. Botanicals as a Beacon of Hope on the Horizon

Herbal preparations used in humans demonstrate their effectiveness in the treatment of rheumatoid arthritis. One of the main positive effects is anti-inflammatory and antioxidant activity [53][54][55]. For example, such an herb as Boswellia spp., which has been used in Ayurvedic medicine since ancient times. Activity of boswellic acid and other active natural compounds of Boswellia spp. includes inhibition of microsomal prostaglandin E2 synthase-1 (PGE2) and 5-lipoxygenase, reducing the production or activation of inflammatory mediators such as matrix metalloproteinase (MMP)-9, MMP-13, cyclooxygenase (COX)-2, and nitric oxide (NO) and also has analgesic and anti-arthritic effects [56][57]. It has been found to reduce the number of osteophytes by attenuating inflammatory mediators such as C-reactive protein and hyaluronic acid [58][59][60]. Research by scientists has demonstrated the safety of using Boswellia serrata R. [59]. However, knee-related activities of daily living and quality of life did not improve significantly [61].
Polyphenols constitute an alternative direction of using natural compounds against rheumatoid arthritis. Application of an extract from Curcuma spp. containing polyphenol curcumin also showed its anti-inflammatory and antioxidant actions [62][63]. Curcumin inhibits the production of inflammatory mediators, such as a variety of MMPs, tumor necrosis factor-alpha (TNF-α), interleukin (IL)-8, IL-1, NO, via diminishing the activation of MAPK signaling pathways protein kinase B (Akt) and NF-κB [64][65] and leads to a decrease in prostaglandin synthesis [66]. Studies have shown that when compared with the treatment of ibuprofen, turmeric showed more effective results [67], and compared with diclofenac, a small number of side effects [68]. The dried plant Matricaria chamomilla L. has been used for many centuries in the treatment of joint pain [69][70]. The plant contains several phenolic compounds: apigenin, patuletin, luteolin, and glycosides [71] that reduce inflammation by reducing the levels of cytokines and PGE2, which play a role in the pathogenesis of arthritis. Epigallocatechin-3-gallate (EGCG), a catechin monomer that has antioxidant and anti-inflammatory effects, is isolated from tea leaves [72]. Green tea extract can inhibit the expression of IL-1β-induced chemokines This was studied in an experiment with rats having arthritis [73][74].
Date seeds of Phoenix dactylifera L. are a well-known traditional Moroccan remedy for pathological conditions involving inflammation such as RA [75]. The wild pomegranate Punica granatum L. has been used as a traditional medicine for various conditions, including pain and inflammation [76]. Pomegranate demonstrated potential inhibition of NO as well as reduction in paw edema in carrageenan-induced mice after administration of 100 mg/kg [77]. Pomegranate juice is one of the natural products that has also shown promising results in clinical trials for the treatment of RA symptoms, which may also be due to polyphenolic compounds with antioxidant and anti-inflammatory effects [78].
Glycosides from different plants show anti-inflammatory effect during rheumatoid arthritis. The root of Paeonia lactiflora P. has been used in Chinese medicine since ancient times. Inhibition of the production of leukotriene B4, PGE2, ROS, NO, and other pro-inflammatory mediators by paeoniflorin and total glucosides of paeony has been proven [79]. Combined treatment using this plant and methotrexate has shown beneficial effects in RA with fewer side effects [80]. Decoctions from Eremostachys laciniata (L.) are also used for arthritis [81][82][83]. It is assumed that the iridoid glycosides of this plant exhibited an anti-inflammatory effect. Curculigoorchioides G. contains curculiglycoside, which improves arthritis symptoms in rats [75] induced by collagen type II (CIA) and reduces levels of inflammatory factors (TNF-α, IL-1β, IL-6, IL-10, IL-12 and IL-17A). Its antiarthritic molecular mechanism may be related to the JAK/STAT/NF-Κb signaling pathway [84]. Extracts from the root of Tripterygium wilfordii Hook F. also has an immunosuppressive effect and inhibits the expression of pro-inflammatory mediators and cytokines, adhesion molecules and matrix metalloproteinases by macrophages [85][86][87].
Alkaloids of some plants show promising results against rheumatoid arthritis too. Synomenine, which is contained in the Chinese medicinal stem Sinomenium acutum Thunb., is used in the treatment of rheumatic diseases [88][89]. Synomenine can phosphorylate p62 Ser351, degrade Keap1 and increase Nrf2 expression, and play a role in protecting against bone destruction by increasing p62 expression and activating the p62-Keap1-Nrf2 axis. Research shows that sinomenine has an immunoregulatory effect on RA [90][91][92]
In recent years, scientists have agreed that a mixed herbal decoction can actively manifest itself in the fight against RA. For example, wutou decoction which consists of ephedra, peony, astragalus, licorice and Sichuan aconite [93]. In a study, wutou decoction was shown to effectively inhibit the expression of iNOS, TNF-α, and IL-6. Another example would be GuiZhiShaoYaoZhiMu Decoction (GSZD), which consists of Ramuluscinnamomi, P. lactiflora root, Radix GlycyrrhizaePreparata, Ephedra sp., Anemarrhenaasphodeloides Bunge root, Atractylodesmacrocephala and Zingiber officinale [94][95]. Combination treatment with GSZD and methotrexate was more effective and safer than RA treatment with methotrexate alone [96].

5. But How Many Joints Will We Save Tomorrow?

Today, different approaches exist that help reach successful regeneration, ranging from a gene-manipulated stem cell laden scaffold for cartilage regeneration [97] to a material-free cell therapy [98]. A promising strategy in medicine is the use of antisense techniques. Antisense technology has been widely and thoroughly explored. The perspective of antisense technology is that specific DNA or RNA can connect to target mRNA and afterward turn the ‘undesired’ gene off. According to complementarity principle, designed DNA or RNA molecules target mRNA of interest, effectively resulting in the degradation of the target mRNA or blocking initiation of translation [99][100].

Oberemok’s research group conceptualized and conducted research with the phosphorothioate antisense oligonucleotide (ASO) Cytos-11 that targets TNF-α mRNA. Cytos-11 selectively lowered levels of TNF-α in the peripheral blood and reduced swelling of joints in rats with RA with an efficacy similar to that of adalimumab. Generally, Cytos-11 was well tolerated by the rats, showing a low frequency of immunological reactions. Obtained results showed the potential of ASO to be used in combination with other drugs or as a monotherapy [101].
In an attempt to develop oligonucleotide therapy aimed at synovitis (an inflammatory disease of the synovial membrane of joints or ligaments with the accumulation of inflammatory effusion in the cavity), modulation of the phenotype of activated proliferative inflammatory synovial fibroblasts using antisense oligonucleotides was documented. For example, antisense oligonucleotides developed by Nakazawa et al. targeting the Notch-1 protein have been reported to inhibit both basal and TNF-α-caused proliferation of human synovial fibroblasts isolated from the synovial membrane of a patient with RA [102][103]. It has also been reported that antisense knockdown of the PTPN11 gene that encodes SHP-2 (a known proto-oncogene) inhibits the migration and survival of synovial fibroblasts [102][104].
In the future, better treatment for rheumatic diseases is indeed possible through the use of new methods of genetic engineering or cell therapy, such as autologous stem cell transplantation [105][106] and CAR-T-cell therapy [105][107]. Although these therapies are currently still risky and costly, the need for effective methods of therapy for autoimmune diseases is urgent, which is why other avenues should be explored as well. In the future, a new goal may be to find a cure, not just remission, for the disease [105].
Funds have already begun to appear for predicting the outcome of therapy and treatment of the disease [108]. With the help of artificial intelligence, it is possible to investigate changes in arthritis using X-rays [109] and can be used in silico trials to develop new treatments [110]. These new technologies using computerization will help to make more accurate predictions for patients for the future. The P4 (predictive, preventive, personalized and participatory) medicine approach has also appeared for early diagnosis of the disease and prevention [111].
Another innovative method is the use of gene therapy technology. An innovative method of treating rheumatoid arthritis using histone modification is a reliable alternative to traditional approaches. Histones are proteins that pack DNA inside a cell and regulate gene activity. Histone modification consists of changing the chemical structure of these proteins, which allows you to control the activity of certain genes. Found that in PDGF-induced FLS, the expression of Jumonji C family of histone demethylases (JMJD3) was increased through the Akt signaling pathway, and the proliferation and migration ability of FLS was weakened after inhibition or silence of JMJD3, and the symptoms of DBA/1 mice by collagen-induced arthritis were alleviated [112]
In recent years, a method of treating rheumatoid arthritis based on the use of phosphates has been developed. Studies have shown that phosphates can have an anti-inflammatory effect by reducing the production of inflammatory cytokines. They are also able to improve the function of the immune system. Dexamethasone sodium phosphate (DSP) is another anti-inflammatory and immunosuppressive glucocorticoid known to be used frequently in treating RA by decreasing cytokines expression and impeding functions of leukocyte, fibroblast and endothelial cells. Delivered using the beta-cyclodextrin nanocarriers (DSP-loaded H-βCD nanoparticles) was effective for RA in the AIA rat model. Thus, the use of nanocarriers has overcome the limited applications of DSP alone in chronic diseases and has been reported to be able to reduce arthritic score, paw thickness, and cytokine level [113].

6. Conclusions

Rheumatoid arthritis does not kill quickly but today it looks like a sentence to a hard-to-treat disease. This, coupled with the many different factors that can trigger RA, may explain why this disease is not treated successfully, with continued high incidence. Now the range of drugs is extensive, and nevertheless has its negative sides. Namely, side effects such as nausea, headaches, fatigue, allergic reactions, and hair loss.
A beacon of hope on the horizon is the use botanicals (polyphenols, glycosides, alkaloids, etc.) and antisense technologies, which have already proven themselves in a number of studies on the treatment of rheumatoid arthritis [114], as well as on the relief of pain in the treatment of diseases such as neurological dysfunction.
Two other useful proactive strategies are vaccination and early diagnosis of the disease, both of which can help prevent and forestall the development of any disease that has already begun. As far as treatment of ongoing disease, the ideal drug has not yet been found, and all of those in use have their side effects. But the prospects for the treatment of rheumatoid arthritis, despite its tortuous solutions, are bright, with promising areas of research fueled by people determined to help these patients. Though today’s battle is lost to rheumatoid arthritis, humanity prepares its joints for the win in the war for movement without pain.

References

  1. Berardi, S.; Corrado, A.; Maruotti, N.; Cici, D.; Cantatore, F.P. Osteoblast role in the pathogenesis of rheumatoid arthritis. Mol. Biol. Rep. 2021, 48, 2843–2852.
  2. Cheng, C.; Liao, H.; Wu, C. Tissue microenvironment dictates inflammation and disease activity in rheumatoid arthritis. J. Formos. Med. Assoc. 2022, 121, 1027–1033.
  3. Go, F.G.; Midwood, K.S. Internal danger: Activation of Toll-like receptors in rheumatoid arthritis. Rheumatology 2011, 51, 7–23.
  4. Klarenbeek, N.B.; Kerstens, P.J.; Huizinga, T.W.; Dijkmans, B.A.; Allaart, C.F. Recent advances in the management of rheumatoid arthritis. Br. Med. J. 2010, 341, c6942.
  5. Chauhan, K.; Jandu, J.S.; Brent, L.H.; Al-Dhahir, M.A. Rheumatoid Arthritis; StatPearls Publishing LLC: Treasure Island, FL, USA, 2023. Available online: https://www.ncbi.nlm.nih.gov/books/NBK441999/ (accessed on 25 May 2023).
  6. Sugiyama, D.; Nishimura, K.; Tamaki, K.; Tsuji, G.; Nakazawa, T.; Morinobu, A.; Kumagai, S. Impact of smoking as a risk factor for developing rheumatoid arthritis: A meta-analysis of observational studies. Ann. Rheum. Dis. 2010, 69, 70–81.
  7. Makrygiannakis, D.; Hermansson, M.; Ulfgren, A.K.; Nicholas, A.P.; Zendman, A.J.; Eklund, A.; Grunewald, J.; Skold, C.M.; Klareskog, L.; Catrina, A.I. Smoking increases peptidylarginine deiminase 2 enzyme expression in human lungs and increases citrullination in BAL cells. Ann. Rheum. Dis. 2008, 67, 1488–1492.
  8. Ishikawa, Y.; Terao, C. The Impact of Cigarette Smoking on Risk of Rheumatoid Arthritis: A Narrative Review. Cells 2020, 9, 475.
  9. Alsalahy, M.M.; Nasser, H.S.; Hashem, M.M.; Elsayed, S.M. Effect of tobacco smoking on tissue protein citrullination and disease progression in patients with rheumatoid arthritis. Saudi Pharm. J. 2010, 18, 75–80.
  10. Jiang, X.; Alfredsson, L.; Klareskog, L.; Bengtsson, C. Smokeless tobacco (moist snuff) use and the risk of developing rheumatoid arthritis: Results from a case-control study. Arthritis Care Res. 2014, 66, 1582–1586.
  11. Smolen, J.S.; Aletaha, D.; McInnes, I.B. Rheumatoid arthritis. Lancet 2016, 10055, 2023–2038.
  12. Yin, X.; Cheng, F.; Wang, X.; Mu, J.; Ma, C.; Zhai, C.; Wang, Q. Top 100 cited articles on rheumatoid arthritis: A bibliometric analysis. Medicine 2019, 98, 14523.
  13. Abhishek, A.; Doherty, M.; Kuo, C.F.; Mallen, C.D.; Zhang, W.; Grainge, M.J. Rheumatoid arthritis is getting less frequent-results of a nationwide population-based cohort study. Rheumatology 2017, 56, 736–744.
  14. Myasoedova, E.; Davis, J.M., 3rd; Achenbach, S.J.; Matteson, E.L.; Crowson, C.S. Trends in Prevalence of Functional Disability in Rheumatoid Arthritis Compared with the General Population. Mayo Clin. Proc. 2019, 94, 1035–1039.
  15. Scott, I.C.; Mount, J.; Barry, J.; Kirkham, B. Factors associated with disability in patients with rheumatoid arthritis with persistent moderate disease activity: A retrospective cohort study. BMC Rheumatol. 2020, 4, 63.
  16. Hsieh, P.; Wu, O.; Geue, C.; McIntosh, E.; McInnes, I.B.; Siebert, S. Economic burden of rheumatoid arthritis: A systematic review of literature in biologic era. Ann. Rheum. Dis. 2020, 79, 771–777.
  17. Gaitonde, P.; Shaya, F.T. Economic and Productivity Consequences Associated with Rheumatoid Arthritis Among Non-Institutionalized Individuals in The United States. Res. Methods–Cost Methods 2016, 19, 3.
  18. Firestein, G.S. Evolving concepts of rheumatoid arthritis. Nature 2003, 423, 356–361.
  19. Rudan, I.; Sidhu, S.; Papana, A.; Meng, S.J.; Xin-Wei, Y.; Wang, W.; Campbell-Page, R.M.; Demaio, A.R.; Nair, H.; Sridhar, D.; et al. Global Health Epidemiology Reference Group (GHERG). Prevalence of rheumatoid arthritis in low- and middle-income countries: A systematic review and analysis. J. Glob. Health 2015, 5, 010409.
  20. Van Vollenhoven, R.F. Sex differences in rheumatoid arthritis: More than meets the eye. BMC Med. 2009, 7, 12.
  21. Gerosa, M.; de Angelis, V.; Riboldi, P.; Meroni, P. Rheumatoid Arthritis: A Female Challenge. Women’s Health 2008, 4, 195–201.
  22. Kanaan, S.B.; Onat, O.E.; Balandraud, N.; Azzouz, D.F.; Roudier, J.; Ozcelik, T.; Lambert, N.C. Does telomere shortening in women with rheumatoid arthritis predict x chromosome inactivation bias? Ann. Rheum. Dis. 2013, 72, A51.
  23. Arend, W.P.; Dayer, J.M. Inhibition of the production and effects of interleukin-1 and tumor necrosis factor α in rheumatoid arthritis. Arthritis Rheum. 1995, 38, 151–160.
  24. Furst, D.E.; Breedveld, F.C.; Kalden, J.R.; Smolen, J.S.; Burmester, G.R.; Bijlsma, J.W.; Dougados, M.; Emery, P.; Keystone, E.C.; Klareskog, L.; et al. Updated consensus statement on biological agents, specifically tumour necrosis factor α (TNFα) blocking agents and interleukin-1 receptor antagonist (IL-1ra), for the treatment of rheumatic diseases, 2004. Ann. Rheum. Dis. 2004, 63 (Suppl. S2), ii2–ii12.
  25. Holdsworth, E.A.; Donaghy, B.; Fox, K.M.; Desai, P.; Collier, D.H.; Furst, D.E. Biologic and Targeted Synthetic DMARD Utilization in the United States: Adelphi Real World Disease Specific Programme for Rheumatoid Arthritis. Rheumatol. Ther. 2021, 8, 1637–1649.
  26. Fassbender, H.G. The clinical presentation of rheumatoid arthritis: The results from three separate pathogenetic mechanisms in adults and children. Acta Clin. Croat. 2008, 47, 50–55.
  27. Ibrahim, F.; Ma, M.; Scott, D.L.; Scott, I.C. Defining the relationship between pain intensity and disease activity in patients with rheumatoid arthritis: A secondary analysis of six studies. Arthritis Res. Ther. 2022, 24, 218.
  28. Kronzer, V.L.; Davis, J.M., 3rd. Etiologies of Rheumatoid Arthritis: Update on Mucosal, Genetic, and Cellular Pathogenesis. Curr. Rheumatol. Rep. 2021, 23, 21.
  29. Klockars, M.; Koskela, R.S.; Järvinen, E.; Kolari, P.J.; Rossi, A. Silica exposure and rheumatoid arthritis: A follow up study of granite workers 1940–81. Br. Med. J. (Clin. Res. Ed.) 1987, 294, 997–1000.
  30. Khanna, S.; Jaiswal, K.S.; Gupta, B. Managing Rheumatoid Arthritis with Dietary Interventions. Front. Nutr. 2017, 4, 52.
  31. Tiwari, V.; Jandu, J.S.; Bergman, M.J. Rheumatoid Factor; StatPearls Publishing: Treasure Island, FL, USA, 2022. Available online: https://www.ncbi.nlm.nih.gov/books/NBK532898/ (accessed on 24 July 2023).
  32. Holm, B.E.; Sandhu, N.; Tronstrom, J.; Lydolph, M.; Trier, N.H.; Houen, G. Species cross-reactivity of rheumatoid factors and implications for immunoassays. Scand. J. Clin. Lab. Investig. 2015, 75, 51–63.
  33. Gehin, J.E.; Klaasen, R.A.; Norli, E.S.; Warren, D.J.; Syversen, S.W.; Goll, G.L.; Bjøro, T.; Kvien, T.K.; Mjaavatten, M.D.; Bolstad, N.; et al. Rheumatoid factor and falsely elevated results in commercial immunoassays: Data from an early arthritis cohort. Rheumatol. Int. 2021, 41, 1657–1665.
  34. Romão, V.C.; Fonseca, J.E. Etiology and Risk Factors for Rheumatoid Arthritis: A State-of-the-Art Review. Front. Med. 2021, 8, 689698.
  35. Van Boxel, J.A.; Paget, S.A. Predominantly T-cell infiltrate in rheumatoid synovial membranes. N. Engl. J. Med. 1975, 293, 517–520.
  36. Morimoto, C.; Romain, P.L.; Fox, D.A.; Anderson, P.; DiMaggio, M.; Levine, H.; Schlossman, S.F. Abnormalities in CD4+ T-lymphocyte subsets in inflammatory rheumatic diseases. Am. J. Med. 1988, 84, 817–825.
  37. Simon, A.K.; Seipelt, E.; Sieper, J. Divergent T-cell cytokine patterns in inflammatory arthritis. Proc. Natl. Acad. Sci. USA 1994, 91, 8562–8566.
  38. Ziolkowska, M.; Koc, A.; Luszczykiewicz, G.; Ksiezopolska-Pietrzak, K.; Klimczak, E.; Chwalinska-Sadowska, H.; Maslinski, W. High levels of IL-17 in rheumatoid arthritis patients: IL-15 triggers in vitro IL-17 production via cyclosporin A-sensitive mechanism. J. Immunol. 2000, 164, 2832–2838.
  39. Duong, S.Q.; Crowson, C.S.; Athreya, A.; Atkinson, E.J.; Davis, J.M., III; Warrington, K.J.; Matteson, E.L.; Weinshilboum, R.; Wang, L.; Myasoedova, E. Clinical predictors of response to methotrexate in patients with rheumatoid arthritis: A machine learning approach using clinical trial data. Arthritis Res. Ther. 2022, 24, 162.
  40. Haraoui, B.; Bykerk, V. Etanercept in the treatment of rheumatoid arthritis. Ther. Clin. Risk Manag. 2007, 3, 99–105.
  41. Feist, E.; Baraliakos, X.; Behrens, F.; Thaçi, D.; Klopsch, T.; Plenske, A.; Blindzellner, L.K.; Klaus, P.; Meng, T.; Löschmann, P.A. Effectiveness of Etanercept in Rheumatoid Arthritis: Real-World Data from the German Non-interventional Study ADEQUATE with Focus on Treat-to-Target and Patient-Reported Outcomes. Rheumatol. Ther. 2022, 9, 621–635.
  42. Stacy, J.M.; Greenmyer, J.R.; Beal, J.R.; Sahmoun, A.E.; Diri, E. The efficacy of low dose short-term prednisone therapy for remission induction in newly diagnosed rheumatoid arthritis patients. Adv. Rheumatol. 2021, 61, 50.
  43. Guadagnin, D.A.; Mazzali, L.V.; Skare, T.L.; Kahlow, B.S. Treating rheumatoid arthritis with leflunomide monotherapy versus combination therapy with methotrexate. Eur. J. Rheumatol. 2021, 8, 12–15.
  44. Smolen, J.S.; Landewé, R.B.M.; Bergstra, S.A.; Kerschbaumer, A.; Sepriano, A.; Aletaha, D.; Caporali, R.; Edwards, C.J.; Hyrich, K.L.; Pope, J.E.; et al. EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2022 update. Ann. Rheum. Dis. 2023, 82, 3–18.
  45. Tanaka, Y. Subcutaneous injection of methotrexate: Advantages in the treatment of rheumatoid arthritis. Mod. Rheumatol. 2022, 33, roac156.
  46. Brown, P.M.; Pratt, A.G.; Isaacs, J.D. Mechanism of action of methotrexate in rheumatoid arthritis, and the search for biomarkers. Nat. Rev. Rheumatol. 2016, 12, 731–742.
  47. Conway, R.; Carey, J.J. Risk of liver disease in methotrexate treated patients. World J. Hepatol. 2017, 9, 1092–1100.
  48. Bedoui, Y.; Guillot, X.; Sélambarom, J.; Guiraud, P.; Giry, C.; Jaffar-Bandjee, M.C.; Ralandison, S.; Gasque, P. Methotrexate an Old Drug with New Tricks. Int. J. Mol. Sci. 2019, 20, 5023.
  49. Bashar, T.; Apu, M.N.H.; Mostaid, M.S.; Islam, M.S.; Hasnat, A. Pharmacokinetics and bioavailability study of a prednisolone tablet as a single oral dose in Bangladeshi healthy volunteers. Dose Response 2018, 16, 1559325818783932.
  50. Cain, D.; Cidlowski, J. Immune regulation by glucocorticoids. Nat. Rev. Immunol. 2017, 17, 233–247.
  51. Huscher, D.; Thiele, K.; Gromnica-Ihle, E.; Hein, G.; Demary, W.; Dreher, R.; Zink, A.; Buttgereit, F. Dose-related patterns of glucocorticoid-induced side effects. Ann. Rheum. Dis. 2009, 68, 1119–1124.
  52. Breedveld, F.C.; Dayer, J.M. Leflunomide: Mode of action in the treatment of rheumatoid arthritis. Ann. Rheum. Dis. 2000, 59, 841–849.
  53. Lindler, B.N.; Long, K.E.; Taylor, N.A.; Lei, W. Use of Herbal Medications for Treatment of Osteoarthritis and Rheumatoid Arthritis. Medicines 2020, 7, 67.
  54. Siemoneit, U.; Koeberle, A.; Rossi, A.; Dehm, F.; Verhoff, M.; Reckel, S.; Maier, T.J.; Jauch, J.; Northoff, H.; Bernhard, F.; et al. Inhibition of microsomal prostaglandin E2 synthase-1 as a molecular basis for the anti-inflammatory actions of boswellic acids from frankincense. Br. J. Pharm. 2010, 162, 147–162.
  55. Safayhi, H.; Mack, T.; Sabieraj, J.; Anazodo, M.I.; Subramanian, L.R.; Ammon, H.P. Boswellic acids: Novel, specific, nonredox inhibitors of 5-lipoxygenase. J. Pharm. Exp. Ther. 1992, 261, 1143–1146.
  56. Xu, X.; Blinder, L.; Shen, J.; Gong, H.; Finnegan, A.; Williams, J.W.; Chong, A.S. In vivo mechanism by which leflunomide controls lymphoproliferative and autoimmune disease in MRL/MpJ-Ipr/Ipr mice. J. Immunol. 1997, 159, 167–174.
  57. Elder, R.T.; Xu, X.; Williams, J.W.; Gong, H.; Finnegan, A.; Chong, A.S.F. The immunosuppressive metabolite of leflunomide, A77 1726, a Vects murine T cells through two biochemical mechanisms. J. Immunol. 1997, 159, 22–27.
  58. Majeed, M.; Majeed, S.; Narayanan, N.K.; Nagabhushanam, K. A pilot, randomized, double-blind, placebo-controlled trial to assess the safety and efficacy of a novel Boswellia serrata extract in the management of osteoarthritis of the knee. Phytother. Res. 2019, 33, 1457–1468.
  59. Majeed, M.; Vaidyanathan, P.; Natarajan, S.; Majeed, S.; Vuppala, K.K. Effect of Boswellin® Super on knee pain in Japanese adults: A randomized, double-blind, placebo-controlled trial. Eur. J. Biomed. 2016, 3, 293–298.
  60. Chopra, A.; Lavin, P.; Patwardhan, B.; Chitre, D. Randomized double blind trial of an ayurvedic plant derived formulation for treatment rheumatoid arthritis. J. Rheumatol. 2000, 27, 1365–1372.
  61. Razavi, S.Z.E.; Karimi, M.; Kamalinejad, M. The efficacy of topical oliban oil (Boswellia carterii, B.) in relieving the symptoms of knee osteoarthritis. Phys. Med. Rehabil. Electrodiagn. 2019, 1, 7–13.
  62. Goel, A.; Kunnumakkara, A.B.; Aggarwal, B.B. Curcumin as “Curecumin”: From kitchen to clinic. Biochem. Pharmacol. 2008, 75, 787–809.
  63. Chin, K.-Y. The spice for joint inflammation: Anti-inflammatory role of curcumin in treating osteoarthritis. Drug Des. Dev. Ther. 2016, 10, 3029–3042.
  64. Prasad, S.; Gupta, S.C.; Tyagi, A.K.; Aggarwal, B.B. Curcumin, a component of golden spice: From bedside to bench and back. Biotechnol. Adv. 2014, 32, 1053–1064.
  65. Aggarwal, B.B.; Surh, Y.-J.; Shishodia, S. The Molecular Targets and Therapeutic Uses of Curcumin in Health and Disease; Springer Science & Business Media: Advances in Experimental Medicine and Biology (AEMB): Berlin/Heidelberg, Germany, 2007; Volume 595.
  66. Shep, D.; Khanwelkar, C.; Gade, P.; Karad, S. Efficacy and safety of combination of curcuminoid complex and diclofenac versus diclofenac in knee osteoarthritis. Medicine 2020, 99, e19723.
  67. Kuptniratsaikul, V.; Dajpratham, P.; Taechaarpornkul, W.; Buntragulpoontawee, M.; Lukkanapichonchut, P.; Chootip, C.; Saengsuwan, J.; Tantayakom, K.; Laongpech, S. Efficacy and safety of Curcuma domestica extracts compared with ibuprofen in patients with knee osteoarthritis: A multicenter study. Clin. Interv. Aging 2014, 9, 451–458.
  68. Shep, D.; Khanwelkar, C.; Gade, P.; Karad, S. Safety and efficacy of curcumin versus diclofenac in knee osteoarthritis: A randomized open-label parallel-arm study. Trials 2019, 20, 214.
  69. Shoara, R.; Hashempur, M.H.; Ashraf, A.; Salehi, A.; Dehshahri, S.; Habibagahi, Z. Efficacy and safety of topical Matricaria chamomilla L. (chamomile) oil for knee osteoarthritis: A randomized controlled clinical trial. Complement. Ther. Clin. Pract. 2015, 21, 181–187.
  70. El Mihyaoui, A.; Esteves da Silva, J.C.G.; Charfi, S.; Candela Castillo, M.E.; Lamarti, A.; Arnao, M.B. Chamomile (Matricaria chamomilla L.): A Review of Ethnomedicinal Use, Phytochemistry and Pharmacological Uses. Life 2022, 12, 479.
  71. Pirouzpanah, S.; Mahboob, S.; Sanayei, M.; Hajaliloo, M.; Safaeiyan, A. The effect of chamomile tea consumption on inflammation among rheumatoid arthritis patients: Randomized clinical trial. Prog. Nutr. 2017, 19, 27–33.
  72. Yamagata, K. Protective effect of epigallocatechin gallate on endothelial disorders in atherosclerosis. J. Cardiovasc. Pharmacol. 2020, 75, 292–298.
  73. Karatas, A.; Dagli, A.F.; Orhan, C.; Gencoglu, H.; Ozgen, M.; Sahin, N.; Sahin, K.; Koca, S.S. Epigallocatechin 3-gallate attenuates arthritis by regulating Nrf2, HO-1, and cytokine levels in an experimental arthritis model. Biotechnol. Appl. Biochem. 2020, 67, 317–322.
  74. Srirangan, S.; Choy, E.H. The role of interleukin 6 in the pathophysiology of rheumatoid arthritis. Ther. Adv. Musculoskelet. Dis. 2010, 2, 247–256.
  75. Bouhlali, E.d.T.; Hmidani, A.; Bourkhis, B.; Khouya, T.; Ramchoun, M.; Filali-Zegzouti, Y.; Alem, C. Phenolic profile and anti-inflammatory activity of four Moroccan date (Phoenix dactylifera L.) seed varieties. Heliyon 2020, 6, e03436.
  76. Zahin, M.; Ahmad, I.; Gupta, R.C.; Aqil, F. Punicalagin and Ellagic Acid Demonstrate Antimutagenic Activity and Inhibition of Benzopyrene Induced DNA Adducts. Biomed Res. Int. 2014, 2014, 467465.
  77. Lee, C.J.; Chen, L.G.; Liang, W.L.; Wang, C.C. Anti-inflammatory effects of Punica granatum Linne in vitro and in vivo. Food Chem. 2010, 118, 315–322.
  78. Ghavipour, M.; Sotoudeh, G.; Tavakoli, E.; Mowla, K.; Hasanzadeh, J.; Mazloom, Z. Pomegranate extract alleviates disease activity and some blood biomarkers of inflammation and oxidative stress in Rheumatoid Arthritis patients. Eur. J. Clin. Nutr. 2017, 71, 92–96.
  79. Zhang, W.; Dai, S.-M. Mechanisms involved in the therapeutic effects of Paeonia lactiflora Pallas in rheumatoid arthritis. Int. Immunopharmacol. 2012, 14, 27–31.
  80. Zhang, L.; Wei, W. Anti-inflammatory and immunoregulatory effects of paeoniflorin and total glucosides of paeony. Pharmacol. Ther. 2020, 207, 107452.
  81. Delazar, A.; Sarker, S.D.; Nahar, L.; Jalali, S.B.; Modarresi, M.; Hamedeyazdan, S.; Babaei, H.; Javadzadeh, Y.; Asnaashari, S.; Moghadam, S.B. Rhizomes of Eremostachys laciniata: Isolation and Structure Elucidation of Chemical Constituents and a Clinical Trial on Inflammatory Diseases. Adv. Pharm. Bull. 2013, 3, 385–393.
  82. Erdemoglu, N.; Turan, N.N.; Cakõcõ, I.; Şener, B.; Aydõn, A. Antioxidant activities of some Lamiaceae plant extracts. Phytother. Res. 2006, 20, 9–13.
  83. Tan, S.; Xu, J.; Lai, A.; Cui, R.; Bai, R.; Li, S.; Liang, W.; Zhang, G.; Jiang, S.; Liu, S.; et al. Curculigoside exerts significant anti–arthritic effects in vivo and in vitro via regulation of the JAK/STAT/NF–κB signaling pathway. Mol. Med. Rep. 2019, 19, 2057–2064.
  84. Han, J.; Wan, M.; Ma, Z.; Hu, C.; Yi, H. Prediction of targets of curculigoside a in osteoporosis and rheumatoid arthritis using network pharmacology and experimental verification. Drug Des. Dev. Ther. 2020, 14, 5235–5250.
  85. Pyatt, D.W.; Yang, Y.; Mehos, B.; Le, A.; Stillman, W.; Irons, R.D. Hematotoxicity of the Chinese Herbal Medicine Tripterygium wilfordii Hook f in CD34-Positive Human Bone Marrow Cells. Mol. Pharm. 2000, 57, 512–518.
  86. Tang, W.; Zuo, J.-P. Immunosuppressant discovery from Tripterygium wilfordii Hook f: The novel triptolide analog (5R)-5-hydroxytriptolide (LLDT-8). Acta Pharm. Sin. 2012, 33, 1112–1118.
  87. Cibere, J.; Deng, Z.; Lin, Y.; Ou, R.; He, Y.; Wang, Z.; Thorne, A.; Lehman, A.J.; Tsang, I.K.; Esdaile, J.M. A randomized double blind, placebo controlled trial of topical Tripterygium wilfordii in rheumatoid arthritis: Reanalysis using logistic regression analysis. J. Rheumatol. 2003, 30, 465–467.
  88. Liu, X.; Wang, Z.; Qian, H.; Tao, W.; Zhang, Y.; Hu, C.; Mao, W.; Guo, Q. Natural medicines of targeted rheumatoid arthritis and its action mechanism. Front. Immunol. 2022, 13, 945129.
  89. Liu, W.; Qian, X.; Ji, W.; Lu, Y.; Wei, G.; Wang, Y. Effects and safety of sinomenine in treatment of rheumatoid arthritis contrast to methotrexate: A systematic review and meta-analysis. J. Tradit. Chin. Med. 2016, 36, 564–577.
  90. Zeng, M.Y.; Tong, Q.Y. Anti-inflammation effects of sinomenine on macrophages through suppressing activated TLR4/NF-κB signaling pathway. Curr. Med. Sci. 2020, 40, 130–137.
  91. Tong, B.; Yu, J.; Wang, T.; Dou, Y.; Wu, X.; Kong, L.; Dai, Y.; Xia, Y. Sinomenine suppresses collagen-induced arthritis by reciprocal modulation of regulatory T cells and Th17 cells in gut-associated lymphoid tissues. Mol. Immunol. 2015, 65, 94–103.
  92. Feng, Z.T.; Yang, T.; Hou, X.Q.; Wu, H.Y.; Feng, J.T.; Ou, B.J.; Cai, S.-J.; Li, J.; Mei, Z.-G. Sinomenine mitigates collagen-induced arthritis mice by inhibiting angiogenesis. BioMed. Pharmacother. 2019, 113, 108759.
  93. Hilvo, M.; Baranauskiene, L.; Salzano, A.M.; Scaloni, A.; Matulis, D.; Innocenti, A.; Scozzafava, A.; Monti, S.M.; Di Fiore, A.; De Simone, G.; et al. Biochemical characterization of CA IX, one of the most active carbonic anhydrase isozymes. J. Biol. Chem. 2008, 283, 27799–27809.
  94. Tian, Y.; Maosheng, Q.; Wei, X. Experimental Study of GuizhiShaoyaoZhimu Decoction on Gene Regulation of Synovial Cell Apoptosis in Rheumatoid Arthritis. Contemp. Med. 2010, 16, 18–20.
  95. Guo, Q.; Mao, X.; Zhang, Y.; Meng, S.; Xi, Y.; Ding, Y.; Zhang, X.; Dai, Y.; Liu, X.; Wang, C.; et al. Guizhi-Shaoyao-Zhimu decoction attenuates rheumatoid arthritis partially by reversing inflammation-immune system imbalance. J. Transl. Med. 2016, 14, 165.
  96. Feng, C.; Chen, R.; Wang, K.; Wen, C.; Xu, Z. Chinese traditional medicine (GuiZhi-ShaoYao-ZhiMu decoction) as an add-on medication to methotrexate for rheumatoid arthritis: A meta-analysis of randomized clinical trials. Ther. Adv. Chronic Dis. 2021, 12, 2040622321993438.
  97. Wang, D.A.; Williams, C.G.; Yang, F.; Elisseeff, J.H. Enhancing the tissue-biomaterial interface: Tissue-initiated integration of biomaterials. Adv. Funct. Mater. 2004, 14, 1152–1159.
  98. Federici, T.; Boulis, N. Gene therapy for peripheral nervous system diseases. Curr. Gene Ther. 2007, 7, 239–248.
  99. Sazani, P.; Vacek, M.M.; Kole, R. Short-term and long-term modulation of gene expression by antisense therapeutics. Curr. Opin. Biotechnol. 2002, 13, 468–472.
  100. Gopi, C.; Dhanaraju, M.D.; Dhanaraju, K. Antisense oligonucleotides: Recent progress in the treatment of various diseases. Beni-Suef Univ. J. Basic Appl. Sci. 2022, 11, 19.
  101. Makalish, T.P.; Golovkin, I.O.; Oberemok, V.V.; Laikova, K.V.; Temirova, Z.Z.; Serdyukova, O.A.; Novikov, I.A.; Rosovskyi, R.A.; Gordienko, A.I.; Zyablitskaya, E.Y.; et al. Anti-Rheumatic Effect of Antisense Oligonucleotide Cytos-11 Targeting TNF-α Expression. Int. J. Mol. Sci. 2021, 22, 1022.
  102. Wijesinghe, S.N.; Lindsay, M.A.; Jones, S.W. Oligonucleotide Therapies in the Treatment of Arthritis: A Narrative Review. Biomedicines 2021, 9, 902.
  103. Nakazawa, M.; Ishii, H.; Aono, H.; Takai, M.; Honda, T.; Aratani, S.; Fukamizu, A.; Nakamura, H.; Yoshino, S.; Kobata, T.; et al. Role of Notch-1 intracellular domain in activation of rheumatoid synoviocytes. Arthritis Rheum. 2001, 44, 1545–1554.
  104. Stanford, S.M.; Maestre, M.F.; Campbell, A.M.; Bartok, B.; Kiosses, W.B.; Boyle, D.L.; Arnett, H.A.; Mustelin, T.; Firestein, G.S.; Bottini, N. Protein tyrosine phosphatase expression profile of rheumatoid arthritis fibroblast-like synoviocytes: A novel role of SH2 domain-containing phosphatase 2 as a modulator of invasion and survival. Arthritis Rheum. 2013, 65, 1171–1180.
  105. Schett, G.; Tanaka, Y.; Isaacs, J.D. Why remission is not enough: Underlying disease mechanisms in RA that prevent cure. Nat. Rev. Rheumatol. 2021, 17, 135–144.
  106. Alexander, T.; Thiel, A.; Rosen, O.; Massenkeil, G.; Sattler, A.; Kohler, S.; Mei, H.; Radtke, H.; Gromnica-Ihle, E.G.; Arnold, R.; et al. Depletion of autoreactive immunologic memory followed by autologous hematopoietic stem cell transplantation in patients with refractory SLE induces long-term remission through de novo generation of a juvenile and tolerant immune system. Blood 2009, 113, 214–223.
  107. Hsieh, M.C.; Lee, J.J. Preliminary study of VR and AR applications in medical and healthcare education. J. Nurs. Health Stud. 2018, 3, 1.
  108. Tao, W.; Concepcion, A.N.; Vianen, M.; Marijnissen, A.C.A.; Lafeber, F.P.G.J.; Radstake, T.R.D.J.; Pandit, A. Multiomics and machine learning accurately predict clinical response to adalimumab and etanercept therapy in patients with rheumatoid arthritis. Arthritis Rheumatol. 2021, 73, 212–222.
  109. Bressem, K.K.; Vahldiek, J.L.; Adams, L.; Niehues, S.M.; Haibel, H.; Rodriguez, V.R.; Torgutalp, M.; Protopopov, M.; Proft, F.; Rademacher, J.; et al. Deep learning for detection of radiographic sacroiliitis: Achieving expert-level performance. Arthritis Res. Ther. 2021, 23, 106.
  110. Vodencarevic, A.; Tascilar, K.; Hartmann, F.; Reiser, M.; Hueber, A.J.; Haschka, J.; Bayat, S.; Meinderink, T.; Knitza, J.; Mendez, L.; et al. Advanced machine learning for predicting individual risk of flares in rheumatoid arthritis patients tapering biologic drugs. Arthritis Res. Ther. 2021, 23, 67.
  111. Sagner, M.; McNeil, A.; Puska, P.; Auffray, C.; Price, N.D.; Hood, L.; Lavie, C.J.; Han, Z.-G.; Chen, Z.; Brahmachari, S.K.; et al. The P4 health spectrum—A predictive, preventive, personalized and participatory continuum for promoting healthspan. Prog. Cardiovasc. Dis. 2017, 59, 506–521.
  112. Wu, W.; Qin, M.; Jia, W.; Huang, Z.; Li, Z.; Yang, D.; Huang, M.; Xiao, C.; Long, F.; Mao, J.; et al. Cystathionine-γ-lyase ameliorates the histone demethylase JMJD3-mediated autoimmune response in rheumatoid arthritis. Cell. Mol. Immunol. 2019, 16, 694–705.
  113. Ben Mrid, R.; Bouchmaa, N.; Ainani, H.; El Fatimy, R.; Malka, G.; Mazini, L. Anti-rheumatoid drugs advancements: New insights into the molecular treatment of rheumatoid arthritis. Biomed. Pharmacother. 2022, 151, 113126.
  114. Nicholson, T.A.; Sagmeister, M.; Wijesinghe, S.N.; Farah, H.; Hardy, R.S.; Jones, S.W. Oligonucleotide Therapeutics for Age-Related Musculoskeletal Disorders: Successes and Challenges. Pharmaceutics 2023, 15, 237.
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: 241
Revisions: 2 times (View History)
Update Date: 30 Oct 2023
1000/1000
Video Production Service