Next Article in Journal
Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation
Next Article in Special Issue
Efficient Generation of Pancreatic Progenitor Cells from Induced Pluripotent Stem Cells Derived from a Non-Invasive and Accessible Tissue Source—The Plucked Hair Follicle
Previous Article in Journal
WISP-1 Regulates Cardiac Fibrosis by Promoting Cardiac Fibroblasts’ Activation and Collagen Processing
Previous Article in Special Issue
Emerging Strategies in Mesenchymal Stem Cell-Based Cardiovascular Therapeutics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Mesenchymal Stem Cell-Based Therapies for Temporomandibular Joint Repair: A Systematic Review of Preclinical Studies

1
Faculty of Dentistry, National University of Singapore, 9 Lower Kent Ridge Road, Singapore 119085, Singapore
2
Center for Cleft Lip and Palate Treatment, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 33 Badachu Road, Shijingshan District, Beijing 100144, China
3
Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
4
Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, Singapore 117510, Singapore
5
Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore
6
Integrative Sciences and Engineering Program, NUS Graduate School, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
*
Author to whom correspondence should be addressed.
Cells 2024, 13(11), 990; https://doi.org/10.3390/cells13110990
Submission received: 1 April 2024 / Revised: 17 May 2024 / Accepted: 29 May 2024 / Published: 6 June 2024
(This article belongs to the Collection Stem Cells in Tissue Engineering and Regeneration)

Abstract

:
Temporomandibular disorders (TMDs) are a heterogeneous group of musculoskeletal and neuromuscular conditions involving the temporomandibular joint (TMJ), masticatory muscles, and associated structures. Mesenchymal stromal/stem cells (MSCs) have emerged as a promising therapy for TMJ repair. This systematic review aims to consolidate findings from the preclinical animal studies evaluating MSC-based therapies, including MSCs, their secretome, and extracellular vesicles (EVs), for the treatment of TMJ cartilage/osteochondral defects and osteoarthritis (OA). Following the PRISMA guidelines, PubMed, Embase, Scopus, and Cochrane Library databases were searched for relevant studies. A total of 23 studies involving 125 mice, 149 rats, 470 rabbits, and 74 goats were identified. Compliance with the ARRIVE guidelines was evaluated for quality assessment, while the SYRCLE risk of bias tool was used to assess the risk of bias for the studies. Generally, MSC-based therapies demonstrated efficacy in TMJ repair across animal models of TMJ defects and OA. In most studies, animals treated with MSCs, their derived secretome, or EVs displayed improved morphological, histological, molecular, and behavioral pain outcomes, coupled with positive effects on cellular proliferation, migration, and matrix synthesis, as well as immunomodulation. However, unclear risk in bias and incomplete reporting highlight the need for standardized outcome measurements and reporting in future investigations.

Graphical Abstract

1. Introduction

The temporomandibular joint (TMJ) is a ginglymoarthrodial (hinging and gliding) joint that connects the mandibular condyle with the temporal articular surface and is one of the most frequently used joints in the human body. Many pathological stimuli such as injury/trauma, joint overload, malocclusion, stress, and psychiatric illness can impact the joint structure and function, potentially leading to temporomandibular disorders (TMDs), a heterogeneous group of musculoskeletal and neuromuscular conditions involving the TMJ, masticatory muscles, and associated structures [1]. Among these conditions, osteoarthritis (OA) of the TMJ is an important subtype of TMDs and is characterized by synovial inflammation, disc degeneration, cartilage degradation, and subchondral bone erosion [2]. Patients with TMJ-OA suffer joint pain and dysfunction with a reduced quality of life. Based on clinical and magnetic resonance imaging (MRI) examination, the prevalence of TMJ-OA was found to be approximately 25% in the 20–49 age group, affecting more females than males [3].
There is presently no curative treatment for TMJ-OA, and current management is focused on conservative and non-invasive modalities, including physical therapies, occlusal splints/orthotics, non-steroidal anti-inflammatory drugs (NSAIDs), arthrocentesis, and intra-articular injections of hyaluronic acid (HA) [1,2]. Despite providing symptomatic relief to some extent, these treatments are unable to repair and restore the damaged cartilage and subchondral bone. Thus, restoring the integrity and function of the joint tissues, including cartilage and subchondral bone, is critical in halting or reversing the OA progression, where the final treatment option is prosthetic replacement [1]. However, repair and regeneration of the joint tissues, in particular the damaged cartilage, have been challenging as the cartilage generally has a poor healing capacity due primarily to it being a poorly vascularized, aneural, and alymphatic load-bearing tissue overlying the supporting subchondral bone.
In recent years, stem cells, particularly mesenchymal stem/stromal cells (MSCs), have emerged as a promising cell source for TMJ repair [4]. Residing as multipotent cells within several adult tissues, MSCs have been isolated from diverse tissues, including bone marrow, adipose tissue, skeletal muscle, synovium, dental tissues, peripheral blood, and others [5]. They can replicate as undifferentiated cells and have the potential to differentiate into multiple lineages of the mesenchymal tissues, including bone, cartilage, fat, and muscle [5].
MSCs have been reported to be safe and effective for articular cartilage repair of the knee, with the added benefits of lower cost and lesser donor site morbidity [6]. Other clinical studies have also reported the efficacy of intra-articular MSC injections to improve pain and functions for knee OA, as recently reviewed [7]. In the context of TMJ repair, intra-articular injections of autologous bone marrow nucleated cells [8] or fat-derived stem cells containing MSCs [9] reported improved clinical outcomes in patients with TMJ-OA or derangement. Despite their therapeutic potential, the clinical translation of such an approach using MSCs remains somewhat limited.
Although MSCs were thought to act therapeutically as stem cells via cellular differentiation and replacement, it is now accepted that these cells mediate tissue repair through their paracrine secretion and may be more appropriately termed “medicinal signaling cells” [10]. The secreted factors, collectively known as the secretome, are composed of soluble proteins, free nucleic acids and lipids, and extracellular vesicles (EVs). Being a heterogeneous class of lipid membrane vesicles released by cells into the extracellular space, EVs can be broadly classified into different classes, namely, exosomes, microvesicles/microparticles, and apoptotic bodies [11]. They are thought to function primarily as intercellular communication vehicles to transfer bioactive cargoes to elicit diverse biological responses in recipient cells, and with MSC-EVs, many of these biological responses culminate in a therapeutic outcome in injured or diseased cells [12].
The current systematic review aims to summarize the results of the existing animal studies that were conducted to evaluate the therapeutic effects of MSCs and their secreted factors, including EVs, in animal models of TMJ cartilage/osteochondral defects and OA.

2. Materials and Methods

2.1. Purpose

The purpose of this study is to systematically review and evaluate the efficacy of MSCs, their secretome, and their secreted factors, including EVs, for TMJ repair in animal studies.

2.2. Systematic Review

This systematic review follows the recommendations of the Cochrane Handbook for Systematic Reviews of Intervention [13] and was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines (Figure 1) [14,15].

2.3. Search Strategy

The literature search for eligible studies was conducted in PubMed, Embase, Scopus, and the Cochrane Library through 31 July 2023. The medical subject heading (MeSH) terms and keywords used were “temporomandibular joint” or “temporo-mandibular joint” or “temporo mandibular joint” or “craniomandibular” or “cranio-mandibular” or “cranio mandibular” or “TMJ” or “TMD” or “CMD” and “mesenchymal stem cell” or “mesenchymal stromal cell” or “marrow stem cell” or “marrow stromal cell” or “stromal cell” or “MSC” or “secretome” or “exosome” or “extracellular vesicle”.

2.4. Eligibility Criteria

The PICO framework was used to identify the components of evidence, which are as follows: Population: animals with TMJ condylar defects (cartilage, osteochondral) or TMJ-OA. Intervention: MSC-based therapies comprising MSCs, their secretome, or secreted factors, including EVs for TMJ repair. Comparison: healthy/naive controls, sham, and other groups (untreated, vehicle, scaffold). Outcome: the histological repair of TMJ and the associated structures as the primary outcome, and the morphological, molecular, and pain improvements as the secondary outcomes.

2.5. Study Selection

All search results were loaded into the EndNote 20 reference management software (Clarivate, Philadelphia, PA, USA), and duplicate records were removed. In the first stage, all abstracts and titles were retrieved and screened using the inclusion and exclusion criteria. After the initial screening, the full texts of the articles were further evaluated for eligibility. Three reviewers (Y.J., J.S., and W.D.) independently assessed all titles and abstracts for inclusion. Studies were included if they met the following criteria: in vivo animal studies that used MSCs, their secretome, or EVs for TMJ repair. Language was restricted to English. Studies were excluded if they were not related to TMJ or MSCs, not related to TMJ repair, or presented as case report, conference abstract, review, or purely in vitro study. As illustrated in Figure 1, a PRISMA flow diagram was used to document the study’s selection process [16].

2.6. Data Extraction

After identifying all the included studies, the following information was recorded: author; year of publication; animal species; age (weeks, months); sample size; gender (male or female); weight (grams or kilograms); animal model (cartilage/osteochondral defect, osteoarthritis); method of induction (surgically, mechanically, or chemically induced); source of MSCs, secretome, or EVs; isolation method; characterization, size distribution, and marker expression; experimental groups; study timepoints; concentration; volume; frequency; delivery; methods of analyses, including gross examination, imaging, histology, immunohistochemistry, molecular analysis, and pain measurement; as well as key in vitro and in vivo therapeutic outcomes. All qualitative and quantitative outcomes of the studies were recorded where available. Fate and biodistribution of MSCs, their secretome, or EVs, and their effects on cell proliferation, migration, and matrix synthesis, as well as immunomodulation, were also evaluated (Figure 2). Attempts were made to contact the corresponding author of the paper when certain details of the study were not reported.

2.7. Quality and Risk of Bias Assessments

The quality assessment for each included study was evaluated through their compliance with the ARRIVE (Animal Research: Reporting of In Vivo Experiments) guidelines [17]. Twenty items were evaluated for each study. The ratio for each item was calculated by dividing the category score (number of points assigned) by the total score (total points for the item). The quality of the item was assessed as “excellent” for values of the ratio between 0.8 and 1.0, “average” for values of the ratio between 0.5 and 0.79, and “low” for values of the ratio below 0.5. The risk of bias was evaluated using the Systematic Review Center for Laboratory Animal Experimentation (SYRCLE) risk of bias assessment tool for animal studies [18] that assessed selection bias, performance bias, detection bias, attrition bias, reporting bias, and other biases. The parameters assessed include sequence generation, baseline characteristics, allocation concealment, random housing, blinding of investigators, random outcome assessment, blinding of outcome assessor for the different analyses, incomplete outcome data, selective outcome reporting, and other sources of bias. All included studies were assessed as having high, unclear, or low risk of bias for the different parameters. This assessment was performed by first author (Y.J.) and independently checked by the second and third authors (J.S. and W.D.). Any discrepancy was resolved by discussion with the fourth author (K.Y.W.T.) and the senior author (W.S.T.).

3. Results

3.1. Search Process and Study Selection

The initial database search retrieved 2340 articles, which included 682 in PubMed, 1398 in Embase, 241 in Scopus, and 19 in Cochrane Library. A total of 749 duplicates were removed, and 1560 studies were excluded after screening the title and abstract. After reading the full texts of 31 articles that might meet the inclusion criteria, 6 articles on age-related degeneration, rheumatoid arthritis, osteoporosis, TMJ condylar resorption, growth, and replacement, as well as 2 articles of other languages, were excluded. A PRISMA flow chart showing the study-selection process is presented in Figure 1. A total of 23 studies that fulfilled the inclusion criteria were included in this systematic review [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. They were published between 2011 and 2022 and more frequently (>50% of papers) between 2020 and 2022 (Figure 3). These 23 included studies were divided according to the nature of the assessed TMDs to enable reliable and equivalent comparisons between studies. These included 11 studies that evaluated MSC-based therapies for the repair of cartilage/osteochondral defects [19,20,21,22,23,24,25,26,27,28,29] and another 12 studies that evaluated MSC-based therapies for TMJ-OA treatment [30,31,32,33,34,35,36,37,38,39,40,41]. The outcomes of the studies were largely evaluated qualitatively as there were insufficient quantitative data across the studies for pooling of data quantitatively.

3.2. Basic Characteristics of the Included Studies

The details of all studies are described in Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6. Table 1 details the animal species, age, sample size, gender, weight, animal model used, and method of induction. Table 2 details the source of MSCs, secretome, or EVs; isolation method; characterization; size distribution; and marker expression. Table 3 and Table 4 detail the comparison groups; study timepoints; concentration of MSCs, secretome, or EVs; volume; frequency; delivery; and methods of analysis, including gross examination, imaging, histology, immunohistochemistry, molecular analysis, and pain measurement. Table 5 and Table 6 summarize the key in vitro and in vivo outcomes reported in each paper.

3.3. Animal Models

To evaluate the efficacy of MSC-based therapies for the repair of cartilage/osteochondral defects, 2 main types of animal models were used. Namely, the condylar cartilage defect model was used in 1 study [20], and osteochondral defect models were used in 10 studies [19,21,22,23,24,25,26,27,28,29]. The defects were all surgically created, and the size of osteochondral defects ranged from a 3–5 mm diameter and 5 mm depth in goats [19,25] to a 2–3 mm diameter and 2–3 mm depth in rabbits [22,24,26,29] and 1 mm diameter in rats [23]. As for TMJ-OA, the efficacy of MSC-based therapies was most commonly evaluated in animal models that were chemically induced to develop OA via intra-articular injection of various agents such as bovine collagen II [32], mono-iodoacetate (MIA) [34,35,40], collagenase II [38,39], and combinations of Complete Freund’s adjuvant (CFA) and MIA [37] or interleukin (IL)-1β [41]. One study surgically induced OA via partial disc resection [30], whereas three studies mechanically induced TMJ-OA via unilateral anterior cross-bite (UAC) [31,33] or forced mouth opening [36]. Among the various animal species, rabbits were the most common animal species used in 10 studies [21,22,24,26,29,30,32,34,38,39] and accounted for 70.4% of animals used to create cartilage/osteochondral defects and 41.9% of animals used to induce experimental OA (Figure 4).

3.4. Source of MSCs, Their Derived Secretome, or EVs

A total of 10 studies used human MSCs or their derivatives [21,23,28,29,34,35,36,37,38,39], 3 studies used goat MSCs [19,20,25], 5 studies derived MSCs from rabbits [22,24,26,30,32], 2 studies used rat MSCs or their derivatives [40,41], and three studies derived MSCs from mice [27,31,33]. Among the various MSC sources that included bone marrow, adipose tissue, dental pulp, exfoliated deciduous teeth, umbilical cord, and embryonic stem cells, bone marrow stem/stromal cells (BMSCs) were the most commonly used cell source for the treatment of TMJ cartilage/osteochondral defects [19,20,22,24,25,26,28] and OA [30,31,32,33,38,40,41]. In terms of the use of cells, secretome, or EVs, eighteen studies tested MSCs as cell-based therapies [19,20,21,22,23,24,25,26,27,28,30,31,32,33,34,37,39,41], of which two studies reported the co-culture of MSCs and chondrocytes [20,26]. The other five studies investigated the use of MSC-derived secretome [36] and EVs [29,35,38,40]. Regardless of the origin or source of MSCs, all studies reported the therapeutic efficacy of MSCs, their derived secretome, or EVs in treating TMJ cartilage/osteochondral defects [19,20,21,22,23,24,25,26,27,28,29] and OA [30,31,32,33,34,35,36,37,38,39,40,41] in vivo. In comparing the different sources of MSCs, Kim et al. [34] compared human umbilical cord MSCs (UCMSCs) and BMSCs in their proliferation and chondrogenic differentiation capabilities in vitro. Of note, human UCMSCs had a comparable proliferation capacity as the human BMSCs and demonstrated a marked chondrogenic differentiation capacity, characterized by the presence of lacunae containing chondrocytes embedded in the matrix. Additionally, MSCs may also be co-cultured with additional cell sources such as chondrocytes [20,26]. By co-culturing with chondrocytes, a chondrogenic microenvironment was fostered to induce chondrogenic differentiation of MSCs without the additional factors [20].

3.5. Isolation and Characterization of MSCs, Their Derived Secretome, or EVs

Most of the studies used centrifugation with or without density gradients for the isolation of mononuclear cells, followed by the separation of MSCs via plastic adherence to tissue culture flasks [19,20,22,24,25,26,28,30,32,34,37,41]. One study, however, used a commercially available isolation system (Celution 800/CRS system) to isolate human adipose-derived regenerative cells (ADRCs) with varying proportions of stem cells, smooth muscle cells, endothelial cells, hematopoietic cells, nucleated cells, and other stromal cells from the human adipose tissue [21]. For the characterization of MSCs, flow cytometry and/or multi-lineage differentiation assays coupled with staining and gene expression analysis were frequently performed to analyze MSC marker expression and differentiation to osteogenic, chondrogenic, and/or adipogenic lineages [22,23,25,26,27,28,29,32,34,39]. Despite some variability among studies, the MSCs were generally positive for CD29, CD44, CD73, CD90, and CD105 and negative for CD31 and CD45 [22,23,28,29,32,34,39,40]. Unlike MSCs, secretome was characterized by liquid chromatography–tandem mass spectrometry (LC-MS/MS) analysis, whereas EVs were typically characterized by nanoparticle tracking analysis (NTA), transmission electron microscope (TEM), and/or Western blotting in the analysis of their size distribution, morphology, and presence of EV markers [29,35,38,40]. These EVs generally appeared as bi-lipid membrane structures under TEM, displayed a size distribution range of approximately 79–200 nm, and expressed endosomal proteins, ALIX and TSG101, and tetraspanin proteins, CD9, CD63, and CD81 [29,35,38,40].

3.6. Concentration

A wide range of concentrations of MSCs and their derived secretome or EVs were employed across different studies. The concentration of MSCs ranged from 105 cells/mL [28,31,33] to 7.5 × 107 cells/mL [19,20,24,25,26,27,28,31,33,34,37,39]. Of these, one study evaluated the therapeutic efficacy of human UCSCs in a rabbit model of TMJ-OA, comparing 5 × 105, 2.5 × 106, and 5 × 106 cells/mL [34]. All three cell concentrations at single injections showed anti-inflammatory effects and cartilage regenerative capacity, although the medium concentration of MSCs (2.5 × 106 cells/mL) was most effective in regenerating the articular cartilage and showed the highest gene expression levels of growth factors, including insulin growth factor (IGF)-1 and fibroblast growth factor (FGF)-2 [34].
EVs are commonly measured by their particle and/or protein concentration. Among the studies that investigated the use of EVs, one study reported particle concentration of EVs around 2–4 × 109 particles/mL [38], two studies reported protein concentration of about 2 mg/mL [35,40], and one study loaded 5 µg EVs per scaffold for implantation [29]. With the use of secretome, Ogasawara et al. [36] adjusted the protein concentration of the conditioned medium to 3 μg/mL with serum-free Dulbecco’s Modified Eagle Medium (DMEM).

3.7. Delivery

A variety of biomaterial scaffolds/carriers, ranging from platelet derivatives [22,23,28], collagen, gelatin, and hyaluronic acid (HA)-based hydrogels [24,26,27] to silk fibroin [29], poly(lactic-co-glycolic acid) (PLGA), and polycaprolactone (PCL)-based polymeric scaffolds [19,25] have been used to deliver MSCs or their EVs for the repair of cartilage/osteochondral defects. Biphasic scaffolds were also designed to simulate the specific structures and matrix compositions of the osteochondral tissues [25,26]. Of note, Wang and colleagues designed the biomimetic biphasic scaffold comprising self-crosslinking thiolated hyaluronic acid (HA-SH) and collagen I blend hydrogel and biphasic calcium phosphate (BCP) ceramics [26]. The blend hydrogel was layered and embedded with chondrocytes and BMSCs to simulate stratifications of the cartilage tissue and BCP ceramics to facilitate bone formation [26]. Interestingly, Putnova et al. flushed the defect with the human ADRC solution without using any scaffold [21].
For the treatment of TMJ-OA, most of the studies delivered MSCs, their derived secretome, or EVs by injections using various carriers, ranging from DMEM/10% fetal calf serum (FCS) [31,33] and serum-free DMEM [36] to phosphate-buffered saline (PBS)/saline [32,34,35,37,38,40], and hylartin/HA [30,39]. One study, however, used the gelatin methacryloyl (GelMA) hydrogel microspheres loaded with transforming growth factor (TGF)-β for coating with rat BMSCs [41]. In that study, the sustained release of TGF-β induced chondrogenic differentiation of BMSCs for enhanced TMJ repair in a rat model of TMJ-OA [41].
All the studies that evaluated scaffold/carrier delivery reported superior therapeutic outcomes in TMJ repair with scaffold/carrier delivery of MSCs, their derived secretome, or EVs against scaffolds alone [19,20,22,23,24,25,26,29,30,39,41]. Regarding the frequency of treatment, most of the studies applied a single scaffold implantation with MSCs [19,20,22,23,24,25,26,27,28] or MSC-EVs [29] or a single injection of MSCs [30,32,34,37,39,41] or MSC-EVs [38,40]. Other studies applied multiple injections [31,33,35,36]. None of the studies compared the efficacy of different biomaterial scaffolds or varying treatment frequencies for cartilage regeneration in their animal models. The optimal delivery method and frequency of treatment remain to be determined, and further studies for optimization might be needed.

3.8. Morphological Outcomes

Morphological analyses were mainly performed via gross/macroscopic assessment, micro-computed tomography (micro-CT), and X-ray examination. Eight studies performed gross examination and reported improved defect filling, macroscopic appearance, and tissue integration in cartilage/osteochondral defects [19,20,21,22,24,25,26,29], whereas one study noted improved integrity of the osteoarthritic condyles [38] following treatment with MSCs [19,20,21,22,24,25,26] or their EVs [29,38]. Micro-CT or X-ray examinations further observed improved subchondral bone reconstitution and restoration in cartilage/osteochondral defects [19,20,24,26,29] and in OA [30,31,34,35,36,37,38,41]. In these studies, a significantly higher bone mineral density (BMD), higher percentage of bone volume over total volume (BV/TV), increased trabecular thickness (Tb.Th), increased trabecular bone number (Tb.N), decreased trabecular separation (Tb.Sp), and/or reduced bone surface over bone volume ratio (BS/BV) were observed in animals treated with MSCs [19,20,24,26,30,31,37,41], secretome [36], or EVs [29,35,38].

3.9. Histological Outcomes

All studies performed histological analysis where specimens were stained with hematoxylin and eosin [20,21,22,23,25,26,27,28,29,31,32,33,34,35,36,37,38,40,41] for the observation of the general morphology; with safranin O [19,24,25,26,27,29,30,31,32,33,34,38,39], alcian blue [21,28,38], and toluidine blue [22,30,35,36,37] for glycosaminoglycan (GAG); and with Masson [26] or picrosirius red [39] for collagen deposition. Tartrate-resistant acid phosphatase (TRAP) [31,36] staining was also performed to detect bone-absorbing osteoclasts and terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling (TUNEL) [36] for apoptotic cells. Immunohistochemistry was frequently performed to detect cartilage matrix proteins, namely, collagen I, collagen II, and aggrecan [19,20,23,24,25,26,29,30,31,33,34,38]; proliferative cell nuclear antigen (PCNA) or antigen Kiel 67 (Ki-67) for proliferative cells [23,33,35,36,38]; and matrix metalloproteinases (MMPs) for matrix degradation [35,36,37,38] and pro-inflammatory cytokines, including interleukin (IL)-1β, interferon (IFN)-γ, and/or tumor necrosis factor (TNF)-α for inflammation [35,36,37].
In the TMJ cartilage/osteochondral defect models, animals treated with MSCs or their EVs generally showed improved defect filling with cartilaginous tissue, smooth surface regularity, and integration with adjacent condylar cartilage, as well as subchondral bone reconstitution [19,20,22,23,24,25,26,27,28,29]. Of these studies, six studies conducted histological scorings and reported significantly improved scores in groups treated with MSCs compared with untreated and/or scaffold groups [19,20,22,24,26,28].
In the TMJ-OA models, animals treated with MSCs, their derived secretome, or EVs had reduced degenerative changes and showed enhanced cartilage and subchondral bone restoration with increased matrix deposition and improved structural integrity [30,31,32,34,35,36,37,38,39,40,41]. These therapeutic effects were often associated with increased levels of PCNA for proliferation [35,36,38]; decreased levels of IL-1β, TNF-α, IFN-γ for inflammation [35,36,37]; MMP3 and MMP13 for matrix degradation [35,36,37,38]; TRAP for subchondral bone resorption [31,36]; as well as reduced levels of TUNEL, cleaved caspase (CCP)3, and death-associated protein (DAP)3 for apoptosis and anoikis [33,35,36]. Of these studies, six studies performed histological scorings to assess synovial inflammation and cartilage and subchondral bone destruction. They reported significantly improved scores in animals treated with MSCs, their derived secretome, or EVs, with reduced synovial inflammation, improved cartilage cellularity, and matrix deposition compared with the vehicle-treated and/or untreated animals [30,34,35,36,37,38].

3.10. Molecular Outcomes

Six studies performed gene expression analysis of the condylar cartilage specimens [22,30,31,34,35,40]. Using a rabbit TMJ osteochondral defect model, Cheng et al. reported significantly upregulated expression of cartilage-specific genes, including Sox9, Col II, and ACAN, in the repaired cartilage following treatment with a cell sheet of BMSCs, and this upregulation was further enhanced when the cells were pre-treated with hydrostatic pressure and delivered in a platelet-rich fibrin (PRF) scaffold [22]. In the TMJ-OA models, five studies reported a significantly upregulated expression of cartilage-specific genes (Sox9, Col2a1, and ACAN) and downregulated expression of genes associated with inflammation (IL-1β, IL-17, TNF-α, and NF-κB) and matrix degradation (MMP3, MMP13, and ADAMTS5) in animals treated with MSCs or their derived EVs compared with the vehicle-treated and/or untreated animals [30,31,34,35,40]. Notably, there was also increased expression of genes such as TGF-β1, IGF-1, and FGF-2, which are growth factors involved in chondrogenic differentiation and cartilage anabolism [34]. One study also reported downregulated levels of genes associated with fibrosis (α-SMA), apoptosis (BAX), and pain (Substance P, CGRP, NGF, p75NTR, and TrkA) with EV treatment compared to those of vehicle treatment [35].

3.11. Pain Behavioral Outcomes

In three studies, the head withdrawal threshold was measured to evaluate the nociceptive responses of the animals during the course of treatment. In these studies, regardless of the route of administration (intra-articular vs. intravenous), MSCs and their derived secretome, including EVs, were all found to be effective in relieving hyperalgesia of progressive TMJ-OA in mice and rats [35,36,37]. However, it is important to note that the secretome and EVs were applied via multiple injections, whereas the cells (i.e., DPSCs) were applied via a single intra-articular injection [35,36,37].

3.12. Fate and Biodistribution of MSCs, Their Derived Secretome, or EVs

Among the 18 studies that conducted implantation or intra-articular injections of MSCs, 6 studies performed in vivo tracing of MSCs [19,20,24,31,33,34]. In cartilage/osteochondral defects, transplanted MSCs were found in the newly formed tissues for up to 6 weeks [19] and 12 weeks post-surgery [20] in goats and up to 12 weeks in rabbits [24]. On the other hand, two studies reported a rapid decline in transplanted MSCs after 7 days and complete loss of the transplanted cells by 20 days following transplantation in a UAC-induced OA mouse model [31,33]. One study, however, reported the presence of transplanted MSCs up to 4 weeks post-injection in a rabbit TMJ-OA model induced by MIA [34]. Based on these observations, the persistence of MSCs following administration may vary in cartilage/osteochondral defects and in OA in the different animal models. In contrast to the MSC studies, none of the secretome or EV studies performed in vivo tracing of the secretome factors or EVs [29,35,36,38,40].

3.13. Cellular Proliferation, Migration, and Matrix Synthesis

Five studies reported increased proliferation and/or decreased apoptosis or anoikis following treatment with MSCs, their derived secretome, or EVs [23,33,35,36,38]. One study reported the upregulated expression of stromal-derived factor (SDF)-1 and regulated upon activation normal T-cell expression and secreted RANTES via osteoarthritic cartilage, and thus a higher capacity of attracting the migration of MSCs toward the degraded cartilage in OA [31]. These in vivo findings were supported by the in vitro studies where EV treatment was found to enhance the proliferation and migratory activity of BMSCs and chondrocytes in vitro [29,38]. Notably, EVs derived from inflammation-stimulated MSCs significantly enhanced the migration of BMSCs over their unstimulated counterpart [29].
Regardless of cartilage/osteochondral defects or OA, most of the studies reported increased matrix synthesis and deposition following treatment with MSCs, their derived secretome, or EVs [19,20,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39]. In TMJ-OA studies, concurrent attenuation of MMP3 [37,40], MMP13 [30,31,35,36,37,38,40], and a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) [35] were also observed. In support of the in vivo findings, three studies also reported significant inhibition of MMP3 and/or MMP13 expression in condylar chondrocytes and synoviocytes under inflammation following treatment with MSCs or their EVs [35,37,38]. These studies further identified protein kinase B (AKT), extracellular signal-regulated kinase (ERK), adenosine monophosphate-activated protein kinase (AMPK), signal transducer and activator of transcription 1 (STAT1), and Hippo-YAP (Yes-associated protein) signaling pathways in regulating the anabolic activities of MSCs and their EVs [35,37,38]. For instance, co-culture with DPSCs inhibited the expression of MMP3 and MMP13 in synoviocytes under inflammation, but these effects were abolished by the STAT1 inhibitor, implicating the role of the STAT1 signaling pathway in regulating the expression of MMP3 and MMP13 [37]. In another study, MSC-EVs were found to enhance GAG synthesis and inhibited nitric oxide and MMP13 production in IL-1β-treated chondrocytes through adenosine receptor activation of AKT, ERK, and AMPK phosphorylation [35].

3.14. Immunomodulation

Two studies examined the immune cell infiltration during TMJ repair [21,37]. Reduced infiltration of CD4+ T cells was observed in the synovial tissue and subchondral bone marrow of OA rats treated with human DPSCs compared to those treated with saline [37]. On the other hand, Putnova et al. observed significant immune cell infiltration and inflammatory reaction in both soft and hard TMJ tissues in rabbits treated with human ADRCs despite prior immunosuppression, and these were attributed to the heterogeneous cell populations present in ADRCs [21]. In other studies, pro-inflammatory mediators, including IL-1β, TNF-α, IFN-γ, IL-17, and inducible nitric oxide synthase (iNOS) [31,34,35,36,37,40] were downregulated, while anti-inflammatory cytokines such as TGF-β1 and IL-10 [34] were upregulated in animals treated with MSCs, their derived secretome, or EVs compared with the vehicle-treated and/or untreated animals. In vitro, EVs derived from inflammation-stimulated MSCs demonstrated augmented efficacy in suppressing inflammation and repolarizing the THP-1 cells toward the M2-like phenotype with the expression of M2 markers, including CD206, IL-10, and CCL22 [29]. These effects of MSC-EVs on the macrophage phenotype and polarization were attributed to the high expression level of miR-27b-3p that regulated macrophage polarization by targeting macrophage colony-stimulating factor (CSF)-1 [29].

3.15. Compliance with the ARRIVE Guidelines

The compliance with the ARRIVE guidelines for all the studies was evaluated and is detailed in Table 7 and Table 8. Based on the category score/total score ratio, three items (1, 2, and 20) were assessed as excellent, eleven items (3, 4, 5, 6, 7, 8, 10, 11, 13, 16, and 19) were marked as average, and the remaining six items (9, 12, 14, 15, 17, and 18) were considered low for studies on TMJ cartilage/osteochondral defects. Consequently, the overall quality score was 0.586. As for the studies on TMJ-OA, five items (1, 5, 6, 13, and 20) were assessed as excellent, nine items (2, 3, 7, 8, 10, 11, 14, 16, and 19) were marked average, and the remaining items (4, 9, 12, 15, 17, and 18) were considered low. Consequently, the overall quality score for the TMJ-OA studies was 0.609, considered average quality. Among all, five items (9, 12, 15, 17, and 18) were consistently marked low in both studies for cartilage/osteochondral defects and TMJ-OA.

3.16. Risk of Bias Assessment

The SYRCLE risk of bias tool was used to assess the risk of bias for all the included studies, and the assessment is summarized in Table 9. Most domains had unclear risks due to the lack of information (Table 9). None of the studies reported random sequence generation under selection bias and random housing and blinding under performance bias, and therefore were assigned unclear risk [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41]. Most studies reported some form of allocation concealment and baseline characteristics, including the species, strain, sex, or weight of the animals used, and were therefore assigned low risk. Only three studies were assigned a low risk for detection bias secondary to the random selection of animals for outcome assessment [22,24,30], while the rest of the studies were classified as unclear risk [19,20,21,23,25,26,27,28,29,31,32,33,34,35,36,37,38,39,40,41]. Only one study reported blinding of the assessor when performing gross assessment [29], whereas nine studies stated that the assessor was blinded when performing the histological analysis [19,22,29,30,34,35,37,38,39], and therefore were classified as low risk under detection bias. However, those studies that performed imaging, immunohistochemical, and/or molecular analyses did not report blinding for detection bias and therefore presented unclear risk. Notably, all studies were free of selective outcome reporting and presented no apparent issues that could result in a high risk of bias [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41].

4. Discussion

The principal finding of this systematic review is that MSC-based therapies are efficacious in the treatment of TMJ cartilage/osteochondral defects and OA. This therapeutic efficacy of MSC-based therapies in TMJ repair is demonstrated by cartilage regeneration and subchondral bone restoration, with overall improvements in the morphological, histological, molecular, and behavioral pain outcomes in the studies reviewed [19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41].
Of 23 studies reviewed, 18 studies tested MSCs as cell-based therapies [19,20,21,22,23,24,25,26,27,28,30,31,32,33,34,37,39,41], and the other five studies investigated the use of MSC-derived secretome [36] and EVs [29,35,38,40] as “cell-free” therapies. Of note, these five studies were published in recent years from 2019 to 2022 and signal a paradigm shift in the therapeutic mechanism of MSCs in tissue repair from one based on cellular differentiation and replacement to one based on secretion and paracrine signaling. Indeed, the role of paracrine secretion, particularly EVs, in mediating the wide-ranging therapeutic efficacy of MSCs has been increasingly reported [42], and it is now accepted that MSCs exert many if not most of their paracrine effects through the release of EVs [43], membrane vesicles that are secreted by all cell types.
Despite the therapeutic efficacy of MSC-based therapies, several factors/variables have been identified to influence the outcome of intervention with the use of MSCs, their secretome, or EVs for TMJ repair. These factors primarily include the cell source, dosage/concentration, and delivery. Several MSC sources were demonstrated to be efficacious in TMJ repair in both cartilage/osteochondral defects and OA, but the ideal cell source is yet to be identified. Kim et al. demonstrated comparable proliferation and differentiation capabilities of human UCMSCs and BMSCs in vitro; however, this remains to be verified in an animal model in vivo [34]. A head-to-head comparison of relevant sources of MSCs, their derived secretome and/or EVs would be required in future studies to determine the most effective source from a TMJ repair perspective. Similarly, a wide range of concentrations of MSCs and their derived EVs was reported. One study evaluated a single intra-articular injection of human UCSCs at varying concentrations in a rabbit model of TMJ-OA and identified the medium concentration of MSCs (2.5 × 106 cells/mL) as the optimal dose/concentration for regenerating the articular cartilage [34]. However, this dose/concentration remains to be validated using other sources of MSCs.
The frequency of treatment is another factor/variable that should be considered and ideally tested. While some studies recorded the long-term survival of the transplanted BMSCs up to 12 weeks in the osteochondral defects of rabbit and goat [20,24], other studies reported a rapid decline in transplanted cells after 1 week in OA in mice and required multiple weekly injections to constantly supply the BMSCs [31,33]. Based on these observations, the persistence of MSCs following implantation or intra-articular injection may vary in cartilage/osteochondral defects and in OA in different animal models. In contrast, none of the secretome or EV studies performed in vivo tracing of the secretome factors or EVs. As such, it would be necessary to determine in future studies the fate and bio-distribution of MSCs, their secretome, and EVs following injection into the joint space to determine their site of action and persistence and to optimize the number of injections for optimal treatment.
Other factors, such as the culture condition and the use of a scaffold, would also influence the treatment outcome in TMJ repair. The culture condition affects the potency of MSCs, their derived secretome, and EVs. For instance, in vitro chondrogenic pre-differentiation or hydrostatic pressure stimulation substantially enhanced the chondrogenic potential of MSCs and, henceforth, their therapeutic efficacy in TMJ repair [22,30]. The overexpression of NEL-like protein-1 (NELL-1) or hypoxia-inducible factor (HIF)-1α also reportedly enhanced the efficacy of MSCs in osteochondral repair. In those studies, animals treated with NELL-1 or HIF-1α overexpressing BMSCs displayed enhanced fibrocartilage regeneration and subchondral bone restoration with increased matrix deposition and improved structural organization compared to those treated with native MSCs [19,24]. Notably, HIF-1α overexpression was found to enhance the survival of transplanted BMSCs, contributing to improved cartilage repair [24]. Alternatively, EVs derived from MSCs stimulated under inflammation in the presence of TNF-α and IFN-γ demonstrated augmented efficacy for osteochondral repair compared to those derived from unstimulated counterparts [29]. Other studies also reported the delivery of MSCs and their derived EVs in different scaffolds/carriers and demonstrated synergistic enhancements in cartilage and subchondral bone repair [19,20,22,23,24,25,26,27,28,29,30,39,41]. For example, Cheng et al. fabricated a BMSC sheet in the PRF scaffold and showed that the BMSC/PRF construct outperformed the BMSC sheet or PRF alone in cartilage regeneration by enhancing matrix synthesis and promoting the mechanical properties and integration of the neocartilage [22]. These studies offer possible strategies to engineer and/or coax the MSCs and their derived EVs to possess the desired therapeutic properties for enhanced TMJ repair. However, it is also important to note that these findings of a particular modification or scaffold being superior to another are all derived from single studies [19,20,22,23,24,25,26,27,28,29,30,39,41], and the conclusions remain to be verified by more studies.
Of the 23 studies reviewed, nine studies applied human MSCs, their derived secretome, or EVs in immunocompetent animals, and no adverse responses were reported [23,28,29,34,35,36,37,38,39]. These findings are consistent with the well-established immune privilege property of MSCs. The cells and their derived secretome, including EVs, did not elicit any adverse responses from the animals, including mice, rats, and rabbits tested in these studies [23,28,29,34,35,36,37,38,39]. These findings suggest the use of MSCs, their derived secretome, and EVs as potential allogeneic therapies.
Although the therapeutic mechanisms underlying MSC-based therapies in TMJ repair remain to be elucidated, it could be summarized from this systematic review of 23 studies that MSCs, including their secretome and EVs, likely exert a multifaceted activity in TMJ repair by enhancing cellular proliferation, migration, and matrix synthesis and modulating the immune reactivity (Figure 5). However, in the 23 studies reviewed, only 3 studies delved into the underlying signaling pathways [35,37,38], and only 2 studies investigated immune cell infiltration during TMJ repair [21,37]. These clearly reflect our nascent understanding of the therapeutic mechanisms, particularly the immunomodulatory mechanisms, underlying the effects of MSCs, their derived secretome, or EVs in TMJ repair, which warrants further investigation. The identification of key factors/variables influencing the therapeutic outcomes of MSCs, their secretome, and EVs in TMJ repair and improved understanding of the underlying mechanisms, as summarized in this timely review, will undoubtedly inform the planning and design of future studies.
This systematic review has several limitations. Although most of the studies included in this review appeared to be methodologically sound, the lack of proper reporting rendered many of these studies as having unclear risk in several domains, such as random sequence generation; random housing during the experiment; the blinding of caregivers and investigators; and the blinding of outcome assessors in gross, imaging, and immunohistochemical and molecular analyses, according to the SYRCLE risk of bias assessment tool. This highlighted the importance of detailed reporting by adherence to the ARRIVE guidelines so as to improve the credibility and reliability of studies as having high-quality evidence. Several factors/variables, including the cell source, dosage/concentration, scaffold, and delivery route, were also identified to influence the therapeutic outcome of MSCs, their secretome, or EVs in TMJ repair. However, none of these studies systematically evaluated these variables to determine the optimal cell source, concentration, scaffold, delivery route, and/or frequency of treatment, which would likely require optimization for a specific TMJ cartilage/osteochondral defect or OA condition. The deficiency in quantitative data and heterogeneity in the assessment and reporting of outcomes in many studies also precluded a more rigorous meta-analysis of the studies in this review. Therefore, more efforts to standardize the reporting of methodology and outcomes are required in future studies. This would then allow future studies to have better uniformity and validity, with the possible pooling of results through meta-analysis for more robust data analysis and conclusion.

5. Conclusions

In this review, we systematically assessed the existing preclinical animal studies and broadly demonstrated the effectiveness of MSC-based therapies for TMJ repair in animal models of TMJ cartilage/osteochondral defects and OA. In general, MSC-based therapies were found to exert positive effects on cell proliferation, migration, matrix synthesis, and immunomodulation, leading to improvements in morphological, histological, molecular, and behavioral pain outcomes.

Author Contributions

Conceptualization, W.S.T.; Data curation, W.S.T.; Formal analysis, Y.J., J.S. and W.D.; Funding acquisition, W.S.T.; Investigation, Y.J., J.S. and W.D.; Methodology, W.S.T.; Resources, W.S.T.; Supervision, W.S.T.; Visualization, Y.J. and J.S.; Writing—original draft, Y.J. and W.S.T.; Writing—review and editing, K.Y.W.T. and W.S.T. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Education (MOE) Tier 2 grant (Project ID: MOE-T2EP30122-0008) and MOE Tier 1 grant (Project ID: NUHSRO/2022/091/T1/Seed-Sep/04).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are available upon reasonable request to the corresponding authors.

Conflicts of Interest

All authors declare no potential conflicts of interest with respect to the authorship and/or publication of this article.

References

  1. Tanaka, E.; Detamore, M.S.; Mercuri, L.G. Degenerative disorders of the temporomandibular joint: Etiology, diagnosis, and treatment. J. Dent. Res. 2008, 87, 296–307. [Google Scholar] [CrossRef] [PubMed]
  2. Wang, X.D.; Zhang, J.N.; Gan, Y.H.; Zhou, Y.H. Current understanding of pathogenesis and treatment of TMJ osteoarthritis. J. Dent. Res. 2015, 94, 666–673. [Google Scholar] [CrossRef] [PubMed]
  3. Bernhardt, O.; Biffar, R.; Kocher, T.; Meyer, G. Prevalence and clinical signs of degenerative temporomandibular joint changes validated by magnetic resonance imaging in a non-patient group. Ann. Anat. 2007, 189, 342–346. [Google Scholar] [CrossRef]
  4. Zhang, S.; Yap, A.U.; Toh, W.S. Stem Cells for Temporomandibular Joint Repair and Regeneration. Stem Cell Rev. Rep. 2015, 11, 728–742. [Google Scholar] [CrossRef] [PubMed]
  5. Huang, G.T.; Gronthos, S.; Shi, S. Mesenchymal stem cells derived from dental tissues vs. those from other sources: Their biology and role in regenerative medicine. J. Dent. Res. 2009, 88, 792–806. [Google Scholar] [CrossRef]
  6. Teo, A.Q.A.; Wong, K.L.; Shen, L.; Lim, J.Y.; Toh, W.S.; Lee, E.H.; Hui, J.H.P. Equivalent 10-Year Outcomes After Implantation of Autologous Bone Marrow-Derived Mesenchymal Stem Cells Versus Autologous Chondrocyte Implantation for Chondral Defects of the Knee. Am. J. Sports Med. 2019, 47, 2881–2887. [Google Scholar] [CrossRef] [PubMed]
  7. Tan, S.H.S.; Kwan, Y.T.; Neo, W.J.; Chong, J.Y.; Kuek, T.Y.J.; See, J.Z.F.; Wong, K.L.; Toh, W.S.; Hui, J.H.P. Intra-articular Injections of Mesenchymal Stem Cells Without Adjuvant Therapies for Knee Osteoarthritis: A Systematic Review and Meta-analysis. Am. J. Sports Med. 2021, 49, 3113–3124. [Google Scholar] [CrossRef] [PubMed]
  8. De Riu, G.; Vaira, L.A.; Carta, E.; Meloni, S.M.; Sembronio, S.; Robiony, M. Bone marrow nucleated cell concentrate autograft in temporomandibular joint degenerative disorders: 1-year results of a randomized clinical trial. J. Craniomaxillofac. Surg. 2019, 47, 1728–1738. [Google Scholar] [CrossRef] [PubMed]
  9. Carboni, A.; Amodeo, G.; Perugini, M.; Arangio, P.; Orsini, R.; Scopelliti, D. Temporomandibular Disorders Clinical and Anatomical Outcomes After Fat-Derived Stem Cells Injection. J. Craniofac. Surg. 2019, 30, 793–797. [Google Scholar] [CrossRef] [PubMed]
  10. Caplan, A.I. Mesenchymal Stem Cells: Time to Change the Name! Stem Cells Transl. Med. 2017, 6, 1445–1451. [Google Scholar] [CrossRef] [PubMed]
  11. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): A position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef] [PubMed]
  12. Toh, W.S.; Lai, R.C.; Hui, J.H.P.; Lim, S.K. MSC exosome as a cell-free MSC therapy for cartilage regeneration: Implications for osteoarthritis treatment. Semin Cell Dev. Biol. 2017, 67, 56–64. [Google Scholar] [CrossRef] [PubMed]
  13. Higgins, J.P.; Green, S. Cochrane Handbook for Systematic Reviews of Interventions, 2nd ed.; John Wiley & Sons: Chichester, UK, 2019. [Google Scholar]
  14. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Rev. Esp. Cardiol. 2021, 74, 790–799. [Google Scholar] [CrossRef]
  15. Haddaway, N.R.; Page, M.J.; Pritchard, C.C.; McGuinness, L.A. PRISMA2020: An R package and Shiny app for producing PRISMA 2020-compliant flow diagrams, with interactivity for optimised digital transparency and Open Synthesis. Campbell Syst. Rev. 2022, 18, e1230. [Google Scholar] [CrossRef] [PubMed]
  16. Moher, D.; Shamseer, L.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; Group, P.-P. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 2015, 4, 1. [Google Scholar] [CrossRef] [PubMed]
  17. Kilkenny, C.; Browne, W.J.; Cuthill, I.C.; Emerson, M.; Altman, D.G. Improving bioscience research reporting: The ARRIVE guidelines for reporting animal research. PLoS Biol. 2010, 8, e1000412. [Google Scholar] [CrossRef] [PubMed]
  18. Hooijmans, C.R.; Rovers, M.M.; de Vries, R.B.; Leenaars, M.; Ritskes-Hoitinga, M.; Langendam, M.W. SYRCLE’s risk of bias tool for animal studies. BMC Med. Res. Methodol. 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
  19. Zhu, S.; Zhang, B.; Man, C.; Ma, Y.; Hu, J. NEL-like molecule-1-modified bone marrow mesenchymal stem cells/poly lactic-co-glycolic acid composite improves repair of large osteochondral defects in mandibular condyle. Osteoarthr. Cartil. 2011, 19, 743–750. [Google Scholar] [CrossRef]
  20. Sun, H.; Huang, Y.; Zhang, L.; Li, B.; Wang, X. Co-culture of bone marrow stromal cells and chondrocytes in vivo for the repair of the goat condylar cartilage defects. Exp. Ther. Med. 2018, 16, 2969–2977. [Google Scholar] [CrossRef]
  21. Putnová, B.; Hurník, P.; Jekl, V.; Žiak, D.; Machoň, V.; Škorič, M.; Stránský, J.; Štembírek, J. Effect of human adipose-derived regenerative cells on temporomandibular joint healing in immunodeficient rabbits. Acta Vet. Brno 2019, 88, 49–56. [Google Scholar] [CrossRef]
  22. Cheng, B.; Tu, T.; Shi, X.; Liu, Y.; Zhao, Y.; Zhao, Y.; Li, Y.; Chen, H.; Chen, Y.; Zhang, M. A novel construct with biomechanical flexibility for articular cartilage regeneration. Stem Cell Res. Ther. 2019, 10, 298. [Google Scholar] [CrossRef] [PubMed]
  23. Sumarta, N.P.M.; Kamadjaja, D.B.; Hendrijantini, N.; Danudiningrat, C.P.; Rantam, F.A. Human umbilical cord mesenchymal stem cells over platelet rich fibrin scaffold for mandibular cartilage defects regenerative medicine. Pesqui. Bras. Odontopediatria Clínica Integr. 2021, 21, e0034. [Google Scholar] [CrossRef]
  24. Cheng, M.S.; Yi, X.; Zhou, Q. Overexpression of HIF-1alpha in Bone Marrow Mesenchymal Stem Cells Promote the Repair of Mandibular Condylar Osteochondral Defect in a Rabbit Model. J. Oral Maxillofac. Surg. 2021, 79, 345.e1–345.e15. [Google Scholar] [CrossRef] [PubMed]
  25. Yu, X.; Hu, Y.; Zou, L.; Yan, S.; Zhu, H.; Zhang, K.; Liu, W.; He, D.; Yin, J. A bilayered scaffold with segregated hydrophilicity-hydrophobicity enables reconstruction of goat hierarchical temporomandibular joint condyle cartilage. Acta Biomater. 2021, 121, 288–302. [Google Scholar] [CrossRef] [PubMed]
  26. Wang, H.; Xu, Y.; Wang, P.; Ma, J.; Wang, P.; Han, X.; Fan, Y.; Bai, D.; Sun, Y.; Zhang, X. Cell-mediated injectable blend hydrogel-BCP ceramic scaffold for in situ condylar osteochondral repair. Acta Biomater. 2021, 123, 364–378. [Google Scholar] [CrossRef] [PubMed]
  27. Guastaldi, F.; Hakim, M.; Liapaki, A.; Lowe, B.; Faquin, W.; Thamm, J. Are stem cells useful in the regeneration and repair of cartilage defects in the TMJ condyle? An In Vivo Study. J. Dent. Oral Disord. 2021, 7, 1159. [Google Scholar]
  28. Gomez, M.; Wittig, O.; Diaz-Solano, D.; Cardier, J.E. Mesenchymal Stromal Cell Transplantation Induces Regeneration of Large and Full-Thickness Cartilage Defect of the Temporomandibular Joint. Cartilage 2021, 13, 1814S–1821S. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, Y.; Zhang, Z.; Wang, B.; Dong, Y.; Zhao, C.; Zhao, Y.; Zhang, L.; Liu, X.; Guo, J.; Chen, Y.; et al. Inflammation-Stimulated MSC-Derived Small Extracellular Vesicle miR-27b-3p Regulates Macrophages by Targeting CSF-1 to Promote Temporomandibular Joint Condylar Regeneration. Small 2022, 18, e2107354. [Google Scholar] [CrossRef]
  30. Chen, K.; Man, C.; Zhang, B.; Hu, J.; Zhu, S.S. Effect of in vitro chondrogenic differentiation of autologous mesenchymal stem cells on cartilage and subchondral cancellous bone repair in osteoarthritis of temporomandibular joint. Int. J. Oral Maxillofac. Surg. 2013, 42, 240–248. [Google Scholar] [CrossRef] [PubMed]
  31. Lu, L.; Zhang, X.; Zhang, M.; Zhang, H.; Liao, L.; Yang, T.; Zhang, J.; Xian, L.; Chen, D.; Wang, M. RANTES and SDF-1 Are Keys in Cell-based Therapy of TMJ Osteoarthritis. J. Dent. Res. 2015, 94, 1601–1609. [Google Scholar] [CrossRef] [PubMed]
  32. Zaki, A.A.; Zaghloul, M.; Helal, M.E.; Mansour, N.A.; Grawish, M.E. Impact of Autologous Bone Marrow-Derived Stem Cells on Degenerative Changes of Articulating Surfaces Associated with the Arthritic Temporomandibular Joint: An Experimental Study in Rabbits. J. Oral Maxillofac. Surg. 2017, 75, 2529–2539. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, M.; Yang, H.; Lu, L.; Wan, X.; Zhang, J.; Zhang, H.; Liu, X.; Huang, X.; Xiao, G.; Wang, M. Matrix replenishing by BMSCs is beneficial for osteoarthritic temporomandibular joint cartilage. Osteoarthr. Cartil. 2017, 25, 1551–1562. [Google Scholar] [CrossRef] [PubMed]
  34. Kim, H.; Yang, G.; Park, J.; Choi, J.; Kang, E.; Lee, B.K. Therapeutic effect of mesenchymal stem cells derived from human umbilical cord in rabbit temporomandibular joint model of osteoarthritis. Sci. Rep. 2019, 9, 13854. [Google Scholar] [CrossRef] [PubMed]
  35. Zhang, S.; Teo, K.Y.W.; Chuah, S.J.; Lai, R.C.; Lim, S.K.; Toh, W.S. MSC exosomes alleviate temporomandibular joint osteoarthritis by attenuating inflammation and restoring matrix homeostasis. Biomaterials 2019, 200, 35–47. [Google Scholar] [CrossRef] [PubMed]
  36. Ogasawara, N.; Kano, F.; Hashimoto, N.; Mori, H.; Liu, Y.; Xia, L.; Sakamaki, T.; Hibi, H.; Iwamoto, T.; Tanaka, E.; et al. Factors secreted from dental pulp stem cells show multifaceted benefits for treating experimental temporomandibular joint osteoarthritis. Osteoarthr. Cartil. 2020, 28, 831–841. [Google Scholar] [CrossRef] [PubMed]
  37. Cui, S.J.; Zhang, T.; Fu, Y.; Liu, Y.; Gan, Y.H.; Zhou, Y.H.; Yang, R.L.; Wang, X.D. DPSCs Attenuate Experimental Progressive TMJ Arthritis by Inhibiting the STAT1 Pathway. J. Dent. Res. 2020, 99, 446–455. [Google Scholar] [CrossRef]
  38. Wang, Y.; Zhao, M.; Li, W.; Yang, Y.; Zhang, Z.; Ma, R.; Wu, M. BMSC-Derived Small Extracellular Vesicles Induce Cartilage Reconstruction of Temporomandibular Joint Osteoarthritis via Autotaxin-YAP Signaling Axis. Front. Cell Dev. Biol. 2021, 9, 656153. [Google Scholar] [CrossRef] [PubMed]
  39. Kohnke, R.; Ahlers, M.O.; Birkelbach, M.A.; Ewald, F.; Krueger, M.; Fiedler, I.; Busse, B.; Heiland, M.; Vollkommer, T.; Gosau, M.; et al. Temporomandibular Joint Osteoarthritis: Regenerative Treatment by a Stem Cell Containing Advanced Therapy Medicinal Product (ATMP)—An In Vivo Animal Trial. Int. J. Mol. Sci. 2021, 22, 443. [Google Scholar] [CrossRef] [PubMed]
  40. AbuBakr, N.; Fares, A.E.; Mostafa, A.; Farag, D.B.E. Mesenchymal stem cells-derived microvesicles versus platelet-rich plasma in the treatment of monoiodoacetate-induced temporomandibular joint osteoarthritis in Albino rats. Heliyon 2022, 8, e10857. [Google Scholar] [CrossRef] [PubMed]
  41. Yang, Y.; Huang, C.; Zheng, H.; Meng, Z.; Heng, B.C.; Zhou, T.; Jiang, S.; Wei, Y. Superwettable and injectable GelMA-MSC microspheres promote cartilage repair in temporomandibular joints. Front. Bioeng. Biotechnol. 2022, 10, 1026911. [Google Scholar] [CrossRef] [PubMed]
  42. Teo, K.Y.W.; Tan, R.; Wong, K.L.; Hey, D.H.W.; Hui, J.H.P.; Toh, W.S. Small extracellular vesicles from mesenchymal stromal cells: The next therapeutic paradigm for musculoskeletal disorders. Cytotherapy 2023, 25, 837–846. [Google Scholar] [CrossRef] [PubMed]
  43. Witwer, K.W.; Van Balkom, B.W.M.; Bruno, S.; Choo, A.; Dominici, M.; Gimona, M.; Hill, A.F.; De Kleijn, D.; Koh, M.; Lai, R.C.; et al. Defining mesenchymal stromal cell (MSC)-derived small extracellular vesicles for therapeutic applications. J. Extracell. Vesicles 2019, 8, 1609206. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Prisma flow diagram. Search strategy and selection process of the included studies.
Figure 1. Prisma flow diagram. Search strategy and selection process of the included studies.
Cells 13 00990 g001
Figure 2. Key factors and outcomes identified in preclinical studies of MSC-based therapies for TMJ repair. BCP, biphasic calcium phosphate; EVs, extracellular vesicles; MSCs, mesenchymal stem/stromal cells; PCL, polycaprolactone; PEG, polyethylene glycol; PLGA, poly(L-glutamic acid); PRF, platelet-rich fibrin; PRP, platelet-rich plasma; SF, silk fibroin; TMJ-OA, temporomandibular joint osteoarthritis.
Figure 2. Key factors and outcomes identified in preclinical studies of MSC-based therapies for TMJ repair. BCP, biphasic calcium phosphate; EVs, extracellular vesicles; MSCs, mesenchymal stem/stromal cells; PCL, polycaprolactone; PEG, polyethylene glycol; PLGA, poly(L-glutamic acid); PRF, platelet-rich fibrin; PRP, platelet-rich plasma; SF, silk fibroin; TMJ-OA, temporomandibular joint osteoarthritis.
Cells 13 00990 g002
Figure 3. Bar chart showing the number of published studies included in the systematic review sorted by the year of publication.
Figure 3. Bar chart showing the number of published studies included in the systematic review sorted by the year of publication.
Cells 13 00990 g003
Figure 4. Animal species used for models of (A) TMJ cartilage/osteochondral defects and (B) OA.
Figure 4. Animal species used for models of (A) TMJ cartilage/osteochondral defects and (B) OA.
Cells 13 00990 g004
Figure 5. MSC-based therapies alleviate pathological processes in TMJ cartilage/osteochondral defects and OA through a multifaceted mechanism of enhancing cellular proliferation, migration, and matrix synthesis and modulating the immune reactivity.
Figure 5. MSC-based therapies alleviate pathological processes in TMJ cartilage/osteochondral defects and OA through a multifaceted mechanism of enhancing cellular proliferation, migration, and matrix synthesis and modulating the immune reactivity.
Cells 13 00990 g005
Table 1. Summary of characteristics of animal models.
Table 1. Summary of characteristics of animal models.
Author, YearAnimalAgeSample SizeGenderWeightAnimal ModelMethod of Induction
Zhu, 2011 [19]Eastern cross goats25–28 months50Male32–37 kgOsteochondral defectSurgical; 3 mm diameter, 5 mm depth
Sun, 2018 [20]Goats6–8 months12Male10–22 kgCondylar cartilage defectSurgical; full-thickness condylar cartilage defect
Putnova, 2019 [21]NZ rabbits10 months17MaleNROsteochondral defectSurgical; NR
Cheng, 2019 [22]NZ rabbits6 months180MaleNROsteochondral defectSurgical; 3 mm diameter, 3 mm depth
Sumarta, 2021 [23]Wistar rats3 months20Male200–300 gOsteochondral defectSurgical; 1 mm diameter
Cheng, 2021 [24]NZ rabbits4 months45Male2–3 kgOsteochondral defectSurgical; 3 mm diameter, 2 mm depth
Yu, 2021 [25]Goats12 months12NR20 kgOsteochondral defectSurgical; 5 mm diameter, 5 mm depth
Wang, 2021 [26]NZ rabbits7 months16Male3 kgOsteochondral defectSurgical; 2 mm diameter, 3 mm depth
Guastaldi, 2021 [27]C57BL/6 mice8–10 weeks30FemaleNROsteochondral defectSurgical; linear condylar cartilage defect
Gomez, 2021 [28]C57BL/6 mice8–10 weeks8FemaleNROsteochondral defectSurgical; focal cartilage defect
Liu, 2022 [29]NZ rabbitsNR56NR2.2–2.5 kgOsteochondral defectSurgical; 2 mm diameter, 2 mm depth
Chen, 2013 [30]NZ rabbits6 months46NR2.5–3.2 kgTMJ-OASurgical; disc resection
Lu, 2015 [31]C57BL/6J mice6 weeks27Female17–19 gTMJ-OAMechanical; UAC
Zaki, 2017 [32]NZ rabbitsNR50Male1–1.5 kgTMJ-OAChemical; bovine collagen II injection
Zhang, 2017 [33]C57BL/6J mice6 weeks40Female17–19 gTMJ-OAMechanical; UAC
Kim, 2019 [34]NZ rabbitsNR25Male2.5–2.8 kgTMJ-OAChemical; MIA injection
Zhang, 2019 [35]SD rats8 weeks48Female198–271 gTMJ-OAChemical; MIA injection
Ogasawara, 2020 [36]Mice11 weeks20MaleNRTMJ-OAMechanical; forced mouth opening
Cui, 2020 [37]SD rats7 weeks15Female180–200 gTMJ-OAChemical; CFA and MIA injection
Wang, 2021 [38]NZ rabbits12–18 weeks7Male and femaleNRTMJ-OAChemical; Collagenase II injection
Köhnke, 2021 [39]Rabbits12 weeks28Female800 gTMJ-OAChemical; Collagenase II injection
AbuBakr, 2022 [40]Albino rats3–4 months48Male150–170 gTMJ-OAChemical; MIA injection
Yang, 2022 [41]SD rats6 weeks18MaleNRTMJ-OAChemical; CFA combined with IL-1β injection
CFA, Complete Freund’s adjuvant; IL-1β, interleukin-1β; MIA, monosodium iodoacetate; NZ, New Zealand; NR, not reported; SD, Sprague Dawley; TMJ-OA, temporomandibular joint osteoarthritis; UAC, unilateral anterior cross-bite.
Table 2. Characterization of MSCs and their derived secretome and EVs in vitro.
Table 2. Characterization of MSCs and their derived secretome and EVs in vitro.
Author, YearSourceIsolation MethodCharacterizationSize DistributionMarker Expression
Zhu, 2011 [19]Goat BMSCsDensity gradient centrifugation, adherence to tissue culture flaskNRNAND
Sun, 2018 [20]Goat BMSCs and auricular chondrocytesCentrifugation, adherence to tissue culture flaskPhase contrast microscopyNAND
Putnova, 2019 [21]Human ADRCsCelution 800/CRS systemNRNACD146+, CD34+, CD31+, CD45+
Cheng, 2019 [22]Rabbit BMSCsCentrifugation, adherence to tissue culture flaskFlow cytometry, adipogenic and osteogenic differentiation, ARS, ALP, oil red O staining, TEM, SEMNACD34−, CD45−, CD29+ and CD44+
Sumarta, 2021 [23]Human UCMSCsNRImmunocytochemical staining, flow cytometryNACD45−, CD73+, CD90+, CD105+
Cheng, 2021 [24]Rabbit BMSCsCentrifugation, adherence to tissue culture flaskImmunofluorescence, WB analysis for HIF-1α expressionNAHIF-1α
Yu, 2021 [25]Goat BMSCsCentrifugation, adherence to tissue culture flaskChondrogenic induction,
CLSM, SEM, gene expression analysis
NACol I, Col II, Sox9
Wang, 2021 [26]Rabbit BMSCs and articular chondrocytesCentrifugation, adherence to tissue culture flaskLive/dead, osteogenic and chondrogenic differentiation, ALP, ARS, SEM, GAG/DNA, gene expression analysisNAACAN, Sox9, Col1a2, Col2a1, Col10a1
Guastaldi, 2021 [27]Mouse TMJ condyle-derived MSCsNRLive/dead, flow cytometryNANR
Gomez, 2021 [28]Human BMSCsFicoll-Hypaque isolation, adherence to tissue culture flaskFlow cytometry, osteogenic and chondrogenic differentiation, ARS, AB stainingNACD90+, CD105+
Liu, 2022 [29]Human ADSC-sEVsUltracentrifugationHuman ADSCs: flow cytometry
human ADSC-sEVs: TEM, NTA, nanoflow cytometry
~130 nmHuman ADSCs: CD73+, CD90+, CD105+, CD14−, CD19−, CD34−, CD45−, HLA-DR- Human ADSC-sEVs: CD9+, CD81+
Chen, 2013 [30]Rabbit BMSCsCentrifugation, adherence to tissue culture flaskNDNAND
Lu, 2015 [31]GFP-labeled mouse BMSCsNRNRNAND
Zaki, 2017 [32]Rabbit BMSCsCentrifugation, adherence to tissue culture flaskFlow cytometryNACD90+, CD105+, CD106+, CD45−
Zhang, 2017 [33]GFP-labeled mouse BMSCsNRNRNAND
Kim, 2019 [34]Human UCMSCsDensity gradient centrifugation, adherence to tissue culture flaskFlow cytometry, proliferation, chondrogenic differentiation, HE, AB staining, gene expression analysisNACD34−, CD45−, CD90+, CD105+
FGF-2, TGF-β1, IGF-1, Col1α1, Col2α1, ACAN, OCT4, NANOG, Sox2
Zhang, 2019 [35]Human ESC-MSC exosomesSize fractionationWB, NTA, protein concentration100–200 nmCD81, ALIX, TSG101
Ogasawara, 2020 [36]Human SHED-CMSupernatants after centrifugationLC-MS/MS analysisNASPON2, IGF2, SDC4, SDC1, SFRP1, PTN, MDK, TGFb2, PDGFD, HGF
Cui, 2020 [37]Human DPSCsCentrifugation, adherence to tissue culture flaskNRNANR
Wang, 2021 [38]Human BMSC-derived sEVsCentrifugation, microfiltration, ultrafiltrationNTA, TEM, immunoblotting~106 nmCD81+, CD63+, Rab5+, ALIX+, GRP94−
Köhnke, 2021 [39]Human adipose-derived MSCsNRFlow cytometry, adipogenic, chondrogenic and osteogenic differentiationNACD13+, CD44+, CD73+, CD90+, CD105+, CD31−, CD45−, CD235a−, HLA-II−
AbuBakr, 2022 [40]Rat BMSC-derived MVsUltracentrifugationTEM, FACS, ELISA~79 nmBMSCs: CD90+, CD105+, CD14−
BMSC-derived MVs: CD63+, CD81+
Yang, 2022 [41]Rat BMSCsCentrifugation, adherence to tissue culture flaskSEM, immunocytochemical staining, ELISA, gene expression analysisNASox9, Col2a1, ACAN
AB, alcian blue; ACAN, aggrecan; ADRCs, adipose-derived regenerative cells; ADSC, adipose-derived stem cell; AE, ADSC-derived sEV; ALIX, ALG2-interacting protein; ALP, alkaline phosphatase; ARS, Alizarin Red S; BMSCs, bone marrow-derived mesenchymal stem cells; CLSM, confocal laser scanning microscopy; CM, conditioned media; SHED, human exfoliated deciduous teeth stem cells; DPSCs, dental pulp stem cells; ELISA, enzyme-linked immunosorbent assay; ESC, embryonic stem cell; FACS, fluorescence-activated cell sorting; FGF-2, fibroblast growth factor-2; GFP, green fluorescence protein; GRP94, glucose-regulated protein 94; HGF, hepatocyte growth factor; HIF-1α, hypoxia-inducible factor-1-alpha; HLA-DR, MHC class II cell surface receptor encoded by the human leukocyte antigen complex; IGF-1, insulin-like growth factor-1 IHC, immunohistochemistry; LC-MS/MS analysis, liquid chromatography tandem mass spectrometry; MDK, midkine; MSC, mesenchymal stem/stromal cell; MVs, microvesicles; NA, not applicable; NANOG, nanog homeobox; ND, not done; NR, not reported; NTA, nanoparticle tracking analysis; OCT, octamer-binding transcription factor; PDGF, platelet derived growth factor; PTN, pleiotrophin; Rab5, ras-related protein 5; SEM, scanning electron microscopy; sEVs, small extracellular vesicles; SDC, syndecan; SFRP, secreted frizzled related protein; Sox, sex-determining region Y-box; SPON, R-Spondin; TEM, transmission electron microscopy; TGF-β, transforming growth factor-beta; TSG101, tumor susceptibility gene 101; UCMSCs, umbilical cord mesenchymal stem cells; WB, Western blot.
Table 3. Treatment parameters and analyses for included studies evaluating MSC-based therapies for cartilage/osteochondral defects.
Table 3. Treatment parameters and analyses for included studies evaluating MSC-based therapies for cartilage/osteochondral defects.
Author, YearGroupsStudy TimepointsConc.Volume/
Frequency
DeliveryMethod of Analysis
GrossImagingHistologyIHCMolecularPain
Zhu, 2011 [19]PLGA scaffold with NELL-1-modified BMSCs group, BMSCs group, PLGA group, empty defect group6, 24 weeks7.5 × 107 cells/mL40 μL/
one time
Implantation/PLGA scaffoldGross morphologyMicro-CT
(BV/TV)
SO, histological scoringCol IINDND
Sun, 2018 [20]Gel-cell group, gel group4, 8, 12 weeks5 × 107 cells/mLNR/
one time
Implantation/Pluronic F-127 gelGross morphologyX-rayHE, Wakitani scoringCol IINDND
Putnova, 2019 [21]Human ADRC group, control group11, 28 daysNR1 mL/
one time
Flushing/
ADRC solution
Gross morphologyNDHE, AB, HistomorphometryNDNDND
Cheng, 2019 [22]Blank control group, PRF group, BMSC sheet group, BMSC/PRF construct group, pressure-pretreated BMSC/PRF construct group2, 4, 8 weeksNRNR/
one time
Implantation/PRF scaffoldGross morphologyNDHE, TB, histological scoringNDSox9, Col II, ACANND
Sumarta, 2021 [23]Untreated defects, defects treated with UCMSCs, defects treated with PRF scaffold, defects treated with UCMSCs and PRF scaffold6 weeks2 × 106 cellsNR/
one time
Implantation/PRFNDNDHEKi67, FGF18, Sox9, Col II, ACANNDND
Cheng, 2021 [24]Empty group, collagen scaffold group, collagen scaffold with BMSCs group, collagen scaffold with HIF-1α overexpressing BMSCs group, sham group12 weeks2 × 105 cells/mLNR/
one time
Implantation/rat tail collagen scaffoldGross morphologyMicro-CT
(BV/TV, Tb.Th, Tb.Sp, Tb.N)
SO/FG,
histological scoring
Col II,
HIF-1α
NDND
Yu, 2021 [25]Healthy control, defect group, bi-layered scaffold group, bi-layered scaffold with induced 14-day BMSCs group2 months5 × 107 cells/mL200 μL/
one time
Implantation/PEG crosslinked-PLGA-g-PCL scaffoldGross morphologyNDHE,
SO/FG
Col I,
Col II
NDND
Wang, 2021 [26]Empty defect group, bi-layer scaffold group, bi-layer scaffold with BMSCs/chondrocyte group6, 24 weeks5 × 106 cells/mLNR/
one time
Implantation/HA-SH-Col I hydrogel-BCP ceramic scaffoldGross morphologyMicro-CT
(BMD, BV/TV, Tb.Th, Tb.N)
HE, Masson, SO/FG, O’Driscoll scoringCol I,
Col II,
Col X
NDND
Guastaldi, 2021 [27]Sham group, defect group, defect treated with MSCs + hydrogel + biosilica group4, 8 weeks1 × 106 cells/mL20 μL/
one time
Implantation/gelatine/biosilica-based hydrogel scaffoldNDNDHE, SO/FGNDNDND
Gomez, 2021 [28]BMSC/PRP treated group, control group (untreated), sham group6 weeks105 cells/mL100 µL/
one time
Implantation/PRPNDNDHE, AB, Mankin scoringNDNDND
Liu, 2022 [29]Non-implanted group, scaffold group, scaffold with AE group, scaffold with IAE group4, 8 weeks5 µg sEV/scaffoldNR/
one time
Implantation/SF scaffoldGross morphology (ICRS macro-scopic scoring)Micro-CT
(BV/TV, Tb.Th, Tb.Sp,
Tb.N)
HE, SO/FGCol I,
Col II
NDND
AB, alcian blue; AE, ADSC-derived sEV; BCP, biphasic calcium phosphate; BMD, bone mineral density; BV/TV, bone volume over total volume; Conc., concentration; Col, collagen; FG, fast green; HA, hyaluronic acid; HIF-1α, HA-SH, self-crosslinking thiolated hyaluronic acid; hypoxia-inducible factor-1-alpha; HE, hematoxylin and eosin; IAE, inflammation-stimulated ADSC-derived sEV; ICRS, International Cartilage Regeneration & Joint Preservation Society; IHC, immunohistochemistry; Ki-67, Kiel 67; Micro-CT, micro-computed tomography; NELL-1, NEL-like protein-1; PCL, polycaprolactone; PEG, polyethylene glycol; PLGA, poly(L-glutamic acid); PRF, platelet-rich fibrin; PRP, platelet-rich plasma; ND, not done; NR, not reported; SO, safranin O; SF, silk fibroin; TB, toluidine blue; Tb.N, trabecular number; Tb.Th, trabecular thickness; Tb.Sp, trabecular spacing; TB, toluidine blue.
Table 4. Treatment parameters and analyses for included studies evaluating the efficacy of MSC-based therapies for OA treatment.
Table 4. Treatment parameters and analyses for included studies evaluating the efficacy of MSC-based therapies for OA treatment.
Author, YearGroupsStudy TimepointsConc.Volume/
Frequency
DeliveryMethod of Analysis
GrossImagingHistologyIHCMolecularPain
Chen, 2013 [30]Non-chondrogenic MSCs group, chondrogenic differentiated MSCs group, vehicle (hylartin) group, normal control group4, 12, 24 weeks2 × 106 cells/mL100 µL/
one time
I.A. injection/
hylartin solution
NDMicro-CT
(BV/TV, Tb.Th, Tb.Sp)
SO, TB, histological gradingCol IISox9, Col2a1, ACAN, MMP13ND
Lu, 2015 [31]Control group, BMSCs group, UAC group, UAC with BMSCs group4, 8, 12 weeks105 cells/mL20 µL/
weekly
I.A. injection/DMEM with 10% FCSNDMicro-CT (BV/TV, Tb.Th, Tb.Sp, Tb.N)HE, SO, TRAPCol II, SDF-1, RANTES, GFPCol1a1,2a1,10a1, ACAN, OCN, MMP13, TNF-α, IL-1βND
Zaki, 2017 [32]Untreated group, PBS group, BMSCs group, Arthritis with PBS group, Arthritis with BMSCs group3 weeksNR200 µL/
one time
I.A. injection/
PBS
NDNDHE, SO, histomorphometryNDNDND
Zhang, 2017 [33]Control group, UAC group, UAC with BMSCs group4, 8, 12 weeks105 cells/mL20 µL/
weekly
I.A. injection/
DMEM with 10% FCS
NDTEMHE, SOCol I,
Col II, Ki67, DAP3, CD163, GFP
NDND
Kim, 2019 [34]Healthy control group; TMJ-OA group; TMJ-OA treated with DEX group; TMJ-OA treated with low, medium, or high dose of human UCMSCs groups8 weeks5 × 105, 2.5 × 106, 5 × 106 cells/mL200 µL/
one time
I.A. injection/
saline
NDMicro-CTHE, SO/FG, Mankin scoringACAN, Col ITNF-α, IL-1β,-6,-10,-17, TGF-β1, IGF-1, FGF-2, Col1a1,2a1, ACANND
Zhang, 2019 [35]OA + PBS group, OA + exosome group, sham group2, 4, 8 weeks2 mg/mL50 µL/
weekly
I.A. injection/
PBS
NDMicro-CT (BV/TV Tb.Th Tb.Sp
Tb.N)
HE, TB, Mankin scoring, histomorphometryα-SMA, MMP13, IL-1β, iNOS, PCNA, CCP3IL-1β,-4,-10 iNOS, TNF-α, TGF-β1, Col1a1,2a1, Sox9, COMP, ACAN, MMP3,9,13 TIMP1,2,3, ADAMTS4,5, BAX, Casp3,8,9, Survivin, PCNA, α-SMA, SP, CGRP, NGF, P75NTR, TrkAHWT
Ogasawara, 2020 [36]Sham group, pre-treatment group, DMEM group, SHED-CM group,12 days3 μg/mL0.5 mL/
5 days
Tail vein injection/
serum-free DMEM
NDMicro-CT (BV/TV, Tb.Th, Tb.Sp)HE, TB, Mankin scoring, TUNEL, TRAPMMP13, iNOS, IL-1β, PCNANDHWT
Cui, 2020 [37]Control group, TMJ-OA with saline group, TMJ-OA with DPSCs group2, 4 weeks4 × 106 cells/mL50 µL/
one time
I.A. injection/
saline
NDMicro-CT (BV/TV
BS/BV
Tb.Sp
Tb.N)
HE, TB, Mankin scoringCD4, IFN-γ, TNF-α, MMP3, MMP13NDHWT
Wang, 2021 [38]Control group, OA group, sEV group, OA + sEV group4, 6, 8 weeks(2–4) × 109 particles/mL200 µL/
one time
I.A. injection/
PBS
Gross morphologyMicro-CT (BV/TV
BS/BV BMD
Tb.Th)
Tb.Sp
HE, SO/FG, AB, Wakitani scoringPCNA, Col I, Col II, ACAN, Sox9, MMP13, RUNX2NDND
Köhnke, 2021 [39]AB serum group, HA group, MSCs group, MSCs + HA group4 weeks106 cells/mL150 µL/
one time
I.A. injection/
HA
NDCT, SEMSO, Picrosirius redNDNDND
AbuBakr, 2022 [40]OA group, OA + MVs group, OA + PRP group2, 4 weeks.2 mg/mL50 µL/
one time
I.A. injection/
PBS
NDNDHENDIL-1β, TNF-α, NF-κB, MMP3,13, Col IIND
Yang, 2022 [41]BMSC-coated microspheres group, microspheres group, control group1, 2 weeksNR200 µL/
one time
I.A. injection/
GelMA microspheres
NDSEM, Micro-CT (BMD, BV/TV)HESox9NDND
ADAMTS, A disintegrin and metalloproteinase with thrombospondin motifs; AB, alcian blue; α-SMA, α-smooth muscle actin; BAX, bcl2 associated x; BMD, bone mineral density; BS/BV; bone surface over bone volume; BV/TV; bone volume over total volume; CCP3, cleaved caspase-3; CGRP, calcitonin gene-related peptide; COMP, cartilage oligomeric matrix protein; CT, computed tomography; DAP3, death-associated protein 3; DEX, dexamethasone; DMEM, Dulbecco’s Modified Eagle Medium; FG, fast green; FCS, fetal calf serum; FGF-2, fibroblast growth factor-2; GelMA, gelatin methacryloyl; HA, hyaluronic acid; HE, hematoxylin and eosin; HWT, head withdrawal threshold; I.A., intra-articular; IFN- γ, interferon-γ; IGF, insulin-like growth factor-1; IL-1β, interleukin-1beta; iNOS, inducible nitric oxide synthase; MMP, matrix metalloproteinase; ND, not done; NF-κB, nuclear factor kappa B; NGF, nerve growth factor; OCN, osteocalcin; P75NTR, p75 neurotrophin receptor; PCNA, proliferative cell nuclear antigen; RANTES, regulated on activation normal T-cell expressed and secreted; RUNX2, RUNX family transcription factor 2; SDF-1, stromal cell-derived factor 1; SO, safranin O; SP, substance P; TB, toluidine blue; Tb.N, trabecular number; Tb.Th, trabecular thickness; Tb.Sp, trabecular spacing; TGF-β1, transforming growth factor-beta-1; TIMP, tissue inhibitor metalloproteinase; TNF-α, tumor necrosis factor-alpha; TRAP, tartrate-resistant acid phosphatase; TrkA, tropomyosin receptor kinase A; TUNEL, terminal deoxynucleotidyl transferase (TdT) dUTP nick-end labeling.
Table 5. Summary of key outcomes of included studies evaluating MSC-based therapies for cartilage/osteochondral defects.
Table 5. Summary of key outcomes of included studies evaluating MSC-based therapies for cartilage/osteochondral defects.
Author, YearKey Outcomes (In Vitro)Key Outcomes (In Vivo)
Zhu, 2011 [19]NDNELL-1-modified BMSCs/PLGA composite rapidly repaired large osteochondral defect in the mandibular condyle with regeneration of fibrocartilage and subchondral bone.
Sun, 2018 [20]NDCo-culture of goat BMSCs and chondrocytes at 7:3 ratio in hydrogel induced chondrogenic differentiation of BMSCs to enhance TMJ repair.
Putnova, 2019 [21]NDHuman ADRCs supported soft tissue repair and promoted bone remodeling in hard tissues.
Cheng, 2019 [22]Hydrostatic pressure pre-treatment (120 kPa/1 h for 4 days) optimally promoted BMSC proliferation and chondrogenic differentiation in the BMSC/PRF construct.Pressure-pretreated BMSC/PRF construct enhanced cartilage regeneration with improved mechanical properties and integration of the neocartilage.
Sumarta, 2021 [23]NDHuman UCMSCs in PRF scaffold proved capable of regenerating mandibular cartilage defect through increased expression of FGF-18, Sox9, Ki67, Col II, ACAN, and cartilage thickness
Cheng, 2021 [24]NDTransplantation of HIF-1α overexpressed BMSCs combined with a collagen scaffold promoted cartilaginous repair of condylar cartilage and inhibited subchondral bone sclerosis in TMJ osteochondral defect.
Yu, 2021 [25]The bi-layered PLGA-g-PCL scaffold with segregated hydrophilicity–hydrophobicity facilitated chondrogenic differentiation of BMSCs toward top fibrocartilage layer and bottom hyaline cartilage layer.The bi-layered PLGA-g-PCL-PEG scaffold with segregated hydrophilicity–hydrophobicity carrying induced 14-day BMSCs enabled reconstruction of goat hierarchical TMJ condylar cartilage.
Wang, 2021 [26]The HA-SH/Col I blend hydrogel-BCP ceramic bi-layered scaffold enhanced proliferation and matrix synthesis of rabbit BMSCs and chondrocytes, as well as osteogenic differentiation of BMSCs.Rabbit BMSCs/chondrocytes-loaded bi-layer scaffold could effectively promote the regeneration of both fibrocartilage and subchondral bone.
Guastaldi, 2021 [27]NDThe MSCs + hydrogel + biosilica was effective in promoting TMJ condylar cartilage regeneration, as evidenced by intact articular surfaces, maturation, and distribution of chondrocytes along the condyle.
Gomez, 2021 [28]NDBMSC/PRP implantation promoted repair of the articular surface with the presence of cartilage-like tissue and subchondral bone filling the defect area.
Liu, 2022 [29]Both IAE and AE showed comparable effects on proliferation, whereas IAE outperformed AE on BMSC migration and M2 macrophage polarization in vitro. RNA sequencing identified high miR-27b-3p expression levels in IAE that may regulate macrophage polarization by targeting CSF-1.IAE loaded onto SF scaffold outperformed AE-loaded scaffold in TMJ osteochondral regeneration, with newly formed cartilage stained for GAG, collagen I and II, and reconstituted subchondral bone.
AE, ADSC-derived sEV; BMSCs, bone marrow-derived mesenchymal stem cells; CSF-1, colony-stimulating factor-1; GAG, glycosaminoglycan; HA-SH, self-crosslinking thiolated hyaluronic acid; HIF-1α, hypoxia-inducible factor-1-alpha; ND, not done; NELL-1, NEL-like protein-1; PCL, polycaprolactone; PEG, polyethylene glycol; PLGA, poly-lactic-co-glycolic acid; PLGA-g-PCL-PEG, poly(L-glutamic acid)-graft-poly(Ɛ-caprolactone)-poly(ethylene glycol); PRF, platelet-rich fibrin; PRP, platelet-rich plasma; IAE, inflammation-stimulated ADSC-derived sEV; SF, silk fibroin.
Table 6. Summary of key outcomes for included studies evaluating MSC-based therapies for OA treatment.
Table 6. Summary of key outcomes for included studies evaluating MSC-based therapies for OA treatment.
Author, YearKey Outcomes (In Vitro)Key Outcomes (In Vivo)
Chen, 2013 [30]NDI.A injection of MSCs could delay the progression of TMJ-OA, and in vitro chondrogenic differentiation of MSCs could enhance the therapeutic effects.
Lu, 2015 [31]SDF-1 and RANTES were significantly increased in the UAC cartilage compared to the control cartilage. Migration of BMSCs was enhanced when cocultured with a UAC TMJ condyle and was attenuated in the presence of AMD3100 (CXCR4 antagonist) or BX471 (CCR1 antagonist).BMSC injections improved cartilage repair and subchondral bone restoration in TMJ-OA mice induced by UAC. The locally injected BMSCs were found to implant and differentiate into chondrocytes in OA cartilage. These effects of BMSCs were inhibited by AMD3100 and BX471.
Zaki, 2017 [32]NDRabbit BMSCs could safely and effectively repair degenerative changes of rabbit TMJs with bovine collagen II-induced arthritis.
Zhang, 2017 [33]Fluid flow shear stress (FFSS) stimulation induced cell death of superficial and deep zone chondrocytes. Genes associated with chondrocyte hypertrophy and fibrosis were upregulated in deep zone chondrocytes with FFSS stimulation.BMSCs rescued the damaged cartilage by increasing matrix production and scavenging activity.
Kim, 2019 [34]Human UCMSC lines isolated from different donors showed comparable proliferation ability but varying in vitro capacities for chondrogenesis and expression of marker genes for growth factors and ECM compared to that of BMSCs.Human UCMSCs exerted anti-inflammatory effects and promoted cartilage regeneration in a rabbit model of TMJ-OA. Medium dose of MSCs was most effective in regenerating the articular cartilage with the highest gene expression levels of growth factors.
Zhang, 2019 [35]Human ESC-MSC exosomes suppressed inflammation and restored matrix synthesis in IL-1β-treated chondrocytes via adenosine receptor activation of AKT, ERK, and AMPK pathways.Human ESC-MSC exosomes suppressed pain and inflammation and reduced cell apoptosis and matrix degradation while enhancing cell proliferation and matrix synthesis to promote overall TMJ repair and regeneration.
Ogasawara, 2020 [36]LC-MS/MS analysis identified several factors present in SHED-CM that could be involved in processes such as anti-fibrosis, anti-apoptosis, anti-inflammation, proliferation, differentiation, and migration of chondrocytes.SHED-CM contained multiple therapeutic factors with the potential to promote the regeneration and repair of mechanical-stress-induced mouse TMJ-OA.
Cui, 2020 [37]DPSCs downregulated the expression of MMP3 and MMP13 in fibroblast-like synoviocytes by suppressing STAT1 activation under the inflammatory condition.DPSC local injection relieved pain, suppressed synovial inflammation, and reduced cartilage degradation and subchondral bone destruction in rats.
Wang, 2021 [38]BMSC-sEVs enhanced proliferation and migration of mandibular condylar chondrocytes, possibly through activation of the Hippo pathway.BMSC-sEVs promoted cartilage reconstruction in TMJ-OA via the autotaxin–YAP signaling axis.
Köhnke, 2021 [39]NDHuman adipose-derived MSCs with or without HA were more effective than serum and HA alone in restoring the cartilage thickness in a rabbit TMJ-OA model.
AbuBakr. 2022 [40]NDBMSC-derived MVs restored damaged condylar structure by suppressing inflammation and matrix degradation in a rat model of TMJ-OA.
Yang, 2022 [41]GelMA microspheres loaded with TGF-β enhanced chondrogenic differentiation of BMSCs coated on the microspheres.Rat BMSC-coated GelMA microspheres endowed with superwettable properties and sustained TGF-β release, can efficiently colonize the bone defect site, release cytokine, and promote cartilage healing.
BMSCs, bone marrow-derived mesenchymal stem cells; CCR, C-C chemokine receptor; CXCR, C-X-C chemokine receptor; DPSCs, dental pulp stem cells; GelMA, gelatin methacryloyl; ESC, embryonic stem cell; ECM, extracellular matrix; FFSS, fluid flow shear stress; HA, hyaluronic acid; I.A., intra-articular; MVs, microvesicles; ND, not done; RANTES, regulated on activation normal T-cell expressed and secreted; SDF-1, stromal cell-derived factor 1; SHED-CM, conditioned media from human exfoliated deciduous teeth stem cells; TMJ-OA, temporomandibular joint osteoarthritis; TGF-β; transforming growth factor-beta; UAC, unilateral anterior cross-bite; UCMSCs, umbilical cord mesenchymal stem cells; YAP, yes-associated protein.
Table 7. ARRIVE checklist to evaluate the quality of the included studies for the repair of cartilage/osteochondral defects.
Table 7. ARRIVE checklist to evaluate the quality of the included studies for the repair of cartilage/osteochondral defects.
Item/Item NumberZhu, 2011 [19]Sun,
2018 [20]
Putnova, 2019 [21]Cheng, 2019 [22]Sumarta, 2021 [23]Cheng, 2021 [24]Yu,
2021 [25]
Wang, 2021 [26]Guastaldi, 2021 [27]Gomez, 2021 [28]Liu, 2022 [29]Cat. ScoreTotal ScoreRatio
1. Title (0, inaccurate/not concise; 1, accurate/concise)1111111111111111
2. Abstract
Summary of the background; research objectives, including details of the species or strain of animal used; key methods; principal findings; and conclusions of the study (0, clearly inaccurate; 1, possibly accurate; 2, clearly accurate)
2112221122218220.82
3. Introduction
Background: objectives, experimental approach and rationale, and relevance to human biology (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
1121111111213220.59
4. Introduction
Objectives: primary and secondary (0, not clear; 1, clear)
111100110017110.64
5. Methods
Ethical statement: nature of the review permission, relevant licenses, and national and institutional guidelines for the care and use of animals (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
1122211122116220.73
6. Methods
Study design: number of experimental and control groups and any steps taken to minimize bias (i.e., allocation concealment, randomization, blinding) (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
1222122210015220.68
7. Methods
Experimental procedure: precise details (i.e., how, when, where, why) (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
1121112122115220.68
8. Methods
Experimental animals: species, strain, sex, developmental stage, weight, and source of animals (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
1111111111111220.5
9. Methods
Housing and husbandry: conditions and welfare-related assessments and interventions (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
012100111209220.41
10. Methods
Sample size: total number of animals used in each experimental group, details of calculation (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
1211111121012220.55
11. Methods
Allocation animals to experimental groups: randomization or matching, order in which animals were treated and assessed (0, no; 1, yes)
011111110007110.64
12. Methods
Experimental outcomes: definition of primary and secondary outcomes (0, no; 1, unclear/not complete; 2, yes)
000000000000220
13. Methods
Statistical methods: details and unit of analysis (0, no; 1, unclear/ not complete; 2, yes)
2212122200216220.73
14. Results
Baseline data: characteristics and health status of animals (0, no; 1, yes)
000101111005110.45
15. Results
Numbers analyzed: absolute numbers in each group included in each analysis, explanation for exclusion (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
100101112018220.36
16. Results
Outcomes and estimation: results for each analysis with a measure of precision (0, no; 1, unclear/not complete; 2, yes)
2111211111214220.64
17. Results
Adverse events: details and modifications for reduction (0, no; 1, unclear/not complete; 2, yes)
000000002103220.14
18. Discussion
Interpretation/scientific implications: study limitations including animal model, and implications for the 3Rs (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
1101011211110220.45
19. Discussion
Generalizability/translation: relevance to human biology (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
2222111211217220.77
20. Discussion
Funding: resources and role of the funders (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
2222122222221220.95
Table 8. ARRIVE checklist evaluating the quality of the included studies for OA treatment.
Table 8. ARRIVE checklist evaluating the quality of the included studies for OA treatment.
Item/Item NumberChen, 2013 [30]Lu, 2015 [31]Zaki, 2017 [32]Zhang, 2017 [33]Kim, 2019 [34]Zhang, 2019 [35]Ogasawara, 2020 [36]Cui, 2020 [37]Wang, 2021 [38]Köhnke, 2021 [39]AbuBakr, 2022 [40]Yang, 2022 [41]Cat. ScoreTotal ScoreRatio
1. Title (0, inaccurate/not concise; 1, accurate/concise)11111111111112121
2. Abstract
Summary of the background; research objectives, including details of the species or strain of animal used; key methods; principal findings; and conclusions of the study (0, clearly inaccurate; 1, possibly accurate; 2, clearly accurate)
11121111112114240.58
3. Introduction
Background: objectives, experimental approach and rationale, and relevance to human biology (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
11121211111114240.58
4. Introduction
Objectives: primary and secondary (0, not clear; 1, clear)
0100010110015120.42
5. Methods
Ethical statement: nature of the review permission, relevant licenses, and national and institutional guidelines for the care and use of animals (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
11112222222220240.83
6. Methods
Study design: number of experimental and control groups and any steps taken to minimize bias (i.e., allocation concealment, randomization, blinding) (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
10202222222219240.79
7. Methods
Experimental procedure: precise details (i.e., how, when, where, why) (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
21111211122116240.67
8. Methods
Experimental animals: species, strain, sex, developmental stage, weight, and source of animals (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
12121111111114240.58
9. Methods
Housing and husbandry: conditions and welfare-related assessments and interventions (0, clearly insufficient; 1, possibly sufficient; 2, clearly sufficient)
10110211022011240.46
10. Methods
Sample size: total number of animals used in each experimental group and details of calculation (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
20202121101012240.5
11. Methods
Allocation of animals to experimental groups: randomization or matching and order in which animals were treated and assessed (0, no; 1, yes)
0010111111119120.75
12. Methods
Experimental outcomes: definition of primary and secondary outcomes (0, no; 1, unclear/not complete; 2, yes)
0000000000000240
13. Methods
Statistical methods: details and unit of analysis (0, no; 1, unclear/ not complete; 2, yes)
22222211212221240.88
14. Results
Baseline data: characteristics and health status of animals (0, no; 1, yes)
1110111101008120.67
15. Results
Numbers analyzed: absolute numbers in each group included in each analysis and explanation for exclusion (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
0100121110018240.33
16. Results
Outcomes and estimation: results of each analysis with a measure of precision (0, no; 1, unclear/not complete; 2, yes)
11211221111115240.63
17. Results
Adverse events: details and modifications for reduction (0, no; 1, unclear/not complete; 2, yes)
00102210220010240.42
18. Discussion
Interpretation/scientific implications: study limitations including animal model, and implications for the 3Rs (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
11111111111011240.46
19. Discussion
Generalizability/translation: relevance to human biology (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
11112212221117240.71
20. Discussion
Funding: resources and role of the funders (0, clearly inadequate; 1, possibly inadequate; 2, clearly adequate)
22022222222222240.92
Table 9. Risk of bias assessed using SYRCLE risk of bias assessment tool.
Table 9. Risk of bias assessed using SYRCLE risk of bias assessment tool.
Author, YearSelection BiasPerformance BiasDetection BiasAttrition BiasReporting BiasOther
Sequence GenerationBaseline Characte-risticsAllocation Conceal-mentRandom HousingBlindingRandom Outcome AssessmentGross BlindingImaging BlindingHistology BlindingIHC BlindingMolecular BlindingIncomplete Outcome DataSelective Outcome ReportingOther Bias
Zhu, 2011 [19]Unclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskLow RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Sun, 2018 [20]Unclear RiskUnclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Putnova, 2019 [21]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskNDNDUnclear RiskLow RiskLow Risk
Cheng, 2019 [22]Unclear RiskUnclear RiskLow RiskUnclear RiskUnclear RiskLow RiskUnclear RiskNDLow RiskNDUnclear RiskUnclear RiskLow RiskLow Risk
Sumarta, 2021 [23]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDNDUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Cheng, 2021 [24]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Yu, 2021 [25]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Wang, 2021 [26]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Guastaldi, 2021 [27]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDNDUnclear RiskNDNDLow RiskLow RiskLow Risk
Gomez, 2021 [28]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskNDNDLow RiskLow RiskLow Risk
Liu, 2022 [29]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskLow RiskUnclear RiskLow RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Chen, 2013 [30]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskLow RiskUnclear RiskUnclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskLow RiskLow Risk
Lu, 2015 [31]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskLow RiskLow Risk
Zaki, 2017 [32]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDNDUnclear RiskNDNDLow RiskLow RiskLow Risk
Zhang, 2017 [33]Unclear RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Kim, 2019 [34]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskUnclear RiskUnclear RiskLow RiskLow RiskLow Risk
Zhang, 2019 [35]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskUnclear RiskUnclear RiskLow RiskLow RiskLow Risk
Ogasawara, 2020 [36]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskUnclear RiskUnclear RiskNDLow RiskLow RiskLow Risk
Cui, 2020 [37]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
Wang, 2021 [38]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskUnclear RiskLow RiskUnclear RiskNDLow RiskLow RiskLow Risk
Köhnke, 2021 [39]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskNDNDLow RiskLow RiskLow Risk
AbuBakr, 2022 [40]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDNDUnclear RiskNDUnclear RiskUnclear RiskLow RiskLow Risk
Yang, 2022 [41]Unclear RiskLow RiskLow RiskUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskUnclear RiskUnclear RiskNDUnclear RiskLow RiskLow Risk
IHC, immunohistochemistry; ND, not done.
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

Jiang, Y.; Shi, J.; Di, W.; Teo, K.Y.W.; Toh, W.S. Mesenchymal Stem Cell-Based Therapies for Temporomandibular Joint Repair: A Systematic Review of Preclinical Studies. Cells 2024, 13, 990. https://doi.org/10.3390/cells13110990

AMA Style

Jiang Y, Shi J, Di W, Teo KYW, Toh WS. Mesenchymal Stem Cell-Based Therapies for Temporomandibular Joint Repair: A Systematic Review of Preclinical Studies. Cells. 2024; 13(11):990. https://doi.org/10.3390/cells13110990

Chicago/Turabian Style

Jiang, Yuanyuan, Jiajun Shi, Wenjun Di, Kristeen Ye Wen Teo, and Wei Seong Toh. 2024. "Mesenchymal Stem Cell-Based Therapies for Temporomandibular Joint Repair: A Systematic Review of Preclinical Studies" Cells 13, no. 11: 990. https://doi.org/10.3390/cells13110990

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