Abstract
Tendon disorders, which are commonly presented in the clinical setting, disrupt the patients’ normal work and life routines, and they damage the careers of athletes. However, there is still no effective treatment for tendon disorders. In the field of tissue engineering, the potential of the therapeutic application of exogenous stem cells to treat tendon pathology has been demonstrated to be promising. With the development of stem cell biology and chemical biology, strategies that use inductive tenogenic factors to program stem cell fate in situ are the most easily and readily translatable to clinical applications. In this review, we focus on bioactive molecules that can potentially induce tenogenesis in adult stem cells, and we summarize the various differentiation factors found in comparative studies. Moreover, we discuss the molecular regulatory mechanisms of tenogenesis, and we examine the various challenges in developing standardized protocols for achieving efficient and reproducible tenogenesis. Finally, we discuss and predict future directions for tendon regeneration. Stem Cells Translational Medicine 2018;7:404–414
Keywords: Adult stem cells, Bone marrow stromal cells, Differentiation, Tissue regeneration, Transforming growth factor β
Significance Statement.
Tendon disorder represents a major clinical challenge in orthopedic surgery. Currently there is no effective drug system for tendon disease treatments. A better understanding of bioactive molecules for tenogenesis effect on stem cells in vitro and in situ will lead to better treatments for tendon injuries and tendinopathy. Current challenges and future directions are discussed and suggested in order to develop effective chemical cocktails for stepwise inducing tendon regeneration.
Introduction
Tendon and ligament injuries comprise almost 50% of sports and physical activity‐related injuries, causing 30–50 million new incidents annually worldwide 1. Tendon injuries following acute trauma or physical strain constitute a major clinical challenge because of the poor intrinsic healing capacity of tendons. Recent advances indicate that the transplantation or grafting of exogenous or endogenous stem cell populations exert regenerative effects on diseased or injured tendons 2. Moreover, the precise regulation of the tenogenic differentiation of stem cells is essential for the positive outcome of stem cell‐based therapy applied to tendon tissues.
Among the various strategies used for tendon differentiation, mechanical stimulation is considered an important regulator 3, 4. However, this approach is heavily dependent on equipment, and it is mainly suitable for three‐dimensional (3D) constructs 5. Additionally, because many previous studies utilized custom‐made bioreactors, it is difficult to define the inductive conditions 6. In addition to mechanical stimulation, other physical parameters, such as extracellular matrix (ECM) topography, stiffness, and the influence of decellularized matrices on tenogenesis, have shown promise 7, 8, 9, 10. However, the fabrication of nanometer‐to‐micrometer scale topography usually requires sophisticated instrumentation, and the high inter‐batch variability is another concern. Another important approach in treating ligament and skeletal muscle injuries is the use of endogenous platelet rich plasma (PRP) 11, which was recently demonstrated to promote the proliferation and myogenic differentiation of myoblasts in vitro when combined with a TGF‐β inhibitor 12. However, controversial conclusions and variable clinical outcomes following PRP treatment for tendon repair have been reported 13, 14, 15. Additionally, variability in the preparation and composition of PRP makes it difficult to compare results across studies 16. In contrast, bioactive molecules, particularly recombinant growth factors, are readily available, stable, and dose‐controllable. Moreover, they are widely utilized in tissue engineering, and they have demonstrated much potential for translational applications.
With respect to osteogenesis and chondrogenesis, we reached the consensus that specific growth factor formulations exert potent therapeutic effects. However, formulations that are reproducible and can induce tenogenesis consistently remain elusive. Because stem cells can differentiate into specific lineages in vitro, they are potential stable source for regenerative medicine. Therefore, to address the requirements of tenogenic induction protocols, this review will focus on the current progress in the utilization of bioactive molecules in tenogenesis, which may have critical implications for the fields of tendon tissue engineering and future clinical translation.
Bioactive Molecules Regulating Tenogenesis of Stem Cells
Connective Tissue Growth Factor (CTGF/CCN2)
Connective tissue growth factor (CTGF) has been demonstrated to induce the differentiation of human bone marrow stromal cells (BMSC) into fibroblasts, which is evidenced by the marked increase in type I collagen (COL I) and tenascin‐C (TN‐C) synthesis as well as the elevated expression of typical fibroblastic markers and the reduced capacity to differentiate into non‐fibroblastic lineages, including osteocytes, chondrocytes, and adipocytes 17, 18. In rats, the in vivo control‐released delivery of CTGF enabled postnatal connective tissues to undergo fibrogenesis instead of ectopic mineralization 18. However, it was also reported that CTGF led to mineralization in the human periodontal ligament stem cell line, but the expression of fibroblast‐related genes was enhanced when it was combined with TGF‐β1 19. These results suggested the biphasic effects of CTGF, which could induce either fibroblastic or osteoblastic differentiation, depending on specific micro‐environmental conditions. These findings have implications for engineering fibrous tissue because they provide evidence for a reproducible protocol for the fibroblastic differentiation of mesenchymal stem cells (MSCs). The fibroblastic differentiation of stem cells is highly significant in the engineering of tendons.
Rat tendon stem/progenitor cells (TSPC) treated with ascorbic acid and CTGF for 2 weeks exhibited the increased expression of tendon‐specific markers and the significantly decreased expression of chondrogenic and osteogenic markers 20, 21. Additionally, engineered scaffold‐free tendon tissues formed by TSPC sheets after treatment with CTGF and ascorbic acid, distinctly enhanced tendon healing in a rat patellar tendon window injury model 20. Similarly, TSPC formed cell sheets after treatment with ascorbic acid and CTGF were evaluated in a rat anterior cruciate ligament reconstruction model, and the improved early graft healing was evidenced by better graft osteo‐integration and enhanced integrity of the intra‐articular graft recovery 22. CTGF delivery has also been used selectively to enrich tendon‐resident CD146+ stem cells during the early phase of tendon healing in rats. Subsequently, CTGF‐pretreated CD146+ progenitor cells were transplanted to exert positive effects on tendon regeneration 23.
FAK/ERK1/2 signaling pathway was reported to regulate the CTGF‐induced proliferation and differentiation of CD146+ stem/progenitor cells 23. However, it was unclear that how CTGF‐regulated tenogenic differentiation was linked with TGF‐β signaling pathway. Further investigation of CTGF and its potential crosstalk with TGF‐β will likely provide an in‐depth understanding of its roles in tendon regeneration.
Hence, CTGF may contribute to the preservation of the structural integrity of tendon tissue, which implies its potential use as a therapeutic agent in tendon regeneration. After treatment with CTGF, the TSPC sheets could be used to promote graft healing in reconstruction, and the engineered scaffold‐free tendon tissue could be a novel approach to tendon repair and regeneration. Treatment with 100 ng/ml of recombinant human CTGF and 50 μg/ml ascorbic acid for 4 weeks was demonstrated to have the best effect on monolayer‐cultured MSCs 17, 18. When the TSPCs were treated, the treatment could be adjusted to 25 ng/ml for 2 weeks 20, 21.
Transforming Growth Factor Beta (TGF‐β)
The transcriptome analysis of developing mouse limb tendon cells showed that TGF‐β was the most significant signaling pathway and exhibited the largest upregulation in differentiated tendon cells compared with TSPCs 24. In addition, Scx‐GFP signaling, which gradually disappeared when TSPCs were cultured in vitro, was reported to reappear upon TGF‐β treatment 25, 26. Moreover, it was demonstrated that the tendon‐specific transcription factors Mkx and Egr1 acted partially through the TGF‐β signaling pathway 25, 27. Combined, these results demonstrated the pivotal role of TGF‐β signaling in tendon induction.
TGF‐β1 enabled equine tenocytes to retain a differentiated state 28. It also highly upregulated neotendon transcription factors Scleraxis (Scx) and Mohawk (Mkx) expression 29, and significantly impeded the decline of mechanical properties 30. Treatment with TGF‐β2 was able to increase the expression of Col1a1 and Scx genes 24, 25, 27, which were significantly downregulated by specific TGF‐β inhibitors 24. It was also demonstrated that TGF‐β2‐loaded affigel beads that were grafted into limb buds in an organ culture, robustly induced Scx expression during tendon development 31. TGF‐β3 was reported to promote tendon differentiation of equine embryo‐derived stem cells 32. Moreover, in vivo studies showed that human MSC and bone marrow‐derived mononuclear cells had the capacity to generate tendon‐like tissues when treated with TGF‐β3 33.
The TGF‐β signaling pathway is involved in multiple cellular functions, including cell growth, cell differentiation, and cellular homeostasis. TGF‐β1 and the insulin‐like growth factor 1 (IGF‐1) were reported to enhance the mechanical properties of rabbit patellar tendons at 2 weeks post‐surgery 34. TGF‐β was also reported to facilitate differentiation of human keratocytes into myofibroblasts, but TGF‐β‐mediated improper fibrosis and scar formation limited its use in human application. Recently, single‐cell analysis reveals the potential of IGF‐1 to inhibit the TGF‐β/Smad pathway of fibrosis in human keratocytes in vitro 35. Additionally, TGF‐β signaling was also reported to play essential roles in cartilage formation and maintenance 36. Thus combined administration of growth factors and guided tenogenesis has gained significant interest in recent years. Recently, it was demonstrated that the combination of tendon‐derived ECM extract with TGF‐β3 enhanced tenogenic differentiation of human adipose‐derived mesenchymal stem cells (ADSCs) 37.
The TGF‐β/Smad signaling axis is one of the main TGF‐β downstream cascades. It was demonstrated that TGF‐β signaling was sufficient and required via Smad2/3 to drive mouse mesodermal stem cells toward the tendon lineage 24. The embedment of Smad8/BMP2–engineered MSCs was also reported to result in higher effective stiffness than in the control groups in a full‐thickness Achilles tendon defect model at 3 weeks post‐surgery 38. Moreover, although TNF‐α inhibited the proliferation and tenogenic differentiation of TSPCs, the expression of tenogenic‐related marker genes and the proliferation of TSPCs were significantly increased after simultaneous or sequential treatment with TGF‐β1 and TNF‐α. 39. During the processes, the TGF‐β and BMP signaling pathways were highly activated as evidenced by highly phosphorylated Smad2/3 and Smad1/5/8 39. It was also demonstrated that the addition of TGF‐β3 to tenocytes minimized extrinsic scarring, decreased tendon adhesion and promoted tendon healing by significantly downregulating the expression of Smad3 and upregulating the expression of Smad7 40.
These results indicated that the local delivery of TGF‐β may accelerate the healing process and play a significant role in tendon‐to‐bone healing. Treatment with 20 ng/ml of TGF‐β for 24 hours was demonstrated to be sufficient to induce the tenogenic differentiation of monolayer‐cultured MSCs 24, 27. We can concluded that adult stem cells are able to differentiate into a therapeutically relevant cell type and that the TGF‐β driven differentiation of stem cells may provide a model for studying tendon development and better understanding the transcriptional networks that are involved in tendon cell differentiation in different developmental stages.
The Growth Differentiation Factor (GDF) Family
GDF‐5 (BMP‐14), GDF‐6 (BMP‐13), and GDF‐7 (BMP‐12), which belong to the TGF‐β superfamily, are essential in tendon development and differentiation 41. GDF‐5 was shown to induce the tenogenesis of rat ADSCs, resulting in an enhanced ECM and tenogenic markers 42, 43. Similar effects of GDF‐5 were reported on human BMSCs 44, 45 and periodontal ligament‐derived cells 46. Additionally, following GDF‐5 induction, the obvious downregulation of the non‐tenogenic marker genes (RUNX2 and SOX9) was observed, which excluded human BMSCs differentiation into other lineages 44. GDF‐6 was reported to have a positive effect on the tenogenic differentiation of rat BMSCs with the significant upregulation of specific markers (Scx and Tnmd) 47. It was demonstrated that treatment of the C3H10T1/2 mouse mesenchymal progenitor cell line with GDF‐6 or GDF‐7 led to the dose‐dependent induction of Thbs4 expression 48. Moreover, different mesenchymal stem cell lineages exhibited different tenogenic differentiation capacities in the presence of GDF‐7, in which ADSCs exhibited inferior capacity 49. However, GDF‐7 stimulated the expression of tenocyte lineage markers and was used to promote tenogenic differentiation in rat TSPCs 50 and BMSCs 51, 52, as well as in canine and mouse ADSCs 53. In horse, BMSCs also differentiated into tenocytes after treatment with GDF‐7 54. The GDF‐7‐releasing sutures also enhanced Achilles tendon healing and reduced adhesions and scars 55.
GDF‐5 also promoted the osteogenic‐lineage differentiation of stem cells in vitro, which was demonstrated by histology and biochemical assays for alkaline phosphatase activity as well as the analysis of gene expression 56. Although the local delivery of GDF‐5 by coating sutures in rats showed a promising beneficial effect on tendon repair, which was evidenced by the improved histological and mechanical properties in an early stage, the subsequent appearance of cartilage‐like structures demonstrated the chondroinductive capacity of GDF‐5 57, 58. The effect of GDF‐5 in rat ADSCs is similar to that of GDF‐7, but it is marginally less effective (based on the expression of Scx and Tnmd), and the effects with selective (based on downregulation of Bglap) 53. However, neither GDF‐7 nor GDF‐5 promoted Col1a1 and Col3a1 expression in ADSCs 53.
To date, the molecular signaling mechanisms which regulate GDFs‐induced tenogenic differentiation remain largely unknown. BMPs are known to signal through the canonical Smad pathway and the non‐canonical mitogen‐activated protein kinase (MAPK) pathway. Although some research investigated the downstream signaling pathways of GDF‐5, GDF‐6, and GDF‐7, such as changes in Smad‐1/5/8 expression or activation level 48, 53, few studies have focused on the underlying molecular mechanism of tenogenesis. Recent research demonstrated that the reorganization of the cytoskeleton plays an important role in tenogenic differentiation induced by GDF‐5. The genes that are mainly related to cytoskeleton reorganization signaling were identified as potentially associated with tenogenic differentiation 59.
Hopefully, the adverse side effect might be overcome by modifying the dose, the suture material, or the micro‐environment. It was reported that GDF‐5 was capable of inducing tendon‐like tissues in a synthetic 3D microenvironment in vivo, based on tendon‐specific genes and the histological and morphological characteristics of the tissue 45. The optimal concentration of GDFs 5, 6, 7 varies from 10 to 100 ng/ml, and the treatment time is difficult to determine because of the different cell types. It was shown that low‐dose GDF‐7 within an absorbable collagen sponge was feasible and likely safe as an adjuvant therapy for rotator cuff repair 60. The clinical relevance of these results is that accelerated repair might reduce rehabilitation time, leading to an earlier return to work and recreational activities.
Basic Fibroblast Growth Factor (bFGF/FGF‐2)
FGF‐2 combined with platelet‐derived growth factors (PDGF‐BB) has been reported to promote cell proliferation and collagen production significantly in canine flexor tendon fibroblasts 61. Low dosages of FGF‐2 triggered a biphasic human BMSC response. Cell proliferation was significantly stimulated in the early stage, and cell differentiation was stimulated in the late stage with upregulated COL I, COL III, and FN 62. The delivery of FGF‐2 through various scaffolds was also shown to induce the tenogenic differentiation of MSCs 63, 64, 65. In the process of healing rat rotator, the FGF‐2‐treated group demonstrated a significant enhancement of biomechanical strength and histological appearance 66. Additionally, FGF‐2 stimulated the proliferation of the tenogenic stem cells in tendon‐to‐bone healing, which led to the better recovery of the repaired rotator cuff 66.
FGF signaling has been reported to induce the formation of a tendon progenitor population that expressed Scx during somite development 67. The application of FGF‐2 on flexor tendon healing in a canine model resulted in enhanced neovascularization and reduced inflammation in the earliest stages, but it did not result in any positive effect on the mechanical and functional properties of the repaired tendon 68. Moreover, it was also reported that FGF‐4 did not increase Scx expression in mouse limbs in both early and late developmental stages and that it showed negative effects on Scx and Col1a1 gene expression in C3H10T1/2 cells 69, 70.
FGF signaling pathway played distinct roles in limb tendon development between the chick and mouse models. ERK inhibition was reported to prime mouse stem cells for the tendon lineage sufficiently 24. More researches are needed to understand the differences of FGF/ERK pathway in tendon development and regeneration.
In summary, FGF‐2 plays a crucial role in cell proliferation and collagen production. And it is usually used at a low concentration of less than 10 ng/ml 62. Low‐dose FGF‐2 may be useful for tissue engineering of tendons by promoting stem cell proliferation and increasing mRNA expression of specific ECM proteins and cytoskeletal elements. But its effect on the mechanical and functional properties of the repaired tendon remains elusive.
Others
Adiponectin, an adipocyte‐secreted hormone that increases cell sensitivity to insulin and protects against type II diabetes, was found to promote progenitor cell differentiation and proliferation in tendons concurrently. After exposure to recombinant adiponectin in vitro, diabetic‐ridden human TSPCs exhibited significantly upregulated tenogenic gene expression with unaltered levels of adipogenic, chondrogenic, and osteogenic gene expression 71.
The intra‐tendon delivery of PDGF‐BB in a rat Achilles tendinopathy model was reported to increase maximum load‐to‐failure and stiffness with respect to the control and the steroid treatment 72, 73. However, there were no significant differences in histology, and a long‐term evaluation was not conducted. Although PDGF‐BB delivery was demonstrated to promote early healing in a rat rotator‐cuff repair model, there was an absence of either a better structure or stronger mechanical properties in later stages of healing 74.
Because the expression of MAPK or MAP kinase and MAP kinase phosphatases was downregulated in developing tendon cells, a reduction in MAPK activity during tendon differentiation was implicated. The PD18 inhibitor blocked the ERK/MAPK pathway and induced the activation of Col1a1, Col1a2, Scx, and Thbs2 expression in mouse E9.5 limb explants and MSCs 24. However, another MAPK signaling inhibitor U0126 did not show any positive effect on tenomodulin (Tnmd) expression of MSCs 75.
The loss of mesenchymal Wls, which is necessary for the secretion of multifarious Wnts, prevents the differentiation of distal mesenchyme. The regulatory role of Wnts in undifferentiated distal limb mesenchyme cells was investigated 76. Wnts were demonstrated to activate the Scx promoter and to be crucial for the induction of distal tendon/ligament 76. Hence, the Wnt protein is a potent inducer of Scx expression and tendon differentiation. Another study of Wnt signaling involved the treatment of BMSCs with the glycogen synthase kinase‐3 (GSK‐3) inhibitor‐BIO. In 3D BIO‐containing collagen gel, BIO induced the expression of Tnmd, Col14a1, Decorin (Dcn), and fibromodulin (Fmod), but not in monolayer BMSCs 75. The inhibition of GSK‐3 with siRNA corresponding to GSK‐3 also resulted in the upregulation of Tnmd, which suggested that Wnt/β‐catenin signaling mediated the expression of Tnmd.
Comparative Studies of Bioactive Molecules
Although many studies have used growth factor supplements to induce tenogenesis with varying degrees of success, their efficacy is difficult to evaluate because of the different cell types, varying durations, and different readout parameters. Hence, it is necessary to compare the bioactive molecules directly in order to evaluate and determine which is the most potent in inducing tenogenic differentiation.
Recently, the tenogenic potential of FGF‐4, TGF‐β3, TGF‐β2, and PD18 was evaluated in MSCs. It indicated that TGF‐β signaling was more effective than the other two growth factors in inducing tenogenic differentiation 24. Additionally, TGF‐β2/3 signaling suppressed the cartilage marker Sox9 significantly. However, neither differences in the effects of TGF‐β2 versus TGF‐β3 nor synergetic effects were observed 24. Similarly, another study showed negligible effects of FGF‐4 on the differentiation of TSPCs that were isolated from the axial and limb in a series of developmental stages 69. This study also analyzed the gene response profiles of TSPCs to TGF‐β2, FGF‐4, mechanical loading, and their combinations. Based on Scx expression, TGF‐β2 was demonstrated to induce tenogenesis of TSPCs in all stages, whereas mechanical loading affected the late‐stage TSPCs. When the growth factors were applied in combination, TGF‐β2 continued to be tenogenic, whereas FGF‐4 appeared anti‐tenogenic 69. In contrast, it was revealed recently that FGF‐4 significantly increased Scx expression, whereas PD18 decreased Scx expression in undifferentiated cells of chick limb. However, TGF‐β2 positively regulated Scx expression as previously described. This result demonstrated that the function of TGF‐β in tendon development was similar in chick and mouse limbs, whereas FGF‐4 and TGF‐β2 showed independent effects 77. Another study on the tenogenic differentiation capacity of human ADSCs and amniotic fluid stem cells (AFSC) evaluated the growth factors EGF, bFGF, PDGF‐BB, and TGF‐β1 78. Based on the results of Col I, Col III, Dcn, Tn‐C, and Scx expression, it was shown that TGF‐β1 induced Scx expression 10 times more than other growth factors did on day 7; EGF induced the greatest amount of Tn‐C expression on day 7 and Col III expression on day 14; TGF‐β1 and PDGF‐BB induced Col I expression 10 times more than other growth factors did on day 21. The expression of genetic markers showed different patterns in human AFSCs and ADSCs, which could indicate that the tenogenic lineage commitment induced by biochemical signals might be affected by the origin of the stem cells 78.
Our recent study investigated the treatment of rat BMSCs with TGF‐β1, BMP‐12, CTGF, and their combinations, which showed that TGF‐β1 treatment led to the greatest upregulation of tenogenic genes. It was observed that TGF‐β1 combined with CTGF elevated Tnmd mRNA and protein expression in the late stages, so a stepwise differentiation strategy was developed 79. It was also reported that Scx‐GFP expression levels decreased over time in primary tenocytes after culture in vitro 26. In addition to the induction of lineage‐specific differentiation, the maintenance of a tenocyte lineage phenotype should receive increased attention and consideration in the research. It was demonstrated that treatment with TGF‐β and GDF‐8 resulted in the maintenance of Scx‐GFP expression in adult tenocytes. 26.
However, the varying patterns of gene expression induced by growth factors may be too complicated to provide any valuable use in future applications. Because the immunofluorescence results for the detection of tendon ECM proteins exhibited no significant difference between each culture condition, it was difficult to reach a conclusion without quantitative data. Although the general conclusion was that both stem cell types could be biochemically induced to the tenogenic lineage, the most suitable growth factors and the proper duration for stimulation are still unclear. Hence, further research is needed to provide comprehensive evidence for the tenogenic differentiation efficacy of a range of bioactive molecules in order to reach a consensus on future therapeutic applications.
Challenges and Future Directions
Elucidation of Molecular Regulatory Mechanisms of Tenogenesis
The formation of highly organized tendon tissue structures relies on the precise temporal‐spatial regulation of signaling pathways. The knowledge about the mechanisms of the signaling pathways that regulate the tenogenic master gene Scx is limited. It has been demonstrated that TGF‐β/Smad2/3 signaling pathways play an important part in this activity. Upregulated Scx by canonical TGF‐β2 signaling was attenuated by MAPK activity, which indicated that the convergence of MAPK signaling to this pathway had a negative effect 80. Moreover, the bioinformatics analysis of tendon transcriptome in diverse developmental stages showed that the two most crucial signaling pathways for tenogenesis were TGF‐β and MAPK 24. TGF‐β signaling was demonstrated to be either pro‐tenogenic or pro‐chondrogenic depending on the cell type and culture conditions 31, 81, 82, 83.
The roles of TGF‐β signaling in tendon homeostasis and tendon development have been elucidated, while the roles of Wnt/β‐catenin signaling in tendon remain mostly elusive. Recently, a novel regulatory mechanism was found in equine BMSCs, suggesting that Wnt/β‐catenin signaling mediated the expression of Tnmd, whereas Scx and Mkx were not affected 75. During this process, tendon‐related ECM components were also upregulated, which showed that the activation of Wnt/β‐catenin signaling induced the tenogenic differentiation of BMSCs. However, it was also demonstrated that the activation of Wnt/β‐catenin signaling in TSPCs suppressed the expression of Scx, Mkx, and Tnmd and reduced the amounts of Smad2 and Smad3, which are intracellular mediators in TGF‐β signaling 84. Combined, these results indicated that Wnt/β‐catenin signaling might have completely different effects on different type of cells, which warrants further elucidation.
Identification of Developmental Stage‐Specific Tendon Markers
The deeper understanding of tendon biology in terms of development and morphogenesis would advance the establishment of an optimal inductive condition for tenogenesis in vitro 85. The identification and characterization of TSPCs in tendon tissues would constitute a milestone step in understanding tendon physiology, thus facilitating progress in this field 86. Currently, there is a dire need for specific and definitive markers to identify TSPCs. At present, TSPCs have almost all the same markers that are present in MSCs, such as the positive expression of CD73, CD90, and CD105, as well as the negative expression of CD31, CD34, and CD45. Although TSPCs express higher levels of the commonly utilized tenogenic‐related markers, such as Scx, Tnmd, and Col I, these genes could not distinguish TSPCs from tenocytes. Because of the lack of definitive developmental stage‐specific markers, it is difficult to evaluate the effects of tenogenic inducers.
Advances in high‐throughput screening technologies have brought about the revolution of big data. Combined with bioinformatics analysis, these technologies provide powerful tools for identifying developmental mechanisms and potential markers for each developmental stage. Several public released datasets are available on the Gene Expression Omnibus (GEO) database. These microarrays and RNA sequencing (RNA‐seq) datasets thus provide a valuable resource for tendon research. The extensive analysis of regulated genes during limb development will provide scientists with the in‐depth understanding of the expression dynamics that govern limb morphogenesis. However, the currently available whole‐genome microarray and RNA‐seq datasets mainly comprise the initial stages of limb development. However, postnatal development stages, including the critical maturation transition from fetal tendon to adult tendon tissues, should be taken into consideration. Because adult tissue regeneration is generally considered to recapitulate developmental processes, insights gained from research in developmental biology have been considered to be important for teno‐lineage differentiation in vitro 87. A recent study successfully developed a protocol that recapitulated the various differentiation stages of developmental skeletal myogenesis, which represents an advance in the field by improving the efficiency and timing of the generation of striated, contractile fibers from mouse and human pluripotent stem cells 85. Additionally, the stage‐specific markers established in developmental biology will provide useful information for evaluating the various tenogenesis stages in vitro. The comparison of RNA‐seq in the gene expression profiles of Scx‐GFP positive cells and Scx‐GFP negative cells from E13.5 forelimb samples identified the novel transcription factor Foxf2 as a specific marker of differentiated limb tendon 88. The comparison of genes that were expressed in adult rat and human tendon tissue with those in other musculoskeletal tissues showed that Thbs4 and Tnmd exhibited the most tendon‐specific expression 89. Based on these findings, definitive markers could serve as useful reporters of tenogenesis evaluation in vitro and in vivo.
Moreover, recent molecular studies found that individual cells exhibited substantial differences in gene expression, even in a seemingly homogenous population. The use of imaging multi‐color flow cytometry has been demonstrated to help researchers to visualize and quantify the reactions of individual cultured cells to bioactives or other physiological impulses and to evaluate the potency of these bioactives in stimulating muscle commitment 90. The cutting‐edge technology used in single‐cell quantitative gene analysis combined with high‐throughput arrays has allowed the analysis of gene expression profiles to investigate the heterogeneity of cell populations 91, 92, 93. This high‐content microfluidic real‐time platform is useful in comparative studies on the regulation of developmental processes in single cells 94, 95. This method overcomes the limitations of heterogeneous cell populations and small sample quantities, and it is potentially useful for analyzing the differential regulation of gene expression in normal versus diseased tissues and organs. The nestin+ tendon stem cells subpopulation was identified by single‐cell gene analysis 96. Technical improvements have enabled the RNA sequencing of single cells, and an increasing number of studies have utilized the power of single‐cell analysis in reconstructing cellular lineages in several tissues and revealing sequential stem cell states 97, 98, 99. Hence, high‐throughput single‐cell qRT‐PCR and single‐cell RNA sequencing have important implications for the characterization of previously unidentified sub‐populations and for the further analysis of the mechanisms of stem cell self‐renewal and differentiation in tendon biology 100.
Development of Small Molecule Cocktails for Stepwise Tenogenic Induction
Induced pluripotent stem cells (iPSC) is currently a “hot” topic in stem cell research. These stem cells are obtained by reprogramming differentiated somatic cells through the recombinant overexpression of specific transcription factors. Such advances and breakthroughs in cellular reprogramming indicate the reversibility of cell lineage fate 101, 102. However, genetic manipulation and the use of viral vectors in reprogramming have evoked concern about the safety of iPSCs in therapeutic applications. Recently, an increasing number of studies have identified small molecules that can substitute reprogramming factors as well as enhance reprogramming efficiency, which could overcome the limitations of traditional reprogramming protocols and achieve cellular reprogramming efficiently and safely 103, 104. Recent reports showed that neural progenitor cells and neurons were induced from fibroblasts by using the appropriate chemical cocktails 105. Moreover, small molecule‐based direct lineage‐specific conversion from mouse or human fibroblasts to various cell lineages has been achieved 106, 107, thus demonstrating the feasibility of a pure small molecule‐based approach to reprogramming. These studies have led to new possibilities for reprogramming cell lineage fate by using chemical agents. To date, only a few of these small molecules have demonstrated tenogenic potential. Recently, the retinoic acid receptor agonist was found to maintain tendon stem cell characteristics 108. The strategy for inducing tendon differentiation from pluripotent or multipotent stem cells could be designed based on developmental processes in tendons. In order to recapitulate the stage‐specific regulatory signaling during tendon morphogenesis, stepwise approaches should be taken into consideration. In the future, we anticipate that chemical screening will be employed to identify small molecule cocktails for teno‐lineage‐specific differentiation, which will provide a step toward therapeutic applications.
Conclusion
In this review, we described the biochemical strategies of tenogenic differentiation (Table 1) and the direct comparative studies on them (Table 2). The delivery method, timing, and duration of the administration of the bioactive factors should be considered in order to achieve the best healing results and eliminate adverse side effects. Furthermore, the efficiency and efficacy of soluble factor treatment was shown by the confirmatory results of pairwise studies. We also discussed potential molecular regulatory mechanisms. The data obtained in these studies demonstrated that TGF‐β/Smad, MAPK/ERK, Wnt/β‐catenin, and so on establish a complex signaling network that directs tenogenic differentiation. Because of its potential relevance in medicine, we highlight the importance of the combined influence of the TGF‐β and MAPK pathways in tenogenic differentiation. We hope that our review will give readers a better understanding of the regulation of bioactive molecules in tendon regeneration. We also hope that this review will guide the development of optimal small molecule cocktails for the stepwise induction of tenogenic differentiation. Based on the findings of this review, the role of bioactive molecules in guiding stem cell fate in tendon regeneration warrants further exploration.
Table 1.
Supplementation | Target cell | Optimal concentration | Treatment time | Outcome | References |
---|---|---|---|---|---|
CTGF with ascorbic acid | Human BMSC | 100 ng/ml | 4 weeks | COL I and TN‐C contents↑ | Lee CH, et al. Conf Proc IEEE Eng Med Biol Soc 2006 |
Human BMSC | 100 ng/ml | 4 weeks | COL I, COL III, TN‐C, FN, VIM, MMP‐1 (4–10 folds↑); COL I and TN‐C contents↑ | Lee CH, et al. J Clin Invest 2010 | |
Rat TSPC | 25 ng/ml | 2 weeks | Tnmd, Scx, Thbs4 (1–2 folds↑); Col1a1, Eln, Dcn, Bgn (1–3 folds↑) | Ni M, et al. Biomaterials 2013 | |
Rat TSPC | 25 ng/ml | 2 weeks | Col1a1, Tnmd ↑; chondrogenic (Acan, Col2a1) and osteogenic (Alpl) markers↓ | Lui PP, et al. Cytotherpy 2016 | |
Rat CD146+ tendon cell | 100 ng/ml | 1 week | Col1a1, Col3a1, Tn‐C, Vim, Tnmd and Scx (3–25 folds↑) | Lee CH, et al. J Clin Invest 2015 | |
TGF‐β1 | Mouse flexor tendon tenocyte | 10 or 100 ng/ml | 24, 48 hours | Mmp‐16, Dcn↓; Bgn, Col V, Col XII, Serpine1, Scx, and Mkx (5–10 folds↑) | Farhat YM, et al. PLoS One 2012 |
TGF‐β2 | Mouse MSC | 20 ng/ml | 1, 24 hours | Scx (6–8 folds↑) and Col1a1 (3–4 folds↑) | Guerquin MJ, et al. J Clin Invest 2013 |
Mouse TSPC and MSC | / | 12, 24, 48 hours | Scx (6–8 folds↑), Col1a1 (3–4 folds↑), Col5a1 (2–3 folds↑) at different time points | Liu H, et al. Stem Cells 2015 | |
TGF‐β2/3 | Mouse MSC (C3H10T1/2) | 20 ng/ml | 24 hours | Scx (6–8 folds↑), Col1a1(3–4 folds↑), Sox9 (4–6 folds↓) | Havis E, et al. Development 2014 |
TGF‐β3 | Human bone marrow‐derived mononuclear cells | 20 ng/ml | 7 days | Synthesis of collagen fibrils and appearance of fibripositors | Kapacee Z, et al. Matrix Biol 2010 |
Equine embryo‐derived stem cells | 20ng/ml | 72 hours | Scx, Tn‐C, Col1a1, and cartilage oligomeric matrix protein (Comp) ↑ | Barsby T, et al. Tissue Eng Part A 2013 | |
GDF‐5 (BMP‐14) | Rat ADSC | 100 ng/ml | 3, 6, 9, and 12 days |
ECM (Col I, Dcn, Acan) and tenogenic marker gene (Scx, Tnmd, and Tn‐C) expression↑ Tnmd, Tn‐C, Smad‐8, and Mmp‐13 protein expression↑ |
Park A, et al. Tissue Eng Part A 2010 |
Human BMSC | 100 ng/ml | 4 days | COL I, SCX, and TN‐C expression (2.31 ± 0.27, 2.30 ± 1.81 and 3.55 ± 0.27 folds↑) | Tan SL, et al. Cells Tissues Organs 2012 | |
P4–6 periodontal ligament‐derived cells | 10 ng/ml | 3 weeks | SCX (2 folds↑), TNMD markedly, ACAN and COL III (4–8 folds↑) | Xia D, et al. Growth Factors 2013 | |
Human BMSC | / | 3 days | COL1A1, COL3A1, DCN, SCX, and TN‐C↑ | Govoni M, et al. Tissue Eng Part A 2017 | |
GDF‐6 (BMP‐13) | Mouse BMSC | 100 nM | 3 days | Col I (3 folds↑), Thbs4 (10 folds↑) | Berasi SP, et al. Growth Factors 2011 |
Rat BMSC | 20 ng/ml | 2 weeks |
Tnmd (5.61 folds↑) and Scx (4.18 folds↑), Tnmd protein expression↑ |
Chai W, et al. Chin Med J (Engl) 2013 | |
GDF‐7 (BMP‐12) | Horse BMSC | 50 ng/ml | 14–21 days | Tnmd and Dcn expression | Violini S, et al. BMC Cell Biol 2009 |
Mouse BMSC | 100 nM | 3 days | Col I (3 folds↑), Thbs4 (12 folds↑) | Berasi SP, et al. Growth Factors 2011 | |
Rat BMSC | 10 ng/ml | 12 hours | Scx, Tnmd, Tn‐C, Col I (2–4 folds↑) | Lee JY, et al. PLoS One 2011 | |
Rat BMSC | 50 ng/ml | 2 weeks | Scx (2.56 folds↑), Tnmd (3.07 folds↑), Tn‐C (1.88 folds↑), Tnmd protein expression↑ | Ni M, et al. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2011 | |
Canine/Mouse ADSC | 100 or 1,000 ng/ml | 3, 7, 14 days | Scx, Tnmd gene, and protein expression↑ | Shen H, et al. PLoS One 2013 | |
Rat TSPC | 50 ng/ml | 14 days | Scx, Tnmd, Col I, and Tn‐C gene, and protein expression↑ | Liu J, et al. Cell Physiol Biochem 2015 | |
FGF‐2 | Human BMSC | 3 ng/ml | 14 days | COL I, COL III, FN expression (1–2 folds↑) | Hankemeier S, et al. Tissue Eng 2005 |
FGF‐2 or PDGF‐BB | Canine flexor tendon fibroblast | 10 ng/ml | 24 hours | Cell proliferation and collagen production↑ | Thomopoulos S, et al. J Hand Surg Am 2005 |
PD18 (a mitogen‐activated protein kinase inhibitor) | Mouse MSC | 3.3 µM | 24 hours | Scx and Col1a1 (∼2 folds↑) | Havis E, et al. Development 2014 |
Wnt 3a | Mouse MSC | / | 48 hours | Scx promoter, luciferase assay | Zhu X, et al. Development Biology 2012 |
BIO (a glycogen synthase kinase‐3 inhibitor) | Equine BMSC | 10 μM | 7 days | Tnmd (∼3 folds↑), Col14a1, Dcn, and Fmod gene expression↑ | Miyabara S, et al. J Equine Sci 2014 |
BMP2 + Smad8 | Mouse MSC | / | 4, 7, 10, 14 days | Morphological characteristics and gene expression profile of tendon cells both in vitro and in vivo | Hoffmann A, et al. J Clin Invest 2006 |
Recombinant human adiponectin | Diabetic ridden human TSPC | 10 μg/ml | 48 hours | COL I, III, TNMD, and SCX expression (2.0 ± 0.3, 1.73 ± 0.25, 1.63 ± 0.21 and 1.74 ± 0.24 folds↑) | Rothan HA, et al. Int J Med Sci 2013 |
Abbreviations: /, not determined; ADSC, adipose‐derived mesenchymal stem cells; BMSC, bone marrow stromal cells; COL I, type I collagen; CTGF, connective tissue growth factor; FGF, fibroblast growth factor; GDF, growth differentiation factor; MSC, mesenchymal stem cells; PDGF‐BB, platelet‐derived growth factors; TGF‐β, transforming growth factor beta; TN‐C, tenascin‐C; TSPC, tendon stem/progenitor cells.
Table 2.
Comparison | Target cell | Outcome | References |
---|---|---|---|
FGF‐4, PD18 versus TGF‐β2/3 | Mouse MSC |
TGF‐β signaling was sufficient to drive mouse mesodermal stem cells toward the tendon lineage ex vivo and in vitro Examination of the tendon markers Scx (6–8 folds↑), Col1a1 (3–4 folds↑) and the cartilage marker Sox9 (3–4 folds↓) ERK inhibition by PD18 activated Scx and Col1a1 (∼2 folds↑), and had no effect on Sox9 expression FGF does not have a tenogenic effect |
Havis E, et al. Development 2014 |
Mechanical forces, FGF‐4 versus TGF‐β2 | Mouse TSPC | Based on Scx expression, TGF‐β2 was tenogenic for TSPCs at all stages, while loading was for late‐stage cells only, and FGF‐4 had no effect | Brown JP, et al. J Biomech 2014 |
Mechanical forces, FGF‐4 versus TGF‐β2 | Mouse MSC and TSPC | TGF‐β2 alone and combined with loading were tenogenic based on increased Scx levels in both MSCs and TSPCs. Loading alone had minimal effect. FGF‐4 downregulated tendon marker levels in MSCs but not in TSPCs | Brown JP, et al. Stem Cell Res Ther 2015 |
BMP‐2 versus TGF‐β1 versus GDF‐5 | Human MSC |
TGF‐β1 and GDF‐5 show similar COL I (3–5 folds↑), COL II (1–2 folds↑), and SOX9 (1–2 folds↑) expression BMP‐2: COL I (2–3 folds↓), COL II (100 folds↑), SOX9 (1–2 fold↑) |
Murphy MK, et al. Stem Cells 2015 |
EGF, bFGF, PDGF‐BB, and TGF‐β1 | Human ASC |
TGF‐β1 induced SCX expression 10 folds more than other growth factors on day 7 EGF induced TN‐C (10 folds↑) and COL III (3–5 folds↑) expression on day 14 TGF‐β1 and PDGF‐BB induced COL I expression 10 folds more than other growth factors on day 21 |
Gonçalves AI, et al. PLoS One 2013 |
TGF‐β1, −2 and −3; GDF‐5, −7 and −8; FGF‐4; IGF‐1; PDGF; EGF and VEGF | Scx‐GFP tenocyte |
TGF‐β1, −2 and −3 were the most potent and induced similar levels of Scx‐GFP expression. GDF‐8 was ∼4 folds less potent than the TGF‐βs Any other cytokines/growth factors examined had no effect |
Maeda T, et al. Curr Biol 2012 |
IGF‐1 versus TGF‐β3 | Human tenocyte |
Collagen synthesis and expression of Scx and Tnmd TGF‐β3 is more effective than IGF‐1 |
Qiu Y, et al. Cells Tissues Organs 2013 |
TGF‐β1, BMP‐12, CTGF, and their combinations | Rat BMSC | TGF‐β1 alone significantly and efficiently induced Scx expression and collagen production. TGF‐β1 combined with CTGF elevated Tnmd mRNA and protein expression on day 7 | Yin Z, et al. Stem Cells Translational Medicine 2016 |
Mechanical forces, FGF, TGF‐β versus PD18 | Chick undifferentiated limb cell |
Both FGF‐4 and TGF‐β2 promote tendon commitment and act downstream of mechanical forces PD18 decreased Scx expression |
Havis E, et al. Development 2016 |
Abbreviations: bFGF, basic fibroblast growth factor; CTGF, connective tissue growth factor; FGF, fibroblast growth factor; IGF‐1, insulin‐like growth factor 1; MSC, mesenchymal stem cells; PDGF‐BB, platelet‐derived growth factors; TGF‐β, transforming growth factor beta; TSPC, tendon stem/progenitor cells.
Author Contributions
Z.Y., G.L., and K.M.C.: conception and design; Y.J.Z., Z.Y., and X.C.: manuscript writing and editing, final approval of manuscript; H.W.O. and X.C.: critical discussion for manuscript; B.C.H.: final approval of manuscript.
Disclosure of Potential Conflicts of Interest
The authors indicated no potential conflicts of interest.
Acknowledgments
This work was supported by the National key research and development program of China (2017YFA0104902, 2016YFC1100204), NSFC grants (81330041, 81522029, 81772418, 31570987, 81401781, 81572157), Fundamental Research Funds for the Central Universities.
References
- 1. Ljungqvist A, Schwellnus MP, Bachl N et al. International Olympic Committee consensus statement: Molecular basis of connective tissue and muscle injuries in sport. Clin Sports Med 2008;27:231–239, x–xi. [DOI] [PubMed] [Google Scholar]
- 2. Meyerrose T, Olson S, Pontow S et al. Mesenchymal stem cells for the sustained in vivo delivery of bioactive factors. Adv Drug Deliv Rev 2010;62:1167–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Chen JL, Yin Z, Shen WL et al. Efficacy of hESC‐MSCs in knitted silk‐collagen scaffold for tendon tissue engineering and their roles. Biomaterials 2010;31:9438–9451. [DOI] [PubMed] [Google Scholar]
- 4. Chen X, Yin Z, Chen JL et al. Force and scleraxis synergistically promote the commitment of human ES cells derived MSCs to tenocytes. Sci Rep 2012;2:977. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Chen X, Yin Z, Chen JL et al. Scleraxis‐overexpressed human embryonic stem cell‐derived mesenchymal stem cells for tendon tissue engineering with knitted silk‐collagen scaffold. Tissue Eng Part A 2014;20:1583–1592. [DOI] [PubMed] [Google Scholar]
- 6. Chen JL, Zhang W, Liu ZY et al. Physical regulation of stem cells differentiation into teno‐lineage: Current strategies and future direction. Cell Tissue Res 2015;360:195–207. [DOI] [PubMed] [Google Scholar]
- 7. Yin Z, Chen X, Chen JL et al. The regulation of tendon stem cell differentiation by the alignment of nanofibers. Biomaterials 2010;31:2163–2175. [DOI] [PubMed] [Google Scholar]
- 8. Zhang C, Yuan H, Liu H et al. Well‐aligned chitosan‐based ultrafine fibers committed teno‐lineage differentiation of human induced pluripotent stem cells for Achilles tendon regeneration. Biomaterials 2015;53:716–730. [DOI] [PubMed] [Google Scholar]
- 9. Yin Z, Chen X, Zhu T et al. The effect of decellularized matrices on human tendon stem/progenitor cell differentiation and tendon repair. Acta Biomater 2013;9:9317–9329. [DOI] [PubMed] [Google Scholar]
- 10. Yin Z, Chen X, Song HX et al. Electrospun scaffolds for multiple tissues regeneration in vivo through topography dependent induction of lineage specific differentiation. Biomaterials 2015;44:173–185. [DOI] [PubMed] [Google Scholar]
- 11. Kon E, Filardo G, Di MA et al. Platelet‐rich plasma (PRP) to treat sports injuries: Evidence to support its use. Knee Surg Sports Traumatol Arthrosc 2011;19:516–527. [DOI] [PubMed] [Google Scholar]
- 12. Kelc R, Trapecar M, Gradisnik L et al. Platelet‐rich plasma, especially when combined with a TGF‐β inhibitor promotes proliferation, viability and myogenic differentiation of myoblasts in vitro. PLoS One 2015;10:e0117302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Kampa RJ, Connell DA. Treatment of tendinopathy: Is there a role for autologous whole blood and platelet rich plasma injection. Int J Clin Pract 2010;64:1813–1823. [DOI] [PubMed] [Google Scholar]
- 14. de Vos RJ, van Veldhoven PL, Moen MH et al. Autologous growth factor injections in chronic tendinopathy: A systematic review. Br Med Bull 2010;95:63–77. [DOI] [PubMed] [Google Scholar]
- 15. Dietrich F, Hammerman M, Blomgran P et al. Effect of platelet‐rich plasma on rat Achilles tendon healing is related to microbiota. Acta Orthop 2017;88:416–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Yan R, Gu Y, Ran J et al. Intratendon delivery of leukocyte‐poor platelet‐rich plasma improves healing compared with leukocyte‐rich platelet‐rich plasma in a rabbit Achilles tendinopathy model. Am J Sports Med 2017;45:1909–1920. [DOI] [PubMed] [Google Scholar]
- 17. Lee CH, Moioli EK, Mao JJ. Fibroblastic differentiation of human mesenchymal stem cells using connective tissue growth factor. Conf Proc IEEE Eng Med Biol Soc 2006;1:775–778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Lee CH, Shah B, Moioli EK et al. CTGF directs fibroblast differentiation from human mesenchymal stem/stromal cells and defines connective tissue healing in a rodent injury model. J Clin Invest 2010;120:3340–3349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Yuda A, Maeda H, Fujii S et al. Effect of CTGF/CCN2 on osteo/cementoblastic and fibroblastic differentiation of a human periodontal ligament stem/progenitor cell line. J Cell Physiol 2015;230:150–159. [DOI] [PubMed] [Google Scholar]
- 20. Ni M, Rui YF, Tan Q et al. Engineered scaffold‐free tendon tissue produced by tendon‐derived stem cells. Biomaterials 2013;34:2024–2037. [DOI] [PubMed] [Google Scholar]
- 21. Lui PP, Wong OT, Lee YW. Transplantation of tendon‐derived stem cells pre‐treated with connective tissue growth factor and ascorbic acid in vitro promoted better tendon repair in a patellar tendon window injury rat model. Cytotherapy 2016;18:99–112. [DOI] [PubMed] [Google Scholar]
- 22. Lui PP, Wong OT, Lee YW. Application of tendon‐derived stem cell sheet for the promotion of graft healing in anterior cruciate ligament reconstruction. Am J Sports Med 2014;42:681–689. [DOI] [PubMed] [Google Scholar]
- 23. Lee CH, Lee FY, Tarafder S et al. Harnessing endogenous stem/progenitor cells for tendon regeneration. J Clin Invest 2015;125:2690–2701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Havis E, Bonnin MA, Olivera‐Martinez I et al. Transcriptomic analysis of mouse limb tendon cells during development. Development 2014;141:3683–3696. [DOI] [PubMed] [Google Scholar]
- 25. Liu H, Zhang C, Zhu S et al. Mohawk promotes the tenogenesis of mesenchymal stem cells through activation of the TGFβ signaling pathway. Stem Cells 2015;33:443–455. [DOI] [PubMed] [Google Scholar]
- 26. Maeda T, Sakabe T, Sunaga A et al. Conversion of mechanical force into TGF‐β‐mediated biochemical signals. Curr Biol 2011;21:933–941. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Guerquin MJ, Charvet B, Nourissat G et al. Transcription factor EGR1 directs tendon differentiation and promotes tendon repair. J Clin Invest 2013;123:3564–3576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Theiss F, Mirsaidi A, Mhanna R et al. Use of biomimetic microtissue spheroids and specific growth factor supplementation to improve tenocyte differentiation and adaptation to a collagen‐based scaffold in vitro. Biomaterials 2015;69:99–109. [DOI] [PubMed] [Google Scholar]
- 29. Farhat YM, Al‐Maliki AA, Chen T et al. Gene expression analysis of the pleiotropic effects of TGF‐β1 in an in vitro model of flexor tendon healing. PLoS One 2012;7:e51411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Katsura T, Tohyama H, Kondo E et al. Effects of administration of transforming growth factor (TGF)‐beta1 and anti‐TGF‐beta1 antibody on the mechanical properties of the stress‐shielded patellar tendon. J Biomech 2006;39:2566–2572. [DOI] [PubMed] [Google Scholar]
- 31. Pryce BA, Watson SS, Murchison ND et al. Recruitment and maintenance of tendon progenitors by TGFbeta signaling are essential for tendon formation. Development 2009;136:1351–1361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Barsby T, Guest D. Transforming growth factor beta3 promotes tendon differentiation of equine embryo‐derived stem cells. Tissue Eng Part A 2013;19:2156–2165. [DOI] [PubMed] [Google Scholar]
- 33. Kapacee Z, Yeung CY, Lu Y et al. Synthesis of embryonic tendon‐like tissue by human marrow stromal/mesenchymal stem cells requires a three‐dimensional environment and transforming growth factor β3. Matrix Biol 2010;29:668–677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Lyras DN, Kazakos K, Verettas D et al. Effect of combined administration of transforming growth factor‐b1 and insulin‐like growth factor I on the mechanical properties of a patellar tendon defect model in rabbits. Acta Orthop Belg 2010;76:380–386. [PubMed] [Google Scholar]
- 35. Sarenac T, Trapecar M, Gradisnik L et al. Single‐cell analysis reveals IGF‐1 potentiation of inhibition of the TGF‐β/Smad pathway of fibrosis in human keratocytes in vitro. Sci Rep 2016;6:34373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Wang W, Rigueur D, Lyons KM. TGFβ signaling in cartilage development and maintenance. Birth Defects Res C Embryo Today 2014;102:37–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Yang G, Rothrauff BB, Lin H et al. Tendon‐derived extracellular matrix enhances transforming growth factor‐β3‐induced tenogenic differentiation of human adipose‐derived stem cells. Tissue Eng Part A 2017;23:166–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Pelled G, Snedeker JG, Ben‐Arav A et al. Smad8/BMP2‐engineered mesenchymal stem cells induce accelerated recovery of the biomechanical properties of the Achilles tendon. J Orthop Res 2012;30:1932–1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Han P, Cui Q, Yang S et al. Tumor necrosis factor‐α and transforming growth factor‐β1 facilitate differentiation and proliferation of tendon‐derived stem cells in vitro. Biotechnol Lett 2017;39:711–719. [DOI] [PubMed] [Google Scholar]
- 40. Jiang K, Chun G, Wang Z et al. Effect of transforming growth factor‐β3 on the expression of Smad3 and Smad7 in tenocytes. Mol Med Rep 2016;13:3567–3573. [DOI] [PubMed] [Google Scholar]
- 41. Wolfman NM, Hattersley G, Cox K et al. Ectopic induction of tendon and ligament in rats by growth and differentiation factors 5, 6, and 7, members of the TGF‐beta gene family. J Clin Invest 1997;100:321–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Park A, Hogan MV, Kesturu GS et al. Adipose‐derived mesenchymal stem cells treated with growth differentiation factor‐5 express tendon‐specific markers. Tissue Eng Part A 2010;16:2941–2951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. James R, Kumbar SG, Laurencin CT et al. Tendon tissue engineering: Adipose‐derived stem cell and GDF‐5 mediated regeneration using electrospun matrix systems. Biomed Mater 2011;6:025011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Tan SL, Ahmad RE, Ahmad TS et al. Effect of growth differentiation factor 5 on the proliferation and tenogenic differentiation potential of human mesenchymal stem cells in vitro. Cells Tissues Organs 2012;196:325–338. [DOI] [PubMed] [Google Scholar]
- 45. Govoni M, Berardi AC, Muscari C et al. *An engineered multiphase three‐dimensional microenvironment to ensure the controlled delivery of cyclic strain and human growth differentiation factor 5 for the tenogenic commitment of human bone marrow mesenchymal stem cells. Tissue Eng Part A 2017;23:811–822. [DOI] [PubMed] [Google Scholar]
- 46. Xia D, Sumita Y, Liu Y et al. GDFs promote tenogenic characteristics on human periodontal ligament‐derived cells in culture at late passages. Growth Factors 2013;31:165–173. [DOI] [PubMed] [Google Scholar]
- 47. Chai W, Ni M, Rui YF et al. Effect of growth and differentiation factor 6 on the tenogenic differentiation of bone marrow‐derived mesenchymal stem cells. Chin Med J (Engl) 2013;126:1509–1516. [PubMed] [Google Scholar]
- 48. Berasi SP, Varadarajan U, Archambault J et al. Divergent activities of osteogenic BMP2, and tenogenic BMP12 and BMP13 independent of receptor binding affinities. Growth Factors 2011;29:128–139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Dai L, Hu X, Zhang X et al. Different tenogenic differentiation capacities of different mesenchymal stem cells in the presence of BMP‐12. J Transl Med 2015;13:200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Liu J, Tao X, Chen L et al. CTGF positively regulates BMP12 induced tenogenic differentiation of tendon stem cells and signaling. Cell Physiol Biochem 2015;35:1831–1845. [DOI] [PubMed] [Google Scholar]
- 51. Lee JY, Zhou Z, Taub PJ et al. BMP‐12 treatment of adult mesenchymal stem cells in vitro augments tendon‐like tissue formation and defect repair in vivo. PLoS One 2011;6:e17531. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Ni M, Rui Y, Chen Q et al. [Effect of growth differentiation factor 7 on tenogenic differentiation of bone marrow mesenchymal stem cells of rat in vitro]. Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi 2011;25:1103–1109. [PubMed] [Google Scholar]
- 53. Shen H, Gelberman RH, Silva MJ et al. BMP12 induces tenogenic differentiation of adipose‐derived stromal cells. PLoS One 2013;8:e77613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Violini S, Ramelli P, Pisani LF et al. Horse bone marrow mesenchymal stem cells express embryo stem cell markers and show the ability for tenogenic differentiation by in vitro exposure to BMP‐12. BMC Cell Biol 2009;10:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chamberlain CS, Lee JS, Leiferman EM et al. Effects of BMP‐12‐releasing sutures on Achilles tendon healing. Tissue Eng Part A 2015;21:916–927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zeng Q, Li X, Choi L et al. Recombinant growth/differentiation factor‐5 stimulates osteogenic differentiation of fat‐derived stromal cells in vitro. Connect Tissue Res 2006;47:264–270. [DOI] [PubMed] [Google Scholar]
- 57. Rickert M, Jung M, Adiyaman M et al. A growth and differentiation factor‐5 (GDF‐5)‐coated suture stimulates tendon healing in an Achilles tendon model in rats. Growth Factors 2001;19:115–126. [DOI] [PubMed] [Google Scholar]
- 58. Dines JS, Weber L, Razzano P et al. The effect of growth differentiation factor‐5‐coated sutures on tendon repair in a rat model. J Shoulder Elbow Surg 2007;16:S215–S221. [DOI] [PubMed] [Google Scholar]
- 59. Tan SL, Ahmad TS, Ng WM et al. Identification of pathways mediating growth differentiation factor5‐induced tenogenic differentiation in human bone marrow stromal cells. PLoS One 2015;10:e0140869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Greiner S, Ide J, Van Noort A et al. Local rhBMP‐12 on an absorbable collagen sponge as an adjuvant therapy for rotator cuff repair ‐ a phase 1, randomized, standard of care control, multicenter study: Safety and feasibility. Am J Sports Med 2015;43:1994–2004. [DOI] [PubMed] [Google Scholar]
- 61. Thomopoulos S, Harwood FL, Silva MJ et al. Effect of several growth factors on canine flexor tendon fibroblast proliferation and collagen synthesis in vitro. J Hand Surg Am 2005;30:441–447. [DOI] [PubMed] [Google Scholar]
- 62. Hankemeier S, Keus M, Zeichen J et al. Modulation of proliferation and differentiation of human bone marrow stromal cells by fibroblast growth factor 2: Potential implications for tissue engineering of tendons and ligaments. Tissue Eng 2005;11:41–49. [DOI] [PubMed] [Google Scholar]
- 63. Sahoo S, Ang LT, Cho‐Hong GJ et al. Bioactive nanofibers for fibroblastic differentiation of mesenchymal precursor cells for ligament/tendon tissue engineering applications. Differentiation 2010;79:102–110. [DOI] [PubMed] [Google Scholar]
- 64. Ker ED, Nain AS, Weiss LE et al. Bioprinting of growth factors onto aligned sub‐micron fibrous scaffolds for simultaneous control of cell differentiation and alignment. Biomaterials 2011;32:8097–8107. [DOI] [PubMed] [Google Scholar]
- 65. Sahoo S, Ang LT, Goh JC et al. Growth factor delivery through electrospun nanofibers in scaffolds for tissue engineering applications. J Biomed Mater Res A 2010;93:1539–1550. [DOI] [PubMed] [Google Scholar]
- 66. Tokunaga T, Shukunami C, Okamoto N et al. FGF‐2 stimulates the growth of tenogenic progenitor cells to facilitate the generation of tenomodulin‐positive tenocytes in a rat rotator cuff healing model. Am J Sports Med 2015;43:2411–2422. [DOI] [PubMed] [Google Scholar]
- 67. Brent AE, Tabin CJ. FGF acts directly on the somitic tendon progenitors through the Ets transcription factors Pea3 and Erm to regulate scleraxis expression. Development 2004;131:3885–3896. [DOI] [PubMed] [Google Scholar]
- 68. Thomopoulos S, Kim HM, Das R et al. The effects of exogenous basic fibroblast growth factor on intrasynovial flexor tendon healing in a canine model. J Bone Joint Surg Am 2010;92:2285–2293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Brown JP, Finley VG, Kuo CK. Embryonic mechanical and soluble cues regulate tendon progenitor cell gene expression as a function of developmental stage and anatomical origin. J Biomech 2014;47:214–222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Brown JP, Galassi TV, Stoppato M et al. Comparative analysis of mesenchymal stem cell and embryonic tendon progenitor cell response to embryonic tendon biochemical and mechanical factors. Stem Cell Res Ther 2015;6:89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Rothan HA, Suhaeb AM, Kamarul T. Recombinant human adiponectin as a potential protein for treating diabetic tendinopathy promotes tenocyte progenitor cells proliferation and tenogenic differentiation in vitro. Int J Med Sci 2013;10:1899–1906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Solchaga LA, Bendele A, Shah V et al. Comparison of the effect of intra‐tendon applications of recombinant human platelet‐derived growth factor‐BB, platelet‐rich plasma, steroids in a rat Achilles tendon collagenase model. J Orthop Res 2014;32:145–150. [DOI] [PubMed] [Google Scholar]
- 73. Shah V, Bendele A, Dines JS et al. Dose‐response effect of an intra‐tendon application of recombinant human platelet‐derived growth factor‐BB (rhPDGF‐BB) in a rat Achilles tendinopathy model. J Orthop Res 2013;31:413–420. [DOI] [PubMed] [Google Scholar]
- 74. Kovacevic D, Gulotta LV, Ying L et al. rhPDGF‐BB promotes early healing in a rat rotator cuff repair model. Clin Orthop Relat Res 2015;473:1644–1654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Miyabara S, Yuda Y, Kasashima Y et al. Regulation of tenomodulin expression via Wnt/β‐catenin signaling in equine bone marrow‐derived mesenchymal stem cells. J Equine Sci 2014;25:7–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Zhu X, Zhu H, Zhang L et al. Wls‐mediated Wnts differentially regulate distal limb patterning and tissue morphogenesis. Dev Biol 2012;365:328–338. [DOI] [PubMed] [Google Scholar]
- 77. Havis E, Bonnin MA, de Lima JE et al. TGFβ and FGF promote tendon progenitor fate and act downstream of muscle contraction to regulate tendon differentiation during chick limb development. Development 2016;143:3839–3851. [DOI] [PubMed] [Google Scholar]
- 78. Gonçalves AI, Rodrigues MT, Lee SJ et al. Understanding the role of growth factors in modulating stem cell tenogenesis. PLoS One 2013;8:e83734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yin Z, Guo J, Wu TY et al. Stepwise differentiation of mesenchymal stem cells augments tendon‐like tissue formation and defect repair in vivo. Stem Cells Translational Medicine 2016;5:1106–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Barnette DN, Hulin A, Ahmed AS et al. Tgfβ‐Smad and MAPK signaling mediate scleraxis and proteoglycan expression in heart valves. J Mol Cell Cardiol 2013;65:137–146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Lorda‐Diez CI, Montero JA, Martinez‐Cue C et al. Transforming growth factors beta coordinate cartilage and tendon differentiation in the developing limb mesenchyme. J Biol Chem 2009;284:29988–29996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Lorda‐Diez CI, Montero JA, Garcia‐Porrero JA et al. Divergent differentiation of skeletal progenitors into cartilage and tendon: Lessons from the embryonic limb. ACS Chem Biol 2014;9:72–79. [DOI] [PubMed] [Google Scholar]
- 83. Lorda‐Diez CI, Montero JA, Diaz‐Mendoza MJ et al. βig‐h3 potentiates the profibrogenic effect of TGFβ signaling on connective tissue progenitor cells through the negative regulation of master chondrogenic genes. Tissue Eng Part A 2013;19:448–457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Kishimoto Y, Ohkawara B, Sakai T et al. Wnt/β‐catenin signaling suppresses expressions of Scx, Mkx, and Tnmd in tendon‐derived cells. PLoS One 2017;12:e0182051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Hicks M, Pyle A. The path from pluripotency to skeletal muscle: Developmental myogenesis guides the way. Cell Stem Cell 2015;17:255–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Bi Y, Ehirchiou D, Kilts TM et al. Identification of tendon stem/progenitor cells and the role of the extracellular matrix in their niche. Nat Med 2007;13:1219–1227. [DOI] [PubMed] [Google Scholar]
- 87. Nourissat G, Berenbaum F, Duprez D. Tendon injury: From biology to tendon repair. Nat Rev Rheumatol 2015;11:223–233. [DOI] [PubMed] [Google Scholar]
- 88. Liu H, Xu J, Liu CF et al. Whole transcriptome expression profiling of mouse limb tendon development by using RNA‐seq. J Orthop Res 2015;33:840–848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Jelinsky SA, Archambault J, Li L et al. Tendon‐selective genes identified from rat and human musculoskeletal tissues. J Orthop Res 2010;28:289–297. [DOI] [PubMed] [Google Scholar]
- 90. Trapecar M, Kelc R, Gradisnik L et al. Myogenic progenitors and imaging single‐cell flow analysis: A model to study commitment of adult muscle stem cells. J Muscle Res Cell Motil 2014;35:249–257. [DOI] [PubMed] [Google Scholar]
- 91. Bendall SC, Simonds EF, Qiu P et al. Single‐cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science 2011;332:687–696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Guo G, Luc S, Marco E et al. Mapping cellular hierarchy by single‐cell analysis of the cell surface repertoire. Cell Stem Cell 2013;13:492–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Grün D, Lyubimova A, Kester L et al. Single‐cell messenger RNA sequencing reveals rare intestinal cell types. Nature 2015;525:251–255. [DOI] [PubMed] [Google Scholar]
- 94. DeLaughter DM, Bick AG, Wakimoto H et al. Single‐cell resolution of temporal gene expression during heart development. Dev Cell 2016;39:480–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Li J, Luo H, Wang R et al. Systematic reconstruction of molecular cascades regulating GP development using single‐cell RNA‐seq. Cell Rep 2016;15:1467–1480. [DOI] [PubMed] [Google Scholar]
- 96. Yin Z, Hu JJ, Yang L et al. Single‐cell analysis reveals a nestin(+) tendon stem/progenitor cell population with strong tenogenic potentiality. Sci Adv 2016;2:e1600874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Linnarsson S. Sequencing single cells reveals sequential stem cell states. Cell Stem Cell 2015;17:251–252. [DOI] [PubMed] [Google Scholar]
- 98. Shapiro E, Biezuner T, Linnarsson S. Single‐cell sequencing‐based technologies will revolutionize whole‐organism science. Nat Rev Genet 2013;14:618–630. [DOI] [PubMed] [Google Scholar]
- 99. Llorens‐Bobadilla E, Zhao S, Baser A et al. Single‐cell transcriptomics reveals a population of dormant neural stem cells that become activated upon brain injury. Cell Stem Cell 2015;17:329–340. [DOI] [PubMed] [Google Scholar]
- 100. Pollen AA, Nowakowski TJ, Shuga J et al. Low‐coverage single‐cell mRNA sequencing reveals cellular heterogeneity and activated signaling pathways in developing cerebral cortex. Nat Biotechnol 2014;32:1053–1058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Ladewig J, Koch P, Brüstle O. Leveling Waddington: The emergence of direct programming and the loss of cell fate hierarchies. Nat Rev Mol Cell Biol 2013;14:225–236. [DOI] [PubMed] [Google Scholar]
- 102. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006;126:663–676. [DOI] [PubMed] [Google Scholar]
- 103. Li W, Tian E, Chen ZX et al. Identification of Oct4‐activating compounds that enhance reprogramming efficiency. Proc Natl Acad Sci USA 2012;109:20853–20858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Shi Y, Desponts C, Do JT et al. Induction of pluripotent stem cells from mouse embryonic fibroblasts by Oct4 and Klf4 with small‐molecule compounds. Cell Stem Cell 2008;3:568–574. [DOI] [PubMed] [Google Scholar]
- 105. Cheng L, Hu W, Qiu B et al. Generation of neural progenitor cells by chemical cocktails and hypoxia. Cell Res 2014;24:665–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Pennarossa G, Maffei S, Campagnol M et al. Brief demethylation step allows the conversion of adult human skin fibroblasts into insulin‐secreting cells. Proc Natl Acad Sci USA 2013;110:8948–8953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Sayed N, Wong WT, Ospino F et al. Transdifferentiation of human fibroblasts to endothelial cells: Role of innate immunity. Circulation 2015;131:300–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Webb S, Gabrelow C, Pierce J et al. Retinoic acid receptor signaling preserves tendon stem cell characteristics and prevents spontaneous differentiation in vitrox. Stem Cell Res Ther 2016;7:45. [DOI] [PMC free article] [PubMed] [Google Scholar]