Next Article in Journal
Design, Characterization, and Preliminary Assessment of a Two-Degree-of-Freedom Powered Ankle–Foot Prosthesis
Previous Article in Journal
Biomimetic Design of a New Semi-Rigid Spatial Mesh Antenna Reflector
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Vasculature and Angiogenic Strategies in Bone Regeneration

by
Hye-Jeong Jang
and
Jeong-Kee Yoon
*
Department of Systems Biotechnology, Chung-Ang University, Anseong-si 17546, Gyeonggi-do, Republic of Korea
*
Author to whom correspondence should be addressed.
Biomimetics 2024, 9(2), 75; https://doi.org/10.3390/biomimetics9020075
Submission received: 6 January 2024 / Revised: 18 January 2024 / Accepted: 22 January 2024 / Published: 26 January 2024
(This article belongs to the Section Biomimetics of Materials and Structures)

Abstract

:
Bone regeneration is a complex process that involves various growth factors, cell types, and extracellular matrix components. A crucial aspect of this process is the formation of a vascular network, which provides essential nutrients and oxygen and promotes osteogenesis by interacting with bone tissue. This review provides a comprehensive discussion of the critical role of vasculature in bone regeneration and the applications of angiogenic strategies, from conventional to cutting-edge methodologies. Recent research has shifted towards innovative bone tissue engineering strategies that integrate vascularized bone complexes, recognizing the significant role of vasculature in bone regeneration. The article begins by examining the role of angiogenesis in bone regeneration. It then introduces various in vitro and in vivo applications that have achieved accelerated bone regeneration through angiogenesis to highlight recent advances in bone tissue engineering. This review also identifies remaining challenges and outlines future directions for research in vascularized bone regeneration.

Graphical Abstract

1. Introduction

Bones play a crucial role in the human body as they provide structural support, protect vital organs, and store essential minerals [1,2]. The process of bone regeneration involves the regrowth of osteocytes, as well as other functional cells such as endothelial cells (ECs) and neurons for revascularization and innervation, restoring the bone’s original shape and function [3,4,5]. These regenerative cascades are activated in response to bone defects, caused by traumatic injury, bone tumors, cysts, or infections, when the inherent adaptability of bone allows it to undergo spontaneous remodeling [6,7]. However, critical-size bone defects, which are clinically defined as defects with a length exceeding 1–2 cm in length and involving a loss of more than 50% of bone circumference, can cause non-healing fractures and reduced mobility, posing a significant challenge in clinical settings [8,9]. Inappropriate treatment of critical-size bone defects can exacerbate the condition, potentially causing malunion (abnormal or misaligned healing of the fractured bone ends) or nonunion (failure of the fractured bone ends to heal) [10,11,12]. These complications can result in persistent pain, deformity, impaired joint function, and, in some cases, limb shortening, adversely impacting the quality of life [13,14,15].
Conventional bone tissue engineering centered on autologous bone grafts is the gold standard for replacing fractured bone with the highest potential for healing [16,17]. However, the surgery required for bone harvest is repeatedly reported to result in sequential complications [18,19]. As an alternative, allogenic bone grafts offer improved safety, size and shape diversity, and reduced donor-site morbidity, as well as reducing the time required for graft preparation [20,21,22]. However, inadequate supply, risks of immune responses due to allograft rejection, and a decrease in osteoinductivity and osteoconductivity during storage, resulting from alterations in mechanical and biological properties, have been reported [23,24,25]. These disadvantages in autologous and allogeneic bone grafts highlight the need for artificial bone substitutes that closely resemble natural bone, particularly in terms of mechanical and biological properties, which can significantly impact the success rate of implants.
Artificial bone substitutes are composed of a diversity of various biomaterials, such as metals or bioceramics with precisely controlled structural properties, to regulate the biomechanical regenerative mechanism and enhance the osteoinductivity, osteoconductivity, and osseointegrity. Since bone is a highly vascularized tissue [26], a recent approach for bone regeneration involves treating bone defects by inducing angiogenesis in the bone complexes. Promoting the formation of blood vessels in new bone is essential for achieving high levels of biostability and biosafety, as it allows for the establishment of connections between newly generated vascular structures at the defect sites and the pre-existing blood vessels of the host, eventually nourishing the new bone and regulating immune responses [27]. Moreover, the angiogenic factors, including the vascular cells (e.g., ECs), growth factors (e.g., vascular endothelial growth factor; VEGF), or the hypoxic condition, are known to directly contribute to osteogenesis by stimulating mesenchymal stem cells (MSCs) and osteoblasts [28,29,30].
Here, we review the recent strategies for inducing angiogenesis to promote bone regeneration. Advances in tissue engineering technologies have enabled the coupling of osteogenesis and angiogenesis during bone regeneration through the use of mechanical and biological factors, including their mechanistic studies. From in vitro to in vivo applications, our review emphasizes the critical role of angiogenesis as a new strategy in designing artificial bone substitutes for successful bone regeneration, providing insights that span from controlled in vitro environments to the promising landscape of in vivo interventions.

2. Angiogenesis in Bone Regeneration

Bone regeneration occurs spontaneously in response to bone injury, involving multiple factors such as growth factors, osteoblasts, osteoclasts, inflammatory cytokines, and the extracellular matrix [31]. Since the bone microenvironment is a sophisticated system involving physiological, chemical, and physical factors, bone regeneration strategies utilize various aspects such as scaffolding [32], cell therapy [33], growth factor delivery [34], cytokine incorporation [35], immunomodulation [36], and angiogenesis [37] to target the microenvironment. The process of bone regeneration is a dynamic and organized procedure that typically consists of three phases: the inflammatory phase, the bone production phase, and the bone remodeling phase [38,39,40]. When a bone defect occurs, an acute inflammatory response follows bleeding, resulting in hematoma formation with a hypoxic, low pH, calcium-rich microenvironment [41,42]. This recruits inflammatory cells to the defect site and initiates the pro- and anti-inflammatory cascades, which further stimulate the angiogenic physiology [43]. During the bone production phase, the hematoma is replaced by soft tissue such as fibrous tissue or cartilage tissue (e.g., soft callus), which gradually hardens over several weeks [44,45]. MSCs differentiate into osteoprogenitor cells, and vascularization occurs in this phase [46]. Finally, the bone remodeling phase then restores the structural and mechanical properties, achieving a balance between osteoblastic and osteoclastic activity.
Bone fractures can cause damage to blood vessels and bleeding, leading to a hypoxic condition that can induce inflammation and hinder the regenerative process [47]. Therefore, angiogenesis, which involves endothelial sprouting from pre-existing blood vessels, is crucial for enhancing the regenerative capability during bone regeneration by salvaging the new bone tissue from hypoxia and supplying oxygen and nutrients through a consistent perfusion [48]. Moreover, a mechanism known as the coupling of osteogenesis and angiogenesis suggests that the neovasculature also stimulates the osteoprogenitor cells via angiogenic paracrine signaling, promoting their proliferation or differentiation [49,50].
The precise mechanisms of bone vasculature formation and the osteogenic–angiogenic coupling still remain poorly understood. Several studies suggest that type H vessels, which express high levels of Endomucin and CD31 on the endothelium, modulate osteogenesis during the bone production to bone remodeling phase (Figure 1) [51,52]. Type H vessels are surrounded by osteoprogenitor cells and guide their proliferation and differentiation into osteocytes [53]. For instance, in endochondral ossification, Type H ECs secrete a high level of VEGF into the cartilage to induce angiogenesis, recruiting chondroclasts and osteoblasts to remodel the soft cartilage into hard bone tissue [54]. In addition, platelet-derived growth factor (PDGF) secreted from osteoclasts is known to recruit such ECs to induce bone formation [55]. In intramembranous ossification, a group of MSCs (i.e., mesenchymal condensation) secrete a high level of VEGF, attracting endothelial cells [56,57]. The ECs stimulate the MSCs to initiate osteogenic differentiation, leading to ossification and mineralization [58,59].

3. Conventional Strategies for Vascularized Bone Tissue Engineering

Current bone tissue engineering methods primarily rely on the use of autologous or allogenic bone grafts. These grafts exhibit good biomechanical properties, including an ideal elastic modulus similar to natural bone, as well as good osteoconductivity and osteoinductivity, making them suitable for replacing the bone defect. However, limitations such as poor availability and donor site morbidity in autologous bone grafts, as well as graft rejection in allogeneic bone grafts, may lead to implant failure and cause significant complications [59,60,61]. Therefore, the development of artificial bone substitutes using synthetic materials has emerged as a viable alternative. Metals such as titanium, stainless steel, or chromium alloys have been widely used due to their high mechanical strength, wear resistance, and durability [62,63]. However, the non-biodegradable nature of metal implants may require secondary surgery for removal, and their mechanical properties may not match those of the surrounding bone, leading to stress shielding and subsequent bone resorption [64,65,66,67]. Bioceramics, such as hydroxyapatite, tricalcium phosphate, or their mixture, known as biphasic calcium phosphate, are biocompatible and support bone ingrowth due to their osteoconductive properties, as they consist of a high amount of calcium and phosphate ions [68,69,70,71,72,73]. However, the brittleness and lower mechanical strength of ceramic implants compared to metals limit their use to non-load-bearing sites [74,75].
The material properties of artificial bone substitutes are recently being improved by synthesizing optimal materials using biodegradable polymers, or various composites of metal, ceramic, and polymeric materials with innovative techniques. However, the biological response, such as angiogenesis induction, has not been resolved. Conventional tissue engineering strategies have enabled vascularization within the implanted scaffolds by creating interconnected micro- or macroporous structures. When designing porous materials, the selection of an optimal pore size range is important to control the biomolecule diffusion, cell infiltration, and the behavior of the adhered cells. For example, smaller pores have been shown to induce cell differentiation, whereas larger pores induce cell proliferation [76,77]. In contrast, smaller pores may lead to a hypoxic condition within the implant due to low diffusion efficiency and stimulate chondrogenesis rather than osteogenesis [78,79].
In terms of vascularization, the natural hypoxic environment in the bone defect will stimulate MSCs to secrete proangiogenic factors that promote angiogenesis [80]. Typically, macropores with a size larger than 100 μm are preferred over micropores with a size smaller than 20 μm, since larger pores better induce the infiltration of ECs, resulting in a higher osseointegration of the implant [81,82]. However, the need for micropores should also be considered to modify the surface properties, such as capillarity, which can lead to a more homogeneous cell distribution and higher bone volume fraction. Therefore, scaffolds with a variety of pore sizes are currently being developed. Regarding porosity, a higher porosity level (>80%) facilitates better cell infiltration and higher interconnectivity but decreased mechanical properties, leading to material failure under high stress [78,83]. Overall, porous scaffolds can improve osseointegration through vascular infiltration [84]. However, spontaneous angiogenesis may not be fast enough to form a dense vascular network in a bone defect larger than a critical-size defect. Therefore, it is important to consider vascular engineering in addition to osteogenesis. This can be achieved by utilizing proangiogenic cells or growth factors, or through advanced material design (Table 1).

4. Angiogenic Strategies for Bone Regeneration

4.1. Angiogenic Factor Delivery

The administration of angiogenic growth factors is typically used in bone regeneration to promote the formation of new bone tissue and blood vessels. Commonly used growth factors, such as VEGF and bone morphogenetic protein 2 (BMP-2), play a critical role in stimulating angiogenesis and osteogenesis [29,113]. By delivering these growth factors directly to the site of bone defects, it can provide necessary signals to promote bone and blood vessel formation, ultimately aiding in the successful regeneration of bone tissue. In one example, the simultaneous release effects of VEGF and BMP-2 on bone regeneration in rat critical-size defect models were evaluated [85]. The results showed that dual administration of VEGF and BMP-2 significantly increased bone formation and bone bridging compared to BMP-2 alone or no growth factor at both 4 and 12 weeks [85]. Additionally, the dual group had a higher percentage of blood vessel volume within the defect at 4 weeks compared to the other groups in microCT imaging results, indicating enhanced angiogenesis [85]. These findings suggest that delivering an angiogenic and an osteogenic factor simultaneously provides a synergistic response that would promote bone regeneration in critical-size defects [85]. Another study investigated the potential of a VEGF-coated scaffold to induce vascularization for bone regeneration [37]. VEGFs were released from the scaffolds in a sustained manner for more than 14 days, and microCT imaging results at 12 weeks showed near-complete bridging of the bone defect by the newly formed mineralized tissue in the coated VEGF-releasing scaffolds [37]. A quantitative analysis showed an increase in bone volume fraction and bone mineral density in the coated VEGF-releasing scaffolds compared to the control scaffolds [37]. In conclusion, the study demonstrated the angiogenic capacity of growth factor-coated scaffolds and their potential to induce vascularization, which is critical for bone formation and healing [37]. However, the osteogenic effect was not as significant, probably due to the low concentration of scaffold material that was used in the study [37]. A different study demonstrated the efficacy of administering angiogenin in vascularized bone regeneration [86]. In vitro cell culture studies showed that angiogenin-loaded scaffolds enhanced cell proliferation and adhesion, indicating that angiogenin stimulates EC proliferation and angiogenesis [86]. In addition, the study found that angiogenin enhanced the adhesion of ECs to the scaffold, suggesting enhanced bone formation through angiogenesis [86]. The in vivo rabbit calvarial defect model demonstrated that angiogenin-containing scaffolds induced an increased number of blood vessels with increasing angiogenin concentrations, and also showed an extensive neo-bone structure around the defect area that fused with the host’s bone 8 weeks post-implantation (Figure 2A) [86]. Collectively, the results indicate that angiogenin promotes angiogenesis and bone formation, suggesting that it is a potential growth factor for bone tissue engineering applications [86]. Another example used a polyhedral oligomeric silsesquioxane (POSS)-modified gelatin hydrogel to promote vascularization in bone regeneration (Figure 3A) [87]. Compared to 0% POSS hydrogels, the 3% POSS hydrogel coupled with VEGF/BMP-2 demonstrated a significantly higher degree of vascularization in microCT imaging and CD31 immunohistochemical staining [87]. In addition, the 3% POSS hydrogel coupled with VEGF/BMP-2 showed enhanced bone regeneration in the in vivo rat calvarial defect model, with complete healing of the defect and no gaps observed, as well as the largest number of newly formed blood vessels, indicating effective bone repair and angiogenesis effects (Figure 2B) [87]. Morphometric analysis results showed that the 3% POSS hydrogel coupled with VEGF/BMP-2 also had the highest bone mineral density, bone volume to total volume, and trabecular number [87]. This suggests that the POSS-modified hydrogel is highly effective in promoting vascularized bone repair [87]. In a different study, whitlockite (WH) nanoparticles and VEGF were utilized to show how the combined microenvironment of these two substances synergistically enhanced the osteogenesis and angiogenesis of MSCs [88]. As a result, enhanced osteogenic differentiation and vascularization were observed in the WH and VEGF scaffold in vitro [88]. In a rat calvarial defect model, the WH and VEGF synergistic group showed the most significant effect in promoting bone regeneration and blood vessel formation at the defect site [88]. In another study, 3D-printed scaffolds with a hollow tube structure and bioactive ions, referred to as BRT-H scaffolds, were fabricated using a coaxial 3D-printing technique (Figure 3B,C) [89]. The hollow tube microstructure and the release of bioactive ions synergistically enhanced angiogenesis and osteogenesis, as evidenced by the stimulation of EC migration and blood vessel formation [89]. The BRT-H scaffolds exhibited high compressive strength and facilitated blood vessel ingrowth and an enhanced delivery of stem cells and growth factors [89]. In a rabbit radial defect model, the scaffolds promoted early angiogenesis and subsequent bone regeneration compared to the control group, evidenced by the microCT image of enhanced blood vessel formation (Figure 2C) [89]. The study showed that the coaxial printing of hollow-tube-structured BRT-H scaffolds was effective in promoting vascularized bone regeneration in large segmental bone defects [89]. Another study developed a nanofibrous scaffold with interconnected microchannels for mimicking the bone microenvironment and facilitated the sequential release of the pro-angiogenic drug dimethyloxalylglycine [90]. In vitro results showed an increased expression of angiogenic and osteogenic genes, increased alkaline phosphatase (ALP) activity, and increased mineral deposition, indicating the potential of the scaffold to stimulate both angiogenesis and osteogenesis [90]. In vivo, the scaffold significantly promoted vascularization and bone regeneration in a rat calvarial defect model, suggesting that the interconnected microchannel structures successfully supported an effective drug delivery system within the scaffold for enhanced vascularization and bone regeneration (Figure 2D) [90]. A different example developed a 3D-bioprinted scaffold composed of silk and hydroxyapatite loaded with BMP-2, VEGF, and NGF to enhance angiogenesis, osteoblast differentiation, and bone regeneration [91]. The results showed that the 3D-bioprinted bone constructs were cytocompatible and osteoconductive, as evidenced by the enhanced osteogenic differentiation of MSCs. In particular, the incorporation of VEGF within the construct was effective in enhancing HUVEC migration, thereby improving the vascularization potential for bone repair and regeneration [91].

4.2. Cell Delivery

Conventional approaches to bone regeneration rely on a single cell type to promote bone formation. Yet, these methods fail to encapsulate the intricacies inherent in the complex processes of bone regeneration, where osteogenesis and angiogenesis engage in a harmonious interplay. In particular, angiogenesis is essential for osteogenesis to ensure an adequate supply of nutrients and oxygen [114]. Some cases have highlighted the significance of co-culturing different cell types, demonstrating their pivotal role in both osteogenesis and angiogenesis. Most commonly, it is known that ECs and mesenchymal stem cells (MSCs) work in concert to increase the efficiency of osteogenesis, which speeds up the vascularization process in bone regeneration. One example performed a co-transplantation of ECs and bone marrow stromal cells (BMSCs) on biodegradable polymer scaffolds to investigate whether Ecs could directly influence the osteogenic potential of BMSCs [92]. When BMSCs were co-cultured with Ecs, an early osteogenic marker (ALP expression) and late osteogenic marker (osteocalcin) greatly increased compared to the culture of BMSCs alone. In addition, the blood vessels were examined using CD31 immunostaining to identify functional vessels to determine if transplanted the ECs improved neovascularization [92]. The proportion of transplanted EC-derived vessels among the total number of vessels showed a significant increase in scaffolds co-cultured with BMSCs and ECs compared to scaffolds with BMSCs alone [92]. However, the overall number of vessels generated within the scaffolds throughout the 8-week observation period did not differ statistically significantly between these two groups [92]. Another study investigated the bone regeneration effects of incorporating ECs along with osteoblasts into the PCL-HA composite (Figure 2E) [93]. In vivo experiments showed that scaffolds seeded with ECs and osteoblasts exhibited enhanced vascularization and osteogenesis, as well as improved mechanical properties of the engineered bone tissue compared to scaffolds containing only osteoblasts (Figure 2E) [93]. Three-dimensional microCT imaging results further supported the efficacy of the EC–osteoblast co-culture group in promoting bone defect repair compared to the osteoblast-only group, highlighting the significant contribution of ECs in the repair process [93]. Overall, these results suggest that the co-culture of ECs with osteoblasts has potential for clinical use in the treatment of large bone defects [93]. A different research study used a co-culture approach between MSCs and MSC-derived ECs in a porous β-tricalcium phosphate scaffold for repairing large segmental bone defects in rabbits [94]. The results showed that the co-culture group containing ECs promoted the osteogenic effect of MSCs with improved mechanical properties and a more abundant capillary network, suggesting that it is an effective approach to enhance osteogenesis and angiogenesis in bone regeneration [94].

4.3. Gene Delivery

The gene delivery method is a pivotal component in bone regeneration, as it facilitates the targeted delivery and expression of angiogenic or osteogenic genes within the defect site. This process leads to the production of vessel-forming or bone-inducing proteins, significantly enhancing the bone-repairing process. Through the use of gene-activated matrices and other delivery platforms, sustained release and localized expression of these genes can be achieved, providing a controlled environment for bone tissue formation and vascularization. In one study, researchers developed a bioactive scaffold for gene delivery to enhance bone repair and vascularization [95]. The scaffold was designed to deliver plasmid DNA encoding for VEGF and BMP-2 using non-viral vectors [95]. The in vitro results demonstrated that the dual delivery scaffold made of polyethylenimine and nano-hydroxyapatite significantly enhanced calcium deposition and mineralization [95]. In vivo, the gene-activated scaffolds recruited and transfected host cells, as demonstrated by the presence of GFP-expressing cells in a rat calvarial defect model. This method effectively enhanced vessel formation in the bone defect region by providing a localized and sustained delivery of the angiogenic gene (VEGF) [95]. In another paper, the researchers delivered the VEGF gene to enhance angiogenesis and bone regeneration [96]. They found that VEGF gene delivery, particularly through a gene-activated matrix, effectively bridged bone gaps and increased vessel formation in a rabbit radial defect model [96]. The utilization of a collagen sponge as a delivery system for the non-viral VEGF gene application resulted in increased angiogenesis and osteoid formation, as confirmed through a radiographic evaluation, μCT scans, histology, and immunohistochemical staining for blood vessels [96]. This approach was shown to be particularly effective in promoting vascularization and bone regeneration in large segmental defects, suggesting its potential as a valuable tool for treating nonunion [96]. Another study investigated the use of angiopoietin 1 (Ang1) gene-transfected MSCs seeded onto beta-tricalcium phosphate (β-TCP) scaffolds for repairing segmental bone defects in rabbits [97]. The results showed that the experimental group receiving the Ang1-transfected MSCs exhibited increased vessel growth, faster bone union, and higher bone formation compared to the control group, resulting in successful repair of the segmental bone defect within 12 weeks [97]. The study showed that the experimental group’s expression of Ang1 significantly improved angiogenesis by promoting capillary regeneration within the porous network [97]. In a different study, the proangiogenic gene miR-21 and the pro-osteogenic gene miR-5106 were delivered to the site of the bone defect using zeolitic imidazolate framework 8 as a non-viral vector for promoting angiogenesis and bone regeneration [98]. The in vivo experiments, using a rat calvarial defect model, demonstrated significant improvements in blood vessel formation, bone mineral density, trabecular number, and thickness in the experimental group [98]. The success of the method in enhancing angiogenesis for bone regeneration was attributed to the efficient delivery and release of therapeutic miRNAs within cells, facilitated by the miRNA delivery nanocomposites [98]. This was demonstrated through an in vitro scratch assay and tube formation assay, which showed the enhanced angiogenic ability of the gene-incorporated nanocomposites [98].

4.4. Perfusable 3D Vascular Network

Mimicking a perfusable 3D vascular network is crucial in osteogenesis and angiogenesis since it closely replicates the natural environment of bone tissue, which is essential for transporting nutrients, oxygen, and waste removal, as well as providing mechanical cues to cells [48]. A perfusable 3D vascular network within a scaffold ensures that these critical processes can occur throughout the entire construct, thereby supporting the growth of viable tissue constructs. In addition, the dynamic fluid flow within the vascular network can simulate the physiological shear stress that cells experience in the in vivo environment. In one study, a nanocoating system was integrated with a biomimetic, 3D-bioprinted, perfused microstructure to create a vascularized bone complex [99]. This construct was designed to mimic the hierarchical architecture of native bone and to provide a dynamic environment for cell growth, with a focus on regulating angiogenesis and osteogenesis through a matrix metalloprotease 2-responsive mechanism [99]. The perfused microstructure system was critical in simulating in vivo conditions and allowing for the dynamic culture of cells [99]. The results demonstrated that the nanocoating-modified 3D-bioprinted scaffolds had great bioactivity and the potential to form a vascularized bone complex [99]. In a different study, 3D-bioprinted scaffolds with perfusable vascular channels were created to enhance bone regeneration by promoting osteogenesis and angiogenesis [100]. The bioprinted scaffolds significantly promoted the osteogenic differentiation of MSCs, and the presence of human umbilical vein endothelial cells (HUVECs) within the scaffolds resulted in an enhanced formation of vascular networks, particularly under dynamic culture conditions that mimic the in vivo environment [100]. The use of GelMA hydrogel bioink was also instrumental in forming these vascular networks and providing the necessary mechanical robustness within the scaffold [100]. This approach effectively enhanced angiogenesis in bone regeneration by creating a conducive environment for vascularization through the strategic use of bioink formulations, co-culture systems, and 3D-printing techniques together [100].

4.5. Hydrogels Inducing Angiogenesis

Hydrogel-based vascularized bone models mimic the intricate function of the natural bone extracellular matrix (ECM). These hydrogel platforms can be precisely patterned or modified to encapsulate different cell types and loaded with different particles to promote bone regeneration. The ability to engineer constructs with spatially organized vascular networks and osteogenic niches hold promise for the treatment of complex bone defects, offering a more tailored and potentially effective healing strategy compared to traditional bone repair methods. In one study, researchers engineered a hydrogel-based vascularized bone model that demonstrated the effective formation of mineralized areas surrounded by vasculature, crucial for bone tissue engineering [101]. The existence of osteogenic and angiogenic niches within the model resulted in enhanced bone formation, with the angiogenic niche significantly improving this regeneration [101]. The hydrogel platform, which included endothelial and mesenchymal stem cells, exhibited self-assembly into cord-like structures, indicating the potential for these engineered tissues to connect with the host’s vasculature upon implantation and support cell survival and function [101]. In another study, a hydrogel-microsphere system was developed for the delivery of integrated biological signal peptides to enhance bone repair [102]. The synthesized composite microspheres, particularly the gelatin methacrylate (GelMA)-S-B microspheres, demonstrated good biocompatibility, sustained osteogenic capacity, and preserved vascular properties [102]. When tested in a rat femoral defect model, the microspheres effectively promoted bone formation, with histologic evaluations confirming increased new bone formation in the GelMA-S-B group compared to controls [102]. These results indicate that the hydrogel-microsphere system is an effective platform for delivering biological signals that can significantly enhance bone regeneration, with promising potential for clinical applications in bone repair and tissue engineering [102]. In another example, they developed a bilayer hydrogel designed to achieve vascularized bone regeneration in bone defects [103]. The hydrogel platform achieved vascularization within the bone structure by simultaneously incorporating magnesium-modified black phosphorus nanosheets to promote angiogenesis and β-tricalcium phosphate nanocrystals to promote osteogenic differentiation [103]. The magnesium ions in the nanosheets promoted angiogenesis and greatly enhanced the in vitro adhesion and proliferation of ECs [103]. The in vivo results indicated that the hydrogel scaffold facilitated bone healing in rat calvarial defect models based on increased bone mineral density and bone score values, with a complete coverage of the defect site with newly formed bone at 12 weeks after hydrogel bilayer transplantation [103]. These results suggest that the hydrogel bilayer scaffold is effective in promoting bone regeneration, with the added benefit of promoting angiogenesis [103]. In a different study, a biomimetic periosteum consisting of a diselenide-containing gelatin and calcium alginate (Gel-Se/Alg-Ca) hydrogel was developed for effective bone regeneration [104]. The biomimetic periosteum was effective in promoting bone regeneration through the continuous release of nitric oxide (NO), which activated the NO-cGMP signaling pathway, thereby enhancing both osteogenesis and angiogenesis [104]. The in vivo results in a rat calvarial defect model showed that the Gel-Se/Alg-Ca group exhibited the most efficient bone tissue repair effect, with a higher bone volume fraction and bone mineral density compared to other groups [104].

4.6. Extracellular Vesicle (EV) Delivery

Extracellular vesicles (EVs) are released by living cells, including mammalian cells and microorganisms, and consist of membranous and intracellular components. EVs play a critical role in intercellular communication, making them important in regenerative medicine applications. In contrast to the challenges of common cell delivery methods, such as unpredictable cell fates and entrapment in pulmonary capillaries, exosomes, although non-living, carry biological information, evade entrapment, and can be cryopreserved for efficient storage and later use. In one study, bone marrow-derived EVs were preconditioned under hypoxia to enhance bone regeneration [105]. These EVs were effectively delivered using an injectable bioactive polypeptide-based hydrogel, which not only demonstrated good biocompatibility and a sustained release of EVs, but also resulted in excellent bone regeneration in a rat calvarial defect model [105]. The study also highlighted that hypoxic EVs were superior at promoting vascularization, osteoblast proliferation, migration, and differentiation compared to EVs from normoxic conditions [105]. Another study used exosomes derived from the stem cells of human exfoliated deciduous teeth (SHEDs) to investigate their potential in bone regeneration (Figure 3D) [106]. The SHEDs preconditioned under hypoxic conditions produced exosomes with an enhanced ability to promote angiogenesis and osteogenesis [106]. These SHED-derived hypoxic exosomes were then successfully loaded onto poly(lactic-co-glycolic acid) microspheres coated with polydopamine to facilitate sustained release [106]. In vivo experiments using a rat calvarial defect model demonstrated that the microsphere delivery of exosomes significantly promoted new bone formation and vascularization [106]. High-throughput RNA sequencing indicated that these preconditioned exosomes could regulate pathways related to bone tissue regeneration, and the released exosomes maintained their biological functions, effectively enhancing angiogenesis and bone regeneration [106].

4.7. Three-Dimensional-Bioprinted Models

Three-dimensional-bioprinted vascularized bone models enable the precise fabrication of bone tissue with integrated vascular networks through advanced 3D-bioprinting techniques that allow for the simultaneous deposition of cells and biomaterials in a spatially controlled manner. By incorporating bioactive factors such as growth factors into the constructs, these models can also provide the targeted stimulation of both osteogenesis and angiogenesis at specific regions within the tissue, and the combination of these factors within the 3D-bioprinted constructs can lead to improved therapeutic outcomes for bone regeneration. In addition, the dynamic culture of these 3D-bioprinted constructs allows for better nutrient exchange, which is beneficial for the maturation of the vascular network and overall bone regeneration [115,116]. In one study, 3D-bioprinting technology was used to create vascularized bone constructs using GelMA hydrogel, HUVECs, and MSCs (Figure 3E) [107]. The incorporation of MSCs was found to stabilize the endothelial tubes, resulting in improved cell adhesion and proliferation [107]. The bioprinted construct showed an increased secretion of VEGFs and collagen I content, along with enhanced osteogenic differentiation due to the presence of bioactive peptides within the construct [107]. Using a custom-designed flow bioreactor system, the authors were able to dynamically culture the constructs, resulting in extensive capillary networks and increased osteogenic and angiogenic differentiation [107]. The results highlight the potential of this novel 3D-bioprinting approach, combined with regional bioactive peptide immobilization, to produce complex, vascularized bone constructs with significant therapeutic efficacy for bone regeneration [107]. Another example presented a 3D-bioprinted scaffold loaded with deferoxamine (DFO) liposomes to enhance bone regeneration through osteogenic and angiogenic properties [108]. DFO is known to promote both osteogenesis and angiogenesis by inducing the expression of hypoxia-inducible factor 1-alpha and VEGF [108]. The scaffold promoted osteogenesis in BMSCs and upregulated osteogenic-related gene and protein expression [108]. In vivo experiments using a rat femoral defect model demonstrated new bone growth and enhanced angiogenesis due to the controlled release of DFO [108]. This release mechanism stimulated the early-stage internal vascularization and maturation of the vascular network, which is critical for coupling angiogenesis with osteogenesis and ultimately promoting bone regeneration [108]. In a different research study, they developed and evaluated gelatin-based bioinks for an extrusion-based method to create bioprinted vascularized bone equivalents [109]. They modified gelatin hydrogel formulation to enhance 3D vascularization and successfully bioprinted co-culture hydrogels containing human dermal microvascular endothelial cells (HDMECs) and adipose-derived stem cells (ASCs) [109]. The resulting hydrogels demonstrated an enhanced ability to form capillary-mimicking structures and supported the deposition of bone-specific proteins [109]. This method effectively enhanced angiogenesis, which is critical for bone regeneration, by providing a conducive environment for angiogenesis and the development of vasculature within the engineered tissue constructs [109].

4.8. Other Synthetic Models

Among various other synthetic models, microfluidic vascularized bone models are recently emerging in vitro platforms that integrate microfluidic technology with 3D tissue engineering to create a controlled environment for studying the interactions between bone cells and blood vessels. By incorporating materials such as hydroxyapatite into the ECM materials, these models can mimic the mineralized matrix of natural bone while also allowing paracrine communication between cells [117]. While most bone-on-a-chip models cannot mimic the vasculature in the system, microfluidic vascularized bone models can closely resemble the dynamic in vivo microenvironment. One example demonstrated a microfluidic vascularized bone model with the integration of hydroxyapatite (HA) and a fibrin ECM, resulting in enhanced angiogenic properties [110]. The presence of HA improved angiogenic characteristics such as the sprout length, velocity, number, and lumen diameter, indicating a more robust and dynamic formation of vascular networks [110]. The microfluidic device enabled paracrine communication between ECs and stromal cells during vessel formation, which is critical for mimicking in vivo bone angiogenesis [110]. In addition, the biocompatibility of HA with the microvascular endothelium was confirmed, as it maintained healthy endothelial markers and supported the expression of functional endothelial markers that contribute to angiogenesis [110]. In particular, this experiment did not include additional in vivo studies, but microfluidic models have great potential for in vivo applications, as shown in other examples of successful transplantation of vascularized microfluidic models [118]. In addition to microfluidic models, other synthetic models have used innovative methods to induce angiogenesis in bone regeneration. A different study developed a biomimetic membrane with HA nanoparticle micropatterns to mimic the natural periosteum and promote bone regeneration [111]. These membranes showed efficient cell adhesion and successfully induced the cell alignment and differentiation in vitro, and MSCs cultured on the membranes showed greater osteogenesis and angiogenesis [111]. The in vivo experiments in a rat calvarial defect model demonstrated that the biomimetic membranes not only enhanced bone regeneration but also significantly promoted blood vessel formation and ossification at the defect site by providing a conducive microenvironment for cell orientation and the sustained release of growth factors from calcium phosphate, which are key to effective vascularization and bone tissue regeneration [111]. Another study investigated the effects of a static magnetic field and magnetic scaffolds on osteoblast differentiation, bone formation, and angiogenesis [112]. Their results showed that the application of a magnetic field in combination with magnetic scaffolds significantly stimulated osteoblast functions, resulting in enhanced bone regeneration [112]. This combination was also found to promote VEGF expression and capillary tube formation [112]. When implanted in a mouse calvarial defect model for 6 weeks, the magnetic scaffold filled the defect gap more densely with newly generated bone tissue and showed a highly calcified structure, suggesting that the use of a magnetic field may be a potential tool for inducing angiogenesis in bone tissue regeneration [112].

5. Conclusions and Future Perspectives

In this review, we provide an overview of the angiogenic strategies that promote bone regeneration. Since bone, as a highly vascularized tissue, undergoes hypoxia due to bleeding in bone fractures, creating new vasculatures connected with host blood vessels has emerged as a promising strategy for bone tissue engineering. The neovasculature in the bone defect provides oxygen and the required nutrients necessary for osteogenesis, and also secretes angiogenic factors that stimulate osteogenesis. Conventional tissue engineering indirectly induces bone angiogenesis by fabricating porous scaffolds that lead to EC migration due to the hypoxic condition inside the scaffold. Meanwhile, recent strategies reviewed in this paper focus on the direct induction of angiogenesis by releasing proangiogenic proteins, genes, cells, or EVs, as well as the incorporation of specific mechanical and structural properties. It is important to note that each approach has its own advantages and disadvantages, which can impact therapeutic efficacy and safety. For instance, angiogenic growth factors such as VEGF are a primary means of inducing angiogenesis, while their application raises concerns about the potential for inducing aberrant physiologies, including leaky vasculature or tumorigenesis, particularly at high dosages [119]. Cell therapy and gene therapy present alternative strategies to growth factors, yet their long-term safety remains inadequately evaluated, and their therapeutic efficacy has not demonstrated superiority over VEGF treatment. EV treatment is emerging as a potential angiogenic strategy with its high safety and therapeutic efficacy, but is hindered by low productivity and high costs, which pose challenges for clinical translation. Scaffold engineering represents another avenue for future advancement in bone angiogenesis without the need for angiogenic factors, such as the NO-releasing hydrogel or 3D microchannel structures demonstrating the ability to recruit ECs and induce angiogenesis [120]. Nevertheless, the therapeutic effectiveness of mechanical and structural cues still appears to be comparatively insufficient compared to biochemical treatments. We further expect the integration of nanostructures to be a transformative force in bone regeneration, as well as microfluidic devices that replicate perfusable vascularized bone as potential implantable devices, which are currently being explored with better regenerative efficacy. Overall, ongoing research into the development and application of angiogenesis in bone regeneration holds tremendous potential for advancing the relevant fields and establishing novel strategies for targeted and efficient therapeutic interventions.

Author Contributions

Writing—original draft preparation, H.-J.J.; Writing—review and editing, J.-K.Y.; Project administration, J.-K.Y.; Funding acquisition, J.-K.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This study was financially supported by a grant from the National Research Foundation of Korea (NRF), funded by the Korea government (MSIT) (RS-2023-00213691 and 2021K1A3A1A74099704), as well as the Chung-Ang University Graduate Research Scholarship in 2022.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zhou, R.; Guo, Q.; Xiao, Y.; Guo, Q.; Huang, Y.; Li, C.; Luo, X. Endocrine role of bone in the regulation of energy metabolism. Bone Res. 2021, 9, 25. [Google Scholar] [CrossRef]
  2. Heini, P.F.; Berlemann, U.; Kaufmann, M.; Lippuner, K.; Fankhauser, C.; van Landuyt, P. Augmentation of mechanical properties in osteoporotic vertebral bones—A biomechanical investigation of vertebroplasty efficacy with different bone cements. Eur. Spine J. 2001, 10, 164–171. [Google Scholar] [CrossRef]
  3. Kenkre, J.; Bassett, J. The bone remodelling cycle. Ann. Clin. Biochem. 2018, 55, 308–327. [Google Scholar] [CrossRef]
  4. Kanczler, J.; Oreffo, R. Osteogenesis and angiogenesis: The potential for engineering bone. Eur. Cell Mater. 2008, 15, 100–114. [Google Scholar] [CrossRef]
  5. Arvidson, K.; Abdallah, B.; Applegate, L.; Baldini, N.; Cenni, E.; Gomez-Barrena, E.; Granchi, D.; Kassem, M.; Konttinen, Y.; Mustafa, K. Bone regeneration and stem cells. J. Cell. Mol. Med. 2011, 15, 718–746. [Google Scholar] [CrossRef]
  6. Weinans, H.; Huiskes, R.; Van Rietbergen, B.; Sumner, D.R.; Turner, T.M.; Galante, J.O. Adaptive bone remodeling around bonded noncemented total hip arthroplasty: A comparison between animal experiments and computer simulation. J. Orthop. Res. 1993, 11, 500–513. [Google Scholar] [CrossRef] [PubMed]
  7. Kim, T.; See, C.W.; Li, X.; Zhu, D. Orthopedic implants and devices for bone fractures and defects: Past, present and perspective. Eng. Regen. 2020, 1, 6–18. [Google Scholar] [CrossRef]
  8. Wüster, J.; Hesse, B.; Rothweiler, R.; Bortel, E.; Gross, C.; Bakhtiyari, S.; King, A.; Boller, E.; Gerber, J.; Rendenbach, C.; et al. Comparison of the 3D-microstructure of human alveolar and fibula bone in microvascular autologous bone transplantation: A synchrotron radiation μ-CT study. Front. Bioeng. Biotechnol. 2023, 11, 1169385. [Google Scholar] [CrossRef] [PubMed]
  9. Ebina, H.; Hatakeyama, J.; Onodera, M.; Honma, T.; Kamakura, S.; Shimauchi, H.; Sasano, Y. Micro-CT analysis of alveolar bone healing using a rat experimental model of critical-size defects. Oral Dis. 2009, 15, 273–280. [Google Scholar] [CrossRef] [PubMed]
  10. Cooper, G.M.; Mooney, M.P.; Gosain, A.K.; Campbell, P.G.; Losee, J.E.; Huard, J. Testing the “critical-size” in calvarial bone defects: Revisiting the concept of a critical-sized defect (CSD). Plast. Reconstr. Surg. 2010, 125, 1685. [Google Scholar] [CrossRef] [PubMed]
  11. Roddy, E.; DeBaun, M.R.; Daoud-Gray, A.; Yang, Y.P.; Gardner, M.J. Treatment of critical-sized bone defects: Clinical and tissue engineering perspectives. Eur. J. Orthop. Surg. Traumatol. 2018, 28, 351–362. [Google Scholar] [CrossRef]
  12. Schmitz, J.P.; Hollinger, J.O. The critical size defect as an experimental model for craniomandibulofacial nonunions. Clin. Orthop. Relat. Res. (1976–2007) 1986, 205, 299–308. [Google Scholar] [CrossRef]
  13. Masquelet, A.C.; Kishi, T.; Benko, P.E. Very long-term results of post-traumatic bone defect reconstruction by the induced membrane technique. Orthop. Traumatol. Surg. Res. 2019, 105, 159–166. [Google Scholar] [CrossRef]
  14. Lasanianos, N.G.; Kanakaris, N.K.; Giannoudis, P.V. Current management of long bone large segmental defects. Orthop. Trauma 2010, 24, 149–163. [Google Scholar] [CrossRef]
  15. Laubach, M.; Suresh, S.; Herath, B.; Wille, M.-L.; Delbrück, H.; Alabdulrahman, H.; Hutmacher, D.W.; Hildebrand, F. Clinical translation of a patient-specific scaffold-guided bone regeneration concept in four cases with large long bone defects. J. Orthop. Transl. 2022, 34, 73–84. [Google Scholar] [CrossRef] [PubMed]
  16. Pape, H.C.; Evans, A.; Kobbe, P. Autologous bone graft: Properties and techniques. J. Orthop. Trauma 2010, 24, S36–S40. [Google Scholar] [CrossRef]
  17. Sakkas, A.; Wilde, F.; Heufelder, M.; Winter, K.; Schramm, A. Autogenous bone grafts in oral implantology—Is it still a “gold standard”? A consecutive review of 279 patients with 456 clinical procedures. Int. J. Implant Dent. 2017, 3, 23. [Google Scholar] [CrossRef] [PubMed]
  18. O’Malley, M.J.; Sayres, S.C.; Saleem, O.; Levine, D.; Roberts, M.; Deland, J.T.; Ellis, S. Morbidity and complications following percutaneous calcaneal autograft bone harvest. Foot Ankle Int. 2014, 35, 30–37. [Google Scholar] [CrossRef]
  19. Schwartz, C.E.; Martha, J.F.; Kowalski, P.; Wang, D.A.; Bode, R.; Li, L.; Kim, D.H. Prospective evaluation of chronic pain associated with posterior autologous iliac crest bone graft harvest and its effect on postoperative outcome. Health Qual. Life Outcomes 2009, 7, 49. [Google Scholar] [CrossRef] [PubMed]
  20. Suchomel, P.; Barsa, P.; Buchvald, P.; Svobodnik, A.; Vanickova, E. Autologous versus allogenic bone grafts in instrumented anterior cervical discectomy and fusion: A prospective study with respect to bone union pattern. Eur. Spine J. 2004, 13, 510–515. [Google Scholar] [CrossRef]
  21. Eppley, B.L.; Pietrzak, W.S.; Blanton, M.W. Allograft and alloplastic bone substitutes: A review of science and technology for the craniomaxillofacial surgeon. J. Craniofac. Surg. 2005, 16, 981–989. [Google Scholar] [CrossRef]
  22. Bauer, T.W.; Muschler, G.F. Bone graft materials: An overview of the basic science. Clin. Orthop. Relat. Res. 2000, 371, 10–27. [Google Scholar] [CrossRef]
  23. Baldwin, P.; Li, D.J.; Auston, D.A.; Mir, H.S.; Yoon, R.S.; Koval, K.J. Autograft, allograft, and bone graft substitutes: Clinical evidence and indications for use in the setting of orthopaedic trauma surgery. J. Orthop. Trauma 2019, 33, 203–213. [Google Scholar] [CrossRef] [PubMed]
  24. Aguirre, L.E.; Guzman, M.E.; Lopes, G.; Hurley, J. Immune checkpoint inhibitors and the risk of allograft rejection: A comprehensive analysis on an emerging issue. Oncologist 2019, 24, 394–401. [Google Scholar] [CrossRef] [PubMed]
  25. Kowalski, R.J.; Post, D.R.; Mannon, R.B.; Sebastian, A.; Wright, H.I.; Sigle, G.; Burdick, J.; Elmagd, K.A.; Zeevi, A.; Lopez-Cepero, M. Assessing relative risks of infection and rejection: A meta-analysis using an immune function assay. Transplantation 2006, 82, 663–668. [Google Scholar] [CrossRef] [PubMed]
  26. Lafage-Proust, M.-H.; Roche, B.; Langer, M.; Cleret, D.; Bossche, A.V.; Olivier, T.; Vico, L. Assessment of bone vascularization and its role in bone remodeling. BoneKEy Rep. 2015, 4, 662. [Google Scholar] [CrossRef] [PubMed]
  27. Schmid, J.; Wallkamm, B.; Hämmerle, C.H.; Gogolewski, S.; Lang, N.P. The significance of angiogenesis in guided bone regeneration. A case report of a rabbit experiment. Clin. Oral. Implant. Res. 1997, 8, 244–248. [Google Scholar] [CrossRef] [PubMed]
  28. Bouland, C.; Philippart, P.; Dequanter, D.; Corrillon, F.; Loeb, I.; Bron, D.; Lagneaux, L.; Meuleman, N. Cross-talk between mesenchymal stromal cells (MSCs) and endothelial progenitor cells (EPCs) in bone regeneration. Front. Cell Dev. Biol. 2021, 9, 674084. [Google Scholar] [CrossRef] [PubMed]
  29. Zhang, W.; Zhu, C.; Wu, Y.; Ye, D.; Wang, S.; Zou, D.; Zhang, X.; Kaplan, D.L.; Jiang, X. VEGF and BMP-2 promote bone regeneration by facilitating bone marrow stem cell homing and differentiation. Eur. Cell Mater. 2014, 27, 1–11; discussion 11–12. [Google Scholar] [CrossRef]
  30. Potier, E.; Ferreira, E.; Andriamanalijaona, R.; Pujol, J.-P.; Oudina, K.; Logeart-Avramoglou, D.; Petite, H. Hypoxia affects mesenchymal stromal cell osteogenic differentiation and angiogenic factor expression. Bone 2007, 40, 1078–1087. [Google Scholar] [CrossRef]
  31. Mistry, A.S.; Mikos, A.G. Tissue engineering strategies for bone regeneration. In Regenerative Medicine II: Clinical and Preclinical Applications; Springer: Berlin/Heidelberg, Germany, 2005; pp. 1–22. [Google Scholar]
  32. Adachi, T.; Osako, Y.; Tanaka, M.; Hojo, M.; Hollister, S.J. Framework for optimal design of porous scaffold microstructure by computational simulation of bone regeneration. Biomaterials 2006, 27, 3964–3972. [Google Scholar] [CrossRef]
  33. Kaigler, D.; Pagni, G.; Park, C.H.; Braun, T.M.; Holman, L.A.; Yi, E.; Tarle, S.A.; Bartel, R.L.; Giannobile, W.V. Stem cell therapy for craniofacial bone regeneration: A randomized, controlled feasibility trial. Cell Transplant. 2013, 22, 767–777. [Google Scholar] [CrossRef] [PubMed]
  34. Subbiah, R.; Ruehle, M.A.; Klosterhoff, B.S.; Lin, A.S.; Hettiaratchi, M.H.; Willett, N.J.; Bertassoni, L.E.; García, A.J.; Guldberg, R.E. Triple growth factor delivery promotes functional bone regeneration following composite musculoskeletal trauma. Acta Biomater. 2021, 127, 180–192. [Google Scholar] [CrossRef] [PubMed]
  35. Katagiri, W.; Sakaguchi, K.; Kawai, T.; Wakayama, Y.; Osugi, M.; Hibi, H. A defined mix of cytokines mimics conditioned medium from cultures of bone marrow-derived mesenchymal stem cells and elicits bone regeneration. Cell Prolif. 2017, 50, e12333. [Google Scholar] [CrossRef] [PubMed]
  36. Liang, H.; Jin, C.; Ma, L.; Feng, X.; Deng, X.; Wu, S.; Liu, X.; Yang, C. Accelerated bone regeneration by gold-nanoparticle-loaded mesoporous silica through stimulating immunomodulation. ACS Appl. Mater. Interfaces 2019, 11, 41758–41769. [Google Scholar] [CrossRef] [PubMed]
  37. Kent Leach, J.; Kaigler, D.; Wang, Z.; Krebsbach, P.H.; Mooney, D.J. Coating of VEGF-releasing scaffolds with bioactive glass for angiogenesis and bone regeneration. Biomaterials 2006, 27, 3249–3255. [Google Scholar] [CrossRef]
  38. Zhou, J.; See, C.W.; Sreenivasamurthy, S.; Zhu, D. Customized Additive Manufacturing in Bone Scaffolds—The Gateway to Precise Bone Defect Treatment. Research 2023, 6, 0239. [Google Scholar] [CrossRef] [PubMed]
  39. Raisz, L.G. Physiology and pathophysiology of bone remodeling. Clin. Chem. 1999, 45, 1353–1358. [Google Scholar]
  40. Duda, G.N.; Geissler, S.; Checa, S.; Tsitsilonis, S.; Petersen, A.; Schmidt-Bleek, K. The decisive early phase of bone regeneration. Nat. Rev. Rheumatol. 2023, 19, 78–95. [Google Scholar] [CrossRef]
  41. Li, J.; Ma, J.; Feng, Q.; Xie, E.; Meng, Q.; Shu, W.; Wu, J.; Bian, L.; Han, F.; Li, B. Building Osteogenic Microenvironments with a Double-Network Composite Hydrogel for Bone Repair. Research 2023, 6, 0021. [Google Scholar] [CrossRef]
  42. Kurkalli, B.G.S.; Gurevitch, O.; Sosnik, A.; Cohn, D.; Slavin, S. Repair of bone defect using bone marrow cells and demineralized bone matrix supplemented with polymeric materials. Curr. Stem Cell Res. Ther. 2010, 5, 49–56. [Google Scholar] [CrossRef]
  43. Schmidt-Bleek, K.; Schell, H.; Lienau, J.; Schulz, N.; Hoff, P.; Pfaff, M.; Schmidt, G.; Martin, C.; Perka, C.; Buttgereit, F. Initial immune reaction and angiogenesis in bone healing. J. Tissue Eng. Regen. Med. 2014, 8, 120–130. [Google Scholar] [CrossRef]
  44. Shiu, H.T.; Leung, P.C.; Ko, C.H. The roles of cellular and molecular components of a hematoma at early stage of bone healing. J. Tissue Eng. Regen. Med. 2018, 12, e1911–e1925. [Google Scholar] [CrossRef]
  45. Oryan, A.; Monazzah, S.; Bigham-Sadegh, A. Bone injury and fracture healing biology. Biomed. Environ. Sci. 2015, 28, 57–71. [Google Scholar]
  46. Wang, L.; Fan, H.; Zhang, Z.-Y.; Lou, A.-J.; Pei, G.-X.; Jiang, S.; Mu, T.-W.; Qin, J.-J.; Chen, S.-Y.; Jin, D. Osteogenesis and angiogenesis of tissue-engineered bone constructed by prevascularized β-tricalcium phosphate scaffold and mesenchymal stem cells. Biomaterials 2010, 31, 9452–9461. [Google Scholar] [CrossRef]
  47. Kolar, P.; Gaber, T.; Perka, C.; Duda, G.N.; Buttgereit, F. Human early fracture hematoma is characterized by inflammation and hypoxia. Clin. Orthop. Relat. Res. 2011, 469, 3118–3126. [Google Scholar] [CrossRef] [PubMed]
  48. Hankenson, K.D.; Dishowitz, M.; Gray, C.; Schenker, M. Angiogenesis in bone regeneration. Injury 2011, 42, 556–561. [Google Scholar] [CrossRef]
  49. Schott, N.G.; Friend, N.E.; Stegemann, J.P. Coupling osteogenesis and vasculogenesis in engineered orthopedic tissues. Tissue Eng. Part B Rev. 2021, 27, 199–214. [Google Scholar] [CrossRef] [PubMed]
  50. Pearson, H.B.; Mason, D.E.; Kegelman, C.D.; Zhao, L.; Dawahare, J.H.; Kacena, M.A.; Boerckel, J.D. Effects of bone morphogenetic protein-2 on neovascularization during large bone defect regeneration. Tissue Eng. Part A 2019, 25, 1623–1634. [Google Scholar] [CrossRef]
  51. Wang, L.; Zhou, F.; Zhang, P.; Wang, H.; Qu, Z.; Jia, P.; Yao, Z.; Shen, G.; Li, G.; Zhao, G. Human type H vessels are a sensitive biomarker of bone mass. Cell Death Dis. 2017, 8, e2760. [Google Scholar] [CrossRef]
  52. Peng, Y.; Wu, S.; Li, Y.; Crane, J.L. Type H blood vessels in bone modeling and remodeling. Theranostics 2020, 10, 426–436. [Google Scholar] [CrossRef]
  53. Kusumbe, A.P.; Ramasamy, S.K.; Adams, R.H. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014, 507, 323–328. [Google Scholar] [CrossRef] [PubMed]
  54. Zelzer, E.; Mamluk, R.; Ferrara, N.; Johnson, R.S.; Schipani, E.; Olsen, B.R. VEGFA is necessary for chondrocyte survival during bone development. Development 2004, 131, 2161–2171. [Google Scholar] [CrossRef]
  55. Xie, H.; Cui, Z.; Wang, L.; Xia, Z.; Hu, Y.; Xian, L.; Li, C.; Xie, L.; Crane, J.; Wan, M.; et al. PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat. Med. 2014, 20, 1270–1278. [Google Scholar] [CrossRef] [PubMed]
  56. Percival, C.J.; Richtsmeier, J.T. Angiogenesis and intramembranous osteogenesis. Dev. Dyn. 2013, 242, 909–922. [Google Scholar] [CrossRef] [PubMed]
  57. Hu, K.; Olsen, B.R. The roles of vascular endothelial growth factor in bone repair and regeneration. Bone 2016, 91, 30–38. [Google Scholar] [CrossRef]
  58. Saleh, F.A.; Whyte, M.; Genever, P.G. Effects of endothelial cells on human mesenchymal stem cell activity in a three-dimensional in vitro model. Eur. Cell Mater. 2011, 22, 242–257; discussion 257. [Google Scholar] [CrossRef] [PubMed]
  59. Pennings, I.; van Dijk, L.A.; van Huuksloot, J.; Fledderus, J.O.; Schepers, K.; Braat, A.K.; Hsiao, E.C.; Barruet, E.; Morales, B.M.; Verhaar, M.C.; et al. Effect of donor variation on osteogenesis and vasculogenesis in hydrogel cocultures. J. Tissue Eng. Regen. Med. 2019, 13, 433–445. [Google Scholar] [CrossRef]
  60. Younger, E.M.; Chapman, M.W. Morbidity at Bone Graft Donor Sites. J. Orthop. Trauma 1989, 3, 192–195. [Google Scholar] [CrossRef]
  61. William Jr, G.; Einhorn, T.A.; Koval, K.; McKee, M.; Smith, W.; Sanders, R.; Watson, T. Bone grafts and bone graft substitutes in orthopaedic trauma surgery: A critical analysis. JBJS 2007, 89, 649–658. [Google Scholar]
  62. Yang, K.; Ren, Y. Nickel-free austenitic stainless steels for medical applications. Sci. Technol. Adv. Mater. 2010, 11, 014105. [Google Scholar] [CrossRef]
  63. Yasuoka, S.; Usukura, Y.; Fuse, M.; Okada, H.; Hayakawa, T.; Kato, T. Stainless and titanium fibers as non-degradable three-dimensional scaffolds for bone reconstruction. J. Hard Tissue Biol. 2014, 23, 407–414. [Google Scholar] [CrossRef]
  64. Zhang, Q.-H.; Cossey, A.; Tong, J. Stress shielding in periprosthetic bone following a total knee replacement: Effects of implant material, design and alignment. Med. Eng. Phys. 2016, 38, 1481–1488. [Google Scholar] [CrossRef] [PubMed]
  65. Noyama, Y.; Miura, T.; Ishimoto, T.; Itaya, T.; Niinomi, M.; Nakano, T. Bone loss and reduced bone quality of the human femur after total hip arthroplasty under stress-shielding effects by titanium-based implant. Mater. Trans. 2012, 53, 565–570. [Google Scholar] [CrossRef]
  66. Mi, Z.; Shuib, S.; Hassan, A.; Shorki, A.; Ibrahim, M. Problem of stress shielding and improvement to the hip Implat designs: A review. J. Med. Sci. 2007, 7, 460–467. [Google Scholar]
  67. Glenske, K.; Donkiewicz, P.; Köwitsch, A.; Milosevic-Oljaca, N.; Rider, P.; Rofall, S.; Franke, J.; Jung, O.; Smeets, R.; Schnettler, R. Applications of metals for bone regeneration. Int. J. Mol. Sci. 2018, 19, 826. [Google Scholar] [CrossRef] [PubMed]
  68. Blázquez-Carmona, P.; Mora-Macías, J.; Martínez-Vázquez, F.J.; Morgaz, J.; Domínguez, J.; Reina-Romo, E. Mechanics Predicts Effective Critical-Size Bone Regeneration Using 3D-Printed Bioceramic Scaffolds. Tissue Eng. Regen. Med. 2023, 20, 893–904. [Google Scholar] [CrossRef] [PubMed]
  69. Bohner, M.; Santoni, B.L.G.; Döbelin, N. β-tricalcium phosphate for bone substitution: Synthesis and properties. Acta Biomater. 2020, 113, 23–41. [Google Scholar] [CrossRef] [PubMed]
  70. Jang, H.L.; Zheng, G.B.; Park, J.; Kim, H.D.; Baek, H.R.; Lee, H.K.; Lee, K.; Han, H.N.; Lee, C.K.; Hwang, N.S. In vitro and in vivo evaluation of whitlockite biocompatibility: Comparative study with hydroxyapatite and β-tricalcium phosphate. Adv. Healthc. Mater. 2016, 5, 128–136. [Google Scholar] [CrossRef] [PubMed]
  71. Szcześ, A.; Hołysz, L.; Chibowski, E. Synthesis of hydroxyapatite for biomedical applications. Adv. Colloid Interface Sci. 2017, 249, 321–330. [Google Scholar] [CrossRef]
  72. Heimann, R.B. Structure, properties, and biomedical performance of osteoconductive bioceramic coatings. Surf. Coat. Technol. 2013, 233, 27–38. [Google Scholar] [CrossRef]
  73. Eliaz, N.; Metoki, N. Calcium Phosphate Bioceramics: A Review of Their History, Structure, Properties, Coating Technologies and Biomedical Applications. Materials 2017, 10, 334. [Google Scholar] [CrossRef]
  74. Thompson, I.D.; Hcnch, L.L. Mechanical properties of bioactive glasses, glass-ceramics and composites. Proc. Inst. Mech. Eng. Part H J. Eng. Med. 1998, 212, 127–136. [Google Scholar] [CrossRef]
  75. Mala, R.; Celsia, A.R. Bioceramics in orthopaedics: A review. In Fundamental Biomaterials: Ceramics; Woodhead Publishing: Sawston, UK, 2018; pp. 195–221. [Google Scholar]
  76. Lin, T.-H.; Wang, H.-C.; Cheng, W.-H.; Hsu, H.-C.; Yeh, M.-L. Osteochondral tissue regeneration using a tyramine-modified bilayered PLGA scaffold combined with articular chondrocytes in a porcine model. Int. J. Mol. Sci. 2019, 20, 326. [Google Scholar] [CrossRef]
  77. Wang, C.; Xu, D.; Li, S.; Yi, C.; Zhang, X.; He, Y.; Yu, D. Effect of pore size on the physicochemical properties and osteogenesis of Ti6Al4V porous scaffolds with bionic structure. ACS Omega 2020, 5, 28684–28692. [Google Scholar] [CrossRef] [PubMed]
  78. Woodard, J.R.; Hilldore, A.J.; Lan, S.K.; Park, C.J.; Morgan, A.W.; Eurell, J.A.C.; Clark, S.G.; Wheeler, M.B.; Jamison, R.D.; Wagoner Johnson, A.J. The mechanical properties and osteoconductivity of hydroxyapatite bone scaffolds with multi-scale porosity. Biomaterials 2007, 28, 45–54. [Google Scholar]
  79. Makihara, T.; Sakane, M.; Noguchi, H.; Tsukanishi, T.; Suetsugu, Y.; Yamazaki, M. Formation of osteon-like structures in unidirectional porous hydroxyapatite substitute. J. Biomed. Mater. Res. Part B Appl. Biomater. 2018, 106, 2665–2672. [Google Scholar]
  80. Quade, M.; Münch, P.; Lode, A.; Duin, S.; Vater, C.; Gabrielyan, A.; Rösen-Wolff, A.; Gelinsky, M. The secretome of hypoxia conditioned hMSC loaded in a central depot induces chemotaxis and angiogenesis in a biomimetic mineralized collagen bone replacement material. Adv. Healthc. Mater. 2020, 9, 1901426. [Google Scholar] [CrossRef]
  81. Song, P.; Hu, C.; Pei, X.; Sun, J.; Sun, H.; Wu, L.; Jiang, Q.; Fan, H.; Yang, B.; Zhou, C. Dual modulation of crystallinity and macro-/microstructures of 3D printed porous titanium implants to enhance stability and osseointegration. J. Mater. Chem. B 2019, 7, 2865–2877. [Google Scholar] [CrossRef]
  82. Klompmaker, J.; Jansen, H.W.; Veth, R.H.; Nielsen, H.K.; de Groot, J.H.; Pennings, A.J. Porous implants for knee joint meniscus reconstruction: A preliminary study on the role of pore sizes in ingrowth and differentiation of fibrocartilage. Clin. Mater. 1993, 14, 1–11. [Google Scholar] [CrossRef]
  83. Abbasi, N.; Hamlet, S.; Love, R.M.; Nguyen, N.-T. Porous scaffolds for bone regeneration. J. Sci. Adv. Mater. Devices 2020, 5, 1–9. [Google Scholar] [CrossRef]
  84. Klenke, F.M.; Liu, Y.; Yuan, H.; Hunziker, E.B.; Siebenrock, K.A.; Hofstetter, W. Impact of pore size on the vascularization and osseointegration of ceramic bone substitutes in vivo. J. Biomed. Mater. Res. Part A Off. J. Soc. Biomater. Jpn. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 2008, 85, 777–786. [Google Scholar] [CrossRef]
  85. Patel, Z.S.; Young, S.; Tabata, Y.; Jansen, J.A.; Wong, M.E.; Mikos, A.G. Dual delivery of an angiogenic and an osteogenic growth factor for bone regeneration in a critical size defect model. Bone 2008, 43, 931–940. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, B.S.; Kim, J.S.; Yang, S.S.; Kim, H.W.; Lim, H.J.; Lee, J. Angiogenin-loaded fibrin/bone powder composite scaffold for vascularized bone regeneration. Biomater. Res. 2015, 19, 18. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, M.; Zhang, Y.; Zhang, W.; Li, J. Polyhedral Oligomeric Silsesquioxane-Incorporated Gelatin Hydrogel Promotes Angiogenesis during Vascularized Bone Regeneration. ACS Appl. Mater. Interfaces 2020, 12, 22410–22425. [Google Scholar] [CrossRef] [PubMed]
  88. Kim, H.D.; Park, J.; Amirthalingam, S.; Jayakumar, R.; Hwang, N.S. Bioinspired inorganic nanoparticles and vascular factor microenvironment directed neo-bone formation. Biomater. Sci. 2020, 8, 2627–2637. [Google Scholar] [CrossRef] [PubMed]
  89. Zhang, W.; Feng, C.; Yang, G.; Li, G.; Ding, X.; Wang, S.; Dou, Y.; Zhang, Z.; Chang, J.; Wu, C.; et al. 3D-printed scaffolds with synergistic effect of hollow-pipe structure and bioactive ions for vascularized bone regeneration. Biomaterials 2017, 135, 85–95. [Google Scholar] [CrossRef] [PubMed]
  90. Ha, Y.; Ma, X.; Li, S.; Li, T.; Li, Z.; Qian, Y.; Shafiq, M.; Wang, J.; Zhou, X.; He, C. Bone Microenvironment-Mimetic Scaffolds with Hierarchical Microstructure for Enhanced Vascularization and Bone Regeneration. Adv. Funct. Mater. 2022, 32, 2200011. [Google Scholar] [CrossRef]
  91. Fitzpatrick, V.; Martín-Moldes, Z.; Deck, A.; Torres-Sanchez, R.; Valat, A.; Cairns, D.; Li, C.; Kaplan, D.L. Functionalized 3D-printed silk-hydroxyapatite scaffolds for enhanced bone regeneration with innervation and vascularization. Biomaterials 2021, 276, 120995. [Google Scholar] [CrossRef] [PubMed]
  92. Kaigler, D.; Krebsbach, P.H.; West, E.R.; Horger, K.; Huang, Y.-C.; Mooney, D.J. Endothelial cell modulation of bone marrow stromal cell osteogenic potential. FASEB J. 2005, 19, 1–26. [Google Scholar] [CrossRef]
  93. Yu, H.; VandeVord, P.J.; Mao, L.; Matthew, H.W.; Wooley, P.H.; Yang, S.-Y. Improved tissue-engineered bone regeneration by endothelial cell mediated vascularization. Biomaterials 2009, 30, 508–517. [Google Scholar] [CrossRef]
  94. Zhou, J.; Lin, H.; Fang, T.; Li, X.; Dai, W.; Uemura, T.; Dong, J. The repair of large segmental bone defects in the rabbit with vascularized tissue engineered bone. Biomaterials 2010, 31, 1171–1179. [Google Scholar] [CrossRef]
  95. Curtin, C.M.; Tierney, E.G.; McSorley, K.; Cryan, S.A.; Duffy, G.P.; O’Brien, F.J. Combinatorial gene therapy accelerates bone regeneration: Non-viral dual delivery of VEGF and BMP2 in a collagen-nanohydroxyapatite scaffold. Adv. Healthc. Mater. 2015, 4, 223–227. [Google Scholar] [CrossRef]
  96. Geiger, F.; Bertram, H.; Berger, I.; Lorenz, H.; Wall, O.; Eckhardt, C.; Simank, H.G.; Richter, W. Vascular endothelial growth factor gene-activated matrix (VEGF165-GAM) enhances osteogenesis and angiogenesis in large segmental bone defects. J. Bone Miner. Res. 2005, 20, 2028–2035. [Google Scholar] [CrossRef] [PubMed]
  97. Cao, L.; Liu, X.; Liu, S.; Jiang, Y.; Zhang, X.; Zhang, C.; Zeng, B. Experimental repair of segmental bone defects in rabbits by angiopoietin-1 gene transfected MSCs seeded on porous β-TCP scaffolds. J. Biomed. Mater. Res. B Appl. Biomater. 2012, 100, 1229–1236. [Google Scholar] [CrossRef]
  98. Feng, H.; Li, Z.; Xie, W.; Wan, Q.; Guo, Y.; Chen, J.; Wang, J.; Pei, X. Delivery of therapeutic miRNAs using nanoscale zeolitic imidazolate framework for accelerating vascularized bone regeneration. Chem. Eng. J. 2022, 430, 132867. [Google Scholar] [CrossRef]
  99. Cui, H.; Zhu, W.; Holmes, B.; Zhang, L.G. Biologically Inspired Smart Release System Based on 3D Bioprinted Perfused Scaffold for Vascularized Tissue Regeneration. Adv. Sci. 2016, 3, 1600058. [Google Scholar] [CrossRef]
  100. Hann, S.Y.; Cui, H.; Esworthy, T.; Zhou, X.; Lee, S.-j.; Plesniak, M.W.; Zhang, L.G. Dual 3D printing for vascularized bone tissue regeneration. Acta Biomater. 2021, 123, 263–274. [Google Scholar] [CrossRef] [PubMed]
  101. Kazemzadeh-Narbat, M.; Rouwkema, J.; Annabi, N.; Cheng, H.; Ghaderi, M.; Cha, B.-H.; Aparnathi, M.; Khalilpour, A.; Byambaa, B.; Jabbari, E.; et al. Engineering Photocrosslinkable Bicomponent Hydrogel Constructs for Creating 3D Vascularized Bone. Adv. Healthc. Mater. 2017, 6, 1601122. [Google Scholar] [CrossRef] [PubMed]
  102. Zhao, Z.; Li, R.; Ruan, H.; Cai, Z.; Zhuang, Y.; Han, Z.; Zhang, M.; Cui, W.; Cai, M. Biological signal integrated microfluidic hydrogel microspheres for promoting bone regeneration. Chem. Eng. J. 2022, 436, 135176. [Google Scholar] [CrossRef]
  103. Xu, Y.; Xu, C.; He, L.; Zhou, J.; Chen, T.; Ouyang, L.; Guo, X.; Qu, Y.; Luo, Z.; Duan, D. Stratified-structural hydrogel incorporated with magnesium-ion-modified black phosphorus nanosheets for promoting neuro-vascularized bone regeneration. Bioact. Mater. 2022, 16, 271–284. [Google Scholar] [CrossRef] [PubMed]
  104. Zhou, Z.; Liu, Y.; Li, W.; Zhao, Z.; Xia, X.; Liu, J.; Deng, Y.; Wu, Y.; Pan, X.; He, F.; et al. A Self-Adaptive Biomimetic Periosteum Employing Nitric Oxide Release for Augmenting Angiogenesis in Bone Defect Regeneration. Adv. Healthc. Mater. 2023, e2302153. [Google Scholar] [CrossRef] [PubMed]
  105. Deng, J.; Wang, X.; Zhang, W.; Sun, L.; Han, X.; Tong, X.; Yu, L.; Ding, J.; Yu, L.; Liu, Y. Versatile Hypoxic Extracellular Vesicles Laden in an Injectable and Bioactive Hydrogel for Accelerated Bone Regeneration. Adv. Funct. Mater. 2023, 33, 2211664. [Google Scholar] [CrossRef]
  106. Gao, Y.; Yuan, Z.; Yuan, X.; Wan, Z.; Yu, Y.; Zhan, Q.; Zhao, Y.; Han, J.; Huang, J.; Xiong, C.; et al. Bioinspired porous microspheres for sustained hypoxic exosomes release and vascularized bone regeneration. Bioact. Mater. 2022, 14, 377–388. [Google Scholar] [CrossRef]
  107. Cui, H.; Zhu, W.; Nowicki, M.; Zhou, X.; Khademhosseini, A.; Zhang, L.G. Hierarchical Fabrication of Engineered Vascularized Bone Biphasic Constructs via Dual 3D Bioprinting: Integrating Regional Bioactive Factors into Architectural Design. Adv. Healthc. Mater. 2016, 5, 2174–2181. [Google Scholar] [CrossRef]
  108. Han, X.; Sun, M.; Chen, B.; Saiding, Q.; Zhang, J.; Song, H.; Deng, L.; Wang, P.; Gong, W.; Cui, W. Lotus seedpod-inspired internal vascularized 3D printed scaffold for bone tissue repair. Bioact. Mater. 2021, 6, 1639–1652. [Google Scholar] [CrossRef]
  109. Leucht, A.; Volz, A.C.; Rogal, J.; Borchers, K.; Kluger, P.J. Advanced gelatin-based vascularization bioinks for extrusion-based bioprinting of vascularized bone equivalents. Sci. Rep. 2020, 10, 5330. [Google Scholar] [CrossRef]
  110. Jusoh, N.; Oh, S.; Kim, S.; Kim, J.; Jeon, N.L. Microfluidic vascularized bone tissue model with hydroxyapatite-incorporated extracellular matrix. Lab Chip 2015, 15, 3984–3988. [Google Scholar] [CrossRef]
  111. Yang, G.; Liu, H.; Cui, Y.; Li, J.; Zhou, X.; Wang, N.; Wu, F.; Li, Y.; Liu, Y.; Jiang, X.; et al. Bioinspired membrane provides periosteum-mimetic microenvironment for accelerating vascularized bone regeneration. Biomaterials 2021, 268, 120561. [Google Scholar] [CrossRef]
  112. Yun, H.-M.; Ahn, S.-J.; Park, K.-R.; Kim, M.-J.; Kim, J.-J.; Jin, G.-Z.; Kim, H.-W.; Kim, E.-C. Magnetic nanocomposite scaffolds combined with static magnetic field in the stimulation of osteoblastic differentiation and bone formation. Biomaterials 2016, 85, 88–98. [Google Scholar] [CrossRef]
  113. Rady, A.A.M.; Hamdy, S.M.; Abdel-Hamid, M.A.; Hegazy, M.G.A.; Fathy, S.A.; Mostafa, A.A. The role of VEGF and BMP-2 in stimulation of bone healing with using hybrid bio-composite scaffolds coated implants in animal model. Bull. Natl. Res. Cent. 2020, 44, 131. [Google Scholar] [CrossRef]
  114. Diomede, F.; Marconi, G.D.; Fonticoli, L.; Pizzicanella, J.; Merciaro, I.; Bramanti, P.; Mazzon, E.; Trubiani, O. Functional relationship between osteogenesis and angiogenesis in tissue regeneration. Int. J. Mol. Sci. 2020, 21, 3242. [Google Scholar] [CrossRef] [PubMed]
  115. Park, J.; Lee, S.J.; Jo, H.H.; Lee, J.H.; Kim, W.D.; Lee, J.Y.; Park, S.A. Fabrication and characterization of 3D-printed bone-like β-tricalcium phosphate/polycaprolactone scaffolds for dental tissue engineering. J. Ind. Eng. Chem. 2017, 46, 175–181. [Google Scholar] [CrossRef]
  116. Kim, Y.; Lim, J.Y.; Yang, G.H.; Seo, J.-H.; Ryu, H.-S.; Kim, G. 3D-printed PCL/bioglass (BGS-7) composite scaffolds with high toughness and cell-responses for bone tissue regeneration. J. Ind. Eng. Chem. 2019, 79, 163–171. [Google Scholar] [CrossRef]
  117. Bajuri, M.Y.; Selvanathan, N.; Dzeidee Schaff, F.N.; Abdul Suki, M.H.; Ng, A.M.H. Tissue-Engineered Hydroxyapatite Bone Scaffold Impregnated with Osteoprogenitor Cells Promotes Bone Regeneration in Sheep Model. Tissue Eng. Regen. Med. 2021, 18, 377–385. [Google Scholar] [CrossRef] [PubMed]
  118. Lee, J.B.; Park, J.S.; Shin, Y.M.; Lee, D.H.; Yoon, J.-K.; Kim, D.-H.; Ko, U.H.; Kim, Y.; Bae, S.H.; Sung, H.-J. Implantable Vascularized Liver Chip for Cross-Validation of Disease Treatment with Animal Model. Adv. Funct. Mater. 2019, 29, 1900075. [Google Scholar] [CrossRef]
  119. Thurston, G.; Suri, C.; Smith, K.; McClain, J.; Sato, T.N.; Yancopoulos, G.D.; McDonald, D.M. Leakage-Resistant Blood Vessels in Mice Transgenically Overexpressing Angiopoietin-1. Science 1999, 286, 2511–2514. [Google Scholar] [CrossRef]
  120. Lee, J.B.; Kim, D.H.; Yoon, J.K.; Park, D.B.; Kim, H.S.; Shin, Y.M.; Baek, W.; Kang, M.L.; Kim, H.J.; Sung, H.J. Microchannel network hydrogel induced ischemic blood perfusion connection. Nat. Commun. 2020, 11, 615. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of the mechanism of bone regeneration and angiogenesis within a bone defect site. Type H vessels, characterized by the expression of Endomucin and CD31, play an important role in the modulation of osteogenesis. Osteoprogenitor cells undergo sequential differentiation, transforming into osteoblasts and subsequently maturing into osteocytes. The interplay of VEGF secreted by endothelial cells within type H vessels, along with MSCs, serves as a potent inducer of angiogenesis and ultimately contributes to bone regeneration.
Figure 1. Schematic illustration of the mechanism of bone regeneration and angiogenesis within a bone defect site. Type H vessels, characterized by the expression of Endomucin and CD31, play an important role in the modulation of osteogenesis. Osteoprogenitor cells undergo sequential differentiation, transforming into osteoblasts and subsequently maturing into osteocytes. The interplay of VEGF secreted by endothelial cells within type H vessels, along with MSCs, serves as a potent inducer of angiogenesis and ultimately contributes to bone regeneration.
Biomimetics 09 00075 g001
Figure 2. (A) Hematoxylin–Eosin staining image of enhanced vascularization at the defect site after implantation, showing an increased number of blood vessels [86]. Reprinted with permission from Kim et al., Copyright © 2024 Springer Nature. (B) MicroCT image showing the effects of POSS hydrogels on in vivo vascularization in the rat calvarial defect model with a higher number of blood vessels [87]. Reprinted with permission from Chen et al., Copyright © 2024 American Chemical Society. (C) MicroCT image showing enhanced vessel formation in the defect sites after BRT-H scaffold implantation in a rabbit radial defect model (red arrows) [89]. Reprinted with permission from Zhang et al., Copyright © 2024 Elsevier Ltd. (D) Digital images of harvested samples and microCT results to evaluate in vivo vascularization and bone regeneration in a rat calvarial defect model showing increased neovascularization after nanofibrous scaffold implantation [90]. Reprinted with permission from He et al., Copyright © 2024 John Wiley & Sons, Inc. (E) (a) PCL-HA composite and its (b) implantation surgery, as well as (c) observation of harvested implants showing bone integration and improved vascularization [93]. Reprinted with permission from Yu et al., Copyright © 2024 Elsevier Ltd.
Figure 2. (A) Hematoxylin–Eosin staining image of enhanced vascularization at the defect site after implantation, showing an increased number of blood vessels [86]. Reprinted with permission from Kim et al., Copyright © 2024 Springer Nature. (B) MicroCT image showing the effects of POSS hydrogels on in vivo vascularization in the rat calvarial defect model with a higher number of blood vessels [87]. Reprinted with permission from Chen et al., Copyright © 2024 American Chemical Society. (C) MicroCT image showing enhanced vessel formation in the defect sites after BRT-H scaffold implantation in a rabbit radial defect model (red arrows) [89]. Reprinted with permission from Zhang et al., Copyright © 2024 Elsevier Ltd. (D) Digital images of harvested samples and microCT results to evaluate in vivo vascularization and bone regeneration in a rat calvarial defect model showing increased neovascularization after nanofibrous scaffold implantation [90]. Reprinted with permission from He et al., Copyright © 2024 John Wiley & Sons, Inc. (E) (a) PCL-HA composite and its (b) implantation surgery, as well as (c) observation of harvested implants showing bone integration and improved vascularization [93]. Reprinted with permission from Yu et al., Copyright © 2024 Elsevier Ltd.
Biomimetics 09 00075 g002
Figure 3. (A) Schematic illustration of structural design of POSS hydrogels and their application for in vivo bone defect repair [87]. Reprinted with permission from Chen et al., Copyright © 2024 American Chemical Society. (B,C) Schematic illustration of the printing and fabrication process of hollow- tube scaffolds for vascularized bone regeneration using coaxial 3D- printing [89]. Reprinted with permission from Zhang et al., Copyright © 2024 Elsevier Ltd. (D) Schematic illustration of SHED-derived hypoxic exosome generation process and their application in the rat calvarial de-fect model for vascularized bone regeneration [106]. Reprinted with permission from Gao et al., Copyright © 2024 Elsevier Ltd. (E) Schematic illustration of the structural design and fabrication process of a 3D-printed vascularized bone construct using GelMA hydrogel, HUVECs, and MSCs [107]. Reprinted with permission from Cui et al., Copyright © 2024 John Wiley & Sons, Inc.
Figure 3. (A) Schematic illustration of structural design of POSS hydrogels and their application for in vivo bone defect repair [87]. Reprinted with permission from Chen et al., Copyright © 2024 American Chemical Society. (B,C) Schematic illustration of the printing and fabrication process of hollow- tube scaffolds for vascularized bone regeneration using coaxial 3D- printing [89]. Reprinted with permission from Zhang et al., Copyright © 2024 Elsevier Ltd. (D) Schematic illustration of SHED-derived hypoxic exosome generation process and their application in the rat calvarial de-fect model for vascularized bone regeneration [106]. Reprinted with permission from Gao et al., Copyright © 2024 Elsevier Ltd. (E) Schematic illustration of the structural design and fabrication process of a 3D-printed vascularized bone construct using GelMA hydrogel, HUVECs, and MSCs [107]. Reprinted with permission from Cui et al., Copyright © 2024 John Wiley & Sons, Inc.
Biomimetics 09 00075 g003
Table 1. A summary of vascularization/angiogenic strategies for bone regeneration.
Table 1. A summary of vascularization/angiogenic strategies for bone regeneration.
StrategyMain FactorsFindings and ObservationsRef.
Angiogenic factor deliveryVEGF, BMP-2Vessel formation ↑/Bone formation, bone bridging ↑[85]
VEGFBone bridging, bone volume fraction, bone mineral density ↑[37]
AngiogeninEC proliferation, adhesion ↑/Vessel formation ↑/Bone formation ↑[86]
VEGF, BMP-2Vessel number ↑/Bone formation ↑[87]
VEGFOsteogenic differentiation ↑/Vessel formation ↑/Bone formation ↑[88]
Bioactive ionsEC migration ↑/Vessel formation ↑/Stem cell, growth factor delivery ↑/Bone formation ↑[89]
DimethyloxalylglycineAngiogenic, Osteogenic marker expression ↑/ALP activity ↑/Vessel formation ↑/Bone formation ↑[90]
BMP-2, VEGF, NGFOsteogenic differentiation ↑/HUVEC migration ↑[91]
Cell deliveryECs, BMSCsOsteogenic marker expression ↑/Vessel formation ↑ [92]
ECs, osteoblastsVessel formation ↑/Bone formation ↑[93]
MSCs, MSC-derived ECsMechanical properties ↑/Capillary network ↑[94]
Gene deliveryVEGF, BMP-2 geneCalcium deposition, mineralization ↑/Host cell recruitment, transfection ↑/Vessel formation ↑[95]
VEGF geneBone gap ↓/Vessel formation ↑ Osteoid formation ↑[96]
Ang1 geneVessel growth ↑/Faster bone union/Bone formation ↑[97]
miR-21, miR-5106Vessel formation ↑/Bone mineral density, trabecular number, thickness ↑/Angiogenic ability ↑[98]
Perfusable 3D vascular networkPerfused microstructureOsteogenic differentiation ↑/Vascular like network ↑[99]
Perfusable vascular channelsOsteogenic differentiation ↑/Vessel formation ↑[100]
Angiogenesis-inducing hydrogelosteogenic and angiogenic niche-incorporated hydrogelEC, MSC self-assembly/Bone formation ↑[101]
Biological signal peptideBone formation ↑/Vascularization ↑[102]
Bilayer hydrogelEC proliferation, adhesion ↑/Bone mineral density, bone score values, bone formation ↑/Angiogenesis ↑[103]
NO-releasing biomimetic periosteumAngiogenesis ↑/Bone volume fraction, bone mineral density ↑[104]
EV deliveryBone marrow-derived hypoxic EVsVascularization ↑/Osteoblast proliferation, migration and differentiation ↑/Bone formation ↑[105]
SHED-derived hypoxic exosomesVascularization ↑/Bone formation ↑[106]
3D-bioprintingHUVECs, MSCs, GelMA hydrogelCell adhesion, proliferation ↑/VEGF, collagen I ↑/Osteogenic, angiogenic differentiation ↑/Capillary network ↑[107]
DFO liposome-containing scaffoldOsteogenic marker expression ↑/Bone growth ↑/Early-stage internal vascularization ↑/Vascular network maturation ↑[108]
HDMECs, ASCs, gelatin-based bioinkCapillary-mimicking structure formation ↑/Bone-specific protein ↑/Vascularization ↑[109]
Other synthetic methodsHA-incorporated microfluidic modelVessel sprout length, velocity, number, and lumen diameter ↑/ECs–stromal cells paracrine communication/Functional endothelial marker expression ↑[110]
Biomimetic membrane with HA nanoparticleCell adhesion, alignment, differentiation ↑/Vessel formation ↑/Ossification ↑ [111]
Static magnetic field, magnetic scaffoldOsteoblast function ↑/VEGF expression ↑/Capillary tube formation ↑/Bone formation ↑[112]
↑, indicating an increase; ↓, indicating a decrease; VEGF, vascular endothelial growth factor; BMP-2, bone morphogenetic protein 2; EC, endothelial cell; ALP, alkaline phosphatase; NGF, neural growth factor; HUVEC, human umbilical vein endothelial cell; BMSC, bone marrow stromal cell; MSC, mesenchymal stem cell; Ang1, angiopoietin 1; EV, extracellular vesicle; DFO, deferoxamine; HDMEC, human dermal microvascular endothelial cell; ASC, adipose-derived stem cell; HA, hydroxyapatite.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jang, H.-J.; Yoon, J.-K. The Role of Vasculature and Angiogenic Strategies in Bone Regeneration. Biomimetics 2024, 9, 75. https://doi.org/10.3390/biomimetics9020075

AMA Style

Jang H-J, Yoon J-K. The Role of Vasculature and Angiogenic Strategies in Bone Regeneration. Biomimetics. 2024; 9(2):75. https://doi.org/10.3390/biomimetics9020075

Chicago/Turabian Style

Jang, Hye-Jeong, and Jeong-Kee Yoon. 2024. "The Role of Vasculature and Angiogenic Strategies in Bone Regeneration" Biomimetics 9, no. 2: 75. https://doi.org/10.3390/biomimetics9020075

Article Metrics

Back to TopTop