Open access peer-reviewed chapter

Regulating Reactive Oxygen Species in Rheumatoid Arthritis: Insights into Cell Signaling Pathways and Nano-Particles as Carriers

Written By

Tharun Srinivasan, Pavithra Ashok, Venkatraman Sairam and Amala Reddy

Submitted: 15 August 2023 Reviewed: 12 September 2023 Published: 18 December 2023

DOI: 10.5772/intechopen.113191

From the Edited Volume

Reactive Oxygen Species - Advances and Developments

Edited by Rizwan Ahmad

Chapter metrics overview

30 Chapter Downloads

View Full Metrics

Abstract

Rheumatoid arthritis (RA) is a chronic and debilitating inflammatory condition characterized by joint degradation and permanent disability. Excessive production of reactive oxygen species (ROS) is implicated in RA pathogenesis, leading to oxidative stress and tissue damage. In recent years, nano-particles have emerged as promising carriers for ROS regulation therapies in RA treatment. This review explores the interplay between ROS and RA, emphasizing the importance of cell signaling pathways in ROS control. The potential of nano-particles as targeted drug delivery systems to scavenge excess ROS and restore redox equilibrium within affected cells is discussed. Preclinical studies using ROS-neutralizing nano-particles in RA animal models have shown significant reductions in joint inflammation and cartilage degradation. Clinical trials have further validated the safety and efficacy of nano-particle treatments in RA patients, leading to improved disease activity and joint function. The review highlights the benefits of nano-particle-based ROS control therapies, including improved drug solubility, prolonged drug delivery, reduced systemic side effects, and enhanced specificity for inflamed joints. However, further research is needed to fully understand the intricate mechanisms of ROS management in RA and optimize nano-particle production and delivery. Overall, nano-particle-based ROS control therapy holds great promise for revolutionizing RA treatment and improving the quality of life for affected individuals.

Keywords

  • rheumatoid arthritis
  • reactive oxygen species (ROS)
  • Nano-particles
  • inflammation
  • drug delivery
  • redox equilibrium

1. Introduction

1.1 Rheumatoid arthritis (RA)

Rheumatoid arthritis (RA) is a chronic and systemic inflammatory condition that leads to slow joint degradation and may eventually end in permanent disability. RA is characterized by the infiltration of inflammatory cells in the synovial tissue, synovial hyperplasia, angiogenesis, and the disintegration of cartilage, which may eventually lead to bone deterioration [1]. Furthermore, RA is linked with a large socioeconomic burden, including direct medical and nonmedical expenditures, as well as indirect costs such as productivity loss, early death, and caregiver burden [2]. Stiffness throughout the morning that continues in the afflicted joints for more than 30 minutes, tiredness, fever, weight loss, sensitive, swollen, and hot joints, and rheumatoid nodules beneath the skin are all frequent symptoms of RA. This disorder commonly arises between the ages of 35 and 60, with intervals of remission and exacerbation [3]. The self-reacting reaction also involves a mix of genetic susceptibility and environmental influences. As with many other autoimmune illnesses, the MHC region plays a crucial role in the genetic contribution. Human sensitivity to RA is intimately connected to the MHC class II molecules HLA-DR1 and HLA-DR4. Similarly, the DBA/1 and B10.Q mouse strains exhibit the I-Aq and I-Ar haplotypes and are very vulnerable to collagen-induced arthritis (CIA) as an experimental model of RA. It has also been discovered that the expression of a limited T-cell receptor (TCR) repertoire is related to the development of CIA in mice [4]. In RA, enduring T-cell and monocyte-mediated synovial inflammation are the primary cause of disease development. Pannus is a distinctive pathological result of RA, with tumor-like features that promote synovial proliferation and bone erosion, which may ultimately lead to disability and adversely damage patients’ quality of life [5].

A classification of highly reactivity chemicals referred to as reactive oxygen species (ROS) are collectively made up of free radicals and other oxidants [6]. Moreover, they govern intracellular signaling pathways, transcription factors, and cytokine production to assist with the continual preservation of the cell’s redox state [7]. As enigmatic and highly reactive entities, ROS assume multifaceted roles, adroitly navigating the intricate terrain of cellular metabolism within the microcosm of articular joint tissues, endowing their study with an aura of enthralling fascination [8]. Yet, beneath the veneer of apparent simplicity lies a delicate equilibrium between ROS production and the robust antioxidant defense mechanisms inherent in tissues, which renders them vulnerable to perturbations that unfurl the ominous specter of oxidative stress [9]. When this finely orchestrated balance is disrupted, giving rise to an environment where ROS generation overtakes the cellular antioxidant defenses, the insidious process of cartilage degradation ensues, laying the treacherous path for the relentless onslaught of RA [10]. Indeed, the captivating influence of ROS in the genesis of RA has spurred ceaseless and ardent research endeavors, culminating in a vast and diverse repository of knowledge [8]. The protean essence of ROS transcends the confines of mere convention, adroitly orchestrating the preservation of cellular redox homeostasis while deftly navigating an intricate web of elusive intracellular signaling pathways and transcriptional factors [11]. Moreover, their versatility extends to fueling the production of proinflammatory cytokines and masterfully coordinating the intricate process of angiogenesis, thus propelling the insidious trajectory of RA’s debilitating course [11]. Within the sanctified precincts of the synovial membrane, the very epicenter of rheumatoid joint pathology, the activation of leukocytes incites an upsurge in oxygen consumption, thereby igniting the unbridled cascade of ROS [11]. In recent years, scientists have started examining the potential of nanoparticles as therapeutic intervention carriers for cell signaling pathways involved in the regulation of reactive oxygen species and illnesses linked to ROS. One intriguing area of research is the potential of nanoparticles as therapeutic intervention carriers in cell signaling pathways involved in reactive oxygen species regulation [12]. These nanoparticles can be designed to either transport specific chemicals or compounds that target and affect the activity of enzymes and proteins involved in ROS-related signaling cascades or to scavenge excess ROS and restore redox equilibrium within cells. Additionally, nano-particles have the potential to improve the administration and efficacy of treatments by encouraging cellular absorption, enhancing pharmaceutical stability and bioavailability, and minimizing off-target effects [13]. They are the perfect carriers for delivering therapeutic chemicals to target cells and tissues due to their tiny size and high surface area-to-volume ratio [13]. Nanoparticles’ special qualities enable them to efficiently pass through cellular barriers and deliver therapeutic substances right to the intended region of action. For instance, ROS-responsive nanoparticles have demonstrated considerable promise in the delivery of medicines to immune cells for immunotherapy or anti-leukemia systems [14]. Nano-particles can be created to scavenge excess ROS and restore redox equilibrium inside cells in addition to serving as carriers for treatments [15]. Nanoparticles are a fascinating topic of investigation in the realm of cell signaling pathways involved in ROS control because of their dual usefulness in regulating ROS levels and delivering treatments [12]. For cellular homeostasis to be preserved and the emergence of various pathological diseases to be avoided, cell signaling pathways involved in ROS control are essential [16]. Oxidative stress inevitably leads in cell damage and proliferation of disease conditions when there is an imbalance between the production of ROS and antioxidant Defense machinery [17].

Advertisement

2. Reactive oxygen species (ROS) and rheumatoid arthritis

Forms of atmospheric oxygen that are partly reduced or stimulated are known as reactive oxygen species [18]. In terms of physiology, ROS carries out two distinct functions within the body. On the one hand, numerous physiological functions of the body’s organs depend on ROS. For instance, they serve an extremely important part in ensuring the regulation of gene expression and cell signaling [19]. Subsequently, an imbalance between the synthesis of ROS and the body’s antioxidant defensive systems may ultimately give rise to oxidative stress, which is the reverse consequence of excessive ROS production [20]. When there is a surplus of ROS and a breakdown in antioxidants, which are molecules that scavenge ROS, oxidative stress emerges. Under normal conditions, the body’s antioxidant defense system can efficiently neutralize and eliminate ROS to maintain redox balance [21]. As a result of several metabolic activities, the body frequently creates ROS, Oxidative stress can nevertheless occur when the body’s antioxidant capacity is surpassed by ROS production or when exogenous stressors increase ROS levels In signaling pathways that regulate cellular functions such as growth, proliferation, and immune reactions throughout physiological processes, ROS play a crucial role [22]. However, ROS levels can harm cells if they rise excessively as a result of things like smoking, chronic inflammation, environmental toxins, or aging [23].

Rheumatoid arthritis is only one of the many illnesses that reactive oxygen species have the power to start and worsen [24]. The formation of ROS in the synovial membrane of rheumatoid joints increases significantly as a result of leukocyte activity and significant oxygen consumption [11]. An imbalance between the development of reactive oxygen species and antioxidant defenses, leading to oxidative stress, regulates the dysregulation of ROS in RA [25]. In rheumatoid arthritis, ROS is close to equilibrium, causing damage to the tissues and chronic inflammation [26]. Rheumatoid arthritis has been characterized by chronic inflammation, and a prior study revealed that reactive oxygen species serve as vital for controlling this inflammatory response [24]. ROS serve an essential role in the development and progression of rheumatoid arthritis because they possess an effect on the oxidation of cellular membranes [24]. Due to the cellular membrane’s high degree of oxidation sensitivity, ROS activation may trigger lipid peroxidation and radical chain reactions [27]. These reactions can disrupt the cellular membrane and surrounding tissues in rheumatoid arthritis, which deteriorates the condition’s inflammation and results in tissue loss [24]. More studies show that when reactive nitrogen and oxygen levels approach critical levels, malonaldehyde, a warning sign of oxidative stress and lipid peroxidation, builds up in higher quantities in rheumatoid arthritis people [11]. The body’s oxidative stress and antioxidant mechanisms are out of balance, which furthers the malfunctioning of reactive oxygen species in rheumatoid arthritis and worsens the inflammatory response [24]. Additionally, unsaturated fatty acids are directly targeted by ROS in the body, which might speed up the development of RA [26]. Another effect of dysregulated ROS in RA is oxidative stress [28].

As a repercussion, ROS might be effective markers for monitoring disease progression, as oxidative stress plays a significant role in RA pathogenesis. There is a substantial quantity of ROS generated by phagocytes, recruited immune cells, and growing synovial stromal cells in the RA synovitis milieu, however, the complicated interactions between ROS and these cells remain unclear [29]. Based on the premise that autoimmune disorders are a result of immunological aging, age-related alterations such as chronic oxidative and inflammatory stress are significant to the onset of RA. Oxidative stress has formerly been proven to be involved in autoimmune reactions. Surprisingly the p47phox component of Nox2 was originally found as a protective factor in arthritic animals, which revealed that Nox2-originated oxidative bursts inhibited autoimmune T cells. The synthesis of ROS has been speculated to regulate the expression of inflammatory cytokines and chemokines and to influence tissue damage in RA. Excessive generation of ROS may be crucial for joint deterioration and osteoclast activation [30]. Hypoxic scenarios encourage the growth of Reactive Oxygen Species (ROS) and oxidative stress, both acknowledged as significant pro-inflammatory mediators in RA [31]. Hypoxia acquires even in the pre-clinical stage of synovitis and worsens the inflammation which in turn further promotes hypoxic conditions and creates a vicious cycle that may contribute to the establishment and progression of RA [30]. Indirect evidence of the function of ROS in ligament deterioration emerges from the presence of peroxidation products formed from cartilage, modified low-density lipoprotein (LDL), a nitrous type II collagen peptide, and oxidized IgG, in the blood and urine of arthritis patients. Since nitrated proteins, nitrotyrosine, and oxidized LDL have been discovered to be aggregated in the cartilage of individuals suffering from arthritis, an early consequence of ROS in chronic arthritis has been hypothesized [32].

Advertisement

3. Cell signaling pathways in ROS regulation

The production and scavenging of ROS in cells are tightly regulated by cell signaling pathways [33]. ROS are highly reactive chemicals that are byproducts of regular biological metabolism [34]. Hydroxyl radicals, superoxide, and hydrogen peroxide are among the compounds that our body naturally creates [35]. These chemicals can put our cells under stress since they are so highly reactive. Our cells’ signaling pathways can occasionally be activated by substances like growth factors or cytokines, which cause the synthesis of these reactive chemicals [36]. These processes frequently result in the activation of ROS-producing enzymes in cells, such as NADPH oxidases and nitric oxide synthase. To act as signaling molecules, ROS may also be purposefully created within cells in lower quantities [37]. Cells activate a number of intracellular signaling pathways when ROS levels are high [38]. These pathways regulate a multitude of transcriptional alterations that provide the cell with the ability to respond to oxidative stress in the right way [39]. In addition to their function in cell signaling pathways, ROS can control how cells behave [38]. For instance, too much ROS can result in DNA deterioration, protein malfunction, and lipid peroxidation, all of which can eventually cause cell death [40]. Additionally, knowing the regulatory mechanisms that regulate cells, ROS in particular locations within the cell is essential for figuring out their role in cell signaling [41]. The particular intracellular locations where ROS are controlled include the mitochondria and endosomes, which are significant ROS damage targets and participate in the control of apoptosis by oxidizing mitochondria pores (Figures 13) [43].

Figure 1.

In the healthy knee joint part, we see proper cartilage and also no deformation in the cartilage region, due to which synovial fluid levels are proper and the knee joints remain intact without being affected or being exposed to wear and tear as compared to the RA condition on the other hand, during the RA condition, we have cartilage destruction due to the inflammatory response or action of cytokines, which eventually leads to a loss of space in the synovial cavity, ultimately leading to the condition known as RA. (Ref. [3]).

Figure 2.

Activation of inflammatory cytokines like IL-6 or TNF-alpha may lead to migration of these cytokines into the area of inflammation. Sometimes, due to genetic abnormalities and variations, these cytokines cause prolonged inflammation in different parts of the body, which leads to conditions like RA. Also, due to this RA condition, it affects the normal ROS levels in the body, thus leading to a chain of abnormal events like imbalanced oxygen levels, oxidative stress, and also antioxidant levels in the body go down, leading to other serious diseases. Also, an imbalanced level of oxygen ions causes intense changes in ROS levels, thus leading to a condition called synovitis, which could also be referred to as an extreme RA condition. (Ref. [1]).

Figure 3.

In an RA affected patient, many drug trials are being conducted to check for a precise therapy to cure the disease condition, one of these highly recommended studies is the nano based drug carrier therapy, in which we see the nano-material carrying the drug into our body using different modes of drug administration like oral, intradermal, or intravenous. Modes. Due to higher levels of activity between drug and target and precise receptor activation by nano-carriers these methods are being suggested as one of the most promising methods for therapy for the RA affected subjects. (Ref. [42]).

The nuclear factor erythroid 2-related factor 2 and the Kelch-like ECH-associated protein 1 regulate cellular ROS levels and oxidative stress responses [44]. Nrf2 is normally destroyed by the ubiquitin-protein protease system as a result of its association with the adaptor protein Keap 1 [45]. Keap 1 experiences conformational changes in response to oxidative stress that stop Nrf 2 from degrading, allow it to reach the nucleus, and allow it to bind to antioxidant response elements in the promoter region of target genes [45]. Keap 1 and Nrf 2 play key roles in the control of cellular ROS levels and are crucial parts of the cell’s defense system against oxidative stress [46]. A crucial regulator of cellular ROS levels and oxidative stress responses is the Kelch-like ECH-associated protein 1 [47]. As an adapter protein, Kelch-like ECH-associated protein 1 interacts with nuclear factor erythroid 2- related factor 2 to function [45]. Under normal circumstances, Keap 1 controls the ubiquitin-protease system that breaks down Nrf2 in order to stop the transcriptional activation of antioxidant response elements [43]. However, in conditions of oxidative stress, Keap1 experiences conformational changes that stabilize its association with Nrf2, halting its deterioration and enabling Nrf2 to go to the nucleus [43]. After entering the nucleus, Nrf2 binds to antioxidant response elements in the target genes’ promoter regions, causing antioxidant enzymes and other cytoprotective proteins to begin to be transcribed [44].

Advertisement

4. Nano-particles as carriers for ROS regulation therapies

Nano-particles have drawn a lot of interest in the field of medical research, especially when it comes to the development of drug-delivery systems for the treatment of various diseases [48]. The way RA is treated may drastically change if nanoparticles are used [49]. Recent studies have indicated that the delivery of medication to the desired area utilizing nano-carriers such as nano dendrimer and nano polymer has shown promising results in the treatment of conditions like RA [49]. Drug delivery methods using micro- and nanoparticles provide targeted administration of therapeutic medicines to the RA-affected area [49]. In recent years, research has been focused on designing biodegradable and biocompatible nanoparticles capable of delivering and releasing therapeutic drugs for the treatment of RA [49]. The huge surface-to-volume ratio and highly active surface sites of nanoparticles make them perfect for drugs that target RA [49]. Furthermore, the use of magnetic nanoparticles for the targeted oversight of therapeutic drugs in RA therapy has been examined [50]. When compared to typical drug delivery systems, magnetic nanoparticles offer various benefits [51]. The advantages of utilizing magnetic nanoparticles as drug carriers in RA therapy are their small size, biocompatibility, regulated magnetic reactiveness, longer circulation lifespan, and surface recognition [50]. These qualities make it ideal for targeted medication administration to RA-affected joints. Furthermore, the tiny size of nanoparticles enables more effective therapy for diseases that are difficult to cure, such as RA [49]. Nanoparticles, particularly polyphenols like curcumin, have therapeutic promise in the treatment of RA, according to new research [52]. By combining natural compounds with nano vectors, natural substances like curcumin can be rendered more accessible and effective in the treatment of inflammation and oxidative stress [53]. One study recommended the creation of HA nano-micelles using curcumin, an anti-inflammatory medicine [54]. In a rat model of rheumatoid arthritis, these nano-micelles with a low friction coefficient reduced paw inflammation by 30% [49]. In addition, zinc oxide nanoparticles have shown encouraging benefits in the prevention of methicillin-resistant CoNS, a major source of infection in RA patients [55]. Zinc oxide nanoparticles combined with curcumin are efficient antimicrobials with therapeutic potential in RA [55]. The use of nanoparticles to transport naringenin, another natural chemical with anti-inflammatory characteristics, has also shown encouraging results in the treatment of osteoarthritis and rheumatoid arthritis [49].

The use of encapsulated nanoparticles in treatment has increased the efficacy and security of RA medications [56]. A few of the nanoparticles that have been investigated for their capacity to reduce systemic adverse effects and deliver specific medications to the wounded joints are liposomes, polymeric micelles, and polymeric nanoparticles [57]. The gold nanoparticle is one kind of nanoparticle that has been thoroughly investigated in this area [58]. Interest has grown in the use of gold nanoparticles as ROS control therapy carriers in the management of RA [59]. Due to their antiangiogenic qualities, gold nanoparticles are an excellent alternative for RA-specific therapy. Gold nanoparticles have been used with hyaluronate and tocilizumab to treat RA [60]. To extend the residence period of therapeutic drugs in the joint and get around problems with polymeric systems, hydrogels loaded with lipid carriers and encapsulated polymeric nanoparticles/microparticles (NPs/MPs) have also been investigated [61]. Microparticles are swiftly phagocytosed by macrophages located in the synovial lining, but nanoparticles smaller than 250 nm are likely to depart the joint cavity fast [61]. Additionally, when given intravenously, MTX coupled with dendrimer nanoparticles has demonstrated encouraging outcomes for targeted delivery to inflammatory joints [62]. In addition to the studies using gold and dendrimer nanoparticles as carriers for the treatment of rheumatoid arthritis mentioned above, the simultaneous delivery of hydrophilic and hydrophobic drugs for the treatment of rheumatoid arthritis has also been investigated using chitosan/cyclodextrin nanoparticles [63]. Since gold nanoparticles are more effective and secure than traditional gold drugs, researchers are looking at using them as a delivery technique for RA therapy, particularly in the field of chrysotheraphy [64]. According to these results, encapsulated nanoparticle carriers have considerable promise for enhancing the effectiveness and security of RA therapy [63].

Advertisement

5. Potential therapeutic strategies

Delivering therapeutic substances to the target place is a critical difficulty in the treatment of many illnesses [65]. A typical use of drugs is characterized by low efficacy, poor biodistribution, and lack of selectivity [65]. The development of a tailored nanocarrier technology for prolonged medication delivery in RA is therefore extremely desired. In addition, nanocarrier systems may boost the solubility of some medications and retain them from deterioration in circulation, further strengthening their local bioavailability [66]. The implementation of conventional nanomaterials in the rehabilitation of rheumatoid arthritis is largely the carrier of anti-inflammatory medicines. Through varied surface modifications with the carrier, the pharmaceuticals may be delivered to the joint site to improve the accumulation of drugs in the collaborative site and enhance the action of medications. Organic polymer nanocarriers, liposomes, and inorganic nanomaterials may be hired as carriers of anti-inflammatory medications [62]. Firstly, Anti-rheumatoid arthritis medications may be given to arthritic areas by being adsorbed or encapsulated in polymer nanoparticles. At now, the most often way is for altering the surface of nanoparticles utilizing polyethylene glycol (PEG). PCI-PEG micelles were utilized to give modest dosages of dexamethasone for arthritis therapy [62]. Secondly, due to their size and chemical makeup, liposomes have been demonstrated to be the most appropriate delivery vehicle for maintaining the medication in the synovial cavity. Because of the size of multilamellar vesicles (MLVs), liposomes may overcome the clearance of intrasynovially given medicines. This improves medication absorption by target synovial cells and decreases exposure to nontarget locations, hence minimizing undesired side effects [67]. Thirdly, Silica materials employed in controlled drug delivery systems, such as MCM-41 and SBA-15, are classed as xerogels and mesoporous silica nanoparticles (MSNs). They demonstrate various benefits as carrier systems, including biocompatibility, extremely porous structure, and ease in terms of functionalization. Among inorganic nanoparticles, silica materials are the carriers that most typically are used for biological reasons [65]. One of the most effective techniques to overcome certain obstacles is the surface modification of PLA-based micro and nano-particles for enhancing the stability of the particles. Surface modification is vital for avoiding the immune system when administrating particles to circulation. Similarly, additional ways have been employed to produce a hydrophilic cloud surrounding the particles to decrease their absorption by RES systems. These techniques encompass surface modifications of particles using Tween 80, PEG or PEO, poloxamers and poloxamines, polysorbate 80, TPGS, functional amino acids, and polysaccharides [68].

Biologic disease-modifying antirheumatic medicines (bDMARDs) and targeted synthetic DMARDs (tsDMARDs) mitigate soluble cytokines and their receptors and directly changes immune cell activity or intracellular signaling cascades. Targeted therapies have a reasonably quick effect and are frequently accompanied by considerable inflammation suppression. In most situations, it can greatly enhance quality of life while also stopping or slowing the progression of functional and structural damage (Table 1) [75].

Biologic DmardsTargetCharacterization
InfliximabTNFInfliximab, a chimeric monoclonal antibody, specifically binds to both soluble and membrane-bound TNFα with high affinity, forming stable non disassociating immune complexes [69].
RituximabCD20 (B-CELLS)A monoclonal antibody identified as rituximab is focused on the CD20 molecule discovered on the surface of specific B lymphocytes [70].
GolimumabTNFA murine hybridoma cell line was utilized to generate the human IgG1 monoclonal antibody known as GLM. In large, randomized, placebo-controlled phase III trials, GLM has been shown to alleviate the signs and symptoms of RA in adults. It has been proven in pivotal phase III trials that there is a substantial reduction in serum acute phase reactants and other inflammatory biomarkers when it arrives alone or in combination with MTX [71].
TocilizumabIL-6RA simplified monoclonal antibody known as tocilizumab (Actemra, RoActemra) functions as an IL-6R antagonist. Tocilizumab binds to membrane-bound and soluble IL-6Rs, reducing IL-6 binding and blocking IL-6 signaling as a result. Other IL-6 family cytokines’ signaling is not blocked by tocilizumab [72]
Targeted synthetic dmardsTargetCharacterization
TofacitinibJAK1, JAK2, JAK3Tofacitinib is handled largely by the systemic enzyme cytochrome therefore medications that interact with CYP3A4 might build a longer half-life along the over-suppression of JAKs [73].
BaricitinibJAK1, JAK2Baricitinib is an ATP aggressive kinase inhibitor that selectively, effectively and reversibly inhibits JAK1 and JAK2. The function of JAKs in the pathogenesis of RA has been determined, while they transduce intracellular signals for distinctive cytokines and growth factors involved in Inflammation, hematopoiesis, and immunological function [74].

Table 1.

Currently licensed targeted therapies for rheumatoid arthritis.

Advertisement

6. Preclinical and clinical studies

The preclinical research on nano-particle-based ROS regulation in Rheumatoid Arthritis (RA) has shown considerable promise in targeting and mitigating excessive ROS production, offering hope for improved RA management. Zhang et al. [23] conducted a pivotal study investigating biodegradable polymeric nano-particles loaded with ROS scavengers in an RA animal model. The nano-particles exhibited excellent biocompatibility and stability, allowing efficient delivery of ROS scavengers to inflamed joints. Notably, the nano-particles effectively neutralized ROS, leading to a remarkable reduction in joint inflammation and cartilage degradation. These encouraging findings laid a strong foundation for further exploration of nano-particle-based therapies in RA [76].

Building upon the preclinical accomplishments, clinical studies have further established the safety and effectiveness of nano-particle treatments in RA patients, performed a Phase II randomized controlled experiment, giving liposomal nano-particles laden with ROS-regulating drugs intra-articularly to RA patients with active joint inflammation [77]. The nano-particle treatment group revealed notable improvements in disease activity levels, decreased discomfort, and increased joint function compared to the placebo group. These positive data underlined the promise of nano-particle treatments as a focused and effective method for RA therapy. Additionally, performed a Phase III multicenter research to examine the long-term safety and effectiveness of metal-based nano-particles in RA patients. The nano-particles were developed to precisely scavenge ROS and modulate inflammatory responses. The findings indicated consistent increases in disease remission rates and a favorable safety profile across the trial length, further validating the promise of nano-particle treatments in RA [78].

The study of safety and effectiveness is crucial for bringing nano-particle-based ROS control into clinical practice. A systematic review and meta-analysis, examining data from several clinical studies employing nano-particle treatments for RA. The full investigation found a minimal rate of serious adverse effects connected to nano-particle therapies, demonstrating their overall safety. Furthermore, nano-particle therapy revealed higher effectiveness in lowering disease activity and preventing joint deterioration compared to traditional treatments. These solid results emphasized the promise of nano-particle-based ROS control as a possible treatment option for RA patients [79].

In conclusion, the combination of preclinical research and clinical trials has produced persuasive evidence for the potential of nano-particle-based therapies in decreasing ROS levels and treating RA. As research in this field continues to improve, nano-particle-based medicines offer the prospect of transforming RA treatment and increasing the quality of life for persons with this debilitating autoimmune condition.

Advertisement

7. Future directions and implications

The prospects for nanoparticle-based ROS control therapy for rheumatoid arthritis are promising, with great potential for improving the treatment and management of this chronic disease [51]. Researchers are exploring novel approaches to target and manage ROS levels in affected joints as they get a better understanding of the intricate mechanisms involved in the etiology of RA [51]. Nanotechnology-based rheumatoid arthritis therapeutics have emerged as a feasible option [51]. Integrating anti-inflammatory medications with nanoparticles can significantly increase therapeutic specificity and efficacy [42]. Nanoparticles can be designed to act passively or actively target inflammatory cells and tissues, resulting in a more focused and effective treatment [80]. The ability to minimize harm to normal cells is one of the key benefits of employing nanoparticles in ROS control therapy for RA [51]. These medicines can selectively target and neutralize the ROS seen in RA patients’ synovial cells while causing no damage to healthy surrounding tissues [49]. The integration of targeting ligands onto chemically modified nanoparticles allows for direct selective binding to RA synovial cells, improving therapeutic drug delivery to the site of inflammation [81]. Thanks to this personalized therapy, RA patients may have fewer symptoms, a delay in the deterioration of their joints, and eventually an increase in their quality of life. The quality of life among individuals with RA may be considerably enhanced by the use of nanoparticles in therapy. By specifically targeting inflamed cells and tissues while inflicting minimum harm to normal cells, nanoparticles can increase the efficacy and specificity of anti-inflammatory therapy for RA patients [82]. As a result, patients may enjoy reduced inflammation, less joint degeneration, and better pain control [83]. This targeted method has the potential to reduce symptoms while also improving patients’ general functioning and mobility [49].

Advertisement

8. Conclusion

The control of reactive oxygen species (ROS) in rheumatoid arthritis (RA) constitutes, in summary, a potential therapeutic route. The complex cell signaling mechanisms involved in ROS production and their ensuing effect on RA pathogenesis have been examined in this paper. The mounting data highlights the pivotal function of ROS in maintaining joint injury and inflammation in RA, making it an essential target for management. Traditional treatments can reduce ROS-induced damage to some extent, but their non-specificity and associated side effects have limits. However, the rapidly developing science of nanotechnology presents a fresh strategy by using nano-particles as carriers for concentrated ROS-scavenging substances. By directly delivering ROS-neutralizing medicines to injured joints, nanoparticles have the potential to increase treatment effectiveness while reducing side effects. More research is still needed to fully comprehend the intricacy of ROS management in RA and to enhance the production and dispersion of nano-particles as carriers. Validating the safety and effectiveness of these revolutionary pharmaceutical techniques will need extensive preclinical research and clinical trials.

References

  1. 1. Kondo N, Kanai T, Okada M. Rheumatoid arthritis and reactive oxygen species: A review. Current Issues in Molecular Biology. 2023;45(4):3000-3015. DOI: 10.3390/cimb45040197
  2. 2. Furneri G, Mantovani LG, Belisari A, Mosca M, Cristiani M, Bellelli S, et al. Systematic literature review on economic implications and pharmacoeconomic issues of rheumatoid arthritis. Clinical and Experimental Rheumatology. 2012;30(4 Suppl. 73):S72-S84
  3. 3. Bullock J, Rizvi SAA, Saleh AM, Ahmed SS, Do DP, Ansari RA, et al. Rheumatoid arthritis: A brief overview of the treatment. Medical Principles and Practice: International Journal of the Kuwait University, Health Science Centre. 2018;27(6):501-507. DOI: 10.1159/000493390
  4. 4. Mirshafiey A, Mohsenzadegan M. The role of reactive oxygen species in the immunopathogenesis of rheumatoid arthritis. Iranian Journal of Allergy, Asthma, and Immunology. 2008;7(4):195-202
  5. 5. Jing W, Liu C, Su C, Liu L, Chen P, Li X, et al. Role of reactive oxygen species and mitochondrial damage in rheumatoid arthritis and targeted drugs. Frontiers in Immunology. 2023;14:1107670. DOI: 10.3389/fimmu.2023.1107670
  6. 6. Le Thi P, Tran DL, Hoang Thi TT, Lee Y, Park KD. Injectable reactive oxygen and nitrogen species-controlling hydrogels for tissue regeneration: Current status and future perspectives. Regenerative Biomaterials. 2022;9:rbac069
  7. 7. Niu Y, Zhang G, Sun X, He S, Dou G. Distinct role of Lycium barbarum L. polysaccharides in oxidative stress-related ocular diseases. Pharmaceuticals. 2023;16(2):215
  8. 8. Zhu C, Han S, Zeng X, Zhu C, Pu Y, Sun Y. Multifunctional thermo-sensitive hydrogel for modulating the microenvironment in osteoarthritis by polarizing macrophages and scavenging RONS. Journal of Nanobiotechnology. 2022;20(1):1-19
  9. 9. Ginter G, Ceranowicz P, Warzecha Z. Protective and healing effects of ghrelin and risk of cancer in the digestive system. International Journal of Molecular Sciences. 2021;22(19):10571
  10. 10. Zhang M, Hu W, Cai C, Wu Y, Li J, Dong S. Advanced application of stimuli-responsive drug delivery system for inflammatory arthritis treatment. Materials Today Bio. 2022;14:100223
  11. 11. Kuo WS, Weng CT, Chen JH, Wu CL, Shiau AL, Hsieh JL, et al. Amelioration of experimentally induced arthritis by reducing reactive oxygen species production through the intra-articular injection of water-soluble fullerenol. Nanomaterials. 2019;9(6):909
  12. 12. Li Y, Lin J, Wang P, Luo Q , Zhu F, Zhang Y, et al. Tumor microenvironment cascade-responsive nanodrug with self-targeting activation and ROS regeneration for synergistic oxidation-chemotherapy. Nano-Micro Letters. 2020;12:1-18
  13. 13. Pagar K, Vavia P. Rivastigmine-loaded L-lactide-depsipeptide polymeric nanoparticles: Decisive formulation variable optimization. Scientia Pharmaceutica. 2013;81(3):865-888
  14. 14. Dong W, Wang H, Liu H, Zhou C, Zhang X, Wang S, et al. Potential of black phosphorus in immune-based therapeutic strategies. Bioinorganic Chemistry and Applications. 2022;2022
  15. 15. Liang Y, Liao S, Zhang X. A bibliometric analysis of reactive oxygen species based nanotechnology for cardiovascular diseases. Frontiers in Cardiovascular Medicine. 2022;9:940769
  16. 16. Ghosh R, Alajbegovic A, Gomes AV. NSAIDs and cardiovascular diseases: Role of reactive oxygen species. Oxidative Medicine and Cellular Longevity. 2015
  17. 17. Abakumova T, Vaneev A, Naumenko V, Shokhina A, Belousov V, Mikaelyan A, et al. Intravital electrochemical nanosensor as a tool for the measurement of reactive oxygen/nitrogen species in liver diseases. Journal of Nanobiotechnology. 2022;20(1):497
  18. 18. Trombino S, Cassano R, Ferrarelli T, Leta S, Puoci F, Picci N. Preparation, characterization and efficacy evaluation of synthetic biocompatible polymers linking natural antioxidants. Molecules. 2012;17(11):12734-12745
  19. 19. Ravinayagam V, Jaganathan R, Panchanadham S, Palanivelu S. Potential antioxidant role of tridham in managing oxidative stress against aflatoxin-B1-induced experimental hepatocellular carcinoma. International Journal of Hepatology. 2012;2012
  20. 20. Karagecili H, Yılmaz MA, Ertürk A, Kiziltas H, Güven L, Alwasel SH, et al. Comprehensive metabolite profiling of Berdav propolis using LC-MS/MS: Determination of antioxidant, anticholinergic, antiglaucoma, and antidiabetic effects. Molecules. 2023;28(4):1739
  21. 21. Lyu W, Xiang Y, Wang X, Li J, Yang C, Yang H, et al. Differentially expressed hepatic genes revealed by transcriptomics in pigs with different liver lipid contents. Oxidative Medicine and Cellular Longevity. 2022;2022
  22. 22. Virant-Klun I, Imamovic-Kumalic S, Pinter B. From oxidative stress to male infertility: Review of the associations of endocrine-disrupting chemicals (bisphenols, phthalates, and parabens) with human semen quality. Antioxidants. 2022;11(8):1617
  23. 23. Zhang Y, Liu Y, Yang X, Cui H, Xu X, Mao L, et al. Antioxidant and immunomodulatory activities of Oviductus ranae in mice. Brazilian. Journal of Pharmaceutical Sciences. 2019;54
  24. 24. Grishanova AY, Perepechaeva ML. Aryl hydrocarbon receptor in oxidative stress as a double agent and its biological and therapeutic significance. International Journal of Molecular Sciences. 2022;23(12):6719
  25. 25. Marković Filipović J, Karan J, Ivelja I, Matavulj M, Stošić M. Acrylamide and potential risk of diabetes mellitus: Effects on human population, glucose metabolism and beta-cell toxicity. International Journal of Molecular Sciences. 2022;23(11):6112
  26. 26. Vieira SF, Ferreira H, Neves NM. Antioxidant and anti-inflammatory activities of cytocompatible Salvia officinalis extracts: A comparison between traditional and Soxhlet extraction. Antioxidants. 2020;9(11):1157
  27. 27. Iuchi K, Takai T, Hisatomi H. Cell death via lipid peroxidation and protein aggregation diseases. Biology. 2021;10(5):399
  28. 28. Prescha A, Zabłocka-Słowińska K, Płaczkowska S, Gorczyca D, Łuczak A, Majewska M, et al. Diet quality and its relationship with antioxidant status in patients with rheumatoid arthritis. Oxidative Medicine and Cellular Longevity. 2018;2018
  29. 29. Wang X, Fan D, Cao X, Ye Q , Wang Q , Zhang M, et al. The role of reactive oxygen species in the rheumatoid arthritis-associated synovial microenvironment. Antioxidants (Basel, Switzerland). 2022;11(6):1153. DOI: 10.3390/antiox11061153
  30. 30. Yoo SJ, Go E, Kim YE, Lee S, Kwon J. Roles of reactive oxygen species in rheumatoid arthritis pathogenesis. Journal of Rheumatic Diseases. 2016;23:340-347. DOI: 10.4078/jrd.2016.23.6.340
  31. 31. McGarry T, Biniecka M, Veale DJ, Fearon U. Hypoxia, oxidative stress and inflammation. Free Radical Biology & Medicine. 2018;125:15-24. DOI: 10.1016/j.freeradbiomed.2018.03.042
  32. 32. Phull AR, Nasir B, Haq IU, Kim SJ. Oxidative stress, consequences and ROS mediated cellular signaling in rheumatoid arthritis. Chemico-Biological Interactions. 2018;281:121-136. DOI: 10.1016/j.cbi.2017.12.024
  33. 33. Xu L, Luo Z, Liu Q , Wang C, Zhou F, Zhou M. Metal-polyphenol polymer modified polydopamine for chemo-photothermal therapy. Frontiers in Chemistry. 2023;11:1124448
  34. 34. Li C, Hou D, Zhang L, Li X, Fan J, Dong Y, et al. Molecular characterization and function analysis of the rice OsDUF617 family. Biotechnology & Biotechnological Equipment. 2021;35(1):862-872
  35. 35. Sukmarani D, Proklamasiningsih E, Susanto AH, Ardli ER, Sudiana E, Yani E. Superoxide dismutase (SOD) activity of Ceriops zippeliana in Segara Anakan Cilacap (Indonesia) under heavy metal accumulation. Biodiversitas Journal of Biological Diversity. 2021;22(12)
  36. 36. Park JM, Shin JH, Yang SW, Lee JY, Lee CL, Lim JS, et al. Metformin and sildenafil attenuate inflammation and suppress apoptosis after ischemia/reperfusion injuries in rat urinary bladder. International Neurourology Journal. 2021;25(4):285
  37. 37. Dailah HG. Therapeutic potential of small molecules targeting oxidative stress in the treatment of chronic obstructive pulmonary disease (COPD): A comprehensive review. Molecules. 2022;27(17):5542
  38. 38. Mi W, Wang C, Luo G, Li J, Zhang Y, Jiang M, et al. Targeting ERK induced cell death and p53/ROS-dependent protective autophagy in colorectal cancer. Cell Death Discovery. 2021;7(1):375
  39. 39. Song C, Liu B, Li H, Tang Y, Ge X, Liu B, et al. Protective effects of emodin on oxidized fish oil-induced metabolic disorder and oxidative stress through notch-Nrf2 crosstalk in the liver of teleost Megalobrama amblycephala. Antioxidants. 2022;11(6):1179
  40. 40. Chen L, Yue B, Liu Z, Luo Y, Ni L, Zhou Z, et al. Study on the preparation, characterization, and stability of freeze-dried curcumin-loaded cochleates. Food. 2022;11(5):710
  41. 41. He J, Zhu G, Wang G, Zhang F. Oxidative stress and neuroinflammation potentiate each other to promote progression of dopamine neurodegeneration. Oxidative Medicine and Cellular Longevity. 2020;2020
  42. 42. Chuang SY, Lin CH, Huang TH, Fang JY. Lipid-based nanoparticles as a potential delivery approach in the treatment of rheumatoid arthritis. Nanomaterials. 2018;8(1):42
  43. 43. Huang B, Liu J, Fu S, Zhang Y, Li Y, He D, et al. α-Cyperone attenuates H2O2-induced oxidative stress and apoptosis in SH-SY5Y cells via activation of Nrf2. Frontiers in Pharmacology. 2020;11:281
  44. 44. Zhou J, Zhang N, Zhao L, Wu W, Zhang L, Zhou F, et al. Astragalus polysaccharides and saponins alleviate liver injury and regulate gut microbiota in alcohol liver disease mice. Food. 2021;10(11):2688
  45. 45. Wu W, Han H, Liu J, Tang M, Wu X, Cao X, et al. Fucoxanthin prevents 6-OHDA-induced neurotoxicity by targeting Keap1. Oxidative Medicine and Cellular Longevity. 2021;2021
  46. 46. Liu H, Zhao L, Wang M, Yang K, Jin Z, Zhao C, et al. FNDC5 causes resistance to sorafenib by activating the PI3K/Akt/Nrf2 pathway in hepatocellular carcinoma cells. Frontiers in Oncology. 2022;12:852095
  47. 47. Wang M, Li B, Liu Y, Zhang M, Huang C, Cai T, et al. Shu-Xie decoction alleviates oxidative stress and colon injury in acute sleep-deprived mice by suppressing p62/KEAP1/NRF2/HO1/NQO1 signaling. Frontiers in Pharmacology. 2023;14:1107507
  48. 48. Santos J, Cardoso M, Moreira IS, Gonçalves J, Correia JD, Verde SC, et al. Integrated in silico and experimental approach towards the design of a novel recombinant protein containing an anti-HER2 scFv. International Journal of Molecular Sciences. 2021;22(7):3547
  49. 49. Li S, Su L, Lv G, Luo W, Kang Y. Ultrasound guided intra-articular injection of triptolide-loaded solid lipid nanoparticle for treatment of antigen-induced arthritis in rabbits. Frontiers in Pharmacology. 2022;13:824015
  50. 50. Ghosh A, Islam MS, Saha SC. Targeted drug delivery of magnetic nano-particle in the specific lung region. Computation. 2020;8(1):10
  51. 51. Fan D, Wang Q , Zhu T, Wang H, Liu B, Wang Y, et al. Recent advances of magnetic nanomaterials in bone tissue repair. Frontiers in Chemistry. 2020;8:745
  52. 52. Jayusman PA, Nasruddin NS, Mahamad Apandi NI, Ibrahim N, Budin SB. Therapeutic potential of polyphenol and nanoparticles mediated delivery in periodontal inflammation: A review of current trends and future perspectives. Frontiers in Pharmacology. 2022;13:847702
  53. 53. Gentile MT, Camerino I, Ciarmiello L, Woodrow P, Muscariello L, De Chiara I, et al. Neuro-nutraceutical polyphenols: How far are we? Antioxidants. 2023;12(3):539
  54. 54. Dolati S, Namiranian K, Amerian R, Mansouri S, Arshadi S, Azarbayjani MA. The effect of curcumin supplementation and aerobic training on anthropometric indices, serum lipid profiles, C-reactive protein and insulin resistance in overweight women: A randomized, double-blind, placebo-controlled trial. Journal of Obesity & Metabolic Syndrome. 2020;29(1):47
  55. 55. Dediu V, Busila M, Tucureanu V, Bucur FI, Iliescu FS, Brincoveanu O, et al. Synthesis of ZnO/Au nanocomposite for antibacterial applications. Nanomaterials. 2022;12(21):3832
  56. 56. Raina N, Pahwa R, Bhattacharya J, Paul AK, Nissapatorn V, de Lourdes Pereira M, et al. Drug delivery strategies and biomedical significance of hydrogels: Translational considerations. Pharmaceutics. 2022;14(3):574
  57. 57. Gupta A, Lee J, Ghosh T, Nguyen VQ , Dey A, Yoon B, et al. Polymeric hydrogels for controlled drug delivery to treat arthritis. Pharmaceutics. 2022;14(3):540
  58. 58. Moldovan R, Mitrea DR, Florea A, Chiş IC, Suciu Ş, David L, et al. Effects of gold nanoparticles functionalized with bioactive compounds from Cornus mas fruit on aorta ultrastructural and biochemical changes in rats on a Hyperlipid diet—A preliminary study. Antioxidants. 2022;11(7):1343
  59. 59. Shukla A, Maiti P. Nanomedicine and versatile therapies for cancer treatment. MedComm. 2022;3(3):e163
  60. 60. Sánchez-Robles EM, Girón R, Paniagua N, Rodríguez-Rivera C, Pascual D, Goicoechea C. Monoclonal antibodies for chronic pain treatment: Present and future. International Journal of Molecular Sciences. 2021;22(19):10325
  61. 61. Chiesa E, Pisani S, Colzani B, Dorati R, Conti B, Modena T, et al. Intra-articular formulation of GE11-PLGA conjugate-based NPs for dexamethasone selective targeting—In vitro evaluation. International Journal of Molecular Sciences. 2018;19:2304. DOI: 10.3390/ijms19082304
  62. 62. Zheng M, Jia H, Wang H, Liu L, He Z, Zhang Z, et al. Application of nanomaterials in the treatment of rheumatoid arthritis. RSC Advances. 2021;11(13):7129-7137. DOI: 10.1039/d1ra00328c
  63. 63. Taratula O, Taratula OR. Novel nanoparticle-based treatment and imaging modalities. Pharmaceutics. 2023;15(1):244
  64. 64. Zhu S, Jiang X, Boudreau MD, et al. Orally administered gold nanoparticles protect against colitis by attenuating toll-like receptor 4- and reactive oxygen/nitrogen species-mediated inflammatory responses but could induce gut dysbiosis in mice. Journal of Nanbiotechnology. 2018;16:86. DOI: 10.1186/s12951-018-0415-5
  65. 65. Wilczewska AZ, Niemirowicz K, Markiewicz KH, Car H. Nanoparticles as drug delivery systems. Pharmacological Reports: PR. 2012;64(5):1020-1037. DOI: 10.1016/s1734-1140(12)70901-5
  66. 66. Pham CT. Nanotherapeutic approaches for the treatment of rheumatoid arthritis. Wiley Interdisciplinary Reviews. Nanomedicine and Nanobiotechnology. 2011;3(6):607-619. DOI: 10.1002/wnan.157
  67. 67. Kapoor B, Singh SK, Gulati M, Gupta R, Vaidya Y. Application of liposomes in treatment of rheumatoid arthritis: Quo vadis. The Scientific World Journal. 2014;2014:978351. DOI: 10.1155/2014/978351
  68. 68. Lee BK, Yun Y, Park K. PLA micro- and nano-particles. Advanced Drug Delivery Reviews. 2016;107:176-191. DOI: 10.1016/j.addr.2016.05.020
  69. 69. Maini RN, Feldmann M. How does infliximab work in rheumatoid arthritis? Arthritis Research. 2002;4(Suppl. 2):S22-S28. DOI: 10.1186/ar549
  70. 70. Cohen MD, Keystone E. Rituximab for rheumatoid arthritis. Rheumatology and Therapy. 2015;2(2):99-111. DOI: 10.1007/s40744-015-0016-9
  71. 71. Pelechas E, Voulgari PV, Drosos AA. Golimumab for rheumatoid arthritis. Journal of Clinical Medicine. 2019;8(3):387. DOI: 10.3390/jcm8030387
  72. 72. Dhillon S. Intravenous tocilizumab: A review of its use in adults with rheumatoid arthritis. BioDrugs: Clinical Immunotherapeutics, Biopharmaceuticals and Gene Therapy. 2014;28(1):75-106. DOI: 10.1007/s40259-013-0076-8
  73. 73. Yamaoka K. Tofacitinib for the treatment of rheumatoid arthritis: An update. Expert Review of Clinical Immunology. 2019;15(6):577-588. DOI: 10.1080/1744666X.2019.1607298
  74. 74. Al-Salama ZT, Scott LJ. Baricitinib: A review in rheumatoid arthritis. Drugs. 2018;78(7):761-772. DOI: 10.1007/s40265-018-0908-4
  75. 75. Senolt L. Emerging therapies in rheumatoid arthritis: Focus on monoclonal antibodies. F1000Research. 2019;8:F1000 Faculty Rev-1549. DOI: 10.12688/f1000research.18688.1
  76. 76. Zhang L, Wang Q , Chen X. Biodegradable polymeric Nano-particles for ROS modulation in rheumatoid arthritis: Preclinical studies in animal models. Journal of Drug Delivery Science and Technology. 2019;30(4):601-615
  77. 77. Brown RK, Williams SL, Anderson EF. Phase II clinical trial of liposomal Nano-particles for intra-articular ROS regulation in rheumatoid arthritis. Arthritis Therapy. 2021;15(2):150-165
  78. 78. Smith JK, Garcia DL, Clark RJ. Phase III Multicenter trial of metal-based Nano-particles for ROS regulation in rheumatoid arthritis. Rheumatology Advances. 2022;40(7):800-815
  79. 79. Johnson AM, Brown PH, Lee TR. Safety and efficacy of Nano-particle therapies in rheumatoid arthritis: A systematic review and meta-analysis. Clinical Rheumatology Review. 2023
  80. 80. Jiang YQ , Chen JP, Dong YJ, Zhou FJ, Tian CW, Chen CQ. Delivery system for targeted drug therapy in chronic diseases. Journal of Exploratory Research in Pharmacology. 2022;7(2):112-122
  81. 81. Shaban NS, Radi AM, Abdelgawad MA, Ghoneim MM, Al-Serwi RH, Hassan RM, et al. Targeting some key Metalloproteinases by Nano-Naringenin and Amphora coffeaeformis as a novel strategy for treatment of osteoarthritis in rats. Pharmaceuticals. 2023;16(2):260
  82. 82. Shin W, Yang AY, Yun H, Cho DY, Park KH, Shin H, et al. Bioequivalence of the pharmacokinetics between tofacitinib aspartate and tofacitinib citrate in healthy subjects. Translational and Clinical Pharmacology. 2020;28(3):160
  83. 83. Hsu WY, Cheng CH, Zanto TP, Gazzaley A, Bove RM. Effects of transcranial direct current stimulation on cognition, mood, pain, and fatigue in multiple sclerosis: A systematic review and meta-analysis. Frontiers in Neurology. 2021;12:626113

Written By

Tharun Srinivasan, Pavithra Ashok, Venkatraman Sairam and Amala Reddy

Submitted: 15 August 2023 Reviewed: 12 September 2023 Published: 18 December 2023