We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research ArticleOpen Accesscc iconby iconnc iconnd icon

Early delivery of human umbilical cord mesenchymal stem cells improves healing in a rat model of Achilles tendinopathy

    Ze Yuan‡

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    ‡These authors made contributions equally to this work

    Search for more papers by this author

    ,
    Ding Yu‡

    Department of Rehabilitation Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology

    ‡These authors made contributions equally to this work

    Search for more papers by this author

    ,
    Yanxue Wang

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    ,
    Lijiaqi Liu

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    ,
    Junchao Wang

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    ,
    Chao Ma

    *Author for correspondence:

    E-mail Address: machao@mail.sysu.edu.cn

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    &
    Shaoling Wu

    **Author for correspondence:

    E-mail Address: wushl@mail.sysu.edu.cn

    Department of Rehabilitation Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China

    Published Online:https://doi.org/10.2217/rme-2023-0222

    Abstract

    Objective: This study aimed to explore the efficacy and optimal delivery time of human umbilical cord mesenchymal stem cells (hUC-MSCs) in treating collagenase-induced Achilles tendinopathy. Methods: Achilles tendinopathy in rats at early or advanced stages was induced by injecting collagenase I into bilateral Achilles tendons. A total of 28 injured rats were injected with a hUC-MSC solution or normal saline into bilateral tendons twice and sampled after 4 weeks for histological staining, gene expression analysis, transmission electron microscope assay and biomechanical testing analysis. Results: The results revealed better histological performance and a larger collagen fiber diameter in the MSC group. mRNA expression of TNF-α, IL-1β and MMP-3 was lower after MSC transplantation. Early MSC delivery promoted collagen I and TIMP-3 synthesis, and strengthened tendon toughness. Conclusion: hUC-MSCs demonstrated a therapeutic effect in treating collagenase-induced Achilles tendinopathy, particularly in the early stage of tendinopathy.

    Tendinopathy is a common musculoskeletal disorder that arises due to overuse, injury and excessive use of certain medications, such as corticosteroids and fluoroquinolones [1,2]. Clinical manifestations of tendinopathy include pain and swelling around the tendon, often accompanied by functional limitations that significantly impact daily activities [1]. The Achilles tendon, being the largest tendon in the human body, plays a crucial role in functions such as standing and walking [3]. Achilles tendinopathy has a lifetime prevalence of 23.9% in athletes and 5.9% in the general population [4]. Unfortunately, the healing ability of tendon tissue is limited due to the absence of adequate cells and blood vessels [5]. Traditional conservative therapies, including nonsteroidal anti-inflammatory drugs, glucocorticoids, extracorporeal shockwave therapy and eccentric exercise, can alleviate symptoms but fall short of repairing the injured tendon [6–8].

    Mesenchymal stem cells (MSCs) derived from human umbilical cords exhibit strong cloning and proliferation abilities, along with multiple differentiation and immune regulation capabilities, and possess an immune-exempt status [9,10]. Numerous clinical studies have corroborated their safety [11]. Furthermore, the use of these stem cells encounters less controversy, given recognition of the human umbilical cord as medical waste. These advantages render hUC-MSCs suitable for diseases resulting from chronic inflammation and associated tissue damage [12].

    While some studies suggest that MSC transplantation alone may not effectively repair Achilles tendon injury [13,14], others report contrasting results [15–17]. These discrepancies may be attributed to variations in the timing of stem cell delivery. Positive effects were consistently observed in studies where injections were administered during the early stages of tendinopathy, while injections given during advanced stages demonstrated limited efficacy. Consequently, the experiments in this study were designed to explore the efficacy of MSC intervention at different stages and its potential therapeutic mechanism.

    Methods

    Achilles tendinopathy model

    In this study, 39 male Sprague–Dawley rats weighing between 250 and 300 g were used. All the animals were sourced from the Animal Laboratory Center of Sun Yat-sen University and housed under controlled conditions, with a 12-h light–dark cycle, a laboratory temperature maintained at 25 ± 1°C, and a relative humidity range of 50–70%. The study received approval from the Animal Ethics Committee of Sun Yat-sen University (SYSU-IACACC-2022-000200). Treatment of the rats adhered to pertinent guidelines and regulations, ensuring ethical standards were maintained throughout the study. Furthermore, the research strictly complied with ARRIVE guidelines, promoting transparency and rigor in reporting.

    Two rats were accommodated per cage; a 1-week acclimatization period was allowed, during which all rats had unrestricted access to food and water. Of the 39 rats, 32 underwent the chemical injury method [18,19] to establish Achilles tendinopathy models, while the remaining seven served as normal controls. The Achilles tendinopathy models were established by inducing anesthesia with 2% sodium pentobarbital and carefully inserting a 4.5-UI needle into the Achilles tendon, positioned 3 mm above the calcaneal and advanced 5 mm along the long axis. Subsequently, 50 μl of collagenase I solution (10 mg/ml; Sigma-Aldrich, Shanghai, China) was injected, which was based on previous relevant studies [20,21] and our pre-experiments. The needle was retracted slowly during the injection process, and this procedure was performed on both sides of the Achilles tendons. A successful model was established after 1 week and 4 weeks of one injection (Figure 1). To ensure the robustness of the modeling results, two rats were utilized for each time point (Supplementary File 1.1). All the models completed strictly according to this protocol were considered successful.

    Figure 1. Processes involved in the establishment of Achilles tendinopathy models, treatment and sampling.

    MSC: Mesenchymal stem cell.

    MSC delivery & sampling

    Following the successful establishment of the tendinopathy model, 28 rats were randomly allocated into four groups using a random-number table: MSC-1 (MSC transplantation 1 week after collagenase I injection), control-1 (saline solution injection 1 week after collagenase I injection), MSC-2 (MSC transplantation 4 weeks after collagenase I injection) and control-2 (saline solution injection 4 weeks after collagenase I injection). Fifth-generation hUC-MSCs sourced from the SALIAI Stem Cells Company (G05003) were employed, with 5 × 105 stem cells dissolved in 50 μl of normal saline injected into each Achilles tendon using 4.5 UI needles for the MSC groups. The control group received a 50-μl injection of normal saline. Stem cells or saline were delivered into and around bilateral Achilles tendons twice, with a 1-week interval between administrations. To mitigate subjective bias, the operator performing the injections was blinded to the injection contents. Sampling was conducted 4 weeks after the conclusion of the second treatment (Figure 1). During sampling, the right Achilles tendons of seven rats in each group underwent a biomechanics assay. Four left Achilles tendons from each group were dissected in the middle, with one-half allocated for histological analysis and the other half for gene expression analysis. Another three left Achilles tendons from each group were designated for transmission electron microscopy. It is noteworthy that all assessors involved in subsequent analyses were blinded to the group affiliations of the samples or sections.

    Hematoxylin & eosin staining

    Upon sampling, the Achilles tendons were promptly immersed in a dedicated tissue-specific fixative (Servicebio, Wuhan, China) for 48 h. Subsequently, the tendons underwent dehydration using sucrose solution and paraffin embedding, and were sliced into 10-μm sections for staining with hematoxylin and eosin reagent. Histological assessment of the tendon involved the evaluation of six parameters [22,23]: collagen fiber structure, collagen fiber arrangement, nuclear roundness, inflammation, vascular proliferation and cell density. The cumulative histological score, ranging from 0 to 18, serves as an indicator of the tendon's condition, with lower scores indicative of a closer resemblance to a normal Achilles tendon. Three expert assessors, blinded to the group assignments, conducted the assessments. This comprehensive histological scoring system provides a multifaceted understanding of the tendon's histopathological characteristics.

    RT-qPCR

    For RT-qPCR analysis, Achilles tendon samples were meticulously minced and ground with grinding beads in a frozen mill. Total RNA extraction was carried out using Trizol (Invitrogen, Shanghai, China). Subsequently, first-Strand cDNA was synthesized using the SuperScript III First-Strand Synthesis Kit (Invitrogen) following the manufacturer's protocol. The resultant cDNA served as the template for subsequent RT-qPCR, employing iQ SYBR Green Supermix (Bio-Rad, Guangzhou, China). Primers (Table 1) were synthesized by Shanghai Sangon Biotechnology Co., Ltd (Shanghai, China). The relative expression levels of PCR products were determined using the 2-ΔΔCT method, with calculations carried out relative to the Control-1 group.

    Table 1. RT-qPCR primer sequences.
    No.PrimerSequence (5′–3′)
    1IL-1β-FORWARDAATCTCACAGCAGCATCTCGACAAG
    2IL-1β-REVERSETCCACGGGCAAGACATAGGTAGC
    3TNF-α-FORWARDAAAGGACACCATGAGCACGGAAAG
    4TNF-α-REVERSECGCCACGAGCAGGAATGAGAAG
    5MMP-3-FORWARDATGGAAGGCGTCGTGTGTTTCAG
    6MMP-3-REVERSETCAGAGGAAGAGCTATCAGGGTCAG
    7TIMP-3-FORWARDACTCAGCACCCTCTTGTCCCTATC
    8TIMP-3-REVERSECTAACACTCACACTCACCTGCCATC

    Immunohistochemical examinations

    The 10-μm sections underwent deparaffinization and were subsequently treated with 10% hydrogen peroxide. Antigen retrieval was performed by heating the sections in a 0.01-mol/l sodium citrate solution at multiple intervals. Following this, blocking with a blocking solution ensued for 1 h. Subsequently, the sections were incubated overnight at 4°C with primary antibodies targeting collagen I, collagen III, MMP-3 and TIMP-3 (Supplementary File 1.2). After thorough washing to remove the primary antibodies, the cells were stained using the DAB chromogenic kit (Gene Tech, Beijing, China), and the nuclei were counterstained with hematoxylin. Hematoxylin-stained nuclei appeared blue, while positive DAB expression manifested as brown. Each type of antibody underwent staining simultaneously in one batch, and all samples were imaged with identical microscope settings at the same time for consistency. The mean optical density of positive expression in four randomly selected fields at 200× magnification was analyzed using ImageJ 1.53c. The optical density of the whole staining area and the integrated density of positive staining were measured. The mean optical density (integrated density/whole staining area) was then calculated as the outcome.

    Transmission electron microscope assay

    Fresh tissue underwent fixation using a transmission electron microscope-specific fixative (Servicebio), followed by dehydration, embedding and sectioning into 60-nm ultrathin sections. These sections were stained with 2% uranium acetate-saturated alcohol solution for 8 min in the absence of light. Subsequently, they were rinsed three times in 70% ethanol and then three times in ultrapure water. A second staining step involved immersion in 2.6% lead citrate for 8 min, followed by another three rinses in ultrapure water. After drying with filter paper, the cuprum grids were carefully placed on a grid board and air-dried overnight at room temperature. The stainied sections were then examined and photographed using a transmission electron microscope (HITACHI, Tokyo, Japan). ImageJ 1.53c was employed for the quantitative analysis of collagen density, diameter and area in the cross-section of the Achilles tendon. Three randomly selected images were analyzed for each sample, with approximately 40 fibrils measured per image.

    Biomechanical testing

    The Achilles tendon was detached from the calcaneus and immersed in saline. Both ends of the tendon were secured onto a jig, with gauze evenly wrapped to prevent slippage and breakage at the clamps. Tensile testing was performed using a Zhiqu ZQ990 tensile machine. The Achilles tendon was stretched at a controlled speed of 1 mm/s until fracture occurred, with a preload of 2N applied. The stress–strain curve derived from the testing provided essential parameters such as the maximum force and stiffness (calculated as the maximum force divided by the deformation at maximum force).

    Statistical analysis

    Statistical analysis of the data was conducted using R language (version 4.3.2). GraphPad Prism (version 9.4.1) was employed for the visualization of statistics charts. Data were presented as mean ± standard deviation, and qualitative data were expressed as numbers and percentages. The nlme package in R was utilized for the implementation of the linear mixed effects model analysis, treating individuals as random effects and groups as fixed effects. Subsequently, the emmeans package was used for post-pairwise comparisons among MSC and control groups, with values adjusted based on the false discovery rate method. A significance level of p < 0.05 was considered statistically significant.

    Results

    Histological performance improvement due to MSC delivery

    As depicted in Figure 2, the normal Achilles tendon exhibited a characteristic parallel collagen arrangement with elongated fusiform nuclei interspersed among the fibrils. In the control groups, collagen arrangement became disordered, accompanied by inflammatory cell infiltration and the emergence of calcified regions, leading to an increased number of tendon cells. Following the injection of hUC-MSCs, a notable improvement in fiber arrangement was observed compared with the control groups. The cell count was reduced and a remarkable absence of calcium salt deposition was noted in the MSC groups in contrast to the control groups. Semiquantitative scores for the MSC groups were significantly lower than those for the control groups (both p < 0.01), with the MSC-1 group yielding lower scores than the MSC-2 group (p < 0.05).

    Figure 2. H&E staining and score of Achilles tendons.

    (A) Gross specimen and hematoxylin and eosin staining section of the Achilles tendon. (B) Semiquantitative scores of different groups.

    **p < 0.01 for the MSC groups compared with the control groups. #p < 0.05 for the MSC-1 group compared with the MSC-2 group.

    MSC: Mesenchymal stem cell.

    mRNA level changes induced by MSC delivery

    Figure 3 illustrates the PCR results, revealing distinctive expression patterns. In both MSC-1 and MSC-2 groups, the expressions of TNF-α and IL-1β were significantly lower than those in the control-1 and control-2 groups (p < 0.01, p < 0.05 for MSC-1 and both p < 0.05 for MSC-2). Notably, the expression in the MSC-1 group was slightly lower than that in the MSC-2 group, although this difference did not reach significance. This collective data suggest a substantial reduction in inflammation following the administration of stem cells, both in the early and advanced stages of Achilles tendinopathy. Examining MMP3 expression, the MSC-1 group exhibited significantly lower levels compared with the control-1 (p < 0.05) and MSC-2 (p < 0.05) groups. In addition, the MSC-2 group demonstrated markedly lower expression than the control-2 group (p < 0.01). In contrast, TIMP3 expression in the MSC-1 group was significantly higher than that in the control-1 (p < 0.01) and MSC-2 (p < 0.05) groups. The MSC-2 group also exhibited higher expression compared with the control-2 group (p < 0.05).

    Figure 3. mRNA expression of TNF-α, IL-1β, MMP-3 and TIMP-3 in the Achilles tendon.

    *(or #) p < 0.05; **p < 0.01.

    *Represents the MSC groups compared with the control groups, while # represents the MSC-1 group compared with the MSC-2 group.

    MSC: Mesenchymal stem cell.

    Promotion of normal collagen matrix synthesis by MSC delivery

    As depicted in Figure 4, collagen I, being the primary component of normal tendon tissue, exhibited higher expression in both the MSC-1 (p < 0.01) and MSC-2 (p < 0.01) groups compared with the control-1 and control-2 groups, respectively. Remarkably, the MSC-1 group demonstrated significantly greater expression than the MSC-2 group (p < 0.05). Conversely, collagen III, associated with Achilles tendon fibrosis, exhibited reduced expression in both MSC-1 (p < 0.01) and MSC-2 (p < 0.05) groups compared with their respective control groups. It is noteworthy that exercising the collagen antibody faced challenges binding to mature collagen fibrils within the matrix, thereby enabling mainly staining for newly synthesized collagen, but the effect of staining for part mature collagen should be considered. MMP-3 expression followed a similar trend, being lower in the MSC groups (both p < 0.01) compared with their respective control groups. In contrast, TIMP-3 expression showed an opposing trend with higher expressions in the MSC-1 (p < 0.01) and MSC-2 groups (p < 0.05) compared with their respective control groups. Notably, the MSC-1 group exhibited higher expression compared with the MSC-2 group, but the difference was not statistically significant (p > 0.05).

    Figure 4. Protein expression of collagen and relevant matrix synthesis.

    (A) Tissue sections subjected to immunostaining with collagen I, collagen III, MMP-3 and TIMP-3, respectively. (B) Mean density of positive areas. *(or #) represents p < 0.05, whereas ** represents p < 0.01.

    *Represents the MSC groups compared with the control groups, while # represents the MSC-1 group compared with the MSC-2 group.

    MSC: Mesenchymal stem cell.

    Effect of MSC delivery on tendon fibrils thickness

    Collagen I, characterized by larger fibers than collagen III, was examined in a transverse section of the Achilles tendon using transmission electron microscopy, as illustrated in Figure 5. In normal and MSC tissues, most fibrils exhibited a larger size, while control groups generally featured smaller fibrils. The results demonstrated that both the MSC-1 (both p < 0.01) and MSC-2 (both p < 0.01) groups displayed significantly larger diameters and a larger average cross-sectional area of collagen fibrils compared with their respective control groups. Moreover, the MSC-1 group exhibited larger diameters and a larger average cross-sectional area of collagen fibrils than the MSC-2 group (both p < 0.05). The density of fibrils in both the MSC-1 (p < 0.01) and MSC-2 (p < 0.01) groups were lower than that of their respective control groups, with no significant difference observed between the MSC groups.

    Figure 5. Transmission electron microscope assay of Achilles tendons.

    (A) Representative transmission electron microscope pictures of Achilles tendon samples from the MSC and control groups. (B) The density, diameter and area of collagens in the cross-section of the Achilles tendon.

    **p < 0.01 for the MSC groups compared with the control groups.

    #p < 0.05 for the MSC-1 group compared with the MSC-2 group.

    MSC: Mesenchymal stem cell.

    Enchancement of tendon strength by MSC delivery

    All tendon ruptures occurred between the two clips. As illustrated in Figure 6, the maximum force and stiffness observed in the MSC and control groups were notably lower than those in the normal group. However, the MSC-1 group exhibited a significantly higher maximum force than the control-1 group (p < 0.05), and the MSC-2 group demonstrated a slightly higher maximum force than the control-2 group (p > 0.05). The difference in maximum force between MSC-1 and MSC-2 groups was not significant (p > 0.05). Furthermore, the stiffness of the MSC-1 group was significantly higher than the both the control-1 (p < 0.01) and MSC-2 (p < 0.05) groups.

    Figure 6. Biomechanical testing of the Achilles tendon.

    Comparing maximum force and stiffness among the normal, MSC and control groups.

    **p < 0.01 for the MSC-1 group compared with the control-1 group.

    #p < 0.05 for the MSC-1 group compared with the MSC-2 group.

    MSC: Mesenchymal stem cell.

    Discussion

    Tendon injuries generally include acute rupture and chronic tendinopathy (or tendinosis) [24]. Regarding this study, we established collagenase-induced tendinopathy models through chemical injury. Tendinopathy arises from the failure of cumulative microinjuries to heal, primarily due to the scarcity of cells and blood vessels [5,25]. The injection of collagenase into the Achilles tendon triggers an inflammatory response, characterized by an acute phase within 3–7 days (early) followed by a proliferative phase lasting approximately 3 weeks, with subsequent remodeling occurring at 4–8 weeks after injury (advanced) [24,26]. The timing of treatment in this study was strategically chosen based on this process [19]. Previous research has demonstrated that the application of MSCs can enhance tendon healing through paracrine effects, influencing the associated inflammatory response and matrix remodeling [27]. Our study revealed that the administration of hUC-MSCs, both in the early and advanced stages of tendinopathy, ameliorated the pathological features of the Achilles tendon. Following early intervention, the Achilles tendon exhibited elevated expression of type I collagen, larger collagen fibrils and improved biomechanical properties.

    During the early stages of tendon injury, there is a decrease in type I collagen and an increase in type III collagen, leading to scar tissue formation, eventually replaced by type I collagen in the advanced stage [27,28]. Prolonged absence of quick repairs can result in an abnormal collagen I/III ratio, potentially compromising tendon strength, causing mechanical microdamage and inducing degeneration [29]. Studies have suggested that stem cell therapy can stimulate tendon cells, accelerate scar tissue formation, restore the collagen ratio, normalize tendon structure early and facilitate the recovery of tendon strength, thus preventing mechanical microinjury and further degenerative changes [16,30]. In this study, the synthesis of type I collagen increased, and the proportion of type III collagen decreased in the stem cell group, with a more pronounced effect observed with early delivery of hUC-MSCs.

    Our findings demonstrate that treatment with hUC-MSCs effectively reduced the expression of inflammatory factors IL-1β and TNF-α in injured Achilles tendons, with early MSC delivery exhibiting a potentially greater efficacy in mitigating inflammation. Following tendon injury, M1 macrophages predominantly assume a proinflammatory role by releasing cytokines such as IL-1β, TNF-α and IL-6. This response is followed by the accumulation of M2 macrophages, which play an anti-inflammatory role by releasing cytokines such as IL-10 and TGF-β [28]. Prior studies have indicated that MSCs possess the ability to regulate the inflammatory response by inhibiting the pro-M1 phenotype and promoting the polarization of macrophages to the M2 phenotype [31,32]. Moreover, matrix synthesis and degradation in healthy tendons are tightly regulated by MMP and TIMP. Overexpression of MMP-3 can stimulate collagen matrix degradation, whereas TIMP-3 inhibits the activity of several MMPs [33,34]. Our results indicate that the MSC group exhibited lower MMP-3 expression and higher TIMP-3 expression compared with the control group. This trend was more prominent in the MSC-1 group than in the MSC-2 group. Therefore, it may be inferred that early intervention not only reduces inflammation but also influences the expression of MMP and TIMP, ultimately promoting the synthesis of type I collagen.

    Injured tendons harbor progenitor cells with robust chondrogenic potential, rendering them susceptible to heterotopic ossification in an inflammatory milieu [35]. Histological assessments revealed degenerative changes in tendons of the control group. The transplantation of MSCs demonstrated a noteworthy reduction in inflammation and a potential decrease in calcification deposition in the MSC groups during tendon repair.

    The microenvironment plays a pivotal role in inflammation and matrix synthesis during various stages of tendinopathy. While numerous studies have investigated MSC delivery for treating tendinopathy [36], our research introduces a novel aspect by examining the impact of MSC delivery time, representing a significant innovation. Our study provides a preliminarily exploration of the therapeutic effects of MSCs in both early and advanced tendinopathy stages, shedding light on their impact on inflammation and collagen synthesis. In future investigations, we intend to delve into the interactive effects between the microenvironment at different stages of tendinopathy and MSCs, paving the way to identify potential strategies for further enhancing the efficacy of MSCs.

    Despite the valuable insights gained from this study, certain limitations should be acknowledged. First, while the collagenase model can serve as a reference, it is not a perfect simulation of human Achilles tendinopathy. Second, the study design lacked a sufficient number of time points, limiting the assessment of both early and prolonged performances of hUC-MSCs in treating Achilles tendinopathy. An early time point to explore inflammation and a prolonged time point to assess therapeutic effect should be added in the future work. Finally, young collagenase-induced tendinopathy rats may not fully mirror the overuse-induced tendinopathy seen in humans. Although tendon degeneration is a common manifestation in both cases, including features such as cell and vascular hyperplasia, disordered collagen fibers and heterotopic ossification [16,37], caution should be exercised in extrapolating the results of this research to human diseases.

    Conclusion

    The administration of hUC-MSCs proves to be a promising intervention for ameliorating the pathological features of collagenase-induced Achilles tendinopathy in rats. This therapeutic approach effectively enhances collagen matrix synthesis and remodeling, mitigates local inflammation and improves the biomechanical properties of the Achilles tendon. Importantly, our findings underscore the greater efficacy hUC-MSC interventional therapy in the early stage of Achilles tendinopathy compared with the advanced stage.

    Summary points
    • Mesenchymal stem cell (MSC) therapy for tendinopathy has been explored in several studies. Notably, this study pioneers the investigation into the impact of MSC delivery stages on the treatment outcomes of tendinopathy.

    • The administration of human umbilical cord MSCs (hUC-MSCs) demonstrates a significant improvement in the pathological manifestations of Achilles tendinopathy, concurrently enhancing tendon strength.

    • Early delivery of hUC-MSCs exhibits superior efficacy compared with advanced delivery in the treatment of Achilles tendinopathy. This is evidenced by increased type I collagen synthesis and enhanced biomechanical properties.

    • Early intervention plays a pivotal role in reducing inflammation, particularly pronounced in the early stages of tendinopathy. It also influences the expression of matrix metalloproteinase and tissue inhibitor of metalloproteinase, potentially facilitating collagen synthesis.

    • This study lays a foundation for guiding the clinical application of hUC-MSCs in treating Achilles tendinopathy. The encouraging results prompt further exploration through rigorous clinical studies to establish robust evidence.

    Supplementary data

    To view the supplementary data that accompany this paper please visit the journal website at: www.futuremedicine.com/doi/suppl/10.2217/rme-2023-0222

    Author contributions

    Z Yuan and D Yu: conceptualization, design, operation, drafting and revision; Y Wang: assessment and revision; L Liu: analysis; J Wang: assessment; C Ma and S Wu: organization, conceptualization, analysis, revision and corresponding. All authors reviewed and approved the final manuscript.

    Acknowledgments

    The authors would like to thank the staff of the Experimental Animal Center of Sun Yat-sen University for animal feeding and model-making support.

    Financial disclosure

    This work received support from the National Natural Science Foundation of Guangdong Province (2023A1515010503), the program of Guangdong Provincial Clinical Research Center for Rehabilitation Medicine (2023B110003), the Guangzhou Key Laboratory of Neuromodulation and Regenerative Medicine Rehabilitation (2024A03J0699), and the Sun Yat-sen Medical-Industrial Integration Project (YXYGRH202201). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

    Competing interests disclosure

    The authors have no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, stock ownership or options and expert testimony.

    Writing disclosure

    No writing assistance was utilized in the production of this manuscript.

    Open access

    This work is licensed under the Attribution-NonCommercial-NoDerivatives 4.0 Unported License. To view a copy of this license, visit http://creativecommons.org/licenses/by-nc-nd/4.0/

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Millar NL, Silbernagel KG, Thorborg K et al. Tendinopathy. Nat. Rev. Dis. Primers 7(1), 1 (2021).
    • 2. Millar NL, Siebert S, Mcinnes IB. Europe rules on harm from fluoroquinolone antibiotics. Nature 566(7744), 326 (2019).
    • 3. Kayce J. Gross anatomy: Achilles tendon. Clin. Podiatr. Med. Surg. 39(3), 405–410 (2022).
    • 4. Kujala UM, Sarna S, Kaprio J. Cumulative incidence of Achilles tendon rupture and tendinopathy in male former elite athletes. Clin. J. Sport Med. 15(3), 133–135 (2005).
    • 5. Benjamin M, Ralphs JR. Tendons and ligaments – an overview. Histol. Histopathol. 12(4), 1135–1144 (1997).
    • 6. Van Der Vlist AC, Winters M, Weir A et al. Which treatment is most effective for patients with Achilles tendinopathy? A living systematic review with network meta-analysis of 29 randomised controlled trials. Br. J. Sports Med. 55(5), 249–256 (2021).
    • 7. Malliaras P. Physiotherapy management of Achilles tendinopathy. J. Physiother. 68(4), 221–237 (2022).
    • 8. Cooper MT. Common painful foot and ankle conditions: a review. JAMA 330(23), 2285–2294 (2023).
    • 9. Li T, Xia M, Gao Y, Chen Y, Xu Y. Human umbilical cord mesenchymal stem cells: an overview of their potential in cell-based therapy. Expert Opin. Biol. Ther. 15(9), 1293–1306 (2015).
    • 10. Ahani-Nahayati M, Niazi V, Moradi A et al. Umbilical cord mesenchymal stem/stromal cells potential to treat organ disorders; an emerging strategy. Curr. Stem Cell Res. Ther. 17(2), 126–146 (2022).
    • 11. Couto PS, Shatirishvili G, Bersenev A, Verter F. First decade of clinical trials and published studies with mesenchymal stromal cells from umbilical cord tissue. Regen. Med. 14(4), 309–319 (2019).
    • 12. Mebarki M, Abadie C, Larghero J, Cras A. Human umbilical cord-derived mesenchymal stem/stromal cells: a promising candidate for the development of advanced therapy medicinal products. Stem Cell Res. Ther. 12(1), 152 (2021). • Review about the characteristics of human umbilical cord mesenchymal stem cells.
    • 13. Chen L, Liu J-P, Tang K-L et al. Tendon derived stem cells promote platelet-rich plasma healing in collagenase-induced rat achilles tendinopathy. Cell. Physiol. Biochem. 34(6), 2153–2168 (2014).
    • 14. Valencia Mora M, Antuña Antuña S, García Arranz M, Carrascal MT, Barco R. Application of adipose tissue-derived stem cells in a rat rotator cuff repair model. Injury 45(Suppl. 4), S22–S27 (2014).
    • 15. 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. 14, 103–110 (2020). • Study on stem cells in treating early tendinopathy.
    • 16. Oshita T, Tobita M, Tajima S, Mizuno H. Adipose-derived stem cells improve collagenase-induced tendinopathy in a rat model. Am. J. Sports Med. 44(8), 1983–1989 (2016).
    • 17. Wei B, Ji M, Lin Y et al. Mitochondrial transfer from bone mesenchymal stem cells protects against tendinopathy both in vitro and in vivo. Stem Cell Res. Ther. 14(1), 104 (2023).
    • 18. Ueda Y, Inui A, Mifune Y et al. Molecular changes to tendons after collagenase-induced acute tendon injury in a senescence-accelerated mouse model. BMC Musculoskelet. Dis. 20(1), 120 (2019). •• Method of collagenase-induced Achilles tendinopathy model-making.
    • 19. Li S, Wu Y, Jiang G et al. Intratendon delivery of leukocyte-rich platelet-rich plasma at early stage promotes tendon repair in a rabbit Achilles tendinopathy model. J. Tissue Eng. Regen. Med. 14(3), 452–463 (2020). •• Study on early and advanced delivery in treating tendinopathy.
    • 20. Ge Z, Li W, Zhao R et al. Programmable DNA hydrogel provides suitable microenvironment for enhancing TSPCS therapy in healing of tendinopathy. Small 19(32), e2207231 (2023).
    • 21. Semis HS, Gur C, Ileriturk M, Kandemir FM, Kaynar O. Evaluation of therapeutic effects of quercetin against Achilles tendinopathy in rats via oxidative stress, inflammation, apoptosis, autophagy, and metalloproteinases. Am. J. Sports Med. 50(2), 486–498 (2022).
    • 22. Movin T, Gad A, Reinholt FP, Rolf C. Tendon pathology in long-standing achillodynia. Biopsy findings in 40 patients. Acta Orthop. Scand. 68(2), 170–175 (1997).
    • 23. Chen J, Yu Q, Wu B et al. Autologous tenocyte therapy for experimental Achilles tendinopathy in a rabbit model. Tissue Eng. Part A. 17(15–16), 2037–2048 (2011).
    • 24. Voleti PB, Buckley MR, Soslowsky LJ. Tendon healing: repair and regeneration. Ann. Rev. Biomed. Eng. 14, 47–71 (2012).
    • 25. Tran PHT, Malmgaard-Clausen NM, Puggaard RS et al. Early development of tendinopathy in humans: sequence of pathological changes in structure and tissue turnover signaling. FASEB J. 34(1), 776–788 (2020).
    • 26. Yang G, Rothrauff BB, Tuan RS. Tendon and ligament regeneration and repair: clinical relevance and developmental paradigm. Birth Defects Res. C. Embryo Today 99(3), 203–222 (2013).
    • 27. Migliorini F, Tingart M, Maffulli N. Progress with stem cell therapies for tendon tissue regeneration. Expert Opin. Biol. Ther. 20(11), 1373–1379 (2020).
    • 28. Millar NL, Murrell GaC, Mcinnes IB. Inflammatory mechanisms in tendinopathy – towards translation. Nat. Rev. Rheumatol. 13(2), 110–122 (2017).
    • 29. Xu Y, Murrell GaC. The basic science of tendinopathy. Clin. Orthopaed. Relat. Res. 466(7), 1528–1538 (2008).
    • 30. Quintero D, Perucca Orfei C, Kaplan LD, De Girolamo L, Best TM, Kouroupis D. The roles and therapeutic potential of mesenchymal stem/stromal cells and their extracellular vesicles in tendinopathies. Front. Bioeng. Biotechnol. 11, 1040762 (2023).
    • 31. Manning CN, Martel C, Sakiyama-Elbert SE et al. Adipose-derived mesenchymal stromal cells modulate tendon fibroblast responses to macrophage-induced inflammation in vitro. Stem Cell Res. Ther. 6(1), 74 (2015).
    • 32. Chen Z, Jin M, He H et al. Mesenchymal stem cells and macrophages and their interactions in tendon-bone healing. J. Orthopaed. Translat. 39, 63–73 (2023).
    • 33. Rees JD, Stride M, Scott A. Tendons – time to revisit inflammation. Br. J. Sports Med. 48(21), 1553–1557 (2014).
    • 34. Cabral-Pacheco GA, Garza-Veloz I, Castruita-De La Rosa C et al. The roles of matrix metalloproteinases and their inhibitors in human diseases. Int. J. Mol. Sci. 21(24), (2020).
    • 35. Shen H, Cheng L, Zheng Q, Liu W, Wang Y. Scavenging of reactive oxygen species can adjust the differentiation of tendon stem cells and progenitor cells and prevent ectopic calcification in tendinopathy. Acta Biomater. 152, 440–452 (2022).
    • 36. Li W, Lin MN, Lu YP, Sui Y, Jiang M. Feasibility and advantages of mesenchymal stem cells for repairing Achilles tendon injury. Chin. J. Tissue Eng. Res. 27(15), 2404–2411 (2023).
    • 37. Nourissat G, Berenbaum F, Duprez D. Tendon injury: from biology to tendon repair. Nat. Rev. Rheumatol. 11(4), 223–233 (2015). • Review on biological therapies and mechanisms of tendon injury.