ArticlePDF AvailableLiterature Review

Target Role of Monocytes as Key Cells of Innate Immunity in Rheumatoid Arthritis

MDPI
Diseases
Authors:

Abstract and Figures

Rheumatoid arthritis (RA) is a chronic, systemic, and inflammatory autoimmune condition characterized by synovitis, pannus formation (with adjacent bone erosion), and joint destruction. In the perpetuation of RA, fibroblast-like synoviocytes (FLSs), macrophages, B cells, and CD4+ T-cells—specifically Th1 and Th17 cells—play crucial roles. Additionally, dendritic cells, neutrophils, mast cells, and monocytes contribute to the disease progression. Monocytes, circulating cells primarily derived from the bone marrow, participate in RA pathogenesis. Notably, CCR2 interacts with CCL2, and CX3CR1 (expressed by monocytes) cooperates with CX3CL1 (produced by FLSs), facilitating the migration involved in RA. Canonical “classical” monocytes predominantly acquire the phenotype of an “intermediate” subset, which differentially expresses proinflammatory cytokines (IL-1β, IL-6, and TNF) and surface markers (CD14, CD16, HLA-DR, TLRs, and β1- and β2-integrins). However, classical monocytes have greater potential to differentiate into osteoclasts, which contribute to bone resorption in the inflammatory milieu; in RA, Th17 cells stimulate FLSs to produce RANKL, triggering osteoclastogenesis. This review aims to explore the monocyte heterogeneity, plasticity, antigenic expression, and their differentiation into macrophages and osteoclasts. Additionally, we investigate the monocyte migration into the synovium and the role of their cytokines in RA.
Content may be subject to copyright.
Citation: Salnikova, D.I.; Nikiforov,
N.G.; Postnov, A.Y.; Orekhov, A.N.
Target Role of Monocytes as Key Cells
of Innate Immunity in Rheumatoid
Arthritis. Diseases 2024,12, 81.
https://doi.org/10.3390/
diseases12050081
Academic Editor: Maurizio Battino
Received: 30 March 2024
Revised: 21 April 2024
Accepted: 22 April 2024
Published: 25 April 2024
Copyright: © 2024 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
diseases
Review
Target Role of Monocytes as Key Cells of Innate Immunity in
Rheumatoid Arthritis
Diana I. Salnikova 1, * , Nikita G. Nikiforov 2,3,4 , Anton Y. Postnov 3and Alexander N. Orekhov 2,3,5
1Laboratory of Oncoproteomics, Department of Experimental Tumor Biology, Institute of Carcinogenesis,
Blokhin N.N. National Medical Research Center of Oncology, 24 Kashirskoe Highway, 115522 Moscow, Russia
2
Laboratory of Angiopathology, The Institute of General Pathology and Pathophysiology, 8 Baltiyskaya Street,
125315 Moscow, Russia; nikiforov.mipt@googlemail.com (N.G.N.); ano.inat@mail.ru (A.N.O.)
3
Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Federal State Budgetary Scientific
Institution “Petrovsky National Research Centre of Surgery”, 3 Tsyurupa Street, 117418 Moscow, Russia;
anton-5@mail.ru
4Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology,
Russian Academy of Sciences, 34/5 Vavilova Street, 119334 Moscow, Russia
5Institute for Atherosclerosis Research, Osennyaya Street 4-1-207, 121609 Moscow, Russia
*Correspondence: dianasalnikova08@yandex.ru
Abstract: Rheumatoid arthritis (RA) is a chronic, systemic, and inflammatory autoimmune condition
characterized by synovitis, pannus formation (with adjacent bone erosion), and joint destruction. In
the perpetuation of RA, fibroblast-like synoviocytes (FLSs), macrophages, B cells, and CD4
+
T-cells—
specifically Th1 and Th17 cells—play crucial roles. Additionally, dendritic cells, neutrophils, mast
cells, and monocytes contribute to the disease progression. Monocytes, circulating cells primarily
derived from the bone marrow, participate in RA pathogenesis. Notably, CCR2 interacts with CCL2,
and CX3CR1 (expressed by monocytes) cooperates with CX3CL1 (produced by FLSs), facilitating the
migration involved in RA. Canonical “classical” monocytes predominantly acquire the phenotype
of an “intermediate” subset, which differentially expresses proinflammatory cytokines (IL-1
β
, IL-6,
and TNF) and surface markers (CD14, CD16, HLA-DR, TLRs, and
β
1- and
β
2-integrins). However,
classical monocytes have greater potential to differentiate into osteoclasts, which contribute to bone
resorption in the inflammatory milieu; in RA, Th17 cells stimulate FLSs to produce RANKL, triggering
osteoclastogenesis. This review aims to explore the monocyte heterogeneity, plasticity, antigenic
expression, and their differentiation into macrophages and osteoclasts. Additionally, we investigate
the monocyte migration into the synovium and the role of their cytokines in RA.
Keywords: monocyte subsets; macrophages; innate immunity; rheumatoid arthritis; chemokines;
proinflammatory cytokines
1. Introduction
RA is a chronic, systemic, and autoimmune disease. The mechanisms of this condition
are still being investigated. The illness is characterized by an inflammatory process in
the synovial joints, which is called synovitis [
1
]. Moreover, RA is accompanied by the
hyperplasia of the synovial tissue. The disease is complicated by the destruction of the
cartilage structure and the formation of pannus [
2
]. Throughout the course of the pathology,
bone erosion occurs and predominantly surrounds the peripheral synovial joints. The
clinical symptoms of RA include pain, edema development, and tenderness of the synovial
joints located symmetrically and peripherally [
3
]. A great deal of progress has been made in
the treatment of RA with antagonists of proinflammatory cytokines, such as tumor necrosis
factor (TNF), interleukin-6 (IL-6), and IL-1
β
. However, the disease remains refractory
in some patients. Studies have demonstrated that prostaglandins, leukotrienes, reactive
oxygen species, nitric oxide, lipoxins, and platelet-activating factors as small-molecule
Diseases 2024,12, 81. https://doi.org/10.3390/diseases12050081 https://www.mdpi.com/journal/diseases
Diseases 2024,12, 81 2 of 20
inflammatory mediators play an important role in RA development. Such compounds
assist in inducing, maintaining, or reducing inflammation and could therefore be potential
therapeutic targets [1].
Innate and adaptive autoimmune cells promote the continuation of RA pathogenesis.
Among them are B cells and CD4
+
T cells, which include the T helper 1 (Th1), Th2, and
Th17 cells. Moreover, monocytes/macrophages and resident macrophages participate
in the disease development [
4
]. In addition, synovial cells and FLSs contribute to RA.
Furthermore, dendritic cells (DCs), neutrophils, mast cells, and immunoregulatory cells,
for example, regulatory T (Treg) cells, are involved in the complex interaction that leads
to the preservation of chronic inflammation [
5
]. The above-mentioned cells stimulate the
formation of the modulators of local inflammation, such as cytokines, chemokine ligands,
and the appropriate receptors. These modulators take part in synovial inflammation and
destruction [6].
Monocytes are a type of blood cell containing a horseshoe-shaped nucleus in their
cytoplasm. They relate to the mononuclear phagocyte system and are much smaller than
macrophages, which are in turn much smaller than multinucleated osteoclasts. Mono-
cytes originate from bone marrow precursors [
7
]. Together with lymphocytes, monocytes
were previously known as agranulocytes. Moreover, monocytes are defense cells of the
second line after neutrophils in an innate immune system and are able to phagocytose
foreign particles. They have the capability of differentiating into their own subsets and
macrophages based on the necessity of the microenvironment. Additionally, monocytes
produce cytokines and act as antigen-presenting cells (APCs) [8].
Monocytes differ from other autoimmune cells due to several reasons. First of all, they
are capable of triggering, maintaining, and increasing the activity of inflammation in the
synovial joints in RA. Moreover, chemokine receptors CCR2 (CD192) of CD14
+
monocytes
and CX3CR1 (fractalkine receptor, GPR13) of CD16
+
monocytes interact with the corre-
sponding ligands CCL2 (MCP-1) and CX3CL1 (fractalkine) released from FLSs, and this
process contributes to the migration of monocytes from circulation and their recruitment
into the RA synovium [
9
]. These chemoalkine receptors represent the differentiation mark-
ers of monocytes. A disintegrin and metalloproteinase domain-containing protein 10 and
products of FLSs such as granulocyte–macrophage colony-stimulating factor (GM-CSF),
TNF, and IL-1
β
promote monocyte migration. Next, monocytes are characterized by the
increased expression of chemokine receptors and antigens on their cell surface [
6
]. Among
them are CD14, CD16, toll-like receptors (TLRs), human leukocyte antigen DR (HLA-DR),
and
β
1- and
β
2-integrins that are adhesion molecules. Moreover, they produce proinflam-
matory cytokines such as TNF, IL-1
β
, and IL-6. The differentiation of monocytes leads
to the development of classical, intermediate, and non-classical monocyte subsets [
10
].
The intermediate monocytes are prevalent in RA, and they undergo differentiation into
proinflammatory M1 macrophages. Proinflammatory macrophages are induced by TNF,
GM-CSF, interferon (IFN)-
γ
, and lipopolysaccharide (LPS). M1 macrophages produce
proinflammatory cytokines TNF, IL-1β, IL-6, and IL-12 and stimulate inflammation in the
synovial joints [
11
]. Furthermore, classical monocytes expressing CD14
++
and CD16
have
relatively greater potential to differentiate into osteoclasts and induce the erosion of the
surrounding bones in the synovial joints in RA [
12
]. Osteoclastogenesis is the process by
which osteoclasts are differentiated. Th17 cells trigger the formation of the receptor activa-
tor of the NF-
κ
B (RANK) ligand (RANKL) by FLSs, which contributes to the differentiation
of monocytes into osteoclasts [
13
]. Osteoclastogenesis is increased by cytokines such as
TNF, IL-1β, and IL-6 [12,14].
The overview reveals the formation, classification, and specific markers of monocytes,
their functioning as APCs and progenitors of macrophages and osteoclasts, the expression
of chemokine receptors, and the production of cytokines in RA.
Diseases 2024,12, 81 3 of 20
2. Monocytes in RA
2.1. Development of Monocytes
Monocytopoiesis, the process of monocyte production, originates from human stem
cells, predominantly found in the bone marrow. Monocytes are part of the mononuclear
phagocyte system and act as key innate immune cells in the organism. The process of mono-
cytopoiesis occurs at a stage of embryonic development via definitive hematopoiesis. The
ventral wall of the aorta is a site for the aorta gonad mesonephros emergence following three
weeks of pregnancy in women and at the E-7 stage in mice. The aorta gonad mesonephros
is the first location in the embryo where monocytes appear [
15
]. Hematopoietic stem cells
differentiate into monoblasts under the influence of various cytokines, including GM-CSF
and macrophage colony-stimulating factor (M-CSF). Hematopoietic stem cells undergo
various stages of multipotent progenitor transformation into monocyte/macrophage and
DC progenitors. These progenitors lose the ability to generate granulocytes and either
give rise to “common monocyte progenitors” restricted to monocytes and their progeny
or are directed to a common DC progenitor [
15
]. The subsequent stages involve further
maturation and differentiation, giving rise to promonocytes and finally monocytes. These
differentiated monocytes are released into the bloodstream, where they execute crucial
functions such as phagocytosis, antigen presentation, and cytokine secretion, playing a piv-
otal role in immune surveillance and tissue homeostasis. Moreover, monocyte production
happens in the liver, thymus, and spleen of the fetus. Since the late period of embryonic
development, the formation of monocytes relocates to the bone marrow, which remains the
main site for the monocyte production in adults and older people [16].
2.2. Human Monocyte Subsets and Their Role in RA
The expression of CD14 and CD16 markers on the cell surface of monocytes was
used to identify and categorize the subsets of monocytes. According to the classification
developed by Ziegler-Heitbrock et al., three types of monocyte subsets are presented in
humans [7].
2.2.1. Classical Monocytes
Primarily, there are classical monocytes, which have high expression of the CD14
marker and, more importantly, do not express the CD16 marker. Furthermore, they pro-
duce the cytokines TNF, IL-1
β
, IL-6, and IL-10 in high concentrations. Several forms of
inflammatory stimulus are capable of eliciting the cytokine expression from monocytes. For
instance, this process occurs as a consequence of the LPS stimulation and activation of the
immune complex [17]. This type of monocyte undergoes differentiation into intermediate
monocytes, inflammatory macrophages, and osteoclasts in the regions of inflammation.
Classical monocytes function as phagocytic scavenger cells and promote synovial inflam-
mation and the osteoclastic erosion of bones. CD16
, but not CD16
+
, monocyte subsets
represent circulating osteoclast progenitors that differentiate into osteoclasts [
12
]. Conse-
quently, classical monocytes are able to be the circulating predecessors of osteoclasts in
erosive RA.
2.2.2. Intermediate Monocytes
The second group refers to intermediate monocytes, which are characterized by the
high expression of the CD14 marker and diminished expression of the CD16 marker. The
number of these monocytes is highly upregulated in the peripheral blood and synovia in
RA. They secrete TNF, IL-1
β
, and IL-6 in high quantities in the synovial joints in RA [
18
].
The differentiation of intermediate monocytes results in inflammatory monocyte subsets
and proinflammatory M1 macrophages. They maintain the inflammation of the synovial
joints and enhance the disease development. Additionally, intermediate monocytes lead
to the direct activation of Th17 cells in the inflamed synovial joints and their expansion.
The amount of these monocytes increases in the blood circulation over the course of the
RA pathogenesis [
19
]. The enhanced level of classical and intermediate monocytes in an
Diseases 2024,12, 81 4 of 20
untreated RA patient is a prognostic factor for the decline or absence of susceptibility to
methotrexate therapy [20].
2.2.3. Non-Classical Monocytes
In addition, there are non-classical monocytes that have diminished expression of
the CD14 marker and high expression of the CD16 marker. Non-classical monocytes
preferentially associated with the endothelial wall engage in ”patrolling behavior” urgently
in cases of tissue injury [
21
]. This process provides an early inflammatory response. Non-
classical monocytes stimulate the adhesion of monocytes in the microvessels of joints.
Resident anti-inflammatory M2 macrophages represent another product of monocyte
differentiation and help to resolve inflammation. Among the monocyte subsets, the non-
classical ones proliferate the least [
10
] and undergo senescence to a greater degree [
22
].
They are capable of acquiring a senescence-associated secreted phenotype (SASP) that
transforms into a proinflammatory type and causes inflammation. Purchasing a phenotype
occurs due to age-related chronic inflammation called inflammaging [23].
2.2.4. Disease-Modifying Antirheumatic Drugs
The prevalence of circulating monocytes subsets, particularly the CD16 expressing
subsets, is associated with RA progression and assists in estimating treatment effectiveness,
for example, in the case of disease-modifying antirheumatic drugs (DMARDs). Com-
monly prescribed DMARDs include methotrexate, sulfasalazine, and hydroxychloroquine.
Methotrexate inhibits dihydrofolate reductase, an enzyme that converts dihydrofolic acid
into tetrahydrofolic acid. This enzyme, in turn, is a donor of one-carbon groups in the
synthesis of purine nucleotides and thymidylate required for DNA synthesis. As a re-
sult, methotrexate inhibits DNA synthesis and repair, cell mitosis, and, to a lesser extent,
affects RNA and protein synthesis [
24
]. Sulfasalazine dissociates into 5-aminosalicylic
acid in the connective tissue of the intestinal wall, and 5-aminosalicylic acid promotes
the anti-inflammatory properties of sulfasalazine and sulfapyridine [
25
]. Sulfapyridine
is a competitive antagonist of para-aminobenzoic acid. This drug stops the synthesis of
folate in the cells of microorganisms and causes antibacterial activity. Hydroxychloroquine
seals lysosomal membranes and prevents the exit of lysosomal enzymes. Moreover, this
drug disrupts DNA replication, RNA synthesis, and hemoglobin utilization by erythrocytic
forms of plasmodium. Hydroxychloroquine weakens the activity of proteolytic enzymes,
which are protease and collagenase. Additionally, this drug decreases the activity of leuko-
cytes and the chemotaxis of lymphocytes. DMARDs help to prevent joint deformity and
functional impairment [26].
2.3. Murine Monocyte Subsets in RA
Murine monocytes are subdivided into Ly6C
++
CD43
+
, LY6C
++
CD43
++
, and Ly6C
types. The first one is an analogue of human classical monocytes. Another type resembles
the intermediate monocytes. Both of these monocyte types trigger synovial inflammation
under sterile conditions. The third type is an equivalent of non-classical monocytes and
manages the progression and termination of sterile synovial inflammation [
27
]. In the
beginning, Ly6C
monocytes develop into proinflammatory M1 macrophages, which
induce and maintain the inflammation in the synovial joints. Further, M1 macrophages are
exposed to polarization and acquire an alternative phenotype, M2, that suppresses synovial
inflammation [28].
2.4. Monocyte Markers
Monocytes are characterized by numerous antigen expression types on their cell
surface, with some alterations in dependence on the monocyte subset. These antigens
manage RA development [8].
Diseases 2024,12, 81 5 of 20
2.4.1. Markers of Double-Positive Monocytes/Macrophages
Double-positive monocytes/macrophages were revealed in human and rat periph-
eral blood. These cells express both CD4 and CD8. In addition, they possess activated
phenotypes expressing M1 and M2 markers, producing proinflammatory cytokines and
contributing to chronic inflammation. Double-positive monocytes/macrophages may be
transferred to the inflammation area and be involved in the immune response of Th1
type [
29
]. These cells promote synovial hyperplasia and facilitate joint damage in RA
(Figure 1).
Diseases 2024, 12, x FOR PEER REVIEW 5 of 21
2.4. Monocyte Markers
Monocytes are characterized by numerous antigen expression types on their cell sur-
face, with some alterations in dependence on the monocyte subset. These antigens manage
RA development [8].
2.4.1. Markers of Double-Positive Monocytes/Macrophages
Double-positive monocytes/macrophages were revealed in human and rat peripheral
blood. These cells express both CD4 and CD8. In addition, they possess activated pheno-
types expressing M1 and M2 markers, producing proinammatory cytokines and contrib-
uting to chronic inammation. Double-positive monocytes/macrophages may be trans-
ferred to the inammation area and be involved in the immune response of Th1 type [29].
These cells promote synovial hyperplasia and facilitate joint damage in RA (Figure 1).
Figure 1. Double-positive monocytes/macrophages, expressed markers, proinammatory (IL-1β,
IL-6, TNF, M-CSF, and GM-CSF) cytokines, role in RA. CD, cluster of dierentiation; IL, interleukin;
TNF, tumor necrosis factor; GM-CSF, granulocyte–macrophage colony-stimulating factor.
2.4.2. Clusters of Dierentiation
CD14 represents the LPS receptor, which is able to be linked to an LPS-binding pro-
tein. A CD14 interaction with an LPS–LPS binding protein complex initiates signaling
pathways with the involvement of TLR-4. Further, TLR-4 triggers the formation and se-
cretion of the proinammatory chemokine IFN-γ inducible protein 10 and the cytokines
TNF, IL-1β, and IL-6 [30]. CD16 designates the immunoglobulin (Ig) Fc-gamma receptor
III (FcγRIII), which has a specic phagocytic activity. CD16
+
monocytes, mostly the inter-
mediate ones, are more abundant than non-classical monocytes in RA [19]. IgG-containing
immune complexes, for instance, containing anti-citrullinated protein antibody (ACPA),
provoke a higher level of TNF in the case of FcγRIII/CD16
+
hyperexpression on CD14
++
monocytes in RA. Magnetic microbeads for negative and positive selection with the use
of anti-CD14 and anti-CD16 monoclonal antibodies allow separating the CD14 and CD16
markers [31]. CD56 is an adhesion molecule in neural cells. CD56
+
monocytes mainly be-
long to classical monocyte subsets and spread in RA. Under the inuence of LPS stimula-
tion, CD14
bright
CD56
+
monocytes secrete greater amounts of TNF, IL-10, and IL-23. Anti-
TNF therapy, for example, with the use of etanercept, diminishes the level of these cyto-
kines [17]. Monocytes as well as myeloid series cells express the myeloid identication
marker CD115 [32].
Figure 1. Double-positive monocytes/macrophages, expressed markers, proinflammatory (IL-1
β
,
IL-6, TNF, M-CSF, and GM-CSF) cytokines, role in RA. CD, cluster of differentiation; IL, interleukin;
TNF, tumor necrosis factor; GM-CSF, granulocyte–macrophage colony-stimulating factor.
2.4.2. Clusters of Differentiation
CD14 represents the LPS receptor, which is able to be linked to an LPS-binding protein.
A CD14 interaction with an LPS–LPS binding protein complex initiates signaling pathways
with the involvement of TLR-4. Further, TLR-4 triggers the formation and secretion of
the proinflammatory chemokine IFN-
γ
inducible protein 10 and the cytokines TNF, IL-1
β
,
and IL-6 [
30
]. CD16 designates the immunoglobulin (Ig) Fc-gamma receptor III (Fc
γ
RIII),
which has a specific phagocytic activity. CD16
+
monocytes, mostly the intermediate ones,
are more abundant than non-classical monocytes in RA [
19
]. IgG-containing immune
complexes, for instance, containing anti-citrullinated protein antibody (ACPA), provoke a
higher level of TNF in the case of FcγRIII/CD16+hyperexpression on CD14++ monocytes
in RA. Magnetic microbeads for negative and positive selection with the use of anti-CD14
and anti-CD16 monoclonal antibodies allow separating the CD14 and CD16 markers [
31
].
CD56 is an adhesion molecule in neural cells. CD56
+
monocytes mainly belong to classical
monocyte subsets and spread in RA. Under the influence of LPS stimulation, CD14
bright
CD56
+
monocytes secrete greater amounts of TNF, IL-10, and IL-23. Anti-TNF therapy, for
example, with the use of etanercept, diminishes the level of these cytokines [
17
]. Monocytes
as well as myeloid series cells express the myeloid identification marker CD115 [32].
In addition to ACPAs, antibodies targeting various post-translational modifications
such as carbamylation [
33
], acetylation, and malondialdehyde are found in RA. Approx-
imately half of the ACPA clones in RA patients react with carbamylated antigens. Some
antigens even show reactivity to acetylated peptides. The cross-reactivity may be due to
similar electron density distributions at key recognized residues. Anti-malondialdehyde
antibodies, although not cross-reactive with citrullinated antigens, can activate osteoclasts
Diseases 2024,12, 81 6 of 20
similar to ACPAs [
34
]. As a result, targeting antigens from different protein modifications
trigger various pathways. Co-existing mechanisms and overlapping protein residues com-
plicate the understanding of the key protein modifications for specific autoantibody effects.
Various protein modifications can co-exist in the same tissues, cells, proteins, and even
at the same peptide epitope. This co-existence explains the presence of autoantibodies
targeting various post-translational modifications in RA and their similar functional effects.
Colasanti T. et al. (2020) described the detection of antibodies targeting homocysteiny-
lated alpha 1 antitrypsin in RA patients seropositive for the “classical” ACPA and RF.
Alpha 1 antitrypsin is a protease inhibitor protein of the serpin superfamily and protects
tissues from the effects of many enzymes secreted by inflammatory cells. Variation in the
alpha 1 antitrypsin gene is associated with increased ACPA production, which leads to RA
progression and joint destruction [35].
2.4.3. HLA-DR and Co-Stimulatory Molecules
The HLA-DR marker refers to the group of MHC II (CD68) molecules. HLA-DR acti-
vates CD4
+
cells via interaction with T cell receptors [
36
]. Intermediate monocytes produce
a higher level of HLA-DR in comparison with other monocyte subsets in RA [
37
]. Fur-
thermore, intermediate monocytes secrete a significant amount of TNF through Pam3Cys
activation and binding to TLR2/TLR1. Classical monocytes increase the expression of HLA-
DR over a period of low RA activity. Monocytes of the synovial joints are characterized by
higher expression of HLA-DR than monocytes of the peripheral blood. Additionally, DCs
demonstrate HLA-DR expression to a high degree and participate in antigen presentation
to CD4
+
T cells. HLA-DR interacts with an antigen and passes the first signal to the T cell
receptor on a CD4
+
T cell. Co-stimulatory molecules CD80 (B7-1) and CD86 (B7-2) are
expressed on the monocyte surface and work as a second signal. To sum up, both signals
stimulate CD4
+
T-cells and enable adaptive autoimmune responses that happen with the
involvement of Th1 and Th17 cells in RA. The expression of HLA-DR and CD80 is enhanced
on the intermediate monocyte surface in the RA synovial joints, and this process contributes
to the activation of IL-17+and IFN-γ+CD4+T-cells under conditions of cocultivation [38].
Moreover, activated Th1 cells promote the expression of HLA-DR and CD80 on the surface
of the monocytes in RA.
2.4.4. Toll-like Receptors
TLRs are expressed on the surface of innate immune cells, for instance, monocytes,
macrophages, DCs, and neutrophils. TLRs identify inflammatory tissues, microbial prod-
ucts, and cellular debris that remains after cell necrosis. Lipoteichoic acid and peptidoglycan
recognition activate TLR2. LPS identification provokes TLR4 functioning [
39
]. Moreover,
signal transduction through TLR2 and TLR4 is stimulated by molecules of heat shock
protein 60, hyaluronic acid, and fibronectin products that are synthesized endogenously.
The TLR level intensifies in resident and infiltrating cells in the RA synovium. Furthermore,
TNF secretion is enhanced by increasing the TLR2 expression in CD16
+
macrophages of
blood and synovial origin in RA. The production of TLR2 grows with the assistance of
classical and intermediate monocytes in the RA peripheral blood and synovial joints. All
three monocyte subsets demonstrate a high TLR9 level on their surface independently of
blood or synovial localization. TLR2 and TLR9 promote the early secretion of proinflamma-
tory cytokines TNF and IL-1
β
[
40
]. Thereby, they induce inflammation that is maintained
by their other products, such as the cytokine IL-6 and the chemokine macrophage inflam-
matory protein-1. Various levels of these proinflammatory factors are correlated with the
involvement of TLR agonists. High expression of TLR3 and TLR4 is observed on fibroblasts
in the RA synovial joints. In particular, continuous inflammation and joint damage cause
this secretion [39].
Monocytes express the genes of type 1 interferons, especially in response to TLR
signaling. Roelofs M.F. et al. (2009) described the co-expression of synovial TLR3/7
expression with IFN-
α
, IL-1
β
, and IL-18, but not with TNF, IL-12, or IL-17 [
41
]. The
Diseases 2024,12, 81 7 of 20
stimulation of TLR3/TLR7 on monocytes, monocyte-derived DCs, or synovial fibroblasts
contributed to the secretion of type I IFN. However, no biologically active IL-1
β
or IL-
18 could be revealed. Type I IFN-
α
increased the TLR3/7 mRNA expression, whereas
IL-1
β
and IL-18 did not. In spite of the fact that the mRNA level of TLR4 remained
unchanged, IFN-
α
enhanced the response to TLR4 agonists, a phenomenon that was clearly
more marked in patients with RA. To conclude, type I IFNs are highly co-expressed with
TLR3/TLR7 in RA synovium. They enhance TLR3/TLR7-mediated cytokine production
and TLR4-mediated responses [41].
2.4.5. β1- and β2-Integrins
The migration of circulating monocytes proceeds after their adhesion into the vascular
endothelium.
β
1- and
β
2-integrins are expressed in high concentrations on monocytes
in RA and participate in the adhesion of monocytes to activated endothelial cells. The
β
1 subfamily of integrins includes very late antigen-4 (
α
4
β
1) and -5 (
α
5
β
1) that bind
to fibronectin and vascular cell adhesion molecule-1 in the endothelium and provoke
monocyte adhesion [
42
].
β
2-integrins take part in the formation of CD11
a,b,c
(CD18) com-
plexes in RA, which are mostly presented by CD11
b
molecules and associated with the
ligands in the endothelial lining. Moreover, intercellular adhesion molecule-1 (CD54),
-2 (CD102), and -3 (CD50) cause monocyte adhesion to endothelial cells. Inflammatory
monocytes/macrophages migrate into the synovial joints in RA and result in local inflam-
mation [43].
2.4.6. A Proliferation-Inducing Ligand
Two separate forms of a proliferation-inducing ligand (APRIL) are enhanced in the pro-
cess of RA progression. The first form is a soluble one, which refers to the TNF superfamily
and is secreted by myeloid cells, for instance, DCs, monocytes, macrophages, activated
T cells, and B cells. Soluble APRIL operates as a proinflammatory cytokine and binds to
receptors of two types. The first one is a transmembrane activator, calcium modulator
and cyclophilin ligand interactor receptor (TACI), which is produced in B cells [
44
]. The
second one is a B cell maturation antigen receptor (BCMA) that is secreted in plasma cells.
The second form of APRIL is a receptor on the cell surface that is highly produced by the
above-mentioned myeloid cells, including all the circulating monocyte subsets in RA. In
the process of binding to TACI and BCMA, a surface form of APRIL functions as a ligand
or receptor and provokes the formation and viability of B cells and plasma cells in RA. As a
result, a B cell-mediated autoimmune response is provided [45].
2.4.7. Sialic Acid Binding Ig-like Lectin
Siglec-1 is a representative of the sialic acid binding Ig-like lectin family. These proteins
undergo upregulation by the circulating monocytes and macrophages in RA, and their
concentration rises with the development of RA and increasing level of C-reactive protein,
erythrocyte sedimentation rate, and IgM-rheumatoid factor. Singlec-1 operates as an
autoantigen, takes part in a cell-to-cell contact of autoimmunity, and induces inflammation
in RA [46].
2.4.8. Transmembrane TNF
Transmembrane TNF is overexpressed on the monocyte surface in RA. Etanercept is
a TNF inhibitor that binds to transmembrane TNF. Consequently, the anti-inflammatory
cytokine IL-10 and soluble decoy receptors are produced. Thus, reverse signaling through
transmembrane TNF occurs in monocytes and represents a prognostic factor for the phar-
macological effect of anti-TNF agents in RA [
47
]. CD147 is an extracellular matrix metallo-
proteinase inducer, which promotes the production of matrix metalloproteinases (MMPs)
by monocytes, for instance, MMP2, MMP3, and MMP9. MMPs provoke the dissolution of
extracellular matrix, which leads to the destruction of synovium in RA. CD147 is hyperex-
pressed in CD14
+
monocytes in the RA peripheral blood and synovial joints. Infliximab
Diseases 2024,12, 81 8 of 20
is a TNF inhibitor that is supposed to block the CD147 secretion on the surface of CD14
+
monocytes in RA [48].
2.4.9. Other Monocyte Markers
α
-Enolase is an anti-citrullinated autoantigen for anti-citrullinated protein autoanti-
bodies and is also expressed on the surface of monocytes/macrophages in RA. The main
function of
α
-enolase is to initiate the secretion of proinflammatory cytokines, for example,
TNF, IFN-
γ
, prostaglandin E2, and IL-1
α
/
β
[
49
]. Proteinase-activated receptor-2 is a mem-
ber of the G protein receptor family and is produced by monocytes and T lymphocytes. The
expression of proteinase-activated receptor-2 is directly proportional to the RA activity. In
addition, the C-reactive protein and erythrocyte sedimentation rate concentrations increase
over the course of the disease. Proteinase-activated receptor-2 initiates the production of
the proinflammatory cytokine IL-6 [
50
]. Allograft inflammatory factor-1 is produced in the
cytoplasm of monocytes/macrophages, lymphocytes, and synovial fibroblasts. Its function
is to initiate and preserve inflammation in the synovial joints in RA. Hyperexpressed allo-
graft inflammatory factor-1 participates in the proliferation of inflammatory macrophages
and activated T lymphocytes in the RA synovium [
51
]. Moreover, 50–60% of CD14
+
CD16
++
non-classical monocytes have been shown to express the 6-sulfo LacNAc (SLAN) antigen,
which is an O-linked glycosylated variant of P-selectin glycoprotein ligand-1 recognized
by specific monoclonal antibodies, including macrophage-derived chemokine 8 and anti-
D-dimers 1 and 2 [
52
,
53
]. In clinical studies, Hofer Th.P. et al. (2015) showed a fivefold
depletion of SLAN-positive monocytes in patients with chronic inflammation [
54
]. SLAN is
one of the monocyte markers stimulating the differentiation of non-classical monocytes into
anti-inflammatory M2 macrophages, which, in turn, provide phagocytosis and resolution
of inflammation.
Monocytes of the peripheral blood are progenitors of macrophages, and both of them
mediate inflammation. Cardiovascular complications of RA are similar to synovitis and
subclinical atherosclerosis, which have an inflammatory mechanism in the endothelium.
Altered levels of the following receptor expression in circulating monocytes cause RA,
accompanied by atherosclerotic complications, for instance, localized in a coronary artery.
Specifically, low secretion of the scavenger receptor (CD36) is observed [
55
]. Moreover, a
comorbidity is characterized by the high production of the calcium-sensing receptor and
low-density lipoprotein-related receptor protein-1. The functional IL-25 receptor is secreted
on the monocyte surface in RA and interacts with the cytokine IL-25, which is produced
mostly by T cells. As a result, proinflammatory cytokine formation through the p38 mitogen-
activated protein kinase (MAPK)-dependent Soc-3 pathway occurs. Additionally, this
process signifies that IL-25 performs a negative control of monocyte-associated autoimmune
disorders [56].
2.5. Cytokines
The cytokines participating in the RA pathways are subdivided into two types: proin-
flammatory and anti-inflammatory cytokines. The first type is responsible for the occur-
rence and development of synovial inflammation [
57
]. Conversely, the functioning of
anti-inflammatory cytokines leads to a reduction in the inflammatory process. The cytokine
environment of the RA synovium is formed by the cells of two groups. The first one
includes resident synovial cells. The second group is represented by innate and adaptive
autoimmunity cells. A higher secretion of cytokines is provided in the RA synovium to a
greater extent by monocytes/macrophages and synovial fibroblasts than by T cells. The
cytokines are involved in all the stages of RA development. Moreover, they unite the
processes of immune regulation and inflammation that mediate the clinical symptoms and
pathogenesis of RA [58].
Diseases 2024,12, 81 9 of 20
2.5.1. Proinflammatory Cytokines
IL-2 and IFN-
γ
are the main proinflammatory cytokines of Th1 cells. IL-2 stimulates
the maturation and viability of T cells. IFN-
γ
initiates the functioning of HLA-DR. Moreover,
IFN-
γ
localized in the synovial joints in RA triggers the development of inflammatory M1
macrophages [
59
]. IL-17 is a pleiotropic cytokine secreted by Th17 cells. This cytokine
maintains the synovial inflammation. In addition, IL-17 participates in the activation
of monocytes and promotes their migration. Furthermore, the Th17 cytokine stimulates
the formation of cytokines by monocytes. There is a synergistic effect between IL-17
and TNF participating in the distribution of RA processes. Moreover, TNF increases
osteoclastogenesis, as well as between IL-17 and IL-1
β
. These advantageous interactions
lead to the activation of fibroblasts in the synovial joints, resulting in the production of
proinflammatory cytokines and MMPs. Additionally, the Th17 cytokine provokes the
erosion in the RA synovial joints. IL-17, in addition to TGF-
β
, IL-1
β
, and IL-23, increases
osteoclastogenesis in RA [60].
TNF is mostly secreted by activated synovial FLSs and inflammatory macrophages in
RA. Moreover, this cytokine is secreted by intermediate monocytes, T cells, and B cells. TNF
participates in the distribution of RA processes. Moreover, TNF increases osteoclastogenesis
in synovial inflammation. Infliximab is an effective anti-TNF agent that demonstrates the
central role of TNF in RA development [
61
]. The B cell activator factor is a representative
of the TNF family. Along with a soluble form of APRIL secreted by peripheral blood
mononuclear cells, the B cell activator factor is able to be linked to B cells, resulting in B
cell proliferation. Next, plasma cells are developed from autoreactive B cells and lead to
the formation of RF and ACPA. Consequently, a B cell-mediated response occurs in RA,
particularly in the early stages. Atacicept is an antagonist of APRIL and the B cell activator
factor and is undergoing clinical trials at the moment [62].
In addition, FLSs release other proinflammatory cytokines in the RA synovial joints,
for example, GM-CSF, IL-1
β
, IL-6, and IL-18. M1 macrophages ordinarily produce high
concentrations of TNF, IL-1
β
, IL-6, and IL-12. With the secretion of TNF, Il-1
β
, and IL-6,
monocytes stimulate an inflammatory process in the RA synovium. Adhesion molecules
β
1-
and
β
2-integrins are expressed on the monocyte surface under the effect of proinflammatory
cytokines. Intermediate monocytes are prevalent among the monocyte subsets that form
the proinflammatory cytokines Il-1
β
, IL-6, and TNF in RA. Moreover, classical monocytes
secrete the above-mentioned cytokines in accordance with the TLR agonists functioning [
63
].
The cytokine IL-18 is released by mononuclear cells in RA and is able to synergize with
Il-1
β
, IL-12, and IL-15. Next, these molecules trigger the IFN-
γ
formation by activated
synovial T cells and mediate the realization of the Th1 response. Moreover, IL-18 is capable
of acting as a straight proinflammatory cytokine in RA and causing the secretion of TNF
and IL-1βunder the control of macrophages [64].
NF-kB signaling plays an important role in RA pathogenesis. This statement is con-
firmed by the NF-kB participation in the production of macrophage inflammatory protein-
1a and the chemokine IL-8 [
65
]. Additionally, IL-15 stimulates neutrophils and delays the
apoptosis of FLSs and endothelial cells over the period of synovial inflammation. Synovial
fibroblasts are the main cells producing IL-23 in RA. Moreover, monocytes secrete IL-23
and initiate Th17 cell distribution and IL-17 expression in the RA synovial tissue. TNF,
IL-1
β
, and IL-32 trigger inflammation in the murine synovium that is supposed to indicate
their proinflammatory activity. IL-33 is a cytoplasmic representative of the IL-1 family and
is expressed in activated monocytes and fibroblasts in the RA synovium. IL-33 undergoes
emission from injured or necrotic cells and participates in the inflammation process. For
instance, this protein controls the formation of mast cells. Furthermore, it manages the
secretion of Th2 cytokines, such as IL-4, IL-5, and IL-13. Next, IL-33 promotes Th2-regulated
pathological conditions [66].
Diseases 2024,12, 81 10 of 20
2.5.2. Anti-Inflammatory Cytokines
IL-1Ra is an IL-1 receptor antagonist formed by monocytes in RA. IL-1Ra and IL-
10 are anti-inflammatory cytokines; therefore, their antagonistic action presupposes the
decline and termination of inflammation. Nevertheless, IL-10 and TGF-
β
prevail among
the products of Breg cells, Treg cells, and suppressor cells of myeloid origin. Il-4 and IL-10
are also secreted by Th2 cells. Moreover, Th2 cells enhance IL-1Ra expression. In addition,
M2 macrophages secrete IL-10 and contribute to wound healing, tissue remodeling, and
inflammation arrest. IL-10, as an anti-inflammatory cytokine, decreases inflammation.
Therefore, a decrease in IL-10 production leads to the development of the inflammatory
process and its transformation into a chronic form. IL-10 and M-CSF added to the cultivation
medium promote monocyte survival, growth, and differentiation into macrophages [
67
].
Furthermore, monocytes express the cytokines IL-11, IL-19, IL-20, and IL-24. Additionally,
the products of monocyte secretion are GM-CSF, G-CSF, and oncostatin M [68].
2.6. Chemokine Receptors
Chemokines binding to their receptors on the monocyte surface cause the return and
migration of monocytes from the blood into the synovial tissue. CCR2 and CX3CR1 are
chemokine receptors that are located on monocytes and trigger their chemotaxis. The first
receptor is overexpressed on classical CD14
++
CD16
monocytes and binds to CCL2, which
is a monocyte chemoattractant protein [
69
]. The second receptor undergoes high expression
on predominantly intermediate CD14
++
CD16
+
monocytes and also non-classical CD14
+
CD16
++
monocytes in RA and interacts with CX3CL1. IL-1
β
cytokines and TNF induce
the synovial fibroblasts to produce mainly CCL2, CX3CL1, and CCL20 in RA. CCL20,
or macrophage inflammatory protein-3 alpha, is a different monocyte chemoattractant.
Moreover, the c-Jun N-terminal kinase, extracellular signal-regulated kinase, and phos-
phatidylinositol 3
–kinase signaling pathways participate in the chemoattractant secretion
by the Th17-associated cytokine IL-17. In addition, IL-17 has an indirect influence on
monocyte migration [70].
Chemokine receptors undergo constitutive expression on peripheral blood and syn-
ovial monocytes/macrophages, mediate the monocyte chemotaxis, and maintain their
level. The migration of peripheral blood monocytes to the synovial tissue during the in-
flammatory process promotes the increased concentration of chemokine receptors on their
surface in RA [
6
]. Therefore, the regional inflammation and monocyte preservation in the
synovium are defined by the overexpression of chemokine receptors on the RA monocytes.
The increased expression of CCR1, CCR2, and CCR4 receptors is provided on peripheral
blood monocytes, whereas synovial monocytes demonstrate high production of CCR3 and
CCR5 in RA. Furthermore, the circulating monocytes secrete CCR9, which interacts with
the CCL25 chemokine, or thymus-expressed chemokine. This binding results in monocytes’
development into macrophages in RA. The CCR7 chemokine receptor is secreted by not
only B lymphocytes, CD4
+
T lymphocytes, DCs, and FLSs but also circulating monocytes in
RA. CCR7 binds to the CCL19 and CCL21 ligands that are characterized by hyperexpression
in RA. The regulation of CCR7 and CCL19 is directly proportional to the RA progression.
Moreover, the CCL19 concentration in plasma indicates the radiographic development of
joint destruction. The DMARD therapy maintains the normal level of both components of
the CCR7/CCL19 system [26].
3. Monocyte Functions in RA
The RA etiopathogenesis and its sensitivity to therapy are defined by genetic pre-
disposition, environmental factors, infections, and the decline of autoimmune regulation.
Frequent inflammation induces the development of autoimmune conditions. The patho-
genesis of RA involves the activation and polarization of monocytes into proinflammatory
M1 macrophages within the joint environment [
71
]. In the arthritic joint, monocytes are
recruited by proinflammatory cytokines such as TNF, IL-1, and IL-6, which are produced
by resident cells, including FLSs and macrophages. These cytokines promote the activation
Diseases 2024,12, 81 11 of 20
of monocytes through the binding of their receptors, leading to the upregulation of adhe-
sion molecules and chemoattractant receptors on the monocyte surface. As a result, the
monocytes adhere to the endothelium of blood vessels and migrate into the synovial tissue.
Activated monocytes express increased levels of cell surface markers, such as CD80 and
CD86, which facilitate interactions with T cells and promote antigen presentation [
72
]. M1-
like macrophages are characterized by the production of proinflammatory cytokines and
reactive oxygen species, which contribute to tissue damage and perpetuate inflammation in
the joint. Furthermore, M1-like macrophages express high levels of surface markers, such
as CD14 and CD16, which enhance their phagocytic and antigen-presenting capabilities.
3.1. Monocyte Interactions with Other Cells
Cells of innate and adaptive immunity work in cooperation to cause and maintain
an inflammatory process in RA. The main cells taking part in this signaling are DCs,
monocytes/macrophages, FLSs, CD4
+
T cells (Th1, Th2, and Th17 cells), B cells, mast
cells, and neutrophils. When immunoregulatory cells are not capable of attenuating acute
inflammation, a chronic form of the disease develops. DCs, macrophages, and FLSs are
activated at the beginning of RA pathogenesis [73].
Circulating monocytes are precursors of macrophages. Additionally, monocytes are
able to operate as APCs and activate T cells. Moreover, they are classified as monocyte-
derived DCs due to their capacity to behave as classical DCs [
7
]. Monocyte-derived DCs
perform hyperexpression of IL-6 and IL-23. Tolerogenic monocyte-derived DCs are another
type of DC that are capable of triggering the formation of Foxp3
+
Treg cells in RA. Myeloid-
derived suppressor cells (MDSCs), Breg cells, and Treg cells control and decrease the
inflammatory process [74].
Among CD4
+
T cells, the role of the Th1 and Th2 cells in the RA progression was ini-
tially revealed. The cytokine IL-12 mediates the participation of monocytes/macrophages
in the polarization of CD4
+
Th1 cells. Th2 cells and their cytokines mainly decrease Th1
activation. Intermediate monocytes prevail in the RA peripheral blood and synovium and
are supposed to be the main monocyte subsets that modulate the Th17 cell functioning.
Th17 cells were investigated as effectors in RA because of the ability of their pleiotropic
cytokine IL-17 to synergize with TNF. IL-17A triggers the secretion of proinflammatory
cytokines IL-1
β
, IL-6, and TNF in macrophages through the AP-1, NF-kB, and MAPK
pathways [
60
]. IL-1
β
, IL-6, and IL-23 initiate the activation and control the polarization of
Th17 cells. Activated T lymphocytes take part in cell-to-cell contact and trigger the secretion
of TNF and IL-1
β
. Moreover, T lymphocytes release cytokine inhibitor secretory IL-1Ra in
monocytes/macrophages [75].
Mast cells are a source of small-molecule inflammatory mediators. Neutrophils un-
dergo chemotaxis into the RA synovial joints, mostly in the early period of inflammation,
and release inflammatory mediators and enzymes. Over the period of joint effusion,
the level of neutrophils predominates in the synovial fluid in comparison with the syn-
ovium [76].
A cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) is a receptor for co-stimulatory
molecules and is expressed on the surface of activated T cells [
77
]. CTLA4 binds with a high
avidity to CD80 (B7-1) and CD86 (B7-2) on APCs. They are expressed on the APC surface
and interact with T cell co-stimulatory receptor CD28 through signal 2 in order to activate
the T cells. Moreover, CTLA4 promotes CD80 and CD86 transport to the CD28 receptor
located on T cells for further activation of these co-stimulatory molecules [
72
]. CTLA4-Ig is
a CTLA4 inhibitor and is registered as Abatacept. A blocking effect of Abatacept indicates
a cell-to-cell contact mechanism of antigen presentation via T cell-monocyte/macrophage
cooperation [77].
Moreover, a surface form of APRIL secreted by monocytes promotes cell communica-
tion with B cells and plasma cells that produce BCMA and TACI [
44
]. In addition, FLSs
secrete soluble APRIL and, consequently, release BCMA. Furthermore, FLSs and synovial
macrophages express CXCL16 and CCL20 chemokines, which interact with CCR6
+
mem-
Diseases 2024,12, 81 12 of 20
ory T cells. This communication, along with IL-15 cytokines, mainly contributes to CD4
+
T cell migration and their involvement in the synovial tissue. Moreover, the enhanced
transport of circulating monocytes into the RA synovial joints stimulates the participation
of CXCR6
+
T cells in the RA progression. Additionally, the CCL20 expression by activated
monocytes occurs in the synovium of mice. Stimulated monocytes secrete CCL20, which
contacts the CCR6 chemokine receptors, a product of Th17 cells. This interaction increases
the CCL20 involvement in the synovial tissue. To sum up, cooperation between mono-
cytes/macrophages, DCs, B cells, Th1, and Th17 cells is related to a positive feedback loop
and has an effect on the continuation of inflammation in the synovial joints [60].
FLSs are the resident cells presenting in the intimal lining of the synovium along with
resident macrophages, which are synoviocytes of type A. FLSs are highly proliferated in
the synovial tissue, thus stimulating the hyperplasia of the synovium and damage to the
joints. FLSs express RANKL and participate in the differentiation of osteoclasts and bone
erosion [
78
]. CCL2 and CX3CL1 are FLS chemokines and behave as chemoattractants for
monocytes/macrophages. Moreover, the corresponding MMPs eliminate the extracellular
matrix. IL-6, IL-18, TNF, and GM-CSF, as the FLS cytokines, promote the stimulation of
innate and adaptive immune cells [61].
Treg cells primarily decrease inflammation and regulate adaptive immunity. The Treg
cells are determined as CD4
+
CD25
+
CD127
low
FOXP3
+
cells. There are two ways in which
they cooperate with APCs and effector T cells. The first one is direct communication through
cell-to-cell contact. The second way occurs with the help of anti-inflammatory cytokines, for
instance, TGF-
β
, IL-4/IL-13, and IL-10. Furthermore, IL-10, as a potent anti-inflammatory
cytokine, influences both the innate and adaptive immune cells [
5
]. CD8
+
FoxP3
+
Treg cells
induced by anti-CD3 monoclonal antibodies decrease Th1 and Th17 cell activation through
P38 phosphorylation inhibition and halt RA inflammation [
74
]. Monocytes promote the
FoxP3 expression in CD8
+
FoxP3
+
Treg cells through direct cell contact. IL-10 decreases the
Th17 cell amount and the IL-17 cytokine generation while simultaneously increasing the
Foxp3
+
Treg cell development. Thus, IL-10 modifies the ratio of Th17 cells and Treg cells in
the CD4
+
T cell population. Activated monocytes/macrophages are able to have both a
positive and negative effect on the Treg cell phenotype and function, with the predominance
of a negative one [74].
MDSCs are tumor-associated cells and take part in the formation of tumor-induced
T regulatory cells and T cell anergy. There are two subsets of MDSCs. The first one is
presented by monocytic MDSCs. Consequently, monocytes can be expanded and polarized
towards MDSCs [
13
]. The second group includes granulocytic MDSCs. Both of these sub-
sets suppress Th1 and Th17 cell proliferation. Moreover, they secrete IL-10 and contribute
to the expansion of Treg cells in mice. To sum up, MDSCs possess immunoregulatory
activity and repress RA synovial inflammation [4].
3.2. Monocyte Transport to the RA Synovial Tissue
Monocytes are the important innate effectors in the progression of RA. Circulating
monocytes undergo chemotaxis and infiltration into the synovial tissue and stimulate
synovial inflammation. Intermediate monocytes, and, to a high extent, the classical ones,
are constantly involved in the local synovial inflammation development. This statement is
confirmed by the increased presence of both of them in the RA synovium as well as the
declining concentration of classical monocytes in the RA peripheral blood and a slightly
enhanced number of intermediate monocytes there. M-CSF promotes the differentiation of
classical monocytes into the intermediate subset, which indicates the heterogeneous charac-
ter of the monocyte subsets and their possible polarization into their own other type [
13
].
The cooperation between CCL2/CCR2 and CX3CL1/CX3CR1 contributes to the migration
of circulating monocytes and their functioning in the RA synovium. CCL2 is released by
bone marrow mesenchymal stem cells and precursor cells. The CCR2/CCl2 complex on
monocytes triggers their recruitment from the bone marrow into the bloodstream. The
CX3CL1 production by FLSs is supported by disintegrin and metalloproteinase domain-
Diseases 2024,12, 81 13 of 20
containing protein 10 and disintegrin; therefore, they mediate the monocyte migration in
RA [79].
3.3. Differentiation of Monocytes into Macrophages
In RA, monocytes are replaced by circulating monocytes, which constantly enter
the synovium to support the local inflammation, although their part among the synovial
macrophages is small. GM-CSF is released by activated synovial fibroblasts under the influ-
ence of IL-1
β
and TNF and provokes the survival of monocytes more than their polarization
into macrophages [
67
]. The human stem cells from the fetal liver and erythro-myeloid
predecessors supply the resident macrophages over the period of embryogenesis. However,
circulating monocytes supply the recruited inflammatory macrophages when it is necessary,
for example, in acute synovial inflammation. When the niche in the synovial tissue is free
or unsaturated, macrophage development from circulating monocytes occurs. When there
is no available niche, the macrophages are self-replenished by their own turnover. Interme-
diate monocyte subsets (CD14++ CD16+) are the prevailing monocytes in the RA synovial
tissue, and they mainly differentiate into inflammatory M1 macrophages [
30
]. Synovial
macrophages are located in the sub-lining and lining layers at the cartilage–pannus junction.
The activated macrophages of the intimal lining release cytokines and provoke articular de-
struction. The rheumatoid factor autoantibodies and antigens form IgG-containing immune
complexes, which stimulate the macrophages. Furthermore, innate immune cells, T cells,
and fibroblasts secrete cytokines that have an impact on the stimulation of macrophages
via cell-to-cell contact. The involved TLR-2 and TLR-4 activate macrophages and maintain
their level in RA [39].
There are two major phenotypes of macrophages that are differentiated from mono-
cytes in humans. The first is a proinflammatory type represented by classically activated
M1 M
φ
macrophages. They secrete high levels of IL-1
β
, IL-6, IL-12, IL-23, TNF, and
reactive oxygen species, which have the main impact on the continuation of the RA in-
flammation. The second type is anti-inflammatory and refers to alternatively activated
M
φ
M2 macrophages [
80
]. They, in turn, provide high concentrations of TGF-
β
, IL-1Ra,
decoy IL-1RII, IL-10, and low IL-12 content. The M
φ
M2 macrophage functions are anti-
inflammatory activity, tissue remodeling, and wound healing. Both the proinflammatory
and anti-inflammatory macrophage phenotypes take part in inflammation regulation. Their
differentiation from monocytes with the use of the GM-CSF and M-CSF induction cytokines
leads to M1 and Mø2 M2 formation, correspondingly [
68
]. Moreover, activated M
φ
M2 macrophages have three subsets according to their stimulating cytokines. There are
three M2 forms. The first one is an alternative M2 form called M2a and triggered by IL-4
and IL-13. The second one is M2b, which is initiated by the impact of the IgG-containing
immune complex and the TLR agonists or IL-1R. The third M2 form is M2c, which is
induced by IL-10 and glucocorticoid hormones [60].
Th1 cells, Th2 cells, and Th17 cells, along with their associated cytokines, play
a crucial role in the polarization, recruitment, activation, and differentiation of mono-
cytes/macrophages [
59
]. The activation and differentiation of M1 macrophages are driven
by the Th1 cytokine IFN-
γ
in the presence of LPS and TNF. On the other hand, M2
macrophages are driven by Th2 cytokines such as IL-4, IL-10, and IL-13. CXCL5, CXCL8,
CXCL9, CXCL10, and CXCL13 are typical chemokines expressed by M1 macrophages. No-
tably,
in vitro
studies have demonstrated that M-CSF polarized M2 macrophages can pro-
duce proinflammatory cytokines in response to an IgG immune complex containing ACPA.
This finding is also relevant in the context of RA since the synovium exhibits elevated levels
of M-CSF [
67
]. Furthermore, activin A, found in RA synovial fluid, induces increased ex-
pression of MMP12 (a proinflammatory polarization marker) on monocytes/macrophages,
polarizing them into proinflammatory M1 phenotypes. Given these findings, activin A
levels may serve as a potential biomarker for various therapies.
The overexpression of the Silent Information Regulator-2 homolog, SIRT1, on mono-
cytes has been shown to downregulate PU.1 phosphorylation, thereby inhibiting their
Diseases 2024,12, 81 14 of 20
differentiation into macrophages. SIRT1 overexpression can inhibit the production of proin-
flammatory cytokines by blocking the NF-kB pathway in RA patients. Consequently, SIRT1
has emerged as a significant therapeutic target of interest due to its role in regulating the
inflammatory process in RA [
81
]. It is worth mentioning that monocytes/macrophages
secrete the cytosolic phospholipase A2a enzyme, which plays a crucial role in generating
prostaglandin E2 from cell membrane phospholipids [
82
]. Prostaglandin E2, in turn, con-
tributes to the induction and maintenance of RA inflammation. In addition, under the
influence of LPS, TNF, and IL-1
β
, osteoblastic stromal cells produce cytosolic phospholi-
pase A2a, ultimately leading to the generation of prostaglandin E2. This process has been
associated with the promotion of inflammatory bone resorption in RA.
3.4. Monocytes Are Osteoclast Predecessors
Periarticular osteopenia and bone erosion adjacent to the pannus formation of synovial
joints are two of the most destructive events observed in RA with high disease activity. The
main culprits responsible for this erosion are osteoclasts, the primary bone resorbing cells.
Osteoclasts originate from circulating monocytes and resident macrophages, which contin-
uously migrate into the inflamed synovium and differentiate into osteoclasts, particularly
in active disease and bone erosion. Recent research has shown that monocytes expressing
CD14
+
and lacking CD16
are the main precursors of osteoclast progenitors, as opposed to
CD16
+
subsets [
12
]. These monocytes upregulate the RANK on their surface and interact
with RANKL, which is predominantly expressed by FLSs, osteoblasts, and activated T cells.
Th17 cells also play a significant role in promoting osteoclastogenesis as they stimulate the
expression of RANKL, which preferably interacts with the CD14
+
monocytes expressing
the surface marker CCR6, characteristic of Th17 cells [
83
]. Furthermore, the production of
IL-17, along with TNF, IL-1
β
, and IL-6, can amplify osteoclastogenesis. Additionally, the
mesenchymal stromal cells secreted by FLSs, when synergized with RANKL, also aid in
promoting osteoclastogenesis [56].
The mature activated osteoclasts release hydrochloric acid near their ruffled border
to dissolve the calcium from the bone matrix and also produce MMPs and cathepsin K,
which break down the remaining bone matrix, resulting in bone erosion adjacent to the
joints [
48
]. RA monocytes exhibit increased expression of the co-stimulatory osteoclast-
associated receptor, which, upon interaction with the collagen type II and collagen type
I ligands in the inflamed synovial articular cartilage, further promotes the formation of
osteoclasts. However, the osteoblasts’ soluble decoy receptor osteoprotegerin negatively
affects osteoclastogenesis and disrupts the osteoprotegerin/RANK ratio, consequently
favoring osteogenesis [
12
]. The activated T cells expressing the inducible costimulator,
when interacting with the inducible costimulator ligand (CD275) expressed by monocyte-
derived osteoclast-like cells, obstruct their differentiation into osteoclasts. This interaction
suppresses the expression of tartrate-resistant acid phosphatase, the nuclear factor of
activated T cells, and the osteoclast-associated receptors in monocyte-derived osteoclast-
like cells during their maturation into osteoclasts. Thus, the CD275-inducible costimulator
system directly interferes with osteoclastogenesis [84].
Clinical trials in RA focused on the monocytes that are of interest. The Chinese
government approved sinomenine as an anti-inflammation drug for RA treatment. Liu W.
et al. (2018) screened the various secretory cytokines in both LPS-induced and sinomenine-
treated RAW264.7 cells, followed by an estimation of the sinomenine ability to modulate
the cytokine secretion in a cell model, a collagen-induced arthritis mouse model, and RA
patients [
2
]. The results demonstrated that sinomenine regulated the IL-6, GM-CSF, IL-12
p40, IL-1
α
, TNF, IL-1
β
, CXCL1, Eotaxin-2, IL-10, M-CSF, RANTES, and CCL2 secretion
in vivo
and
in vitro
and reduced the RA activity and the 28-joint disease activity score in
a clinical setting. Moreover, sinomenine attenuated the CD11b
+
F4/80
+
CD64
+
resident
macrophages in the synovial tissue, the CD11b
+
Ly6C
+
CD43
+
macrophages in the spleen,
and draining the lymph nodes in collagen-induced arthritic mice. The percentage of
CD14
+
CD16
+
peripheral blood mononuclear cells was reduced by sinomenine in the
Diseases 2024,12, 81 15 of 20
RA patients. In conclusion, sinomenine controls the secretion of multiple inflammatory
cytokines and monocyte/macrophage subsets, thus decreasing RA development. Along
with methotrexate, sinomenine could be an alternative as a cost-effective anti-inflammatory
agent for treating RA [2].
4. Discussion
The innate and adaptive autoimmunity cells within the interconnected network drive
the continuous progression of RA pathogenesis. Numerous studies conducted to date, as
well as targeted therapies, have demonstrated that FLSs, macrophages, Th1 cells, and B
cells play a pivotal role in initiating and advancing synovial inflammation [
85
]. Recently,
Th17 cells have been implicated as the primary drivers of immune cell activation and
cytokine production [
60
]. Furthermore, they are primarily responsible for inducing RANKL
production by FLSs and osteoblasts, thereby promoting osteoclastogenesis [
78
]. Presently,
the data suggest that monocyte activation occurs within the circulation of individuals with
RA. These activated monocytes respond to chemotactic ligands such as CCL2 and CX3CL1,
migrating from the circulation into the RA synovium and releasing proinflammatory
cytokines [
83
]. Thus, their involvement in perpetuating synovial inflammation in RA
cannot be disregarded. Additionally, they function as APCs, activating other autoimmune
cells and stimulating the production of their respective cytokines. Evidently, they partake
in a positive feedback loop with the other key cells involved in RA pathogenesis. The
increased expression of various antigens on the monocyte membrane surface can be a
potential target for therapy in RA. Evaluating the levels of total monocytes and their
subsets in the peripheral blood of the RA patients, both before and after therapy, can
provide rheumatologists with guidance in determining the preference for treatment with
DMARDs or biologic agents [
26
]. Elevated levels of monocytes in the peripheral blood of
RA patients may serve as an additional biomarker for high disease activity, as evidenced by
the correlation with the disease activity score-28 scores and the serum levels of C-reactive
protein and the erythrocyte sedimentation rate.
The display of a heterogeneous character by the monocytes enables the differentiation
into subsets based on the microenvironment or inflammatory stages and, more impor-
tantly, the ability to transform into intermediate monocytes. This emphasizes the high
plasticity of the monocytes in the pathogenesis of RA. Inflammatory M1 macrophages,
which are crucial players in the development of RA, are primarily derived from circulating
monocytes, specifically intermediate monocytes [
2
]. This raises the question of which
chemical events, signals, or specific factors, apart from the arthritic joint environment,
trigger the conversion of CD14 monocytes to CD14
++
CD16
+
intermediate inflammatory
macrophages [
86
]. Focusing on these studies would not only benefit RA patients but also
shed light on other diseases where CD14
++
CD16
+
macrophages play a significant role,
such as atherosclerosis. Classical monocytes expressing CD14
++
and CD16
are responsible
for the bone erosion observed in erosive RA [
87
]. Osteoclastogenesis, the formation of
osteoclasts, is influenced by the presence of FLSs and Th17 cells. FLSs produce RANKL
and M-CSF to promote osteoclastogenesis [
67
]. Additionally, IL-17, produced by Th17 cells,
induces the production of RANKL by FLSs. Furthermore, IL-17, along with TNF, IL-1
β
,
and IL-6, amplifies osteoclastogenesis [
88
]. However, it is equally important to consider
the availability of classical monocytes in the RA synovium or microcirculation near RA
synovial joints in erosive rheumatoid arthritis as they have been shown to be precursor
cells capable of differentiating into osteoclasts.
5. Conclusions
Recent discussions have highlighted the significant advancements in our understand-
ing of how innate immune cells and signaling play a crucial role in driving the pathogenesis
of RA. This complexity of the disease underscores the importance of targeting the innate
immune system and its components for potential therapeutic interventions that can reduce
the incidence and enhance the quality of life for RA patients. To further explore this,
Diseases 2024,12, 81 16 of 20
additional research is needed to investigate how the heterogeneity and plasticity of mono-
cytes, the antigenic expressions on their surface, the differentiation into macrophages and
osteoclasts, the migration into the synovium, and the role of their cytokines in RA influence
their function in RA development. Moreover, studying how macrophage polarization in
inflamed joints changes during the course of RA can provide valuable insights into the
intricate and pivotal role of these cells in the disease process.
Author Contributions: Conceptualization, D.I.S. and N.G.N.; methodology, A.N.O.; software, D.I.S.;
validation, D.I.S., N.G.N. and A.N.O.; formal analysis, A.Y.P.; investigation, D.I.S.; resources, N.G.N.;
data curation, D.I.S.; writing—original draft preparation, D.I.S.; writing—review and editing, N.G.N.;
visualization, D.I.S.; supervision, N.G.N.; project administration, A.Y.P.; funding acquisition, A.Y.P.
All authors have read and agreed to the published version of the manuscript.
Funding: This research was funded by the Russian Scientific Foundation, grant number 20-15-00337.
The APC was funded by 20-15-00337.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Data available from the authors.
Acknowledgments: The authors would like to thank Alina I. Salnikova, translator and interpreter at
the European Medical Center (Moscow, Russia), for her kind assistance in English editing. Figure
was created with Biorender.com, accessed on 1 January 2024.
Conflicts of Interest: The authors declare no conflicts of interest. The funders had no role in the design
of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or
in the decision to publish the results.
References
1.
Cheng, Q.; Wu, H.; Du, Y. The roles of small-molecule inflammatory mediators in rheumatoid arthritis. Scand. J. Immunol. 2021,
93, e12982. [CrossRef] [PubMed]
2.
Liu, W.; Zhang, Y.; Zhu, W.; Ma, C.; Ruan, J.; Long, H.; Wang, Y. Sinomenine Inhibits the Progression of Rheumatoid Arthritis
by Regulating the Secretion of Inflammatory Cytokines and Monocyte/Macrophage Subsets. Front. Immunol. 2018,26, 2228.
[CrossRef] [PubMed]
3. Balchin, C.; Tan, A.L.; Golding, J.; Bissell, L.-A.; Wilson, O.J.; McKenna, J.; Stavropoulos-Kalinoglou, A. Acute effects of exercise
on pain symptoms, clinical inflammatory markers and inflammatory cytokines in people with rheumatoid arthritis: A systematic
literature review. Ther. Adv. Musculoskelet. Dis. 2022,14, 1759720X221114104. [CrossRef] [PubMed]
4.
Morell, M.; Varela, N.; Marañón, C. Myeloid Populations in Systemic Autoimmune Diseases. Clin. Rev. Allergy Immunol. 2017,53,
198–218. [CrossRef] [PubMed]
5.
Mishra, S.; Srinivasan, S.; Ma, C.; Zhang, N. CD8+ Regulatory T Cell—A Mystery to Be Revealed. Front. Immunol. 2021,12,
708874. [CrossRef] [PubMed]
6.
Choi, J.; Selmi, C.; Leung, P.S.C.; Kenny, T.P.; Roskams, T.; Gershwin, M.E. Chemokine and chemokine receptors in autoimmunity:
The case of primary biliary cholangitis. Expert Rev. Clin. Immunol. 2016,12, 661–672. [CrossRef] [PubMed]
7.
Ziegler-Heitbrock, L.; Ancuta, P.; Crowe, S.; Dalod, M.; Grau, V.; Hart, D.N.; Leenen, P.J.M.; Liu, Y.-J.; MacPherson, G.; Randolph,
G.J.; et al. Nomenclature of monocytes and dendritic cells in blood. Blood 2010,116, e74–e80. [CrossRef] [PubMed]
8.
Liu, E.G.; Yin, X.; Swaminathan, A.; Eisenbarth, S.C. Antigen-Presenting Cells in Food Tolerance and Allergy. Front. Immunol.
2021,11, 616020. [CrossRef] [PubMed]
9.
Li, L.; Huang, L.; Sung, S.-S.J.; Vergis, A.L.; Rosin, D.L.; Rose, C.E., Jr.; Lobo, P.I.; Okusa, M.D. The chemokine receptors CCR2
and CX3CR1 mediate monocyte/macrophage trafficking in kidney ischemia-reperfusion injury. Kidney Int. 2008,74, 1526–1537.
[CrossRef]
10.
Wolf, A.A.; Yáñez, A.; Barman, P.K.; Goodridge, H.S. The Ontogeny of Monocyte Subsets. Front. Immunol. 2019,10, 1642.
[CrossRef]
11.
Mushenkova, N.V.; Nikiforov, N.G.; Shakhpazyan, N.K.; Orekhova, V.A.; Sadykhov, N.K.; Orekhov, A.N. Phenotype Diversity of
Macrophages in Osteoarthritis: Implications for Development of Macrophage Modulating Therapies. Int. J. Mol. Sci. 2022,23,
8381. [CrossRef] [PubMed]
12.
Komano, Y.; Nanki, T.; Hayashida, K.; Taniguchi, K.; Miyasaka, N. Identification of a human peripheral blood monocyte subset
that differentiates into osteoclasts. Arthritis Res. Ther. 2006,8, R152. [CrossRef] [PubMed]
Diseases 2024,12, 81 17 of 20
13.
Chen, S.; Guo, C.; Wang, R.; Feng, Z.; Liu, Z.; Wu, L.; Zhao, D.; Zheng, S.; Chen, F.; Zhang, D.; et al. Monocytic MDSCs skew
Th17 cells toward a pro-osteoclastogenic phenotype and potentiate bone erosion in rheumatoid arthritis. Rheumatology 2021,60,
2409–2420. [CrossRef] [PubMed]
14.
Luukkonen, J.; Huhtakangas, J.; Palosaari, S.; Tuukkanen, J.; Vuolteenaho, O.; Lehenkari, P. Preliminary Report: Osteoarthritis
and Rheumatoid Arthritis Synovial Fluid Increased Osteoclastogenesis In Vitro by Monocyte Differentiation Pathway Regulating
Cytokines. Mediators Inflamm. 2022,2022, 2606916. [CrossRef] [PubMed]
15.
De Kleer, I.; Willems, F.; Lambrecht, B.; Goriely, S. Ontogeny of myeloid cells. Front. Immunol. 2014,5, 423. [CrossRef] [PubMed]
16.
Weaver, L.K.; Chu, N.; Behrens, E.M. TLR9-mediated inflammation drives a Ccr2-independent peripheral monocytosis through
enhanced extramedullary monocytopoiesis. Proc. Natl. Acad. Sci. USA 2016,113, 10944–10949. [CrossRef] [PubMed]
17.
Krasselt, M.; Baerwald, C.; Wagner, U.; Rossol, M. CD56+ monocytes have a dysregulated cytokine response to lipopolysaccharide
and accumulate in rheumatoid arthritis and immunosenescence. Arthritis Res. Ther. 2013,15, R139. [CrossRef] [PubMed]
18.
Sørensen, A.S.; Andersen, M.N.; Juul-Madsen, K.; Broksø, A.D.; Skejø, C.; Schmidt, H.; Vorup-Jensen, T.; Kragstrup, T.W. Tumor
necrosis factor alpha neutralization attenuates immune checkpoint inhibitor-induced activation of intermediate monocytes in
synovial fluid mononuclear cells from patients with inflammatory arthritis. Arthritis Res. Ther. 2022,24, 43. [CrossRef] [PubMed]
19.
Tsukamoto, M.; Seta, N.; Yoshimoto, K.; Suzuki, K.; Yamaoka, K.; Takeuchi, T. CD14brightCD16+ intermediate monocytes are
induced by interleukin-10 and positively correlate with disease activity in rheumatoid arthritis. Arthritis Res. Ther. 2017,19, 28.
[CrossRef]
20.
Chara, L.; Sánchez-Atrio, A.; Pérez, A.; Cuende, E.; Albarrán, F.; Turrión, A.; Chevarria, J.; del Barco, A.A.; Sánchez, M.A.;
Monserrat, J.; et al. The number of circulating monocytes as biomarkers of the clinical response to methotrexate in untreated
patients with rheumatoid arthritis. J. Transl. Med. 2015,13, 2. [CrossRef]
21.
Buscher, K.; Marcovecchio, P.; Hedrick, C.C.; Ley, K. Patrolling Mechanics of Non-Classical Monocytes in Vascular Inflammation.
Front. Cardiovasc. Med. 2017,4, 80. [CrossRef]
22.
Ong, S.-M.; Hadadi, E.; Dang, T.-M.; Yeap, W.-H.; Tan, C.T.-Y.; Ng, T.-P.; Larbi, A.; Wong, S.-C. The pro-inflammatory phenotype
of the human non-classical monocyte subset is attributed to senescence. Cell Death Dis. 2018,9, 266. [CrossRef] [PubMed]
23.
Rodríguez-Vargas, G.-S.; Santos-Moreno, P.; Rubio-Rubio, J.-A.; Bautista-Niño, P.-K.; Echeverri, D.; Gutiérrez-Castañeda, L.-D.;
Sierra-Matamoros, F.; Navarrete, S.; Aparicio, A.; Saenz, L.; et al. Vascular Age, Metabolic Panel, Cardiovascular Risk and
Inflammaging in Patients with Rheumatoid Arthritis Compared with Patients with Osteoarthritis. Front. Cardiovasc. Med. 2022,9,
894577. [CrossRef] [PubMed]
24.
Frouin, I.; Prosperi, E.; Denegri, M.; Negri, C.; Donzelli, M.; Rossi, L.; Riva, F.; Stefanini, M.; Scovassi, A.I. Different effects of
methotrexate on DNA mismatch repair proficient and deficient cells. Eur. J. Cancer 2001,37, 1173–1180. [CrossRef] [PubMed]
25.
Greenstein, R.J.; Su, L.; Shahidi, A.; Brown, S.T. On the action of 5-amino-salicylic acid and sulfapyridine on M. avium including
subspecies paratuberculosis. PLoS ONE 2007,2, e516. [CrossRef] [PubMed]
26.
Deng, G.; Chen, X.; Shao, L.; Wu, Q.; Wang, S. Effectiveness and safety of 99Tc-methylene diphosphonate as a disease-modifying
anti-rheumatic drug (DMARD) in combination with conventional synthetic (cs) DMARDs in the treatment of rheumatoid arthritis:
A systematic review and meta-analysis of 34 randomized controlled trials. Heliyon 2023,9, e21691. [CrossRef] [PubMed]
27.
Ammari, M.; Presumey, J.; Ponsolles, C.; Roussignol, G.; Roubert, C.; Escriou, V.; Toupet, K.; Mausset-Bonnefont, A.-L.; Cren,
M.; Robin, M.; et al. Delivery of miR-146a to Ly6Chigh Monocytes Inhibits Pathogenic Bone Erosion in Inflammatory Arthritis.
Theranostics 2018,8, 5972–5985. [CrossRef] [PubMed]
28.
Ledesma-Colunga, M.G.; Baschant, U.; Weidner, H.; Alves, T.C.; Mirtschink, P.; Hofbauer, L.C.; Rauner, M. Transferrin receptor 2
deficiency promotes macrophage polarization and inflammatory arthritis. Redox Biol. 2023,60, 102616. [CrossRef] [PubMed]
29.
Roberts, C.A.; Dickinson, A.K.; Taams, L.S. The Interplay Between Monocytes/Macrophages and CD4(+) T Cell Subsets in
Rheumatoid Arthritis. Front. Immunol. 2015,6, 571. [CrossRef]
30.
Zamani, F.; Shahneh, F.Z.; Aghebati-Maleki, L.; Baradaran, B. Induction of CD14 Expression and Differentiation to Monocytes or
Mature Macrophages in Promyelocytic Cell Lines: New Approach. Adv. Pharm. Bull. 2013,3, 329–332. [CrossRef]
31.
Rodrigues, C.P.; Ferreira, A.C.F.; Pinho, M.P.; de Moraes, C.J.; Bergami-Santos, P.C.; Barbuto, J.A.M. Tolerogenic IDO(+) Dendritic
Cells Are Induced by PD-1-Expressing Mast Cells. Front. Immunol. 2016,7, 9. [CrossRef] [PubMed]
32.
Forget, A.; Gianni-Barrera, R.; Uccelli, A.; Sarem, M.; Kohler, E.; Fogli, B.; Muraro, M.G.; Bichet, S.; Aumann, K.; Banfi, A.; et al.
Mechanically Defined Microenvironment Promotes Stabilization of Microvasculature, Which Correlates with the Enrichment of a
Novel Piezo-1+ Population of Circulating CD11b+ /CD115+ Monocytes. Adv. Mater. 2019,31, e1808050. [CrossRef] [PubMed]
33.
Spinelli, F.R.; Pecani, A.; Conti, F.; Mancini, R.; Alessandri, C.; Valesini, G. Post-translational modifications in rheumatoid arthritis
and atherosclerosis: Focus on citrullination and carbamylation. J. Int. Med. Res. 2016,44, 81–84. [CrossRef] [PubMed]
34.
Sakuraba, K.; Krishnamurthy, A.; Sun, J.; Zheng, X.; Xu, C.; Peng, B.; Engström, M.; Jakobsson, P.-J.; Wermeling, F.; Catrina,
S.; et al. Autoantibodies targeting malondialdehyde-modifications in rheumatoid arthritis regulate osteoclasts via inducing
glycolysis and lipid biosynthesis. J. Autoimmun. 2022,133, 102903. [CrossRef] [PubMed]
35.
Colasanti, T.; Sabatinelli, D.; Mancone, C.; Giorgi, A.; Pecani, A.; Spinelli, F.R.; Giamberardino, A.D.; Navarini, L.; Speziali, M.;
Vomero, M.; et al. Homocysteinylated alpha 1 antitrypsin as an antigenic target of autoantibodies in seronegative rheumatoid
arthritis patients. J. Autoimmun. 2020,113, 102470. [CrossRef]
36.
Costantino, C.M.; Ploegh, H.L.; Hafler, D.A. Cathepsin S regulates class II MHC processing in human CD4+ HLA-DR+ T cells. J.
Immunol. 2009,183, 945–952. [CrossRef]
Diseases 2024,12, 81 18 of 20
37.
Wahyurini, D.; Wibowo, H. Monocytic HLA-DR expression in type 2 diabetes mellitus: Impact on disease susceptibility. World J.
Adv. Res. Rev. 2023,18, 684–689. [CrossRef]
38.
Iwamoto, N.; Kawakami, A. The monocyte-to-osteoclast transition in rheumatoid arthritis: Recent findings. Front. Immunol. 2022,
13, 998554. [CrossRef]
39.
Hwang, S.; Sung, D.K.; Kim, Y.E.; Yang, M.; Ahn, S.Y.; Sung, S.I.; Chang, Y.S. Mesenchymal Stromal Cells Primed by Toll-like
Receptors 3 and 4 Enhanced Anti-Inflammatory Effects against LPS-Induced Macrophages via Extracellular Vesicles. Int. J. Mol.
Sci. 2023,24, 16264. [CrossRef]
40.
Wu, X.-Y.; Li, K.-T.; Yang, H.-X.; Yang, B.; Lu, X.; Zhao, L.-D.; Fei, Y.-Y.; Chen, H.; Wang, L.; Li, J.; et al. Complement C1q
synergizes with PTX3 in promoting NLRP3 inflammasome over-activation and pyroptosis in rheumatoid arthritis. J. Autoimmun.
2020,106, 102336. [CrossRef]
41.
Roelofs, M.F.; Wenink, M.H.; Brentano, F.; Abdollahi-Roodsaz, S.; Oppers-Walgreen, B.; Barrera, P.; van Riel, P.L.C.M.; Joosten,
L.A.B.; Kyburz, D.; van den Berg, W.B.; et al. Type I interferons might form the link between Toll-like receptor (TLR) 3/7 and
TLR4-mediated synovial inflammation in rheumatoid arthritis (RA). Ann. Rheum. Dis. 2009,68, 1486–1493. [CrossRef] [PubMed]
42.
Park, J.-Y.; Park, H.-M.; Kim, S.; Jeon, K.-B.; Lim, C.-M.; Hong, J.T.; Yoon, D.-Y. Human IL-32
θ
A94V mutant attenuates monocyte-
endothelial adhesion by suppressing the expression of ICAM-1 and VCAM-1 via binding to cell surface receptor integrin
α
V
β
3
and αVβ6 in TNF-α-stimulated HUVECs. Front. Immunol. 2023,14, 1160301. [CrossRef] [PubMed]
43.
Luo, Z.; The, E.; Zhang, P.; Zhai, Y.; Yao, Q.; Ao, L.; Zeng, Q.; Fullerton, D.A.; Meng, X. Monocytes augment inflammatory
responses in human aortic valve interstitial cells via
β
2-integrin/ICAM-1-mediated signaling. Inflamm. Res. 2022,71, 681–694.
[CrossRef] [PubMed]
44.
Mantchev, G.T.; Cortesão, C.S.; Rebrovich, M.; Cascalho, M.; Bram, R.J. TACI is required for efficient plasma cell differentiation in
response to T-independent type 2 antigens. J. Immunol. 2007,179, 2282–2288. [CrossRef] [PubMed]
45.
Nagatani, K.; Itoh, K.; Nakajima, K.; Kuroki, H.; Katsuragawa, Y.; Mochizuki, M.; Aotsuka, S.; Mimori, A. Rheumatoid arthritis
fibroblast-like synoviocytes express BCMA and are stimulated by APRIL. Arthritis Rheum. 2007,56, 3554–3563. [CrossRef]
[PubMed]
46.
Xiong, Y.-S.; Cheng, Y.; Lin, Q.-S.; Wu, A.-L.; Yu, J.; Li, C.; Sun, Y.; Zhong, R.-Q.; Wu, L.-J. Increased expression of Siglec-1 on
peripheral blood monocytes and its role in mononuclear cell reactivity to autoantigen in rheumatoid arthritis. Rheumatology 2014,
53, 250–259. [CrossRef]
47.
Meusch, U.; Krasselt, M.; Rossol, M.; Baerwald, C.; Klingner, M.; Wagner, U.
In vitro
response pattern of monocytes after tmTNF
reverse signaling predicts response to anti-TNF therapy in rheumatoid arthritis. J. Transl. Med. 2015,13, 256. [CrossRef] [PubMed]
48.
Huang, J.; Xie, B.; Li, Q.; Xie, X.; Zhu, S.; Wang, M.; Peng, W.; Gu, J. Infliximab reduces CD147, MMP-3, and MMP-9 expression in
peripheral blood monocytes in patients with active rheumatoid arthritis. Eur. J. Pharmacol. 2013,698, 429–434. [CrossRef]
49.
Ebrahimi-Rad, M.; Khatami, S.; Akhbari, H.; Mahmoudzadeh-Niknam, H.; Valadbeigi, S.; Mahmoudi, M.; Jamshidi, A.; Riazi-Rad,
F.; Saghiri, R. Evaluation of autoantibodies against vimentin and
α
-enolase in rheumatoid arthritis patients. Reumatologia 2020,58,
350–356. [CrossRef]
50.
Crilly, A.; Burns, E.; Nickdel, M.B.; Lockhart, J.C.; Perry, M.E.; Ferrell, P.W.; Baxter, D.; Dale, J.; Dunning, L.; Wilson, H.; et al.
PAR(2) expression in peripheral blood monocytes of patients with rheumatoid arthritis. Ann. Rheum. Dis. 2012,71, 1049–1054.
[CrossRef]
51.
Piotrowska, K.; Słuczanowska-Głabowska, S.; Kurzawski, M.; Dziedziejko, V.; Kopytko, P.; Paczkowska, E.; Rogi´nska, D.;
Safranow, K.; Machali ´nski, B.; Pawlik, A. Over-Expression of Allograft Inflammatory Factor-1 (AIF-1) in Patients with Rheumatoid
Arthritis. Biomolecules 2020,10, 1064. [CrossRef] [PubMed]
52.
Hofer, T.P.; van de Loosdrecht, A.A.; Stahl-Hennig, C.; Cassatella, M.A.; Ziegler-Heitbrock, L. 6-Sulfo LacNAc (Slan) as a Marker
for Non-classical Monocytes. Front. Immunol. 2019,10, 2052. [CrossRef] [PubMed]
53.
Tamassia, N.; Bianchetto-Aguilera, F.; Gasperini, S.; Grimaldi, A.; Montaldo, C.; Calzetti, F.; Gardiman, E.; Signoretto, I.;
Castellucci, M.; Barnaba, V.; et al. The slan antigen identifies the prototypical non-classical CD16+-monocytes in human blood.
Front. Immunol. 2023,14, 1287656. [CrossRef] [PubMed]
54.
Hofer, T.P.; Zawada, A.M.; Frankenberger, M.; Skokann, K.; Satzl, A.A.; Gesierich, W.; Schuberth, M.; Levin, J.; Danek, A.; Rotter,
B.; et al. slan-defined subsets of CD16-positive monocytes: Impact of granulomatous inflammation and M-CSF receptor mutation.
Blood 2015,126, 2601–2610. [CrossRef] [PubMed]
55.
Gómez-Bañuelos, E.; Martín-Márquez, B.T.; Martínez-García, E.A.; Figueroa-Sanchez, M.; Nuñez-Atahualpa, L.; Rocha-Muñoz,
A.D.; Sánchez-Hernández, P.E.; Navarro-Hernandez, R.E.; Madrigal-Ruiz, P.M.; Saldaña-Millan, A.A.; et al. Low levels of CD36 in
peripheral blood monocytes in subclinical atherosclerosis in rheumatoid arthritis: A cross-sectional study in a Mexican population.
Biomed. Res. Int. 2014,2014, 736786. [CrossRef] [PubMed]
56.
Min, H.K.; Won, J.-Y.; Kim, B.-M.; Lee, K.-A.; Lee, S.-J.; Lee, S.-H.; Kim, H.-R.; Kim, K.-W. Interleukin (IL)-25 suppresses IL-22-
induced osteoclastogenesis in rheumatoid arthritis via STAT3 and p38 MAPK/I
κ
B
α
pathway. Arthritis Res. Ther. 2020,22, 222.
[CrossRef] [PubMed]
57.
Pan, S.; Wu, S.; Wei, Y.; Liu, J.; Zhou, C.; Chen, T.; Zhu, J.; Tan, W.; Huang, C.; Feng, S.; et al. Exploring the causal relationship
between inflammatory cytokines and inflammatory arthritis: A Mendelian randomization study. Cytokine 2024,173, 156446.
[CrossRef] [PubMed]
Diseases 2024,12, 81 19 of 20
58.
Filali, S.; Noack, M.; Géloën, A.; Pirot, F.; Miossec, P. Effects of pro-inflammatory cytokines and cell interactions on cell area and
cytoskeleton of rheumatoid arthritis synoviocytes and immune cells. Eur. J. Cell Biol. 2023,102, 151303. [CrossRef] [PubMed]
59.
Gloyer, L.; Golumba-Nagy, V.; Meyer, A.; Yan, S.; Schiller, J.; Breuninger, M.; Jochimsen, D.; Kofler, D.M. Adenosine receptor A2a
blockade by caffeine increases IFN-gamma production in Th1 cells from patients with rheumatoid arthritis. Scand. J. Rheumatol.
2022,51, 279–283. [CrossRef]
60.
Park, J.-S.; Kim, N.-R.; Lim, M.-A.; Kim, S.-M.; Hwang, S.-H.; Jung, K.-A.; Choi, J.W.; Park, S.-H.; Cho, M.-L. Deficiency of IL-1
receptor antagonist suppresses IL-10-producing B cells in autoimmune arthritis in an IL-17/Th17-dependent manner. Immunol.
Lett. 2018,199, 44–52. [CrossRef]
61.
Yan, J.; Yao, L.; Tan, Y.; Wang, Y. The protective effects of Phoenixin-20 in tumor necrosis factor
α
(TNF-
α
)-induced cell senescence
of rheumatoid arthritis fibroblast-like synoviocytes (FLS). Aging 2023,15, 14607–14616. [CrossRef] [PubMed]
62.
Shabgah, A.G.; Shariati-Sarabi, Z.; Tavakkol-Afshari, J.; Ghasemi, A.; Ghoryani, M.; Mohammadi, M. A significant decrease of
BAFF, APRIL, and BAFF receptors following mesenchymal stem cell transplantation in patients with refractory rheumatoid
arthritis. Gene 2020,732, 144336. [CrossRef]
63.
Okamato, Y.; Ghosh, T.; Okamoto, T.; Schuyler, R.P.; Seifert, J.; Charry, L.L.; Visser, A.; Feser, M.; Fleischer, C.; Pedrick, C.; et al.
Subjects at-risk for future development of rheumatoid arthritis demonstrate a PAD4-and TLR-dependent enhanced histone H3
citrullination and proinflammatory cytokine production in CD14hi monocytes. J. Autoimmun. 2021,117, 102581. [CrossRef]
[PubMed]
64.
Stangl, H.; Krammetsvogl, A.; Lesiak, M.; Wolff, C.; Straub, R.H. MHC/class-II-positive cells inhibit corticosterone of adrenal
gland cells in experimental arthritis: A role for IL-1
β
, IL-18, and the inflammasome. Sci. Rep. 2020,10, 17071. [CrossRef] [PubMed]
65.
Murphy, E.P.; Crean, D. Molecular Interactions between NR4A Orphan Nuclear Receptors and NF-
κ
B Are Required for Appropri-
ate Inflammatory Responses and Immune Cell Homeostasis. Biomolecules 2015,5, 1302–1318. [CrossRef] [PubMed]
66.
Yamamoto, T.; Endo, Y.; Onodera, A.; Hirahara, K.; Asou, H.K.; Nakajima, T.; Kanno, T.; Ouchi, Y.; Uematsu, S.; Nishimasu, H.;
et al. DUSP10 constrains innate IL-33-mediated cytokine production in ST2hi memory-type pathogenic Th2 cells. Nat. Commun.
2018,9, 4231. [CrossRef] [PubMed]
67.
Chung, S.; Ranjan, R.; Lee, Y.G.; Park, G.Y.; Karpurapu, M.; Deng, J.; Xiao, L.; Kim, J.Y.; Unterman, T.G.; Christman, J.W. Distinct
role of FoxO1 in M-CSF- and GM-CSF-differentiated macrophages contributes LPS-mediated IL-10: Implication in hyperglycemia.
J. Leukoc. Biol. 2015,97, 327–339. [CrossRef] [PubMed]
68.
Elbjeirami, W.M.; Donnachie, E.M.; Burns, A.R.; Smith, C.W. Endothelium-derived GM-CSF influences expression of oncostatin
M. Am. J. Physiol. Cell Physiol. 2011,301, C947–C953. [CrossRef]
69.
Haringman, J.J.; Gerlag, D.M.; Smeets, T.J.M.; Baeten, D.; van den Bosch, F.; Bresnihan, B.; Breedveld, F.C.; Dinant, H.J.; Legay, F.;
Gram, H.; et al. A randomized controlled trial with an anti-CCL2 (anti-monocyte chemotactic protein 1) monoclonal antibody in
patients with RA. Arthritis Rheum. 2006,54, 2387–2392. [CrossRef]
70.
Tsai, C.-H.; Liu, S.-C.; Wang, Y.-H.; Su, C.-M.; Huang, C.-C.; Hsu, C.-J.; Tang, C.-H. Osteopontin inhibition of miR-129-3p enhances
IL-17 expression and monocyte migration in rheumatoid arthritis. Biochim. Biophys. Acta Gen. Subj. 2017,1861, 15–22. [CrossRef]
71.
Wu, Y.-J.; Zhang, S.-S.; Yin, Q.; Lei, M.; Wang, Q.-H.; Chen, W.-G.; Luo, T.-T.; Zhou, P.; Ji, C.-L.
α
-Mangostin Inhibited M1
Polarization of Macrophages/Monocytes in Antigen-Induced Arthritis Mice by Up-Regulating Silent Information Regulator 1
and Peroxisome Proliferators-Activated Receptor
γ
Simultaneously. Drug Des. Devel Ther. 2023,17, 563–577. [CrossRef] [PubMed]
72.
Vasilevko, V.; Ghochikyan, A.; Holterman, M.J.; Agadjanyan, M.G. CD80 (B7-1) and CD86 (B7-2) are functionally equivalent in
the initiation and maintenance of CD4+ T-cell proliferation after activation with suboptimal doses of PHA. DNA Cell Biol. 2002,
21, 137–149. [CrossRef] [PubMed]
73.
Moghaddami, M.; Cleland, L.G.; Radisic, G.; Mayrhofer, G. Recruitment of dendritic cells and macrophages during T cell-mediated
synovial inflammation. Arthritis Res. Ther. 2007,9, R120. [CrossRef] [PubMed]
74. König, M.; Rharbaoui, F.; Aigner, S.; Dälken, B.; Schüttrumpf, J. Tregalizumab—A Monoclonal Antibody to Target Regulatory T
Cells. Front. Immunol. 2016,7, 11. [CrossRef] [PubMed]
75. Bossi, G. Mutant p53 and sIL-1Ra. Aging 2015,7, 742–743. [CrossRef] [PubMed]
76.
Wang, M.; Zhao, H.; Zhao, H.; Huo, C.; Yuan, Y.; Zhu, Y. Moxibustion-mediated alleviation of synovitis in rats with rheumatoid
arthritis through the regulation of NLRP3 inflammasome by modulating neutrophil extracellular traps. Heliyon 2023,10, e23633.
[CrossRef] [PubMed]
77. Jiang, F.-Y.; Zhang, Y.-Z.; Tai, Y.-H.; Chou, C.-Y.; Hsieh, Y.-C.; Chang, Y.-C.; Huang, H.-C.; Li, Z.-Q.; Hsieh, Y.-C.; Chen, I.-J.; et al.
A lesion-selective albumin-CTLA4Ig as a safe and effective treatment for collagen-induced arthritis. Inflamm. Regen. 2023,43, 13.
[CrossRef] [PubMed]
78.
Tian, Y.; Ming, J. Melatonin inhibits osteoclastogenesis via RANKL/OPG suppression mediated by Rev-Erb
α
in osteoblasts. J.
Cell Mol. Med. 2022,26, 4032–4047. [CrossRef] [PubMed]
79.
Isozaki, T.; Nishimi, S.; Nishimi, A.; Saito, M.; Miwa, Y.; Toyoshima, Y.; Inagaki, K.; Kasama, T. A disintegrin and metalloproteinase
(ADAM)-10 as a predictive factor for tocilizumab effectiveness in rheumatoid arthritis. Mod. Rheumatol. 2017,27, 782–786.
[CrossRef]
80.
Li, S.; Wang, Y.; Wu, M.; Younis, M.H.; Olson, A.P.; Barnhart, T.E.; Engle, J.W.; Zhu, X.; Cai, W. Spleen-Targeted Glabridin-Loaded
Nanoparticles Regulate Polarization of Monocyte/Macrophage (Mo/M
φ
) for the Treatment of Cerebral Ischemia-Reperfusion
Injury. Adv. Mater. 2022,34, e2204976. [CrossRef]
Diseases 2024,12, 81 20 of 20
81.
Wang, D.-D.; He, C.-Y.; Wu, Y.-J.; Xu, L.; Shi, C.; Olatunji, O.J.; Zuo, J.; Ji, C.-L. AMPK/SIRT1 Deficiency Drives Adjuvant-Induced
Arthritis in Rats by Promoting Glycolysis-Mediated Monocytes Inflammatory Polarization. J. Inflamm. Res. 2022,15, 4663–4675.
[CrossRef] [PubMed]
82.
Pang, X.; Yin, P.; Han, J.; Wang, Z.; Zheng, F.; Chen, X. cPLA2a correlates with metastasis and poor prognosis of osteosarcoma by
facilitating epithelial-mesenchymal transition. Pathol. Res. Pract. 2019,215, 152398. [CrossRef] [PubMed]
83.
Lee, A.Y.S.; Eri, R.; Lyons, A.B.; Grimm, M.C.; Korner, H. CC chemokine ligand 20 and its cognate receptor CCR6 in mucosal T
cell immunology and inflammatory bowel disease: Odd couple or axis of evil? Front. Immunol. 2013,4, 194. [CrossRef] [PubMed]
84.
Riella, L.V.; Dada, S.; Chabtini, L.; Smith, B.; Huang, L.; Dakle, P.; Mfarrej, B.; D‘Addio, F.; Adams, L.-T.; Kochupurakkal, N.; et al.
B7h (ICOS-L) maintains tolerance at the fetomaternal interface. Am. J. Pathol. 2013,182, 2204–2213. [CrossRef] [PubMed]
85.
Blagov, A.V.; Grechko, A.V.; Nikiforov, N.G.; Zhuravlev, A.D.; Sadykhov, N.K.; Orekhov, A.N. Effects of Metabolic Disorders in
Immune Cells and Synoviocytes on the Development of Rheumatoid Arthritis. Metabolites 2022,12, 634. [CrossRef] [PubMed]
86.
Amoruso, A.; Sola, D.; Rossi, L.; Obeng, J.A.; Fresu, L.G.; Sainaghi, P.P.; Pirisi, M.; Brunelleschi, S. Relation among anti-rheumatic
drug therapy, CD14(+)CD16(+) blood monocytes and disease activity markers (DAS28 and US7 scores) in rheumatoid arthritis: A
pilot study. Pharmacol. Res. 2016,107, 308–314. [CrossRef] [PubMed]
87.
Batko, B.; Schramm-Luc, A.; Skiba, D.S.; Mikolajczyk, T.P.; Siedlinski, M. TNF-
α
Inhibitors Decrease Classical CD14hiCD16-
Monocyte Subsets in Highly Active, Conventional Treatment Refractory Rheumatoid Arthritis and Ankylosing Spondylitis. Int. J.
Mol. Sci. 2019,20, 291. [CrossRef]
88.
Du, Z.; Cong, W.; Tang, K.; Zheng, Q.; Song, Z.; Chen, Y.; Yang, S.; Zhang, C.; Ye, T. Electroacupuncture stimulating Zusanli
(ST36), Sanyinjiao (SP6) in mice with collagen-induced arthritis leads to adenosine A2A receptor-mediated alteration of p38
α
mitogen-activated protein kinase signaling and inhibition of osteoclastogenesis. J. Tradit. Chin. Med. 2023,43, 1103–1109.
[CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... При этом активация резидентных макрофагов сопровождается синтезом ряда провоспалительных цитокинов и хемокинов. Последние -CXCL5, CXCL8, CXCL9, CXCL10 и CXCL13 -являются мощными хемоаттрактантами, вызывающими направленное перемещение в область формирующегося воспаления моноцитов и миелоидных клеток (нейтрофилов, базофилов, эозинофилов) периферической крови [18,[35][36][37]. ...
... Моноциты и клетки миелоидного ряда под воздействием провоспалительных цитокинов осуществляют продукцию основных провоспалительных медиаторов -ПГ Е2, ЛТ В4, ФРН, фосфатидилинозитола (ФИТ), 5-гидроксиэйкозотетраеновой кислоты (5(S)-HETE, 5-hydroxyeicosatetraenoic acid) и др. [18,[35][36][37]. ...
Article
Chronic pain is the main manifestation of rheumatoid arthritis (RA), determining the severity of suffering and functional impairment. Although pain in RA is primarily associated with autoimmune inflammation, it can persist against the background of low activity and even remission of the disease. This makes it necessary to search for the causes and peculiarities of the development of chronic pain in RA. It seems that the classification of pain types in RA can help in personalizing approaches to its medication control. In this regard, the evaluation of the relationship between pain and the cellular composition (pathotype) of synovitis in RA is of great interest. Three main pathotypes are known: lymphoid (with predominance of T and B lymphocytes, plasmocytes), myeloid or diffuse-myeloid (with predominance of macrophages, monocytes, granulocytes) and pauci-immune (mainly consisting of fibroblast-like synoviocytes (FLS)). The lymphoid pathotype is characterised by high positivity for rheumatoid factor and anti-citrullinated protein antibodies, severe RA activity and intense pain, including that associated with polyneuropathy and dysfunctional disorders; the myeloid pathotype is characterized by less severe activity and local nociceptive pain; the pauci-immune pathotype is characterized by moderately severe pain and peripheral hyperalgesia against a background of moderate/low disease activity. The last pathotype can determine chronic pain in seronegative RA and at late stages of the disease, in which marked structural changes are noted. Currently, there is no clear view on drug approaches for the different pathotypes of synovitis in RA. There is limited evidence for the use of CD20 inhibitors (rituximab) and interleukin (IL) 6 inhibitors in the lymphoid pathotype, and IL-6 and tumour necrosis factor α inhibitors in the myeloid pathotype. Currently, active development of drugs to target FLS is underway. The data of some studies indicate higher efficacy of IL-6 inhibitors in pauci-immune pathotype.
... While acute inflammation is crucial for healing and recovery, chronic inflammation can lead to a variety of diseases, including cancer, cardiovascular disorders, and autoimmune conditions [1][2][3]. The recent Special Issue, 'Inflammation: The cause of all diseases 2.0' [4], along with the participating journals-Cells, Diseases, Healthcare, International Journal of Molecular Sciences, and Vaccines-sheds light on the complex interplay between inflammation and disease mechanisms, providing valuable insights into potential therapeutic strategies targeting inflammatory pathways [5][6][7][8][9][10][11][12][13][14][15][16]. Inflammation is characterized by a series of physiological responses involving immune cells, blood vessels, and molecular and cellular mediators. ...
... A review was published in this Special Issue describing the key players in rheumatoid arthritis pathogenesis. Immune cells, fibroblast-like synoviocytes, macrophages, monocytes, B cells, CD4+ T cells, Th1, Th17 cells, and pro-inflammatory mediators, CCR2, CX3CR1, RANKL, IL-1β, IL-6, and TNF-α, were described in the context of inflammatory responses associated with disease [12]. Another review described cellular senescence and mitochondrial dysfunction in CVD and provided advancements in therapies aimed at targeting mitochondrial dysfunction, such as energy starvation, oxidative stress, and mitophagy [8]. ...
Article
Full-text available
Inflammation is an essential biological process that serves as the body’s first line of defence against harmful stimuli, including pathogens, damaged cells, and irritants [...]
... Then, as an autoimmune disease, the pathogenesis of RA is relatively complex. Despite of adaptive immunity, innate immunity also participates in the progression of RA [85,86]. Since proinflammatory cytokines have been reported to activate various innate immune cells, including natural killer cells, mast cells, dendritic cells and so on [87][88][89]. ...
Article
Full-text available
Overproduction of reactive oxygen species (ROS), elevated synovial inflammation, synovial hyperplasia and fibrosis are the main characteristic of microenvironment in rheumatoid arthritis (RA). Macrophages and fibroblast-like synoviocytes (FLSs) play crucial roles in the progression of RA. Hence, synergistic combination of ROS scavenging, macrophage polarization from pro-inflammatory M1 phenotype towards M2 anti-inflammatory phenotype, and restoring homeostasis of FLSs will provide a promising therapeutic strategy for RA. In this study, we successfully synthesized metformin-derived carbon dots (MCDs), and investigated the antirheumatic effect in vivo and in vitro. Designed MCDs could target inflamed cells and accumulate at the inflammatory joints of collagen-induced arthritis (CIA) rats. In vivo therapeutic investigation suggested that MCDs reduced synovial inflammation and hyperplasia, ultimately prevented cartilage destruction, bone erosion, and synovial fibrosis in CIA rats. In addition, MCDs eliminated the cellular ROS in M1 phenotype macrophages in RA microenvironment through the enzyme-like catalytic activity as well as inhibiting NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome signaling pathway, effectively polarizing them into the M2 phenotype to realize the anti-inflammatory effect. Furthermore, MCDs could inhibit the proliferation, migration, and fibrosis of inflamed FLSs. Mechanistically, MCDs restored the homeostasis of FLSs while reducing the level of synovial inflammation by blocking IL-6/gp130 signaling pathway. Combined with preferable biocompatibility, MCDs offer a prospective treatment approach for RA.
... Importantly, CD14 and CD16 are surface markers that play a crucial role in monocyte and macrophage function in RA. Monocytes are categorized based on these markers, and they are differentially involved in the inflammatory processes observed in RA [42,43]. ...
Article
Full-text available
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by symmetrical joint inflammation, cartilage degradation, and bone erosion. This review explores the multifaceted aspects of RA pathogenesis, focusing on the dynamic interplay between innate and adaptive immune responses, genetic predisposition, and environmental triggers. The development of RA involves genetic susceptibility and trigger events such as infections, trauma, smoking, obesity, and microbiome alterations, fostering autoimmune reactions and tissue/organ destruction. The innate immune response, including toll-like receptor activation and synovial fibroblasts’ roles, contributes to the acceleration of inflammatory processes in joint tissues. Monocytes and macrophages organize and sustain chronic joint inflammation, leading to tissue damage and bone resorption, while highlighting the significance of CD14 and CD16 subsets in RA pathogenesis. In the adaptive immune response, aberrant activation and proliferation of CD4+ T cells and the role of regulatory T cells in maintaining immune tolerance are discussed. Target cytokines like TNF-α, IL-6, IL-1, IL-17, and BAFF, as well as chemokines such as CCL2, CXCL10, CCL5, and CXCL12, have emerged as critical components in managing chronic inflammation and joint damage in RA. This comprehensive overview provides insights into the pathophysiology of RA and potential therapeutic avenues, emphasizing the importance of understanding these complex immunological and genetic mechanisms for developing more effective treatment strategies.
Article
Objective The objective of study is to clarify the relationship between monocyte‐to‐lymphocyte ratio (MLR) and lumbar spine bone mineral density (BMD) in type 2 diabetes mellitus (T2DM), and analyse the mediating role of fat distribution, providing a new indicator for the early diagnosis of osteoporosis in T2DM. Methods A total of 430 T2DM patients over 50 years were included. Subjects were divided into T1, T2 and T3 three groups based on MLR. The adjusted regression models, subgroup analysis, generalised additive model (GAM), smoothed curve fitting, and mediated effects analysis methods were used. Results The results indicated a significant negative correlation between MLR and lumbar spine BMD in T2DM ( β = −0.701; 95%CI: −0.112, −0.290). Subgroup analysis, smoothed curve fitting and threshold effect analysis showed a nonlinear relationship between MLR and BMD, with an optimal inflection point at an MLR value of 0.1266. Additionally, the ratio of visceral to subcutaneous fat area (VSR) was negatively correlated with lumbar spine BMD ( β = −0.775; 95%CI: −1.007, −0.504). Mediation effect analysis of VSR between MLR and lumbar spine BMD showed that the total effect estimate was −0.0717, the direct effect estimate was −0.0524, the mediation effect estimate was −0.0193, with the mediated proportion accounting for 26.88% of the total effect. Conclusion MLR is significantly associated with lumbar spine BMD in T2DM. Higher MLR levels correlated with reduced BMD. VSR plays a significant mediating role in this relationship, highlighting MLR's potential utility in osteoporosis diagnosis and importance of fat management for bone health.
Article
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by complex immune interactions. Elucidating the causal relationships between blood cell perturbations, immune cell subsets, and RA can provide valuable insights into its pathogenesis. This study employed bidirectional two-sample Mendelian Randomization (MR) to explore the causal effects of blood cell perturbations on RA risk, with a focus on immune cell mediation. Genetic data from large-scale Genome-Wide Association Studies (GWAS) were utilized to select instrumental variables (IVs) for exposure, mediator, and outcome. Inverse Variance Weighted (IVW) analysis was applied, supplemented by sensitivity tests. Mediation analysis was conducted to assess the indirect effects mediated by immune cells. Significant causal associations were identified between perturbations in reticulocytes, monocytes, and lymphocytes and specific immune cell subsets, including CD3 + CD39 + regulatory T cells (Tregs) and CD45RA + terminally differentiated CD8 + T cells (CD45RA + TD CD8 + cells). Erythropoiesis perturbation was associated with a reduced RA risk, while perturbations in monocytes and lymphocytes were found to facilitate RA progression through immune-mediated mechanisms. This study underscores the pivotal role of immune cell subsets in mediating the effects of blood cell perturbations on RA development. These findings suggest that targeting immune cell-mediated pathways, particularly those involving Tregs and CD8 + T cells, can provide new therapeutic strategies for RA management. Key Points • Causal Relationships: Mendelian randomization (MR) analysis identified significant causal relationships between specific blood cell disturbances (e.g., reticulocytes, monocytes, and lymphocytes) and rheumatoid arthritis (RA). • Role of Immune Cells: CD3 + CD39 + regulatory T cells (Tregs) and CD45RA + Terminally Differentiated CD8 + T cells (CD45RA + TD CD8 + cells) mediate the association between blood cell disturbances and RA. • Protective Role of Reticulocytes: Reticulocyte disturbances under potassium chloride (KCl) conditions are negatively associated with RA, potentially protecting joints from inflammatory damage by reducing oxidative stress. • Protective Role of Non-Classical Monocytes: Baseline disturbances in monocyte median side scatter are negatively associated with RA, suggesting non-classical monocytes may reduce RA-related inflammation. • Positive Association of Lymphocyte Disturbances with RA: Lymphocyte side scatter standard deviation under colchicine disturbances shows a significant positive association with RA, indicating abnormal T cell activation may exacerbate RA progression.AQ
Article
Full-text available
BACKGROUND Depression is a significant psychiatric disorder with particularly high prevalence among adolescents. This mental health condition can have severe consequences, including academic failure, social withdrawal, and suicidal behavior. Given the increasing rate of depression in this age group, understanding the underlying biological mechanisms is essential for early detection and intervention. Recent studies have suggested that immune markers play a role in the pathophysiology of depression, prompting further investigation of their potential association with depressive symptoms in adolescents. AIM To investigate the relationship between immune markers (monocytes, lymphocytes, and direct bilirubin) and the incidence and severity of depression among adolescents. METHODS This cross-sectional study recruited 145 adolescent patients with depression [male (M)/female (F) = 38/107] from Jiangbin Hospital in Guangxi, Zhuang and 163 healthy controls (M/F = 77/86) from routine health check-ups. Blood samples were collected after an overnight fast. Depression severity was measured using the Zung Self-Rating Depression Scale. The inclusion criteria were age 12-24 years, diagnosis of depressive disorder (ICD-10), and no recent antidepressant use. The exclusion criteria included psychiatric comorbidities and serious somatic diseases. Key statistical methods included group comparisons and correlation analyses. RESULTS There was a higher prevalence of females in the depression group (P < 0.001). Significant age differences were observed between the groups (Z = 9.43, P < 0.001). The depression group had higher monocyte (Z = 3.43, P < 0.001) and lymphocyte (t = 2.29, P < 0.05) counts, and higher serum direct bilirubin levels (Z = 4.72, P < 0.001). Monocyte count varied significantly according to depression severity, with lower counts in the mild group (Z = -2.90, P < 0.05). A negative correlation between age and lymphocyte counts was observed (ρ = -0.22, P < 0.01). Logistic regression analysis showed that serum direct bilirubin levels significantly predicted depression. CONCLUSION The potential role of elevated levels of immune markers in the early detection of depression in adolescents has been highlighted. Therefore, it is necessary to explore further the relationships between these immune markers and depression.
Article
Full-text available
Purpose This study investigates the potential mechanism of moxibustion in the treatment of rheumatoid arthritis (RA) by regulating the neutrophil extracellular trap (NET)/NOD-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome axis with the use of a rat model with adjuvant arthritis (AA). Methods Four groups, including normal control (NC), AA, moxibustion (MOX), and chlor-amidine (Cl-amidine) were created from 24 Wistar male rats (6 rats/group). After the intervention and treatment respectively, the joint swelling degree (JSD) and arthritis index (AI) were compared. The pathological changes of synovium were observed with hematoxylin and eosin staining and transmission electron microscopy. The formation of NETs in synovial tissues was detected with immunofluorescence staining. The protein expression of myeloperoxidase (MPO), neutrophil elastase (NE), citrullinated histone (Cit-H3), acyl arginine deiminase 4 (PAD-4), and NLRP3 was measured in the synovium of rat ankle joints with western blotting, and the levels of anti-cyclic citrullinated peptide antibody (CCP-Ab) and interleukin (IL)-1β were examined in rat serum with ELISA. Results AA modeling markedly increased JSD, AI, synovial protein expression of MPO, NE, Cit-H3, PAD-4, and NLRP3, and serum levels of CCP-Ab and IL-1β in rats (P < 0.01). JSD and AI, as well as the levels of MPO, NE, Cit-H3, PAD-4, NLRP3, CCP-Ab, and IL-1β, were significantly lowered in AA rats by MOX and Cl-amidine (P < 0.01). In addition, AA modeling caused severe pathological injury in the synovium of rats, which was annulled by MOX and Cl-amidine. The formation of NETs in synovium was substantially promoted in rats by AA modeling and was significantly reduced in AA rats after the treatment. Conclusion Moxibustion can markedly alleviate synovitis and repress inflammatory factor release in AA rats, which may be achieved by diminished synthesis of NETs or their constituents and the blocked formation of NLRP3 inflammasome.
Article
Full-text available
Rheumatoid arthritis (RA) is an age-related joint destruction disease that markedly impacts the normal life of patients. Currently, the clinical treatment strategies are far from satisfactory with severe side effects. Cellular senescence of fibroblast-like synoviocytes (FLS) has been reported to be involved in the pathological process of arthritis, which may provide an important research direction for RA treatment. Phoenixin-20 (PNX-20) is a peptide targeting G-protein-coupled receptor 173 (GPR173) with promising anti-inflammatory properties. Our study will probe into the function of PNX-20 on tumor necrosis factor α (TNF-α)- induced rheumatoid arthritis (RA) FLS cell senescence to provide a theoretical basis for treating RA with PNX-20. RA-FLSs were handled with 10 ng/mL TNF-α, followed by introducing Phoenixin-20 (10, 20 nM) or not for 7 days. Enhanced release of inflammatory cytokines, increased proportion of senescence-associated β-galactosidase (SA-β-gal) positive cells, and declined telomerase activity were all observed in TNF-α-treated RA-FLSs, accompanied by a noticeable decline in the p21 and p53 level, which were notably reversed by 10 and 20 nM PNX-20. Furthermore, the increased signal transducer and activator of transcription 6 (STAT6) level observed in TNF-α-treated RA-FLSs were signally repressed by PNX-20. Moreover, the impact of PNX-20 on TNF-α-induced cellular senescence in RA-FLSs was abrogated by the overexpression of STAT6. Collectively, PNX-20 protected the TNF-α-induced cell senescence in RA-FLSs by downregulating STAT6. Based on these findings, we speculate that PNX-20 might be a promising agent for the treatment of RA.
Article
Full-text available
Although it has been suggested that toll-like receptor (TLR) 3 and TLR4 activation alters mesenchymal stromal cells (MSCs)’ immunoregulatory function as anti- or pro-inflammatory phenotypes, we have previously confirmed that TLR4-primed hUCB-MSCs alleviate lung inflammation and tissue injury in an E. coli-induced acute lung injury (ALI) mouse model. Therefore, we hypothesized that strong stimulation of TLR3 or TLR4 prompts hUCB-MSCs to exhibit an anti-inflammatory phenotype mediated by extracellular vesicles (EVs). In this study, we compared the anti-inflammatory effect of TLR3-primed and TLR4-primed hUCB-MSCs against an LPS-induced ALI in vitro model by treating MSCs, MSC-derived conditioned medium (CM), and MSC-derived extracellular vesicles (EVs). LPS-induced rat primary alveolar macrophage and RAW 264.7 cells were treated with naïve, TLR3-, and TLR4-primed MSCs and their derived CM and EVs. Flow cytometry and ELISA were used to evaluate M1-M2 polarization of macrophages and pro-inflammatory cytokine levels, respectively. LPS-stimulated macrophages showed significantly increased pro-inflammatory cytokines compared to those of the normal control, and the percentage of M2 macrophage phenotype was predominantly low. In reducing the inflammatory cytokines and enhancing M2 polarization, TLR3- and TLR4-primed MSCs were significantly more effective than the naïve MSCs, and this finding was also observed with the treatment of MSC-derived CMs and EVs. No significant difference between the efficacy of TLR3- and TLR-primed MSCs was observed. Strong stimulation of TLR3- and TLR4-stimulated hUCB-MSCs significantly reduced pro-inflammatory cytokine secretion from LPS-induced macrophages and significantly enhanced the M2 polarization of macrophages. We further confirmed that TLR-primed MSC-derived EVs can exert anti-inflammatory and immunosuppressive effects alone comparable to MSC treatment. We hereby suggest that in the LPS-induced macrophage in vitro model, EVs derived from both TLR3 and TLR4-primed MSCs can be a therapeutic candidate by promoting the M2 phenotype.
Article
Full-text available
Introduction Peripheral monocytes in humans are conventionally divided into classical (CL, CD14⁺⁺CD16⁻), intermediate (INT, CD14⁺⁺CD16⁺) and non-classical (NC, CD14dim/−CD16⁺⁺) cells, based on their expression levels of CD14 and CD16. A major fraction of the NC-monocytes has been shown to express the 6-sulfo LacNAc (slan) antigen, but whether these slan⁺/NC-monocytes represent the prototypical non-classical monocytes or whether they are simply a sub-fraction with identical features as the remainder of NC monocytes is still unclear. Methods We analyzed transcriptome (by bulk and single cell RNA-seq), proteome, cell surface markers and production of discrete cytokines by peripheral slan⁺/NC- and slan⁻/NC-monocytes, in comparison to total NC-, CL- and INT- monocytes. Results By bulk RNA-seq and proteomic analysis, we found that slan⁺/NC-monocytes express higher levels of genes and proteins specific of NC-monocytes than slan⁻/NC-monocytes do. Unsupervised clustering of scRNA-seq data generated one cluster of NC- and one of INT-monocytes, where all slan⁺/NC-monocytes were allocated to the NC-monocyte cluster, while slan⁻/NC-monocytes were found, in part (13.4%), within the INT-monocyte cluster. In addition, total NC- and slan⁻/NC-monocytes, but not slan⁺/NC-monocytes, were found by both bulk RNA-seq and scRNA-seq to contain a small percentage of natural killer cells. Conclusion In addition to comparatively characterize total NC-, slan⁻/NC- and slan⁺/NC-monocyte transcriptomes and proteomes, our data prove that slan⁺/NC-, but not slan⁻/NC-, monocytes are more representative of prototypical NC-monocytes.
Article
Full-text available
Background Technetium [99Tc] methylene diphosphonate injection (99Tc-MDP) is widely used for the treatment of rheumatoid arthritis (RA), but there is still insufficient evidence for its application. Through the utilization of meta-analysis and systematic reviews, this study aimed to evaluate the effectiveness and safety of 99 TC-MDP in combination with conventional synthetic disease-modifying anti-rheumatic drugs (csDMARDs) for RA. Methods This study was registered on PROSPERO in advance (CRD42021220780). A systematic search was conducted in PubMed, Embase, the Cochrane Library, and multiple international public databases from their inception to April 2023 to identify clinical randomized controlled trials exploring the use of 99Tc-MDP combined with csDMARDs in the treatment of RA. Each outcome was subjected to meta-analysis, and the quality of evidence was assessed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. The American College of Rheumatology's 50 %/70 % response criteria scores (ACR50/70) scores were utilized as the primary effectiveness outcomes, and risks were measured by assessing the rates of AEs. Moreover, secondary efficacy outcomes were evaluated, including the Disease Activity Score 28 (DAS28) and bone mineral density (BMD) as joint function indicators and the erythrocyte sedimentation rate (ESR) and interleukin-17 (IL-17) as inflammatory indicators. Results In this meta-analysis, a total of 34 studies (2296 patients) were included out of 1149 retrieved studies. The summarized results showed that the treatment group treated with the combination of 99Tc-MDP and csDMARDs had significantly higher ACR50 (RR = 1.32, 95 % CI: 1.13–1.55, P = 0.0004) and ACR70 (RR = 1.40, 95 % CI: 1.07–1.82, P = 0.01) scores than the control group receiving csDMARDs alone. In addition, the overall incidence of AEs was lower with the combination of 99Tc-MDP and csDMARDs than with csDMARDs alone (RR = 0.75, 95 % CI: 0.60–0.93, P = 0.009), but the possibility of phlebitis was higher in the treatment group (RR = 4.15, 95 % CI: 1.04–16.50, P = 0.04). In addition, the combination of 99Tc-MDP and csDMARDs had advantages over csDMARDs alone in improving DAS28 (WMD = 1.56, 95 % CI: 0.86–2.25, P < 0.0001), BMD (SMD = 1.12, 95 % CI 0.46–1.78, P = 0.0008), ESR (SMD = 0.71, 95 % CI 0.45–0.97, P < 0.00001), and IL-17 (WMD = 5.82, 95 % CI 3.86–7.77, P < 0.00001). However, the above results might have been influenced by the 99Tc-MDP dosage, csDMARD category, and treatment duration. Combining methotrexate and leflunomide, administering continuous treatment for 24 weeks, or using 3 sets of 99Tc-MDP doses (16.5 mg) may be the optimal 99Tc-MDP treatment plan for RA. Conclusion Compared with csDMARD therapy alone, the combination therapy with 99Tc-MDP is more effective for RA patients and is associated with a lower overall incidence of adverse events, although the possibility of phlebitis was higher. However, due to the inherent limitations of the included RCTs, high-quality clinical trials are still needed to further assess the effectiveness and safety of this combination therapy.
Article
Full-text available
Monocytes are innate immune cells that act as antigen-presenting cells. HLA-DR is a cell surface protein that plays a crucial role in the immune system by presenting antigens to CD4 T Cells (T helper). Based on surface expression, monocytes can differentiate into three subsets; classical monocytes (CD14+CD16-), intermediates monocytes (CD14+ CD16+), and non-classical monocytes (CD14dim CD16+). Among these subsets, classical monocytes have the lowest, and intermediate monocytes have the highest expression of HLA-DR. Monocytes are very sensitive to environmental changes, like hyperglycemia in Type 2 Diabetes Mellitus (T2DM). Metabolic changes in T2DM conditions trigger changes in immune cells including monocytes. Chronic low-grade inflammation in T2DM decreased HLA-DR expression in all monocyte subsets. Monocytes that do not express or lose HLA-DR expression are known as CD14+HLA-DR-/low monocyte and is immunosuppressive. This increase in monocytes in T2DM conditions is related to the ability of monocytes as antigen-presenting cells. Decreased HLA-DR expression increased the risk of severity and susceptibility to several diseases such as COVID-19 infection, TB, cancer, or sepsis. The alterations in HLA-DR expression are one of the factors that make T2DM a comorbid disease. This literature review aims to explore monocytic HLA-DR in T2DM conditions hence increasing the risk of severity and susceptibility to disease. This literature is expected to be the basis for research on the potential of HLA-DR as a prevention or treatment for T2DM patients.
Article
Full-text available
Interleukin-32 (IL-32), first reported in 2005, and its isoforms have been the subject of numerous studies investigating their functions in virus infection, cancer, and inflammation. IL-32θ, one of the IL-32 isoforms, has been shown to modulate cancer development and inflammatory responses. A recent study identified an IL-32θ mutant with a cytosine to thymine replacement at position 281 in breast cancer tissues. It means that alanine was also replaced to valine at position 94 in amino acid sequence (A94V). In this study, we investigated the cell surface receptors of IL-32θA94V and evaluated their effect on human umbilical vein endothelial cells (HUVECs). Recombinant human IL-32θA94V was expressed, isolated, and purified using Ni-NTA and IL-32 mAb (KU32-52)-coupled agarose columns. We observed that IL-32θA94V could bind to the integrins αVβ3 and αVβ6, suggesting that integrins act as cell surface receptors for IL-32θA94V. IL-32θA94V significantly attenuated monocyte-endothelial adhesion by inhibiting the expression of Intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) in tumor necrosis factor (TNF)-α-stimulated HUVECs. IL-32θA94V also reduced the TNF-α-induced phosphorylation of protein kinase B (AKT) and c-jun N-terminal kinases (JNK) by inhibiting phosphorylation of focal adhesion kinase (FAK). Additionally, IL-32θA94V regulated the nuclear translocation of nuclear factor kappa B (NF-κB) and activator protein 1 (AP-1), which are involved in ICAM-1 and VCAM-1 expression. Monocyte-endothelial adhesion mediated by ICAM-1 and VCAM-1 is an important early step in atherosclerosis, which is a major cause of cardiovascular disease. Our findings suggest that IL-32θA94V binds to the cell surface receptors, integrins αVβ3 and αVβ6, and attenuates monocyte-endothelial adhesion by suppressing the expression of ICAM-1 and VCAM-1 in TNF-α-stimulated HUVECs. These results demonstrate that IL-32θA94V can act as an anti-inflammatory cytokine in a chronic inflammatory disease such as atherosclerosis.
Article
Rheumatoid synovitis is infiltrated by immune cells that interact with synoviocytes, leading to the pannus formation. Inflammation or cell interaction effects are mainly evaluated with cytokine production, cell proliferation or migration. Few studies interest on cell morphology. Here, the purpose was to deepen some morphological changes of synoviocytes or immune cells under inflammatory conditions. Inflammatory cytokines, IL-17 and TNF that are largely involved in RA pathogenesis, induced a change in synoviocyte morphology, inducing a retracted cell with higher number of pseudopodia. Several morphological parameters decreased in inflammatory conditions: cell confluence, area and motility speed. The same impact on cell morphology was observed in co-culture of synoviocytes and immune cells in inflammatory/non-inflammatory conditions or with cell activation (miming the in vivo situation), affecting both cell types: synoviocytes were retracted and inversely immune cells proliferated, indicating that cell activation induced a morphological change of cells. In contrast, with RA but not control synoviocytes, cell interactions were not sufficient to affect PBMC and synoviocyte morphology. The morphological effect came only from the inflammatory environment. These findings reveal that the inflammatory environment or cell interactions induced massive changes in control synoviocytes, with cell retraction and increase of pseudopodia number, leading to better interactions with other cells. Except in the case of RA, the inflammatory environment was absolutely required for such changes.