Open Access

Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review)

  • Authors:
    • Yongjia Zhu
    • Jiapeng Yan
    • Hongfei Zhang
    • Guanxing Cui
  • View Affiliations

  • Published online on: November 3, 2023     https://doi.org/10.3892/ijmm.2023.5324
  • Article Number: 121
  • Copyright: © Zhu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In sports medicine, injuries related to the insertion of tendons into bones, including rotator cuff injuries, anterior cruciate ligament injuries and Achilles tendon ruptures, are commonly observed. However, traditional therapies have proven to be insufficient in achieving satisfactory outcomes due to the intricate anatomical structure associated with these injuries. Adult bone marrow mesenchymal stem cells possess self‑renewal and multi‑directional differentiation potential and can generate various mesenchymal tissues to aid in the recovery of bone, cartilage, adipose tissue and bone marrow hematopoietic tissue. In addition, extracellular vesicles derived from bone marrow mesenchymal stem cells known as exosomes, contain lipids, proteins and nucleic acids that govern the tissue microenvironment, facilitate tissue repair and perform various biological functions. Studies have demonstrated that bone marrow mesenchymal stem cell‑derived exosomes can function as natural nanocapsules for drug delivery and can enhance tendon‑bone healing strength. The present review discusses the latest research results on the role of exosomes released by bone marrow mesenchymal stem cells in tendon‑bone healing and provides valuable information for implementing these techniques in regenerative medicine and sports health.

1. Introduction

The tendon-bone interface is a transitional region of soft tissue-to-bone transformation composed of the tendon, uncalcified fibrocartilage, calcified fibrocartilage and bone. It is a highly specialized structure (1). The connection between the tendon or ligament and the bone is established. The tendon-bone interface transmits the force generated by skeletal muscle contraction to the bone, providing strong support for joint movement and facilitating the gradual transfer of intricate mechanical loads in an efficient manner. In addition, it also plays an essential role in maintaining homeostasis and intercellular communications. However, due to the enormous difference in elastic modulus between tendon and bone, the tendon is a stress-bearing area, which significantly increases the risk of injury (2). Tendon-bone insertion (TBI) injuries are a common musculoskeletal disorder, including the anterior cruciate ligament (ACL) and the rotator cuff (RC) and Achilles tendon injuries. Injuries of the ACL and RC are the most common (3). Survey data indicate that the incidence of RC tears in the general population is ~20.7%, and increases with age (4). ACL injuries occur in >2 million patients annually in the United States, accounting for more than half of all knee injuries (5). Local tendon swelling, pain and functional impairment typically accompany TBI injury. This condition severely affects the quality of life of affected individuals and is a significant cause of pain and disability (6).

Bone marrow mesenchymal stem cells (BMSCs) are adult stem cells derived from bone marrow with self-replication and multiple differentiation potential that can differentiate into various cell types, including osteoblasts, chondrocytes and adipocytes, and these entities possess the capacity to efficiently restore impaired tissues, thus rendering them ideal for regenerative medicine and tissue engineering repair (7,8). BMSCs have the advantages of convenient sampling, a potent proliferative ability, low immunogenicity, easy gene transfection and strong differentiation potential, and they are the optimal type of stem cells used in current research and clinical applications. With further research and the development of related technologies, BMSCs are widely used in the treatment of orthopedic clinical diseases. Previous studies have indicated that BMSCs can promote bone, cartilage, muscle, ligament and tendon healing (9,10). BMSCs possess the ability to modulate the expression and release of growth factors and cytokines in the vicinity of the injury site, thereby facilitating the process of wound healing and regeneration of damaged tissues (11,12).

Exosomes (EXOs) are nanoparticles with lipid bilayers and membrane structures that are naturally released by cells through cytoplasmic exocytosis (13). Research has indicated that BMSCs release EXOs containing various bioactive substances in a paracrine manner to regulate the local microenvironment of tissues and physiological and pathological activities in cells and may be critical factors by which BMSCs play a therapeutic role (14).

2. Exosomes

EXO biogenesis and uptake

EXOs are derived from intracellular bodies termed multivesicular bodies (MVBs) and are nanoscale extracellular vesicles, with a diameter of ~30 to 150 nm (15,16) and a density between 1.1 and 1.2 g/ml. They are found in various cell types and extracellular fluids, such as plasma, synovial fluid, urine, amniotic fluid, saliva, cerebrospinal fluid and breast milk (17-21). The cell membrane invaginates to form early endosomes, which turn into late endosomes under the control of various cellular signaling pathways (22-24). Stage 2 refers to the budding of late endosomes to form MVBs. Stage 3 of the process entails the amalgamation of MVBs with the plasma membrane, followed by the selective recruitment of cytoplasmic elements, including proteins, RNA and lipids, to form intact EXOs. The process of EXO formation is relatively complex and is regulated by a number of factors, such as tetraspanins, cholesterol, endosomal sorting complexes responsible for transport, sphingomyelinases and adhesion molecules, after which they are released into the microenvironment (25,26). When EXOs were first discovered, scholars considered them a way to expel unwanted components from cells (27). As research progressed, receptor-ligand interactions with EXOs, and direct membrane fusions for the delivery of biomolecules to target cells and mediate intercellular communications have been verified (28).

Structure and function of EXOs

Exosomal vesicles are rich in lipids, proteins and various types of nucleic acids, such as DNA, mRNAs, microRNAs (miRNAs/miRs) and long non-coding RNAs, transcription factors and cytokine receptors. Organelles, such as ribosomes and mitochondria are typically absent. EXOs mediate cell-cell and cell-extracellular matrix (ECM) communications through the biologically active molecules within them to induce the necessary growth signals and transcriptional changes to cause phenotypic changes in the local environment (29,30). Various physiological and pathological mechanisms have been observed to exert an effect on recipient cells, including the amelioration of ischemic brain injury and the stimulation of angiogenesis, which leads to the acceleration of skin wound healing, thus holding promise as a potential therapeutic approach for addressing osteoarthritis (31-33). Depending on the cell type that produces the EXOs and changes in the cellular microenvironment, such as physiological and pathological states, the number of EXOs secreted and the composition of the contents of EXOs varies (34). For example, BMSC-derived EXOs (BMSC-EXOs) contain a variety of anti-inflammatory factors and growth factors, such as transforming growth factor-β (TGF-β), interleukin (IL)-10 and tumor necrosis factor (TNF)-stimulated gene ((TSG)-6 (35). It has been demonstrated that BMSC-EXOs contain miR-301a, miR-22 and miR-let-7, which are involved in immune-related pathways (36). Given their significant osmotic impact, EXOs possess the ability to traverse the blood-brain and blood-spinal cord barriers effectively. Moreover, they exhibit a selective propensity to infiltrate sites of inflammation within bodily tissues, thereby exerting regulatory control over the inflammatory response (37,38). In addition, the membrane transport process is involved in the production of EXOs. Several proteins that are closely related to this process, such as tetraspanins (CD9, CD63 and CD81), the membrane proteins, annexin and flotillin, heat shock proteins (HSP70 and HSP90) and markers of the endosomal sorting complex required for transport pathway, have been identified. Several proteins, including lysosomal-associated membrane protein 1 and TSG101, are highly enriched in various cell-derived EXOs (39). Therefore, they are often used as the marker proteins of EXOs and are widely used to extract and purify EXOs. The morphological and structural pattern of EXOs is illustrated in Fig. 1.

3. TBI

Physical structure and physiopathology of TBI

There are two types of TBI: Indirect insertion and direct insertion. For example, indirect insertion involves the insertion of the medial collateral ligament into the tibia. The ACL and RC are anatomical structures that are intricately integrated into the bone through a gradient structure comprising four consecutive layers of distinct tissue. These layers can be categorized from soft to hard, starting with the tendon, followed by noncalcified fibrocartilage, calcified fibrocartilage and ultimately bone tissue (40-42). Once the tendon-bone connection is damaged, restoring the natural anatomical structure is difficult (Fig. 2).

Tendon cells are responsible for synthesizing and secreting collagen, and are the basic units of tendon function. Healthy tendons primarily contain type I collagen, which provides mechanical strength and durability. Type II collagen is found in lower amounts and is typically concentrated where tendons connect to bones. Following a tendon injury, type III collagen is abundant in scar tissue and is crucial for tendon healing. An imbalance in type III and type I collagen in the tendons can cause mechanical damage. Notably, in addition to altering the phenotype and function of various cells, MSC-EXOs can also increase the content of type III collagen by increasing the level of TGF-β1, thus promoting tendon healing (43).

Tendon cells are closely associated with RC disease; studies have shown that they are involved in tendon repair through proliferation and migration (44). For example, tendon stem cell-derived EXOs induce tendon differentiation in mesenchymal stem cells via TGF-β (45), promote healing in injured tendons by balancing the synthesis and degradation of tendon extracellular matrix (6), and can regulate inflammation and promote high-quality healing of injured tendons (46).

TBI injuries are typically classified as acute and chronic. If not repaired in time, acute injury can gradually transform into chronic injury. The overuse or overload of tendons often leads to injury (42), while other factors such as age, metabolism and blood pressure can also exacerbate tendon injury (43). This condition severely affects the quality of life of affected individuals, and is a major cause of pain and disability (6). The distinguishing characteristics of the healing process between tendons and bones encompass the fusion of bone, heightened mechanical thresholds and the constricting of the bone tunnel (47,48). Tendon healing is divided into four phases: Inflammatory, proliferative, remodeling and maturation (49). During tendon healing, the levels of several growth factors, including TGF-β, platelet-derived growth factor, vascular endothelial growth factor (VEGF) and insulin-like growth factor-1, are increased and play critical roles in all stages of the recovery process (50).

TBI healing is very complex for several reasons. First, the region of fibrocartilage at the site of injury exhibits a deficiency in cells and blood supply, leading to a delayed or impaired healing process in TBI (51). Second, the regeneration of new bone is typically very slow, and strength and stiffness are correspondingly reduced (2). Third, during the healing of TBI, an initial inflammatory response occurs at the junction between the tendon and bone, and a large number of macrophages infiltrate, fibroblasts release extracellular matrix to form granulomas, and a large amount of collagen is deposited; the occurrence of fibrovascular scar tissue formation in the local area is a consequence that can be observed (52). In contrast to the initial anatomical arrangement, scar tissue has a significant impact on the process of new bone generation and the development of the interface between the tendon and bone. In comparison to the original physiological system, the presence of scar tissue has notable implications for the regeneration of new bone. The development of the tendon-bone interface and its ability to mediate load transmission and stress dispersion experiences a notable decline, which leads to decreased biomechanical properties in poorly regenerated tendons, impaired motor function, a shortened service life and a significantly reduced quality of healing (8,53).

Conventional treatment of TBI injuries
Surgical treatment

Non-surgical treatments often need to be revised. These strategies can only be used to control pain (54). Therefore, in the majority of cases, TBI injury requires surgical intervention. Thus, for decades, surgical reconstruction has been considered the standard treatment for attachment point injuries. The clinical treatment of ACL and RC injuries often involves tendon/ligament reconstruction surgery, in which the insertion of the tendon graft into the bone tunnel is essential for promoting tendon-bone healing. A critical aspect of this healing process is the regeneration of the fibrocartilage zone (55,56). During the surgical procedure of reconstruction, the graft is carefully positioned and subsequently maneuvered through a bone tunnel. The indigenous and directly implanted transitional tissue fails to undergo regeneration, instead giving rise to fibrovascular scar tissue at the interface of the graft and tunnel (57). Initially, this tissue exhibits a lack of organization. After 3-4 weeks, vertical fibers that resemble indirectly inserted threads begin to form. At ~1 year after surgery, the bone gradually grows into the graft-tunnel interface, enhancing the strength of graft attachment and facilitating the integration of the graft with the adjacent bone (57).

The process of fibrocartilage healing involves the formation of scar tissue following an injury, which is rich in type I collagen (40,58,59). The fibrocartilage zone plays a vital role in the absorption of stress that occurs between the tendon and bone (2). Nevertheless, scar tissue does not possess the inherent gradient structure and organized alignment of collagen fibers present in the native tendon-bone interface, resulting in low biomechanical properties, weak mechanical strength and increased re-tearing rate (60). There are data indicating high recurrence rates following TBI; for example, recurrent retear rates after RC repair are typically 20 to 30%, and can be as high as 94%, and ACL reconstruction has an average failure rate of 11.7% (40,61,62). For other commonly injured tendons and ligaments, such as the distal biceps tendon and lateral ankle sprain (anterior talofibular ligament, calcaneofibular ligament and posterior talofibular ligament), the rates of re-rupture were 1.6 and 18.1%, respectively (63). The most important cause of recurrent retears suboptimal tendon-bone healing. Promoting the healing of the connection between tendons and bones is crucial in the treatment of TBI injury and in preventing the occurrence of another tear after reconstruction. In contrast to interosseous recovery, the regeneration of fibrocartilage transitions is restricted, and TBI regeneration is particularly difficult. Insufficient osseointegration following surgical reconstruction is one of the main reasons for unsatisfactory clinical results (64). In addition, joint stiffness and pain caused by long-term post-operative immobilization can significantly affect the outcome of surgical treatment.

Tissue engineering therapy

Over the past few years, there has been a significant application of growth factors/cytokines, platelet-rich plasma, physiotherapy, tissue engineering, and different delivery and induction techniques (65-68). Currently, a diverse range of growth factors and cytokines, such as TGF-β, bone morphogenetic protein (BMP) and granulocyte colony-stimulating factor, have been employed to enhance the healing of tendon grafts within bone tunnels in different animal models. However, the implementation of these methods in clinical settings still has certain restrictions. Of note, retaining these biological factors at the specific location where the tendon or ligament injury occurred is challenging, and they are lost or rapidly removed (62).

Platelet-rich plasma (PRP) therapy

In recent years, multiple preclinical studies have demonstrated the role of PRP in promoting TBI interface healing and improving biomechanical properties (69-71). Several systematic reviews have reported that PRP can enhance tendon and ligament tissue by promoting tendon cell proliferation and angiogenesis, thus significantly promoting healing and controlling pain, and this treatment has exhibited sound therapeutic effects on RC and lateral elbow injuries (72-74). However, some studies have reported the potential limitations of PRP therapy; for example, the results of a randomized controlled trial suggested that injecting platelet-rich plasma did not improve tendon function or the quality of life in patients with severe acute Achilles tendon rupture. There was no evidence of any benefit from the injection of platelet-rich plasma (75). In addition, Bennell et al (76) used PRP therapy to treat patients with knee osteoarthritis and did not achieve significant efficacy. In summary, as a new technology, PRP therapy has the advantages of less trauma and fewer complications. It is a promising treatment strategy for TBI injury; however, its application scope, indications and efficacy warrant further investigation.

Stem cell therapy

Stem cell-based cell therapy is anticipated to emerge as a viable substitute for tissue engineering therapy. Mesenchymal stem cells can self-renew and differentiate into various cell types, including adipocytes, osteoblasts and chondrocytes, and exhibit low immunogenicity when transplanted. In the context of healing tendons and bones, the stem cells that are frequently utilized are typically obtained from either the bone marrow (BMSCs) or adipose tissue (adipose tissue-derived stem cells). It has been demonstrated that mesenchymal stem cells derived from the periosteum, synovium and tendon have been extensively employed to enhance the healing process at locations where tendons and bones meet (77). Although the results of cell therapies are promising, they also have some limitations. For instance, the absence of a standardized protocol for the dosage and frequency of stem cell therapy hinders the attainment of optimal outcomes. This poses a significant challenge to the widespread clinical implementation of stem cells (55,78). The clinical application of stem cell transplantation therapy still needs to be improved, including factors such as cell dedifferentiation, low cell survival rate, immune rejection and ethical issues (79). To address these limitations, it is necessary to investigate an innovative treatment method that can effectively meet the clinical requirements and enhance the healing process of the tendon and bone. The conventional treatment methods for TBI injuries, as well as their advantages, disadvantages and clinical applications are presented in Table I.

Table I

Conventional treatment of TBI injuries.

Table I

Conventional treatment of TBI injuries.

TreatmentAdvantageDisadvantageClinical application
Surgical treatmentSurgical treatment is by far the most widely used.
After years of development, the surgical technique is mature.
It is almost the only effectivereatment for tsevere TBI injury.
The trauma is relatively large.
Post-operativecomplications, such as infection, pain andjoint stiffness are common.
Traditional surgical treatment.
Arthroscopic treatment.
Tissue engineering therapyThe trauma is minimal.Bioactive factors have poor stability and are rapidly degraded and inactivated at the site of tendon/ligament injury.Tissue engineering scaffold.
Biological factor injection.
Platelet-rich plasma therapyIt has the advantages of low cost, simple operation and easy clinical promotion.
There was no infection and immune rejection.
The efficacy of treatment has not been well established.
The mechanism of action is not clear.
Autologous platelet-rich plasma injection.
Stem cell therapyStem cells can self-renew and have the potential of multidirectional differentiation.
Stem cells can promote tissue repair through paracrine.
Stem cells have a wide range of tissue sources and are relatively easy to isolate.
The biological mechanism of action remains unclear.
The dose and duration of stem cell therapy have not been achieved to ensure the optimal result.
The immune rejection of stem cells and the safety of genetically engineered stem cells cannot be completely guaranteed.
Bone marrow mesenchymal stem cells (BMSCs).
Synovium-derived mesenchymal stem cells (SMSCs).
Adipose-derived stem cells (ADSCs).
Periosteum-derived periosteal stem cells (PSCs).
ACL-derived stem cells.

4. Research progress on BMSC-EXOs in the repair of TBI injury

BMSCs-EXOs have been used to treat diseases of the respiratory system (such as the treatment of acute lung injury (ALI), circulatory system (such as the treatment of ischemic myocardial infarction), digestive system (such as the repair of liver damage), nervous system diseases (such as the treatment of brain injury and stroke), urinary system diseases (such as the treatment of ischemia reperfusion kidney injury), reproductive system diseases (such as repairing ovarian function) and breast diseases (such as breast cancer treatment). At present, BMSC-EXOs have been used in the research of diseases of multiple systems, providing new insight for the research and application of clinical drugs in the future. With extensive research on mesenchymal stem cells in recent years, BMSC-EXOs have also been widely used in the treatment of motor system diseases, and BMSC-EXOs have been used to treat TBI injury.

As natural cell products, EXOs have good biocompatibility, can penetrate biological barriers, exert therapeutic effects and can be used to treat bone-related diseases. It has been reported that mesenchymal stem cells from different sources can be used to promote tendon-bone healing, and in recent times, a substantial amount of evidence has indicated that the beneficial impacts of stem cell therapy could potentially occur through the release of extracellular vesicles termed EXOs from MSCs via a process known as paracrine signaling (2,58,80-83). This paracrine mechanism, which is known as stem cell conditioned medium (CM), secretome CM or secretome, is the medium in which stem cells are cultured and contains soluble proteins, lipids, nucleic acids and extracellular vesicles or microvesicles (84,85). These vesicles are further divided into EXOs and shedding vesicles. A summary of the advances in the promotion of tendon and bone healing by BMSC-EXOs is presented in Table II.

Table II

Recent advances in the promotion of tendon and bone healing by BMSC-EXOs.

Table II

Recent advances in the promotion of tendon and bone healing by BMSC-EXOs.

Author(s)Affected tissueStudy typeMain resultsMechanism(Refs.)
Huang et alRCIn vivo and in vitroBMSC-EXOs inhibited inflammation by inhibiting the polarization of M1 macrophages and the secretion of proinflammatory factors, confirming that BMSC-EXOs can increase the fracture load and stiffness of the RC after reconstruction, induce angiogenesis and inhibit inflammation around RC endpoints, thereby promoting tendonbone healing after RC reconstruction in rats.Regulation of macrophage phenotypic polarization.(100)
Li et alACLIn vivo and in vitroBMSC-EXOs promoted the polarization of M1 macrophages to M2 macrophages through miR-23a-3p, suggesting that early BMSC-EXOs treatment could inhibit the inflammatory response at the tendon-bone interface, promote fibrocartilage regeneration, and accelerate tendon-bone healing afterACL reconstruction. Moreover, the overexpression of miR-23a-3p could enhance the therapeutic effect.Regulation of macrophage phenotypic polarization.(8)
Shi et alTBIIn vivo and in vitroBMSC-EXOs improved the inflammatory microenvironment and promoted fibrocartilage regeneration at the tendon-bone interface by increasing M2 macrophage polarization, reducing the expression of proinflammatory cytokines IL-1β and IL-6, and enhancing the expression of anti-inflammatory cytokines IL-10, TGF-β, and insulinlike growth factor (IGF) during tendon healing. Thus, the biomechanical properties of tendon-to bone healing can be improved.Regulation of macrophage phenotypic polarization.(101)
Takayama et alACLIn vivo and in vitroInhibition of VEGF expression impairs revascularization and inhibits the biomechanical strength of tendon grafts after ACL reconstruction surgery. At the same time, overexpression of VEGF affects the enhancement of the biomechanical strength of tendon grafts.Increase promotes angiogenesis.(105)
Huang et alRCIn vivo and in vitroBMSC-EXOs can promote angiogenesis around the tendon-bone interface, increase the load and stiffness of RC rupturein rats after reconstruction, and promote the growth of thetendon-bone interface. BMSC-EXOs can activate the VEGF and Hippo signaling pathways and promote the proliferation,migration, and angiogenic tube formation of human umbilical vein endothelial cells (HUVECs) under in vitro culture conditions.Increase promotes angiogenesis.(100)
Zhang et alACLIn vivo and in vitroBMSC-EXOs promoted the formation of blood vessels around the graft and improved bone microstructure, which accelerated the healing of the transplanted tendonbone tunnel after ACL reconstruction.Regulation of bone metabolism.(107)
Fang et alBMSCsIn vitroBMSC-EXOs containing tRNA-10277 could regulate the adipogenic and osteogenic potential of BMSCs.Regulation of bone metabolism.(108)
Zhang et alBMSCsIn vivo and in vitromiR-935-enriched EXOs produced by BMSCs directly enhanced osteoblast proliferation and activity by targeting signal transducer and transcription 1 (STAT1) activator to promote osteoblast proliferation and differentiation.Regulation of bone metabolism.(109)
Han et alRCIn vivo and in vitroBMP-2 and poly lactic acid (PLA) delivered by BMSC-EXOs of PASP-PLGA microcapsules promoted chondrogenic differentiation through the Smad/RUNX2 pathway, enhanced tendon interface stiffness and ultimate load strength, and promoted tendon-bone healing in rabbits with acute RC tears (RCT).Regulation of bone metabolism.(112)
Wu et alRCIn vivo and in vitroBMSC-EXOs pre-treated with low-intensity pulsed ultrasound stimulation (LIPUS) can improve fibrocartilage regeneration at the tendon-bone interface and reduce supraspinatus fat infiltration in a mouse RC repair model by delivering miR-140Regulation of bone metabolism.(113)
Cai et alRCIn vivo and in vitroLocal injection of SAH with sustained KGN-EXOs release could effectively promote cartilage formation as well as collagen maturation and organization for enthesis regeneration, contributing to enhanced biomechanical properties after RCR.Regulation of bone metabolism.(114)
Li et alRCIn vitroEXOs derived from BMSCs promote the proliferation, migration, and fibrotic activity of RC tenocytes, and TGF-β1 is the crucial molecule mediating the effects of EXOs, pretreatment of BMSCs with TGF-β1 can significantly promote the secretion and release of EXOs.Promotes tendon regeneration.(117)
Li et alRCIn vitroTGF-β1 treatment promoted BMSCs-EXOs secretion, in which miR-29a promoted tendon cell proliferation, migration, and fibrosis by targeting FABP3, thus improving tendon injury and RCT.Promotes tendon regeneration.(118)
Yu et alBMSCsIn vivo and in vitroBMSCs-EXOs significantly promoted the proliferation, migration and tenogenic differentiation ability of TSPCs. Moreover, by embedding BMSCs-EXOs into the fibrin gel, the control-released exosomes retained the ability of being internalized by TSPCs in vivo and promoted the regeneration of patellar tendon tissue in the defect area in rats.Promotes tendon regeneration.(116)
Li et alBMSCsIn vitroBMSC-derived exosomes promote fibroblast activation possibly through the PTEN and PI3K/Akt signaling pathways, which may serve as potential targets to further promote tendon-bone healing.Promotes tendon regeneration.(122)

[i] BMSCs, bone marrow mesenchymal stem cells; EXOs, exosomes; BMSC-EXOs, exosomes derived from BMSCs; RC, rotator cuff; TBI, tendon-bone insertion; ACL, anterior cruciate ligament; IL, interleukin; TGF, transforming growth factor; miR, microRNA.

EXOs appear to be a more promising treatment option than stem cell therapy. The benefits of EXOs are mainly characterized by several aspects: i) EXOs are smaller and have a simple structure and composition, rendering them easier to separate and preserve, while exhibiting a lower immunogenicity; ii) EXOs can block the metastasis of cells that may contain immunogenic molecules or even mutated or damaged DNA; iii) EXOs can penetrate any organ and pass through it effortlessly due to their nanoscale size, unlike large cells that cannot migrate through capillaries to the injury site; iv) EXOs have the ability to move to different parts of the body as they have specific molecules on their surfaces that guide their migration; v) EXOs, being an integral part of the human body, possess biochemical characteristics that are akin to their originating cells. As a result, they are capable of evading phagocytosis, merging with cell membranes and lysosomal fusion (86,87).

5. Mechanisms through which BMSC-EXOs promote tendon-bone healing

Regulation of macrophage phenotypic polarization

Macrophages are derived from the bone marrow mononuclear cell line, are essential to the innate immune system and play an indispensable role in the immune response. Macrophages are divided into classically activated (M1) macrophages and alternately activated (M2) macrophages in response to various environmental stimuli. These two types of macrophages exhibit significant heterogeneity in phenotype and function. M1 and M2 macrophages are considered to have pro-inflammatory and fibrotic phenotypes, respectively (88,89). Previous studies have demonstrated that macrophages play a non-negligible role in the occurrence and development of TBI injury (90-92). In the early stages following tissue injury or tendon/ligament reconstruction, macrophages are recruited in large numbers to the graft-tunnel interface and are polarized toward the M1 type, inducing inflammatory responses and engulfing apoptotic cells, and removing cell debris (90). During this period, the expression of various pro-inflammatory factors, including TNF-α, IL-12, and inducible nitric oxide synthase, is significantly increased, thereby amplifying the inflammatory response in the affected area (90). The presence of inflammation in the surrounding environment helps attract additional cells from various locations to travel to the injured area and assist in the preparation of future tissue healing. In the advanced phase of the injury, M2 macrophages replace M1 macrophages in large numbers and secrete anti-inflammatory factors, such as IL-10 and TGF-β; by doing so, this decreases the localized inflammation and enhances the local regeneration and repair of tissues (93). Thus, accelerating macrophage polarization from M1 to M2 can accelerate tissue repair (94). If this is not achieved, it can lead to a prolonged inflammatory phase, increased apoptosis and decreased cell proliferation, resulting in slow healing. In addition, this condition can induce the excessive secretion of ECM by fibroblasts, leading to soft tissue fibrosis and scar tissue formation at the injury site, which hinders cartilage regeneration and graft remodeling (95-97). Therefore, regulating the polarization of macrophages may be key to promoting early tendon-bone healing.

Previous studies have reported that EXOs regulate inflammation through macrophage polarization, reduce cell infiltration and matrix deposition, promote collagen formation, and improve fiber continuity and alignment during tendon healing remodeling (46,98,99). In addition, EXOs derived from mesenchymal stem cells have been shown to modulate macrophage polarization in several in vitro and in vivo studies. For example, Huang et al (100) discovered that BMSC-EXOs were able to suppress inflammation by preventing the activation of M1 macrophages and the release of pro-inflammatory substances. Their study confirmed that BMSC-EXOs have the potential to enhance the fracture load and stiffness in the reconstructed RC, thereby inducing angiogenesis and inhibiting inflammation around RC endpoints, and promoting the healing of tendons and bones following RC reconstruction in rats. Li et al (8), in their groundbreaking study, demonstrated that BMSC-EXOs have the ability to induce the transformation of M1 macrophages into M2 macrophages through the involvement of miR-23a-3p. This finding suggests that early treatment with BMSC-EXOs may effectively suppress the inflammatory response at the interface between the tendon and bone. Furthermore, it was observed that BMSC-EXOs facilitated the regeneration of fibrocartilage and expedited the healing process of the tendon-bone junction following ACL reconstruction. Moreover, miR-23a-3p overexpression enhanced the therapeutic effect (8). Shi et al (101) demonstrated that BMSC-EXOs improved the inflammatory microenvironment and promoted fibrocartilage regeneration at the tendon-bone interface by increasing M2 macrophage polarization, thereby reducing the expression of the pro-inflammatory cytokines. IL-1β and IL-6. and enhancing the expression of the anti-inflammatory cytokines, IL-10, TGF-β and insulin-like growth factor during tendon healing. Furthermore, it is possible to enhance the biomechanical characteristics of the healing process between tendons and bones.

The characteristics of EXOs exhibit potential for reducing initial inflammatory reactions, which is necessary for effective tissue healing (102). However, the specific mechanism through which BMSC-EXOs control the polarization and function of M2 macrophages in TBI injury is not yet completely understood and warrants further investigation. The mechanisms of BMSC-EXOs in promoting tendon-bone healing are illustrated in Fig. 3.

An increase in VEGF promotes angiogenesis

After experiencing a traumatic brain injury, there is a decrease in blood flow to the area where the tendon connects to the bone. This decrease in blood flow means that important nutrients that are necessary for the healing of the tendon and bone are not being delivered properly. As a result, the biomechanical properties of the tendon and bone are negatively affected, which in turn affects the overall recovery of the tendon and bone (103). Numerous research studies have provided evidence that neovascularization plays a crucial role in facilitating the healing process of tendon-bone. It has been consistently demonstrated that following RC reconstruction, tendon-bone healing quality can be effectively improved by improving the blood supply at the tenodesis point or trough, where the blood supply is relatively poor (104).

VEGF is a key factor that regulates the natural formation of new blood vessels in the body. It plays a crucial role in activating, increasing in number and facilitating the movement of endothelial cells which line the inside of blood vessels and improve the circulation of blood to the transplanted tendon, which in turn facilitates the healing of the tendon-bone connection (103). Takayama et al (105) provided evidence that suppressing the expression of VEGF hindered the process of blood vessel formation and also hindered the mechanical integrity of tendon grafts following ACL reconstruction surgery. Furthermore, the overexpression of VEGF affected the enhancement of the biomechanical strength of tendon grafts (105). Huang et al (100) discovered that BMSC-EXOs have the ability to enhance the formation of new blood vessels around the area where the tendon and bone meet. Additionally, they found that these exosomes can increase the strength and rigidity of the tendon in rats following reconstruction, as well as stimulate the growth of the tendon-bone junction (100). During further mechanistic analyses, the researchers verified that BMSC-EXOs have the ability to stimulate the VEGF and Hippo signaling pathways. Additionally, they can enhance the growth, movement and formation of blood vessels in human umbilical vein endothelial cells in vitro (100). Notably, the researchers discovered that the stimulation of the VEGF and Hippo signaling pathways by BMSC-EXOs could be separate. This indicates that the activation of the Hippo signaling pathway by BMSC-EXOs does not solely rely on the VEGF signaling pathway. This suggests that BMSC-EXOs have extensive and beneficial impacts on enhancing angiogenesis (Fig. 4).

Regulation of bone metabolism

Bone is a dynamic tissue that undergoes continuous remodeling by a delicate equilibrium between the creation of new bone by osteoblasts and the breakdown of old bone by osteoclasts. The process of bone formation is carefully regulated and involves the direct transformation of BMSCs into osteoblasts. Following surgical reconstruction, up to 25% of patients require revision surgery, partly due to traumatic, technical, bacterial and biological factors. A significant factor contributing to this issue is the deterioration of bone surrounding the graft, which directly affects the secure connection of the transplanted tendon to the bone tunnel. Therefore, reducing bone loss around the graft may be a therapeutic strategy with which to promote tendon-bone tunnel healing and reduce the rate of reconstruction failure.

The healing quality of the grafted tendon-to-bone tunnel is closely related to the formation of bone around the graft, which is inseparable from early post-operative angiogenesis. The blood vessels located at the boundary between the transplanted tendon and the bone tunnel supply an ample amount of oxygen and nutrients to the cells. As a result, this has a direct impact on the formation of new bone around the graft (100,106). Zhang et al (107) examined a rat ACL reconstruction model and demonstrated that BMSC-EXOs promoted the formation of blood vessels around the graft and improved bone microstructure, which accelerated the healing of the transplanted tendon-bone tunnel following ACL reconstruction.

BMSC-EXOs can participate in bone remodeling by directly regulating the proliferation and activity of osteoblasts. Fang et al (108) found that BMSC-EXOs containing tsRNA-10277 altered the adipogenic and osteogenic potential of BMSCs. Zhang et al (109) found that miR-935-enriched EXOs produced by BMSCs directly enhanced osteoblast proliferation and activity by targeting signal transducer and transcription 1 activation to promote osteoblast proliferation and differentiation. In addition, miR-218, miR-196a and miR-181a contained in BMSC-EXOs have been confirmed to exert positive regulatory effects on osteoblast differentiation (110).

In addition to targeting osteoblasts, BMSC-EXOs can regulate the activity of osteoclasts, thereby regulating bone metabolism at the tendon-bone interface. Feng et al (111) demonstrated that EXOs containing a high amount of miR-6924-5p, derived from platelet derived growth factor receptor α (+) BMSCs overexpressing Scleraxis, could be used as a novel type of nanotherapeutic agent. These EXOs were able to prevent the formation of osteoclasts by targeting two specific proteins: Osteoclast stimulatory transmembrane protein and chemokine (C-X-C motif) ligand 1. In addition, this treatment could effectively hinder the process of tunnel osteolysis and enhance the biomechanical stability of tendon-bone healing (111).

Fibrocartilage is an essential component of the tendon-bone interface. Recently, Han et al (112) reported that BMP-2 and polylactic acid delivered by BMSC-EXOs in polyaspartic acid-polylactic acid-glycolic acid copolymer microcapsules promoted chondrogenic differentiation through the Smad/RUNX2 pathway, enhanced tendon interface stiffness and ultimate load strength, and promoted tendon-bone healing in rabbits with acute RC tears. BMSC-EXOs that have undergone low-intensity pulsed ultrasound stimulation (LIPUS) have the potential to enhance the regeneration of fibrocartilage at the interface between the tendon and bone. Additionally, they can help reduce the infiltration of fat in the supraspinatus muscle in a mouse model of RC repair. This is achieved by delivering miR-140 (113). Cai et al (114) reported that the local injection of EXOs derived from kartogenin-preconditioned BMSCs had the potential to efficiently stimulate the development of cartilage, enhance the maturation of collagen, and facilitate the regeneration of tissues in the rotator cuffs of rats with chronic RC tears, and enhance biomechanical properties following RC repair (Fig. 5).

Promotion of tendon regeneration

The proliferation and migration of tendon cells are involved in tendon tissue repair and tendon-bone healing (115). Ample research has validated the positive impact of BMSC-EXOs in facilitating the restoration of tendons. For example, Yu et al (116) demonstrated that BMSC-EXOs promoted the proliferation, migration and tendon differentiation of tendon stem/progenitor cells (TSPCs) in vitro, and subsequent in vivo analyses further confirmed that BMSC-EXOs could be taken up and internalized by rat TSPCs, thereby promoting the proliferation and migration of TSPCs. This effect was characterized by improved histological scores of patellar tendons, the enhanced expression of Mohawk, tending-regulatory protein and type I collagen, and improved mechanical properties of the new tendons (116). Subsequent studies confirmed that exosomes derived from BMSCs promoted the proliferation, migration and fibrotic activity of rotator cuff tendon cells, and TGF-β1 was a key molecule that mediated the effect of exosomes (117). Pre-treatment of BMSCs with TGF-β1 can also significantly promote the secretion and release of EXOs from BMSCs. Li et al (118) first described that TGF-β1 treatment promoted BMSC-EXO secretion, and miR-29a promoted tendon cell proliferation, migration and fibrosis by targeting fatty acid binding protein 3, thereby improving tendon injury and RC tear. Several previous studies have shown that BMSCs-EXOs can promote skin wound healing by regulating the activation and proliferation of fibroblasts (119-121). Recently, Li et al (122) confirmed that BMSC-EXOs also play a critical role in promoting tendon-bone healing by promoting the proliferation and differentiation of fibroblasts. In addition, the downstream targets of miRNA, which is an active molecule in BMSCs-EXOs, have also been more fully verified in their study. According to the study conducted by Li et al (122), it was confirmed that miR-144-3p and miR-23b-3p enhanced the proliferation, migration and collagen synthesis of NIH3T3 fibroblasts through both bioinformatics analysis and in vitro experiments. Further analyses demonstrated that miR-144-3p and miR-23b-3p promoted fibroblast activation through the upregulation of the PTEN and PI3K/Akt signaling pathways (122). This discovery lays a theoretical foundation for RC tear therapy and provides a new avenue for further research (Fig. 6).

6. Conclusions and future perspectives

EXOs are small soluble vesicles secreted by various cells that can be found in cell cultures and bodily fluids. Their lipid bilayers protect proteins and nucleic acids from degradation in the extracellular environment, and facilitate intercellular communication by carrying contents across the cell membrane to the cytoplasm of recipient cells. EXOs play a crucial role in regulating life activities (123,124). BMSCs have been known to promote tissue healing and regeneration due to their multipotent stem cell properties. BMSC-EXOs, which are key products secreted by BMSCs, have also been shown to possess regenerative qualities similar to those of parental BMSCs. BMSC-EXOs exhibit great promise in serving as diagnostic or prognostic biomarkers, drug delivery systems and carriers for gene therapy in the clinical setting. These findings provide a new perspective for the study of promoting tendon healing and bring new opportunities.

In recent years, with increasingly extensive and in-depth research on the treatment of TBI injury, an increasing number of new methods have been developed. Numerous studies have suggested that purified BMSC-EXOs offer several unique advantages over BMSCs in repairing damaged tissues. They have stable biological activity, long-term preservation, fast transportation, strong permeability, good biocompatibility, ease of engineering and the ability to avoid immune responses and tumorigenesis (8,14,125). Therefore, BMSC-EXO-based decellularized therapy is a promising therapeutic approach for maintaining the regenerative properties of BMSCs, while avoiding the potential downsides associated with cell therapy (126,127).

While the beneficial effects of BMSC-EXOs on tendon-bone healing have been initially confirmed, further extensive and in-depth studies are warranted. Clinical trials for safety and efficacy should be accelerated and performed as soon as possible. Additionally, the extraction and purification of EXOs needs to improved, as factors such as high cost, complex technology, low yield and the ease of destruction limit their broad clinical applications (128). The future of EXOs in biomedical engineering is an area that requires further investigation.

In summary, BMSC-EXOs have a vast potential for use in tendon-bone healing and repair. This breakthrough presents fresh possibilities for fundamental scientific investigation, the diagnosis of medical conditions, and the management of associated illnesses. However, further research is required in order to fully comprehend the mechanisms through which BMSC-EXOs promote healing and improve the clinical application of EXOs (Fig. 7).

Availability of data and materials

Not applicable.

Authors' contributions

HZ provided a brief introduction to the article. YZ and GC were responsible for the writing of the manuscript. YZ prepared the tables. HZ and JY revised the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the Shandong Traditional Chinese Medicine Technology Project (grant no. 2021M156).

References

1 

Li M, Tang Y, Chen C, Zhou J, Zheng C, Chen H, Lu H and Qu J: Comparison of bone surface and trough fixation on bone-tendon healing in a rabbit patella-patellar tendon injury model. J Orthop Translat. 21:49–56. 2020.

2 

Zou J, Yang W, Cui W, Li C, Ma C, Ji X, Hong J, Qu Z, Chen J, Liu A and Wu H: Therapeutic potential and mechanisms of mesenchymal stem cell-derived exosomes as bioactive materials in tendon-bone healing. J Nanobiotechnology. 21:142023.

3 

Zou M, Wang J and Shao Z: Therapeutic potential of exosomes in tendon and tendon-bone healing: A systematic review of preclinical studies. J Funct Biomater. 14:2992023.

4 

Yamamoto A, Takagishi K, Osawa T, Yanagawa T, Nakajima D, Shitara H and Kobayashi T: Prevalence and risk factors of a rotator cuff tear in the general population. J Shoulder Elbow Surg. 19:116–120. 2010.

5 

Musahl V and Karlsson J: Anterior cruciate ligament tear. N Engl J Med. 380:2341–2348. 2019.

6 

Wang Y, He G, Guo Y, Tang H, Shi Y, Bian X, Zhu M, Kang X, Zhou M, Lyu J, et al: Exosomes from tendon stem cells promote injury tendon healing through balancing synthesis and degradation of the tendon extracellular matrix. J Cell Mol Med. 23:5475–5485. 2019.

7 

Arabpour M, Saghazadeh A and Rezaei N: Anti-inflammatory and M2 macrophage polarization-promoting effect of mesenchymal stem cell-derived exosomes. Int Immunopharmacol. 97:1078232021.

8 

Li Z, Li Q, Tong K, Zhu J, Wang H, Chen B and Chen L: BMSC-derived exosomes promote tendon-bone healing after anterior cruciate ligament reconstruction by regulating M1/M2 macrophage polarization in rats. Stem Cell Res Ther. 13:2952022.

9 

Yokoya S, Mochizuki Y, Natsu K, Omae H, Nagata Y and Ochi M: Rotator cuff regeneration using a bioabsorbable material with bone marrow-derived mesenchymal stem cells in a rabbit model. Am J Sports Med. 40:1259–1268. 2012.

10 

Rodeo SA, Potter HG, Kawamura S, Turner AS, Kim HJ and Atkinson BL: Biologic augmentation of rotator cuff tendon-healing with use of a mixture of osteoinductive growth factors. J Bone Joint Surg Am. 89:2485–2497. 2007.

11 

Anz AW, Hackel JG, Nilssen EC and Andrews JR: Application of biologics in the treatment of the rotator cuff, meniscus, cartilage, and osteoarthritis. J Am Acad Orthop Surg. 22:68–79. 2014.

12 

Jungebluth P, Alici E, Baiguera S, Blomberg P, Bozóky B, Crowley C, Einarsson O, Gudbjartsson T, Le Guyader S, Henriksson G, et al: Tracheobronchial transplantation with a stem-cell-seeded bioartificial nanocomposite: A proof-of-concept study. Lancet. 378:1997–2004. 2011.

13 

Nooshabadi VT, Mardpour S, Yousefi-Ahmadipour A, Allahverdi A, Izadpanah M, Daneshimehr F, Ai J, Banafshe HR and Ebrahimi-Barough S: The extracellular vesicles-derived from mesenchymal stromal cells: A new therapeutic option in regenerative medicine. J Cell Biochem. 119:8048–8073. 2018.

14 

Kourembanas S: Exosomes: Vehicles of intercellular signaling, biomarkers, and vectors of cell therapy. Annu Rev Physiol. 77:13–27. 2015.

15 

Abels ER and Breakefield XO: Introduction to extracellular vesicles: Biogenesis, RNA cargo selection, content, release, and uptake. Cell Mol Neurobiol. 36:301–312. 2016.

16 

Xiang XN, Zhu SY, He HC, Yu X, Xu Y and He CQ: Mesenchymal stromal cell-based therapy for cartilage regeneration in knee osteoarthritis. Stem Cell Res Ther. 13:142022.

17 

Lobb RJ, Becker M, Wen SW, Wong CS, Wiegmans AP, Leimgruber A and Möller A: Optimized exosome isolation protocol for cell culture supernatant and human plasma. J Extracell Vesicles. 4:270312015.

18 

Xu X, Liang Y, Li X, Ouyang K, Wang M, Cao T, Li W, Liu J, Xiong J, Li B, et al: Exosome-mediated delivery of kartogenin for chondrogenesis of synovial fluid-derived mesenchymal stem cells and cartilage regeneration. Biomaterials. 269:1205392021.

19 

McKiernan J, Donovan MJ, O'Neill V, Bentink S, Noerholm M, Belzer S, Skog J, Kattan MW, Partin A, Andriole G, et al: A novel urine exosome gene expression assay to predict high-grade prostate cancer at initial biopsy. JAMA Oncol. 2:882–889. 2016.

20 

Lässer C, Alikhani VS, Ekström K, Eldh M, Paredes PT, Bossios A, Sjöstrand M, Gabrielsson S, Lötvall J and Valadi H: Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J Transl Med. 9:92011.

21 

Jia L, Qiu Q, Zhang H, Chu L, Du Y, Zhang J, Zhou C, Liang F, Shi S, Wang S, et al: Concordance between the assessment of Aβ42, T-tau, and P-T181-tau in peripheral blood neuronal-derived exosomes and cerebrospinal fluid. Alzheimers Dement. 15:1071–1080. 2019.

22 

Latifkar A, Hur YH, Sanchez JC, Cerione RA and Antonyak MA: New insights into extracellular vesicle biogenesis and function. J Cell Sci. 132:jcs2224062019.

23 

Zakeri Z, Salmaninejad A, Hosseini N, Shahbakhsh Y, Fadaee E, Shahrzad MK and Fadaei S: MicroRNA and exosome: Key players in rheumatoid arthritis. J Cell Biochem. 120:10930–10944. 2019.

24 

van Niel G, D'Angelo G and Raposo G: Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 19:213–228. 2018.

25 

Raposo G and Stoorvogel W: Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013.

26 

Baglio SR, Rooijers K, Koppers-Lalic D, Verweij FJ, Pérez Lanzón M, Zini N, Naaijkens B, Perut F, Niessen HW, Baldini N and Pegtel DM: Human bone marrow- and adipose-mesenchymal stem cells secrete exosomes enriched in distinctive miRNA and tRNA species. Stem Cell Res Ther. 6:1272015.

27 

Johnstone RM, Adam M, Hammond JR, Orr L and Turbide C: Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem. 262:9412–9420. 1987.

28 

Mosquera-Heredia MI, Morales LC, Vidal OM, Barceló E, Silvera-Redondo C, Vélez JI and Garavito-Galofre P: Exosomes: Potential disease biomarkers and new therapeutic targets. Biomedicines. 9:10612021.

29 

Kalluri R and LeBleu VS: The biology, function, and biomedical applications of exosomes. Science. 367:eaau69772020.

30 

Théry C, Zitvogel L and Amigorena S: Exosomes: Composition, biogenesis and function. Nat Rev Immunol. 2:569–579. 2002.

31 

Qiu X, Liu J, Zheng C, Su Y, Bao L, Zhu B, Liu S, Wang L, Wang X, Wang Y, et al: Exosomes released from educated mesenchymal stem cells accelerate cutaneous wound healing via promoting angiogenesis. Cell Prolif. 53:e128302020.

32 

Keshtkar S, Azarpira N and Ghahremani MH: Mesenchymal stem cell-derived extracellular vesicles: Novel frontiers in regenerative medicine. Stem Cell Res Ther. 9:632018.

33 

Ren Y, Zhang S, Wang Y, Jacobson DS, Reisdorf RL, Kuroiwa T, Behfar A, Moran SL, Steinmann SP and Zhao C: Effects of purified exosome product on rotator cuff tendon-bone healing in vitro and in vivo. Biomaterials. 276:1210192021.

34 

Fang WH, Agrawal DK and Thankam FG: 'Smart exosomes': A smart approach for tendon regeneration. Tissue Eng Part B Rev. 28:613–625. 2022.

35 

Wang Z, Wu Y, Zhao Z, Liu C and Zhang L: Study on transorgan regulation of intervertebral disc and extra-skeletal organs through exosomes derived from bone marrow mesenchymal stem cells. Front Cell Dev Biol. 9:7411832021.

36 

Ma X, Becker Buscaglia LE, Barker JR and Li Y: MicroRNAs in NF-kappaB signaling. J Mol Cell Biol. 3:159–166. 2011.

37 

van den Boorn JG, Schlee M, Coch C and Hartmann G: SiRNA delivery with exosome nanoparticles. Nat Biotechnol. 29:325–326. 2011.

38 

EL Andaloussi S, Mäger I, Breakefield XO and Wood MJ: Extracellular vesicles: Biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 12:347–357. 2013.

39 

Vlassov AV, Magdaleno S, Setterquist R and Conrad R: Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta. 1820:940–948. 2012.

40 

Chen H, Li S, Xiao H, Wu B, Zhou L, Hu J and Li H: Effect of exercise intensity on the healing of the bone-tendon interface: A mouse rotator cuff injury model study. Am J Sports Med. 49:2064–2073. 2021.

41 

Chen W, Sun Y, Gu X, Cai J, Liu X, Zhang X, Chen J, Hao Y and Chen S: Conditioned medium of human bone marrow-derived stem cells promotes tendon-bone healing of the rotator cuff in a rat model. Biomaterials. 271:1207142021.

42 

Cheng P, Han P, Zhao C, Zhang S, Wu H, Ni J, Hou P, Zhang Y, Liu J, Xu H, et al: High-purity magnesium interference screws promote fibrocartilaginous entheses regeneration in the anterior cruciate ligament reconstruction rabbit model via accumulation of BMP-2 and VEGF. Biomaterials. 81:14–26. 2016.

43 

Hu J, Chen Y, Huang Y and Su Y: Human umbilical cord mesenchymal stem cell-derived exosomes suppress dermal fibroblasts-myofibroblats transition via inhibiting the TGF-β1/Smad 2/3 signaling pathway. Exp Mol Pathol. 115:1044682020.

44 

Lundgreen K, Lian OB, Engebretsen L and Scott A: Tenocyte apoptosis in the torn rotator cuff: A primary or secondary pathological event? Br J Sports Med. 45:1035–1039. 2011.

45 

Xu T, Xu M, Bai J, Lin J, Yu B, Liu Y, Guo X, Shen J, Sun H, Hao Y and Geng D: Tenocyte-derived exosomes induce the tenogenic differentiation of mesenchymal stem cells through TGF-β. Cytotechnology. 71:57–65. 2019.

46 

Zhang M, Liu H, Cui Q, Han P, Yang S, Shi M, Zhang T, Zhang Z and Li Z: Tendon stem cell-derived exosomes regulate inflammation and promote the high-quality healing of injured tendon. Stem Cell Res Ther. 11:4022020.

47 

Sun Y, Chen W, Hao Y, Gu X, Liu X, Cai J, Liu S, Chen J and Chen S: Stem cell-conditioned medium promotes graft remodeling of midsubstance and intratunnel incorporation after anterior cruciate ligament reconstruction in a rat model. Am J Sports Med. 47:2327–2337. 2019.

48 

Kuang GM, Yau WP, Lu WW and Chiu KY: Osteointegration of soft tissue grafts within the bone tunnels in anterior cruciate ligament reconstruction can be enhanced. Knee Surg Sports Traumatol Arthrosc. 18:1038–1051. 2010.

49 

Kovacevic D and Rodeo SA: Biological augmentation of rotator cuff tendon repair. Clin Orthop Relat Res. 466:622–633. 2008.

50 

Molloy T, Wang Y and Murrell G: The roles of growth factors in tendon and ligament healing. Sports Med. 33:381–394. 2003.

51 

Sharma P and Maffulli N: Tendon injury and tendinopathy: Healing and repair. J Bone Joint Surg Am. 87:187–202. 2005.

52 

Chamberlain CS, Leiferman EM, Frisch KE, Duenwald-Kuehl SE, Brickson SL, Murphy WL, Baer GS and Vanderby R: Interleukin-1 receptor antagonist modulates inflammation and scarring after ligament injury. Connect Tissue Res. 55:177–186. 2014.

53 

Xu Y and Murrell GAC: The basic science of tendinopathy. Clin Orthop Relat Res. 466:1528–1538. 2008.

54 

Sevivas N, Teixeira FG, Portugal R, Araújo L, Carriço LF, Ferreira N, Vieira da Silva M, Espregueira-Mendes J, Anjo S, Manadas B, et al: Mesenchymal stem cell secretome: A potential tool for the prevention of muscle degenerative changes associated with chronic rotator cuff tears. Am J Sports Med. 45:179–188. 2017.

55 

Xu Y, Zhang WX, Wang LN, Ming YQ, Li YL and Ni GX: Stem cell therapies in tendon-bone healing. World J Stem Cells. 13:753–775. 2021.

56 

Shengnan Q, Bennett S, Wen W, Aiguo L and Jiake X: The role of tendon derived stem/progenitor cells and extracellular matrix components in the bone tendon junction repair. Bone. 153:1161722021.

57 

Hao ZC, Wang SZ, Zhang XJ and Lu J: Stem cell therapy: A promising biological strategy for tendon-bone healing after anterior cruciate ligament reconstruction. Cell Prolif. 49:154–162. 2016.

58 

Lui PPY: Mesenchymal stem cell-derived extracellular vesicles for the promotion of tendon repair-an update of literature. Stem Cell Rev Rep. 17:379–389. 2021.

59 

Patel S, Caldwell JM, Doty SB, Levine WN, Rodeo S, Soslowsky LJ, Thomopoulos S and Lu HH: Integrating soft and hard tissues via interface tissue engineering. J Orthop Res. 36:1069–1077. 2018.

60 

Connor DE, Paulus JA, Dabestani PJ, Thankam FK, Dilisio MF, Gross RM and Agrawal DK: Therapeutic potential of exosomes in rotator cuff tendon healing. J Bone Miner Metab. 37:759–767. 2019.

61 

Diebold G, Lam P, Walton J and Murrell GAC: Relationship between age and rotator cuff retear: A study of 1,600 consecutive rotator cuff repairs. J Bone Joint Surg Am. 99:1198–1205. 2017.

62 

Wang J, Xu J, Wang X, Sheng L, Zheng L, Song B, Wu G, Zhang R, Yao H, Zheng N, et al: Magnesium-pretreated periosteum for promoting bone-tendon healing after anterior cruciate ligament reconstruction. Biomaterials. 268:1205762021.

63 

Lim WL, Liau LL, Ng MH, Chowdhury SR and Law JX: Current progress in tendon and ligament tissue engineering. Tissue Eng Regen Med. 16:549–571. 2019.

64 

Ménétrey J, Duthon VB, Laumonier T and Fritschy D: 'Biological failure' of the anterior cruciate ligament graft. Knee Surg Sports Traumatol Arthrosc. 16:224–231. 2008.

65 

Mihelic R, Pecina M, Jelic M, Zoricic S, Kusec V, Simic P, Bobinac D, Lah B, Legovic D and Vukicevic S: Bone morphogenetic protein-7 (osteogenic protein-1) promotes tendon graft integration in anterior cruciate ligament reconstruction in sheep. Am J Sports Med. 32:1619–1625. 2004.

66 

Murray MM, Spindler KP, Ballard P, Welch TP, Zurakowski D and Nanney LB: Enhanced histologic repair in a central wound in the anterior cruciate ligament with a collagen-platelet-rich plasma scaffold. J Orthop Res. 25:1007–1017. 2007.

67 

Cervellin M, de Girolamo L, Bait C, Denti M and Volpi P: Autologous platelet-rich plasma gel to reduce donor-site morbidity after patellar tendon graft harvesting for anterior cruciate ligament reconstruction: A randomized, controlled clinical study. Knee Surg Sports Traumatol Arthrosc. 20:114–120. 2012.

68 

Lu H, Liu F, Chen C, Wang Z, Chen H, Qu J, Zhang T, Xu D and Hu J: Low-intensity pulsed ultrasound stimulation for tendon-bone healing: A dose-dependent study. Am J Phys Med Rehabil. 97:270–277. 2018.

69 

Ersen A, Demirhan M, Atalar AC, Kapicioğlu M and Baysal G: Platelet-rich plasma for enhancing surgical rotator cuff repair: Evaluation and comparison of two application methods in a rat model. Arch Orthop Trauma Surg. 134:405–411. 2014.

70 

Zhang M, Zhen J, Zhang X, Yang Z, Zhang L, Hao D and Ren B: Effect of autologous platelet-rich plasma and gelatin sponge for tendon-to-bone healing after rabbit anterior cruciate ligament reconstruction. Arthroscopy. 35:1486–1497. 2019.

71 

Zhang J, Li F, Augi T, Williamson KM, Onishi K, Hogan MV, Neal MD and Wang JH: Platelet HMGB1 in platelet-rich plasma (PRP) promotes tendon wound healing. PLoS One. 16:e02511662021.

72 

Chen X, Jones IA, Park C and Vangsness CT Jr: The efficacy of platelet-rich plasma on tendon and ligament healing: A systematic review and meta-analysis with bias assessment. Am J Sports Med. 46:2020–2032. 2018.

73 

Kim CH, Park YB, Lee JS and Jung HS: Platelet-rich plasma injection vs operative treatment for lateral elbow tendinosis: A systematic review and meta-analysis. J Shoulder Elbow Surg. 31:428–436. 2022.

74 

Chen X, Jones IA, Togashi R, Park C and Vangsness CT Jr: Use of platelet-rich plasma for the improvement of pain and function in rotator cuff tears: A systematic review and meta-analysis with bias assessment. Am J Sports Med. 48:2028–2041. 2020.

75 

Keene DJ, Alsousou J, Harrison P, Hulley P, Wagland S, Parsons SR, Thompson JY, O'Connor HM, Schlüssel MM, Dutton SJ, et al: Platelet rich plasma injection for acute Achilles tendon rupture: PATH-2 randomised, placebo controlled, superiority trial. BMJ. 367:l61322019.

76 

Bennell KL, Paterson KL, Metcalf BR, Duong V, Eyles J, Kasza J, Wang Y, Cicuttini F, Buchbinder R, Forbes A, et al: Effect of intra-articular platelet-rich plasma vs placebo injection on pain and medial tibial cartilage volume in patients with knee osteoarthritis: The RESTORE randomized clinical trial. JAMA. 326:2021–2030. 2021.

77 

Lui PPY, Wong OT and Lee YW: Application of tendon-derived stem cell sheet for the promotion of graft healing in anterior cruciate ligament reconstruction. Am J Sports Med. 42:681–689. 2014.

78 

Akbari A, Jabbari N, Sharifi R, Ahmadi M, Vahhabi A, Seyedzadeh SJ, Nawaz M, Szafert S, Mahmoodi M, Jabbari E, et al: Free and hydrogel encapsulated exosome-based therapies in regenerative medicine. Life Sci. 249:1174472020.

79 

Lu V, Tennyson M, Zhang J and Khan W: Mesenchymal stem cell-derived extracellular vesicles in tendon and ligament repair-A systematic review of in vivo studies. Cells. 10:25532021.

80 

Liu Q, Yu Y, Reisdorf RL, Qi J, Lu CK, Berglund LJ, Amadio PC, Moran SL, Steinmann SP, An KN, et al: Engineered tendon-fibrocartilage-bone composite and bone marrow-derived mesenchymal stem cell sheet augmentation promotes rotator cuff healing in a non-weight-bearing canine model. Biomaterials. 192:189–198. 2019.

81 

Rothrauff BB, Smith CA, Ferrer GA, Novaretti JV, Pauyo T, Chao T, Hirsch D, Beaudry MF, Herbst E, Tuan RS, et al: The effect of adipose-derived stem cells on enthesis healing after repair of acute and chronic massive rotator cuff tears in rats. J Shoulder Elbow Surg. 28:654–664. 2019.

82 

Utsunomiya H, Sekiya I and Uchida S: Editorial commentary: Are we ready to apply stem cell therapy in rotator cuff tear surgery? Arthroscopy. 36:86–87. 2020.

83 

Sun H, Pratt RE, Hodgkinson CP and Dzau VJ: Sequential paracrine mechanisms are necessary for the therapeutic benefits of stem cell therapy. Am J Physiol Cell Physiol. 319:C1141–C1150. 2020.

84 

Pawitan JA: Prospect of stem cell conditioned medium in regenerative medicine. Biomed Res Int. 2014:9658492014.

85 

Driscoll J and Patel T: The mesenchymal stem cell secretome as an acellular regenerative therapy for liver disease. J Gastroenterol. 54:763–773. 2019.

86 

Riau AK, Ong HS, Yam GHF and Mehta JS: Sustained delivery system for stem cell-derived exosomes. Front Pharmacol. 10:13682019.

87 

Malekpour K, Hazrati A, Zahar M, Markov A, Zekiy AO, Navashenaq JG, Roshangar L and Ahmadi M: The potential use of mesenchymal stem cells and their derived exosomes for orthopedic diseases treatment. Stem Cell Rev Rep. 18:933–951. 2022.

88 

Gordon S, Plüddemann A and Martinez Estrada F: Macrophage heterogeneity in tissues: Phenotypic diversity and functions. Immunol Rev. 262:36–55. 2014.

89 

McWhorter FY, Wang T, Nguyen P, Chung T and Liu WF: Modulation of macrophage phenotype by cell shape. Proc Natl Acad Sci USA. 110:17253–17258. 2013.

90 

Kawamura S, Ying L, Kim HJ, Dynybil C and Rodeo SA: Macrophages accumulate in the early phase of tendon-bone healing. J Orthop Res. 23:1425–1432. 2005.

91 

Geng R, Lin Y, Ji M, Chang Q, Li Z, Xu L, Zhang W and Lu J: MFG-E8 promotes tendon-bone healing by regualting macrophage efferocytosis and M2 polarization after anterior cruciate ligament reconstruction. J Orthop Translat. 34:11–21. 2022.

92 

Chen Z, Jin M, He H, Dong J, Li J, Nie J, Wang Z, Xu J and Wu F: Mesenchymal stem cells and macrophages and their interactions in tendon-bone healing. J Orthop Translat. 39:63–73. 2023.

93 

Klinkert K, Whelan D, Clover AJP, Leblond AL, Kumar AHS and Caplice NM: Selective M2 macrophage depletion leads to prolonged inflammation in surgical wounds. Eur Surg Res. 58:109–120. 2017.

94 

Sindrilaru A, Peters T, Wieschalka S, Baican C, Baican A, Peter H, Hainzl A, Schatz S, Qi Y, Schlecht A, et al: An unrestrained proinflammatory M1 macrophage population induced by iron impairs wound healing in humans and mice. J Clin Invest. 121:985–997. 2011.

95 

Mirza R, DiPietro LA and Koh TJ: Selective and specific macrophage ablation is detrimental to wound healing in mice. Am J Pathol. 175:2454–2462. 2009.

96 

Janssen RP and Scheffler SU: Intra-articular remodelling of hamstring tendon grafts after anterior cruciate ligament reconstruction. Knee Surg Sports Traumatol Arthrosc. 22:2102–2108. 2014.

97 

Li S, Xu Z, Wang Z, Xiang J, Zhang T and Lu H: Acceleration of bone-tendon interface healing by low-intensity pulsed ultrasound is mediated by macrophages. Phys Ther. 101:pzab0552021.

98 

Chamberlain CS, Kink JA, Wildenauer LA, McCaughey M, Henry K, Spiker AM, Halanski MA, Hematti P and Vanderby R: Exosome-educated macrophages and exosomes differentially improve ligament healing. Stem Cells. 39:55–61. 2021.

99 

Wang C, Zhang Y, Zhang G, Yu W and He Y: Adipose stem cell-derived exosomes ameliorate chronic rotator cuff tendinopathy by regulating macrophage polarization: From a mouse model to a study in human tissue. Am J Sports Med. 49:2321–2331. 2021.

100 

Huang Y, He B, Wang L, Yuan B, Shu H, Zhang F and Sun L: Bone marrow mesenchymal stem cell-derived exosomes promote rotator cuff tendon-bone healing by promoting angiogenesis and regulating M1 macrophages in rats. Stem Cell Res Ther. 11:4962020.

101 

Shi Y, Kang X, Wang Y, Bian X, He G, Zhou M and Tang K: Exosomes derived from bone marrow stromal cells (BMSCs) Enhance tendon-bone healing by regulating macrophage polarization. Med Sci Monit. 26:e9233282020.

102 

Fatima F, Ekstrom K, Nazarenko I, Maugeri M, Valadi H, Hill AF, Camussi G and Nawaz M: Non-coding RNAs in mesenchymal stem cell-derived extracellular vesicles: Deciphering regulatory roles in stem cell potency, inflammatory resolve, and tissue regeneration. Front Genet. 8:1612017.

103 

Yoshikawa T, Tohyama H, Katsura T, Kondo E, Kotani Y, Matsumoto H, Toyama Y and Yasuda K: Effects of local administration of vascular endothelial growth factor on mechanical characteristics of the semitendinosus tendon graft after anterior cruciate ligament reconstruction in sheep. Am J Sports Med. 34:1918–1925. 2006.

104 

Fealy S, Adler RS, Drakos MC, Kelly AM, Allen AA, Cordasco FA, Warren RF and O'Brien SJ: Patterns of vascular and anatomical response after rotator cuff repair. Am J Sports Med. 34:120–127. 2006.

105 

Takayama K, Kawakami Y, Mifune Y, Matsumoto T, Tang Y, Cummins JH, Greco N, Kuroda R, Kurosaka M, Wang B, et al: The effect of blocking angiogenesis on anterior cruciate ligament healing following stem cell transplantation. Biomaterials. 60:9–19. 2015.

106 

Sivaraj KK and Adams RH: Blood vessel formation and function in bone. Development. 143:2706–2715. 2016.

107 

Zhang T, Yan S, Song Y, Chen C, Xu D, Lu B and Xu Y: Exosomes secreted by hypoxia-stimulated bone-marrow mesenchymal stem cells promote grafted tendon-bone tunnel healing in rat anterior cruciate ligament reconstruction model. J Orthop Translat. 36:152–163. 2022.

108 

Fang S, He T, Jiang J, Li Y and Chen P: Osteogenic effect of tsRNA-10277-loaded exosome derived from bone mesenchymal stem cells on steroid-induced osteonecrosis of the femoral head. Drug Des Devel Ther. 14:4579–4591. 2020.

109 

Zhang Y, Cao X, Li P, Fan Y, Zhang L, Ma X, Sun R, Liu Y and Li W: microRNA-935-modified bone marrow mesenchymal stem cells-derived exosomes enhance osteoblast proliferation and differentiation in osteoporotic rats. Life Sci. 272:1192042021.

110 

Xie Y, Chen Y, Zhang L, Ge W and Tang P: The roles of bone-derived exosomes and exosomal microRNAs in regulating bone remodelling. J Cell Mol Med. 21:1033–1041. 2017.

111 

Feng W, Jin Q, Ming-Yu Y, Yang H, Xu T, You-Xing S, Xu-Ting B, Wan C, Yun-Jiao W, Huan W, et al: MiR-6924-5p-rich exosomes derived from genetically modified Scleraxis-overexpressing PDGFRα(+) BMMSCs as novel nanotherapeutics for treating osteolysis during tendon-bone healing and improving healing strength. Biomaterials. 279:1212422021.

112 

Han L, Liu H, Fu H, Hu Y, Fang W and Liu J: Exosome-delivered BMP-2 and polyaspartic acid promotes tendon bone healing in rotator cuff tear via Smad/RUNX2 signaling pathway. Bioengineered. 13:1459–1475. 2022.

113 

Wu B, Chen H, Shi X, Wang L, Zhang T, Guan C, Huang T, Yang Y, Hu J and Lu H: Exosomes derived from bone marrow mesenchymal stem cell preconditioned by low-intensity pulsed ultrasound stimulation promote bone-tendon interface fibrocartilage regeneration and ameliorate rotator cuff fatty infiltration. Res Sq. 2021.

114 

Cai J, Xu J, Ye Z, Wang L, Zheng T, Zhang T, Li Y, Jiang J and Zhao J: Exosomes derived from kartogenin-preconditioned mesenchymal stem cells promote cartilage formation and collagen maturation for enthesis regeneration in a rat model of chronic rotator cuff tear. Am J Sports Med. 51:1267–1276. 2023.

115 

Berger DR, Centeno CJ and Steinmetz NJ: Platelet lysates from aged donors promote human tenocyte proliferation and migration in a concentration-dependent manner. Bone Joint Res. 8:32–40. 2019.

116 

Yu H, Cheng J, Shi W, Ren B, Zhao F, Shi Y, Yang P, Duan X, Zhang J, Fu X, et al: Bone marrow mesenchymal stem cell-derived exosomes promote tendon regeneration by facilitating the proliferation and migration of endogenous tendon stem/progenitor cells. Acta Biomater. 106:328–341. 2020.

117 

Li J, Liu ZP, Xu C and Guo A: TGF-β1-containing exosomes derived from bone marrow mesenchymal stem cells promote proliferation, migration and fibrotic activity in rotator cuff tenocytes. Regen Ther. 15:70–76. 2020.

118 

Li J, Wang ZH and Sun YH: TGF-β1 stimulated mesenchymal stem cells-generated exosomal miR-29a promotes the proliferation, migration and fibrogenesis of tenocytes by targeting FABP3. Cytokine. 162:1560902023.

119 

Xiong QH, Zhao L, Wan GQ, Hu YG and Li XL: Engineered BMSCs-derived exosomal miR-542-3p promotes cutaneous wound healing. Endocr Metab Immune Disord Drug Targets. 23:336–346. 2023.

120 

Wu D, Kang L, Tian J, Wu Y, Liu J, Li Z, Wu X, Huang Y, Gao B, Wang H, et al: Exosomes derived from bone mesenchymal stem cells with the stimulation of Fe3O4 nanoparticles and static magnetic field enhance wound healing through upregulated miR-21-5p. Int J Nanomedicine. 15:7979–7993. 2020.

121 

Pomatto M, Gai C, Negro F, Cedrino M, Grange C, Ceccotti E, Togliatto G, Collino F, Tapparo M, Figliolini F, et al: Differential therapeutic effect of extracellular vesicles derived by bone marrow and adipose mesenchymal stem cells on wound healing of diabetic ulcers and correlation to their cargoes. Int J Mol Sci. 22:38512021.

122 

Li FQ, Chen WB, Luo ZW, Chen YS, Sun YY, Su XP, Sun JM and Chen SY: Bone marrow mesenchymal stem cell-derived exosomal microRNAs target PI3K/Akt signaling pathway to promote the activation of fibroblasts. World J Stem Cells. 15:248–267. 2023.

123 

Gatti S, Bruno S, Deregibus MC, Sordi A, Cantaluppi V, Tetta C and Camussi G: Microvesicles derived from human adult mesenchymal stem cells protect against ischaemia-reperfusion-induced acute and chronic kidney injury. Nephrol Dial Transplant. 26:1474–1483. 2011.

124 

Kordelas L, Rebmann V, Ludwig AK, Radtke S, Ruesing J, Doeppner TR, Epple M, Horn PA, Beelen DW and Giebel B: MSC-derived exosomes: A novel tool to treat therapy-refractory graft-versus-host disease. Leukemia. 28:970–973. 2014.

125 

Di Rocco G, Baldari S and Toietta G: Towards therapeutic delivery of extracellular vesicles: Strategies for in vivo tracking and biodistribution analysis. Stem Cells Int. 2016:50296192016.

126 

Sevivas N, Teixeira FG, Portugal R, Direito-Santos B, Espregueira-Mendes J, Oliveira FJ, Silva RF, Sousa N, Sow WT, Nguyen LTH, et al: Mesenchymal stem cell secretome improves tendon cell viability in vitro and tendon-bone healing in vivo when a tissue engineering strategy is used in a rat model of chronic massive rotator cuff tear. Am J Sports Med. 46:449–459. 2018.

127 

Gaspar D, Spanoudes K, Holladay C, Pandit A and Zeugolis D: Progress in cell-based therapies for tendon repair. Adv Drug Deliv Rev. 84:240–256. 2015.

128 

Gao H, Zhang L, Wang Z, Yan K, Zhao L and Xiao W: Research progress on transorgan regulation of the cardiovascular and motor system through cardiogenic exosomes. Int J Mol Sci. 23:57652022.

Related Articles

Journal Cover

December-2023
Volume 52 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhu Y, Yan J, Zhang H and Cui G: Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review). Int J Mol Med 52: 121, 2023
APA
Zhu, Y., Yan, J., Zhang, H., & Cui, G. (2023). Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review). International Journal of Molecular Medicine, 52, 121. https://doi.org/10.3892/ijmm.2023.5324
MLA
Zhu, Y., Yan, J., Zhang, H., Cui, G."Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review)". International Journal of Molecular Medicine 52.6 (2023): 121.
Chicago
Zhu, Y., Yan, J., Zhang, H., Cui, G."Bone marrow mesenchymal stem cell‑derived exosomes: A novel therapeutic agent for tendon‑bone healing (Review)". International Journal of Molecular Medicine 52, no. 6 (2023): 121. https://doi.org/10.3892/ijmm.2023.5324