Defining the extracellular matrix in non-cartilage soft tissues in osteoarthritis – a systematic review ========================================================================================================= * Jolet Y. Mimpen * Iwan G. A. Raza * Sarah J. B. Snelling ## Abstract **Objective** Osteoarthritis (OA) is increasingly seen as a disease of global joint dysfunction, affecting not only cartilage but also the other joint tissues. Extracellular matrix (ECM) is a critical determinant of tissue mechanobiology, but ECM is poorly understood in osteoarthritic joint tissues beyond cartilage in human OA and animal models of OA. Therefore, we aimed to define the structural composition and architecture of non-cartilage soft joint tissue ECM in human OA, and to compare the ECM changes observed in humans to those seen in animal models of OA. **Design** A systematic search strategy, devised using relevant matrix, tissue, and disease nomenclature, was run through the MEDLINE, EMBASE, and Scopus databases. Demographic, clinical, and biological data were extracted from eligible studies. Bias analysis was performed. **Results** 142 studies were included, which covered capsule, ligaments, meniscus, skeletal muscle, synovium, and tendon in both humans and animals, and fat pad and intervertebral disc in humans only. Overall, included studies show that the expression of structural ECM components changes in disease within an ECM that becomes disorganised with increasing joint degeneration. **Conclusions** This systematic review consolidates existing knowledge of a poorly defined aspect of OA pathophysiology. Changes in ECM composition and architecture occur across soft joint tissues in OA, but most of these remain poorly defined due to the low number of studies and lack of healthy comparator groups. Further research to better understand the context within which cartilage is damaged in OA may enable a better understanding of OA and its potential treatments. **What is already known on this topic** * Extracellular matrix (ECM) is a critical determinant of tissue mechanobiology and cell behaviour, but it is poorly described in osteoarthritic joint tissues beyond cartilage. **What this study adds** * Our study highlights the global nature of ECM dysregulation across the osteoarthritic joint. In addition, this study describes practical and methodological challenges that should be addressed to improve the contribution of future studies to define the role of ECM in non-cartilage soft tissues in osteoarthritis. **How this study might affect research, practice or policy** * A better understanding of ECM changes and their underlying mechanisms throughout the osteoarthritic joint may assist with disease classification and patient stratification and also holds promise for the development of ECM-targeting treatments which could modify the pathogenic cell behaviour that may drive osteoarthritis progression. Keywords * osteoarthritis * extracellular matrix * human * animal models * synovium * meniscus ## 1 Introduction Osteoarthritis (OA) is the most common joint disease globally, affecting over 500 million people. OA is typically attributed to mechanically-driven joint damage and is characterised by articular cartilage degeneration and subchondral bone remodelling[1]. However, these tissues are not affected in isolation from the wider joint, with pathology in other soft joint tissues contributing to the symptoms and progression of OA[2, 3]. Damage to menisci and ligaments disrupts joint biomechanics, while inflammation, fibrosis, and distension of the synovium and joint capsule are associated with joint pain and stiffness[4–8]. Despite significant clinical need and substantial efforts to identify disease modifying OA drugs, there is no effective way of inhibiting or decelerating OA-related joint damage by targeting cartilage directly. Given the important role of other soft tissues in joint biomechanics and the release of pro-inflammatory and matrix-degrading mediators into the synovial fluid[9], understanding the whole joint context of OA might provide novel therapeutic strategies and prognostic markers. Joint tissues are rich in extracellular matrix (ECM), a network of structural and regulatory macromolecules within which cells are embedded[10]. The role of ECM as a major determinant of the biophysical properties of a tissue has clear relevance in a disease such as OA[11, 12]. ECM not only provides structure to the tissue, but can also affect cell function through receptor engagement, mechanical cues, and the sequestration of growth factors and cytokines[13–16]. Significant crosstalk occurs between cells and matrix components, such that pathological ECM may exacerbate cellular dysfunction in disease[15, 17]. Therefore, ECM composition and architecture cannot be disregarded when attempting to understand OA pathophysiology. However, outside of cartilage, ECM remodelling in OA has received relatively little attention. Studying OA in the clinical setting is challenging due to the slow and unpredictable nature of the course of the disease. In addition, clinical symptoms often appear late in the disease process, making it difficult to study its onset and early progression. Therefore, many animal models for OA have been developed and are used to overcome these issues and to facilitate the development and evaluation of new therapies and diagnostic tools[18]. However, since there is no single “gold standard” animal model that accurately reflects all aspects of human disease, a major challenge is selecting the “right” model for each study[19]. The main aim of this systematic review is to consolidate existing data describing ECM architecture and structural composition in the synovium, joint capsule, skeletal muscle, tendon, ligament, meniscus, intervertebral disc, and fat pad of osteoarthritic joints. Secondly, we aim to define the changes in ECM architecture and structural composition in these tissues in animal models of OA to address their ability to replicate human disease pathophysiology. ## 2 Methods This review was conducted according to a protocol registered on the PROSPERO database [CRD42021231241] and guidelines set out in the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement[20]. ### Database and search strategy The search strategy, written by JM and a medical librarian, can be found in the ***Supplementary Information*.** ECM components and architectural features were defined using National Centre for Biotechnology Information Medical Subject Heading terms. Non-cartilage soft joint tissues and disease nomenclature were also specified. The search strategy was validated against relevant papers identified in a preliminary literature search. The search strategy was run on the Ovid MEDLINE, Ovid EMBASE and Scopus platforms on 30 October 2020 and repeated on 1 October 2021. ### Eligibility criteria and screening Abstracts were de-duplicated in Mendeley Reference Manager (Elsevier B.V., Netherlands) before importing into the Covidence platform. The remaining studies were screened independently at title/abstract and full-text stages by two reviewers (IR & JM), with conflicts resolves through consensus or a third reviewer (SS). Included studies were required to have ≥3 OA participants. In human studies, eligible patients and controls were ≥18 years. Non-OA diseases, including inflammatory arthrides and crystalline arthropathies, were excluded. The presence of a valid control group was not a requirement for human studies. However, control groups were included if present and a minimum of 3 participants were included in this group. Valid control groups include tissues from healthy people or near-healthy tissues, including cadavers, individuals with osteosarcoma, and traumatic joint injuries provided that the comparator tissue was not directly damaged by the trauma. In contrast to human studies, all animal studies required a control group. Studies that induced OA unilaterally and only used a contralateral control joint were excluded, as non-physiological loading of the contralateral joint induces ECM remodelling[21, 22]. Excluded animal models included the genetic deletion of ECM components, the introduction of matrix-degrading enzymes into the joint, surgical damage of a tissue subsequently reported on, and the ovariectomised rat model, as this is more commonly used as a model for osteoporosis[23, 24]. Regarding outcome measures, included studies evaluated at least one of the following tissues: intervertebral disc, ligament, skeletal muscle, tendon, meniscus, articular capsule, synovium, and fat pad. Papers that only studied these tissues after treatment, including – but not limited to – surgical or drug treatment, or after these tissues were purposely injured to induce the development of OA, were excluded. Papers evaluating non-ECM tissue components (cells, cytokines, matrix-degrading enzymes) were ineligible for inclusion. Given the focus on structural ECM, regulatory matricellular proteins, as well as neoepitopes generated during ECM turnover, were not included. Studies using *in vitro* or *ex vivo* culture systems were excluded as the ECM proteins cells synthesise differ in culture and *in vivo*. Transcriptomic analyses were excluded as gene expression is a determinant, not a measure, of protein abundance. Finally, only English-language articles were included. ### Data extraction and bias analysis Data were extracted from all included studies by one reviewer (JM or IR) using a standardised extraction form in Microsoft Excel; the extraction was verified by the other reviewer. Where there was uncertainty, extraction was performed in duplicate by both reviewers. Number of participants (or animals) in each group was recorded as well as the presence/absence of a control group; if a control group was present, the control population and control tissue were described. For animal studies, the species, strain, and type of OA model were recorded. When available, participant age, sex, body mass index (BMI) and disease severity were recorded, as were the joint and tissue being studied. Relevant ECM components and architectural features were described; comparisons to control tissues and statistical analysis were noted when applicable. Results were grouped by tissue, followed by ECM feature, and finally the direction of change compared to control (increase, no change, decrease, or no control group present) and presented in Tables 1 (human studies) and 2 (animal studies). Due to the large number of different included ECM features, accepted research methods, and accepted measures of effect, a quantitative meta-analysis was not deemed appropriate. Bias analysis was performed by IR, with all included studies assessed using the 2015 Office of Health Assessment and Translation (OHAT) Risk of Bias Rating Tool for Human and Animal Studies. The results of the bias analysis can be found in Supplementary Table 3. ## 3 Results ### 3.1. Study overview 18,133 potentially relevant articles were identified by the search strategy (Figure 1). Following the removal of duplicates, 8,378 abstracts were screened. Of the 399 studies assessed for eligibility at full-text screening, 142 met all criteria for inclusion in this review. The characteristics of all included studies are summarised in ***Supplementary Tables 1 and 2 (human and animal studies, respectively).*** A schematic overview of the included studies can be found in Figure 2. ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/31/2023.08.31.23294625/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2023/08/31/2023.08.31.23294625/F1) Figure 1. PRISMA 2022 flow diagram. ![Figure 2.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2023/08/31/2023.08.31.23294625/F2.medium.gif) [Figure 2.](http://medrxiv.org/content/early/2023/08/31/2023.08.31.23294625/F2) Figure 2. Schematic overview of the study population, anatomical locations, and ECM features studied in the included studies. Created with BioRender.com. ### 3.2 Human studies Most studies investigated meniscus (n = 41) and synovium (n = 40), followed by ligaments (n = 14), tendon (n = 5), capsule (n = 5), skeletal muscle (n = 4), fat pad (n = 2) and intervertebral disc (n = 1) (Supplementary Table 1). Studies mostly commonly investigated the knee joint (n = 77), but papers on hip (n = 10), thumb (n = 2), temporomandibular joint (TMJ) (n = 2), shoulder (n = 2), and spine OA (n = 1) were also identified. While most studies on synovium, tendon, and capsule focussed on the presence/absence and distribution of specific ECM components, a large proportion of the papers on meniscus and ligaments investigated ECM architecture and viscoelastic properties (Table 1). View this table: [Table 1.](http://medrxiv.org/content/early/2023/08/31/2023.08.31.23294625/T1) Table 1. Structural ECM components and architectural features in non-cartilage soft tissues of human osteoarthritic joints. **Abbreviations:** AA = amino acid, ACL = anterior cruciate ligament, CILP = cartilage intermediate layer protein, CHS = chondroitin sulphate, CPPD = calcium pyrophosphate deposition, CT = computed tomography, DS = dermatan sulphate, ECM = extracellular matrix, EDA = ethylenediamine, ED + F = enzymatic digestion and fractionation, EM = electron microscopy, FCCIS = fine cationic colloidal iron staining, FITR = Fourier-transformed infrared spectroscopy, Fn = fibronectin, F-UAA-EP = Fractination - uronic acid assay – electrophoresis, GAG = glycosaminoglycan, HA = hyaluronic acid, H&E = haematoxylin and eosin, HS = heparan sulphate, IB = immunoblotting, IEC = ion exchange chromatography, IHC = immunohistochemistry, IP = immunoprecipitation, IRT = indentation relaxation tests, KL-grade = Kellgren-Lawrence grade, LC = liquid chromatography, LM = light microscopy, N/A = not applicable, NI = nanoindentation, NR = not reported, OA = osteoarthritis, PAS = periodic acid Schiff, PCL = posterior cruciate ligament, PLM = polarised light microscopy, qual = qualitative, quant = quantitative, s-quant = semi-quantitative, SPA = sandwich-binding protein assay, TEM = transmission electron microscopy, WB = western blot. #### 3.2.1 Capsule in human OA Of 5 studies which assessed the capsule (hip (n = 3), knee (n = 2)) [25–29], 4 were published before the year 2000. These studies covered both ECM components and architecture, but no two studies investigated the same ECM feature. Of note, DiFrancesco *et al.* (1995) studied several different ECM features (calcification, collagen fibre organisation, elastic fibres, and GAG/proteoglycan content) in parallel[27], providing an overview of this tissue in OA. #### 3.2.2 Fat pad in human OA Two studies were identified for infrapatellar fat pad[30, 31]. Grevenstein *et al.* (2020) showed no change in COMP content between OA and control fat pads[30] and Belluzzi *et al.* (2020) showed that there was a decrease in collagen I and III expression in OA fat pads compared to controls [31]. #### 3.2.3 Intervertebral disc in human OA One study was identified for intervertebral disc. Cheng *et al.* (1996) showed an increase in calcification with increasing OA grade in intervertebral discs[32]. #### 3.2.4 Ligaments in human OA Out of the 14 studies on ligaments, 12 studies focussed on ACL and/or PCL of the knee[33–44], and two studies looked at ligaments in the thumb, one covering the palmar beak ligament and one covering volar anterior oblique (AOL) and dorsoradial (DRL) ligaments[45, 46]. Studies mostly focussed on collagen fibre organisation, which generally decreased in OA compared to control[38, 39, 41]. Studies without controls report disorganised and irregular collagen fibre organisation in OA ligaments. Other identified studies confirmed the presence of aggrecan, collagens I, II, and III, and decorin, suggest an increase in calcification and GAG/proteoglycan content, and no change in overall collagen content. #### 3.2.5 Meniscus in human OA Studies on human meniscus (n = 41) covered a wide range of ECM components, architectural changes, as well as measurements of viscoelastic properties[47–87]. Most studies concur on an increase in calcification and GAG/proteoglycan content, and consistently show a decrease in collagen fibre diameter and organisation. The presence or change in many other ECM components has been studied, including aggrecan, biglycan, cartilage intermediate layer protein, collagens and collagen crosslinks, COMP, decorin, fibromodulin, GAG components, hydroxyproline, keratocan, lubricin, and lumican. Most notable, all proteomics studies included in this systematic review evaluated human OA meniscus, identifying a range of ECM and ECM-associated proteins[52, 76, 79]. Two of these studies (Folkesson *et al.* (2020) and Roller *et al. (*2015)) also analysed control samples and found several proteins to be changed in OA compared to control tissue. For example, both studies report an increase in collagen type VI alpha 1 and collagen type VI alpha 2 in OA, and Folkesson *et al.* (2020) found a change in protein abundance in several small leucin-rich proteoglycans, such as an increase lumican and decrease in decorin, an increase in the proteoglycans aggrecan and versican, and a decrease in collagens type III and V [52, 79]. Finally, the results on viscoelastic properties are conflicting: whereas some studies show an increase in elastic modulus [66] and instantaneous modulus[77], another study showed a decrease in these moduli [47]. #### 3.2.6 Skeletal muscle in human OA All four studies on human skeletal muscle studied the ECM components in the vastus medialis or vastus lateralis of the quadriceps muscle[88–91]. These studies demonstrated the presence [89] or increase [91] in collagens type I, III, and IV compared to control. In addition, these studies show the presence of calcification and laminin[88, 91], and an increase in collagen and GAG content[90]. #### 3.2.7 Synovium in human OA Synovial tissue was studied in several joints, including 17 studies on the knee[68, 92–107], 5 studies on hip[108–112], 6 studies used a combination of knee and hip synovium [113–118], 2 studies on TMJ[119, 120], and 10 studies did not report the joint site[121–130]. The ECM components that were most often studied in human synovium were collagens, fibronectins, and laminins. Other ECM features covered by the included studies are calcification, collagen content, collagen fibre organisation, collagen cross-links, COMP, elastin, fibromodulin, GAG components, latent TGF- β - binding protein 1, lumican, reticulin, and vitronectin. While the presence and tissue distribution of these components has been clearly shown by several studies, the changes between OA and normal tissue remains unclear with most studies lacking healthy control groups; instead, OA is often the comparator group in studies investigating rheumatoid arthritis. #### 3.2.8 Tendon in human OA Human tendon studies covered a range of different tendons across the body, including Achilles, (long head of) biceps, subscapularis, gluteus medius, and internal obturator[131–135]. Discordant results between studies of anatomically distinct tendons are unsurprising, but disagreement was also seen for two studies on biceps tendon. For example, GAG/proteoglycan content was increased in the long head of biceps and internal obturator tendon[134, 135], unchanged in another study on biceps tendon and subscapularis tendon[132], and decreased in gluteus medius tendon in OA compared to control[133]. Similarly, increased calcification was seen in obturator tendon[135], while there was no difference in subscapularis, and a decrease in biceps tendon[132]. In terms of architecture, 3 out of 4 studies reporting on collagen fibre organisation report a decrease in organisation[132, 133, 135], while the last reported no difference compared to control[134]. An increase in collagen fibre diameter was found in internal obturator and biceps tendon[132, 135], while no difference was seen in subscapularis and gluteus medius tendons[132, 133]. Finally, no difference was found in the protein expression of collagen I and II and decorin[134]. ### 3.3 Animal studies Animal studies followed a similar pattern as human studies regarding the most studied tissues: synovium (n = 15), meniscus (n = 12), ligament (n = 6), skeletal muscle (n = 1), tendon (n = 1), and capsule (n = 1) (Supplementary Table 2). A broad range of species, strains, and models were used, all looking at the stifle joint of these animals. Overall, these studies generally found increases in ECM components such as collagen and disrupted ECM architecture, including a decrease in collagen fibre organisation in most tissues (Table 2). Viscoelastic properties were mainly studied in meniscus, where the elastic and instantaneous modulus tended to decrease. View this table: [Table 2.](http://medrxiv.org/content/early/2023/08/31/2023.08.31.23294625/T2) Table 2. Structural ECM composition and architecture in animal models of OA. The studied joint in all studies is the stifle joint. Abbreviations: AA = amino acid, ACL = anterior cruciate ligament, ACLT = anterior cruciate ligament transection, AFM = atomic force microscopy, CT = computed tomography, DMM = destabilisation of the medial meniscus, ECM = extracellular matrix, EM = electron microscopy, GAG = glycosaminoglycan, H&E = haematoxylin and eosin, IHC = immunohistochemistry, IRT = indentation relaxation tests, LCL = lateral cruciate ligament, MCL = medial cruciate ligament, MIA = monoiodoacetate, Mx = meniscectomy, N/A = not applicable, NR = not reported, NZW = New Zealand white, OA = osteoarthritis, PAS = periodic acid Schiff, PCL = posterior cruciate ligament, qual = qualitative, quant = quantitative, s-quant = semi-quantitative, WB = western blot. #### 3.3.1 Capsule in animal models of OA Only one study was identified on capsule. Loeser *et al.* (2013) studied capsule in the DMM model in CD57BL/6 mice[136]. Collagen III was found to be diffusely expressed in OA capsule, and mostly present in vascular endothelium. Interestingly, this study also assessed the meniscus, ligament, and synovium, taking a whole-joint approach to OA; they report a diffuse distribution of collagen type III similar to capsule in ligaments and synovium, while there was a pericellular distribution in meniscus. #### 3.3.2 Ligament in animal models of OA Ligaments were studied in animal models of OA in 4 studies using mouse models[136–139], 1 study using a rabbit model[140], and 2 studies using sheep models[141, 142]. A decrease in collagen fibre organisation was reported by two studies[140, 141]. All other reported ECM features were only present in one study. These features include calcification, mineralisation, collagen II and III, collagen cross-links, collagen fibre diameter, GAG/proteoglycan content, and mechanical strength. #### 3.3.3. Meniscus in animal models of OA ECM changes in meniscus in animal models of OA were investigated by 5 studies using mouse models [136, 138, 143–145], 5 studies using rabbit models [146–150], 1 study using a rat model[151], and 1 study using a pig model[152]. Overall, these studies show an increase in calcification and collagen types I, II, III, and X, and a decrease in collagen fibre organisation. Most studies show a decrease in GAG/proteoglycan content and viscoelastic properties in at least parts of the meniscus. In addition, thickening of the collagen fibres and no change in fibromodulin were found. #### 3.3.4 Skeletal muscle in animal models of OA One study was identified that investigated skeletal muscle. Shi *et al.* (2020) studied the elastic modulus in biceps femoris and rectus femoris muscles in an adapted Videman method in rabbits; they report an increase in elastic modulus in OA compared to control[153]. #### 3.3.5 Synovium in animal models of OA Synovium was investigated in 2 studies using mouse models[136, 143], 10 studies using rat models[154–163], and 2 studies using rabbit models[164, 165], and 1 study using a sheep model of OA[141]. All studies on calcification, collagen content, and collagen I showed an increase in OA compared to control. However, results on collagen fibre organisation and collagen fibre diameter were less clear, with some studies reporting no change, while others reported a decrease in collagen fibre organisation and increase in collagen fibre diameter. Other studied features included type III collagen, COMP, fibromodulin, lubricin, and viscoelastic properties (elastic modulus), which were all only reported on by a single study. #### 3.3.6 Tendon in animal models of OA Tendon was investigated in one study by McErlain *et al.* (2008) using an ACL transection model in rats. They found calcification of the patellar tendon to be more common in OA than control animals[166]. ### 3.4 Bias analysis The risk of bias varied between studies but was generally high (Supplementary Table 3). The potential for confounding bias was common, with many human studies failing to report on the age, sex and BMI of participants. Frequently, OA diagnoses were stated without reference to the diagnostic criteria used. Most studies failed to report on the blinding of assessors, even when qualitative histological observations were made. Purely qualitative observations were common, although semi-quantitative scoring systems were increasingly used in more recent studies. However, many quantitative and semi-quantitative differences between healthy and osteoarthritic tissues were not statistically analysed. ## 4 Discussion Despite OA becoming more widely accepted as a whole joint disease, the role of and the changes to non-cartilage soft joint tissues remain underexplored. This study aimed to collate current knowledge on the structural ECM of these tissues to summarise and highlight gaps in existing knowledge. For instance, tissues such as the joint capsule and fat pad are very poorly defined, perhaps reflecting their perceived importance in OA. Overall, the studies included in this review show that the expression of many structural ECM components changes in disease, within an ECM that becomes less organised with increasing joint degeneration. Human studies covered a range of tissues and ECM features, but focused mainly on calcification, the expression of proteoglycans, and the expression, fibre diameter, and fibre organisation of collagens. While recent studies begin to define the presence and distribution of many ECM components, a frequent lack of well-defined controls limit understanding of the changes in disease. Most ECM features are only described by one or a few studies, highlighting the broader studies on the subject. While studies that did look at the same ECM feature mostly agreed, this was not always the case. This included studies with control groups that investigated the collagen content in meniscus[78, 83], elastic modulus in meniscus[47, 66], chondroitin sulphate in synovium[102, 118], and calcification and GAG/proteoglycan content in tendon[132–135], which all contradict each other in terms of the direction of change. The summary and results tables highlight several potential factors for these differences already, including different analysis methods, differences in tissue joint origin, and differences between the microanatomical area of tissue studied. This emphasizes the importance of in-depth reporting of tissue metadata and methods. Several recent human studies, mostly in ligaments, tendon, and meniscus, have begun to study both compositional and architectural ECM features within a single tissue. Importantly, such studies can begin to dissect the relationship, including causality, between changes in ECM composition, ECM architecture, and viscoelastic properties. For example, calcification of tendon has been shown to change its viscoelastic properties[167], while the mechanical properties of fibril-forming collagens are dependent on covalent cross-linking[168], and different matrix proteoglycans differ in their effects on cell-mediated collagen reorganisation[169]. Whole tissue proteomics, which can be used to study the ECM composition of a tissue holistically, was performed in three studies which all investigated OA meniscus[52, 76, 79]. While the study of ECM proteins using proteomic techniques are subject to methodological biases due to the fact that ECM proteins are large, heavily post-translationally modified, and highly insoluble[170], they are a powerful tool to better understand overall tissue composition and formulate new research questions. The application of this technique to other osteoarthritic tissues is likely to provide important insights. In animal models, OA is induced in a range of species using varied surgical techniques and pharmacological interventions, with no animal model truly replicating human disease[171, 172]. Joint mechanics, inflammatory responses, and disease chronicity all vary between animal models [172, 173]. If ECM remodelling also differs between species and procedures, it can be assumed that not all animal models are equally suited to the study of changes in osteoarthritic ECM. Certain models may be generally more representative of changes seen in human OA, or better suited to the study of particular joint tissues or ECM features. This review covers a range of ECM changes in several different musculoskeletal soft tissues across different species and models. Although limited animal studies were eligible for inclusion in this review, some changes in ECM features could be compared between human OA and animal models. Generally similar trends could be seen as in humans, including a decrease in collagen fibre organisation and an increase in calcification across ligaments, meniscus, and synovium. However, other observations seem to contradict those in humans; for example, the decreased expression of collagens in human osteoarthritic menisci [54, 83, 86] is not reflected in data from any animal models in this review [138, 144, 146]. Therefore, the models used by these studies, namely the mouse STR/ort, rabbit ACLT, and mouse DMM models, respectively, might not be suitable to infer OA-related changes in human menisci. These results emphasize that more studies on ECM changes in non-cartilage soft-joint tissues in human OA and animal models must be compared before the validity of the latter can be accurately defined. The strength of any systematic review is partly contingent on the quality of included studies. As discussed in section 3.4, the methodology of many studies conferred a high risk of bias, resulting in a low confidence in the evidence provided. In basic science studies utilising human samples, the baseline characteristics and clinical characterisation of OA patients are often missing, or lack necessary detail. Clinical background is a particularly important consideration in the context of soft tissue calcification, given that crystal depositional diseases, such as pseudogout, can drive OA pathology[174]. Patients’ clinical background is poorly reported throughout the literature, as is disease severity, despite ECM and other tissue components differing more from the physiological state with OA progression[38]. Although the search strategy covered many non-cartilage soft joint tissues, some tissues, such as the temporomandibular joint disc and acetabular labrum, were not included. In addition, the focus of this review was on structural components of the ECM, which are the elements that are studied most extensively and make up the majority of tissue ECM. However, this does mean that this work does not provide a complete account of all OA ECM, as non-structural matrix elements such as matricellular proteins or neoepitopes have not been reported on. Finally, a limitation of the review process is the data extraction, which was not done by two independent reviewers, but rather extracted by one reviewer and verified by the other reviewer. However, the effect of this is likely limited as a previous study has reported that while extraction by two independent reviewers is preferable, extraction by one reviewer with verification by a second reviewer has limited influence on the conclusions of a systematic review, especially considering a meta-analysis was not performed in the current work[175]. In the process of consolidating the current literature on this topic, this work highlights several practical and methodological challenges that have limited advances in knowledge of the understanding of structural ECM components, architectural features, and viscoelastic properties in non-cartilage soft tissues in OA. One of these problems, is the cross-sectional nature of studies. Cross-sectional studies are common across the OA field as tissues are only accessible at the time of joint replacement. Since OA can take decades to progress, the study of end-stage or advanced OA might not be very informative of the processes that are driving these changes. In addition, the lack of a healthy, or non-OA, comparator group in combination with the fact that many studies only report qualitative results, vastly reduces the depth of knowledge that can be gained from these studies. Finally, while many screened human and animal studies investigated both cartilage and other soft joint tissues, ECM is often studied exclusively in cartilage, with other features, such as cellularity and inflammatory markers being the focus in other tissues. This shows that while there is access to both the tissues and the methods to study ECM changes in non-cartilage soft tissues, their analysis is not seen as a priority. However, due to the limited definition of ECM in these tissues and their unknown contribution to disease development and progression, it is also possible that it remains unclear which ECM features should be focused on. Structural ECM encompasses a wide range of features that can be investigated with a plethora of different methods. To evaluate the most critical ECM features and applicable methods, studies investigating multiple ECM features in non-cartilage soft tissues across different stages of disease are required. Recent studies have started to highlight the importance of ECM as a determinant of tissue architecture and cell behaviour in disease. For example, a recent review has highlighted the changes in microenvironment in rheumatoid arthritis synovium that occur in early in the development of the disease, which form important extracellular cues that shape the pathogenic cell behaviour during the onset and progression of disease[176]. Therefore, they argue that understanding the ECM changes across different tissues in a particular disease might not only be able to help with disease classification and patient stratification but could also hold promise for the development of treatments that target ECM[176]. These treatments might not only be able to modify pathogenic cell behaviour that could be driving the disease, but also impact on joint stiffness, which is one of the most common symptoms of OA[177]. All in all, more research is needed to unravel the presence and distribution of different ECM components and architectural features in joint tissues in health and in (different stages of) OA and interplay with tissue-resident and tissue-infiltrating cells. Future research will also help to differentiate between the remodelling process in different joint tissues, which contain unique cell populations and are exposed to different mechanical and inflammatory stimuli in OA. ECM remodelling may also differ between synovial joints, given their varied anatomical locations, mechanical functions, and the presence of joint-specific tissues such as menisci. Potential variation in pathophysiology between osteoarthritic joints has received little attention, with the predominance of studies on knee OA likely due to high disease prevalence in this joint and tissue being relatively accessible during commonly performed knee replacements. Therefore, the future of this field is both dependent on the thorough investigation of ECM features in soft joint tissues across multiple OA joints and varied stages of disease progression, as well as the rigorous reporting of patient characteristics of all tissue donors. In conclusion, this systematic review gives an overview of the current knowledge of the presence and distribution of structural ECM components, as well as the changes in ECM components and architecture that occur throughout the osteoarthritic joint. Overall, the studies included in this review show that the expression of many structural ECM components changes in disease and that the ECM architecture becomes more disorganised with increasing joint degeneration. Furthermore, this review highlights practical and methodological challenges that have limited progress in the understanding of changes in ECM composition and architecture in non-cartilage soft tissues during OA development and progression, including the fact that historically studies have mainly focussed on other aspects of the disease in these tissues, such as inflammation. Given the role of ECM in influencing cell behaviour, further research to better understand the broad context within which cartilage is damaged in OA may enable a better understanding of the disease as well as potential treatments. ## Supporting information Supplementary [[supplements/294625_file02.pdf]](pending:yes) ## Author contributions Conceptualization, J.Y.M. and S.J.B.S.; Formal analysis, J.Y.M. and I.G.A.R.; Investigation, J.Y.M. and I.G.A.R.; Data curation, J.Y.M. and I.G.A.R.; Writing - original draft, J.Y.M. and I.G.A.R.; Writing - review & editing, S.J.B.S; Project Administration, J.Y.M. and S.J.B.S. ## Funding statement This work was supported by the National Institute for Health Research Oxford Biomedical Research Centre. JYM is funded by Versus Arthritis (22873) and was supported the Chan Zuckerberg Initiative (CZIF2019-002426). SJBS is funded by the Chan Zuckerberg Initiative (CZIF2019-002426 and CZIF2021-240342) and supported by the National Institute for Health Research Oxford Biomedical Research Centre. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript. ## Data availability statement The authors confirm that the data supporting the findings of this study are available within the article and its supplementary materials. In addition, the raw data from the data extraction process, which was used to populate Tables 1 and 2 and Supplementary Tables 1 and 2, is available upon reasonable request from the corresponding author (JYM). ## Competing interest The authors declare that they have no competing interests. ## Acknowledgements We would like to thank Oxford University medical librarian Eli Harriss for her support in generating and executing the search strategy. We would like to thank Dr Mathew Baldwin for his helpful feedback on the design of this study. * Received August 31, 2023. * Revision received August 31, 2023. * Accepted August 31, 2023. * © 2023, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial 4.0 International), CC BY-NC 4.0, as described at [http://creativecommons.org/licenses/by-nc/4.0/](http://creativecommons.org/licenses/by-nc/4.0/) ## References 1. 1.Goldring SR, Goldring MB (2016) Changes in the osteochondral unit during osteoarthritis: structure, function and cartilage–bone crosstalk. Nature Reviews Rheumatology 2016 12:11 12:632–644 2. 2.Mimpen JY, Snelling SJB (2019) Chondroprotective Factors in Osteoarthritis: a Joint Affair. Curr Rheumatol Rep 21:1–14 3. 3.Poole AR (2012) Osteoarthritis as a Whole Joint Disease. HSS Journal 8:4–6 4. 4.Zhang K, Li L, Yang L, Shi J, Zhu L, Liang H, Wang X, Yang X, Jiang Q (2019) The biomechanical changes of load distribution with longitudinal tears of meniscal horns on knee joint: A finite element analysis. J Orthop Surg Res 14:1–12 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13018-019-1165-2&link_type=DOI) 5. 5.Shirazi R, Shirazi-Adl A (2009) Analysis of partial meniscectomy and ACL reconstruction in knee joint biomechanics under a combined loading. Clinical Biomechanics 24:755–761 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.clinbiomech.2009.07.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19656595&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 6. 6.Wellsandt E, Gardinier ES, Manal K, Axe MJ, Buchanan TS, Snyder-Mackler L (2015) Decreased Knee Joint Loading Associated With Early Knee Osteoarthritis After Anterior Cruciate Ligament Injury. doi:101177/0363546515608475 44:143–151 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=101177/0363546515608475&link_type=DOI) 7. 7.Hill CL, Hunter DJ, Niu J, Clancy M, Guermazi A, Genant H, Gale D, Grainger A, Conaghan P, Felson DT (2007) Synovitis detected on magnetic resonance imaging and its relation to pain and cartilage loss in knee osteoarthritis. Ann Rheum Dis 66:1599–1603 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjEwOiI2Ni8xMi8xNTk5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 8. 8.Hill CL, Gale DG, Chaisson CE, Skinner K, Kazis L, Gale ME, Felson DT (2001) Knee effusions, popliteal cysts, and synovial thickening: association with knee pain in osteoarthritis. J Rheumatol 28: 9. 9.Sanchez-Lopez E, Coras R, Torres A, Lane NE, Guma M (2022) Synovial inflammation in osteoarthritis progression. Nature Reviews Rheumatology 2022 18:5 18:258–275 10. 10.Theocharis AD, Skandalis SS, Gialeli C, Karamanos NK (2016) Extracellular matrix structure. Adv Drug Deliv Rev 97:4–27 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.addr.2015.11.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26562801&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 11. 11.Urbanczyk M, Layland SL, Schenke-Layland K (2020) The role of extracellular matrix in biomechanics and its impact on bioengineering of cells and 3D tissues. Matrix Biology 85– 86:1–14 12. 12.Felson DT (2013) Osteoarthritis as a disease of mechanics. Osteoarthritis Cartilage 21:10–15 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2012.09.012&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23041436&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000314480200002&link_type=ISI) 13. 13.Klees RF, Salasznyk RM, Kingsley K, Williams WA, Boskey A, Plopper GE (2005) Laminin-5 Induces Osteogenic Gene Expression in Human Mesenchymal Stem Cells through an ERK-dependent Pathway. Mol Biol Cell 16:881 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6Im1vbGJpb2xjZWxsIjtzOjU6InJlc2lkIjtzOjg6IjE2LzIvODgxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 14. 14.Du J, Zu Y, Li J, Du S, Xu Y, Zhang L, Jiang L, Wang Z, Chien S, Yang C (2016) Extracellular matrix stiffness dictates Wnt expression through integrin pathway. Scientific Reports 2016 6:1 6:1– 12 15. 15.Allen JL, Cooke ME, Alliston T (2012) ECM stiffness primes the TGFβ pathway to promote chondrocyte differentiation. Mol Biol Cell 23:3731–3742 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6Im1vbGJpb2xjZWxsIjtzOjU6InJlc2lkIjtzOjEwOiIyMy8xOC8zNzMxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 16. 16.Wijelath ES, Rahman S, Namekata M, Murray J, Nishimura T, Mostafavi-Pour Z, Patel Y, Suda Y, Humphries MJ, Sobel M (2006) Heparin-II Domain of Fibronectin Is a Vascular Endothelial Growth Factor-Binding Domain. Circ Res 99:853–860 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNpcmNyZXNhaGEiO3M6NToicmVzaWQiO3M6ODoiOTkvOC84NTMiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wOC8zMS8yMDIzLjA4LjMxLjIzMjk0NjI1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 17. 17.Thomas CM, Murray R, Sharif M (2011) Chondrocyte apoptosis determined by caspase-3 expression varies with fibronectin distribution in equine articular cartilage. Int J Rheum Dis 14:290–297 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21816026&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 18. 18.McCoy AM (2015) Animal Models of Osteoarthritis: Comparisons and Key Considerations. Vet Pathol 52:803–818 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0300985815588611&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26063173&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 19. 19.Teeple E, Jay GD, Elsaid KA, Fleming BC (2013) Animal models of osteoarthritis: Challenges of model selection and analysis. AAPS Journal 15:438–446 20. 20.Page MJ, McKenzie JE, Bossuyt PM, et al. (2021) The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. doi:10.1136/BMJ.N71 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1136/BMJ.N71&link_type=DOI) 21. 21.Poulet B, de Souza R, Kent A V., Saxon L, Barker O, Wilson A, Chang YM, Cake M, Pitsillides AA (2015) Intermittent applied mechanical loading induces subchondral bone thickening that may be intensified locally by contiguous articular cartilage lesions. Osteoarthritis Cartilage 23:940–948 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2015.01.012&link_type=DOI) 22. 22.Zhu J, Zhu Y, Xiao W, Hu Y, Li Y (2020) Instability and excessive mechanical loading mediate subchondral bone changes to induce osteoarthritis. Ann Transl Med 8:350–350 23. 23.Kalu DN (1991) The ovariectomized rat model of postmenopausal bone loss. Bone Miner 15:175–191 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/0169-6009(91)90124-I&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1773131&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1991GV01200001&link_type=ISI) 24. 24.Yousefzadeh N, Kashfi K, Jeddi S, Ghasemi A (2020) Ovariectomized rat model of osteoporosis: a practical guide. EXCLI J 19:89 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.17179/excli2019-1990&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32038119&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 25. 25.Cameron HU, Macnab I (1973) Scanning electron microscopic studies of the hip joint capsule and synovial membrane. Canadian Journal of Surgery 16:388–392 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=4748419&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 26. 26.Campbell TM, Trudel G, Laneuville O (2015) Knee flexion contractures in patients with osteoarthritis: clinical features and histologic characterization of the posterior capsule. Pm & R 7:466–473 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.pmrj.2014.12.001&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25511691&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 27. 27.DiFrancesco L, Sokoloff L (1995) Lipochondral degeneration of capsular tissue in osteoarthritic hips. American Journal of Surgical Pathology 19:278–283 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7872426&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QJ44700005&link_type=ISI) 28. 28.Heinegård D, Hernborg J, Lundberg BJ, Heinegard D, Hernborg J, Lundberg BJ, Heinegård D, Hernborg J, Lundberg BJ (1968) The glycosaminoglycans of the human joint capsule: isolation and characterizaion. Arthritis Rheum 11:787–795 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=4236075&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 29. 29.Herbert C, Jayson MI V, Bailey AJ (1973) Joint capsule collagen in osteoarthrosis. Ann Rheum Dis 32:510–514 [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6MzoiUERGIjtzOjExOiJqb3VybmFsQ29kZSI7czoxMToiYW5ucmhldW1kaXMiO3M6NToicmVzaWQiO3M6ODoiMzIvNi81MTAiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wOC8zMS8yMDIzLjA4LjMxLjIzMjk0NjI1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 30. 30.Grevenstein D, Heilig J, Dargel J, Oppermann J, Eysel P, Brochhausen C, Niehoff A (2020) COMP in the Infrapatellar Fat Pad—Results of a Prospective Histological, Immunohistological, and Biochemical Case–Control Study. Journal of Orthopaedic Research 38:747–758 31. 31.Belluzzi E, Macchi V, Fontanella CG, et al. (2020) Infrapatellar Fat Pad Gene Expression and Protein Production in Patients with and without Osteoarthritis. Int J Mol Sci 21:21 32. 32.Cheng XG, Brys P, Nijs J, Nicholson P, Jiang Y, Baert AL, Dequeker J (1996) Radiological prevalence of lumbar intervertebral disc calcification in the elderly: an autopsy study. Skeletal Radiol 25:231–235 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s002560050070&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8741057&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 33. 33.Abdul Sahib NS, Al-Sharqi SAH, Wahab MS (2017) Study histopathological changes in the anterior and posterior cruciate ligament after knee replacement: Correlations with Vitamin D, calcium and c-reactive protein in Iraqi patients with osteoarthritis. Pakistan Journal of Biotechnology 14:393–400 34. 34.Akisue T, Stulberg BN, Bauer TW, McMahon JT, Wilde AH, Kurosaka M (2002) Histologic evaluation of posterior cruciate ligaments from osteoarthritic knees. Clin Orthop Relat Res 165–173 35. 35.Allain J, Goutallier D, Voisin MC (2001) Macroscopic and histological assessments of the cruciate ligaments in arthrosis of the knee. Acta Orthop Scand 72:266–269 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1080/00016470152846592&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11480602&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000169749500010&link_type=ISI) 36. 36.Komro J, Gonzales J, Marberry K, Main DC, Cramberg M, Kondrashov P (2020) Fibrocartilaginous metaplasia and neovascularization of the anterior cruciate ligament in patients with osteoarthritis. Clinical Anatomy 33:899–905 37. 37.Kumagai K, Sakai K, Kusayama Y, Akamatsu Y, Sakamaki K, Morita S, Sasaki T, Saito T, Sakai T (2012) The extent of degeneration of cruciate ligament is associated with chondrogenic differentiation in patients with osteoarthritis of the knee. Osteoarthritis Cartilage 20:1258– 1267 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2012.07.013&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22846713&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 38. 38.Levy YD, Hasegawa A, Patil S, Koziol JA, Lotz MK, D’Lima DD (2013) Histopathological changes in the human posterior cruciate ligament during aging and osteoarthritis: correlations with anterior cruciate ligament and cartilage changes. Ann Rheum Dis 72:271–277 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6IjcyLzIvMjcxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 39. 39.Marczak D, Kowalczewski J, Okon T, Synder M, Sibinski M (2017) An evaluation of the posterior cruciate ligament function in total knee arthroplasty with regard to its morphology and clinical properties. Folia Morphologica (Warszawa) 76:94–99 40. 40.Martins GC, Camanho G, Rodrigues MI, Filho LFM, Demange MK (2018) Histopathological analysis of the posterior cruciate ligament in primary osteoarthritis. European journal of orthopaedic surgery & traumatologie 28:691–699 41. 41.Nakahara H, Hasegawa A, Otabe K, Ayabe F, Matsukawa T, Onizuka N, Ito Y, Ozaki T, Lotz MK, Asahara H (2013) Transcription factor Mohawk and the pathogenesis of human anterior cruciate ligament degradation. Arthritis Rheum 65:2081–2089 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.38020&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23686683&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 42. 42.Nelissen RGHH, Hogendoorn PCW (2001) Retain or sacrifice the posterior cruciate ligament in total knee arthroplasty? A histopathological study of the cruciate ligament in osteoarthritic and rheumatoid disease. J Clin Pathol 54:381–384 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6OToiamNsaW5wYXRoIjtzOjU6InJlc2lkIjtzOjg6IjU0LzUvMzgxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 43. 43.Rajgopal A, Vasdev N, Pathak A, Gautam D, Vasdev A (2014) Histological changes and neural elements in the posterior cruciate ligament in osteoarthritic knees. Journal of Orthopaedic Surgery 22:142–145 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/230949901402200204&link_type=DOI) 44. 44.Zhu J, Zhang X, Ma Y, Zhou C, Ao Y (2012) Ultrastructural and Morphological Characteristics of Human Anterior Cruciate Ligament and Hamstring Tendons. Anatomical Record 295:1430– 1436 45. 45.Doerschuk SH, Hicks DG, Chinchilli VM, Pellegrini Jr. VD (1999) Histopathology of the palmar beak ligament in trapeziometacarpal osteoarthritis. Journal of Hand Surgery - American Volume 24:496–504 46. 46.Mobargha N, Ludwig C, Ladd AL, Hagert E (2014) Ultrastructure and innervation of thumb carpometacarpal ligaments in surgical patients with osteoarthritis. Clin Orthop Relat Res 472:1146–1154 47. 47.Abraham AC, Pauly HM, Donahue TL, Haut Donahue TL (2014) Deleterious effects of osteoarthritis on the structure and function of the meniscal enthesis. Osteoarthritis Cartilage 22:275–283 48. 48.Atik OS, Erdogan D, Seymen CM, Bozkurt HH, Kaplanoglu GT (2016) Is there crosstalk between subchondral bone, cartilage, and meniscus in the pathogenesis of osteoarthritis? Eklem Hastalik Cerrahisi 27:62–67 49. 49.Battistelli M, Favero M, Burini D, et al. (2019) Morphological and ultrastructural analysis of normal, injured and osteoarthritic human knee menisci. European Journal of Histochemistry 63:11 50. 50.Dessombz A, Nguyen C, Ea HK, et al. (2013) Combining μX-ray fluorescence, μXANES and μXRD to shed light on Zn2+ cations in cartilage and meniscus calcifications. Journal of Trace Elements in Medicine and Biology 27:326–333 51. 51.Fischenich KM, Lewis J, Kindsfater KA, Bailey TS, Haut Donahue TL (2015) Effects of degeneration on the compressive and tensile properties of human meniscus. J Biomech 48:1407–1411 52. 52.Folkesson E, Turkiewicz A, Ali N, Ryden M, Hughes H V, Tjornstrand J, Onnerfjord P, Englund M (2020) Proteomic comparison of osteoarthritic and reference human menisci using data-independent acquisition mass spectrometry. Osteoarthritis Cartilage 28:1092–1101 53. 53.Fuhrmann IK, Steinhagen J, Ruther W, Schumacher U, Rüther W, Schumacher U, Ruther W, Schumacher U (2015) Comparative immunohistochemical evaluation of the zonal distribution of extracellular matrix and inflammation markers in human meniscus in osteoarthritis and rheumatoid arthritis. Acta Histochem 117:243–254 54. 54.Ghosh P, Ingman AM, Taylor TK (1975) Variations in collagen, non-collagenous proteins, and hexosamine in menisci derived from osteoarthritic and rheumatoid arthritic knee joints. Journal of Rheumatology 2:100–107 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1185729&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1975AB82500014&link_type=ISI) 55. 55.Haut Donahue TL, Pauly HM (2021) Osteoarthritic meniscal entheses exhibit altered collagen fiber orientation. Connect Tissue Res 1–5 56. 56.Hino T, Furumatsu T, Miyazawa S, Fujii M, Kodama Y, Kamatsuki Y, Okazaki Y, Masuda S, Okazaki Y, Ozaki T (2020) A histological study of the medial meniscus posterior root tibial insertion. Connect Tissue Res 61:546–553 57. 57.Ishizuka S, Sakai T, Hiraiwa H, et al. (2016) Hypoxia-inducible factor-2α induces expression of type X collagen and matrix metalloproteinases 13 in osteoarthritic meniscal cells. Inflammation Research 65:439–448 58. 58.Jacquet C, Erivan R, Argenson JN, Parratte S, Ollivier M (2018) Effect of 3 Preservation Methods (Freezing, Cryopreservation, and Freezing + Irradiation) on Human Menisci Ultrastructure: An Ex Vivo Comparative Study With Fresh Tissue as a Gold Standard. American Journal of Sports Medicine 46:2899–2904 59. 59.Jacquet C, Erivan R, Sharma A, Pithioux M, Parratte S, Argenson JN, Ollivier M (2019) Preservation Methods Influence the Biomechanical Properties of Human Lateral Menisci: An Ex Vivo Comparative Study of 3 Methods. Orthop J Sports Med 7:2325967119841622 60. 60.Johnson K, Hashimoto S, Lotz M, Pritzker K, Goding J, Terkeltaub R (2001) Up-regulated expression of the phosphodiesterase nucleotide pyrophosphatase family member PC-1 is a marker and pathogenic factor for knee meniscal cartilage matrix calcification. Arthritis Rheum 44:1071–1081 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/1529-0131(200105)44:5<1071::AID-ANR187>3.0.CO;2-3&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11352238&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000171751000012&link_type=ISI) 61. 61.Karjalainen VP, Kestila I, Finnila MA, Folkesson E, Turkiewicz A, Onnerfjord P, Hughes V, Tjornstrand J, Englund M, Saarakkala S (2021) Quantitative three-dimensional collagen orientation analysis of human meniscus posterior horn in health and osteoarthritis using micro-computed tomography. Osteoarthritis Cartilage 29:762–772 62. 62.Karube S, Shoji H (1981) Compositional changes of glycosaminoglycans of the human menisci with age and degenerative joint disease. Journal of the Japanese Orthopaedic Association 56:51–57 63. 63.Katsuragawa Y, Saitoh K, Tanaka N, et al. (2010) Changes of human menisci in osteoarthritic knee joints. Osteoarthritis Cartilage 18:1133–1143 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2010.05.017&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20633672&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000282459500004&link_type=ISI) 64. 64.Kiraly AJ, Roberts A, Cox M, Mauerhan D, Hanley E, Sun Y (2017) Comparison of Meniscal Cell-Mediated and Chondrocyte-Mediated Calcification. Open Orthop J 11:225–233 65. 65.Kodama Y, Furumatsu T, Maehara A, Ozaki T (2018) Composition of Cell Clusters in Torn Menisci and Their Extracellular Matrix Components. Acta Med Okayama 72:499–506 66. 66.Kwok J, Grogan S, Meckes B, Arce F, Lal R, D’Lima D (2014) Atomic force microscopy reveals age-dependent changes in nanomechanical properties of the extracellular matrix of native human menisci: Implications for joint degeneration and osteoarthritis. Nanomedicine 10:1777–1785 67. 67.Lopez-Franco M, Lopez-Franco O, Murciano-Anton MA, Canamero-Vaquero M, Fernandez-Acenero MJ, Herrero-Beaumont G, Gomez-Barrena E (2016) Meniscal degeneration in human knee osteoarthritis: in situ hybridization and immunohistochemistry study. Arch Orthop Trauma Surg 136:175–183 68. 68.Masuda I, Ishikawa K, Usuku G (1991) A histologic and immunohistochemical study of calcium pyrophosphate dihydrate crystal deposition disease. Clin Orthop Relat Res 272–287 69. 69.McDaniel D, Tilton E, Dominick K, Flory K, Ernest T, Johnson JC, Main DC, Kondrashov P (2017) Histological characteristics of knee menisci in patients with osteoarthritis. Clinical Anatomy 30:805–810 70. 70.Melrose J, Fuller ES, Roughley PJ, Smith MM, Kerr B, Hughes CE, Caterson B, Little CB (2008) Fragmentation of decorin, biglycan, lumican and keratocan is elevated in degenerate human meniscus, knee and hip articular cartilages compared with age-matched macroscopically normal and control tissues. Arthritis Res Ther. doi:10.1186/ar2453 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/ar2453&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=18620607&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 71. 71.Mine T, Ihara K, Kawamura H, Date R, Umehara K (2013) Collagen expression in various degenerative meniscal changes: an immunohistological study. Journal of Orthopaedic Surgery 21:216–220 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/230949901302100221&link_type=DOI) 72. 72.Monibi FA, Pannellini T, Otero M, Warren RF, Rodeo SA (2021) Histologic and molecular features in pathologic human menisci from knees with and without osteoarthritis. Journal of Orthopaedic Research 01:1 73. 73.Musumeci G, Trovato FM, Loreto C, Leonardi R, Szychlinska MA, Castorina S, Mobasheri A (2014) Lubricin expression in human osteoarthritic knee meniscus and synovial fluid: a morphological, immunohistochemical and biochemical study. Acta Histochem 116:965–972 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.acthis.2014.03.011&link_type=DOI) 74. 74.Nagata N, Koshino T, Saito T (2000) Up-regulation of CD44-positive cells in medial meniscus of medial compartmental osteoarthritis of the knee. Knee 7:3–9 75. 75.Park DY, Min BH, Choi BH, et al. (2015) The Degeneration of Meniscus Roots Is Accompanied by Fibrocartilage Formation, Which May Precede Meniscus Root Tears in Osteoarthritic Knees. American Journal of Sports Medicine 43:3034–3044 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0363546515605086&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26430056&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 76. 76.Park J, Lee HS, Go EB, Lee JY, Kim JY, Lee SY, Lee DH (2021) Proteomic analysis of the meniscus cartilage in osteoarthritis. Int J Mol Sci 22:30 77. 77.Pordzik J, Bernstein A, Mayr HO, Latorre SH, Maks A, Schmal H, Seidenstuecker M (2020) Analysis of proteoglycan content and biomechanical properties in arthritic and arthritis-free menisci. Applied Sciences (Switzerland) 10:1–12 78. 78.Roller BL, Monibi FA, Stoker AM, Kuroki K, Bal BS, Cook JL (2015) Characterization of knee meniscal pathology: correlation of gross, histologic, biochemical, molecular, and radiographic measures of disease. J Knee Surg 28:175–182 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24807193&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 79. 79.Roller BL, Monibi F, Stoker AM, Bal BS, Stannard JP, Cook JL (2015) Characterization of Meniscal Pathology Using Molecular and Proteomic Analyses. J Knee Surg 28:496–505 80. 80.Sladojevic I, Krivokuca Z, Gajanin V, Manojlovic S (2016) Expression of Collagen Type I in Unaltered and Osteoarthritic Menisci of Knee Joint. Med Pregl 69:16–23 81. 81.Son M, Goodman SB, Chen W, Hargreaves BA, Gold GE, Levenston ME (2013) Regional variation in T1rho and T2 times in osteoarthritic human menisci: correlation with mechanical properties and matrix composition. Osteoarthritis Cartilage 21:796–805 82. 82.Sun Y, Mauerhan DR, Honeycutt PR, Kneisl JS, Norton HJ, Zinchenko N, Hanley Jr EN, Gruber HE (2010) Calcium deposition in osteoarthritic meniscus and meniscal cell culture. Arthritis Res Ther. doi:10.1186/ar2968 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/ar2968&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20353559&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 83. 83.Sun Y, Mauerhan DR, Kneisl JS, James Norton H, Zinchenko N, Ingram J, Hanley Jr. EN, Gruber HE (2012) Histological examination of collagen and proteoglycan changes in osteoarthritic menisci. Open Rheumatol J 6:24–32 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22550551&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 84. 84.Takahashi M, Suzuki M, Kushida K, Hoshino H, Inoue T (1998) The effect of aging and osteoarthritis on the mature and senescent cross-links of collagen in human meniscus. Arthroscopy - Journal of Arthroscopic and Related Surgery 14:366–372 85. 85.Wang J, Roberts S, Kuiper JH, Zhang W, Garcia J, Cui Z, Wright K (2020) Characterization of regional meniscal cell and chondrocyte phenotypes and chondrogenic differentiation with histological analysis in osteoarthritic donor-matched tissues. Sci Rep 10:21658 86. 86.Warnecke D, Balko J, Haas J, et al. (2020) Degeneration alters the biomechanical properties and structural composition of lateral human menisci. Osteoarthritis Cartilage 28:1482–1491 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2020.07.004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=32739340&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 87. 87.Zhang D, Cheriyan T, Martin SD, Schmid TM, Spector M (2012) Lubricin Distribution in the Menisci and Labra of Human Osteoarthritic Joints. Cartilage 3:165–172 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/1947603511429699&link_type=DOI) 88. 88.Fink B, Egl M, Singer J, Fuerst M, Bubenheim M, Neuen-Jacob E (2007) Morphologic changes in the vastus medialis muscle in patients with osteoarthritis of the knee. Arthritis Rheum 56:3626–3633 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.22960&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17968889&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000250891000015&link_type=ISI) 89. 89.Mattiello-Sverzut AC, Petersen SG, Kjaer M, Mackey AL (2013) Morphological adaptation of muscle collagen and receptor of advanced glycation end product (RAGE) in osteoarthritis patients with 12 weeks of resistance training: influence of anti-inflammatory or glucosamine treatment. Rheumatol Int 33:2215–2224 90. 90.Noehren B, Kosmac K, Walton RG, Murach KA, Lyles MF, Loeser RF, Peterson CA, Messier SP (2018) Alterations in quadriceps muscle cellular and molecular properties in adults with moderate knee osteoarthritis. Osteoarthritis Cartilage 26:1359–1368 91. 91.Serrao PRMS, Vasilceac FA, Gramani-Say K, Lessi GC, Reiff RBM, Mattiello-Sverzut AC, Mattiello SM (2014) Expression of receptors of advanced glycation end product (RAGE) and types I, III and IV collagen in the vastus lateralis muscle of men in early stages of knee osteoarthritis. Connect Tissue Res 55:331–338 92. 92.Cillero-Pastor B, Eijkel GB, Blanco FJ, Heeren RM (2015) Protein classification and distribution in osteoarthritic human synovial tissue by matrix-assisted laser desorption ionization mass spectrometry imaging. Anal Bioanal Chem 407:2213–2222 93. 93.Cutolo M, Picasso M, Ponassi M, Sun MZ, Balza E (1992) Tenascin and fibronectin distribution in human normal and pathological synovium. Journal of Rheumatology 19:1439–1447 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1279171&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1992JN26900026&link_type=ISI) 94. 94.Di Cesare PE, Fang C, Leslie MP, Della Valle CJ, Gold JM, Tulli H, Perris R, Carlson CS (1999) Localization and expression of cartilage oligomeric matrix protein by human rheumatoid and osteoarthritic synovium and cartilage. Journal of Orthopaedic Research 17:437–445 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jor.1100170321&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10376735&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 95. 95.Ea HK, Chobaz V, Nguyen C, et al. (2013) Pathogenic Role of Basic Calcium Phosphate Crystals in Destructive Arthropathies. PLoS ONE [Electronic Resource]. doi:10.1371/journal.pone.0057352 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0057352&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23468973&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 96. 96.Ene R, Sinescu RD, Ene P, Cirstoiu MM, Cirstoiu FC (2015) Synovial inflammation in patients with different stages of knee osteoarthritis. Rom J Morphol Embryol 56:169–173 97. 97.Fan L, Wang Q, Liu R, Zong M, He D, Zhang H, Ding Y, Ma J (2012) Citrullinated fibronectin inhibits apoptosis and promotes the secretion of pro-inflammatory cytokines in fibroblast-like synoviocytes in rheumatoid arthritis. Arthritis Res Ther. doi:10.1186/ar4112 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/ar4112&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23217276&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 98. 98.Kaufmann J, Mueller A, Voigt A, Carl HD, Gursche A, Zacher J, Stein G, Hein G (2003) Hydroxypyridinium collagen crosslinks in serum, urine, synovial fluid and synovial tissue in patients with rheumatoid arthritis compared with osteoarthritis. Rheumatology 42:314–320 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/rheumatology/keg102&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12595629&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 99. 99.Kragstrup TW, Sohn DH, Lepus CM, Onuma K, Wang Q, Robinson WH, Sokolove J (2019) Fibroblast-like synovial cell production of extra domain A fibronectin associates with inflammation in osteoarthritis. BMC Rheumatol 3:46 100.100.Nakashima K, Koshino T, Saito T (1998) Synovial immunohistochemical changes after high tibial osteotomy for osteoarthritis of the knee. Two-year prospective follow-up. Bull Hosp Joint Dis 57:187–194 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9926257&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 101.101.Nikkari L, Haapasalmi K, Aho H, Torvinen A, Sheppard D, Larjava H, Heino J (1995) Localization of the αV subfamily of integrins and their putative ligands in synovial lining cell layer. Journal of Rheumatology 22:16–23 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7535359&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QA69300005&link_type=ISI) 102.102.Nishida K, Inoue H, Toda K, Murakami T (1995) Localization of the glycosaminoglycans in the synovial tissues from osteoarthritic knees. Acta Med Okayama 49:287–294 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8770237&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 103.103.Rafael MS, Cavaco S, Viegas CSB, Santos S, Ramos A, Willems BAG, Herfs M, Theuwissen E, Vermeer C, Simes DC (2014) Insights into the association of Gla-rich protein and osteoarthritis, novel splice variants and gamma-carboxylation status. Mol Nutr Food Res 58:1636–1646 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/mnfr.201300941&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 104.104.Richardot P, Charni-Ben Tabassi N, Toh L, Marotte H, Bay-Jensen AC, Miossec P, Garnero P (2009) Nitrated type III collagen as a biological marker of nitric oxide-mediated synovial tissue metabolism in osteoarthritis. Osteoarthritis Cartilage 17:1362–1367 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19467351&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 105.105.Saito I, Koshino T, Nakashima K, Uesugi M, Saito T (2002) Increased cellular infiltrate in inflammatory synovia of osteoarthritic knees. Osteoarthritis Cartilage 10:156–162 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/joca.2001.0494&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11869075&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000173602700008&link_type=ISI) 106.106.Takahashi M, Kushida K, Hoshino H, Suzuki M, Sano M, Miyamoto S, Inoue T (1996) Concentrations of pyridinoline and deoxypyridinoline in joint tissues from patients with osteoarthritis or rheumatoid arthritis. Ann Rheum Dis 55:324–327 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6IjU1LzUvMzI0IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 107.107.Wang X, Dong C, Li N, Ma Q, Yun Z, Cai C, An M, Ma B (2018) Modulation of TGF-β activity by latent TGF-β-binding protein 1 in human osteoarthritis fibroblast-like synoviocytes. Mol Med Rep 17:1893–1900 108.108.Christensen AF, Sorensen GL, Junker K, Revald P, Varnum C, Issa SF, Junker P, Sorensen FB (2019) Site-specific absence of microfibrillar-associated protein 4 (MFAP4) from the internal elastic membrane of arterioles in the rheumatoid arthritis synovial membrane: an immunohistochemical study in patients with advanced rheumatoid arthritis versus osteoar. APMIS 127:588–593 109.109.Konttinen YT, Li TF, Xu JW, et al. (1999) Expression of laminins and their integrin receptors in different conditions of synovial membrane and synovial membrane-like interface tissue. Ann Rheum Dis 58:683–690 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjk6IjU4LzExLzY4MyI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIzLzA4LzMxLzIwMjMuMDguMzEuMjMyOTQ2MjUuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 110.110.Konttinen YT, Li TF, Mandelin J, Ainola M, Lassus J, Virtanen I, Santavirta S, Tammi M, Tammi R (2001) Hyaluronan synthases, hyaluronan, and its CD44 receptor in tissue around loosened total hip prostheses. Journal of Pathology 194:384–390 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/1096-9896(200107)194:3<384::AID-PATH896>3.0.CO;2-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11439372&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 111.111.Li TF, Xu JW, Santavirta S, Nordsletten L, Michelsson O, Takagi M, Virtanen I, Konttinen YT (2000) Distribution of fibronectins and their integrin receptors in interface tissue from aseptic loosening of hip prostheses. Clin Exp Rheumatol 18:221–225 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10812495&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 112.112.Turdean SG, Jung I, Gurzu S, Zazgyva A, Fetyko A, Roman CO, Turcu M, Pop TS (2017) Histopathological evaluation and expression of the pluripotent mesenchymal stem cell-like markers CD105 and CD44 in the synovial membrane of patients with primary versus secondary hip osteoarthritis. Journal of Investigative Medicine 65:363–369 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamltIjtzOjU6InJlc2lkIjtzOjg6IjY1LzIvMzYzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 113.113.Mapp PI, Revell PA (1985) Fibronectin production by synovial intimal cells. Rheumatol Int 5:229–237 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF00541341&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=4070925&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1985ATN8000007&link_type=ISI) 114.114.Pollock LE, Lalor P, Revell PA (1990) Type IV collagen and laminin in the synovial intimal layer: an immunohistochemical study. Rheumatol Int 9:277–280 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF00541324&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2315607&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1990CL76000005&link_type=ISI) 115.115.Rinaldi N, Barth TF, Weis D, Schwarz-Eywill M, Pezzutto A, Lukoschek M, Brocai D, Brado B (1998) Loss of laminin and of the laminin receptor integrin subunit alpha 6 in situ correlates with cytokine induced down regulation of alpha 6 on fibroblast-like synoviocytes from rheumatoid arthritis. Ann Rheum Dis 57:559–565 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6IjU3LzkvNTU5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 116.116.Scott DL, Wainwright AC, Walton KW, Williamson N (1981) Significance of fibronectin in rheumatoid arthritis and osteoarthrosis. Ann Rheum Dis 40:142–153 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6IjQwLzIvMTQyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 117.117.van Linthoudt D, Beutler A, Clayburne G, Sieck M, Fernandes L, Schumacher Jr HR, Schumacher Jr. HR, Schumacher Jr HR (1997) Morphometric studies on synovium in advanced osteoarthritis: is there an association between apatite-like material and collagen deposits? Clin Exp Rheumatol 15:493–497 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9307856&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 118.118.Worrall JG, Wilkinson LS, Bayliss MT, Edwards JCW (1994) Zonal distribution of chondroitin-4-sulphate/dermatan sulphate and chondroitin-6-sulphate in normal and diseased human synovium. Ann Rheum Dis 53:35–38 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjc6IjUzLzEvMzUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMy8wOC8zMS8yMDIzLjA4LjMxLjIzMjk0NjI1LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 119.119.Dijkgraaf LC, Liem RSB, de Bont LGM (1997) Ultrastructural characteristics of the synovial membrane in osteoarthritic temporomandibular joints. Journal of Oral & Maxillofacial Surgery 55:1269–1280 120.120.Okamoto K, Kiga N, Shinohara Y, Tojyo I, Fujita S (2015) Effect of interleukin-1beta and dehydroepiandrosterone on the expression of lumican and fibromodulin in fibroblast-like synovial cells of the human temporomandibular joint. European Journal of Histochemistry 59:2440 121.121.Chang X, Yamada R, Suzuki A, Kochi Y, Sawada T, Yamamoto K (2005) Citrullination of fibronectin in rheumatoid arthritis synovial tissue. Rheumatology 44:1374–1382 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/rheumatology/kei023&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16105911&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000232900900009&link_type=ISI) 122.122.Hino K, Shiozawa S, Kuroki Y, Ishikawa H, Shiozawa K, Sekiguchi K, Hirano H, Sakashita E, Miyashita K, Chihara K (1995) EDA-containing fibronectin is synthesized from rheumatoid synovial fibroblast-like cells. Arthritis Rheum 38:678–683 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.1780380516&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=7748223&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1995QX39200015&link_type=ISI) 123.123.Itokazu M, Shinozaki M, Ohno T (1998) Quantitative analysis of hyaluronan in the synovial tissues of patients with joint disorders. Clin Rheumatol 17:261–262 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF01451063&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9694068&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000074986600022&link_type=ISI) 124.124.Klareskog L, Johnell O, Hulth A, Holmdahl R, Rubin K (1986) Reactivity of monoclonal anti-type II collagen antibodies with cartilage and synovial tissue in rheumatoid arthritis and osteoarthritis. Arthritis Rheum 29:730–738 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/art.1780290605&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2424461&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1986C913700005&link_type=ISI) 125.125.Kriegsmann J, Berndt A, Hansen T, et al. (2004) Expression of fibronectin splice variants and oncofetal glycosylated fibronectin in the synovial membranes of patients with rheumatoid arthritis and osteoarthritis. Rheumatol Int 24:25–33 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s00296-003-0316-1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=12712258&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000188669100005&link_type=ISI) 126.126.Poduval P, Sillat T, Virtanen I, Dabagh M, Konttinen YT (2010) Immigration check for neutrophils in RA lining: Laminin α5 low expression regions act as exit points. Scand J Rheumatol 39:132–140 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20059371&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 127.127.Santiago B, Baleux F, Palao G, Gutiérrez-Cañas I, Ramírez JC, Arenzana-Seisdedos F, Pablos JL (2006) CXCL12 is displayed by rheumatoid endothelial cells through its basic amino-terminal motif on heparan sulfate proteoglycans. Arthritis Res Ther. doi:10.1186/ar1900 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/ar1900&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16507142&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 128.128.Schneider M, Voss B, Rauterberg J, Menke M, Pauly T, Miehlke RK, Friemann J, Gerlach U (1994) Basement membrane proteins in synovial membrane: distribution in rheumatoid arthritis and synthesis by fibroblast-like cells. Clin Rheumatol 13:90–97 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/BF02229873&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8187452&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 129.129.Scott DL, Salmon M, Morris CJ, Wainwright AC, Walton KW (1984) Laminin and vascular proliferation in rheumatoid arthritis. Ann Rheum Dis 43:551–555 [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6ImFubnJoZXVtZGlzIjtzOjU6InJlc2lkIjtzOjg6IjQzLzQvNTUxIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjMvMDgvMzEvMjAyMy4wOC4zMS4yMzI5NDYyNS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 130.130.Worrall JG, Bayliss MT, Edwards JCW (1991) Morphological localization of hyaluronan in normal and diseased synovium. Journal of Rheumatology 18:1466–1472 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=1765969&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1991GP89600009&link_type=ISI) 131.131.Exposito Molinero MR, de Miguel Mendieta E, Expósito Molinero MR, de Miguel Mendieta E (2016) Discriminant validity study of Achilles enthesis ultrasound. Reumatol Clin 12:206–209 132.132.Ibrahim M, Kartus JT, Steigen SE, Olsen R, Meknas K (2019) More tendon degeneration in patients with shoulder osteoarthritis. Knee Surgery, Sports Traumatology, Arthroscopy 27:267–275 133.133.Ibrahim M, Hedlundh U, Sernert N, Meknas K, Haag L, Movin T, Papadogiannakis N, Kartus JT (2021) Histological and ultrastructural degenerative findings in the gluteus medius tendon after hip arthroplasty. J Orthop Surg Res 16:339 134.134.Mazzocca AD, McCarthy MBR, Ledgard FA, et al. (2013) Histomorphologic changes of the long head of the biceps tendon in common shoulder pathologies. Arthroscopy - Journal of Arthroscopic and Related Surgery 29:972–981 135.135.Meknas K, Johansen O, Steigen SE, Olsen R, Jorgensen L, Kartus J (2012) Could tendinosis be involved in osteoarthritis? Scand J Med Sci Sports 22:627–634 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/j.1600-0838.2010.01287.x&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21410541&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 136.136.Loeser RF, Olex AL, McNulty MA, Carlson CS, Callahan M, Ferguson C, Fetrow JS (2013) Disease Progression and Phasic Changes in Gene Expression in a Mouse Model of Osteoarthritis. PLoS ONE [Electronic Resource]. doi:10.1371/journal.pone.0054633 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1371/journal.pone.0054633&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23382930&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 137.137.Anderson-Mackenzie JM, Billingham ME, Bailey AJ (1999) Collagen remodeling in the anterior cruciate ligament associated with developing spontaneous murine osteoarthritis. Biochem Biophys Res Commun 258:763–767 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1006/bbrc.1999.0713&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10329460&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000080596100048&link_type=ISI) 138.138.Ramos-Mucci L, Javaheri B, Van THR, Bou-Gharios G, Pitsillides AA, Comerford E, Poulet B (2020) Meniscal and ligament modifications in spontaneous and post-traumatic mouse models of osteoarthritis. Arthritis Res Ther. doi:10.1186/s13075-020-02261-5 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13075-020-02261-5&link_type=DOI) 139.139.Walton M (1977) Degenerative joint disease in the mouse knee; radiological and morphological observations. Journal of Pathology 123:97–107 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/path.1711230206&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=592018&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1977DZ71200005&link_type=ISI) 140.140.Miller D, Desutter C, Scott A, Koglin L, Hart DA, Salo P, Leonard C, Mammoto T, Bray RC (2014) Vascular structure and function in the medial collateral ligament of anterior cruciate ligament transected rabbit knees. Journal of Orthopaedic Research 32:1104–1110 141.141.Barton KI, Heard BJ, Kroker A, et al. (2021) Structural Consequences of a Partial Anterior Cruciate Ligament Injury on Remaining Joint Integrity: Evidence for Ligament and Bone Changes Over Time in an Ovine Model. American Journal of Sports Medicine 49:637–648 142.142.Funakoshi Y, Hariu M, Tapper JE, Marchuk LL, Shrive NG, Kanaya F, Rattner JB, Hart DA, Frank CB (2007) Periarticular ligament changes following ACL/MCL transection in an ovine stifle joint model of osteoarthritis. Journal of Orthopaedic Research 25:997–1006 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/jor.20370&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17436314&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000248200300003&link_type=ISI) 143.143.Bedingfield SK, Colazo JM, Di Francesco M, et al. (2021) Top-Down Fabricated microPlates for Prolonged, Intra-articular Matrix Metalloproteinase 13 siRNA Nanocarrier Delivery to Reduce Post-traumatic Osteoarthritis. ACS Nano 19:14475–14491 144.144.Lee KI, Gamini R, Olmer M, et al. (2020) Mohawk is a transcription factor that promotes meniscus cell phenotype and tissue repair and reduces osteoarthritis severity. Sci Transl Med 12:28 145.145.Muschter D, Fleischhauer L, Taheri S, Schilling AF, Clausen-Schaumann H, Grassel S (2020) Sensory neuropeptides are required for bone and cartilage homeostasis in a murine destabilization-induced osteoarthritis model. Bone. doi:10.1016/j.bone.2019.115181 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bone.2019.115181&link_type=DOI) 146.146.Hellio Le Graverand MP, Vignon E, Otterness IG, Hart DA (2001) Early changes in lapine menisci during osteoarthritis development: Part I: cellular and matrix alterations. Osteoarthritis Cartilage 9:56–64 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1053/joca.2000.0350&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11178948&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000166911300008&link_type=ISI) 147.147.Le Graverand MPH, Sciore P, Eggerer J, et al. (2001) Formation and phenotype of cell clusters in osteoarthritic meniscus. Arthritis Rheum 44:1808–1818 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/1529-0131(200108)44:8<1808::AID-ART318>3.0.CO;2-B&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=11508433&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 148.148.Levillain A, Magoariec H, Boulocher C, Decambron A, Viateau V, Hoc T (2017) Effects of a viscosupplementation therapy on rabbit menisci in an anterior cruciate ligament transection model of osteoarthritis. J Biomech 58:147–154 149.149.Levillain A, Magoariec H, Boulocher C, Decambron A, Viateau V, Hoc T (2017) Viscoelastic properties of rabbit osteoarthritic menisci: A correlation with matrix alterations. J Mech Behav Biomed Mater 65:1–10 150.150.Zhao J, Huang S, Zheng J, Zhong C, Tang C, Zheng L, Zhang Z, Xu J (2014) Changes of rabbit meniscus influenced by hyaline cartilage injury of osteoarthritis. Int J Clin Exp Med 7:2948– 2956 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25356168&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 151.151.Endo J, Sasho T, Akagi R, Muramatsu Y, Watanabe A, Akatsu Y, Fukawa T, Tahara M, Yamaguchi S (2018) Comparative Analysis of Gene Expression between Cartilage and Menisci in Early-Phase Osteoarthritis of the Knee-An Animal Model Study. Journal of Knee Surgery 31:664–669 152.152.Bansal S, Miller LM, Patel JM, et al. (2020) Transection of the medial meniscus anterior horn results in cartilage degeneration and meniscus remodeling in a large animal model. Journal of Orthopaedic Research 14:14 153.153.Shi X, Yu W, Wang T, Battulga O, Wang C, Shu Q, Yang X, Liu C, Guo C (2020) Electroacupuncture alleviates cartilage degradation: Improvement in cartilage biomechanics via pain relief and potentiation of muscle function in a rabbit model of knee osteoarthritis. Biomedicine and Pharmacotherapy. doi:10.1016/j.biopha.2019.109724 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.biopha.2019.109724&link_type=DOI) 154.154.Almasry SM, Soliman HM, El-Tarhouny SA, Algaidi SA, Ragab EM (2015) Platelet rich plasma enhances the immunohistochemical expression of platelet derived growth factor and vascular endothelial growth factor in the synovium of the meniscectomized rat models of osteoarthritis. Annals of Anatomy 197:38–49 155.155.Bryk M, Chwastek J, Mlost J, Kostrzewa M, Starowicz K (2021) Sodium Monoiodoacetate Dose-Dependent Changes in Matrix Metalloproteinases and Inflammatory Components as Prognostic Factors for the Progression of Osteoarthritis. Front Pharmacol 12:643605 156.156.Castrogiovanni P, Di Rosa M, Ravalli S, Castorina A, Guglielmino C, Imbesi R, Vecchio M, Drago F, Szychlinska MA, Musumeci G (2019) Moderate physical activity as a prevention method for knee osteoarthritis and the role of synoviocytes as biological key. Int J Mol Sci. doi:10.3390/ijms20030511 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/ijms20030511&link_type=DOI) 157.157.Dai S, Liang T, Fujii T, He S, Zhang F, Jiang H, Liu B, Shi X, Luo Z, Yang H (2020) Increased elastic modulus of the synovial membrane in a rat ACLT model of osteoarthritis revealed by atomic force microscopy. Brazilian Journal of Medical & Biological Research 53:e10058 158.158.Gamal N, Abou-Rabia NM, El Ebiary FH, Khalaf G, Raafat MH (2019) The possible therapeutic role of platelet rich plasma on a model of osteoarthritis in male albino rat. Histological and immunohistochemical study. Egyptian Journal of Histology 42:554–566 159.159.Li X, Mei W, Huang Z, et al. (2020) Casticin suppresses monoiodoacetic acid-induced knee osteoarthritis through inhibiting HIF-1α/NLRP3 inflammasome signaling. Int Immunopharmacol. doi:10.1016/j.intimp.2020.106745 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.intimp.2020.106745&link_type=DOI) 160.160.Zhang L, Li X, Zhang H, Huang Z, Zhang N, Zhang L, Xing R, Wang P (2021) Agnuside Alleviates Synovitis and Fibrosis in Knee Osteoarthritis through the Inhibition of HIF-1 α and NLRP3 Inflammasome. Mediators Inflamm. doi:10.1155/2021/5534614 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2021/5534614&link_type=DOI) 161.161.Zhang L, Li M, Li X, Liao T, Ma Z, Xing R, Wang P, Mao J (2021) Characteristics of sensory innervation in synovium of rats within different knee osteoarthritis models and the correlation between synovial fibrosis and hyperalgesia. J Adv Res. doi:10.1016/j.jare.2021.06.007 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.jare.2021.06.007&link_type=DOI) 162.162.Zhang L, Xing R, Huang Z, Zhang N, Zhang L, Li X, Wang P (2019) Inhibition of Synovial Macrophage Pyroptosis Alleviates Synovitis and Fibrosis in Knee Osteoarthritis. Mediators Inflamm. doi:10.1155/2019/2165918 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2019/2165918&link_type=DOI) 163.163.Zhang L, Zhang L, Huang Z, et al. (2019) Increased HIF-1α in knee osteoarthritis aggravate synovial fibrosis via fibroblast-like synoviocyte pyroptosis. Oxid Med Cell Longev. doi:10.1155/2019/6326517 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1155/2019/6326517&link_type=DOI) 164.164.Lapadula G, Nico B, Cantatore FP, La Canna R, Roncali L, Pipitone V (1995) Early ultrastructural changes of articular cartilage and synovial membrane in experimental vitamin A-induced osteoarthritis. Journal of Rheumatology 22:1913–1921 [PubMed](http://medrxiv.org/lookup/external-ref?access_num=8991991&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 165.165.Wei Q, Kong N, Liu X, Tian R, Jiao M, Li Y, Guan H, Wang K, Yang P (2021) Pirfenidone attenuates synovial fibrosis and postpones the progression of osteoarthritis by anti-fibrotic and anti-inflammatory properties in vivo and in vitro. J Transl Med 19:157 166.166.McErlain DD, Appleton CTG, Litchfield RB, Pitelka V, Henry JL, Bernier SM, Beier F, Holdsworth DW (2008) Study of subchondral bone adaptations in a rodent surgical model of OA using in vivo micro-computed tomography. Osteoarthritis Cartilage 16:458–469 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2007.08.006&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17900933&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000255194100009&link_type=ISI) 167.167.Dabrowska S, Ekiert-Radecka M, Karbowniczek J, Weglarz WP, Heljak M, Lojkowski M, Obuchowicz R, Swieszkowski W, Mlyniec A (2023) Calcification alters the viscoelastic properties of tendon fascicle bundles depending on matrix content. Acta Biomater. doi:10.1016/j.actbio.2023.05.010 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.actbio.2023.05.010&link_type=DOI) 168.168.Ricard-Blum S (2011) The Collagen Family. Cold Spring Harb Perspect Biol 3:1–19 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1101/cshperspect.a005298&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21441587&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 169.169.Chen D, Smith LR, Khandekar G, Patel P, Yu CK, Zhang K, Chen CS, Han L, Wells RG (2020) Distinct effects of different matrix proteoglycans on collagen fibrillogenesis and cell-mediated collagen reorganization. Sci Rep. doi:10.1038/s41598-020-76107-0 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41598-020-76107-0&link_type=DOI) 170.170.Naba A (2023) Ten Years of Extracellular Matrix Proteomics: Accomplishments, Challenges, and Future Perspectives. Mol Cell Proteomics 22:100528 171.171.McCoy AM (2015) Animal Models of Osteoarthritis: Comparisons and Key Considerations. Vet Pathol 52:803–818 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0300985815588611&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26063173&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 172.172.Cope PJ, Ourradi K, Li Y, Sharif M (2019) Models of osteoarthritis: the good, the bad and the promising. Osteoarthritis Cartilage 27:230–239 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.joca.2018.09.016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) 173.173.Proffen BL, McElfresh M, Fleming BC, Murray MM (2012) A comparative anatomical study of the human knee and six animal species. Knee 19:493–499 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.knee.2011.07.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21852139&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000305852300049&link_type=ISI) 174.174.Derfus BA, Kurian JB, Butler JJ, Daft LJ, Carrera GF, Ryan LM, Rosenthal AK (2002) The high prevalence of pathologic calcium crystals in pre-operative knees. J Rheumatol 29: 175.175.Mathes T, Klaßen P, Pieper D (2017) Frequency of data extraction errors and methods to increase data extraction quality: a methodological review. BMC Med Res Methodol. doi:10.1186/S12874-017-0431-4 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/S12874-017-0431-4&link_type=DOI) 176.176.Buckley CD, Ospelt C, Gay S, Midwood KS (2021) Location, location, location: how the tissue microenvironment affects inflammation in RA. Nature Reviews Rheumatology 2021 17:4 17:195–212 177.177.Sharma L (2021) Osteoarthritis of the Knee. New England Journal of Medicine 384:51–59 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMcp1903768&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2023%2F08%2F31%2F2023.08.31.23294625.atom)