You are viewing the site in preview mode

Skip to main content

Beyond symptomatic alignment: evaluating the integration of causal mechanisms in matching animal models with human pathotypes in osteoarthritis research

Abstract

Osteoarthritis (OA) is a highly prevalent and disabling condition lacking curative treatments, with only symptomatic relief available. Recognizing OA as a heterogenous disorder with diverse aetiologies and molecular foundations underscores the need to classify patients by both phenotypes and molecular pathomechanisms (endotypes). Such stratification could enable the development of targeted therapies to surmount existing treatment barriers. From a scientific, economic, and ethical perspective, it is crucial to employ animal models that accurately represent the endotype of the target patient population, not merely their clinical symptoms. These models must also account for intrinsic and extrinsic factors, like age, sex, metabolic status, and comorbidities, which impact OA's pathogenesis and its clinical and molecular variability and can profoundly influence not only structural and symptomatic disease severity and progression but also the underlying molecular pathophysiology. The molecular definition of the OA subpopulation must also be reflected in the read-outs, as the traditional methods—macroscopic and histological scoring, along with limited gene expression profiling of established biomarkers for cartilage degradation, extracellular matrix (ECM) turnover, and synovial inflammation—are inadequate for discovering new, phenotype- and endotype-specific biomarkers or therapeutic targets. Thus, animal model characterisation should evolve to include both clinically and pathophysiologically pertinent measures of disease progression and response to treatment. This review evaluates the utility and accuracy of current animal models in OA research, focusing on their capacity to replicate the disease’s pathophysiological processes.

Graphical Abstract

Introduction

Osteoarthritis (OA) currently affects approximately 500 million people worldwide and is projected to become the leading cause of disability by 2030 [1,2,3]. Despite this widespread prevalence, there are still no curative treatments available, amplifying the burden on individuals, societies and healthcare systems alike. The notable absence of therapeutic breakthroughs can be attributed to several factors, with the lacking consideration of disease heterogeneity in preclinical and clinical trials prime among them [4,5,6,7].

OA is now recognized as a heterogenous syndrome comprising distinct subtypes, each arising from different aetiologies and involving diverse cellular, molecular, and biomechanical pathways. Despite this variability, these pathways ultimately converge in a common end-stage pathology characterized by similar clinical and radiographic features. Acknowledging this complexity paves the way for targeted therapies, which may overcome the current therapeutic limitations. However, while traditional treatments, limited to pain management at the symptomatic stage of the disease, were universally applicable to OA, the development of disease-modifying therapies requires the stratification of patients based not only on observable characteristics (phenotypes) but also on molecular pathomechanisms (endotypes) that may warrant distinct targeted therapeutic strategies [8]. Indeed, meta analyses revealed that only two of 199 reported candidate genes were consistently associated with OA, emphasizing the importance of appropriate categorization and selection of endotype-specific patient populations for successful biomarker and therapeutic target discovery [9,10,11,12]. Disease stratification and endotype-based personalized therapies have been successfully implemented in allergy and asthma treatment and are expected to also significantly enhance OA therapeutic success. Accordingly, over the past decade, there has been a surge in efforts to stratify OA. This has resulted in a multitude of proposed OA subtypes, variably defined by clinical and/or molecular characteristics [13, 14]. However, significant overlap exists between these subtypes, and further research is necessary to establish a unified framework of well-characterised molecular endotypes that correlate with distinct clinical phenotypes and therapies [13].

Diagnosing OA in its early stages remains challenging due to the insensitivity of current clinical diagnostic methods to pre-symptomatic cellular and molecular changes and the weak correlation between clinical symptoms and pathological changes [15,16,17,18]. Consequently, elucidating the molecular pathogenetic and pathophysiological events that occur during OA initiation and progression remains difficult, hindering the identification of therapeutic targets and biomarkers for early OA diagnosis and monitoring [19,20,21]. To address these limitations and account for the multifaceted disease mechanisms and the complex interaction between articular tissues and systemic influences, animal models are essential. However, a critical disconnect often exists between preclinical models and the OA patient population, hindering scientific progress and contributing to the low translational success rate of biomedical research. Notably, the prevalent use of young, normal-weight, male animals to model OA stands in massive contrast to the demographics of the human patient population, where ageing, obesity and female sex are predisposing factors [22]. Moreover, while age is the primary risk factor for OA and traumatic joint injury accounts for only 12% of cases [23], joint injury remains the most widely used method to induce OA in animal models. This discrepancy between experimental models and clinical reality is exacerbated by the distinct molecular pathophysiology of post-traumatic (secondary) OA compared to spontaneous OA [24].

Optimising the selection of animal models and the design of preclinical trials is essential to improving their predictive utility, minimising the number of animal lives that are avoidably wasted under the 3Rs principle [20, 22]. To facilitate the matching of OA endotypes with relevant in vivo models, this perspective review reflects on the utility and validity of currently available mammalian animal models in the context of their stratification into subtypes, focusing on ageing and senescence-driven, metabolic syndrome-associated and traumatic-injury-driven OA (see [13, 16, 25], for comprehensive discussions of other putative OA endotypes) [11, 14, 26, 27].

General considerations for OA animal models

Evidence-based medicine, scientific rigour, and the 3Rs urgently require the careful selection of animal models based on the OA subtype of interest, the pathological feature of OA one wishes to investigate, and the study objectives. To improve the clinical translatability of preclinical findings, the chosen animal model should mimic the clinical and structural outcomes and molecular pathomechanisms of the OA phenotype and endotypes of interest, including the level of joint inflammation, cartilage and/or bone alterations as well as the severity and type of pain [19, 22, 24, 28,29,30,31,32,33,34,35]. This model-patient alignment also requires the consideration of intrinsic and extrinsic factors, such as age, metabolic status, sex, and co-morbid health conditions, that confound or contribute to OA pathogenesis and variability and can profoundly influence not only structural and symptomatic disease severity and progression but also the associated molecular pathophysiology [20, 36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57]. For example, the importance of age for the selection of animal models is demonstrated by studies investigating age-dependent responses to IL- 6 ablation, which is chondroprotective in young mouse models but can worsen age-associated OA in older mice [22]. Furthermore, even basal gene expression of a variety of joint tissues diverges between 12-week and 12-month-old mice, and medial meniscus destabilisation results in more severe OA in the older mice [22, 58]. The divergent outcomes in animal models representing different disease and patient characteristics highlight the importance of interpretation and translating preclinical trial results only within the framework of the specific OA subtype the model was selected to mimic.

In addition, an animal model should reflect the broad range and temporal progression of joint tissue pathology and allow quantifiable assessment of clinical outcomes. A fully sequenced and publicly available genome and proteome should be available to facilitate pathophysiological studies and the identification of treatment targets [19]. The species-specific variability in disease severity and time course following the same OA induction method is also a factor to be accounted for. Anterior cruciate ligament transection, as a frequently used OA induction method, induces slow progression to mild to moderate OA in sheep, goats and dogs but rapid progression to severe OA phenotype in mice [22, 59]. In addition, analogous to the distinct OA phenotype and endotypes observed in different appendicular joints in humans, different joints in animals show substantial variance in their response to the same insult [19, 60] which must be critically reflected during model selection.

Due to the heterogeneity of OA, there is no “gold-standard” animal model. Furthermore, as the pathophysiology of human OA is poorly understood and OA phenotype/endotype characterisation is still in progress, the validation of animal models against the human condition is difficult and typically limited to structural or biomechanical comparisons of articular cartilage [61], which fail to reflect the differences in pathogenetic mechanisms and corresponding OA phenotypes. Consequently, there are more than 50 different OA models utilising a wide range of species and disease induction approaches (see Table 1 for a detailed overview), each with a variety of advantages and shortcomings (Table 2). Commonly used species for OA research include foremost mice, which, together with guinea pigs, rabbits, and rats, represent over two-thirds of the animals used for OA research. Among large animals, dogs are most employed next to sheep, goats, and horses [19, 22, 29, 57]. OA in animal models can either arise spontaneously in naturally occurring and genetic models of disease or be induced using a variety of methods, including surgery, intra-articular chemical injection, mechanical overload, obesity, and high-fat diets [19]. Veterinary clinical populations suffering from naturally occurring disease are likely the best fit for a variety of phenotype and endotypes (post-traumatic, ageing, metabolic, etc.) of human OA, but are rarely used due to the time and resources required to recruit sufficient veterinary patients, inherently diverse patient populations, disease states and environmental conditions and the variable and protracted disease progression analogous to human disease. Veterinary patients with naturally occurring OA, following rigorous enrollment criteria and ethical protocols aligned with human clinical trials, could help bridge the gap between basic science and clinical application in humans, benefiting both animal and human patients.

Table 1 Overview of currently available in vivo models matched with the six specific OA phenotypes
Table 2 Advantages and disadvantages of the currently used OA animal models

OA represents a spectrum of molecularly distinct subtypes, but current classification efforts remain fragmented. While rheumatoid arthritis (RA) has benefited from endotype-specific classification based on transcriptomic and proteomic markers, OA endotyping is still in its infancy. A unified framework should integrate multi-omics data (genomics, transcriptomics, metabolomics, and proteomics) with imaging and clinical characteristics to establish reproducible OA endotypes. Studies have identified age-related differences in transcriptomic profiles of OA cartilage [172] and distinct metabolic signatures in synovial fluid of patients with different OA subtypes [173,174,175]. However, inconsistencies in patient cohorts and the lack of large-scale, standardized datasets hinder efforts to consolidate these findings. To address this, Artificial Intelligence (AI)-driven clustering methods could stratify OA into robust subtypes, analogous to approaches used in RA [16]. Establishing a global OA endotype database, akin to RA biomarker repositories, could facilitate cross-study validation and enhance preclinical model selection. Recent advances in omics technologies have revolutionized OA endotyping, enabling for instance precise molecular classification utilizing RNA sequencing and single-cell transcriptomics revealing distinct inflammatory, senescent, and metabolic endotypes [11]. Metabolomics and proteomics identify biochemical signatures in synovial fluid and serum [10]. Imaging tools like MRI and PET scans provide structural and metabolic insights, helping to correlate molecular changes with phenotypic severity [15]. Standardizing these methodologies will enhance cross-study comparability and preclinical-to-clinical translation. In addition to standard omics and imaging technologies, recent studies combine metabolomic profiling and machine learning algorithms to refine OA endotype classification further. For instance, Carlson et al. used synovial fluid metabolomics to identify metabolic phenotypes associated with structural cartilage changes, suggesting distinct biochemical environments that underpin specific OA subtypes [176]. Similarly, deep metabolic profiling of temporomandibular joint (TMJ) OA enabeled the identification of over 1,400 metabolites, including amino acids, lipids, and benzene derivatives, several of which correlated with disease severity [177]. Additionally, integrated metabolomic and transcriptomic analyses highlighted significant metabolic alterations, including disruptions in the tricarboxylic acid (TCA) cycle and amino acid metabolism in TMJ OA [178]. These findings underscore the potential of stage-related metabolic markers as non-invasive indicators of progression. Machine learning approaches, as shown by Nelson et al., who applied unsupervised clustering to biomarker data from the FNIH cohort, revealed progression-associated phenotypes that may inform patient stratification and therapeutic response [179]. Additionally, global metabolomic profiling has been shown to differentiate OA from other joint pathologies, offering diagnostic specificity at the biochemical level [180]. Spatial proteomic imaging (e.g., FTIR, Raman, nano-FTIR, MSI) supported mapping of cartilage and bone microenvironments at near-cellular resolution in a study conducted by Fan et al. [181]. These technologies captured biochemical gradients associated with inflammation, fibrosis, and mineralization. Combined with AI-based pattern recognition, these methods enable tissue-level phenotyping beyond histology. Rockel et al. introduced a variational autoencoder (OmicVAE) to integrate microRNA and metabolomic data from multiple biofluids [182]. This unsupervised model stratified patients into three molecularly defined endotypes, which predicted differential pain and function outcomes post-arthroplasty. Such integrative models demonstrate how latent omic features can outperform traditional clinical or single-omic classifiers in outcome prediction.

Similarly, Angelini et al. used clustering on biochemical markers from the IMI-APPROACH cohort to define three reproducible endotypes: one with low tissue turnover (C1), a second driven by structural degradation (C2), and a thrid driven by systemic inflammation (C3) [16]. These endotypes correlated with radiographic and symptomatic progression and were validated in an external cohort. The use of explainable AI (SHAP values) clarified key features per cluster, facilitating interpretability and potential trial stratification. The STEpUP OA consortium optimized a proteomic pipeline for synovial fluid (SF) using SomaScan aptamer technology [183]. An unprecedented number of SF samples (n = 1746) from diverse cohorts were analyzed, applying variance analyses (PCA, UMAP) revealing distinct proteomic patterns separating OA from injury. This provides a robust resource for future endotype discovery and supports SF as a local and disease-relevant matrix for biomarker identification. Mobasheri et al. emphasized the strategic importance of deep phenotyping using multi-omics and single-cell approaches to refine the cellular taxonomy of OA [1]. They advocate for harmonized data structures and open-access databases to integrate datasets from cartilage, bone, synovium, and immune cell compartments. Such efforts are key to redefining OA not as a single disease but as a set of overlapping molecular subtypes that may respond to targeted therapies.

These studies converge on a shared message, that endotype discovery in OA requires multimodal integration of omics data, spatial resolution, and computational modeling enabling fine-grained molecular phenotyping. They also allow reverse translation using human omics data to select or refine animal models that best reflect specific OA subtypes. They further shift the focus from morphological staging to mechanism-driven classification with the incentive to identify translatable biomarkers and therapeutic targets. A future OA classification framework should combine tissue-specific molecular signatures, joint-level imaging, and biofluid-derived biomarkers, supported by machine learning algorithms trained on harmonized datasets. Such a categorization may have the potential to improve patient stratification, refine animal models, and to accelerate disease-modifying drug development.

Mind the patient: Do we have the right animal models for our human OA phenotypes and endotypes?

Overall, post-traumatic OA models are by far the most validated and accepted for their phenotype authenticity [152]. In contrast, the relevance and validity of models for other OA subtypes, for which the cause-and-effect relationship for disease onset and progression has not yet been unraveled, remain inherently ambiguous [184].

Ageing and Senescence-driven OA

Ageing and cell senescence are associated with declining joint tissue functionality and regenerative capacity in cartilage and other tissues [22, 185, 186]. While ageing is the predominant risk factor for OA, the quiescent chondrocytes predominantly undergo stress-induced rather than replicative senescence due to accumulating damage from extrinsic and intrinsic stressors, such as reactive oxygen species (ROS) [187]. Accordingly, senescent cells have also been observed in articular tissues of young post-traumatic OA patients [188], highlighting the possible overlap of different endotypes that have to be considered in individualised targeted therapies.

Ageing and age-related diseases such as OA are characterised by cellular senescence and chronic systemic and local inflammation (inflammageing) and progenitor cell dysfunction [1]. Age-related increases in local and systemic proinflammatory mediators exacerbate cellular senescence of joint tissue-resident cells, such as chondrocytes and synoviocytes, and the subsequent release of senescence-associated secretory factors (SASP) with elevated pro-inflammatory cytokines [9,10,11, 22, 36, 185, 186, 189]. This, in turn, intensifies synovial inflammation, creating a vicious cycle where senescent cells promote inflammageing and inflammageing accelerates cellular senescence.

To accurately mimic ageing and senescence-driven OA, animal models should replicate the hallmarks of ageing, including senescence, inflammageing and progenitor cell exhaustion. Aging remains the predominant risk factor for OA, yet the pathophysiological mechanisms of aging-associated OA are underrepresented in preclinical models. However, in ageing research, models merely phenocopying selected ageing manifestations are often erroneously used to conclude the mechanisms of ageing [190]. Notably, a significant portion of studies cited for cellular senescence (63%) and stem cell exhaustion (62%) utilise models with unclear relevance to ageing [190]. Caution is warranted when using progeroid mouse models to study age-driven OA pathomechanisms. For example, XpdTTD mice, a commonly used progeroid model exhibiting severe osteoporosis, do not display accelerated cartilage ageing, underscoring the highly compartmentalised nature of ageing phenotypes in progeroid syndromes [191]. The STR/ORT (Strain 1/Old Retirement) mouse is one of the most widely studied spontaneous models for age-related OA, developing progressive joint degeneration without surgical induction, closely mimicking the multifactorial onset observed in humans [62]. The STR/ORT mouse model spontaneously develops OA. Disease onset typically begins between 18 and 24 weeks of age and presents with hallmark features such as cartilage erosion, osteophyte formation, and subchondral bone sclerosis [192, 193]. Importantly, STR/ORT mice display a polygenic susceptibility to OA, mimicking the multifactorial nature of human disease, and show upregulation of matrix metalloproteinases (e.g., MMP-3, MMP-13), chondrocyte hypertrophy, and reduced aggrecan content [40, 194]. Despite these strengths, the model has limitations. The murine cartilage matrix composition differs from humans in terms of glycosaminoglycan content and thickness, and mice experience distinct mechanical joint loading patterns due to quadrupedal gait and reduced body mass [57, 195]. Moreover, STR/ORT mice exhibit a marked sexual dimorphism, with males showing a higher and more consistent OA incidence most likely linked to androgenic signaling and growth plate dynamics [40, 196]. Although immune mechanisms are not the primary focus of this model, age-associated changes in immune cell profiles, including altered macrophage polarization and T cell senescence, have been reported in aged mice generally and may be relevant for STR/ORT pathogenesis [197, 198]. Overall, the STR/ORT mouse remains one of the few models that recapitulates slow, spontaneous OA progression in the absence of overt trauma, making it uniquely suited for studies on age-related joint degeneration. For instance, elderly STR/ORT mice were already used to explore the potential therapeutic potential of peptides targeting cartilage degradation. Improved cartilage integrity and decreasing expression of OA markers, MMP-13 and COL10A1, suggest that Gly-Arg-Gly-Asp-Ser (GRGDS) administration may enable ageing-related cartilage damage [199].

In contrast to genetically predisposed murine models such as the STR/ORT mouse, canine and equine models, which naturally develop OA due to age and mechanical stress, offer advantages in biomechanical similarity and joint size, allowing for longitudinal imaging and biomarker assessments akin to human patients [68]. Aged canines, particularly in larger breeds such as Labrador Retrievers or Beagles, frequently develop OA in the stifle and hip joints with age. Disease onset is often exacerbated by congenital dysplasia or joint instability but progresses in the absence of experimental induction. Histopathological features mirror human OA, including fibrillation and erosion of the articular cartilage, subchondral bone remodeling, and low-grade synovial inflammation. Notably, dogs exhibit gait patterns, joint kinetics, and cartilage thickness that are more similar to humans than rodents, enhancing their relevance for longitudinal imaging and therapeutic testing [200, 201]. In addition, aging dogs with OA diagnosis and PTOA models display exhibited levels of MMP- 13, IL- 1β, IL- 6, and TNF-α in synovial fluid and tissue, aligning with known catabolic cascades in human disease [202, 203]. Horses develop naturally occurring OA in high-load joints such as the carpus, fetlock, and tarsus, particularly in older performance horses. These animals present subchondral sclerosis, osteophyte formation, and early cartilage matrix changes, including depletion of proteoglycans and increased collagen II fragmentation [68]. Equine cartilage shares key features with human cartilage, such as comparable chondrocyte density, ECM structure, and cartilage thickness (~ 1.5–2 mm). Moreover, aged horses with OA show significant elevations of biomarkers such as CTX-II, COMP, and CPII, which are widely used in clinical trials for monitoring cartilage turnover [167, 204]. Their large joint size and tolerance for serial arthroscopy further enable longitudinal assessments, making them ideal for evaluating disease-modifying OA drugs (DMOADs) in aging contexts. Thus, animal models that spontaneously develop age-related joint tissue changes and naturally occurring OA offer distinct advantages for studying the complex interplay between senescence, inflammation, and joint degeneration, investigating the molecular pathomechanisms of the disease and identifying potential therapeutic targets [62, 63, 66,67,68]. However, challenges such as high cost, genetic variability, and ethical constraints limit their widespread use. Future research should focus on cross-validating spontaneous and induced models with human endotypes to refine model selection.

Metabolic syndrome associated OA

Obesity doubles the risk of symptomatic OA in both weight-bearing (i.e., knee) and non-weight-bearing (i.e., hand) joints, indicating contributions beyond biomechanical overload with adipokine levels correlating with OA severity. Obesity triggers inflammation and remodeling of white adipose tissue, adipocyte hypertrophy and hyperplasia and the pro-inflammatory phenotype of adipose tissue-resident immune cells [205,206,207]. Altered adipokine signatures in obesity, especially with concurrent metabolic syndrome, are characterised by decreased levels of anti-inflammatory adipokines (e.g., adiponectin and omentin-1) and the upregulation of pro-inflammatory adipokines (e.g., leptin, resistin, and visfatin) and foster a pro-inflammatory milieu akin to inflammageing, driving metabolic syndrome-related chronic diseases, such as OA. In addition, metabolic syndrome is associated with an increased chronic cellular senescence burden, including senescence of mesenchymal stromal cells (MSCs), derived not only from fat but also from bone marrow [51, 52, 205,206,207,208].

Metabolically induced OA models are crucial for exploring the relationship between metabolism and OA development [56]. Beyond systemic inflammation, obesity-induced OA also involves intrinsic metabolic changes within articular cartilage, promoting disturbance in the intracellular lipid homeostasis [209]. A recent study performed by Liu et al. demonstrated that adipokines such as leptin and resistin not only exacerbate synovial inflammation but also directly alter chondrocyte lipid metabolism, leading to increased lipid droplet accumulation and oxidative stress [210]. This dysregulated fat metabolism impairs chondrocyte function, promoting catabolic enzyme activity (e.g., MMP-13) and accelerating cartilage breakdown. The high-fat diet-induced OA model effectively replicates these intra-articular changes, supporting its validity for studying metabolic OA endotypes [211, 212].

In addition to veterinary patients suffering from naturally occurring metabolic syndrome associated with OA, high-fat diet-induced OA has been established as a good model for metabolic OA [211]. It mimics not only the relevant OA pathogenesis characterised by local and systemic inflammation with elevation of specific cytokines, chemokines and adipokines but also induces typical general alterations, including anxiety and hyperalgesia, as well as decreased muscle function and locomotor activity [211, 213, 214]. These striking similarities to the human disease progression render this in vivo model for this particular OA subtype a valuable tool for basic and applied research [22, 184, 211, 213,214,215,216,217]. Nonetheless, a clear separation and stratification between metabolic and chronic mechanical overloading subtypes is difficult as global analysis of structure (number of lesions & Mankin score) and secretion profiles (i.e., adipokines) for obesity-related disease onset and progression in combination with mechanical altercations is heterogenous among study reports [184].

Traumatic-injury driven OA

Acute trauma to joint structures, such as cartilage, subchondral bone, ligaments and meniscus, initiates molecular cascades leading to post-traumatic OA, exacerbating inflammatory and catabolic responses (e.g., ROS, toll-like receptor activation).

Post-traumatic OA in animal models can be induced through various methods, including non-invasive mechanical loading, surgical induction of cartilage lesions or surgical destabilisation of the joint, e.g. in the knee by transection of the cranial cruciate ligament, collateral ligaments, or meniscotibial ligament with or without removing all or part of the meniscus [79, 103, 108, 124, 218, 219]. Trauma-induced OA models mimic injuries also commonly observed in human patients and reflect the mechanical and biological changes observed in human OA, providing a controlled environment to study disease progression and possible therapeutic options. While surgical models offer the benefit of precisely targeting the tissue of interest, they may not fully capture the damage to other joint structures often seen in real-world injuries. For example, impact-induced bone bruises, evident in 80% of human ACL tears, are not typically replicated in surgical models [220, 221]. In addition, opening the joint introduces compounding effects by contributing to articular inflammation and pain [220]. Closed-joint impact models, on the other hand, damage multiple articular structures, offering a more realistic representation of clinical injuries, but they are inherently more variable due to the less controlled nature of the injury [220].

Matchmaking of OA models against specific human phenotypes requires deeper analysis

As the value of animal models also depends on the reproducibility and validity of the outcome measures [36, 184], an expansion of the assessment method repertoire (e.g., omics approaches, pain assessment) is urgently required to maximise the usefulness of preclinical research. Interestingly, while multi-modal analyses and omics approaches have been successfully applied for molecular research in rheumatoid arthritis for over a decade, they are new to the OA research field. Currently, read-outs are typically limited to macroscopic and histological scoring as well as gene expression and protein secretion screening of a few established biomarkers of cartilage degeneration, ECM turnover, and synovial inflammation, such as collagen type 2 (Col2), type 1 (Col1), type 10 (Col10), matrix metalloproteinases (MMP)-1, -3 and -6, interleukin (IL)-1 and -6, and tumor necrosis factor-alpha (TNFa). The limitation of analysis approaches to well-established procedures and molecular indicators hampers the discovery of novel, phenotype, and endotype-specific biomarkers and therapeutic target identification. OA molecular pheno- and endotyping and model-OA-subtype matching can benefit from the increasing availability and affordability of omics methods and annotation of animal species [174], as shown by recent studies identifying age-related differences in murine post-traumatic knee OA transcriptome and metabolomic characteristics of anterior cruciate reconstructed versus sham-operated ovine synovial fluid [172, 174]. In general, a comprehensive molecular characterisation of phenotypes in animals and humans will help to identify where great overlaps exist, which in turn will indicate the most suitable animal models for preclinical studies. Once the appropriate animal model is identified, the next step is to successfully treat the condition in this model. Following the success in preclinical studies, the corresponding patient cohort can be targeted for clinical trials.

Additionally, it may be beneficial to revisit previously unsuccessful clinical trials. By redefining patient sub-cohorts, there is an opportunity to reanalyse the data to determine if the treatment might have significant effects that were previously overlooked. This iterative approach can refine the understanding and improve the success rates of clinical interventions.

As pain relief and joint function are the most important outcomes for clinicians and patients, more emphasis should be placed on pain assessment in OA models. Regular pain assessment using species-specific pain scores (i.e., grimace scale), weight-bearing, functional gait analysis, and lameness scores should be mandatory for each preclinical trial to ensure proper analgesia for the animals and provide clinically relevant read-outs of OA progression. In addition, anxiety and depression are increasingly used to monitor OA symptoms and progression also in animals [222]. For example, in mice that underwent partial medial meniscectomy as a disease trigger to mimic human post-traumatic OA, a longitudinal, multiparametric assessment of pain revealed distinct time-dependent and disease-progression-related pain levels and mechanisms [73]. Another comprehensive study investigating the association between OA disease phenotypes, joint pathology, gene expression, and pain behaviour revealed phenotype-specific pain and peripheral sensory neuronal responses [163]. By combining gene expression analysis with a wide range of pain evaluation modalities, the study demonstrated that the molecular pathophysiology of pain and joint-tissue pathology is influenced by the underlying disease model even in the later stages of the disease. This highlights the importance of considering phenotype- and disease-stage-specific factors when interpreting animal model studies and extrapolating their findings to human disease. Additionally, the study emphasises the significance of pain assessment for animal welfare and relevant clinical outcomes [163].

In rheumatoid arthritis (RA) research, multi-modal analyses have significantly advanced our understanding of disease mechanisms, patient stratification, and treatment responses [5, 223, 224]. These approaches integrate various data types, such as genomic, transcriptomic, proteomic, and imaging data, to provide a comprehensive view of the disease. One prominent example is the integration of single-cell RNA sequencing (scRNA-seq) and single-cell chromatin accessibility profiling (scATAC-seq) to dissect the transcriptional and epigenetic landscape of synovial fibroblast subpopulations [225,226,227,228]. This combined strategy revealed conserved gene regulatory networks across human RA tissue and mouse models, highlighting distinct fibroblast endotypes that drive inflammation and joint damage [229]. In parallel, photoacoustic imaging combined with ultrasound (PA-US) has emerged as a powerful tool to non-invasively assess inflammatory activity in RA joints, capturing both vascular changes and tissue structure with high resolution [230,231,232,233,234,235]. Moreover, machine learning techniques, such as dynamic deep neural networks, have been employed to integrate clinical and biomarker data for more accurate prediction of RA progression and treatment response [236, 237]. These multi-modal approaches, linking molecular, cellular, imaging, and computational layers, have redefined disease stratification in RA and provide a conceptual and technical framework for OA endotyping.

Histopathological grading remains a cornerstone in OA research; however, traditional scoring systems like OARSI and Mankin exhibit critical limitations. These semi-quantitative methods are inherently subjective, relying on visual interpretation of morphological features such as cartilage surface integrity, matrix staining, and cellularity, which leads to considerable inter-observer variability and poor reproducibility [238,239,240,241]. Moreover, they predominantly assess late-stage structural changes in cartilage and fail to integrate molecular, cellular, or biomechanical alterations that are essential to distinguishing OA endotypes [25]. Artificial intelligence-driven histopathology and deep learning-based imaging analysis are efforts to refine histopathological assessment and to modernize histopathological evaluation in general [242].

Studies in RA have demonstrated AI’s ability to integrate histopathological, omics, and imaging data for more accurate disease classification [5, 239, 241]. Similarly, AI-powered deep learning models have achieved 93% accuracy in grading knee OA from radiographic images, significantly outperforming human raters [240]. Such methods could be applied to automate OA scoring systems, integrating cellular-level changes with structural histopathology for a more comprehensive disease assessment.

Conclusion & Prospects

The poor reproducibility of biomedical research and lack of translatability of basic science to clinical applications call for a critical evaluation of preclinical models regarding their alignment with the clinical trial population’s OA phenotype and endotype, age, sex, confounding comorbidities, and evaluation parameters [22]. To successfully identify biomarkers and therapeutic targets for novel treatment approaches, animal models need to accurately mimic the aspects of human OA pathophysiology that are relevant to the OA subtype of interest and the study objectives. Longitudinal and multiparametric assessment of biomarkers in a variety of tissues, in serum, urine, or synovial fluid, will provide more meaningful information about the disease stage, structural pathology, and underlying molecular mechanisms. For example, the integration of AI-driven tools into OA classification and model evaluation offers a promising route to enhance the fidelity and translatability of preclinical research. Deep learning approaches applied to radiographic and histological images have demonstrated superior accuracy and consistency compared to human-assisted scoring, particularly in the grading of structural joint changes. However, their full potential lies beyond automation. To modernise histopathological OA scoring, a multi-dimensional refinement is warranted: a) the incorporation of molecular biomarkers into standard grading systems would enable a more mechanistic understanding of histological changes and better reflect the underlying disease biology; b) AI-based digital pathology pipelines could support automated, high-throughput, and reproducible analysis of tissue samples, including the identification of histological signatures specific to OA phenotypes; and c) machine learning models for multimodal data integration can combine histological imaging with transcriptomic, proteomic, and imaging datasets, supporting phenotype- and endotype-specific classification. By transitioning from morphology-based assessments to AI-enhanced, biomarker-informed classification systems, histopathology can become a more powerful tool for aligning animal models with specific human OA subtypes. This paradigm shift would not only improve model selection but also enhance the resolution of treatment effects, biomarker discovery, and translational success. The currently predominant evidence-based selection of animal models subtypes of interest is currently hindered by ambiguous definitions of the different OA patient subtypes and inadequate characterisation of existing animal models, which is typically limited to radiographic, macroscopic, and histological features and qPCRs of a few select ECM factors, proteases, and inflammatory mediators. However, our growing understanding of the cellular and molecular mechanisms of OA, combined with technical advancements in molecular imaging and omics technologies, is rapidly expanding the repertoire of methods available for the pathophysiologic stratification of both human patients and animals. This allows for a more accurate classification based on the underlying molecular mechanisms of the disease [20]. Hence, the characterisation of animal models should progress to include the criteria used to differentiate between different OA phenotypes and endotypes [243]. Similarly, clinically and pathophysiologically relevant readouts of disease progression and treatment response, and corresponding reporting guidelines, need to be established and standardised for models of each OA subtype. In addition to standardised evaluation criteria, consolidation of the current plethora of animal models may also aid in improving the comparability of preclinical data. Currently, post-traumatic knee OA, for example, is induced in species including mice, rats, rabbits, dogs, sheep, goats, and horses, using either non-invasive mechanical loading, surgical induction of cartilage lesions, or surgical destabilisation of the joint by transection of the cranial cruciate ligament, collateral ligaments, or meniscotibial ligament, with or without removing all or part of the meniscus. This profusion of models, all aiming to mimic the same OA subtype, spreads the data characterising each model thin and limits the comparability of study results. Limiting the number of models to those best reflecting each OA phenotype and endotype would greatly increase research synergies and hence contribute to reducing the number of animals needed for OA research, optimising research economy and ethics. Using reverse translation as a fidelity check will ensure that molecules and pathways identified in vitro and in vivo models of OA can reflect the naturally occurring pathology and could help identify the best-suited model species and OA induction methods. Uniform reporting standards of preclinical trials should also document and publish negative study outcomes. Initiatives such as the ‘One Health Initiative’ [89] can further facilitate the multi-directional flow of knowledge and synergistic gains for multiple disciplines, which will contribute to a better and more holistic interpretation and validation of data in the field of OA as well as OA phenotyping.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

OA:

Osteoarthritis

PTs:

Phenotype

ETs:

Endotype

CT:

Computed tomography

MRI:

Magnetic resonance imaging

Col2:

Collagen type 2

MMP:

Matrix metalloproteinase

IL:

Interleukin

TNFa:

Tumor necrosis factor-alpha

SASP:

Senescence-associated secretory phenotype

RA:

Rheumatoid arthritis

SF:

Synovial fluid

TCA:

The tricarboxylic acid cycle

TMJ:

Temporomandibular joint

DMOADs:

Disease-modifying osteoarthritis drug

FTIR:

Fourier transform infrared spectroscopy

MSI:

Mass spectrometry imaging

PCA:

Principal Component Analysis

UMAP:

Uniform Manifold Approximation and Projection

SHAP:

Shapley Additive exPlanations

3Rs:

Replacement, Reduction and Refinement

ACL:

Anterior cruciate ligament

AI:

Artificial intelligence

COMP:

Cartilage Oligomeric Matrix Protein

ECM:

Extracellular matrix

MSCs:

Mesenchymal stromal cells

OARSI:

Osteoarthritis Research Society International

PT:

Post traumatic

ROS:

Reactive oxygen species

qPCR:

Quantitative polymerase chain reaction

References

  1. Mobasheri A, Kapoor M, Ali SA, Lang A, Madry H. The future of deep phenotyping in osteoarthritis: How can high throughput omics technologies advance our understanding of the cellular and molecular taxonomy of the disease? Osteoarthr Cartil Open. 2021;3: 100144.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Thomas E, Peat G, Croft P. Defining and mapping the person with osteoarthritis for population studies and public health. Rheumatology (Oxford). 2014;53:338.

    Article  PubMed  Google Scholar 

  3. Makaram NS, Simpson AHRW. Disease-modifying agents in osteoarthritis: where are we now and what does the future hold? Bone Joint Res. 2023;12:654.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Yuan C, Pan Z, Zhao K, Li J, Sheng Z, Yao X, et al. Classification of four distinct osteoarthritis subtypes with a knee joint tissue transcriptome atlas. Bone Res. 2020;8:38.

  5. Steinberg J, Ritchie GRS, Roumeliotis TI, Jayasuriya RL, Clark MJ, Brooks RA, et al. Integrative epigenomics, transcriptomics and proteomics of patient chondrocytes reveal genes and pathways involved in osteoarthritis. Sci Rep. 2017;7:8935.

  6. Katsoula G, Kreitmaier P, Zeggini E. Insights into the molecular landscape of osteoarthritis in human tissues. Curr Opin Rheumatol. 2022;34:79–90.

    Article  CAS  PubMed  Google Scholar 

  7. Steinberg J, Southam L, Fontalis A, Clark MJ, Jayasuriya RL, Swift D, et al. Linking chondrocyte and synovial transcriptional profile to clinical phenotype in osteoarthritis. Ann Rheum Dis. 2021;80:1070–4.

    Article  CAS  PubMed  Google Scholar 

  8. Werdyani S, Liu M, Zhang H, Sun G, Furey A, Randell EW, et al. Endotypes of primary osteoarthritis identified by plasma metabolomics analysis. Rheumatology. 2021;60:2735–44.

    Article  CAS  PubMed  Google Scholar 

  9. Boer CG, Hatzikotoulas K, Southam L, Stefánsdóttir L, Zhang Y, de Coutinho Almeida R, et al. Deciphering osteoarthritis genetics across 826,690 individuals from 9 populations. Cell. 2021;184:4784–4818.e17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Soul J, Dunn SL, Anand S, Serracino-Inglott F, Schwartz JM, Boot-Handford RP, et al. Stratification of knee osteoarthritis: two major patient subgroups identified by genome-wide expression analysis of articular cartilage. Ann Rheum Dis. 2018;77:423–30.

    Article  CAS  PubMed  Google Scholar 

  11. Lv Z, Yang YX, Li J, Fei Y, Guo H, Sun Z, et al. Molecular Classification of Knee Osteoarthritis. Front cell Dev Biol. 2021;9:725568.

  12. Zaki S, Blaker CL, Little CB. OA foundations - experimental models of osteoarthritis. Osteoarthr Cartil. 2022;30:357–80.

    Article  CAS  Google Scholar 

  13. Mobasheri A, Loeser R. Clinical phenotypes, molecular endotypes and theratypes in OA therapeutic development. Nat Rev Rheumatol. 2024;20:525–6.

    Article  PubMed  Google Scholar 

  14. Mobasheri A, Van Spil WE, Budd E, Uzieliene I, Bernotiene E, Bay-Jensen AC, et al. Molecular taxonomy of osteoarthritis for patient stratification, disease management and drug development: biochemical markers associated with emerging clinical phenotypes and molecular endotypes. Curr Opin Rheumatol. 2019;31:80–9.

    Article  PubMed  Google Scholar 

  15. Henrotin Y. Osteoarthritis in year 2021: biochemical markers. Osteoarthr Cartil. 2022;30:237–48.

    Article  CAS  Google Scholar 

  16. Angelini F, Widera P, Mobasheri A, Blair J, Struglics A, Uebelhoer M, et al. Osteoarthritis endotype discovery via clustering of biochemical marker data. Ann Rheum Dis. 2022;81:666–75.

    Article  PubMed  Google Scholar 

  17. Karsdal MA, Christiansen C, Ladel C, Henriksen K, Kraus VB, Bay-Jensen AC. Osteoarthritis - a case for personalized health care? Osteoarthr Cartil. 2014;22:7–16.

    Article  CAS  Google Scholar 

  18. Bierma-Zeinstra SMA, Verhagen AP. Osteoarthritis subpopulations and implications for clinical trial design. Arthritis Res Ther. 2011;13:213.

  19. Zaki S, Blaker CL, Little CB. OA foundations – experimental models of osteoarthritis. Osteoarthr Cartil. 2022;30:357–80.

    Article  CAS  Google Scholar 

  20. Frangogiannis NG. Why animal model studies are lost in translation. J Cardiovasc Aging 2022;2:22. https://dx.doi.org/10.20517/jca.2022.10.

  21. Drevet S, Favier B, Brun E, Gavazzi G, Lardy B. Mouse Models of Osteoarthritis: A Summary of Models and Outcomes Assessment. Comp Med. 2022;72:3–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Malfait AM, Little CB. On the predictive utility of animal models of osteoarthritis. Arthritis Res Ther. 2015;17:225.

  23. Brown TD, Johnston RC, Saltzman CL, Marsh JL, Buckwalter JA. Posttraumatic osteoarthritis: a first estimate of incidence, prevalence, and burden of disease. J Orthop Trauma. 2006;20:739–44.

  24. Little CB, Hunter DJ. Post-traumatic osteoarthritis: from mouse models to clinical trials. Nat Rev Rheumatol. 2013;9:485–97.

    Article  CAS  PubMed  Google Scholar 

  25. Mobasheri A, Saarakkala S, Finnilä M, et al. Recent advances in understanding the phenotypes of osteoarthritis [version 1; peer review: 2 approved]. F1000Res. 2019;8(F1000 Faculty Rev):2091. https://doiorg.publicaciones.saludcastillayleon.es/10.12688/f1000research.20575.1.

  26. Mobasheri A, Bay-Jensen AC, van Spil WE, Larkin J, Levesque MC. Osteoarthritis Year in Review 2016: biomarkers (biochemical markers). Osteoarthr Cartil. 2017;25:199–208.

    Article  CAS  Google Scholar 

  27. Mobasheri A, Rayman MP, Gualillo O, Sellam J, Van Der Kraan P, Fearon U. The role of metabolism in the pathogenesis of osteoarthritis. Nat Rev Rheumatol. 2017;13:302–11.

    Article  CAS  PubMed  Google Scholar 

  28. Wendler A, Wehling M. The translatability of animal models for clinical development: biomarkers and disease models. Curr Opin Pharmacol. 2010;10:601–6.

    Article  CAS  PubMed  Google Scholar 

  29. Little CB, Zaki S, Blaker CL, Clarke EC. Animal models of osteoarthritis : the benefits and costs of reducing variability. Bone Joint Res. 2022;11:514–7.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Medicine M, Aaron Ericsson by C, Crim MJ, Franklin CL, Ericsson AC. A Brief History of Animal Modeling. Mo Med. 2013;110:201.

    Google Scholar 

  31. Blaker CL, Clarke EC, Little CB. Using mouse models to investigate the pathophysiology, treatment, and prevention of post-traumatic osteoarthritis. J Orthop Res. 2017;35:424–39.

    Article  PubMed  Google Scholar 

  32. Little CB, Zaki S. What constitutes an “animal model of osteoarthritis” - the need for consensus? Osteoarthr Cartil. 2012;20:261–7.

    Article  CAS  Google Scholar 

  33. Hummel M, Whiteside GT. Measuring and realizing the translational significance of preclinical in vivo studies of painful osteoarthritis. Osteoarthr Cartil. 2017;25:376–84.

    Article  CAS  Google Scholar 

  34. Leenaars CHC, Kouwenaar C, Stafleu FR, Bleich A, Ritskes-Hoitinga M, De Vries RBM, et al. Animal to human translation: a systematic scoping review of reported concordance rates. J Transl Med. 2019;17:223.

  35. McGonigle P, Ruggeri B. Animal models of human disease: challenges in enabling translation. Biochem Pharmacol. 2014;87:162–71.

    Article  CAS  PubMed  Google Scholar 

  36. Im GIL, Moon JY. Emerging Concepts of Endotypes Phenotypes in Regenerative Medicine for Osteoarthritis. Tissue Eng Regen Med. 2022;19:321–4.

    Article  CAS  PubMed  Google Scholar 

  37. Pound P. Are Animal Models Needed to Discover, Develop and Test Pharmaceutical Drugs for Humans in the 21st Century? Anim. 2020;10:2455.

    Google Scholar 

  38. Tuncel M, Halici M, Canoz O, Yildirim Turk C, Oner M, Ozturk F, et al. Role of insulin like growth factor-I in repair response in immature cartilage. Knee. 2005;12:113–9.

    Article  PubMed  Google Scholar 

  39. Mastbergen SC, Marijnissen AC, Vianen ME, van Roermund PM, Bijlsma JW, Lafeber FP. The canine “groove” model of osteoarthritis is more than simply the expression of surgically applied damage. Osteoarthr Cartil. 2006;14:39–46.

    Article  Google Scholar 

  40. Poulet B, Hamilton RW, Shefelbine S, Pitsillides AA. Characterizing a novel and adjustable noninvasive murine joint loading model. Arthritis Rheum. 2011;63:137–47.

    Article  PubMed  Google Scholar 

  41. De Souza RL, Matsuura M, Eckstein F, Rawlinson SCF, Lanyon LE, Pitsillides AA. Non-invasive axial loading of mouse tibiae increases cortical bone formation and modifies trabecular organization: A new model to study cortical and cancellous compartments in a single loaded element. Bone. 2005;37:810–8.

    Article  PubMed  Google Scholar 

  42. Ogbonna AC, Clark AK, Gentry C, Hobbs C, Malcangio M. Pain-like behaviour and spinal changes in the monosodium iodoacetate model of osteoarthritis in C57Bl/6 mice. Eur J Pain (United Kingdom). 2013;17:514–26.

    CAS  Google Scholar 

  43. Hadipour-Jahromy M, Mozaffari-Kermani R. Chondroprotective Effects of Pomegranate Juice on Monoiodoacetate-induced Osteoarthritis of the Knee Joint of Mice. Phyther Res. 2010;24:182–5.

    Article  Google Scholar 

  44. Van Der Kraan PM, Vitters EL, Van Beuningen HM, Van De Putte LBA, Van Den Berg WB. Degenerative knee joint lesions in mice after a single intra-articular collagenase injection. A new model of osteoarthritis. J Exp Pathol. 1990;71:19–31.

    Google Scholar 

  45. Van Osch GJVM, Van Der Kraan PM, Blankevoort L, Huiskes R, Van Den Berg WB. Relation of ligament damage with site specific cartilage loss and osteophyte formation in collagenase induced osteoarthritis in mice. J Rheumatol. 1996;23:1227–32.

    PubMed  Google Scholar 

  46. Panicker S, Borgia J, Fhied C, Mikecz K, Oegema TR. Oral glucosamine modulates the response of the liver and lymphocytes of the mesenteric lymph nodes in a papain-induced model of joint damage and repair. Osteoarthr Cartil. 2009;17:1014–21.

    Article  CAS  Google Scholar 

  47. Shane Anderson A, Loeser RF. Why is Osteoarthritis an Age-Related Disease? Best Pract Res Clin Rheumatol. 2010;24:15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Guingamp C, Gegout-Pottie P, Philippe L, Terlain B, Netter P, Gillet P. Mono-iodoacetate-induced experimental osteoarthritis: A dose-response study of loss of mobility, morphology, and biochemistry. Arthritis Rheum. 1997;40:1670–9.

    Article  CAS  PubMed  Google Scholar 

  49. Marker CL, Pomonis JD. The Monosodium Iodoacetate Model of Osteoarthritis. Pain Res. 2012;851:239–48.

    Article  CAS  Google Scholar 

  50. Guzman RE, Evans MG, Bove S, Morenko B, Kilgore K. Mono-Iodoacetate-Induced Histologic Changes in Subchondral Bone and Articular Cartilage of Rat Femorotibial Joints: AN Animal Model of Osteoarthritis. Toxicol Pathol. 2003;31:619–24.

    Article  CAS  PubMed  Google Scholar 

  51. Xu X, Li X, Liang Y, Ou Y, Huang J, Xiong J, et al. Estrogen Modulates Cartilage and Subchondral Bone Remodeling in an Ovariectomized Rat Model of Postmenopausal Osteoarthritis. Med Sci Monit. 2019;25:3146–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Clockaerts S, Bastiaansen-Jenniskens YM, Runhaar J, Van Osch GJVM, Van Offel JF, Verhaar JAN, et al. The infrapatellar fat pad should be considered as an active osteoarthritic joint tissue: a narrative review. Osteoarthr Cartil. 2010;18:876–82.

    Article  CAS  Google Scholar 

  53. Roman-Blas JA, Castañeda S, Largo R, Herrero-Beaumont G. Osteoarthritis associated with estrogen deficiency. Arthritis Res Ther. 2009;11:1–14.

    Article  Google Scholar 

  54. Bertoldo E, Adami G, Rossini M, Giollo A, Orsolini G, Viapiana O, et al. The Emerging Roles of Endocrine Hormones in Different Arthritic Disorders. Front Endocrinol (Lausanne). 2021;12:510.

    Article  Google Scholar 

  55. Bliddal H, Leeds AR, Christensen R. Osteoarthritis, obesity and weight loss: evidence, hypotheses and horizons – a scoping review. Obes Rev. 2014;15:578.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chen L, Zheng JJY, Li G, Yuan J, Ebert JR, Li H, et al. Pathogenesis and clinical management of obesity-related knee osteoarthritis: Impact of mechanical loading. J Orthop Transl. 2020;24:66–75.

    CAS  Google Scholar 

  57. McCoy AM. Animal Models of Osteoarthritis: Comparisons and Key Considerations. Vet Pathol. 2015;52:803–18.

    Article  CAS  PubMed  Google Scholar 

  58. Ryu JH, Yang S, Shin Y, Rhee J, Chun CH, Chun JS. Interleukin-6 plays an essential role in hypoxia-inducible factor 2α-induced experimental osteoarthritic cartilage destruction in mice. Arthritis Rheum. 2011;63:2732–43.

    Article  CAS  PubMed  Google Scholar 

  59. Little CB, Smith MM. Animal Models of Osteoarthritis. Curr Rheumatol Rev. 2008;4:175–82. https://doiorg.publicaciones.saludcastillayleon.es/10.2174/157339708785133523.

    Article  Google Scholar 

  60. Ängeby Möller K, Klein S, Seeliger F, Finn A, Stenfors C, Svensson CI. Monosodium iodoacetate-induced monoarthritis develops differently in knee versus ankle joint in rats. Neurobiol Pain. 2019;6: 100036.

    Article  PubMed  PubMed Central  Google Scholar 

  61. Mastbergen S, Lafeber F, Osteoarthritis. Animal Models of Osteoarthritis—Why Choose a Larger Model? US Musculoskelet Rev. 2009;4.

  62. Kyostio-Moore S, Nambiar B, Hutto E, Ewing PJ, Piraino S, Berthelette P, et al. STR/ort mice, a model for spontaneous osteoarthritis, exhibit elevated levels of both local and systemic inflammatory markers. Comp Med. 2011;61:346–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Pasold J, Osterberg A, Peters K, Taipaleenmäki H, Säämänen AM, Vollmar B, et al. Reduced expression of Sfrp1 during chondrogenesis and in articular chondrocytes correlates with osteoarthritis in STR/ort mice. Exp Cell Res. 2013;319:649–59.

    Article  CAS  PubMed  Google Scholar 

  64. Jaeger K, Selent C, Jaehme W, Mahr S, Goebel U, Ibrahim S, et al. The genetics of osteoarthritis in STR/ort mice. Osteoarthr Cartil. 2008;16:607–14.

    Article  CAS  Google Scholar 

  65. Bendele AM, Hulman JF. Effects of Body Weight Restriction on the Development and Progression of Spontaneous Osteoarthritis in Guinea Pigs. Arthritis Rheum. 1991;34:1180–4.

    Article  CAS  PubMed  Google Scholar 

  66. Arzi B, Wisner ER, Huey DJ, Kass PH, Hu J, Athanasiou KA. A proposed model of naturally occurring osteoarthritis in the domestic rabbit. Lab Anim (NY). 2012;41:20–5.

    Article  Google Scholar 

  67. Bland SD. Canine osteoarthritis and treatments: a review. Vet Sci Dev. 2015;5(2). https://doiorg.publicaciones.saludcastillayleon.es/10.4081/vsd.2015.5931.

  68. McIlwraith CW, Frisbie DD, Kawcak CE. The horse as a model of naturally occurring osteoarthritis. Bone Joint Res. 2012;1:297–309.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ross M, Dyson S. Diagnosis and Management of Lameness in the Horse: Second Edition. 2010.

  70. Macfadyen MA, Daniel Z, Kelly S, Parr T, Brameld JM, Murton AJ, et al. The commercial pig as a model of spontaneously-occurring osteoarthritis. BMC Musculoskelet Disord. 2019;20:1–12.

    Article  Google Scholar 

  71. Hargrave-Thomas EJ, Thambyah A, Mcglashan SR, Broom ND. The bovine patella as a model of early osteoarthritis. J Anat. 2013;223:651–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Cook JL, Kuroki K, Visco D, Pelletier JP, Schulz L, Lafeber FPJG. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the dog. Osteoarthr Cartil. 2010;18(SUPPL. 3):S66–79.

    Article  Google Scholar 

  73. Knights CB, Gentry C, Bevan S. Partial medial meniscectomy produces osteoarthritis pain-related behaviour in female C57BL/6 mice. Pain. 2012;153:281–92.

    Article  PubMed  Google Scholar 

  74. Glasson SS, Blanchet TJ, Morris EA. The surgical destabilization of the medial meniscus (DMM) model of osteoarthritis in the 129/SvEv mouse. Osteoarthr Cartil. 2007;15:1061–9.

    Article  CAS  Google Scholar 

  75. Mak J, Jablonski CL, Leonard CA, Dunn JF, Raharjo E, Matyas JR, et al. Intra-articular injection of synovial mesenchymal stem cells improves cartilage repair in a mouse injury model. Sci Rep. October 2015;2016(6):1–12.

    Google Scholar 

  76. Matsuoka M, Onodera T, Sasazawa F, Momma D, Baba R, Hontani K, et al. An Articular Cartilage Repair Model in Common C57Bl/6 Mice. Tissue Eng - Part C Methods. 2015;21:767–72.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Bridgette D. Furman, Jens Strand, W. Chad Hembree, Benjamin D. Ward, Farshid Guilak SAO. Joint Degeneration following Closed Intraarticular Fracturein the Mouse Knee: A Model of Posttraumatic Arthritis. J Orthop Res Sept. 2007; May. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jor.20331.

  78. Christiansen BA, Anderson MJ, Lee CA, Williams JC, Yik JHN, Haudenschild DR. Musculoskeletal changes following non-invasive knee injury using a novel mouse model of post-traumatic osteoarthritis. Osteoarthr Cartil. 2012;20:773–82.

    Article  CAS  Google Scholar 

  79. Hayami T, Pickarski M, Zhuo Y, Wesolowski GA, Rodan GA, Duong LT. Characterization of articular cartilage and subchondral bone changes in the rat anterior cruciate ligament transection and meniscectomized models of osteoarthritis. Bone. 2006;38:234–43.

    Article  PubMed  Google Scholar 

  80. Hayami T, Pickarski M, Wesolowski GA, McLane J, Bone A, Destefano J, et al. The Role of Subchondral Bone Remodeling in Osteoarthritis: Reduction of Cartilage Degeneration and Prevention of Osteophyte Formation by Alendronate in the Rat Anterior Cruciate Ligament Transection Model. Arthritis Rheum. 2004;50:1193–206.

    Article  CAS  PubMed  Google Scholar 

  81. Iijima H, Aoyama T, Ito A, Tajino J, Nagai M, Zhang X, et al. Destabilization of the medial meniscus leads to subchondral bone defects and site-specific cartilage degeneration in an experimental rat model. Osteoarthr Cartil. 2014;22:1036–43.

    Article  CAS  Google Scholar 

  82. Baragi VM, Becher G, Bendele AM, Biesinger R, Bluhm H, Boer J, et al. A new class of potent matrix metalloproteinase 13 inhibitors for potential treatment of osteoarthritis: Evidence of histologic and clinical efficacy without musculoskeletal toxicity in rat models. Arthritis Rheum. 2009;60:2008–18.

    Article  CAS  PubMed  Google Scholar 

  83. Bove SE, Laemont KD, Brooker RM, Osborn MN, Sanchez BM, Guzman RE, et al. Surgically induced osteoarthritis in the rat results in the development of both osteoarthritis-like joint pain and secondary hyperalgesia. Osteoarthr Cartil. 2006;14:1041–8.

    Article  CAS  Google Scholar 

  84. Fernihough J, Gentry C, Malcangio M, Fox A, Rediske J, Pellas T, et al. Pain related behaviour in two models of osteoarthritis in the rat knee. Pain. 2004;112:83–93.

    Article  PubMed  Google Scholar 

  85. Gelse K, Von der Mark K, Aigner T, Park J, Schneider H. Articular cartilage repair by gene therapy using growth factor-producing mesenchymal cells. Arthritis Rheum. 2003;48:430–41.

    Article  CAS  PubMed  Google Scholar 

  86. Kuroda R, Usas A, Kubo S, Corsi K, Peng H, Rose T, et al. Cartilage repair using bone morphogenetic protein 4 and muscle-derived stem cells. Arthritis Rheum. 2006;54:433–42.

    Article  CAS  PubMed  Google Scholar 

  87. Rodrigues-Neto HR, Andrade-Junior EF, Feitosa-Junior DJS, Valente AL, Xavier TC, Alho do BCN, et al. Comparison of Three Experimental Models for Rat Osteoarthritis Induction. J Biosci Med. 2016;04:62–9.

    CAS  Google Scholar 

  88. Abbud Lozoya KA, Kouri Flores JB. A novel rat osteoarthrosis model to assess apoptosis and matrix degradation. Pathol Res Pract. 2000;196:729–45.

    Article  Google Scholar 

  89. Du G, Zhan H, Ding D, Wang S, Wei X, Wei F, et al. Abnormal Mechanical Loading Induces Cartilage Degeneration by Accelerating Meniscus Hypertrophy and Mineralization after ACL Injuries in Vivo. Am J Sports Med. 2016;44:652–63.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Bouchgua M, Alexander K, André d’Anjou M, Girard CA, Carmel EN, Beauchamp G, et al. Use of routine clinical multimodality imaging in a rabbit model of osteoarthritis - part I. Osteoarthr Cartil. 2009;17:188–96.

    Article  CAS  Google Scholar 

  91. Levillain A, Boulocher C, Kaderli S, Viguier E, Hannouche D, Hoc T, et al. Meniscal biomechanical alterations in an ACLT rabbit model of early osteoarthritis. Osteoarthr Cartil. 2015;23:1186–93.

    Article  CAS  Google Scholar 

  92. Zellner J, Pattappa G, Koch M, Lang S, Weber J, Pfeifer CG, et al. Autologous mesenchymal stem cells or meniscal cells: What is the best cell source for regenerative meniscus treatment in an early osteoarthritis situation? Stem Cell Res Ther. 2017;8:1–12.

    Article  Google Scholar 

  93. Atarod M, Frank CB, Shrive NG. Increased meniscal loading after anterior cruciate ligament transection in vivo: A longitudinal study in sheep. Knee. 2015;22:11–7.

    Article  PubMed  Google Scholar 

  94. Ghosh P, Sutherland J, Bellenger C, Read R, Darvodelsky A. The influence of weight-bearing exercise on articular cartilage of meniscectomized joints. An experimental study in sheep. Clinical orthopaedics and related research. 1990;252:101–13.

    Article  Google Scholar 

  95. Armstrong SJ, Read RA, Ghosh P, Wilson DM. Moderate exercise exacerbates the osteoarthritic lesions produced in cartilage by meniscectomy: a morphological study. Osteoarthr Cartil. 1993;1:89–96.

    Article  CAS  Google Scholar 

  96. Beveridge JE, Shrive NG, Frank CB. Meniscectomy causes significant in vivo kinematic changes and mechanically induced focal chondral lesions in a sheep model. J Orthop Res. 2011;29:1397–405.

    Article  PubMed  Google Scholar 

  97. Cake MA, Read RA, Corfield G, Daniel A, Burkhardt D, Smith MM, et al. Comparison of gait and pathology outcomes of three meniscal procedures for induction of knee osteoarthritis in sheep. Osteoarthr Cartil. 2013;21:226–36.

    Article  CAS  Google Scholar 

  98. Pearce SG, Hurtig MB, Clarnette R, Kalra M, Cowan B, Miniaci A. An investigation of 2 techniques for optimizing joint surface congruency using multiple cylindrical osteochondral autografts. Arthroscopy. 2001;17:50–5.

    Article  CAS  PubMed  Google Scholar 

  99. Burks RT, Greis PE, Arnoczky SP, Scher C. The use of a single osteochondral autograft plug in the treatment of a large osteochondral lesion in the femoral condyle: An experimental study in sheep. Am J Sports Med. 2006;34:247–55.

    Article  PubMed  Google Scholar 

  100. Karakilic B, Taskiran E, Doganavsargil B, Uzun B, Celik S, Kaya Bicer E. Central defect type partial ACL injury model on goat knees: The effect of infrapatellar fat pad excision. J Orthop Surg Res. 2015;10:137.

  101. Murphy JM, Fink DJ, Hunziker EB, Barry FP. Stem Cell Therapy in a Caprine Model of Osteoarthritis. Arthritis Rheum. 2003;48:3464–74.

    Article  PubMed  Google Scholar 

  102. Pallante AL, Görtz S, Chen AC, Healey RM, Chase DC, Ball ST, et al. Treatment of articular cartilage defects in the goat with frozen versus fresh osteochondral allografts: Effects on cartilage stiffness, zonal composition, and structure at six months. J Bone Jt Surg - Ser A. 2012;94:1984–95.

    Article  Google Scholar 

  103. Shoemaker RS, Bertone AL, Martin GS, McIlwraith CW, Roberts ED, Pechman R, et al. Effects of intra-articular administration of methylprednisolone acetate on normal articular cartilage and on healing of experimentally induced osteochondral defects in horses. Am J Vet Res. 1992;53:1446–53.

    Article  CAS  PubMed  Google Scholar 

  104. Reesink HL, Nixon AJ, Su J, Liu S, Sutton RM, Mann S, et al. Galectins-1 and-3 increase in equine post-traumatic osteoarthritis. Front Vet Sci. 2018;5:1–11.

    Article  Google Scholar 

  105. Maninchedda U, Lepage OM, Gangl M, Hilairet S, Remandet B, Meot F, et al. Development of an equine groove model to induce metacarpophalangeal osteoarthritis: A pilot study on 6 horses. PLoS ONE. 2015;10:1–18.

    Article  Google Scholar 

  106. Markus M. Wilke, Daryl V. Nydam AJN. Enhanced Early Chondrogenesis in Articular Defectsfollowing Arthroscopic Mesenchymal Stem CellImplantation in an Equine Model. J Orthop Res. 2007. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jor.20382.

  107. Salgado C, Jordan O, Allémann E. Osteoarthritis in vitro models: Applications and implications in development of intra-articular drug delivery systems. Pharmaceutics. 2021;13:1–23.

    Article  Google Scholar 

  108. Cruz R, Ramírez C, Rojas OI, Casas-Mejía O, Kouri JB, Vega-López MA. Menisectomized miniature Vietnamese pigs develop articular cartilage pathology resembling osteoarthritis. Pathol Res Pract. 2015;211:829–38.

    Article  PubMed  Google Scholar 

  109. Ahern BJ, Parvizi J, Boston R, Schaer TP. Preclinical animal models in single site cartilage defect testing: a systematic review. Osteoarthr Cartil. 2009;17:705–13.

    Article  CAS  Google Scholar 

  110. Chiang H, Kuo TF, Tsai CC, Lin MC, She BR, Huang YY, et al. Repair of porcine articular cartilage defect with autologous chondrocyte transplantation. J Orthop Res. 2005;23:584–93.

    Article  PubMed  Google Scholar 

  111. Gál P, Nečas A, Adler J, Teyschl O, Fabián P, Bibrová Š. Transplantation of the Autogenous Chondrocyte Graft to Physeal Defects: An Experimental Study in Pigs. Acta Vet Brno. 2002;71:327–32.

    Article  Google Scholar 

  112. Harman BD, Weeden SH, Lichota DK, Brindley GW. Osteochondral autograft transplantation in the porcine knee. Am J Sports Med. 2006;34:913–8.

    Article  PubMed  Google Scholar 

  113. Gotterbarm T, Richter W, Jung M, Berardi Vilei S, Mainil-Varlet P, Yamashita T, et al. An in vivo study of a growth-factor enhanced, cell free, two-layered collagen-tricalcium phosphate in deep osteochondral defects. Biomaterials. 2006;27:3387–95.

    Article  CAS  PubMed  Google Scholar 

  114. Flanigan DC, Harris JD, Brockmeier PM, Lathrop RL, Siston RA. The effects of defect size, orientation, and location on subchondral bone contact in oval-shaped experimental articular cartilage defects in a bovine knee model. Knee Surgery, Sport Traumatol Arthrosc. 2014;22:174–80.

    Article  Google Scholar 

  115. Boyd SK, Müller R, Leonard T, Herzog W. Long-term periarticular bone adaptation in a feline knee injury model for post-traumatic experimental osteoarthritis. Osteoarthr Cartil. 2005;13:235–42.

    Article  CAS  Google Scholar 

  116. Ogawa S, Awaga Y, Takashima M, Hama A, Matsuda A, Takamatsu H. Knee osteoarthritis pain following medial meniscectomy in the nonhuman primate. Elsevier Ltd; 2016.

  117. Bendele AM. Progressive Chronic Osteoarthritis in Femorotibial Joints of Partial Medial Meniscectomized Guinea Pigs. Vet Pathol. 1987;24:444–8.

    Article  CAS  PubMed  Google Scholar 

  118. Arunakul M, Tochigi Y, Goetz JE, Diestelmeier BW, Heiner AD, Rudert J, et al. Replication of chronic abnormal cartilage loading by medial meniscus destabilization for modeling osteoarthritis in the rabbit knee in vivo. J Orthop Res. 2013;31:1555–60.

    Article  PubMed  PubMed Central  Google Scholar 

  119. Kuroki K, Cook CR, Cook JL. Subchondral bone changes in three different canine models of osteoarthritis. Osteoarthr Cartil. 2011;19:1142–9.

    Article  CAS  Google Scholar 

  120. Szeto SG, Narimatsu M, Lu M, He X, Sidiqi AM, Tolosa MF, et al. YAP/TAZ Are Mechanoregulators of TGF-b-Smad Signaling and Renal Fibrogenesis. J Am Soc Nephrol. 2016;27:3117–28.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Luther JK, Cook CR, Cook JL. Meniscal release in cruciate ligament intact stifles causes lameness and medial compartment cartilage pathology in dogs 12 weeks postoperatively. Vet Surg. 2009;38:520–9.

    Article  PubMed  Google Scholar 

  122. Simmons EJ, Bertone AL, Weisbrode SE. Instability-induced osteoarthritis in the metacarpophalangeal joint of horses. Am J Vet Res. 1999;60:7–13.

    Article  CAS  PubMed  Google Scholar 

  123. Auer JA, Fackelman GE, Gingerich DA, Fetter AW. Effect of hyaluronic acid in naturally occurring and experimentally induced osteoarthritis. Am J Vet Res. 1980;41:568–74.

    Article  CAS  PubMed  Google Scholar 

  124. Bolam CJ, Hurtig MB, Cruz A, McEwen BJE. Characterization of experimentally induced post-traumatic osteoarthritis in the medial femorotibial joint of horses. Am J Vet Res. 2006;67:433–47.

    Article  CAS  PubMed  Google Scholar 

  125. Frisbie DD, Ghivizzani SC, Robbins PD, CM CE. Treatment of experimental equine osteoarthritis by invivo delivery of the equine interleukin 1 receptorantagonist gene. Gene Ther. 2002;9:12–20.

    Article  CAS  PubMed  Google Scholar 

  126. Crawford WH. Intra-Articular Replacement of Bovine Cranial Cruciate Ligaments with an Autogenous Fascial Graft. Vet Surg. 1990;19:380–8.

    Article  CAS  PubMed  Google Scholar 

  127. Williams JM, Brandt KD. Benoxaprofen reduces osteophyte formation and fibrillation after articular cartilage injury. J Rheumatol. 1985;12:27–32.

    CAS  PubMed  Google Scholar 

  128. Kopp S, Mejersjö C, Clemensson E. Induction of osteoarthrosis in the guinea pig knee by papain. Oral Surgery, Oral Med Oral Pathol. 1983;55:259–66.

    Article  CAS  Google Scholar 

  129. Goranov NV. Clinical changes in sodium monoiodoacetate-induced stifle osteoarthritis model in dogs. Vet World. 2012;5:138–44.

    Article  CAS  Google Scholar 

  130. Frisbie DD, Kawcak CE, Werpy NM, Park RD, Mcilwraith CW. Clinical, biochemical, and histologic effects of intra-articular administration of autologous conditioned serum in horses with experimentally induced osteoarthritis. Am J Vet Res. 2007;68.

  131. Kawcak CE, Frisbie DD, McIlwraith W, Werpy NM, Park RD. Evaluation of avocado and soybean unsaponifiable extracts for treatment of horses with experimentally induced osteoarthritis. Am J Vet Res. 2007;68:598–604.

    Article  PubMed  Google Scholar 

  132. Uilenreef J, van der Staay FJ, Meijer E. A monosodium iodoacetate osteoarthritis lameness model in growing pigs. Animals. 2019;9:1–19.

    Article  Google Scholar 

  133. Li Jiang, Anlun Ma, Lijun Song, Yanxin Hu, Hao Dun, Pierre Daloze, Yonglin Yu, Jianyuan Jiang MZ and HC. Cartilage regeneration by selected chondrogenicclonal mesenchymal stem cells in the collagenase-induced monkey osteoarthritis model. J Tissue Eng Regen Med. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.1002/term.1676.

  134. Hülser ML, Luo Y, Frommer K, Hasseli R, Köhler K, Diller M, et al. Systemic versus local adipokine expression differs in a combined obesity and osteoarthritis mouse model. Sci Reports. 2021;11:1–13.

    Google Scholar 

  135. Warmink K, Kozijn AE, Bobeldijk I, Stoop R, Weinans H, Korthagen NM. High-fat feeding primes the mouse knee joint to develop osteoarthritis and pathologic infrapatellar fat pad changes after surgically induced injury. Osteoarthr Cartil. 2020;28:593–602.

    Article  CAS  Google Scholar 

  136. Adães S, Mendonça M, Santos TN, Castro-Lopes JM, Ferreira-Gomes J, Neto FL. Intra-articular injection of collagenase in the knee of rats as an alternative model to study nociception associated with osteoarthritis. Arthritis Res Ther. 2014;16:1–17.

    Article  Google Scholar 

  137. Rainsford KD, Skerry TM, Chindemi P, Delaney K. Effects of the NSAIDs Meloxicam and Indomethacin on Cartilage Proteoglycan Synthesis and Joint Responses to Calcium Pyrophosphate Crystals in Dogs. Vet Res Commun. 1999;23:101–13.

    Article  CAS  PubMed  Google Scholar 

  138. Sniekers YH, Weinans H, van Osch GJVM, van Leeuwen JPTM. Oestrogen is important for maintenance of cartilage and subchondral bone in a murine model of knee osteoarthritis. Arthritis Res Ther. 2010;12:R182.

  139. Oestergaard S, Sondergaard BC, Hoegh-Andersen P, Henriksen K, Qvist P, Christiansen C, et al. Effects of ovariectomy and estrogen therapy on type II collagen degradation and structural integrity of articular cartilage in rats: Implications of the time of initiation. Arthritis Rheum. 2006;54:2441–51.

    Article  CAS  PubMed  Google Scholar 

  140. Dai G, Li J, Liu X, Liu Q, Liu C. The relationship of the expression of estrogen receptor in cartilage cell and osteoarthritis induced by bilateral ovariectomy in guinea pig. J Huazhong Univ Sci Technolog Med Sci. 2005;25:683–6.

    Article  CAS  PubMed  Google Scholar 

  141. Calvo E, Castañeda S, Largo R, Fernández-Valle ME, Rodríguez-Salvanés F, Herrero-Beaumont G. Osteoporosis increases the severity of cartilage damage in an experimental model of osteoarthritis in rabbits. Osteoarthr Cartil. 2007;15:69–77.

    Article  CAS  Google Scholar 

  142. Kreipke TC, Rivera NC, Garrison JG, Easley JT, Turner AS, Niebur GL. Alterations in trabecular bone microarchitecture in the ovine spine and distal femur following ovariectomy. J Biomech. 2014;47:1918–21.

    Article  PubMed  PubMed Central  Google Scholar 

  143. Carlson CS, Loeser RF, Jayo MJ, Weaver DS, Adams MR, Jerome CP. Osteoarthritis in cynomolgus macaques: A primate model of naturally occurring disease. J Orthop Res. 1994;12:331–9.

    Article  CAS  PubMed  Google Scholar 

  144. Blair-Levy JM, Watts CE, Fiorientino NM, Dimitriadis EK, Marini JC, Lipsky PE. A type I collagen defect leads to rapidly progressive osteoarthritis in a mouse model. Arthritis Rheum. 2008;58:1096–106.

    Article  CAS  PubMed  Google Scholar 

  145. van der Kraan PM. Relevance of zebrafish as an OA research model. Osteoarthr Cartil. 2013;21:261–2.

    Article  Google Scholar 

  146. Salminen H, Perälä M, Lorenzo P, Saxne T, Heinegård D, Säämänen AM, et al. Up-regulation of cartilage oligomeric matrix protein at the onset of articular cartilage degeneration in a transgenic mouse model of osteoarthritis. Arthritis Rheum. 2000;43:1742–8.

    Article  CAS  PubMed  Google Scholar 

  147. Fässler R, Schnegelsberg PNJ, Dausman J, Shinya T, Muragaki Y, Mccarthy MT, et al. Mice lacking α1(IX) collagen develop noninflammatory degenerative joint disease. Proc Natl Acad Sci U S A. 1994;91:5070–4.

    Article  PubMed  PubMed Central  Google Scholar 

  148. Modeling osteoarthritis genetics in zebrafish. Bonekey Rep. 2013;2:349. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/bonekey.2013.83.

  149. Lawrence EA, Hammond CL, Blain EJ. Potential of zebrafish as a model to characterise MicroRNA profiles in mechanically mediated joint degeneration. Histochem Cell Biol. 2020;154:521–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Gregory MH, Capito N, Kuroki K, Stoker AM, Cook JL, Sherman SL. A Review of Translational Animal Models for Knee Osteoarthritis. Arthritis. 2012;2012:1–14.

    Article  Google Scholar 

  151. Lorenz J, Grässel S. Experimental Osteoarthritis Models in Mice. Mouse Genet. 2014;1194 July:401–19.

    Article  Google Scholar 

  152. Bendele AM. Animal models of osteoarthritis. J Musculoskel Neuron Interact. 2001;1(4):363–76.

    CAS  PubMed  Google Scholar 

  153. Glasson SS, Chambers MG, Van Den Berg WB, Little CB. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the mouse. Osteoarthr Cartil. 2010;18(SUPPL. 3):S17–23.

    Article  Google Scholar 

  154. Smale G, Bendele A, Horton WE. Comparison of age-associated degeneration of articular cartilage in Wistar and Fischer 344 rats. Lab Anim Sci. 1995;45:191–4.

    CAS  PubMed  Google Scholar 

  155. Kraus VB, Huebner JL, DeGroot J, Bendele A. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the guinea pig. Osteoarthr Cartil. 2010;18(SUPPL. 3):S35–52.

    Article  Google Scholar 

  156. Schuelert N, Johnson MP, Oskins JL, Jassal K, Chambers MG, McDougall JJ. Local application of the endocannabinoid hydrolysis inhibitor URB597 reduces nociception in spontaneous and chemically induced models of osteoarthritis. Pain. 2011;152:975–81.

    Article  CAS  PubMed  Google Scholar 

  157. Santangelo KS, Pieczarka EM, Nuovo GJ, Weisbrode SE, Bertone AL. Temporal expression and tissue distribution of interleukin-1β in two strains of guinea pigs with varying propensity for spontaneous knee osteoarthritis. Osteoarthr Cartil. 2011;19:439–48.

    Article  CAS  Google Scholar 

  158. Laverty S, Girard CA, Williams JM, Hunziker EB, Pritzker KPH. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the rabbit. Osteoarthr Cartil. 2010;18(SUPPL. 3):S53–65.

    Article  Google Scholar 

  159. BROAD Institute. Rabbit Genome Project.

  160. Garner BC, Stoker AM, Kuroki K, Evans R, Cook CR, Cook JL. Using animal models in osteoarthritis biomarker research. J Knee Surg. 2011;24:251–64.

    Article  PubMed  Google Scholar 

  161. Sanderson RO, Beata C, Flipo RM, Genevois JP, Macias C, Tacke S, et al. Systematic review of the management of canine osteoarthritis. Vet Rec. 2009;164:418–24.

    Article  CAS  PubMed  Google Scholar 

  162. Libicher M, Ivancic M, Hoffmann V, Wenz W. Early changes in experimental osteoarthritis using the Pond-Nuki dog model: Technical procedure and initial results of in vivo MR imaging. Eur Radiol. 2005;15:390–4.

    Article  PubMed  Google Scholar 

  163. Zaki S, Smith MM, Little CB. Pathology-pain relationships in different osteoarthritis animal model phenotypes: it matters what you measure, when you measure, and how you got there. Osteoarthr Cartil. 2021;29:1448–61.

    Article  CAS  Google Scholar 

  164. Little CB, Smith MM, Cake MA, Read RA, Murphy MJ, Barry FP. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in sheep and goats. Osteoarthr Cartil. 2010;18(SUPPL. 3):S80–92.

    Article  Google Scholar 

  165. Cake MA, Appleyard RC, Read RA, Smith MM, Murell GAC, Ghosh P. Ovariectomy alters the structural and biomechanical properties of ovine femoro-tibial articular cartilage and increases cartilage iNOS. Osteoarthr Cartil. 2005;13:1066–75.

    Article  CAS  Google Scholar 

  166. Takroni T, Laouar L, Adesida A, Elliott JAW, Jomha NM. Anatomical study: comparing the human, sheep and pig knee meniscus. J Exp Orthop. 2016;3:35.

  167. McIlwraith CW, Frisbie DD, Kawcak CE, Fuller CJ, Hurtig M, Cruz A. The OARSI histopathology initiative - recommendations for histological assessments of osteoarthritis in the horse. Osteoarthr Cartil. 2010;18(SUPPL. 3):S93–105.

    Article  Google Scholar 

  168. Lindblad-Toh K, Wade CM, Mikkelsen TS, Karlsson EK, Jaffe DB, Kamal M, et al. Genome sequence, comparative analysis and haplotype structure of the domestic dog. Nature. 2005;438:803–19.

    Article  CAS  PubMed  Google Scholar 

  169. Vierboom MPM, Jonker M, Bontrop RE, ’t Hart B. Modeling human arthritic diseases in nonhuman primates. Arthritis Res Ther. 2005;7:145.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  170. Bi X, Li T, Li M, Xiang S, Li J, Ling B, et al. A New Method to Develop the Primate Model of Knee Osteoarthritis With Focal Cartilage Defect. Front Bioeng Biotechnol. 2021;9:1072.

    Article  Google Scholar 

  171. Kalbfleisch TS, Rice ES, DePriest MS, Walenz BP, Hestand MS, Vermeesch JR, et al. Improved reference genome for the domestic horse increases assembly contiguity and composition. Commun Biol. 2018;1:1–8.

    Article  CAS  Google Scholar 

  172. Sebastian A, Murugesh DK, Mendez ME, Hum NR, Rios-Arce ND, McCool JL, Christiansen BA, Loots GG. Global Gene Expression Analysis Identifies Age-Related Differences in Knee Joint Transcriptome during the Development of Post-Traumatic Osteoarthritis in Mice. Int J Mol Sci. 2020;21(1):364. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/ijms21010364.

  173. de Sousa EB, Dos Santos GC, Duarte MEL, Moura Neto V, Aguiar DP. Metabolomics as a promising tool for early osteoarthritis diagnosis. Brazilian J Med Biol Res. 2017;50:1–7.

    Article  Google Scholar 

  174. Mickiewicz B, Heard BJ, Chau JK, Chung M, Hart DA, Shrive NG, et al. Metabolic profiling of synovial fluid in a unilateral ovine model of anterior cruciate ligament reconstruction of the knee suggests biomarkers for early osteoarthritis. J Orthop Res. 2015;33:71–7.

    Article  CAS  PubMed  Google Scholar 

  175. Carlson AK, Rawle RA, Adams E, Greenwood MC, Bothner B, June RK. Application of global metabolomic profiling of synovial fluid for osteoarthritis biomarkers. Biochem Biophys Res Commun. 2018;499:182–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Carlson AK, Rawle RA, Wallace CW, Brooks EG, Adams E, Greenwood MC, et al. Characterization of synovial fluid metabolomic phenotypes of cartilage morphological changes associated with osteoarthritis. Osteoarthr Cartil. 2019;27:1174–84.

    Article  CAS  Google Scholar 

  177. Zhang D, Zhang Y, Xia S, Shen P, Yang C. Metabolic profiling of synovial fluid in human temporomandibular joint osteoarthritis. Front Immunol. 2024;15 March:1–11.

    Google Scholar 

  178. Tuerxun P, Ng T, Zhao K, Zhu P. Integration of metabolomics and transcriptomics provides insights into the molecular mechanism of temporomandibular joint osteoarthritis. PLoS One. 2024;19 5 May:1–17.

    Google Scholar 

  179. Nelson AE, Fang F, Arbeeva L, Cleveland RJ, Schwartz TA, Callahan LF, et al. A machine learning approach to knee osteoarthritis phenotyping: data from the FNIH Biomarkers Consortium. Osteoarthr Cartil. 2019;27:994–1001.

    Article  CAS  Google Scholar 

  180. Kokkotis C, Moustakidis S, Papageorgiou E, Giakas G, Tsaopoulos DE. Machine learning in knee osteoarthritis: A review. Osteoarthr Cartil Open. 2020;2: 100069.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Fan X, Sun AR, Young RSE, Afara IO, Hamilton BR, Ong LJY, Crawford R, Prasadam I. Spatial analysis of the osteoarthritis microenvironment: techniques, insights, and applications. Bone Res. 2024;12(1):7. https://doiorg.publicaciones.saludcastillayleon.es/10.1038/s41413-023-00304-6.

  182. Rockel JS, Sharma D, Espin-Garcia O, Hueniken K, Sandhu A, Pastrello C, et al. Deep learning–based clustering for endotyping and post-arthroplasty response classification using knee osteoarthritis multiomic data. Ann Rheum Dis. 2025;000.

  183. Deng Y, Perry TA, Hulley P, Maciewicz RA, Mitchelmore J, Perry D, et al. Development of methodology to support molecular endotype discovery from synovial fluid of individuals with knee osteoarthritis: The STEpUP OA consortium. PLoS One. 2024;19 11 November:1–24.

    Google Scholar 

  184. Collins KH, Reimer RA, Seerattan RA, Leonard TR, Herzog W. Using diet-induced obesity to understand a metabolic subtype of osteoarthritis in rats. Osteoarthr Cartil. 2015;23:957–65.

    Article  CAS  Google Scholar 

  185. Martel-Pelletier J, Barr AJ, Cicuttini FM, Conaghan PG, Cooper C, Goldring MB, et al. Osteoarthritis. Nat Rev Dis Prim. 2016;2:16072.

  186. Pearce JMS. Disease, diagnosis or syndrome? Pract Neurol. 2011;11:91–7.

    Article  CAS  PubMed  Google Scholar 

  187. Mobasheri A, Matta C, Zákány R, Musumeci G. Chondrosenescence: Definition, hallmarks and potential role in the pathogenesis of osteoarthritis. Maturitas. 2015;80:237–44.

    Article  CAS  PubMed  Google Scholar 

  188. McCulloch K, Litherland GJ, Rai TS. Cellular senescence in osteoarthritis pathology. Aging Cell. 2017;16:210.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  189. Deveza LA, Loeser RF. Is osteoarthritis one disease or a collection of many? Rheumatology (Oxford). 2018;57 suppl_4:iv34–42.

    Article  Google Scholar 

  190. Keshavarz M, Xie K, Schaaf K, Bano D, Ehninger D. Targeting the “hallmarks of aging” to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry. 2022;28:242–55.

    Article  PubMed  PubMed Central  Google Scholar 

  191. Botter SM, Zar M, Van Osch GJVM, Van Steeg H, Dollé MET, Hoeijmakers JHJ, et al. Analysis of osteoarthritis in a mouse model of the progeroid human DNA repair syndrome trichothiodystrophy. Age (Dordr). 2011;33:247–60.

    Article  CAS  PubMed  Google Scholar 

  192. Mason RM, Chambers MG, Flannelly J, Gaffen JD, Dudhia J, Bayliss MT. The STR/ort mouse and its use as a model of osteoarthritis. Osteoarthr Cartil. 2001;9:85–91.

    Article  CAS  Google Scholar 

  193. Evans RG, Collins C, Miller P, Ponsford FM, Elson CJ. Radiological scoring of osteoarthritis progression in STR/ORT mice. Osteoarthr Cartil. 1994;2:103–9.

    Article  CAS  Google Scholar 

  194. Samvelyan HJ, Hughes D, Stevens C, Staines KA. Models of Osteoarthritis: Relevance and New Insights. Calcif Tissue Int. 2021;109:243–56.

    Article  CAS  PubMed  Google Scholar 

  195. Bapat S, Hubbard D, Munjal A, Hunter M, Fulzele S. Pros and cons of mouse models for studying osteoarthritis. Clin Transl Med. 2018;7(1):36. https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40169-018-0215-4.

  196. Staines KA, Poulet B, Wentworth DN, Pitsillides AA. The STR/ort mouse model of spontaneous osteoarthritis – an update. Osteoarthr Cartil. 2017;25:802–8.

    Article  CAS  Google Scholar 

  197. Lv J, Zhang C, Liu X, Gu C, Liu Y, Gao Y, et al. An aging-related immune landscape in the hematopoietic immune system. Immun Ageing. 2024;21:1–20.

    Article  Google Scholar 

  198. Lewis ED, Wu D, Meydani SN. Age-associated alterations in immune function and inflammation. Prog Neuro-Psychopharmacology Biol Psychiatry. 2022;118 May:110576.

    Article  Google Scholar 

  199. Chen MF, Hu CC, Hsu YH, Chiu YT, Chen KL, Ueng SWN, et al. Characterization and Advancement of an Evaluation Method for the Treatment of Spontaneous Osteoarthritis in STR/ort Mice: GRGDS Peptides as a Potential Treatment for Osteoarthritis. Biomedicines. 2023;11(4):1111. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/biomedicines11041111.

  200. Dhollander A, Malone A, Price J, Getgood A. Determination of knee cartilage volume and surface area in beagle dogs: a pilot study. J Exp Orthop. 2017;4:1–8.

    Article  Google Scholar 

  201. Fu Y, Torres BT, Budsberg SC. Model for Canine Gait Analysis. Am J Vet Res. 2010;71:1118–22.

    Article  PubMed  Google Scholar 

  202. Wang Y, Gludish DW, Hayashi K, Todhunter RJ, Krotscheck U, Johnson PJ, et al. Synovial fluid lubricin increases in spontaneous canine cruciate ligament rupture. Sci Rep. 2020;10:1–10.

    CAS  Google Scholar 

  203. Yamazaki A, Edamura K, Tomo Y, Seki M, Asano K. Variations in gene expression levels with severity of synovitis in dogs with naturally occurring stifle osteoarthritis. PLoS One. 2021;16 1 January:1–11.

    Google Scholar 

  204. Frisbie DD, Cross MW, McIlwraith CW. A comparative study of articular cartilage thickness in the stifle of animal species used in human pre-clinical studies compared to articular cartilage thickness in the human knee. Vet Comp Orthop Traumatol. 2006;19(3):142–6.

    Article  CAS  PubMed  Google Scholar 

  205. Frasca D. Several areas of overlap between obesity and aging indicate obesity as a biomarker of accelerated aging of human B cell function and antibody responses. Immun Ageing. 2022;19:48.

  206. Nunan E, Wright CL, Semola OA, Subramanian M, Balasubramanian P, Lovern PC, et al. Obesity as a premature aging phenotype - implications for sarcopenic obesity. GeroScience. 2022;44:1393–405.

    Article  PubMed  PubMed Central  Google Scholar 

  207. Nedunchezhiyan U, Varughese I, Sun ARJ, Wu X, Crawford R, Prasadam I. Obesity, Inflammation, and Immune System in Osteoarthritis. Front Immunol. 2022;13:3505.

    Article  Google Scholar 

  208. Andersson M, Haglund E, Aili K, Bremander A, Bergman S. Associations between metabolic factors and radiographic knee osteoarthritis in early disease - a cross-sectional study of individuals with knee pain. BMC Musculoskelet Disord. 2022;23:1–11.

    Article  Google Scholar 

  209. Chen X, Liu J, Wang G, Sun Y, Ding X, Zhang X. Regulating lipid metabolism in osteoarthritis: a complex area with important future therapeutic potential. Ann Med. 2024;56(1):2420863. https://doiorg.publicaciones.saludcastillayleon.es/10.1080/07853890.2024.

  210. Liu H, Witzigreuter L, Sathiaseelan R, Agbaga MP, Brush RS, Stout MB, et al. Obesity promotes lipid accumulation in mouse cartilage—A potential role of acetyl-CoA carboxylase (ACC) mediated chondrocyte de novo lipogenesis. J Orthop Res. 2022;40:2771–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Iwata M, Ochi H, Hara Y, Tagawa M, Koga D, Okawa A, et al. Initial Responses of Articular Tissues in a Murine High-Fat Diet-Induced Osteoarthritis Model: Pivotal Role of the IPFP as a Cytokine Fountain. PLoS ONE. 2013;8:60706.

    Article  Google Scholar 

  212. Asou Y, Iwata M, Ochi H, Ailixiding M, Aibibula Z, Piao J, et al. Pleiotropic functions of high fat diet in the etiology of osteoarthritis. PLoS ONE. 2016;11:1–14.

    Article  Google Scholar 

  213. van der Kraan PM. Osteoarthritis and a high-fat diet: The full “OA syndrome” in a small animal model. Arthritis Res Ther. 2010;12:1–2.

    Google Scholar 

  214. Griffin TM, Fermor B, Huebner JL, Kraus VB, Rodriguiz RM, Wetsel WC, et al. Diet-induced obesity differentially regulates behavioral, biomechanical, and molecular risk factors for osteoarthritis in mice. Arthritis Res Ther. 2010;12:R130.

    Article  PubMed  PubMed Central  Google Scholar 

  215. Brunner AM, Henn CM, Drewniak EI, Lesieur-Brooks A, Machan J, Crisco JJ, et al. High dietary fat and the development of osteoarthritis in a rabbit model. Osteoarthr Cartil. 2012;20:584–92.

    Article  CAS  Google Scholar 

  216. Gierman LM, van der Ham F, Koudijs A, Wielinga PY, Kleemann R, Kooistra T, et al. Metabolic stress-induced inflammation plays a major role in the development of osteoarthritis in mice. Arthritis Rheum. 2012;64:1172–81.

    Article  CAS  PubMed  Google Scholar 

  217. Gierman LM, Kühnast S, Koudijs A, Pieterman EJ, Kloppenburg M, Van Osch GJVM, et al. Osteoarthritis development is induced by increased dietary cholesterol and can be inhibited by atorvastatin in APOE*3Leiden.CETP mice--a translational model for atherosclerosis. Ann Rheum Dis. 2014;73:921–7.

    Article  CAS  PubMed  Google Scholar 

  218. Brandt KD, Braunstein EM, Visco DM, O’Connor B, Heck D, Albrecht M. Anterior (cranial) cruciate ligament transection in the dog: A bona fide model of osteoarthritis, not merely of cartilage injury and repair. J Rheumatol. 1991;18:436–46.

    CAS  PubMed  Google Scholar 

  219. Flanigan DC, Harris JD, Brockmeier PM, Siston RA. The effects of lesion size and location on subchondral bone contact in experimental knee articular cartilage defects in a bovine model. Arthrosc - J Arthrosc Relat Surg. 2010;26:1655–61.

    Article  Google Scholar 

  220. Narez GE, Fischenich KM, Donahue TLH. Experimental animal models of post-traumatic osteoarthritis of the knee. Orthop Rev (Pavia). 2020;12:95–103.

    Article  Google Scholar 

  221. Atkinson PJ, Cooper TG, Anseth S, Walter NE, Kargus R, Haut RC. Association of knee bone bruise frequency with time postinjury and type of soft tissue injury. Orthopedics. 2008;31(5):440. https://doiorg.publicaciones.saludcastillayleon.es/10.3928/01477447-20080501-01.

  222. Loeser RF. Aging processes and the development of osteoarthritis. Curr Opin Rheumatol. 2013;25:108–13.

    Article  PubMed  PubMed Central  Google Scholar 

  223. Rivellese F, Pitzalis C. Cellular and molecular diversity in Rheumatoid Arthritis. Semin Immunol. 2021;58 January:101519.

    Article  Google Scholar 

  224. Lliso-Ribera G, Humby F, Lewis M, Nerviani A, Mauro D, Rivellese F, et al. Synovial tissue signatures enhance clinical classification and prognostic/treatment response algorithms in early inflammatory arthritis and predict requirement for subsequent biological therapy: Results from the pathobiology of early arthritis cohort (PEAC). Ann Rheum Dis. 2019;78:1642–52.

    Article  CAS  PubMed  Google Scholar 

  225. Li W, Li Z, Zou Z, Liu X, Li X. Integrated single-cell and bulk RNA sequencing identifies POSTN as a potential biomarker and therapeutic target for rheumatoid arthritis. Gene. 2024;928 April:148798.

    Article  Google Scholar 

  226. Figueiredo ML. Applications of single-cell RNA sequencing in rheumatoid arthritis. 2024;November:1–9.

    Google Scholar 

  227. Smith MH, Gao VR, Periyakoil PK, Kochen A, DiCarlo EF, Goodman SM, et al. Drivers of heterogeneity in synovial fibroblasts in rheumatoid arthritis. Nat Immunol. 2023;24:1200–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Armaka M, Konstantopoulos D, Tzaferis C, Lavigne MD, Sakkou M, Liakos A, et al. Single-cell multimodal analysis identifies common regulatory programs in synovial fibroblasts of rheumatoid arthritis patients and modeled TNF-driven arthritis. Genome Med. 2022;14:1–25.

    Article  Google Scholar 

  229. Cheng L, Wang Y, Wu R, Ding T, Xue H, Gao C, et al. New Insights From Single-Cell Sequencing Data: Synovial Fibroblasts and Synovial Macrophages in Rheumatoid Arthritis. Front Immunol. 2021;12 July:1–11.

    Google Scholar 

  230. Jo J, Xu G, Cao M, Marquardt A, Francis S, Gandikota G, et al. A Functional Study of Human Inflammatory Arthritis Using Photoacoustic Imaging. Sci Rep. 2017;7:1–9.

    Article  Google Scholar 

  231. Hügle M, Kalweit G, Hügle T, Boedecker J. A Dynamic Deep Neural Network for Multimodal Clinical Data Analysis. Stud Comput Intell. 2021;914:79–92.

    Article  Google Scholar 

  232. Wang Z, Tong Z, Chen H, Nie G, Hu J, Liu W, et al. Photoacoustic/ultrasonic dual-mode imaging for monitoring angiogenesis and synovial erosion in rheumatoid arthritis. Photoacoustics. 2023;29: 100458.

    Article  PubMed  PubMed Central  Google Scholar 

  233. Huang Z, Liu D, Mo S, Hong X, Xie J, Chen Y, et al. Multimodal PA/US imaging in Rheumatoid Arthritis: Enhanced correlation with clinical scores. Photoacoustics. 2024;38: 100615.

    Article  PubMed  PubMed Central  Google Scholar 

  234. Yang M, Zhao C, Wang M, Wang Q, Zhang R, Bai W, et al. Synovial Oxygenation at Photoacoustic Imaging to Assess Rheumatoid Arthritis Disease Activity. Radiology. 2023;306:220–8.

    Article  PubMed  Google Scholar 

  235. Melanie H Smith, Vianne R Gao, Michail Schizas, Alejandro Kochen EFD, Susan M Goodman, Thomas M Norman, Laura T Donlin, Christina S Leslie and A, Rudensky Y. Heterogeneity of Inflammation-associated Synovial Fibroblasts in Rheumatoid Arthritis and Its Drivers. 2022;:1–40.

  236. Salehi F, Lopera Gonzalez LI, Bayat S, Kleyer A, Zanca D, Brost A, et al. Machine Learning Prediction of Treatment Response to Biological Disease-Modifying Antirheumatic Drugs in Rheumatoid Arthritis. J Clin Med. 2024;13(13):3890. https://doiorg.publicaciones.saludcastillayleon.es/10.3390/jcm13133890.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  237. Lee S, Kang S, Eun Y, Won HH, Kim H, Lee J, et al. Machine learning-based prediction model for responses of bDMARDs in patients with rheumatoid arthritis and ankylosing spondylitis. Arthritis Res Ther. 2021;23:1–12.

    Article  CAS  Google Scholar 

  238. Zhao H, Ou L, Zhang Z, Zhang L, Liu K, Kuang J. The value of deep learning-based X-ray techniques in detecting and classifying K-L grades of knee osteoarthritis: a systematic review and meta-analysis. Eur Radiol. 2024;:327–40.

  239. Momtazmanesh S, Nowroozi A, Rezaei N. Artificial Intelligence in Rheumatoid Arthritis: Current Status and Future Perspectives: A State-of-the-Art Review. Rheumatol Ther. 2022;9(5):1249–304. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s40744-022-00475-4.

    Article  PubMed  PubMed Central  Google Scholar 

  240. Tiwari A, Poduval M, Bagaria V. Evaluation of artificial intelligence models for osteoarthritis of the knee using deep learning algorithms for orthopedic radiographs. World J Orthop. 2022;13:603–14.

    Article  PubMed  PubMed Central  Google Scholar 

  241. Gharibshahian M, Torkashvand M, Bavisi M, Aldaghi N, Alizadeh A. Recent advances in artificial intelligent strategies for tissue engineering and regenerative medicine. Ski Res Technol. 2024;30:1–15.

    Google Scholar 

  242. Tariq T, Suhail Z, Nawaz Z. A Review for automated classification of knee osteoarthritis using KL grading scheme for X-rays. Biomed Eng Lett. 2024;15(1):1–35. https://doiorg.publicaciones.saludcastillayleon.es/10.1007/s13534-024-00437-5.

  243. Lascelles BDX, Brown DC, Maixner W, Mogil JS. Spontaneous painful disease in companion animals can facilitate the development of chronic pain therapies for humans. Osteoarthr Cartil. 2018;26:175–83.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The graphical abstract was created with BioRender.com.

Use of AI

The AI tools ChatGPT- 4 and Grammarly (basic version) were used for syntax and grammar correction.

Funding

Open access funding provided by TU Wien (TUW). The Medical University of Vienna funded this work.

Author information

Authors and Affiliations

Authors

Contributions

Eva Reihs (ER), Anita Fischer (AF), Iris Gerner (IG), Reinhard Windhager (RW), Stefan Toegel (ST), Frank Zaucke (FZ), Mario Rothbauer (MR), and Florien Jenner (FJ) have made substantial contributions to the conception and the design of the review. (ER), (AF), (IG), (RW), (ST), (FZ), (MR), and (FJ) performed either literature research, interpretation of data, and/or provided substantial information. (ER), (IG), (ST), (FZ), (MR), and (FJ) have drafted the work and ER), (AF), (IG), (RW), (ST), (FZ), (MR), and (FJ) substantively revised it. All authors reviewed the manuscript. (ER), (AF), (IG), (RW), (ST), (FZ), (MR), and (FJ) have approved the submitted version.

Corresponding author

Correspondence to Mario Rothbauer.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reihs, E., Fischer, A., Gerner, I. et al. Beyond symptomatic alignment: evaluating the integration of causal mechanisms in matching animal models with human pathotypes in osteoarthritis research. Arthritis Res Ther 27, 109 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-025-03561-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s13075-025-03561-4

Keywords