Open access peer-reviewed chapter

Pathogenesis, Pathology and Genetics of Osteoarthritis

Written By

Ferhat Ege

Submitted: 02 July 2021 Reviewed: 05 July 2021 Published: 29 August 2021

DOI: 10.5772/intechopen.99238

From the Edited Volume

Rheumatoid Arthritis

Edited by Hechmi Toumi

Chapter metrics overview

493 Chapter Downloads

View Full Metrics

Abstract

Osteoarthritis (OA) is a condition with high prevalence worldwide. OA affects not only the articular cartilage, but the entire joint, including the subchondral bone, ligaments, capsule, synovial membrane and the periarticular muscles. Despite the fact that the risks associated with OA increase with age, it is not a part of the natural aging process. It typically involves the knee, hip, spine, hand and foot joints. Several factors play an important role in the pathogenesis of OA, including biomechanical factors, proinflammatory mediators and proteases. On the other hand, it was mostly the results of the studies conducted on the genetic, genomic and epigenetic aspects of OA, from among many of its underlying etiological factors, which shed light on the molecular processes involved in the etiopathogenesis of OA. As the mechanisms that cause joint tissue damage in OA come to light, the treatment of OA will go beyond just providing symptomatic relief. Consequentially, new treatments will emerge that will either slow or completely stop the progression of OA.

Keywords

  • Osteoarthritis
  • Genetics
  • Epigenetics
  • Etiopathogenesis
  • Pathology

1. Introduction

Osteoarthritis (OA) is a chronic disease that affects all structures of the joint as well as the periarticular tissues. In the past, OA was considered simply as a degenerative joint disease, yet the pathogenesis of OA is in fact much more complex than just wear and tear. Hence, the term “osteoarthritis” is indeed a pertinent term, as the suffix “itis” is indicative of an inflammatory process. It is estimated that approximately 50% of the world population over 65 years of age is affected by OA. The symptomatic treatment of this common disease provides regression of symptoms, nevertheless it often does not constitute an effective treatment option thus causing an increase in the OA-related health expenditures. The elucidation of the etiopathogenesis of OA and the molecular studies to be carried out in respect thereof are likely to allow early diagnosis of OA and also contribute to the development of new treatment options.

Advertisement

2. Pathology and pathogenesis of osteoarthritis

Articular cartilage degeneration, which develop as a result of the deterioration of the balance between the production and destruction of cartilage, new bone formation, sclerosis of subchondral bone, ligament and meniscus damage, periarticular muscle weakness, synovial inflammation and fibrosis are all involved in the pathogenesis of OA [1]. Hence, the pathogenesis of OA would be better understood provided that the structure of the joint and the related histopathological features are reviewed.

2.1 Structure of the joint

Synovial joints consist of an articular cartilage that covers the ends of the opposing bones, the synovial fluid that nourishes and lubricates the tissues, the synovium that secretes the synovial fluid, the ligaments that hold the skeletal elements together, the tendons that connect the bones with the muscles, and the joint capsule surrounding the joint. In order to have normal joint functions, it is necessary that the opposing joint surfaces move over each other painlessly, that the load on the joint tissues is homogeneously distributed, and that the stability to that effect is sustained [2].

Articular cartilage is a connective tissue located at the bone ends and which has a thickness of 0.2 mm to 6 mm depending on the location. Articular cartilage provides a smooth and low-friction surface that primarily allows for normal gliding motion of the articular surfaces [3]. Cartilage consists of an extracellular matrix, 65–80% of which is water and 20–35% of which is solid matter, and of chondrocytes dispersed in this matrix. 5–6% of the tissue is composed of inorganic material consisting mostly of hydroxyapatite. The organic matter on the other hand is composed of fibrous proteins (collagen), hydrophilic sulfated proteoglycans (chondroitin sulfate, keratan sulfate I and II) and unsulfated proteins (hyaluronic acid). 90% of the collagen is type II collagen, whereas the remaining collagen consists of smaller amounts of type IX, XI, III, VI, XII and XIV collagen [4].

A proteoglycan consists of a protein and glycosaminoglycan chains attached to this protein. The most abundant type of proteoglycan is ‘aggrecan’ [5]. Type II collagen plays a role in maintaining the volume and shape of the content it is part of, whereas proteoglycans play a role in maintaining the hardness and elasticity [6].

Hyaluronic acid is the substance that maintains the viscosity in synovial fluid. Nonetheless, it requires the presence of a large mucinous protein, which is called lubrisin (proteoglycan-4), in order to maintain a low-friction environment and protect the surface of the joint [7].

Articular cartilage is a avascular heterogeneous structure with four different layers which has no nerve innervation and is fed by a bidirectional diffusion system. These layers are the superficial zone, transitional zone, deep zone and calcified zone. The calcified line between the deep zone and the calcified zone is called the Tide mark [8].

The extracellular matrix is synthesized by chondrocytes. Chondrocytes synthesize cartilage matrix molecules and the metalloproteinases which breakdown the matrix. The cartilage metabolism is based on the balance between the anabolic processes and the catabolic processes carried out by the matrix metalloproteinases (collagenase, gelatinase, stromelysin, cathepsin B and D) and the adamalysins [a disintegrin and metalloproteinase (ADAM), a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), aggrecanase] [5]. This balance is regulated by anabolic cytokines such as transforming growth factor beta (TGF-β), insulin-like growth factor-1 (IGF-1) and bone morphogenetic proteins (BMPs) and catabolic cytokines such as interleukin 1 alpha (IL-1α), interleukin 1 beta (IL-1β) and tumor necrosis factor-alpha (TNF-α) [6].

The synovium, which produces the synovial fluid, consists of two layers as inner and outer layers. It is firmly attached to the joint capsule and prevents the synovial fluid from leaving the joint. The inner and outer layers of the synovium are composed of the synovial membrane and fibrous connective tissue, respectively. The inner layer includes two types of cells. Type A synoviocytes have the characteristics of macrophages, whereas type B synoviocytes are cells with proliferative capacity and produce hyaluronic acid, collagen, lubricin and fibronectin [9].

The joint capsule is a tissue that contains vascular and nervous tissues and is rich in collagen fibers. It protects the whole joint both passively by restricting the movements of the joint and actively through the proprioceptive sensation triggered by the nerve endings.

2.2 Pathological changes that occur in connection with osteoarthritis

2.2.1 Changes that occur in the articular cartilage

Chondrocytes are active cells that maintain cartilage through normal anabolic/catabolic activities. The earliest pathological changes observed in association with OA are the fibrillations seen on the surface of the cartilage. Fibrillations are more common at parts of the cartilage exposed to higher loads. Loosening of the collagen network and loss of aggregate occurs in the cartilage at the onset of OA. This loosening of the collagen network allows the hydrophilic proteoglycans to attract water and expand.

The activity of chondrocytes, the only cell type found in cartilage, accelerates significantly as OA develops, that is, chondrocytes begin to proliferate moderately. Nevertheless, the reasons that trigger this premature aging and changes in the chondrocyte cycle such as inflammation, proteoglycan loss, collagen degeneration and chondrocyte failure, as well as the order of occurrence of these changes are still not fully known [10].

As OA progresses, extensive matrix breakdown and loss occur due to the continued production of the proteases driven by proinflammatory cytokines. Fragmented matrix proteins give rise to the further production of cytokine and protease by chondrocytes through autocrine and paracrine stimulations. Cartilage has limited regeneration capacity, hence once collagen is broken down and lost, regeneration does not occur at a measurable degree [11].

There are various histopathological staging-grading systems that are used to categorize the changes associated with OA according to their severity, extent or order of occurrence. These classification systems classically address the changes that occur in articular cartilage, since OA primarily targets the articular cartilage. One of these systems, the histological evaluation system proposed by Osteoarthritis Research Society International (OARSI) is a grading, staging and a scoring system. The grades used in the said OARSI system to classify the changes occur in articular cartilage, key features of these grades and the associated criteria in terms of tissue reactions are shown in Table 1 [12].

Grade #Key featuresAssociated criteria (tissue reaction)
Grade 0Intact surface and cartilage morphologyMatrix: normal architecture
Cells: intact with appropriate orientation
Grade 1Intact surfaceMatrix: intact superficial zone, oedema and/or superficial fibrillation (abrasion), focal superficial matrix condensation
Cells: death, proliferation (clusters), hypertrophy, superficial zone reaction must be more than superficial fibrillation only
Grade 2Surface discontinuityAs above CMatrix discontinuity at superficial zone (deep fibrillation) GCationic stain matrix depletion (Safranin O or Toluidine Blue) upper 1/3 of cartilage GFocal perichondronal increased stain (transitional zone) GDisorientation of chondron columns
Cells: death, proliferation (clusters), hypertrophy
Grade 3Vertical fissuresAs above Matrix vertical fissures into transitional zone, branched fissures GCationic stain depletion (Safranin O or Toluidine Blue) into lower 2/3 of cartilage (deep zone) GNew collagen formation (polarized light microscopy, Picro Sirius Red stain)
Cells: death, regeneration (clusters), hypertrophy, cartilage domains adjacent to fissures
Grade 4ErosionCartilage matrix loss: delamination of superficial layer, transitional zone cyst formation
Excavation: matrix loss superficial and transitional zones
Grade 5DenudationSurface: sclerotic bone or reparative tissue including fibrocartilage within denuded surface. Microfracture with repair limited to bone surface
Grade 6DeformationBone remodeling (more than osteophyte formation only) including microfracture with fibrocartilaginous and osseous repair extending above the previous surface

Table 1.

A cartilage histopathology grading methodology.

2.2.2 Changes that occur in the bone

Thickening of the subchondral bone (bone sclerosis) occurs due to increased production of improperly mineralized collagen. Osteophytes occur at the margins of the joints, usually at the insertion sites of tendons or ligaments. Osteophytes seen in the distal interphalangeal joints of the hand are called “Heberden’s nodes”, whereas the osteophytes seen in the proximal interphalangeal joints are called “Bouchard’s nodes”. Bone cysts form in the advanced stages of the disease, but bone erosions are not typically seen. Erosive OA is commonly seen in the distal joints of the hands (distal interphalangeals and proximal interphalangeals) and central erosions are also seen as opposed to the marginal erosions seen in rheumatoid arthritis (RA) and gout [13].

2.2.3 Changes that occur in the synovium

Four patterns have been described in OA-related synovial pathology, which are hyperplastic, inflammatory, fibrotic and detritic patterns. Hyperplastic pattern is the most common manifestation in all stages of OA. Hyperplastic pattern is considered as an early OA finding in its isolated form. Inflammatory pattern is seen equally in both the early and late stages of OA. Inflammatory cell density in the inflammatory pattern is not as much as it is in rheumatoid arthritis. Fibrotic pattern is characterized by capsular fibrosis reflecting late-stage OA. Detrital pattern is characterized by macromolecular cartilages and debris within the synovium, and reflects late-stage OA [14].

2.2.4 Changes that occur in the meniscus

The changes that occur in the meniscus in connection with OA are first observed in the medial part of the meniscus. Meniscal tears are both a cause and effect of OA. Meniscal tears further increase the matrix degeneration through the inflammatory mediators which emerge as a result of the damage to the meniscus and may lead to the development of OA [15]. The regeneration capacity of the meniscus is limited. The red zone of the meniscus, which is peripherally located, is the area with the best blood supply and the best regeneration capacity, whereas the white zone of the meniscus, which is more centrally located, is largely avascular and its regeneration is very slow and inadequate [16].

2.3 Etiopathogenesis of osteoarthritis

OA refers to a dynamic process, which is triggered by various biochemical and mechanical factors and in which destruction and regeneration both take place. In the past, OA was thought to be a degenerative joint disease that emerged with aging. Yet, it is known today that various factors such as biomechanical factors, proinflammatory mediators and proteases play a role in the pathogenesis of OA [17]. The release of biomarkers indicates that the findings that emerge in the earliest detectable stage of knee OA are bone and cartilage metabolisms that are impaired as a result inflammation [18].

2.3.1 Factors involved in the etiopathogenesis of osteoarthritis

2.3.1.1 Inflammation

The number of proinflammatory mediators included in the synovial fluid and tissue and which play a role in OA and is increasing by the day. Early studies on OA were focused on interleukin-1 (IL1), which stimulates cartilage catabolic activity. Nevertheless, the role of IL1 in OA has been questioned over the years, since the IL1 levels in OA joints are much lower than the levels that cause cartilage deterioration. It has been shown in the relevant clinical studies that the inhibition of IL1 in knee [19] and hand OA [20] have not improved the structure and symptoms of the disease.

Cytokines such as IL6, interferon-gamma inducible-protein-10 (IP-10), monocyte chemoattractant protein-1 (MCP-1) and monokine induced by gamma interferon (MIG) were found to be more abundant in OA synovial fluid than IL1 or TNF-α [21]. This finding suggests that these proinflammatory cytokines play a role in inflammation. It has been demonstrated in experimental animal models that there may be a relationship between IL-6 level and increased cartilage loss. The results of all these studies support the hypothesis that IL-6, as a regulatory cytokine, plays a role in the development of OA [22]. Other cytokines and chemokines involved in cartilage degeneration caused by inhibition of the anabolic process and induction of the catabolic process are IL-7, IL-15, IL-17, IL-18, oncostatin M (OSM), growth related oncogene-alpha (GRO-alpha), chemokine (C-C motif) ligand 19 (CCL19) and macrophage inflammatory protein-1beta (MIP-1beta) [23].

It was demonstrated in some studies that there is complement activity in OA joints. In one of these studies, which was conducted on mice, it was demonstrated that complement activation was inhibited by gene deletion or pharmacological modulation and that, as a result of this inhibition, the joint is protected from surgery-induced OA [24].

Adipokines secreted by adipose tissue cause cartilage damage by activating the inflammatory cytokines along with the matrix metalloproteinases (MMPs) triggered by the inflammatory cytokines. These adipokines include leptin, adiponectin, visfatin and resistin [25].

Prostaglandin E2 (PGE-2) has been shown to inhibit proteoglycan synthesis and increase matrix degradation. Additionally, it was shown that patients with OA have high levels of PGE-2 in cartilage [26]. Furthermore, leukotriene B4, a strong leukocyte chemotaxis, has been shown to stimulate proinflammatory cytokines in human synovial fluid samples [27].

2.3.1.2 Proteases

Proteases are mediators that play a primary role in the catabolic process of OA. There are several proteases that have a role in the pathogenesis of OA. These proteases are collagenase-containing matrix metalloproteinases, cathepsin K-containing proteases, and serine-containing proteases. As the proteases degrade collagen, the related catabolic process results in the progression of matrix loss, since the cartilage’s response to damaged matrix repair is limited [28].

‘Agreccan’, the largest proteoglycan, provides cartilage elasticity. The ADAMTS family of enzymes, also called aggrecanases (ADAMTS-4-5), is involved in the early stage of OA degeneration and is responsible for aggrecan degradation [28].

Type-2 collagen, the most abundant type of collagen found in the cartilage tissue, provides cartilage tensile strength. It is broken down by collagenase-containing matrix metalloproteinases. MMP13 is considered to be the main collagenase responsible for cartilage destruction in OA [28].

Aggrecanase-2 (ADAMTS-5) and MMP-13 have an important place in the pathogenesis of OA. The development of specific inhibitors to these proteases in the context of the development of potential modifying treatments for OA has been of interest [29].

2.3.1.3 Molecular patterns associated with cartilage damage

Damage-associated molecular patterns (DAMPs) are molecules released from the chondrocytes in the damaged cartilage. DAMPs include extracellular matrix proteins, high mobility group box 1 protein (HMGB1), advanced glycation end products (AGEs) and receptor for advanced glycation endproducts (RAGEs), and alarmins [S100 calcium-binding protein A8 (S100A8) and S100 calcium-binding protein A9 (S100A9)].

It has been demonstrated that DAMPs have important roles in the pathogenesis of OA. DAMPs activate intercellular signaling pathways such as RAGE, toll-like receptors (TLR) and mitogen-activated protein kinases (MAPKs), thereby inducing the expression of catabolic proteases and inflammation-related genes [30]. DAMPs give rise to the increase in MMPs and activated macrophages which in turn lead to chondrocyte apoptosis and cause damage to the extracellular matrix (ECM) and cartilage [31, 32].

Fragmented matrix proteins such as cartilage oligomeric matrix protein (COMP), fibromodulin, proteoglycan, collagen, tenascin C, fibronectin, biglycan and aggregate are released from the damaged matrix. These fragmented matrix proteins stimulate the immune response. Consequentially, TLR and integrin are activated and the upregulation of the degenerative pathway is achieved [23, 33, 34, 35].

RAGE is a member of the immunoglobulin family. It is expressed in chondrocytes and macrophages. RAGE has been demonstrated to increase in OA joints. This increase causes the production of MMPs, which play a direct role in the pathogenesis of OA [36].

Alarmins are intracellular proteins secreted from bone cartilage or synovium in OA. It is an important member of DAMP family that has a role in the pathogenesis of OA [33].

HMGB1 is a nonhistone nuclear protein. Its release from the nucleus is associated with the apoptosis or necrosis or inflammatory stimulation of cells [37]. A significant increase occurs in the secretion of proinflammatory cytokines, chemokines and MMPs with the increase in HMGB1 secretion [38].

S100A8 and S100A9 are secreted from granulocytes, macrophages and monocytes. There is evidence that these proteins play a role in the cartilage damage and OA progression [39]. In addition to their catabolic effect, S100A8 and S100A9 lead to the formation of bone/osteophyte [40].

It has been demonstrated in the literature that the basic calcium phosphate (BCP) and calcium pyrophosphate dihydrate (CPPD) crystals, from among the inorganic calcium crystals, accumulate in the synovial fluid [41]. Calcium-containing crystals trigger the inflammatory process by either directly stimulating the chondrocytes or indirectly stimulating the immune system [30]. Additionally, it has been reported in the literature that monosodium urate crystals also trigger inflammation and cause cartilage damage [42].

2.3.1.4 Free oxygen radicals

The amount of free oxygen radicals and the extent of the DNA damage they cause are higher in OA cartilages than in cartilages without OA. Free oxygen radicals have an important place in OA progression, since they increase the synovial inflammation and cartilage destruction [43].

2.3.1.5 Biomechanical factors

Abnormal mechanical loading has an important role in the onset and progression of OA [44]. Abnormal mechanical loading may be caused by various factors such as obesity, joint alignment disorders or joint instability. Abnormal mechanical loading leads to mechanical damage in the joint and result in an increase in the release of matrix-degrading enzymes. Cartilage destruction products trigger inflammation and damage to the joint cartilage occurs through cytokine activation.

2.4 Genetics of osteoarthritis

The molecular processes underlying OA, which have a complex etiology, have become clearer through genetic and epigenetic studies. OA has been categorized as early-onset OA and late-onset OA. Genetic factors are more prominent in the early-onset OA. Genetic studies on the early-onset OA will provide a better understanding of the etiopathogenesis of the disease.

Family and twin studies have been conducted to reveal the genetic factors in OA [45]. To give a few examples, in the family studies conducted by Kellgren in UK and US, it was determined that there is a genetic component of the hand and knee OA [46], whereas in the study conducted by Lanyon et al., it was shown that the risk of radiographic hip OA is higher in the siblings of the patients with advanced hip OA [47].

Studies, in which monozygotic (MZ) and dizygotic (DZ) twins were compared, have shown that genetic factors are effective in OA. In one of these studies, it was shown that genetic factors are 39–65% effective on hand and knee OA radiographs, independently of environmental and demographic factors [48]. In another study, knee OA progression was investigated in 114 MZ and 195 DZ female twin couples. Consequentially, a higher correlation was found in the MZ twins than in the DZ twins in terms of both osteophyte and joint space narrowing, and the heritability was calculated as 62% for osteophyte progression and 72% for joint space narrowing progression. Additionally, it has been reported that the genetic effect on knee OA progression is more prominent in the medial compartment [49]. Furthermore, it has also been reported that the genetic effect differs according to the affected area in OA. Accordingly, the heritability was reported as 40%, 60%, 65% and 70% in the knee, hip, hand and spine regions, respectively [48].

Candidate gene studies have focused on many gene groups such as cartilage structural genes [collagen type II alpha 1 (COL2A1), collagen type IX alpha 3 (COL9A3), collagen type XI alpha 1 (COL11A1)], genes associated with bone mineral density (BMD) [vitamin D receptor (VDR), estrogen receptor 1 (ESR1)], genes associated with chondrocyte cell signal transduction (bone morphogenetic protein 5 (BMP5), frizzled-related protein B (FRZB), interleukin-4 receptor alpha (IL-4Rα)], inflammatory cytokine genes (IL-1, IL-10, TGFβ1, IL-6, TNFα) [50].

The finding that VDR gene polymorphism is associated with BMD lead to the studies on the possible relationship of VDR gene polymorphism with OA [51]. In this context, it was shown in a study conducted on 543 women in Finland that VDR polymorphism plays a role in the etiology of symmetrical hand OA [52].

The prevalence of knee OA is significantly higher in women than in men. This difference was atrributed to the estrogen receptor α (ERα), which is encoded by ESR1. Several polymorphisms in ESR1 [PvuII (rs2234693) and BtgI (rs2228480)] have been confirmed as risk factors for OA [53].

FRZB is a glycoprotein and plays a role in chondrocyte maturation and bone development. In Rotterdam and Genetics, Osteoarthritis and Progression (GARP) studies, R324G single nucleotide polymorphism (SNP) of the FRZB gene was found to be associated with generalized OA, whereas rs7775 and rs2888326 SNPs were found to be associated with knee and hip OA [50]. BMPs are bone-derived factors that can induce new bone formation. In a study conducted by Sharma et al. on BMP5 gene, rs1470527 and rs9382564 polymorphisms were shown to be significantly associated with knee OA [54].

The hypothesis put forward in candidate gene studies is still being investigated in terms of the genetic variant. Researchers favor genome-wide association studies (GWAS), which is a hypothesis-free approach, in the event that they think that candidate gene studies do not contribute much to the etiopathogenesis of the disease. GWAS allows the identification of genetic loci and the discovery of new genetic variants. In this context, GWAS contributes to the discovery of prognostic biomarkers that can contribute to early diagnosis and the identification of new areas that can be targeted by medical treatments [55, 56, 57, 58]. The number of OA genetic risk loci, most of which have small effect sizes, has increased to 90 in the GWAS studies carried out up till 2019 [59]. 56 new loci were identified in the two major OA analyzes published recently [59, 60]. First of these two studies, that is the deCODE (Decode Genetics, Iceland)-UKBB (UK Biobank, England) study, was conducted with more than 650,000 British and Icelandic citizens. 11.6 million genotype variants were examined within the scope of the said study, and 23 significant variants were detected in 22 loci [60]. Second of these studies, that is the Arthritis Research UK Osteoarthritis Genetics (arcOGEN)-UKBB study, was conducted with more than 455,000 British citizens. 17.5 million genotype variants were examined within the scope of the said study, and 65 significant variants were detected in 64 loci [61]. These studies, which were conducted via performing separate meta-analyses for the hip and knee OAs, are the largest OA GWAS studies published to date (Figure 1) [59].

Figure 1.

The new OA risk loci identified in either or/both of the deCODE-UKBB and arcOGEN-UKBB studies.

Genetic variations are grouped into single nucleotide substitutions (mutations and single nucleotide polymorphisms (SNPs)], insertions and deletions, copy number variations or short tandem repeats [62]. Variations in the genome underlie the differences between the individuals. The most common of these variations are SNPs. SNPs are considered to be associated with susceptibility to diseases [63]. The majority of the common diseases that give rise to SNPs, including OA, are considered to affect the transcription of nearby genes by altering the transcription factor binding [59].

Epigenetics plays an important role in the regulation of gene expression and is associated with the pathogenesis of a number of human diseases. The term epigenetics encompasses DNA and chromatin modifications and the functions related thereto, in addition to non-coding RNAs (ncRNAs). Epigenetic control of gene expression is necessary and essential for typical organism development and cell control [63]. Epigenetic changes are transmissible and reversible changes that do not change the nucleotide sequence but cause changes in gene expression [64]. Changes that occur within the gene itself cause structural changes in some synthesized proteins. These changes lead up to early onset-OA. Given the above considerations, epigenetics is a very important area in the diagnosis, prognosis and treatment of OA [63]. Three different epigenetic regulation are involved in the molecular pathogenesis of OA. These include DNA methylation, expression of non-coding RNAs [ncRNAs, microRNAs (miRNAs), long non-coding RNAs (lncRNAs), small nucleolar RNAs (snoRNAs)], histone modifications that regulate gene expression at transcriptional and/or post-transcriptional levels [65]. DNA methylation is the most studied epigenetic control mechanism. 5-methylcytosine is formed as a result of the addition of a methyl group to the 5′ position of cytosine in the CpG dinucleotide by DNA methyltransferase [DNA Mtase (DNMT)]. Methylation at gene promoter regions is associated with suppression of gene expression. On the other hand, methylation within the gene bodies is associated with increased gene expression [66, 67]. The candidate gene study conducted to examine DNA methylation of matrix-degrading proteases such as MMP3, MMP9, MMP13 and ADAMTS4 was the first study to describe the possible effect of DNA methylation in OA. In the said study, hypomethylation was demonstrated in the promoter regions of selected catabolic genes in OA chondrocytes, and it was found that this hypomethylation was associated with increased expression of the gene [68].

miRNAs are small ncRNAs, which consist of 19 to 25 nucleotides and function at the post-transcriptional level by binding and repressing the expression of specific mRNA targets. miRNAs are involved in different cellular pathways and play a role in OA and in maintaining cartilage homeostasis [69]. Despite the constantly increasing number of publications and miRs related to the pathogenesis of OA, there is still no miR biomarker, which has been validated for use in the early diagnosis of the disease. This has been atrributed in part to the fact that OA is a multifactorial heterogeneous disease [63].

lncRNAs are large RNA molecules comprising more than 200 nucleotides. Deregulated expression of lncRNAs plays an important role in inflammatory diseases. lncRNAs have been shown to be associated with OA progression and cartilage degeneration [70]. LncRNAs regulate gene expression at the post-transcriptional level via micro-RNAs and modulate transcriptional gene silencing through chromatin regulation [71].

2.5 New treatments and future in osteoarthritis

Since chronic low-severity inflammation is involved in OA, the development of drugs that act on pro-inflammatory cytokines has also become a new hope in the treatment of OA [72]. In a study, an intra-articular (IA) IL-1 receptor antagonist (IL-1Ra) was applied to the canine knee and it was reported that it reduced the number and size of osteophytes in the femoral condyle in the follow-ups [73]. However, in another study, anakinra, which is IL-1Ra, was applied IA to the knees of patients with OA. In this randomized controlled trial, they found no superior effect to placebo on pain and WOMAC scores [74].

It has been reported that the serum TNF levels of patients with OA are elevated. The positive results obtained with the use of TNF-α inhibitors especially in erosive hand osteoarthritis are promising. In the study of Magnano et al., 12 patients with erosive hand OA were treated with adalimumab (ADA) and reported a significant improvement in symptoms after 3 months [75].

Proteases are mediators that play a primary role in the catabolic process of OA. ‘Agreccan’, the largest proteoglycan, provides cartilage elasticity. The ADAMTS family of enzymes, also called aggrecanases (ADAMTS-4-5), is involved in the early stage of OA degeneration and is responsible for aggrecan degradation [28]. Preclinical studies of the molecule GSK2394002, which effectively inhibits ADAMTS 4 and 5, were discontinued because serious cardiovascular side effects were encountered in animal experiments with systemic use [76]. However, phase II studies on 114810, an IA administration molecule developed to reduce systemic side effects, are ongoing [77].

OA is a dynamic process triggered by various biochemical and mechanical factors, where destruction and repair are together. The fibroblast growth factor 3 (FGF-3) family, especially FGF 18, has an anabolic effect on human chondrocytes [78]. In a study including 549 patients with stage 2 and 3 knee OA, FGF-18 (sprifermin) IA was administered. An increase in tibiofemoral joint cartilage thickness has been reported up to 12 months. In the light of this information, it can be said that Sprifermin is currently one of the promising candidates for disease-modifying OA drug (DMOAD) [79].

The mechanism of action of platelet-rich plasma (PRP) is suggested to be that bioactive growth factors released from α granules in platelets stimulate tissue healing at high concentrations. In a meta-analysis of 16 studies, 1543 patients were examined; PRP and IA hyaluronic acid (HA) were compared. In terms of pain and functionality, it was found to be more effective than intra-articular HA injection [80]. However, the 2019 OARSI guidelines state that there is low-level evidence of the use of PRP in patients with knee, hip, and polyarticular OA and should not be used [81]. Larger randomized controlled studies with long-term follow-up are needed to elucidate its effects on tissue regeneration and delaying surgery.

In the light of these, the aim of OA treatment is to prevent disease formation or to provide regeneration of damaged tissue rather than eliminating the symptom. It would be more logical for DMOADs to be developed in the future to target the early stages of disease pathogenesis. For this reason, randomized double-blind controlled studies will contribute to the development of OA treatment.

References

  1. 1. Man GS, Mologhianu G. Osteoarthritis pathogenesis - a complex process that involves the entire joint. J Med Life. 2014 Mar 15;7(1):37-41. Epub 2014 Mar 25
  2. 2. Felson DT. An update on the pathogenesis and epidemiology of osteoarthritis. Radiol Clin North Am. 2004 Jan;42(1):1-9, v
  3. 3. van den Bosch MH, Blom AB, Schelbergen RF, Koenders MI, van de Loo FA, van den Berg WB, Vogl T, Roth J, van der Kraan PM, van Lent PL. Alarmin S100A9 Induces Proinflammatory and Catabolic Effects Predominantly in the M1 Macrophages of Human Osteoarthritic Synovium. J Rheumatol. 2016 Oct;43(10):1874-1884
  4. 4. Goldring MB. Cartilage and Chondrocytes. In: Firestein GBRG, SE, McInnes IB, O'dell, JR., ed. Kelley’s Textbook of Rheumatology. 9th ed: Saunders; 2013:33-60
  5. 5. Sophia Fox AJ, Bedi A, Rodeo SA. The basic science of articular cartilage: structure, composition, and function Sports Health. 2009;1(6):461-468
  6. 6. Sulzbacher I. Osteoarthritis: histology and pathogenesis. Wiener Medizinische Wochenschrift 2013;163(9-10):212-219
  7. 7. Waller KA, Zhang LX, Elsaid KA, Fleming BC, Warman ML, Jay GD. Role of lubricin and boundary lubrication in the prevention of chondrocyte apoptosis. Proc Natl Acad Sci U S A. 2013 Apr 9;110(15):5852-5857
  8. 8. Heinegard D, Lorenzo P, Saxne T. The articular cartilage. In: Hochberg M, Silman AJ, Smolen JS, Weinblatt ME, Weisman MH, eds. Rheumatology. 5th ed: Mosby; 2011:57-66
  9. 9. Mathiessen A, Conaghan PG. Synovitis in osteoarthritis: current understanding with therapeutic implications Arthritis Res Ther.2017; 19: 18
  10. 10. Doğanavşargil Yakut B. Osteoartritte patoloji.Hepgüler AS, editör. Osteoartrit. 1. Baskı. Ankara:Türkiye Klinikleri; 2020. p.16-26
  11. 11. Heinemeier KM, Schjerling P, Heinemeier J, et al. Radiocarbon dating reveals minimal collagen turnover in both healthy and osteoarthritic human cartilage. Sci Transl Med 2016; 8:346ra90
  12. 12. Pritzker KP, Gay S, Jimenez SA, Ostergaard K, Pelletier JP, Revell PA, et al. Osteoarthritis cartilage histopathology: grading and staging. Osteoarthritis Cartilage. 2006;14(1):13-29
  13. 13. Taljanovic MS, Graham AR, Benjamin JB, et al. Bone marrow edema pattern in advanced hip osteoarthritis: quantitative assessment with magnetic resonance imaging and correlation with clinical examination, radiographic findings, and histopathology. Skeletal Radiol 2008; 37:423
  14. 14. Oehler S, Neureiter D, Meyer-Scholten C, Aigner T. Subtyping of osteoarthritic synoviopathy. Clin Exp Rheumatol. 2002;20:633-
  15. 15. Battistelli M, Favero M, Burini D, Trisolino G, Dallari D, De Franceschi L, et al. Morphological and ultrastructural analysis of normal, injured and osteoarthritic human knee menisci. Eur J Histochem. 2019;63(1):11
  16. 16. Jarraya M, Roemer FW, Englund M, Crema MD, Gale HI, Hayashi D, et al. Meniscus morphology: Does tear type matter? A narrative review with focus on relevance for osteoarthritis research. Semin Arthritis Rheum. 2017;46(5):552-561
  17. 17. Huang Z, Ding C, Li T, Yu SP. Current status and future prospects for disease modification in osteoarthritis. Rheumatology (Oxford) 2018; 57:iv108
  18. 18. Petersson IF, Boegård T, Dahlström J, Svensson B, Heinegård D, Saxne T. Bone scan and serum markers of bone and cartilage in patients with knee pain and osteoarthritis. Osteoarthritis Cartilage. 1998 Jan;6(1):33-39
  19. 19. Fleischmann RM, Bliddal H, Blanco FJ, et al. A Phase II Trial of Lutikizumab, an Anti-Interleukin-1α/β Dual Variable Domain Immunoglobulin, in Knee Osteoarthritis Patients With Synovitis. Arthritis Rheumatol 2019; 71:1056
  20. 20. Kloppenburg M, Peterfy C, Haugen IK, et al. Phase IIa, placebo-controlled, randomised study of lutikizumab, an anti-interleukin-1α and anti-interleukin-1β dual variable domain immunoglobulin, in patients with erosive hand osteoarthritis. Ann Rheum Dis 2019; 78:413
  21. 21. Sohn DH, Sokolove J, Sharpe O, et al. Plasma proteins present in osteoarthritic synovial fluid can stimulate cytokine production via Toll-like receptor 4. Arthritis Res Ther 2012; 14:R7
  22. 22. de Hooge AS, van de Loo FA, Bennink MB, Arntz OJ, de Hooge P, van den Berg WB. Male IL-6 gene knock out mice developed more advanced osteoarthritis upon aging. Osteoarthritis Cartilage. 2005;13(1):66-73
  23. 23. Liu-Bryan R, Terkeltaub R. Emerging regulators of the inflammatory process in osteoarthritis. Nat Rev Rheumatol. 2015;11(1): 35-44
  24. 24. Wang Q, Rozelle AL, Lepus CM, et al. Identification of a central role for complement in osteoarthritis. Nat Med 2011; 17:1674
  25. 25. Hui W, Litherland GJ, Elias MS, Kitson GI, Cawston TE, Rowan AD, et al. Leptin produced by joint white adipose tissue induces cartilage degradation via upregulation and activation of matrix metalloproteinases. Ann Rheum Dis. 2012;71(3):455-462
  26. 26. Attur M, Al-Mussawir HE, Patel J, Kitay A, Dave M, Palmer G, et al. Prostaglandin E2 exerts catabolic effects in osteoarthritis cartilage: evidence for signaling via the EP4 receptor. J Immunol. 2008;181(7):5082-5088
  27. 27. He W, Pelletier JP, Martel-Pelletier J, Laufer S, Di Battista JA. Synthesis of interleukin 1beta, tumor necrosis factor-alpha, and interstitial collagenase (MMP-1) is eicosanoid dependent in human osteoarthritis synovial membrane explants: interactions with antiinflammatory cytokines. J Rheumatol. 2002; 29(3):546-553
  28. 28. Troeberg L, Nagase H. Proteases involved in cartilage matrix degradation in osteoarthritis. Biochim Biophys Acta 2012; 1824:133
  29. 29. Tonge DP, Pearson MJ, Jones SW. The hallmarks of osteoarthritis and the potential to develop personalised disease-modifying pharmacological therapeutics. Osteoarthritis Cartilage 2014; 22:609
  30. 30. Sokolove J, Lepus CM (2013) Role of inflammation in the pathogenesis of osteoarthritis: latest findings and interpretations. Ther Adv Musculoskelet Dis 5(2):77-94
  31. 31. Attur M, Belitskaya-Le’vy I, Oh C, Krasnokutsky S, Greenberg J, Samuels J, Smiles S, Lee S, Patel J, Al-Mussawir H, McDaniel G, Kraus VB, Abramson SB (2011) Increased interleukin-1b gene expression in peripheral blood leukocytes is associated with increased pain and predicts risk for progression of symptomatic knee osteoarthritis. Arthritis Rheumatol 63(7):1908-1917
  32. 32. Sun XH, Liu Y, Han Y, Wang J (2016) Expression and significance of high-mobility group protein B1 (HMGB1) and the receptor for advanced glycation end-product (RAGE) in knee osteoarthritis. Med Sci Monit 22:2105-2112
  33. 33. van den Bosch MHJ. Inflammation in osteoarthritis: is it time to dampen the alarm(in) in this debilitating disease? Clin Exp Immunol. 2019;195(2):153-166
  34. 34. Loeser RF, Goldring SR, Scanzello CR, Goldring MB. Osteoarthritis: a disease of the joint as an organ. Arthritis Rheum 2012; 64:1697
  35. 35. Sofat N. Analysing the role of endogenous matrix molecules in the development of osteoarthritis. Int J Exp Pathol 2009; 90:463
  36. 36. Loeser RF, Yammani RR, Carlson CS, Chen H, Cole A, Im HJ, Bursch LS, Yan SD (2005) Articular chondrocytes express the receptor for advanced glycation end products: potential role in osteoarthritis. Arthritis Rheumatol 52(8):2376-2385
  37. 37. Ke X, Jin G, Yang Y, Cao X, Fang R, Feng X,et al. Synovial Fluid HMGB-1 levels are associated with osteoarthritis severity. Clin Lab. 2015;61(7):809-818
  38. 38. Garcia-Arnandis I, Guillen MI, Gomar F, Pelletier JP, Martel-Pelletier J, Alcaraz MJ. High mobility group box 1 potentiates the pro-inflammatory effects of interleukin-1beta in osteoarthritic synoviocytes. Arthritis Res Ther. 2010;12(4):165
  39. 39. Zreiqat H, Belluoccio D, Smith MM, Wilson R, Rowley LA, Jones K, et al. S100A8 and S100A9 in experimental osteoarthritis. Arthritis Res Ther. 2010;12(1):16
  40. 40. Schelbergen RF, Geven EJ, van den Bosch MH, Eriksson H, Leanderson T, Vogl T, et al. Prophylactic treatment with S100A9 inhibitor paquinimod reduces pathology in experimental collagenase-induced osteoarthritis. Ann Rheum Dis. 2015;74(12):2254-2258
  41. 41. Fuerst M, Bertrand J, Lammers L, Dreier R, Echtermeyer F, Nitschke Y, et al. Calcification of articular cartilage in human osteoarthritis. Arthritis Rheum. 2009;60(9):2694-2703
  42. 42. Martinon F, Petrilli V, Mayor A, Tardivel A, Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature. 2006;440(7081):237-241
  43. 43. Lepetsos P, Papavassiliou AG. ROS/oxidative stress signaling in osteoarthritis. Biochim Biophys Acta. 2016;1862(4):576-591
  44. 44. Houard X, Goldring MB, Berenbaum F. Homeostatic mechanisms in articular cartilage and role of inflammation in osteoarthritis. Curr Rheumatol Rep. 2013;15(11):375
  45. 45. Spector TD, MacGregor AJ. Risk factorsforosteoarthritis: genetics.OsteoarthritisCartilage 2004;12: 39-44
  46. 46. Felson DT, Couropmitree NN, Chaisson CE et al. Evidencefor a Mendeliangene in a segregationanalysis of generalized radiographi costeoarthritis: theFraminghamStudy. ArthritisRheum 1998; 41: 1064-1071
  47. 47. Lanyon P, Muir K, Doherty S, Doherty M. Assessment of a geneticcontributiontoosteoarthritis of thehip: siblingstudy. BMJ. 2000;321(7270):1179-1183
  48. 48. Spector TD, Cicuttini F, Baker J et al. Geneticinfluences on osteoarthritisinwomen: a twinstudy. BMJ 1996; 312: 940-943
  49. 49. Zhai G, Hart DJ, Kato BS, MacGregor A, Spector TD. Geneticinfluence on theprogression of radiographickneeosteoarthritis: a longitudinaltwinstudy.Osteoarthritis Cartilage. 2007;15(2):222-5
  50. 50. Yucesoy B, Charles LE, Baker B, Burchfiel CM. Occupational and geneticrisk factors for osteoarthritis: a review. Work. 2015;50(2):261-273
  51. 51. Brandi ML, Gennari L, Cerinic MM et al. Geneticmarkers of osteoarticulardisorders: factsandhopes. ArthritisRes 2001; 3: 270-280
  52. 52. Solovieva S, Hirvonen A, Siivola P et al. Vitamin D receptor gene polymorphismsandsusceptibility of handosteoarthritis in Finnishwomen. ArthritisResTher 2006; 8: R20
  53. 53. Piva SR, Susko AM, Khoja SS, Josbeno DA, Fitzgerald GK, Toledo FG. Links between osteoarthritis and diabetes: implications for management from a physical activity perspective. Clin Geriatr Med. 2015 Feb;31(1):67-87
  54. 54. Sharma AC, Srivastava RN, Srivastava SR, Agrahari A, Singh A, Parmar D. Evaluation of theassociationbetween a single-nucleotidepolymorphism of bone morphogeneticproteins 5 gene and risk of kneeosteoarthritis. J PostgradMed. 2017 Jul-Sep;63(3):151-156
  55. 55. Kunkle BW, Grenier-Boley B, Sims R, Bis JC, Damotte V, NajAC,et al. Genetic meta-analysis of diagnosedAlzheimer'sdiseaseidentifiesnew risk lociandimplicatesAbeta, tau, immunityandlipidprocessing. NatGenet 2019;51(3):414e30
  56. 56. Sims R, van der Lee SJ, Naj AC, Bellenguez C, BadarinarayanN, Jakobsdottir J, et al. Rarecodingvariants in PLCG2, ABI3, andTREM2 implicatemicroglial-mediatedinnateimmunityinAlzheimer'sdisease. NatGenet 2017;49(9):1373e84
  57. 57. Schizophrenia Working Group of thePsychiatricGenomics C. Biologicalinsightsfrom 108 schizophrenia-associatedgeneticloci. Nature 2014;511(7510):421e7
  58. 58. Westra HJ, Martinez-Bonet M, Onengut-Gumuscu S, Lee A,Luo Y, Teslovich N, et al. Fine-mapping and functional studies high light potential causal variants for rheumatoid arthritis and type 1 diabetes. NatGenet 2018;50(10):1366e74
  59. 59. Reynard LN, Barter MJ. Osteoarthritisyear in review 2019: genetics, genomicsandepigenetics. OsteoarthritisCartilage. 2020 Mar;28(3):275-284
  60. 60. Styrkarsdottir U, Lund SH, Thorleifsson G, ZinkF,Stefansson OA, Sigurdsson JK, et al. Meta-analysis of Icelandicand UK datasets identifiesmissensevariants in SMO,IL11,COL11A1 and 13 more new loci associated witho steoarthritis.NatGenet 2018;50(12):1681e7
  61. 61. Tachmazidou I, Hatzikotoulas K, Southam L, Esparza-Gordillo J, Haberland V, Zheng J, Johnson T, Koprulu M, Zengini E, Steinberg J, Wilkinson JM, Bhatnagar S, Hoffman JD, Buchan N, Süveges D; arcOGENConsortium, Yerges-Armstrong L, Smith GD, Gaunt TR, Scott RA, McCarthy LC, Zeggini E. Identification of new therapeuti c targets foroste oarthritis through genome-wideanalyses of UK Biobankdata. NatGenet. 2019 Feb;51(2):230-236
  62. 62. Murphy K, Cooper A, Tobias ES. The Human Genome, Gene Regulation and Genomic Variation. In: Padmanabhan S, ed. Handbook of Farmaco genomics and Stratified Medicine: Elsevier; 2014:41-56
  63. 63. Peffers MJ, Balaskas P, Smagul A. Osteoarthritisyear in review 2017: geneticsandepigenetics. Osteoarthritis Cartilage. 2018 Mar;26(3):304-311
  64. 64. Nussbaum RL, McInnes RR, Willard HF. (2016). The Human Genome: Gene StructureandFunction. In. Thompson&Thompson Genetics in Medicine(8th ed.,pp. 21-43). Canada: ElsevierInc
  65. 65. Khan NM, Haqqi TM. Epigenetics in osteoarthritis: Potential of HDAC inhibitors as therapeutics. Pharmacol. Res. 2018, 128(1): 73-79
  66. 66. G. LevMaor, A. Yearim, G. Ast, Thealternative role of DNA methylation in splicingregulation, Trends in genetics : TIG 31(5) (2015) 274-280
  67. 67. A.K. Maunakea, R.P. Nagarajan, M. Bilenky, T.J. Ballinger, C. D'Souza, S.D. Fouse, B.E. Johnson, C. Hong, C. Nielsen, Y. Zhao, G. Turecki, A. Delaney, R. Varhol, N. Thiessen, K. Shchors, V.M. Heine, D.H. Rowitch, X. Xing, C. Fiore, M. Schillebeeckx, S.J. Jones, D. Haussler, M.A. Marra, M. Hirst, T. Wang, J.F. Costello, Conserved role of intragenic DNA methylation in regulatingalternativepromoters, Nature 466(7303) (2010) 253-257
  68. 68. H.I. Roach, N. Yamada, K.S. Cheung, S. Tilley, N.M. Clarke, R.O. Oreffo, S. Kokubun, F. Bronner, Association between the abnormal expression of matrix-degrading enzymes by human osteoarthriti cchondrocytes and demethylation of specificCpGsites in the promoter regions, Arthritisandrheumatism 52(10) (2005) 3110-3124
  69. 69. Del Real A, Perez-Campo FM, Fernandez AF, Sanudo C, Ibarbia CG, Perez-Nunez MI, et al. Differentialanalysis of genome-wide methylationand gene expression in mesenchymal stem cells of patients with fractures and osteoarthritis. Epigenetics 2017; 12: 113-122
  70. 70. Jiang SD, Lu J, Deng ZH, Li YS, Lei GH. LongnoncodingRNAs in osteoarthritis. Joint Bone Spine 2016
  71. 71. Kang M, Ren M, Li Y, Fu Y, Deng M, Li C. Exosome-mediated transfer of lncRNA PART1 inducesge fitinib resistance in esophageal squamous cell carcinoma via functioning as a competingendogenous RNA. J. Exp. Clin. CancerRes. 2018, 37(1): 1-16
  72. 72. Goldring MB, Otero M, Tsuchimochi K, Ijiri K, Li Y. Defining the roles of inflammatory and anabolic cytokines in cartilage metabolism. Ann Rheum Dis. 2008; 67 Suppl 3:iii75-iii82
  73. 73. Caron JP, Fernandes JC, Martel-Pelletier J, Tardif G, Mineau F, Geng C, et al. Chondroprotective effect of intraarticular injections of interleukin-1 receptor antagonist in experimental osteoarthritis. Suppression of collagenase- 1 expression. Arthritis Rheum. 1996; 39:1535-1544
  74. 74. Chevalier X, Goupille P, Beaulieu AD, Burch FX, Bensen WG, Conrozier T, et al. Intraarticular injection of anakinra in osteoarthritis of the knee: a multicenter, randomized, double-blind, placebo-controlled study. Arthritis Rheum. 2009; 61:344-352
  75. 75. Magnano MD, Chakravarty EF, Broudy C, Chung L, Kelman A, Hillygus J, et al. A pilot study of tumor necrosis factor inhibition in erosive inflammatory osteoarthritis of the hands. J Rheumatol. 2007; 34:1323-1327
  76. 76. Dubail J, Apte S. Insights on ADAMTS proteases and ADAMTS-like proteins from mammalian genetics Matrix Biol. 2015,44-46: 24-37
  77. 77. Blanqué R, Mollat P, Brebion F, et al. GLPG1972: A potent, selective, orally available ADAMTS- 5 inhibitor for the treatment of OA. Osteoarthritis Cartilage. 2018;25:S58
  78. 78. Davidson D, Blanc A, Filion D, Wang H, Plut P, Pfeffer G, et al. Fibroblast growth factor (FGF) 18 signals through FGF receptor 3 to promote chondrogenesis. J. Biol. Chem. 2005; 280:20509-20515
  79. 79. Lohmander LS, Hellot S, Dreher D, Krantz EF, Kruger DS, Guermazi A, et al. Intraarticular sprifermin (recombinant human fibroblast growth factor 18) in knee osteoarthritis: a randomized, double-blind, placebo-controlled trial. Arthritis Rheumatol. 2014;66: 1820-1831
  80. 80. Chang KV, Hung CY, Aliwarga F, Wang TG, Han DS, Chen WS. Comparative effectiveness of platelet-rich plasma injections for treating knee joint cartilage degenerative pathology: a systematic review and metaanalysis. Arch Phys Med Rehabil. 2014; 95:562-575
  81. 81. Bannuru RR, Osani MC, Vaysbrot EE, Arden NK, Bennell K, Bierma-Zeinstra SM, et al. OARSI guidelines for the non-surgical management of knee, hip, and polyarticular osteoarthritis. Osteoarthr cartilage 2019. S1063-4584(19)31116-1

Written By

Ferhat Ege

Submitted: 02 July 2021 Reviewed: 05 July 2021 Published: 29 August 2021