Abstract Bone defect healing is a multi-factorial process involving the inflammatory microenvironment, bone regeneration and the formation of blood vessels, and remains a great challenge in clinical practice. Combined use of three-dimensional (3D)-printed scaffolds and bioactive factors is an emerging strategy for the treatment of bone defects. Scaffolds can be printed using 3D cryogenic printing technology to create a microarchitecture similar to trabecular bone. Melatonin (MT) has attracted attention in recent years as an excellent factor for promoting cell viability and tissue repair. In this study, porous scaffolds were prepared by cryogenic printing with poly(lactic-co-glycolic acid) and ultralong hydroxyapatite nanowires. The hierarchical pore size distribution of the scaffolds was evaluated by scanning electron microscopy (SEM) and micro-computed tomography (micro-CT). Sleep-inspired small extracellular vesicles (MT-sEVs) were then obtained from MT-stimulated cells and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol)-inorganic pyrophosphate (DSPE-PEG-PPi) was used to modify the membrane of MT-sEVs to obtain PPi-MT-sEVs. RNA sequencing was performed to explore the potential mechanisms. The results demonstrated that PPi-MT-sEVs not only enhanced cell proliferation, migration and angiogenesis, but also regulated the osteogenic/adipogenic fate determination and M1/M2 macrophage polarization switch in vitro. PPi-MT-sEVs were used to coat scaffolds, enabled by the capacity of PPi to bind to hydroxyapatite, and computational simulations were used to analyze the interfacial bonding of PPi and hydroxyapatite. The macrophage phenotype-modulating and osteogenesis–angiogenesis coupling effects were evaluated in vivo. In summary, this study suggests that the combination of hierarchical porous scaffolds and PPi-MT-sEVs could be a promising candidate for the clinical treatment of bone defects. Graphical abstract [48]graphic file with name 12951_2024_2977_Figa_HTML.jpg Supplementary Information The online version contains supplementary material available at 10.1186/s12951-024-02977-5. Keywords: 3D printing, Extracellular vesicles, Vascularized bone regeneration, Surface functionalization, Macrophage polarization Introduction Many clinical conditions, such as high-energy trauma and tumor resection, result in bone defects that are difficult to heal spontaneously [[49]1]. Autografts and allografts have excellent biocompatibility, mechanical properties and osteoinductivity, but there are still issues including limited bone volume, donor site damage and potential immune rejection [[50]2, [51]3]. Conventional bone substitutes exhibit deficiencies in growth factors and hierarchical pore architecture, leading to suboptimal outcomes in repairing large bone defects [[52]4–[53]6]. The ideal bone substitutes should have excellent biodegradability and biocompatibility, while simultaneously evading inflammatory responses. Additionally, the osteointegration and proangiogenic capacity of ideal bone substitutes emerge as crucial factors that influence their therapeutic outcomes [[54]7]. Bone tissue engineering provides a promising strategy to effectively repair bone defects. Tissue engineering scaffolds are made from materials with excellent biocompatibility. By incorporating bioactive factors, the osteointegration and osteoinductivity of scaffolds are enhanced, effectively promoting bone and vascular regeneration [[55]8]. Poly (lactic-co-glycolic acid) (PLGA) exhibits superior biocompatibility and degradability, and has been approved for use in basic and clinical medical research [[56]9, [57]10]. The degradation products of PLGA do not cause immune rejection, making it widely applicable for use in bone tissue engineering and drug delivery [[58]11]. Hydroxyapatite is the main inorganic component of bone and has excellent osteoinductive properties that promote osteogenic differentiation of mesenchymal stem cells (MSCs) to enhance bone regeneration [[59]12]. Hydroxyapatite nanowires have excellent osteoinductive properties and are easier to mix uniformly in scaffolds than hydroxyapatite nanoparticles [[60]13]. However, short hydroxyapatite nanowires (tens of micrometers in length) are brittle [[61]14], whereas ultralong hydroxyapatite nanowires (uW) with lengths of several hundred micrometers have superior toughness and mechanical strength [[62]15]. Further, uW can interweave with each other in scaffolds to promote vascular regeneration and bone defect repair [[63]16]. Three-dimensional (3D) printing is an additive manufacturing technology that allows control of the spatial distribution of biomaterials as well as the layered architecture [[64]17]. In recent years, 3D printing has been used in the field of tissue engineering, driving research progress in regenerative medicine [[65]18–[66]21]. For bone defect regeneration, 3D printing technology can create personalized scaffolds to promote cell migration and adhesion around the bone defect region by enabling precise design of the structural parameters of the scaffolds [[67]22]. In addition, it is well known that bone defect repair requires neovascularization to deliver oxygen and exchange metabolites, and therefore the ability to promote vascular regeneration is one of the critical properties of scaffolds required to induce bone formation [[68]23]. Scaffolds fabricated by 3D cryogenic printing possess a hierarchical porous structure [[69]24], which is similar to that of trabecular bone and has the capacity to promote vascular regeneration [[70]25]. Macrophages, which can be polarized into two major types–M1 and M2–are closely associated with neovascularization and bone formation in the process of bone defect repair [[71]26]. In the early stage of bone repair, macrophages polarize toward the M1 phenotype and secrete pro-inflammatory cytokines [[72]26]. In the late stage of bone repair, macrophage polarization is shifted toward the M2 phenotype which secrete anti-inflammatory cytokines and growth factors, promoting bone healing [[73]27]. The shift of macrophage polarization from the M1 to the M2 phenotype is essential for bone repair. Sleep has an immunoregulatory effect and sleep deprivation promotes the expression of inflammatory factors, which are associated with chronic inflammation [[74]28]. In recent years, MT, a sleep-inducing molecule, has been found to inhibit the expression of inflammatory cytokines and reduce the exaggerated inflammatory response to restore tissue function [[75]29]. A previous study found that MT enhances bone repair and increases bone strength by promoting the expression of angiogenic markers [[76]30]. MT promotes the proliferation and osteogenic differentiation of bone mesenchymal stem cells (BMSCs) by regulating the HGF/PTEN/Wnt/β-catenin pathway, thereby alleviating bone loss [[77]31]. Furthermore, MT upregulates the expression of antioxidant enzymes and reduces the levels of superoxide, thereby restoring mitochondrial function and improving the repair of bone defects [[78]32]. MT is a lipophilic molecule with a poor absorption rate when administered orally or by intravenous injection [[79]33]. In addition, its clinical application is limited because of poor oral bioavailability [[80]34]. To improve the bioavailability, several nanotechnology-based drug delivery systems have been developed for efficient loading of MT [[81]35]. However, these drug delivery systems also present limitations such as tedious synthetic steps, low production rates and small-scale production [[82]36]. Small extracellular vesicles (sEVs), which contain RNA and protein, originate from the endosomal compartment and activate a variety of signaling pathways to regulate cellular functions in many physiological processes [[83]37]. In addition, sEVs can be modified to carry specific sets of RNA or proteins, showing great advantages and application prospects as cargo vehicles for targeted drug delivery [[84]38]. Some studies have shown that engineered sEVs loaded with bioactive factors have promise for use in clinical applications, with a promotional effect on bone defect regeneration [[85]39]. Our previous study found that sEVs loaded with sleep-related circular RNA promoted chondrocyte proliferation and migration as well as showing potential for preventing osteoarthritis [[86]40]. However, it has not been elucidated whether MT-treated sEVs, in other words, sleep-inspired sEVs, facilitate bone defect regeneration. Surface functionalization can promote the targeting capacity of sEVs [[87]41]. Distearoyl phosphatidylethanolamine-polyethylene glycol (DSPE-PEG), an amphiphilic molecule, can be conjugated to targeting ligands for surface functionalization of sEVs to enable molecular interaction. Inorganic pyrophosphate (PPi) is an excellent targeting ligand due to its ability to bind hydroxyapatite [[88]42], and sEVs functionalized with DSPE-PEG-PPi may bind to scaffolds containing hydroxyapatite. Additionally, it has also been reported that PPi promotes osteogenic differentiation while downregulating adipogenic differentiation [[89]43]. In this study, we used 3D cryogenic printing technology to print scaffolds containing PLGA and uW (PL-uW scaffold) and then investigated the morphology and biocompatibility of the PL-uW scaffold. Sleep-inspired sEVs (MT-sEVs) were obtained from MT-stimulated fibroblast-like synoviocytes. We used DSPE-PEG-PPi to functionalize the MT-sEVs (PPi-MT-sEVs) so that PPi-MT-sEVs could bind to a PL-uW scaffold (to form a PL-uW@PPi-MT-sEV scaffold). RNA sequencing was performed to explore the molecular mechanisms. The effects of PPi-MT-sEVs on osteogenesis, angiogenesis and the regulation of macrophage phenotype were investigated by in vitro and in vivo experiments. Materials and methods Details of the materials and methods are provided in Supplemental Information. Results Characterization of 3D cryo-printed scaffolds A schematic of the overall experimental strategy is shown in Fig. [90]1. Previous studies demonstrated that uW were hundreds of micrometers in length and tens of nanometers in diameter [[91]44]. Our results also confirmed the high aspect ratio of uW using scanning electron microscopy (SEM) and transmission electron microscopy (TEM), as shown in Fig. [92]2A. PLGA and PL-uW scaffolds were then printed using a 3D cryogenic printer. The surface and internal structure of the scaffolds were observed by SEM, and the micrographs showed that both scaffolds had regular shapes with specific parameters as initially set (Fig. [93]2B). In addition, interconnected microporous architecture was observed inside the scaffolds printed using 3D cryogenic printing technology, forming connected pipelines to facilitate cell migration and transmission of nutrients [[94]45]. The images showed that there were smaller pores on the pore walls of the microporous structure (Fig. [95]2B). To verify the presence of uW, energy dispersive spectroscopy (EDS) was performed. The results of EDS point analysis showed that Ca and P were present on the surface of a PL-uW scaffold but not a PLGA scaffold, indicating the presence of uW in the PL-uW scaffold (Fig. [96]2C). Fig. 1. [97]Fig. 1 [98]Open in a new tab Schematic illustration of the process of preparing PPi-MT-sEVs and treatment of the PL-uW@PPi-MT-sEV scaffold. (A) Printing of the PL-uW scaffold using 3D cryogenic printing technology. (B) Modification of PPi-MT-sEVs. MT was used to produce MT-sEVs, which were then surface-functionalized by DSPE-PEG-PPi (PPi-MT-sEVs). (C) The PL-uW@PPi-MT-sEV scaffold facilitated vascularized bone regeneration based on angiogenesis–osteogenesis coupling and modulation of the macrophage phenotype. In the early stage, PPi-MT-sEVs not only promoted cell migration, proliferation and osteogenic differentiation, but also inhibited M1 polarization; in the late stage, PPi-MT-sEVs promoted cell proliferation, osteogenic differentiation and M2 polarization. Illustration credit: Lina Cao Fig. 2. [99]Fig. 2 [100]Open in a new tab Characterization of porous scaffolds printed using 3D cryogenic printing. (A) Representative SEM and TEM micrographs of uW. Scale bars of SEM images are 10 μm and 2 μm. For TEM images, scale bars are 2 μm and 400 nm. (B) Representative surface and cross-sectional SEM images of PLGA and PL-uW scaffolds. For surface images, scale bars are 600 μm, 60 μm and 15 μm, respectively. For cross-sectional images, scale bars are 40 μm, 20 μm and 10 μm, respectively. (C) Results of energy-dispersive spectroscopy (EDS) analysis of PLGA and PL-uW scaffolds Scaffolds produced by 3D cryogenic printing consisted of extruded and deposited bioink filaments. Both global and local (microstructure of deposited layer) 3D reconstructions of PLGA and PL-uW scaffolds were produced by micro-CT and are indicated in blue. The pore 3D reconstruction of PLGA and PL-uW scaffolds are shown in pseudocolors to distinguish micropores with different diameters (Fig. [101]S1). The results of micro-CT analysis showed that the total (including macropore and micropore) and local (micropore only) porosity of the PLGA scaffold was 75.55% and 59.49%, respectively, while the total and local porosity of the PL-uW scaffold was 72.91% and 53.03% (Fig. [102]S1). Further, the compressive modulus of the PL-uW scaffold (mean = 4.654 MPa, n = 3) was higher than that of the PLGA scaffold (mean = 2.752 MPa, n = 3), which suggested that uW enhanced the mechanical strength of the PL-uW scaffold (Fig. [103]S2). Biocompatibility of scaffolds In order to assess the biocompatibility of scaffolds, human microvascular endothelial cells (HMEC-1) were seeded onto scaffolds and cultured for 3 d. Cell adhesion on both PLGA and PL-uW scaffolds was observed using SEM, and the results revealed that HMEC-1 grew well on both types of scaffolds (Fig. [104]3A). Live/Dead assay was performed to evaluate the viability of HMEC-1 (Fig. [105]3B), and demonstrated that HMEC-1 grew on the surface of the scaffolds, and were able to survive for a long time on PLGA and PL-uW scaffolds, with the addition of uW having no effect on cell activity. These results demonstrated that both PLGA and PL-uW scaffolds have good biocompatibility. Fig. 3. [106]Fig. 3 [107]Open in a new tab Biocompatibility of the scaffolds. (A) SEM images of co-culture experiments. The images show HMEC-1 cultured on PLGA and PL-uW scaffolds. Scale bar: 100 μm in low magnification, 10 μm in high magnification. (B) Fluorescence images of calcein-AM (green) and propidium iodide (red) captured under confocal microscopy. Scale bar: 200 μm Adsorption of sEVs to the scaffolds by surface functionalization DSPE-PEG, an amphiphilic material, is widely used in drug delivery, and can be conjugated to different molecules [[108]46]. PPi has high affinity for hydroxyapatite because phosphate and hydroxyl groups can bind to the cluster of calcium ions [[109]47]. DSPE-PEG-PPi was used to modify the surface of MT-sEVs, and its structural formula is shown in Fig. [110]4A. To provide a clear view of the binding, hydroxyapatite and PPi-terminus of DSPE-PEG-PPi were modeled using Materials Studio (Biovia, San Diego, CA, USA) (Fig. [111]4B). The structure and differential electron charge density were calculated to obtain the charge transfer at the interface. The results demonstrated that the electron cloud was shifted in the interfacial region after PPi and hydroxyapatite came into contact (Fig. [112]4C). All these results supported the hypothesis that DSPE-PEG-PPi had the ability to bind to hydroxyapatite. Fig. 4. [113]Fig. 4 [114]Open in a new tab PPi-MT-sEVs coated onto scaffolds. (A) Chemical structure of DSPE-PEG-PPi polymers. (B) 3D charge density difference before and after contact between hydroxyapatite and PPi-terminus of DSPE-PEG-PPi based on density functional theory calculations. (C) The z-axis plane-averaged electron charge density difference of hydroxyapatite/PPi-terminus. (D) The adsorption of different sEVs labeled with DiI onto the PLGA and PL-uW scaffolds. Scale bar: 100 μm. (E) A schematic illustration of PPi-MT-sEVs. (F) A schematic illustration of PL-uW@PPi-MT-sEVs. Illustrations were created using BioRender.com The isolated sEVs were divided into four groups: (I) The sEV group: sEVs directly isolated from the supernatant; (II) The MT-sEV group: sEVs isolated from the supernatant of MT-stimulated cells; (III) The PPi-sEV group: sEVs isolated from the supernatant and functionalized by DSPE-PEG-PPi; (IV) The PPi-MT-sEV group: sEVs isolated from the supernatant of MT-stimulated cells and functionalized with DSPE-PEG-PPi. The isolated sEVs were characterized using cryo-TEM, atomic force microscopy (AFM) and dynamic light scattering (DLS). The morphology of sEVs was observed by cryo-TEM prior to or after surface functionalization and the results demonstrated that spheroid sEVs possessed a compact membrane structure (Fig. [115]S3). The AFM topography images showed that the isolated sEVs had a distinct rounded morphology without aggregates. The size distribution of sEVs was measured by DLS and the results showed that the particle size of different sEVs was in the range of 30–200 nm (Fig. [116]S4), which was consistent with previous reports [[117]48]. PLGA and PL-uW scaffolds were placed in 24-well plates and different sEVs labeled with DiI (a lipophilic fluorescent dye) were added. The results of adsorption experiments are shown in Fig. [118]4D. As can be seen, no sEVs attached to the PLGA scaffold regardless of whether the surface was functionalized by DSPE-PEG-PPi or not. PPi-sEVs and PPi-MT-sEVs were attached to the PL-uW scaffold whereas sEVs and MT-sEVs were not attached to the PL-uW scaffold. A schematic illustration of the process of PPi-MT-sEV production is provided in Fig. [119]4E, while Fig. [120]4F shows the binding process of PPi-MT-sEVs to the PL-uW scaffold. Bioinformatic analysis of the influence of MT on gene expression BMSCs were divided into two groups according to whether MT was added to the culture medium. RNA sequencing (RNA-seq) was performed to identify differentially-expressed genes (DEGs) between the MT-treated group and the control group (Fig. [121]5A). KEGG, Reactome, Wikipathway and GO pathway enrichment analysis were performed to identify some signaling pathways and the data were visualized as an interaction network by Cytoscape ([122]https://cytoscape.org/) (Fig. [123]5B–E, Fig. [124]S5–[125]S6): (I) “Osteoblast differentiation and related diseases”, “WNT ligand biogenesis and trafficking” and “Hippo signaling pathway”, implied that MT might have an influence on osteogenic/adipogenic differentiation [[126]49]; (II) “Phosphatidylinositol-3,4,5-trisphosphate binding”, “Protein kinase B signaling pathway” and “Phosphatidylinositol-4,5-bisphosphate binding”, suggested that MT might have some influence on angiogenesis and cell proliferation [[127]50]; (III) “Cytoskeletal anchor activity”, suggested that MT might have an influence on cell migration [[128]51]; (IV) “Cytokine–cytokine receptor interaction” and “Cytokine-mediated signaling pathway”, implied that MT might have an influence on the interplay between cells and their microenvironment [[129]52]; (V) “Cyclooxygenase pathway”, “Peroxidase activity”, “NAD(P)H oxidase H[2]O[2]-forming activity” and “Cytokines and inflammatory response signaling pathway”, suggested that MT might have some influence on inflammatory and anti-inflammatory responses [[130]53]. Based on these results of bioinformatic analysis, observation of the effects of PPi-MT-sEVs on cell proliferation, migration, osteogenic/adipogenic differentiation and macrophage polarization seemed a promising investigation. Fig. 5. [131]Fig. 5 [132]Open in a new tab RNA sequencing and bioinformatics analysis. (A) Heatmap of clustering analysis. (B–E) The interaction networks identified by KEGG (B), Reactome (C), Wikipathway (D) and GO (E) pathway enrichment analysis Osteogenic and adipogenic modulation by PPi-MT-sEVs Studies have found that MT promotes osteogenic differentiation of MSCs, with the gene expression levels of osteogenic differentiation markers increased [[133]54]. To investigate the effect of PPi-MT-sEVs on osteogenic differentiation, BMSCs were cultured in osteogenic induction medium with four different sEVs as previously described. Alkaline phosphatase (ALP) is an early-stage enzyme marker of osteoblasts, and ALP staining was performed after 7 d of osteogenic differentiation. As shown in Fig. [134]6A and B, BMSCs cultured with PPi-MT-sEVs or MT-sEVs exhibited the highest expression levels of ALP, while the control group exhibited the least ALP activity. Alizarin red staining was performed to examine calcium deposition after 14 d of osteogenic induction, and the results demonstrated that the four groups cultured with sEVs developed more calcium nodules than the control group (Fig. [135]6C). Notably, the MT-sEV and PPi-MT-sEV groups displayed the most mineral deposition (Fig. [136]6D). BMSCs possess the capacity to differentiate into adipocytes and MT inhibits adipogenic differentiation [[137]55]. Oil red O staining was conducted to investigate the effect of PPi-MT-sEVs on adipogenic differentiation after 21 d of induction. As shown in Fig. [138]6E, the MT-sEV and PPi-MT-sEV groups contained fewer lipid droplets than the control group. The results of reverse transcription quantitative PCR (RT-qPCR) experiments indicated that PPi-MT-sEVs promote the expression of osteogenic markers, including type I collagen (COL I), ALP, and runt-related transcription factor 2 (RUNX2) (Fig. [139]6F). These results demonstrated that MT-sEVs and PPi-MT-sEVs exhibited the capacity to promote osteogenic differentiation while suppressing adipogenic differentiation. Fig. 6. [140]Fig. 6 [141]Open in a new tab Effect of EVs on cell differentiation. (A) ALP staining. BMSCs were cultured in osteogenic medium for 7 d before ALP staining was performed. Scale bar: 400 μm. (B) Relative ALP activity (versus control group). (C) Alizarin red staining. Alizarin red staining was performed after 14 d of osteogenic differentiation. Scale bar: 400 μm. (D) Relative mineral deposition (versus control group). (E) Oil Red O staining. Oil Red O staining was performed after 21 d of adipogenic differentiation. Scale bar: 400 μm. (F) RT-qPCR results of osteogenic markers. *p < 0.05 compared with control group. **p < 0.01 compared with control group. ***p < 0.001 compared with control group. ****p < 0.0001 compared with control group Effects of PPi-MT-sEVs on cell proliferation, cell migration and angiogenesis MT facilitates cell proliferation and inhibits apoptosis through multiple signaling pathways [[142]56]. EdU cell proliferation assay was performed to investigate the effect of sEVs on the cell proliferation of HMEC-1. Flow cytometry showed that EdU-positive cells in the MT-sEV (62.0%) and PPi-MT-sEV (60.5%) groups were significantly increased compared with the control (50.0%), sEV (51.2%) and PPi-sEV (54.4%) groups (Fig. [143]7A). Fig. 7. [144]Fig. 7 [145]Open in a new tab The effects of PPi-MT-sEVs on cell proliferation, cell migration and the formation of tubule-like structures. (A, B) Flow cytometry results of EdU cell proliferation assay in HMEC-1 (A) and BMSCs (B). (C, D) Transwell migration experiments of BMSCs (C) and HMEC-1 (D). Scale bar: 500 μm. (E) Representative images of angiogenesis assay. Scale bar: 500 μm. (F) Schematic illustration of the Transwell migration assay. (G) Schematic illustration of the effects of PPi-MT-sEVs on bone and neovascular regeneration. Illustrations were created using BioRender.com We further explored the role of PPi-MT-sEVs on cell proliferation of BMSCs. As shown in Fig. [146]7B, flow cytometry indicated that EdU-positive BMSCs in the MT-sEV and PPi-MT-sEV groups accounted for 15.9% and 16.1% of total cells, a higher proportion than in the control (8.27%), sEV (9.08%) or PPi-sEV groups (12.5%). Cell migration to the bone defect region is an important factor that affects bone regeneration, and MT has been shown to enhance cell migration [[147]30]. A Transwell migration assay was performed to evaluate the impact of PPi-MT-sEVs on cell migration of BMSCs and HMEC-1. The results, shown in Fig. [148]7C and D, indicated that significantly more cells migrated to the lower chambers in the PPi-MT-sEV and MT-sEV groups than in the control group. The number of migrated BMSCs in the MT-sEV and PPi-MT-sEV groups was approximately 2.94 times and 3.13 times higher than the control group, respectively. Similarly, the migration of HMEC-1 in the MT-sEV and PPi-MT-sEV groups was approximately 2.74 times and 2.96 times higher than the control group (Fig. [149]S7). These results showed that PPi-MT-sEVs promoted the migratory capacity of BMSCs and HMEC-1. Neovascularization is favorable for the transmission of nutrients and cytokines, which is important for bone regeneration [[150]57]. In order to explore the influence of PPi-MT-sEVs on angiogenesis, HMEC-1 adhered on Matrigel were cultured in cell medium with the different sEVs. As can be seen in Fig. [151]7E and Fig. [152]S8, after 6 h, there were more tubule-like structures formed in the MT-sEV and PPi-MT-sEV groups compared with the control group. This result indicated that PPi-MT-sEVs have a promotional effect on angiogenesis, which is beneficial for bone defect repair. A schematic illustration of the Transwell assay is provided in Fig. [153]7F, while Fig. [154]7G shows a schematic representation of the promotional effects of PPi-MT-sEVs on bone regeneration and neovascularization. Modulation of macrophage polarization by PPi-MT-sEVs Immunofluorescent staining was performed to investigate the effect of PPi-MT-sEVs on macrophage polarization. Tumor necrosis factor α (TNFα), C-C motif chemokine ligand 3 (CCL3), and inducible nitric oxide synthase (iNOS) are M1 macrophage markers, while arginase 1 (ARG1), suppressor of cytokine signaling 2 (SOCS2), transforming growth factor β (TGFβ), and CD206 are markers of M2 macrophages. The results showed that the MT-sEV and PPi-MT-sEV groups expressed lower levels of iNOS (Fig. [155]8A and Fig. [156]S9). Meanwhile CD206 expression showed the opposite trend among these groups, with CD206 expression in the MT-sEV and PPi-MT-sEV groups being significantly greater than in the other groups (Fig. [157]8B and Fig. [158]S10). Schematic illustrations of the effect of PPi-MT-sEVs on macrophage polarization are provided in Fig. [159]8C and D. RT-qPCR experiments were performed to detect the expression of M1 and M2 macrophage markers. The MT-sEV and PPi-MT-sEV groups showed decreased expression of M1 markers (iNOS, TNFα, CCL3) and increased expression of M2 markers (ARG1, SOCS2, TGFβ) (Fig. [160]S11). These results showed that PPi-MT-sEVs induced a shift in macrophage polarization from the M1 to the M2 phenotype, preventing excessive inflammation and promoting tissue repair. Fig. 8. [161]Fig. 8 [162]Open in a new tab The effect of PPi-MT-sEVs on macrophage polarization. (A) Immunofluorescent images of M1 macrophage polarization stained to show iNOS (red) and nuclei (blue). Scale bar: 100 μm. (B) Immunofluorescent images of M2 macrophage polarization stained to show CD206 (green) and nuclei (blue). Scale bar: 100 μm. (C) Schematic illustration showing the effect of PPi-MT-sEVs on M1 polarization. (D) Schematic illustration showing the effect of PPi-MT-sEVs on M2 polarization. Schematic illustrations were created using BioRender.com The ability of the PL-uW@PPi-MT-sEV scaffold to repair bone defects in vivo The effects of the PL-uW@PPi-MT-sEV scaffold on bone regeneration and revascularization were evaluated in a rat cranial defect model. As shown in Fig. [163]9A, there was only a small amount of neovascularization in the defect region of the PLGA, PL-uW and PL-uW@PPi-sEV groups. More neovascularization was observed in the PL-uW@PPi-MT-sEV group, revealing that PPi-MT-sEVs have a strong pro-angiogenic effect. 3D reconstruction and pseudocolor processing of cross-sections were performed with reference to previous studies [[164]58–[165]60]. The results (Fig. [166]9B and C) indicated that there was almost no new bone formation in the PLGA group. A limited number of bone islands were formed in the PL-uW and PL-uW@PPi-sEV groups. In the PL-uW@PPi-MT-sEV group, a large number of bone islands were generated, and the density of the regenerated bone was close to that of native bone. Fig. 9. [167]Fig. 9 [168]Open in a new tab Effects on bone regeneration and vascularization in vivo. (A) Micro-CT images of neovascularization. (B) Micro-CT 3D images of the defect areas. (C) Micro-CT sagittal images presented in a pseudocolor mode with distinct colors indicating different gray values The effects of the PL-uW@PPi-MT-sEV scaffold on bone defect regeneration were further assessed by histological staining. H&E staining showed that little new bone was formed in the PLGA group (Fig. [169]10A), and there was limited bone regeneration within the defect area in the PL-uW and PL-uW@PPi-sEV groups. A noteworthy difference was observed in the bone defect area of the PL-uW@PPi-MT-sEV group, where a substantial amount of new bone formation was evident, indicating that PL-uW@PPi-MT-sEVs had a pro-osteogenic effect. Masson’s staining showed that there were significantly more collagen fibers in the PL-uW@PPi-MT-sEV group compared with the other groups (Fig. [170]10B). In addition, the collagen fibers were interconnected throughout the whole defect region of the PL-uW@PPi-MT-sEV group. The result of COL I staining was consistent with that of Masson’s staining (Fig. [171]10C). The PL-uW@PPi-MT-sEV group exhibited significantly higher expression of COL I in the defect region, indicating the capability of PL-uW@PPi-MT-sEVs to facilitate bone defect repair (Fig. [172]S12). In addition, the highest expression of osteocalcin (OCN) was observed in the PL-uW@PPi-MT-sEV group (Fig. [173]10D). Immunofluorescent staining of CD206 and iNOS demonstrated that the PL-uW@PPi-MT-sEV group had the greatest capacity to inhibit M1 polarization and promote M2 polarization compared with the other groups (Fig. [174]10E and F). In summary, Fig. [175]1 depicts the primary principle that the PL-uW@PPi-MT-sEV scaffold promotes vascularized bone regeneration via angiogenesis–osteogenesis coupling and modulation of the macrophage phenotype. Fig. 10. [176]Fig. 10 [177]Open in a new tab Histological analysis of bone defect repair in a rat model. (A) H&E and (B) Masson’s trichrome staining of the bone defect area implanted with different scaffolds. Low magnification scale bar: 1 mm and high magnification scale bar: 200 μm. (C) Immunohistochemical staining of COL I. Low magnification scale bar: 1 mm and high magnification scale bar: 200 μm. (D) Immunofluorescence staining of OCN (green) and nuclei (blue). Scale bar: 100 μm. (E) Immunofluorescence staining of iNOS (green) and nuclei (blue). Scale bar: 100 μm. (F) Immunofluorescence staining of CD206 (green) and nuclei (blue). Scale bar: 100 μm Discussion Bone defect healing is a complex process that requires appropriate substitute materials and a pro-regenerative microenvironment [[178]61]. 3D-printed scaffolds have a precise structure and can be personalized to meet the needs of bone regeneration, and thus provide a potential tailored treatment strategy [[179]62]. Many synthetic materials have been used for 3D-printed scaffolds [[180]63]. In this study, we used PLGA and uW to print scaffolds. Cells adhered to scaffolds were in a good state of growth demonstrating that the PL-uW scaffold was an ideal substrate for bone defect regeneration due to its excellent biocompatibility. Porosity is the focus of scaffold design and high porosity facilitates cell adhesion, cell migration and the exchange of metabolites. The pores of 3D-printed scaffolds can be classified into two types: macropores and micropores. Macropores of appropriate diameter improve the osteogenic capacity of scaffolds [[181]64]. Scaffolds with uniform pore sizes of 378–435 μm are known to promote better cell proliferation, neovascularization and osteogenic differentiation than scaffolds without macropores [[182]65]. Hence, we designed scaffolds with 400 μm diameter macropores in this work. The presence of micropores is one of the main requirements for scaffolds. Some studies demonstrated that 3D cryo-printed scaffolds had high porosity and interior micropores, which provided a suitable microenvironment for cell growth [[183]66]. Porous scaffolds with controllable micropores have been shown to promote cellular adhesion and bone formation [[184]45]. Herein, we printed a PL-uW scaffold using 3D cryogenic printing technology. Our study suggested that the PL-uW scaffold had a hierarchical porous structure with interconnected micropores, and cells adhered well to the surface of the PL-uW scaffold. In addition, the PL-uW scaffold had high mechanical strength, another property essential for bone regeneration [[185]67]. Compared with bone tissue, 3D cryo-printed scaffolds possess a structure resembling bone microarchitecture but lack substantial amounts of bioactive factors [[186]68]. Thus, supplementation with bioactive factors is one of the effective approaches to improve the osteogenic induction capacity of scaffolds. Bone defect repair is a complex and dynamic process involving osteogenic differentiation, cell proliferation, angiogenesis and coordination of the inflammatory response [[187]69]. Consequently, bioactive factors with extensive effects are urgently needed for bone defect healing [[188]70]. Circadian rhythms, one of the physiological human rhythms, regulate multiple cellular functions and have important implications for tissue homeostasis and repair [[189]71]. Circadian genes affect the regulation of bone formation and bone resorption through their impact on osteoblast functionality as well as intercellular communication [[190]72]. Circadian disruption has adverse effects on bone mineral density and strength of the skeleton [[191]73]. Based on this background, we combined 3D cryo-printed scaffolds with bioactive factors related to the circadian rhythm and investigated whether the combination could enhance bone defect regeneration. MT is released by the pineal gland and implicated in regulation of the circadian rhythm, which has been proven to be a regulator of bone formation [[192]74]. It has been demonstrated that MT induces osteogenic differentiation and inhibits adipogenic differentiation of stem cells [[193]75]. In addition, MT enhances anti-inflammatory polarization of macrophages and reduces tissue inflammation [[194]76]. Functionally, MT is an apparent bioactive factor for bone defect regeneration. We further considered how to combine MT with hierarchical porous scaffolds printed using 3D cryogenic printing technology. Modularized sEVs have attracted attention in the field of bone tissue engineering as a drug delivery system [[195]77]. The lipid bilayer membrane of sEVs can be modified with targeting ligands while pretreatment of cells regulates the internal contents of sEVs [[196]78]. It has been demonstrated that sEVs carrying specific microRNAs promote osteogenic differentiation of BMSCs and enhance bone regenerative ability [[197]79]. In our previous study, chondrocytes were cultured with MT and a sleep-related circular RNA was identified by RNA sequencing. These sEVs combined with the sleep-related circular RNAs have been proven to be beneficial in preventing osteoarthritis progression [[198]40]. Herein, we further isolated MT-sEVs, sleep-inspired sEVs, from the supernatant of MT-stimulated cells. RNA sequencing was performed and we found that MT influenced multiple signaling pathways involved in osteogenic differentiation, angiogenesis and inflammation, which revealed the potential of PPi-MT-sEVs to enhance bone defect regeneration. Immune modulation is an essential component of bone defect healing [[199]69]. M1 macrophages, which are pro-inflammatory, promote bone defect repair at the inception, but long-term M1 macrophage infiltration can result in delayed healing [[200]26], while anti-inflammatory M2 macrophages play an essential role in bone regeneration due to their capacity to promote tissue repair and angiogenesis [[201]80]. It has been demonstrated that facilitating M2 phenotype polarization promotes recruitment and osteogenic differentiation of MSCs. In addition, anti-inflammatory cytokines secreted by M2 macrophages contribute to bone repair [[202]81]. In this study, we demonstrated that PPi-MT-sEVs promoted macrophage polarization towards the M2 phenotype and inhibited M1 polarization in vitro. In addition, the PL-uW@PPi-MT-sEV scaffold induced M2 polarization to facilitate bone defect repair in vivo. Newly-formed vessels have the ability to deliver nutrients to, and clear metabolites from, bone, which is another important factor in bone regeneration [[203]82]. MT has the ability to induce M2 polarization, which has been proven to be beneficial for angiogenesis [[204]83]. In addition, the migration of endothelial cells is one of the important prerequisites for angiogenesis [[205]84]. In this study, we printed porous scaffolds incorporating various sEVs and our results showed that PPi-MT-sEVs promoted M2 polarization, cell migration and the formation of tubule-like structures in vitro. We also demonstrated that the PL-uW@PPi-MT-sEV scaffold induced the formation of more neovessels in the bone defect region in vivo compared to other groups. Cell migration to the defect site and osteogenic differentiation of BMSCs have a significant effect on bone defect healing [[206]85]. BMSCs have the potential to differentiate into adipocytes and osteoblasts [[207]86]. Differentiation of BMSCs to adipocytes may lead to bone loss and delay bone regeneration [[208]87]. Herein, for the first time, our research indicated that PPi-MT-sEVs have the ability to enhance osteogenic differentiation and inhibit adipogenic differentiation. In addition, our research indicated that PPi-MT-sEVs have the ability to enhance cell migration, facilitating bone repair in a calvarial defect model. sEVs can be loaded onto scaffolds in several different ways depending on the materials and printing technique [[209]88]. sEVs can be adsorbed onto the surface of positively-charged scaffolds due to the negative potential of the membrane [[210]89]. In addition, sEVs can be directly encapsulated into a hydrogel which preserves their biological activity due to its water solubility [[211]90]. Notably, PLGA is usually dissolved in organic solvents, which might disrupt the bioactivity of sEVs, and the scaffolds also experience a dramatic change in temperature during the 3D cryogenic printing process [[212]91]. The modularized EVs provide a solution to this problem, and the membrane of sEVs can be modified via targeting ligands for binding ability [[213]38]. Thus, we considered binding sEVs to scaffolds via surface modification. The lipid bilayer of sEVs can be modified by amphiphilic molecules. DSPE-PEG is an amphiphilic molecule that includes a lipid tail and can be conjugated to various ligands [[214]92]. In this work, we combined DSPE-PEG with PPi and used DSPE-PEG-PPi to modify the MT-sEVs, enhancing the binding ability of PPi-MT-sEVs to the PL-uW scaffold. The uW provided binding sites for modified PPi-MT-sEVs and the porosity improved the loading efficiency. Quantum chemical simulation was performed to confirm the interaction between PPi and uW, and adsorption experiments demonstrated that adsorptive interactions existed between the PL-uW scaffold and PPi-MT-sEVs. Conclusion Briefly, these results showed that PPi-MT-sEVs have the ability to promote cell proliferation, migration and angiogenesis. In addition, PPi-MT-sEVs regulate the macrophage M1/M2 polarization shift and osteogenic/adipogenic differentiation. Our work is the first to report that sleep-inspired sEVs can be modified with DSPE-PEG-PPi to incorporate into hierarchical porous scaffolds printed using 3D cryogenic printing technology. The in vivo experiments demonstrated that the combination of PPi-MT-sEVs and porous scaffolds had the therapeutic effects of osteogenesis–angiogenesis coupling and macrophage phenotype regulation, promoting bone defect regeneration. In summary, our study highlights the pivotal roles of a PL-uW@PPi-MT-sEV scaffold for bone defect regeneration, which we believe represents a novel therapeutic strategy for the treatment of bone defects. Electronic supplementary material Below is the link to the electronic supplementary material. [215]Supplementary Material 1^ (17.1MB, docx) Acknowledgements