Next Article in Journal
Development of a Novel Medical Device for Mucositis and Peri-Implantitis Treatment
Next Article in Special Issue
Mechanical Enhancement of Cytocompatible 3D Scaffolds, Consisting of Hydroxyapatite Nanocrystals and Natural Biomolecules, Through Physical Cross-Linking
Previous Article in Journal
Skeletal Muscle Tissue Engineering: Biomaterials-Based Strategies for the Treatment of Volumetric Muscle Loss
Previous Article in Special Issue
Material-Dependent Formation and Degradation of Bone Matrix—Comparison of Two Cryogels
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair

by
Salomi Desai
and
Chathuraka T. Jayasuriya
*
Department of Orthopaedics, Warren Alpert Medical School of Brown University and the Rhode Island Hospital, Providence, RI 02903, USA
*
Author to whom correspondence should be addressed.
Bioengineering 2020, 7(3), 86; https://doi.org/10.3390/bioengineering7030086
Submission received: 21 June 2020 / Revised: 25 July 2020 / Accepted: 30 July 2020 / Published: 4 August 2020
(This article belongs to the Special Issue Biomaterials for Bone Tissue Engineering)

Abstract

:
Harnessing adult mesenchymal stem/progenitor cells to stimulate skeletal tissue repair is a strategy that is being actively investigated. While scientists continue to develop creative and thoughtful ways to utilize these cells for tissue repair, the vast majority of these methodologies can ultimately be categorized into two main approaches: (1) Facilitating the recruitment of endogenous host cells to the injury site; and (2) physically administering into the injury site cells themselves, exogenously, either by autologous or allogeneic implantation. The aim of this paper is to comprehensively review recent key literature on the use of these two approaches in stimulating healing and repair of different skeletal tissues. As expected, each of the two strategies have their own advantages and limitations (which we describe), especially when considering the diverse microenvironments of different skeletal tissues like bone, tendon/ligament, and cartilage/fibrocartilage. This paper also discusses stem/progenitor cells commonly used for repairing different skeletal tissues, and it lists ongoing clinical trials that have risen from the implementation of these cells and strategies. Lastly, we discuss our own thoughts on where the field is headed in the near future.

1. Introduction

Repair/regeneration of damaged tissue is fundamental in the maintenance of normal homeostasis and it is fine-tuned at the cellular and molecular levels. The process of wound healing is highly conserved across the animal kingdom from some of the most primitive organisms, such as hydra [1] and amoeba [2], to advanced organisms like mammals [3,4]. However, the regenerative capabilities of advanced vertebrates are comparatively limited. In humans, certain tissue types, such as blood vessels and epidermis or the gastrointestinal track, can repair within hours, whereas in parts of the nervous system and skeletal system, repair and remodeling may take several weeks to months [5].
Musculoskeletal tissue is on the front line of exposure to injury in any active individual. It comprises nearly 40% of the total body mass and it is vital for body movements, which are tightly regulated by the coordinative efforts of its different components, such as bone, cartilage, muscle, ligament, and tendon. Skeletal tissue injuries occur by various means, which include, but are not limited to: overuse (i.e., wear and tear), trauma (i.e., accidental/sport injuries), and degenerative diseases (i.e., muscular dystrophy, osteoarthritis, etc. [6,7]). In most tissues, wound healing begins with the formation of a blood clot [8,9], followed by the recruitment of mesenchymal stem cells (MSCs). Studies have shown that MSCs are integral to the repair process as they help replace damaged tissue by differentiating and directly replacing damaged tissue [10], as well as by mediating indirect paracrine actions that regulate the migration and behavior of mature cells to promote healing [11,12]. These paracrine effects are mediated by factors secreted into the extracellular space by MSCs that are collectively referred to as the secretome, which regulates immunomodulation, angiogenesis, migration, cell proliferation/survival features. For further reading on the MSC secretome and its influences on tissue healing, please refer to Daneshmandi et al., 2020 [13].
The skeletal system consists of different tissues which exist in varying micro-environments that inherently have, or do not have, access to specific resources [14]. Due to these complexities, not all tissues have the same level of access to MSCs, hence factoring into why repair efficacy can vary dramatically between different skeletal tissues. In efforts to account for this disparity, different cell-based approaches have been implemented in order to jumpstart the repair process, including administering (or physically relocating) exogenous MSCs as biologic therapies to injury sites, or administering chemokines, growth factors, scaffolds (and combinations) to stimulate activation and migration of native endogenous MSCs to these locations (Figure 1). Surgical procedures (such as tibial microfracture surgery for knee cartilage focal defect repair) have also been developed to facilitate the migration of native MSCs from one tissue to another (i.e., bone to cartilage), without having to physically relocate the cells, in order to facilitate repair. Here, we will review the repair efficacies of both “endogenous” and “exogenous” cell-based repair approaches as they have been reported in different pre-clinical models. We will also touch on the limitations of these strategies.

2. Common Progenitor/Stem Cells Utilized for Skeletal Tissue Repair

Stem cell-based therapies have gained great interest from scientific communities in the last two decades, not only for their dynamic potential to treat infectious diseases and cancers, but also for their ability to stimulate tissue healing. Strategies to enhance healing by mimicking the natural cellular processes that occur during skeletal development are actively being explored. Most notably, transplantation of tissue specific progenitor cells or stimulating the recruitment of endogenous/native progenitors are approaches that are utilized to enhance healing of skeletal tissues in preclinical models [15,16,17,18,19]. The self-renewal capacities of skeletal tissues diminish with aging. This is partly attributed to the reduced abundance and efficiency (i.e., loss of proliferative capacity and/or differentiation potential) of native adult stem cells that are normally involved in repairing these tissues. The importance of recruiting and/or administrating cells with the greatest potential to rebuild the specific skeletal tissue in question have been highlighted in previous reviews [20,21,22,23,24].
Human clinical trials involving stem/progenitor cells for the treatment of musculoskeletal injuries are increasing [25,26,27,28,29]. Many of these trials involve somatic (adult) stem cells, with mesenchymal stem/progenitor cells being the most commonly used variety. MSCs are essential for the development and repair of the skeletal system—from embryonic bone formation to adult fracture healing and remodeling [30]. MSCs are heterogeneous populations of multipotent cells residing in adult tissues [30,31]. These cells are scattered throughout the skeletal system and they aid in the restoration of damaged tissue (Figure 2).
MSCs can be isolated from marrow [32], periosteal bone [33,34], cartilage [35,36,37], fibrocartilage [38,39], adipose tissue [40], tendon [41], ligament [42,43,44], and synovium [45]. MSCs isolated from these different tissues may vary in phenotype, morphology, differentiation, and proliferation capacity, but the consensus is that they all possess similar characteristics to bone marrow derived stromal cells (BM-MSCs), suggesting that MSC populations found across all skeletal tissues share a similar ontogeny [46]. Veritably, a set of minimum criteria have been described for the identification of these cells [47]; although, these criteria still need further refinement as some adult cells have been found to be capable of de-differentiating into stem-like cells in culture, further blurring the line between native MSCs and culture inspired MSC-like cells [48,49]. Regardless, however, both native MSCs and de-differentiated MSC-like cells exhibit demonstrated usefulness for tissue engineering applications as well as stimulating and/or enhancing tissue healing [50,51,52,53].
MSCs secrete a large variety of bioactive molecules that facilitate a regenerative environment. This trophic activity deem them immunosuppressive, especially to T-cells, and as a result, allogeneic MSCs can be used for therapeutic purposes [54]. MSCs exhibit significant immunomodulatory capacity and play an important role in maintaining immune homeostasis by interacting with chemokines, cytokines and cell surface molecules [55]. Besides considering which type of MSC is optimal for therapy, another equally important consideration is the microenvironment of the injured tissue in question, which also dictates overall repair success. The local tissue microenvironment can influence how cells integrate into the existing tissue matrix and how they restore the damaged tissue [14]. Tissue structure, organization, extracellular matrix (density and content), cellularity, and vasculature are all determining factors of how quickly and how well it will heal.
Alongside MSCs, human induced pluripotent stem cells (iPSCs) have also emerged as a potential cell source for skeletal tissue repair. iPSCs are cells that have been dedifferentiated into the state of pluripotency by the induction of select transcription factors including octamer-binding transcription factor-3/4 (OCT3/4), SRY-related high-mobility-group (HMG)-box protein-2 (SOX2), c-MYC, and Kruppel-like factor-4 (KLF4) [56,57]. These cells represent an inexhaustible cell source for tissue repair and regeneration; however, their incredible plasticity leads to concerns about their tendency to differentiate resulting in unexpected and undesirable phenotypic changes, which are now being addressed [58].

3. Stimulating Bone Repair

Bone is the most rigid and vital tissue of the skeletal system since it provides our bodies with structure, protects vital organs, and facilitates hematopoiesis occurring in the bone marrow. The structural components that shape the microenvironment of long bones are nanocrystals of hydroxyapatite (referred as mineral phase); collagen type-I (referred as organic phase); cellular components that include osteoclast, osteoblast, and osteocytes; ions (primarily calcium); and lastly, growth factors and cytokines [59]. The most common broken bone injury is the fracture. Compared to the other tissues of the skeletal system, bones are equipped with sophisticated microvasculature regularly supplying necessary components such as nutrients for growth and maintenance of homeostasis. Bone marrow, which is the primary site for hematopoiesis, attracts not only cytokines and growth factors, but it also attracts metastatic cells (in some cancers), hence stability of the bone microenvironment is critical during injury repair and metastatic disease [60].
Although the architecture and microenvironment of bone tissue allows fracture repair to efficiently occur, often healing to pre-injury state, about 10% of fractures will not heal normally [61]. Bone repair after fracture is a complex process involving a series of cellular and molecular events leading to new bone formation through systemic and local factors [59]. In general, fracture healing mainly involves five steps: hematoma formation, inflammation, angiogenesis, fibrocartilaginous callus formation to bony callus formation, and bone remodeling—with stem/progenitor cells contributing to each stage of healing [62,63]. Following bone fracture, the bone marrow is exposed and results in the rapid formation of a hematoma due to damaged blood vessel. This involves an inflammatory response where specific cytokines like interleukin-1 (IL-1), IL-6 and tumor necrosis factor-α (TNF-α) [64], neutrophils, macrophages, and other inflammatory cells initiate the bone healing mechanism, towards endochondral bone formation and remodeling [65]. Growth and differentiation factors like TGF-β superfamily including bone morphogenetic protein (BMPs), as well as insulin-like growth factors (IGFs), fibroblast growth factors (FGFs), and platelet-derived growth factors (PDGF), orchestrate MSC cell proliferation and differentiation (i.e., chondrogenesis, osteogenesis) [66]. Lastly, during revascularization of the injury site (angiogenesis), BMPs stimulate the expression of vascular endothelial growth factor (VEGF) by osteoblasts [67]. Due to these orchestrated events, bones have a high capacity for healing but repairing comminuted fractures that result in bone loss remains a major challenge [68]. Bone is the most commonly transplanted tissue, leading to 1.5 million annual grafts in the United States [69,70]. However, due to hurdles such as the limited availability of donor tissue for grafts [71], donor site morbidity [72], possibility of allograft rejection [73], and ethical dilemmas concerning putting potential donors at risk make this approach challenging [74,75]. Hence, alternative approaches to heal fractured or damaged bone tissue become necessary.
Previous studies have documented the use of cells and other biologics in the treatment of injured bone. Induction of osteogenesis by co-culturing C3H10T1⁄2 mesenchymal stem cells with chondrocytes stimulated well-known bone formation markers including osteopontin, osterix, and osteocalcin, to enhance the bone healing [76,77,78,79]. Other treatment strategies such as osteoanabolic and antiresorptive treatment strategies (including Wnt/ß-catenin signal activation to promote cell proliferation, differentiation, and microcirculation) have also been utilized [80]. Additionally, parathyroid hormone treatment and bisphosphonate injections have had promising results, but they have several limitations since long-term ablation of bone resorption interferes with the ongoing renewal of the bone matrix and affects skeletal integrity, ultimately affecting the fracture healing process [81,82,83,84,85]. These studies collectively suggest that current approaches still need refinement.
Hematopoietic stem cells (HSCs) have been widely used as a potential candidate for the bone injury healing. Kumar et al. demonstrated using a rat model that the treatment of IGF1 in combination with AMD3100 elevates the growth of fractured bone that is primarily regulated by cell signaling pathways, Akt and Erk [86]. Further, using a clonal cell transplantation model, Malhotra et al. demonstrated that HSCs migrate and give rise to osteocytes and chondrocytes aiding the healing process of fractured bones [87]. Recently, Chan et al. identified human skeletal stem cells (hSSCs) that undergo local expansion upon injury to the bone. Interestingly, upon comparative analysis, the study also identified evolutionary differences between mouse and human skeletogenesis [57]. Furthermore, the periosteum is considered to be the primary source of mesenchymal progenitor cells, giving rise to the fracture callus [88], but there is evidence that BM-MSCs and muscle progenitor cells also contribute to the bone repair process [63,89,90].
Exogenous injection/transplantation of MSCs seems to be the most common and most effective avenue of cell-motivated bone fracture repair that has been reported to date. In late 1960, Friedenstein et al. [91,92] first demonstrated the osteogenic potential of BM-MSCs. The authors filled a diffusion chamber with bone marrow and bone marrow fragments, which was placed in filtered culture containers, showing that the ectopic transplantation of bone marrow cells result in osteogenesis [91,92]. These findings were then further validated in the 1980s when it was documented that the injection of bone marrow aspirants directly on to the sites of bone fractures successfully promoted healing [93]. In the past two decades, several preclinical studies documenting the positive role of bone marrow aspirant injections on the healing of fractured bones have been conducted using rodent models [86,94,95]. Clinical studies have also been conducted using bone marrow aspirant to stimulate healing of fractured bones. Independent case studies conducted on 20 patients by Garg et al. and Sim et al. in 1993 showed 90% and 85% radiographic healing of the fractured bones, respectively [96,97]. Kim and colleagues conducted an open randomized clinical trial consisting of 64 patients with a long bone fracture, that were treated with a local injection of 3.0 × 107 osteogenically differentiated autologous BM-MSCs mixed with fibrin [98]. After 2 months, injected patients showed no complications and exhibited significant fracture healing. Overall, these studies demonstrate the efficacy of using BM-MSCs in a clinical setting for bone repair. However, due to the limitations of BM-MSC abundance (especially in older patients [99]) and their reduced regenerative capacity with continuous cell expansion [100], there is a need for a detailed clinical trial considering factors such as age and gender for their therapeutic applications.
Several groups have shown the potential of exogenous stem cells such as circulating skeletal progenitors as potential therapeutic candidates for the healing of fractured bones. Almost two decades ago, Kuznestove et al. isolated fibroblast-like skeletal progenitors from rodents and humans and demonstrated the significant osteogenic potential of these cells upon transplantation [101]. However, these circulating skeletal progenitors were shown to be rare in humans [102]. Other studies in nude mice have demonstrated that administering human adipose stem/progenitors (ASCs) directly on to sites of calvarial bone injuries induces the expression of osteoblast markers (including Alpl, Col1a1, Runx2) and stimulates bone formation [103,104]. Interestingly, the exogenous cells persisted only for 14–15 days and were eventually replaced by the host’s own cells.
Progenitor-like cells isolated from dental pulp, referred to as dental pulp stem cells (DPSCs), were also investigated for bone healing due to their potential for differentiating down the osteoblast lineage (in vitro) and their ability to stimulate bone formation (in vivo) [105,106,107]. In 2000, Gronthos et al., first isolated DPSCs from adult human dental pulp, and determined their in vitro and in vivo characteristics [108]. Recently, Fuji et al. demonstrated the osteogenic differentiation of DPSCs in vitro and bone regeneration in vivo in mouse calvaria defects using a derivative of helioxanthin, which is known to induce osteogenic differentiation of pre-osteoblasts and MSCs [108]. Lee et al. compared the osteogenic and bone regeneration potential of DPSCs and BM-MSCs in vitro and in vivo in a rabbit model [109]. Although, DPSCs have been used pre-clinically for repairing various tissues including cardiovascular tissue, cornea, and muscular, further clinical studies are needed to evaluate their therapeutic potential [110,111,112].
Many preclinical studies demonstrate the positive effects of a variety of exogenous and endogenous stem/progenitor cells on bone fracture repair (Table 1). On the other hand, only two ongoing US clinical trials testing the efficacy of human MSCs on bone fracture repair and osteochondral defect repair is being carried out at the time of this review (Table 2) suggesting that there is a fairly large gap that separates bench and bedside. This does not necessarily imply that pre-clinical cell-based strategies for bone repair translate poorly, but rather it is more likely that such strategies are difficult to translate into clinical approaches that comply with established regulatory standards set by the Center for Biologics Evaluation and Research (CBER) branch of the Food and Drug Administration (FDA), which take into careful consideration factors such as cell source, purity/homogeneity, and culturing conditions. Furthermore, this field would benefit greatly from an influx of clinical studies that thoroughly evaluate different factors such as donor age, sex, and the number of cells to be injected as this would improve our understanding of therapeutic applications of exogenous stem/progenitor cells for bone healing.

4. Stimulating Tendon and Ligament Repair

Tendon and ligament are the dense connective tissue that connects bone to muscle and bone to bone, respectively. Both are highly prone to injury and can be difficult to repair due to their structural organization, hypo-cellularity and hypo-vascularity [138,139,140,141]. Tendon tissue primarily consists of collagens I, II, III, V, XI, XII, XIV, elastin, and glycosaminoglycans (GAGs) [142,143,144,145]. Type I collagen is a most robust component of the tendon extracellular matrix (ECM) network [146]. Tendon cells, or tenocytes, are fibroblast-like in appearance and they synthesize ECM components including collagen fibers, elastin, proteoglycans [142,147]. The proportion of tendon/ligament ECM constituents can vary among individuals, mainly due to differences in the mechanical loading environment; however, they tend to have relatively consistent physiology and structure [148,149].
The healing response to tendon/ligament injury can be divided into three overlapping stages—(1) inflammation, (2) proliferation, and (3) remodeling [150]. These stages are ushered by specific cytokines and cellular processes. In the inflammatory phase, a blood clot is formed after tendon injury, which is followed by activation of chemoattractant inflammatory cells like neutrophils, monocytes, and lymphocytes [151]. The clot primarily serves as a scaffold to harbor the cells and releases important growth factors like TGF-β, IGF-1, PDGF, and VEGF causing inflammation [152]. The second phase, proliferative phase begins roughly after 2 days following injury. This phase is directed by macrophages and tenocytes of the endotenon and epitenon region of the tendon. Macrophages release growth factors to direct cell recruitment and tenocytes help in synthesis of a matrix, primarily consisting of type III collagen [153,154,155]. The proliferative phase is identified by increased cellularity, synthesis of ECM, and the deposition of scar tissue by fibroblasts [156]. After about 1–2 months following injury, the remodeling phase begins with reorganization of tenocytes and collagen fibers, aligning in the direction of stress [156] with a decrease in type III collagen and GAG content [157] and increased synthesis of type I collagen [158]. This process continues for months after injury; however, the newly formed tissue gradually changes to scar-like tendon tissue. The repaired tissue also lacks biomechanical, biochemical, and ultrastructural properties of native uninjured tendon tissue [159,160].
This complex microenvironment of tendon and ligament tissues makes the healing process slow [161,162]. In the US alone, tendon and ligament injuries account for almost half of the 32 million musculoskeletal injuries incurred each year [163]. These rates are rising due to the increasing aged population and also due to increased participation in sports activities [156]. Rotator cuff tears increase with age, from 9.7% in patients 20 years and younger to 62% in patients that are 80 years and older [164], which represents a significant burden [165]. The main challenge in tendon healing is the failure to functionally attach tendon to bone. The attachment, called enthesis, consists of the transitional gradient of tendon, fibrocartilage, calcified fibrocartilage and bone; and it mainly allows dissipation of stress between these tissues of different properties [166,167]. Although the enthesis can be reattached surgically, the gradient is disrupted and replaced with scar tissue exhibiting impaired mechanical properties [160,168]. As with tendon, anterior cruciate ligament (ACL) surgical repair results in failure due to the lack of blood clot formation, intra-articular hypo-vascularity, loss of intrinsic cell migration, and poor healing capacity of ACL [169,170]. There is even a high risk of failure when the ACL is repaired surgically by suturing in adolescent patients [171]. Severe tendon/ligament injuries that result in lost or unsalvageable tissue require tissue autografts, which often lead to donor site morbidity [172,173]. Ultimately, current surgical paradigms fail to restore the functional, biochemical, and structural properties of the native tissue [156].
Tenocytes and ligamentocytes (ligament cells) express Scleraxis (Scx)—a transcription factor that regulates the expression of the glycoprotein Tenomodulin, which is a specific marker of these mature cells [174,175,176]. Like other connective tissues, tendon also contains a unique population of heterogeneous tissue-specific MSCs. They are capable of differentiating into tenocytes, and true to the nature of all MSCs, they can differentiate along the chondrogenic, osteogenic, and adipogenic lineages upon in vitro induction [177]. Further, these cells are capable of generating tendon, cartilage, and tendon-bone junction-like tissues in rats [178], rabbits [117], and equine animal models [179]. Over the years, regenerative approaches for treating tendon and ligament injuries have been explored. These include: (i) cell based therapies [180,181,182] (ii) gene therapy [24,183] (iii) orthobiologics/platelet rich plasma injections (PRP) [119,120,184], and recently, (iv) cell free strategies that implement sustained released growth factors to recruit endogenous stem cell that promote healing [121]. Biochemical factors such as cytokines (IL-6, IL-10, IL-1β,TNF-α) [185,186,187] and growth factors (transforming growth factor (TGF)-β, basic fibroblast growth factor (bFGF), IGF-1, PDGF) play a crucial role in maintaining tissue homeostasis [152,188]. Many endogenous and exogenous cell-based therapies have been used to repair tendons and ligaments in pre-clinical models (Table 1).
It has been reported that tendon derived stem/progenitor cells (TDSCs) exhibited a higher regenerative potential towards ruptured Achilles tendon, compared to BM-MSCs at 4 weeks in a rat model [122]. However, due to the limited availability of these cells, they must be culture expanded. Unfortunately, in vitro expansion has been reported to cause the cells to lose their phenotypic markers [189] and studies have demonstrated that the number of TDSCs are greatly reduced with aging, showing diminished proliferative capacity [190,191]. With this in mind, other cell sources are being investigated for therapeutic use.
Bone marrow is a widely explored alternative cell source for tendon and ligament tissue engineering [123,124,125,192]. Gulotta et al. explored the use of BM-MSCs for treating unilateral detachment of the supraspinatus tendon in the rats [193]. Although there were no significant differences found between the treated and untreated groups, they showed that cells were present and metabolically active at the repair site following treatment. The same group found that cell-based strategies alone may not be sufficient to improve the structure, composition, and strength of the healing tendon tissue [193]. Two years later, they showed that administering BM-MSCs transduced with adenoviral-mediated Scleraxis improved rotator cuff repair in a rat model [194]. There seemed to be no difference in the histological appearance between the Scx and MSC group, but the Scx transduced cell treated group had more fibrocartilage, higher load-to-failure and stress-to-failure ratio at 4 weeks, compared to BM-MSC group alone. Genetic modifications of administered BM-MSCs and ACL fibroblast with bone morphogenetic factor (BMP)-12 and BMP-13 has also been demonstrated to induce ligamentogenic differentiation, in vitro [195].
ASCs are also interesting for tissue engineering due to their accessibility and great abundance when extracted from human subcutaneous adipose tissue [196,197]. Park et al. demonstrated that rat ASCs, when treated with growth differentiation factor-5 (GDF-5), exhibited enhanced ECM production and tendonogenic differentiation of cells, in vitro [198]. Multiple studies have reported the in vivo efficacy of using ASCs to stimulate tendon healing and improve biomechanical properties and normal collagen fiber organization, compared to the control groups [126,199,200]. Recently, Kokubu et al. demonstrated that ASCs improved tendon healing by stimulating collagen fiber reformation and preventing ectopic ossification of tendons in mice, compared to the control group at 2 and 4 weeks after injury [201]. This study suggested that ASCs can modulate inflammation and induce neovascularization at the site of injury. Tracking of transplanted ASCs revealed that they were present at 2 and 7 days post transplantation, but no longer present by 3 weeks, post transplantation [201].
When used in combination with fibrin sealant or hydrogels, ASC treatment has been reported improved tendon healing with increased expression of Col1, Scx, and Tenomodulin in the damaged tendon tissue. In vivo survival of ASCs injected with scaffolds (fibrin sealant or hydrogel) into the defect region was confirmed at day 14 [127] and day 31, post injury [202]. While these studies effectively demonstrate that there are several different approaches for utilizing exogenous cell treatments (with/without bioactive scaffolds or growth factors) to stimulate tendon and ligament repair [119,120,127,183,184,202], further comprehensive studies are needed to assess the safety and efficacy of these approaches to take the research from bench-to-bedside.
Endogenous cell-based approaches for tendon regeneration have also emerged as a promising strategy consisting of applying different growth factors and biomaterials to effectively recruit native stem/progenitor cell population. Solaiman et al. demonstrated a tissue engineering integrated approach, utilizing both in vitro and in vivo models, by a precise spatiotemporal growth factor delivery system embedded in 3D-printed flexible scaffolds, which resulted in regeneration of the tendon-to-bone interface by recruitment of endogenous stem/progenitor cells [121]. Immunofluorescence analysis 7 days post-implantation showed the infiltration of CD146+ cells into the growth factor embedded scaffold, suggesting an endogenous cell source does exist for tendon-to-bone healing in a rat model [121]. The regenerative potential of tissue-resident stem/progenitor cells (CD146+) in a rat model was evaluated by Lee et al., [128] These cells, after enriching with connective tissue growth factor (CTGF), were mixed with fibrin gel and delivered at the injury site. By the end of week 1 and 2, CTGF led to a dense, aligned collagen fibers, compared to the group without CTGF delivery. By post-operative week 4, CTGF generated tendon exhibited dense collagen structure compared to the scar-like tissue in the fibrin-alone group [128]. Furthermore, expression of different growth factors are evaluated in early phases of tendon healing [203,204].
Several ongoing clinical studies are trying to better understand the implications of treating injuries with MSCs alone or in combination with bioactive scaffolds to repair tendon and ligament (Table 2). Hernigou et al. showed 10-year follow up results, after injecting MSCs to have enhanced rate of healing and reduced number of re-tears over time [205]. Kim et al. revealed that an injection of ASCs in combination with fibrin sealant, significantly improved re-tear rates of rotator cuff injuries [206]. Ligament injuries like ACL tears have also shown promising results in small number of patients using autologous injection of bone marrow nucleated cells [207]. Murray et al. developed a bridge enhanced ACL repair (BEAR) which combines suture repair with an extra cellular matrix scaffold to bridge the gap between the ligament ends [27] and presented the first-in-human study to show no graft or repair failures following a two year follow up [28].

5. Stimulating Articular Cartilage Repair

Articular cartilage is a resilient yet flexible tissue which is a vital component of the skeletal system. The extracellular matrix (ECM) of articular cartilage is mainly composed of water, proteoglycans, and collagen fibers. The chondrocyte is the primary type of cell found in cartilage, which almost completely lacks blood vasculatures and neural architecture [208]. This means that the sole mode of nutrient supplements to the chondrocytes is though adjacent tissues and fluids such as subchondral bone and synovial fluid, respectively [209,210]. In comparison to other connective tissues, articular cartilage has the lowest turnover of ECM and it has a very limited capacity for healing after injury [211]. Chondrocytes reside in small compartmented cavities called lacunae that are surrounded by large areas of dense cartilage ECM making it difficult for these cells to move freely or migrate in response to biological cues of damaged tissue. Moreover, the lack of blood vasculature prevents the infiltration of articular cartilage by circulating immune cells [212]. Multiple approaches have been documented to help restore cartilage without surgical proceedings, but the fact remains that there are virtually no clinical treatment options in existence that completely restore damaged articular cartilage to its native state [213,214,215,216,217].
Given its poor capacity for healing, knee injury resulting in cartilage damage is a great clinical challenge [218,219,220,221,222]. Widely practiced clinical procedures for attempting cartilage restoration include endogenous cell-based methods like bone marrow stimulation by creating microfractures [223,224], and exogenous transplantation of tissue and cells such as the use of osteochondral grafts [225,226], and autologous chondrocyte implantation (ACI) [227,228,229,230,231,232]. ACI was first described by Brittberg and colleagues in 1994 where patients’ own cartilage tissues were harvested in one surgery to extract the chondrocytes, followed by cell culture expansion and injection into the defect site during a second surgery [231]. This treatment, along with the matrix induced autologous chondrocyte implantation (MACI) method [233], has shown positive clinical results for larger cartilage defects [234,235]. However, each method has its own limitations that have been previously discussed to great lengths [236,237,238,239,240].
With regards to novel methodologies that are still in development for cartilage repair, pre-clinical strategies fall into one (or both) of two avenues: (1) utilizing biomaterials that can replace/fill lost cartilage and restore some loadbearing capacity to defect region; (2) utilizing/stimulating cells to assist with rebuilding lost cartilage ECM [241]. Cellular approaches involve BM-MSCs and ASCs along with the induction of chondrogenesis. Chondrogenesis is thought to be more functional in a 3D culture system as chondrocytes tend to lose their original characteristics, dedifferentiate in the monolayer and acquire a fibroblastic morphology [242,243]. Studies have demonstrated the chondrogenic differentiation of BM-MSCs and ASCs in monolayer culture as well as in 3D aggregate cultures [40,45,244,245,246]. The preclinical use of stem/progenitor cells for cartilage repair has been documented in different animal models (Table 1). Ko et al. investigated the chondrogenic and hypertrophic potential of human iPSCs compared to BM-MSCs and also demonstrated the healing of damaged cartilage in rats by grafting iPSCs [130]. Additionally, Xing et al. demonstrated that a single injection of human umbilical cord derived MSCs into sprague dawley rat knee joint, 4 weeks post-surgery, resulted in a significant retardation of OA progression compared to controls [131]. Other studies have also reported that stem cells induce cartilage repair in rodents [132,133], porcine [133] as well as the equine models [134]. Although cell/tissue transplantation-based therapeutic strategies seem promising, there are drawbacks such as donor-site morbidity, the possibility of graft rejection and rapid cell ‘wash-out’ from synovial fluid [247]. Due to these limitations, scientists are actively considering the guided delivery of the biologics such as proteins, nucleic acid, and growth factors to redirect the native/endogenous cells to the site of cartilage tissue injury [248,249,250]. These strategies are intended to motivate cartilage stem/progenitor cells (CPCs) and mature chondrocytes to help accelerate healing [251].
For large cartilage defects, growth factors are used in combination with biomaterials/scaffolds that have the dual role of providing the necessary structural support while also facilitating the recruitment of endogenous cells over time [135]. Scaffold can also be laced with other bioactive molecules, like chemokines, to promote cartilage repair. Chen et al. demonstrated a silk fibroin-porous gelatin scaffold (capable of sustained release) loaded with growth factors stromal-derived factor-1α (SDF-1α) and TGF-β1 can promote in vitro MSC homing, migration and chondrogenesis and cartilage regeneration in vivo [136]. Other bioactive molecules like IGF-1 and FGF-1 encapsulated in bio-scaffolds, were reported to accelerate the repair process ex vivo and in vivo [137,252,253,254,255].
Current clinically practiced methods to reduce pain, increase joint lubrication, and subside inflammation include: intra-articular injections of platelet rich plasma (PRP), hyaluronic acid, and corticosteroids. Administering specific growth factors have also showed promise for stimulating healing [248,249]. An early phase clinical trial [26] seem to suggest that intra-articular injections of ASCs induce a degree of cartilage regeneration, as they can differentiate along the chondrogenic lineage [256,257,258]. However, ASCs maintain their chondrogenesis potential up to 15–16 passages with diminishing efficiency to differentiate into chondrocytes, comparatively less than that of BM-MSCs [246,259]. A clinical trial comparing the efficacy of using microfracture vs. application of ASCs on a collagen scaffold (NCT02090140) for articular cartilage defect repair is currently ongoing. Simultaneously, randomized phase III clinical trials are being conducted to evaluate which source of stem cells are superior (NCT03818737). In addition, ongoing is a trial comparing the outcomes in patients with osteoarthritis (OA) injected with fragmented adipose tissue (NCT03467919) in comparison to the current standard treatment of treating with corticosteroid to alleviate pain. Refer to Table 2 for more details on ongoing clinical trials.
Although studies to date suggest that the stem/progenitor cells are capable of cartilage repair, the combination approach of using cells with different growth factors and bio-scaffolds may be beneficial for future pre-clinical studies seeking to improve tissue repair efficacy. While facilitating the migration of endogenous cells to areas of injury is clinically appealing from a regulatory standpoint, aging ensures that these cells have dwindling capacity for self-renewal and healing. Unfortunately, elderly individuals make up the largest population suffering from cartilage degeneration. Taking the road less traveled by genetically modifying these cells, in order to enhance their regenerative potential, may change the therapeutic paradigm for cartilage repair.

6. Conclusions

Skeletal tissues like bone, cartilage, tendon, and ligament are difficult to repair completely upon injury due to their complex microenvironment. Surgical approaches to repair these damaged tissues are widely used but due to their limitations, cell-based approaches have emerged, and they are being tested in both pre-clinical models and in current ongoing clinical trials. The approach of stimulating recruitment of endogenous cells to the injury site is preferable and advantageous in order to avoid donor site morbidity and graft/cell rejection that is associated with use of exogenous cell-based approaches detailed here. Unfortunately, in the aging population, the existing native stem/progenitors may have diminished capacity for proliferation and repair, hence the use of exogenous cells may be preferable. An intriguing solution that should be more deeply explored in the future is to combine these approaches—by administering both exogenous cells and growth factors that will also help recruit native cells to aid in repair.

Author Contributions

S.D. and C.T.J: Conceptualization and investigation; S.D.: Composition of first draft; S.D. and C.T.J.: Revision and editing; C.T.J.: Funding acquisition; S.D. and C.T.J.: Review and final approval of manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

Jayasuriya’s research laboratory receives funding support from Award number NIH R21AR07732601 (NIAMS) and by the Rhode Island Hospital Department of Orthopaedics.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Steele, R.; Collins, E.-M. Wnt signaling determines body axis polarity in regenerating Hydra tissue. bioRxiv 2020. [Google Scholar] [CrossRef] [Green Version]
  2. Mohri, K.; Tanaka, R.; Nagano, S. Live cell imaging of cell movement and transdifferentiation during regeneration of an amputated multicellular body of the social amoeba Dictyostelium discoideum. Dev. Biol. 2020, 457, 140–149. [Google Scholar] [CrossRef]
  3. Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef] [PubMed]
  4. Galliot, B.; Crescenzi, M.; Jacinto, A.; Tajbakhsh, S. Trends in tissue repair and regeneration. Developmenmt 2017, 144, 357–364. [Google Scholar] [CrossRef] [Green Version]
  5. Stroncek, J.D.; Reichert, W.M. Overview of wound healing in different tissue types. In Indwelling Neural Implants: Strategies for Contending with the in Vivo Environment; CRC Press/Taylor & Francis: Boca Raton, FL, USA, 2007; ISBN 9781420009309. [Google Scholar] [CrossRef]
  6. Tidball, J.G. Mechanisms of muscle injury, repair, and regeneration. Compr. Physiol. 2011, 1, 2029–2062. [Google Scholar] [CrossRef] [PubMed]
  7. Vannella, K.M.; Wynn, T.A. Mechanisms of Organ Injury and Repair by Macrophages. Annu. Rev. Physiol. 2017, 79, 593–617. [Google Scholar] [CrossRef] [PubMed]
  8. Ulivi, V.; Tasso, R.; Cancedda, R.; Descalzi, F. Mesenchymal stem cell paracrine activity is modulated by platelet lysate: Induction of an inflammatory response and secretion of factors maintaining macrophages in a proinflammatory phenotype. Stem Cells Dev. 2014, 23, 1858–1869. [Google Scholar] [CrossRef]
  9. Shiu, H.T.; Goss, B.; Lutton, C.; Crawford, R.; Xiao, Y. Formation of Blood Clot on Biomaterial Implants Influences Bone Healing. Tissue Eng. Part B Rev. 2014, 20, 697–712. [Google Scholar] [CrossRef] [PubMed]
  10. Hoogduijn, M.J.; Lombardo, E. Mesenchymal Stromal Cells Anno 2019: Dawn of the Therapeutic Era? Concise Review. Stem Cells Transl. Med. 2019, 8, 1126–1134. [Google Scholar] [CrossRef] [Green Version]
  11. Linero, I.; Chaparro, O. Paracrine effect of mesenchymal stem cells derived from human adipose tissue in bone regeneration. PLoS ONE 2014, 9, e107001. [Google Scholar] [CrossRef] [Green Version]
  12. Chen, L.; Tredget, E.E.; Wu, P.Y.G.; Wu, Y.; Wu, Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008, 3, e1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Daneshmandi, L.; Shah, S.; Jafari, T.; Bhattacharjee, M.; Momah, D.; Saveh-Shemshaki, N.; Lo, K.W.H.; Laurencin, C.T. Emergence of the Stem Cell Secretome in Regenerative Engineering. Trends Biotechnol. 2020. [Google Scholar] [CrossRef] [PubMed]
  14. Jayasuriya, C.T.; Chen, Y.; Liu, W.; Chen, Q. The influence of tissue microenvironment on stem cell–based cartilage repair. Ann. N. Y. Acad. Sci. 2016, 1383, 21–33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Embree, M.C.; Chen, M.; Pylawka, S.; Kong, D.; Iwaoka, G.M.; Kalajzic, I.; Yao, H.; Shi, C.; Sun, D.; Sheu, T.J.; et al. Exploiting endogenous fibrocartilage stem cells to regenerate cartilage and repair joint injury. Nat. Commun. 2016, 7, 13073. [Google Scholar] [CrossRef] [Green Version]
  16. Ozeki, N.; Muneta, T.; Koga, H.; Nakagawa, Y.; Mizuno, M.; Tsuji, K.; Mabuchi, Y.; Akazawa, C.; Kobayashi, E.; Matsumoto, K.; et al. Not single but periodic injections of synovial mesenchymal stem cells maintain viable cells in knees and inhibit osteoarthritis progression in rats. Osteoarthr. Cartil. 2016, 24, 1061–1070. [Google Scholar] [CrossRef] [Green Version]
  17. Meeson, R.; Sanghani-Keri, A.; Coathup, M.; Blunn, G. VEGF with AMD3100 endogenously mobilizes mesenchymal stem cells and improves fracture healing. J. Orthop. Res. 2019, 37, 1294–1302. [Google Scholar] [CrossRef]
  18. Devana, S.K.; Kelley, B.V.; McBride, O.J.; Kabir, N.; Jensen, A.R.; Park, S.J.; Eliasberg, C.D.; Dar, A.; Mosich, G.M.; Kowalski, T.J.; et al. Adipose-derived human perivascular stem cells may improve achilles tendon healing in rats. Clin. Orthop. Relat. Res. 2018, 476, 2091–2100. [Google Scholar] [CrossRef]
  19. Black, C.; Kanczler, J.M.; de Andrés, M.C.; White, L.J.; Savi, F.M.; Bas, O.; Saifzadeh, S.; Henkel, J.; Zannettino, A.; Gronthos, S.; et al. Characterisation and evaluation of the regenerative capacity of Stro-4+ enriched bone marrow mesenchymal stromal cells using bovine extracellular matrix hydrogel and a novel biocompatible melt electro-written medical-grade polycaprolactone scaffold. Biomaterials 2020, 247, 119998. [Google Scholar] [CrossRef]
  20. Loebel, C.; Burdick, J.A. Engineering Stem and Stromal Cell Therapies for Musculoskeletal Tissue Repair. Cell Stem Cell 2018, 22, 325–339. [Google Scholar] [CrossRef] [Green Version]
  21. De Bari, C.; Roelofs, A.J. Stem cell-based therapeutic strategies for cartilage defects and osteoarthritis. Curr. Opin. Pharmacol. 2018, 40, 74–80. [Google Scholar] [CrossRef]
  22. Zhang, R.; Ma, J.; Han, J.; Zhang, W.; Ma, J. Mesenchymal stem cell related therapies for cartilage lesions and osteoarthritis. Am. J. Transl. Res. 2019, 11, 6275–6289. [Google Scholar] [PubMed]
  23. Scarfì, S. Use of bone morphogenetic proteins in mesenchymal stem cell stimulation of cartilage and bone repair. World J. Stem Cells 2016, 8, 1–12. [Google Scholar] [CrossRef] [PubMed]
  24. Docheva, D.; Müller, S.A.; Majewski, M.; Evans, C.H. Biologics for tendon repair. Adv. Drug Deliv. Rev. 2015, 84, 222–239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Steinert, A.F.; Rackwitz, L.; Gilbert, F.; Nöth, U.; Tuan, R.S. Concise Review: The Clinical Application of Mesenchymal Stem Cells for Musculoskeletal Regeneration: Current Status and Perspectives. Stem Cells Transl. Med. 2012, 1, 237–247. [Google Scholar] [CrossRef]
  26. Jo, C.H.; Lee, Y.G.; Shin, W.H.; Kim, H.; Chai, J.W.; Jeong, E.C.; Kim, J.E.; Shim, H.; Shin, J.S.; Shin, I.S.; et al. Intra-articular injection of mesenchymal stem cells for the treatment of osteoarthritis of the knee: A proof-of-concept clinical trial. Stem Cells 2014, 32, 1254–1266. [Google Scholar] [CrossRef]
  27. Murray, M.M.; Flutie, B.M.; Kalish, L.A.; Ecklund, K.; Fleming, B.C.; Proffen, B.L.; Micheli, L.J. The Bridge-Enhanced Anterior Cruciate Ligament Repair (BEAR) Procedure: An Early Feasibility Cohort Study. Orthop. J. Sport. Med. 2016, 4. [Google Scholar] [CrossRef] [Green Version]
  28. Murray, M.M.; Kalish, L.A.; Fleming, B.C.; Flutie, B.; Freiberger, C.; Henderson, R.N.; Perrone, G.S.; Thurber, L.G.; Proffen, B.L.; Ecklund, K.; et al. Bridge-Enhanced Anterior Cruciate Ligament Repair: Two-Year Results of a First-in-Human Study. Orthop. J. Sport. Med. 2019, 7. [Google Scholar] [CrossRef]
  29. Hurd, J.L.; Facile, T.R.; Weiss, J.; Hayes, M.; Hayes, M.; Furia, J.P.; Maffulli, N.; Winnier, G.E.; Alt, C.; Schmitz, C.; et al. Safety and efficacy of treating symptomatic, partial-thickness rotator cuff tears with fresh, uncultured, unmodified, autologous adipose-derived regenerative cells (UA-ADRCs) isolated at the point of care: A prospective, randomized, controlled first-in-human pilot study. J. Orthop. Surg. Res. 2020, 15, 122–128. [Google Scholar] [CrossRef] [Green Version]
  30. Bruder, S.P.; Fink, D.J.; Caplan, A.I. Mesenchymal stem cells in bone development, bone repair, and skeletal regenaration therapy. J. Cell. Biochem. 1994, 56, 283–294. [Google Scholar] [CrossRef]
  31. Caplan, A.I. Mesenchymal stem cells: Cell-based reconstructive therapy in orthopedics. Tissue Eng. 2005, 11, 1198–1211. [Google Scholar] [CrossRef] [Green Version]
  32. Nöth, U.; Osyczka, A.M.; Tuli, R.; Hickok, N.J.; Danielson, K.G.; Tuan, R.S. Multilineage mesenchymal differentiation potential of human trabecular bone-derived cells. J. Orthop. Res. 2002, 20, 1060–1069. [Google Scholar] [CrossRef]
  33. Colnot, C. Skeletal cell fate decisions within periosteum and bone marrow during bone regeneration. J. Bone Miner. Res. 2009, 24, 274–282. [Google Scholar] [CrossRef] [PubMed]
  34. Owston, H.E.; Ganguly, P.; Tronci, G.; Russell, S.J.; Giannoudis, P.V.; Jones, E.A. Colony Formation, Migratory, and Differentiation Characteristics of Multipotential Stromal Cells (MSCs) From “Clinically Accessible” Human Periosteum Compared to Donor-Matched Bone Marrow MSCs. Stem Cells Int. 2019, 2019, 6074245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Williams, R.; Khan, I.M.; Richardson, K.; Nelson, L.; McCarthy, H.E.; Analbelsi, T.; Singhrao, S.K.; Dowthwaite, G.P.; Jones, R.E.; Baird, D.M.; et al. Identification and clonal characterisation of a progenitor cell sub-population in normal human articular cartilage. PLoS ONE 2010, 5, e13246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Candela, M.E.; Yasuhara, R.; Iwamoto, M.; Enomoto-Iwamoto, M. Resident mesenchymal progenitors of articular cartilage. Matrix Biol. 2014, 39, 44–49. [Google Scholar] [CrossRef] [PubMed]
  37. Jayasuriya, C.T.; Twomey-Kozak, J.; Newberry, J.; Desai, S.; Feltman, P.; Franco, J.R.; Li, N.; Terek, R.; Ehrlich, M.G.; Owens, B.D. Human Cartilage-Derived Progenitors Resist Terminal Differentiation and Require CXCR4 Activation to Successfully Bridge Meniscus Tissue Tears. Stem Cells 2019, 37, 102–114. [Google Scholar] [CrossRef] [Green Version]
  38. Ding, Z.; Huang, H. Mesenchymal stem cells in rabbit meniscus and bone marrow exhibit a similar feature but a heterogeneous multi-differentiation potential: Superiority of meniscus as a cell source for meniscus repair Evolutionary developmental biology and morphology. BMC Musculoskelet. Disord. 2015, 16, 65. [Google Scholar] [CrossRef] [Green Version]
  39. Liu, S.; Liang, H.; Lee, S.M.; Li, Z.; Zhang, J.; Fei, Q. Isolation and Identification of Stem Cells From Degenerated Human Intervertebral Discs and Their Migration Characteristics. Acta Biochim. Biophys. Sin. 2017, 49, 101–109. [Google Scholar] [CrossRef]
  40. Zuk, P.A.; Zhu, M.; Mizuno, H.; Huang, J.; Futrell, J.W.; Katz, A.J.; Benhaim, P.; Lorenz, H.P.; Hedrick, M.H. Multilineage cells from human adipose tissue: Implications for cell-based therapies. Tissue Eng. 2001, 7, 211–228. [Google Scholar] [CrossRef] [Green Version]
  41. Jo, C.H.; Lim, H.J.; Yoon, K.S. Characterization of Tendon-Specific Markers in Various Human Tissues, Tenocytes and Mesenchymal Stem Cells. Tissue Eng. Regen. Med. 2019, 16, 151–159. [Google Scholar] [CrossRef]
  42. Lee, K.J.; Clegg, P.D.; Comerford, E.J.; Canty-Laird, E.G. Ligament-Derived Stem Cells: Identification, Characterisation, and Therapeutic Application. Stem Cells Int. 2017, 2017, 1919845. [Google Scholar] [CrossRef]
  43. Ogata, Y.; Mabuchi, Y.; Shinoda, K.; Horiike, Y.; Mizuno, M.; Otabe, K.; Suto, E.G.; Suzuki, N.; Sekiya, I.; Akazawa, C. Anterior cruciate ligament-derived mesenchymal stromal cells have a propensity to differentiate into the ligament lineage. Regen. Ther. 2018, 8, 20–28. [Google Scholar] [CrossRef] [PubMed]
  44. Seo, B.M.; Miura, M.; Gronthos, S.; Bartold, P.M.; Batouli, S.; Brahim, J.; Young, M.; Robey, P.G.; Wang, C.Y.; Shi, S. Investigation of multipotent postnatal stem cells from human periodontal ligament. Lancet 2004, 364, 149–155. [Google Scholar] [CrossRef]
  45. De Bari, C.; Dell’Accio, F.; Tylzanowski, P.; Luyten, F.P. Multipotent mesenchymal stem cells from adult human synovial membrane. Arthritis Rheum. 2001, 44, 1928–1942. [Google Scholar] [CrossRef]
  46. Liu, Z.-J.; Zhuge, Y.; Velazquez, O.C. Trafficking and differentiation of mesenchymal stem cells. J. Cell. Biochem. 2009, 106, 984–991. [Google Scholar] [CrossRef]
  47. Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.C.; Krause, D.S.; Deans, R.J.; Keating, A.; Prockop, D.J.; Horwitz, E.M. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
  48. Kachroo, U.; Vinod, E. Comparative analysis of gene expression between articular cartilage-derived cells to assess suitability of fibronectin adhesion assay to enrich chondroprogenitors. Knee 2020, 27, 755–759. [Google Scholar] [CrossRef]
  49. Grogan, S.P.; Duffy, S.F.; Pauli, C.; Lotz, M.K.; D’Lima, D.D. Gene expression profiles of the meniscus avascular phenotype: A guide for meniscus tissue engineering. J. Orthop. Res. 2018, 36, 1947–1958. [Google Scholar] [CrossRef]
  50. Iaquinta, M.R.; Mazzoni, E.; Bononi, I.; Rotondo, J.C.; Mazziotta, C.; Montesi, M.; Sprio, S.; Tampieri, A.; Tognon, M.; Martini, F. Adult Stem Cells for Bone Regeneration and Repair. Front. Cell Dev. Biol. 2019, 7. [Google Scholar] [CrossRef] [Green Version]
  51. Liang, Y.; Idrees, E.; Andrews, S.H.J.; Labib, K.; Szojka, A.; Kunze, M.; Burbank, A.D.; Mulet-Sierra, A.; Jomha, N.M.; Adesida, A.B. Plasticity of Human Meniscus Fibrochondrocytes: A Study on Effects of Mitotic Divisions and Oxygen Tension. Sci. Rep. 2017, 7, 12148. [Google Scholar] [CrossRef] [Green Version]
  52. Angelozzi, M.; Penolazzi, L.; Mazzitelli, S.; Lambertini, E.; Lolli, A.; Piva, R.; Nastruzzi, C. Dedifferentiated chondrocytes in composite microfibers as tool for cartilage repair. Front. Bioeng. Biotechnol. 2017, 5, 35. [Google Scholar] [CrossRef] [Green Version]
  53. Lin, L.; Zhou, C.; Wei, X.; Hou, Y.; Zhao, L.; Fu, X.; Zhang, J.; Yu, C. Articular cartilage repair using dedifferentiated articular chondrocytes and bone morphogenetic protein 4 in a rabbit model of articular cartilage defects. Arthritis Rheum. 2008, 58, 1067–1075. [Google Scholar] [CrossRef]
  54. Caplan, A.I. Adult mesenchymal stem cells for tissue engineering versus regenerative medicine. J. Cell. Physiol. 2007, 213, 341–347. [Google Scholar] [CrossRef] [PubMed]
  55. Li, H.; Shen, S.; Fu, H.; Wang, Z.; Li, X.; Sui, X.; Yuan, M.; Liu, S.; Wang, G.; Guo, Q. Immunomodulatory functions of mesenchymal stem cells in tissue engineering. Stem Cells Int. 2019, 2019, 9671206. [Google Scholar] [CrossRef] [Green Version]
  56. Takahashi, K.; Yamanaka, S. Induction of Pluripotent Stem Cells from Mouse Embryonic and Adult Fibroblast Cultures by Defined Factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Chan, C.K.F.; Gulati, G.S.; Sinha, R.; Tompkins, J.V.; Lopez, M.; Carter, A.C.; Ransom, R.C.; Reinisch, A.; Wearda, T.; Murphy, M.; et al. Identification of the Human Skeletal Stem Cell. Cell 2018, 175, 43–56.e21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Liang, G.; Zhang, Y. Genetic and epigenetic variations in iPSCs: Potential causes and implications for application. Cell Stem Cell 2013, 13, 149–159. [Google Scholar] [CrossRef] [Green Version]
  59. Arvidson, K.; Abdallah, B.M.; Applegate, L.A.; Baldini, N.; Cenni, E.; Gomez-Barrena, E.; Granchi, D.; Kassem, M.; Konttinen, Y.T.; Mustafa, K.; et al. Bone regeneration and stem cells. J. Cell. Mol. Med. 2011, 15, 718–746. [Google Scholar] [CrossRef] [PubMed]
  60. Bussard, K.M.; Gay, C.V.; Mastro, A.M. The bone microenvironment in metastasis; what is special about bone? Cancer Metastasis Rev. 2008, 27, 41–55. [Google Scholar] [CrossRef] [PubMed]
  61. Einhorn, T.A.; Gerstenfeld, L.C. Fracture healing: Mechanisms and interventions. Nat. Rev. Rheumatol. 2015, 11, 45–54. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Phillips, A.M. Overview of the fracture healing cascade. Injury 2005, 36 (Suppl. 3), S5–S7. [Google Scholar] [CrossRef] [PubMed]
  63. Bragdon, B.C.; Bahney, C.S. Origin of Reparative Stem Cells in Fracture Healing. Curr. Osteoporos. Rep. 2018, 16, 490–503. [Google Scholar] [CrossRef] [PubMed]
  64. Einhorn, T.A.; Majeska, R.J.; Rush, E.B.; Levine, P.M.; Horowitz, M.C. The expression of cytokine activity by fracture callus. J. Bone Miner. Res. 1995, 10, 1272–1281. [Google Scholar] [CrossRef] [PubMed]
  65. Gerstenfeld, L.C.; Cho, T.J.; Kon, T.; Aizawa, T.; Tsay, A.; Fitch, J.; Barnes, G.L.; Graves, D.T.; Einhorn, T.A. Impaired fracture healing in the absence of TNF-α signaling: The role of TNF-α in endochondral cartilage resorption. J. Bone Miner. Res. 2003, 18, 1584–1592. [Google Scholar] [CrossRef] [PubMed]
  66. Tsiridis, E.; Upadhyay, N.; Giannoudis, P. Molecular aspects of fracture healing:Which are the important molecules? Injury 2007, 38 (Suppl. 1), S11–S25. [Google Scholar] [CrossRef]
  67. Deckers, M.M.L.; Van Bezooijen, R.L.; Van Geertje Horst, D.E.R.; Hoogendam, J.; Van Chris Bent, D.E.R.; Papapoulos, S.E.; Löwik, C.W.G.M. Bone morphogenetic proteins stimulate angiogenesis through osteoblast-derived vascular endothelial growth factor A. Endocrinology 2002, 143, 1545–1553. [Google Scholar] [CrossRef]
  68. Keating, J.F.; Simpson, A.H.; Robinson, C.M. The management of fractures with bone loss. J. Bone Jt. Surg. 2005, 87, 142–150. [Google Scholar] [CrossRef] [Green Version]
  69. Hak, D.J.; Fitzpatrick, D.; Bishop, J.A.; Marsh, J.L.; Tilp, S.; Schnettler, R.; Simpson, H.; Alt, V. Delayed union and nonunions: Epidemiology, clinical issues, and financial aspects. Injury 2014, 45, S3–S7. [Google Scholar] [CrossRef]
  70. Marsh, D. Concepts of fracture union, delayed union, and nonunion. Clin. Orthop. Relat. Res. 1998, 355, S22–S30. [Google Scholar] [CrossRef]
  71. Roberts, T.T.; Rosenbaum, A.J. Bone grafts, bone substitutes and orthobiologics the bridge between basic science and clinical advancements in fracture healing. Organogenesis 2012, 8, 114–124. [Google Scholar] [CrossRef] [Green Version]
  72. Younger, E.M.; Chapman, M.W. Morbidity at bone graft donor sites. J. Orthop. Trauma 1989, 3, 192–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Aro, H.T.; Aho, A.J. Clinical use of bone allografts. Ann. Med. 1993, 25, 403–412. [Google Scholar] [CrossRef] [PubMed]
  74. Fernández, R.F.; Bucchi, C.; Navarro, P.; Beltrán, V.; Borie, E. Bone grafts utilized in dentistry: An analysis of patients’ preferences. BMC Med. Ethics 2015, 16, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Offner, D.; de Grado, G.F.; Meisels, I.; Pijnenburg, L.; Fioretti, F.; Benkirane-Jessel, N.; Musset, A.-M. Bone Grafts, Bone Substitutes and Regenerative Medicine Acceptance for the Management of Bone Defects Among French Population: Issues about Ethics, Religion or Fear? Cell Med. 2019, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Gerstenfeld, L.C.; Shapiro, F.D. Expression of bone-specific genes by hypertrophic chondrocytes: Implications of the complex functions of the hypertrophic chondrocyte during endochondral bone development. J. Cell. Biochem. 1996, 62. [Google Scholar] [CrossRef]
  77. Gerstenfeld, L.C.; Cruceta, J.; Shea, C.M.; Sampath, K.; Barnes, G.L.; Einhorn, T.A. Chondrocytes Provide Morphogenic Signals That Selectively Induce Osteogenic Differentiation of Mesenchymal Stem Cells. J. Bone Miner. Res. 2002, 17, 221–230. [Google Scholar] [CrossRef]
  78. Gerstenfeld, L.C.; Barnes, G.L.; Shea, C.M.; Einhorn, T.A. Osteogenic differentiation is selectively promoted by morphogenetic signals from chondrocytes and synergized by a nutrient rich growth environment. Connect. Tissue Res. 2003, 44, 85–91. [Google Scholar] [CrossRef]
  79. Bahney, C.S.; Zondervan, R.L.; Allison, P.; Theologis, A.; Ashley, J.W.; Ahn, J.; Miclau, T.; Marcucio, R.S.; Hankenson, K.D. Cellular biology of fracture healing. J. Orthop. Res. 2019, 37, 35–50. [Google Scholar] [CrossRef] [Green Version]
  80. Liu, D.; Qin, H.; Yang, J.; Yang, L.; He, S.; Chen, S.; Bao, Q.; Zhao, Y.; Zong, Z. Different effects of Wnt/β-catenin activation and PTH activation in adult and aged male mice metaphyseal fracture healing. BMC Musculoskelet. Disord. 2020, 21, 110–113. [Google Scholar] [CrossRef] [Green Version]
  81. Sandberg, O.; Bernhardsson, M.; Aspenberg, P. Earlier effect of alendronate in mouse metaphyseal versus diaphyseal bone healing. J. Orthop. Res. 2017, 35, 793–799. [Google Scholar] [CrossRef] [Green Version]
  82. Bernhardsson, M.; Aspenberg, P. Abaloparatide versus teriparatide: A head to head comparison of effects on fracture healing in mouse models. Acta Orthop. 2018, 89, 674–677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Bjørgul, K.; Reigstad, A. Atypical fracture of the ulna associated with alendronate use. Acta Orthop. 2011, 82, 761–763. [Google Scholar] [CrossRef] [PubMed]
  84. Odvina, C.V.; Zerwekh, J.E.; Rao, D.S.; Maalouf, N.; Gottschalk, F.A.; Pak, C.Y.C. Severely suppressed bone turnover: A potential complication of alendronate therapy. J. Clin. Endocrinol. Metab. 2005, 90, 1294–1301. [Google Scholar] [CrossRef] [PubMed]
  85. Ang, B.F.H.; Koh, J.S.B.; Ng, A.C.M.; Howe, T.S. Bilateral ulna fractures associated with bisphosphonate therapy. Osteoporos. Int. 2013, 24, 1523–1525. [Google Scholar] [CrossRef]
  86. Kumar, S.; Ponnazhagan, S. Mobilization of bone marrow mesenchymal stem cells in vivo augments bone healing in a mouse model of segmental bone defect. Bone 2012, 50, 1012–1018. [Google Scholar] [CrossRef] [Green Version]
  87. Mehrotra, M.; Williams, C.R.; Ogawa, M.; LaRue, A.C. Hematopoietic stem cells give rise to osteo-chondrogenic cells. Blood Cells Mol. Dis. 2013, 50, 41–49. [Google Scholar] [CrossRef] [Green Version]
  88. Murao, H.; Yamamoto, K.; Matsuda, S.; Akiyama, H. Periosteal cells are a major source of soft callus in bone fracture. J. Bone Miner. Metab. 2013, 31, 390–398. [Google Scholar] [CrossRef] [Green Version]
  89. Hadjiargyrou, M.; O’Keefe, R.J. The convergence of fracture repair and stem cells: Interplay of genes, aging, environmental factors and disease. J. Bone Miner. Res. 2014, 29, 2307–2322. [Google Scholar] [CrossRef]
  90. Palombella, S.; Lopa, S.; Gianola, S.; Zagra, L.; Moretti, M.; Lovati, A.B. Bone Marrow-Derived Cell Therapies to Heal Long-Bone Nonunions: A Systematic Review and Meta-Analysis-Which Is the Best Available Treatment? Stem Cells Int. 2019, 2019, 3715964. [Google Scholar] [CrossRef]
  91. Friedenstein, A.J.; Petrakova, K.V.; Kurolesova, A.I.; Frolova, G.P. Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 1968, 6, 230. [Google Scholar] [CrossRef]
  92. Friedenstein, A.J.; Piatetzky-Shapiro, I.I.; Petrakova, K.V. Osteogenesis in transplants of bone marrow cells. J. Embryol. Exp. Morphol. 1966, 16, 381–390. [Google Scholar] [PubMed]
  93. Takagi, K.; Urist, M.R. The role of bone marrow in bone morphogenetic protein-induced repair of femoral massive diaphyseal defects. Clin. Orthop. Relat. Res. 1982, 171, 224–231. [Google Scholar] [CrossRef]
  94. Kadiyala, S. Culture expanded canine mesenchymal stem cells possess osteochondrogenic potential in vivo and in vitro. Cell Transplant. 1997, 6, 125–134. [Google Scholar] [CrossRef] [PubMed]
  95. Kumar, S.; Chanda, D.; Ponnazhagan, S. Therapeutic potential of genetically modified mesenchymal stem cells. Gene Ther. 2008, 15, 711–715. [Google Scholar] [CrossRef] [PubMed]
  96. Garg, N.K.; Gaur, S.; Sharma, S. Percutaneous autogenous bone marrow grafting in 20 cases of ununited fracture. Acta Orthop. 1993, 64, 671–672. [Google Scholar] [CrossRef] [Green Version]
  97. Sim, R.; Liang, T.S.; Tay, B.K. Autologous marrow injection in the treatment of delayed and non-union in long bones. Singapore Med. J. 1993, 34, 412–417. [Google Scholar]
  98. Kim, S.J.; Shin, Y.W.; Yang, K.H.; Kim, S.B.; Yoo, M.J.; Han, S.K.; Im, S.A.; Won, Y.D.; Sung, Y.B.; Jeon, T.S.; et al. A multi-center, randomized, clinical study to compare the effect and safety of autologous cultured osteoblast(OssronTM) injection to treat fractures. BMC Musculoskelet. Disord. 2009, 10, 20. [Google Scholar] [CrossRef] [Green Version]
  99. Mareschi, K.; Ferrero, I.; Rustichelli, D.; Aschero, S.; Gammaitoni, L.; Aglietta, M.; Madon, E.; Fagioli, F. Expansion of mesenchymal stem cells isolated from pediatric and adult donor bone marrow. J. Cell. Biochem. 2006, 97, 744–754. [Google Scholar] [CrossRef] [Green Version]
  100. Both, S.K.; Van Apeldoorn, A.A.; Jukes, J.M.; Englund, M.C.O.; Hyllner, J.; Van Blitterswijk, C.A.; De Boer, J. Differential bone-forming capacity of osteogenic cells from either embryonic stem cells or bone marrow-derived mesenchymal stem cells. J. Tissue Eng. Regen. Med. 2011, 5, 180–190. [Google Scholar] [CrossRef]
  101. Kuznetsov, S.A.; Mankani, M.H.; Gronthos, S.; Satomura, K.; Bianco, P.; Robey, P.G. Circulating skeletal stem cells. J. Cell Biol. 2001, 153, 1133–1140. [Google Scholar] [CrossRef] [Green Version]
  102. Kuznetsov, S.A.; Mankani, M.H.; Leet, A.I.; Ziran, N.; Gronthos, S.; Robey, P.G. Circulating Connective Tissue Precursors: Extreme Rarity in Humans and Chondrogenic Potential in Guinea Pigs. Stem Cells 2007, 25, 1830–1839. [Google Scholar] [CrossRef]
  103. Levi, B.; James, A.W.; Nelson, E.R.; Vistnes, D.; Wu, B.; Lee, M.; Gupta, A.; Longaker, M.T. Human adipose derived stromal cells heal critical size mouse calvarial defects. PLoS ONE 2010, 5, e11177. [Google Scholar] [CrossRef] [PubMed]
  104. Cowan, C.M.; Shi, Y.Y.; Aalami, O.O.; Chou, Y.F.; Mari, C.; Thomas, R.; Quarto, N.; Contag, C.H.; Wu, B.; Longaker, M.T. Adipose-derived adult stromal cells heal critical-size mouse calvarial defects. Nat. Biotechnol. 2004, 22, 560–567. [Google Scholar] [CrossRef] [PubMed]
  105. Kerkis, I.; Kerkis, A.; Dozortsev, D.; Stukart-Parsons, G.C.; Gomes Massironi, S.M.; Pereira, L.V.; Caplan, A.I.; Cerruti, H.F. Isolation and characterization of a population of immature dental pulp stem cells expressing OCT-4 and other embryonic stem cell markers. Cells Tissues Organs 2007, 184, 105–116. [Google Scholar] [CrossRef] [PubMed]
  106. Miura, M.; Gronthos, S.; Zhao, M.; Lu, B.; Fisher, L.W.; Robey, P.G.; Shi, S. SHED: Stem cells from human exfoliated deciduous teeth. Proc. Natl. Acad. Sci. USA 2003, 100, 5807–5812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. D’Aquino, R.; Papaccio, G.; Laino, G.; Graziano, A. Dental pulp stem cells: A promising tool for bone regeneration. Stem Cell Rev. 2008, 4, 21–26. [Google Scholar] [CrossRef]
  108. Gronthos, S.; Mankani, M.; Brahim, J.; Robey, P.G.; Shi, S. Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc. Natl. Acad. Sci. USA 2000, 97, 13625–13630. [Google Scholar] [CrossRef] [Green Version]
  109. Lee, Y.C.; Chan, Y.H.; Hsieh, S.C.; Lew, W.Z.; Feng, S.W. Comparing the osteogenic potentials and bone regeneration capacities of bone marrow and dental pulp mesenchymal stem cells in a rabbit calvarial bone defect model. Int. J. Mol. Sci. 2019, 20, 5015. [Google Scholar] [CrossRef] [Green Version]
  110. Gandia, C.; Armiñan, A.; García-Verdugo, J.M.; Lledó, E.; Ruiz, A.; Miñana, M.D.; Sanchez-Torrijos, J.; Payá, R.; Mirabet, V.; Carbonell-Uberos, F.; et al. Human Dental Pulp Stem Cells Improve Left Ventricular Function, Induce Angiogenesis, and Reduce Infarct Size in Rats with Acute Myocardial Infarction. Stem Cells 2008, 26, 638–645. [Google Scholar] [CrossRef] [Green Version]
  111. Kerkis, I.; Ambrosio, C.E.; Kerkis, A.; Martins, D.S.; Zucconi, E.; Fonseca, S.A.S.; Cabral, R.M.; Maranduba, C.M.C.; Gaiad, T.P.; Morini, A.C.; et al. Early transplantation of human immature dental pulp stem cells from baby teeth to golden retriever muscular dystrophy (GRMD) dogs: Local or systemic? J. Transl. Med. 2008, 6, 35. [Google Scholar] [CrossRef] [Green Version]
  112. Yamada, Y.; Nakamura-Yamada, S.; Kusano, K.; Baba, S. Clinical potential and current progress of dental pulp stem cells for various systemic diseases in regenerative medicine: A concise review. Int. J. Mol. Sci. 2019, 20, 1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Huang, S.; Xu, L.; Zhang, Y.; Sun, Y.; Li, G. Systemic and local administration of allogeneic bone marrow-derived mesenchymal stem cells promotes fracture healing in rats. Cell Transplant. 2015, 24, 2643–2655. [Google Scholar] [CrossRef] [Green Version]
  114. Wang, X.; Wang, C.; Gou, W.; Xu, X.; Wang, Y.; Wang, A.; Xu, W.; Guo, Q.; Liu, S.; Lu, Q.; et al. The optimal time to inject bone mesenchymal stem cells for fracture healing in a murine model 11 Medical and Health Sciences 1103 Clinical Sciences. Stem Cell Res. Ther. 2018, 9, 272. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, G.; Fang, T.; Qi, Y.; Yin, X.; Di, T.; Feng, G.; Lei, Z.; Zhang, Y.; Huang, Z. Combined use of mesenchymal stromal cell sheet transplantation and local injection of SDF-1 for bone repair in a rat nonunion model. Cell Transplant. 2016, 25, 1801–1817. [Google Scholar] [CrossRef] [PubMed]
  116. Guan, M.; Yao, W.; Liu, R.; Lam, K.S.; Nolta, J.; Jia, J.; Panganiban, B.; Meng, L.; Zhou, P.; Shahnazari, M.; et al. Directing mesenchymal stem cells to bone to augment bone formation and increase bone mass. Nat. Med. 2012, 18, 456–462. [Google Scholar] [CrossRef] [Green Version]
  117. Zhang, J.; Wang, J.H.C. Characterization of differential properties of rabbit tendon stem cells and tenocytes. BMC Musculoskelet. Disord. 2010, 11, 10. [Google Scholar] [CrossRef] [Green Version]
  118. Huang, T.F.; Yew, T.L.; Chiang, E.R.; Ma, H.L.; Hsu, C.Y.; Hsu, S.H.; Hsu, Y.T.; Hung, S.C. Mesenchymal stem cells from a hypoxic culture improve and engraft achilles tendon repair. Am. J. Sports Med. 2013, 41, 1117–1125. [Google Scholar] [CrossRef]
  119. Murray, M.M.; Spindler, K.P.; Devin, C.; Snyder, B.S.; Muller, J.; Takahashi, M.; Ballard, P.; Nanney, L.B.; Zurakowski, D. Use of a collagen-platelet rich plasma scaffold to stimulate healing of a central defect in the canine ACL. J. Orthop. Res. 2006, 24, 820–830. [Google Scholar] [CrossRef]
  120. Murray, M.M.; Spindler, K.P.; Abreu, E.; Muller, J.A.; Nedder, A.; Kelly, M.; Frino, J.; Zurakowski, D.; Valenza, M.; Snyder, B.D.; et al. Collagen-platelet rich plasma hydrogel enhances primary repair of the porcine anterior cruciate ligament. J. Orthop. Res. 2007, 25, 81–91. [Google Scholar] [CrossRef]
  121. Tarafder, S.; Brito, J.A.; Minhas, S.; Effiong, L.; Thomopoulos, S.; Lee, C.H. In situ tissue engineering of the tendon-to-bone interface by endogenous stem/progenitor cells. Biofabrication 2019, 12, 015008. [Google Scholar] [CrossRef]
  122. Al-Ani, M.K.; Xu, K.; Sun, Y.; Pan, L.; Xu, Z.; Yang, L. Study of Bone Marrow Mesenchymal and Tendon-Derived Stem Cells Transplantation on the Regenerating Effect of Achilles Tendon Ruptures in Rats. Stem Cells Int. 2015, 2015, 984146. [Google Scholar] [CrossRef] [PubMed]
  123. Awad, H.A.; Butler, D.L.; Boivin, G.P.; Smith, F.N.L.; Malaviya, P.; Huibregtse, B.; Caplan, A.I. Autologous mesenchymal stem cell-mediated repair of tendon. Tissue Eng. 1999, 5, 267–277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fan, H.; Liu, H.; Wong, E.J.W.; Toh, S.L.; Goh, J.C.H. In vivo study of anterior cruciate ligament regeneration using mesenchymal stem cells and silk scaffold. Biomaterials 2008, 29, 3324–3337. [Google Scholar] [CrossRef] [PubMed]
  125. Han, L.; Fang, W.L.; Jin, B.; Xu, S.C.; Zheng, X.; Hu, Y.G. Enhancement of tendon-bone healing after rotator cuff injuries using combined therapy with mesenchymal stem cells and platelet rich plasma. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 9075–9084. [Google Scholar] [PubMed]
  126. Lee, S.Y.; Kwon, B.; Lee, K.; Son, Y.H.; Chung, S.G. Therapeutic Mechanisms of Human Adipose-Derived Mesenchymal Stem Cells in a Rat Tendon Injury Model. Am. J. Sports Med. 2017, 45, 1429–1439. [Google Scholar] [CrossRef] [PubMed]
  127. Frauz, K.; Teodoro, L.F.; Carneiro, G.; da Veiga, F.C.; Ferrucci, D.L.; Bombeiro, A.L.; Simões, P.W.; Alvares, L.E.; de Oliveira, L.R.; Vicente, C.P.; et al. Transected Tendon Treated with a New Fibrin Sealant Alone or Associated with Adipose-Derived Stem Cells. Cells 2019, 8, 56. [Google Scholar] [CrossRef] [Green Version]
  128. Lee, C.H.; Lee, F.Y.; Tarafder, S.; Kao, K.; Jun, Y.; Yang, G.; Mao, J.J. Harnessing endogenous stem/progenitor cells for tendon regeneration. J. Clin. Investig. 2015, 125, 2690–2701. [Google Scholar] [CrossRef]
  129. Uggen, C.; Dines, J.; McGarry, M.; Grande, D.; Lee, T.; Limpisvasti, O. The effect of recombinant human platelet-derived growth factor BB-coated sutures on rotator cuff healing in a sheep model. J. Arthrosc. Relat. Surg. 2010, 26, 1456–1462. [Google Scholar] [CrossRef]
  130. Ko, J.Y.; Kim, K.I.; Park, S.; Im, G.I. In vitro chondrogenesis and in vivo repair of osteochondral defect with human induced pluripotent stem cells. Biomaterials 2014, 35, 3571–3581. [Google Scholar] [CrossRef]
  131. Xing, D.; Wu, J.; Wang, B.; Liu, W.; Liu, W.; Zhao, Y.; Wang, L.; Li, J.J.; Liu, A.; Zhou, Q.; et al. Intra-articular delivery of umbilical cord-derived mesenchymal stem cells temporarily retard the progression of osteoarthritis in a rat model. Int. J. Rheum. Dis. 2020, 23, 778–787. [Google Scholar] [CrossRef]
  132. Liu, Y.; Li, M.; Yin, Z.; Zhou, S.; Qiu, Y. SUMO-Modified bone marrow mesenchymal stem cells promoted the repair of articular cartilage in rats. Cell Biol. Int. 2020, 44, 560–568. [Google Scholar] [CrossRef] [PubMed]
  133. Tang, C.; Jin, C.; Li, X.; Li, J.; Du, X.; Yan, C.; Lu, S.; Wei, B.; Xu, Y.; Wang, L. Evaluation of an autologous bone mesenchymal stem cell-derived extracellular matrix scaffold in a rabbit and minipig model of cartilage repair. Med. Sci. Monit. 2019, 25, 7342–7350. [Google Scholar] [CrossRef] [PubMed]
  134. Zayed, M.; Newby, S.; Misk, N.; Donnell, R.; Dhar, M. Xenogenic implantation of equine synovial fluid-derived mesenchymal stem cells leads to articular cartilage regeneration. Stem Cells Int. 2018, 2018, 1073705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Lee, C.H.; Cook, J.L.; Mendelson, A.; Moioli, E.K.; Yao, H.; Mao, J.J. Regeneration of the articular surface of the rabbit synovial joint by cell homing: A proof of concept study. Lancet 2010, 376, 440–448. [Google Scholar] [CrossRef] [Green Version]
  136. Chen, Y.; Wu, T.; Huang, S.; Suen, C.W.W.; Cheng, X.; Li, J.; Hou, H.; She, G.; Zhang, H.; Wang, H.; et al. Sustained Release SDF-1α/TGF-β1-Loaded Silk Fibroin-Porous Gelatin Scaffold Promotes Cartilage Repair. ACS Appl. Mater. Interfaces 2019, 11, 14608–14618. [Google Scholar] [CrossRef]
  137. Maehara, H.; Sotome, S.; Yoshii, T.; Torigoe, I.; Kawasaki, Y.; Sugata, Y.; Yuasa, M.; Hirano, M.; Mochizuki, N.; Kikuchi, M.; et al. Repair of large osteochondral defects in rabbits using porous hydroxyapatite/collagen (HAp/Col) and fibroblast growth factor-2 (FGF-2). J. Orthop. Res. 2010, 28, 677–686. [Google Scholar] [CrossRef]
  138. Nourissat, G.; Berenbaum, F.; Duprez, D. Tendon injury: From biology to tendon repair. Nat. Rev. Rheumatol. 2015, 11, 223–233. [Google Scholar] [CrossRef]
  139. Fenwick, S.A.; Hazleman, B.L.; Riley, G.P. The vasculature and its role in the damaged and healing tendon. Arthritis Res. 2002, 4, 252–260. [Google Scholar] [CrossRef] [Green Version]
  140. Bray, R.C.; Rangayyan, R.M.; Frank, C.B. Normal and healing ligament vascularity: A quantitative histological assessment in the adult rabbit medial collateral ligament. J. Anat. 1996, 188, 87. [Google Scholar]
  141. Tozer, S.; Duprez, D. Tendon and ligament: Development, repair and disease. Birth Defects Res. Part C Embryo Today Rev. 2005, 75, 226–236. [Google Scholar] [CrossRef]
  142. Kannus, P. Structure of the tendon connective tissue. J. Med. Sci. Sport. 2000, 10, 312–320. [Google Scholar] [CrossRef] [PubMed]
  143. Kastelic, J.; Galeski, A.; Baer, E. The multicomposite structure of tendon. Connect. Tissue Res. 1978, 6, 11–23. [Google Scholar] [CrossRef] [PubMed]
  144. Riley, G.P.; Harrall, H.; Constant, C.R.; Chard, M.D.; Cawston, T.E.; Hazleman, B.L. Tendon degeneration and chronic shoulder pain: Changes in the collagen composition of the human rotator cuff tendons in rotator cuff tendinitis. Ann. Rheum. Dis. 1994, 53, 359–366. [Google Scholar] [CrossRef]
  145. Thorpe, C.T.; Birch, H.L.; Clegg, P.D.; Screen, H.R. The Role of the Non-Collagenous Matrix in Tendon Function. Int. J. Exp. Pathol. 2013, 94, 248–259. [Google Scholar] [CrossRef] [PubMed]
  146. Buckley, M.R.; Evans, E.B.; Matuszewski, P.E.; Chen, Y.L.; Satchel, L.N.; Elliott, D.M.; Soslowsky, L.J.; Dodge, G.R. Distributions of types I, II and III collagen by region in the human supraspinatus tendon. Connect. Tissue Res. 2013, 54, 374–379. [Google Scholar] [CrossRef]
  147. J.H., Y.; J., H. Tendon proteoglycans: Biochemistry and function. J. Musculoskelet. Neuronal Interact. 2005, 5, 22–34. [Google Scholar]
  148. Leong, N.L.; Kator, J.L.; Clemens, T.L.; James, A.; Enamoto-Iwamoto, M.; Jiang, J. Tendon and Ligament Healing and Current Approaches to Tendon and Ligament Regeneration. J. Orthop. Res. 2020, 38, 7–12. [Google Scholar] [CrossRef]
  149. Benjamin, M.; Ralphs, J.R. The cell and developmental biology of tendons and ligaments. Int. Rev. Cytol. 2000, 196, 85–130. [Google Scholar]
  150. Hope, M.; Saxby, T.S. Tendon Healing. Foot Ankle Clin. 2007, 12, 553–567. [Google Scholar] [CrossRef]
  151. Voleti, P.B.; Buckley, M.R.; Soslowsky, L.J. Tendon Healing: Repair and Regeneration. Annu. Rev. Biomed. Eng. 2012, 14, 47–71. [Google Scholar] [CrossRef]
  152. Molloy, T.; Wang, Y.; Murrell, G.A. The roles of growth factors in tendon and ligament healing. Sport. Med. 2003, 33, 381–394. [Google Scholar] [CrossRef] [PubMed]
  153. Massimino, M.L.; Rapizzi, E.; Cantini, M.; Libera, L.D.; Mazzoleni, F.; Arslan, P.; Carraro, U. ED2+ macrophages increase selectively myoblast proliferation in muscle cultures. Biochem. Biophys. Res. Commun. 1997, 235, 754–759. [Google Scholar] [CrossRef] [PubMed]
  154. Leadbetter, W.B. Cell-matrix response in tendon injury. Clin. Sports Med. 1992, 11, 533–578. [Google Scholar] [PubMed]
  155. James, R.; Kesturu, G.; Balian, G.; Chhabra, A.B. Tendon: Biology, Biomechanics, Repair, Growth Factors, and Evolving Treatment Options. J. Hand Surg. Am. 2008, 33, 102–112. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, G.; Rothrauff, B.B.; Tuan, R.S. Tendon and ligament regeneration and repair: Clinical relevance and developmental paradigm. Birth Defects Res. Part C Embryo Today Rev. 2013, 99, 203–222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Sharma, P.; Maffulli, N.; Maffulli, N. Tendon Structure Biology of Tendon Injury: Healing, Modeling and Remodeling. J. Musculoskelet. Neuronal Interact. 2006, 6, 181–190. [Google Scholar]
  158. Abrahamsson, S.-O.; Lundborg, G.; Lohmander, L.S. Long-term explant culture of rabbit flexor tendon: Effects of recombinant human insulin-like growth factor-I and serum on matrix metabolism. J. Orthop. Res. 1991, 9, 503–515. [Google Scholar] [CrossRef]
  159. Miyashita, H.; Ochi, M.; Ikuta, Y. Histological and biomechanical observations of the rabbit patellar tendon after removal of its central one-third. Arch. Orthop. Trauma Surg. 1997, 116, 454–462. [Google Scholar] [CrossRef]
  160. Carpenter, J.E.; Thomopoulos, S.; Flanagan, C.L.; DeBano, C.M.; Soslowsky, L.J. Rotator cuff defect healing: A biomechanical and histologic analysis in an animal model. J. Shoulder Elb. Surg. 1998, 7, 599–605. [Google Scholar] [CrossRef]
  161. Liu, C.F.; Aschbacher-Smith, L.; Barthelery, N.J.; Dyment, N.; Butler, D.; Wylie, C. What we should know before using tissue engineering techniques to repair injured tendons: A developmental biology perspective. Tissue Eng. Part B Rev. 2011, 17, 165–176. [Google Scholar] [CrossRef] [Green Version]
  162. Uchida, R.; Jacob, G.; Shimomura, K.; Horibe, S.; Nakamura, N. Biological Augmentation of ACL Repair and Reconstruction: Current Status and Future Perspective. Sports Med. Arthrosc. 2020, 28, 49–55. [Google Scholar] [CrossRef] [PubMed]
  163. Butler, D.L.; Juncosa, N.; Dressler, M.R. Functional Efficacy of Tendon Repair Processes. Annu. Rev. Biomed. Eng. 2004, 6, 303–329. [Google Scholar] [CrossRef] [PubMed]
  164. Teunis, T.; Lubberts, B.; Reilly, B.T.; Ring, D. A systematic review and pooled analysis of the prevalence of rotator cuff disease with increasing age. J. Shoulder Elb. Surg. 2014, 23, 1913–1921. [Google Scholar] [CrossRef] [PubMed]
  165. Oh, L.S.; Wolf, B.R.; Hall, M.P.; Levy, B.A.; Marx, R.G. Indications for rotator cuff repair: A systematic review. Clin. Orthop. Relat. Res. 2007, 455, 52–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Benjamin, M.; Evans, E.J.; Copp, L. The histology of tendon attachments to bone in man. J. Anat. 1986, 149, 89–100. [Google Scholar] [PubMed]
  167. Benjamin, M.; Toumi, H.; Ralphs, J.R.; Bydder, G.; Best, T.M.; Milz, S. Where tendons and ligaments meet bone: Attachment sites (‘entheses’) in relation to exercise and/or mechanical load. J. Anat. 2006, 208, 471–490. [Google Scholar] [CrossRef]
  168. Gerber, C.; Schneeberger, A.G.; Perren, S.M.; Nyffeler, R.W. Experimental rotator cuff repair. A preliminary study. J. Bone Jt. Surg. Ser. A 1999, 81, 1281–1290. [Google Scholar] [CrossRef]
  169. Frank, C.B.; Jackson, D.W. Current Concepts Review—The Science of Reconstruction of the Anterior Cruciate Ligament. J. Bone Jt. Surg. 2005, 1556–1576. [Google Scholar] [CrossRef]
  170. Murray, M.M.; Martin, S.D.; Martin, T.L.; Spector, M. Histological changes in the human anterior cruciate ligament after rupture. J. Bone Jt. Surg. Ser. A 2000, 82, 1387–1397. [Google Scholar] [CrossRef]
  171. Gagliardi, A.G.; Carry, P.M.; Parikh, H.B.; Traver, J.L.; Howell, D.R.; Albright, J.C. ACL Repair With Suture Ligament Augmentation Is Associated With a High Failure Rate Among Adolescent Patients. Am. J. Sports Med. 2019, 47, 560–566. [Google Scholar] [CrossRef]
  172. Buss, D.D.; Warren, R.F.; Wickiewicz, T.L.; Galinat, B.J.; Panariello, R. Arthroscopically assisted reconstruction of the anterior cruciate ligament with use of autogenous patellar-ligament grafts. Results after twenty-four to forty-two months. J. Bone Jt. Surg. Ser. A 1993, 75, 1346–1355. [Google Scholar] [CrossRef] [PubMed]
  173. Chiou, H.M.; Chang, M.C.; Lo, W.H. One-stage reconstruction of skin defect and patellar tendon rupture after total knee arthroplasty: A new technique. J. Arthroplast. 1997, 12, 575–579. [Google Scholar] [CrossRef]
  174. Schweitzer, R.; Chyung, J.H.; Murtaugh, L.C.; Brent, A.E.; Rosen, V.; Olson, E.N.; Lassar, A.; Tabin, C.J. Analysis of the tendon cell fate using Scleraxis, a specific marker for tendons and ligaments. Development 2001, 128, 3855–3866. [Google Scholar] [PubMed]
  175. Murchison, N.D.; Price, B.A.; Conner, D.A.; Keene, D.R.; Olson, E.N.; Tabin, C.J.; Schweitzer, R. Regulation of tendon differentiation by scleraxis distinguishes force-transmitting tendons from muscle-anchoring tendons. Development 2007, 134, 2697–2708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Shukunami, C.; Takimoto, A.; Nishizaki, Y.; Yoshimoto, Y.; Tanaka, S.; Miura, S.; Watanabe, H.; Sakuma, T.; Yamamoto, T.; Kondoh, G.; et al. Scleraxis is a transcriptional activator that regulates the expression of Tenomodulin, a marker of mature tenocytes and ligamentocytes. Sci. Rep. 2018, 8, 3155. [Google Scholar] [CrossRef] [Green Version]
  177. Bi, Y.; Ehirchiou, D.; Kilts, T.M.; Inkson, C.A.; Embree, M.C.; Sonoyama, W.; Li, L.; Leet, A.I.; Seo, B.-M.; Zhang, L.; et al. Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nature Med. 2007, 13, 1219–1227. [Google Scholar] [CrossRef]
  178. Rui, Y.-F.; Lui, P.P.Y.; Li, G.; Fu, S.C.; Lee, Y.W.; Chan, K.M. Isolation and characterization of multipotent rat tendon-derived stem cells. Tissue Eng. Part A 2010, 16, 1549–1558. [Google Scholar] [CrossRef] [Green Version]
  179. Lovati, A.B.; Corradetti, B.; Lange Consiglio, A.; Recordati, C.; Bonacina, E.; Bizzaro, D.; Cremonesi, F. Characterization and differentiation of equine tendon-derived progenitor cells. J. Biol. Regul. Homeost. Agents 2020, 25 (Suppl. 2), S75–S84. [Google Scholar]
  180. Sutter, W.W. Autologous Cell-Based Therapy for Tendon and Ligament Injuries. Clin. Tech. Equine Pract. 2007, 6, 198–208. [Google Scholar] [CrossRef]
  181. Gaspar, D.; Spanoudes, K.; Holladay, C.; Pandit, A.; Zeugolis, D. Progress in cell-based therapies for tendon repair. Adv. Drug Deliv. Rev. 2015, 84, 240–256. [Google Scholar] [CrossRef] [PubMed]
  182. Hirzinger, C.; Tauber, M.; Korntner, S.; Quirchmayr, M.; Bauer, H.C.; Traweger, A.; Tempfer, H. ACL injuries and stem cell therapy. Arch. Orthop. Trauma Surg. 2014, 134, 1573–1578. [Google Scholar] [CrossRef] [PubMed]
  183. Evans, C.H.; Huard, J. Gene therapy approaches to regenerating the musculoskeletal system. Nat. Rev. Rheumatol. 2015, 11, 234–242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. Dhillon, M.; Behera, P.; Patel, S.; Shetty, V. Orthobiologics and platelet rich plasma. Indian J. Orthop. 2014, 48, 1–9. [Google Scholar] [CrossRef]
  185. Bedi, A.; Maak, T.; Walsh, C.; Rodeo, S.A.; Grande, D.; Dines, D.M.; Dines, J.S. Cytokines in rotator cuff degeneration and repair. J. Shoulder Elb. Surg. 2012, 21, 218–227. [Google Scholar] [CrossRef] [PubMed]
  186. Morita, W.; Dakin, S.G.; Snelling, S.J.B.; Carr, A.J. Cytokines in tendon disease: A systematic review. Bone Jt. Res. 2017, 6, 656–664. [Google Scholar] [CrossRef] [Green Version]
  187. Irie, K.; Uchiyama, E.; Iwaso, H. Intraarticular inflammatory cytokines in acute anterior cruciate ligament injured knee. Knee 2003, 10, 93–96. [Google Scholar] [CrossRef]
  188. Kobayashi, D.; Kurosaka, M.; Yoshiya, S.; Mizuno, K. Effect of basic fibroblast growth factor on the healing of defects in the canine anterior cruciate ligament. Knee Surg. Sports Traumatol. Arthrosc. 1997, 5, 189–194. [Google Scholar] [CrossRef]
  189. Vermeulen, S.; Vasilevich, A.; Tsiapalis, D.; Roumans, N.; Vroemen, P.; Beijer, N.R.M.; Dede Eren, A.; Zeugolis, D.; de Boer, J. Identification of topographical architectures supporting the phenotype of rat tenocytes. Acta Biomater. 2019, 83, 277–290. [Google Scholar] [CrossRef]
  190. Lui, P.P.Y.; Wong, C.M. Biology of Tendon Stem Cells and Tendon in Aging. Front. Genet. 2020, 10, 1338. [Google Scholar] [CrossRef] [Green Version]
  191. Zhou, Z.; Akinbiyi, T.; Xu, L.; Ramcharan, M.; Leong, D.J.; Ros, S.J.; Colvin, A.C.; Schaffler, M.B.; Majeska, R.J.; Flatow, E.L.; et al. Tendon-derived stem/progenitor cell aging: Defective self-renewal and altered fate. Aging Cell 2010, 9, 911–915. [Google Scholar] [CrossRef] [Green Version]
  192. Jang, K.-M.; Lim, H.; Bae, J. Mesenchymal Stem Cells for Enhancing Biologic Healing after Anterior Cruciate Ligament Injuries. Curr. Stem Cell Res. Ther. 2015, 10, 535–547. [Google Scholar] [CrossRef] [PubMed]
  193. Gulotta, L.V.; Kovacevic, D.; Ehteshami, J.R.; Dagher, E.; Packer, J.D.; Rodeo, S.A. Application of bone marrow-derived mesenchymal stem cells in a Rotator cuff repair model. Am. J. Sports Med. 2009, 37, 2126–2133. [Google Scholar] [CrossRef] [PubMed]
  194. Gulotta, L.V.; Kovacevic, D.; Packer, J.D.; Deng, X.H.; Rodeo, S.A. Bone marrow-derived mesenchymal stem cells transduced with scleraxis improve rotator cuff healing in a rat model. Am. J. Sports Med. 2011, 39, 1282–1289. [Google Scholar] [CrossRef] [PubMed]
  195. Haddad-Weber, M.; Prager, P.; Kunz, M.; Seefried, L.; Jakob, F.; Murray, M.M.; Evans, C.H.; Nöth, U.; Steinert, A.F. BMP12 and BMP13 gene transfer induce ligamentogenic differentiation in mesenchymal progenitor and anterior cruciate ligament cells. Cytotherapy 2010, 12, 505–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Bunnell, B.A.; Flaat, M.; Gagliardi, C.; Patel, B.; Ripoll, C. Adipose-derived stem cells: Isolation, expansion and differentiation. Methods 2008, 45, 115–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. De Francesco, F.; Ricci, G.; D’Andrea, F.; Nicoletti, G.F.; Ferraro, G.A. Human Adipose Stem Cells: From Bench to Bedside. Tissue Eng. Part B Rev. 2015, 21, 572–584. [Google Scholar] [CrossRef] [Green Version]
  198. Park, A.; Hogan, M.V.; Kesturu, G.S.; James, R.; Balian, G.; Chhabra, A.B. Adipose-derived mesenchymal stem cells treated with growth differentiation factor-5 express tendon-specific markers. Tissue Eng. Part A 2010, 16, 2941–2951. [Google Scholar] [CrossRef] [Green Version]
  199. Uysal, C.A.; Tobita, M.; Hyakusoku, H.; Mizuno, H. Adipose-derived stem cells enhance primary tendon repair: Biomechanical and immunohistochemical evaluation. J. Plast. Reconstr. Aesthetic Surg. 2012, 65, 1712–1719. [Google Scholar] [CrossRef]
  200. Oshita, T.; Tobita, M.; Tajima, S.; Mizuno, H. Adipose-Derived Stem Cells Improve Collagenase-Induced Tendinopathy in a Rat Model. Am. J. Sports Med. 2016, 44, 1983–1989. [Google Scholar] [CrossRef]
  201. Kokubu, S.; Inaki, R.; Hoshi, K.; Hikita, A. Adipose-derived stem cells improve tendon repair and prevent ectopic ossification in tendinopathy by inhibiting inflammation and inducing neovascularization in the early stage of tendon healing. Regen. Ther. 2020, 14, 103–110. [Google Scholar] [CrossRef]
  202. Norelli, J.B.; Plaza, D.P.; Stal, D.N.; Varghese, A.M.; Liang, H.; Grande, D.A. Tenogenically differentiated adipose-derived stem cells are effective in Achilles tendon repair in vivo. J. Tissue Eng. 2018, 9. [Google Scholar] [CrossRef] [Green Version]
  203. Würgler-Hauri, C.C.; Dourte, L.A.M.; Baradet, T.C.; Williams, G.R.; Soslowsky, L.J. Temporal expression of 8 growth factors in tendon-to-bone healing in a rat supraspinatus model. J. Shoulder Elb. Surg. 2007, 16, S198–S203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Heisterbach, P.E.; Todorov, A.; Flückiger, R.; Evans, C.H.; Majewski, M. Effect of BMP-12, TGF-β1 and autologous conditioned serum on growth factor expression in Achilles tendon healing. Knee Surgery Sport. Traumatol. Arthrosc. 2012, 20, 1907–1914. [Google Scholar] [CrossRef] [PubMed]
  205. Hernigou, P.; Flouzat Lachaniette, C.H.; Delambre, J.; Zilber, S.; Duffiet, P.; Chevallier, N.; Rouard, H. Biologic augmentation of rotator cuff repair with mesenchymal stem cells during arthroscopy improves healing and prevents further tears: A case-controlled study. Int. Orthop. 2014, 38, 1811–1818. [Google Scholar] [CrossRef] [PubMed]
  206. Kim, Y.S.; Sung, C.H.; Chung, S.H.; Kwak, S.J.; Koh, Y.G. Does an Injection of Adipose-Derived Mesenchymal Stem Cells Loaded in Fibrin Glue Influence Rotator Cuff Repair Outcomes? A Clinical and Magnetic Resonance Imaging Study. Am. J. Sports Med. 2017, 45, 2010–2018. [Google Scholar] [CrossRef]
  207. Centeno, C.J.; Pitts, J.; Al-Sayegh, H.; Freeman, M.D. Anterior cruciate ligament tears treated with percutaneous injection of autologous bone marrow nucleated cells: A case series. J. Pain Res. 2015, 8, 437–447. [Google Scholar]
  208. Sophia Fox, A.J.; Bedi, A.; Rodeo, S.A. The basic science of articular cartilage: Structure, composition, and function. Sports Health 2009, 1, 461–468. [Google Scholar] [CrossRef]
  209. Malinin, T.; Ouellette, E.A. Articular cartilage nutrition is mediated by subchondral bone: A long-term autograft study in baboons. Osteoarthr. Cartil. 2000, 8, 483–491. [Google Scholar] [CrossRef] [Green Version]
  210. Wang, Y.; Wei, L.; Zeng, L.; He, D.; Wei, X. Nutrition and degeneration of articular cartilage. Knee Surgery Sport. Traumatol. Arthrosc. 2013, 21, 1751–1762. [Google Scholar] [CrossRef] [Green Version]
  211. Akkiraju, H.; Nohe, A. Role of chondrocytes in cartilage formation, progression of osteoarthritis and cartilage regeneration. J. Dev. Biol. 2015, 3, 177–192. [Google Scholar] [CrossRef] [Green Version]
  212. Smith, B.; Sigal, I.R.; Grande, D.A. Immunology and cartilage regeneration. Immunol. Res. 2015, 63, 181–186. [Google Scholar] [CrossRef] [PubMed]
  213. Huey, D.J.; Hu, J.C.; Athanasiou, K.A. Unlike bone, cartilage regeneration remains elusive. Science 2012, 338, 917–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  214. Liao, J.; Shi, K.; Ding, Q.; Qu, Y.; Luo, F.; Qian, Z. Recent developments in scaffold-guided cartilage tissue regeneration. J. Biomed. Nanotechnol. 2014, 10, 3085–3104. [Google Scholar] [CrossRef] [PubMed]
  215. Yuan, T.; Zhang, L.; Li, K.; Fan, H.; Fan, Y.; Liang, J.; Zhang, X. Collagen hydrogel as an immunomodulatory scaffold in cartilage tissue engineering. J. Biomed. Mater. Res. Part B Appl. Biomater. 2014, 102, 337–344. [Google Scholar] [CrossRef] [PubMed]
  216. Vilela, C.A.; Correia, C.; Oliveira, J.M.; Sousa, R.A.; Espregueira-Mendes, J.; Reis, R.L. Cartilage Repair Using Hydrogels: A Critical Review of in Vivo Experimental Designs. ACS Biomater. Sci. Eng. 2015, 1, 726–739. [Google Scholar] [CrossRef] [Green Version]
  217. Muchedzi, T.A.; Roberts, S.B. A systematic review of the effects of platelet rich plasma on outcomes for patients with knee osteoarthritis and following total knee arthroplasty. Surgeon 2018, 16, 250–258. [Google Scholar] [CrossRef]
  218. Cancedda, R.; Dozin, B.; Giannoni, P.; Quarto, R. Tissue engineering and cell therapy of cartilage and bone. Matrix Biol. 2003, 22, 81–91. [Google Scholar] [CrossRef]
  219. Hjelle, K.; Solheim, E.; Strand, T.; Muri, R.; Brittberg, M. Articular cartilage defects in 1,000 knee arthroscopies. Arthroscopy 2002, 18, 730–734. [Google Scholar] [CrossRef]
  220. Liu, M.; Zeng, X.; Ma, C.; Yi, H.; Ali, Z.; Mou, X.; Li, S.; Deng, Y.; He, N. Injectable hydrogels for cartilage and bone tissue engineering. Bone Res. 2017, 5, 17014. [Google Scholar] [CrossRef]
  221. Walker, K.J.; Madihally, S.V. Anisotropic temperature sensitive chitosan-based injectable hydrogels mimicking cartilage matrix. J. Biomed. Mater. Res. Part B Appl. Biomater. 2015, 103, 1149–1160. [Google Scholar] [CrossRef]
  222. Chahla, J.; Cinque, M.E.; Godin, J.A.; Sanchez, G.; Lebus, G.F.; Whalen, J.M.; Price, M.D.; Kennedy, N.I.; Moatshe, G.; LaPrade, R.F.; et al. Meniscectomy and Resultant Articular Cartilage Lesions of the Knee Among Prospective National Football League Players: An Imaging and Performance Analysis. Am. J. Sports Med. 2018, 46, 200–207. [Google Scholar] [CrossRef] [PubMed]
  223. Dorotka, R.; Windberger, U.; Macfelda, K.; Bindreiter, U.; Toma, C.; Nehrer, S. Repair of articular cartilage defects treated by microfracture and a three-dimensional collagen matrix. Biomaterials 2005, 26, 3617–3629. [Google Scholar] [CrossRef] [PubMed]
  224. Mithoefer, K.; Mcadams, T.; Williams, R.J.; Kreuz, P.C.; Mandelbaum, B.R. Clinical efficacy of the microfracture technique for articular cartilage repair in the knee: An evidence-based systematic analysis. Am. J. Sports Med. 2009, 37, 2053–2063. [Google Scholar] [CrossRef] [PubMed]
  225. Benazzo, F.; Cadossi, M.; Cavani, F.; Fini, M.; Giavaresi, G.; Setti, S.; Cadossi, R.; Giardino, R. Cartilage repair with osteochondral autografts in sheep: Effect of biophysical stimulation with pulsed electromagnetic fields. J. Orthop. Res. 2008, 26, 631–642. [Google Scholar] [CrossRef]
  226. McCarty, E.C.; Fader, R.R.; Mitchell, J.J.; Glenn, R.E.; Potter, H.G.; Spindler, K.P. Fresh osteochondral allograft versus autograft: Twelve-month results in isolated canine knee defects. Am. J. Sports Med. 2016, 44, 2354–2365. [Google Scholar] [CrossRef]
  227. DiBartola, A.C.; Wright, B.M.; Magnussen, R.A.; Flanigan, D.C. Clinical Outcomes After Autologous Chondrocyte Implantation in Adolescents’ Knees: A Systematic Review. Arthrosc. J. Arthrosc. Relat. Surg. 2016, 32, 1905–1916. [Google Scholar] [CrossRef]
  228. Niemeyer, P.; Pestka, J.M.; Kreuz, P.C.; Erggelet, C.; Schmal, H.; Suedkamp, N.P.; Steinwachs, M. Characteristic complications after autologous chondrocyte implantation for cartilage defects of the knee joint. Am. J. Sports Med. 2008, 36, 2091–2099. [Google Scholar] [CrossRef]
  229. Selmi, T.A.S.; Verdonk, P.; Chambat, P.; Dubrana, F.; Potel, J.F.; Barnouin, L.; Neyret, P. Autologous chondrocyte implantation in a novel alginate-agarose hydrogel: Outcome at two years. J. Bone Jt. Surg. 2008, 90, 597–604. [Google Scholar] [CrossRef]
  230. Harris, J.D.; Siston, R.A.; Pan, X.; Flanigan, D.C. Autologous chondrocyte implantation: A systematic review. J. Bone Jt. Surg. Ser. 2010, 92, 2220–2233. [Google Scholar] [CrossRef]
  231. Brittberg, M.; Lindahl, A.; Nilsson, A.; Ohlsson, C.; Isaksson, O.; Peterson, L. Treatment of deep cartilage defects in the knee with autologous chondrocyte transplantation. N. Engl. J. Med. 1994, 331, 889–895. [Google Scholar] [CrossRef] [PubMed]
  232. Peterson, L.; Vasiliadis, H.S.; Brittberg, M.; Lindahl, A. Autologous chondrocyte implantation: A long-term follow-up. Am. J. Sports Med. 2010, 38, 1117–1124. [Google Scholar] [CrossRef] [PubMed]
  233. Jacobi, M.; Villa, V.; Magnussen, R.A.; Neyret, P. MACI—A new era? Sport. Med. Arthrosc. Rehabil. Ther. Technol. 2011, 3, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  234. Devitt, B.M.; Bell, S.W.; Webster, K.E.; Feller, J.A.; Whitehead, T.S. Surgical treatments of cartilage defects of the knee: Systematic review of randomised controlled trials. Knee 2017, 24, 508–517. [Google Scholar] [CrossRef] [PubMed]
  235. Na, Y.; Shi, Y.; Liu, W.; Jia, Y.; Kong, L.; Zhang, T.; Han, C.; Ren, Y. Is implantation of autologous chondrocytes superior to microfracture for articular-cartilage defects of the knee? A systematic review of 5-year follow-up data. Int. J. Surg. 2019, 68, 56–62. [Google Scholar] [CrossRef] [PubMed]
  236. Song, S.J.; Park, C.H. Microfracture for cartilage repair in the knee: Current concepts and limitations of systematic reviews. Ann. Transl. Med. 2019, 7, S108. [Google Scholar] [CrossRef] [PubMed]
  237. Aae, T.F.; Randsborg, P.H.; Lurås, H.; Årøen, A.; Lian, Ø.B. Microfracture is more cost-effective than autologous chondrocyte implantation: A review of level 1 and level 2 studies with 5 year follow-up. Knee Surgery Sport. Traumatol. Arthrosc. 2018, 26, 1044–1052. [Google Scholar] [CrossRef] [Green Version]
  238. Sherman, S.L.; Thyssen, E.; Nuelle, C.W. Osteochondral Autologous Transplantation. Clin. Sports Med. 2017, 36, 489–500. [Google Scholar] [CrossRef]
  239. Batty, L.; Dance, S.; Bajaj, S.; Cole, B.J. Autologous chondrocyte implantation: An overview of technique and outcomes. ANZ J. Surg. 2011, 81, 18–25. [Google Scholar] [CrossRef]
  240. Kizaki, K.; El-Khechen, H.A.; Yamashita, F.; Duong, A.; Simunovic, N.; Musahl, V.; Ayeni, O.R. Arthroscopic versus Open Osteochondral Autograft Transplantation (Mosaicplasty) for Cartilage Damage of the Knee: A Systematic Review. J. Knee Surg. 2019, 9, 2019. [Google Scholar] [CrossRef]
  241. Bao, W.; Li, M.; Yang, Y.; Wan, Y.; Wang, X.; Bi, N.; Li, C. Advancements and Frontiers in the High Performance of Natural Hydrogels for Cartilage Tissue Engineering. Front. Chem. 2020, 8. [Google Scholar] [CrossRef] [Green Version]
  242. Schnabel, M.; Marlovits, S.; Eckhoff, G.; Fichtel, I.; Gotzen, L.; Vécsei, V.; Schlegel, J. Dedifferentiation-associated changes in morphology and gene expression in primary human articular chondrocytes in cell culture. Osteoarthr. Cartil. 2002, 10, 62–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  243. Roberts, S.; Menage, J.; Sandell, L.J.; Evans, E.H.; Richardson, J.B. Immunohistochemical study of collagen types I and II and procollagen IIA in human cartilage repair tissue following autologous chondrocyte implantation. Knee 2009, 16, 398–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Pittenger, M.F.; Mackay, A.M.; Beck, S.C.; Jaiswal, R.K.; Douglas, R.; Mosca, J.D.; Moorman, M.A.; Simonetti, D.W.; Craig, S.; Marshak, D.R. Multilineage potential of adult human mesenchymal stem cells. Science 1999, 284, 143–147. [Google Scholar] [CrossRef] [Green Version]
  245. Johnstone, B.; Hering, T.M.; Caplan, A.I.; Goldberg, V.M.; Yoo, J.U. In vitro chondrogenesis of bone marrow-derived mesenchymal progenitor cells. Exp. Cell Res. 1998, 238, 265–272. [Google Scholar] [CrossRef] [PubMed]
  246. Winter, A.; Breit, S.; Parsch, D.; Benz, K.; Steck, E.; Hauner, H.; Weber, R.M.; Ewerbeck, V.; Richter, W. Cartilage-like gene expression in differentiated human stem cell spheroids: A comparison of bone marrow-derived and adipose tissue-derived stromal cells. Arthritis Rheum. 2003, 48, 418–429. [Google Scholar] [CrossRef] [PubMed]
  247. Fuggle, N.R.; Cooper, C.; Oreffo, R.O.C.; Price, A.J.; Kaux, J.F.; Maheu, E.; Cutolo, M.; Honvo, G.; Conaghan, P.G.; Berenbaum, F.; et al. Alternative and complementary therapies in osteoarthritis and cartilage repair. Aging Clin. Exp. Res. 2020, 32, 547–560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  248. Ohba, S.; Hojo, H.; Chung, U. Il Bioactive factors for tissue regeneration: State of the art. Muscles Ligaments Tendons J. 2012, 2, 193–203. [Google Scholar] [PubMed]
  249. Fortier, L.A.; Barker, J.U.; Strauss, E.J.; McCarrel, T.M.; Cole, B.J. The role of growth factors in cartilage repair. Clin. Orthop. Relat. Res. 2011, 469, 2706–2715. [Google Scholar] [CrossRef] [Green Version]
  250. Censi, R.; Dubbini, A.; Matricardi, P. Bioactive Hydrogel Scaffolds—Advances in Cartilage Regeneration Through Controlled Drug Delivery. Curr. Pharm. Des. 2015, 21, 1545–1555. [Google Scholar] [CrossRef]
  251. Dowthwaite, G.P.; Bishop, J.C.; Redman, S.N.; Khan, I.M.; Rooney, P.; Evans, D.J.R.; Haughton, L.; Bayram, Z.; Boyer, S.; Thomson, B.; et al. The surface of articular cartilage contains a progenitor cell populations. J. Cell Sci. 2004, 117, 889–897. [Google Scholar] [CrossRef] [Green Version]
  252. Yang, S.S.; Jin, L.H.; Park, S.H.; Kim, M.S.; Kim, Y.J.; Choi, B.H.; Lee, C.T.; Park, S.R.; Min, B.H. Extracellular matrix (ECM) multilayer membrane as a sustained releasing growth factor delivery system for rhTGF-β3 in articular cartilage repair. PLoS ONE 2016, 11, e0156292. [Google Scholar] [CrossRef] [PubMed]
  253. Park, M.S.; Kim, Y.H.; Jung, Y.; Kim, S.H.; Park, J.C.; Yoon, D.S.; Kim, S.H.; Lee, J.W. In Situ Recruitment of Human Bone Marrow-Derived Mesenchymal Stem Cells Using Chemokines for Articular Cartilage Regeneration. Cell Transplant. 2015, 24, 1067–1083. [Google Scholar] [CrossRef] [PubMed]
  254. Yu, Y.; Brouillette, M.J.; Seol, D.; Zheng, H.; Buckwalter, J.A.; Martin, J.A. Use of recombinant human stromal cell-derived factor 1α-loaded fibrin/hyaluronic acid hydrogel networks to achieve functional repair of full-thickness bovine articular cartilage via homing of chondrogenic progenitor cells. Arthritis Rheumatol. 2015, 67, 1274–1285. [Google Scholar] [CrossRef] [PubMed]
  255. Boushell, M.K.; Mosher, C.Z.; Suri, G.K.; Doty, S.B.; Strauss, E.J.; Hunziker, E.B.; Lu, H.H. Polymeric mesh and insulin-like growth factor 1 delivery enhance cell homing and graft−cartilage integration. Ann. N. Y. Acad. Sci. 2019, 1442, 138–152. [Google Scholar] [CrossRef] [PubMed]
  256. Gimble, J.M.; Guilak, F. Adipose-derived adult stem cells: Isolation, characterization, and differentiation potential. Cytotherapy 2003, 5, 362–369. [Google Scholar] [CrossRef] [PubMed]
  257. Wickham, M.Q.; Erickson, G.R.; Gimble, J.M.; Vail, T.P.; Guilak, F. Multipotent stromal cells derived from the infrapatellar fat pad of the knee. Clin. Orthop. Relat. Res. 2003, 412, 196–212. [Google Scholar] [CrossRef]
  258. Estes, B.T.; Diekman, B.O.; Gimble, J.M.; Guilak, F. Isolation of adipose-derived stem cells and their induction to a chondrogenic phenotype. Nat. Protoc. 2010, 5, 1294–1311. [Google Scholar] [CrossRef] [Green Version]
  259. Huang, J.I.; Zuk, P.A.; Jones, N.F.; Zhu, M.; Lorenz, H.P.; Hedrick, M.H.; Benhaim, P. Chondrogenic potential of multipotential cells from human adipose tissue. Plast. Reconstr. Surg. 2004, 113, 585–594. [Google Scholar] [CrossRef]
Figure 1. Conceptual diagrams of the two main approaches used to stimulate skeletal tissue healing and repair by sequestering the help of stem/progenitor cells. (A) Biomaterials/scaffolds and/or bioactive agents, such as growth factors, chemokines, and small molecules are placed at the site of injury to stimulate the migration and differentiation of endogenous native mesenchymal stem cells for aiding in the repair process. (B) Cells are physically administered exogenously, from one anatomical location of the patient to the site of injury, or from a donor, to aid in the repair process. This can be done with or without a scaffold or biomaterial to hold the newly administered cells in place.
Figure 1. Conceptual diagrams of the two main approaches used to stimulate skeletal tissue healing and repair by sequestering the help of stem/progenitor cells. (A) Biomaterials/scaffolds and/or bioactive agents, such as growth factors, chemokines, and small molecules are placed at the site of injury to stimulate the migration and differentiation of endogenous native mesenchymal stem cells for aiding in the repair process. (B) Cells are physically administered exogenously, from one anatomical location of the patient to the site of injury, or from a donor, to aid in the repair process. This can be done with or without a scaffold or biomaterial to hold the newly administered cells in place.
Bioengineering 07 00086 g001
Figure 2. Diagram depicting various sources of Mesenchymal Stem Cells (MSCs) and their cellular microenvironments of adjacent skeletal tissues that make up a joint. These include MSCs from synovium, cartilage, bone marrow, periosteum, and ligament/tendon. Additionally, MSCs from adipose tissue such as fat pad (not depicted here) are also present. This represents how each tissue is not a closed system but rather that they are constantly in the direct proximity of MSCs from adjacent tissues.
Figure 2. Diagram depicting various sources of Mesenchymal Stem Cells (MSCs) and their cellular microenvironments of adjacent skeletal tissues that make up a joint. These include MSCs from synovium, cartilage, bone marrow, periosteum, and ligament/tendon. Additionally, MSCs from adipose tissue such as fat pad (not depicted here) are also present. This represents how each tissue is not a closed system but rather that they are constantly in the direct proximity of MSCs from adjacent tissues.
Bioengineering 07 00086 g002
Table 1. Pre-clinical studies of endogenous and exogenous use of stem/progenitor cells in skeletal tissue.
Table 1. Pre-clinical studies of endogenous and exogenous use of stem/progenitor cells in skeletal tissue.
TissueApproachAnimal ModelInjury ModelExperimental TreatmentOutcomes/ResultsReferences
BoneEndogenousMiceLong Segmental DefectGrowth factor + AMD3100 treatment 2 weeks:
IGF1 showed increased proliferation and migration of isolated MSC as well as augmented bone growth.
Kumar et al., [86] 2012
BoneExogenousNude miceCalvarial defect (4 mm)Undifferentiated Human ASCs + PLGA scaffold + (rh) BMP28 weeks:
Gross analysis, microCT, and histological examination showed complete healing and trabecular bone formation in the hASCs treated group compared to the scaffold only group and empty defects.
Levi et al., [103] 2010
BoneExogenousRatsFemoral fractureBMSCs + skin fibroblasts5 weeks:
Callus size and mechanical properties were significantly higher in the MSC injected group compared to fibroblast and the PBS control. Quantitative analysis showed GFP-positive MSCs were present in callus in MSC group at 5 weeks after fracture.
Huang et al., [113] 2015
BoneExogenousMiceFemur fractureMouse BMSCs/RFP42 days:
BMSCs injected on day 7 post-fracture accelerated fracture healing with improved callus and bone quality.
Wang et al., [114] 2018
BoneExogenousRatBone nonunionPrimary MSCs sheet + SDF1 injection4 and 8 weeks:
At 4 weeks, new formed bone tissue united the distal and proximal sites in the MSC sheet/SDF group compared to 5 other groups. At 8 weeks, the MSC sheet/SDF group showed complete bridging of the fracture site, forming hard bony union.
Chen et al., [115] 2016
BoneExogenous + EndogenousMiceOsteogenesis impairmentPeptidomimetic ligand (LLP2A) + Alendronate (LLP2A-Ale) injection3 and 12 weeks:
At 3 weeks, the hMSC cells injected intravenously in the xenotransplantation model were observed at the bone surface in the LLP2A-Ale group. At 12 weeks, the LLP2A-Ale group could augment bone formation in mice.
Guan et al., [116] 2012
TendonExogenousRatsMulti-differentiation potentialTSCs + Matrigel (gel-cells)8 weeks:
Transplantation of TSCs subcutaneously resulted in the formation of tendon, cartilage and bone-like tissues.
Zhang et al., [117] 2010
TendonExogenousRatAchilles TendonMSCs cultured in hypoxic and normoxic condition2 and 4 weeks:
Tendon rupture site and biomechanical properties were superior in hypoxic MSC group compared to the normoxic and control.
Huang et al., [118] 2013
LigamentEndogenous; Growth factorsCanineACL defectCollagen-Platelet Rich Plasma (PRP) Scaffold3 and 6 weeks:
The percent filling defect was significantly higher in the treated group at both 3 and 6 weeks compared to the untreated defects. Mechanically, the ACL treated group had 40% increase in strength at 6 weeks, compared to untreated defects.
Murray et al., [119] 2006
LigamentEndogenous; Growth factorsPorcineACL defectSuture + Collagen-Platelet Rich Plasma (PRP) hydrogel4 weeks:
At 4 weeks, the collagen-PRP hydrogel group stimulated healing and improved biomechanical properties after suture repair, compared to suture repair alone. However, both groups remained significantly inferior to the intact ligament group.
Murray et al., [120] 2007
TendonEndogenous; Growth factorsRatRotator Cuff3D printed scaffold + Growth factors (CTGF, CTGF + TGF-b + BMP2)1 and 4 weeks
GF embedded (+GF) scaffolds promoted recruitment of endogenous tendon progenitor cells and healed tendon-to-bone via formation of cartilaginous interface compared to –GF scaffold.
Solaiman et al., [121] 2019
TendonExogenousRatAchilles TendonTDSCs and BMMSCs1, 2 and 4 weeks:
TDSCs showed higher regenerative potential with high mechanical strength, better appearance density and well-organized longitudinal fibrous structure and BMSCs also showed positive results.
Al-Ani MK et al., [122] 2015
TendonExogenousRabbitPatellar Tendon defectBMSCs + Type I bovine collagen gel4 weeks:
Mechanically and histologically, the MSC + gel group showed significantly greater material and structural properties compared to the collagen gel alone control. However, treatment group improvements were not impressive compared to the normal healthy tendon.
Awad et al., [123] 1999
LigamentExogenousRabbitACL ReconstructionBMSCs + Silk scaffold8, 16 and 24 weeks:
The MSC/Scaffold group showed abundant ligament ECM (Col I was more prominent compared to Col III and Tenascin-C), compared to the scaffold alone control. The tensile strength was comparable to the mechanical properties of daily activities.
Fan et al., [124] 2008
TendonExogenousRatRotator Cuff injuryBMSCs + PRP4 and 8 weeks:
Gene and protein detection at 4 weeks, showed that combined therapy enhanced the expression of growth factors and genes related to tendon repair (Col I, Tenomodulin, Scx). At 8 weeks, mechanical testing demonstrated that combined therapy was most efficient to promote tissue regeneration, compared to single therapy control (PRP alone and MSC alone).
Han et al., [125] 2019
TendonExogenousRatTendon injuryhASC + fibrin glue4 weeks:
Treatment group of hASCs demonstrated enhanced tendon healing biomechanically, compared to the fibrin alone and sham group. Cells were showed to survive for 4 weeks, in vivo and secreted human-specific Col I and Tenascin-C.
Lee et al., [126] 2017
TendonExogenousRatPartial Transection of Achilles TendonASCs + Fibrin Sealant (FS) from serpent venom21 days:
In vivo analysis at day 14 revealed higher quantification of the transplanted fluorescent ASCs in the tendon treated with ASCs + FS compared to ASC alone. The ASCs group up-regulated Tenomodulin expression compared to normal (without transection), transection alone and the FS group. TIMP-2 and Scx expression compared to N group. FS group demonstrated great organization of collagen fibers followed by ASCs + FS and ASCs alone in comparison to N
Frauz et al., [127] 2019
TendonEndogenous;
Growth factor
RatPatellar TendonTSCs (CD146+) + Fibrin glue + CTGF1, 2 and 4 weeks:
CTFG treated CD146+ cells led to tendon regeneration with dense collagen fibers, compared to the untreated CD146+. By week 4, the CTGF group generated tendon with dense collagen fibers compared to the fibrin alone group and tensile property on the level of native tendon compared to CD146- and untreated CD146+.
Lee et al., [128] 2015
TendonEndogenous;
Growth factor
SheepRotator Cuff injuryrhPDGF-BB coated sutures6 weeks:
rhPDGF-BB coated sutures enhanced histologic scores of sheep rotator injury and enhanced tendon healing. However, load to failure was equivalent to standard suture repair.
Uggen et al., [129] 2010
CartilageExogenousRatOsteo-chondral
Defect
hiPSCs pellet or hiPSCs + alginate hydrogel12 weeks:
Defects treated with chondro-induced hiPSCs implantation had smooth, firm tissue with good restoration of articular surface compared to control or alginate alone. However, histological appearance showed reduced amount of proteoglycan compared to the normal cartilage.
Ko et al., [130] 2014
CartilageExogenousRatOsteoarthritis Human umbilical MSCs + Hyaluronic acid (HA)6 and 12 weeks:
Macroscopic observation of the femur surface at 6 weeks, showed signs of OA progression with cartilage surface roughness and osteophyte formation compared to preserved cartilage in MSC + HA group; at 12 weeks, joint surface showed OA progression in all 3 groups.
Histologically at 6 weeks, the MSC + HA group showed abundant proteoglycan and reduced cartilage loss, whereas at 12 weeks, Saf-O staining was significantly reduced compared to 6 weeks Hence, single injection of hUC-MSCs had temporary effects to decelerate OA progression.
Xing et al., [131] 2020
CartilageExogenousRatFull thickness cartilage defect (2mm)BMSCs + SUMO1/SUMO2,3/SUMO1,2,34 weeks:
BMSCs overexpressing SUMO1 differentiated into articular cartilage with hard surface; BMSCs overexpressing SUMO1,2 reduced inflammation and improved damaged cartilage microenvironment; BMSCs overexpressing SUMO1,2,3 showed better survival, less inflammatory response, and improved tissue repair.
Liu et al., [132] 2020
CartilageExogenousRabbit and MinipigsOsteo-chondral defectECM group: autologous MSC-derived ECM scaffold;
BMS group:
Bone marrow stimulation
Rabbits: 6hrs, 3 and 7 days:
Macroscopic appearance of exudate healing wounds showed less fibrosis and histology showed evenly distributed chondrocyte in the EMS group compared to the BMS. The CFU-F assay showed increased number of bone MSCs in the ECM group.
Minipigs: 6 months:
Macroscopic and MRI finding improved in the ECM compared to BMS group. Repaired tissue in ECM had similar histological characteristic to normal hyaline cartilage.
Tang et al., [133] 2019
CartilageExogenousRatFull thickness cartilage defect (2 mm)Equine BMSCs and Synovial Fluid-Derived MSC (SFMSCs) + agarose gel1 and 12 weeks:
At 1 week, the knee joint showed the presence of MSCs at the injured site.Macroscopic and histological analysis demonstrated better healing of cartilage in MSC treated knees at 12 weeks, compared to the control. SFMSC treated showed significantly higher Col II, suggesting presence of hyaline cartilage at the defect site.
Zayed et al., [134] 2018
CartilageEndogenous;
Growth factors
RabbitHumeral Head incisionTGF-β3 adsorbed or TGF-β3-free + collagen hydrogel4 months:
The TGF-β3 treated group had significantly greater matrix and articular cartilage thickness compared to the TGF-β3-free group, showing that the articular cartilage of the synovial joint was regenerated by homing endogenous cells. The TGF-β3 treated group also had consistent distribution of Col II and Aggrecan.
Lee et al., [135] 2010
CartilageEndogenous;
Growth factors
RatsOsteo-chondral Defect (1.6mm)Silk fibroin scaffold + SDF-1α + TGF-β112 weeks:
Scaffold treated with + SDF-1α and TGF-β1 (GSTS) had the most significant cartilage regeneration compared to 4 other control groups. The GSTS group also produced more type II collagen compared to other groups, which generated fibrocartilage.
Chen et al., [136] 2019
CartilageEndogenous;
Growth factors
RabbitOsteo-chondral Defect (5 mm)Hydroxyapatite collagen (Hap/Col) scaffold + FGF-2 with 10 and 100 µg/mL concentration collagen (HAp/Col) scaffold 3,6, 12 and 24 weeks:
Abundant bone formation observed in the Hap/Col group compared to the defect group. The FGF10 group demonstrated abundant bone regeneration as well as satisfactory cartilage regeneration with a hyaline-like appearance.
Maehara et al., [137] 2010
Table 2. List of few ongoing clinical trials for skeletal tissue.
Table 2. List of few ongoing clinical trials for skeletal tissue.
ConditionNCT IdentifierTitleStatusIntervention
Non Union FractureNCT03325504A Comparative Study of 2 Doses of BM Autologous H-MSC + Biomaterial vs Iliac Crest AutoGraft for Bone Healing in Non-Union RecruitingBiological: Cultured Mesenchymal Stem Cells
Procedure: Autologous iliac crest graft
Osteochondral Fracture of TalusNCT03905824The Effectiveness of Adding Allogenic Stem Cells After Traditional Treatment of Osteochondral Lesions of the TalusRecruitingBiological: Allogenic stromal mesenchymal cells derived from the umbilical cord
Procedure: Debridement and microfracture
Full Thickness Rotator Cuff TearNCT02484950Mesenchymal Stem Cell Augmentation in Patients Undergoing Arthroscopic Rotator Cuff RepairRecruitingBiological: Mesenchymal stem cell augmentation in rotator cuff repair
Procedure: Standard arthroscopic rotator cuff repair
Rotator Cuff Tear
Rotator Cuff Tendinitis
NCT03752827Autologous Adult Adipose-Derived Regenerative Cell Injection into Chronic Partial-Thickness Rotator Cuff TearsRecruitingDevice: Adipose Derived Regenerative Cells
Drug: Corticosteroid
Rotator Cuff TearNCT03688308Bone Marrow Derived Stem Cells for the Treatment of Rotator Cuff TearsRecruitingProcedure: Arthroscopic rotator cuff repair with bone marrow aspirate concentrate
Rotator Cuff TearNCT03551509LifeNet: Extracellular Matrix Graft in Rotator Cuff RepairRecruitingBiological: ArthroFLEX ECM scaffold graft
Procedure: Control
Biological: Crossover
Rotator Cuff TearsNCT04325789Rotator Cuff Healing Using a Nanofiber Scaffold in Patients Greater Than 55 YearsRecruitingDevice: Rotium nanofiber graft
ACL—Anterior Cruciate Ligament RuptureNCT03294720BioACL Reconstruction with Amnion Collagen Matrix Wrap and Stem Cells Case Series Active, not recruitingProcedure: Bio-ACL
Device: amnion wrap and BMAC
ACL—Anterior Cruciate Ligament RuptureNCT03294759Bio ACL Reconstruction Amnion Collagen Matrix Wrap and Stem CellsActive, not recruitingOther: Bio ACL
Other: Control
Anterior Cruciate Ligament TearNCT02664545Bridge-Enhanced ACL Repair vs. ACL ReconstructionActive, not recruitingDevice: BEAR Scaffold
Procedure: Tendon Graft
Defect of Articular Cartilage
Cartilage Injury
Osteoarthritis, Knee
NCT02696876Synovium Brushing to Augmented Microfracture for Improved Cartilage RepairRecruitingDevice: Arthroscopic synovial brushing
Procedure: Microfracture
Degenerative Lesion of Articular Cartilage of KneeNCT02090140Microfracture Versus Adipose Derived Stem Cells for the Treatment of Articular Cartilage DefectsRecruitingProcedure: ADSC Application
Procedure: Microfracture
Osteoarthritis, KneeNCT04205656Prospective Evaluation of PRP and BMC Treatment to Accelerate Healing After ACL ReconstructionRecruitingBiological: Leukocyte-Poor Platelet Rich Plasma (LP-PRP)
Biological: Bone Marrow Concentrate (BMC)
Other: Control group (Placebo)
Osteoarthritis, Knee NCT02805855Autologous Culture Expanded Mesenchymal Stromal Cells for Knee Osteoarthritis Recruiting Drug: Autologous Adipose-Derived Mesenchymal Stromal Cells
Knee OsteoarthritisNCT03014401The Effect of Adipose-Derived Stem Cells for Knee OsteoarthritisRecruitingProcedure: Arthroscopic debridement with stem cell transplantation
Procedure: Arthroscopic debridement only
Osteoarthritis, Knee
Knee Pain
NCT03467919The Effect of Micro Fragmented Adipose Tissue (MFAT) on Knee OsteoarthritisRecruitingProcedure: Micro Fragmented Adipose Tissue
Procedure: Corticosteroid injection
Post-Traumatic Osteoarthritis of KneeNCT04222140Early Regenerative Intervention for Post-Traumatic OsteoarthritisNot yet recruitingCombination Product: ERIPTO Protocol
Biological: BMAC Only
Knee OsteoarthritisNCT04043819Evaluation of Safety and Exploratory Efficacy of an Autologous Adipose-derived Cell Therapy Product for Treatment of Single Knee OsteoarthritisActive, not recruitingDrug: PSC-01
Musculoskeletal Pain
Knee Osteoarthritis
Cartilage Injury
Cartilage Degeneration
NCT03477942Impact of Mesenchymal Stem Cells in Knee OsteoarthritisRecruitingBiological: Autologous Mesenchymal Stem Cells
Articular Cartilage Disorder of Knee
Articular Cartilage; Degeneration
NCT03101163Efficacy and Safety Study of Intra-Articular Injections of Autologous Peripheral Blood Stem Cells Following Subchondral Drilling Surgery for the Treatment of Articular Cartilage Injury in the KneeRecruitingBiological: Autologous peripheral blood stem cells and hyaluronic acid
Other: Hyaluronic acid
Osteoarthritis, HipNCT03608579Autologous Culture Expanded Adipose Derived MSCs for Treatment of Painful Hip OARecruitingDrug: Autologous Adipose Derived Mesenchymal Stromal Cells
OsteoarthritisNCT03818737Multicenter Trial of Stem Cell Therapy for Osteoarthritis (MILES)RecruitingBiological: Autologous Bone Marrow Concentrate (BMAC)
Biological: Adipose-derived Stromal Vascular Fraction (SVF)
Biological: Umbilical Cord Tissue
Drug: Depomedrol and Normal saline (Corticosteroid injection)

Share and Cite

MDPI and ACS Style

Desai, S.; Jayasuriya, C.T. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering 2020, 7, 86. https://doi.org/10.3390/bioengineering7030086

AMA Style

Desai S, Jayasuriya CT. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering. 2020; 7(3):86. https://doi.org/10.3390/bioengineering7030086

Chicago/Turabian Style

Desai, Salomi, and Chathuraka T. Jayasuriya. 2020. "Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair" Bioengineering 7, no. 3: 86. https://doi.org/10.3390/bioengineering7030086

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