Skip to main content
Stem Cell Research & Therapy logoLink to Stem Cell Research & Therapy
. 2025 Oct 31;16:600. doi: 10.1186/s13287-025-04699-3

Functionalized mesenchymal stem cells for enhanced bone regeneration: advances and challenges

Wentao Huang 1,2,#, Chao Zhou 3,#, Yijun Yu 4,#, Shu Qin 2, Lin Chen 2, Hongming Lin 2, Songou Zhang 5, Linying Xia 4,, Wenqing Liang 1,2,
PMCID: PMC12577075  PMID: 41174641

Abstract

Bone fracture continues to pose a significant clinical challenge in regenerative medicine due to limited repair capacity and inadequate therapeutic options. Among the various therapeutic strategies, mesenchymal stem cells have shown strong potential due to their ability to promote bone formation, modulate inflammation, and migrate to injury sites. However, clinical outcomes have been limited by issues like low survival rates, poor integration, and non-specific distribution after transplantation. Functionalized mesenchymal cells enhanced through genetic, chemical, or material-based modifications have emerged as an advanced strategy to overcome these limitations and significantly improve bone regeneration. This review explores recent developments in the functionalization of stem cells to increase their bone-forming potential. It covers techniques of gene modification, preconditioning, nanoparticle integration, and scaffold-based delivery. The role of these engineered cells is in activating key pathways involved in bone repair, including bone morphogenetic proteins and Wnt signaling. Furthermore, the study highlights current delivery platforms, including injectable gels, printed scaffolds, and bioactive coatings that support targeted, sustained cell activity. Despite encouraging preclinical outcomes, unresolved challenges in manufacturing, immune compatibility, and regulatory pathways persist, prompting the exploration of emerging solutions like precision-engineered implants and artificial intelligence-driven design to guide the future of advanced bone regeneration therapies.

Keywords: Mesenchymal stem cells, Osteogenesis, Bone fracture healing

Introduction

Bone fractures are among the most common injuries encountered in clinical practice, yet their treatment becomes particularly challenging in cases involving critical-sized defects, delayed union, non-union, or patients with systemic conditions such as osteoporosis, diabetes, or cancer [1]. Conventional therapeutic strategies including autologous and allogeneic bone grafts, present significant limitations like donor site morbidity, limited graft availability, immune rejection, and variable rates of graft integration [2, 3]. Moreover, synthetic biomaterials, while useful as temporary scaffolds, often lack osteoinductive potential and fail to fully recapitulate the complex cellular signaling required for robust bone regeneration [4]. In recent years, regenerative medicine has shifted its focus toward biologically active approaches that support and stimulate the body’s intrinsic healing mechanisms [5]. Among these, stem cell–based therapies have emerged as a transformative strategy to promote bone repair. Mesenchymal stem cells (MSCs), in particular, have gained widespread attention due to their multipotent nature, ease of isolation from various tissues (bone marrow, adipose tissue, umbilical cord), and their ability to differentiate into osteoblasts, chondrocytes, and adipocytes [6]. More importantly, MSCs exert strong paracrine effects by secreting cytokines, growth factors, and exosomes that promote angiogenesis, immunomodulation, and extracellular matrix remodeling key components in the bone regeneration cascade [7].

The biological rationale for using MSCs in bone healing is rooted in their capacity to participate in both direct and indirect mechanisms of osteogenesis [8]. Directly, MSCs contribute by differentiating into bone-forming osteoblasts under appropriate biochemical and mechanical stimuli. Indirectly, they secrete trophic factors such as bone morphogenetic proteins (BMPs), vascular endothelial growth factor (VEGF), transforming growth factor-beta (TGF-β), and insulin-like growth factors (IGFs), which modulate local cell behavior and promote tissue repair [9]. Furthermore, MSCs display immunosuppressive properties, which make them particularly valuable in allogeneic applications, reducing the risk of host immune rejection [10]. Despite their promise, the clinical translation of MSC-based therapies faces significant challenges. Unmodified or “naïve” MSCs often exhibit poor survival and low engraftment rates following transplantation, especially in ischemic or inflammatory environments such as those present in non-union fractures [11]. Additionally, their osteogenic potential may be inconsistent due to donor variability, passage number, and culture conditions. These limitations have prompted the exploration of MSC modification a broad term encompassing various strategies aimed at enhancing the therapeutic efficacy of MSCs [12]. The field is also evolving to integrate these modified MSCs within bioactive delivery platforms such as injectable hydrogels, electrospun scaffolds, and 3D-printed constructs that provide mechanical support while promoting cell viability and spatial control [13, 14]. Moreover, advances in nanotechnology and biofabrication have enabled the development of “smart” scaffolds that can respond to environmental cues (pH, temperature, enzymatic activity) to release bioactive molecules in a controlled fashion, further improving the regenerative microenvironment [15]. Understanding how preconditioned MSCs interact with their microenvironment, deliver osteogenic cues, and integrate with host tissues is essential for developing next-generation bone regeneration therapies [16].

This review aims to provide a comprehensive synthesis of recent developments in the optimization of mesenchymal stem cells for bone fracture healing. It explores the biological underpinnings of MSC-mediated osteogenesis, analyzes emerging strategies for enhancing their regenerative performance, and evaluates the efficacy of various delivery platforms. In doing so, the review highlights major challenges including immune safety, large-scale production, and regulatory considerations while also discussing future directions such as the integration of artificial intelligence, precision-engineered implants, and patient-specific therapies that promise to redefine the clinical landscape of bone regeneration.

Biological and therapeutic profile of mesenchymal stem cells

MSCs are multipotent stromal cells derived from various adult and perinatal tissues, including bone marrow, adipose tissue, and umbilical cord [17] (Fig. 1). While bone marrow-derived MSCs (BM-MSCs) remain the most studied due to their high osteogenic potential, adipose-derived MSCs (AD-MSCs) and umbilical cord-derived MSCs (UC-MSCs) offer significant clinical advantages. AD-MSCs are easier to harvest via minimally invasive procedures (e.g., liposuction), yield substantially higher cell numbers (over 5000 cells per gram of adipose tissue vs. 100–1000 MSCs per mL of bone marrow), and exhibit greater proliferative capacity in vitro. UC-MSCs can be collected painlessly from discarded umbilical cords, are ethically unencumbered, and demonstrate the highest proliferation rates and clonogenic potential among MSC sources, with reduced expression of senescence markers (p53, p21, p16). These unique attributes make AD-MSCs and UC-MSCs highly attractive for scalable regenerative therapies, particularly when large cell doses are required [1821].

Fig. 1.

Fig. 1

MSCs can be isolated from different sources, neonatal, fetal, and adult tissues. These cells possess the potential, under specific conditions, to differentiate into a wide range of cell types, including osteoblasts, chondrocytes, and adipocytes, making them a promising candidate for regenerative therapies across multiple tissue types.

Adapted from the reference [37] under the terms and conditions of a Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Recent studies recognize dental pulp stem cells (DPSCs) as a promising MSC source for bone regeneration due to their multipotency, abundant availability, and minimally invasive harvestability. DPSCs exhibit robust osteogenic differentiation in vitro, expressing markers such as ALP, type I collagen, BMP-2/4, osteonectin, osteopontin, and osteocalcin and can form mineralized nodules under osteoinductive culture conditions [22]. In vivo meta-analyses have demonstrated that DPSC-based interventions significantly enhance new bone area and volume compared to controls [23]. A 2024 review further emphasizes DPSCs’ clinical appeal, citing their easy accessibility, low immunogenicity, and potential for both cell-based and cell-free bone regenerative strategies [24]. Moreover, scaffold-based in vivo studies reveal that combining DPSCs with biomaterials can achieve nearly complete structural restoration of critical bone defects, resulting in well-vascularized lamellar bone formation, increased TGF-β1 levels, and a conducive bone microenvironment [25]. MSCs are typically identified by the expression of surface markers including CD73, CD90, and CD105, along with the absence of hematopoietic markers like CD34, CD45, and HLA-DR [26, 27]. However, variations in these markers have been observed depending on the tissue source and culture conditions, indicating phenotypic heterogeneity that may influence therapeutic performance [28, 29]. MSCs possess robust differentiation capacity, particularly into osteogenic lineages under osteoinductive conditions, making them highly attractive for bone tissue engineering [30, 31]. This osteogenic differentiation is tightly regulated by transcription factors such as RUNX2 and signaling pathways including Wnt/β-catenin and BMP [3234]. In addition to their regenerative capacity, MSCs play a pivotal role in immunomodulation, exerting anti-inflammatory effects by inhibiting T-cell proliferation, inducing regulatory T-cells, and secreting immunosuppressive molecules like prostaglandin E2, TGF-β, and interleukin-10 [35, 36]. These properties enhance their therapeutic utility in bone healing, particularly in inflammatory or immune-compromised conditions.

MSCs exhibit notable variability in proliferative potential depending on their tissue of origin, which significantly affects their suitability for clinical-scale applications. For example, umbilical cord-derived MSCs (UC-MSCs) display markedly higher population doubling limits and faster expansion rates compared to BM-MSCs and AD-MSCs [38]. Conversely, in a comparative study of canine MSCs, adipose-derived MSCs (AD-MSCs) demonstrated the shortest population doubling time, indicating superior in vitro growth rates [39]. Donor characteristics such as age profoundly influence MSC proliferation. Placenta-derived MSCs from donors aged 22–35 showed enhanced growth rates and colony-forming efficiency relative to cells from younger or older donors [40]. Moreover, aging reduces MSC proliferative potential across tissues; younger donors retain superior self-renewal capacity [20]. Culture conditions and source-dependent heterogeneity also modulate proliferative behavior. Factors such as culture density, oxygen tension, and cell isolation methods significantly affect replicative potential and senescence onset [41].

Clinically, MSCs have been investigated for various orthopaedic applications, including non-union fractures, critical-sized bone defects, osteonecrosis of the femoral head, spinal fusion, and bone cysts (Fig. 2) [42, 43]. Early-phase clinical trials have reported encouraging results, with MSC-based therapies promoting bone regeneration, reducing pain, and minimizing the need for autologous grafts [44, 45]. However, despite these promising outcomes, several challenges persist, particularly regarding donor-to-donor variability, large-scale manufacturing, immune compatibility, and regulatory approval [46]. Moreover, the translation of MSC therapies from bench to bedside is limited by the lack of standardized protocols and variability in delivery methods [47]. Nonetheless, ongoing developments in biomaterial engineering, scaffold design, and cellular functionalization continue to improve the osteogenic performance and clinical applicability of MSCs in bone tissue repair [48].

Fig. 2.

Fig. 2

Schematic representation of the functions of mesenchymal stem cells (MSCs) in biological systems.

Adapted from the reference [49] under the terms and conditions of Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Osteogenesis and bone healing cascade

Following a fracture, the repair cascade begins with an inflammatory phase characterized by hematoma formation and infiltration of immune cells, which release cytokines and chemokines that recruit MSCs to the injury site [50]. Immune cells release a range of cytokines and chemokines, including interleukins (IL-1, IL-6, IL-11, IL-18), tumor necrosis factor-alpha (TNF-α), monocyte chemoattractant protein-1 (MCP-1), and platelet-derived growth factor (PDGF), among others, to initiate the inflammatory phase of fracture healing (Fig. 3). The subsequent repair phase involves proliferation and osteogenic differentiation of MSCs, as well as angiogenesis and extracellular matrix deposition. MSCs play a central role by differentiating into osteoblasts, which are responsible for new bone formation [51]. This differentiation is governed by complex signaling networks, including the activation of RUNX2, a master transcription factor required for commitment to the osteogenic lineage [52, 53]. Other critical pathways include Wnt/β-catenin, which promotes osteoblast maturation and mineralization; bone morphogenetic protein-2 (BMP-2), which enhances early osteogenic induction; and the Notch signaling pathway, which contributes to osteogenic regulation through interaction with the bone microenvironment [54, 55]. In addition to their osteogenic role, MSCs support neovascularization by secreting pro-angiogenic factors such as VEGF, thereby ensuring adequate nutrient and oxygen supply essential for bone tissue regeneration [56]. They also contribute to matrix remodelling by producing enzymes like MMP-2 and MMP-9, facilitating the transition from a soft callus to a mineralized bone structure [57]. Through their combined effects on bone formation, angiogenesis, and remodelling, MSCs function as orchestrators of the bone healing cascade, making them key targets for enhancing fracture repair and addressing non-union defects [58].

Fig. 3.

Fig. 3

Schematic representation of the sequential phases of fracture healing—namely inflammation, repair, and remodeling—highlighting the dynamic involvement of mesenchymal stem cells (MSCs) and key growth factors. The healing cascade includes hematoma formation, immune cell recruitment, soft and hard callus formation, and eventual bone remodeling. PDGF facilitates MSC recruitment and proliferation; VEGF drives angiogenesis; BMP-2 promotes osteoblast and chondrocyte differentiation; FGF-2 supports matrix synthesis and mitogenic activity; IGF and TGF-β regulate osteoblast and osteoclast function during bone regeneration.

Adapted from the reference [63] under the terms and conditions of a Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Following the early phases of inflammation and osteogenesis, the consolidation phase (days 21–42) transitions the soft callus into a mechanically stable, mineralized hard callus, driven by robust angiogenic signaling notably VEGF-A and angiopoietin-2 which promote vascular invasion and matrix mineralization. Thereafter, during the remodeling phase (from ~ day 42 to and beyond day 64), woven bone is gradually replaced by organized lamellar bone through tightly regulated resorption and formation dynamics mediated by the RANKL–OPG axis; specifically, VEGF enhances osteoblast proliferation and upregulates RANKL while downregulating OPG, thereby indirectly promoting osteoclastogenesis and coordinated bone turnover. Meanwhile, paracrine/autocrine VEGF signaling via VEGFR1/VEGFR2 influences osteoblast and endothelial cross-talk, further supporting remodeling and angiogenesis of the callus [5962].

Strategies for functionalizing MSCs to enhance osteogenesis

To enhance the therapeutic efficiency of MSCs in bone regeneration, a range of functionalization strategies have been designed to modulate their osteogenic potential through genetic, epigenetic, biophysical, and nanoengineering approaches [64].

Genetic engineering has emerged as a cornerstone strategy, wherein MSCs are transfected with osteoinductive genes such as BMP-2, RUNX2, and VEGF using viral or non-viral vectors (Fig. 4) [65]. These genetic interventions not only amplify the cells’ intrinsic differentiation capacity but also enhance local angiogenesis, which is critical for mineralized tissue formation. Viral delivery methods, such as lentiviral and adenoviral vectors, have demonstrated high transfection efficiency; however, they also raise concerns regarding insertional mutagenesis and long-term safety, which remains a key regulatory challenge in clinical translation [6668].

Fig. 4.

Fig. 4

Through transfection-based approaches, MSCs-derived small extracellular vesicles (sEVs) serve as carriers for miRNAs. By introducing miRNA mimics or inhibitors into stem cells, the expression levels of specific miRNAs within MSCs-sEVs can be upregulated or suppressed. These miRNA-enriched sEVs subsequently promote angiogenesis and facilitate bone regeneration.

Adapted from the reference [73] under the terms and conditions of a Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Epigenetic modulation offers a non-integrative strategy for altering MSC behavior. MicroRNAs such as miR-26a, miR-29b, and miR-21 have been identified as potent regulators of osteogenesis through their ability to suppress inhibitory signaling pathways (GSK-3β, Smad7), thereby promoting the expression of osteogenic genes [69, 70]. Synthetic miRNA mimics or inhibitors, delivered via lipid nanoparticles or exosomes, have shown efficacy in preclinical fracture models, enhancing matrix mineralization and osteoblast marker expression [71, 72]. Recent studies underscore the therapeutic potential of mesenchymal stem cell-derived small extracellular vesicles (MSCs-sEVs) as delivery vehicles for microRNAs (miRNAs) in bone regeneration [73, 74]. By modulating gene expression, these miRNAs influence key cellular processes such as osteoblast proliferation, migration, and differentiation. For instance, sEVs enriched with miR-21-5p, miR-126-5p, miR-29a, and miR-27a-5p have demonstrated efficacy in enhancing osteogenesis, angiogenesis, and matrix mineralization through the regulation of targets like KLF3, VASH1, Atg4B, and IGFBP3 [73]. Several miRNAs within MSCs-sEVs have been shown to activate osteogenic signaling pathways, including Wnt/β-catenin, PI3K/AKT, NF-κB, and RhoA/ROCK, while others inhibit osteogenesis by downregulating osteogenic markers (RUNX2, ALP, OCN) via pathways such as SMAD2/ERK and BMPR2/Smad1/5/9 [73]. Notably, long non-coding RNAs (lncRNAs) within sEVs further refine osteogenic responses by acting as competitive endogenous RNAs, sequestering miRNAs and thus promoting the expression of osteoinductive genes [75, 76].

Beyond osteogenesis, MSCs-sEVs play pivotal roles in suppressing osteoclast differentiation and promoting angiogenesis—critical for bone remodeling. miRNAs such as miR-21, miR-27a, and miR-6924-5p inhibit bone resorption, while others (miR-21/NOTCH1/DLL4 axis and miR-126/PI3K/AKT pathway) facilitate vascular development [73]. Additionally, sEVs contribute to immunomodulation by inducing M2 macrophage polarization and attenuating inflammation, thus creating a favorable microenvironment for bone repair [77]. These findings highlight the multifaceted role of MSCs-sEVs in orchestrating osteogenesis, angiogenesis, and immunomodulation via the targeted delivery of miRNAs and lncRNAs—underscoring their promise in regenerative medicine and bone tissue engineering [73].

Biophysical and biochemical priming strategies further enhance osteogenic readiness by exposing MSCs to tailored microenvironments [78]. Mechanical stimulation through cyclic strain or fluid shear stress has been shown to upregulate mechanosensitive pathways such as YAP/TAZ and integrin-FAK signaling, reinforcing osteogenic lineage commitment [79, 80]. Hypoxic preconditioning (1–5% O₂) mimics in vivo niche conditions and has been associated with enhanced secretion of angiogenic factors and improved cell survival [81, 82]. Biochemical cues, such as dexamethasone, β-glycerophosphate, and ascorbic acid, remain standard in vitro additives that direct osteogenic priming [83].

Nanoparticle-mediated functionalization introduces a versatile toolkit for MSC engineering, enabling both targeted delivery and real-time imaging [84]. Gold and silica nanoparticles modified with osteoinductive molecules (BMP-2, RGD peptides) allow for controlled release and localized activity at the defect site [85, 86]. Magnetic nanoparticles, particularly those coated with poly-L-lysine or gelatin, offer magnetically guided delivery, improving retention and spatial control (Fig. 5) [87, 88]. Recent studies also demonstrate that such nanoparticles can activate mechanotransduction pathways through magneto-mechanical stimulation, further augmenting osteogenesis [89]. However, the long-term safety of these nanomaterials requires more thorough discussion. Gold nanoparticles, despite their osteoinductive potential, exhibit dose-dependent cytotoxicity and can induce reactive oxygen species generation and membrane damage at high concentrations. Silica nanoparticle interactions with bone cells are highly dependent on size, surface chemistry, and protein corona formation, which can unpredictably alter cellular responses and toxicity profiles [90, 91]. Moreover, while magnetic nanoparticles such as SPIONs enable enhanced cell homing, tracking, and mechanotransduction, concerns remain regarding aggregation, potential iron-mediated oxidative stress, immune activation, and limited in vivo localization, particularly in deep tissues. Therefore, before predicting clinical viability, these long-term biocompatibility and safety considerations must be rigorously evaluated through dose–response studies, biodegradation assessments, and preclinical trials [91].

Fig. 5.

Fig. 5

Schematic representation of nanoparticle-mediated functionalization strategies. Nanoparticles serve as delivery platforms for bioactive molecules, enabling targeted modulation of cellular behavior and enhancing tissue regeneration through improved stability, cellular uptake, and controlled release of therapeutic agents.

Adapted from the reference [95] under the terms and conditions of a Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/).

Beyond nanomaterials, calcium-based biomolecules like calcium chloride (CaCl₂) and amino-calcium (AC) are gaining attention for bone regeneration due to their osteoconductive properties. Amino-calcium has demonstrated significant bone-preserving effects in ovariectomized rats, enhancing bone volume and trabecular connectivity, while upregulating osteogenic markers such as IBSP and BGLAP in MC3T3-E1 cells. Similarly, CaCl₂, when integrated into bioceramic or polymeric scaffolds, improves compressive strength, mineralization, and cell adhesion. The long-term biodegradation, immunogenicity, and systemic effects of both AC and CaCl₂ remain insufficiently characterized. Thus, rigorous in vivo studies are essential to validate their biocompatibility and clinical safety [9294].

Cell surface modification techniques have been developed to enhance homing efficiency and interaction with bone tissue [96]. Ligand-receptor strategies, such as conjugating MSC surfaces with RGD motifs, CXCR4, or integrin-targeting peptides, improve adherence to inflamed or injured endothelium and facilitate site-specific accumulation [97]. Biomimetic coatings using extracellular matrix proteins like fibronectin or osteopontin have been shown to increase integrin-mediated adhesion and downstream osteogenic signaling [98]. These multifaceted strategies underscore the progress toward precision MSC therapies. By integrating genetic, biochemical, physical, and surface-engineering tools, researchers are now able to fine-tune the therapeutic profile of MSCs to match the complex demands of bone healing, particularly in challenging clinical scenarios such as critical-sized defects or osteoporotic fractures [99].

MSC delivery platforms and bioengineering integration

Advanced delivery platforms and bioengineering strategies have been developed to optimize the efficacy of stem cell therapies for bone regeneration given in Table 1. 3D bioprinted scaffolds offer precise control over structural features, enhancing cell attachment, proliferation, and differentiation [100]. These scaffolds are often combined with osteoinductive factors to further support bone formation [101]. Hydrogel-based and injectable carriers provide flexibility and enable minimally invasive delivery, offering protection to cells while facilitating sustained release of bioactive molecules [102]. Microcarrier-based systems assist in cell expansion during culture and promote efficient transplantation, improving cell survival [103]. Biofunctionalized implant surfaces guide cell migration and differentiation, promoting tissue integration and osseointegration [104]. Additionally, exosome-loaded cells are emerging as a novel approach, using the bioactive molecules in exosomes to enhance tissue regeneration and improve therapeutic outcomes [105].

Table 1.

Various MSC delivery platforms and bioengineering integration strategies for enhanced bone Regeneration, highlighting key features, advantages, and associated challenges in each approach

Delivery Platform Key Features Advantages Challenges References
3D Bioprinted Scaffolds Customizable 3D-printed scaffolds for precise bone defect repair. High precision, supports cell growth, osteogenesis. High cost, technical complexity. [106]
Hydrogel and Injectable Carriers Hydrogels used as injectable carriers for MSC delivery. Easy to inject, adaptable to various defect shapes, biocompatible. Limited mechanical strength, burst release. [107]
Microcarrier-Based MSC Expansion Biodegradable microcarriers for MSC expansion in culture. Efficient cell expansion, scalable for clinical applications. Limited long-term stability, handling challenges. [108]
Biofunctionalized Implant Surfaces Implants functionalized with bioactive molecules to enhance MSC adhesion. Enhanced cell attachment and differentiation, improved integration. Difficulty in maintaining bioactivity in vivo. [109]
Exosome-Loaded Functionalized MSCs MSCs loaded with exosomes for enhanced therapeutic effects. Enhanced regenerative potential, targeted therapy. Difficulty in exosome isolation, scalability issues. [110]
Electrospun Nanofiber Scaffolds Nanofibers mimicking the extracellular matrix for MSC culture. High surface area, customizable fiber alignment, promotes osteogenesis. Low scalability, issues with fiber orientation. [111]
Microsphere-Based Drug Delivery Biodegradable microspheres for controlled release of MSCs or growth factors. Sustained release, enhanced cell survival, and targeted delivery. Burst release, limited release control. [112]
Nanostructured Biomaterial Coatings Coating of implants with nanomaterials to enhance MSC interactions. Improved MSC adhesion and osteogenic differentiation. Coating consistency, immune responses. [113]
Photopolymerized Hydrogels Hydrogels that solidify under UV light, encapsulating MSCs. Precise control over scaffold formation, biocompatible. UV sensitivity, limited to certain applications. [114]
Nanofiber-Encapsulated Cell Therapy MSCs encapsulated in nanofiber mats for enhanced interaction with matrix. Promotes differentiation and cell migration. Production difficulty, cell viability concerns. [115]
Self-assembled Peptide Hydrogels Hydrogels that self-assemble from peptides, supporting MSC encapsulation. Biocompatible, promotes osteogenesis, easy to functionalize. Limited mechanical strength, stability issues. [116]
ECM Mimicking Scaffolds Scaffolds designed to mimic natural bone extracellular matrix. Better MSC differentiation and integration into host tissue. Difficulty in mimicking natural ECM properties. [117]
Microfluidic Platforms for MSC Culture Microfluidic devices providing controlled culture environments for MSCs. Precise nutrient and waste control, supports MSC differentiation. Complex design, scalability issues. [118]

Pharmacokinetic and pharmacodynamic perspectives in optimized MSC-based bone regeneration therapies

Understanding the pharmacokinetics (PK) and pharmacodynamics (PD) of functionalized MSC therapies is essential for optimizing their clinical translation in bone regeneration [119]. Following systemic or local administration, MSC biodistribution is influenced by various factors, including cell surface modifications, delivery route, and the presence of inflammatory or hypoxic microenvironments [119121]. A major pharmacokinetic limitation involves first-pass pulmonary entrapment, where over 80% of infused cells are sequestered in the lungs, limiting bioavailability at osseous defect sites [121]. Functionalization strategies such as SDF-1/CXCR4 axis modulation, integrin ligand incorporation, and biomaterial-assisted cell delivery have shown promise in enhancing homing efficiency and tissue-specific retention [122]. MSCs primarily exert their therapeutic effects via a paracrine mechanism, secreting a repertoire of cytokines, growth factors, and extracellular vesicles that modulate osteogenesis, angiogenesis, and immune responses [123, 124]. These enhancements such as gene editing (BMP-2 overexpression), miRNA modulation, and nanoparticle conjugation have been employed to prolong and potentiate these effects in vivo [125]. However, the transient viability of transplanted MSCs and the temporally limited nature of their secretome necessitate integration with sustained-release delivery systems to maintain therapeutic efficacy [126]. The delivery route critically affects both pharmacokinetics and regenerative efficacy. Local delivery via intraosseous injection, hydrogel scaffolds, or composite carriers—offers superior outcomes in bone healing contexts by enabling direct cell deposition at the defect site, enhancing cell retention, and promoting sustained paracrine signaling within the bone microenvironment. This is especially important given bone’s relatively low vascularity compared to neural or spinal tissues, which limits systemic MSC migration and engraftment in orthopedic indications [127]. Systemic delivery, while non-invasive and suitable for diffuse skeletal conditions, often results in off-target cell accumulation (especially in the lungs and liver) and requires strategies to enhance homing capacity to bone tissue. Moreover, systemic routes may exhibit unpredictable pharmacodynamics in poorly perfused osseous lesions, underscoring the importance of site-specific delivery platforms [128, 129].

PK/PD profiling standardization for cell-based products remains a critical gap in current translational pipelines [130]. Quantitative imaging, biodistribution assays, and systems pharmacology models are being developed to better predict cell fate, optimal dosing, and therapeutic windows [131]. As regulatory agencies move toward stricter guidelines for advanced therapy medicinal products (ATMPs), incorporating robust PK/PD frameworks will be pivotal in ensuring safety, efficacy, and batch-to-batch consistency in modified MSC therapies for bone repair [132].

Preclinical and clinical evidence

Preclinical studies play a pivotal role in evaluating the efficacy and safety of MSC-based therapies for bone regeneration. Animal models have often been used to assess MSC transplantation in bone fractures, critical-sized defects, and osteoporotic bone healing [133]. Bone Defects (Fractures, Critical-sized defects).

In fracture and defect models, MSCs have been shown to enhance callus formation, bridge critical-sized gaps, accelerate mineralization, and improve biomechanical strength, as assessed through BMD, histology, µCT, and mechanical testing. Notably, a systematic meta-analysis of systemic MSC administration in preclinical bone regeneration revealed significant increases in bone mineral density (SMD 3.02), bone volume/tissue volume (SMD 2.10), and new bone area (SMD 7.03) [129]. Additionally, both systemic and local MSC delivery enhanced healing in murine fracture models, although systemic delivery was more feasible in diffuse skeletal injuries [134].

Osteoporosis-related bone regeneration

In osteoporotic models, MSCs have demonstrated potential in restoring bone mass and microarchitecture. Transplantation of BM-MSCs in OVX-induced osteoporotic animals resulted in significant improvements in BMD, trabecular thickness, and osteogenic marker expression (ALP, osteocalcin), along with reduced TNF-α and enhanced trabecular microstructure [135]. However, systemic administration of unmodified allogeneic MSCs failed to halt bone loss in osteoporotic rats—and, in some cases, repeated injections accelerated bone loss, underscoring the need for targeted homing enhancements, such as integrin modification or scaffold-based delivery [136]. Furthermore, the regenerative capacity of MSCs in osteoporotic individuals appears preserved: MSCs derived from osteoporotic patients exhibited comparable or superior consolidation in fracture models, particularly after systemic delivery, compared to MSCs from osteoarthritic donors [137]. These findings underscore the necessity for delineated strategies local delivery may be more appropriate for isolated bone defects, while osteoporosis-related applications may benefit from engineered MSC homing mechanisms to enhance systemic targeting efficacy.

Challenges and limitations

Despite their regenerative potential, the clinical translation of MSCs for bone repair remains constrained by several key limitations (Table 2). One of the foremost issues is poor in vivo survival and engraftment, with the majority of transplanted cells failing to persist or integrate due to the harsh microenvironment of bone injury sites. Immunogenicity, particularly in allogeneic applications, poses risks of host rejection, while genetic or epigenetic modifications raise concerns about tumorigenicity [138]. MSCs surface modification protocols vary widely across laboratories, leading to inconsistency in therapeutic outcomes. From a manufacturing standpoint, challenges in large-scale expansion, maintaining phenotypic stability, and meeting Good Manufacturing Practice (GMP) requirements further complicate clinical translation [139]. Regulatory uncertainty, coupled with ethical concerns about cell sourcing and manipulation, continues to hinder standardized approval pathways [140]. Additionally, insufficient vascularization, the complexity of scaffold integration, and lack of long-term safety data limit broader adoption. Collectively, these obstacles emphasize the need for robust, reproducible, and safe manipulation strategies that align with regulatory and clinical frameworks [141].

Table 2.

Summary of major limitations associated with different types of MSC-based therapies for bone regeneration, highlighting clinical and translational barriers

Type of MSC-Based Therapy Limitation Description References
Unmodified MSC Transplantation Low in vivo survival Transplanted MSCs exhibit poor viability due to harsh bone injury environments. [145]
Allogeneic MSC Therapy Immunogenicity Risk of host immune rejection remains significant in allogeneic applications. [146]
Gene-Modified MSCs Tumorigenicity Genetic manipulation may lead to uncontrolled growth or oncogenic mutations. [147]
MSCs with Scaffold Integration Scaffold-cell mismatch Mechanical or biochemical incompatibility can impair MSC attachment and differentiation. [148]
MSC Expansion on Microcarriers Limited scalability Difficult to scale up while preserving phenotype and osteogenic capacity. [103]
Epigenetically Modified MSCs Protocol variability Variability in modification protocols affects reproducibility across labs. [149]
MSCs with Angiogenic Preconditioning Poor vascularization Despite angiogenic cues, inadequate vascular support often hampers full integration. [150]
Donor-Derived MSCs (BM, AD, UC) Donor variability Cell potency and immunomodulation potential vary with donor age and health. [151]
MSC-Loaded Injectable Hydrogels Inconsistent osteogenic differentiation Variable responses due to material properties or local tissue conditions. [152]
GMP-Grade MSC Expansion Systems GMP compliance challenges Complex documentation, traceability, and sterility requirements increase regulatory burden. [153]
Scaffold-Free MSC Sheets Limited engraftment Lack of 3D support can reduce the structural integration in large defects. [154]
Exosome-Loaded MSCs Lack of long-term efficacy data Therapeutic benefits are promising but long-term studies are insufficient. [155]
Genetically Enhanced MSCs (BMP-2) Regulatory uncertainty No clear approval pathways for gene-edited cell therapies in most regions. [156]
Bioactive Surface-Coated MSC Implants High production costs Functionalization and sterilization increase economic barriers. [157]
MSCs from Fetal/Perinatal Sources Ethical concerns Use of certain cell sources may raise ethical issues regarding consent and origin. [158]

Moreover, genetic modification of MSCs introduces unique and critical biosafety challenges. Prolonged ex vivo expansion has been shown to lead to spontaneous malignant transformation in a significant proportion of human bone marrow‑derived MSC cultures 45.8% in one study resulting in tumorigenic phenotypes when transplanted into immunodeficient mice (cells formed rapidly growing lung tumors) [142]. Even in cultures without overt malignant transformation, chromosomal abnormalities including DNA copy number variations (CNVs) and aneuploidy have been detected upon long-term passaging, raising concerns over genomic stability [143]. Further compounding these risks, the introduction of immortalizing or transgene constructs such as hTERT, SV40 large T antigen, or oncogenes like c‑Myc and BMI‑1 is associated with potential loss of cell cycle checkpoint control, karyotypic instability, and insertional mutagenesis, all of which may drive neoplastic transformation [144]. Preclinical studies also report that murine MSCs are particularly susceptible to oncogenic transformation: passage‑3 MSCs implanted into mice have developed sarcoma-like tumors, a risk that underscores the need for cautious translation of gene-modified MSCs from animal models to human clinical applications [19].

Emerging trends and future directions

Emerging directions in MSC-based bone regeneration are rapidly evolving through the integration of precision gene editing, smart biomaterials, computational biology, and personalized medicine frameworks [159]. At the forefront, gene-editing technologies such as CRISPR-Cas9, base editing, and prime editing are being harnessed to selectively enhance osteogenic gene expression in MSCs without disrupting genomic integrity. These tools allow fine-tuned regulation of transcription factors like RUNX2 or SP7, as well as immune-modulatory genes to improve allogeneic cell acceptance [160]. However, these approaches are not without risks. CRISPR-Cas9 has been associated with off-target effects, where unintended genomic sites are edited, potentially leading to harmful mutations or genomic instability. Moreover, the ethical implications of human germline editing remain contentious, with concerns about informed consent, potential misuse for eugenics, and the long-term impact on human evolution. Therefore, while CRISPR-based strategies offer promising avenues for enhancing osteogenesis, their clinical application necessitates rigorous safety assessments and ethical considerations [161]. Compared to traditional viral transduction, these non-integrative systems minimize insertional mutagenesis risks, offering greater translational safety [162]. Simultaneously, artificial intelligence into stem cell engineering has transformed MSC formulation design. AI-driven models trained on multi-omics datasets—combining transcriptomic, proteomic, and metabolomic profiles can now predict MSC behavior in response to scaffold composition, bioactive loading, and environmental conditions. This capability enables the rapid development of optimized, patient-specific regenerative formulations while minimizing empirical trial-and-error [163]. In the domain of biomaterials, smart bioresponsive implants represent a significant leap forward. These platforms incorporate embedded biosensors that monitor microenvironmental parameters—such as pH, inflammatory cytokines, or mechanical stress—and adjust the release of therapeutic agents (growth factors or anti-inflammatory molecules) accordingly. Such closed-loop systems are particularly relevant in post-traumatic or osteoporotic environments where dynamic regulation of healing is required [164]. Moreover, personalized MSC grafts is reshaping clinical delivery strategies. Using patient-derived imaging data (CT or MRI), computer-aided design and additive biomanufacturing enable the creation of anatomically tailored scaffolds loaded with tailored MSCs. Advanced bioinks incorporating osteoconductive and angiogenic cues further support spatial patterning of osteogenesis and vascular integration, especially in irregular or critical-sized bone defects [165].

On the translational front, there is increasing emphasis on multi-arm, multicenter clinical trial design to accommodate inter-patient variability and accelerate regulatory validation. Such designs allow simultaneous comparison of delivery modes, cell sources, or MSC phenotypic modulation strategies, enhancing data robustness while reducing time to market. Additionally, the development of Good Manufacturing Practice (GMP)-compliant bioprocessing platforms is critical for scalable MSC expansion and quality control, which remains a major bottleneck in clinical readiness [153]. Looking forward, integrating quantitative pharmacokinetic and pharmacodynamic (PK/PD) modeling of MSC-based therapies, which can inform dosing regimens, biodistribution, and therapeutic windows—currently underexplored in cell-based regenerative medicine [153]. Furthermore, the use of multi-omics-integrated digital twins—virtual patient-specific models predicting therapeutic outcomes may enable pre-intervention simulations to enhance treatment planning and regulatory approval processes. These innovations mark a transformative phase in MSC-mediated bone healing. The convergence of high-precision gene modulation, intelligent delivery systems, predictive modeling, and personalized bioengineering is poised to overcome longstanding translational barriers, moving MSC therapies from bench to bedside with greater efficiency, safety, and patient specificity [166].

Despite advances in MSC therapies, economic barriers limit their broad clinical adoption. High manufacturing and quality control costs hinder reimbursement and accessibility, especially in low- and middle-income countries where healthcare funding is limited. While some studies suggest MSC treatments may be cost-effective compared to standard care in specific conditions, comprehensive long-term economic data remain scarce. These financial challenges restrict MSC therapies mainly to affluent regions and specialized centers, adding to the regulatory hurdles faced globally [167, 168]. Mesenchymal stem cell (MSC) therapies face significant regulatory and standardization challenges across global jurisdictions, notably under agencies like the FDA (USA) and EMA (Europe). In the U.S., MSCs are regulated as biologics requiring Investigational New Drug (IND) applications under strict cGMP conditions, while the EMA classifies them as Advanced Therapy Medicinal Products (ATMPs), demanding complex clinical and quality documentation through centralized procedures [44, 169, 170]. However, regulatory pathways remain heterogeneous, with countries like Japan adopting conditional approval systems and others lacking robust oversight, contributing to inconsistent clinical access and proliferation of unregulated therapies [171]. A major bottleneck lies in the lack of standardized stem cell characterization current identity criteria (CD73, CD90, CD105 expression) fail to predict therapeutic efficacy, and validated potency assays (immunomodulation or angiogenesis) are not universally implemented in clinical-grade production [172]. Furthermore, reporting across clinical trials is inconsistent; many studies omit critical viability, function, or batch-specific data, limiting reproducibility [173]. Although efforts by bodies like ISO, NIST, and the Standards Coordinating Body are underway to harmonize manufacturing and quality assessment protocols, the absence of universally accepted benchmarks continues to hinder global regulatory convergence and clinical translation of MSC-based therapies [174].

Future clinical integration framework

The integration of MSC-based therapies into orthopedic clinical practice requires a structured and multifaceted approach. The first step in this framework involves scalable, GMP-compliant production of MSCs, ensuring the cells are produced at a high standard for safety, consistency, and efficacy. The need for large-scale expansion of MSCs, coupled with efficient storage and transport systems, will be essential to meet the demands of clinical applications, particularly in managing large patient populations. Once MSCs are produced, the next challenge lies in clinical translation and regulatory approval. In many countries, this requires thorough preclinical data supporting the safety and efficacy of MSC-based therapies, including robust evidence from animal models and early-phase human trials. Regulatory agencies like the FDA or EMA require clear demonstrations of cell viability, immunogenicity, and potential for tumorigenicity, which necessitates meticulous quality control protocols during production and administration. The use of standardized clinical protocols, including clear guidelines for cell dosage, administration route, and follow-up care, will be key to ensuring widespread acceptance [168, 175].

Another critical component is the training and integration of healthcare professionals, including orthopedic surgeons, cell biologists, and medical technologists, who will need to familiarize themselves with MSC therapies, novel delivery platforms, and post-infusion care procedures. Furthermore, healthcare facilities will need to establish specialized units capable of handling the advanced nature of these treatments, including 3D bioprinting technology, bioactive scaffolds, and injectable hydrogels that support MSC function during in vivo integration. Multi-center clinical trials will be required to assess the safety and efficacy of MSC-based therapies across diverse patient populations. The clinical integration framework must account for the biological variability seen in different patient groups (e.g., age, comorbidities), and testing should include multi-arm trials designed to optimize MSC activation and delivery methods for specific subpopulations. Once clinical validation is achieved, insurance and reimbursement pathways must be defined, ensuring that these therapies are accessible to patients. The emergence of digital health tools as real-time monitoring systems and AI-driven patient stratification models could enhance clinical decision-making. These technologies would support clinicians in adjusting MSC-based treatments in response to real-time feedback from patients, thus maximizing the therapeutic outcomes. By addressing these components in a cohesive, step-by-step framework, MSC-based therapies can be effectively integrated into orthopedic clinics, providing a robust solution for bone regeneration and joint repair in patients suffering from degenerative diseases, fractures, and other musculoskeletal disorders [168].

Conclusion

The functionalization of mesenchymal stem cells presents an exciting frontier in the field of bone regeneration therapies, with the potential to revolutionize current treatment strategies for bone defects and fractures. Significant advancements have been made through various techniques such as gene modification, preconditioning, nanoparticle integration, and scaffold-based delivery systems. These methods have shown considerable promise in enhancing the osteogenic potential of MSCs, promoting bone repair, and improving overall healing outcomes. Delivery platforms have been designed to optimize cell activity, facilitate targeted release of bioactive molecules, and support sustained healing at defect sites. Moreover, while delivery platforms have shown promise in controlled release, the optimization of these systems for precise, patient-specific therapeutic outcomes remains an ongoing area of research. Looking ahead, emerging technologies, such as precision-engineered implants, 3D bioprinting, and artificial intelligence-driven design, hold great potential to address these challenges. These innovations can facilitate the development of more efficient and scalable manufacturing processes, as well as better-targeted therapies that align with the individual needs of patients. The convergence of these advancements signals a transformative shift in bone healing strategies, offering a promising future for the clinical application of MSC-based therapies. As the field progresses, continued research focusing on overcoming current limitations, such as large-scale production, immune compatibility, and precise delivery, will be essential to unlock the full therapeutic potential of MSCs and establish them as a cornerstone in regenerative medicine for bone repair.

Acknowledgements

This work was supported by Zhejiang Provincial Natural Science Foundation of China (QN25H040001 to LYX), Medical and Health Research Project of Zhejiang Province (2025KY1804 to LYX), Traditional Chinese Medicine Science and Technology Projects of Zhejiang Province (2025ZR218 to LYX), Science and Technology Project of Zhoushan (2025C31060 to SQ, 2025C31079 to LC, 2024C31019 to LYX), Research Fund Projects of The Affiliated Hospital of Zhejiang Chinese Medicine University (2024FSYYZZ14 to YJY, 2024FSYYZY34 to HML).

Author contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work. All the authors listed meet the criteria for authorship as per the ICMJE guidelines, read the final manuscript and agree to publish this work.

Funding

None.

Data availability

No datasets were generated or analysed during the current study.

Declarations

Ethical approval and consent to participate

Not Applicable. This is a review paper and does not involve direct research on humans or animals.

Consent for publication

“Not applicable” as this manuscript does not contain data from any person.

Competing interests

The authors declare no competing interests.

Footnotes

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Wentao Huang, Chao Zhou and Yijun Yu contributed equally to this work.

Contributor Information

Linying Xia, Email: xialinying1224@163.com.

Wenqing Liang, Email: liangwq@usx.edu.cn.

References

  • 1.Bowers KM, Anderson DE. Delayed union and nonunion: current concepts, prevention, and correction: a review. Bioengineering. 2024;11(6):525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Georgeanu VA, Gingu O, Antoniac IV, Manolea HO. Current options and future perspectives on bone graft and biomaterials substitutes for bone repair, from clinical needs to advanced biomaterials research. Appl Sci. 2023;13(14):8471. [Google Scholar]
  • 3.Miron RJ. Optimized bone grafting. Periodontology 2000, 2024. 94(1): pp. 143–160. [DOI] [PubMed]
  • 4.Szwed-Georgiou A, et al. Bioactive materials for bone regeneration: biomolecules and delivery systems. ACS Biomaterials Sci Eng. 2023;9(9):5222–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kantaros A, Ganetsos T. From static to dynamic: smart materials pioneering additive manufacturing in regenerative medicine. Int J Mol Sci. 2023;24(21):15748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Gholami Farashah MS, et al. Bone marrow mesenchymal stem cells’ osteogenic potential: superiority or non-superiority to other sources of mesenchymal stem cells? Cell Tissue Banking. 2023;24(3):663–81. [DOI] [PubMed] [Google Scholar]
  • 7.Maacha S, et al. Paracrine mechanisms of mesenchymal stromal cells in angiogenesis. Stem Cells Int. 2020;2020(1):4356359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Hayrapetyan A, Jansen JA, van den Beucken JJ. Signaling pathways involved in osteogenesis and their application for bone regenerative medicine. Tissue Eng Part B: Reviews. 2015;21(1):75–87. [DOI] [PubMed] [Google Scholar]
  • 9.González-González A, et al. Engineering therapeutic mesenchymal stem cells secretome for bone Regeneration, in Handbook of regenerative medicine. CRC; 2025. pp. 181–98.
  • 10.Zheng Q, Zhang S, Guo W-Z, Li X-K. The unique Immunomodulatory properties of MSC-derived exosomes in organ transplantation. Front Immunol. 2021;12:659621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Alagesan S, et al. Enhancement strategies for mesenchymal stem cells and related therapies. Stem Cell Res Ther. 2022;13(1):75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kouroupis D, Correa D. Increased mesenchymal stem cell functionalization in three-dimensional manufacturing settings for enhanced therapeutic applications. Front Bioeng Biotechnol. 2021;9:621748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pishavar E, et al. Advanced hydrogels as exosome delivery systems for osteogenic differentiation of mscs: application in bone regeneration. Int J Mol Sci. 2021;22(12):6203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Han D, et al. Controlled delivery of mesenchymal stem cells via biodegradable scaffolds for fracture healing. Nanomedicine. 2025;20(2):207–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Percival KM, Paul V, Husseini GA. Recent advancements in bone tissue engineering: integrating smart scaffold technologies and Bio-Responsive systems for enhanced regeneration. Int J Mol Sci. 2024;25(11):6012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Xiong Y, Mi B, Liu G, Zhao Y. Microenvironment-sensitive nanozymes for tissue regeneration. Biomaterials, 2024: p. 122585. [DOI] [PubMed]
  • 17.Kulus M, et al. Mesenchymal stem/stromal cells derived from human and animal perinatal tissues—origins, characteristics, signaling pathways, and clinical trials. Cells. 2021;10(12):3278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Zhu C, Wu W, Qu X. Mesenchymal stem cells in osteoarthritis therapy: a review. Am J Translational Res. 2021;13(2):448. [PMC free article] [PubMed] [Google Scholar]
  • 19.Xiao W, Mohseny AB, Hogendoorn PC, Cleton-Jansen A-M. Mesenchymal Stem Cell Transformation Sarcoma Genesis Clin Sarcoma Res. 2013;3(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Via AG, Frizziero A, Oliva F. Biological properties of mesenchymal stem cells from different sources. Muscles Ligaments Tendons J. 2012;2(3):154. [PMC free article] [PubMed] [Google Scholar]
  • 21.Jin HJ, et al. Comparative analysis of human mesenchymal stem cells from bone marrow, adipose tissue, and umbilical cord blood as sources of cell therapy. Int J Mol Sci. 2013;14(9):17986–8001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Bai X, et al. Dental pulp stem cells for bone tissue engineering: a literature review. Stem Cells Int. 2023;2023(1):7357179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moeenzade N, Naseri M, Osmani F, Razavi FE. Dental pulp stem cells for reconstructing bone defects: a systematic review and meta-analysis. J Dent Res Dent Clin Dent Prospects. 2022;16(4):204. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liu Y, et al. Application of dental pulp stem cells for bone regeneration. Front Med. 2024;11:1339573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Hamad-Alrashid H, et al. Bone regeneration with dental pulp stem cells in an experimental model. J Personalized Med. 2024;14(11):1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Maslennikov S, Maksym G. The most commonly used cell surface markers for determining mesenchymal stromal cells in stromal vascular fraction and bone marrow autologous concentrate: a systematic review. J Biotech Res, 2023. 14.
  • 27.Alvarez-Viejo M, Haider KH. Mesenchymal stem cells: from identification and characterization to clinical applications, in Handbook of stem cell therapy. Springer; 2022. pp. 127–62.
  • 28.Costa LA, et al. Functional heterogeneity of mesenchymal stem cells from natural niches to culture conditions: implications for further clinical uses. Cell Mol Life Sci. 2021;78:447–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Dunn CM, Kameishi S, Grainger DW, Okano T. Strategies to address mesenchymal stem/stromal cell heterogeneity in Immunomodulatory profiles to improve cell-based therapies. Acta Biomater. 2021;133:114–25. [DOI] [PubMed] [Google Scholar]
  • 30.Zha K, et al. Recent advances in enhancement strategies for osteogenic differentiation of mesenchymal stem cells in bone tissue engineering. Front Cell Dev Biology. 2022;10:824812. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Shi H, et al. Integrating physicomechanical and biological strategies for BTE: biomaterials-induced osteogenic differentiation of MSCs. Theranostics. 2023;13(10):3245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Tang C-Y, et al. Runx1 is a central regulator of osteogenesis for bone homeostasis by orchestrating BMP and WNT signaling pathways. PLoS Genet. 2021;17(1):e1009233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chan WCW, Tan Z, To MKT, Chan D. Regulation and role of transcription factors in osteogenesis. Int J Mol Sci. 2021;22(11):5445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Arya PN, Saranya I, Selvamurugan N. Crosstalk between Wnt and bone morphogenetic protein signaling during osteogenic differentiation. World J Stem Cells. 2024;16(2):102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kulesza A, Paczek L, Burdzinska A. The role of COX-2 and PGE2 in the regulation of Immunomodulation and other functions of mesenchymal stromal cells. Biomedicines. 2023;11(2):445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Ahn J, et al. Regenerative functions of regulatory T cells and current strategies utilizing mesenchymal stem cells in Immunomodulatory tissue regeneration. Tissue Engineering and Regenerative Medicine; 2025. pp. 1–14. [DOI] [PMC free article] [PubMed]
  • 37.Merimi M, et al. The therapeutic potential of mesenchymal stromal cells for regenerative medicine: current knowledge and future Understandings. Front Cell Dev Biology. 2021;9:661532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Hori A, et al. Superior migration ability of umbilical cord-derived mesenchymal stromal cells (MSCs) toward activated lymphocytes in comparison with those of bone marrow and adipose-derived MSCs. Front Cell Dev Biology. 2024;12:1329218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhan X-S, et al. A comparative study of biological characteristics and transcriptome profiles of mesenchymal stem cells from different canine tissues. Int J Mol Sci. 2019;20(6):1485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Guerrero EN, et al. Increased proliferation and differentiation capacity of placenta-derived mesenchymal stem cells from women of median maternal age correlates with telomere shortening. Aging. 2021;13(22):24542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Trivanović D, et al. Mesenchymal stem cells of different origin: comparative evaluation of proliferative capacity, telomere length and pluripotency marker expression. Life Sci. 2015;141:61–73. [DOI] [PubMed] [Google Scholar]
  • 42.Pal D, et al. Recent trends of stem cell therapies in the management of orthopedic surgical challenges. Int J Surg. 2024;110(10):6330–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kirankumar S, et al. Modern approaches on stem cells and scaffolding technology for osteogenic differentiation and regeneration. Experimental Biology Med. 2022;247(5):433–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jovic D, et al. A brief overview of global trends in MSC-based cell therapy. Stem Cell Reviews Rep. 2022;18(5):1525–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Galderisi U, Peluso G, Di G, Bernardo. Clinical trials based on mesenchymal stromal cells are exponentially increasing: where are we in recent years? Stem Cell Reviews Rep. 2022;18(1):23–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Česnik AB, Švajger U. The issue of heterogeneity of MSC-based advanced therapy medicinal products–a review. Front Cell Dev Biology. 2024;12:1400347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Lui PPY, Leung YT. Practical considerations for translating mesenchymal stromal cell-derived extracellular vesicles from bench to bed. Pharmaceutics. 2022;14(8):1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Battafarano G, et al. Strategies for bone regeneration: from graft to tissue engineering. Int J Mol Sci. 2021;22(3):1128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kalkal M, et al. Mesenchymal stem cells: A novel therapeutic approach to enhance protective Immunomodulation and erythropoietic recovery in malaria. Stem Cell Reviews Rep. 2021;17(6):1993–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Zhu M, et al. Orchestration of mesenchymal stem/stromal cells and inflammation during wound healing. Stem Cells Translational Med. 2023;12(9):576–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Costela-Ruiz VJ, et al. Different sources of mesenchymal stem cells for tissue regeneration: a guide to identifying the most favorable one in orthopedics and dentistry applications. Int J Mol Sci. 2022;23(11):6356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Al-Sharabi N, et al. Osteogenic human MSC-derived extracellular vesicles regulate MSC activity and osteogenic differentiation and promote bone regeneration in a rat calvarial defect model. Stem Cell Res Ther. 2024;15(1):33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lee E-J, Jain M, Alimperti S. Bone microvasculature: stimulus for tissue function and regeneration. Tissue Eng Part B: Reviews. 2021;27(4):313–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jing Z, et al. Bone formation and bone repair: the roles and crosstalk of osteoinductive signaling pathways. Process Biochem. 2022;118:252–62. [Google Scholar]
  • 55.Zhu S, Chen W, Masson A, Li Y-P. Cell signaling and transcriptional regulation of osteoblast lineage commitment, differentiation, bone formation, and homeostasis. Cell Discovery. 2024;10(1):71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Nambiar J, Jana S, Nandi SK. Strategies for enhancing vascularization of Biomaterial-Based scaffold in bone regeneration. Volume 22. The Chemical Record; 2022. p. e202200008. 6. [DOI] [PubMed]
  • 57.Khoswanto C. Role of matrix metalloproteinases in bone regeneration: narrative review. J Oral Biology Craniofac Res. 2023;13(5):539–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Song C, et al. Immunomodulation pathogenesis and treatment of bone nonunion. Orthop Surg. 2024;16(8):1770–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Grosso A, et al. It takes two to tango: coupling of angiogenesis and osteogenesis for bone regeneration. Front Bioeng Biotechnol. 2017;5:68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hankenson KD, Dishowitz M, Gray C, Schenker M. Angiogenesis in bone regeneration. Injury. 2011;42(6):556–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Noh J-Y, Yang Y, Jung H. Molecular mechanisms and emerging therapeutics for osteoporosis. Int J Mol Sci. 2020;21(20):7623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Huang H, Ma L, Kyrkanides S. Effects of vascular endothelial growth factor on osteoblasts and osteoclasts. Am J Orthod Dentofac Orthop. 2016;149(3):366–73. [DOI] [PubMed] [Google Scholar]
  • 63.Wang H, et al. Advances in the role and mechanism of fibroblasts in fracture healing. Front Endocrinol. 2024;15:1350958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Wan Z, et al. Programmed biomolecule delivery orchestrate bone tissue regeneration via MSC recruitment and epigenetic modulation. Chem Eng J. 2022;438:135518. [Google Scholar]
  • 65.Attia N, Mashal M, Puras G, Pedraz JL. Mesenchymal stem cells as a gene delivery tool: promise, problems, and prospects. Pharmaceutics. 2021;13(6):843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.LEK TK. Genetically engineered mesenchymal stem cells using viral vectors: A new frontier in Anti-Angiogenic therapy. Sains Malaysiana. 2024;53(1):63–86. [Google Scholar]
  • 67.Dick TA, Sone ED, Uludağ H. Mineralized vectors for gene therapy. Acta Biomater. 2022;147:1–33. [DOI] [PubMed] [Google Scholar]
  • 68.Watson-Levings RS, et al. Gene therapy in orthopaedics: progress and challenges in pre-clinical development and translation. Front Bioeng Biotechnol. 2022;10:901317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Loh H-Y, et al. Post-transcriptional regulatory crosstalk between MicroRNAs and canonical TGF-β/BMP signalling cascades on osteoblast lineage: a comprehensive review. Int J Mol Sci. 2023;24(7):6423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Shams R, et al. Developed bone biomaterials incorporated with MicroRNAs to promote bone regeneration: A systematic Review, Bioinformatics, and Meta-Analysis study. ACS Biomaterials Sci Eng. 2023;9(9):5186–204. [DOI] [PubMed] [Google Scholar]
  • 71.Komatsu DE, Duque E, Hadjiargyrou M. MicroRNAs and fracture healing: pre-clinical studies. Bone. 2021;143:115758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Shi D et al. Nanomaterials-Based Drug Delivery Systems for Therapeutic Applications in Osteoporosis. Advanced Biology: p. 2400721. [DOI] [PubMed]
  • 73.Liu R, et al. MicroRNAs delivered by small extracellular vesicles in MSCs as an emerging tool for bone regeneration. Front Bioeng Biotechnol. 2023;11:1249860. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Li Y, et al. The application of extracellular vesicles in orthopedic diseases. Interdisciplinary Med. 2024;2(3):e20230055. [Google Scholar]
  • 75.Tang J, Wang X, Lin X, Wu C. Mesenchymal stem cell-derived extracellular vesicles: a regulator and carrier for targeting bone-related diseases. Cell Death Discovery. 2024;10(1):212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lanzillotti C, et al. Long non-coding RNAs and MicroRNAs interplay in osteogenic differentiation of mesenchymal stem cells. Front Cell Dev Biology. 2021;9:646032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Li N, et al. MSC-derived small extracellular vesicles attenuate autoimmune dacryoadenitis by promoting M2 macrophage polarization and inducing Tregs via miR-100-5p. Front Immunol. 2022;13:p888949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Luo G, et al. Glucose metabolism: optimizing regenerative functionalities of mesenchymal stromal cells postimplantation. Tissue Eng Part B: Reviews. 2023;29(1):47–61. [DOI] [PubMed] [Google Scholar]
  • 79.Hu W, Guo Z, Tang W, Long J. Mechanoresponsive regulation of tissue regeneration during distraction osteogenesis. FASEB J. 2024;38(18):e70056. [DOI] [PubMed] [Google Scholar]
  • 80.Wang L et al. Integrins in the regulation of mesenchymal stem cell differentiation by mechanical signals. Stem Cell Reviews Rep, 2022: pp. 1–16. [DOI] [PubMed]
  • 81.Liu Y, et al. The effects of hypoxia-preconditioned dental stem cell-derived secretome on tissue regeneration. Tissue Eng Part B: Reviews. 2025;31(1):44–60. [DOI] [PubMed] [Google Scholar]
  • 82.Garcia JP, et al. Hypoxia-preconditioning of human adipose-derived stem cells enhances cellular proliferation and angiogenesis: A systematic review. J Clin Translational Res. 2022;8(1):61. [PMC free article] [PubMed] [Google Scholar]
  • 83.Schott NG, Friend NE, Stegemann JP. Coupling osteogenesis and vasculogenesis in engineered orthopedic tissues. Tissue Eng Part B: Reviews. 2021;27(3):199–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Wang Y, et al. Nanoengineered endocytic biomaterials for stem cell therapy. Adv Funct Mater. 2024;34(52):2410714. [Google Scholar]
  • 85.Bi Z, et al. Strategies of immobilizing BMP-2 with 3D-printed scaffolds to improve osteogenesis. Regen Med. 2023;18(5):425–41. [DOI] [PubMed] [Google Scholar]
  • 86.Frączek W, Kotela A, Kotela I, Grodzik M. Nanostructures in orthopedics: advancing Diagnostics, targeted Therapies, and tissue regeneration. Materials. 2024;17(24):6162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wychowaniec JK, Brougham DF. Emerging magnetic fabrication technologies provide controllable Hierarchically-Structured biomaterials and stimulus response for biomedical applications. Adv Sci. 2022;9(34):2202278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mehta KJ. Iron oxide nanoparticles in mesenchymal stem cell detection and therapy. Stem Cell Reviews Rep. 2022;18(7):2234–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Rotherham M, et al. Remote magnetic actuation of cell signalling for tissue engineering. Curr Opin Biomedical Eng. 2022;24:100410. [Google Scholar]
  • 90.Farjaminejad S, Farjaminejad R, Garcia-Godoy F. Nanoparticles in bone regeneration: a narrative review of current advances and future directions in tissue engineering. J Funct Biomaterials. 2024;15(9):241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ha S-W, Viggeswarapu M, Habib MM, Beck GR Jr. Bioactive effects of silica nanoparticles on bone cells are size, surface, and composition dependent. Acta Biomater. 2018;82:184–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ali M, et al. The bone-protective benefits of amino-conjugated calcium in an ovariectomized (OVX) rat model. Life Sci. 2023;328:121927. [DOI] [PubMed] [Google Scholar]
  • 93.Kuzmenka D, et al. Sustained calcium (II)-release to impart bioactivity in hybrid glass scaffolds for bone tissue engineering. Pharmaceutics. 2020;12(12):1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pereira MM, Jones JR, Hench LL. Bioactive glass and hybrid scaffolds prepared by sol–gel method for bone tissue engineering. Adv Appl Ceram. 2005;104(1):35–42. [Google Scholar]
  • 95.Xu H, et al. Nanoparticle-based drug delivery systems for enhancing bone regeneration. ACS Biomaterials Sci Eng. 2024;10(3):1302–22. [DOI] [PubMed] [Google Scholar]
  • 96.Ren X, Chen X, Geng Z, Su J. Bone-targeted biomaterials: strategies and applications. Chem Eng J. 2022;446:137133. [Google Scholar]
  • 97.Sabir F, et al. Onco-receptors targeting in lung cancer via application of surface-modified and hybrid nanoparticles: A cross-disciplinary review. Processes. 2021;9(4):621. [Google Scholar]
  • 98.Azadi S, et al. Bioinspired synthetic peptide-based biomaterials regenerate bone through biomimicking of extracellular matrix. J Tissue Eng. 2024;15:20417314241303818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hussen BM, et al. Revolutionizing medicine: recent developments and future prospects in stem-cell therapy. Int J Surg. 2024;110(12):8002–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Adhikari J, et al. Effects of surface patterning and topography on the cellular functions of tissue engineered scaffolds with special reference to 3D Bioprinting. Biomaterials Sci. 2023;11(4):1236–69. [DOI] [PubMed] [Google Scholar]
  • 101.Collins MN, et al. Scaffold fabrication technologies and structure/function properties in bone tissue engineering. Adv Funct Mater. 2021;31(21):2010609. [Google Scholar]
  • 102.Liu J, Du C, Huang W, Lei Y. Injectable smart stimuli-responsive hydrogels: pioneering advancements in biomedical applications. Biomaterials Sci. 2024;12(1):8–56. [DOI] [PubMed] [Google Scholar]
  • 103.Major GS, et al. Mapping the microcarrier design pathway to modernise clinical mesenchymal stromal cell expansion. Trends Biotechnol. 2024;42(7):859–76. [DOI] [PubMed] [Google Scholar]
  • 104.Zhu G, Wang G, Li JJ. Advances in implant surface modifications to improve osseointegration. Mater Adv. 2021;2(21):6901–27. [Google Scholar]
  • 105.Khazaei F, et al. Exosomes and exosome-loaded scaffolds: characterization and application in modern regenerative medicine. Tissue Cell. 2023;80:102007. [DOI] [PubMed] [Google Scholar]
  • 106.Ivanovski S, et al. 3D printing for bone regeneration: challenges and opportunities for achieving predictability. Periodontol 2000. 2023;93(1):358–84. [DOI] [PubMed] [Google Scholar]
  • 107.Zheng Z, Yu C, Wei H. Injectable hydrogels as three-dimensional network reservoirs for osteoporosis treatment. Tissue Eng Part B: Reviews. 2021;27(5):430–54. [DOI] [PubMed] [Google Scholar]
  • 108.Ornelas-González A, González-González M, Rito-Palomares M. Microcarrier-based stem cell bioprocessing: GMP-grade culture challenges and future trends for regenerative medicine. Crit Rev Biotechnol. 2021;41(7):1081–95. [DOI] [PubMed] [Google Scholar]
  • 109.Liu Z, Liu X, Ramakrishna S. Surface engineering of biomaterials in orthopedic and dental implants: strategies to improve osteointegration, bacteriostatic and bactericidal activities. Biotechnol J. 2021;16(7):2000116. [DOI] [PubMed] [Google Scholar]
  • 110.Mishra A, et al. Current strategies in tailoring methods for engineered exosomes and future avenues in biomedical applications. J Mater Chem B. 2021;9(32):6281–309. [DOI] [PubMed] [Google Scholar]
  • 111.Huang T, et al. Application and development of electrospun nanofiber scaffolds for bone tissue engineering. ACS Biomaterials Sci Eng. 2024;10(7):4114–44. [DOI] [PubMed] [Google Scholar]
  • 112.Li Q, Chang B, Dong H, Liu X. Functional microspheres for tissue regeneration. Bioactive Mater. 2023;25:485–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Sartori M, et al. Nanostructure and biomimetics orchestrate mesenchymal stromal cell differentiation: an in vitro bioactivity study on new coatings for orthopedic applications. Mater Sci Engineering: C. 2021;123:112031. [DOI] [PubMed] [Google Scholar]
  • 114.Tan G, et al. Photo-crosslinkable hydrogels for 3d Bioprinting in the repair of osteochondral defects: A review of present applications and future perspectives. Micromachines. 2022;13(7):1038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Mamidi N, et al. Recent advances in designing fibrous biomaterials for the domain of biomedical, clinical, and environmental applications. ACS Biomaterials Sci Eng. 2022;8(9):3690–716. [DOI] [PubMed] [Google Scholar]
  • 116.Najafi H, et al. Recent advances in design and applications of biomimetic self-assembled peptide hydrogels for hard tissue regeneration. Bio-design Manuf. 2021;4:735–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Lemos R, Maia FR, Reis RL, Oliveira JM. Engineering of extracellular matrix-like biomaterials at nano‐and macroscale toward fabrication of hierarchical scaffolds for bone tissue engineering. Adv NanoBiomed Res. 2022;2(2):2100116. [Google Scholar]
  • 118.Cardoso BD, Castanheira EM, Lanceros-Méndez S, Cardoso VF. Recent advances on cell culture platforms for in vitro drug screening and cell therapies: from conventional to microfluidic strategies. Adv Healthc Mater. 2023;12(18):2202936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Shan Y, et al. Pharmacokinetic characteristics of mesenchymal stem cells in translational challenges. Signal Transduct Target Therapy. 2024;9(1):242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Matsuzaka Y, Yashiro R. Current strategies and therapeutic applications of mesenchymal stem Cell-Based drug delivery. Pharmaceuticals. 2024;17(6):707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bagno LL, Salerno AG, Balkan W, Hare JM. Mechanism of action of mesenchymal stem cells (MSCs): impact of delivery method. Expert Opin Biol Ther. 2022;22(4):449–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Shafiq M, Ali O, Han S-B, Kim D-H. Mechanobiological strategies to enhance stem cell functionality for regenerative medicine and tissue engineering. Front Cell Dev Biology. 2021;9:747398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Hoseini S, Hosseini E, Abessi P, Montazeri F. Paracrine secretions and immunological activities of human mesenchymal stem cells; the key regenerative factors of microenvironment. J Stem Cell Res Therapy. 2024;14(1):636. [Google Scholar]
  • 124.Jia Z, Zhang S, Li W. Harnessing stem cell-derived extracellular vesicles for the regeneration of degenerative bone conditions. Int J Nanomed, 2023: pp. 5561–78. [DOI] [PMC free article] [PubMed]
  • 125.Qi J, Wu H, Liu G. Novel strategies for Spatiotemporal and controlled BMP-2 delivery in bone tissue engineering. Cell Transplant. 2024;33:09636897241276733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Umar AK. Stem cell’s secretome delivery systems. Adv Pharm Bull. 2022;13(2):244–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Artamonov MY, Sokov EL. Intraosseous delivery of mesenchymal stem cells for the treatment of bone and hematological diseases. Curr Issues Mol Biol. 2024;46(11):12672–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Huang S, et al. Systemic and local administration of allogeneic bone marrow-derived mesenchymal stem cells promotes fracture healing in rats. Cell Transplant. 2015;24(12):2643–55. [DOI] [PubMed] [Google Scholar]
  • 129.Fu J, et al. Systemic therapy of MSCs in bone regeneration: a systematic review and meta-analysis. Stem Cell Res Ther. 2021;12(1):377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Mody H, Kowthavarapu VK, Betts A. Recent Advances in PK/PD and Quantitative Systems Pharmacology (QSP) Models for Biopharmaceuticals. Biopharmaceutical Informatics, 2025: pp. 307–343.
  • 131.Murias-Closas A et al. Computational modelling of CAR T-cell therapy: from cellular kinetics to patient-level predictions. EBioMedicine, 2025. 113. [DOI] [PMC free article] [PubMed]
  • 132.Joly J, et al. Clinical translation. Tissue engineering. Elsevier; 2023. pp. 717–46.
  • 133.Wei J et al. Significance and considerations of Establishing standardized critical values for critical size defects in animal models of bone tissue regeneration. Heliyon, 2024. 10(13). [DOI] [PMC free article] [PubMed]
  • 134.Yi H, Wang Y, Liang Q, Mao X. Preclinical and clinical amelioration of bone fractures with mesenchymal stromal cells: a systematic review and meta-analysis. Cell Transplant. 2022;31:09636897211051743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Jiang Y, et al. Advances in mesenchymal stem cell transplantation for the treatment of osteoporosis. Cell Prolif. 2021;54(1):e12956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Huang S, et al. Systemic administration of allogeneic mesenchymal stem cells does not halt osteoporotic bone loss in ovariectomized rats. PLoS ONE. 2016;11(10):e0163131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Laguna E, et al. Effects of systemic or local administration of mesenchymal stem cells from patients with osteoporosis or osteoarthritis on femoral fracture healing in a mouse model. Biomolecules. 2022;12(5):722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Chow SK-H, et al. The advantages and shortcomings of stem cell therapy for enhanced bone healing. Tissue Eng Part C: Methods. 2024;30(10):415–30. [DOI] [PubMed] [Google Scholar]
  • 139.Wang Z, et al. Translational challenges and prospective solutions in the implementation of biomimetic delivery systems. Pharmaceutics. 2023;15(11):2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ding S, et al. Synthetic and biogenic materials for oral delivery of biologics: from bench to bedside. Chemical Reviews; 2025. pp. 1966–78. [DOI] [PubMed]
  • 141.Gupta DK, et al. Ensuring safety and efficacy in combination products: regulatory challenges and best practices. Front Med Technol. 2024;6:1377443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Røsland GV, et al. Long-term cultures of bone marrow–derived human mesenchymal stem cells frequently undergo spontaneous malignant transformation. Cancer Res. 2009;69(13):5331–9. [DOI] [PubMed] [Google Scholar]
  • 143.Wang Y, et al. Long-term cultured mesenchymal stem cells frequently develop genomic mutations but do not undergo malignant transformation. Cell Death Dis. 2013;4(12):e950–950. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Momin N, Vela EG, Zaidi HA, Quiñones-Hinojosa A. The oncogenic potential of mesenchymal stem cells in the treatment of cancer: directions for future research. Curr Immunol Rev. 2010;6(2):137–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Huerta CT, Ortiz YY, Liu Z-J, Velazquez OC. Methods and limitations of augmenting mesenchymal stem cells for therapeutic applications. Adv Wound Care. 2023;12(8):467–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Haworth R, Sharpe M. Accept or reject: the role of immune tolerance in the development of stem cell therapies and possible future approaches. Toxicol Pathol. 2021;49(7):1308–16. [DOI] [PubMed] [Google Scholar]
  • 147.Moeinabadi-Bidgoli K, et al. Genetic modification and preconditioning strategies to enhance functionality of mesenchymal stromal cells: a clinical perspective. Expert Opin Biol Ther. 2023;23(6):461–78. [DOI] [PubMed] [Google Scholar]
  • 148.Liu S, et al. Advancements in scaffold for treating ligament injuries; in vitro evaluation. Biotechnol J. 2024;19(1):2300251. [DOI] [PubMed] [Google Scholar]
  • 149.Giebel B, Lim SK. Overcoming challenges in MSC-sEV therapeutics: insights and advances after a decade of research. Cytotherapy, 2025. [DOI] [PubMed]
  • 150.Ulpiano C, da Silva CL, Monteiro GA. Mesenchymal stromal cells (MSCs): a promising tool for cell-based angiogenic therapy. Curr Gene Ther. 2021;21(5):382–405. [DOI] [PubMed] [Google Scholar]
  • 151.Zhang C, et al. Eradication of specific donor-dependent variations of mesenchymal stem cells in Immunomodulation to enhance therapeutic values. Cell Death Dis. 2021;12(4):357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Tian B, et al. Macromolecule-based hydrogels nanoarchitectonics with mesenchymal stem cells for regenerative medicine: A review. Int J Biol Macromol. 2023;243:125161. [DOI] [PubMed] [Google Scholar]
  • 153.Jayaraman P, Lim R, Ng J, Vemuri MC. Acceleration of translational mesenchymal stromal cell therapy through consistent quality GMP manufacturing. Front Cell Dev Biology. 2021;9:648472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.De Pieri A, Rochev Y, Zeugolis DI. Scaffold-free cell-based tissue engineering therapies: advances, shortfalls and forecast. NPJ Regenerative Med. 2021;6(1):18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Lu Y, Mai Z, Cui L, Zhao X. Engineering exosomes and biomaterial-assisted exosomes as therapeutic carriers for bone regeneration. Stem Cell Res Ther. 2023;14(1):55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Zheng D, et al. Advances in extracellular vesicle functionalization strategies for tissue regeneration. Bioactive Mater. 2023;25:500–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Mohammadpour S, Mokhtarzade A, Jafari-Ramiani A, Solati-Hashjin M. Tailoring surface properties of Poly (caprolactone)/Hydroxyapatite scaffolds through aminolysis and multi-walled carbon nanotube coating for bone tissue engineering. Mater Today Commun. 2024;39:109056. [Google Scholar]
  • 158.Bieback K, Brinkmann I. Mesenchymal stromal cells from human perinatal tissues: from biology to cell therapy. World J Stem Cells. 2010;2(4):81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Dalle Carbonare L, et al. The bone microenvironment: new insights into the role of stem cells and cell communication in bone regeneration. Stem Cell Res Ther. 2025;16(1):169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Reviews Endocr Metabolic Disorders. 2024;25(2):339–67. [DOI] [PubMed] [Google Scholar]
  • 161.Shinwari ZK, Tanveer F, Khalil AT. Ethical issues regarding CRISPR mediated genome editing. Curr Issues Mol Biol. 2018;26(1):103–10. [DOI] [PubMed] [Google Scholar]
  • 162.Rony R. Transcriptional characterization of osteogenic and adipogenic differentiation of human bone marrow derived mesenchymal stem cells in 2D and 3D peptide hydrogel culture system. Wright State University; 2018.
  • 163.Wu Y, Xie L. AI-driven multi-omics integration for multi-scale predictive modeling of causal genotype-environment-phenotype relationships. arXiv preprint arXiv:2407.06405, 2024. [DOI] [PMC free article] [PubMed]
  • 164.Raheem A et al. Smart biomaterials in healthcare: breakthroughs in tissue engineering, immunomodulation, patient-specific therapies, and biosensor applications. Appl Phys Reviews, 2025. 12(1).
  • 165.Lee SJ, Jeong W, Atala A. 3D Bioprinting for engineered tissue constructs and Patient-Specific models: current progress and prospects in clinical applications. Adv Mater. 2024;36(49):2408032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Saliev T, Singh PB. From Bench to Bedside: Translating Cellular Rejuvenation Therapies into Clinical Applications. Cells, 2024. 13(24): p. 2052. [DOI] [PMC free article] [PubMed]
  • 167.Lloyd-Williams H, Hughes DA. A systematic review of economic evaluations of advanced therapy medicinal products. Br J Clin Pharmacol. 2021;87(6):2428–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Zhidu S, Ying T, Rui J, Chao Z. Translational potential of mesenchymal stem cells in regenerative therapies for human diseases: challenges and opportunities. Stem Cell Res Ther. 2024;15(1):266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Olesti E, et al. Academic challenges on advanced therapy medicinal products’ development: A regulatory perspective. Cytotherapy. 2024;26(3):221–30. [DOI] [PubMed] [Google Scholar]
  • 170.Kamisetti RR. Strategies Followed for the Assessment of the Advanced Therapies by European Medicines Agency. 2023.
  • 171.Arato T, Nomura K. Cell and gene therapy approvals in Japan and the need for international harmonization. Nat Biotechnol. 2024;42(1):13–7. [DOI] [PubMed] [Google Scholar]
  • 172.Capelli C, et al. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front Immunol. 2023;14:1186224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Wilson AJ, Rand E, Webster AJ, Genever PG. Characterisation of mesenchymal stromal cells in clinical trial reports: analysis of published descriptors. Stem Cell Res Ther. 2021;12(1):360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Sawarkar S, Bapat A. Global regulatory frameworks and quality standards for stem cells therapy and regenerative Medicines, in Stem cell production: Processes, practices and regulations. Springer; 2022. pp. 69–111.
  • 175.Maumus M, et al. Mesenchymal stem cell-derived extracellular vesicles: opportunities and challenges for clinical translation. Front Bioeng Biotechnol. 2020;8:997. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Stem Cell Research & Therapy are provided here courtesy of BMC

RESOURCES