ArticlePDF AvailableLiterature Review

Comprehensive insights into rheumatoid arthritis: Pathophysiology, current therapies and herbal alternatives for effective disease management

Authors:

Abstract

Rheumatoid arthritis is a chronic autoimmune inflammatory disease characterized by immune response overexpression, causing pain and swelling in the synovial joints. This condition is caused by auto-reactive antibodies that attack self-antigens due to their incapacity to distinguish between self and foreign molecules. Dysregulated activity within numerous signalling and immunological pathways supports the disease's development and progression, elevating its complexity. While current treatments provide some alleviation, their effectiveness is accompanied by a variety of adverse effects that are inherent in conventional medications. As a result, there is a deep-rooted necessity to investigate alternate therapeutic strategies capable of neutralizing these disadvantages. Medicinal herbs display a variety of potent bioactive phytochemicals that are effective in the complementary management of disease, thus generating an enormous potency for the researchers to delve deep into the development of novel phytomedicine against autoimmune diseases, although additional evidence and understanding are required in terms of their efficacy and pharmacodynamic mechanisms. This literature-based review highlights the dysregulation of immune tolerance in rheumatoid arthritis, analyses the pathophysiology, elucidates relevant signalling pathways involved, evaluates present and future therapy options and underscores the therapeutic attributes of a diverse array of medicinal herbs in addressing this severe disease.
REVIEW
Comprehensive insights into rheumatoid arthritis:
Pathophysiology, current therapies and herbal alternatives
for effective disease management
Amrita Chatterjee
1
| Monisha Jayaprakasan
1
| Anirban Kr Chakrabarty
2
|
Naga Rajiv Lakkaniga
1
| Bibhuti Nath Bhatt
2
| Dipankar Banerjee
2
|
Avinash Narwaria
2
| Chandra Kant Katiyar
2
| Sunil Kumar Dubey
2
1
Department of Chemistry and Chemical Biology, Indian Institute of Technology (Indian School of Mines), Dhanbad, India
2
R&D Healthcare Division, Emami Ltd, Kolkata, India
Correspondence
Sunil Kumar Dubey, R&D Healthcare Division,
Emami Ltd, Kolkata 700056, India.
Email: sunilbit2014@gmail.com
Funding information
Science and Engineering Research Board,
Grant/Award Number: SRG/2022/000091
Abstract
Rheumatoid arthritis is a chronic autoimmune inflammatory disease characterized by
immune response overexpression, causing pain and swelling in the synovial joints.
This condition is caused by auto-reactive antibodies that attack self-antigens due to
their incapacity to distinguish between self and foreign molecules. Dysregulated
activity within numerous signalling and immunological pathways supports the dis-
ease's development and progression, elevating its complexity. While current treat-
ments provide some alleviation, their effectiveness is accompanied by a variety of
adverse effects that are inherent in conventional medications. As a result, there is a
deep-rooted necessity to investigate alternate therapeutic strategies capable of neu-
tralizing these disadvantages. Medicinal herbs display a variety of potent bioactive
phytochemicals that are effective in the complementary management of disease, thus
generating an enormous potency for the researchers to delve deep into the develop-
ment of novel phytomedicine against autoimmune diseases, although additional
evidence and understanding are required in terms of their efficacy and pharmacody-
namic mechanisms. This literature-based review highlights the dysregulation of
immune tolerance in rheumatoid arthritis, analyses the pathophysiology, elucidates
relevant signalling pathways involved, evaluates present and future therapy options
and underscores the therapeutic attributes of a diverse array of medicinal herbs in
addressing this severe disease.
Abbreviations: ACPA, anti-citrullinated protein antibody; APC, antigen-presenting cells; BCR, B-cell receptor; bDMARDs, biological DMARDs; CAR-T cell, chimeric antigen receptor-T cell; CD,
cluster of differentiation; CIA, collagen induced arthritis; COX, cyclooxygenase; CRP, C-reactive protein; CSF, colony stimulating factor; CTLA, cytotoxic t lymphocyte antigen; DC, dendritic cells;
DCM, dichloromethane; DMARDs, disease-modifying anti-rheumatic drug; ECM, extracellular matrix; ERK, extracellular signal-regulated kinase; FLS, fibroblast-like synoviocytes; FOXP3,
forkhead box P3; Fz, frizzled; GFR, growth factor receptor; GMSCs, gingiva-derived MSCs; HLA, human leukocyte antigen; IDO, indoleamine 2,3-dioxygenase; IFN, interferon; Ig,
immunoglobulin; IKK, inhibitor of κB Kinase; IκB, inhibitor of kappa-B; IL, interleukin; JAK, Janus kinase; LPS, lipopolysaccharide; LRP, low-density lipoprotein receptor-related protein; MAPK,
mitogen-activated protein kinase; MDSCs, myeloid derived suppressor cells; MEK, map kinase kinase; MEKK, MAP kinase kinase kinase; MHC, major histocompatibility complex; MMP, matrix-
metalloproteases; MMPs, metalloproteinases; mRNA, messenger RNA; MSC, mesenchymal stem cell; mTOR, mammalian target of rapamycin; NIK, NF-κB inducing kinase; NSAIDs, non-steroidal
anti-inflammatory drugs; PAD, peptidylarginine deiminase; PG, prostaglandin; PI3K, phosphatidylinositol 3-kinase; PIP, plasma membrane intrinsic protein; PPAD, Porphyromonas gingivalis PAD;
PTPN, non-receptor protein tyrosine phosphatases; RA, rheumatoid arthritis; RANK, receptor activator of nuclear factorkappa beta; RF, rheumatoid factor; RNA, ribonucleic acid; sDMARDs,
synthetic DMARDs; SE, shared epitope; STAT, signal transducer and activator of transcription; T-reg, T-regulatory; TCF, transcription factor; TCR, T cell receptor; TGF, transforming growth
factor; Th, T-helper; TLR, toll-like receptor; TNF, tumour necrosis factor; TX, thromboxane; TYK, tyrosine kinase; VEGF, vascular endothelial growth factor; Wnt, Wingless-related integration site.
Amrita Chatterjee, Monisha Jayaprakasan, and Anirban Kr Chakrabarty contributed equally to this study.
Received: 1 November 2023 Revised: 14 February 2024 Accepted: 1 March 2024
DOI: 10.1002/ptr.8187
Phytotherapy Research. 2024;136. wileyonlinelibrary.com/journal/ptr © 2024 John Wiley & Sons Ltd. 1
KEYWORDS
autoimmunity, herbals, inflammation, joints, rheumatoid arthritis, tolerance
1|INTRODUCTION
Rheumatoid arthritis (RA) is a systemic (van Delft & Huizinga, 2020),
chronic, inflammatory autoimmune disease (Giannini et al., 2020)
characterized by pain and swelling in the diarthrodial joints of the
body due to symmetric bone and articular cartilage destruction, lead-
ing to permanent loss of function and deformation of the joints (Bus-
tamante et al., 2017; Felix et al., 2021). This disease manifests in the
form of synovial hyperplastic inflammation, that is, hyperproliferation
of macrophage-like synoviocytes and fibroblast-like synoviocytes
(FLS) in the synovial membrane inside the joint capsule (Masoumi
et al., 2021; Mousavi et al., 2021). This condition witnesses a large
influx of immune cells like activated neutrophils, CD4
+
T cells, B cells
and macrophages into the inflamed synovium, along with a massive
production of matrix metalloproteinases (MMPs), collagenases, serine
proteases and aggrecanases that digest and disintegrate the articular
components of the joints (Abdel-Rafei et al., 2022; Guo et al., 2018).
In 2020, the global estimated number of people suffering from RA
was 17.6 million with a prevalence of 208.8 cases per million individ-
uals worldwide. The systematic analysis also reported a death rate of
0.47 per million, showcasing a 23.8% reduction in the last 30 years
(GBD 2021 Rheumatoid arthritis collaborators, 2023). Another study
reported the global average point and period prevalence of RA as
0.51% and 0.56%, respectively (Almutairi et al., 2021).
Scientific studies have established that RA has deep-seated early
signs of immune response much before the clinical onset of the dis-
ease. The rheumatoid factor (RF) and the anti-citrullinated peptide
antibodies (ACPAs) are the two categories of antibodies, which can be
detected in patients' sera as early as 14 years before the onset of clin-
ical RA symptoms. Magnetic resonance imaging scans and synovial
biopsy of ACPA and RF-positive patients with no history of RA, some
of whom developed RA symptoms eventually, have shown regular
healthy synovium. This suggested that autoimmunological responses
precede synovial inflammation in the progression of this disease,
which is an advantage in terms of pre-diagnosis and management for
individuals at risk of developing RA (Derksen et al., 2017; Scherer
et al., 2020). Researchers have concluded that a second hitis needed
to convert autoantibody presence to clinical inflammation (Petrovská
et al., 2021). Current treatment options for RA include non-steroidal
anti-inflammatory drugs (NSAIDs) and disease-modifying anti-rheu-
matic drugs (DMARDs) in combination with steroids (immunosuppres-
sants), biologics and surgery (Radu & Bungau, 2021; Sardana
et al., 2023). Recent therapeutic advances constitute mesenchymal
stem cell (MSC) therapy (Lv et al., 2021), dendritic cell (DC) therapy
(Alahdal et al., 2021; Han et al., 2020), adenosine augmentation and
T-cell modulation (Li & Chen, 2022). These therapies work by inhibit-
ing inflammatory responses at different levels of the immunological
cascades and targeting different immune cells involved in the
progression of the disease for instance pro-inflammatory cytokines,
cell surface receptors and adhesion molecules, B and T cells, chemo-
kines and cell signalling pathways. However, the success of these
treatment options is limited by adverse events, poor solubility, immu-
nogenicity (for biologics), toxicity, poor patient compliance, resistance,
and so forth. Therefore, there is an immediate need to develop more
efficient and highly specific immunomodulators (Bullock et al., 2019;
Costello et al., 2019). Anti-inflammatory and anti-arthritic herbs have
been used to treat various arthritis ailments for eternity. Over time,
mounting evidence highlights numerous herbs possessing anti-inflam-
matory potential, urging the scientific community to delve deeper into
this field (Ghasemian et al., 2016; Wang, Chen, et al., 2021). There are
several studies of the anti-rheumatic effects of East Asian herbal med-
icine showing anti-inflammatory activity comparable with current
drugs (Jo et al., 2022). The attention of the scientific community has
shifted from modern small-molecule drugs back to herb-derived medi-
cines in recent decades. Researchers are diving deep into phytochemi-
cals that can serve as safer, non-toxic, free from synthetic
constituents and efficiently soluble entities that can show effective
inhibition of pathogenic mechanisms of RA. It is high time that the
focus of pharmaceuticals is transitioned towards phytopharmaceutical
entities to establish non-invasive but efficacious therapeutic
regimens.
In this review, we have explored the mechanisms of the body's
immune tolerance. In addition, we've examined the breakdown of tol-
erance in the case of RA. Next, we have listed the signalling pathways
involved in the progression of RA, which are responsible for chronic
inflammation and damage to the synovial joints. Moreover, we've pro-
vided an overview of current treatment approaches for the manage-
ment of RA, followed by a discussion on emerging therapeutic
strategies that show promise for more effective management of the
disease. Finally, we've mentioned herbal sources and their potential
anti-inflammatory role in modulating immune responses and alleviat-
ing RA symptoms. This comprehensive perspective aims to promote
the utilisation of natural effective therapeutic sources towards
addressing this disease.
1.1 |Risk factors for RA
1.1.1 | Genetic factors
The human leukocyte antigen (HLA) allele plays a role in antigen pre-
sentation by antigen-presenting cells (APCs). This allele is found
within the major histocompatibility complex (MHC) and is responsible
for the expression of the MHC molecules on the cell surface for cell-
to-cell immune signalling functions like antigen presentation to T-cells.
The shared epitope (SE) of the HLA-DR beta locus is a specific genetic
2CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
determinant for the ACPA phenotype of RA (Larid et al., 2021;
Wysocki et al., 2020). The peptide corresponding to the HLA/DR4/1
allele or the SE is responsible for presenting the autoantigen APC to T
lymphocytes (Jung et al., 2022; Zhuo et al., 2022). The non-HLA genes
associated with RA are constituted by the peptidylarginine deiminase
(PAD)-I4, CD28 and LIMK1 (Mathebula et al., 2022). Genetic varia-
tions, especially single nucleotide polymorphisms of non-receptor pro-
tein tyrosine phosphatases 22 (PTPN22), cytotoxic T lymphocyte
antigen 4 (CTLA4) and PADI4 genes have been associated with the
risk of developing RA (Padyukov, 2022). These are the genetic predis-
positions that dictate the chances of developing RA in most cases.
1.1.2 | Epigenetic factors
Epigenetics forms the bridge between genetically inherited
factors leading to causative roles and environmental stimulations
manifesting the disease. Although this makes comprehension of the
disease more complex, it is important to point out that these different
aspects crosstalk with each other during the development of
RA. Researchers have shown that epigenetic alterations are associ-
ated with RA through RA FLS, where these hyperplastic cells show-
case a unique deoxy ribonucleic acid hypomethylation pattern in the
early onset RA that evolves as the disease progresses. The hypo-
methylation was observed in the promoter of the C-X-C motif ligand-
12 gene and long interspersed nuclear element-1 retrotransposons,
which are normally repressed by deoxy ribonucleic acid methylation.
Histone acetylation in the nuclei of RA FLS is also abnormal due to
disharmony between acetylation enzymes like histone acetyltrans-
ferases and histone deacetylase. Micro-ribonucleic acids (micro-RNAs)
and other non-coding RNAs exhibit disorganized erratic expression in
FLS (Glant et al., 2014; Karami et al., 2020). These epigenetic modifi-
cations may determine the possibility of suffering from this chronic
disease.
1.1.3 | Gender
Autoimmune disorders portray an evident gender bias with a preva-
lence twice in women as compared to men. Pregnancy, menopause
and other hormonally critical and stressful stages make women vul-
nerable to distortions of immune regulation (Angum et al., 2020; Ngo
et al., 2014). This gender disparity in the prevalence of autoimmune
diseases is attributed to sex hormone-dependent signalling, and the
complex expression of genes encoded on the X chromosome
(Yuen, 2020). The prevalence of RA in women is approximately three
times more than that in men (Dedmon, 2020; Maranini et al., 2022).
Hormones and cytokines evidently affect bone metabolism (Bertoldo
et al., 2021). Sex hormones such as oestrogens, androgens and prolac-
tin have been associated with immune responses and rheumatic dis-
eases (Raine & Giles, 2022). Women are at a greater risk of
developing autoimmune disorders, especially between ages 40 and
60, when the female body undergoes significant menopausal
hormonal alterations (Shah et al., 2020). Menopausal women suffering
from RA are associated with an increased deterioration in joint func-
tionality, as compared to women in pre-menopausal or post-meno-
pausal age groups (Mollard et al., 2018; Ramachandran et al., 2023).
In RA, decreasing oestrogen and progesterone levels during meno-
pause might aid the pathogenesis of the disease since these hor-
mones have anti-inflammatory and protective effects on bones and
joints (Angum et al., 2020). The difference in the complex interactions
between genes, environment and hormones may cause increased dis-
ease activity in women (Carmona et al., 2023). This is how gender
plays a role in the risk of development and severity of disease
manifestation.
1.1.4 | Lifestyle-related and environmental factors
Smoking is an environmental trigger that invokes the risk of the devel-
opment of RA and is one of the leading causes of RA mortality (Gwin-
nutt et al., 2020; Ishikawa & Terao, 2020). Smoking leads to
subsequent asthma and chronic obstructive pulmonary disorder,
which increases the risk of developing RA (Ford et al., 2020). Accord-
ing to a study, there is an independent correlation between the risk of
developing RA and the presence of DRB1-SE genes in a patient's
genome as well as with patients having a smoking habit. Moreover,
there is a greater relative risk of developing RF-seropositive RA in
smokers having the SE gene by more than two times as compared to
the independent relations (Padyukov et al., 2004). A study proved that
hydroquinone, a benzene metabolite present in cigarettes deteriorates
RA conditions and worsens the chronic inflammatory conditions of
the body (Heluany et al., 2018). Along with smoking, obesity is a con-
tributing risk factor to the development of RA (De Hair et al., 2012).
Adipocytes, or fat-storing cells, secrete chemicals called adipokines,
which can upregulate pro-inflammatory mediators such as T cells, B
cells, macrophages, neutrophils, DCs and a repertoire of pro-inflam-
matory cytokines, which is why obese RA patients have more severe
conditions and altered response profiles to treatment (Poudel
et al., 2020; Tang et al., 2021). Scientists have identified obesity as an
inflammatory state and associated it with increased pain, inflamma-
tion, comorbidities and treatment failure (Kadhim, 2023). Diabetes is
also associated with the development of RA. A 51% of recent onset
RA patients and 58% of long-term RA patients developed insulin resis-
tance, which is also correlated with obesity and adipose content. Insu-
lin resistance was reported higher in RA patients than in non-RA
patients (Giles et al., 2015). Since Type 1 diabetes is also autoimmune,
it was hypothesized that the genetic determining factors of these two
diseases arising from genes like HLA-DRβ1, PTPN22, CTLA-4, inter-
leukin (IL) 23 receptor, tyrosine kinase 2 (TYK2) and T cell activation
rho GTPase activating protein may have conjoint aspects (Nicolau
et al., 2017; Theofilopoulos et al., 2017). Inhalation of notorious che-
micals like smoke, silica dust, nano-sized silica and carbon-based
nanoparticles has exhibited activation of mucosal toll-like receptors
(TLRs) that activate PAD enzymes leading to unusual citrullination,
dendritic and B cells (Guo et al., 2018). Diet has also been associated
CHATTERJEE ET AL.3
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
with RA (Rondanelli et al., 2021). Foods containing Porphyromonas gin-
givalis, high-fat content and plant-derived lectins can trigger ACPA
production (Skoczyńska & Swierkot, 2018). Emphasis on the impor-
tance of one's environment and daily habits is a concept that holds no
novelty. The discussed lifestyle-related causes may play a massive role
in the break of tolerance and subsequent deterioration of joint health
in RA. It is very important to lead a holistically healthy life to prevent
the onset of diseases such as RA.
1.1.5 | Pathogenic factors
Microbial infections like P. gingivalis, EpsteinBarr virus, Aggregatibac-
ter actinomycetemcomitans,Proteus mirabilis and mycobacteria are
known to trigger the onset of RA (Bo et al., 2020; Bourgeois
et al., 2019). These organisms are capable of encouraging peptide-
citrullination and the development of ACPAs. It is reported that
P. gingivalis synthesizes the PAD enzyme, which makes it a major
pathogen capable of inflicting the disease. P. gingivalis biofilm was pro-
nounced in RA patients suffering from periodontitis and synovial fluid
samples of RA patients also showed the presence of the microbe
(Ahmadi et al., 2023; Li et al., 2022). A correlation was found between
anti-P. gingivalis PAD (anti-PPAD) immunoglobulin G (IgG) and anti-
cyclic citrullinated peptide IgG, and among anti-PPAD, C-reactive
protein and IL-6, indicating a causative role of PPAD in RA peptide
citrullination and systemic inflammation (Shimada et al., 2016). The
pathogen that causes periodontitis, A. actinomycetemcomitans,
secretes a toxin, leukotoxin A, which can cause abnormal citrullination
of antigens in the RA-affected joint and can also alter the neutrophil
structure in the synovium (Konig et al., 2016). An imbalance in the
homeostasis of the gut microbiome can have detrimental effects on
various pro-inflammatory and immune pathways (Coradduzza
et al., 2023). Hence, pathogenic factors should be considered with
equal importance while discussing risk factors for RA.
Figure 1consolidates the risk factors of RA and the consequent
symptoms that gradually manifest after the onset of the disease.
FIGURE 1 Causes and consequences of rheumatoid arthritis (RA).
4CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
2|INTERPLAY OF TOLERANCE AND
AUTOIMMUNITY
Immunological tolerance is an intricate set of processes that facilitate
non-responsiveness to autoantigens in the body. Tolerance is classi-
fied based on location into central and peripheral tolerance (Wald-
mann, 2016). T and B cells both originate in the bone marrow, while T
cells are transported to the thymus for maturation (Brugman
et al., 2015; Owen et al., 2019), B cells remain in the marrow and tol-
erance is mainly regulated in these organs (Theofilopoulos
et al., 2017). When an antigen encounters an APC, there are three
possible fates that it can undergo: (1) non-recognition by the immune
cell if the antigen is non-immunogenic (ignorance), (2) recognition and
clonal expansion of the lymphocyte if the antigen is immunogenic
and (3) anergy which is the suppression of any immune response (neg-
ative selection; Yang et al., 2020).
It is well established that the body produces auto-reactive B and
T cells under normal conditions, but these entities are either removed
or suppressed through a variety of regulatory mechanisms like anergy
(non-responsiveness by inactivation) and deletion in both primary and
secondary lymphoid organs to render them harmless (Jumbo, 2021).
Auto-reactive B cells produce poly-reactive natural autoantibodies
that circulate in the normal human serum. These antibodies play an
important role in maintaining immunological homeostasis and mediat-
ing innate responses due to their broad specificity against antigens
(Miossec, 2022). Further, we delve into details about the two major
mechanisms of the development of immune tolerance.
2.1 |Central tolerance
In central tolerance, a collection of lymphocytes of both types: B and
T cells are generated that contain specific receptors for all the proba-
ble types of antigens that may encounter the immune system. B cell
surface receptors, or Igs and T cell receptors (TCRs), upon MHC pep-
tide presentation and co-stimulation, can respond to antigens. The
thymic training of T lymphocytes is divided into two phases. The initial
part occurs in the cortex where T cells undergo positive selection. The
T lymphocytes are constantly presented with self-antigens and
the ones that do not show any response eventually die (death by
neglectAlberti & Handel, 2021). The next phase takes place as the sur-
viving cells migrate to the medullary area of the thymus and are pre-
sented with peptideMHC complexes by thymic medullary epithelial
cells that serve as APCs (Owen et al., 2019). The immune cells that
show recognition and elicit an immune response against these self-
peptides are eliminated (negative selection; Theofilopoulos
et al., 2017).
The thymus releases both self-reacting T cells and T-regulatory
(T-reg) cells, the latter being in quantitative excess. Early research on
tolerance suggested that self-antigens were simply ignored by auto-
reactive immune complexes (Wing & Sakaguchi, 2010). The discovery
of T-reg cells brought a different aspect of tolerance into the picture
where T-reg cells actively suppressed the autoantibodies that would
otherwise respond to autoantigens in circulation. Tipping the balance
in favour of T-reg cells by suppressing T-effector cell-stimulating
cytokines can prove to be an effective strategy for the alleviation of
autoimmunity (Ni et al., 2019).
Researchers have identified a gene called autoimmune regulator
that plays an important role in medullary thymic epithelial promiscu-
ous gene expression, crucial for central tolerance. This gene can regu-
late the transcription of certain proteins that are expressed and used
for antigen presentation during thymic training (Shevyrev et al., 2022;
Theofilopoulos et al., 2017; Ucar & Rattay, 2015).
B cell central tolerance comprises apoptosis of B cells that show
reactivity towards self-antigens in the bone marrow or modification
of the B-cell receptor (BCR) to render it non-reactive towards the
self-peptide, a process known as receptor editing (Castañeda
et al., 2021). Tolerance occurs in two phases: passive tolerance that
comprises anergy of autoreactive B lymphocytes and active toler-
ance that consists of CD4
+
T cell-mediated suppression of autoreac-
tive B lymphocytes. B cell development witnesses gene
rearrangement to express a repertoire of specific Igs (Meng
et al., 2023). Auto-reactive B cells are eliminated (negative selection)
in these ways during maturation in the bone marrow before they
develop into naïve B cells (Singh et al., 2021). Central B-cell tolerance
is controlled by BCR and TLR signalling. An abnormal PTPN22 gene
causes anomalous BCR signalling (Yap et al., 2018). IL-2 cytokine pro-
duction stimulates the differentiation of naïve T cells into T effector
cells. After this, they are released and exported for exhibiting periph-
eral tolerance (Kumar et al., 2018). Central tolerance is thus the pri-
mary process undertaken by our body to evade the occurrence of
autoimmunity.
2.2 |Peripheral tolerance
Despite the rigorous training at the thymus, some auto-reactive lym-
phocytes still bypass elimination and are released into the circulation
(Gallegos & Bevan, 2006; Theofilopoulos et al., 2017). These are kept
in check by peripheral tolerance that takes place in secondary lym-
phoid organs. Peripheral tolerance of B cells mainly occurs in two
ways: the B cells come across self-peptides but are not activated due
to the absence of activator T helper cells, or second, partially activated
B lymphocytes are excluded from the follicles (Singh et al., 2021).
There are three mechanisms of T cells exhibiting peripheral tolerance:
anergy, deletion and immune suppression. Anergy can be demon-
strated by two pathways, the first being the lack of co-stimulators
such as CD80/CD86 on the non-professional APC or CD28 on the T-
helper (Th) cell, leading to failure of T cell activation. The aftermath of
this phenomenon is that the T helper cell will not be activated even
on subsequent encounters with that specific antigen. The second
mechanism of anergy comprises CTLA-4 suppressor co-stimulation
instead of activator CD28 by the respective APC, rendering the Th
cells as merely persisting functionally inactive cells. The deletion
method is characterised by caspase-induced apoptotic Th cell death
due to over-activation by high antigen concentration, called
CHATTERJEE ET AL.5
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
activation-induced cell death, mediated by the Fas (CD95) ligand and
receptor. The final mode of peripheral tolerance, immune suppression,
is exhibited by the T-reg cells, which were initially named T-suppres-
sors (Owen et al., 2019). T-reg family of cells exist in the form of some
subtypes. The first is natural T-regs that express CD4, CD25, forkhead
box P3 (Foxp3), CTLA-4 and membrane transforming growth factor
(TGF)-β. They do not produce cytokines and show immune suppres-
sion by a cell-contact-dependent mechanism. The second type of Treg
cells is called adaptive Treg cells, which are further classified into
(1) Th3 cells that act by producing soluble TGF-β, and (2) Tr1 cells that
are induced by the presence of, and produce IL-10, and do not
express Foxp3. These adaptive Treg cells are found in the periphery
which also act through TGF-βand IL-10 (Meng et al., 2023). Peripheral
T-cells consist of naïve T cells, memory T cells and Treg cells having
different immune functions (Kumar et al., 2018). Autoreactive B cells
are removed from the B cell repertoire by undergoing deletion in the
T cell zones of the spleen and lymph nodes. Loss of B cell tolerance is
believed to occasionally result from a lack of assistance from T lym-
phocytes, especially T-reg cells (Kamradt & Mitchison, 2001; van de
Veen et al., 2016). This is how the event of peripheral tolerance works
to prevent the possibility of the development of autoimmune
conditions.
2.3 |Loss of tolerance and development of
autoimmune diseases
The tolerance-related process of auto-reactive T lymphocytes getting
deleted, inactivated or anergised, commences shortly after birth and
this is how any autoimmune conditions are prevented. These mecha-
nisms are referred to as recessive tolerance actions that alleviate the
odds of highly auto-reactive lymphocytes being circulated. However,
self-reactive lymphocytes with low affinity for autoantigens might
escape these measures and need a more dominant arsenal like Foxp3
+
T-reg cells to be silenced. Studies have shown that autoimmune dis-
eases display variations of IL-2, its receptor IL2RA, tumour necrosis
factor (TNF) alpha induced protein 3 and CTLA4, which can poten-
tially disrupt usual T-reg manufacture and activity and aggravate the
inflammatory environment (Law et al., 2014).
Scrutinized research has highlighted a subset of IL-10 and TGF-
βsecreting CD1d CD5
+
B cells called B-regulatory or B10 cells that
develop in the spleen upon IL-21 stimulation. These cells are trig-
gered by lipopolysaccharide (LPS) and TLR4, are predominantly pre-
sent in aged mice and can be activated irrespective of T cell
presence in the proximity of a diversity of BCRs, pointing towards
the activity of these cells in autoimmune circumstances. These cells
were considered to modulate T-reg cell activity as well as Th cell
polarizations (Luu et al., 2014). Despite these tolerance mechanisms
set in place, the body might overcome them to lead to autoimmu-
nity. The probable causes for this are the variable extent of intrathy-
mic deletion of autoimmune T cells and the activation of self-
reactive T cells upon interaction with pathogenic entities
(Jumbo, 2021).
2.4 |Loss of tolerance in RA
T-reg cells are vital for peripheral immune tolerance, and their dys-
function has been associated with the onset of autoimmune disorders
like RA due to the imbalance of T-reg and self-reactive T-effector
cells. One of the characteristic features of RA is the presence of RF in
the serum of the patient. This antibody is sensitive to the fragment
crystallizable region of the IgG antibodies (Kuligowska-Prusińska &
Odrowąż-Sypniewska, 2018; Yamada et al., 2005). It is known that
IgG acts as an autoantigen in this autoimmune condition. More
recently, however, the focus has shifted to citrullinated proteins as
the actual autoantigens triggering the onset of RA (Wegner
et al., 2010). According to a study, B lymphocytes have a pivotal role
in the progression of RA. B cells, demonstrate an over-expression of
HLA-DR cell surface marker (needed for auto-antigen presentation)
and CD22 (BCR co-receptor) in seropositive RA patients. The sera of
RA patients were found to contain abnormally high levels of memory
B cells, IgM and IgA antibodies (Wang et al., 2019). In RA, both central
and peripheral tolerance checkpoints are aberrant, resulting in an
excessive generation of autoreactive mature naïve B cells. This causes
T-reg cell dysregulation and the generation of polyreactive autoanti-
bodies, resulting in B cell evading suppression and apoptosis. B cells
also exacerbate T cell differentiation into memory cells (Yap
et al., 2018). However, it is not clear what causes the transition of
ACPA presence to clinically evident RA. The exact trigger for the
break of tolerance against self-antigens needs more clarity and is
surely multifaceted, but is associated with genetic, pathogenic, envi-
ronmental and lifestyle-related factors (Catrina et al., 2021; Page
et al., 2021). The mechanisms of central and peripheral tolerance of
both B and T lymphocytes are depicted in Figure 2, along with the
break of immunological tolerance and generation of autoantibodies in
RA-affected patients.
3|PATHOPHYSIOLOGY AND DISEASE
PROGRESSION OF RA
RA manifests in three consecutive stages: pre-clinical or triggering
stage RA, early clinically evidentRA and chronic establishedRA
(Singh et al., 2022). Pre-clinical RA encompasses autoimmune priming
in healthy individuals, which is characterised by the detection of auto-
antibodies in the patient's serum (Greenblatt et al., 2020). The second
stage is the maturation stage where the joints are attacked by the vast
network of mediators of the immune system, though the specificity of
the type of ACPAs towards a certain inflammatory after-effect is yet
to be clearly understood. The final targeting stage represents chronic
inflammation, and most patients do not visit the clinic before the
arrival of this stage. The nature of this phase is progressive and
exhibits irreversible destruction and deformation of the affected joints
(Holmdahl et al., 2014; Yarwood et al., 2015).
A synovial joint or a diarthrosis enables smooth movement at the
junction of two or more cartilage-covered ends of bones by encapsu-
lation of the synovial cavity with a fibrous joint capsule that is
6CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
continuous with the periosteum of the consecutive bones. The syno-
vial cavity contains synovial fluid, which lubricates the joint (Marian
et al., 2021). The synovial membrane lines the inner side of the cap-
sule and is composed of two layers: the intimal and the subintimal
layer. The intimal layer is further comprised of two types of cellsFLS
and macrophage-like synoviocytes (Kiener & Karonitsch, 2011). The
intimal stratum of the synovium contains spaces and lacks tight junc-
tions and desmosomes (Wu et al., 2021). The vascularised subintimal
layer composed of connective tissue is more tightly packed with little
room for porosity, consisting of macrophages, mast cells, adipocytes,
fibroblasts, lymphatic cells and nerve endings. FLS is responsible for
the maintenance of the synovial extracellular matrix (ECM) by secret-
ing matrix proteins such as fibronectin, collagens, tenascin, proteogly-
cans, laminins and ECM-degrading enzymes such as proteases, MMPs
and cathepsins. They also secrete hyaluronic acid as well as lubricin
for the lubrication of the joint (Nygaard & Firestein, 2020; Smith &
Wechalekar, 2015; Zhang et al., 2022).
In RA, the FLS undertake an aggressive phenotype and undergo
excessive proliferation as an inflammatory response (Masoumi
et al., 2020; Xu et al., 2019), probably because of MSCs in the syno-
vium, followed by aggregation of abnormal hyperproliferative FLS
(pannus). FLS secrete MMPs that manifest ECM destruction (Wu
et al., 2021). FLS also secrete a range of pro-inflammatory and pro-
angiogenic cytokines and chemokines, prostaglandins (PGs), cyclooxy-
genases (COXs) (Umar et al., 2012), which in turn trigger the recruit-
ment of other immune cells, such as neutrophils, macrophages and
fibroblasts, as well as B and T cells, which altogether activate destruc-
tive participants, such as osteoclasts to promote inflammation and
ultimately, joint destruction (Masoumi et al., 2020). Cartilage destruc-
tion and bone resorption take place because of the generation of
osteoclasts and chondrocytes. Under normal conditions, chondrocytes
secrete and maintain the components of the cartilaginous layer
(Prathap Kumar et al., 2020). This causes parts of the cartilage to wear
away and the bone to be exposed, causing friction, and aggravating
the pain and inflammation. The pannus infiltrates into the cartilage
and subchondral bone (Masoumi et al., 2021). Receptor activator of
nuclear factorkappa beta (RANK)-induced differentiation of osteo-
blasts into osteoclasts and consequently bone resorption is regulated
by a family of transmembrane proteins called Siglecs or Sialic acid-
binding Ig-like lectins. These are expressed by immune cells and help
in the regulation of tolerance and bone homeostasis and can prove to
be effective targets for treatment against RA (Ye et al., 2023).
This condition originates from the immunological response of
autoantibodies against proteins that are citrullinated as a part of post-
translational modifications of proteins. Citrullinated proteins are char-
acteristic of inflammation, but the phenomenon of antibodies target-
ing these proteins is highly specific to RA (Vossenaar et al., 2003). The
positively charged amino acid arginine is converted to citrulline, a neu-
tral amino acid, by a subtype of enzymes called PADs in a calcium-
dependant reaction (Curran et al., 2020; Fuhrmann et al., 2015;
Yamada et al., 2005). PADs are secreted by granulocytes and macro-
phages present in the synovium. They can be of five isoforms: PAD1,
FIGURE 2 Tolerance mechanisms in healthy individuals and loss of tolerance in rheumatoid arthritis (RA)-affected patients. APC, antigen-
presenting cells; IgG, immunoglobulin G.
CHATTERJEE ET AL.7
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
PAD2, PAD3, PAD4 and PAD6 (Fuhrmann et al., 2015; Kuligowska-
Prusińska & Odrowąż-Sypniewska, 2018). It has been established that
dysregulated citrullination is the key driving factor for triggering
RA. The targets of these ACPAs are identified as proteins like vimen-
tin, α-enolase, fibronectin, heterogeneous nuclear ribonucleoproteins,
EpsteinBarr nuclear antigen-1, type II collagen, specific histone pro-
teins and fibrin (Curran et al., 2020; Darrah & Andrade, 2018; Guo
et al., 2018). Moreover, modified IgG can also trigger the production
of autoantibodies (Islam et al., 2018). Citrulline being a non-natural
amino acid is recognised as foreign, which leads to the generation of
several ACPAs (anti-citrullinated protein antibodies) that include but
are not limited to RFs, anti-perinuclear factor or anti-keratin antibody,
anti-Sa, anti-PAD antibodies, anti-cyclic citrullinated peptide, anti-car-
bamylated protein antibodies, anti-binding Ig protein antibodies, anti-
RA33 antibodies and anti-glucose-6-phosphate isomerase antibodies,
which are reported to respond to citrullinated self-antigens. These
autoantibodies form immune complexes and stimulate a cascade of
downstream immunological responses at the inflamed synovial layer.
The RF and the ACPA are the most prominent specific markers used
for the diagnosis of RA (Kelmenson et al., 2020; Schaller et al., 2001;
Steiner & Smolen, 2002; Yamada et al., 2005; Yee et al., 2015). How-
ever, a study established that the rate of destruction of the joints of
RA-diagnosed patients was not associated with the antibodies against
citrullinated proteins (Nieto-Colonia et al., 2008).
Earlier, there was evidence to believe that the progression of RA
was driven by the MHC class II region on the 6th chromosome (Strand
et al., 2007). A recent study has modified this hypothesis and has
shown how citrullination modulates the antigen processing and MHC
presentation of the proteins that could potentially serve as autoanti-
gens. The repertoire of peptides generated from the unmodified pro-
tein forms of vimentin, fibrinogen and heterogeneous nuclear
ribonucleoprotein after proteolysis by a cocktail of cathepsins (mim-
icking MHC class II-associated proteases) was observed to be differ-
ent than the PAD-citrullinated variations of the same proteins (Curran
et al., 2023).
The inception of RA is characterised by an elevated expression of
the inflammatory cytokine and chemokine profiles in the circulation
even before the detection of autoantibodies. The profile of these sig-
nalling molecules and their receptors such as IL-1β, IL-2, IL-1RA, IL-
17, IL-4, IL-15, TNF-α, interferon α(IFN), monocyte chemoattractant
protein-1, macrophage inflammatory protein-1αand IL-2R is altered
in the system, a clear indication of immune activation and the onset of
RA. Citrullination of peptides triggers the invocation of RFs and
ACPAs, two important diagnostic markers for RA. However, there is
also a subtype of RA patients who do not show ACPAs in their sys-
temic circulation (33% of RA patients), leading to two categories of
patients: the ACPA-seropositive and the ACPA-seronegative subsets
of patients. Citrullinated antigens can activate MHC class II co-stimu-
lated T cells that activate B cells to synthesize ACPAs (Radu
et al., 2022).
These factors cumulatively form an auto-antibody storm that
largely precedes the manifestation of joint symptoms of RA. ACPAs
are the evidence of loss of immunological tolerance. The
autoantibodies bind to the sensed antigens and form anti-antibody
complexes. These immune complexes bind to fragment crystallizable
receptors present on the surface of various immune cells and activate
the complement system, bridging the gap between the innate and
adaptive immune systems (Ji et al., 2002). Consequently, immune cells
namely B lymphocytes, T lymphocytes and macrophages are activated
to elucidate immune responses (Hogarth & Pietersz, 2012). These cells
can either already be present in the synovium or can enter the joint
from the peripheral blood. Like macrophages, B and T lymphocytes
also produce a range of cytokines and chemokines enhancing the pro-
gression of RA. B and T cells co-stimulate and communicate with one
another to secrete more cytokines. Synovial macrophages and fibro-
blasts metamorphosize into tissue-invasive cells and aggravate joint
inflammation. Auto-reactive B cells attack self-antigens and worsen
joint damage. TNF-α, IL-1, IL-6, IL-12, IL-15, IL-17 and IL-18 are pro-
inflammatory cytokines (Stolina et al., 2009) secreted by natural killer
cells, neutrophils, macrophages, monocytes, fibroblasts and mast cells,
which frequently dominate the anti-inflammatory cytokines IL-4, IL-
10 and IL-13's protective effects, leading to cytokine-mediated
inflammation. IL-17, secreted by CD4
+
Th17 cells, can give rise to
MMP1 and MMP3 secreted by FLS that participate in the degenera-
tion of the ECM, increasing immune cell infiltration into the synovium.
These cytokines also induce the activation of osteoclasts by acting as
ligands for the RANK receptor on the surface of osteoblasts, causing
bone-resorption activity (Molendijk et al., 2018; Yap et al., 2018). The
FLS in the synovial layer become hyper-inflamed during the progres-
sion of RA upon signalling from pro-inflammatory cytokines, resulting
in a pannus-like hyperproliferative lining. The increased volume of
cells breaks into the narrowing joint space and invades the cartilage,
damaging it (Abdel-Rafei et al., 2022). Integrin-related processes
emerge, upregulating proliferative and infiltrative cellular pathways
like the mammalian target of rapamycin (mTOR) pathway (Bustamante
et al., 2017). Thus, the progression of the disease is an extremely intri-
cate series of events whose cumulative effects result in the painful
manifestations of the disease. The pathophysiological changes seen in
RA have been demonstrated in Figure 3.
4|SIGNALLING CASCADES AND
CELLULAR CROSSTALK IN RA
Herein, we have described the different cell signalling pathways that
are directly involved and upregulated during the progression of
RA. All these pathways have also been represented in Figure 4.
4.1 |JAKSignal transducer and activator of
transcription pathway
The Janus kinases (JAKs) belong to an intracellular protein family of
TYKs. The four subtypesJAK1/2/3 and TYK2 associate with intra-
cellular domains of transmembrane cytokine receptors (Rapalli,
Waghule, et al., 2020). Two out of the four types of JAKs get
8CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
activated at once and they recruit cytoplasm-residing signal trans-
ducer and activator of transcription (STAT) proteins (Tanaka
et al., 2022) that dimerize and get activated by phosphorylation, trans-
locate to the nucleus and regulate transcription. Different members of
the STAT family identified are STAT1/2/3/4/5a/5b/6 (Ivashkiv &
Hu, 2003). Research studies have shown that levels of native as well
as phosphorylated forms of the transcription factor (TCF) STAT3 are
substantially elevated in the cytokine-induced rheumatoid synovium,
and this phosphorylation process is JAK dependent. One of the con-
tributions of this cell-signalling pathway to RA progression is through
IL-17-induced STAT3 that elevates the production of anti-apoptotic
genes like B-cell lymphoma-2 and RA-FLS proliferation is aggravated.
JAK-inhibitors such as gefitinib, tofacitinib and baricitinib downregu-
lated STAT3 in the synoviocytes. The study went on to show that JAK
inhibitors expedite sodium nitroprusside-induced apoptosis in hyper-
plastic RA synovium, along with curtailing the thickening of the syno-
vial layer. Some JAK-inhibiting drugs also suppressed the generation
of vascular endothelial growth factor (VEGF)-A, a few MMPs and
cytokines like IL-6 and TNF, pointing to the role of the JAKSTAT
pathway in RA pathogenesis (Emori et al., 2020; Lee et al., 2013). The
JAK pathway is triggered by various pro-inflammatory cytokines and
the pathway in turn results in the production of more cytokines and
this vicious cycle contributes to the severity of the disease. This was
supported by observing the reduction in disease severity and joint
destruction on treatment with JAK inhibitors like tofacitinib and pefi-
citinib (Ishikawa et al., 2023). The profusion of cytokines that are cir-
culated during RA advancement is perceived by nociceptive receptors
and this phenomenon induces the sensation of pain. This pain is not
only due to inflammation but also indicative of the changes occurring
in the RA synovium that are partially caused by the JAKSTAT signal-
ling cascade (Simon et al., 2021). Hence, the JAKSTAT mechanism is
proved to have a primary function as the disease progresses.
4.2 |Stress-activated protein kinase/MAPK
pathway
Stress-activated protein kinase or mitogen-activated protein kinase
(MAPK) pathway is constituted by a cascade of kinases that are acti-
vated by stimuli such as growth factors, inflammatory cytokines,
FIGURE 3 Pathophysiology of rheumatoid arthritis. ACPA, anti-citrullinated peptide antibodies; APC, antigen-presenting cells; ECM,
extracellular matrix; IL, interleukin; MHC, major histocompatibility complex; MMP, matrix-metalloproteases; RF, rheumatoid factor; TCR, T cell
receptor; TGF, transforming growth factor; TNF, tumour necrosis factor.
CHATTERJEE ET AL.9
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
stress signals and G protein-coupled receptor agonists. These extra-
cellular signals stimulate the activation of MAPK kinase kinases that in
turn phosphorylate MAPK kinases, followed by phosphorylation of
different isoforms of MAPKs that facilitate the activation of certain
TCFs that mediate transcription of genes responsible for cell growth,
survival, differentiation and development (Pradhan et al., 2019). Raf,
Ras, Mos and MAP kinase kinase kinase (MEKK)1 and 4 are various
subtypes of MAPK kinase kinases that activate MAPK kinases like
MEKs. MEK1 and 2 further trigger extracellular signal-regulated
kinase 1 and 2 (ERK1 and 2), while MEK4, 7 and MEK3, 6 trigger apo-
ptotic mediators like JNK1, 2, 3 and p38 MAPKs, respectively (Morri-
son, 2012). Multiple studies have shown the connection between RA
and the MAPK signalling pathway. MEK4, MEK7 and JNK (c-Jun N-
terminal kinase) can be detected in the IL-1-induced RA synovium,
FIGURE 4 Cellular signalling pathways involved in the progression of rheumatoid arthritis. APC, antigen-presenting cells; ECM, extracellular
matrix; IFN, interferon; IKK, inhibitor of κB kinase; IL, interleukin; IκB, inhibitor of kappa-B; JAK, Janus kinase; LRP, low-density lipoprotein
receptor-related protein; MMPs, matrix metalloproteinases; RANK, receptor activator of nuclear factorkappa beta; STAT, signal transducer and
activator of transcription; TCF, transcription factor; TLR, toll-like receptor; VEGF, vascular endothelial growth factor; Wnt, Wingless-related
integration site.
10 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
along with the phosphorylated (activated) forms of MEK4 and MEK7
with the help of western blotting and immunohistochemistry using
anti-MEK antibodies. MEK4, 7 and JNK form a complex called a signa-
losome localised in the RA FLS cytoplasm, part of these complexes
moving to the FLS nucleus after cytokine stimulation (Sundarrajan
et al., 2003). There is evidence that IL-22-induced osteoclastogenesis
is mediated through the MAPK pathway by phosphorylation of the
p38 kinase. On the other hand, the anti-inflammatory cytokine IL-25
downregulated this phosphorylation induced by IL-22 (Min
et al., 2020). The MAPK pathway induces apoptosis by activating cas-
pases through MAP kinases, JNK and p38. Liquiritin, a natural flavo-
noid derived from the roots of Glycyrrhiza uralensis, an Asian plant,
caused apoptosis in IL-1βinduced RA-FLS and inhibited inflammation,
angiogenesis by downregulating phosphorylated p38 and JNK (Zhai
et al., 2019). Conclusively, the MAPK pathway is a principal process
during the pathogenesis of RA.
4.3 |NF-κB pathway
The NF-κB complex is localised in the cytoplasm, comprised of sub-
units: NF-κB1 (p50), NF-κB2 (p52), RelA (p65), RelB and c-Rel. This
complex is blocked by the inhibitor of kappa-B (IκB) family compo-
nents, which secludes the complex to the peripheral cytoplasm. The
pathway can be stimulated in two ways: canonical and non-canonical.
The canonical route encompasses stimuli-induced ubiquitinproteo-
some mediated degradation of the IκB, releasing the NF-κB complex,
which is then free to translocate into the nucleus and perform tran-
scriptional regulation (Kunnumakkara et al., 2021). It is activated at
the cell membrane through receptors like TNF receptor, IL-1 receptor
(IL1R), TLR, BCR, TCR and growth factor receptors (GFRs). The degra-
dation of IκB is triggered by its phosphorylation by the inhibitor of κB
kinase (IKK) complex, which is stimulated by different effectors like
cytokines, growth factors, stress signals and microbial fragments. The
non-canonical pathway works slightly differently. It is stimulated by
the lymphotoxin-αand β, CD40 and B cell activating factor (Ramadass
et al., 2020). This pathway is characterised by the degradation of the
precursor protein of the NF-κB2, called p100. This protein is phos-
phorylated by the NF-κB inducing kinase in association with IKKα
(a subtype of IKK) and is eventually ubiquitinylated. Then, it
undergoes further maturation by alteration of its c-terminal IκB-like
conformation, followed by the generation of a non-canonical NF-κB
complex that translocates to the nucleus (Liu et al., 2017). The NF-
κB pathway affects the transcription of genes for cytokines such as
IL-1, IL-6 and TNF-α, which are known to be upregulated in RA
patients' serum. The NF-κB cascade is prompted by the RANK ligand
receptor binding on cytokine signalling, which leads to osteoclasto-
genesis-mediated bone resorption. The RANK ligand, a member of
the TNF family, is secreted by activated T cells and is over-expressed
in RA FLS, consequently, NF-κB is hyper-activated in RA synovial tis-
sue (Ilchovska & Barrow, 2021). Therefore, the NF-κB pathway has a
predominant function during the development and advance-
ment of RA.
4.4 |mTOR/Aβt pathway
The phosphoinositide-3 kinase (PI3K)/Akt/mTOR pathway is a pivotal
signalling pathway for cellular maintenance responsible for regulating
cell growth, proliferation, differentiation, messenger RNA (mRNA)
ribosomal synthesis, angiogenesis, apoptosis and cellular homeostasis.
This pathway can be activated by a range of growth factors as well as
insulin, herceptin and glucose. The basic series of events in this path-
way comprises the growth factor-induced activation of membrane-
bound TYK, which leads to the stimulation of the PI3K through an
exchange of phosphate group with Kirsten rat sarcoma virus. PI3K is
responsible for phosphorylating plasma membrane intrinsic protein
1 to 3 (PIP2 to PIP3). This, or phosphorylated phosphoinositide-
dependent kinase-1, can stimulate Akt, producing activated mTOR,
which moves into the nucleus to trigger the transcription of growth-
related genes. mTOR is detected in the form of two complexes,
named mTORC1 and mTORC2. mTORC1 exists in combination with
regulatory-associated protein of mTOR and regulates translation initi-
ation protein 4E binding protein 1 and is also associated with ribo-
somal S6 kinase-1. Alternatively, mTORC2, together with a factor
called rapamycin-insensitive companion of mTOR, demonstrates
kinase activity. This pathway is regulated by a tumour suppressor lipid
phosphatase, phosphatase and tensin homolog, that suppresses con-
sequent phosphorylation of PIP3 (Malemud, 2015). Inhibition of the
mTOR pathway by mTOR inhibitor sirolimus (also called rapamycin,
after which the name mTOR is derived) attenuated paw swelling in
human-TNF transgenic mice, proving that mTOR is involved in inflam-
matory progression. Moreover, it downregulated the osteoclast levels,
cartilaginous destruction and bone erosion at the synovial site. Osteo-
clast survival was established to be mTOR dependent, owing to dimin-
ished mature osteoclasts when cultured in vitro in the presence of
sirolimus, M-colony stimulating factor (CSF) and the RANK ligand.
Downstream components of the mTOR cascade such as Akt, mTOR,
S6 and 4E-BP are in their active phosphorylated state in the induced
RA environment (Cejka et al., 2010). Thus, the mTOR signalling path-
way plays a major role in the disease progression of RA.
4.5 |Wingless-related integration site/β-catenin
pathway
The Wingless-related integration site (Wnt)/β-catenin intercellular sig-
nalling pathway is an evolutionarily conserved pathway that plays a
role in the survival, homeostasis, regeneration and sustenance of cells,
which is why it is responsible for the cell proliferation of FLS in the
RA synovial environment (Dinesh et al., 2020). The pathway is acti-
vated by glycoprotein ligands of the Wnt family secreted by certain
cells that are recognised by receptors on the respective receiving cells.
There are 19 Wnt ligands present in animals and respective receptor
proteins for these ligands: 10 heterodimeric frizzled (Fz) receptors,
along with 1 low-density lipoprotein receptor-related protein (LRP)-5
and LRP6 as co-receptors (Ma et al., 2020). This signalling mechanism
may undergo one of two pathways: the canonical (β-catenin-
CHATTERJEE ET AL.11
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
dependant) pathway or the non-canonical (β-catenin-independent)
pathway (Rim et al., 2022). The canonical pathway proceeds in the fol-
lowing manner: in the inactivated state, aberrant signalling is pre-
vented by phosphorylation and subsequent degradation of the main
effector protein, which is a transcriptional co-activator, β-catenin.
These receptor components do not exist in physical contact with each
other when the pathway is not active. The phosphorylation and prote-
olysis of cytoplasmic β-catenin are led by a multimeric destruction
complex comprised of axin, adenomatous polyposis coli, glycogen
synthase kinase-3βand casein kinase 1α(Lorzadeh et al., 2021). Wnt
ligand (namely Wnt1, Wnt2b, Wnt3a, Wnt10b and Wnt16) binding
facilitates dimerization of the Fz and LRP5/6 to form the Fz-LRP5/6
receptor complex, activating the pathway. This is followed by the
assembling of the destruction complex at the cell membrane, thus dis-
engaging the proteosome-dependant ubiquitylation process. Β-cate-
nin stabilises and accumulates in the cytoplasm, and consequently
regulates transcriptional activation with the help of TCF/lymphoid
enhancer-binding factor (Bian et al., 2020). β-catenin target gene acti-
vation dictates cell fate, survival, proliferation and stem cell differenti-
ation, especially bone tissue formation osteoblastic differentiation
(Sharma & Pruitt, 2020). The non-canonical Wnt cascade can be fur-
ther divided into the Wnt/planar cell polarity pathway and the Wnt/
Ca
2+
pathway. The Wnt/Ca
2+
pathway results in the discharge of
intracellular calcium and promotes calcium-dependant mechanisms,
while the Wnt/planar cell polarity pathway is responsible for the
development of cell polarity, cytoskeletal re-arrangement, cell migra-
tion and adhesion (Sánchez & Flores, 2023). Instead of LRP5/6, non-
canonical Wnt ligands (namely Wnt3a, Wnt4, Wnt5a, Wnt5b, Wnt7b,
Wnt10b and Wnt16) bind to different co-receptors like TYKs and
receptor TYK-like orphan receptor (Lojk & Marc, 2021). A study
proved that the neuron navigator 2, which is upregulated in RA
patients, aggravates the condition through the Wnt/β-catenin path-
way, by activating inflammatory mediators like IL-8, IL-1βand IL-6 at
mRNA levels (Wang, Li, et al., 2021). Wnt5a upregulates pro-inflam-
matory mediators such as IL-1β, IL-8 and IL-6 (Mahmoud et al., 2021).
Wnt5a is reported to promote migration and invasion of RA FLS and
stimulate the expression of IL-6, IL-8, C-X-C motif ligand-5, MMPs-
1,3,9 and 13 predominantly through the Wnt/Ca
2+
pathway. This
pathway also interacts with other signalling entities like p38, ERK,
AKT and glycogen synthase kinase-3βto enhance FLS invasion into
the synovium (Trillo et al., 2020). This is how the Wnt/β-catenin path-
way contributes to the aggravation of RA.
4.6 |VEGF pathway
The glycoprotein family (VEGFs) is responsible for the formation of
new blood vessels, wound healing, endothelial proliferation and
embryonic development. These dimeric proteins are produced in
fibroblasts, endothelial cells, T cells, platelets, macrophages, smooth
muscle cells and neutrophils (Mumtaz & Hussain, 2020). Comprised of
five related forms, VEGF-A, VEGF-B, VEGF-C, VEGF-D and placental
growth factor, this pathway regulates also regulates vessel
permeability and angiogenesis. These ligand isoforms form dimers
when active and bind to their respective TYK-like vascular endothelial
GFRs on the surface of endothelial cells. This dimerization and binding
process instigates a cascade of channels such as the MAPK, PI3K and
phospholipase c-γsignalling pathways through phosphorylation of
tyrosine residues in the intracellular domain of the transmembrane
receptors (Shaw et al., 2024; Wang, Bove, et al., 2020). In RA, VEGF
regulates synovial angiogenesis during the progression of RA through
the interaction between the RA-FLS and the human dermal microvas-
cular endothelial cells. The newly formed blood vessels help feed the
inflamed synovial mass, thus aggravating the proliferative process. In
addition, VEGF prevents synoviocyte apoptosis, controls osteoclast
differentiation, instigates RANKL secretion and activates IL-6 and the
JAKSTAT pathway (Van Le & Kwon, 2021). The circular RNA homeo-
domain-interacting protein kinase-3 is upregulated and plays an
important role in the pathogenesis of RA by induction of VEGF-medi-
ated angiogenesis (Zhang, Ma, et al., 2021). A research group sug-
gested that β-sitosterol, a plant-derived sterol, suppressed the
migration and proliferation of endothelial cells, thus arresting synovial
hyperplasia and joint destruction by inhibiting the VEGF signalling
pathway (Qian et al., 2022). Another study reported that a single
nucleotide polymorphism in the VEGF gene can increase the chances
of developing RA. While the allele A of VEGF was found to be harm-
ful, the occurrence of allele C was found to have a protective effect
against RA (Hussain et al., 2023). Clearly, the VEGF pathway plays a
substantial role in increasing the severity of the disease.
5|CURRENT TREATMENTS AND THEIR
LIMITATIONS
Herein, we present an overview of the conventional treatment strate-
gies and the associated adverse effects that these therapies cause.
Some commonly prescribed drugs of each therapeutic category and
their reported adverse drug reactions have been summarized in
Table 1.
5.1 |Non-steroidal anti-inflammatory drugs
NSAIDs exhibit anti-inflammatory activity through the inhibition of
COX enzymes (Willoughby et al., 2000), which exist in three isoforms:
COX-1, COX-2 and COX-3 (Chandrasekharan et al., 2002), as well as
by suppressing TNF-α, inducible nitric oxide synthase and phospholi-
pase A2 or phospholipase C. All of these membrane-bound COX
enzymes convert arachidonic acid into PGH2 via PGG2, which is
eventually transformed into PGs like PGD2, PGE2, PGF2 and PGI2
(prostacyclin) along with thromboxane TXA2, each responsible for dif-
ferent physiological responses (Dwivedi et al., 2015). While COX-1
acts as a housekeeping enzyme being synthesized under all conditions
and regulating mucosal gastroprotection, vasodilation, bronchocon-
striction and platelet accumulation, COX-2 is induced during pain,
pyrexia and inflammatory responses (Willoughby et al., 2000).
12 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Commonly prescribed NSAIDs include phenoprofen, paracetamol
(acetaminophen), ibuprofen, indomethacin, ketoprofen, ketorolac,
paracetamol and naproxen. Some NSAIDs are COX-2 selective inhibi-
tors, while others work by non-specifically inhibiting both COX
enzymes (Liu et al., 2005). Therefore, it is logically inferred that gen-
eral NSAIDs perform their expected anti-inflammatory activity by
inhibiting COX-2, and the undesirable side effects arise due preven-
tion of COX-1 activity (Kumar Vishwakarma & Negi, 2020).
An accumulation of adverse drug reactions originates from the
consumption of NSAIDs, ranging from gastrointestinal bleeding, car-
diovascular complications, thrombo-genetic risks to convulsions (Liu
et al., 2005), bronchospasm, renal disturbance and surge in haemoglo-
bin A1c in Type 2 diabetes patients. Aspirin, a commonly prescribed
NSAID, causes gastrointestinal bleeding (Dwivedi et al., 2015). The
plethora of adverse events associated with NSAIDs ranges from mild
symptoms like heartburn, digestive issues and abdominal pain to seri-
ous conditions like gastroduodenal ulcerations causing bleeding, and
perforation that may manifest into mortality. COX-1 inhibition by
NSAIDs may even suppress protective mucosal flow and cause ische-
mic damage. Over recent years, the rate of hospitalisation and treat-
ment discontinuation has spiked due to these adverse events (Davis &
Robson, 2016; Vonkeman & van de Laar, 2010).
5.2 |Disease-modifying anti-rheumatic drugs
The European Alliance of Associations for Rheumatology recommends
DMARDs as the first line of treatment upon diagnosis of RA (Smolen
et al., 2020). DMARDs are a family of drugs intended for inflammatory
diseases such as RA, psoriatic arthritis and ankylosing spondylitis;
TABLE 1 Current therapeutic options in practise for management of rheumatoid arthritis and their reported adverse events.
Treatment Options Adverse events reported References
NSAIDs Diclofenac
Celecoxib
Ibuprofen
Indomethacin
Ketorolac
Naproxen
Salsalate
Gastrointestinal bleeding and ulceration
Cardiovascular complications
Risk of thrombosis
Bronchospasm
Renal impairment
Heart burn
Davis and Robson (2016), Harirforoosh and
Jamali (2009), Jiang et al. (2012), Liu et al.
(2005), Vonkeman and van de Laar (2010)
DMARDs
(not including biological
DMARDs)
Methotrexate
Hydroxychloroquine
Sulfasalazine
Leflunomide
Azathioprine
Opportunistic infections
Anorexia
Erythema
Ultra-violet sensitivity, blurred vision
Venous thrombosis
Neurological complications
Cardiovascular difficulties
Psoriasis
Tuberculosis
Cancer
Methylenetetrahydrofolate reductase
gene polymorphism
Berkun et al. (2004), Caporali et al. (2008),
Dhir and Aggarwal (2012), Goodman
(2015b), Ramiro et al. (2014)
Corticosteroids Prednisone
Dexamethasone
Hydrocortisone
Betamethasone
Osteoporosis
Cataract
Poor bone mineral density
Cardiovascular risks: hypertension,
atheroschlerosis, myocardial infarctions,
cardiac failure
Vulnerable to infections
Psychiatric reactions
Alopecia
Diabetes mellitus, obesity
Ethgen et al. (2013), Luís et al. (2019); Mitra
(2011), Stone et al. (2021), Wang, Zhao,
et al. (2020), Warrington and Bostwick
(2006), Wilson et al. (2019)
Biologics Etanercept
Adalimumab
Infliximab
Rituximab
Anakinra
Increased susceptibility to infections
Malignancies: lymphoma and skin cancer
Goodman (2015b), Scott (2012), Smolen
et al. (2020), Strand et al. (2007), Tarp
et al. (2017), Woodrick and Ruderman
(2011)
Surgery Arthroplasty
Arthrodesis
Synovectomy
Arthroscopy
Joint infections
Deep vein thrombosis
Nerve and tissue damage
Pulmonary embolism
Ossification
Athanasiou (2022), Burn et al. (2018),
Goodman (2015a,2015b)
Abbreviations: DMARDs, disease-modifying anti-inflammatory drugs; NSAIDs, non-steroidal anti-inflammatory drugs.
CHATTERJEE ET AL.13
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
connective tissue abnormalities and some cancers. These immuno-
modulatory entities possess unique mechanisms of action, that is,
each DMARD alters immune responses through a different mechanis-
tic fashion. For example, methotrexate is a DMARD that mimics folic
acid structure that inhibits enzymes pivotal for de-novo nucleotide
biosynthesis, such as dihydrofolate reductase, thymidylate synthase,
aminoimidazole carboxamide ribonucleotide transformylase and
amido phosphoribosyltransferase. Aminoimidazole carboxamide ribo-
nucleotide transformylase has been postulated as the primary target
of methotrexate (Benjamin et al., 2022; Inoue & Yuasa, 2014).
DMARDs are broadly classified into biological DMARDs
(bDMARDs) and synthetic DMARDs (sDMARDs). bDMARDS are fur-
ther of two types: biological originator DMARDs and biosimilar
DMARDs. Similarly, sDMARDs can be subdivided into conventional
sDMARDs like methotrexate, sulfasalazine and targeted sDMARDs
which include the recently introduced Jak-inhibitor drugs (Köhler
et al., 2019). A systematic literature review revealed that adverse
effects of bDMARD treatments may range from opportunistic infec-
tions to cancer, neurological, cardiovascular and GI complications,
venous thrombosis, upregulation of hepatic enzymes and manifesta-
tion of psoriasis. sDMARD treatment also posed similar aftermaths
such as skin infections, tuberculosis, cancer, cardiovascular conditions,
GI perforation and neurological problems (Ramiro et al., 2014).
JAK inhibitors are kinase-inhibiting targeted sDMARDs capable
of inhibiting the JAKSTAT pathway, hence suppressing inflammation,
antibody generation, synovial hyperplasia and joint damage. JAK
inhibitors approved by the Food and Drug Administration for RA man-
agement are tofacitinib, upadacitinib, peficitinib, baricitinib and filgoti-
nib. Another similar molecule called ritlecitinib is currently undergoing
phase II clinical trials for this disease (Tanaka et al., 2022). These are
very potent molecules that inhibit the different JAKs and provide
effective arthritic and neuropathic pain relief within 24 h (Harrington
et al., 2020).
Adverse events associated with DMARDs resulting in cessation
of therapy include nausea, abdominal pain and rashes. Moreover,
symptoms that surfaced early within an average of 3.1 months were
sweating and anorexia while latent effects include erythema, sensitiv-
ity to the sun and blurred vision taking a period as long as
32.5 months (Aletaha et al., 2003). Treatment with methotrexate, a
common DMARD, has been associated with an increased risk of infec-
tions, especially in the urinary and respiratory tracts. Leflunomide
usage has also been reported to cause upper respiratory tract infec-
tions in 10% or more of the patient population, as well as demanding
hospitalisation due to pneumonia and tuberculosis manifestation
(Caporali et al., 2008).
5.3 |Corticosteroids
Glucocorticoids or corticosteroids are a predominant immunosuppres-
sive mode of treatment against inflammatory diseases like RA (Huovi-
nen et al., 2023). They mostly elucidate action by interacting with key
signalling receptors. It is also detected in free form or combined with
corticosteroid-binding globulin, or transcortin. The free steroid mole-
cules can passively transport across the biological membranes, and
bind to intracellular glucocorticoid receptors, and the bound forms
can affect ligand-dependant TCFs and modulate transcription, transla-
tion and other genetic modifications. The anti-inflammatory effect of
corticosteroids is mainly mediated by inhibition of cytokine, chemo-
kine and adhesion molecule production and activity (Stone
et al., 2021). Some steroids recommended in the RA treatment regi-
men are prednisone, betamethasone, dexamethasone and hydrocorti-
sone (Hecht, 2022).
Glucocorticoids projected negative effects on glucose and cortisol
metabolism, along with catabolic effects on bone growth (Huovinen
et al., 2023). Steroid use was reported to be associated with a higher
risk of decreased bone mineral density, vertebral deformity, bone frac-
ture and osteoporosis, the extent of which was dependent on collat-
eral parameters like age, steroid dose and body mass index.
Prednisone treatment was linked to increased cases of bone loss,
especially in post-menopausal women. A study inferred that women
RA patients on corticosteroid treatment had a higher chance of devel-
oping osteoporosis. Glucocorticoid treatment has also shown elevated
chances of cardiovascular risks in RF-seropositive RA patients in the
form of carotid plaques and arterial incompressibility, and these symp-
toms were not a result of RA disease manifestation. Hypertension,
myocardial infarction and cardiac failure were also associated strongly
with patients on a long-term regimen of medium steroidal doses (Eth-
gen et al., 2013).
5.4 |Biologics
Biologic agents targeted against pro-inflammatory cytokines and
mediators of immune flare-ups have revolutionised therapy for inflam-
matory disorders like RA. These specific modulators interfere with
selective niches of immune responses and thus produce a highly spe-
cific but powerful impact. The major classes of biologics currently
being recommended for RA therapy are TNF inhibitors (adalimumab,
certolizumab pegol, etanercept and infliximab), IL1R antagonists (Ana-
kinra, Canakinumab), CD30 chimeric monoclonal antibodies (brentuxi-
mab, vedotin), RANK inhibitors (denosumab), B cell CD20 inhibitors
(rituximab), T cell co-stimulation inhibitors (abatacept) and IL-6 inhibi-
tors (two available drugs, e.g., tocilizumab; Ramadass et al., 2020;
Scott, 2012). ABT-122, a bispecific Ig designed to inhibit TNF and IL-
17A, is yet to be approved and has undergone clinical trials on
patients showing insufficient response to methotrexate (Genovese
et al., 2018).
TNF-αis a vital upstream cytokine regulator having both auto-
crine and paracrine nature that in turn stimulates the release of vari-
ous other cytokines such as ILs, facilitating angiogenesis, cell adhesion
and infiltration in the RA synovium, partially mediated by the NF-ĸB
cascade (Jang et al., 2021; Lee, Lee, Kim, et al., 2008). Etanercept, a
dimeric fusion protein specific for TNF; Infliximab, a chimeric mono-
clonal antibody comprised of human constant and mouse variable
fragments and Adalimumab, another TNF targeting recombinant
14 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
human monoclonal antibody, are some examples of TNF-specific bio-
logics used in RA treatment. These medications are generally pre-
scribed to be administered in combination with conventional
DMARDs like methotrexate and have shown effective alleviation of
rheumatoid symptoms, and advancement of joint destruction with
improvement in joint mobility (Tak & Kalden, 2011). Generally, the
therapeutic targets of biological medications in inflammation are T
and B lymphocytes, as well as cytokines like TNF-α, IL-1β, IL-6 and IL-
15 (Strand et al., 2007; van Loo & Bertrand, 2023).
However, like all other modern medicinal therapeutic strategies,
biologics also pose a threat of adverse events. TNF-suppressors like
etanercept, infliximab and adalimumab increase the risk of serious
infections, making the situation more complex since TNF is a pivotal
component for tackling infections. Logically, the rate of viral and other
opportunistic infections arising in populations undergoing IL-6 and IL-
1 inhibitor treatment has shown high susceptibility to infections
owing to the functional similarity of these cytokines to TNF. Random-
ised controlled trials of some biological therapies also revealed a con-
cerning increase in the incidence of malignancies like lymphoma and
skin cancer (Woodrick & Ruderman, 2011).
5.5 |Surgery
A common surgical approach to RA is to perform arthroplasty or joint
replacement surgery. This is a complicated procedure due to eventual
comorbidities, immunosuppressive therapeutic schemes and deformi-
ties. According to a study, 58% of RA patients choose orthopaedic
surgery during their anti-rheumatic treatment, with five out of
100,000 RA patients undergoing arthroplasty (Goodman, 2015a). Sur-
gery is the last resort in terms of treatment of RA when all medica-
tions such as DMARDs, NSAIDs and biologics have failed to alleviate
the symptoms of RA (Athanasiou, 2022). The other surgical treatment
options are arthrodesis, synovectomy and soft tissue surgery. Non-
operative management by splints and supports is critical (Simmen
et al., 2008). Arthrodesis entails irreversibly fusing the bones and
removing the joint gap and is a preferred option for RA-affected
wrists (Lautenbach et al., 2013).
Patients undergoing total hip or knee arthroplasty are at an
increased risk of dislocation and prosthetic joint infection. In the case
of prosthetic introduction, bacteria can form biofilms promptly at the
boneprosthesis junction with a polysaccharide matrix that is
immune-resistant, making arthroplasty patients a high-risk group for
joint infections. However, there have been advancements in the surgi-
cal sector such as prophylactic antibiotics and laminar airflow on the
wound to prevent contamination, reducing joint infection incidences
to a low 2%2.4% (Goodman, 2015b).
6|NOVEL POTENTIAL THERAPEUTIC
STRATEGIES EMERGING AGAINST RA
New-age therapeutic interventions are surfacing with enhanced experi-
mental technologies over time. A 2022 study revealed the potential role
of a microbial dye in combination with multiple low doses of gamma
radiation exhibiting anti-inflammatory effects and reducing disease
severity in adjuvant induced arthritis rats (Abdel-Rafei et al., 2022).
Alternatively, nano-carriers have also been explored for drug delivery
against chronic inflammatory diseases (Oshi et al., 2022;Gorantlaetal.,
2020; Donthi et al., 2023). Recently, dendrimers have gained attention
for their narrow polydispersity index (Dubey et al., 2021). A dendrimer
containing aza bisphosphonate was found to specifically target mono-
cytes and osteoclasts and suppress inflammation in experimental arthri-
tis rat models (Hayder et al., 2011). Micro-RNAs are also being explored
as potential targets in different diseases, as they regulate cell prolifera-
tion, differentiation and maintenance (Singhvi et al., 2018). These
instances bring scope for newer innovative inventions against RA that
are different from conventional treatment methods. Here, we elaborate
on some research phase therapeutic approaches that show immense
promise in evolving into established treatments.
6.1 |Myeloid-derived suppressor cells-based
therapy
Myeloid derived suppressor cells (MDSCs) are an assembly of heter-
ogenous immature immunosuppressive cells of myeloid origin gener-
ated in the bone marrow. They are classified into polymorphonuclear
(or granulocytic) and monocytic MDSCs (Ben-Meir et al., 2022). Iden-
tified in the 1990s, these cells can be produced and invoked by
pro-inflammatory mediators like granulocyte-CSF, macrophage-CSF,
granulocyte-macrophage-CSF, IL-1β, and IL-6. These immunomodula-
tors can inhibit T-cell mediated immune responses regulate B-cell
activity and be upregulated in the circulation of RA patients (Li
et al., 2021). Hyper-activity of MDSCs is associated with upregulated
granulocyte-macrophage-CSF and IL-6 production. Both types of
MDSCs express surface CD11b surface markers (Tamadaho
et al., 2018). MDSCs were evidenced to have a negative correlation
with activated Th-17 cells in the peripheral blood, indicating that
MDSC might have a role in inhibiting pro-inflammatory Th-17 cells
(Jiao et al., 2013; Park & Kwok, 2022). Detected in the synovial
region, these cells have a neutrophil-like structure and are potent T
cell inhibitors and prevent T cell infiltration into the synovium. So, an
increase in MDSC levels at the site of inflammation is desired and can
be a therapeutic strategy against RA (Kurkó et al., 2014). Male DBA/1
strain of mice induced with arthritis using CFA were used to study the
behaviour of MDSCs in autoimmune arthritis. The study demon-
strated that CD11b +Gr-1 +MDSCs accumulate in murine spleen
after induction of arthritis, suppress CD4
+
T cell expansion and pro-
inflammatory cytokine release, in case of malignancies as well as auto-
immune conditions (Fujii et al., 2013). An attempt at a deeper under-
standing of the mechanistic effect of MDSCs led to the fact that
granulocytic-myeloid-derived suppressor cells release exosomes con-
taining various immunoregulatory molecules, which could decrease
immune aggravation by stimulating B cells to produce anti-inflamma-
tory cytokine IL-10, both in vitro and in vivo. This led to a decrease in
activated plasma and Th cells in the lymph nodes of collagen-induced
arthritis (CIA) mice models (Wu, Zhu, et al., 2020).
CHATTERJEE ET AL.15
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
6.2 |T-cell targeted therapy
T cells play a vital role in the advancement of RA. CD4
+
T cells pre-
dominantly invade the synovial space and aggravate inflammatory
conditions. They trigger multiple signalling pathways offering both
pro-inflammatory and anti-inflammatory responses, i.e., they have a
regulatory role in autoimmune diseases. Cytotoxic T lymphocytes are
amply detected in the RA synovium. A strategy to control the differ-
entiation and functionality of T cells can be a potential treatment for
RA, by inducing the disease-suppressing functions and inhibiting the
disease-aggravating properties (Ni et al., 2019). Cytotoxic T lympho-
cytes are amply detected in the RA synovium. Several T-cell targeted
therapies have been proposed to modulate the functionalities of T
lymphocytes in the body for a desired activity, targeting T-cell surface
markers, TCRs, co-stimulatory cell surface factors, T-cell GFRs and T-
cell-produced cytokines (Weyand & Goronzy, 2006). For example,
cytotoxic T-lymphocyte-associated antigen 4-IgG1 (CTLA4Ig), a fusion
protein, is the initial member of a new family of medications called co-
stimulation blockers that is being tested for the management of
RA. APCs' co-stimulatory cell surface molecules CD80 and CD86 are
bound by CTLA4Ig, which prevents CD28 on T cells from binding and
T-cell activation. CTLA4Ig treatment was safe, well tolerated and did
not elicit an antibody response. It demonstrated effective inhibition of
DCs and macrophages (Kremer et al., 2003). Treatment of autoim-
mune disease with tailored chimeric antigen receptor-T cells (CAR-T)
is another emerging paradigm and is similar to tumour treatment in
that it relies on focused eradication of autoantigenic antibody-produc-
ing B cells, which are identified by their sensitivity to specific autoan-
tigens, without the risk of broad immunosuppression. Although CAR-
T cell therapy has demonstrated promise in the focused treatment of
autoimmune illnesses, its application is constrained by the real-world
difficulties posed by the heterogeneity and complexity of autoimmune
diseases like RA. In order to specifically target different types of auto-
reactive B cell subsets, Zhang et al. developed a specific and modified
approach that used universal anti-fluorescein isothiocyanate CAR-T
cells in combination with fluorescein isothiocyanate-labelled RA-
immunodominant peptides. The study highlighted the promising
potential of CAR T cell therapy against RA (Zhang, Wang, et al., 2021).
6.3 |MSC therapy
MSCs are multipotent stem cells having immunomodulatory, regener-
ative and tissue-protective properties that produce large amounts of
exosomes, which have been indicated for the transport of RNAs for
therapeutic delivery mechanisms (Yang et al., 2023). HAND2-AS1, a
long non-coding anti-sense RNA, is an established tumour suppressor
in cancers. Early studies have shown how these RNA fragments are
present in small quantities in the RA FLS. Upregulation of these same
long non-coding anti-sense RNAs in RA FLS in vitro not only inacti-
vated the proliferation, infiltration and migration of the synoviocytes
but also induced their apoptosis through the impairment of NF-κB
TCF p65. MSC-derived exosome-enclosed HAND2-AS1 when
incubated with RA-FLS suppressed growth and mobility and success-
fully halted the progression of RA (Su et al., 2021). MSCs have been
explored for therapeutic activity in autoimmunity for their immuno-
suppressive characteristics (Sardana et al., 2023). They have been
comprehended to migrate to inflammatory corners and cease the
proliferation of T and B cells (Luque-Campos et al., 2019). B7-H1 is a
co-stimulator found on the surface of activated T cells, B cells, mono-
cytes, and cancer cells. A study established that B7-H1 expressing
human gingiva-derived MSCs (GMSCs) show a stronger immunosup-
pressive potential than B7-H1-low GMSCs in CIA murine models
through interaction with STAT3 signalling pathway (Wu, Xiao,
et al., 2020). The positive effect of GMSCs on autoimmune disorders
was validated by a study showing restoration of the appropriate ratio
of Th-17 cells and upregulation of T-reg cells on GMSC induction in
uveitis-induced mice (Gao et al., 2023). Another study investigated
the transfer of MSC-derived mitochondria to CD4
+
T cells and con-
cluded that mRNA expression of T cell activation and T-reg cell devel-
opment were elevated, thus making the CD25
+
Foxp3
+
T-reg
population exploitable. This made possible the alleviation of tissue
damage in mice and reduced transplantation rejection. This concept
can be utilised in repairing RA-affected joints and preventing further
damage by enhancing the activation of T-reg cells (Court et al., 2020).
6.4 |Tolerogenic DC therapy
DCs are specialized APCs whose role was initially defined by their
ability to stimulate naive T cells and initiate successful T cell
responses. Serving as sentinels of the immune system, these cells link
innate immunity to acquired immunity, patrolling the body for anti-
gens and capturing them. Moreover, it is now understood that DC
play a key role in the development and preservation of peripheral T-
cell tolerance. Tolerogenic DCs induce immunological peripheral toler-
ance by several methods, including inhibiting T cell clonal proliferation
and induction of T cell anergy, deletion of T lymphocytes and induc-
tion of Tregs (Kiss et al., 2013). There are two types of Tregs: naturally
occurring T-reg cells that emerge in the thymus and adaptive T-reg
cells that are induced in the periphery (Haribhai et al., 2009; Hilkens &
Isaacs, 2013). Bianco et al. formerly proved that DCs treated with
immunosuppressive cytokines, as well as exosomes produced from
DCs, can inhibit the onset and severity of murine CIA. Indoleamine
2,3-dioxygenase (IDO) is a tryptophan-hydrolysing enzyme that is
essential for immunological modulation and maintenance of tolerance.
T cells can be inhibited by DCs expressing functioning IDO by deplet-
ing them of critical tryptophan and/or manufacturing toxins, as well as
by producing Treg cells. The purpose of this work was to look at the
immunosuppressive effects of bone marrow-derived DCs that had
been genetically engineered to express IDO, as well as exosomes
formed from IDO-positive DCs. Study findings reveal that IDO-
expressing DCs and DC-derived exosomes are immunosuppressive
and anti-inflammatory and that they can reverse established arthritis.
Exosomes from IDO-positive DCs may thus represent a potential
therapeutic for RA (Bianco et al., 2009). Kim et al. demonstrated that
16 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
periarticular introduction of exosomes purified from bone marrow-
derived DCs transduced ex-vivo with an adenovirus expressing viral
IL-10 or bone marrow-derived DCs treated with recombinant mice IL-
10 suppressed delayed-type hypersensitivity responses in contralat-
eral joints. Furthermore, systemic administration of IL-10-treated DC-
derived exosomes suppressed the onset of CIA model in mice and
reduced the severity of arthritis. These findings imply that immature
DCs can release exosomes that are important in the inhibition of
inflammatory and autoimmune responses. As a result, DC-derived
exosomes could be a potential cell-free therapeutic option for the
management of autoimmune disorders (Kim et al., 2005). A peptide
vaccine developed to induce the generation of tolerogenic DCs in an
ongoing inflammatory condition ameliorated joint destruction and
enhanced citrulline tolerance by upregulating T-reg and IL-10 popula-
tions (Chen et al., 2021). Exosomes secreted by DCs loaded with
MHC class II molecules effectively activated specific T cells. This strat-
egy could be manipulated to engage T-reg cells for the promotion of
self-tolerance (^
Ale ´
A ne Vincent-Schneider et al., 2002). This treat-
ment strategy is an effective targeted approach to the management of
the disease, and with enough clinical support, can be.
7|HERBAL INTERVENTIONS WITH
POTENTIAL ANTI-RHEUMATIC ACTIVITY
Recently, there has been an elevated interest in substances of natural
origin for exploration of anti-arthritic therapeutic activity, owing to
the accumulating evidence regarding the adverse events related
to modern medicine (Lee et al., 2012; Manan et al., 2020; Moon
et al., 2015). Common targets of traditional herbal medicinal com-
pounds against RA are Th cells, FLS, B cells, macrophages, DCs, miR-
NAs, angiogenesis pathways, osteoclasts and so on (Wang, Chen,
et al., 2021). Hence, herbal alternatives are being revisited for phar-
macological properties. In this section, we summarize 10 anti-inflam-
matory, anti-arthritic and anti-rheumatic herbs that have shown
intense potential to be developed into treatments against RA. The
same herbs and their major phytoconstituents have been summarized
in Figure 5. The in vitro and in vivo studies done on these 10 herbs
have been summarized in Tables 2and 3, respectively.
7.1 |Salix nigra Marshall
The bark of the willow tree (genus: Salix) is a sumptuous natural
source of invaluable bioactive compounds like phenolic glycosides, fla-
vonoids and polyphenols (Mahdi, 2010). Salicylic acid (derived from
salicin), one of the oldest medicinal compounds was discovered from
this tree, which went on to become one of the major classes of anti-
inflammatory, anti-rheumatic, antipyretic and analgesic drugs (Gligori
c
et al., 2020). Apart from that they also possess glycemia and pro-
thrombin-modulating aspects (Shara & Stohs, 2015). Overall, 322 com-
pounds are identified from leaf and bark extracts of the Salix herbs
including flavonoids (flavonols, flavones, flavanones, catechins,
procyanidins, chalcones, dihydro flavonols), phenolic glycosides,
organic acids, non-phenolic glycosides, sterols, terpenes, lignans, vola-
tiles and fatty acids (Wang et al., 2014). Phenolic compounds present
are mainly caffeoylhexoses and caffeoylquinic acids. Flavanols and
procyanidins include epicatechins and epigallocatechins. Flavonols
characterized are quercetin derivatives, kaempferol pentoside and iso-
rhamnetin (Piątczak et al., 2020). Methanolic extract of Salix nigra
exhibited strong antioxidant properties by scavenging free radicals of
nitric oxide and hydrogen peroxide and lowering lipid peroxidation in
the CIA model of rats. In vivo, studies proved that rats treated with
this preparation experienced a diminished arthritis score in terms of
reduction in erythema and paw volume. The extent of disease charac-
terized by cartilage injury, immune cell infiltration, joint damage and
synovial hyperplasia was reduced in rats treated with Salix extract, as
compared to control rats treated with CIA (vehicle only). Levels of
pro-inflammatory cytokines TNF-α, IL-1βand IL-6 were lowered to a
greater extent in CIA rats treated with methanolic Salix extract as
compared to CIA rats administered with known NSAID indomethacin
(Sharma et al., 2011). Willow extracts included in the diets of CFA-
induced rats improved levels of anti-inflammatory IL-10 and antioxi-
dants superoxide dismutase and glutathione. Consequently, histopath-
ological evidence showed that rheumatoid symptoms of oxidative
stress and abnormal inflammation were alleviated (Faid, 2021). There-
fore, plant extracts of the Salix genus offer a favourable possibility for
a treatment regimen for RA.
7.2 |Boswellia serrata Roxb.
The resin of Boswellia serrata (also called Frankincense) has been tradi-
tionally used to treat chronic inflammatory disorders. The gum of Bos-
wellia contains monoterpenes, diterpenes, triterpenes, tetracyclic
triterpenic acids and four major pentacyclic triterpenic acids such as
β-boswellic acid, acetyl-β-boswellic acid, 11-keto-β-boswellic acid and
acetyl-11-keto-β-boswellic acid (Sharma et al., 2007; Siddiqui, 2011).
Essential oils derived from leaf extracts of this plant consist of
cymene, limonene, terpinolene, bornyl acetate, pinene, thujene, phel-
landrene and terpineol (Sharma et al., 2007; Sharma et al., 2009). The
gum extract of this plant shows immunomodulatory effects such as
inhibition of the complement system, NF-ĸB and MAPK pathways
(Goswami et al., 2018), pro-inflammatory cytokine IL-1, IL-2, IL-4, IL-6
and IFN-γproduction (Ammon, 2010; Eltahir et al., 2019). The gum
resin of the Boswellia plant reduces symptoms of CFA-induced RA
such as inflammation in Wistar rats. This extract consists of boswellic
acids that can suppress the production of pro-inflammatory cytokines,
along with possessing antipyretic and analgesic activities. RA symp-
toms like pain, swelling, and erythema arise mainly from inflammation
and immunological activation (Afsar et al., 2012). Boswellia extract has
shown a substantial attenuation of inflamed paw volume and arthritic
scores, as well as lowering of cartilage damage, synovial hyperproli-
feration, pannus and vascular infiltration that are characteristic fea-
tures of RA. This in vivo study went on to show weight gain after
arthritis-induced weight loss. However, this concoction failed to
CHATTERJEE ET AL.17
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
effectuate any changes in the TNF-αlevels in the rats' sera (Kumar
et al., 2019). A research study proved the anti-inflammatory property
of B. serrata extract by showing a reduction of cellular proliferation
and paw volume in rats immunized with an emulsion of complete
Freund's adjuvant and type II collagen (CIA model of arthritis). The
preparation of Boswellia extract also diminished the levels of articular
elastase and myeloperoxidase. These enzymes are responsible for
elastin protein degradation, neutrophil activity and synovial infiltration
in the joints. Additionally, the extracted mixture also inhibited lipid
oxidative damage of the cartilage, and most importantly, repressed
the generation of pro-inflammatory cytokines like IL-1β, IL-6, TNF-α,
IFN-γand PGE2, inferred from enzyme linked immunosorbent assay
analyses (Umar et al., 2014). Boswellia herbs can thus be promoted to
be considered as therapeutic alternatives for the treatment of RA.
7.3 |Curcuma longa L.
The classes of phytochemicals present in Curcuma rhizome extract are
composed of carbohydrates, proteins, starch, amino acids, steroids,
glycosides, flavonoids, alkaloids, tannins and saponins (Sawant & God-
ghate, 2013). The high performance liquid chromatography chromato-
gram of Curcuma longa rhizome extract contains peaks for caffeic acid
hexoside, curdione, coumaric acid, caffeic acid, sinapic acid, quercetin-
FIGURE 5 Representative herbs and their major phytoconstituents with potential activity against rheumatoid arthritis.
18 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 2 In vitro studies of the medicinal herbs against rheumatoid arthritis (RA).
Herbs
Part of
plant used Study objective Cell lines used Result and inference Reference
Salix nigra Marshall Bark Evaluation of the anti-inflammatory effect
of salix extract
Primary human monocytes (CD14/CD36
positive peripheral mononuclear cells)
stimulated by LPS
Inhibition of COX-2 mediated PGE2
release
Steady decrease in release of LPS
induced IL-6, TNF-αand IL-1β
Fiebich and Chrubasik
(2004)
Boswellia serrata Roxb. Leaves Investigation of anti-inflammatory activity
of Boswellia leaf extract
Human red blood cells Substantial stabilisation of Human
erythrocyte membranes indicating anti-
inflammatory activity comparable with
NSAIDs
Afsar et al. (2012)
Curcuma longa L. Rhizome Determination of anti-inflammatory
activity of turmeric extract
LPS induced RAW 264.7 Suppressed cell proliferation.
Downregulation of IFN-γ, IL-6, IL-13
and TNF-α
Lee et al. (2020)
Zingiber officinale
Roscoe
Rhizome Evaluation of the anti-inflammatory
efficacy
THP Inhibition of TNF-α, IL-1 and IL-6
Slight induction of production of anti-
inflammatory cytokines TGF-βand
IL-10
Mohammed et al.
(2019)
Piper longum L. Fruits Demonstration of anti-inflammatory
potential of Retrofractamide C, a major
phytocomponent extracted from Piper
longum
LPS-Induced J774A.1 cells Suppression of nitric oxide and PGE2
generation
Lowering of iNOS and COX2 enzyme
production
Inhibition of IL-6 and IL-1βgene
expression
Attenuation of phosphorylated ERK
and NF-κB, p65
Lim et al. (2020)
Pluchea lanceolata
(DC.) C.B.Clarke:
Leaves Evaluation of the immunosuppressive
properties
Peritonial murine macrophages opsonized
with heat killed Candida albicans
Significant reduction in phagocytic
activity of macrophages treated with
Pluchea extract
Bhagwat et al. (2010)
Commiphora wightii
(Arn.) Bhandari
Exudate Determination of the effect of
guggulsterone on LPS induced
inflammation
Murine macrophage RAW 264.7 cell line Guggulsterone inhibited mRNA
expression of TNF-αand iNOS
Guggulsterone inhibited the
phosphorylation of IκB with dose-
dependency
Zhang et al. (2016)
Paederia foetida L. Leaves Investigation of anti-inflammatory
potential
Standard COX-1, COX-2 and LOX kits 72.23 ± 4.53% inhibition of COX-1
82.84 ± 4.85% inhibition of COX-2,
which is significantly higher than
quercetin and comparable with
indomethacin
Kumar et al. (2015)
(Continues)
CHATTERJEE ET AL.19
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
3-D-galactoside, casuarinin, bisdemethoxycurcumin, curcuminol,
dimethoxycurcumin, isorhamnetin, valoneic acid bilactone, curcumin
and curcumin-O-glucuronide (Sabir et al., 2021). C. longa (turmeric) is
traditionally used for ages for its numerous activities. The main bioac-
tive component of C. longa (turmeric) rhizome, curcumin, which
imparts the yellow colour to turmeric, possesses anti-inflammatory,
antioxidant, anti-cancer and anti-arthritic properties, and has dis-
played a wide regulation of various cellular pathways and transcription
processes (Rapalli, Kaul, et al., 2020). Its anti-inflammatory activities
involve the modulation of a spectrum of cytokines, chemokines,
growth factors, adhesion components and enzymes such as COX-2.
These functions are possible by the effectuation of NF-κB, JAKSTAT
MAPK and other kinase pathways by curcumin, which also make it a
potent chemotherapeutic molecule (Goel et al., 2008; Sharma
et al., 2005). In a research study, the efficacy of curcumin and diclofe-
nac sodium were compared in treating RA, and curcumin resulted in
fewer adverse events as compared to the standard NSAID. Curcumin
proved to be a well-tolerated analgesic as compared to diclofenac,
along with bringing overall improvement in RA patients (Chandran &
Goel, 2012). There is evidence to prove that curcumin shows anti-
inflammatory effects by downregulating pro-inflammatory factors like
TNF-α, IL-1β, IL-6, 8 and 12. In a study, IL-6, IL-8 and MMP 1 and
3 production in IL-1βinduced RA FLS was restricted by the introduc-
tion of curcumin. This potent compound also curbed Th cell produc-
tion and enhanced T-reg cell differentiation during immune activation.
The only limitation associated with the use of this molecule is its low
bioavailability, and hence, nanoparticle-encapsulation was investi-
gated for this compound's better delivery at the site in question
(Waghule et al., 2020). Liposomal curcumin shows strong inhibition of
inflammatory responses and increased anti-inflammatory cytokine
production (Feng et al., 2017). Micellar encapsulation of curcumin has
also been explored for effective transport (Gou et al., 2011; Moham-
madian Haftcheshmeh & Momtazi-Borojeni, 2020). The enormous
potential of Curcuma plants makes them a strong prospect to be
developed into management options for the disease.
7.4 |Zingiber officinale Roscoe
The source of the popular edible rhizome, ginger, is the Zingiber offici-
nale plant. The Z. officinale plant contains anti-oxidant and anti-ar-
thritic potential, as suggested by in vitro and in-silico studies
(Murugesan et al., 2020). A study has shown that ginger extract can
bring anti-inflammatory impacts through inhibition of IL-1,6, nitric
oxide, PG (mainly PGE2) and leukotriene synthesis, thus, inhibiting
joint inflammation in streptococcal cell wall-induced arthritis in mice
(Funk et al., 2016). The active constituents identified were mainly
comprised of phenols (gingerols, paradols), sesquiterpenes (zingiber-
enes, zingiberols) and vitamins (thiamine, riboflavin, niacin, pyridoxine,
etc.) (Al-Nahain et al., 2014). Z. officinale juice mainly contains three
gingerols: 4-gingerol, 6-gingediol and 6-gingerol (Bekkouch
et al., 2022). The fresh ginger rhizome is comprised of phytoconstitu-
ents of two categories: volatile and non-volatile compounds. Volatile
TABLE 2 (Continued)
Herbs
Part of
plant used Study objective Cell lines used Result and inference Reference
Vitex negundo L. Seeds Determination of anti-inflammatory and
anti-osteoporotic properties
LPS stimulated RAW 264.7 macrophages
and rat osteoblast-like UMR106 cells
Strong inhibition of nitric oxide
production through interference with
nitric oxide synthase protein
expression in LPS-stimulated
RAW264.7 cells comparable to
indomethacin
11.4% inhibitory effect of osteoclastic
trate-resistant acid phosphatase
activity in UMR106 cells
Zheng, Zhang, et al.
(2014)
Ricinus communis L. Leaves Assessment of anti-inflammatory activities LPS stimulated RAW 264.7 macrophage
cell lines
Reduction of free radicals (ROS) in LPS
stimulated cells, which will lead to
reduction in inflammation
Nemudzivhadi and
Masoko (2014)
Abbreviations: CD, cluster of differentiation; COX, cyclooxygenase; ENA, epithelial cell-derived neutrophil activating peptide; ERK, extracellular signal-regulated kinase; IL, interleukin; iNOS, inducible nitric oxide
synthase; LOX, lipoxygenase; LPS, lipopolysaccharide; MMP, metalloproteinases; MTT, 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide; NF-ĸB, nuclear factor kappa-light-chain-enhancer of
activated B cells; PG: prostaglandin; RA-FLS, rheumatoid arthritis fibroblast-like synoviocytes; RANTES, regulated upon activation, normal t cell expressed and presumably secreted; ROS, reactive oxygen
species; TNF, tumour necrosis factor.
20 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 3 In vivo studies of medicinal herbs against rheumatoid arthritis (RA).
Herb species
Part of
plant used Study objective Animal model Results Reference
Salix nigra Marshall Leaves Exploration of treatment effect of
willow leaves in rheumatoid arthritis
Sprague Dawely Rats with Collagen
Induced Arthritis (administration of
collagen emulsified with CFA)
Increased antioxidant levels of
glutathione and superoxide
dismutase, thus reducing oxidative
stress
Decreased levels of pro-
inflammatory TNF-α, and increased
anti-inflammatory IL-10 levels
Thinning of inflamed synovium,
reduced inflammatory lesions on
joint
Faid (2021)
Boswellia serrata
Roxb.
Exudate (gum
resin extract)
Evaluation of anti-inflammatory
property
Adult female Lewis rats with CFA
induced arthritis
Significant reduction of rat paw
oedema
Decreased arthritic score
Decreased IL-17 levels
Increased anti-oxidant capacity of
blood
St-Pierre et al. (2018)
Curcuma longa L. Rhizome Evaluation of antiarthritic efficiency of
turmeric extract
Adjuvant induced arthritis model in
Male Wistar Rats
Reduced joint swelling by 41.3%
Diminished serum levels of IL-17
Reduced synovial membrane and
leukocyte infiltration
Brad
unaitėet al. (2020)
Zingiber officinale
Roscoe
Rhizome Determination of anti-inflammatory
efficacy of ginger extract
CIA model in Female Wistar albino
rats
Reduced mean arthritis scores
Lowered serum levels of IL-6, IL-17
and TNF-α
Downregulated NF-κB and COX-2
levels
Decreased inflammatory
infiltration, synovial hyperplasia
Improvement in cartilage and bone
destruction
Öz et al. (2021)
Piper longum L. Fruit Investigation of rate of alteration of
RA progression by Piperlongumine
CIA induced Male DBA/1 mice Piperlongumine interfered with
inflammatory cell infiltration into
synovium, cartilage damage, pro-
inflammatory cytokine production,
migration and invasion of RA FLS.
Piperlongumine upregulated levels
of Myeloid derived suppressor cells
Sun et al. (2015)
(Continues)
CHATTERJEE ET AL.21
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
TABLE 3 (Continued)
Herb species
Part of
plant used Study objective Animal model Results Reference
Pluchea lanceolata
(DC.) C.B.Clarke
Whole plant Exploration of the anti-inflammatory
and anti-arthritic activities of four
flavonoids
Adult albino rats of either sex The bio-actives showed anti-
inflammatory and anti-arthritic
effects against pedal inflammation
and granuloma formation.
The concoction showed less
efficacy than betamethasone but
possessed the advantage of
manifesting no adverse gastric
lesions as in case of the steroid
Shankar Pandey and Trigunayat
(2018)
Commiphora wightii
(Arn.) Bhandari
Exudate (gum
resin)
Investigation of anti-inflammatory
effects of aqueous extract of gum
resin
Carrageenan induced rat hind paw
oedema
Decrease in rat hind paw oedema
Significant reduction of
inflammatory biomarkers
Shekhawat et al. (2021)
Paederia foetida L. Whole plant Evaluation of anti-inflammatory
property
Carrageenan induced paw oedema
model in Wistar Albino rats
The butanolic extract showed the
best dose-dependent decrease in
oedema of up to 79.2%
Bandiya et al. (2022)
Vitex negundo L. Seeds Estimation of therapeutic effects CFA induced arthritis in Male Wistar
rats
Subsidence of rat paw oedema,
synovial layer hyperproliferation
and inflammatory cell infiltration,
chronic inflammation, pannus
build-up and bone resorption
Downregulation of TNF-αand IL-
1β, IL-6; upregulation of IL-10
Reversal of weight loss as observed
after induction of arthritis.
Marked reduction of COX-2
and 5-LOX
Zheng, Zhang, et al. (2014),
Zheng, Zhao, et al. (2014)
Ricinus communis L. Leaves Evaluation of anti-arthritic activity CFA induced arthritis in Wistar rats Hydroalcoholic extract of Ricinus
root showed significant decrease in
arthritic scores
Reduction of paw oedema
Improvement in body weight
Hussain et al. (2021)
Abbreviations: CFA, complete Freund's adjuvant; COX, cyclooxygenase; DCM, dichloromethane; GSH, glutathione; IFN, interferon; IL, interleukin; LOX, lipoxygenase; PG, prostaglandin; RA-FLS, rheumatoid
arthritis fibroblast-like synoviocytes; SOD, superoxide dismutase; TBARs, thiobarbituric acid reactive substances.
22 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
compounds consist of sesquiterpene hydrocarbons, which are mainly
zingiberene, curcumene and farnesene (Yousfi et al., 2021), which
contribute to the characteristic gingery taste and odour. Non-volatile
compounds are comprised of gingerols, shogaols, paradols and zinger-
one, which are responsible for the hot sensation on ingestion of the
rhizome. They also contain vitamins and enzymes named zingibain
(Gupta & Sharma, 2014). The dichloromethane (DCM) extract of dried
pulverised ginger was experimentally detected as containing various
gingerols, some oils, terpenes and polar moieties. These compounds
were collectively found to ameliorate the synthesis of PGE2 in vitro,
out of which gingerols and their variations were witnessed to be key
contributors to this anti-PG effect, with complimentary effects by the
non-gingerols. Further in vivo studies compared a phenolic prepara-
tion versus a similar crude DCM extract of the gingerol components
administered intra-peritoneally substantially deferring synovial inflam-
mation and bone damage in streptococcal cell wall-induced arthritis
model in mice, with the DCM extract showing better efficacy (Funk
et al., 2009). Another in vivo study suggested the anti-inflammatory
activity of zingiber by reducing rat paw oedema in arthritic rats (Bek-
kouch et al., 2022). Therefore, it is evident that plants of the genus
Zingiber carry the promise of an established solution for
the management of RA.
7.5 |Piper longum L.
Plants belonging to the genus Piper have famously been reported to
be of pharmacological significance, with Piper longum being one of the
most important species in the genus. P. longum, also identified as long
pepper, is mentioned in ancient literature to be possessing medicinal
properties (Khandhar et al., 2010). The P. longum plant contains bioac-
tive alkaloids such as piperine, methylpiperine, iperonaline, piperet-
tine, asarinine, pellitorine, piperundecalidine, piperlongumine,
piperlonguminine, piperazine, methyl-3,4,5-trimehoxycinnamate and
many more. Other than that, they also contain lignans, esters and vol-
atile oils (Khandhar et al., 2010; Khushbu et al., 2011). Piperine is the
major active component of the Piper family, which can be extracted
from P. longum,orPiper nigrum. Piperine acts as a potent bio-enhancer
to many other bioactives. A systematic review of different
publications on P. longum reported that it exhibits analgesic, anti-
inflammatory, and anti-arthritic behaviour among a vast list of activi-
ties (Yadav et al., 2020). Piperlongumine weakened the pathological
damage in the form of inflammatory cell penetration as well as carti-
laginous damage in the CIA mice model through the attenuation of
serum Ig levels and cytokine production of TNF-α, IL-1β, IL-23 and IL-
17 in the mice sera. The investigation suggested that the Piperlongu-
mine substantially decreased Th-17 cells in the lymph nodes, along
with a sharp rise in myeloid-derived suppressor cells (MDSCs) that are
capable of suppressing T-cell mediated inflammatory responses. How-
ever, it did not affect the T-reg cells. Piperlongumine was also able to
suppress the migratory nature of the human RA-FLS (Sun et al., 2015).
Another study by Xiao et al. (2016) demonstrated the ability of piper-
longumine to ameliorate LPS-induced DC maturation, reactive oxygen
species production by DCs and activation of p38, JNK, NF-ĸB and
PI3K/Akt signalling pathways. A dose between 200 and 400 mg/kg of
aqueous extract of P. longum seeds shows an effective reduction in
rat paw volume in CFA-induced arthritic rats (Yende, 2010). In Com-
plete Freund's Adjuvant-induced arthritis in mice, solid lipid nanoparti-
cles encapsulating piperine also brought a greater reduction in
inflamed paw volume as compared to a suspension of chloroquine as
an anti-rheumatic standard. The researchers suggested the reason
was that topical and oral piperine localised more effectively at the tar-
get site. These piperine preparations also demonstrated marked inhi-
bition of bone disintegration, suppression of TNF-αrelease from
macrophages, angiogenesis, joint infiltration and distortion (Bhalekar
et al., 2017). The anti-inflammatory properties of the Piper plant offer
immense capacity to be expanded to a course of medication for RA.
7.6 |Pluchea lanceolata (DC.) C.B.Clarke
The plant Pluchea lanceolata belongs to the family Asteraceae and is
known to possess anti-inflammatory, analgesic, anti-ulcer, anti-micro-
bial, anti-nociceptive, antipyretic, anti-rheumatic and neuroprotective
characteristics (Kamel & Ahemd, 2013; Srivastava & Shanker, 2012).
The leaves of this plant were discovered to be abundant in bioactive
metabolites such as flavonoids, phenolic compounds, alkaloids, terpe-
noids, sterols, and so forth (Sharma & Goyal, 2011). The major com-
pounds found in the pulverised powder of the stem of this plant
found through gas chromatographymass spectrometryMS analysis
are quercetin, hesperetin-7-rutinoside, taxifolin-3-arabinoside, formo-
nonetin-7-O-glucoside, isorhamnetin, decanoic acid, octadecanoic
acid, cyclopentadecanone, stearic acid, heptadecane, tetracosane,
docosane and hexadecenoic acid (Gour et al., 2012; Kamel &
Ahemd, 2013). The ethanolic extract of the leaves of P. lanceolata
exhibited significant anti-inflammatory behaviour, which was further
fractionated into a hexane extract that showed optimum anti-inflam-
matory suppression of carrageenin-triggered oedema in the rat paw,
owing to psi-taraxasterol acetate acting as the active ingredient
(Garg & Manisha, 2020). The leaf extract of this herb showed inhibi-
tion of both cellular and humoral antibody responses through the
downregulation of pro-inflammatory cytokines and effective suppres-
sion of CD4
+
and CD8
+
T cells (Bhagwat et al., 2010). Another study
similarly proved the anti-inflammatory and anti-arthritic activity of fla-
vonoids present in P. lanceolata extract using albino rats. Although its
activity was not as strong as betamethasone, there was a notable dif-
ference in the incidence of gastric lesions and haemorrhagic ulcers
which was 50% higher in the case of established anti-inflammatory
corticosteroid betamethasone (Shankar Pandey & Trigunayat, 2018).
The Pluchea plant possesses the properties required for the ameliora-
tion of the disease conditions of RA.
7.7 |Commiphora wightii (Arn.) Bhandari
Commiphora wightii has been traditionally utilised for its famously
known effects against arthritis, inflammation, gout, rheumatism and
metabolic disorders (Sarup et al., 2015). The white exudate from the
CHATTERJEE ET AL.23
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Commiphora plant contains a concoction of volatile oils containing ter-
penes and terpenoids, sterols, sterones, steroids, lignans, flavonoids
and flavanones. The major volatile oils are myrecene, dimyrecene,
polymyrecene and resins or guggulsterols (Rani & Mishra, 2013; Sarup
et al., 2015). Guggulsterone (4,17(20)-pregnadiene-3,16-dione) is
named after the common name of the plant, guggul (Kunnumakkara
et al., 2018). Z-guggulsterone and E-guggulsterone, predominant ste-
roids present in this herb, exhibited potent inhibition of nitric oxide
production in LPS-induced murine macrophage cell line J774.1 (IC
50
21.1 and 42.3 mM, respectively) (Meselhy, 2003). Cis and trans iso-
mers of this compound reportedly inhibited the production of chemo-
kines regulated upon activation, normal t cell expressed and
presumably secreted and epithelial cell-derived neutrophil activating
peptide-78 in IL-1βtreated FLS, allegedly by some interference with
the farnesoid X receptor (Bile acid) signalling pathway. 3-(4,5-
Dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide assay of this
setup revealed that guggulsterone also curtailed the hyperprolifera-
tion seen in the IL-1βtreated FLS by 101.3 ± 4.2% and 98.3 ± 3.7%,
respectively. IL-1β-induced MMP secretion by FLS was also sup-
pressed by treatment with this potent natural compound. It is known
that chemokines are secreted by FLS and macrophages through acti-
vation of the NF-κB cascade. Moreover, IL-1βwas proved to increase
the association of TCF NF-κB with its corresponding consensus
sequence. Cis and trans guggulsterone demonstrated inhibition of
MMP production through interference with the NF-κB route, along
with significantly decreasing IκBαdegradation and consequently muf-
fled NF-κB activation (Lee, Lee, Noh, et al., 2008). Electrophoretic
mobility shift assay established that guggulsterone abolished RANKL
mediated NF-κB induction in a dose-dependent manner with optimum
inhibition at 50 μmol/L concentration. Pretreatment of cells with gug-
gulsterone restricted the phosphorylation of IκBα(Inhibitor of κB-α),
which is needed for NF-κB activation without interfering with the
amount of IKKαor IKKβ. Osteoclast differentiation was consequently
tapered (more than 90%) in RAW 264.7 cells cultured in the presence
of a low concentration of 5 μmol/L of this active compound (Ichi-
kawa & Aggarwal, 2006). Plants belonging to the genus Commiphora
as a whole possess anti-inflammatory properties (Dinku et al., 2022).
This plant and its phytoconstituents can thus emerge as a prospective
treatment option for RA.
7.8 |Paederia foetida L.
This plant belonging to the Rubiaceae family has been known for its
anti-inflammatory properties since pre-historic times and was even
used for the treatment of rheumatic diseases, being endowed with
principal bio-actives like iridoid glycosides, flavonoid glycosides, phe-
nolic derivatives, anthraquinones, triterpenoids, phytosterols, essential
oils, sterols and galacturonic acids (Dutta et al., 2023).
Essential medicinal extracts are obtained from multiple parts of this
invaluable plant, such as diuretic functions from a decoction of the
leaves, emetics from roots, dental analgesics from fruits and leuko-
derma suppressors from the seeds, to name a few. A carefully
prepared leaf extract demonstrated a higher dose-dependent inhibi-
tory effect on cotton pellet granuloma formation in Charles Foster
albino rats in comparison to phenylbutazone, an established NSAID.
There also exist reports of methanolic and hexane extracts of this
herb offering an anti-nociceptive effect measured by a reduction in
writhing in rats administered with acetic acid (Wang et al., 2014). A
study by Das et al. (2012) showed that Paederia leaf extract in ethanol
had an anti-nociceptive effect by demonstrating an increase in tail
flicking time, and an anti-inflammatory effect based on rat paw
oedema volume reduction. A 100 mg/kg body weight oral administra-
tion of desiccated methanolic extract of Paederia foetida leaves dimin-
ished paw oedema induced by injecting turpentine oil in Swiss albino
rats by 45%. This same dose also exhibited substantial inhibition of
pro-inflammatory cytokines IL-2, IL-1βand TNF-αin rats with CFA-
induced arthritis. Inflammation was induced by using alternative com-
pounds like arachidonic acid and PGE2, oedema being considerably
restricted in all these inflammatory models by the Paederia-derived
preparation. Additionally, neutrophil infiltration was also hindered
proportionately with dose by treating the arthritis-ridden rats with the
extract (Kumar et al., 2015). As a result, it can be stated that the com-
ponents of this herb can be certainly incorporated into management
regimens for RA.
7.9 |Vitex negundo L.
Traditionally known for its medicinal properties, the Vitex negundo
herb has been reported to possess anti-arthritic, anti-inflammatory
and anti-rheumatic potency (Vishwanathan & Basavaraju, 2010).
Extracts from various parts of the plant contain flavonoids, lignans,
terpenoids, steroids and additional compounds like coumarins and
acids (Zheng et al., 2015). In male Wistar rats injected with Complete
Freund's Adjuvant, ethanolic extract (340 and 85 mg/kg) of the pul-
verised seeds of this plant brought a reduction of oedema in the rat
paws with an impact comparable with the strong drug methotrexate.
Moreover, the time taken to regain weight was lesser in CFA-treated
rats administered with Vitex extract as compared to CFA-treated rats
not administered with any extract or drug. The group of adjuvant-
induced arthritic rats treated with Vitex seed extract witnessed a
sharp improvement in arthritic condition in terms of reduction in
synovial inflammatory infiltration and synovial layer hyper-prolifera-
tion with hindrance in joint destruction. Evidence suggests that this
extract also facilitated resistance to inflammation, pannus generation,
bone damage, downregulation of pro-inflammatory cytokines, upregu-
lation of anti-inflammatory cytokines and a stark reduction in COX-2
and 5-lipoxygenase quantities in the peripheral blood mononuclear
cells (Zheng, Zhao, et al., 2014). Zheng et al. fractioned four lignans
from this extract called vitexdoin F, vitexdoin G, vitexdoin H and
vitexdoin I. One of these compounds, vitexdoin F, demonstrated inhi-
bition against nitric oxide generation in RAW264.7 macrophage cell
lines stimulated with LPS through interference with inducible nitric
oxide synthase enzyme with efficacy comparable with indomethacin,
offering potential inhibitory effect against bone resorption. They also
24 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
showed anti-osteoporotic activity in LPS-stimulated UMR106 osteo-
blastic cells. Vitexdoin H, clearly showed inhibition of osteoclastic tar-
trate-resistant acid phosphatase up to 11.4%, hinting at anti-RA
activity of V. negundo seeds. Thus, these substances offer positive
potential against RA (Zheng, Zhang, et al., 2014). It can be conclusively
suggested that the constituents of this plant can be processed and
developed into a mode of treatment for this autoimmune disease.
7.10 |Ricinus communis L.
Commonly known as the castor plant, Ricinus communis is of great
pharmacological value. The roots of this plant are known for its anti-
rheumatic, anti-nociceptive, immunomodulatory, anti-inflammatory,
wound healing and antioxidant properties (Jena & Gupta, 2012). Anal-
ysis of the raw methanolic extract of the root resulted in the detection
of compounds such as alkaloids, saponins, flavonoids, terpenoids, car-
bohydrates and various proteins. Gas chromatographymass spec-
trometry analysis of Ricinus root extract revealed the presence of
major phytocompounds such as hexadecanoic acid (77.8%), 2-meth-
oxy-4-vinylphenol ethenone (28.4%), sucrose (12.6%) and heptatria-
cotanol (10.5%) among a vast variety of constituents (Pandey &
Tiwari, 2021). Hydroalcoholic extract of the leaves of this plant
(500 mg/kg body weight) reversed weight loss in CFA-induced
arthritic male Wistar rats, significantly attenuated inflammation in
their paws, reduced inflamed thymic and spleen mass, reduced levels
of inflammatory biomarkers such as C-reactive protein and RF as well
as IL-6, IL-17a, IL-1βexpression, upregulated INF-γ, IL-4 quantity,
downregulated RANKL mRNA generation and led to an overall decre-
ment in the arthritic score calibrated by the research group. Histo-
pathological studies revealed that this extract of R. communis was able
to counteract synovial inflammatory infiltration, bone resorption and
pannus build-up. Radiological data suggested that the plant also ame-
liorated joint space obstruction, ankylosis, tissue swelling and osteoly-
sis (Hussain et al., 2021). So, this herb holds substantial potential in
alleviating the symptoms of RA.
8|CONCLUSION AND FUTURE
PERSPECTIVES
This review expands on the risk factors that can potentially trigger the
onset of RA. These triggers can be of genetic, environmental or micro-
bial nature. They can also be dependent on the gender and lifestyle of
the individual. The sequential events of the pathophysiological pro-
gression of the disease are multifaceted. The first sign of this disease
is the break of immunological tolerance due to the above-mentioned
risk factors. This is followed by auto-antibody generation and aggre-
gation of antibodyantigen immune complexes that activate Fcγnoci-
ceptive receptors (on the surface of immune cells) that are highly
specific for IgG protein. This leads to a cascade of inflammatory
responses involving activation of immune cells such as neutrophils,
macrophages and mast cells, and overproduction of pro-inflammatory
cytokines. These cytokines act as messengers of inflammation and
trigger FLS, osteoclast and chondrocytes. The FLS in the synovium
begin to hyper-proliferate due to the upregulation of the cellular sig-
nalling pathways such as the JAKSTAT, MAPK/ERK, VEGF and
mTOR pathways. Consequently, the chondrocytes degrade the carti-
lage, the osteoclasts perform resorption of the bone tissue and the
ECM begins to disintegrate. As a result, there is pain and inflammation
in the joints, leading to loss of joint functionality and disabilities. The
current medications for this chronic condition include DMARDs,
NSAIDs, corticosteroids and biological drugs. When these strategies
fail, patients undergo surgical procedures such as arthroplasty or joint
replacement to restore the functionality of the joint. In this review,
we have highlighted the adverse events that follow the usage of these
therapeutic approaches. Due to these shortcomings, the riskbenefit
ratio of these lines of treatment is depreciating and the scientific com-
munity is turning its attention towards safer alternatives. Compounds
of natural origin have had leverage in the discovery of new drug enti-
ties and have remained pivotal participants of ancient therapy systems
(Sam, 2019). Although it is already established that medicinal herbs
have significant therapeutic potential against inflammatory conditions
like RA (Amalraj & Gopi, 2021), herbal medications are yet to be con-
sidered as a preferred treatment option. This review aims to empha-
size the immense potential of therapeutic herbs in the realm of the
medical world and their superiority in terms of lowered side effects as
compared to conventional treatment profiles. In-depth standardized
analytical procedures, molecular identification, safety and efficacy
evaluation parameters are required to validate the authenticity and
activities of these compounds. Thus, there is immense potential for
natural compounds to be introduced in the healthcare realm. Only
with strong scientific techniques can these herbs penetrate the world
of modern medicine, and that looks very feasible in today's scenario.
AUTHOR CONTRIBUTIONS
Amrita Chatterjee: Conceptualization; data curation; formal analysis;
visualization; writing original draft; writing review and editing.
Monisha Jayaprakasan: Conceptualization; data curation; formal anal-
ysis; visualization; writing original draft; writing review and editing.
Anirban Kr Chakrabarty: Conceptualization; data curation; formal
analysis; investigation; methodology; visualization; writing original
draft; writing review and editing. Naga Rajiv Lakkaniga: Formal anal-
ysis; funding acquisition; resources; software; supervision; validation;
writing review and editing. Bibhuti Nath Bhatt: Data curation;
methodology; writing review and editing. Dipankar Banerjee: Super-
vision; writing review and editing. Avinash Narwaria: Supervision;
writing review and editing. Chandra Kant Katiyar:
Supervision; writing review and editing. Sunil Kumar Dubey: Con-
ceptualization; data curation; formal analysis; investigation; project
administration; resources; supervision; validation; visualization;
writing review and editing.
ACKNOWLEDGEMENTS
The authors acknowledge R&D Healthcare Division, Emami Ltd., Kol-
kata, West Bengal, India. The authors gratefully acknowledge that this
CHATTERJEE ET AL.25
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
work is partially supported by NRL's Science and Engineering
Research Board SRG/2022/000091. All the authors are grateful for
the support.
CONFLICT OF INTEREST STATEMENT
The authors declare that they have no conflict of interest.
DATA AVAILABILITY STATEMENT
Data sharing is not applicable to this article as no new data were cre-
ated or analyzed in this study.
ORCID
Amrita Chatterjee https://orcid.org/0009-0009-8635-2464
Monisha Jayaprakasan https://orcid.org/0009-0003-2453-9609
Anirban Kr Chakrabarty https://orcid.org/0000-0001-7165-7366
Naga Rajiv Lakkaniga https://orcid.org/0000-0001-8370-2224
Dipankar Banerjee https://orcid.org/0000-0002-6276-6816
Avinash Narwaria https://orcid.org/0009-0007-7137-1190
Sunil Kumar Dubey https://orcid.org/0000-0002-7554-3232
REFERENCES
^
Ale ´
A ne Vincent-Schneider, H., Stumptner-Cuvelette, P., Lankar, D.,
Pain, S., Ë Raposo, G., Benaroch, P., & Bonnerot, C. (2002). Exosomes
bearing HLA-DR1 molecules need dendritic cells to efficiently stimu-
late specific T cells. International Immunology,14, 713722. https://
doi.org/10.1093/intimm/dxf048
Abdel-Rafei, M. K., Thabet, N. M., & Amin, M. M. (2022). Concerted regula-
tion of OPG/RANKL/ NF-κB/MMP-13 trajectories contribute to ame-
liorative capability of prodigiosin and/or low dose γ-radiation against
adjuvant- induced arthritis in rats. International Immunopharmacology,
111, 109068. https://doi.org/10.1016/j.intimp.2022.109068
Afsar, V., Mohan Reddy, Y., & Saritha, K. V. (2012). In vitro antioxidant
activity and anti inflammatory activity of methanolic leaf extract of
Boswellia serrata.International Journal of Life Sciences Biotechnology and
Pharma Research,1,1523.
Ahmadi, P., Mahmoudi, M., Kheder, R. K., Faraj, T. A., Mollazadeh, S.,
Abdulabbas, H. S., & Esmaeili, S. A. (2023). Impacts of Porphyromonas
gingivalis periodontitis on rheumatoid arthritis autoimmunity. Interna-
tional Immunopharmacology,118, 109936. https://doi.org/10.1016/j.
intimp.2023.109936
Alahdal, M., Zhang, H., Huang, R., Sun, W., Deng, Z., Duan, L.,
Ouyang, H., & Wang, D. (2021). Potential efficacy of dendritic cell
immunomodulation in the treatment of osteoarthritis. Rheumatology
(United Kingdom),60, 507517. https://doi.org/10.1093/
rheumatology/keaa745
Alberti, P., & Handel, A. E. (2021). The contribution of thymic tolerance to
central nervous system autoimmunity. Seminars in Immunopathology,
43, 135157. https://doi.org/10.1007/s00281-020-00822-z/
Published
Aletaha, D., Kapral, T., & Smolen, J. S. (2003). Toxicity profiles of tradi-
tional disease modifying antirheumatic drugs for rheumatoid arthritis.
Annals of the Rheumatic Diseases,62, 482486. https://doi.org/10.
1136/ard.62.5.482
Almutairi, K. B., Nossent, J. C., Preen, D. B., Keen, H. I., & Inderjeeth, C. A.
(2021). The prevalence of rheumatoid arthritis: A systematic review of
population-based studies. Journal of Rheumatology,48, 669676.
https://doi.org/10.3899/jrheum.200367
Al-Nahain, A., Jahan, R., & Rahmatullah, M. (2014). Zingiber officinale:A
potential plant against rheumatoid arthritis. Art,2014,18. https://
doi.org/10.1155/2014/159089
Amalraj, A., & Gopi, S. (2021). Values of natural products to future antiin-
flammatory pharmaceutical discovery. In S. Gopi, A. Amalraj, & A. B.
Kunnumakkara (Eds.), Inflammation and natural products (pp. 375403).
Elsevier. https://doi.org/10.1016/B978-0-12-819218-4.00009-2
Ammon, H. P. T. (2010). Modulation of the immune system by Boswellia
serrata extracts and boswellic acids. Phytomedicine,17, 862867.
https://doi.org/10.1016/j.phymed.2010.03.003
Angum, F., Khan, T., Kaler, J., Siddiqui, L., & Hussain, A. (2020). The preva-
lence of autoimmune disorders in women: A narrative review. Cureus,
12, e8094. https://doi.org/10.7759/cureus.8094
Athanasiou, K. A. (2022). 7 Rheumatoid Arthritis Surgery Options for You
[WWW document]. Very Well Health. https://www.verywellhealth.
com/rheumatoid-arthritis-surgery-5225734
Bandiya, M., Dashora, K., & Sisodiya, D. (2022). Anti-inflammatory activity
of Paederia foetida Linn in carrageenan induced rat paw edema. AGPE
the Royal GONDWANA Research Journal of History, Science, Economic,
Political and Social Science,3,18.
Bekkouch, O., Zengin, G., Harnafi, M., Touiss, I., Khoulati, A., Saalaoui, E.,
Harnafi, H., Abdellattif, M. H., & Amrani, S. (2022). Anti-inflammatory
study and phytochemical characterization of Zingiber officinale Roscoe
and Citrus limon L. juices and their formulation. ACS Omega,30,
2671526724. https://doi.org/10.1021/acsomega.2c04263
Benjamin, O., Goyal, A., & Lappin, S. L. (2022). Disease-modifying antirheu-
matic drugs (DMARD). StatPearls Publishing.
Ben-Meir, K., Twaik, N., Meirow, Y., & Baniyash, M. (2022). An in vivo
mouse model for chronic inflammationinduced immune suppression:
Afactoryfor myeloid-derived suppressor cells (MDSCs). Current Pro-
tocols,2.https://doi.org/10.1002/cpz1.558
Berkun, Y., Levartovsky, D., Rubinow, A., Orbach, H., Aamar, S.,
Grenader, T., Abou Atta, I., Mevorach, D., Friedman, G., & Ben-
Yehuda, A. (2004). Methotrexate related adverse effects in patients
with rheumatoid arthritis are associated with the A1298C polymor-
phism of the MTHFR gene. Annals of the Rheumatic Diseases,63,
12271231. https://doi.org/10.1136/ard.2003.016337
Bertoldo, E., Adami, G., Rossini, M., Giollo, A., Orsolini, G., Viapiana, O.,
Gatti, D., & Fassio, A. (2021). The emerging roles of endocrine hor-
mones in different arthritic disorders. Frontiers in Endocrinology,12,
620920. https://doi.org/10.3389/fendo.2021.620920
Bhagwat, D. P., Kharya, M. D., Bani, S., Kaul, A., Kour, K., Chauhan, P. S.,
Suri, K. A., & Satti, N. K. (2010). Immunosuppressive properties of Plu-
chea lanceolata leaves. Indian Journal of Pharmacology,42,2126.
https://doi.org/10.4103/0253-7613.62405
Bhalekar, M. R., Madgulkar, A. R., Desale, P. S., & Marium, G. (2017). For-
mulation of piperine solid lipid nanoparticles (SLN) for treatment of
rheumatoid arthritis. Drug Development and Industrial Pharmacy,43,
10031010. https://doi.org/10.1080/03639045.2017.1291666
Bian, J., Dannappel, M., Wan, C., & Firestein, R. (2020). Transcriptional reg-
ulation of Wnt/β-catenin pathway in colorectal cancer. Cells,9, 2125.
https://doi.org/10.3390/cells9092125
Bianco, N. R., Seon, H. K., Ruffner, M. A., & Robbins, P. D. (2009). Thera-
peutic effect of exosomes from indoleamine 2,3-dioxygenase-positive
dendritic cells in collagen-induced arthritis and delayed-type hypersen-
sitivity disease models. Arthritis and Rheumatism,60, 380389.
https://doi.org/10.1002/art.24229
Bo, M., Jasemi, S., Uras, G., Erre, G. L., Passiu, G., & Sechi, L. A. (2020). Role
of infections in the pathogenesis of rheumatoid arthritis: Focus on
mycobacteria. Microorganisms,8, 1459. https://doi.org/10.3390/
microorganisms8101459
Bourgeois, D., Inquimbert, C., Ottolenghi, L., & Carrouel, F. (2019). Peri-
odontal pathogens as risk factors of cardiovascular diseases, diabetes,
rheumatoid arthritis, cancer, and chronic obstructive pulmonary dis-
easeIs there cause for consideration? Microorganisms,7, 424.
https://doi.org/10.3390/microorganisms7100424
Brad
unaitė, R., Leonaviˇ
cienė, L., Akramas, L., Leonaviˇ
ci
utė-
Klimantaviˇ
cienė, M., Vasiliauskas, A., Dumalakienė, I., Jonauskienė, I.,
26 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Baleiˇ
sis, J., & Mackiewicz, Z. (2020). Anti-inflammatory and antioxidant
effects of turmeric extracts in rat adjuvant arthritis. Veterinarski Arhiv,
90, 393402. https://doi.org/10.24099/vet.arhiv.0675
Brugman, M. H., Wiekmeijer, A. S., Van Eggermond, M., Wolvers-
Tettero, I., Langerak, A. W., De Haas, E. F. E., Bystrykh, L. V., Van
Rood, J. J., De Haan, G., Fibbe, W. E., & Staal, F. J. T. (2015). Develop-
ment of a diverse human T-cell repertoire despite stringent restriction
of hematopoietic clonality in the thymus. Proceedings of the National
Academy of Sciences of the United States of America,112, E6020
E6027. https://doi.org/10.1073/pnas.1519118112
Bullock, J., Rizvi, S. A. A., Saleh, A. M., Ahmed, S. S., Do, D. P.,
Ansari, R. A., & Ahmed, J. (2019). Rheumatoid arthritis: A brief over-
view of the treatment. Medical Principles and Practice,27, 501507.
https://doi.org/10.1159/000493390
Burn, E., Edwards, C. J., Murray, D. W., Silman, A., Cooper, C., Arden, N. K.,
Prieto-Alhambra, D., & Pinedo-Villanueva, R. (2018). The impact of
rheumatoid arthritis on the risk of adverse events following joint
replacement: A real-world cohort study. Clinical Epidemiology,10,
697704. https://doi.org/10.2147/CLEP.S160347
Bustamante, M. F., Garcia-Carbonell, R., Whisenant, K. D., & Guma, M.
(2017). Fibroblast-like synoviocyte metabolism in the pathogenesis of
rheumatoid arthritis. Arthritis Research & Therapy,19, 110. https://doi.
org/10.1186/s13075-017-1303-3
Caporali, R., Caprioli, M., Bobbio-Pallavicini, F., & Montecucco, C. (2008).
DMARDS and infections in rheumatoid arthritis. Autoimmunity
Reviews,8, 139143. https://doi.org/10.1016/j.autrev.2008.05.001
Carmona, L., Aurrecoechea, E., & García de Yébenes, M. J. (2023). Tailoring
rheumatoid arthritis treatment through a sex and gender lens. Journal
of Clinical Medicine,13, 55. https://doi.org/10.3390/jcm13010055
Castañeda, J., Hidalgo, Y., Sauma, D., Rosemblatt, M., Bono, M. R., &
Núñez, S. (2021). The multifaceted roles of B cells in the thymus: From
immune tolerance to autoimmunity. Frontiers in Immunology,12,
766698. https://doi.org/10.3389/fimmu.2021.766698
Catrina, A., Krishnamurthy, A., & Rethi, B. (2021). Current view on the
pathogenic role of anti-citrullinated protein antibodies in rheumatoid
arthritis. RMD Open,7, e001228. https://doi.org/10.1136/rmdopen-
2020-001228
Cejka, D., Hayer, S., Niederreiter, B., Sieghart, W., Fuereder, T.,
Zwerina, J., & Schett, G. (2010). Mammalian target of rapamycin sig-
naling is crucial for joint destruction in experimental arthritis and is
activated in osteoclasts from patients with rheumatoid arthritis. Arthri-
tis and Rheumatism,62, 22942302. https://doi.org/10.1002/art.
27504
Chandran, B., & Goel, A. (2012). A randomized, pilot study to assess the
efficacy and safety of curcumin in patients with active rheumatoid
arthritis. Phytotherapy Research,26, 17191725. https://doi.org/10.
1002/ptr.4639
Chandrasekharan, N. V., Dai, H., Lamar, K., Roos, T., Evanson, N. K.,
Tomsik, J., Elton, T. S., & Simmons, D. L. (2002). COX-3, a cyclooxy-
genase-1 variant inhibited by acetaminophen and other analgesic/anti-
pyretic drugs: Cloning, structure, and expression. Proceedings of the
National Academy of Sciences,99, 1392613931. https://doi.org/10.
1073/pnas.162468699
Chen, X., Du, G., Bai, S., Dijia, L., Li, C., Hou, Y., Zhang, Y., Zhang, Z.,
Gong, T., Fu, Y., Bottini, M., & Sun, X. (2021). Restoring immunological
tolerance in established experimental arthritis by combinatorial citrulli-
nated peptides and immunomodulatory signals. Nano Today,41,
101307. https://doi.org/10.1016/j.nantod.2021.101307
Coradduzza, D., Bo, M., Congiargiu, A., Azara, E., De Miglio, M. R., Luca
Erre, G., & Carru, C. (2023). Decoding the microbiome's influence on
rheumatoid arthritis. Microorganisms,11, 2170. https://doi.org/10.
3390/microorganisms11092170
Costello, R., David, T., & Jani, M. (2019). Impact of adverse events associ-
ated with medications in the treatment and prevention of rheumatoid
arthritis. Clinical Therapeutics,41, 13761396. https://doi.org/10.
1016/j.clinthera.2019.04.030
Court, A. C., Le-Gatt, A., Luz-Crawford, P., Parra, E., Aliaga-Tobar, V.,
Bátiz, L. F., Contreras, R. A., Ortúzar, M. I., Kurte, M., Elizondo-Vega, R.,
Maracaja-Coutinho, V., Pino-Lagos, K., Figueroa, F. E., & Khoury, M.
(2020). Mitochondrial transfer from MSCs to T cells induces Treg dif-
ferentiation and restricts inflammatory response. EMBO Reports,21,
e48052. https://doi.org/10.15252/embr.201948052
Curran, A. M., Girgis, A. A., Jang, Y., Crawford, J. D., Thomas, M. A.,
Kawalerski, R., Coller, J., Bingham, C. O., Na, C. H., & Darrah, E. (2023).
Citrullination modulates antigen processing and presentation by
revealing cryptic epitopes in rheumatoid arthritis. Nature Communica-
tions,14, 1061. https://doi.org/10.1038/s41467-023-36620-y
Curran, A. M., Naik, P., Giles, J. T., & Darrah, E. (2020). PAD enzymes in
rheumatoid arthritis: Pathogenic effectors and autoimmune targets.
Nature Reviews Rheumatology,16, 301315. https://doi.org/10.1038/
s41584-020-0409-1
Darrah, E., & Andrade, F. (2018). Rheumatoid arthritis and citrullination.
Current Opinion in Rheumatology,30,7278. https://doi.org/10.1097/
BOR.0000000000000452
Das, S., Bordoloi, P., Saikia, P., & Kanodia, L. (2012). The analgesic and
acute anti-inflammatory effect of the ethanolic extract of the leaves of
Paederia foetida (EEPF) on experimental animal models. Bangladesh
Journal of Medical Science,11, 206211. https://doi.org/10.3329/
bjms.v11i3.11723
Davis, A., & Robson, J. (2016). The dangers of NSAIDs: Look both ways.
British Journal of General Practice,66, 172173. https://doi.org/10.
3399/bjgp16X684433
De Hair, M. J. H., Landewé, R. B. M., Van De Sande, M. G. H., Van
Schaardenburg, D., Van Baarsen, L. G. M., Gerlag, D. M., & Tak, P. P.
(2012). Smoking and overweight determine the likelihood of develop-
ing rheumatoid arthritis. Annals of the Rheumatic Diseases,72, 1654
1658. https://doi.org/10.1136/annrheumdis
Dedmon, L. E. (2020). The genetics of rheumatoid arthritis. Rheumatology
(United Kingdom),59, 26612670. https://doi.org/10.1093/
rheumatology/keaa232
Derksen, V. F. A. M., Huizinga, T. W. J., & van der Woude, D. (2017). The
role of autoantibodies in the pathophysiology of rheumatoid arthritis.
Seminars in Immunopathology,39, 437446. https://doi.org/10.1007/
s00281-017-0627-z
Dhir, V., & Aggarwal, A. (2012). Methotrexate-related minor adverse
effects in rheumatoid arthritis: More than a nuisance. Journal of Clinical
Rheumatology,18,4446. https://doi.org/10.1097/RHU.
0b013e31823ee540
Dinesh, P., Kalaiselvan, S., Sujitha, S., & Rasool, M. (2020). MiR-145-5p
mitigates dysregulated Wnt1/β-catenin signaling pathway in rheuma-
toid arthritis. International Immunopharmacology,82, 106328. https://
doi.org/10.1016/j.intimp.2020.106328
Dinku, W., Park, S. B., Jeong, J. B., Jung, C., & Dekebo, A. (2022). Chemical
composition and anti-inflammatory activity of essential oils from resin
of COMMIPHORA species. Bulletin of the Chemical Society of Ethiopia,
36, 399415. https://doi.org/10.4314/bcse.v36i2.13
Donthi, M. R., Saha, R. N., Singhvi, G., & Dubey, S. K. (2023). Dasatinib-
Loaded topical nano-emulgel for rheumatoid arthritis: formulation
design and optimization by QbD, In vitro, Ex vivo, and In vivo evalua-
tion. Pharmaceutics,15, 736. https://doi.org/10.3390/
pharmaceutics15030736
Dubey, S. K., Kali, M., Hejmady, S., Saha, R. N., Alexander, A., &
Kesharwani, P. (2021). Recent advances of dendrimers as multifunctional
nano-carriers to combat breast cancer. European Journal of Pharmaceuti-
cal Sciences,164, 105890. https://doi.org/10.1016/j.ejps.2021.105890
Dutta, P. P., Marbaniang, K., Sen, S., Dey, B. K., & Talukdar, N. C. (2023). A
review on phytochemistry of Paederia foetida Linn. Phytomedicine Plus,
3, 100411. https://doi.org/10.1016/j.phyplu.2023.100411
CHATTERJEE ET AL.27
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Dwivedi, A. K., Gurjar, V., Kumar, S., & Singh, N. (2015). Molecular basis
for nonspecificity of nonsteroidal anti-inflammatory drugs (NSAIDs).
Drug Discovery Today,20, 863873. https://doi.org/10.1016/j.drudis.
2015.03.004
Eltahir, H., Fawzy, M., Mohamed, E., Alrehany, M., Shehata, A., &
Abouzied, M. (2019). Antioxidant, anti-inflammatory and anti-fibrotic
effects of Boswellia serrate gum resin in CCl4-induced hepatotoxicity.
Experimental and Therapeutic Medicine,19, 13131321. https://doi.
org/10.3892/etm.2019.8353
Emori, T., Kasahara, M., Sugahara, S., Hashimoto, M., Ito, H., Narumiya, S.,
Higashi, Y., & Fujii, Y. (2020). Role of JAK-STAT signaling in the patho-
genic behavior of fibroblast-like synoviocytes in rheumatoid arthritis:
Effect of the novel JAK inhibitor peficitinib. European Journal of Pharma-
cology,882, 173238. https://doi.org/10.1016/j.ejphar.2020.173238
Ethgen, O., De Lemos Esteves, F., Bruyere, O., & Reginster, J. Y. (2013).
What do we know about the safety of corticosteroids in rheumatoid
arthritis? Current Medical Research and Opinion,29, 11471160.
https://doi.org/10.1185/03007995.2013.818531
Faid, S. M. (2021). Treatment effect of willow leaves on rheumatoid arthri-
tis in chronic inflammatory disorders of rats. Journal of Research in the
Fields of Specific Education,7, 345371. https://doi.org/10.21608/
jedu.2021.57432.119
Felix, F. B., Vago, J. P., Fernandes, D. d. O., Martins, D. G., Moreira, I. Z.,
Gonçalves, W. A., Costa, W. C., Araújo, J. M. D., Queiroz-Junior, C. M.,
Campolina-Silva, G. H., Soriani, F. M., Sousa, L. P., Grespan, R.,
Teixeira, M. M., & Pinho, V. (2021). Biochanin a regulates key steps of
inflammation resolution in a model of antigen-induced arthritis via
GPR30/PKA-dependent mechanism. Frontiers in Pharmacology,12,
662308. https://doi.org/10.3389/fphar.2021.662308
Feng, T., Wei, Y., Lee, R. J., & Zhao, L. (2017). Liposomal curcumin and its
application in cancer. International Journal of Nanomedicine,12, 6027
6044. https://doi.org/10.2147/IJN.S132434
Fiebich, B. L., & Chrubasik, S. (2004). Effects of an ethanolic Salix extract
on the release of selected inflammatory mediators in vitro. Phytomedi-
cine,11, 135138. https://doi.org/10.1078/0944-7113-00338
Ford, J. A., Liu, X., Chu, S. H., Lu, B., Cho, M. H., Silverman, E. K.,
Costenbader, K. H., Camargo, C. A., & Sparks, J. A. (2020). Asthma,
chronic obstructive pulmonary disease, and subsequent risk for inci-
dent rheumatoid arthritis among women: A prospective cohort study.
Arthritis and Rheumatology,72, 704713. https://doi.org/10.1002/art.
41194
Fuhrmann, J., Clancy, K. W., & Thompson, P. R. (2015). Chemical biology
of protein arginine modifications in epigenetic regulation. Chemical
Reviews,115, 54135461. https://doi.org/10.1021/acs.chemrev.
5b00003
Fujii, W., Ashihara, E., Hirai, H., Nagahara, H., Kajitani, N., Fujioka, K.,
Murakami, K., Seno, T., Yamamoto, A., Ishino, H., Kohno, M.,
Maekawa, T., & Kawahito, Y. (2013). Myeloid-derived suppressor cells
play crucial roles in the regulation of mouse collagen-induced arthritis.
The Journal of Immunology,191, 10731081. https://doi.org/10.4049/
jimmunol.1203535
Funk, J. L., Frye, J. B., Oyarzo, J. N., Chen, J., Zhang, H., &
Timmermann, B. N. (2016). Anti-inflammatory effects of the essential
oils of ginger (Zingiber officinale Roscoe) in experimental rheumatoid
arthritis. PharmaNutrition,4, 123131. https://doi.org/10.1016/j.
phanu.2016.02.004
Funk, J. L., Frye, J. B., Oyarzo, J. N., & Timmermann, B. N. (2009). Compar-
ative effects of two gingerol-containing Zingiber officinale extracts on
experimental rheumatoid arthritis. Journal of Natural Products,72,
403407. https://doi.org/10.1021/np8006183
Gallegos, A. M., & Bevan, M. J. (2006). Central tolerance: Good but imper-
fect. Immunological Reviews,209, 290296. https://doi.org/10.1111/j.
0105-2896.2006.00348.x
Gao, Y., Duan, R., Li, H., Jiang, L., Tao, T., Liu, X., Zhu, L., Li, Z., Chen, B.,
Zheng, S., Lin, X., & Su, W. (2023). Single-cell analysis of immune cells
on gingiva-derived mesenchymal stem cells in experimental autoim-
mune uveitis. iScience,26, 106729. https://doi.org/10.1016/j.isci.
2023.106729
Garg, N. K., & Manisha. (2020). Therapeutic uses of Rasna (Pluchea lanceo-
lata): A review article. World Journal of Pharmaceutical and Medical
Research,6, 109112.
GBD 2021 Rheumatoid arthritis collaborators. (2023). Global, regional, and
national burden of rheumatoid arthritis, 19902020, and projections
to 2050: A systematic analysis of the global burden of disease study
2021. Lancet Rheumatology,5, 594610.
Genovese, M. C., Weinblatt, M. E., Aelion, J. A., Mansikka, H. T.,
Peloso, P. M., Chen, K., Li, Y., Othman, A. A., Khatri, A., Khan, N. S., &
Padley, R. J. (2018). ABT-122, a bispecific dual variable domain immu-
noglobulin targeting tumor necrosis factor and interleukin-17A, in
patients with rheumatoid arthritis with an inadequate response to
methotrexate: A randomized, double-blind study. Arthritis and Rheuma-
tology,70, 17101720. https://doi.org/10.1002/art.40580
Ghasemian, M., Owlia, S., & Owlia, M. B. (2016). Review of anti-inflamma-
tory herbal medicines. Advances in Pharmacological Sciences,2016,1
11. https://doi.org/10.1155/2016/9130979
Giannini, D., Antonucci, M., Petrelli, F., Bilia, S., Alunno, A., & Puxeddu, I.
(2020). One year in review 2020: Pathogenesis of rheumatoid arthritis.
Clinical and Experimental Rheumatology,38, 387397.
Giles, J. T., Danielides, S., Szklo, M., Post, W. S., Blumenthal, R. S.,
Petri, M., Schreiner, P. J., Budoff, M., Detrano, R., & Bathon, J. M.
(2015). Insulin resistance in rheumatoid arthritis: Disease-related indi-
cators and associations with the presence and progression of subclini-
cal atherosclerosis. Arthritis and Rheumatology,67, 626636. https://
doi.org/10.1002/art.38986
Glant, T. T., Mikecz, K., & Rauch, T. A. (2014). Epigenetics in the pathogen-
esis of rheumatoid arthritis. BMC Medicine,12, 35. https://doi.org/10.
1186/1741-7015-12-35
Gligori
c, E. I., Igi
c, R., Suvajdˇzi
c, L., Teofilovi
c, B. D., & Gruji
c-Leti
c, N. N.
(2020). Salix eleagnos Scop. A novel source of antioxidant and anti-
inflammatory compounds: Biochemical screening and in silico
approaches. South African Journal of Botany,128, 339348. https://
doi.org/10.1016/j.sajb.2019.11.018
Goel, A., Kunnumakkara, A. B., & Aggarwal, B. B. (2008). Curcumin as Cur-
ecumin: From kitchen to clinic. Biochemical Pharmacology,75, 787
809. https://doi.org/10.1016/j.bcp.2007.08.016
Goodman, S. M. (2015a). Optimizing perioperative outcomes for older
patients with rheumatoid arthritis undergoing arthroplasty: Emphasis
on medication management. Drugs & Aging,32, 361369. https://doi.
org/10.1007/s40266-015-0262-0
Goodman, S. M. (2015b). Rheumatoid arthritis: Perioperative management
of biologics and DMARDs. Seminars in Arthritis and Rheumatism,44,
627632. https://doi.org/10.1016/j.semarthrit.2015.01.008
Gorantla, S., Singhvi, G., Rapalli, V. K., Waghule, T., Dubey, S. K., &
Saha, R. N. (2020). Targeted drug-delivery systems in the treatment of
rheumatoid arthritis: recent advancement and clinical status. Therapeu-
tic Delivery,11(4), 269284. https://doi.org/10.4155/tde-2020-0029
Goswami, D., Mahapatra, A. D., Banerjee, S., Kar, A., Ojha, D.,
Mukherjee, P. K., & Chattopadhyay, D. (2018). Boswellia serrata oleo-
gum-resin and β-boswellic acid inhibits HSV-1 infection in vitro
through modulation of NF-кB and p38 MAP kinase signaling. Phytome-
dicine,51,94103. https://doi.org/10.1016/j.phymed.2018.10.016
Gou, M., Men, K., Shi, H., Xiang, M., Zhang, J., Song, J., Long, J., Wan, Y.,
Luo, F., Zhao, X., & Qian, Z. (2011). Curcumin-loaded biodegradable
polymeric micelles for colon cancer therapy in vitro and in vivo. Nano-
scale,3, 15581567. https://doi.org/10.1039/C0NR00758G
Gour, K., Singh Nathawat, R., Arya, D., Patni, V., & Pathology, P. (2012).
GC-MS analysis and identification of daidzein by high performance
thin layer chromatography (HPTLC) of Pluchea lanceolata - A bone
healing plant of semi-arid land. Journal of Pharmacy Research,5,
257260.
28 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Greenblatt, H. K., Kim, H. A., Bettner, L. F., & Deane, K. D. (2020). Preclini-
cal rheumatoid arthritis and rheumatoid arthritis prevention. Current
Opinion in Rheumatology,32, 289296. https://doi.org/10.1097/BOR.
0000000000000708
Guo, Q., Wang, Y., Xu, D., Nossent, J., Pavlos, N. J., & Xu, J. (2018). Rheu-
matoid arthritis: Pathological mechanisms and modern pharmacologic
therapies. Bone Research,6, 15. https://doi.org/10.1038/s41413-018-
0016
Gupta, S. K., & Sharma, A. (2014). Medicinal properties of Zingiber officinale
Roscoe - A review. IOSR Journal of Pharmacy and Biological Sciences,9,
124129. https://doi.org/10.9790/3008-0955124129
Gwinnutt, J. M., Verstappen, S. M., & Humphreys, J. H. (2020). The impact
of lifestyle behaviours, physical activity and smoking on morbidity and
mortality in patients with rheumatoid arthritis. Best Practice &
Research. Clinical Rheumatology,34, 101562. https://doi.org/10.1016/
j.berh.2020.101562
Han, J., Li, X., Luo, X., He, J., Huang, X., Zhou, Q., Han, Y., Jie, H.,
Zhuang, J., Li, Y., Yang, F., Zhai, Z., Wu, S., He, Y., Yang, B., & Sun, E.
(2020). The mechanisms of hydroxychloroquine in rheumatoid arthritis
treatment: Inhibition of dendritic cell functions via toll like receptor
9 signaling. Biomedicine and Pharmacotherapy,132, 110848. https://
doi.org/10.1016/j.biopha.2020.110848
Haribhai, D., Lin, W., Edwards, B., Ziegelbauer, J., Salzman, N. H.,
Carlson, M. R., Li, S.-H., Simpson, P. M., Chatila, T. A., & Williams, C. B.
(2009). A central role for induced regulatory T cells in tolerance induc-
tion in experimental colitis. The Journal of Immunology,182, 3461
3468. https://doi.org/10.4049/jimmunol.0802535
Harirforoosh, S., & Jamali, F. (2009). Renal adverse effects of nonsteroidal
anti-inflammatory drugs. Expert Opinion on Drug Safety,8, 669681.
https://doi.org/10.1517/14740330903311023
Harrington, R., Al Nokhatha, S. A., & Conway, R. (2020). Jak inhibitors in
rheumatoid arthritis: An evidence-based review on the emerging clini-
cal data. Journal of Inflammation Research,13, 519531. https://doi.
org/10.2147/JIR.S219586
Hayder, M., Poupot, M., Baron, M., Nigon, D., Turrin, C.-O., Caminade, A.-
M., Majoral, J.-P., Eisenberg, R. A., Fournié, J.-J., Cantagrel, A.,
Poupot, R., & Davignon, J.-L. (2011). A phosphorus-based dendrimer
targets inflammation and Osteoclastogenesis in experimental arthritis.
Science Translational Medicine,3,3546. https://doi.org/10.1126/
scitranslmed.3002212
Hecht, M. (2022). Steroids for the Treatment of Rheumatoid Arthritis [WWW
document]. Healthline. https://www.healthline.com/health/
rheumatoid-arthritis/steroids-for-rheumatoid-arthritis
Heluany, C. S., Kupa, L. V. K., Viana, M. N., Fernandes, C. M., &
Farsky, S. H. P. (2018). Hydroquinone exposure worsens the symp-
tomatology of rheumatoid arthritis. Chemico-Biological Interactions,
291, 120127. https://doi.org/10.1016/j.cbi.2018.06.016
Hilkens, C. M. U., & Isaacs, J. D. (2013). Tolerogenic dendritic cell therapy
for rheumatoid arthritis: Where are we now? Clinical and Experimental
Immunology,172, 148157. https://doi.org/10.1111/cei.12038
Hogarth, P. M., & Pietersz, G. A. (2012). Fc receptor-targeted therapies for
the treatment of inflammation, cancer and beyond. Nature Reviews.
Drug Discovery,11, 311331. https://doi.org/10.1038/nrd2909
Holmdahl, R., Malmström, V., & Burkhardt, H. (2014). Autoimmune prim-
ing, tissue attack and chronic inflammation - the three stages of rheu-
matoid arthritis. European Journal of Immunology,44, 15931599.
https://doi.org/10.1002/eji.201444486
Huovinen, J., Palosaari, S., Pesonen, P., Huhtakangas, J. A., & Lehenkari, P.
(2023). 1,25(OH)2D3 and its analogue calcipotriol inhibit the migration
of human synovial and mesenchymal stromal cells in a wound healing
model A comparison with glucocorticoids. The Journal of Steroid Bio-
chemistry and Molecular Biology,233, 106373. https://doi.org/10.
1016/j.jsbmb.2023.106373
Hussain, A., Aslam, B., Muhammad, F., Faisal, M. N., Kousar, S.,
Mushtaq, A., & Bari, M. U. (2021). Anti-arthritic activity of Ricinus
communis L. and Withania somnifera L. extracts in adjuvant-induced
arthritic rats via modulating inflammatory mediators and subsiding oxi-
dative stress. Iranian Journal of Basic Medical Sciences,24, 951961.
https://doi.org/10.22038/ijbms.2021.55145.12355
Hussain, N., Mumtaz, M., Adil, M., Nadeem, A. A., Sarwar, A., Aziz, T.,
Alharbi, M., Alsahammari, A., Alasmari, A. F., & Alharbi, M. E. (2023).
Investigation of VEGF (rs 699947) polymorphism in the progression of
rheumatoid arthritis (RA) and in-silico nanoparticle drug delivery
of potential phytochemicals to cure RA. Acta Biochimica Polonica,70,
591598. https://doi.org/10.18388/abp.2020_6654
Ichikawa, H., & Aggarwal, B. B. (2006). Guggulsterone inhibits osteoclasto-
genesis induced by receptor activator of nuclear factor-κB ligand and
by tumor cells by suppressing nuclear factor-κB activation. Clinical
Cancer Research,12, 662668. https://doi.org/10.1158/1078-0432.
CCR-05-1749
Ilchovska, D., & Barrow, D. M. (2021). An overview of the NF-kB mecha-
nism of pathophysiology in rheumatoid arthritis, investigation of the
NF-kB ligand RANKL and related nutritional interventions. Autoimmu-
nity Reviews,20, 102741. https://doi.org/10.1016/j.autrev.2020.
102741
Inoue, K., & Yuasa, H. (2014). Molecular basis for pharmacokinetics and
pharmacodynamics of methotrexate in rheumatoid arthritis therapy.
Drug Metabolism and Pharmacokinetics,29,1219. https://doi.org/10.
2133/dmpk.dmpk-13-rv-119
Ishikawa, G., Kwon, C., & Fujii, Y. (2023). Comparison of the effects of
peficitinib and tofacitinib in the adjuvant-induced arthritis rat model.
European Journal of Pharmacology,941, 175490. https://doi.org/10.
1016/j.ejphar.2023.175490
Ishikawa, Y., & Terao, C. (2020). The impact of cigarette smoking on risk of
rheumatoid arthritis: A narrative review. Cells,9, 475. https://doi.org/
10.3390/cells9020475
Islam, S., Mir, A. R., Abidi, M., Talha, M., Zafar, A., Habib, S., & Moinuddin.
(2018). Methylglyoxal modified IgG generates autoimmune response
in rheumatoid arthritis. International Journal of Biological Macromole-
cules,118,1523. https://doi.org/10.1016/j.ijbiomac.2018.06.040
Ivashkiv, L. B., & Hu, X. (2003). The JAK/STAT pathway in rheumatoid
arthritis: Pathogenic or protective? Arthritis and Rheumatism,48,
20922096. https://doi.org/10.1002/art.11095
Jang, D. I., Lee, A. H., Shin, H. Y., Song, H. R., Park, J. H., Kang, T. B.,
Lee, S. R., & Yang, S. H. (2021). The role of tumor necrosis factor alpha
(Tnf-α) in autoimmune disease and current tnf-αinhibitors in therapeu-
tics. International Journal of Molecular Sciences,22.https://doi.org/10.
3390/ijms22052719
Jena, J., & Gupta, A. K. (2012). Ricinus communis Linn: A phytopharmacolo-
gical review. International Journal of Pharmacy and Pharmaceutical Sci-
ences,4,2529.
Ji, H., Ohmura, K., Mahmood, U., Lee, D. M., Hofhuis, F. M. A.,
Boackle, S. A., Takahashi, K., Holers, V. M., Walport, M., & Gerard, C.
(2002). Arthritis critically dependent on innate immune system players
disease in these animals is focused specifically on the joints but stems
from autoreactivity to a ubiquitously expressed antigen, glucose-6-
phosphate isomerase. Immunity,16, 157168. https://doi.org/10.
1016/s1074-7613(02)00275-3
Jiang, M., Zha, Q., He, Y., & Lu, A. (2012). Risk factors of gastrointestinal
and hepatic adverse drug reactions in the treatment of rheumatoid
arthritis with biomedical combination therapy and Chinese medicine.
Journal of Ethnopharmacology,141, 615621. https://doi.org/10.
1016/j.jep.2011.07.026
Jiao, Z., Hua, S., Wang, W., Wang, H. H., Gao, J., & Wang, X. (2013).
Increased circulating myeloid-derived suppressor cells correlated nega-
tively with Th17 cells in patients with rheumatoid arthritis. Scandina-
vian Journal of Rheumatology,42,8590. https://doi.org/10.3109/
03009742.2012.716450
Jo, H. G., Seo, J., & Lee, D. (2022). Clinical evidence construction of east
Asian herbal medicine for inflammatory pain in rheumatoid arthritis
CHATTERJEE ET AL.29
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
based on integrative data mining approach. Pharmacological Research,
185, 106460. https://doi.org/10.1016/j.phrs.2022.106460
Jumbo, A. (2021). Immunological Tolerance. Immunome Research,17(3), 1.
Jung, S. M., Park, Y. J., Park, K. S., & Kim, K. J. (2022). Clinical implications
of shared epitope and anti-citrullinated peptide antibody in patients
with rheumatoid arthritis. Journal of Rheumatic Diseases,29, 171180.
https://doi.org/10.4078/jrd.2022.29.3.171
Kadhim, H. J. (2023). The relationship between obesity and rheumatoid
arthritis. Research Review,6, 19141926. https://doi.org/10.52845/
CMRO/2023/6-12-3
Kamel, E. M., & Ahemd, S. A. (2013). Phenolic constituents and biological
activity of the genus Pluchea.Der Pharma Chemica,5, 109114.
Kamradt, T., & Mitchison, A. N. (2001). Tolerance and autoimmunity. The
New England Journal of Medicine,344, 655664. https://doi.org/10.
1056/NEJM200103013440907
Karami, J., Aslani, S., Tahmasebi, M. N., Mousavi, M. J., Sharafat Vaziri, A.,
Jamshidi, A., Farhadi, E., & Mahmoudi, M. (2020). Epigenetics in rheu-
matoid arthritis; fibroblast-like synoviocytes as an emerging paradigm
in the pathogenesis of the disease. Immunology and Cell Biology,98,
171186. https://doi.org/10.1111/imcb.12311
Kelmenson, L. B., Wagner, B. D., McNair, B. K., Frazer-Abel, A.,
Demoruelle, M. K., Bergstedt, D. T., Feser, M. L., Moss, L. K.,
Parish, M. C., Mewshaw, E. A., Mikuls, T. R., Edison, J. D.,
Holers, V. M., & Deane, K. D. (2020). Timing of elevations of autoanti-
body isotypes prior to diagnosis of rheumatoid arthritis. Arthritis and
Rheumatology,72, 251261. https://doi.org/10.1002/art.41091
Khandhar, A., Patel, S. G., Zaveri, M., Patel, S., & Patel, A. (2010). Chemistry
and pharmacology of Piper longum L. International Journal of Pharma-
ceutical Sciences Review and Research, 5(1), 6776.
Khushbu, C., Roshni, S., Anar, P., Carol, M., & Mayuree, P. (2011). Phyto-
chemical and therapeutic potential of Piper longum Linn: A review.
International Journal of Research in Ayurveda and Pharmacy,2, 157161.
Kiener, H. P., & Karonitsch, T. (2011). The synovium as a privileged site in
rheumatoid arthritis: Cadherin-11 as a dominant player in synovial
pathology. Best Practice & Research. Clinical Rheumatology,25, 767
777. https://doi.org/10.1016/j.berh.2011.11.012
Kim, S.-H., Lechman, E. R., Bianco, N., Menon, R., Keravala, A., Nash, J.,
Mi, Z., Watkins, S. C., Gambotto, A., & Robbins, P. D. (2005). Exosomes
derived from IL-10-treated dendritic cells can suppress inflammation
and collagen-induced arthritis. The Journal of Immunology,174, 6440
6448. https://doi.org/10.4049/jimmunol.174.10.6440
Kiss, M., Czimmerer, Z., & Nagy, L. (2013). The role of lipid-activated
nuclear receptors in shaping macrophage and dendritic cell function:
From physiology to pathology. Journal of Allergy and Clinical Immunol-
ogy,132, 264286. https://doi.org/10.1016/j.jaci.2013.05.044
Köhler, B. M., Günther, J., Kaudewitz, D., & Lorenz, H. M. (2019). Current
therapeutic options in the treatment of rheumatoid arthritis. Journal of
Clinical Medicine,8.https://doi.org/10.3390/jcm8070938
Konig, M. F., Abusleme, L., Reinholdt, J., Palmer, R. J., Teles, R. P.,
Sampson, K., Rosen, A., Nigrovic, P. A., Sokolove, J., Giles, J. T.,
Moutsopoulos, N. M., & Andrade, F. (2016). Aggregatibacter actinomy-
cetemcomitans-induced hypercitrullination links periodontal infection
to autoimmunity in rheumatoid arthritis. Science Translational Medicine,
8, 369ra176. https://doi.org/10.1126/scitranslmed.aaj1921
Kremer, J. M., Westhovens, R., Leon, M., Di Giorgio, E., Alten, R.,
Steinfeld, S., Russell, A., Dougados, M., Emery, P., Nuamah, I. F.,
Williams, G. R. D., Becker, J.-C., Hagerty, D. T., & Moreland, L. W.
(2003). Treatment of rheumatoid arthritis by selective inhibition of T-
cell activation with fusion protein CTLA4Ig. Boston Medical and Surgi-
cal Journal,349, 19071915. https://doi.org/10.1056/nejmoa035075
Kuligowska-Prusińska, M., & Odrowąż-Sypniewska, G. (2018). Laboratory
diagnostics and pathogenesis of rheumatoid arthritisThe past and
the present. Medical Research Journal,2, 128134. https://doi.org/10.
5603/mrj.2017.0017
Kumar, B. V., Connors, T. J., & Farber, D. L. (2018). Human T cell develop-
ment, localization, and function throughout life. Immunity,48, 202
213. https://doi.org/10.1016/j.immuni.2018.01.007
Kumar, R., Singh, S., Saksena, A., Pal, R., Jaiswal, R., & Kumar, R. (2019).
Effect of Boswellia serrata extract on acute inflammatory parameters
and tumor necrosis factor-αin complete Freund's adjuvant-induced
animal model of rheumatoid arthritis. International Journal of Applied &
Basic Medical Research,9, 100106. https://doi.org/10.4103/ijabmr.
ijabmr_248_18
Kumar, V., Al-Abbasi, F. A., Ahmed, D., Verma, A., Mujeeb, M., & Anwar, F.
(2015). Paederia foetida Linn. Inhibits adjuvant induced arthritis by sup-
pression of PGE2 and COX-2 expression via nuclear factor-κB. Food &
Function,6, 16521666. https://doi.org/10.1039/c5fo00178a
Kumar Vishwakarma, R., & Negi, D. S. (2020). The development of COX-1
and COX-2 inhibitors: A review. International Journal of Pharmaceutical
Sciences and Research,11, 3544. https://doi.org/10.13040/IJPSR.
0975-8232.11(8).3544-55
Kunnumakkara, A. B., Banik, K., Bordoloi, D., Harsha, C., Sailo, B. L.,
Padmavathi, G., Roy, N. K., Gupta, S. C., & Aggarwal, B. B. (2018). Goo-
gling the guggul (Commiphora and Boswellia) for prevention of chronic
diseases. Frontiers in Pharmacology,9.https://doi.org/10.3389/fphar.
2018.00686
Kunnumakkara, A. B., Rana, V., Parama, D., Banik, K., Girisa, S.,
Henamayee, S., Thakur, K. K., Dutta, U., Garodia, P., Gupta, S. C., &
Aggarwal, B. B. (2021). COVID-19, cytokines, inflammation, and
spices: How are they related? Life Sciences,284, 119201. https://doi.
org/10.1016/j.lfs.2021.119201
Kurkó, J., Vida, A., Glant, T. T., Scanzello, C. R., Katz, R. S., Nair, A.,
Mikecz, K., & Szekanecz, Z. (2014). Identification of myeloid-derived
suppressor cells in the synovial fluid of patients with rheumatoid
arthritis: A pilot study. BMC Musculoskeletal Disorders,15, 281.
https://doi.org/10.1186/1471-2474-15-281
Larid, G., Pancarte, M., Offer, G., Clavel, C., Martin, M., Pradel, V., Auger, I.,
Lafforgue, P., Roudier, J., Serre, G., & Balandraud, N. (2021). In rheuma-
toid arthritis patients, HLA-DRB1*04:01 and rheumatoid nodules are
associated with ACPA to a particular fibrin epitope. Frontiers in Immu-
nology,12, 692041. https://doi.org/10.3389/fimmu.2021.692041
Lautenbach, M., Millrose, M., Langner, I., & Eisenschenk, A. (2013). Results
of Mannerfelt wrist arthrodesis for rheumatoid arthritis in relation to
the position of the fused wrist. International Orthopaedics,37, 2409
2413. https://doi.org/10.1007/s00264-013-2063-6
Law, S. C., Benham, H., Reid, H. H., Rossjohn, J., & Thomas, R. (2014). Iden-
tification of self-antigen-specific T cells reflecting loss of tolerance in
autoimmune disease underpins preventative immunotherapeutic strat-
egies in rheumatoid arthritis. Rheumatic Disease Clinics of North Amer-
ica.,40, 735752. https://doi.org/10.1016/j.rdc.2014.07.015
Lee, C. K., Lee, E. Y., Kim, Y. G., Mun, S. H., Moon, H. B., & Yoo, B. (2008).
Alpha-lipoic acid inhibits TNF-αinduced NF-κB activation through
blocking of MEKK1-MKK4-IKK signaling cascades. International Immu-
nopharmacology,8, 362370. https://doi.org/10.1016/j.intimp.2007.
10.020
Lee, S. Y., Cho, S. S., Li, Y. C., Bae, C. S., Park, K. M., & Park, D. H. (2020).
Anti-inflammatory effect of Curcuma longa and Allium hookeri co-treat-
ment via NF-κB and COX-2 pathways. Scientific Reports,10, 5718.
https://doi.org/10.1038/s41598-020-62749-7
Lee, S.-Y., Kwok, S.-K., Son, H.-J., Ryu, J.-G., Kim, E.-K., Oh, H.-J., Cho, M.-
L., Ju, J. H., Park, S.-H., & Kim, H.-Y. (2013). IL-17-mediated Bcl-2
expression regulates survival of fibroblast-like synoviocytes in rheu-
matoid arthritis through STAT3 activation. Arthritis Research & Therapy,
15, R31. https://doi.org/10.1186/ar4179
Lee, Y. R., Hwang, J. K., Koh, H. W., Jang, K. Y., Lee, J. H., Park, J. W., &
Park, B. H. (2012). Sulfuretin, a major flavonoid isolated from Rhus ver-
niciflua, ameliorates experimental arthritis in mice. Life Sciences,90,
799807. https://doi.org/10.1016/j.lfs.2012.04.015
30 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Lee, Y. R., Lee, J. H., Noh, E. M., Kim, E. K., Song, M. Y., Jung, W. S.,
Park, S. J., Kim, J. S., Park, J. W., Kwon, K. B., & Park, B. H. (2008). Gug-
gulsterone blocks IL-1β-mediated inflammatory responses by suppres-
sing NF-κB activation in fibroblast-like synoviocytes. Life Sciences,82,
12031209. https://doi.org/10.1016/j.lfs.2008.04.006
Li, K., Shi, H., Zhang, B., Ou, X., Ma, Q., Chen, Y., Shu, P., Li, D., & Wang, Y.
(2021). Myeloid-derived suppressor cells as immunosuppressive regu-
lators and therapeutic targets in cancer. Signal Transduction and Tar-
geted Therapy,6, 362. https://doi.org/10.1038/s41392-021-00670-9
Li, Y., Guo, R., Oduro, P. K., Sun, T., Chen, H., Yi, Y., Zeng, W., Wang, Q.,
Leng, L., Yang, L., & Zhang, J. (2022). The relationship between Por-
phyromonas gingivalis and rheumatoid arthritis: A meta-analysis. Fron-
tiers in Cellular and Infection Microbiology,12, 956417. https://doi.org/
10.3389/fcimb.2022.956417
Li, Y. J., & Chen, Z. (2022). Cell-based therapies for rheumatoid arthritis:
Opportunities and challenges. Therapeutic Advances in Musculoskeletal
Disease,14, 1759720X2211002. https://doi.org/10.1177/
1759720X221100294
Lim, H. J., Bak, S. G., Park, E. J., Ku, S. K., Lee, S., Woong Lee, S., Min
Lee, K., Lee, S. J., & Rho, M. C. (2020). Retrofractamide C derived from
Piper longum alleviates xylene-induced mouse ear edema and inhibits
phosphorylation of ERK and NF-κB in LPS-induced J774A.1. Mole-
cules,25, 4058. https://doi.org/10.3390/molecules25184058
Liu, L. Y., Fei, X. W., Li, Z. M., Zhang, Z. H., & Mei, Y. A. (2005). Diclofenac,
a nonsteroidal anti-inflammatory drug, activates the transient outward
K+current in rat cerebellar granule cells. Neuropharmacology,48,
918926. https://doi.org/10.1016/j.neuropharm.2004.12.020
Liu, T., Zhang, L., Joo, D., & Sun, S. C. (2017). NF-κB signaling in inflamma-
tion. Signal Transduction and Targeted Therapy,2, 17023. https://doi.
org/10.1038/sigtrans.2017.23
Lojk, J., & Marc, J. (2021). Roles of non-canonical wnt signalling pathways
in bone biology. International Journal of Molecular Sciences,22.https://
doi.org/10.3390/ijms221910840
Lorzadeh, S., Kohan, L., Ghavami, S., & Azarpira, N. (2021). Autophagy and
the Wnt signaling pathway: A focus on Wnt/β-catenin signaling. Bio-
chimica et Biophysica Acta - Molecular Cell Research,1868, 118926.
https://doi.org/10.1016/j.bbamcr.2020.118926
Luís, M., Freitas, J., Costa, F., Buttgereit, F., Boers, M., Jap, D. S., &
Santiago, T. (2019). An updated review of glucocorticoid-related
adverse events in patients with rheumatoid arthritis. Expert Opinion on
Drug Safety,18, 581590. https://doi.org/10.1080/14740338.2019.
1615052
Luque-Campos, N., Contreras-López, R. A., Paredes-Martínez, M. J.,
Torres, M. J., Bahraoui, S., Wei, M., Espinoza, F., Djouad, F., Elizondo-
Vega, R. J., & Luz-Crawford, P. (2019). Mesenchymal stem cells
improve rheumatoid arthritis progression by controlling memory T cell
response. Frontiers in Immunology,10, 798. https://doi.org/10.3389/
fimmu.2019.00798
Luu, V. P., Vazquez, M. I., & Zlotnik, A. (2014). B cells participate in toler-
ance and autoimmunity through cytokine production. Autoimmunity,
47,112. https://doi.org/10.3109/08916934.2013.856006
Lv, X., Wang, L., Zou, X., & Huang, S. (2021). Umbilical cord mesenchymal
stem cell therapy for regenerative treatment of rheumatoid arthritis:
Opportunities and challenges. Drug Design, Development and Therapy,
15, 39273936. https://doi.org/10.2147/DDDT.S323107
Ma, W., Chen, M., Kang, H., Steinhart, Z., Angers, S., He, X., &
Kirschner, M. W. (2020). Single-molecule dynamics of Dishevelled at
the plasma membrane and Wnt pathway activation. The Proceedings of
the National Academy of Sciences,117, 1669016701. https://doi.org/
10.1073/pnas.1910547117/-/DCSupplemental
Mahdi, J. G. (2010). Medicinal potential of willow: A chemical perspective
of aspirin discovery. Journal of Saudi Chemical Society,14, 317322.
https://doi.org/10.1016/j.jscs.2010.04.010
Mahmoud, D. E., Kaabachi, W., Sassi, N., Mokhtar, A., Moalla, M.,
Ammar, L. B., Jemmali, S., Rekik, S., Tarhouni, L., Kallel-Sellami, M.,
Cheour, E., & Laadhar, L. (2021). SFRP5 enhances Wnt5a induced-
inflammation in rheumatoid arthritis fibroblast-like synoviocytes. Fron-
tiers in Immunology,12, 663683. https://doi.org/10.3389/fimmu.
2021.663683
Malemud, C. J. (2015). The PI3K/Akt/PTEN/mTOR pathway: A fruitful tar-
get for inducing cell death in rheumatoid arthritis? Future Medicinal
Chemistry,7, 11371147. https://doi.org/10.4155/fmc.15.55
Manan, M., Saleem, U., Hamid Akash, M. S., Qasim, M., Hayat, M.,
Raza, Z., & Ahmad, B. (2020). Antiarthritic potential of comprehen-
sively standardized extract of alternanthera bettzickiana: In vitro and
in vivo studies. ACS Omega,5, 1947819496. https://doi.org/10.
1021/acsomega.0c01670
Maranini, B., Bortoluzzi, A., Silvagni, E., & Govoni, M. (2022). Focus on sex
and gender: What we need to know in the management of rheumatoid
arthritis. Journal of Personalized Medicine,12, 499. https://doi.org/10.
3390/jpm12030499
Marian, M., Shah, R., Gashi, B., Zhang, S., Bhavnani, K., Wartzack, S., &
Rosenkranz, A. (2021). Exploring the lubrication mechanisms of synovial
fluids for joint longevity A perspective. Colloids and Surfaces. B, Bioin-
terfaces,206, 111926. https://doi.org/10.1016/j.colsurfb.2021.111926
Masoumi, M., Bashiri, H., Khorramdelazad, H., Barzaman, K., Hashemi, N.,
Sereshki, H. A., Sahebkar, A., & Karami, J. (2021). Destructive roles of
fibroblast-like synoviocytes in chronic inflammation and joint damage
in rheumatoid arthritis. Inflammation,44, 466479. https://doi.org/10.
1007/s10753-020-01371-1
Masoumi, M., Mehrabzadeh, M., Mahmoudzehi, S., Mousavi, M. J.,
Jamalzehi, S., Sahebkar, A., & Karami, J. (2020). Role of glucose metab-
olism in aggressive phenotype of fibroblast-like synoviocytes: Latest
evidence and therapeutic approaches in rheumatoid arthritis. Interna-
tional Immunopharmacology,89, 107064. https://doi.org/10.1016/j.
intimp.2020.107064
Mathebula, E. M., Sengupta, D., Govind, N., Laufer, V. A., Bridges, S. L.,
Tikly, M., Ramsay, M., & Choudhury, A. (2022). A genome-wide associ-
ation study for rheumatoid arthritis replicates previous HLA and non-
HLA associations in a cohort from South Africa. Human Molecular
Genetics,31, 42864294. https://doi.org/10.1093/hmg/ddac178
Meng, X., Layhadi, J. A., Keane, S. T., Cartwright, N. J. K., Durham, S. R., &
Shamji, M. H. (2023). Immunological mechanisms of tolerance: Central,
peripheral and the role of T and B cells. Asia Pacific Association of
Allergy, Asthma and Clinical Immunology,13, 175186. https://doi.org/
10.5415/apallergy.0000000000000128
Meselhy, M. R. (2003). Inhibition of LPS-induced NO production by the
oleogum resin of Commiphora wightii and its constituents. Phytochem-
istry,62, 213218. https://doi.org/10.1016/s0031-9422(02)00388-6
Min, H. K., Won, J. Y., Kim, B. M., Lee, K. A., Lee, S. J., Lee, S. H.,
Kim, H. R., & Kim, K. W. (2020). Interleukin (IL)-25 suppresses IL-22-
induced osteoclastogenesis in rheumatoid arthritis via STAT3 and p38
MAPK/IκBαpathway. Arthritis Research & Therapy,22, 222. https://
doi.org/10.1186/s13075-020-02315-8
Miossec, P. (2022). The role of B cells and their interactions with stromal
cells in the context of inflammatory autoimmune diseases. Autoimmu-
nity Reviews,21, 103098. https://doi.org/10.1016/j.autrev.2022.
103098
Mitra, R. (2011). Adverse effects of corticosteroids on bone metabolism: A
review. PM and R,3, 466471. https://doi.org/10.1016/j.pmrj.2011.
02.017
Mohammadian Haftcheshmeh, S., & Momtazi-Borojeni, A. A. (2020).
Immunomodulatory therapeutic effects of curcumin in rheumatoid
arthritis. Autoimmunity Reviews,19, 102593. https://doi.org/10.1016/
j.autrev.2020.102593
Mohammed, A., Ali, A., Elamin, M., El-Nour, M., Mohammad, O., &
Yagi, S. M. (2019). In vitro anti-inflammatory activity of ginger (Zingiber
officinale Rosc.) rhizome, callus and callus treated with some elicitors.
Journal of Medicinal Plants Research,13, 227235. https://doi.org/10.
5897/JMPR2019.6758
CHATTERJEE ET AL.31
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Molendijk, M., Hazes, J. M. W., & Lubberts, E. (2018). From patients with
arthralgia, pre-RA and recently diagnosed RA: What is the current sta-
tus of understanding RA pathogenesis? RMD Open,4, e000256.
https://doi.org/10.1136/rmdopen-2016-000256
Mollard, E., Pedro, S., Chakravarty, E., Clowse, M., Schumacher, R., &
Michaud, K. (2018). The impact of menopause on functional status in
women with rheumatoid arthritis. Rheumatology (Oxford),57, 798
802. https://doi.org/10.1093/rheumatology/kex526
Moon, J. E., Kim, D. M., & Kim, J. Y. (2015). Anti-inflammatory effect of
Rhus verniciflua stokes extract in the murine macrophage cell line,
Raw264.7. Journal of Korean Society for Applied Biological Chemistry,
58, 481486. https://doi.org/10.1007/s13765-015-0065-3
Morrison, D. K. (2012). MAP kinase pathways. Cold Spring Harbor Perspec-
tives in Biology,4, a011254. https://doi.org/10.1101/cshperspect.
a011254
Mousavi, M. J., Karami, J., Aslani, S., Tahmasebi, M. N., Vaziri, A. S.,
Jamshidi, A., Farhadi, E., & Mahmoudi, M. (2021). Transformation of
fibroblast-like synoviocytes in rheumatoid arthritis; from a friend to
foe. Autoimmunity Highlights,12,3.https://doi.org/10.1186/s13317-
020-00145-x
Mumtaz, M., & Hussain, N. (2020). Rheumatoid arthritis and the role of
VEGF gene: An overview. Journal of Scientific Research in Medical and
Biological Sciences,1,7590. https://doi.org/10.47631/jsrmbs.v1i2.93
Murugesan, S., Venkateswaran, M. R., Jayabal, S., & Periyasamy, S. (2020).
Evaluation of the antioxidant and anti-arthritic potential of Zingiber
officinale Rosc. by in vitro and in silico analysis. South African Journal of
Botany,130,4553. https://doi.org/10.1016/j.sajb.2019.12.019
Nemudzivhadi, V., & Masoko, P. (2014). In vitro assessment of cytotoxic-
ity, antioxidant, and anti-inflammatory activities of Ricinus communis
(euphorbiaceae) leaf extracts. Evidence-Based Complementary and
Alternative Medicine,2014, 625961. https://doi.org/10.1155/2014/
625961
Ngo, S. T., Steyn, F. J., & McCombe, P. A. (2014). Gender differences in
autoimmune disease. Frontiers in Neuroendocrinology,35, 347369.
https://doi.org/10.1016/j.yfrne.2014.04.004
Ni, Q., Pham, N. B., Meng, W. S., Zhu, G., & Chen, X. (2019). Advances in
immunotherapy of type I diabetes. Advanced Drug Delivery Reviews,
139,8391. https://doi.org/10.1016/j.addr.2018.12.003
Nicolau, J., Lequerré, T., Bacquet, H., & Vittecoq, O. (2017). Rheumatoid
arthritis, insulin resistance, and diabetes. Joint Bone Spine,84, 411
416. https://doi.org/10.1016/j.jbspin.2016.09.001
Nieto-Colonia, A. M., Santos, W. S., Keusseyan, S. P., Caldana, W.,
Fernandes, A. R. C., & Andrade, L. E. C. (2008). Antibodies to citrulli-
nated peptides are not associated with the rate of joint destruction in
patients with a well-established diagnosis of rheumatoid arthritis. Bra-
zilian Journal of Medical and Biological Research,41, 188192. https://
doi.org/10.1590/S0100-879X2008005000005
Nygaard, G., & Firestein, G. S. (2020). Restoring synovial homeostasis in
rheumatoid arthritis by targeting fibroblast-like synoviocytes. Nature
Reviews Rheumatology,16, 316333. https://doi.org/10.1038/
s41584-020-0413-5
Oshi, M. A., Haider, A., Siddique, M. I., Zeb, A., Jamal, S. B.,
Khalil, A. A. K., & Naeem, M. (2022). Nanomaterials for chronic inflam-
matory diseases: The current status and future prospects. Applied
Nanoscience (Switzerland),12, 30973110. https://doi.org/10.1007/
s13204-021-02019-8
Owen, D. L., Sjaastad, L. E., & Farrar, M. A. (2019). Regulatory T cell devel-
opment in the thymus. The Journal of Immunology,203, 20312041.
https://doi.org/10.4049/jimmunol.1900662
Öz, B., Orhan, C., Tuzcu, M., Şahin, N., Özercan, _
I. H., Demirel Öner, P.,
Koca, S. S., Juturu, V., & Şahin, K. (2021). Ginger extract suppresses
the activations of NF-κB and Wnt pathways and protects inflamma-
tory arthritis. Anatolian Journal of Cardiology,8, 196201. https://doi.
org/10.5152/eujrheum.2020.20192
Padyukov, L. (2022). Genetics of rheumatoid arthritis. Seminars in Immuno-
pathology,44,4762. https://doi.org/10.1007/s00281-022-00912-0
Padyukov, L., Suva, C., Stolt, P., Alfredsson, L., & Klareskog, L. (2004). A
gene-environment interaction between smoking and shared epitope
genes in HLA-DR provides a high risk of seropositive rheumatoid
arthritis. Arthritis and Rheumatism,50, 30853092. https://doi.org/10.
1002/art.20553
Page, A., Fusil, F., & Cosset, F. L. (2021). Antigen-specific tolerance
approach for rheumatoid arthritis: Past, present and future. Joint, Bone,
Spine,88, 105164. https://doi.org/10.1016/j.jbspin.2021.105164
Pandey, P., & Tiwari, S. (2021). Therapeutic role of Ricinus communis
L. root bioactive compounds in rheumatoid arthritis. International Jour-
nal of Pharmacy and Biological Sciences,11,99104. https://doi.org/
10.21276/ijpbs.2021.11.1.13
Park, Y., & Kwok, S. K. (2022). Recent advances in cell therapeutics for sys-
temic autoimmune diseases. Immune Network,22, e10. https://doi.
org/10.4110/in.2022.22.e10
Petrovská, N., Prajzlerová, K., Vencovský, J., ˇ
Senolt, L., & Filková, M.
(2021). The pre-clinical phase of rheumatoid arthritis: From risk factors
to prevention of arthritis. Autoimmunity Reviews,20, 102797. https://
doi.org/10.1016/j.autrev.2021.102797
Piątczak, E., Dybowska, M., Płuciennik, E., Ko
sla, K., Kolniak-ostek, J., &
Kalinowska-lis, U. (2020). Identification and accumulation of phenolic
compounds in the leaves and bark of Salix alba (L.) and their biological
potential. Biomolecules,10,117. https://doi.org/10.3390/
biom10101391
Poudel, D., George, M. D., & Baker, J. F. (2020). The impact of obesity on
disease activity and treatment response in rheumatoid arthritis. Cur-
rent Rheumatology Reports,22, 56. https://doi.org/10.1007/s11926-
020-00933-4
Pradhan, R., Singhvi, G., Dubey, S. K., Gupta, G., & Dua, K. (2019). MAPK
pathway: A potential target for the treatment of non-small-cell lung
carcinoma. Future Medicinal Chemistry,11, 793795. https://doi.org/
10.4155/fmc-2018-0468
Prathap Kumar, J., Arun Kumar, M., & Venkatesh, D. (2020). Healthy gait:
Review of anatomy and physiology of knee joint. International Journal
of Current Research and Review,12,18. doi:10.31782/IJCRR.2020.
12061
Qian, K., Zheng, X. X., Wang, C., Huang, W. G., Liu, X. B., Di Xu, S.,
Liu, D. K., Liu, M. Y., & Lin, C. S. (2022). β-Sitosterol inhibits rheuma-
toid synovial angiogenesis through suppressing VEGF signaling path-
way. Frontiers in Pharmacology,12, 816477. https://doi.org/10.3389/
fphar.2021.816477
Radu, A. F., & Bungau, S. G. (2021). Management of rheumatoid arthritis:
An overview. Cells,10, 2587. https://doi.org/10.3390/cells10112857
Radu, A. F., Bungau, S. G., Negru, P. A., Marcu, M. F., & Andronie-
Cioara, F. L. (2022). In-depth bibliometric analysis and current scien-
tific mapping research in the context of rheumatoid arthritis pharma-
cotherapy. Biomedicine and Pharmacotherapy,154, 113614. https://
doi.org/10.1016/j.biopha.2022.113614
Raine, C., & Giles, I. (2022). What is the impact of sex hormones on the
pathogenesis of rheumatoid arthritis? Frontiers in Medicine,9, 909879.
Ramachandran, A., Sharma, U. R., Haripriya, R. L., Sulthana, S., Borah, R.,
Taj, N., & Mudagal, M. P. (2023). Menopausal transition and the devel-
opment of rheumatoid arthritis: An overview. Defence Life Science
Journal,8, 248254. https://doi.org/10.14429/dlsj.8.18335
Ramadass, V., Vaiyapuri, T., & Tergaonkar, V. (2020). Small molecule NF-
κB pathway inhibitors in clinic. International Journal of Molecular Sci-
ences,21,143. https://doi.org/10.3390/ijms21145164
Ramiro, S., Gaujoux-Viala, C., Nam, J. L., Smolen, J. S., Buch, M., Gossec, L.,
Van Der Heijde, D., Winthrop, K., & Landewé, R. (2014). Safety of syn-
thetic and biological DMARDs: A systematic literature review inform-
ing the 2013 update of the EULAR recommendations for management
of rheumatoid arthritis. Annals of the Rheumatic Diseases,73, 529535.
https://doi.org/10.1136/annrheumdis-2013-204575
Rani, R., & Mishra, S. (2013). Phytochemistry of Guggul (Commiphora
wightii): A review. Asian Journal of Research in Chemistry,6, 413424.
https://doi.org/10.5958/0974-4150
32 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Rapalli, V. K., Kaul, V., Gorantla, S., Waghule, T., Dubey, S. K.,
Pandey, M. M., & Singhvi, G. (2020). UV spectrophotometric method
for characterization of curcumin loaded nanostructured lipid
nanocarriers in simulated conditions: Method development, in-vitro
and ex-vivo applications in topical delivery. Spectrochimica Acta.
Part A, Molecular and Biomolecular Spectroscopy,224, 117392. https://
doi.org/10.1016/j.saa.2019.117392
Rapalli, V. K., Waghule, T., Gorantla, S., Dubey, S. K., Saha, R. N., &
Singhvi, G. (2020). Psoriasis: Pathological mechanisms, current phar-
macological therapies, and emerging drug delivery systems. Drug Dis-
covery Today,25, 22122226. https://doi.org/10.1016/j.drudis.2020.
09.023
Rim, E. Y., Clevers, H., & Nusse, R. (2022). The Wnt pathway: From signal-
ing mechanisms to synthetic modulators. Annual Review of Biochemis-
try,91, 571598. https://doi.org/10.1146/annurev-biochem-040320
Rondanelli, M., Perdoni, F., Peroni, G., Caporali, R., Gasparri, C., Riva, A.,
Petrangolini, G., Faliva, M. A., Infantino, V., Naso, M., Perna, S., &
Rigon, C. (2021). Ideal food pyramid for patients with rheumatoid
arthritis: A narrative review. Clinical Nutrition,40, 661689. https://
doi.org/10.1016/j.clnu.2020.08.020
Sabir, S. M., Zeb, A., Mahmood, M., Abbas, S. R., Ahmad, Z., & Iqbal, N.
(2021). Phytochemical analysis and biological activities of ethanolic
extract of curcuma longa rhizome. Brazilian Journal of Biology,81, 737
740. https://doi.org/10.1590/1519-6984.230628
Sam, S. (2019). Importance and effectiveness of herbal medicines. Journal
of Pharmacognosy and Phytochemistry,8, 354357.
Sánchez, M. A. S., & Flores, M. R. (2023). WNT signaling in stem cells: A
look into the non-canonical pathway. Stem Cell Reviews and Reports,
20,5266. https://doi.org/10.1007/s12015-023-10610-5
Sardana, Y., Bhatti, G. K., Singh, C., Sharma, P. K., Reddy, P. H., &
Bhatti, J. S. (2023). Progression of pre-rheumatoid arthritis to clinical
disease of joints: Potential role of mesenchymal stem cells. Life Sci-
ences,321, 121641. https://doi.org/10.1016/j.lfs.2023.121641
Sarup, P., Bala, S., & Kamboj, S. (2015). Pharmacology and phytochemistry
of oleo-gum resin of Commiphora wightii (Guggulu). Scientifica (Cairo),
2015,114. https://doi.org/10.1155/2015/138039
Sawant, R. S., & Godghate, A. G. (2013). Qualitative phytochemical screen-
ing of rhizomes of Curcuma longa linn. International Journal of Environ-
mental Science and Technology,2, 634641.
Schaller, M., Burton, D. R., & Ditzel, H. J. (2001). Autoantibodies to GPI in
rheumatoid arthritis: Linkage between an animal model and human
disease. Nature Immunology,2, 746753. https://doi.org/10.1038/
90696
Scherer, H. U., Häupl, T., & Burmester, G. R. (2020). The etiology of rheu-
matoid arthritis. Journal of Autoimmunity,110, 102400. https://doi.
org/10.1016/j.jaut.2019.102400
Scott, D. L. (2012). Biologics-based therapy for the treatment of rheuma-
toid arthritis. Clinical Pharmacology and Therapeutics,91,3043.
https://doi.org/10.1038/clpt.2011.278
Shah, L., Elshaikh, A. O., Lee, R., Joy Mathew, C., Jose, M. T., &
Cancarevic, I. (2020). Do menopause and aging affect the onset and
progression of rheumatoid arthritis and systemic lupus erythemato-
sus? Cureus,12, e10944. https://doi.org/10.7759/cureus.10944
Shankar Pandey, P., & Trigunayat, A. (2018). Anti-inflammatory and anti-
arthritic activities of the mixture of flavonoids isolated from Pluchea
lanceolata.International Journal of Pharmacy and Pharmaceutical Sci-
ences,13,3743.
Shara, M., & Stohs, S. J. (2015). Efficacy and safety of white willow bark
(Salix alba) extracts. Phytotherapy Research,29, 11121116. https://
doi.org/10.1002/ptr.5377
Sharma, A., Gajbhiye, V., & Kharya, M. D. (2007). Phytochemical profile of
Boswellia serrata: An overview. Pharmacognosy Reviews,1, 137142.
Sharma, A., Gajbhiye, V., Namdeo, A., & Vidyapeeth, B. (2009). Phyto-
chemical and pharmacological investigations on Boswellia serrata.Phar-
macognosy Reviews,3, 206215.
Sharma, M., & Pruitt, K. (2020). Wnt pathway: An integral hub for develop-
mental and oncogenic signaling networks. International Journal of
Molecular Sciences,21.https://doi.org/10.3390/ijms21218018
Sharma, R. A., Gescher, A. J., & Steward, W. P. (2005). Curcumin: The story
so far. European Journal of Cancer,41, 19551968. https://doi.org/10.
1016/j.ejca.2005.05.009
Sharma, S., Sahu, D., Das, H. R., & Sharma, D. (2011). Amelioration of colla-
gen-induced arthritis by Salix nigra bark extract via suppression of pro-
inflammatory cytokines and oxidative stress. Food and Chemical Toxi-
cology,49, 33953406. https://doi.org/10.1016/j.fct.2011.08.013
Sharma, S. K., & Goyal, N. (2011). Biological studies of the plants from
genus Pluchea.Annals of Biological Research,2,2534.
Shaw, P., Dwivedi, S. K. D., Bhattacharya, R., Mukherjee, P., & Rao, G.
(2024). VEGF signaling: Role in angiogenesis and beyond. Biochimica et
Biophysica Acta. Reviews on Cancer,1879, 189079. https://doi.org/10.
1016/j.bbcan.2024.189079
Shekhawat, M., Sisodia, S. S., & Shivavedi, N. (2021). Anti-inflammatory
and diuretic activity of Commiphora wightii and Vanda roxburghii.Jour-
nal of Critical Reviews,8, 317333.
Shevyrev, D., Tereshchenko, V., Kozlov, V., & Sennikov, S. (2022). Phylog-
eny, structure, functions, and role of AIRE in the formation of T-cell
subsets. Cells,11, 194. https://doi.org/10.3390/cells11020194
Shimada, A., Kobayashi, T., Ito, S., Okada, M., Murasawa, A.,
Nakazono, K., & Yoshie, H. (2016). Expression of anti-Porphyromonas
gingivalis peptidylarginine deiminase immunoglobulin G and peptidylar-
ginine deiminase-4 in patients with rheumatoid arthritis and periodon-
titis. Journal of Periodontal Research,51, 103111. https://doi.org/10.
1111/jre.12288
Siddiqui, M. Z. (2011). Boswellia serrata, a potential antiinflammatory
agent: An overview. Indian Journal of Pharmaceutical Sciences,73,
255261. https://doi.org/10.4103/0250-474X.93507
Simmen, B. R., Bogoch, E. R., & Goldhahn, J. (2008). Surgery insight: Ortho-
pedic treatment options in rheumatoid arthritis. Nature Clinical Prac-
tice. Rheumatology,4, 266273. https://doi.org/10.1038/
ncprheum0767
Simon, L. S., Taylor, P. C., Choy, E. H., Sebba, A., Quebe, A., Knopp, K. L., &
Porreca, F. (2021). The Jak/STAT pathway: A focus on pain in rheuma-
toid arthritis. Seminars in Arthritis and Rheumatism,51, 278284.
https://doi.org/10.1016/j.semarthrit.2020.10.008
Singh, A., Behl, T., Sehgal, A., Singh, S., Sharma, N., Naved, T., Bhatia, S., Al-
Harrasi, A., Chakrabarti, P., Aleya, L., Vargas-De-La-Cruz, C., &
Bungau, S. (2021). Mechanistic insights into the role of B cells in rheu-
matoid arthritis. International Immunopharmacology,99, 108078.
https://doi.org/10.1016/j.intimp.2021.108078
Singh, V., Sheikh, A., Kesharwani, P., Rajak, H., Talegaonkar, S.,
Hamid, H., & Sarwar Alam, M. (2022). Combination drug delivery
approaches in rheumatoid arthritis. In P. Kesharwani (Ed.), Combination
drug delivery approach as an effective therapy for various diseases
(pp. 81106). Elsevier. https://doi.org/10.1016/B978-0-323-85873-
1.00012-5
Singhvi, G., Manchanda, P., Krishna Rapalli, V., Kumar Dubey, S.,
Gupta, G., & Dua, K. (2018). MicroRNAs as biological regulators in skin
disorders. Biomedicine and Pharmacotherapy,108, 9961004. https://
doi.org/10.1016/j.biopha.2018.09.090
Skoczyńska, M., & Swierkot, J. (2018). The role of diet in rheumatoid
arthritis. Reumatologia,56, 259267. https://doi.org/10.5114/reum.
2018.77979
Smith, M. D., & Wechalekar, M. D. (2015). The synovium (6th ed. Rheuma-
tology). Elsevier.
Smolen, J. S., Landewé, R. B. M., Bijlsma, J. W. J., Burmester, G. R.,
Dougados, M., Kerschbaumer, A., McInnes, I. B., Sepriano, A., Van
Vollenhoven, R. F., De Wit, M., Aletaha, D., Aringer, M., Askling, J.,
Balsa, A., Boers, M., Den Broeder, A. A., Buch, M. H., Buttgereit, F.,
Caporali, R., Van Der Heijde, D. (2020). EULAR recommendations
for the management of rheumatoid arthritis with synthetic and
CHATTERJEE ET AL.33
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
biological disease-modifying antirheumatic drugs: 2019 update. Annals
of the Rheumatic Diseases,79, S685S699. https://doi.org/10.1136/
ard-2022-223356
Srivastava, P., & Shanker, K. (2012). Pluchea lanceolata (Rasana): Chemical
and biological potential of Rasayana herb used in traditional system of
medicine. Fitoterapia,83, 13711385. https://doi.org/10.1016/j.
fitote.2012.07.008
Steiner, G., & Smolen, J. (2002). Autoantibodies in rheumatoid arthritis and
their clinical significance. Arthritis Research & Therapy,4,S1S5.
https://doi.org/10.1186/ar551
Stolina, M., Bolon, B., Middleton, S., Dwyer, D., Brown, H., Duryea, D.,
Zhu, L., Rohner, A., Pretorius, J., Kostenuik, P., Feige, U., & Zack, D.
(2009). The evolving systemic and local biomarker milieu at different
stages of disease progression in rat adjuvant-induced arthritis. Journal
of Clinical Immunology,29, 158174. https://doi.org/10.1007/
s10875-008-9238-8
Stone, S., Malanga, G. A., & Capella, T. (2021). Corticosteroids: Review of
the history, the effectiveness, and adverse effects in the treatment of
joint Pain. Pain Physician,24, 233246.
St-Pierre, A., Desrosiers, V., Dufresne, F., & Blier, P. U. (2018). Anti-inflam-
matory properties of natural ingredients used in combinations on adju-
vant induced arthritis in rats. bioRxiv.https://doi.org/10.1101/325365
Strand, V., Kimberly, R., & Isaacs, J. D. (2007). Biologic therapies in rheu-
matology: Lessons learned, future directions. Nature Reviews. Drug Dis-
covery,6,7592. https://doi.org/10.1038/nrd2196
Su, Y., Liu, Y., Ma, C., Guan, C., Ma, X., & Meng, S. (2021). Mesenchymal
stem cell-originated exosomal lncRNA HAND2-AS1 impairs rheuma-
toid arthritis fibroblast-like synoviocyte activation through miR-143-
3p/TNFAIP3/NF-κB pathway. Journal of Orthopaedic Surgery and
Research,16, 116. https://doi.org/10.1186/s13018-021-02248-1
Sun, J., Xu, P., Du, X., Zhang, Q., & Zhu, Y. (2015). Piperlongumine attenu-
ates collagen-induced arthritis via expansion of myeloid-derived sup-
pressor cells and inhibition of the activation of fibroblast-like
synoviocytes. Molecular Medicine Reports,11, 26892694. https://doi.
org/10.3892/mmr.2014.3001
Sundarrajan, M., Boyle, D. L., Chabaud-Riou, M., Hammaker, D., &
Firestein, G. S. (2003). Expression of the MAPK kinases MKK-4 and
MKK-7 in rheumatoid arthritis and their role as key regulators of JNK.
Arthritis and Rheumatism,48, 24502460. https://doi.org/10.1002/
art.11228
Tak, P. P., & Kalden, J. R. (2011). Advances in rheumatology: New targeted
therapeutics. Arthritis Research & Therapy,13, S5. https://doi.org/10.
1186/1478-6354-13-S1-S5
Tamadaho, R. S. E., Hoerauf, A., & Layland, L. E. (2018). Immunomodula-
tory effects of myeloid-derived suppressor cells in diseases: Role in
cancer and infections. Immunobiology,223, 432442. https://doi.org/
10.1016/j.imbio.2017.07.001
Tanaka, Y., Luo, Y., O'Shea, J. J., & Nakayamada, S. (2022). Janus kinase-
targeting therapies in rheumatology: A mechanisms-based approach.
Nature Reviews Rheumatology,18, 133145. https://doi.org/10.1038/
s41584-021-00726-8
Tang, B., Shi, H., Alfredsson, L., Klareskog, L., Padyukov, L., & Jiang, X.
(2021). Obesity-related traits and the development of rheumatoid
arthritis: Evidence from genetic data. Arthritis and Rheumatology,73,
203211. https://doi.org/10.1002/art.41517
Tarp, S., Furst, D. E., Boers, M., Luta, G., Bliddal, H., Tarp, U.,
Asmussen, K. H., Brock, B., Dossing, A., Jørgensen, T. S.,
Thirstrup, S., & Christensen, R. (2017). Risk of serious adverse effects
of biological and targeted drugs in patients with rheumatoid arthritis:
A systematic review meta-analysis. Rheumatology (United Kingdom),56,
417425. https://doi.org/10.1093/rheumatology/kew442
Theofilopoulos, A. N., Kono, D. H., & Baccala, R. (2017). The multiple path-
ways to autoimmunity. Nature Immunology,18, 716724. https://doi.
org/10.1038/ni.3731
Trillo, A. R., Mosquera, N., Pena, C., Rivas-Tobío, F., Mera-Varela, A.,
Gonzalez, A., & Conde, C. (2020). Non-canonical WNT5A signaling
through RYK contributes to aggressive phenotype of the rheumatoid
fibroblast-like synoviocytes. Frontiers in Immunology,11, 555245.
https://doi.org/10.3389/fimmu.2020.555245
Ucar, O., & Rattay, K. (2015). Promiscuous gene expression in the thymus:
A matter of epigenetics, miRNA, and more? Frontiers in Immunology,6,
93. https://doi.org/10.3389/fimmu.2015.00093
Umar, S., Umar, K., Sarwar, A. H. M. G., Khan, A., Ahmad, N., Ahmad, S.,
Katiyar, C. K., Husain, S. A., & Khan, H. A. (2014). Boswellia serrata
extract attenuates inflammatory mediators and oxidative stress in col-
lagen induced arthritis. Phytomedicine,21, 847856. https://doi.org/
10.1016/j.phymed.2014.02.001
Umar, S., Zargan, J., Umar, K., Ahmad, S., Katiyar, C. K., & Khan, H. A.
(2012). Modulation of the oxidative stress and inflammatory cytokine
response by thymoquinone in the collagen induced arthritis in Wistar
rats. Chemico-Biological Interactions,197,4046. https://doi.org/10.
1016/j.cbi.2012.03.003
van de Veen, W., Stanic, B., Wirz, O. F., Jansen, K., Globinska, A., &
Akdis, M. (2016). Role of regulatory B cells in immune tolerance to
allergens and beyond. Journal of Allergy and Clinical Immunology,138,
654665. https://doi.org/10.1016/j.jaci.2016.07.006
van Delft, M. A. M., & Huizinga, T. W. J. (2020). An overview of autoanti-
bodies in rheumatoid arthritis. Journal of Autoimmunity,110, 102392.
https://doi.org/10.1016/j.jaut.2019.102392
Van Le, T. H., & Kwon, S. M. (2021). Vascular endothelial growth factor
biology and its potential as a therapeutic target in rheumatic diseases.
International Journal of Molecular Sciences,22, 5387. https://doi.org/
10.3390/ijms22105387
van Loo, G., & Bertrand, M. J. M. (2023). Death by TNF: A road to inflam-
mation. Nature Reviews. Immunology,23, 289303. https://doi.org/10.
1038/s41577-022-00792-3
Vishwanathan, A. S., & Basavaraju, R. (2010). A review on Vitex negundo
L. A medicinally important plant. EJBS,3,3042.
Vonkeman, H. E., & van de Laar, M. A. F. J. (2010). Nonsteroidal anti-
inflammatory drugs: Adverse effects and their prevention. Seminars in
Arthritis and Rheumatism,39, 294312. https://doi.org/10.1016/j.
semarthrit.2008.08.001
Vossenaar, E. R., Nijenhuis, S., Helsen, M. M. A., Van der Heijden, A.,
Senshu, T., Van den Berg, W. B., Van Venrooij, W. J., &
Joosten, L. A. B. (2003). Citrullination of synovial proteins in murine
models of rheumatoid arthritis. Arthritis and Rheumatism,48, 2489
2500. https://doi.org/10.1002/art.11229
Waghule, T., Gorantla, S., Rapalli, V. K., Shah, P., Dubey, S. K., Saha, R. N., &
Singhvi, G. (2020). Emerging trends in topical delivery of curcumin
through lipid nanocarriers: Effectiveness in skin disorders. AAPS
PharmSciTech,21, 284. https://doi.org/10.1208/s12249-020-01831-9
Waldmann, H. (2016). Mechanisms of immunological tolerance. Clinical
Biochemistry,49, 324328. https://doi.org/10.1016/j.clinbiochem.
2015.05.019
Wang, L., Jiang, Y., Han, T., Zheng, C., & Qin, L. (2014). A phytochemical,
pharmacological and clinical profile of Paederia foetida and P. scandens.
Natural Product Communications,9, 879886. https://doi.org/10.
1177/1934578X1400900640
Wang, R., Li, M., Wu, W., Qiu, Y., Hu, W., Li, Z., Wang, Z., Yu, Y., Liao, J.,
Sun, W., Mao, J., & Zhu, Y. Z. (2021). NAV2 positively modulates
inflammatory response of fibroblast-like synoviocytes through activat-
ing Wnt/β-catenin signaling pathway in rheumatoid arthritis. Clinical
and Translational Medicine,11, e376. https://doi.org/10.1002/
ctm2.376
Wang, X., Bove, A. M., Simone, G., & Ma, B. (2020). Molecular bases of
VEGFR-2-mediated physiological function and pathological role. Fron-
tiers in Cell and Development Biology,8, 599281. https://doi.org/10.
3389/fcell.2020.599281
Wang, Y., Chen, S., Du, K., Liang, C., Wang, S., Owusu Boadi, E., Li, J.,
Pang, X., He, J., & Chang, Y. X. (2021). Traditional herbal medicine:
Therapeutic potential in rheumatoid arthritis. Journal of Ethnopharma-
cology,279, 114368. https://doi.org/10.1016/j.jep.2021.114368
34 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Wang, Y., Lloyd, K. A., Melas, I., Zhou, D., Thyagarajan, R., Lindqvist, J.,
Hansson, M., Svärd, A., Mathsson-Alm, L., Kastbom, A., Lundberg, K.,
Klareskog, L., Catrina, A. I., Rapecki, S., Malmström, V., & Grönwall, C.
(2019). Rheumatoid arthritis patients display B-cell dysregulation
already in the naïve repertoire consistent with defects in B-cell toler-
ance. Scientific Reports,9, 19995. https://doi.org/10.1038/s41598-
019-56279-0
Wang, Y., Zhao, R., Gu, Z., Dong, C., Guo, G., & Li, L. (2020). Effects of glu-
cocorticoids on osteoporosis in rheumatoid arthritis: A systematic
review and meta-analysis. Osteoporosis International,31, 14011409.
https://doi.org/10.1007/s00198-020-05360-w
Warrington, T. P., & Bostwick, J. M. (2006). Psychiatric adverse effects of
corticosteroids. Mayo Clinic Proceedings,81, 13611367. https://doi.
org/10.4065/81.10.1361
Wegner, N., Lundberg, K., Kinloch, A., Fisher, B., Malmström, V.,
Feldmann, M., & Venables, P. J. (2010). Autoimmunity to specific
citrullinated proteins gives the first clues to the etiology of rheumatoid
arthritis. Immunological Reviews,233,3454. https://doi.org/10.1111/
j.0105-2896.2009.00850.x
Weyand, C. M., & Goronzy, J. J. (2006). T-cell-targeted therapies in rheu-
matoid arthritis. Nature Clinical Practice. Rheumatology,2, 201210.
https://doi.org/10.1038/ncprheum0142
Willoughby, D. A., Moore, A. R., & Colville-Nash, P. R. (2000). COX-1,
COX-2, and COX-3 and the future treatment of chronic inflammatory
disease. Lancet,355, 646648. https://doi.org/10.1016/S0140-6736
(99)12031-2
Wilson, J. C., Sarsour, K., Gale, S., Pethö-Schramm, A., Jick, S. S., &
Meier, C. R. (2019). Incidence and risk of glucocorticoid-associated
adverse effects in patients with rheumatoid arthritis. Arthritis Care &
Research,71, 498511. https://doi.org/10.1002/acr.23611
Wing, K., & Sakaguchi, S. (2010). Regulatory T cells exert checks and bal-
ances on self tolerance and autoimmunity. Nature Immunology,11,7
13. https://doi.org/10.1038/ni.1818
Woodrick, R. S., & Ruderman, E. M. (2011). Safety of biologic therapy in
rheumatoid arthritis. Nature Reviews Rheumatology,7, 639652.
https://doi.org/10.1038/nrrheum.2011.145
Wu, W., Xiao, Z. X., Zeng, D., Huang, F., Wang, J., Liu, Y., Bellanti, J. A.,
Olsen, N., & Zheng, S. G. (2020). B7-H1 promotes the functional effect
of human gingiva-derived mesenchymal stem cells on collagen-
induced arthritis murine model. Molecular Therapy,28, 24172429.
https://doi.org/10.1016/j.ymthe.2020.07.002
Wu, X., Zhu, D., Tian, J., Tang, X., Guo, H., Ma, J., Xu, H., & Wang, S.
(2020). Granulocytic myeloid-derived suppressor cell exosomal prosta-
glandin E2 ameliorates collagen-induced arthritis by enhancing IL-10+
B cells. Frontiers in Immunology,11, 588500. https://doi.org/10.3389/
fimmu.2020.588500
Wu, Z., Ma, D., Yang, H., Gao, J., Zhang, G., Xu, K., & Zhang, L. (2021).
Fibroblast-like synoviocytes in rheumatoid arthritis: Surface markers
and phenotypes. International Immunopharmacology,93, 107392.
https://doi.org/10.1016/j.intimp.2021.107392
Wysocki, T., Olesińska, M., & Paradowska-Gorycka, A. (2020). Current
understanding of an emerging role of HLA-DRB1 gene in rheumatoid
arthritis-from research to clinical practice. Cells,9.https://doi.org/10.
3390/cells9051127
Xiao, Y., Shi, M., Qiu, Q., Huang, M., Zeng, S., Zou, Y., Zhan, Z., Liang, L.,
Yang, X., & Xu, H. (2016). Piperlongumine suppresses dendritic cell
maturation by reducing production of reactive oxygen species and has
therapeutic potential for rheumatoid arthritis. The Journal of Immunol-
ogy,196, 49254934. https://doi.org/10.4049/jimmunol.1501281
Xu, H., Jia, S., & Xu, H. (2019). Potential therapeutic applications of exo-
somes in different autoimmune diseases. Clinical Immunology,205,
116124. https://doi.org/10.1016/j.clim.2019.06.006
Yadav, V., Krishnan, A., & Vohora, D. (2020). A systematic review on Piper
longum L.: Bridging traditional knowledge and pharmacological evi-
dence for future translational research. Journal of Ethnopharmacology,
247, 112255. https://doi.org/10.1016/j.jep.2019.112255
Yamada, R., Suzuki, A., Chang, X., & Yamamoto, K. (2005). Citrullinated
proteins in rheumatoid arthritis. Frontiers in Bioscience,10,5464.
https://doi.org/10.2741/1506
Yang, L., Jin, R., Lu, D., & Ge, Q. (2020). T cell tolerance in early life. Fron-
tiers in Immunology,11, 576261. https://doi.org/10.3389/fimmu.
2020.576261
Yang, X., Xia, H., Liu, C., Wu, Y., Liu, X., Cheng, Y., Wang, Y., Xia, Y.,
Yue, Y., Cheng, X., & Jia, R. (2023). The novel delivery-exosome appli-
cation for diagnosis and treatment of rheumatoid arthritis. Pathology,
Research and Practice,242, 154332. https://doi.org/10.1016/j.prp.
2023.154332
Yap, H. Y., Tee, S. Z. Y., Wong, M. M. T., Chow, S. K., Peh, S. C., &
Teow, S. Y. (2018). Pathogenic role of immune cells in rheumatoid
arthritis: Implications in clinical treatment and biomarker development.
Cells,7, 161. https://doi.org/10.3390/cells7100161
Yarwood, A., Huizinga, T. W. J., & Worthington, J. (2015). The genetics of
rheumatoid arthritis: Risk and protection in different stages of the evo-
lution of RA. Rheumatology (United Kingdom),55, 199209. https://
doi.org/10.1093/rheumatology/keu323
Ye, Z., Wang, Y., Xiang, B., Wang, H., Tao, H., Zhang, C., Zhang, S., Sun, D.,
Luo, F., & Song, L. (2023). Roles of the Siglec family in bone and bone
homeostasis. Biomedicine and Pharmacotherapy,165, 115064. https://
doi.org/10.1016/j.biopha.2023.115064
Yee, A., Webb, T., Seaman, A., Infantino, M., Meacci, F., Manfredi, M.,
Benucci, M., Lakos, G., Favalli, E., Shioppo, T., Meroni, P. L., &
Mahler, M. (2015). Anti-CarP antibodies as promising marker to mea-
sure joint damage and disease activity in patients with rheumatoid
arthritis. Immunologic Research,61,2430. https://doi.org/10.1007/
s12026-014-8560-x
Yende, S. R. (2010). Anti-rheumatoid activity of aqueous extract of Piper
longum on Freunds adjuvant-induced arthritis in rats' evaluation of
Polyhebal formulation for rheumatoid arthritis view project. Interna-
tional Journal of Pharmaceutical Sciences and Research,1, 129133.
https://doi.org/10.13040/IJPSR.0975-8232.1(9-S).129-33
Yousfi, F., Abrigach, F., Petrovic, J. D., Sokovic, M., & Ramdani, M. (2021).
Phytochemical screening and evaluation of the antioxidant and anti-
bacterial potential of Zingiber officinale extracts. South African Journal
of Botany,142, 433440. https://doi.org/10.1016/j.sajb.2021.07.010
Yuen, G. J. (2020). Autoimmunity in women: An eXamination of eXisting
models. Clinical Immunology,210, 108270. https://doi.org/10.1016/j.
clim.2019.108270
Zhai, K. F., Duan, H., Cui, C. Y., Cao, Y. Y., Si, J. L., Yang, H. J., Wang, Y. C.,
Cao, W. G., Gao, G. Z., & Wei, Z. J. (2019). Liquiritin from Glycyrrhiza
uralensis attenuating rheumatoid arthritis via reducing inflammation,
suppressing angiogenesis, and inhibiting MAPK signaling pathway.
Journal of Agricultural and Food Chemistry,67, 28562864. https://doi.
org/10.1021/acs.jafc.9b00185
Zhang, B., Wang, Y., Yuan, Y., Sun, J., Liu, L., Huang, D., Hu, J., Wang, M.,
Li, S., Song, W., Chen, H., Zhou, D., & Zhang, X. (2021). In vitro elimina-
tion of autoreactive B cells from rheumatoid arthritis patients by uni-
versal chimeric antigen receptor T cells. Annals of the Rheumatic
Diseases,80, 176184. https://doi.org/10.1136/annrheumdis-2020-
217844
Zhang, J., Ma, Y., Zhang, Y., Niu, S., Chu, M., & Zhang, Z. (2021). Angiogen-
esis is inhibited by arsenic trioxide through downregulation of the Cir-
cHIPK3/miR-149-5p/FOXO1/VEGF functional module in rheumatoid
arthritis. Frontiers in Pharmacology,12, 751667. https://doi.org/10.
3389/fphar.2021.751667
Zhang, J. H., Shangguan, Z. S., Chen, C., Zhang, H. J., & Lin, Y. (2016). Anti-
inflammatory effects of guggulsterone on murine macrophage by inhi-
biting LPS-induced inflammatory cytokines in NF-κB signaling path-
way. Drug Design, Development and Therapy,10, 18291835. https://
doi.org/10.2147/DDDT.S104602
Zhang, W., Chen, Y., Liu, Q., Zhou, M., Wang, K., Wang, Y., Nie, J., Gui, S.,
Peng, D., He, Z., & Li, Z. (2022). Emerging nanotherapeutics alleviating
rheumatoid arthritis by readjusting the seeds and soils. Journal of
CHATTERJEE ET AL.35
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Controlled Release,345, 851879. https://doi.org/10.1016/j.jconrel.
2022.04.001
Zheng, C. J., Li, H. Q., Ren, S. C., Xu, C. L., Rahman, K., Qin, L. P., &
Sun, Y. H. (2015). Phytochemical and pharmacological profile of Vitex
negundo.Phytotherapy Research,29, 633647. https://doi.org/10.
1002/ptr.5303
Zheng, C. J., Zhang, X. W., Han, T., Jiang, Y. P., Tang, J. Y., Brömme, D., &
Qin, L. P. (2014). Anti-inflammatory and anti-osteoporotic lignans from
Vitex negundo seeds. Fitoterapia,93,3138. https://doi.org/10.1016/j.
fitote.2013.12.006
Zheng, C. J., Zhao, X. X., Ai, H. W., Lin, B., Han, T., Jiang, Y. P., Xing, X., &
Qin, L. P. (2014). Therapeutic effects of standardized Vitex negundo
seeds extract on complete Freund's adjuvant induced arthritis in rats.
Phytomedicine,21, 838846. https://doi.org/10.1016/j.phymed.2014.
02.003
Zhuo, J., Xia, Q., Sharma, N., Gao, S., Lama, S., Cui, J., Feathers, V.,
Shadick, N., & Weinblatt, M. E. (2022). The role of shared epitope in
rheumatoid arthritis prognosis in relation to anti-citrullinated protein
antibody positivity. Rheumatology and Therapy,9, 637647. https://
doi.org/10.1007/s40744-022-00427-y
How to cite this article: Chatterjee, A., Jayaprakasan, M.,
Chakrabarty, A. K., Lakkaniga, N. R., Bhatt, B. N., Banerjee, D.,
Narwaria, A., Katiyar, C. K., & Dubey, S. K. (2024).
Comprehensive insights into rheumatoid arthritis:
Pathophysiology, current therapies and herbal alternatives for
effective disease management. Phytotherapy Research,136.
https://doi.org/10.1002/ptr.8187
36 CHATTERJEE ET AL.
10991573, 0, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/ptr.8187 by Birla Institute of Technology & Science - Pilani, Wiley Online Library on [24/03/2024]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
T and B cells are key components of the adaptive immune system. Through their immune properties and their interactions with other immune cells and cytokines around them, they build a complex network to achieve immune tolerance and maintain homeostasis of the body. This is achieved through mechanisms of central and peripheral tolerance, both of which are associated with advantages and disadvantages. For this reason, the immune system is tightly regulated and their dysregulation can result in the subsequent initiation of various diseases. In this review, we will summarize the roles played by T cells and B cells within immune tolerance with specific examples in the context of different diseases that include allergic disease. In addition, we will also provide an overview on their suitability as biomarkers of allergen-specific immunotherapy.
Article
Full-text available
Rheumatoid arthritis (RA) occurs more frequently in women than in men, and the studies that have addressed clinical and prognostic differences between the sexes are scarce and have contradictory results and methodological problems. The present work aims to evaluate sex- and gender-related differences in the clinical expression and prognosis of RA as well as on the impact on psychosocial variables, coping behavior, and healthcare use and access. By identifying between sex differences and gender-related outcomes in RA, it may be possible to design tailored therapeutic strategies that consider the differences and unmet needs. Being that sex, together with age, is the most relevant biomarker and health determinant, a so-called personalized medicine approach to RA must include clear guidance on what to do in case of differences.
Article
Full-text available
Rheumatoid arthritis (RA) is a chronic immune-mediated inflammatory illness caused by an accumulation of inflammatory infiltration in the synovial membrane, which leads to the progressive deterioration of joint architecture. Women are three times more likely than men to have RA, with more severe functional loss and disability. The purpose of this review is to study the shifts in the hormonal system brought on by menopause and their potential links with RA. Females are more likely to develop RA as a result of hereditary and environmental interactions of sex hormones and their effects on the immune system. Rapid declines in ovarian function and systemic estrogen have been linked to postmenopausal increases in proinflammatory cytokines such as IL-6, TNF-α, and IL-1β. The majority of the data from the literature review imply that female hormone characteristics that can be influenced by hereditary and environmental variables impact the development of autoimmune illnesses, including RA.
Article
Full-text available
Tissue homeostasis is crucial for multicellular organisms, wherein the loss of cells is compensated by generating new cells with the capacity for proliferation and differentiation. At the origin of these populations are the stem cells, which have the potential to give rise to cells with both capabilities, and persevere for a long time through the self-renewal and quiescence. Since the discovery of stem cells, an enormous effort has been focused on learning about their functions and the molecular regulation behind them. Wnt signaling is widely recognized as essential for normal and cancer stem cell. Moreover, β-catenin-dependent Wnt pathway, referred to as canonical, has gained attention, while β-catenin-independent Wnt pathways, known as non-canonical, have remained conspicuously less explored. However, recent evidence about non-canonical Wnt pathways in stem cells begins to lay the foundations of a conceivably vast field, and on which we aim to explain this in the present review. In this regard, we addressed the different aspects in which non-canonical Wnt pathways impact the properties of stem cells, both under normal conditions and also under disease, specifically in cancer. Graphical abstract
Article
Full-text available
Background Rheumatoid arthritis is a chronic autoimmune inflammatory disease associated with disability and premature death. Up-to-date estimates of the burden of rheumatoid arthritis are required for health-care planning, resource allocation, and prevention. As part of the Global Burden of Diseases, Injuries, and Risk Factors Study (GBD) 2021, we provide updated estimates of the prevalence of rheumatoid arthritis and its associated deaths and disability-adjusted life-years (DALYs) by age, sex, year, and location, with forecasted prevalence to 2050. Methods Rheumatoid arthritis prevalence was estimated in 204 countries and territories from 1990 to 2020 using Bayesian meta-regression models and data from population-based studies and medical claims data (98 prevalence and 25 incidence studies). Mortality was estimated from vital registration data with the Cause of Death Ensemble model (CODEm). Years of life lost (YLL) were calculated with use of standard GBD lifetables, and years lived with disability (YLDs) were estimated from prevalence, a meta-analysed distribution of rheumatoid arthritis severity, and disability weights. DALYs were calculated by summing YLLs and YLDs. Smoking was the only risk factor analysed. Rheumatoid arthritis prevalence was forecast to 2050 by logistic regression with Socio-Demographic Index as a predictor, then multiplying by projected population estimates. Findings In 2020, an estimated 17·6 million (95% uncertainty interval 15·8–20·3) people had rheumatoid arthritis worldwide. The age-standardised global prevalence rate was 208·8 cases (186·8–241·1) per 100 000 population, representing a 14·1% (12·7–15·4) increase since 1990. Prevalence was higher in females (age-standardised female-to-male prevalence ratio 2·45 [2·40–2·47]). The age-standardised death rate was 0·47 (0·41–0·54) per 100 000 population (38 300 global deaths [33 500–44 000]), a 23·8% (17·5–29·3) decrease from 1990 to 2020. The 2020 DALY count was 3 060 000 (2 320 000–3 860 000), with an age-standardised DALY rate of 36·4 (27·6–45·9) per 100 000 population. YLDs accounted for 76·4% (68·3–81·0) of DALYs. Smoking risk attribution for rheumatoid arthritis DALYs was 7·1% (3·6–10·3). We forecast that 31·7 million (25·8–39·0) individuals will be living with rheumatoid arthritis worldwide by 2050. Interpretation Rheumatoid arthritis mortality has decreased globally over the past three decades. Global age-standardised prevalence rate and YLDs have increased over the same period, and the number of cases is projected to continue to increase to the year 2050. Improved access to early diagnosis and treatment of rheumatoid arthritis globally is required to reduce the future burden of the disease.
Article
Full-text available
Mutation in the VEGF gene disturbs the production of chondrocytes and angiogenesis which are essential for cartilage health. Cytokines and chemokines produced by auto-activation of B-cells degrade cartilage. Bruton's Tyrosine Kinase (BTK) plays a crucial role in the activation of these B-cells. VEGF has a central part in angiogenesis, in the recruitment of endothelial cells, and is involved in mechanisms that result in tumour formation. The objective of this research is to investigate the potential role of VEGF polymorphism in the development of Rheumatoid Arthritis (RA) and the screening of potential natural, synthetic BTK inhibitor compounds as possible in-silico chemotherapeutic agents to control auto-activation of B-cells and cartilage degrading cytokines. In this study, it had been shown that allele A frequency was significantly higher than that of allele C in RA-positive patients as compared to controls. Hence it depicts that allele A of VEGF (rs699947) can increase the risk of RA while allele C has a protective role. The phytochemicals which showed maximum binding affinity at the inhibitory site of BTK include beta boswellic acid, tanshinone, and baicalin. These phytochemicals as BTK inhibitor give insights to use them as anti-arthritic compounds by nanoparticle drug delivery mechanism.
Article
Full-text available
The aim is better to understand and critically explore and present the available data from observational studies on the pathogenetic role of the microbiome in the development of rheumatoid arthritis (RA). The electronic databases PubMed, Scopus, and Web of Science were screened for the relevant literature published in the last ten years. The primary outcomes investigated included the influence of the gut microbiome on the pathogenesis and development of rheumatoid arthritis, exploring the changes in microbiota diversity and relative abundance of microbial taxa in individuals with RA and healthy controls (HCs). The risk of bias in the included literature was assessed using the GRADE criteria. Ten observational studies were identified and included in the qualitative assessment. A total of 647 individuals with RA were represented in the literature, in addition to 16 individuals with psoriatic arthritis (PsA) and 247 HCs. The biospecimens comprised fecal samples across all the included literature, with 16S rDNA sequencing representing the primary method of biological analyses. Significant differences were observed in the RA microbiome compared to that of HCs: a decrease in Faecalibacterium, Fusicatenibacter, Enterococcus, and Megamonas and increases in Eggerthellales, Collinsella, Prevotella copri, Klebsiella, Escherichia, Eisenbergiella, and Flavobacterium. There are significant alterations in the microbiome of individuals with RA compared to HCs. This includes an increase in Prevotella copri and Lactobacillus and reductions in Collinsella. Collectively, these alterations are proposed to induce inflammatory responses and degrade the integrity of the intestinal barrier; however, further studies are needed to confirm this relationship.
Article
Full-text available
Zingiber officinale and Citrus limon, well known as ginger and lemon, are two vegetals widely used in traditional medicine and the culinary field. The juices of the two vegetals were evaluated based on their inflammation, both in vivo and in vitro. High-performance liquid chromatography (HPLC) was used to characterize different juices from Zingiber of ficinale Roscoe and Citrus limon. After the application of the HPLC method, different compounds were identified, such as 6-gingerol and 6-gingediol from the ginger juice and isorhamnetin and hesperidin from the lemon juice. In addition, the two juices and their formulation were assessed for their anti-inflammatory activity, in vitro by utilizing the BSA denaturation test, in vivo using the carrageenan-induced inflammation test, and the vascular permeability test. Important and statistically significant anti-inflammatory activities were observed for all juices, especially the formulation. The results of our work showed clearly that the Zingiber officinale and Citrus limon juices protect in vivo the development of the rat paw edema, especially the formulation F composed of the Zingiber officinale and Citrus limon juices, which shows an anti-inflammatory activity equal to −35.95% and −44.05% using 10 and 20 mg/kg of the dose, respectively. Our work also showed that the formulation was the most effective tested extract since it inhibits the vascular permeability by −37% and −44% at the doses of 200 and 400 mg/kg, respectively, and in vitro via the inhibition of the denaturation of BSA by giving a synergetic effect with the highest IC 50 equal to 684.61 ± 7.62 μg/mL corresponding to the formulation F. This work aims to develop nutraceutical preparations in the future and furnishes the support for a new investigation into the activities of the various compounds found in Zingiber officinale Roscoe and Citrus limon.
Article
Vitamin D analogue calcipotriol is currently used in the local treatment of psoriasis. However, it also has antiproliferative and anti-inflammatory effects in the cells of the joint - suggesting a possible benefit in local treatment of arthritis. In this study, calcipotriol was studied in different in vitro methods to find out its effect on synovial and mesenchymal stromal cells. Primary human cell lines of osteoarthritis or rheumatoid arthritis patients (five mesenchymal stromal cells, MSC, and four synovial stromal cells, SSC) were cultured to study migration and proliferation of the cells in a wound healing model. The media was supplemented with calcipotriol, 1,25(OH)2D3, dexamethasone, betamethasone, methylprednisolone or control solution in 1 nM-100 nM concentrations. To see possible toxic effects of calcipotriol, concentrations up to 10µM in SSCs and MSCs were studied in apoptosis and necrosis assays in four cell lines. Calcipotriol and 1,25(OH)2D3, as well as the three glucocorticoids, reduced the migration of both SSCs and MSCs. In SSCs, the effect of calcipotriol and 1,25(OH)2D3 was at least as effective as with glucocorticoids, while with MSCs, the glucocorticoids were stronger inhibitors of migration. The antimigratory of calcipotriol and 1,25(OH)2D3 was consistently maintained in 10µM and 1µM. Calcipotriol was not toxic to MSCs and SSCs up to concentrations of 10µM. Calcipotriol, as well as 1,25(OH)2D3, exerts antimigratory and antiproliferative effects on human SSCs and MSCs of the joint. These effects are not caused by apoptosis or necrosis. Both calcipotriol and 1,25(OH)2D3 have similar effects as glucocorticoids without apparent toxicity, suggesting that calcipotriol might be an eligible candidate to the local treatment of arthritis with a broad therapeutic window.