Next Article in Journal
CAR Based Immunotherapy of Solid Tumours—A Clinically Based Review of Target Antigens
Previous Article in Journal
Molecular Mechanisms Behind the Role of Plasmacytoid Dendritic Cells in Systemic Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Induced Models of Osteoarthritis in Animal Models: A Systematic Review

by
Umile Giuseppe Longo
1,2,*,
Rocco Papalia
1,2,
Sergio De Salvatore
1,2,3,
Riccardo Picozzi
1,2,
Antonio Sarubbi
1,2 and
Vincenzo Denaro
1,2
1
Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo 200, 00128 Roma, Italy
2
Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Roma, Italy
3
Department of Orthopedics, Children’s Hospital Bambino Gesù, 00165 Roma, Italy
*
Author to whom correspondence should be addressed.
Biology 2023, 12(2), 283; https://doi.org/10.3390/biology12020283
Submission received: 13 December 2022 / Revised: 5 February 2023 / Accepted: 6 February 2023 / Published: 10 February 2023

Abstract

:

Simple Summary

This study aimed to determine the most appropriate animal model for the study of osteoarthritis (OA). OA is a common joint disease and can be induced in animal models through mechanical, surgical, or chemical methods. The study conducted a literature review of articles from January 2010 to July 2021 on various databases such as Medline, Embase, and Google Scholar. After reviewing 1621 articles, 36 studies were selected that included a total of 1472 animals. The results showed that various induction models, including mechanical, surgical, and chemical, have been proven to be suitable for inducing OA in animals. However, there is still not a clear "gold standard" for choosing the best animal model for OA. This study provides valuable information for researchers to make informed decisions about the appropriate animal model for their specific OA research questions. The findings of this study will be valuable to society by helping to advance our understanding of OA and develop better treatments for this debilitating disease.

Abstract

The most common induction methods for OA are mechanical, surgical and chemical. However, there is not a gold standard in the choice of OA animal models, as different animals and induction methods are helpful in different contexts. Reporting the latest evidence and results in the literature could help researchers worldwide to define the most appropriate indication for OA animal-model development. This review aims to better define the most appropriate animal model for various OA conditions. The research was conducted on the following literature databases: Medline, Embase, Cinahl, Scopus, Web of Science and Google Scholar. Studies reporting cases of OA in animal models and their induction from January 2010 to July 2021 were included in the study and reviewed by two authors. The literature search retrieved 1621 articles, of which 36 met the selection criteria and were included in this review. The selected studies included 1472 animals. Of all the studies selected, 8 included information about the chemical induction of OA, 19 were focused on mechanical induction, and 9 on surgical induction. Nevertheless, it is noteworthy that several induction models, mechanical, surgical and chemical, have been proven suitable for the induction of OA in animals.

1. Introduction

Osteoarthritis (OA) is the most prevalent form of arthritis among middle-aged and elderly individuals [1]. The etiology of OA is multi-factorial, and the underlying pathogenic mechanisms are not fully understood. While the progression of the disease is typically slow, it can result in significant pain and disability, negatively impacting the quality of life. Studying OA in humans can be difficult, hence animal models have been utilized to assess OA development and evaluate novel treatments.
Several animal models have been developed for the study of OA, but it can be challenging to determine the most suitable one for specific research purposes [2]. Ideal experimental models should accurately mimic the disease and enable the joint analysis of relevant tissues using biomechanical, radiographic and microscopic evaluations. Common surgical models of knee OA include the anterior cruciate ligament transection model (ACLT), the partial medial meniscectomy model or a combination of both [3]. Other examples include rat ACLT and partial meniscectomy in rabbits. A widely studied and well-evaluated mouse model involves destabilizing the medial meniscus by surgically transecting the tibial ligament. To examine the potential pathogenic mechanisms leading to morphological changes, researchers have analyzed the morphological responses of the articular cartilage and subchondral bone after surgical instability, mechanical unloading and intra-articular injection [4]. Induction methods for OA include mechanical, surgical and chemical methods; however, there is no gold standard for choosing OA animal models, as different animals and induction methods are useful in different situations.
The aim of this review is to clearly define the most reliable animal model for different OA conditions by presenting the newest evidence and outcomes found in the literature and using a systematic approach. This could assist researchers worldwide in determining the most accurate indication for developing an OA animal model, discover new ways to study this condition and test pharmaceuticals or surgical techniques to combat the disease.

2. Materials and Methods

2.1. Study Selection

The PIOS approach (Patient (P), Intervention (I), Outcome (O) and Study Design (S)) was used to formulate the research question. This research aimed to report the most reliable animal models (P) and their induction methods (I) to better study OA development (O) in different contexts.

2.2. Inclusion Criteria

Only studies that reported cases of OA in animal models made through mechanical, surgical and chemical inductions were included.
Only studies regarding knee, hip, shoulder and spine were included. Only English-language articles were considered. The Oxford categorization was used to filter peer-reviewed publications at each level of evidence.

2.3. Exclusion Criteria

Studies focusing on spontaneous OA animal models were excluded, as were studies regarding human models of OA and other forms of arthritis. Studies that selected genetic manipulations as a method of induction of OA in animal models were also discarded. Only the most recent literature was evaluated; therefore, studies conducted prior to 2010 were also excluded. Technical notes, letters to editors and instructional courses were excluded. Studies with a sample size of less than ten animals were deemed ineligible for the current study. Cases involving temporomandibular joint OA were also excluded. Studies with specific therapeutic purposes and drug tests were excluded, specifically, OA inductions following diets, oral administration of drugs and biological alterations.

2.4. Search

The systematic review was carried out using the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) criteria. Bibliographic databases Medline, Embase, Cinahl, Scopus, Web of Science and Google Scholar were searched. The following was the string-searching used: ((“models, animal”[MeSH Terms] OR (“models”[All Fields] AND “animal”[All Fields]) OR “animal models”[All Fields] OR (“animal”[All Fields] AND “model”[All Fields]) OR “animal model”[All Fields]) AND (“osteoarthritis”[MeSH Terms] OR “osteoarthritis”[All Fields] OR “osteoarthritides”[All Fields]) AND (“chemical”[All Fields] OR “chemicals”[All Fields] OR “chemically”[All Fields] OR “chemicals”[All Fields]) OR “surgical procedures, operative”[MeSH Terms] OR (“surgical”[All Fields] AND “procedures”[All Fields] AND “operative”[All Fields]) OR “operative surgical procedures”[All Fields] OR “surgical”[All Fields] OR “surgically”[All Fields] OR “surgicals”[All Fields] OR (“mechanical”[All Fields] OR “mechanically”[All Fields] OR “mechanicals”[All Fields] OR “mechanics”[MeSH Terms] OR “mechanics”[All Fields] OR “mechanic”[All Fields])) NOT (“mesenchym”[All Fields] OR “mesenchymal”[All Fields] OR “mesenchymalized”[All Fields] OR “mesenchymally”[All Fields] OR “mesenchymals”[All Fields] OR “mesenchymes”[All Fields] OR “mesoderm”[MeSH Terms] OR “mesoderm”[All Fields] OR “mesenchyme”[All Fields]))) AND (english[Filter]). All the studies included were screened for additional references.
Two of the authors (R.P and A.S) performed the searches between May and July 2021. Articles which were published between January 2010 and July 2021 were searched.

2.5. Data Collection Process

Two of the authors (R.P. and A.S.) worked independently on the data collection method. A third reviewer was brought in to resolve any disagreements (S.D.S). The following was the screening procedure: R.P. and A.S. began by reviewing the title and abstract, then moved on to the full-text version. The articles that were not rejected based on titles and abstracts were read in their entirety. In the case of disagreement, S.D.S. intermediated. The number of publications included and excluded was reported using the PRISMA flowchart [5].

2.6. Data Items

Data regarding primary author, year of publication, animal species, level of evidence, sample size, sex, checkpoints, type of induction and classification type (mechanical, surgical and chemical), and joint involved were extracted.

2.7. Risk of Bias

The Risk of Bias in Non-Randomized Studies of Interventions methodological index for non-randomized studies (MINORS) [6] by Cochrane was used to assess the possibility of bias in the included studies. Authors R.P. and A.S. separately scored the articles that were chosen. Any disagreements were resolved by the third reviewer, S.D.S.

3. Results

3.1. Study Selection

Overall, 1621 articles were found from the search of the relevant literature; however, after eliminating duplicates, only 1358 remained. Based on the title and abstract screening, 1298 of the 1358 papers were eliminated. A total of 60 articles were screened in full-text, and 24 were excluded due to a lack of data on population and accuracy (n = 5), incoherent conclusions on OA induction models (n = 5), a number of participants lower than 10 (n = 3) or unspecified data on population (n = 11). As a result, 36 articles passed the final screening and were included in the analysis. Figure 1 depicts the screening procedure.

3.2. Study Characteristics

The 36 selected articles included a total of 1434 animals, which were selected for the study. The final studies selected by the reviewers included the following level of evidence: Level IV 36 case-control studies. Eight of the studies included information about the chemical induction of OA [1,7,8,9,10,11,12,13], nineteen of the studies were focused on mechanical induction [4,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35] and nine of the studies on induced OA through surgery [3,17,20,21,22,35,36,37,38]. None of the included studies adopted the genetic induction model; therefore, this item was not assessed. In addition, 35 studies specified the timing at which the animals were checked, making the longest follow-up at 20 weeks post-induction. All the study characteristics are summarized in Table 1 and Table 2.

3.3. Quality of Evidence

Using the MINORS tool, thirty-six studies were scored as having a “high risk of bias”. The studies examined were all similar in design, with several missing follow-ups [40] or failing to account for potential confounding areas among the variables. The RoB-2 tool was not used, as randomized clinical trials were not found. The MINORS’ screening process is reported in Table 3.

3.4. Diagnostic Procedure

In twenty-three of the studies, the diagnostic procedure involved to evaluate the cartilage severity damage was the histological method [3,4,8,9,11,12,13,15,17,18,19,20,21,22,24,37,38,39]. Eleven studies selected the µCT scan as a radiological investigation method [4,7,8,13,15,21,23,27,28,29,31,32,33,37,38], either in combination with the histological test or individually, in order to diagnose the damage to the joint structures. In only two studies [15,37], magnetic resonance imaging (MRI) [41] was used for the diagnosis. The diagnostic timing varied across the studies considered. All the data are reported in Table 2.

3.5. Animal Models

In the 36 studies selected, several animal species were used for the OA evaluation process, with mice being the most utilized animal model. Fifteen [4,21,23,25,26,27,28,30,31,32,33,34,35,39,42] authors selected male and female C57BL/6N mice, CBA mice, Str/ort mice, FVB strain mice, LGXSM-6 and LGXSM-33 mice. C5BL/6N mice and CBA mice were selected as the most suitable species for the experiments. However, eighteen authors [1,3,4,7,8,9,10,11,12,13,14,15,17,18,19,22,24,36,37,38,39] preferred to study and induce OA in other animal species, such as Wistar Rats, Sprague Dawley rats, Lewis rats, Flemish Giant and New Zealand rabbits. Furthermore, two other authors [16,20] selected pigs and Suffolk-cross sheep, respectively. All the data are reported in Table 2.

3.6. Type of Induction

Of the eight studies that specified OA induction in animals utilizing a chemical approach, five were caused by an intra-articular injection of monoiodoacetate (MIA) [1,7,8,12,13]. In contrast, the other three models were determined by an injection of collagenase [11], a urinary plasminogen activator injection [9] and an injection of complete Freund’s adjuvant [10], respectively. The majority of the studies selected focused on the mechanical inductive approach through in vivo mechanical loadings. Conversely, of the nine studies focused on the surgical approach for the induction of OA in animals, four performed a surgical destabilization by ACLT [17,20,37,38], three performed a medial meniscus debridement (MMD) [3,21,35], one performed a medial collateral ligament tear (MCLT) [36] and one performed sham surgery [22]. All the data are reported in Table 2.

3.7. Type of Joint Involved

In the OA induction process, the authors focused on a specific joint in a specific animal model. As a consequence, the knee joint resulted as the most selected one, remarkably in murine animal models. For the knee joint, 16 studies preferred to induce OA in mice [4,15,21,25,26,27,28,29,30,31,32,34,35,39,43,44], 9 in rats [1,7,11,12,13,14,17,19,36,38], 5 in rabbits [3,18,22,24,37] and only 1 in sheep [20] and pigs [12], respectively. Subsequently, in the rat animal models, two papers elected the lumbar facet joints [18,19] and, similarly, OA was induced in the hip joint [13] by only one author. All the data are reported in Table 2.

4. Discussion

OA is considered to be one of the most significant diseases of the future due to the aging population [45,46,47].
Finding new solutions for studying this condition and testing drugs or surgical methods is crucial. Reporting the latest findings in the international literature can assist researchers worldwide in conducting more accurate studies using the most appropriate animal models for OA conditions. This paper has reviewed the latest literature to gather information about different animal models and induction methods for OA.
The reviewed studies utilized mechanical, surgical and chemical methods to induce OA. Most of the studies used mechanical induction, while surgical and chemical induction were performed less frequently. The studies primarily focused on the mechanical approach for inducing OA in animals using mechanical loading devices. In contrast, the surgery-induced approach focused on trauma caused by (micro)surgery, including ACLT, MMD, MCLT and MMT [18].
Diet, drugs, age, environmental pollution and many other factors have an impact on the synovial fluid’s composition and, therefore, its lubricating properties [48,49]. The major cause of OA, excluding mechanical or surgical injuries, are changes in the chemical composition of the synovial fluid. This is also the reason why there is contact between the articular surfaces; their abrasion and damage is identified as OA. Sulaiman et al. [50] compared the synovial fluid proteomics changes in surgical- and chemical-induced OA rabbit models. This study demonstrated that the surgically induced model showed a wider range of proteome profile; conversely, the chemically induced joints had a slower OA progression, showing inflammatory changes at the early phase but having a decreased expression at the later stages. Although several OA animal models have been reported, it is unknown at this time if any of them truly display these “ideal” characteristics. OA and other musculoskeletal diseases are frequently studied using mice, which are a practical model species [43].

4.1. Mechanical Models

Non-invasive mechanical mouse models of OA that do not require surgery or invasive techniques are a valuable advancement in studying the mechanisms and treatments of OA. Compared with invasive injury models, these non-invasive models initiate joint degradation in a more controlled and accurate manner, replicating human damage circumstances. Mechanical force is applied externally to the joint to cause harm to the bones, cartilage or soft tissues [43]. Some studies, such as that of Rai et al. [15], used a multi-cyclic loading pattern to cause significant cartilage fibrillation and degradation over a prolonged period. On the other hand, Christiansen and colleagues [23] used a single-cycle loading strategy that induced ACL rupture and resulted in mild OA over a longer period, with cartilage lesions observed in all the compartments of the knee joint. This approach released energy that likely reduced direct injury to the cartilage and indicates that joint destabilization, rather than direct cartilage injury, is caused by a single episode of tibial compression. In Rai et al.’s model [15], the loading pattern directly impacted the articular cartilage, resulting in a more accurate representation of human knee trauma. Both models suggest that human OA is usually the result of a series of loading events rather than a single traumatic experience.

4.2. Surgically Induced Models

Surgical damage techniques such as anterior cruciate ligament transection (ACLT) induce joint degradation by rupturing ligaments and menisci, altering the joint’s stability and biomechanics [13,43]. However, as noted by Mohan et al. [13], ACL tears often accompany acute meniscal damage, which is not accounted for in ACLT models. The meniscal transection (mACLT) model destabilizes the knee by transecting the ACL and partially transecting the meniscus. The tibiofemoral compressive impact model (ACLF) causes ACL rupture and injury to adjacent tissues, including the meniscus, through a single blunt-force hit to the tibiofemoral joint [13]. The surgically induced OA model (DMM) in mice, according to McNulty et al. [35], is highly reproducible, inducing moderate to severe articular cartilage lesions in young mice within 8 weeks post-surgery. Lorenz et al. [46] suggest considering gender-specific effects and evaluating data separately for males and females in mouse models, as sex hormones play a crucial role in OA development in DMM surgery models. Foxa et al. [47] described generating an OA-like model in mice through bilateral DMM surgery, with male mice being excellent subjects due to their increased likelihood of developing OA. The shift of the tibia anterior to the femur after DMM surgery, as noted by Moodie and colleagues [21], significantly increases the tibial AP range of motion, potentially damaging the ACL due to the proximity of the medial meniscal tibial ligament to the ACL’s tibial insertion [35].

4.3. Chemical Models

Chemical models of OA have a distinct pathophysiology and bear no resemblance to post-traumatic OA. They are also less invasive than surgical models [48]. The ease of induction and reproducibility make them ideal for short-term trials [48,49]. Pitcher et al. [49] reported that MIA models are responsive to conventional pain-relieving therapies, making it possible to test preventive and therapeutic procedures for OA pain management in compound development. However, Mohan et al. [13] noted that the gradual progression of OA in chemical induction models may be superior, but the data are ambiguous.
Currently, the most utilized type of chemical induction is MIA, which has an inhibitory activity of glyceraldehyde-3-phosphate dehydrogenase glycolysis and induces the death of chondrocytes, according to Morais et al. [7]. In both rodents and non-rodents, the intra-articular injection causes chondrocyte destruction. When administered in rats, it causes cartilage lesions with loss of proteoglycan matrix and functional alterations with stiffness that are analogous to those seen in human osteoarthritis [7]. The study by Miyamoto et al. [8] developed a rat hip OA model induced by an intra-articular injection of MIA. The histological appearance of MIA-injected hips showed an OA-like appearance. The current work describes the temporally dependent course of radiographic and subchondral bone alterations in the rat hip histology. It is very interesting to observe how osteoarthritis progresses over time following the MIA injection [8].

4.4. Animal Models Characteristics

McCoy et al. [51] suggested that, as with any species, only skeletally mature mice should be used in models of OA. Likewise, in rats, the recommended age for this species to participate in any study is 3 months. The earliest age at which it is recommended to include guinea pigs in any study is 6 months of age, by which time spontaneous lesions may already be established. Finally, in sheep and goats, it should be noted that skeletal maturity may not be reached until at least 2 years of age.
For the study of the pathogenesis and pathophysiology of the disease, Serra et al. [52] observed that small animals (mice, rats, guinea pigs and rabbits) are the most frequently utilized models, primarily because these models have a faster disease progression and are relatively cheap and simple to handle. Based on the results proposed by Ding et al. [53], the rat chondrocyte inflammation model may help in the study of the early pathological mechanism of OA, demonstrating considerable differences from mice. Therefore, various animal species have been found to be relevant to the OA evaluation process, with mice being the most used surgical and mechanical induction model. In contrast, rats are equally valuable to the latter for chemical induction.
Moreover, sheep and rabbits have also been considered acceptable for the mechanical method. Even though the heterogeneity of animal species appears as an important tool to evaluate different aspects of the osteoarthritic event, these models’ main drawbacks include their small size, which reduces the quantity of tissue available for biochemical studies, and the differences in tissue structure and joint mechanics between these species and humans. [51].
The fact that mice are much smaller than humans leads to noticeably different biomechanical loads on their joints, which cannot be overlooked. Additionally, the cartilage anatomy of mice differs noticeably from that of humans in, for instance, the overall cartilage thickness (on average, 30 mm; 50-fold thinner than in humans), a thick layer of calcified cartilage and a lack of distinct superficial, transitional and radial zones of chondrocytes [51]. Because rat articular cartilage is thicker (*0.1 mm) than mouse articular cartilage, whole and partial thickness lesions in rat cartilage can be created to examine therapeutic approaches for cartilage healing. However, the biomechanics of the rat’s stifle-joint differs significantly from that of the human joint, resulting in distinct patterns of cartilage stress [51].
Rabbits have been used for decades to induce OA both chemically and surgically despite the marked differences in joint biomechanics and gait when compared to humans. Rabbit stifles have a significantly higher flexion angle than that of humans. In addition, rabbit cartilage is 10 times thinner than human cartilage (0.3–0.7 vs. 2–3 mm) but has a much higher chondrocyte density [51]. Sheep and pigs represent an advantageous model due to their larger size, which means easier maneuverability, amenability to diagnostic imaging, effortless synovial fluid collection, arthroscopic intervention and postoperative management.
Sheep are favored as a large animal species due to the marked similarity between their stifle anatomy and that of the human knee. Pigs, on the other hand, differ somewhat in size with respect to the cruciate ligament and width of the meniscus, but the remarkable similarities with humans in gastrointestinal physiology and the immune system make this species an attractive option for testing pharmaceutical and biological interventions [51].
Throughout this study, it has been highlighted that sixteen authors preferred to induce OA in knee joints predominantly in mice, ten in rats, five in rabbits and only one in sheep and pigs, respectively. Moreover, two studies selected lumbar facet joints in rats, and only one paper chose hip joints in rats. The knee joints and lumbar facet joints resulted in the most used anatomical models. Shuang et al. [10] highlighted them as the most prone to destabilization to induce OA. Moreover, there is only one author [8] who preferred to use the hip joint to induce OA.

4.5. Limitations

The differences between studies in terms of follow-up, animal model and type of induction made it impossible to perform a meta-analysis. While numerous animal models of OA have been described, no consensus exists on the injury methods used to induce OA development, since joint degeneration likely depends on the injury method used. Moreover, various scales have been used to describe the settlement of the osteoarthritic event, showing a preference for the Osteoarthritis Research Society International (OARSI) histopathology grading and the Mankin systems [54]. The OARSI scale assigns a grade of damage from 0 to 6, based on the depth of OA advancement into the cartilage, and a stage of damage from 0 to 4, based on cartilage involvement. The combined value of grade and stage (score range 0–24) determines the final score. On the other hand, the Mankin scoring system has been established for grading OA changes. According to Miyamoto et al., Mankin [8] ratings are based on five factors [7]. These include shape, chondrocyte quantity, chondrocyte clustering, proteoglycan content, subchondral bone plate and/or tidemark modifications, and a vascular invasion into cartilage. Low (1–3 points), moderate (4–7 points) and high (>8 points) total points were assigned. Due to an inhomogeneous or lacking ranking scale, it has been found difficult to classify the most suitable animal model for OA evaluation. Moreover, OA development is not a linear process. As reported in the article published by Longo et al. [55] in 2021, the type of animal model used and the circumstances in the stables and cages may have an impact on the experiment outcomes.
The animal model of OA could be spontaneous or induced; however, the topic of the current review is focused on induced models. Spontaneous models of OA include a wide number of articles and topics of discussion; therefore, these models were not included as they deserve a dedicated article. Further reviews are required to report the most recent findings on spontaneous models.
All the articles included reported a high risk of bias according to the MINORS scale. Lastly, it was not possible to perform a meta-analysis due to the high heterogeneity between the studies.

5. Conclusions

In the field of OA research, animal models are commonly used, but a universally accepted method has yet to be established. The varying OA grading scales make it challenging to determine the severity of OA. While mice are frequently used in studies, there is still a significant diversity in the choice of animals. Despite this, several induction models, including mechanical, surgical and chemical, have been found to be effective in inducing OA in animals, with the mechanical model being the most popular and consistent with the OA process, the surgical model being the most invasive, and the chemical model being the most time-dependent. To better understand OA development, further high-quality studies with consistent sample sizes and classification methods are needed to identify the most valid and reliable animal model.

Author Contributions

Conceptualization, U.G.L. and V.D.; methodology, S.D.S.; software, R.P. (Rocco Papalia); validation, R.P. (Rocco Papalia), A.S. and R.P. (Riccardo Picozzi); formal analysis, A.S.; investigation, S.D.S.; resources, V.D.; data curation, A.S.; writing—original draft preparation, R.P. (Riccardo Picozzi); writing—review and editing, S.D.S.; visualization, A.S.; supervision, R.P. (Rocco Papalia); project administration, U.G.L.; funding acquisition, V.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The datasets generated and/or analyzed during the current study are not publicly available due to analyses being underway for subsequent publications. However, they are available from the corresponding author on reasonable request.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Korostynski, M.; Malek, N.; Piechota, M.; Starowicz, K. Cell-type-specific gene expression patterns in the knee cartilage in an osteoarthritis rat model. Funct. Integr. Genom. 2018, 18, 79–87. [Google Scholar] [CrossRef]
  2. Longo, U.G.; Loppini, M.; Fumo, C.; Rizzello, G.; Khan, W.S.; Maffulli, N.; Denaro, V. Osteoarthritis: New Insights in Animal Models. Open Orthop. J. 2012, 6, 558–563. [Google Scholar] [CrossRef] [PubMed]
  3. Arunakul, M.; Tochigi, Y.; Goetz, J.E.; Diestelmeier, B.W.; Heiner, A.D.; Rudert, J.; Fredericks, D.C.; Brown, T.D.; McKinley, T.O. Replication of chronic abnormal cartilage loading by medial meniscus destabilization for modeling osteoarthritis in the rabbit knee in vivo. J. Orthop. Res. 2013, 31, 1555–1560. [Google Scholar] [CrossRef] [PubMed]
  4. Nomura, M.; Sakitani, N.; Iwasawa, H.; Kohara, Y.; Takano, S.; Wakimoto, Y.; Kuroki, H.; Moriyama, H. Thinning of articular cartilage after joint unloading or immobilization. An experimental investigation of the pathogenesis in mice. Osteoarthr. Cartil. 2017, 25, 727–736. [Google Scholar] [CrossRef] [PubMed]
  5. Haddaway, N.R.; McGuinness, L.A. PRISMA2020: R Package and ShinyApp for Producing PRISMA 2020 Compliant Flow Diagrams, Version 0.0.2; John Wiley & Sons Ltd.: Hoboken, NJ, USA, 2022. [Google Scholar]
  6. Slim, K.; Nini, E.; Forestier, D.; Kwiatkowski, F.; Panis, Y.; Chipponi, J. Methodological index for non-randomized studies (MINORS): Development and validation of a new instrument. ANZ J. Surg. 2003, 73, 712–716. [Google Scholar] [CrossRef] [PubMed]
  7. Morais, S.V.D.; Czeczko, N.G.; Malafaia, O.; Ribas Filho, J.M.; Garcia, J.B.S.; Miguel, M.T.; Zini, C.; Massignan, A.G. Osteoarthritis model induced by intra-articular monosodium iodoacetate in rats knee. Acta Cir. Bras. 2016, 31, 765–773. [Google Scholar] [CrossRef]
  8. Miyamoto, S.; Nakamura, J.; Ohtori, S.; Orita, S.; Omae, T.; Nakajima, T.; Suzuki, T.; Takahashi, K. Intra-articular injection of mono-iodoacetate induces osteoarthritis of the hip in rats. BMC Musculoskelet Disord. 2016, 17, 132. [Google Scholar] [CrossRef]
  9. Shuang, F.; Hou, S.-X.; Zhu, J.-L.; Liu, Y.; Zhou, Y.; Zhang, C.-L.; Tang, J.-G. Establishment of a rat model of lumbar facet joint osteoarthritis using intraarticular injection of urinary plasminogen activator. Sci. Rep. 2015, 5, 9828. [Google Scholar] [CrossRef]
  10. Shuang, F.; Zhu, J.; Song, K.; Hou, S.; Liu, Y.; Zhang, C.; Tang, J. Establishment of a Rat Model of Adjuvant-Induced Osteoarthritis of the Lumbar Facet Joint. Cell Biochem. Biophys. 2014, 70, 1545–1551. [Google Scholar] [CrossRef]
  11. Adães, S.; Mendonça, M.; Santos, T.N.; Castro-Lopes, J.M.; Ferreira-Gomes, J.; Neto, F.L. Intra-articular injection of collagenase in the knee of rats as an alternative model to study nociception associated with osteoarthritis. Thromb. Haemost. 2014, 16, R10. [Google Scholar] [CrossRef] [Green Version]
  12. Ferreira-Gomes, J.; Adães, S.; Sousa, R.M.; Mendonça, M.; Castro-Lopes, J.M. Dose-Dependent Expression of Neuronal Injury Markers during Experimental Osteoarthritis Induced by Monoiodoacetate in the Rat. Mol. Pain 2012, 8, 50. [Google Scholar] [CrossRef] [PubMed]
  13. Mohan, G.; Perilli, E.; Kuliwaba, J.S.; Humphries, J.M.; Parkinson, I.H.; Fazzalari, N.L. Application of in vivo micro-computed tomography in the temporal characterisation of subchondral bone architecture in a rat model of low-dose monosodium iodoacetate-induced osteoarthritis. Thromb. Haemost. 2011, 13, R210. [Google Scholar] [CrossRef] [PubMed]
  14. Ramme, A.J.; Lendhey, M.S.; Strauss, E.J.; Kennedy, O.D. A Biomechanical Study of Two Distinct Methods of Anterior Cruciate Ligament Rupture, and a Novel Surgical Reconstruction Technique, in a Small Animal Model of Posttraumatic Osteoarthritis. J. Knee Surg. 2017, 31, 43–49. [Google Scholar] [CrossRef] [PubMed]
  15. Rai, M.F.; Duan, X.; Quirk, J.D.; Holguin, N.; Schmidt, E.J.; Chinzei, N.; Silva, M.J.; Sandell, L.J. Post-Traumatic Osteoarthritis in Mice Following Mechanical Injury to the Synovial Joint. Sci. Rep. 2017, 7, 45223. [Google Scholar] [CrossRef]
  16. McCulloch, R.S.; Ashwell, M.S.; Maltecca, C.; O’Nan, A.T.; Mente, P.L. Progression of Gene Expression Changes following a Mechanical Injury to Articular Cartilage as a Model of Early Stage Osteoarthritis. Arthritis 2014, 2014, 371426. [Google Scholar] [CrossRef]
  17. Qin, J.; Chow, S.-H.; Guo, A.; Wong, W.-N.; Leung, K.-S.; Cheung, W.-H. Low magnitude high frequency vibration accelerated cartilage degeneration but improved epiphyseal bone formation in anterior cruciate ligament transect induced osteoarthritis rat model. Osteoarthr. Cartil. 2014, 22, 1061–1067. [Google Scholar] [CrossRef]
  18. Horisberger, M.; Fortuna, R.; Valderrabano, V.; Herzog, W. Long-term repetitive mechanical loading of the knee joint by in vivo muscle stimulation accelerates cartilage degeneration and increases chondrocyte death in a rabbit model. Clin. Biomech. 2013, 28, 536–543. [Google Scholar] [CrossRef]
  19. Roemhildt, M.; Beynnon, B.; Gauthier, A.; Gardner-Morse, M.; Ertem, F.; Badger, G. Chronic in vivo load alteration induces degenerative changes in the rat tibiofemoral joint. Osteoarthr. Cartil. 2013, 21, 346–357. [Google Scholar] [CrossRef]
  20. O’Brien, E.J.; Beveridge, J.E.; Huebner, K.D.; Heard, B.J.; Tapper, J.E.; Shrive, N.G.; Frank, C.B. Osteoarthritis develops in the operated joint of an ovine model following ACL reconstruction with immediate anatomic reattachment of the native ACL. J. Orthop. Res. 2012, 31, 35–43. [Google Scholar] [CrossRef]
  21. Moodie, J.; Stok, K.; Müller, R.; Vincent, T.; Shefelbine, S. Multimodal imaging demonstrates concomitant changes in bone and cartilage after destabilisation of the medial meniscus and increased joint laxity. Osteoarthr. Cartil. 2011, 19, 163–170. [Google Scholar] [CrossRef] [Green Version]
  22. Vaseenon, T.; Tochigi, Y.; Heiner, A.D.; Goetz, J.; Baer, T.E.; Fredericks, D.C.; Martin, J.; Rudert, M.J.; Hillis, S.; Brown, T.D.; et al. Organ-level histological and biomechanical responses from localized osteoarticular injury in the rabbit knee. J. Orthop. Res. 2010, 29, 340–346. [Google Scholar] [CrossRef] [PubMed]
  23. Christiansen, B.; Anderson, M.; Lee, C.; Williams, J.; Yik, J.; Haudenschild, D. Musculoskeletal changes following non-invasive knee injury using a novel mouse model of post-traumatic osteoarthritis. Osteoarthr. Cartil. 2012, 20, 773–782. [Google Scholar] [CrossRef]
  24. Killian, M.L.; Isaac, D.I.; Haut, R.C.; Déjardin, L.M.; Leetun, D.; Donahue, T.L.H. Traumatic anterior cruciate ligament tear and its implications on meniscal degradation: A preliminary novel lapine osteoarthritis model. J. Surg. Res. 2010, 164, 234–241. [Google Scholar] [CrossRef] [PubMed]
  25. Ko, F.C.; Dragomir, C.; Plumb, D.A.; Goldring, S.R.; Wright, T.M.; Goldring, M.B.; van der Meulen, M.C.H. In Vivo Cyclic Compression Causes Cartilage Degeneration and Subchondral Bone Changes in Mouse Tibiae. Arthritis Rheum. 2013, 65, 1569–1578. [Google Scholar] [CrossRef] [PubMed]
  26. Ko, F.C.; Dragomir, C.L.; Plumb, D.A.; Hsia, A.W.; Adebayo, O.O.; Goldring, S.R.; Wright, T.M.; Goldring, M.B.; van der Meulen, M.C. Progressive cell-mediated changes in articular cartilage and bone in mice are initiated by a single session of controlled cyclic compressive loading. J. Orthop. Res. 2016, 34, 1941–1949. [Google Scholar] [CrossRef]
  27. Lewis, J.; Hembree, W.; Furman, B.; Tippets, L.; Cattel, D.; Huebner, J.; Little, D.; DeFrate, L.; Kraus, V.; Guilak, F.; et al. Acute joint pathology and synovial inflammation is associated with increased intra-articular fracture severity in the mouse knee. Osteoarthr. Cartil. 2011, 19, 864–873. [Google Scholar] [CrossRef] [PubMed]
  28. Lynch, M.E.; Main, R.P.; Xu, Q.; Walsh, D.J.; Schaffler, M.B.; Wright, T.M.; van der Meulen, M.C.H. Cancellous bone adaptation to tibial compression is not sex dependent in growing mice. J. Appl. Physiol. 2010, 109, 685–691. [Google Scholar] [CrossRef] [PubMed]
  29. Lockwood, K.A.; Chu, B.T.; Anderson, M.J.; Haudenschild, D.R.; Christiansen, B.A. Comparison of loading rate-dependent injury modes in a murine model of post-traumatic osteoarthritis. J. Orthop. Res. 2014, 32, 79–88. [Google Scholar] [CrossRef]
  30. Onur, T.S.; Wu, R.; Chu, S.; Chang, W.; Kim, H.T.; Dang, A.B. Joint instability and cartilage compression in a mouse model of posttraumatic osteoarthritis. J. Orthop. Res. 2014, 32, 318–323. [Google Scholar] [CrossRef]
  31. Poulet, B.; Hamilton, R.W.; Shefelbine, S.; Pitsillides, A.A. Characterizing a novel and adjustable noninvasive murine joint loading model. Arthritis Rheum. 2011, 63, 137–147. [Google Scholar] [CrossRef]
  32. Poulet, B.; Westerhof, T.; Hamilton, R.; Shefelbine, S.; Pitsillides, A. Spontaneous osteoarthritis in Str/ort mice is unlikely due to greater vulnerability to mechanical trauma. Osteoarthr. Cartil. 2013, 21, 756–763. [Google Scholar] [CrossRef] [PubMed]
  33. Poulet, B.; de Souza, R.; Kent, A.; Saxon, L.; Barker, O.; Wilson, A.; Chang, Y.-M.; Cake, M.; Pitsillides, A. Intermittent applied mechanical loading induces subchondral bone thickening that may be intensified locally by contiguous articular cartilage lesions. Osteoarthr. Cartil. 2015, 23, 940–948. [Google Scholar] [CrossRef] [PubMed]
  34. Wu, P.; Holguin, N.; Silva, M.J.; Fu, M.; Liao, W.; Sandell, L.J. Early response of mouse joint tissue to noninvasive knee injury suggests treatment targets. Arthritis Rheumatol. 2014, 66, 1256–1265. [Google Scholar] [CrossRef] [PubMed]
  35. McNulty, M.; Loeser, R.; Davey, C.; Callahan, M.; Ferguson, C.; Carlson, C. Histopathology of naturally occurring and surgically induced osteoarthritis in mice. Osteoarthr. Cartil. 2012, 20, 949–956. [Google Scholar] [CrossRef] [PubMed]
  36. Allen, K.D.; A Mata, B.; Gabr, M.; Huebner, J.L.; Adams, S.B.; Kraus, V.B.; O Schmitt, D.; A Setton, L. Kinematic and dynamic gait compensations resulting from knee instability in a rat model of osteoarthritis. Thromb. Haemost. 2012, 14, R78. [Google Scholar] [CrossRef]
  37. Fischenich, K.; Pauly, H.; Button, K.; Fajardo, R.; DeCamp, C.; Haut, R.; Donahue, T.H. A study of acute and chronic tissue changes in surgical and traumatically-induced experimental models of knee joint injury using magnetic resonance imaging and micro-computed tomography. Osteoarthr. Cartil. 2016, 25, 561–569. [Google Scholar] [CrossRef] [PubMed]
  38. Maerz, T.; Newton, M.D.; Kurdziel, M.D.; Altman, P.; Anderson, K.; Matthew, H.; Baker, K.C. Articular cartilage degeneration following anterior cruciate ligament injury: A comparison of surgical transection and noninvasive rupture as preclinical models of post-traumatic osteoarthritis. Osteoarthr. Cartil. 2016, 24, 1918–1927. [Google Scholar] [CrossRef]
  39. Satkunananthan, P.; Anderson, M.; De Jesus, N.; Haudenschild, D.; Ripplinger, C.; Christiansen, B. In vivo fluorescence reflectance imaging of protease activity in a mouse model of post-traumatic osteoarthritis. Osteoarthr. Cartil. 2014, 22, 1461–1469. [Google Scholar] [CrossRef] [PubMed]
  40. van Dijk, P.A.; Murawski, C.D.; Hunt, K.J.; Andrews, C.L.; Longo, U.G.; McCollum, G.; Simpson, H.; Sofka, C.M.; Yoshimura, I.; Karlsson, J. Post-treatment Follow-up, Imaging, and Outcome Scores: Proceedings of the International Consensus Meeting on Cartilage Repair of the Ankle. Foot Ankle. Int. 2018, 39 (Suppl. 1), 68S–73S. [Google Scholar] [CrossRef]
  41. Longo, U.; Candela, V.; Berton, A.; De Salvatore, S.; Fioravanti, S.; Giannone, L.; Marchetti, A.; De Marinis, M.; Denaro, V. Biosensors for Detection of Biochemical Markers Relevant to Osteoarthritis. Biosensors 2021, 11, 31. [Google Scholar] [CrossRef]
  42. Lockwood, S.; Lopes, D.; McMahon, S.; Dickenson, A. Characterisation of peripheral and central components of the rat monoiodoacetate model of Osteoarthritis. Osteoarthr. Cartil. 2019, 27, 712–722. [Google Scholar] [CrossRef] [PubMed]
  43. Christiansen, B.; Guilak, F.; Lockwood, K.; Olson, S.; Pitsillides, A.; Sandell, L.; Silva, M.; van der Meulen, M.; Haudenschild, D. Non-invasive mouse models of post-traumatic osteoarthritis. Osteoarthr. Cartil. 2015, 23, 1627–1638. [Google Scholar] [CrossRef] [PubMed]
  44. Poulet, B. Non-invasive Loading Model of Murine Osteoarthritis. Curr. Rheumatol. Rep. 2016, 18, 40. [Google Scholar] [CrossRef] [PubMed]
  45. Turkiewicz, A.; Petersson, I.; Björk, J.; Hawker, G.; Dahlberg, L.; Lohmander, L.; Englund, M. Current and future impact of osteoarthritis on health care: A population-based study with projections to year 2032. Osteoarthr. Cartil. 2014, 22, 1826–1832. [Google Scholar] [CrossRef]
  46. Lorenz, J.; Grässel, S. Experimental Osteoarthritis Models in Mice. Mouse Genet. Methods Protoc. 2014, 1194, 401–419. [Google Scholar]
  47. Foxa, G.E.; Liu, Y.; Turner, L.M.; Robling, A.G.; Yang, T.; Williams, B.O. Generation and Characterization of Mouse Models for Skeletal Disease. Methods Mol. Biol. 2021, 2221, 165–191. [Google Scholar] [PubMed]
  48. Kuyinu, E.L.; Narayanan, G.; Nair, L.S.; Laurencin, C.T. Animal models of osteoarthritis: Classification, update, and measurement of outcomes. J. Orthop. Surg. Res. 2016, 11, 19. [Google Scholar] [CrossRef]
  49. Pitcher, T.; Sousa-Valente, J.; Malcangio, M. The Monoiodoacetate Model of Osteoarthritis Pain in the Mouse. J. Vis. Exp. 2016, 111, e53746. [Google Scholar]
  50. Sulaiman, S.Z.S.; Tan, W.M.; Radzi, R.; Shafie, I.N.F.; Ajat, M.; Mansor, R.; Mohamed, S.; Rahmad, N.; Ng, A.M.H.; Lau, S.F. Synovial fluid proteome profile of surgical versus chemical induced osteoarthritis in rabbits. Peerj 2022, 10, e12897. [Google Scholar] [CrossRef]
  51. McCoy, A.M. Animal Models of Osteoarthritis: Comparisons and Key Considerations. Vet. Pathol. 2015, 52, 803–818. [Google Scholar] [CrossRef]
  52. Serra, C.I.; Soler, C. Animal Models of Osteoarthritis in Small Mammals. Veter- Clin. North Am. Exot. Anim. Pr. 2019, 22, 211–221. [Google Scholar] [CrossRef] [PubMed]
  53. Ding, D.-F.; Xue, Y.; Zhang, J.-P.; Zhang, Z.-Q.; Li, W.-Y.; Cao, Y.-L.; Xu, J.-G. Similarities and differences between rat and mouse chondrocyte gene expression induced by IL-1β. J. Orthop. Surg. Res. 2022, 17, 70. [Google Scholar] [CrossRef] [PubMed]
  54. Longo, U.G.; Loppini, M.; Romeo, G.; Maffulli, N.; Denaro, V. Histological scoring systems for tissue-engineered, ex vivo and degenerative meniscus. Knee Surgery Sports Traumatol. Arthrosc. 2012, 21, 1569–1576. [Google Scholar] [CrossRef] [PubMed]
  55. Longo, U.; Forriol, F.; Candela, V.; Tecce, S.; De Salvatore, S.; Altonaga, J.; Wallace, A.; Denaro, V. Arthroscopic Tenotomy of the Long Head of the Biceps Tendon and Section of the Anterior Joint Capsule Produce Moderate Osteoarthritic Changes in an Experimental Sheep Model. Int. J. Environ. Res. Public Heal. 2021, 18, 7471. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Prisma flowchart.
Figure 1. Prisma flowchart.
Biology 12 00283 g001
Table 1. Study characteristics.
Table 1. Study characteristics.
AuthorLevel of
Evidence
YearAnimalSexN of SamplesInductionType of
Induction
Joint
Involved
Endpoint
Adães et al. [11]LOE 42014Wistar ratsM72 (n = 24)CTwo IAI of collagenase Knee 1, 2, 3, 4, 5, 6 wk post-inj.
Allen, Kyle D et al. [36]LOE 42012Lewis ratsU16 (n = 3)SMCL was transected Knee9-24-28 d
Arunakul et al. [3]LOE 42013NZW rabbits M26 (n = 5)SMMDKnee8 wk
Christiansen et al. [23]LOE 42012C57BL/6N miceM48MSingle overload cycle of tibial compressionKnee 1, 3, 7, 14, 28, or 56 d
Etienne J O O’Brien et al. [20]LOE 42012SC sheepF29 (n = 5, n = 7)STransection of ACL with consequent reconstructionKnee 0-4-20 wk
Farooq Rai et al. [15]LOE 42017LGXSM-6 and LGXSM-33 miceM88 (n = 44)MAxial tibial compressionKnee5, 9, 14, 28, 56 d
Fischenich et al. [37]LOE 42016FG rabbitsM and F33SACLT, ACLF and mACLTKnee4, 8, 12 wk
Geetha Mohan et al. [13]LOE 42011Wistar ratsM12CInjection of MIA Knee0-2-6-10 wk
Horisberger et al. [18]LOE 42013NZW rabbitsM24 (n = 8)MJoint loading by muscle stimulationKnee NS
Joana Ferreira-Gomes et al. [12]LOE 42012Wistar ratsM54 (n = 5)CInjection of MIA Knee 3-7-14-21-31 d after MIA injection
Killian et al. [24]LOE 42010FG rabbitsU11MTibiofemoral impaction resulting in ACL rupture or surgical ACL transectionKnee 12 wk for traumatically torn and after 12 wk in ACL transected animals
Ko et al. [25]LOE 42013C57BL/6 miceM63MCyclic compressionKnee 1, 2 and 6 wk
Ko et al. [26]LOE 42016C57Bl/6 miceM21MVCLKnee 0, 1 and 2 wk
Korostynski et al. [1]LOE 42018Wistar ratsM32 (n = 8)CIAI of MIAKnee 2, 14, 28 d
Lewis et al. [27]LOE 42011C57BL/6 miceM50 (n = 25)MLow and high energy fractures after loadingKnee0, 1, 3, 5 and 7 d post-fracture
Lynch et al. [28]LOE 42010C57BL/6 miceM and Fn = 14/sexMDynamic compression of the left tibiaKnee 2 wk of load evaluation
Lockwood et al. [29]LOE 42013C57BL/6N miceM80MTibial compressive overloadKnee 0 d, 10 d, 12 wk or 16 wk
Maerz et al. [38]LOE 42016Lewis ratsF36 (n = 6)SACL rupture or ACL transectionKnee 4 or 10 wk
McCulloch et al. [16]LOE 42014PigU36MShear loading modelKnee 3, 7, 14 d
McNulty et al. [35]LOE 4 2012C57BL/6 miceM11SDDM surgery, Sham surgeryKnee 2 months
Miyamoto et al. [8]LOE 42016SD ratsM60 (n = 30)CIAI of MIAHip 7, 14, 28, 42, 56 d
M.L. Roemhildt et al. [19]LOE 42012SD ratsM25 (n = 5)M VLD implementKnee 0-6-20 wk
Moodie, J.P. et al. [21]LOE 42011C57Bl6 miceM38SDMM surgeryKnee 0-4-8 wk
Morais SV et al. [7]LOE 42016Wistar ratsM48 (n = 24)CIAI of MIAKnee 1, 7, 14, 21, 28 d
Nomura et al. [4]LOE 42017C57BL/6J miceM24 (12 × 2; n = 4)MHindlimb unloading Knee2, 4, 8 wk
Onur et al. [30]LOE 4 2013FVB strain miceU21MAxial compression Knee 1, 8 wk
Poulet et al. [31]LOE 4 2011CBA miceU35MCyclic loading compressionKnee 2, 3 and 5 wk
Poulet et al. [32]LOE 4 2013Str/ort miceMn = 3,
n = 6,
n = 8
MCompression with servo-hydraulic materialsKnee 2 wk
Poulet et al. [33]LOE 4 2015CBA miceMn = 8, n = 8, n = 5MCompression with servo-hydraulic materials Knee 10, 13 wk
Qin et al. [17]LOE 42014SD ratsF88SACLTKnee 6, 12, 18 wk
Ramme et al. [14]LOE 42017SD rats F24MACL biomechanical rupture and surgical transectionKnee U
Satkunananthan et al. [39]LOE 4 2014C57BL/6 miceM and F54MACL rupture induced by tibial compressionKnee 1-7 d
Shuang et al. [10]LOE 42014SD ratsM60CIAI of complete Freund’s adjuvant [9]Spine3, 7, 14, 21 and 28 d
Shuang et al. [9]LOE 42015SD ratsMn = 88CIAI of uPA [9]Spine3, 7, 14, 28, 42, 56 d
Vaseenon, Tanawa et al. [22]LOE 42011NZW rabbitsU40SSham surgery Knee 0-8-16 wk
Wu et al. [34]LOE 42014C57BL/6 miceMn = 54MCompressive joint loadingKnee 5, 9, 14 d
Table 2. Type of induction and joint involved.
Table 2. Type of induction and joint involved.
AuthorOA ScoreConclusions
Adães et al. [11]Knee-bend and catwalkIt induces significant nociceptive alterations associated with OA changes
Allen, Kyle D et al. [36]OARSI score presentFirst description of functional losses after medial meniscus injury in the rat with reported changes in gait dynamics
Arunakul et al. [3]Mankin scoreResults support the MMD technique as being an effective surgical insult modality to replicate injurious cumulative abnormal cartilage loading
Christiansen et al. [23]OARSI scaleThis model is a significant improvement over other mouse models of PTOA, since it induces an injury that is translatable to humans, easy to implement and highly reproducible.
Etienne J O O’Brien et al. [20]Hellio Le Graverand protocolShows how a reconstruction model is not really helpful
Farooq Rai et al. [15] High mechanical loading instigated whole knee-joint changes leading to PTOA
Fischenich et al. [37] Dependency of the model on the location, type and progression of damage over time
Geetha Mohan et al. [13]OARSI score = 17Low-dose MIA-induced OA rat model mimics the human disease condition and clearly demonstrates disease progression in the tibial subchondral bone in a timely manner
Horisberger et al. [18]Mankin scoreMuscular loading of physiological magnitude but excessive intensity caused chondrocyte deaths and onset of early OA in rabbit knees
Joana Ferreira-Gomes et al. [12]Knee-bend and catwalkConcentrated more on the neuronal aspect rather than the causes of OA, suggests that axonal injury and a regeneration response may
be happening in this model of OA
Killian et al. [24] This study has implications for the future use of lapine models for osteoarthritis, as it incorporates traumatic loading as a more realistic progression of osteoarthritis compared with surgically transected models
Ko et al. [25]OARSI scaleNoninvasive loading model, permits dissection of temporal and topographic changes in cartilage and bone
Ko et al. [26]Osteomeasure histomorphometry system, OsteoMetrics, Decatur, GADemonstrate that a single session of noninvasive loading leads to the development of OA
Korostynski et al. [1] The progression of cartilage damage is driven by the complex but precise regulation of gene patterns
Lewis et al. [27]Mankin scoreThis study demonstrates that articular fracture is associated with a loss of chondrocyte viability and increased levels of systemic biomarkers
Lynch et al. [28] For all cancellous measures, the response to tibial compression did not differ between male and female mice
Lockwood et al. [29]OARSI scoreThese studies further characterize the non-invasive knee-injury mouse model
Maerz et al. [38]Mankin scoreAC degeneration is a time-, compartment- and injury-dependent cascade
McCulloch et al. [16] The chondrocytes in the shear specimens are attempting repairs but are unable to mount a successful effort
McNulty et al. [35]Mankin scoreProvided surgically induced models using a comprehensive histological grading scheme
Miyamoto et al. [8]Mankin scoreIntra-articular injection of MIA consistently causes progressive hip OA
M.L. Roemhildt et al. [19] Shows VLD is a better replicate of how OA really develops
Moodie et al. [21]Graded but without scaling gradeIncrease in AP laxity suggests that DMM surgery redistributes loading posteriorly on the medial plateau, resulting in bone and cartilage loss
Morais SV et al. [7] OA induced by intra-articular MIA is a good model to be used in related research
Nomura et al. [4] Thinning of articular cartilage induced by mechanical unloading may be mediated by metabolic changes in chondrocytes
Onur et al. [30]Pritzker grading scaleAxial compression without joint instability does not appear to be sufficient for inducing PTOA
Poulet et al. [31] This model offers opportunities to study the effects of various loading magnitudes and regimens on joint health and disease
Poulet et al. [32] Load application appears to accelerate OA
Poulet et al. [33] Concomitant AC-damage aggravates focal SCB-thickening induced by applied loading
Qin et al. [17]OARSI scoreLMHFV accelerated cartilage degeneration and deterioration of OA and promoted bone formation in affected distal femur epiphysis
Ramme et al. [14] ACL reconstruction and joint assessment will be useful in future joint-injury/PTOA studies in small-animal models
Satkunananthan et al. [39]OARSI scaleDescribes the dynamic protease profile following traumatic knee injury and establishes FRI as a useful analysis method
Shuang et al. [10]OARSI scoreInjection of complete Freund’s adjuvant caused local synovitis in early stage, and the inflammation was similar with osteoarthritis
Shuang et al. [9]OARSI scoreThis animal model is convenient and shows good resemblance of human facet joint OA pathology
Vaseenon, Tanawa et al. [22]Mankin scoreThe accompanying localized incongruity involved the onset of biomechanical abnormality consistent with causing cartilage degeneration
Wu et al. [34] Established a murine model of knee-joint trauma with different degrees of overloading in vivo
Table 3. MINORS.
Table 3. MINORS.
AuthorClearly Stated AimInclusion of Consecutive PatientsProspective Data CollectionCheckpoints Appropriate to Study AimUnbiased Assessment of Study EndpointFollow-Up PeriodAppropriate to <5% Lost to Follow-UpProspective Calculation of Study SizeTotal Score
Adães, Mendonça et al., 2014 [11]2222000210
Allen, Mata et al., 2012 [36]2222000210
Arunakul, Tochigi et al., 2013 [3]2222000210
Christiansen et al., 2012 [23]2222000210
Fischenich, Pauly et al., 2017 [37]2222020212
Mohan, Perilli et al., 2011 [13]2222000210
Horisberger, Fortuna et al., 2013 [18]222000028
Ferreira-Gomes, Adães et al., 2012 [12]2222000210
Killian et al., 2010 [24]2222000210
Ko et al., 2013 [25]2222000210
Ko et al., 2016 [26]2222000210
Korostynski, Malek et al., 2018 [1]2222000210
Lewis et al., 2011 [27]2222000210
Lynch et al., 2010 [28]2222000210
Lockwook et al., 2014 [29]2222000210
Maerz et al., 2016 [38]2222000210
McCulloch et al., 2014 [16]2222000210
McNulty et al., 2012 [35]2222000210
Miyamoto, Nakamura et al., 2016 [8]222100029
Moodie et al., 2011 [21]222000028
Morais, Czeczko et al., 2016 [7] 2222000210
Nomura, Sakitani et al., 2017 [4]2222000210
O’Brien et al., 2012 [20]2222000210
Onur et al., 2013 [30]2222000210
Poulet et al., 2011 [31]2222000210
Poulet et al. 2013 [32]2222000210
Poulet et al., 2015 [33]2222000210
Qin, Chow et al., 2014 [17]2222000210
Ramme, Lendhey et al., 2018 [14]222000028
Rai et al. 2017 [15]2220000210
Roemhildt, Beynnon et al., 2013 [19]2222000210
Satkunananthan et al., 2014 [39]2222000210
Shuang, Hou et al., 2015 [9]2222000210
Shuang, Zhu et al., 2014 [10]2222000210
Vaseenon, Tochigi et al., 2011 [22]2222000210
Wu et al., 2014 [34]2222000210
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Longo, U.G.; Papalia, R.; De Salvatore, S.; Picozzi, R.; Sarubbi, A.; Denaro, V. Induced Models of Osteoarthritis in Animal Models: A Systematic Review. Biology 2023, 12, 283. https://doi.org/10.3390/biology12020283

AMA Style

Longo UG, Papalia R, De Salvatore S, Picozzi R, Sarubbi A, Denaro V. Induced Models of Osteoarthritis in Animal Models: A Systematic Review. Biology. 2023; 12(2):283. https://doi.org/10.3390/biology12020283

Chicago/Turabian Style

Longo, Umile Giuseppe, Rocco Papalia, Sergio De Salvatore, Riccardo Picozzi, Antonio Sarubbi, and Vincenzo Denaro. 2023. "Induced Models of Osteoarthritis in Animal Models: A Systematic Review" Biology 12, no. 2: 283. https://doi.org/10.3390/biology12020283

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

Article Metrics

Back to TopTop