Next Article in Journal
MAPKKKs in Plants: Multidimensional Regulators of Plant Growth and Stress Responses
Next Article in Special Issue
Methylphenidate Promotes Premature Growth Plate Closure: In Vitro Evidence
Previous Article in Journal
Optimization of Primary Human Bronchial Epithelial 3D Cell Culture with Donor-Matched Fibroblasts and Comparison of Two Different Culture Media
Previous Article in Special Issue
METTL3 Regulates Osteoclast Biological Behaviors via iNOS/NO-Mediated Mitochondrial Dysfunction in Inflammatory Conditions
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk

1
Department of Doctoral Studies, Rīga Stradiņš University, LV-1007 Riga, Latvia
2
Joint Laboratory of Electron Microscopy, Institute of Anatomy and Anthropology, Rīga Stradiņš University, LV-1007 Riga, Latvia
3
Department of Internal Diseases, Rīga Stradiņš University, LV-1007 Riga, Latvia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(4), 4120; https://doi.org/10.3390/ijms24044120
Submission received: 27 January 2023 / Revised: 14 February 2023 / Accepted: 16 February 2023 / Published: 18 February 2023
(This article belongs to the Special Issue Inflammation and the Musculoskeletal System)

Abstract

:
Osteoarthritis (OA) is a chronic, progressive, severely debilitating, and multifactorial joint disease that is recognized as the most common type of arthritis. During the last decade, it shows an incremental global rise in prevalence and incidence. The interaction between etiologic factors that mediate joint degradation has been explored in numerous studies. However, the underlying processes that induce OA remain obscure, largely due to the variety and complexity of these mechanisms. During synovial joint dysfunction, the osteochondral unit undergoes cellular phenotypic and functional alterations. At the cellular level, the synovial membrane is influenced by cartilage and subchondral bone cleavage fragments and extracellular matrix (ECM) degradation products from apoptotic and necrotic cells. These “foreign bodies” serve as danger-associated molecular patterns (DAMPs) that trigger innate immunity, eliciting and sustaining low-grade inflammation in the synovium. In this review, we explore the cellular and molecular communication networks established between the major joint compartments—the synovial membrane, cartilage, and subchondral bone of normal and OA-affected joints.

1. Introduction

Osteoarthritis (OA) is a chronic, progressive, highly debilitating, and multifactorial disease of a joint that is acknowledged as the most common form of arthritis, with an incremental rise in prevalence and incidence globally for the past decades [1]. According to the “Global Burden of Disease Study 2019” data, the burden of OA from 1990 to 2019 has increased by 113.5% and could be estimated at more than 500 million individuals [2]. About 10% of people worldwide suffer from OA-induced symptoms, such as functional disability, chronic pain, and mental health problems [3]. The prevalence of depressive symptoms among OA patients is estimated to be around 20% [3,4]. According to Shuang Zheng et al., depression is found in 25.4% of symptomatic knee OA patients with an annual incidence of 11.2% among them [5]. Han Lu et al. explored that there is a bidirectional association between depression and knee OA [6]. The progression of OA is deleterious to the quality of life, working capacity, health, and social welfare [7].
Multiple factors are reported to contribute to the development of OA, such as increasing age, female sex, obesity and insulin resistance, joint mechanical overloading, experienced musculoskeletal trauma, and genetic predisposition [8] (Figure 1).
Telomere length is a biomarker of aging that declines with advancing years, and its shortening is linked to the pathophysiology and development of OA [9]. In addition, senescent cells secrete factors that induce the aging of other cells in a paracrine manner [10]. Gao et al. found that senescence-associated beta-galactosidase expression by articular chondrocytes correlates with the severity of OA [11]. Evidence of gender differences in joint morphometry, kinematics, pain severity, use of healthcare resources, and functional recovery after arthroplasty is reviewed by Tschon et al. [12]. Obesity, defined as a body mass index greater than 30 kg/m2, more than triples the risk of knee OA [8]. According to Joseph et al., weight loss of more than 5% over four years is associated with less pain and slower knee OA radiographic progression on the Kellgren/Lawrence scale [13]. Joseph and colleagues explain these findings as a higher load transferred on the weight-bearing joint such as the knee or hip [13]. Furthermore, adipose tissue is a major source of adipocytokines, which modulate the immune response and inflammation both locally and systemically, thus contributing to joint degeneration [14]. Belluzzi et al. explored that changes in the infrapatellar fat pad (IFP) can predispose patients to OA progression by producing pro-inflammatory cytokines, growth factors, and profibrotic factors [15]. These changes in IFP facilitate prolonged pain in OA patients [16]. Wu and colleagues found that serum levels of adiponectin and leptin are negatively associated with bone mineral density in OA [17]. Type 2 diabetes and insulin resistance have been reported to associate with OA as a multifactorial pathology [18]. Mechanical load and post-traumatic injury both have been identified as undisputed risk factors in the development of OA; the latter is more common at a younger age and is associated with faster progression [19,20]. Recent research indicates that several OA genetic risk signals affect, or at least include, epigenetic regulators [21]. These risk factors create a beneficial background for a pathology. They are necessary, but not sufficient, to develop OA.
The primary attention in OA research thus far has been devoted to investigating joint cartilage [22]. The potential therapeutic approaches are oriented to stop or delay the progression of cartilage structural changes, thus reversing existing defects of the joint tissue [22]. At the same time, the latest available data about joint structural parts’ involvement in OA progression disclose the importance of research that highlights the interplay between components of a joint, respectively, subchondral bone, cartilage, and the synovial membrane. OA begins with joint dysfunction, primarily affecting the articular cartilage, synovium, and subchondral bone, all of which have close cellular and molecular interactions [23]. Moreover, in the case of a knee OA, the onset and progression of disease are strongly associated with meniscal lesions, both, posttraumatic and age-related degenerative [24,25].
Furthermore, even though, it is acknowledged that changes within joint tissue during OA are mainly degenerative, recent studies underline the importance of chronic inflammation [26,27,28,29,30,31,32,33]. The activation of the immune system, whether innate or adaptive, reflects local changes in the tissue. In the joint, it is strongly associated with low-grade systemic inflammation and the production of damage-associated molecular patterns (DAMPs) released during cartilage degradation [34,35,36,37,38,39,40]. Inflamed synovium acts as a trigger for OA progression, recruiting cells for intra-articular changes. To date, various cells have been recognized as potential contributors to synovial inflammation, mainly synoviocytes, macrophages, fibroblasts, and lymphocytes being the most prominent of them [41,42,43]. Activated cells trigger catabolic and pro-inflammatory cellular responses in the synovial membrane and fluid [44]. Finally, synoviocytes, being exposed to intracellular and extracellular DAMPs, activate chondrocytes and mediate the production of various matrix metalloproteinases (MMPs), chemokines, cytokines, and neuropeptides, thus establishing a vicious cycle between all joint tissue compartments [38].
In this review, we explore the cellular and molecular crosstalk established between the major joint compartments—the synovial membrane, cartilage, and subchondral bone of normal and OA-affected joints.

2. Joint as an Organ in a Healthy State

The synovial joints represent the most common type of joint in the human body [45]. They are composed of subchondral bone, articular cartilage, and a two-layered synovial membrane, which surrounds all the mentioned elements, thus making the joint cavity filled with synovial fluid [45]. In physiological conditions, all joint-forming elements remain in balance or homeostasis [46] (Figure 2).

2.1. Synovial Membrane

The synovial membrane is the innermost portion of the joint capsule, which lines all non-articulating joint structures enclosing the cavity. It makes folded protrusions called villi in the joint cavity that increase the surface of the synovium, thus contributing to the joints’ adaptation during movements [47].

2.1.1. Synovial Fluid

The synovial fluid is a viscous, transparent, and relatively hypocellular liquid. It is a blood plasma transudate that contains signaling factors (cytokines, enzymes, growth factors) and biomolecules in response to synovial fluid redistribution-induced sheer stress (hyaluronan, lubricin), which are synthesized by synovial membrane resident cells (mainly fibroblasts) [48,49]. The synovial fluid is the only source of nutrients for avascular, alymphatic, and aneural cartilage. Moreover, it facilitates lubricating properties, regulating biomechanical stress on the joint [49].

2.1.2. Cellularity of the Synovial Membrane

The synovial membrane is composed of two layers: the lining (intima) layer and the sublining (subintima) layer. This delicate tissue represents a selectively porous barrier with intercellular junctions between the lining layer cells. It lacks the basement membrane that separates both layers [50]. On the one hand, it allows free passage for biomechanical, molecular, and cellular signals between synovial tissue and synovial fluid [51]. On the other hand, the synovial membrane has an immune regulatory function, providing homeostasis and integrity of the synovial tissue, adjacent cartilage, and subchondral bone [50,52,53]. The lining layer consists of two distinct cell types: type A and type B synoviocytes, also known as macrophage-like synoviocytes (MLSs) and fibroblast-like synoviocytes (FLSs). In healthy individuals, the synovial lining is two-layered. Close physical and chemical association and communication with synovial fluid are maintained by MLSs and FLSs [26]. The sublining layer is a loose connective tissue, which can be more adipose, areolar, or fibrous in type. This layer is supplied with blood vessels, lymphatic vessels, and nerves. The cellularity of the sublining layer is low. The major cell population is represented by resident cells such as fibroblasts and macrophages, and non-resident cells such as lymphocytes, and plasmocytes [48,54].

2.1.3. Synovial Populations of Macrophages

The healthy synovial membrane niche contains several resident macrophage subsets based on their localization and surface markers [55]. In physiologic conditions, resident macrophages sense tissue damage, take part in the inflammatory response, and support tissue homeostasis. According to parabiotic mouse experiments and single-cell transcriptome sequencing, the healthy synovial membrane contains resident MLSs with Trem2+CX3CR1+ surface markers (phenotypic marker of efferocytosis MerTK+CD206+, subset Trem2, CX3CR1+ in humans) in the lining, accounting for 40% of the macrophage pool, resident interstitial MHCII+CSF1R+ macrophages (MerTK+Folr2highID2+ in humans), and resident Lyve1+Relma+ perivascular macrophages (MerTK+Folr2+Lyve1+ in humans) in the sublining layer [55,56,57].
In the lining layer, MLSs are tissue-specific barrier-providing macrophages with “apical-basal” polarity, that form tight junctions (zonula occludens) and desmosomes, which thus determine the paracellular ion permeability. The main function of MLSs is to protect the adjacent sublining layer from biochemical stress that could be caused by cartilage degradation particles, cellular cleavage components, and pro-inflammatory cytokines, thus preventing spontaneous inflammation within joint tissue [50,52,58]. Unlike the lining layer, which has a single macrophage population (MLSs), the sublining layer is comprised of diverse resident macrophage populations. There are up to four distinct subsets of resident macrophages, classified according to the surface markers they express [50]. The origin of synovial resident macrophages remains obscure. They are blood monocyte-independent cells, and, theoretically, could appear prenatally [50,59]. Though, resident MLSs and perivascular macrophage pools can be restored from “precursor interstitial macrophages”: MHCII+CSF1R+ (MerTK+Folr2+ID2+ in humans) [59].

2.1.4. Synovial Populations of Fibroblasts

Fibroblasts are mesenchymal-derived cells that can execute their lineage functions, such as accommodating the extracellular matrix (ECM) by synthesizing, assembling, and remodeling various types of collagen, proteoglycan, and fibronectin, as well as expressing MMPs and their inhibitors. Apart from coordinating remodeling processes, fibroblasts have resident tissue-specific functions [60]. FLSs are in charge of maintaining internal joint homeostasis. They do this by controlling the composition and turnover of synovial fluid, which lubricates and feeds cells in the deepest joint compartments. This function of FLSs is under the control of mechanical loading and inflammatory stressors [61]. An interaction of FLSs and MLSs supports the immunological barrier in a healthy synovial membrane [52]. Furthermore, all fibroblasts augment other cell functions in both healthy and affected tissue [58].

2.1.5. Synovial Populations of Non-Resident Cells

Other cells that are found in the synovial membrane are vascular endotheliocytes, as well as immune cells such as lymphocytes, neutrophils, mast cells, plasmocytes, and dendritic cells. In healthy synovial membranes, most of them reside in small numbers in the perivascular compartment [62].

2.1.6. Extracellular Matrix of the Synovial Lining

Resident fibroblasts are the main source of ECM. It is composed of fibrillar components such as collagens, fibronectins, elastin, and non-fibrillar components such as proteoglycans and hyaluronan. Cell-to-matrix interactions strictly coordinate the balance between the production, assembly, and degradation of ECM components [63].

2.2. Articular Cartilage

In synovial joints, articular cartilage is a highly specialized, load-bearing, friction-reducing connective tissue that overlays the ends of opposing articulating bone surfaces, providing smooth sliding for a joint during movements and executing properties of resiliency and deformability [46,57,64,65].

2.2.1. The Composition of Articular Cartilage

In the recesses of a joint cavity, articular cartilage is attached to the synovial membrane that lines the joint. Cartilage is an aneural, alymphatic, and avascular tissue that receives nourishment from synovial fluid and the underlying bone [49]. Wang et al. explored that nutrition supplied by the synovial fluid is more significant for cartilage viability than that from subchondral bone [66]. Articular cartilage is made of chondrocytes that produce ECM, which is comprised of water (more than 70%), collagen, non-collagenous proteins, and proteoglycans [49,65,67]. The specific architecture of joint cartilage explains its limited ability to self-repair and its inherent limited healing potential. Unfortunately, both natural and pathological changes in articular cartilage may cause damage to the whole joint’s stability.

2.2.2. Chondrocytes and Their Metabolic State

In mature cartilage, the arrangement of chondrocytes and ECM is more zonal [64]. The chondrocyte distribution, activity, and cellular shape, as well as the orientation and metabolism of the ECM structure within each zone, differ [64,68,69,70]. Histologically, there are four distinct layers in articular cartilage: the superficial, the middle, the deep, and the mineralized cartilage zones [69]. The latter zone is separated from non-mineralized cartilage by a tidemark [71]. In physiological conditions, the tidemark varies in number and thickness all along the cartilage. The tidemark plays a role in preventing the intrusion of mineralized cartilage into the non-calcified cartilage [72,73,74].
Up to 10% of cartilage tissue is occupied by chondrocytes, cells with phenotypic instability and poor regenerative capacity [64,67,68,75]. Chondrocytes are mechanosensitive cells whose cellular responses are influenced by their mechanical and chemical surroundings [61,76,77]. Normally, chondrocytes adapt their metabolic state to the environment by transducing received mechanical signals into chemical responses, which manifest as perturbations in gene expression responsible for remodeling processes, morphogenesis, the ECM, and matrix-degrading enzyme synthesis [78]. The synthetic ability of chondrocytes decreases with cartilage depth [79].

2.2.3. Extracellular Matrix of Articular Cartilage

ECM is a complex network composed primarily of collagen type II (90% of all collagens), with minor pericellular collagens such as type IV, VI, and III, as well as fibril-associated collagens with interrupted triple helices (FACIT) collagens IX, XII, XIV, XVI, and XXI [67,80]. Collagen fibrils of articular cartilage ECM are arranged to resist the mechanical forces: collagen fibril orientation is parallel to the surface in the superficial zone, more randomly arranged in the middle zone, and vertically aligned in the deep zone [68,75]. That alignment is paralleled by chondrocytes’ arrangements and cellular activity profiles within each zone [81,82]. Thus, activated mature chondrocytes are mostly located in the superficial layer and partly in the middle layer; the cells synthesize collagen types II (COL2B), IX, and XI, whereas hypertrophic chondrocytes in the deep layer synthesize collagen type X [64,68,75]. Chondroprogenitor cells produce procollagen type IIA (COL2A) [82]. The pericellular matrix (PCM) buffers stress for chondrocytes and exhibits a different profile. It is mainly made of collagen types IV and VI, fibromodulin, and matrilin-3 [61,65,83]. Taken together, collagens provide articular cartilage with flexibility, compressive resistance, and tensile strength. Simultaneously, pericellular collagens anchor and maintain the integrity of chondrocytes, as well as mediate cell-to-matrix interactions [67]. The ECM collagen network is stabilized by proteoglycan aggregates, mainly aggrecan, with lesser amounts of decorin, biglycan, and fibromodulin, which make collagen type II inaccessible to proteinases and protect it from degradation, as well as non-collagenous proteins such as cartilage oligomeric matrix protein (COMP), link protein and many others [22,46,84].

2.3. Microenvironment in the Subchondral Bone

Together with articular cartilage, subchondral bone forms a morphofunctional unit called an “osteochondral unit” [85]. The osteochondral junction connects the calcified zone of the hyaline cartilage with the cortical plate of the subchondral bone and creates a mechanical and biochemical interplay between articular cartilage and subchondral bone [71,86]. The coupled mechanical interplay of both tissues increases the functionality of a joint, dissipating energy transfer [87].

2.3.1. Architecture of the Subchondral Bone

Subchondral bone is separated from adjacent calcified cartilage by a cement line and is divided into two entities: subchondral cortical bone plate and subchondral trabecular bone [88]. The subchondral cortical bone plate contains channels with nerves and blood vessels that run from the subchondral trabecular bone. These blood vessels and nerves divide into smaller branches and reach into calcified cartilage. Subchondral trabecular bone is a sponge-like supportive structure that absorbs energy and supplies nutrients for remaining bone and cartilage [85].
Subchondral bone is a unique shock-absorbing tissue that dynamically adapts to the mechanical strain, decreasing the transmitted loading to the overlaying articular cartilage [88]. The process of adjusting bone structure to local strain is called “modeling”. The complex mechanism of bone resorption with subsequent formation is known as bone remodeling [89,90]. In physiological conditions, it is activated by biomechanical factors [91].
The microenvironment of subchondral bone depends on resident cells—osteoblasts, mature osteocytes, and osteoclasts—and their interaction with articular cartilage [71,85,92]. Lajeunesse and colleagues reported that prostaglandins, leukotrienes, and growth factors produced by subchondral osteoblasts could reach the calcified layer of articular cartilage [93]. Wu and his team found exosomes that osteoblasts release and thus facilitate intercellular communication between cartilage and bone [94].

2.3.2. Osteoblast Lineage Cells

Osteocytes comprise 42 billion cells in the adult human skeleton with a cellular density of 19,000–28,500 cells/mm3 [95]. Their processes are embedded in the mineralized bone matrix and together with the lacunar–canalicular network, they make a connected system that links the vasculature with the mineralized surface, supplying the bone with nutrients [96]. Bone metabolism is mainly regulated through the wingless (Wnt) signaling pathway and the receptor activator of nuclear factor kappa B (NF-κB)/receptor activator of NF-κB ligand-osteoprotegerin (RANK/RANKL-OPG) pathway [97].
Osteoblast lineage cells release bone metabolic activity regulator proteins such as sclerostin, RANKL, and OPG [98]. RANKL and OPG are mainly expressed by osteoblasts. Osteocytes are the major source of skeletal sclerostin, an inhibitor of the wingless-related MMTV integration site’s (Wnt) canonical signaling pathway. The Wnt signaling pathway induces mesenchymal stem cell differentiation into osteoblasts [99]. Song et al. explored that forced Wnt7b expression in mice results in high bone formation [100]. Sclerostin synthesis is mainly amplified by immobilization [101,102]. Skeletal sclerostin gene knockout mice demonstrate high bone mass [103]. The induction of sclerostin synthesis is a major factor in stopping osteoblast-associated bone remodeling [104].
Osteoblasts are responsible for extracellular bone matrix synthesis, as well as for osteoclastogenesis by RANKL and OPG expression as well as the canonical Wnt signaling pathway [105].

2.3.3. Macrophage Lineage Cells

Osteoclasts are macrophage lineage cells that are responsible for bone resorption. RANKL is a positive regulator of osteoclastogenesis [106]. OPG is a soluble decoy receptor that possesses an antagonizing effect, neutralizing RANKL and maintaining the bone [107].

2.4. Cellular and Molecular Interactions in the Healthy Joints

The cross-tissue interaction in subchondral bone and cartilage and a network-based molecular regulation by the synovium are responsible for healthy environments in joints [24,108]. In Table 1, we collected and represented data focusing on a given healthy condition—molecular activity in each resident or non-resident cell type.

3. Pathological Changes in OA-Affected Joint Tissues

OA has been described for a long time as a mechanically induced degenerative process of the joint [78,114]. OA starts gradually when the disturbance in the joint microenvironment appears with an imbalance between synthesis and destruction processes, pro-inflammatory and anti-inflammatory effects [115]. The pathological processes result in cartilage loss, subchondral bone remodeling, and synovial inflammation (Figure 3). The main joint components—synovial membrane, articular cartilage, and subchondral bone—contribute to the development of OA to varying degrees, thus reflecting various OA subgroups and phenotypes in patients [116].

3.1. OA-Affected Synovial Membrane Triggers and Perpetuates the Pathological Process

There is increasing evidence that the synovial membrane plays the role of “a cornerstone” in the progression of OA. OA presents with pain, joint swelling, and morning stiffness, which are mostly linked to synovial inflammation (synovitis) [40,117]. In combination with mechanical factors, synovitis could be the first trigger for the activation of the immune system and the further perpetrator of the pathologic cycle in OA pathogenesis [118]. The importance of synovitis in OA progression, joint destruction, and associated manifestations has been highlighted over the last decades [119,120].

3.1.1. Changes in the Biochemical Profile of Synovial Fluid in OA

Despite the low number of immune cells found in joint tissues, serum, and synovial fluid in OA patients, the alterations in synovial fluid’s biochemical profile reflect changes in cell metabolism in the joint. Under the influence of joint disease, many cytokines and collagen turnover biomarkers accumulate in synovial fluid, including COMP, CC-chemokine ligand 5 (CCL5), matrix metalloproteinase-1 (MMP-1), MMP-3 and MMP-18, as well as tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and IL-8 [121]. Elevated IL-6 level in synovial fluid may be a predictor of severe OA, and its level correlates with severe pain and is associated with chronic inflammation, while the concentration of COMP found in the serum correlates with the number of OA-affected joints [7].

3.1.2. Alterations in of the Synovial Membrane Reflecting Inflammatory and Fibrotic Changes

The microenvironment of the synovial membrane undergoes substantial changes in the case of joint inflammation. Hyperplasia of the lining layer and increased cellular density of the sublining layer, inflammatory cell infiltration (macrophages, T and B cells), and angiogenesis are all common features of OA-related synovitis [122]. Krenn et al. proposed a histopathological grading system to evaluate the damage to the synovial membrane and define the synovitis grade in inflammatory and non-inflammatory arthritides by using light microscopy [54]. Patients with OA-related synovitis may have different levels of inflammation, but in most cases, it is a low-grade [26,27].
Synovitis occurs in more than 50% of early OA patients [33,48]. It is linked to a higher risk of cartilage damage, thus worsening the disease, and correlates with an increase in pain sensation [120,123,124,125,126,127]. According to arthroscopic and soft tissue ultrasound (US) examination data, synovitis, confirmed by these imaging methods, is seen before OA is revealed in conventional radiographs and cartilage damage is present in magnetic resonance imaging (MRI) [120,128]. Thus, it could be concluded that synovitis could precede cartilage damage or the development of radiographic OA. Riis et al. found a positive correlation between MRI data, cartilage histopathological data, and the severity of synovitis, whereas Abbasi and colleagues revealed a correlation between the severity of ultrasonographic synovitis and pain [129,130]. Synovitis increases the risk of presenting with painful knee OA ninefold [124]. Zhang and colleagues explored that pain in OA patients is positively associated with synovitis and bone marrow lesions (BMLs). The diminution of BMLs and synovitis frequently results in pain resolution [131]. However, Bacon et al. found that a 0.1 mm decrease in cartilage thickness over 2 years increases the Western Ontario and McMaster Universities osteoarthritis index (WOMAC) pain score by only 0.32 points, implying that cartilage loss does not significantly worsen the pain and that the cartilage loss associated with pain is mainly mediated by synovitis [132].
Synovial membrane cells produce mediators with proteolytic and pro-inflammatory activity [33,65]. Approximately 55% of OA-related cytokines are produced by synoviocytes; 38% of them are produced exclusively by synoviocytes [33]. Elevated levels of pro-inflammatory mediators are found in the synovial fluid, synovial tissue, and serum of OA patients [133,134,135]. Nonetheless, all findings from controlled randomized trials on the potential use of biological agents in OA treatment remain controversial, with no significant differences in OA alleviation compared to a placebo [136]. Unfortunately, as the grade of synovitis differs between OA subjects, the synovial membrane’s cellular compartment is highly heterogeneous between patients, thus the dominant effector cell type in OA pathogenesis remains unknown [33,57]. Chou and colleagues performed single-cell RNA sequencing, revealing 12 distinct cell subpopulations in the OA synovium, more than 77% of them being FLSs, and sublining fibroblasts and about 12% being HLA-DRA+ cells, including macrophages, dendritic cells, proinflammatory fibroblasts, and B cells [33]. Macrophages, fibroblasts, and other cellular players can impact each other’s transcriptomes, propagating synovial inflammation, cartilage breakdown, and subchondral bone sclerosis [33].
Synovial fibrosis is another OA manifestation related to joint pain and stiffness [137]. It is a consequence of chronic inflammation and angiogenesis. Synovial fibrosis is caused by an imbalance between collagen synthetic and proteolytic processes as a response to pro-inflammatory factors such as IL-6, IL-1β and pro-fibrotic factors such as transforming growth factor-beta (TGF-β) and connective tissue growth factor (CTGF). MLSs and FLSs are both implicated in synovial fibrosis [138,139]. Interestingly, IFP has been acknowledged as a source of pro-inflammatory factors and adipokines, thus potentially contributing to synovial inflammation and fibrosis [140]. Furthermore, the IFP function is thought to be linked to the synovial membrane [141]. Both can contribute to fibrosis, hypervascularity, and vascular remodeling, resulting in a pro-inflammatory microenvironment, as well as pain sensitization [141,142]. Moreover, Belluzzi et al. highlighted an elevated expression of Piezo1/2 mechanosensitive ion channels in IFP and synovial membrane vasculature that suggests the aforementioned contribution of this anatomo-functional unit to pain in OA [143].

3.1.3. Insights into Synovial Macrophage Diversity Confirmed in OA Patients

Resident and non-resident synovial lining and sublining macrophages are the most prominent cellular component of innate immunity in OA [42]. One of the classical ways to distinguish macrophages is by their state of polarization. The cells are classified as M1 (pro-inflammatory) or M2 (anti-inflammatory) depending on the surface markers. This concept of M1 and M2 macrophage polarization exists since Mills et al. defined two activation poles as M1 and M2 in 2000 [144]. However, a recent study suggests that there are at least three subsets of M2 macrophages: M2a, M2b, and M2c [59]. M2 macrophages contribute to tissue repair and remodeling by releasing IL-10, TGF-β, IL-1 receptor antagonists, and a variety of other mediators [49,57,115]. The transition from M2 to the M1 state accelerates joint destruction via TNF-α, IL-1β, IL-6, and IL-17 [57]. As macrophages have even greater phenotypic heterogeneity and plasticity, characterized by the presence of immunoregulatory and cartilage remodeling proliferative functions, this conception is now considered extreme and imprecise [33].
Upon the onset of synovitis, blood-derived macrophages (MerTK) infiltrate the sublining layer, leading to the formation of a variety of pro-inflammatory non-resident macrophage subtypes [58]. Sublining inflammatory macrophages produce more pro-inflammatory mediators than lining MLSs [33]. Activated macrophages are associated with cartilage breakdown directly and indirectly via the secretion of cytokines and the dynamic interplay between fibroblasts and chondrocytes. They produce MMP-1, MMP-3, MMP-9, MMP-13, and aggrecanase and downregulate anabolic processes within cartilage [57]. Blom et al. identified that macrophage depletion from the synovial membrane causes a significant decrease in MMP-3 and MMP-9 [145]. The abundance of activated macrophages in the sublining layer correlates with knee OA radiographic severity and symptoms [146]. Huo et al. found elevated levels of the macrophage chemoattractant fractalkine (CX3CL1) and CCL2 in OA patients’ synovial fluid and serum as well as their positive association with pain and poor clinical outcome [147]. Interaction with CX3CL1 induces pro-inflammatory and degrading enzyme production by macrophages [148]. In animal models, CCL2/CCR2 blockade results in macrophage number reduction, diminished synovitis, and cartilage damage [149]. According to Daghestani et al., soluble macrophage-associated molecules sCD163 and sCD14 shed from activated macrophages, are found in OA synovial fluid and serum, and reflect OA symptomatic severity, osteophyte formation, and joint space narrowing [150]. In another study, macrophage depletion from the synovial membrane was associated with a reduction in osteophyte formation during experimental OA. Downregulation of both TGF-β and bone morphogenetic protein-2 (BMP-2) plays a role in osteophyte formation in this case [151].
Over time, mechanical and biochemical factors reduce the abundance of MLSs (MerTK+CX3CR1+Trem2+ lining macrophages) from 40% to 10% of all synovial macrophage pools. This results in tight junction disruption between MLSs and the breakdown of the lining layer’s barrier-like structure. According to MRI data, the breakdown of the MLS barrier-like structure is associated with increased intra-articular accumulation of the infused contrast agent [50]. In the arthritic mouse models, the depletion of these cells leads to increased inflammatory cell infiltration into the synovial membrane and a pro-inflammatory microenvironment [50,52,57]. Of note, in inflammatory arthritis, MerTK+ macrophage number inversely correlates with disease activity, angiogenesis, and hyperplasia of the synovial membrane [55].

3.1.4. Distinct Subsets of Synovial Fibroblasts Confirmed in OA Patients

Inflammatory and mechanical factors influence the phenotype and function of FLSs and sublining fibroblasts [152]. The amount of FLS-synthesized lubricin and hyaluronan is decreased in the OA joint under the influence of excessive stress and pro-inflammatory cytokines [152]. This accounts for the diminished volume and viscosity of synovial fluid that is associated with OA-related pain. Interestingly, intra-articular lubricin injections into OA mouse models downregulated the inflammatory environment and alleviated pain [153]. Intra-articular hyaluronan injections may also provide pain relief and improve joint function [154]. Noteworthily, the diminished hyaluronan synthesis by FLSs is attributable to the pro-inflammatory microenvironment in OA, which induces the depolymerization of high molecular weight hyaluronan into small oligosaccharides [155]. FLSs and sublining fibroblasts’ phenotypes switch to destructive and pro-inflammatory, respectively. However, there is no clear evidence for a phenotype switch in OA when compared to typical inflammatory arthritis [156,157]. Nevertheless, some studies describe the activation of synovial fibroblasts with upregulation of the NF-κB pathway under the influence of pro-inflammatory factors [158]. A switch from FLSs to myofibroblast-like cells is associated with synovial fibrosis [159]. Moreover, elevated levels of fractalkine CX3CL1 in the synovium induce FLSs and sublining fibroblasts to synthesize MMP through the NF-κB pathway, particularly MMP-3, MMP-9, MMP-2, which degrade aggrecan and collagen [148,160,161]. Studies on transcriptomics reveal that all synovial fibroblasts demonstrate positional diversity, which makes them respond differently to the same pro-inflammatory signals. As a result, the TNF-α response may result in matrix degradation or an increase in pro-inflammatory responses [162].

3.1.5. Non-Resident Cell Role in the Progression of OA

Trajerova and colleagues identified several immune cell phenotypes of knee OA based on synovial fluid-derived immune cell composition. They found increased infiltration of T cells, monocyte–macrophage lineage cells, NK cells, and neutrophils [163]. A flow cytometry analysis showed the higher infiltration of synovial fluid and synovial tissue by CD4+ T cells with an increased CD4+ to CD8+ ratio, particularly Th1-polarized cells, in early stage and end-stage OA [43,164]. In addition, the abundance of Treg cells is usually reduced [118].

3.1.6. Danger-Associated Molecular Associated Patterns Characteristic of OA-Caused Damage

The immune system is an active player during synovitis. Immune cells react to mechanical and biochemical stimuli that accumulate within joints over time [165]. Schedel and colleagues found that cartilage and subchondral bone cleavage fragments, and ECM degradation products stimulate the synovial membrane. This stimulation results in FLSs and MLSs binding to debris with further absorption of them, producing detritus synovitis. These changes do not lead to an invasion, as it is in RA [166]. The formation of detritus synovitis may have a further aggravating effect on the inflammation process as well as on cartilage loss [166]. Thus, it can be speculated that cartilage and subchondral bone ECM components could stimulate the innate immune system [7]. In that case, ECM components and intracellular proteins from apoptotic or necrotic cells, which occur after prolonged mechanical overuse or during the physiological aging process, are considered foreign bodies. As foreign bodies, they serve as DAMPs that trigger innate immunity, eliciting and sustaining sterile low-grade inflammation [57,117,167,168].
DAMPs or alarmins are endogenous molecules produced during tissue remodeling that link inflammation with immune responses in OA [169]. Physiologically, they induce immune responses to trigger the repair of damaged tissue and the clearance of debris [117]. In OA, this response is altered [117]. ECM cleavage remnants from proteoglycans, fibronectin, low-molecular-weight hyaluronan, tenascin C, intracellular cleavage products from apoptotic or necrotic cells, uric acid, and other crystals, as well as plasma proteins, serve as DAMPs [170,171]. DAMPs, S100A8 and S100A9 proteins, and uric acid are closely related to the age-related OA phenotype [171]. Depolymerized hyaluronan fragments accumulate within the synovial membrane and upregulate membrane-associated Toll-like receptor 4 (TLR4) and CD44 receptors’ expression [172]. Interestingly, cell migration-inducing hyaluronidase-1 (CEMIP), which plays a role in hyaluronan catabolism, is increased in inflamed synovial membranes and overexpressed by FLS [173]. Moreover, the DAMP-like action of hyaluronan is molecular weight-dependent [172]. Low-molecular-weight hyaluronan and small oligosaccharides propagate further degradation of the ECM and amplify the inflammation responses through the NF-kB pathway, thus stimulating the release of pro-inflammatory mediators and destructive enzymes [172].
Calcium-containing crystals, both basic calcium phosphate (BCP) and calcium pyrophosphate dihydrate (CPP), are potential DAMPs that are found in the synovial fluid of OA patients, induce pro-inflammatory responses in synovial tissue, and exacerbate articular cartilage calcification [174,175].
DAMPs are recognized by pattern recognition receptors (PRR), which include TLRs on the surface of immune cells, cytoplasmic PRRs such as nod-like receptors (NLRs), or secreted receptors such as complement receptors [49,176]. TLRs are found on the cell surface (TLR-1, TLR-2, TLR-4, TLR-5, TLR-6) and on the surface of endosomes (TLR-3, TLR-4, TLR-7, TLR-8, TLR-9). Although in smaller numbers than in RA, TLR-2 and TLR-4 are found to be overexpressed in the OA synovial membrane [165]. Stolberg-Stolberg and colleagues studied the contribution of TLR-3 activation to cartilage degeneration. In vitro, it was revealed that TLR-3 is upregulated in chondrocytes of OA cartilage and reacts to apoptotic chondrocytes. The TLR-3 knockout mice were protected from OA-like cartilage breakdown [177]. It is also worth mentioning, that satellite glial cells of the dorsal root ganglia (DRG) express many TLRs. Miller et al. revealed that TLR-2 in the DRG is activated by aggrecan cleavage fragments, promoting OA-associated pain [178]. TLRs activation triggers the production of pro-inflammatory cytokines, chemokines, proteolytic enzymes, and growth factors such as TNF-α, IL-1β, IL-6, IL-8, IL-15, IL-17, IL-18, IL-21, CCL-5, MMP-1, MMP-3, MMP-9, MMP-13, TGF-β, fibroblast growth factors (FGF), vascular endothelial growth factor (VEGF), nerve growth factor (NGF), and many others by activation of NF-κB, activator protein-1 (AP-1), and mitogen-activated protein kinase (MAPK) [117,165,170]. In addition, the complement system may be implicated in endogenous molecules that induce chronic inflammation in OA [117].

3.2. Degradation of the Articular Cartilage in OA

The changes to articular cartilage include alterations to the cartilage matrix and cellular components. Disturbed matrix cellular organization leads to tissue fibrillation, vertical fissures, degradation of the cartilage, and endochondral ossification. On a molecular level, the loss of aggrecan, decreased quantity and quality of collagen type II, collagen type I accumulation, and perturbations of chondrocytes are observed [114]. Overall, it results in decreased tensile strength and energy-storing capacity, limiting joints’ functional abilities.

3.2.1. Chondrocyte Phenotypes in Cartilage Affected by OA

Compositional and organizational changes in ECM and pericellular matrix (PCM) affect chondrocytes’ behavior [67,179,180]. Normally quiescent chondrocytes start clustering to form multicellular chondrons to satisfy anabolic demands [167]. Unfortunately, the composition of newly produced collagen is switched from type II to type I, which weakens cartilage durability and turns hyaline cartilage into a tensile-incompetent fibrotic cartilage-like structure [75,181]. Even though with time, actively proliferating, swollen chondrocytes become more susceptible to hypertrophy and dedifferentiation, it inevitably results in cellular damage and death [182]. In the late stages of OA, chondrocyte anabolic activity cannot compensate for catabolic changes within cartilage, leading to a thin, calcified, and functionally incompetent structure that propagates into the non-mineralized cartilage, reducing its thickness [45,82].
Chondrocytes undergo phenotypic changes in response to mechanical stress, various self-produced cytokines, and growth factors, accompanied by the synovial membrane and subchondral bone cell-produced pro-inflammatory factors. Chou and colleagues explored the phenotypic and functional heterogeneity of articular chondrocytes and their potential upstream regulators during OA by genotyping [33]. They identified seven distinct cell phenotypes such as homeostatic chondrocytes (HomC), prehypertrophic chondrocytes (preHTC), hypertrophic chondrocytes (HTC), regulatory chondrocytes (RegC), prefibrochondrocytes (preFC), fibrochondrocytes (FC) and reparative chondrocytes (RepC). PreFC, FC, preHTC, RepC, and RegC are predominantly seen in the arthritic part of the cartilage. HomC regulates cell metabolism, development, and homeostasis [33,183]. PreHTC expresses genes to control hypertrophic differentiation [183]. HTC expresses genes to control ECM mineralization but also induces angiogenesis and osteoblast invasion into cartilage tissue [183]. RegC expresses genes for antigen-presenting function, as well as modulates different signaling pathways [183]. RepC expresses genes for ECM remodeling, while PreFC and FC express genes for fibroblast-like activity [33].
However, there are studies suggesting the presence of more chondrocyte phenotypes [68,184]. A dedifferentiated chondrocyte is another phenotype of a cartilage cell. In physiological conditions, the microenvironment and other factors such as the master regulator SRY-box transcription factor 9 (SOX-9) restrain chondrocytes from losing their physiological chondrogenic phenotype or dedifferentiation [185]. Morphologically and functionally, dedifferentiated chondrocytes become fibroblast-like cells, which despite their high synthetic ability, produce collagen with reduced mechanical properties (collagen type I, type III, type V), thus contributing to articular cartilage disturbance, remodeling, and fibrosis [64,82]. Synoviocytes also release pro-inflammatory cytokines, such as IL-1α, IL-1β, and TNF-α, which aid in dedifferentiation. Moreover, dedifferentiation also leads to a hypertrophic osteoblast-like phenotype with increased MMP-13 expression in chondrocytes and collagen type X secretion. Nidogen-2 is one of the chondrocyte pericellular matrix components, that regulates the antagonizing action of RUNX2 and SOX-9. Nidogen-2 depletion also contributes to the hypertrophy of chondrocytes and cartilage calcification [186]. As mentioned above, IFP is a major intra-articular source of pro-inflammatory cytokines or adipokines. Gomez and the team reported that adiponectin induces the expression of IL-8 in chondrocytes more potently than IL-1β, thus contributing to the degradation of articular cartilage [187].
Furthermore, available data suggest that chondrocytes express a resistant-to-apoptosis senescent cell phenotype with a proinflammatory secretome in damaged cartilage [188,189,190]. Moreover, senescent chondrocytes are present in osteoarthritic lesions with an increased tendency to accumulate with age but are absent in intact cartilage [189,191]. Jacob and colleagues explored that chondroprogenitor cells obtained from OA cartilage display morphological features of senescent cells, expressing a senescence-associated secretory phenotype with increased levels of reactive oxygen species (ROS), IL-6, and IL-8 [192]. In addition to that, the authors report the main stress-inducers, metabolic changes with impaired mitochondrial function, oxidative stress, and genomic damage with upregulated p16INK4a expression [167,192,193].

3.2.2. Remodeling of Extracellular Matrix during the OA

Cartilage swelling is the first sign of the ECM degradation process, which appears when the osmotic properties of cartilage are changed [194]. It is detected by MRI as cartilage “thickening” and positively correlates with proteoglycan loss in early experimental arthritis [195]. Bank et al. first explored that swelling of the cartilage appears due to the loss of the collagen network [196]. Other studies have highlighted that increased catabolism of proteoglycans, particularly aggrecan, is the main contributor to the increase in water content in cartilage tissue with no significant differences in collagen composition in early stage OA [197,198]. On the molecular level, cartilage hyperhydration is driven by the loss of aggrecan’s G1 domain, which normally binds this proteoglycan to the link protein, hyaluronan, and matrix itself [49,199]. Interestingly, the loss of hyaluronan from cartilage and its depolymerization is also associated with pro-inflammatory conditions and enhanced production of IL-1β and TNF-α that lead to CEMIP overexpression by chondrocytes correlated with OA activity [173]. The perturbations in the composition of the proteoglycan network typically begin in the superficial zone of the cartilage extending to the deep zone as the OA evolves [200]. It is worth mentioning that aggrecan degradation by itself does not lead to OA progression. Major proteoglycan degradation proceeds with the loss of minor ECM components, an irreversible breakdown of microfibrillar collagen, and the depletion of collagen type II, resulting in disruption of the collagen network, known as fibrillations and fissures [46,199].
The changes in ECM influence the composition of PCM [61]. PCM is a part of ECM, which surrounds the groups of chondrocytes, making a common entity called the chondron. Disturbed integrity of PCM is associated with the loss of microfibrillar collagen type VI, the main component of the PCM, as well as collagen type IV, percelan, fibronectin 1, nidogens, and laminins [201]. Schminke et al. found that chondroprogenitor cell stimulation by nidogen-2 stimulation decreases Runt-related transcription factor 2 (RUNX2) and increases both SOX-9 mRNA and aggrecan, whereas stimulation by laminin upregulates type II collagen synthesis and downregulates type I collagen synthesis, thereby supporting chondrogenesis. Thus, the authors conclude that the decrease in PCM components, as it is observed in OA, contributes to the increase in collagen type I and cartilage endochondral ossification [186].
It is reported that the loss of microfibrillar collagen type VI alters the mechanotransduction through the chondrocytes’ primary cilium [202]. Furthermore, PCM collagen type IV depletion aids to change the viability and phenotype of chondrocytes [67,78,203].
Overall, PCM alterations decrease chondrocyte abilities to sense mechanical signals, exposing the cell to swelling, and making them susceptible to phenotypical perturbations.

3.2.3. Cartilage Destruction-Associated Proteases Activated in OA

All catabolic changes within articular cartilage are managed by excessive production of matrix-degrading enzymes, such as matrixins or zinc-dependent endopeptidases, MMPs, and a disintegrin and MMP with thrombospondin motifs (ADAMTS), as well as a diminished synthesis of their inhibitors [204,205]. The key aggrecan-degrading enzymes are ADAMTS-4, ADAMTS-5 and ADAMTS-9, to a lesser extent MMP-1, MMP-3, MMP-9, MMP-13 [206]. The major structural macromolecule in the ECM, collagen type II, is cleaved mainly by MMP-13, MMP-1, and MMP-3. These matrix-degrading enzymes, which act in an autocrine and paracrine fashion, are synthesized by both chondrocytes and synovial FLS [31,45]. MMP hyperproduction and hyperactivity, particularly ADAMTS, are caused by altered post-translational endocytosis of these enzymes [207,208]. Genetic polymorphism in the enzyme that regulates ADAMTS activity, as well as in the enzyme structure, is positively associated with the occurrence of OA [208,209]. OA-related proinflammatory cytokines, which are derived from synovial membrane cells, also contribute to the hyperproduction of matrix enzymes [207].

3.2.4. Cartilage Destruction-Associated Cytokines Contributing to OA Development

A major part of the OA key cytokines that regulate chondrocyte transcription and function originate from synovial membrane resident cells; 38% of OA-related key cytokines are exclusively produced by synoviocytes and none of the cytokines are exclusively produced by chondrocytes [33]. According to recent studies, resident macrophages and non-resident dendritic cells, but not chondrocytes, express interleukin-1 beta (IL-1β), interleukin-6 (IL-6), and TNF-α, all of which are mostly expressed in synovial membranes [33]. ADAMTS-5 expression is mainly activated by IL-1β, TNF-α, and TGF-β [210]. Notably, MMP-13, the main collagen type II cleaving enzyme, is expressed not only by chondrocytes but also by FLSs and osteocytes, thus pointing to a relationship between joint cells and their increased impact on cartilage [7]. Anabolic changes are stimulated by TGF-β and insulin-like growth factor-1 (IGF-1). Catabolic changes are mostly controlled by IL-1α and IL-1β [33,211]. Cai and the team have studied that chondrocyte stimulation with IL-1β elicits an immune responsive gene 1 (IRG1) associated pro-inflammatory response [212].

3.3. Alterations in the Subchondral Bone Affected by OA

The microstructure of the subchondral bone changes during the OA progression. Changes in cortical bone plates: volume, porosity, composition, and mineralization degree; transformation of the trabecular bone compartment; the appearance of bone marrow lesions (BMLs); neovascularization; cyst and osteophyte formation; and sclerosis are examples of typical alterations [45,213]. When the bone’s adaptive capability to mechanical and biological signals is exceeded, bone remodeling decouples [48,214].

3.3.1. Microstructure of Subchondral Bone during the Early Stage OA

In the early stages of OA, a high bone turnover with resorption dominates, resulting in cortical bone thinning, increased porosity, trabecular compartment widening, and separation of trabeculae [22,88]. Findings of high bone turnover are supported by the hyperexpression of bone resorption markers [46,91]. For example, Huebner et al. found that the urinary alpha C-telopeptide of type I collagen (α-CTX) marker, which is a metabolite of type I collagen, was elevated in regions with newly formed bone as confirmed by scintigraphy, corresponding to the sites of high bone turnover in knee OA [215]. According to Zhao et al., high bone turnover is reflected by increased rates of leukemia inhibitory factor (LIF), a cytokine secreted by osteoclasts that upregulate the Wnt signaling pathway by lowering sclerostin in osteocytes [91]. The intensity of LIF expression correlates with the stage of OA evolution [216].

3.3.2. Microstructure of Subchondral Bone during the Late-Stage OA

In the late stages of OA, increased bone synthesis and hypomineralized bone volume are more evident [85]. The relative hypomineralization of bones is explained by the discordance of still high bone turnover and mineralization processes [46]. The cortical bone plate becomes thicker and the rod-to-plate ratio in trabecular bone decreases [88,89,213]. The hallmarks of late OA are osteoid islets and sclerosis [85]. BMLs are one of the results of abnormal reciprocity between cartilage and subchondral bone and are associated with pain and disease progression [217,218]. BMLs occur before the development of radiographic OA and are observed by MRI [218]. BMLs, as well as subchondral bone cysts, express a positive correlation with synovitis severity. BMLs, once present, tend to increase [45,48].

3.3.3. Changes in the Cellular Composition of the Subchondral Bone

On the cellular level, the changes observed are orchestrated by the subchondral bone resident cells, their interaction with chondrocytes and synoviocytes, and the supervision of mechanical and biological signals [214].
In the early stages of OA, osteocytes increase the expression of RANKL and sclerostin and decrease the expression of osteoprotegerin (OPG), thus increasing bone resorption [214,219]. Over the course of the disease, osteocytes shift their phenotype to OA-associated, meaning round-like shape and disarrangement, which interferes with their ability to sense and transduce mechanical stimuli [220].
Impaired osteocyte perilacunar remodeling (PLR) is both the initiator and consequence of OA degenerative processes within the subchondral bone and articular cartilage [221]. Mazur and colleagues found that a deficiency of PLR in OA, particularly MMP-13, causes disruption of subchondral bone homeostasis as well as accelerates articular cartilage lesions [222]. A defect in osteocyte PLR impacts the phenotype of osteocytes and the regulation of osteoblasts and osteoclasts [221]. Fowler et al. explored the hypothesis that glucocorticoid administration inhibits enzymes of the perilacunar matrix, suppressing remodeling and causing the breakdown of the lacunar–canalicular network (LCN) early in the disease [223]. The insult of LCN manifests as decreased lacunar area, which interferes with mechanosensitive functions of osteocytes; a diminished number of osteocyte processes and impaired canalicular flow, which causes undernourishment of subchondral bone cells [97,224,225,226]. It results in osteocytes’ failure to perceive mechanical load and their proneness to secondary necrosis in a state of nutritional deprivation. The death of osteocytes causes new bone formation in both sclerostin-RANKL dependent and independent ways, with the latter being associated with the release of DAMPs during necrosis, which positively stimulates osteoblastogenesis and further bone sclerosis [227,228].
In the late stages of OA, osteocytes induce osteoblast mineralization and enhance osteoblast-mediated collagen type I synthesis [89]. According to Couchourel et al. data, during OA there is an increase in type I collagen α1 chain expression, while α2 chain expression remains stable, thus producing type I collagen, which has a lower affinity to calcium [229,230]. Moreover, the basal expression of impaired type I collagen is increased [230]. TGF-β1 is one of the key cytokines that accelerate new bone formation and angiogenesis, as well as plays an important role in mineralization, osteophyte, and fibrosis formation [214,230]. TGF-β1 in inactivated form is mobilized from the ECM during ongoing bone remodeling. While activated, it stimulates osteoblastogenesis as well as bone–cartilage interaction [214].

4. OA-Affected Impaired Interaction between Joint Compartments

Disturbed cellular and molecular coupling between the synovial membrane, articular cartilage, and subchondral bone promotes the progression of OA. The mutual modulation of the transcriptomes of the cells and unwanted changes in the expression of various mediators affect the stability of the whole joint.

4.1. Altered Interaction between the Synovium and Cartilage

Similarly, to the “osteochondal unit”, the “chondrosynovial unit” deserves just as much attention. Structures interconnected by a synovial fluid are linked molecularly. Together with the synovial membrane, synovial fluid contributes to inflammation and cartilage degradation. Housmans and his group explored the hypothesis that alterations of synovial fluid induce chondrocytes’ dedifferentiation and cartilage degeneration [231]. It is possible to detect the difference between several types of OA based on the immune cells and proteins in the synovial fluid of people with knee OA [163]. As mentioned above, synovial fluid changes its biochemical profile, accumulating various signaling factors that can serve as potential disease biomarkers. Yang and colleagues used antibody array technology to determine the downregulation of 20 proteins and the upregulation of 30 proteins in OA synovial fluid compared to healthy controls, as well as which proteins are involved in OA pathogenesis [232]. Exposure to IL-1α and TNF-α in synovial fluid, for example, modulates the collagen profile of FLSs and promotes the secretion of pro-inflammatory factors such as IL-6, IL-8, prostaglandin E2 (PGE2) and nitric oxide (NO). The expression of pro-inflammatory factors initiates the expression of MMPs and proinflammatory genes by both fibroblasts and chondrocytes, leading to cartilage degeneration [233]. Furthermore, extracellular vesicles derived from the synovial membrane activate chondrocytes through the NF-κB signaling pathway. This leads to the release of proteases degrading cartilage (MMP-9, MMP-13) and pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), which are then found in synovial fluid [234].
By reciprocity, cellular and ECM particle debris in cartilage, which is induced by mechanical and biochemical stress, propagates DAMPs-associated low-grade inflammation in synovial tissue. Common DAMPs such as S100A8, S100A9, and S100A12 upregulate MMP-1, MMP-3, MMP-9, MMP-13, IL-6, ADAMTS-1, ADAMTS-4, ADAMTS-5, ADAMTS-12, VEGF gene expression perpetuating cartilage degradation [235,236,237]. Moreover, synovial cells, particularly fibroblasts, can directly alternate the articular cartilage by releasing proteases that degrade COMPs and other collagenous proteins via the Wnt/β -catenin and RUNX pathways. Furthermore, M1-polarized synovial macrophages from an OA joint negatively regulate cartilage regenerative capacity [238].
Together, altered cellular and molecular communications between the synovial membrane and articular cartilage may serve as amplifiers for the development of OA.

4.2. Impaired Osteochondral Communication during the Development of OA

The deterioration of subchondral bone predisposes to an abnormal distribution of mechanical forces transferred to the joint and precedes the breakdown of cartilage [89,239,240,241,242]. Prior subchondral bone damage increases the risk for cartilage loss within the same anatomical region of a knee joint by 7.5 times [243]. OA affects articular cartilage and subchondral bone as functional units [242]. Alterations in the osteochondral junction occur early in the disease course. They underline the bone–cartilage crosstalk and are pivotal in OA establishment. Pan et al. established that fluorescent dyes are actively transported from subchondral bone to calcified articular cartilage as diffusion capacity increases during the development of OA [71,86]. This is explained by intensive neoangiogenesis in subchondral bone with the later invasion of blood vessels into the osteochondral junction and cartilage [48]. In the pre- and early stages of OA, abnormal neoangiogenesis with type H vessel formation begins. It is supported by IL-6, IL-8, PGE2, TGF-β1, MMP-13, platelet-derived growth factor-BB (PDGF-BB), and vascular endothelial growth factor (VEGF) [229]. In the early stage of OA, one of the main factors for aberrant neoangiogenesis is PDGF-BB, which is secreted by mononuclear preosteoclasts in excessive amounts and acts on endothelial cells and pericytes in a paracrine fashion, as well as stimulating VEGF. VEGF mainly induces neoangiogenesis in late-stage OA [244]. The extent of type H vessels exhibits a positive association with cartilage breakdown [245]. The newly formed vasculature allows cartilage to access bone-released mediators such as TGF-β1, IGF-1, chondrolytic enzymes, bone marrow mononuclear cells that alter chondrocyte metabolism, the evolving calcification of hyaline cartilage, and osteoclast recruitment, which degrades the osteochondral junction. In turn, chondrocytes release proinflammatory cytokines, such as IL-1, which promote RANKL expression in osteoblasts and influence osteoclastogenesis. Furthermore, the expression of IL-6 and TNF-α induces osteoclastogenesis, which destabilizes the bone microenvironment [89]. New vasculature is accompanied by the novel ingrowth of nerve fibers that invade aneural cartilage tissue, causing pain [246]. This crosstalk is primarily mechanical in late-stage OA: cartilage cannot absorb excessive mechanical stress, which promotes abnormal load on the subchondral bone, aggravating remodeling [48].

4.3. Molecular Signaling in OA-Affected Joints

In this subsection, we have presented signaling molecules and their possible actions established between the major joint compartments—the synovial membrane, cartilage, and subchondral bone in OA-affected joints (Table 2).

5. Conclusions and Future Directions

Evidence from many studies on OA shows that the exact causes and effects of the disease are still not clear. Understanding the possible relationship between etiologic factors and the phenotyping of OA based on a synergy of clinical, morphological, and molecular detection methods remains one of the most challenging goals for future studies. Both the “chondrosynovial unit” and the “osteochondral unit” are essentially linked by synovial fluid; therefore, synovial fluid can be the source of important information that can be provided to assist in the creation of a personalized treatment strategy for patients. In addition, future work is needed to create a complex overview of all three joint compartments by confirming the sub-phenotypes of cells and understanding the collaboration between them at the microstructural and molecular levels. The creation of new therapeutics that alter disease-associated joint remodeling before major degeneration takes place, as well as the early identification of those who are at risk of developing OA, are two critical areas for directing future research. By achieving these two goals, we might be able to reduce the cost of OA in our society and improve the quality of life for our aging population.

Author Contributions

Conceptualization, S.S. (Sofija Semenistaja) and S.S. (Sandra Skuja); writing—original draft preparation, S.S. (Sofija Semenistaja) and S.S. (Sandra Skuja); visualizations, S.S. (Sofija Semenistaja) and S.S. (Sandra Skuja); writing—review and editing, S.S. (Sofija Semenistaja), S.S. (Sandra Skuja), A.K. and V.G. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by Rīga Stradiņš University Internal Research Grant Nr. 6-ZD-22/3/2022 “Identification of specific osteoarthritis phenotypes and disease endotypes tackled using the molecular and cellular assessment of the synovium-cartilage-bone interplay by correlation-based network analysis”. The article processing charge was covered by Rīga Stradiņš University Research Department.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

A publicly available bibliographic database, PubMed.gov, was used in this study. The full bibliographic reference list is available upon request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Safiri, S.; Kolahi, A.-A.; Smith, E.; Hill, C.; Bettampadi, D.; Mansournia, M.A.; Hoy, D.; Ashrafi-Asgarabad, A.; Sepidarkish, M.; Almasi-Hashiani, A.; et al. Global, Regional and National Burden of Osteoarthritis 1990-2017: A Systematic Analysis of the Global Burden of Disease Study 2017. Ann. Rheum. Dis. 2020, 79, 819–828. [Google Scholar] [CrossRef] [PubMed]
  2. Long, H.; Liu, Q.; Yin, H.; Wang, K.; Diao, N.; Zhang, Y.; Lin, J.; Guo, A. Prevalence Trends of Site-Specific Osteoarthritis From 1990 to 2019: Findings From the Global Burden of Disease Study 2019. Arthritis Rheumatol. 2022, 74, 1172–1183. [Google Scholar] [CrossRef] [PubMed]
  3. Hunter, D.J.; March, L.; Chew, M. Osteoarthritis in 2020 and beyond: A Lancet Commission. Lancet 2020, 396, 1711–1712. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, S.-T.; Ni, G.-X. Depression in Osteoarthritis: Current Understanding. Neuropsychiatr. Dis. Treat. 2022, 18, 375–389. [Google Scholar] [CrossRef]
  5. Zheng, S.; Tu, L.; Cicuttini, F.; Zhu, Z.; Han, W.; Antony, B.; Wluka, A.E.; Winzenberg, T.; Aitken, D.; Blizzard, L.; et al. Depression in Patients with Knee Osteoarthritis: Risk Factors and Associations with Joint Symptoms. BMC Musculoskelet. Disord. 2021, 22, 40. [Google Scholar] [CrossRef]
  6. Lu, H.; Wang, L.; Zhou, W.; Jin, S.; Chen, H.; Su, Y.; Li, N.; Shang, S. Bidirectional Association between Knee Osteoarthritis and Depressive Symptoms: Evidence from a Nationwide Population-Based Cohort. BMC Musculoskelet. Disord. 2022, 23, 213. [Google Scholar] [CrossRef]
  7. Grässel, S.; Zaucke, F.; Madry, H. Osteoarthritis: Novel Molecular Mechanisms Increase Our Understanding of the Disease Pathology. J. Clin. Med. 2021, 10, 1938. [Google Scholar] [CrossRef]
  8. Allen, K.D.; Golightly, Y.M. State of the Evidence. Curr. Opin. Rheumatol. 2015, 27, 276–283. [Google Scholar] [CrossRef]
  9. Xie, H.; Ma, Y.; Shao, M.; Kong, J.; Zhou, T.; Wang, F.; Cai, G.; Xu, S.; Pan, F. Telomere Length in Patients with Osteoarthritis: A Systematic Review and Meta-Analysis. Aging Clin. Exp. Res. 2022, 34, 495–503. [Google Scholar] [CrossRef]
  10. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.-W.; Lasitschka, F.; Andrulis, M.; et al. A Complex Secretory Program Orchestrated by the Inflammasome Controls Paracrine Senescence. Nat. Cell Biol. 2013, 15, 978–990. [Google Scholar] [CrossRef]
  11. Gao, S.-G.; Zeng, C.; Li, L.-J.; Luo, W.; Zhang, F.-J.; Tian, J.; Cheng, C.; Tu, M.; Xiong, Y.-L.; Jiang, W.; et al. Correlation between Senescence-Associated Beta-Galactosidase Expression in Articular Cartilage and Disease Severity of Patients with Knee Osteoarthritis. Int. J. Rheum. Dis. 2016, 19, 226–232. [Google Scholar] [CrossRef] [PubMed]
  12. Tschon, M.; Contartese, D.; Pagani, S.; Borsari, V.; Fini, M. Gender and Sex Are Key Determinants in Osteoarthritis Not Only Confounding Variables. A Systematic Review of Clinical Data. J. Clin. Med. 2021, 10, 3178. [Google Scholar] [CrossRef] [PubMed]
  13. Joseph, G.B.; McCulloch, C.E.; Nevitt, M.C.; Lynch, J.; Lane, N.E.; Link, T.M. Effects of Weight Change on Knee and Hip Radiographic Measurements and Pain Over Four Years: Data From the Osteoarthritis Initiative. Arthritis Care Res. 2022. [Google Scholar] [CrossRef] [PubMed]
  14. Collins, K.H.; Lenz, K.L.; Pollitt, E.N.; Ferguson, D.; Hutson, I.; Springer, L.E.; Oestreich, A.K.; Tang, R.; Choi, Y.-R.; Meyer, G.A.; et al. Adipose Tissue Is a Critical Regulator of Osteoarthritis. Proc. Natl. Acad. Sci. USA 2021, 118, e2021096118. [Google Scholar] [CrossRef]
  15. Belluzzi, E.; Macchi, V.; Fontanella, C.G.; Carniel, E.L.; Olivotto, E.; Filardo, G.; Sarasin, G.; Porzionato, A.; Granzotto, M.; Pozzuoli, A.; et al. Infrapatellar Fat Pad Gene Expression and Protein Production in Patients with and without Osteoarthritis. Int. J. Mol. Sci. 2020, 21, 6016. [Google Scholar] [CrossRef]
  16. Onuma, H.; Tsuji, K.; Hoshino, T.; Inomata, K.; Udo, M.; Nakagawa, Y.; Katagiri, H.; Miyatake, K.; Watanabe, T.; Sekiya, I.; et al. Fibrotic Changes in the Infrapatellar Fat Pad Induce New Vessel Formation and Sensory Nerve Fiber Endings That Associate Prolonged Pain. J. Orthop. Res. 2020, 38, 1296–1306. [Google Scholar] [CrossRef]
  17. Wu, J.; Xu, J.; Wang, K.; Zhu, Q.; Cai, J.; Ren, J.; Zheng, S.; Ding, C. Associations between Circulating Adipokines and Bone Mineral Density in Patients with Knee Osteoarthritis: A Cross-Sectional Study. BMC Musculoskelet. Disord. 2018, 19, 16. [Google Scholar] [CrossRef] [Green Version]
  18. Cannata, F.; Vadalà, G.; Ambrosio, L.; Napoli, N.; Papalia, R.; Denaro, V.; Pozzilli, P. Osteoarthritis and Type 2 Diabetes: From Pathogenetic Factors to Therapeutic Intervention. Diabetes Metab. Res. Rev. 2020, 36, e3254. [Google Scholar] [CrossRef]
  19. Vincent, T.L. Mechanoflammation in Osteoarthritis Pathogenesis. Semin. Arthritis Rheum. 2019, 49, S36–S38. [Google Scholar] [CrossRef]
  20. Whittaker, J.L.; Roos, E.M. A Pragmatic Approach to Prevent Post-Traumatic Osteoarthritis after Sport or Exercise-Related Joint Injury. Best Pract. Res. Clin. Rheumatol. 2019, 33, 158–171. [Google Scholar] [CrossRef]
  21. Rice, S.J.; Beier, F.; Young, D.A.; Loughlin, J. Interplay between Genetics and Epigenetics in Osteoarthritis. Nat. Rev. Rheumatol. 2020, 16, 268–281. [Google Scholar] [CrossRef] [PubMed]
  22. Grässel, S.; Muschter, D. Recent Advances in the Treatment of Osteoarthritis. F1000Res 2020, 9, 325. [Google Scholar] [CrossRef] [PubMed]
  23. Goldring, S.R.; Goldring, M.B. Changes in the Osteochondral Unit during Osteoarthritis: Structure, Function and Cartilage–Bone Crosstalk. Nat. Rev. Rheumatol. 2016, 12, 632–644. [Google Scholar] [CrossRef]
  24. Badlani, J.T.; Borrero, C.; Golla, S.; Harner, C.D.; Irrgang, J.J. The Effects of Meniscus Injury on the Development of Knee Osteoarthritis: Data From the Osteoarthritis Initiative. Am. J. Sport. Med. 2013, 41, 1238–1244. [Google Scholar] [CrossRef] [PubMed]
  25. Olivotto, E.; Trisolino, G.; Belluzzi, E.; Lazzaro, A.; Strazzari, A.; Pozzuoli, A.; Cigolotti, A.; Ruggieri, P.; Evangelista, A.; Ometto, F.; et al. Macroscopic Synovial Inflammation Correlates with Symptoms and Cartilage Lesions in Patients Undergoing Arthroscopic Partial Meniscectomy: A Clinical Study. J. Clin. Med. 2022, 11, 4330. [Google Scholar] [CrossRef] [PubMed]
  26. Tarasovs, M.; Skuja, S.; Semenistaja, S.; Murovska, M.; Groma, V. Synovitis in Osteoarthritic Patients: Morphological and Virological Evidence of Its Contribution to Development of the Disease. Proc. Latv. Acad. Sci. Sect. B Nat. Exact Appl. Sci. 2019, 73, 317–324. [Google Scholar] [CrossRef] [Green Version]
  27. Groma, V.; Tarasovs, M.; Skuja, S.; Semenistaja, S.; Nora-Krukle, Z.; Svirskis, S.; Murovska, M. Inflammatory Cytokine-Producing Cells and Inflammation Markers in the Synovium of Osteoarthritis Patients Evidenced in Human Herpesvirus 7 Infection. Int. J. Mol. Sci. 2020, 21, 6004. [Google Scholar] [CrossRef]
  28. Masoumi, M.; Bashiri, H.; Khorramdelazad, H.; Barzaman, K.; Hashemi, N.; Sereshki, H.A.; Sahebkar, A.; Karami, J. Destructive Roles of Fibroblast-like Synoviocytes in Chronic Inflammation and Joint Damage in Rheumatoid Arthritis. Inflammation 2021, 44, 466–479. [Google Scholar] [CrossRef]
  29. Bijlsma, J.W.J.; Berenbaum, F.; Lafeber, F.P.J.G. Osteoarthritis: An Update with Relevance for Clinical Practice. Lancet 2011, 377, 2115–2126. [Google Scholar] [CrossRef]
  30. De Lange-Brokaar, B.J.E.; Ioan-Facsinay, A.; van Osch, G.J.V.M.; Zuurmond, A.-M.; Schoones, J.; Toes, R.E.M.; Huizinga, T.W.J.; Kloppenburg, M. Synovial Inflammation, Immune Cells and Their Cytokines in Osteoarthritis: A Review. Osteoarthr. Cartil. 2012, 20, 1484–1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Blüml, S.; Redlich, K.; Smolen, J.S. Mechanisms of Tissue Damage in Arthritis. Semin. Immunopathol. 2014, 36, 531–540. [Google Scholar] [CrossRef] [PubMed]
  32. Rai, M.F.; Cheverud, J.M.; Schmidt, E.J.; Sandell, L.J. Genetic Correlations between Cartilage Regeneration and Degeneration Reveal an Inverse Relationship. Osteoarthr. Cartil. 2020, 28, 1111–1120. [Google Scholar] [CrossRef]
  33. Chou, C.-H.; Jain, V.; Gibson, J.; Attarian, D.E.; Haraden, C.A.; Yohn, C.B.; Laberge, R.-M.; Gregory, S.; Kraus, V.B. Synovial Cell Cross-Talk with Cartilage Plays a Major Role in the Pathogenesis of Osteoarthritis. Sci. Rep. 2020, 10, 10868. [Google Scholar] [CrossRef] [PubMed]
  34. Rahmati, M.; Mobasheri, A.; Mozafari, M. Inflammatory Mediators in Osteoarthritis: A Critical Review of the State-of-the-Art, Current Prospects, and Future Challenges. Bone 2016, 85, 81–90. [Google Scholar] [CrossRef] [PubMed]
  35. Siebuhr, A.S.; Bay-Jensen, A.C.; Jordan, J.M.; Kjelgaard-Petersen, C.F.; Christiansen, C.; Abramson, S.B.; Attur, M.; Berenbaum, F.; Kraus, V.; Karsdal, M.A. Inflammation (or Synovitis)-Driven Osteoarthritis: An Opportunity for Personalizing Prognosis and Treatment? Scand. J. Rheumatol. 2016, 45, 87–98. [Google Scholar] [CrossRef] [PubMed]
  36. Griffin, T.M.; Scanzello, C.R. Innate Inflammation and Synovial Macrophages in Osteoarthritis Pathophysiology. Clin. Exp. Rheumatol. 2019, 37 (Suppl. 120), 57–63. [Google Scholar]
  37. Frevert, C.W.; Felgenhauer, J.; Wygrecka, M.; Nastase, M.V.; Schaefer, L. Danger-Associated Molecular Patterns Derived From the Extracellular Matrix Provide Temporal Control of Innate Immunity. J. Histochem. Cytochem. 2018, 66, 213–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Lambert, C.; Zappia, J.; Sanchez, C.; Florin, A.; Dubuc, J.-E.; Henrotin, Y. The Damage-Associated Molecular Patterns (DAMPs) as Potential Targets to Treat Osteoarthritis: Perspectives From a Review of the Literature. Front. Med. 2021, 7, 607186. [Google Scholar] [CrossRef]
  39. Hamasaki, M.; Terkawi, M.A.; Onodera, T.; Tian, Y.; Ebata, T.; Matsumae, G.; Alhasan, H.; Takahashi, D.; Iwasaki, N. Transcriptional Profiling of Murine Macrophages Stimulated with Cartilage Fragments Revealed a Strategy for Treatment of Progressive Osteoarthritis. Sci. Rep. 2020, 10, 7558. [Google Scholar] [CrossRef]
  40. Berenbaum, F. Osteoarthritis as an Inflammatory Disease (Osteoarthritis Is Not Osteoarthrosis!). Osteoarthr. Cartil. 2013, 21, 16–21. [Google Scholar] [CrossRef] [Green Version]
  41. Li, Y.-S.; Luo, W.; Zhu, S.-A.; Lei, G.-H. T Cells in Osteoarthritis: Alterations and Beyond. Front. Immunol. 2017, 8, 356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Benito, M.J. Synovial Tissue Inflammation in Early and Late Osteoarthritis. Ann. Rheum. Dis. 2005, 64, 1263–1267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Rosshirt, N.; Trauth, R.; Platzer, H.; Tripel, E.; Nees, T.A.; Lorenz, H.-M.; Tretter, T.; Moradi, B. Proinflammatory T Cell Polarization Is Already Present in Patients with Early Knee Osteoarthritis. Arthritis Res. Ther. 2021, 23, 37. [Google Scholar] [CrossRef] [PubMed]
  44. Haynes, M.K.; Hume, E.L.; Smith, J.B. Phenotypic Characterization of Inflammatory Cells from Osteoarthritic Synovium and Synovial Fluids. Clin. Immunol. 2002, 105, 315–325. [Google Scholar] [CrossRef] [PubMed]
  45. Martel-Pelletier, J.; Barr, A.J.; Cicuttini, F.M.; Conaghan, P.G.; Cooper, C.; Goldring, M.B.; Goldring, S.R.; Jones, G.; Teichtahl, A.J.; Pelletier, J.-P. Osteoarthritis. Nat. Rev. Dis. Prim. 2016, 2, 16072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Loeser, R.F.; Goldring, S.R.; Scanzello, C.R.; Goldring, M.B. Osteoarthritis: A Disease of the Joint as an Organ. Arthritis Rheum. 2012, 64, 1697–1707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Smith, M.D. The Normal Synovium. Open Rheumatol. J. 2011, 5, 100–106. [Google Scholar] [CrossRef]
  48. Hügle, T.; Geurts, J. What Drives Osteoarthritis?—Synovial versus Subchondral Bone Pathology. Rheumatology 2017, 56, 1461–1471. [Google Scholar] [CrossRef] [Green Version]
  49. Kulkarni, P.; Martson, A.; Vidya, R.; Chitnavis, S.; Harsulkar, A. Pathophysiological Landscape of Osteoarthritis. Adv. Clin. Chem. 2021, 100, 37–90. [Google Scholar] [CrossRef]
  50. Culemann, S.; Grüneboom, A.; Nicolás-Ávila, J.Á.; Weidner, D.; Lämmle, K.F.; Rothe, T.; Quintana, J.A.; Kirchner, P.; Krljanac, B.; Eberhardt, M.; et al. Locally Renewing Resident Synovial Macrophages Provide a Protective Barrier for the Joint. Nature 2019, 572, 670–675. [Google Scholar] [CrossRef]
  51. Carpintero-Fernandez, P.; Gago-Fuentes, R.; Wang, H.Z.; Fonseca, E.; Caeiro, J.R.; Valiunas, V.; Brink, P.R.; Mayan, M.D. Intercellular Communication via Gap Junction Channels between Chondrocytes and Bone Cells. Biochim. Et Biophys. Acta (BBA)-Biomembr. 2018, 1860, 2499–2505. [Google Scholar] [CrossRef] [PubMed]
  52. Marsh, L.; Kemble, S.; Reis Nisa, P.; Singh, R.; Croft, A.P. Fibroblast Pathology in Inflammatory Joint Disease. Immunol. Rev. 2021, 302, 163–183. [Google Scholar] [CrossRef]
  53. Bartok, B.; Firestein, G.S. Fibroblast-like Synoviocytes: Key Effector Cells in Rheumatoid Arthritis. Immunol. Rev. 2010, 233, 233–255. [Google Scholar] [CrossRef] [PubMed]
  54. Krenn, V.; Morawietz, L.; Burmester, G.-R.; Kinne, R.W.; Mueller-Ladner, U.; Muller, B.; Haupl, T. Synovitis Score: Discrimination between Chronic Low-Grade and High-Grade Synovitis. Histopathology 2006, 49, 358–364. [Google Scholar] [CrossRef] [PubMed]
  55. Alivernini, S.; MacDonald, L.; Elmesmari, A.; Finlay, S.; Tolusso, B.; Gigante, M.R.; Petricca, L.; Di Mario, C.; Bui, L.; Perniola, S.; et al. Distinct Synovial Tissue Macrophage Subsets Regulate Inflammation and Remission in Rheumatoid Arthritis. Nat. Med. 2020, 26, 1295–1306. [Google Scholar] [CrossRef] [PubMed]
  56. Hannemann, N.; Apparailly, F.; Courties, G. Synovial Macrophages: From Ordinary Eaters to Extraordinary Multitaskers. Trends Immunol. 2021, 42, 368–371. [Google Scholar] [CrossRef]
  57. Thomson, A.; Hilkens, C.M.U. Synovial Macrophages in Osteoarthritis: The Key to Understanding Pathogenesis? Front. Immunol. 2021, 12, 678757. [Google Scholar] [CrossRef]
  58. Knab, K.; Chambers, D.; Krönke, G. Synovial Macrophage and Fibroblast Heterogeneity in Joint Homeostasis and Inflammation. Front. Med. 2022, 9, 862161. [Google Scholar] [CrossRef]
  59. 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. [Google Scholar] [CrossRef] [PubMed]
  60. Buechler, M.B.; Pradhan, R.N.; Krishnamurty, A.T.; Cox, C.; Calviello, A.K.; Wang, A.W.; Yang, Y.A.; Tam, L.; Caothien, R.; Roose-Girma, M.; et al. Cross-Tissue Organization of the Fibroblast Lineage. Nature 2021, 593, 575–579. [Google Scholar] [CrossRef]
  61. Guilak, F.; Nims, R.J.; Dicks, A.; Wu, C.-L.; Meulenbelt, I. Osteoarthritis as a Disease of the Cartilage Pericellular Matrix. Matrix Biol. 2018, 71–72, 40–50. [Google Scholar] [CrossRef] [PubMed]
  62. Singh, J.A.; Arayssi, T.; Duray, P.; Schumacher, H.R. Immunohistochemistry of Normal Human Knee Synovium: A Quantitative Study. Ann. Rheum. Dis. 2004, 63, 785–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Muntz, I.; Fenu, M.; van Osch, G.J.V.M.; Koenderink, G.H. The Role of Cell–Matrix Interactions in Connective Tissue Mechanics. Phys. Biol. 2022, 19, 021001. [Google Scholar] [CrossRef] [PubMed]
  64. Charlier, E.; Deroyer, C.; Ciregia, F.; Malaise, O.; Neuville, S.; Plener, Z.; Malaise, M.; de Seny, D. Chondrocyte Dedifferentiation and Osteoarthritis (OA). Biochem. Pharmacol. 2019, 165, 49–65. [Google Scholar] [CrossRef]
  65. Primorac, D.; Molnar, V.; Rod, E.; Jeleč, Ž.; Čukelj, F.; Matišić, V.; Vrdoljak, T.; Hudetz, D.; Hajsok, H.; Borić, I. Knee Osteoarthritis: A Review of Pathogenesis and State-Of-The-Art Non-Operative Therapeutic Considerations. Genes 2020, 11, 854. [Google Scholar] [CrossRef]
  66. Wang, Y.; Wei, L.; Zeng, L.; He, D.; Wei, X. Nutrition and Degeneration of Articular Cartilage. Knee Surg. Sport. Traumatol. Arthrosc. 2013, 21, 1751–1762. [Google Scholar] [CrossRef] [Green Version]
  67. Luo, Y.; Sinkeviciute, D.; He, Y.; Karsdal, M.; Henrotin, Y.; Mobasheri, A.; Önnerfjord, P.; Bay-Jensen, A. The Minor Collagens in Articular Cartilage. Protein Cell 2017, 8, 560–572. [Google Scholar] [CrossRef] [Green Version]
  68. Hall, A.C. The Role of Chondrocyte Morphology and Volume in Controlling Phenotype—Implications for Osteoarthritis, Cartilage Repair, and Cartilage Engineering. Curr. Rheumatol. Rep. 2019, 21, 38. [Google Scholar] [CrossRef] [Green Version]
  69. Wang, X.; Ning, Y.; Zhang, P.; Poulet, B.; Huang, R.; Gong, Y.; Hu, M.; Li, C.; Zhou, R.; Lammi, M.J.; et al. Comparison of the Major Cell Populations among Osteoarthritis, Kashin–Beck Disease and Healthy Chondrocytes by Single-Cell RNA-Seq Analysis. Cell Death Dis. 2021, 12, 551. [Google Scholar] [CrossRef]
  70. Basso, P.R.; Carava’, E.; Protasoni, M.; Reguzzoni, M.; Raspanti, M. The Synovial Surface of the Articular Cartilage. Eur. J. Histochem. 2020, 64, 3146. [Google Scholar] [CrossRef]
  71. Pan, J.; Wang, B.; Li, W.; Zhou, X.; Scherr, T.; Yang, Y.; Price, C.; Wang, L. Elevated Cross-Talk between Subchondral Bone and Cartilage in Osteoarthritic Joints. Bone 2012, 51, 212–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zecca, P.A.; Reguzzoni, M.; Protasoni, M.; Raspanti, M. The Chondro-Osseous Junction Of Articular Cartilage. Tissue Cell 2022, 80, 101993. [Google Scholar] [CrossRef] [PubMed]
  73. Lyons, T.J.; Stoddart, R.W.; McClure, S.F.; McClure, J. The Tidemark of the Chondro-Osseous Junction of the Normal Human Knee Joint. J. Mol. Hist. 2005, 36, 207–215. [Google Scholar] [CrossRef]
  74. Boyde, A. The Bone Cartilage Interface and Osteoarthritis. Calcif. Tissue Int. 2021, 109, 303–328. [Google Scholar] [CrossRef] [PubMed]
  75. Akkiraju, H.; Nohe, A. Role of Chondrocytes in Cartilage Formation, Progression of Osteoarthritis and Cartilage Regeneration. J. Dev. Biol. 2015, 3, 177–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chery, D.R.; Han, B.; Zhou, Y.; Wang, C.; Adams, S.M.; Chandrasekaran, P.; Kwok, B.; Heo, S.-J.; Enomoto-Iwamoto, M.; Lu, X.L.; et al. Decorin Regulates Cartilage Pericellular Matrix Micromechanobiology. Matrix Biol. 2021, 96, 1–17. [Google Scholar] [CrossRef] [PubMed]
  77. Peng, Z.; Sun, H.; Bunpetch, V.; Koh, Y.; Wen, Y.; Wu, D.; Ouyang, H. The Regulation of Cartilage Extracellular Matrix Homeostasis in Joint Cartilage Degeneration and Regeneration. Biomaterials 2021, 268, 120555. [Google Scholar] [CrossRef]
  78. Brandt, K.D. Yet More Evidence That Osteoarthritis Is Not a Cartilage Disease. Ann. Rheum. Dis. 2006, 65, 1261–1264. [Google Scholar] [CrossRef] [Green Version]
  79. Danalache, M.; Kleinert, R.; Schneider, J.; Erler, A.L.; Schwitalle, M.; Riester, R.; Traub, F.; Hofmann, U.K. Changes in Stiffness and Biochemical Composition of the Pericellular Matrix as a Function of Spatial Chondrocyte Organisation in Osteoarthritic Cartilage. Osteoarthr. Cartil. 2019, 27, 823–832. [Google Scholar] [CrossRef]
  80. Wang, C.; Brisson, B.K.; Terajima, M.; Li, Q.; Hoxha, K.; Han, B.; Goldberg, A.M.; Sherry Liu, X.; Marcolongo, M.S.; Enomoto-Iwamoto, M.; et al. Type III Collagen Is a Key Regulator of the Collagen Fibrillar Structure and Biomechanics of Articular Cartilage and Meniscus. Matrix Biol. 2020, 85–86, 47–67. [Google Scholar] [CrossRef]
  81. Eyre, D. Articular Cartilage and Changes in Arthritis: Collagen of Articular Cartilage. Arthritis Res. Ther. 2001, 4, 30. [Google Scholar] [CrossRef] [Green Version]
  82. Sandell, L.J.; Aigner, T. Articular Cartilage and Changes in Arthritis. An Introduction: Cell Biology of Osteoarthritis. Arthritis Res. Ther. 2001, 3, 107–113. [Google Scholar] [CrossRef] [Green Version]
  83. Pullig, O.; Weseloh, G.; Klatt, A.R.; Wagener, R.; Swoboda, B. Matrilin-3 in Human Articular Cartilage: Increased Expression in Osteoarthritis. Osteoarthr. Cartil. 2002, 10, 253–263. [Google Scholar] [CrossRef] [Green Version]
  84. Roseti, L.; Desando, G.; Cavallo, C.; Petretta, M.; Grigolo, B. Articular Cartilage Regeneration in Osteoarthritis. Cells 2019, 8, 1305. [Google Scholar] [CrossRef] [Green Version]
  85. Li, G.; Yin, J.; Gao, J.; Cheng, T.S.; Pavlos, N.J.; Zhang, C.; Zheng, M.H. Subchondral Bone in Osteoarthritis: Insight into Risk Factors and Microstructural Changes. Arthritis Res. Ther. 2013, 15, 223. [Google Scholar] [CrossRef] [Green Version]
  86. Pan, J.; Zhou, X.; Li, W.; Novotny, J.E.; Doty, S.B.; Wang, L. In Situ Measurement of Transport between Subchondral Bone and Articular Cartilage: TRANSPORT BETWEEN BONE AND CARTILAGE. J. Orthop. Res. 2009, 27, 1347–1352. [Google Scholar] [CrossRef] [Green Version]
  87. Mountcastle, S.E.; Allen, P.; Mellors, B.O.L.; Lawless, B.M.; Cooke, M.E.; Lavecchia, C.E.; Fell, N.L.A.; Espino, D.M.; Jones, S.W.; Cox, S.C. Dynamic Viscoelastic Characterisation of Human Osteochondral Tissue: Understanding the Effect of the Cartilage-Bone Interface. BMC Musculoskelet. Disord. 2019, 20, 575. [Google Scholar] [CrossRef]
  88. Hu, W.; Chen, Y.; Dou, C.; Dong, S. Microenvironment in Subchondral Bone: Predominant Regulator for the Treatment of Osteoarthritis. Ann. Rheum. Dis. 2021, 80, 413–422. [Google Scholar] [CrossRef]
  89. Burr, D.B.; Gallant, M.A. Bone Remodelling in Osteoarthritis. Nat. Rev. Rheumatol. 2012, 8, 665–673. [Google Scholar] [CrossRef]
  90. Donell, S. Subchondral Bone Remodelling in Osteoarthritis. EFORT Open Rev. 2019, 4, 221–229. [Google Scholar] [CrossRef]
  91. Zhao, X.; Ma, L.; Guo, H.; Wang, J.; Zhang, S.; Yang, X.; Yang, L.; Jin, Q. Osteoclasts Secrete Leukemia Inhibitory Factor to Promote Abnormal Bone Remodeling of Subchondral Bone in Osteoarthritis. BMC Musculoskelet. Disord. 2022, 23, 87. [Google Scholar] [CrossRef]
  92. Sharma, A.; Jagga, S.; Lee, S.-S.; Nam, J.-S. Interplay between Cartilage and Subchondral Bone Contributing to Pathogenesis of Osteoarthritis. Int. J. Mol. Sci. 2013, 14, 19805–19830. [Google Scholar] [CrossRef] [Green Version]
  93. Lajeunesse, D.; Reboul, P. Subchondral Bone in Osteoarthritis: A Biologic Link with Articular Cartilage Leading to Abnormal Remodeling. Curr. Opin. Rheumatol. 2003, 15, 628–633. [Google Scholar] [CrossRef]
  94. Wu, X.; Crawford, R.; Xiao, Y.; Mao, X.; Prasadam, I. Osteoarthritic Subchondral Bone Release Exosomes That Promote Cartilage Degeneration. Cells 2021, 10, 251. [Google Scholar] [CrossRef]
  95. Buenzli, P.R.; Sims, N.A. Quantifying the Osteocyte Network in the Human Skeleton. Bone 2015, 75, 144–150. [Google Scholar] [CrossRef]
  96. Schurman, C.A.; Verbruggen, S.W.; Alliston, T. Disrupted Osteocyte Connectivity and Pericellular Fluid Flow in Bone with Aging and Defective TGF-β Signaling. Proc. Natl. Acad. Sci. USA 2021, 118, e2023999118. [Google Scholar] [CrossRef]
  97. Zhang, L.; Wen, C. Osteocyte Dysfunction in Joint Homeostasis and Osteoarthritis. Int. J. Mol. Sci. 2021, 22, 6522. [Google Scholar] [CrossRef]
  98. Morrell, A.E.; Brown, G.N.; Robinson, S.T.; Sattler, R.L.; Baik, A.D.; Zhen, G.; Cao, X.; Bonewald, L.F.; Jin, W.; Kam, L.C.; et al. Mechanically Induced Ca2+ Oscillations in Osteocytes Release Extracellular Vesicles and Enhance Bone Formation. Bone Res. 2018, 6, 6. [Google Scholar] [CrossRef] [Green Version]
  99. Chen, J.; Tu, X.; Esen, E.; Joeng, K.S.; Lin, C.; Arbeit, J.M.; Rüegg, M.A.; Hall, M.N.; Ma, L.; Long, F. WNT7B Promotes Bone Formation in Part through MTORC1. PLoS Genet. 2014, 10, e1004145. [Google Scholar] [CrossRef] [Green Version]
  100. Song, D.; He, G.; Song, F.; Wang, Z.; Liu, X.; Liao, L.; Ni, J.; Silva, M.J.; Long, F. Inducible Expression of Wnt7b Promotes Bone Formation in Aged Mice and Enhances Fracture Healing. Bone Res. 2020, 8, 4. [Google Scholar] [CrossRef] [Green Version]
  101. Robling, A.G.; Niziolek, P.J.; Baldridge, L.A.; Condon, K.W.; Allen, M.R.; Alam, I.; Mantila, S.M.; Gluhak-Heinrich, J.; Bellido, T.M.; Harris, S.E.; et al. Mechanical Stimulation of Bone in Vivo Reduces Osteocyte Expression of Sost/Sclerostin. J. Biol. Chem. 2008, 283, 5866–5875. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Gaudio, A.; Pennisi, P.; Bratengeier, C.; Torrisi, V.; Lindner, B.; Mangiafico, R.A.; Pulvirenti, I.; Hawa, G.; Tringali, G.; Fiore, C.E. Increased Sclerostin Serum Levels Associated with Bone Formation and Resorption Markers in Patients with Immobilization-Induced Bone Loss. J. Clin. Endocrinol. Metab. 2010, 95, 2248–2253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Yee, C.S.; Manilay, J.O.; Chang, J.C.; Hum, N.R.; Murugesh, D.K.; Bajwa, J.; Mendez, M.E.; Economides, A.E.; Horan, D.J.; Robling, A.G.; et al. Conditional Deletion of Sost in MSC-Derived Lineages Identifies Specific Cell-Type Contributions to Bone Mass and B-Cell Development. J. Bone Min. Res. 2018, 33, 1748–1759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Van Bezooijen, R.L.; Roelen, B.A.J.; Visser, A.; van der Wee-Pals, L.; de Wilt, E.; Karperien, M.; Hamersma, H.; Papapoulos, S.E.; ten Dijke, P.; Löwik, C.W.G.M. Sclerostin Is an Osteocyte-Expressed Negative Regulator of Bone Formation, But Not a Classical BMP Antagonist. J. Exp. Med. 2004, 199, 805–814. [Google Scholar] [CrossRef] [PubMed]
  105. Glass, D.A.; Bialek, P.; Ahn, J.D.; Starbuck, M.; Patel, M.S.; Clevers, H.; Taketo, M.M.; Long, F.; McMahon, A.P.; Lang, R.A.; et al. Canonical Wnt Signaling in Differentiated Osteoblasts Controls Osteoclast Differentiation. Dev. Cell 2005, 8, 751–764. [Google Scholar] [CrossRef] [Green Version]
  106. Zimmerman, S.M.; Heard-Lipsmeyer, M.E.; Dimori, M.; Thostenson, J.D.; Mannen, E.M.; O’Brien, C.A.; Morello, R. Loss of RANKL in Osteocytes Dramatically Increases Cancellous Bone Mass in the Osteogenesis Imperfecta Mouse (Oim). Bone Rep. 2018, 9, 61–73. [Google Scholar] [CrossRef]
  107. Udagawa, N.; Koide, M.; Nakamura, M.; Nakamichi, Y.; Yamashita, T.; Uehara, S.; Kobayashi, Y.; Furuya, Y.; Yasuda, H.; Fukuda, C.; et al. Osteoclast Differentiation by RANKL and OPG Signaling Pathways. J. Bone Min. Metab. 2021, 39, 19–26. [Google Scholar] [CrossRef]
  108. Pellicore, M.J.; Gangi, L.R.; Murphy, L.A.; Lee, A.J.; Jacobsen, T.; Kenawy, H.M.; Shah, R.P.; Chahine, N.O.; Ateshian, G.A.; Hung, C.T. Toward Defining the Role of the Synovium in Mitigating Normal Articular Cartilage Wear and Tear. J. Biomech. 2023, 148, 111472. [Google Scholar] [CrossRef]
  109. Dy, P.; Wang, W.; Bhattaram, P.; Wang, Q.; Wang, L.; Ballock, R.T.; Lefebvre, V. Sox9 Directs Hypertrophic Maturation and Blocks Osteoblast Differentiation of Growth Plate Chondrocytes. Dev. Cell 2012, 22, 597–609. [Google Scholar] [CrossRef] [Green Version]
  110. Ouyang, Y.; Wang, W.; Tu, B.; Zhu, Y.; Fan, C.; Li, Y. Overexpression of SOX9 Alleviates the Progression of Human Osteoarthritis in Vitro and in Vivo. Drug Des. Devel. 2019, 13, 2833–2842. [Google Scholar] [CrossRef] [Green Version]
  111. Hossain, M.A.; Adithan, A.; Alam, M.J.; Kopalli, S.R.; Kim, B.; Kang, C.-W.; Hwang, K.-C.; Kim, J.-H. IGF-1 Facilitates Cartilage Reconstruction by Regulating PI3K/AKT, MAPK, and NF-KB Signaling in Rabbit Osteoarthritis. J. Inflamm. Res. 2021, 14, 3555–3568. [Google Scholar] [CrossRef] [PubMed]
  112. Wen, C.; Xu, L.; Xu, X.; Wang, D.; Liang, Y.; Duan, L. Insulin-like Growth Factor-1 in Articular Cartilage Repair for Osteoarthritis Treatment. Arthritis Res. Ther. 2021, 23, 277. [Google Scholar] [CrossRef] [PubMed]
  113. Cherifi, C.; Monteagudo, S.; Lories, R.J. Promising Targets for Therapy of Osteoarthritis: A Review on the Wnt and TGF-β Signalling Pathways. Ther. Adv. Musculoskelet. 2021, 13, 1759720X211006959. [Google Scholar] [CrossRef] [PubMed]
  114. Hunter, D.J.; Bierma-Zeinstra, S. Osteoarthritis. Lancet 2019, 393, 1745–1759. [Google Scholar] [CrossRef]
  115. Maldonado, M.; Nam, J. The Role of Changes in Extracellular Matrix of Cartilage in the Presence of Inflammation on the Pathology of Osteoarthritis. BioMed Res. Int. 2013, 2013, 284873. [Google Scholar] [CrossRef] [Green Version]
  116. Berenbaum, F.; Walker, C. Osteoarthritis and Inflammation: A Serious Disease with Overlapping Phenotypic Patterns. Postgrad. Med. 2020, 132, 377–384. [Google Scholar] [CrossRef]
  117. Robinson, W.H.; Lepus, C.M.; Wang, Q.; Raghu, H.; Mao, R.; Lindstrom, T.M.; Sokolove, J. Low-Grade Inflammation as a Key Mediator of the Pathogenesis of Osteoarthritis. Nat. Rev. Rheumatol. 2016, 12, 580–592. [Google Scholar] [CrossRef]
  118. Zhu, W.; Zhang, X.; Jiang, Y.; Liu, X.; Huang, L.; Wei, Q.; Huang, Y.; Wu, W.; Gu, J. Alterations in Peripheral T Cell and B Cell Subsets in Patients with Osteoarthritis. Clin. Rheumatol. 2020, 39, 523–532. [Google Scholar] [CrossRef]
  119. Hayashi, D.; Roemer, F.W.; Katur, A.; Felson, D.T.; Yang, S.-O.; Alomran, F.; Guermazi, A. Imaging of Synovitis in Osteoarthritis: Current Status and Outlook. Semin. Arthritis Rheum. 2011, 41, 116–130. [Google Scholar] [CrossRef]
  120. Atukorala, I.; Kwoh, C.K.; Guermazi, A.; Roemer, F.W.; Boudreau, R.M.; Hannon, M.J.; Hunter, D.J. Synovitis in Knee Osteoarthritis: A Precursor of Disease? Ann. Rheum. Dis. 2016, 75, 390–395. [Google Scholar] [CrossRef]
  121. Boffa, A.; Merli, G.; Andriolo, L.; Lattermann, C.; Salzmann, G.M.; Filardo, G. Synovial Fluid Biomarkers in Knee Osteoarthritis: A Systematic Review and Quantitative Evaluation Using BIPEDs Criteria. Cartilage 2021, 13, 82S–103S. [Google Scholar] [CrossRef] [PubMed]
  122. Manferdini, C.; Paolella, F.; Gabusi, E.; Silvestri, Y.; Gambari, L.; Cattini, L.; Filardo, G.; Fleury-Cappellesso, S.; Lisignoli, G. From Osteoarthritic Synovium to Synovial-Derived Cells Characterization: Synovial Macrophages Are Key Effector Cells. Arthritis Res. Ther. 2016, 18, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Thoenen, J.; MacKay, J.W.; Sandford, H.J.C.; Gold, G.E.; Kogan, F. Imaging of Synovial Inflammation in Osteoarthritis, From the AJR Special Series on Inflammation. AJR Am. J. Roentgenol. 2022, 218, 405–417. [Google Scholar] [CrossRef]
  124. Baker, K.; Grainger, A.; Niu, J.; Clancy, M.; Guermazi, A.; Crema, M.; Hughes, L.; Buckwalter, J.; Wooley, A.; Nevitt, M.; et al. Relation of Synovitis to Knee Pain Using Contrast-Enhanced MRIs. Ann. Rheum. Dis. 2010, 69, 1779–1783. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Roemer, F.W.; Guermazi, A.; Felson, D.T.; Niu, J.; Nevitt, M.C.; Crema, M.D.; Lynch, J.A.; Lewis, C.E.; Torner, J.; Zhang, Y. Presence of MRI-Detected Joint Effusion and Synovitis Increases the Risk of Cartilage Loss in Knees without Osteoarthritis at 30-Month Follow-up: The MOST Study. Ann. Rheum. Dis. 2011, 70, 1804–1809. [Google Scholar] [CrossRef]
  126. Wang, X.; Hunter, D.J.; Jin, X.; Ding, C. The Importance of Synovial Inflammation in Osteoarthritis: Current Evidence from Imaging Assessments and Clinical Trials. Osteoarthr. Cartil. 2018, 26, 165–174. [Google Scholar] [CrossRef] [Green Version]
  127. Felson, D.T.; Niu, J.; Neogi, T.; Goggins, J.; Nevitt, M.C.; Roemer, F.; Torner, J.; Lewis, C.E.; Guermazi, A. Synovitis and the Risk of Knee Osteoarthritis: The MOST Study. Osteoarthr. Cartil. 2016, 24, 458–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ayral, X.; Pickering, E.H.; Woodworth, T.G.; Mackillop, N.; Dougados, M. Synovitis: A Potential Predictive Factor of Structural Progression of Medial Tibiofemoral Knee Osteoarthritis-Results of a 1 Year Longitudinal Arthroscopic Study in 422 Patients. Osteoarthr. Cartil. 2005, 13, 361–367. [Google Scholar] [CrossRef] [Green Version]
  129. Riis, R.G.C.; Gudbergsen, H.; Simonsen, O.; Henriksen, M.; Al-Mashkur, N.; Eld, M.; Petersen, K.K.; Kubassova, O.; Bay Jensen, A.C.; Damm, J.; et al. The Association between Histological, Macroscopic and Magnetic Resonance Imaging Assessed Synovitis in End-Stage Knee Osteoarthritis: A Cross-Sectional Study. Osteoarthr. Cartil. 2017, 25, 272–280. [Google Scholar] [CrossRef] [Green Version]
  130. Abbasi, B.; Pezeshki-Rad, M.; Akhavan, R.; Sahebari, M. Association between Clinical and Sonographic Synovitis in Patients with Painful Knee Osteoarthritis. Int. J. Rheum. Dis. 2017, 20, 561–566. [Google Scholar] [CrossRef]
  131. Zhang, Y.; Nevitt, M.; Niu, J.; Lewis, C.; Torner, J.; Guermazi, A.; Roemer, F.; McCulloch, C.; Felson, D. Fluctuation of Knee Pain and Changes in Bone Marrow Lesions, Effusions and Synovitis on MRI: The MOST Study. Arthritis Rheum. 2011, 63, 573–862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Bacon, K.; LaValley, M.P.; Jafarzadeh, S.R.; Felson, D. Does Cartilage Loss Cause Pain in Osteoarthritis and If so, How Much? Ann. Rheum. Dis. 2020, 79, 1105–1110. [Google Scholar] [CrossRef] [PubMed]
  133. Zhao, Q.; Lin, L.; Guo, Y.; Zou, R.; Wang, Z.; Shi, Z.; Lin, F. Matrix Metalloproteinase-13, NF-κB P65 and Interleukin-1β Are Associated with the Severity of Knee Osteoarthritis. Exp. Ther. Med. 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Stannus, O.; Jones, G.; Cicuttini, F.; Parameswaran, V.; Quinn, S.; Burgess, J.; Ding, C. Circulating Levels of IL-6 and TNF-α Are Associated with Knee Radiographic Osteoarthritis and Knee Cartilage Loss in Older Adults. Osteoarthr. Cartil. 2010, 18, 1441–1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Hoff, P.; Buttgereit, F.; Burmester, G.-R.; Jakstadt, M.; Gaber, T.; Andreas, K.; Matziolis, G.; Perka, C.; Röhner, E. Osteoarthritis Synovial Fluid Activates Pro-Inflammatory Cytokines in Primary Human Chondrocytes. Int. Orthop. (SICOT) 2013, 37, 145–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Li, Y.; Mai, Y.; Cao, P.; Wen, X.; Fan, T.; Wang, X.; Ruan, G.; Tang, S.; Ding, C.; Zhu, Z. Relative Efficacy and Safety of Anti-Inflammatory Biologic Agents for Osteoarthritis: A Conventional and Network Meta-Analysis. J. Clin. Med. 2022, 11, 3958. [Google Scholar] [CrossRef] [PubMed]
  137. Zhang, L.; Li, M.; Li, X.; Liao, T.; Ma, Z.; Zhang, L.; Xing, R.; Wang, P.; Mao, J. Characteristics of Sensory Innervation in Synovium of Rats within Different Knee Osteoarthritis Models and the Correlation between Synovial Fibrosis and Hyperalgesia. J. Adv. Res. 2022, 35, 141–151. [Google Scholar] [CrossRef] [PubMed]
  138. Nanus, D.E.; Badoume, A.; Wijesinghe, S.N.; Halsey, A.M.; Hurley, P.; Ahmed, Z.; Botchu, R.; Davis, E.T.; Lindsay, M.A.; Jones, S.W. Synovial Tissue from Sites of Joint Pain in Knee Osteoarthritis Patients Exhibits a Differential Phenotype with Distinct Fibroblast Subsets. eBioMedicine 2021, 72, 103618. [Google Scholar] [CrossRef]
  139. Wang, M.; Lessard, S.G.; Singh, P.; Pannellini, T.; Chen, T.; Rourke, B.J.; Chowdhury, L.; Craveiro, V.; Sculco, P.K.; van der Meulen, M.C.H.; et al. Knee Fibrosis Is Associated with the Development of Osteoarthritis in a Murine Model of Tibial Compression. J. Orthop. Res. 2021, 39, 1030–1040. [Google Scholar] [CrossRef]
  140. Favero, M.; El-Hadi, H.; Belluzzi, E.; Granzotto, M.; Porzionato, A.; Sarasin, G.; Rambaldo, A.; Iacobellis, C.; Cigolotti, A.; Fontanella, C.G.; et al. Infrapatellar Fat Pad Features in Osteoarthritis: A Histopathological and Molecular Study. Rheumatology 2017, 56, 1784–1793. [Google Scholar] [CrossRef] [Green Version]
  141. Macchi, V.; Stocco, E.; Stecco, C.; Belluzzi, E.; Favero, M.; Porzionato, A.; De Caro, R. The infrapatellar fat pad and the synovial membrane: An anatomo-functional unit. J. Anat. 2018, 233, 146–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Belluzzi, E.; Stocco, E.; Pozzuoli, A.; Granzotto, M.; Porzionato, A.; Vettor, R.; De Caro, R.; Ruggieri, P.; Ramonda, R.; Rossato, M.; et al. Contribution of Infrapatellar Fat Pad and Synovial Membrane to Knee Osteoarthritis Pain. BioMed Res. Int. 2019, 2019, e6390182. [Google Scholar] [CrossRef] [PubMed]
  143. Emmi, A.; Stocco, E.; Boscolo-Berto, R.; Contran, M.; Belluzzi, E.; Favero, M.; Ramonda, R.; Porzionato, A.; Ruggieri, P.; De Caro, R.; et al. Infrapatellar Fat Pad-Synovial Membrane Anatomo-Fuctional Unit: Microscopic Basis for Piezo1/2 Mechanosensors Involvement in Osteoarthritis Pain. Front. Cell. Dev. Biol. 2022, 10, 886604. [Google Scholar] [CrossRef] [PubMed]
  144. Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 Macrophages and the Th1/Th2 Paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Blom, A.B.; van Lent, P.L.; Libregts, S.; Holthuysen, A.E.; van der Kraan, P.M.; van Rooijen, N.; van den Berg, W.B. Crucial Role of Macrophages in Matrix Metalloproteinase–Mediated Cartilage Destruction during Experimental Osteoarthritis: Involvement of Matrix Metalloproteinase 3. Arthritis Rheum. 2007, 56, 147–157. [Google Scholar] [CrossRef]
  146. Kraus, V.B.; McDaniel, G.; Huebner, J.L.; Stabler, T.V.; Pieper, C.F.; Shipes, S.W.; Petry, N.A.; Low, P.S.; Shen, J.; McNearney, T.A.; et al. Direct in Vivo Evidence of Activated Macrophages in Human Osteoarthritis. Osteoarthr. Cartil. 2016, 24, 1613–1621. [Google Scholar] [CrossRef] [Green Version]
  147. Huo, L.W.; Ye, Y.L.; Wang, G.W.; Ye, Y.G. Fractalkine (CX3CL1): A Biomarker Reflecting Symptomatic Severity in Patients With Knee Osteoarthritis. J. Investig. Med. 2015, 63, 626–631. [Google Scholar] [CrossRef]
  148. Ancuta, P.; Wang, J.; Gabuzda, D. CD16+ Monocytes Produce IL-6, CCL2, and Matrix Metalloproteinase-9 upon Interaction with CX3CL1-Expressing Endothelial Cells. J. Leukoc. Biol. 2006, 80, 1156–1164. [Google Scholar] [CrossRef]
  149. Raghu, H.; Lepus, C.M.; Wang, Q.; Wong, H.H.; Lingampalli, N.; Oliviero, F.; Punzi, L.; Giori, N.J.; Goodman, S.B.; Chu, C.R.; et al. CCL2/CCR2, but Not CCL5/CCR5, Mediates Monocyte Recruitment, Inflammation and Cartilage Destruction in Osteoarthritis. Ann. Rheum. Dis. 2017, 76, 914–922. [Google Scholar] [CrossRef] [Green Version]
  150. Daghestani, H.N.; Pieper, C.F.; Kraus, V.B. Soluble Macrophage Biomarkers Indicate Inflammatory Phenotypes in Patients With Knee Osteoarthritis. Arthritis Rheumatol. 2015, 67, 956–965. [Google Scholar] [CrossRef] [Green Version]
  151. Blom, A.B.; van Lent, P.L.E.M.; Holthuysen, A.E.M.; van der Kraan, P.M.; Roth, J.; van Rooijen, N.; van den Berg, W.B. Synovial Lining Macrophages Mediate Osteophyte Formation during Experimental Osteoarthritis. Osteoarthr. Cartil. 2004, 12, 627–635. [Google Scholar] [CrossRef] [Green Version]
  152. Jamal, J.; Roebuck, M.M.; Lee, S.-Y.; Frostick, S.P.; Abbas, A.A.; Merican, A.M.; Teo, S.-H.; Wood, A.; Tan, S.-L.; Bou-Gharios, G.; et al. Modulation of the Mechanical Responses of Synovial Fibroblasts by Osteoarthritis-Associated Inflammatory Stressors. Int. J. Biochem. Cell Biol. 2020, 126, 105800. [Google Scholar] [CrossRef] [PubMed]
  153. Iqbal, S.M.; Leonard, C.; Regmi, C.S.; De Rantere, D.; Tailor, P.; Ren, G.; Ishida, H.; Hsu, C.y.; Abubacker, S.; Pang, D.S.J.; et al. Lubricin/Proteoglycan 4 Binds to and Regulates the Activity of Toll-Like Receptors In Vitro. Sci. Rep. 2016, 6, 18910. [Google Scholar] [CrossRef] [Green Version]
  154. Chavda, S.; Rabbani, S.A.; Wadhwa, T. Role and Effectiveness of Intra-Articular Injection of Hyaluronic Acid in the Treatment of Knee Osteoarthritis: A Systematic Review. Cureus 2022, 14, e24503. [Google Scholar] [CrossRef]
  155. Avenoso, A.; Bruschetta, G.; D’Ascola, A.; Scuruchi, M.; Mandraffino, G.; Saitta, A.; Campo, S.; Campo, G.M. Hyaluronan Fragmentation During Inflammatory Pathologies: A Signal That Empowers Tissue Damage. Mini Rev. Med. Chem. 2020, 20, 54–65. [Google Scholar] [CrossRef] [PubMed]
  156. Mizoguchi, F.; Slowikowski, K.; Wei, K.; Marshall, J.L.; Rao, D.A.; Chang, S.K.; Nguyen, H.N.; Noss, E.H.; Turner, J.D.; Earp, B.E.; et al. Functionally Distinct Disease-Associated Fibroblast Subsets in Rheumatoid Arthritis. Nat. Commun. 2018, 9, 789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Nygaard, G.; Firestein, G.S. Restoring Synovial Homeostasis in Rheumatoid Arthritis by Targeting Fibroblast-like Synoviocytes. Nat. Rev. Rheumatol. 2020, 16, 316–333. [Google Scholar] [CrossRef] [PubMed]
  158. Ni, S.; Miao, K.; Zhou, X.; Xu, N.; Li, C.; Zhu, R.; Sun, R.; Wang, Y. The Involvement of Follistatin-like Protein 1 in Osteoarthritis by Elevating NF-ΚB-Mediated Inflammatory Cytokines and Enhancing Fibroblast like Synoviocyte Proliferation. Arthritis Res. Ther. 2015, 17, 91. [Google Scholar] [CrossRef] [Green Version]
  159. Qadri, M.; Jay, G.D.; Zhang, L.X.; Richendrfer, H.; Schmidt, T.A.; Elsaid, K.A. Proteoglycan-4 Regulates Fibroblast to Myofibroblast Transition and Expression of Fibrotic Genes in the Synovium. Arthritis Res. Ther. 2020, 22, 113. [Google Scholar] [CrossRef]
  160. Wojdasiewicz, P.; Poniatowski, Ł.A.; Kotela, A.; Deszczyński, J.; Kotela, I.; Szukiewicz, D. The Chemokine CX3CL1 (Fractalkine) and Its Receptor CX3CR1: Occurrence and Potential Role in Osteoarthritis. Arch. Immunol. Et Ther. Exp. (Warsz) 2014, 62, 395–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  161. Hou, S.-M.; Hou, C.-H.; Liu, J.-F. CX3CL1 Promotes MMP-3 Production via the CX3CR1, c-Raf, MEK, ERK, and NF-ΚB Signaling Pathway in Osteoarthritis Synovial Fibroblasts. Arthritis Res. Ther. 2017, 19, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Frank-Bertoncelj, M.; Trenkmann, M.; Klein, K.; Karouzakis, E.; Rehrauer, H.; Bratus, A.; Kolling, C.; Armaka, M.; Filer, A.; Michel, B.A.; et al. Epigenetically-Driven Anatomical Diversity of Synovial Fibroblasts Guides Joint-Specific Fibroblast Functions. Nat. Commun. 2017, 8, 14852. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Trajerova, M.; Kriegova, E.; Mikulkova, Z.; Savara, J.; Kudelka, M.; Gallo, J. Knee Osteoarthritis Phenotypes Based on Synovial Fluid Immune Cells Correlate with Clinical Outcome Trajectories. Osteoarthr. Cartil. 2022, 30, 1583–1592. [Google Scholar] [CrossRef]
  164. Rosshirt, N.; Hagmann, S.; Tripel, E.; Gotterbarm, T.; Kirsch, J.; Zeifang, F.; Lorenz, H.-M.; Tretter, T.; Moradi, B. A Predominant Th1 Polarization Is Present in Synovial Fluid of End-Stage Osteoarthritic Knee Joints: Analysis of Peripheral Blood, Synovial Fluid and Synovial Membrane. Clin. Exp. Immunol. 2019, 195, 395–406. [Google Scholar] [CrossRef] [PubMed]
  165. Barreto, G.; Manninen, M.; Eklund, K.K. Osteoarthritis and Toll-Like Receptors: When Innate Immunity Meets Chondrocyte Apoptosis. Biology 2020, 9, 65. [Google Scholar] [CrossRef] [Green Version]
  166. Schedel, J.; Wenglen, C.; Distler, O.; Muller-Ladner, U.; Scholmerich, J.; Heinegard, D.; Krenn, V. Differential Adherence of Osteoarthritis and Rheumatoid Arthritis Synovial Fibroblasts to Cartilage and Bone Matrix Proteins and Its Implication for Osteoarthritis Pathogenesis. Scand. J. Immunol. 2004, 60, 514–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Mobasheri, A.; Rayman, M.P.; Gualillo, O.; Sellam, J.; van der Kraan, P.; Fearon, U. The Role of Metabolism in the Pathogenesis of Osteoarthritis. Nat. Rev. Rheumatol. 2017, 13, 302–311. [Google Scholar] [CrossRef]
  168. Woodell-May, J.E.; Sommerfeld, S.D. Role of Inflammation and the Immune System in the Progression of Osteoarthritis. J. Orthop. Res. 2020, 38, 253–257. [Google Scholar] [CrossRef]
  169. Mobasheri, A.; Fonseca, J.E.; Gualillo, O.; Henrotin, Y.; Largo, R.; Herrero-Beaumont, G.; Rocha, F.A.C. Editorial: Inflammation and Biomarkers in Osteoarthritis. Front. Med. 2021, 8, 727700. [Google Scholar] [CrossRef]
  170. Orlowsky, E.W.; Kraus, V.B. The Role of Innate Immunity in Osteoarthritis: When Our First Line of Defense Goes On the Offensive. J. Rheumatol. 2015, 42, 363–371. [Google Scholar] [CrossRef] [Green Version]
  171. Millerand, M.; Berenbaum, F.; Jacques, C. Danger Signals and Inflammaging in Osteoarthritis. Clin. Exp. Rheumatol. 2019, 37, 48–56. [Google Scholar] [PubMed]
  172. Avenoso, A.; Bruschetta, G.; D’Ascola, A.; Scuruchi, M.; Mandraffino, G.; Gullace, R.; Saitta, A.; Campo, S.; Campo, G.M. Hyaluronan Fragments Produced during Tissue Injury: A Signal Amplifying the Inflammatory Response. Arch. Biochem. Biophys. 2019, 663, 228–238. [Google Scholar] [CrossRef] [PubMed]
  173. Deroyer, C.; Poulet, C.; Paulissen, G.; Ciregia, F.; Malaise, O.; Plener, Z.; Cobraiville, G.; Daniel, C.; Gillet, P.; Malaise, M.G.; et al. CEMIP (KIAA1199) Regulates Inflammation, Hyperplasia and Fibrosis in Osteoarthritis Synovial Membrane. Cell. Mol. Life Sci. 2022, 79, 260. [Google Scholar] [CrossRef] [PubMed]
  174. Favero, M.; Belluzzi, E.; Frallonardo, P.; Peruzzo, L.; Tauro, L.; Oliviero, F.; Ramonda, R.; Punzi, L. Synovial Fluid Fetuin-A Levels in Patients Affected by Osteoarthritis with or without Evidence of Calcium Crystals. Rheumatology 2019, 58, 729–730. [Google Scholar] [CrossRef] [PubMed]
  175. Stücker, S.; Bollmann, M.; Garbers, C.; Bertrand, J. The Role of Calcium Crystals and Their Effect on Osteoarthritis Pathogenesis. Best Pract. Res. Clin. Rheumatol. 2021, 35, 101722. [Google Scholar] [CrossRef] [PubMed]
  176. Berenbaum, F.; Eymard, F.; Houard, X. Osteoarthritis, Inflammation and Obesity. Curr. Opin. Rheumatol. 2013, 25, 114–118. [Google Scholar] [CrossRef] [PubMed]
  177. Stolberg-Stolberg, J.; Boettcher, A.; Sambale, M.; Stuecker, S.; Sherwood, J.; Raschke, M.; Pap, T.; Bertrand, J. Toll-like Receptor 3 Activation Promotes Joint Degeneration in Osteoarthritis. Cell Death Dis. 2022, 13, 224. [Google Scholar] [CrossRef]
  178. Miller, R.E.; Ishihara, S.; Tran, P.B.; Golub, S.B.; Last, K.; Miller, R.J.; Fosang, A.J.; Malfait, A.-M. An Aggrecan Fragment Drives Osteoarthritis Pain through Toll-like Receptor 2. JCI Insight 2018, 3, e95704. [Google Scholar] [CrossRef] [Green Version]
  179. Cescon, M.; Gattazzo, F.; Chen, P.; Bonaldo, P. Collagen VI at a Glance. J. Cell Sci. 2015, 128, 3525–3531. [Google Scholar] [CrossRef] [Green Version]
  180. Zelenski, N.A.; Leddy, H.A.; Sanchez-Adams, J.; Zhang, J.; Bonaldo, P.; Liedtke, W.; Guilak, F. Type VI Collagen Regulates Pericellular Matrix Properties, Chondrocyte Swelling, and Mechanotransduction in Mouse Articular Cartilage. Arthritis Rheumatol. 2015, 67, 1286–1294. [Google Scholar] [CrossRef] [Green Version]
  181. Rim, Y.A.; Ju, J.H. The Role of Fibrosis in Osteoarthritis Progression. Life 2020, 11, 3. [Google Scholar] [CrossRef] [PubMed]
  182. Bush, P.G.; Hodkinson, P.D.; Hamilton, G.L.; Hall, A.C. Viability and Volume of in Situ Bovine Articular Chondrocytes-Changes Following a Single Impact and Effects of Medium Osmolarity. Osteoarthr. Cartil. 2005, 13, 54–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Ji, Q.; Zheng, Y.; Zhang, G.; Hu, Y.; Fan, X.; Hou, Y.; Wen, L.; Li, L.; Xu, Y.; Wang, Y.; et al. Single-Cell RNA-Seq Analysis Reveals the Progression of Human Osteoarthritis. Ann. Rheum. Dis. 2019, 78, 100–110. [Google Scholar] [CrossRef] [PubMed]
  184. Styczynska-Soczka, K.; Amin, A.K.; Hall, A.C. Cell-associated Type I Collagen in Nondegenerate and Degenerate Human Articular Cartilage. J. Cell Physiol. 2021, 236, 7672–7681. [Google Scholar] [CrossRef]
  185. Haseeb, A.; Kc, R.; Angelozzi, M.; de Charleroy, C.; Rux, D.; Tower, R.J.; Yao, L.; Pellegrino da Silva, R.; Pacifici, M.; Qin, L.; et al. SOX9 Keeps Growth Plates and Articular Cartilage Healthy by Inhibiting Chondrocyte Dedifferentiation/Osteoblastic Redifferentiation. Proc. Natl. Acad. Sci. USA 2021, 118, e2019152118. [Google Scholar] [CrossRef]
  186. Schminke, B.; Frese, J.; Bode, C.; Goldring, M.B.; Miosge, N. Laminins and Nidogens in the Pericellular Matrix of Chondrocytes: Their Role in Osteoarthritis and Chondrogenic Differentiation. Am. J. Pathol. 2016, 186, 410–418. [Google Scholar] [CrossRef] [Green Version]
  187. Gómez, R.; Scotece, M.; Conde, J.; Gómez-Reino, J.J.; Lago, F.; Gualillo, O. Adiponectin and Leptin Increase IL-8 Production in Human Chondrocytes. Ann. Rheum. Dis. 2011, 70, 2052–2054. [Google Scholar] [CrossRef]
  188. Salucci, S.; Falcieri, E.; Battistelli, M. Chondrocyte Death Involvement in Osteoarthritis. Cell Tissue Res. 2022, 389, 159–170. [Google Scholar] [CrossRef]
  189. Jeon, O.H.; David, N.; Campisi, J.; Elisseeff, J.H. Senescent Cells and Osteoarthritis: A Painful Connection. J. Clin. Investig. 2018, 128, 1229–1237. [Google Scholar] [CrossRef]
  190. Wiley, C.D.; Velarde, M.C.; Lecot, P.; Liu, S.; Sarnoski, E.A.; Freund, A.; Shirakawa, K.; Lim, H.W.; Davis, S.S.; Ramanathan, A.; et al. Mitochondrial Dysfunction Induces Senescence with a Distinct Secretory Phenotype. Cell Metab. 2016, 23, 303–314. [Google Scholar] [CrossRef] [Green Version]
  191. Shane Anderson, A.; Loeser, R.F. Why Is Osteoarthritis an Age-Related Disease? Best Pract. Res. Clin. Rheumatol. 2010, 24, 15–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Jacob, J.; Aggarwal, A.; Aggarwal, A.; Bhattacharyya, S.; Kumar, V.; Sharma, V.; Sahni, D. Senescent Chondrogenic Progenitor Cells Derived from Articular Cartilage of Knee Osteoarthritis Patients Contributes to Senescence-Associated Secretory Phenotype via Release of IL-6 and IL-8. Acta Histochem. 2022, 124, 151867. [Google Scholar] [CrossRef] [PubMed]
  193. Malaise, O.; Tachikart, Y.; Constantinides, M.; Mumme, M.; Ferreira-Lopez, R.; Noack, S.; Krettek, C.; Noël, D.; Wang, J.; Jorgensen, C.; et al. Mesenchymal Stem Cell Senescence Alleviates Their Intrinsic and Seno-Suppressive Paracrine Properties Contributing to Osteoarthritis Development. Aging 2019, 11, 9128–9146. [Google Scholar] [CrossRef] [PubMed]
  194. Troeberg, L.; Nagase, H. Proteases Involved in Cartilage Matrix Degradation in Osteoarthritis. Biochim. Biophys. Acta 2012, 1824, 133–145. [Google Scholar] [CrossRef] [Green Version]
  195. Calvo, E.; Palacios, I.; Delgado, E.; Sánchez-Pernaute, O.; Largo, R.; Egido, J.; Herrero-Beaumont, G. Histopathological Correlation of Cartilage Swelling Detected by Magnetic Resonance Imaging in Early Experimental Osteoarthritis. Osteoarthr. Cartil. 2004, 12, 878–886. [Google Scholar] [CrossRef] [Green Version]
  196. Bank, R.A.; Soudry, M.; Maroudas, A.; Mizrahi, J.; TeKoppele, J.M. The Increased Swelling and Instantaneous Deformation of Osteoarthritic Cartilage Is Highly Correlated with Collagen Degradation. Arthritis Rheum. 2000, 43, 2202–2210. [Google Scholar] [CrossRef]
  197. Ebrahimi, M.; Turunen, M.J.; Finnilä, M.A.; Joukainen, A.; Kröger, H.; Saarakkala, S.; Korhonen, R.K.; Tanska, P. Structure–Function Relationships of Healthy and Osteoarthritic Human Tibial Cartilage: Experimental and Numerical Investigation. Ann. Biomed. Eng. 2020, 48, 2887–2900. [Google Scholar] [CrossRef]
  198. Saarakkala, S.; Julkunen, P.; Kiviranta, P.; Mäkitalo, J.; Jurvelin, J.S.; Korhonen, R.K. Depth-Wise Progression of Osteoarthritis in Human Articular Cartilage: Investigation of Composition, Structure and Biomechanics. Osteoarthr. Cartil. 2010, 18, 73–81. [Google Scholar] [CrossRef] [Green Version]
  199. Mort, J.S.; Geng, Y.; Fisher, W.D.; Roughley, P.J. Aggrecan Heterogeneity in Articular Cartilage from Patients with Osteoarthritis. BMC Musculoskelet. Disord. 2016, 17, 89. [Google Scholar] [CrossRef] [Green Version]
  200. Lorenzo, P.; Bayliss, M.T.; Heinegård, D. Altered Patterns and Synthesis of Extracellular Matrix Macromolecules in Early Osteoarthritis. Matrix Biol. 2004, 23, 381–391. [Google Scholar] [CrossRef]
  201. Wilusz, R.E.; Sanchez-Adams, J.; Guilak, F. The Structure and Function of the Pericellular Matrix of Articular Cartilage. Matrix Biol. 2014, 39, 25–32. [Google Scholar] [CrossRef] [PubMed]
  202. Zhao, Z.; Li, Y.; Wang, M.; Zhao, S.; Zhao, Z.; Fang, J. Mechanotransduction Pathways in the Regulation of Cartilage Chondrocyte Homoeostasis. J. Cell. Mol. Med. 2020, 24, 5408–5419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Loeser, R.F. Integrins and Chondrocyte–Matrix Interactions in Articular Cartilage. Matrix Biol. 2014, 39, 11–16. [Google Scholar] [CrossRef] [PubMed]
  204. Murphy, G.; Nagase, H. Reappraising Metalloproteinases in Rheumatoid Arthritis and Osteoarthritis: Destruction or Repair? Nat. Clin. Pract. Rheumatol. 2008, 4, 128–135. [Google Scholar] [CrossRef] [PubMed]
  205. Mohammed, F.F.; Smookler, D.S.; Khokha, R. Metalloproteinases, Inflammation, and Rheumatoid Arthritis. Ann. Rheum. Dis. 2003, 62 (Suppl. 2), ii43–ii47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Yang, C.-Y.; Chanalaris, A.; Troeberg, L. ADAMTS and ADAM Metalloproteinases in Osteoarthritis–Looking beyond the ‘Usual Suspects’. Osteoarthr. Cartil. 2017, 25, 1000–1009. [Google Scholar] [CrossRef] [Green Version]
  207. Yamamoto, K.; Troeberg, L.; Scilabra, S.D.; Pelosi, M.; Murphy, C.L.; Strickland, D.K.; Nagase, H. LRP-1-mediated Endocytosis Regulates Extracellular Activity of ADAMTS-5 in Articular Cartilage. FASEB J. 2013, 27, 511–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  208. He, J.; Yang, H.; Xu, Z.; Li, J.; Chen, G.; Jiang, L.; Wu, L.; Zhou, X. A Functional Polymorphism in the Paired Basic Amino Acid-Cleaving Enzyme 4 Gene Confers Osteoarthritis Risk in a Population of Eastern China. Genet. Mol. Biol. 2020, 43, e20190115. [Google Scholar] [CrossRef]
  209. Zhou, X.; Jiang, L.; Zhang, Y.; Zhang, J.; Zhou, D.; Wu, L.; Huang, Y.; Xu, N. Genetic Variation of Aggrecanase-2 (ADAMTS5) in Susceptibility to Osteoarthritis. Braz. J. Med. Biol. Res. 2019, 52, e8109. [Google Scholar] [CrossRef] [Green Version]
  210. Stefik, D.; Vranic, V.; Ivkovic, N.; Abazovic, D.; Maric, D.; Vojvodic, D.; Supic, G. An Insight into Osteoarthritis Susceptibility: Integration of Immunological and Genetic Background. Bosn. J. Basic Med. Sci. 2020. [Google Scholar] [CrossRef]
  211. Sulzbacher, I. Osteoarthritis: Histology and Pathogenesis. Wien. Med. Wochenschr. 2013, 163, 212–219. [Google Scholar] [CrossRef] [PubMed]
  212. Cai, L.; Huang, J.; Huang, D.; Lv, H.; Wang, D.; Wang, H.; Miao, H.; Wu, L.; Wang, F. Deficiency of Immune-Responsive Gene 1 Exacerbates Interleukin-1beta-Elicited the Inflammatory Response of Chondrocytes via Enhancing the Activation of NLRP3 Inflammasome. Int. Immunopharmacol. 2023, 114, 109456. [Google Scholar] [CrossRef] [PubMed]
  213. Aho, O.-M.; Finnilä, M.; Thevenot, J.; Saarakkala, S.; Lehenkari, P. Subchondral Bone Histology and Grading in Osteoarthritis. PLoS ONE 2017, 12, e0173726. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Hu, Y.; Chen, X.; Wang, S.; Jing, Y.; Su, J. Subchondral Bone Microenvironment in Osteoarthritis and Pain. Bone Res. 2021, 9, 20. [Google Scholar] [CrossRef] [PubMed]
  215. Huebner, J.L.; Bay-Jensen, A.C.; Huffman, K.M.; He, Y.; Leeming, D.J.; McDaniel, G.E.; Karsdal, M.A.; Kraus, V.B. Alpha C-Telopeptide of Type I Collagen Is Associated With Subchondral Bone Turnover and Predicts Progression of Joint Space Narrowing and Osteophytes in Osteoarthritis: Urinary α-CTX Predicts OA Progression. Arthritis Rheumatol. 2014, 66, 2440–2449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  216. Jiang, Y.; Xiao, Q.; Hu, Z.; Pu, B.; Shu, J.; Yang, Q.; Lao, H.; Hao, J. Tissue Levels of Leukemia Inhibitory Factor Vary by Osteoarthritis Grade. Orthopedics 2014, 37, e460–e464. [Google Scholar] [CrossRef] [Green Version]
  217. Alliston, T.; Hernandez, C.J.; Findlay, D.M.; Felson, D.T.; Kennedy, O.D. Bone Marrow Lesions in Osteoarthritis: What Lies beneath: BMLs WHAT LIES BENEATH. J. Orthop. Res. 2018, 36, 1818–1825. [Google Scholar] [CrossRef]
  218. Klement, M.R.; Sharkey, P.F. The Significance of Osteoarthritis-Associated Bone Marrow Lesions in the Knee. J. Am. Acad. Orthop. Surg. 2019, 27, 752–759. [Google Scholar] [CrossRef]
  219. Nakashima, T.; Hayashi, M.; Fukunaga, T.; Kurata, K.; Oh-Hora, M.; Feng, J.Q.; Bonewald, L.F.; Kodama, T.; Wutz, A.; Wagner, E.F.; et al. Evidence for Osteocyte Regulation of Bone Homeostasis through RANKL Expression. Nat. Med. 2011, 17, 1231–1234. [Google Scholar] [CrossRef]
  220. Jaiprakash, A.; Prasadam, I.; Feng, J.Q.; Liu, Y.; Crawford, R.; Xiao, Y. Phenotypic Characterization of Osteoarthritic Osteocytes from the Sclerotic Zones: A Possible Pathological Role in Subchondral Bone Sclerosis. Int. J. Biol. Sci. 2012, 8, 406–417. [Google Scholar] [CrossRef] [Green Version]
  221. Yee, C.S.; Schurman, C.A.; White, C.R.; Alliston, T. Investigating Osteocytic Perilacunar/Canalicular Remodeling. Curr. Osteoporos. Rep. 2019, 17, 157–168. [Google Scholar] [CrossRef] [PubMed]
  222. Mazur, C.M.; Woo, J.J.; Yee, C.S.; Fields, A.J.; Acevedo, C.; Bailey, K.N.; Kaya, S.; Fowler, T.W.; Lotz, J.C.; Dang, A.; et al. Osteocyte Dysfunction Promotes Osteoarthritis through MMP13-Dependent Suppression of Subchondral Bone Homeostasis. Bone Res. 2019, 7, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Fowler, T.W.; Acevedo, C.; Mazur, C.M.; Hall-Glenn, F.; Fields, A.J.; Bale, H.A.; Ritchie, R.O.; Lotz, J.C.; Vail, T.P.; Alliston, T. Glucocorticoid Suppression of Osteocyte Perilacunar Remodeling Is Associated with Subchondral Bone Degeneration in Osteonecrosis. Sci. Rep. 2017, 7, 44618. [Google Scholar] [CrossRef] [PubMed]
  224. Hemmatian, H.; Jalali, R.; Semeins, C.M.; Hogervorst, J.M.A.; van Lenthe, G.H.; Klein-Nulend, J.; Bakker, A.D. Mechanical Loading Differentially Affects Osteocytes in Fibulae from Lactating Mice Compared to Osteocytes in Virgin Mice: Possible Role for Lacuna Size. Calcif. Tissue Int. 2018, 103, 675–685. [Google Scholar] [CrossRef] [Green Version]
  225. Busse, B.; Djonic, D.; Milovanovic, P.; Hahn, M.; Püschel, K.; Ritchie, R.O.; Djuric, M.; Amling, M. Decrease in the Osteocyte Lacunar Density Accompanied by Hypermineralized Lacunar Occlusion Reveals Failure and Delay of Remodeling in Aged Human Bone. Aging Cell 2010, 9, 1065–1075. [Google Scholar] [CrossRef]
  226. Bolamperti, S.; Villa, I.; Rubinacci, A. Bone Remodeling: An Operational Process Ensuring Survival and Bone Mechanical Competence. Bone Res. 2022, 10, 48. [Google Scholar] [CrossRef]
  227. Andreev, D.; Liu, M.; Weidner, D.; Kachler, K.; Faas, M.; Grüneboom, A.; Schlötzer-Schrehardt, U.; Muñoz, L.E.; Steffen, U.; Grötsch, B.; et al. Osteocyte Necrosis Triggers Osteoclast-Mediated Bone Loss through Macrophage-Inducible C-Type Lectin. J. Clin. Investig. 2020, 130, 4811–4830. [Google Scholar] [CrossRef]
  228. Bouaziz, W.; Funck-Brentano, T.; Lin, H.; Marty, C.; Ea, H.-K.; Hay, E.; Cohen-Solal, M. Loss of Sclerostin Promotes Osteoarthritis in Mice via β-Catenin-Dependent and -Independent Wnt Pathways. Arthritis Res. Ther. 2015, 17, 24. [Google Scholar] [CrossRef] [Green Version]
  229. Sanchez, C.; Deberg, M.A.; Bellahcène, A.; Castronovo, V.; Msika, P.; Delcour, J.P.; Crielaard, J.M.; Henrotin, Y.E. Phenotypic Characterization of Osteoblasts from the Sclerotic Zones of Osteoarthritic Subchondral Bone. Arthritis Rheum. 2008, 58, 442–455. [Google Scholar] [CrossRef]
  230. Couchourel, D.; Aubry, I.; Delalandre, A.; Lavigne, M.; Martel-Pelletier, J.; Pelletier, J.-P.; Lajeunesse, D. Altered Mineralization of Human Osteoarthritic Osteoblasts Is Attributable to Abnormal Type I Collagen Production. Arthritis Rheum. 2009, 60, 1438–1450. [Google Scholar] [CrossRef]
  231. Housmans, B.A.; van den Akker, G.G.; Neefjes, M.; Surtel, D.A.; Cremers, A.; van der Kraan, P.; van Rhijn, L.; Welting, T. OA Synovial Fluid Induces Chondrocyte Fibrosis and Proliferation via MAPK and RHOGTPASE Signaling Routes. Osteoarthr. Cartil. 2021, 29, S136. [Google Scholar] [CrossRef]
  232. Yang, L.; Chen, Z.; Guo, H.; Wang, Z.; Sun, K.; Yang, X.; Zhao, X.; Ma, L.; Wang, J.; Meng, Z.; et al. Extensive Cytokine Analysis in Synovial Fluid of Osteoarthritis Patients. Cytokine 2021, 143, 155546. [Google Scholar] [CrossRef] [PubMed]
  233. Silverstein, A.M.; Stefani, R.M.; Sobczak, E.; Tong, E.L.; Attur, M.G.; Shah, R.P.; Bulinski, J.C.; Ateshian, G.A.; Hung, C.T. Toward Understanding the Role of Cartilage Particulates in Synovial Inflammation. Osteoarthr. Cartil. 2017, 25, 1353–1361. [Google Scholar] [CrossRef] [PubMed]
  234. Chen, P.; Zhou, J.; Ruan, A.; Guan, H.; Xie, J.; Zeng, L.; Liu, J.; Wang, Q. Synovial Tissue-Derived Extracellular Vesicles Induce Chondrocyte Inflammation and Degradation via NF-ΚB Signalling Pathway: An in Vitro Study. J. Cell. Mol. Med. 2022, 26, 2038–2048. [Google Scholar] [CrossRef]
  235. Zreiqat, H.; Belluoccio, D.; Smith, M.M.; Wilson, R.; Rowley, L.A.; Jones, K.; Ramaswamy, Y.; Vogl, T.; Roth, J.; Bateman, J.F.; et al. S100A8 and S100A9 in Experimental Osteoarthritis. Arthritis Res. Ther. 2010, 12, R16. [Google Scholar] [CrossRef] [Green Version]
  236. Nakashima, M.; Sakai, T.; Hiraiwa, H.; Hamada, T.; Omachi, T.; Ono, Y.; Inukai, N.; Ishizuka, S.; Matsukawa, T.; Oda, T.; et al. Role of S100A12 in the Pathogenesis of Osteoarthritis. Biochem. Biophys. Res. Commun. 2012, 422, 508–514. [Google Scholar] [CrossRef]
  237. Chan, M.W.Y.; Gomez-Aristizábal, A.; Mahomed, N.; Gandhi, R.; Viswanathan, S. A Tool for Evaluating Novel Osteoarthritis Therapies Using Multivariate Analyses of Human Cartilage-Synovium Explant Co-Culture. Osteoarthr. Cartil. 2022, 30, 147–159. [Google Scholar] [CrossRef]
  238. Fahy, N.; Melle, M.L.d.V.; Lehmann, J.; Wei, W.; Grotenhuis, N.; Farrell, E.; van der Kraan, P.M.; Murphy, J.M.; Bastiaansen-Jenniskens, Y.M.; Osch, G.J.V.M. van Human Osteoarthritic Synovium Impacts Chondrogenic Differentiation of Mesenchymal Stem Cells via Macrophage Polarisation State. Osteoarthr. Cartil. 2014, 22, 1167–1175. [Google Scholar] [CrossRef] [Green Version]
  239. Intema, F.; Hazewinkel, H.A.W.; Gouwens, D.; Bijlsma, J.W.J.; Weinans, H.; Lafeber, F.P.J.G.; Mastbergen, S.C. In Early OA, Thinning of the Subchondral Plate Is Directly Related to Cartilage Damage: Results from a Canine ACLT-Meniscectomy Model. Osteoarthr. Cartil. 2010, 18, 691–698. [Google Scholar] [CrossRef] [Green Version]
  240. Chen, Y.; Huang, Y.-C.; Yan, C.H.; Chiu, K.Y.; Wei, Q.; Zhao, J.; Guo, X.E.; Leung, F.; Lu, W.W. Abnormal Subchondral Bone Remodeling and Its Association with Articular Cartilage Degradation in Knees of Type 2 Diabetes Patients. Bone Res. 2017, 5, 17034. [Google Scholar] [CrossRef] [Green Version]
  241. Fell, N.L.A.; Lawless, B.M.; Cox, S.C.; Cooke, M.E.; Eisenstein, N.M.; Shepherd, D.E.T.; Espino, D.M. The Role of Subchondral Bone, and Its Histomorphology, on the Dynamic Viscoelasticity of Cartilage, Bone and Osteochondral Cores. Osteoarthr. Cartil. 2019, 27, 535–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  242. Bellido, M.; Lugo, L.; Roman-Blas, J.A.; Castañeda, S.; Calvo, E.; Largo, R.; Herrero-Beaumont, G. Improving Subchondral Bone Integrity Reduces Progression of Cartilage Damage in Experimental Osteoarthritis Preceded by Osteoporosis. Osteoarthr. Cartil. 2011, 19, 1228–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Neogi, T.; Felson, D.; Niu, J.; Lynch, J.; Nevitt, M.; Guermazi, A.; Roemer, F.; Lewis, C.E.; Wallace, B.; Zhang, Y. Cartilage Loss Occurs in the Same Subregions as Subchondral Bone Attrition: A within-Knee Subregion-Matched Approach from the Multicenter Osteoarthritis Study. Arthritis Rheum. 2009, 61, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
  244. Su, W.; Liu, G.; Liu, X.; Zhou, Y.; Sun, Q.; Zhen, G.; Wang, X.; Hu, Y.; Gao, P.; Demehri, S.; et al. Angiogenesis Stimulated by Elevated PDGF-BB in Subchondral Bone Contributes to Osteoarthritis Development. JCI Insight 2020, 5, e135446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  245. Walsh, D.A.; Bonnet, C.S.; Turner, E.L.; Wilson, D.; Situ, M.; McWilliams, D.F. Angiogenesis in the Synovium and at the Osteochondral Junction in Osteoarthritis. Osteoarthr. Cartil. 2007, 15, 743–751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  246. Aso, K.; Shahtaheri, S.M.; Hill, R.; Wilson, D.; McWilliams, D.F.; Nwosu, L.N.; Chapman, V.; Walsh, D.A. Contribution of Nerves within Osteochondral Channels to Osteoarthritis Knee Pain in Humans and Rats. Osteoarthr. Cartil. 2020, 28, 1245–1254. [Google Scholar] [CrossRef]
  247. Wang, T.; He, C. Pro-Inflammatory Cytokines: The Link between Obesity and Osteoarthritis. Cytokine Growth Factor Rev. 2018, 44, 38–50. [Google Scholar] [CrossRef]
  248. Yu, C.D.; Miao, W.H.; Zhang, Y.Y.; Zou, M.J.; Yan, X.F. Inhibition of MiR-126 Protects Chondrocytes from IL-1β Induced Inflammation via Upregulation of Bcl-2. Bone Jt. Res. 2018, 7, 414–421. [Google Scholar] [CrossRef]
  249. Mehana, E.-S.E.; Khafaga, A.F.; El-Blehi, S.S. The Role of Matrix Metalloproteinases in Osteoarthritis Pathogenesis: An Updated Review. Life Sci. 2019, 234, 116786. [Google Scholar] [CrossRef]
  250. Latourte, A.; Cherifi, C.; Maillet, J.; Ea, H.-K.; Bouaziz, W.; Funck-Brentano, T.; Cohen-Solal, M.; Hay, E.; Richette, P. Systemic Inhibition of IL-6/Stat3 Signalling Protects against Experimental Osteoarthritis. Ann. Rheum. Dis. 2017, 76, 748–755. [Google Scholar] [CrossRef]
  251. Sato, K.; Suematsu, A.; Okamoto, K.; Yamaguchi, A.; Morishita, Y.; Kadono, Y.; Tanaka, S.; Kodama, T.; Akira, S.; Iwakura, Y.; et al. Th17 Functions as an Osteoclastogenic Helper T Cell Subset That Links T Cell Activation and Bone Destruction. J. Exp. Med. 2006, 203, 2673–2682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  252. Sinkeviciute, D.; Aspberg, A.; He, Y.; Bay-Jensen, A.-C.; Önnerfjord, P. Characterization of the Interleukin-17 Effect on Articular Cartilage in a Translational Model: An Explorative Study. BMC Rheumatol. 2020, 4, 30. [Google Scholar] [CrossRef]
  253. Fu, Z.; Liu, P.; Yang, D.; Wang, F.; Yuan, L.; Lin, Z.; Jiang, J. Interleukin-18-Induced Inflammatory Responses in Synoviocytes and Chondrocytes from Osteoarthritic Patients. Int. J. Mol. Med. 2012, 30, 805–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Molnar, V.; Matišić, V.; Kodvanj, I.; Bjelica, R.; Jeleč, Ž.; Hudetz, D.; Rod, E.; Čukelj, F.; Vrdoljak, T.; Vidović, D.; et al. Cytokines and Chemokines Involved in Osteoarthritis Pathogenesis. Int. J. Mol. Sci. 2021, 22, 9208. [Google Scholar] [CrossRef] [PubMed]
  255. Xue, J.; Wang, J.; Liu, Q.; Luo, A. Tumor Necrosis Factor-α Induces ADAMTS-4 Expression in Human Osteoarthritis Chondrocytes. Mol. Med. Rep. 2013, 8, 1755–1760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  256. Wojdasiewicz, P.; Poniatowski, Ł.A.; Szukiewicz, D. The Role of Inflammatory and Anti-Inflammatory Cytokines in the Pathogenesis of Osteoarthritis. Mediat. Inflamm. 2014, 2014, 561459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Chakrabarti, S.; Hore, Z.; Pattison, L.A.; Lalnunhlimi, S.; Bhebhe, C.N.; Callejo, G.; Bulmer, D.C.; Taams, L.S.; Denk, F.; Smith, E.S.T.J. Sensitization of Knee-Innervating Sensory Neurons by Tumor Necrosis Factor-α-Activated Fibroblast-like Synoviocytes: An in Vitro, Coculture Model of Inflammatory Pain. Pain 2020, 161, 2129–2141. [Google Scholar] [CrossRef]
  258. Lepetsos, P.; Papavassiliou, K.A.; Papavassiliou, A.G. Redox and NF-ΚB Signaling in Osteoarthritis. Free Radic. Biol. Med. 2019, 132, 90–100. [Google Scholar] [CrossRef]
  259. Conde, J.; Otero, M.; Scotece, M.; Abella, V.; López, V.; Pino, J.; Gómez, R.; Lago, F.; Goldring, M.B.; Gualillo, O. E74-like Factor 3 and Nuclear Factor-ΚB Regulate Lipocalin-2 Expression in Chondrocytes. J. Physiol. 2016, 594, 6133–6146. [Google Scholar] [CrossRef]
  260. Choi, M.-C.; Jo, J.; Park, J.; Kang, H.K.; Park, Y. NF-ΚB Signaling Pathways in Osteoarthritic Cartilage Destruction. Cells 2019, 8, 734. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Common sites (red circles) and risk factors for OA: age, gender, genetics, metabolic state (obesity, type 2 diabetes), joint overuse, trauma, and injury that contribute to changes in the main joint compartments, namely, the synovial membrane, cartilage, fibrocartilage of meniscus, as well as subchondral bone.
Figure 1. Common sites (red circles) and risk factors for OA: age, gender, genetics, metabolic state (obesity, type 2 diabetes), joint overuse, trauma, and injury that contribute to changes in the main joint compartments, namely, the synovial membrane, cartilage, fibrocartilage of meniscus, as well as subchondral bone.
Ijms 24 04120 g001
Figure 2. Schematic overview of a synovial joint structure. (A) Macroscopic view on the compartments of synovial joint. (B) The synovial membrane (upper part) in a healthy state. Composition of a healthy “osteochondral” unit (lower part), tidemark (red asterisk), and calcified cartilage (yellow asterisk).
Figure 2. Schematic overview of a synovial joint structure. (A) Macroscopic view on the compartments of synovial joint. (B) The synovial membrane (upper part) in a healthy state. Composition of a healthy “osteochondral” unit (lower part), tidemark (red asterisk), and calcified cartilage (yellow asterisk).
Ijms 24 04120 g002
Figure 3. Schematic overview of the OA-affected synovial joint with involvement of both “chondrosynovial” and “osteochondral units”. (A) Synovial joint depicting common changes, such as degradation of the articular cartilage, formation of subchondral bone cysts, and osteophytes (red arrows). (B) The synovial membrane (upper part) reveals hyperplasia of the lining layer, immune cell infiltration, angiogenesis, and fibrosis in the sublining layer. Alterations of the articular cartilage (lower part) include degradation of the non-calcified zone and thickening of the calcified zone (yellow asterisks), formation of free cartilage fragments, duplication of tidemark (red asterisk), alterations in the composition of cellular and extracellular matrix. Remodeling of the subchondral bone (lower part) reveals the thickening of the cortical bone plate, formation of cysts, and angiogenesis.
Figure 3. Schematic overview of the OA-affected synovial joint with involvement of both “chondrosynovial” and “osteochondral units”. (A) Synovial joint depicting common changes, such as degradation of the articular cartilage, formation of subchondral bone cysts, and osteophytes (red arrows). (B) The synovial membrane (upper part) reveals hyperplasia of the lining layer, immune cell infiltration, angiogenesis, and fibrosis in the sublining layer. Alterations of the articular cartilage (lower part) include degradation of the non-calcified zone and thickening of the calcified zone (yellow asterisks), formation of free cartilage fragments, duplication of tidemark (red asterisk), alterations in the composition of cellular and extracellular matrix. Remodeling of the subchondral bone (lower part) reveals the thickening of the cortical bone plate, formation of cysts, and angiogenesis.
Ijms 24 04120 g003
Table 1. The overview of signaling molecules produced by cells in the context of compartments in the healthy joint.
Table 1. The overview of signaling molecules produced by cells in the context of compartments in the healthy joint.
Chemical
Messenger
Signaling
Factor
CellsEffectReferences
Transcription
factor
SOX-9 1Synoviocytes,
chondrocytes,
osteoblast lineage
cells
Maintains the chondrogenic phenotype and functions of the chondrocyte, prevents hypertrophy, antagonizing with RUNX2
Inhibits IL-1β-induced inflammatory response and chondrocyte apoptosis
Inhibits the production of ADAMTS in cartilage tissue
[97,109,110]
Growth
factor
IGF-1 2Synoviocytes,
chondrocytes,
osteoblast lineage
cells
Exhibits anti-inflammatory and anti-catabolic effects in chondrocytes
Maintains articular cartilage anabolism, stimulating ECM production and chondrogenesis
[111,112]
Growth
factor
TGF-β 3Chondrocytes,
synovial fibroblasts, and macrophages
Inhibits chondrocytes phenotype switch to hypertrophic chondrocytes and collagen type X production
Induces proteoglycan synthesis by chondrocytes
Inhibits IL-1β effects
[113]
1 SOX-9—SRY-box transcription factor 9; 2 IGF-1—insulin-like growth factor-1; 3 TGF-β—transforming growth factor-beta; ECM—extracellular matrix.
Table 2. The overview of signaling molecules produced by cells in the major joint compartments of OA-affected joint.
Table 2. The overview of signaling molecules produced by cells in the major joint compartments of OA-affected joint.
Chemical MessengerSignaling FactorCellsEffectReferences
CytokineIL-1β 1Synovial
macrophages and
fibroblasts,
chondrocytes,
osteoblasts
Induces cartilage degradation and inhibits its repair abilities
Stimulates production of MMPs (MMP-1, 3, 9, 13), ADAMTSs 4, 5 by chondrocytes
Suppresses the synthesis of collagen type II and aggrecan
Enhances chondrocytes’ pro-inflammatory response, hypertrophy, dedifferentiation, and apoptosis, inhibits chondrogenesis
Stimulates synovial inflammation and the production of pro-inflammatory cytokines (TNF-α, IL-6, IL-8, IL-17, CCL5), mediators (NO, COX-1), prostaglandins (PGE2)
Induces the formation of pannus-like tissue, fibrosis, and production of pro-fibrotic factors (PDGF, TGF-β)
[126,133,212,247,248,249]
CytokineIL-6Synovial
macrophages and
fibroblasts,
chondrocytes,
osteoblasts
Promotes osteoclast formation and subchondral bone resorption
Increases production of MMPs (MMP-1, 3, 13) and ADAMTS (ADAMTS-4) by chondrocytes
Induces catabolic changes in chondrocytes, as well as promotes cellular senescence
Shows synergic action with IL-1β and TNF-α, sustaining articular cartilage degradation and synovial inflammation
[192,250]
CytokineIL-17CD4+ T cells,
macrophages, NK cells, mast cells
Induces cartilage degradation by upregulating catabolic factors (MMP-1, 3, 13; ADAMTS) and downregulating anabolic factors (SOX-9, COL2A1) in chondrocytes
Promotes recruitment of inflammatory cells and release of pro-inflammatory mediators, induces angiogenesis
Induces RANKL expression and osteoclastogenesis
[251,252]
CytokineIL-18Synovial macrophages, fibroblasts,
Chondrocytes, and
osteoblasts
Promotes articular cartilage degradation by upregulating MMP-1, 3, 13 and suppressing aggrecan synthesis
Stimulates pro-inflammatory conditions by induction of cytokines synthesis through NF-κB and MAPK signaling pathways
Stimulates both bone resorption and osteophyte formation
Enhances gene expression for the synthesis of IL-6, TNF-α
[232,253,254]
CytokineTNF-α 2Synovial macrophages and
fibroblasts,
chondrocytes,
osteoblasts
Levels of expression are associated with radiographic OA cartilage loss
Shows action synergism with IL-1β
Stimulates MMP and ADAMTS production by chondrocytes; inhibits synthesis of collagen type II and aggrecan
Inhibits chondrocyte differentiation by suppressing the expression of SOX-9, and induces apoptosis
Promotes pro-inflammatory signaling pathways in synoviocytes and chondrocytes, stimulating the release of IL-1β, IL-6, IL-8, IL-10
Leads to neuronal sensitization, predisposing to the development of pain in OA
Promotes angiogenesis and aberrant bone formation in subchondral bone by recruiting mesenchymal stem cells
[110,134,248,253,255,256,257]
EnzymeMMPs 3Chondrocytes,
synovial
macrophages, and
fibroblasts
MMP-1 degrades collagen types I, II, III, and aggrecan of articular cartilage
MMP-3 cleaves collagen types II, IV, IX, X, XI, and aggrecan; activates other MMPs (MMP-1, 7, 9)
MMP-9 cleaves non-collagenous matric components
MMP-13 levels correlate with hypertrophic chondrocytes in early stage OA; with OA severity and articular cartilage deterioration, as well as NF-κB expression; the enzyme exhibits higher activity for collagen type II cleavage and degrades aggrecan; it is associated with synovial membrane hyperplasia and cellular senescence
[88,133,249]
EnzymeADAMTSs 4Chondrocytes,
synovial fibroblasts, and
macrophages
ADAMTS-4, 5 are induced by IL-1β, TNF-α in chondrocytes and promote cleavage of aggrecan[206]
Transcription factorNF-κB 5All joint cellsActs alone or in synergy with other signaling pathways
Inhibits anabolic functions of chondrocytes
Triggers chondrocyte hypertrophy, apoptosis, catabolic functions (MMP, ADAMTS, NO, PGE2, COX2 production), production of pro-inflammatory cytokines (IL-1β, TNF-α, IL-6, IL-8) by chondrocytes, which augment the action of NF-κB
Augments activation of other transcription factors such as ELF3, and RUNX2, that stimulate MMP13 and cartilage degradation
In synovial membrane promotes inflammation, angiogenesis, production of cytokines (IL-1β, TNF-α, IL-6), enzymes (MMP-1, MMP-13, ADAMTS-4, ADAMTS-5), VEGF
In subchondral bone promotes resorption
[258,259,260]
Transcription factorSOX-9 6All joint cellsExpression is downregulated in OA-affected joint
Downregulation results in calcified cartilage and osteophyte formation
[110]
Growth
factor
TGF-β 7Chondrocytes,
osteoblasts,
osteoclasts, synovial fibroblasts, and
macrophages
Leads to the osteoid formation and bone sclerosis
Leads to cartilage damage and angiogenesis
[113]
1 IL—interleukin; 2 TNF-α—tumor necrosis factor-alpha; 3 MMP—matrix metalloproteinase; 4 ADAMTS—a disintegrin and metalloproteinase with thrombospondin motifs; 5 NF-κB—nuclear factor kappa B; 6 SOX-9—SRY-box transcription factor 9; 7 TGF-β—transforming growth factor-beta.
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.

Share and Cite

MDPI and ACS Style

Semenistaja, S.; Skuja, S.; Kadisa, A.; Groma, V. Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk. Int. J. Mol. Sci. 2023, 24, 4120. https://doi.org/10.3390/ijms24044120

AMA Style

Semenistaja S, Skuja S, Kadisa A, Groma V. Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk. International Journal of Molecular Sciences. 2023; 24(4):4120. https://doi.org/10.3390/ijms24044120

Chicago/Turabian Style

Semenistaja, Sofija, Sandra Skuja, Anda Kadisa, and Valerija Groma. 2023. "Healthy and Osteoarthritis-Affected Joints Facing the Cellular Crosstalk" International Journal of Molecular Sciences 24, no. 4: 4120. https://doi.org/10.3390/ijms24044120

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop