Abstract
Repairing large bone defects remains a major challenge, as current
strategies—including autografts, allografts, and tissue‐engineered
scaffolds—are limited by donor shortage, suboptimal biocompatibility,
and insufficient control of the regenerative microenvironment. Bone
organoids offer a promising strategy by simulating bone tissue
functions and microenvironments in vitro, showing great potential for
bone repair. However, current bone organoids still face issues such as
insufficient oxygen supply, and limited immune modulation, reducing
their effectiveness for complex bone defect repair, especially when
precise shape conformity is required. To address these challenges, bone
organoid unit based on silk hydrogel microspheres is developed,
introducing oxygen‐releasing components and immune cells to better
mimic the bone repair environment and enable conformal filling of
irregular defects. In vitro, the system formed bone organoid units
through 28 days culture that sustained cell viability, promoted M2
macrophage polarization, and enhanced osteogenesis and angiogenesis. In
a critical‐sized mouse cranial defect model, this strategy outperformed
traditional approaches, showing superior bone regeneration and tissue
integration. These findings indicate that the strategy effectively
addresses key obstacles in bone repair, such as oxygen supply, immune
modulation, and shape conformity, providing a promising solution for
personalized bone regeneration.
Keywords: bone organoid, bone regeneration, macrophage,
osteoimmunomodulation, oxygen release
__________________________________________________________________
Oxygen‐releasing immunomodulatory microspheres are used to construct
conformal bone organoid units that enhance osteogenesis, angiogenesis,
and immune regulation. By integrating BMSCs, macrophages, and CaO₂‐HAp
into a SilMA‐based hydrogel platform, this strategy provides sustained
oxygen supply and anti‐inflammatory microenvironments, leading to
effective bone regeneration and personalized repair of critical‐sized
bone defects.
graphic file with name ADVS-12-e01437-g009.jpg
1. Introduction
Critical‐size bone defects, caused by trauma, tumor resection, or
osteoporosis, are often associated with prolonged recovery periods and
functional impairments, which can increase the risk of nonunion.^[
[48]^1 ^] These defects remain a major challenge in orthopedic clinical
practice and a prominent research focus in regenerative medicine. While
traditional methods of bone repair, such as metal implants, autologous
bone grafts, and allografts, provide partial solutions, each comes with
significant limitations.^[ [49]^2 ^] Metal implants offer mechanical
stability but struggle to integrate well with surrounding bone tissue,
leading to complications such as infection and poor bone integration.^[
[50]^3 ^] Autologous and allogeneic bone grafts, while biologically
active, face challenges related to donor site morbidity,^[ [51]^4 ^]
immune rejection,^[ [52]^5 ^] and limited material availability,^[
[53]^6 ^] particularly when addressing large‐scale bone defects.^[
[54]^7 ^] Therefore, there is an urgent need for more efficient,
flexible, and personalized bone repair strategies that can promote bone
regeneration, especially for critical‐sized bone defects.
With the advancement of regenerative medicine, organoid technology has
emerged as a promising solution and has garnered significant
attention.^[ [55]^8 ^] Organoids are 3D, in vitro reconstructed
miniature organs that replicate the structure and function of human
tissues, significantly advancing tissue regeneration research. Recent
progress in soft tissue organoids, such as liver, neural, and
intestinal organoids, has opened new possibilities for disease modeling
and treatment.^[ [56]^9 ^] These soft tissue organoids have
demonstrated great potential in regenerative medicine, particularly in
drug screening, disease research, and tissue repair.^[ [57]^10 ^]
However, constructing bone tissue, a hard tissue, presents unique
challenges that differ significantly from soft tissues. Bone tissue
requires not only mechanical support and load‐bearing capacity but also
high biological activity, including osteogenesis and angiogenesis.^[
[58]^11 ^] Traditional organoid construction typically relies on
natural matrix gels (such as Matrigel) extracted from mice, but these
materials have several drawbacks, including high cost, undefined
composition, batch‐to‐batch variability, and high immunogenicity.^[
[59]^12 ^] Additionally, their mechanical properties are insufficient
to replicate the mechanical characteristics of bone tissue. Recent
advances in biomaterials have prompted researchers to explore more
suitable alternatives, such as silk fibroin (SilMA) hydrogels, gelatin,
and polylactic acid, which offer unique advantages in biocompatibility,
mechanical properties, and versatility, providing novel strategies for
bone organoid construction.^[ [60]^13 ^] SilMA is a photocrosslinkable
hydrogel derived from natural silk fibroin, widely recognized for its
biocompatibility, mechanical tunability, and mild gelation conditions.
It enables the formation of stable, cell‐friendly 3D scaffolds that
support the survival and differentiation of various cell types. SilMA
also exhibits good permeability to nutrients and oxygen, and its
degradation products‐amino acids, peptides, and low‐molecular‐weight
proteins are naturally metabolized in vivo, reducing cytotoxicity and
inflammation risks. These features make SilMA an ideal base material
for constructing bone organoids, providing both structural support and
a favorable microenvironment for osteogenesis and immune regulation.^[
[61]^14 ^]
Building on previous work in bone organoid engineering, including the
use of HAp‐based hybrid bioinks,^[ [62]^15 ^] dynamic DNA‐hydrogel
systems,^[ [63]^16 ^] and ECM‐DNA‐CPO‐based bionic matrices^[ [64]^17
^] to support osteogenic differentiation and spatial organization, our
team has demonstrated the feasibility of constructing large‐scale
bone‐like tissue in vitro and applying it to bone defect models with
promising outcomes. However, significant limitations remain that hinder
the translational potential of current bone organoid models. One major
challenge is the lack of a sufficient oxygen supply during long‐term in
vitro culture and after implantation. Bone is a highly vascularized
tissue, and oxygen is essential for maintaining cell viability,
promoting osteogenic differentiation, and supporting extracellular
matrix mineralization.^[ [65]^18 ^] Most bone organoid constructs to
date do not incorporate functional vasculature or oxygen delivery
systems, resulting in hypoxic microenvironments that impair tissue
maturation and regenerative capacity.^[ [66]^19 ^] Another key
limitation is the absence of immune components. Recent studies have
highlighted the essential role of macrophage‐mediated immune modulation
in bone healing, particularly the polarization toward the M2 phenotype,
which supports angiogenesis and suppresses inflammation.^[ [67]^20 ^]
However, current bone organoids typically exclude immune‐regulatory
elements, overlooking their synergistic role in orchestrating tissue
regeneration. Finally, existing bone organoid platforms are mostly
based on rigid scaffolds or printed constructs that lack geometric
adaptability, limiting their ability to conform to complex,
patient‐specific bone defects. These challenges collectively restrict
the clinical translation of bone organoid technology and underscore the
urgent need for integrated solutions that address oxygenation, immune
modulation, and morphological flexibility.
To address these challenges, this study proposes an innovative
microfluidic hydrogel microsphere strategy to construct bone organoid
units capable of sustained oxygen release, immune modulation, and
conformal repair (Figure [68]1 ). By utilizing microfluidic technology,
silk fibroin hydrogel is employed as a supporting material to
encapsulate calcium peroxide (CaO₂) particles, bone marrow mesenchymal
stem cells (BMSCs), and macrophages (RAW264.7) within microspheres.
This approach not only ensures continuous oxygen release, alleviating
the hypoxic conditions in bone organoids but also promotes osteogenesis
and controls inflammation through immune modulation. Additionally, this
strategy allows precise control over the size, shape, and composition
of the microspheres, enabling the creation of shape‐matched bone
organoid units tailored for various bone defect sites and sizes. By
filling customized bone organoid units, this system effectively mimics
natural bone healing processes, promoting osteogenesis, angiogenesis,
and immune modulation. In vitro experiments demonstrated significant
osteogenic mineralization and maintained immune regulatory functions
for up to 28 days. In vivo experiments showed that the bone organoid
units using this strategy significantly accelerated new bone formation
at critical‐sized cranial defects, enhanced angiogenesis, and reduced
inflammation, highlighting their potential for large bone defect
repair. This innovative strategy offers a personalized, customizable
approach to bone defect repair, with significant clinical application
potential. By precisely controlling the structure and function of bone
organoids and combining the needs for oxygen supply, immune modulation,
and shape‐matched repair, this approach presents a novel therapeutic
pathway for bone repair, paving the way for future clinical treatments
of large bone defects.
Figure 1.
Figure 1
[69]Open in a new tab
Schematic diagram of the design and application of oxygen‐releasing
immunomodulatory microspheres for bone organoid construction and bone
regeneration.
2. Results and Discussion
2.1. Preparation and Characterization of Hydrogel Microspheres and Oxygen
Releasing Materials
In addressing the multiple challenges of bone organoid construction,
including insufficient oxygen supply and the need for immune
modulation, we observed in previous studies that bone organoids undergo
rapid cell apoptosis at the center due to inadequate oxygen supply
during long‐term culture.^[ [70]^15 ^] Moreover, immune
microenvironment regulation is critical in bone repair.^[ [71]^21 ^] To
address these issues, we developed a microfluidic hydrogel microsphere
strategy capable of precisely controlling the size and shape of the
microspheres and effectively encapsulating oxygen‐releasing materials
to mitigate the detrimental effects of hypoxic environments on
osteogenesis. This strategy also incorporates immune‐modulatory factors
to further promote immune response control during bone repair.
The microfluidic platform was equipped with coaxial needles to produce
uniform hydrogel microspheres (Figure [72] 2a). The water phase,
containing silk fibroin methacryloyl and lithium phenyl
(2,4,6‐trimethylbenzoyl) phosphinate (LAP) was delivered through the
inner needle, while the outer needle transported the oil phase, which
encapsulated the aqueous droplets through shear force.^[ [73]^22 ^] By
carefully controlling the flow rate ratio between the aqueous and oil
phases, we successfully generated microspheres with an average diameter
of ≈622.6 ± 5.23 mm, as shown by particle size distribution analysis
(Figure [74]2b). Under 365 nm UV irradiation, the SilMA in the aqueous
phase underwent crosslinking with the LAP, resulting in the formation
of stable, porous hydrogel microspheres. This porous structure is
beneficial for the encapsulation of functional agents and the delivery
of nutrients (Figure [75]2c).
Figure 2.
Figure 2
[76]Open in a new tab
Characterization of hydrogel microspheres and oxygen‐releasing
materials. a) Optical microscopy images of SilMA microspheres. b) Size
distribution of SilMA microspheres. c) SEM image of SilMA microspheres
showing their porous structure. d,e) SEM images of CaO₂ and CaO₂‐HAp
particles, respectively. f) EDS mapping images of SilMA@CaO₂ and
SilMA@CaO₂‐HAp microspheres, with calcium (Ca) and phosphorus (P)
signals confirming the successful encapsulation of CaO₂‐HAp in the
microspheres. g) Force‐displacement test of SilMA microspheres and
SilMA@CaO₂‐HAp microspheres under compression. h) Oxygen release
profiles of SilMA@CaO₂ and SilMA@CaO₂‐HAp microspheres over 30 days
(n = 3). i) Degradation profiles of SilMA@CaO₂‐HAp microspheres tested
in PBS, DMEM, and type II collagenase (n = 3). j) Swelling behavior of
SilMA microspheres, SilMA@CaO₂, and SilMA@CaO₂‐HAp microspheres
(n = 3). k) Conformal assembly of SilMA@CaO₂‐HAp microspheres into
customizable shapes, including bone, heart, triangle, and rectangle.
To enable oxygen release, we synthesized CaO₂ and calcium
peroxide‐hydroxyapatite (CaO₂‐HAp) particles (Figure [77]2d,e), which
were then incorporated into the aqueous SilMA solution prior to
microsphere fabrication. This approach allowed for the encapsulation of
these particles within the hydrogel microspheres, providing sustained
oxygen release. Energy‐dispersive X‐ray spectroscopy (EDS) confirmed
the successful loading of CaO₂ and CaO₂‐HAp, as indicated by distinct
calcium (Ca) and phosphorus (P) signals within the microspheres
(Figure [78]2f). The Figure [79]2g demonstrates the compressive
deformation and force‐displacement relationship of SilMA microspheres
(left) and SilMA microspheres encapsulating CaO₂‐HAp (right). Images of
the microspheres at the start, under pressing, and at the end of
compression indicate their deformation percentages. SilMA Microspheres:
The deformation analysis shows progressive compression from 100% at the
beginning to 30% at the end. The force‐displacement graph reveals a
peak force of 225.9 µN, fitting a Gaussian curve
[MATH: f(x)=225.9·exp
[−(x−18.76)22·(8.437)2] :MATH]
, reflecting the elastic and subsequent failure characteristics of the
microspheres. SilMA microspheres encapsulating CaO₂‐HAP: These
microspheres demonstrate enhanced resistance to deformation. The peak
force in the force‐displacement graph is lower at 188.4 µN, with a
broader Gaussian curve
[MATH: f(x)=188.4·exp
[−(x−20.34)22·(9.146)2] :MATH]
, suggesting improved structural integrity and resilience under
compression. These results highlight the reinforcement effect of
CaO₂‐HAp encapsulation, enhancing the deformation resistance of SilMA
microspheres. Mechanical testing revealed that the CaO₂‐HAp‐loaded
microspheres exhibited superior resilience under compression compared
to CaO₂‐only microspheres, ensuring structural stability in
physiological environments.
Oxygen release studies demonstrated that CaO₂‐HAp microspheres provided
sustained oxygen delivery over a 30‐day period, significantly
outperforming both CaO₂‐only microspheres and PBS controls
(Figure [80]2h). This long‐term oxygen release helps alleviate the
hypoxic conditions typically encountered in long‐term in vitro bone
organoid cultures and large bone defects, promoting cell survival and
enhancing osteogenesis. Furthermore, degradation and swelling studies
showed that CaO₂‐HAp microspheres exhibited controlled degradation and
moderate swelling behavior, maintaining structural integrity while
offering functional adaptability (Figure [81]2i,j). Finally, these
microspheres demonstrated excellent conformability, enabling them to be
assembled into customized shapes, such as bone‐like structures or
geometric forms, facilitating personalized repair strategies for
irregular bone defects (Figure [82]2k).
This innovative oxygen‐releasing hydrogel microsphere system addresses
key challenges in bone organoid engineering, including oxygen supply
and conformability. It provides a multifunctional and customizable
platform for bone defect repair, with the potential to accelerate bone
regeneration. By integrating mechanical stability and functional oxygen
release, this approach lays a solid foundation for advancing bone
organoid research and clinical applications in large‐scale bone defect
repair.
2.2. Effects of Oxygen‐Releasing SilMA Microspheres on Cell Viability,
Proliferation, Osteogenesis, and Immunomodulation
Building upon the innovative strategy of constructing conformal bone
organoids using oxygen‐releasing silk fibroin hydrogel microspheres,
this section explores the in vitro performance of hydrogel microspheres
encapsulating BMSCs, macrophages (RAW264.7), and oxygen‐releasing
materials (CaO₂ and CaO₂‐HAp) in terms of cell survival, proliferation,
osteogenesis, and immunomodulation. Four experimental groups were
designed: Sil@B (silk fibroin hydrogel microspheres encapsulating
BMSCs), Sil@B/R (microspheres encapsulating BMSCs and RAW264.7),
Sil@B/R/C (microspheres encapsulating BMSCs, RAW264.7, CaO₂, and
catalase), and Sil@B/R/C‐H (microspheres encapsulating BMSCs, RAW264.7,
CaO₂‐HAp, and catalase). The focus was to investigate the effects of
oxygen release and immunomodulation on cellular behaviors.
Cell survival and proliferation of BMSCs within oxygen‐releasing silk
fibroin hydrogel microspheres were assessed under hypoxic (1 day) and
normoxic (2 days) conditions using live/dead staining and CCK‐8 assays.
Live/dead staining results and quantitative analysis (Figure [83]
3a,[84]b) showed that Sil@B/R/C‐H microspheres significantly improved
cell survival compared to other groups, as evidenced by a higher
proportion of live cells (green fluorescence) and a marked reduction in
dead cells (red fluorescence). This indicates that the oxygen‐releasing
material CaO₂‐HAp effectively mitigates hypoxic conditions within the
microspheres, providing a more favorable microenvironment for cell
survival. The CCK‐8 assay further confirmed these findings. Under
hypoxic conditions, Sil@B/R/C and Sil@B/R/C‐H microspheres demonstrated
significantly higher cell viability compared to Sil@B and Sil@B/R
groups (Figure [85]3c), highlighting the critical role of oxygen
regulation. After two days of culture under normoxic conditions,
Sil@B/R/C‐H microspheres exhibited the most pronounced cell
proliferation activity (Figure [86]3c), suggesting a synergistic effect
between sustained oxygen release and bioactive components.
Figure 3.
Figure 3
[87]Open in a new tab
Effects of oxygen‐releasing SilMA microspheres on cell viability,
proliferation, osteogenesis, and immunomodulation. a) Live/dead
staining of cells in Sil@B, Sil@B/R, Sil@B/R/C, and Sil@B/R/C‐H
microspheres under hypoxic conditions (1 day) and normoxic conditions
(2 days). b) Quantitative analysis of cell viability based on live/dead
staining. c) CCK‐8 assay showing the proliferation of cells in
microspheres under hypoxic (1 day) and normoxic (2 days) conditions. d)
ALP staining of microspheres showing osteogenic differentiation. e)
Quantitative analysis of ALP staining. f) ARS staining showing calcium
deposition of cells in microspheres during osteogenesis. g)
Quantitative analysis of ARS staining. h) Gene expression analysis of
inflammatory markers (TNF‐α, CD86, and iNOS), anti‐inflammatory markers
(CD163 and IL‐10), angiogenic markers (VEGF, CD31), and osteogenic
markers (BMP‐2, COL1, and RUNX2) in cells encapsulated within
microspheres. Data are presented as means ± SD (n = 3). ^* p < 0.05,
^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001.
Osteogenic differentiation of BMSCs within the microspheres was
evaluated through Alkaline Phosphatase (ALP) and Alizarin Red S (ARS)
staining, which represent early and late osteogenic markers,
respectively. As shown in Figure [88]3d,e, ALP activity was
significantly enhanced in the Sil@B/R/C and Sil@B/R/C‐H groups, with
Sil@B/R/C‐H microspheres exhibiting the highest osteogenic potential.
ARS staining further revealed a substantial increase in mineralized
matrix formation in the Sil@B/R/C‐H group (Figure [89]3f). Quantitative
analysis (Figure [90]3g) indicated that ARS activity in Sil@B/R/C‐H
microspheres was significantly higher than in other groups,
underscoring the pivotal role of sustained oxygen release in promoting
osteogenic mineralization.
To investigate the immunomodulatory effects of oxygen‐releasing
microspheres, gene expression analysis was performed to assess key
markers related to inflammation, angiogenesis, and osteogenesis
(Figure [91]3h; Figure [92]S1, Supporting Information). The Sil@B/R/C‐H
group significantly downregulated pro‐inflammatory markers such as
TNF‐α, iNOS, CD86, IL‐1β, and IL‐6 while upregulating anti‐inflammatory
markers such as IL‐10, CD163, TGF‐β and Arg‐1,^[ [93]^23 ^] indicating
effective regulation of immune responses and macrophage polarization
toward the M2 phenotype. Additionally, the Sil@B/R/C‐H group
significantly upregulated markers associated with angiogenesis such as
VEGF and CD31, and osteogenesis such as BMP‐2, RUNX2, and COL1
demonstrating that these oxygen‐releasing microspheres create a
regenerative microenvironment conducive to bone repair. By coordinating
immunomodulation with angiogenesis and osteogenesis, Sil@B/R/C‐H
microspheres exhibit multifunctional potential to meet the complex
demands of bone defect repair, including oxygen supply, enhanced
osteogenic differentiation, reduced immune responses, and promotion of
angiogenesis.
These findings demonstrate that Sil@B/R/C‐H microspheres exhibit
superior performance in supporting cell survival, promoting osteogenic
differentiation, and regulating immune responses through sustained
oxygen release. The integration of CaO₂‐HAp not only effectively
alleviates hypoxic conditions but also establishes a regenerative
microenvironment favorable for bone organoid development. Furthermore,
by encapsulating RAW264.7 immune cells, this strategy offers a novel
approach to bone regeneration and immunomodulation. It provides a solid
theoretical and practical foundation for regenerative medicine,
particularly in applications requiring oxygen supply, osteogenesis,
angiogenesis, and immune regulation, such as the construction of bone
organoids and the repair of critical‐sized bone defects.
2.3. Transcriptomic Analysis of Oxygen‐Releasing Immunomodulatory
Microspheres
To investigate the molecular mechanisms underlying the enhanced
osteogenesis, angiogenesis, and immunomodulation induced by
oxygen‐releasing immunomodulatory microspheres, we performed
transcriptomic sequencing on four experimental groups: Sil@B, Sil@B/R,
Sil@B/R/C, and Sil@B/R/C‐H. Each group included three parallel
biological replicates to ensure the reliability of the results. This
comprehensive transcriptomic analysis revealed key differences in gene
expression and enriched pathways, providing mechanistic insights into
the synergistic effects of immune regulation and sustained oxygen
release in promoting bone regeneration and vascularization.
Differential gene expression (DGE) analysis highlighted significant
variations between treatment groups (Figure [94] 4a). Among the
comparisons, notable differences were observed between Sil@B/R and
Sil@B/R/C‐H, as well as between Sil@B/R/C and Sil@B/R/C‐H, with the
latter group exhibiting a substantially higher number of differentially
expressed genes (DEGs). Other group comparisons, such as Sil@B versus
Sil@B/R and Sil@B versus Sil@B/R/C, showed minimal differences,
suggesting that the enhanced osteogenesis and vascularization are
primarily attributed to the Sil@B/R/C‐H microspheres.
Figure 4.
Figure 4
[95]Open in a new tab
Transcriptomic analysis of oxygen‐releasing immunomodulatory
microspheres. a) DEGs among four groups (Sil@B, Sil@B/R, Sil@B/R/C, and
Sil@B/R/C‐H). Volcano plots showing upregulated and downregulated genes
in the Sil@B/R versus Sil@B/R/C‐H b) and Sil@B/R/C versus Sil@B/R/C‐H
c). Pathway enrichment analysis of upregulated DEGs in the Sil@B/R
versus Sil@B/R/C‐H d) and Sil@B/R/C versus Sil@B/R/C‐H e). f) Heatmap
of osteogenesis‐related genes across all groups. g) Heatmap of
angiogenesis‐related genes across all groups. h) Heatmap of M2
macrophage polarization‐related genes across all groups. Data are
presented as means ± SD (n = 3). ^* p < 0.05, ^** p < 0.01, ^***
p < 0.001, ^**** p < 0.0001.
Volcano plots (Figure [96] 4b,c) further demonstrated the significance
of DEGs in Sil@B/R versus Sil@B/R/C‐H and Sil@B/R/C versus Sil@B/R/C‐H
comparisons. The Sil@B/R/C‐H group showed a higher number of
upregulated genes compared to the other groups, indicating its superior
ability to activate genes associated with bone regeneration,
angiogenesis, and immune modulation.
Pathway enrichment analysis of upregulated DEGs identified several
critical signaling pathways involved in bone regeneration and
vascularization. In the Sil@B/R versus Sil@B/R/C‐H comparison
(Figure [97]4d), top enriched pathways included focal adhesion,
ECM‐receptor interaction, TGF‐β signaling, Wnt signaling, and PI3K‐Akt
signaling. These pathways are well‐documented for their roles in
osteogenesis, angiogenesis, and extracellular matrix (ECM) remodeling.
Similarly, the Sil@B/R/C versus Sil@B/R/C‐H comparison (Figure [98]4e)
revealed similar enrichment trends, with additional pathways such as
Rap1 signaling and axon guidance also being activated, further
emphasizing the regulatory role of Sil@B/R/C‐H microspheres in
promoting tissue regeneration.
The osteogenic potential of the microspheres was confirmed by the
expression profiles of key osteogenesis‐related genes (Figure [99]4f).
The Sil@B/R/C‐H group demonstrated significantly higher expression of
pivotal osteogenic markers such as Tgf‐β1, Notch1, and SP7 compared to
other groups.^[ [100]^24 ^] These genes play essential roles in
osteogenic differentiation, bone matrix formation, and mineralization.
The upregulation of these markers strongly supports the enhanced
osteogenic capacity of Sil@B/R/C‐H microspheres.
Effective vascularization is essential for successful bone
regeneration.^[ [101]^25 ^] Gene expression analysis revealed
significantly higher levels of angiogenesis‐related markers such as
Icam1 in the Sil@B/R/C‐H group (Figure [102]4g). These genes are
critical for endothelial cell proliferation, migration, and new blood
vessel formation.^[ [103]^26 ^] The upregulation of these markers in
the Sil@B/R/C‐H group indicates its superior angiogenic potential,
which likely contributes to improved nutrient and oxygen delivery to
the regenerating bone tissue.
Immune regulation is a critical factor in bone regeneration. The
expression of M2 macrophage polarization‐related genes was
significantly upregulated in the Sil@B/R/C‐H group (Figure [104]4h).
Notably, Arg1 and other pro‐healing M2 macrophage markers showed the
highest expression levels in this group, suggesting that the
Sil@B/R/C‐H microspheres effectively promoted an anti‐inflammatory and
pro‐regenerative immune environment.^[ [105]^27 ^] This finding is
consistent with the immunomodulatory effects observed in earlier
sections, where the Sil@B/R/C‐H group demonstrated enhanced macrophage
polarization and reduced inflammation at the bone defect site.
This transcriptomic analysis provides valuable insights into the
molecular mechanisms underlying the enhanced osteogenic, angiogenic,
and immunomodulatory effects of oxygen‐releasing immunomodulatory
microspheres. The Sil@B/R/C‐H group demonstrated significant
upregulation of genes involved in bone formation, vascularization, and
M2 macrophage polarization, suggesting a synergistic interaction
between sustained oxygen release and immune regulation. Key signaling
pathways such as TGF‐β, Wnt, and ECM‐receptor interaction were
identified as central mediators of these effects, further validating
the role of Sil@B/R/C‐H microspheres in creating a regenerative
microenvironment.
Overall, these findings reinforce the hypothesis that the integration
of oxygen release and immune modulation within microspheres can
effectively address the challenges of hypoxia and inflammation in bone
regeneration. By promoting osteogenesis, angiogenesis, and tissue
repair, the Sil@B/R/C‐H microspheres lay a solid foundation for the
construction of functional bone organoid units and offer a promising
platform for advanced bone tissue engineering and regenerative medicine
applications.
2.4. Evaluation of Oxygen‐Releasing Immunomodulatory Microspheres for In Vivo
Bone Regeneration
Based on the in vitro results and material design principles, this
section presents the in vivo evaluation of silk fibroin
oxygen‐releasing immunomodulatory microspheres in bone regeneration.
Using a mouse critical‐sized calvarial defect model, the ability of
microspheres encapsulating BMSCs, RAW264.7 macrophages, and
oxygen‐releasing materials to promote osteogenesis and immunomodulation
was assessed. Experimental groups (Sil@B, Sil@B/R, Sil@B/R/C, and
Sil@B/R/C‐H) were compared with an untreated control group (Con) at 4
and 8 weeks. Micro‐CT imaging, histological staining,
immunohistochemistry, and immunofluorescence analyses were used to
reveal the synergistic effects of sustained oxygen supply and
immunomodulation in bone repair.
Micro‐CT imaging demonstrated clear differences in bone regeneration
among the experimental groups (Figure [106] 5a,[107]b). Consistent with
the in vitro findings, the control group exhibited minimal new bone
formation at both 4 and 8 weeks. Similarly, the Sil@B group, lacking
immune cells and oxygen‐releasing components, showed limited bone
repair, indicating that insufficient bioactivity and oxygen supply
hindered bone regeneration. In contrast, the Sil@B/R/C‐H group
displayed the most significant bone regeneration, with substantial bone
deposition observed as early as 4 weeks and dense, mature bone
structures by 8 weeks. The other experimental groups (Sil@B/R and
Sil@B/R/C) exhibited moderate levels of bone repair, highlighting the
critical role of sustained oxygen release and macrophage‐mediated
immunomodulation in bone regeneration. These findings directly support
the hypothesis that.^[ [108]^28 ^] Quantitative analyses of bone
mineral density (BMD), bone volume‐to‐total volume ratio (BV/TV), and
trabecular number (Tb.N) further confirmed these observations
(Figure [109] 5c–e). The Sil@B/R/C‐H group showed significantly higher
BMD, BV/TV, and Tb.N values at both 4 and 8 weeks compared to the other
groups, demonstrating the crucial role of CaO₂‐HAp in overcoming
hypoxia and supporting immunomodulatory functions. These results
suggest that the inclusion of CaO₂‐HAp in the microspheres ensures
adequate oxygen supply, effectively addressing the typical hypoxic
environment in critical‐sized defects. Additionally, the
immunomodulatory effects of RAW264.7 macrophages may regulate
inflammation and promote angiogenesis, thereby creating a favorable
environment for osteogenesis and fundamentally improving bone
regeneration outcomes.
Figure 5.
Figure 5
[110]Open in a new tab
In vivo evaluation of oxygen‐releasing SilMA microspheres in a mouse
calvarial critical‐sized defect model. a) Schematic illustration of the
experimental design for bone regeneration evaluation, including the
skull defect model, material implantation, and analysis at 4 and 8
weeks. b) Micro‐CT images of bone regeneration in control (Con), Sil@B,
Sil@B/R, Sil@B/R/C, and Sil@B/R/C‐H groups at 4 and 8 weeks. c–e)
Quantitative analysis of BMD, BV/TV, and Tb. N in the defect areas. f)
H&E and Masson's trichrome staining of tissue sections from the defect
areas at 4 and 8 weeks, showing bone regeneration and collagen
deposition. Data are presented as means ± SD (n = 6). ^* p < 0.05, ^**
p < 0.01, ^*** p < 0.001, ^**** p < 0.0001.
Histological staining provided supplementary evidence for the bone
regeneration process. H&E staining (Figure [111]5f) showed disorganized
tissue and persistent defects in the control and Sil@B groups at both 4
and 8 weeks. The Sil@B/R group exhibited some bone formation, but the
tissue structure remained incomplete. The Sil@B/R/C group showed
improved tissue structure, though with limited bone maturity. However,
the Sil@B/R/C‐H group displayed organized and vascularized bone
structures at 4 weeks, with mature bone tissue and reduced fibrotic
tissue observed at 8 weeks. These findings underscore the importance of
oxygen supply and immunomodulation in accelerating matrix remodeling
and bone maturation. Masson's trichrome staining further emphasized
these trends, with the Sil@B/R/C‐H group exhibiting more extensive
collagen deposition and mineralization compared to the control and
Sil@B groups. These results strongly support the hypothesis that the
incorporation of macrophages and oxygen‐releasing materials into the
microspheres promotes ECM remodeling and effective bone repair.^[
[112]^29 ^]
To evaluate in vivo osteogenic differentiation, immunohistochemical
staining for key osteogenic markers‐osteocalcin (OCN), osteopontin
(OPN), and RUNX2‐was performed (Figure [113] 6a,c). Consistent with the
in vitro data, the expression of OCN and OPN was significantly higher
in the Sil@B/R/C and Sil@B/R/C‐H groups compared to the control and
Sil@B groups. RUNX2, as a marker of osteogenic activity, was elevated
in the Sil@B/R/C and Sil@B/R/C‐H groups at 4 weeks and further
increased at 8 weeks, indicating sustained promotion of osteogenesis.
Quantitative analysis of the stained areas (Figure [114]6d,f) showed
the highest marker expression in the Sil@B/R/C‐H group at both time
points, especially at 8 weeks. These findings align with the previous
conclusions that sustained oxygen release improves the hypoxic
environment, supporting BMSC activity and promoting osteogenic
differentiation.
Figure 6.
Figure 6
[115]Open in a new tab
Immunohistochemical analysis of osteogenic markers in defect areas
treated with oxygen‐releasing SilMA microspheres. a–c)
Immunohistochemical staining for OCN, OPN, and RUNX2 in the defect
areas at 4 and 8 weeks in control (Con), Sil@B, Sil@B/R, Sil@B/R/C, and
Sil@B/R/C‐H groups. d–f) Quantitative analysis of the stained areas for
OCN, OPN, and RUNX2. Data are presented as means ± SD (n = 6). ^*
p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001.
Immunofluorescence staining for CD31 and CD206 revealed the role of the
microspheres in promoting vascularization and immunomodulation
(Figure [116] 7a,[117]b). The Sil@B/R/C‐H group exhibited the strongest
angiogenic response, with significantly higher CD31‐positive staining
compared to the other groups (Figure [118]7c). This result aligns with
the emphasis in the introduction on the critical role of angiogenesis
in bone regeneration, as vascularization is essential for nutrient
delivery and tissue remodeling. Simultaneously, the Sil@B/R/C‐H group
showed elevated CD206 expression, indicating macrophage polarization
toward the anti‐inflammatory M2 phenotype. These results demonstrate
that the inclusion of RAW264.7 macrophages and sustained oxygen release
synergistically promoted the formation of an anti‐inflammatory
environment, consistent with the immunomodulatory effects discussed in
Section [119]2.2. Notably, the Sil@B/R/C‐H microspheres effectively
regulated inflammation while promoting angiogenesis, underscoring their
multifunctional potential in bone regeneration. To evaluate the
systemic biocompatibility and potential toxicity of the implanted
oxygen‐releasing immunomodulatory microspheres, H&E staining of major
organs, including the heart, liver, spleen, lung, and kidney, was
performed. No noticeable histopathological abnormalities or
inflammatory infiltration were observed across all groups, indicating
that the microspheres possess favorable systemic biocompatibility
(Figure [120]S2, Supporting Information).
Figure 7.
Figure 7
[121]Open in a new tab
Immunofluorescence analysis of angiogenic and immunomodulatory markers
in defect areas treated with oxygen‐releasing SilMA microspheres. a)
Immunofluorescence staining for CD31 in control (Con), Sil@B, Sil@B/R,
Sil@B/R/C, and Sil@B/R/C‐H groups at 4 and 8 weeks. b)
Immunofluorescence staining for CD206 in the defect areas. c, d)
Quantitative analysis of CD31 and CD206 staining. Data are presented as
means ± SD (n = 6). ^* p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^****
p < 0.0001.
The in vivo results validate the previous design and in vitro findings.
Combining macrophages and CaO₂‐HAp within silk fibroin hydrogel
microspheres effectively addressed the dual challenges of hypoxia and
inflammation, which are key limitations in bone regeneration. The
synergistic effects of immunomodulation and oxygen supply observed in
the Sil@B/R/C‐H group not only satisfied the conformability
requirements for repairing defects of different sizes and shapes but
also enhanced bone repair outcomes. These results reinforce the
hypothesis that immune regulation and sustained oxygen supply are
essential for achieving robust and functional bone regeneration.^[
[122]^30 ^] By creating a regenerative microenvironment that supports
osteogenesis, angiogenesis, and immunomodulation, oxygen‐releasing
immunomodulatory microspheres provide a promising solution for bone
organoid construction and the repair of critical‐sized bone defects.
2.5. Application of Oxygen‐Releasing Immunomodulatory Microspheres in
Subcutaneous Bone Organoid Formation
To further investigate the application of oxygen‐releasing
immunomodulatory microspheres in bone tissue engineering, this section
evaluates their ability to support in vivo bone organoid formation.
Microspheres encapsulating BMSCs, RAW264.7 macrophages, and
oxygen‐releasing materials were subcutaneously implanted into BALB/c
mice, resulting in the formation of bone organoids. The morphology,
histology, and molecular characteristics of the bone organoids were
analyzed to assess their osteogenesis, angiogenesis, and
immunomodulation capabilities, providing further evidence of their
regenerative potential in calvarial defect repair (Figure [123] 8a).
Figure 8.
Figure 8
[124]Open in a new tab
Application of oxygen‐releasing immunomodulatory microspheres in
subcutaneous bone organoid formation. a) Schematic illustration of the
experimental setup, including subcutaneous implantation of microspheres
in BALB/c mice, sample collection at 8 weeks, and subsequent staining
(HE/Masson) and immunohistochemical/immunofluorescence analysis. b)
Morphological comparison of bone organoids formed in control (Con),
Sil@B, Sil@B/R, Sil@B/R/C, and Sil@B/R/C‐H groups. H&E staining c) and
Masson's trichrome staining d) of bone organoids. Immunohistochemical
staining for OPN e) and OCN f) in bone organoids, with quantitative
analysis of the stained areas indicating osteogenic activity in the
experimental groups. Immunofluorescence staining for CD206, g) and
CD31, h) in bone organoids, with quantitative analysis of CD206‐ and
CD31‐positive areas. Data are presented as means ± SD (n = 6). ^*
p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001.
After 8 weeks of subcutaneous implantation, the bone organoids formed
in the Sil@B/R/C‐H group were significantly larger and better
structured than those in other experimental groups (Figure [125]8b).
The control group (Con) and Sil@B group exhibited poor organoid
development with no distinct tissue formation. In contrast, the Sil@B/R
and Sil@B/R/C groups showed moderate growth with early signs of tissue
organization. The superior size and structural integrity of the bone
organoids in the Sil@B/R/C‐H group underscore the importance of the
synergistic effects of sustained oxygen release and immunomodulation in
promoting tissue development.
Histological staining provided detailed insights into the tissue
structure and ECM remodeling within the bone organoids. H&E staining
(Figure [126]8c) revealed well‐organized tissue structures and high
cellular activity in the Sil@B/R/C‐H group, while the control and Sil@B
groups exhibited sparse and disordered tissue. The Sil@B/R and
Sil@B/R/C groups displayed intermediate levels of tissue formation,
further emphasizing the critical roles of immune cells and sustained
oxygen supply in achieving optimal tissue development. Masson's
trichrome staining (Figure [127]8d) demonstrated extensive collagen
deposition in the Sil@B/R/C‐H group, indicating significant progress in
ECM remodeling and tissue maturation. In contrast, the control and
Sil@B groups exhibited limited collagen deposition, reflecting their
restricted regenerative capacity. These findings are consistent with
the results from calvarial defect repair, further validating the
importance of oxygen‐releasing immunomodulatory microspheres in
promoting ECM remodeling and tissue construction.
To assess the osteogenic potential of the bone organoids,
immunohistochemical staining for osteogenic markers, including OPN and
OCN, was performed (Figure [128]8e,f). The expression of OPN and OCN
was significantly higher in the Sil@B/R/C‐H group compared to other
groups (p < 0.0001), indicating strong osteogenic activity. The Sil@B/R
and Sil@B/R/C groups also showed increased expression of these markers
compared to the control and Sil@B groups, albeit to a lesser extent.
Quantitative analysis confirmed the highest levels of OPN and OCN
expression in the Sil@B/R/C‐H group, supporting the hypothesis that
sustained oxygen release and immunomodulation create a microenvironment
conducive to osteogenic differentiation. These findings align with
results from calvarial defect repair and in vitro studies, further
demonstrating the ability of the microspheres to drive osteogenesis
across different models.
To evaluate the angiogenic and immunomodulatory effects of the
microspheres, immunofluorescence staining for CD31 and CD206 was
conducted (Figure [129]8g,h). The Sil@B/R/C‐H group exhibited the
strongest CD31‐positive staining, indicating significantly enhanced
vascularization within the bone organoids. Similarly, CD206 expression
was markedly upregulated in the Sil@B/R/C‐H group, reflecting effective
polarization of macrophages toward the anti‐inflammatory M2 phenotype.
The control and Sil@B groups demonstrated weak angiogenesis and
immunomodulation, highlighting their limited regenerative potential
under these experimental conditions. Quantitative analysis showed
significantly larger CD31‐ and CD206‐positive areas in the Sil@B/R/C‐H
group compared to other groups. These results further reinforce the
conclusions, demonstrating that immunomodulation and angiogenesis are
critical drivers of successful tissue regeneration and validating the
efficacy of oxygen‐releasing immunomodulatory microspheres in promoting
these processes.
The results from the subcutaneous bone organoid model further confirm
the multifunctionality of Sil@B/R/C‐H microspheres in supporting bone
tissue engineering. By effectively combining sustained oxygen supply
with immunomodulation, these microspheres create a regenerative
microenvironment conducive to osteogenesis, angiogenesis, and ECM
remodeling. The outstanding performance of the Sil@B/R/C‐H group in
osteogenesis, vascularization, and anti‐inflammatory effects highlights
the critical role of coordinated oxygen release and immunomodulation in
achieving robust and functional tissue formation.
These findings are consistent with the conclusions from earlier
sections, demonstrating the translational potential of oxygen‐releasing
immunomodulatory microspheres in advanced bone tissue engineering
applications. Beyond repairing critical‐sized defects, this study
indicates that these immunomodulatory oxygen‐releasing microspheres can
be used to construct pre‐vascularized bone organoids, laying a
foundation for future therapeutic strategies in complex bone defect
repair.
2.6. Long‐Term In Vitro Culture and Osteogenic Potential of Bone Organoid
Units
Building on the findings from in vitro cell experiments, in vivo
cranial defect repair, and subcutaneous bone organoid formation, this
section explores the potential of oxygen‐releasing immunomodulatory
microspheres in generating bone organoid units through long‐term in
vitro culture. A comparative study of the Sil@B group and the
Sil@B/R/C‐H group was conducted to evaluate the effects of
oxygen‐releasing immunomodulatory microspheres on cell viability and
osteogenic performance during extended culture. The results
demonstrated that Sil@B/R/C‐H microspheres could maintain high cell
viability and osteogenic capacity throughout a 28‐day culture period,
successfully forming functional bone organoid units. This establishes a
solid foundation for the development of transplantable bone
regeneration units for therapeutic applications.
Microscopic observations revealed significant differences between the
experimental groups during the long‐term culture period (Figure [130]
9a). The Sil@B group exhibited progressive contraction and sparse
cellular structure, indicating limited cell viability and tissue
formation capacity. In contrast, the Sil@B/R/C‐H group maintained
structural integrity throughout the culture period, showing pronounced
cell proliferation and tissue thickening. By day 28, Sil@B/R/C‐H
microspheres exhibited tissue‐like morphology with active cells,
indicating that sustained oxygen release and immunomodulation
significantly supported cell viability and tissue morphology during
long‐term culture.
Figure 9.
Figure 9
[131]Open in a new tab
Long‐term in vitro culture and osteogenic potential of bone organoid
units. a) Microscopic images of Sil@B and Sil@B/R/C‐H bone organoid
units during long‐term in vitro culture (days 7, 14, 21, and 28). b)
Live/dead staining of bone organoid units at different time points. c)
ALP staining of bone organoid units showing osteogenic differentiation.
d) ARS staining showing calcium deposition of bone organoid units
during osteogenesis. Quantitative analysis of live cell area e), ALP
activity f), and ARS staining g) at various time points. Data are
presented as means ± SD (n = 3). ^* p < 0.05, ^** p < 0.01, ^***
p < 0.001, ^**** p < 0.0001.
Cell viability within the microspheres was analyzed using live/dead
staining (Figure [132]9b). Significant differences in cell viability
between the two groups were observed as early as day 7. Over time, the
cell survival rate in the Sil@B group declined, with extensive cell
death by day 28. In contrast, the Sil@B/R/C‐H group maintained high
cell viability throughout the culture period, peaking in activity on
day 21. These findings suggest that oxygen‐releasing immunomodulatory
microspheres mitigate hypoxic conditions, providing sustained metabolic
support and extending cell survival. Quantitative analysis
(Figure [133]9e) confirmed that the Sil@B/R/C‐H group exhibited
significantly higher cell survival areas compared to the Sil@B group
during the long‐term culture period.
To evaluate the osteogenic differentiation potential of the
microspheres, ALP staining, and activity assays were performed
(Figure [134]9c,f). ALP staining in the Sil@B group was weak and did
not show significant enhancement over time. In contrast, the
Sil@B/R/C‐H group exhibited strong ALP staining starting on day 14,
with further intensification by day 28, indicating robust early
osteogenic differentiation activity. Quantitative analysis of ALP
activity revealed that the Sil@B/R/C‐H group peaked on day 21,
significantly outperforming the Sil@B group. These results validate the
synergistic effects of sustained oxygen release and immunomodulation in
promoting osteogenic differentiation under long‐term culture
conditions.
To assess mineralization capacity, ARS staining and mineralization
analysis were conducted (Figure [135]9d,g). The Sil@B group exhibited
minimal mineralization throughout the culture period, whereas the
Sil@B/R/C‐H group showed significant mineral deposition starting on day
14, with extensive calcium accumulation by day 28. Quantitative
analysis of ARS staining indicated that the Sil@B/R/C‐H group
significantly outperformed the Sil@B group in mineralization capacity,
with the highest mineralization observed on day 28. These results
demonstrate that oxygen‐releasing immunomodulatory microspheres not
only support early osteogenic differentiation but also facilitate the
formation of mineralized matrix, thus enabling the development of
functional bone organoid units.
This study validates the potential of oxygen‐releasing immunomodulatory
microspheres to generate functional bone organoid units through
long‐term in vitro culture. The results demonstrated that Sil@B/R/C‐H
microspheres exhibited peak cell activity on day 21, followed by
significantly enhanced osteogenic differentiation and mineralization by
day 28, leading to the formation of mature bone organoid units. These
findings further confirm the mechanisms proposed in Sections [136]2.3
and [137]2.4, highlighting the synergistic effects of sustained oxygen
release and immunomodulation in promoting bone tissue regeneration.
From an immunological perspective, RAW264.7 macrophages polarize toward
the anti‐inflammatory M2 phenotype, modulating inflammatory responses
and creating a supportive immune environment for bone tissue
regeneration.^[ [138]^31 ^] From an oxygenation perspective, sustained
oxygen release from CaO₂‐HAp alleviates hypoxia within the
microspheres, supporting BMSC proliferation and osteogenic
differentiation, ultimately enhancing the functionality of bone
organoid units.
This study not only demonstrates the feasibility of constructing bone
organoid units under long‐term in vitro culture conditions using
oxygen‐releasing immunomodulatory microspheres but also provides a new
approach to bone regeneration. By combining immunomodulation with
sustained oxygen supply, these microspheres establish a reliable
foundation for the creation of bioactive, conformal bone organoid
units, offering new possibilities for the treatment of critical‐sized
bone defects and complex bone injuries.
2.7. Functional Validation of Bone Organoid Units In Vivo
Building upon the results of long‐term in vitro culture and in vivo
bone repair experiments, this section further explores the performance
of bone organoid units both in vitro and after in vivo transplantation.
By transplanting bone organoid units, cultured for 28 days in vitro,
into a critical‐sized cranial defect model in mice, this study
comprehensively evaluated their potential for bone regeneration,
angiogenesis, and immune modulation. The study highlights the
mechanisms by which oxygen‐releasing immunomodulatory microspheres
promote the construction and functional repair capabilities of bone
organoid units.
CT analysis of bone organoid units after 28 days of in vitro culture
revealed significant mineralization and dense matrix formation in the
Sil@B/R/C‐H group (Figure [139] 10a). Compared to the Sil@B group, the
Sil@B/R/C‐H group demonstrated a substantially higher degree of mineral
deposition with uniform distribution. Quantitative analysis confirmed
that BMD and bone volume‐to‐total volume ratio (BV/TV) were
significantly higher in the Sil@B/R/C‐H group than in the Sil@B group
(Figure [140]10b). These results demonstrate that the oxygen‐releasing
immunomodulatory microspheres effectively enhance mineralization and
provide a robust platform for the construction of bone organoid units
in vitro.
Figure 10.
Figure 10
[141]Open in a new tab
Functional Validation of Bone Organoid Units In Vivo. a) CT analysis of
in vitro‐cultured bone organoid units after 28 days. b) Quantitative
analysis of BMD and BV/TV of the in vitro‐cultured bone organoid units.
c) CT images of cranial defects 8 weeks after transplantation of in
vitro‐cultured bone organoid units. d) Quantitative analysis of BMD and
BV/TV of the cranial defect area post‐transplantation.
Immunohistochemical staining for RUNX2 e), OCN f), and OPN g) in
cranial defect sections 8 weeks post‐transplantation. h)
Immunofluorescence staining of CD31 in cranial defect sections. i)
Immunofluorescence staining of CD206 in cranial defect sections. Data
are presented as means ± SD (n = 6). ^* p < 0.05, ^** p < 0.01, ^***
p < 0.001, ^**** p < 0.0001.
To validate the functionality of in vitro‐cultured bone organoid units
in vivo, organoid units from the Sil@B/R/C‐H and Sil@B groups were
transplanted into critical‐sized cranial defects in mice, with the
injection of SilMA microspheres serving as the control group (Con)
(Figure [142]10c). Eight weeks post‐transplantation, CT analysis
demonstrated minimal new bone formation in the Con group, with the
defect remaining largely unhealed. The Sil@B group exhibited limited
bone formation, while the Sil@B/R/C‐H group showed substantial defect
repair, with dense and well‐organized bone tissue fully bridging the
defect. Quantitative analysis further confirmed significantly higher
BMD and BV/TV values in the Sil@B/R/C‐H group compared to the other
groups (Figure [143]10d), highlighting the superior regenerative
potential of the bone organoid units cultivated with oxygen‐releasing
immunomodulatory microspheres.
The histological evaluation further revealed the differences in bone
formation among the groups (Figure [144]10e–g). Immunohistochemical
staining showed significantly higher expression of RUNX2, OCN, and OPN
in the Sil@B/R/C‐H group compared to the Sil@B and Con groups.
Quantitative analysis indicated that the positive staining areas for
RUNX2, OCN, and OPN were significantly larger in the Sil@B/R/C‐H group
(p < 0.0001), confirming the enhanced osteogenic differentiation and
mineralization potential of the Sil@B/R/C‐H microspheres both in vitro
and in vivo.
Angiogenesis and immune modulation are critical factors for successful
bone defect repair.^[ [145]^32 ^] Immunofluorescence staining revealed
significantly higher expression of CD31 and CD206 in the Sil@B/R/C‐H
group compared to the Sil@B and Con groups (Figure [146]10h,i).
Positive CD31 staining demonstrated that the Sil@B/R/C‐H group
facilitated the formation of a dense vascular network within the defect
area, providing essential nutrients and oxygen for tissue repair.
Similarly, the increased CD206 expression indicated effective
polarization of macrophages toward the anti‐inflammatory M2 phenotype,
creating a favorable immune microenvironment for bone regeneration.
Quantitative analysis confirmed that the CD31 and CD206‐positive
staining areas were significantly larger in the Sil@B/R/C‐H group,
underscoring the multifunctional advantages of oxygen‐releasing
immunomodulatory microspheres in promoting vascularization and immune
regulation.
This section validates the functional superiority of oxygen‐releasing
immunomodulatory microspheres in constructing bone organoid units and
repairing cranial bone defects. The Sil@B/R/C‐H group demonstrated
excellent mineralization during in vitro long‐term culture and showed
superior performance in bone regeneration, angiogenesis, and immune
modulation after in vivo transplantation. These findings highlight the
critical roles of sustained oxygen delivery and immune regulation in
enhancing the functionality of bone organoid units. Taken together with
the results from previous sections, this study systematically
demonstrates the effectiveness of oxygen‐releasing immunomodulatory
microspheres in facilitating both in vitro and in vivo bone
regeneration. By addressing the challenges of hypoxia and immune
imbalance, these microspheres provide a powerful and versatile platform
for constructing bone organoid units and repairing complex bone
defects. This innovative strategy offers promising insights and
practical applications for personalized bone regeneration therapies and
advanced bone tissue engineering, paving the way for future clinical
translation.
3. Conclusion
This study presents the innovative design and application of silk
fibroin hydrogel microspheres with sustained oxygen release and
immunomodulatory functions, successfully constructing conformal bone
organoid units and systematically evaluating their role and mechanisms
in bone regeneration. To achieve a biomimetic and functional organoid
microenvironment, BMSCs were incorporated as the primary osteogenic
component due to their well‐established differentiation potential.
RAW264.7 macrophages were co‐encapsulated to provide immunoregulatory
functionality, particularly through their polarization toward the M2
phenotype, which promotes anti‐inflammatory signaling and supports
tissue regeneration. CaO₂‐HAp nanoparticles served as the core
oxygen‐releasing system, not only alleviating hypoxia but also
contributing calcium and phosphate ions for mineralization. Catalase
was introduced to decompose the intermediate hydrogen peroxide, thereby
enhancing biocompatibility and preventing oxidative stress. This
rational integration of cellular and material components enables the
microspheres to simultaneously address hypoxia, inflammation, and
osteogenesis—three major challenges in bone organoid engineering. The
findings demonstrate that these microspheres effectively address the
challenges of insufficient oxygen supply and impaired immune
microenvironment regulation in bone regeneration. By continuously
releasing oxygen to alleviate hypoxia and leveraging the
immunomodulatory effects of RAW264.7 macrophages, the microspheres
create a regenerative microenvironment conducive to osteogenic
differentiation and angiogenesis. In vitro experiments validated the
significant advantages of the oxygen‐releasing immunomodulatory
microspheres in promoting cell viability, early osteogenic
differentiation, and late‐stage mineralization. Results from an in vivo
mouse cranial defect model further confirmed that these microspheres
significantly enhance bone formation efficiency, improve
vascularization, and optimize immune regulation. Notably, under
long‐term culture conditions, these microspheres demonstrated the
ability to form functional bone organoid units at the
single‐microsphere level, offering new possibilities for the repair of
complex bone defects.
Despite these promising outcomes, several limitations and challenges
must be acknowledged. First, the scalability of the microsphere‐based
bone organoid system for large‐scale production and clinical
translation remains to be further validated. Second, while the current
in vivo experiments in mice provide preliminary insights, the long‐term
stability, biodegradation kinetics, and immune responses in larger
animal models are still unknown and warrant further investigation.
Additionally, the integration of bone organoid units into irregular and
load‐bearing defect sites under mechanical stress presents another
technical barrier. Addressing these challenges will require further
refinement of fabrication methods, optimization of material
compositions, and the use of more clinically relevant models. Moreover,
broader accessibility and standardization of the fabrication platform
will be essential to promote knowledge sharing and facilitate
reproducibility across research groups.
The outcomes of this study not only provide an innovative solution for
the treatment of critical‐sized bone defects but also lay a theoretical
and practical foundation for the construction of vascularized bone
organoids. In the future, with further optimization of bone organoid
construction techniques, oxygen‐releasing immunomodulatory bone
organoid units are expected to find widespread applications in bone
tissue engineering and regenerative medicine. They hold the potential
to serve as a novel platform for personalized and transplantable bone
repair, paving the way for transformative advances in clinical bone
regeneration therapies.
4. Experimental Section
Materials
SilMA, LAP (Engineering for Life, Jiangsu). Liquid paraffin, Span 80
(Aladdin Shanghai). Microfluidic Coaxial needles (Nuokang Environmental
Protection Technology, Qingdao). Ultra‐low adsorption culture plate
(NEST, Wuxi). Fetal bovine serum (Sigma, Shanghai). Automatic cell
counter (Countstar, Shanghai).
Preparation of CaO[2], CaO[2]‐HAp
To prepare CaO₂, dissolve 3 g of calcium chloride (CaCl₂) in water to
create a 10% calcium chloride solution. Add 15 mL of ammonia solution
(25–28%, 1 m) and 120 mL of PEG 200 to the solution. Subsequently, add
15 mL of hydrogen peroxide (H₂O₂) solution at a constant rate of
0.2 mL min^−1 to the mixture while stirring continuously for 2 h.
During the precipitation process, adjust the pH to 11.5 by adding 0.1 m
sodium hydroxide (NaOH) solution. The resulting yellow precipitate is
collected by centrifugation and washed three times with 0.4 m NaOH
solution, followed by three washes with double‐distilled water (DDW)
until the pH reaches 10. Finally, the obtained precipitate is dried in
an oven at 80 °C, yielding CaO₂.^[ [147]^33 ^]
To prepare CaO₂‐HAp, the synthesized CaO₂ is added to an appropriate
amount of 100 mm PBS and stirred for 8 h. During this process, CaO₂
reacts with water to form Ca(OH)₂, which subsequently reacts with H₃PO₄
in PBS to form hydroxyapatite (Ca₁₀(PO₄)₆(OH)₂, HAp). Continuous
stirring promotes the gradual deposition of HAp onto the surface of
CaO₂ particles, resulting in a coated structure that enables sustained
oxygen release.^[ [148]^34 ^]
[MATH: 10CaOH2<
mo
linebreak="badbreak">+6H3<
mi>PO4→Ca10PO46OH2<
mo
linebreak="goodbreak">+18H2O :MATH]
(1)
Preparation of SilMA Microspheres and Oxygen‐Releasing Hydrogel Microspheres
Before preparing the hydrogel microspheres, the aqueous and oil phases
were first prepared. In this study, the aqueous phase consisted of 15%
SilMA and 0.05% ALP. For groups containing oxygen‐releasing components,
CaO₂ or CaO₂‐HAp nanoparticles were first dispersed in PBS using a
probe sonicator (5 s on/5 s off, 2 min total) to ensure uniform
dispersion. The homogenized dispersion was then thoroughly mixed with
the SilMA solution to form the functionalized aqueous phase. While the
oil phase is composed of 90% paraffin oil and 10% Span 80.^[ [149]^35
^]
Microfluidic technology was utilized to fabricate the hydrogel
microspheres, using a coaxial microfluidic needle setup, which includes
two concentric needles. The inner needle delivers the aqueous phase,
and the outer needle delivers the oil phase. The generation of
microspheres relies on the shear force produced by the oil phase, which
encapsulates the aqueous droplets in the oil, forming microspheres with
uniform size. By adjusting the flow rate ratio of the oil and aqueous
phases, the size of the microspheres can be precisely controlled. Under
UV light at 365 nm, ALP in the aqueous phase undergoes photochemical
crosslinking with SilMA, resulting in loosely porous hydrogel
microspheres.
Characterization of Oxygen‐Releasing Material, SilMA Microspheres and
Oxygen‐Releasing Hydrogel Microspheres
The Scanning Electron Microscopy (SEM) characterization of CaO₂ and
CaO₂‐HAp was conducted by dispersing nanoparticles in anhydrous
ethanol, dropping onto silicon wafers, and sputter‐coating with a
conductive layer. The SilMA microspheres and oxygen‐releasing hydrogel
microspheres were freeze‐dried, adhered to conductive tape, and
metal‐coated for SEM analysis. SEM (JSM‐7500F, Japan) was used to
visualize the surface morphology. To confirm the incorporation and
spatial distribution of the oxygen‐releasing components,
energy‐dispersive X‐ray spectroscopy (EDX) was performed using the SEM
system. EDX mapping of the SilMA@CaO₂ and SilMA@CaO₂‐HAp microspheres
revealed uniform elemental distributions of calcium (Ca) and phosphorus
(P), indicating successful and homogeneous embedding of CaO₂ and
CaO₂‐HAp nanoparticles within the hydrogel network. This verified the
structural integrity and functional uniformity of the oxygen‐releasing
microspheres.
Oxygen Release Measurement
Samples of CaO₂, CaO₂‐HAp, and catalase were placed in an anaerobic
incubator and left undisturbed for one day to eliminate interference
from residual oxygen. Similarly, pure water was deoxygenated by
bubbling nitrogen gas through it, and then stored in the anaerobic
incubator for one day. Subsequently, 150 mg of CaO₂ or 150 mg of
CaO₂‐HAp were added to 30 mL of deoxygenated pure water containing
catalase at a concentration of 1500 U mL^−1. Deoxygenated pure water
was used as the blank control. The dissolved oxygen levels in the three
groups were measured under anaerobic conditions at 1, 3, 5, 7, 14, and
28 days using a dissolved oxygen meter (AR8606, Smart Sensor).
[MATH:
CaO2+2H2<
mi>O→CaOH2<
mo
linebreak="goodbreak">+H2
O2 :MATH]
(2)
[MATH:
2H2O
2→O2+2H2
O :MATH]
(3)
Characterization of SilMA Microspheres Properties—Swelling Test
The prepared hydrogel microspheres were immersed in PBS and incubated
at 37 °C. The dry weight of the hydrogel microspheres was recorded as
W[0]. At specific time intervals, the microspheres were removed from
the incubator, their surfaces were gently dried, and their wet weight
(W[t] ) was recorded. The swelling ratio was calculated using the
following formula:
[MATH: Swellingratio%=Wt−W0W0×100% :MATH]
(4)
Degradation Test
The prepared hydrogel microspheres were freeze‐dried, and their initial
dry weight was recorded as W[0]. The microspheres were then immersed
in PBS, DMEM medium, or a solution containing collagenase II, with the
soaking solution replaced every three days. At specific time intervals,
the microspheres were removed, and dried, and their dry weight (W[t])
was recorded. The degradation rate was calculated using the following
formula:
[MATH: Degradationratio%=W0−WtW0×100% :MATH]
(5)
Microsphere Mechanical Testing
To evaluate the mechanical properties of the hydrogel microspheres,
single‐microsphere compression tests were performed in PBS using a
micro‐force testing platform. A custom stainless‐steel flat‐end probe
(diameter = 0.56 mm) equipped with a 3 × 3 mm platen was used to apply
vertical compressive force onto individual microspheres. During the
test, each microsphere was submerged in PBS and compressed from its
full height (100%) to 75% of its original height. A high‐sensitivity
force sensor continuously recorded the real‐time force and displacement
at the probe tip. The resulting force–time, displacement–time, and
force–displacement curves were analyzed. Gaussian fitting was performed
using GraphPad Prism 9 software to characterize the mechanical
deformation behavior of the microspheres.
Preparation of SilMA@B, SilMA@B/R, SilMA@B/R/C and SilMA@B/R/C‐H
SilMA@B: For this group, 1 mL of 15% SilMA hydrogel was prepared, and
1 × 10^7 BMSCs were resuspended in the hydrogel. The cell suspension
was carefully mixed with the hydrogel to ensure uniform distribution of
cells. The mixture was then processed using the microfluidic technique
to generate the hydrogel microspheres. SilMA@B/R: In this group, 1 mL
of 15% SilMA hydrogel was prepared, and a mixture of 8 × 10^6 BMSCs and
2 × 10^6 RAW 264.7 cells was resuspended in the hydrogel. The cells
were thoroughly mixed with the hydrogel to ensure a homogeneous
suspension. The final mixture was processed through the microfluidic
system to generate uniform hydrogel microspheres. SilMA@B/R/C: For this
group, 1 mL of 15% SilMA hydrogel was prepared, and a mixture of
8 × 10^6 BMSCs, 2 × 10^6 RAW 264.7 cells, and 1 mg of CaO[2] was added.
Additionally, 1000 U of catalase enzyme is incorporated into the
mixture. The components were well‐mixed to ensure uniform distribution
before being processed using the microfluidic system to generate the
hydrogel microspheres. SilMA@B/R/C‐H: In this group, 1 mL of 15% SilMA
hydrogel is prepared, and a mixture of 8 × 10^6 BMSCs, 2 × 10^6 RAW
264.7 cells, 1 mg of CaO[2]‐HAp, and 1000 U of catalase enzyme was
added. The components are mixed thoroughly to achieve a homogeneous
suspension. This mixture was then processed using the microfluidic
system to produce the desired hydrogel microspheres.
The composition of the engineered microspheres, including the BMSCs to
RAW264.7 cell ratio (4:1), the concentration of CaO₂ or CaO₂‐HAp
(1 mg mL^−1), and the dosage of catalase (1000 U mL^−1), was determined
based on the previous study,^[ [150]^36 ^] which demonstrated that this
combination effectively promoted osteogenesis, immune modulation, and
biocompatibility. In addition, the rational selection of
oxygen‐releasing components and catalase was supported by the work of
Daisuke Tomioka et al.,^[ [151]^34 ^] in which a similar oxygen
delivery strategy significantly improved functional performance and
cellular safety. Therefore, the current formulation adopted in this
study builds upon proven effective designs and ensures a balance
between oxygen supply, immunoregulation, and osteogenic potential for
optimal bone organoid construction.
Cytotoxicity and Proliferation Assay of Oxygen‐Releasing Microspheres
Hydrogel microspheres from the four experimental groups were stained
using a live/dead cell double‐staining kit after culturing under
hypoxic conditions for 1 day followed by normoxic conditions for 2
days. The microspheres were incubated in the dark for 30 min and then
imaged using a confocal microscope (FV3000, Olympus, Japan). Cell
viability for the four groups was assessed using the CCK‐8 assay. After
incubating the samples for 2 h at a constant temperature, absorbance
was measured at a wavelength of 450 nm.
ALP Staining and ARS Staining with Quantitative Analysis of Mineralization in
Oxygen‐Releasing Microspheres
To evaluate the osteogenic differentiation capacity of cells within the
microspheres, an ALP staining assay was performed. Microspheres were
fixed with 4% paraformaldehyde at room temperature for 15 min, washed
with PBS, and incubated with BCIP/NBT ALP staining (Beyotime) solution
at 37 °C for 30 min. After color development, images were captured
using a stereomicroscope. ALP staining results were quantitatively
analyzed using ImageJ software to calculate the percentage of stained
areas, providing an assessment of ALP activity for the different groups
of microspheres.
To assess the formation of mineralized matrices within the
microspheres, an ARS staining assay was conducted. Microspheres were
fixed with 4% paraformaldehyde for 20 min, followed by staining with 2%
alizarin red S solution (pH 4.2) at room temperature for 20 min. After
staining, the microspheres were washed with distilled water, observed,
and imaged using a stereomicroscope. The ARS staining results were
quantitatively analyzed using ImageJ software to calculate the
percentage of stained areas, serving as an indicator of the
mineralization capacity of the microspheres.
To quantitatively assess osteogenic activity and mineralization levels
among different experimental groups, ALP and ARS staining results were
subjected to image‐based analysis. Each experiment was performed with
three biological replicates (n = 3). After staining, images of each
group were acquired under consistent parameters using a microplate
reader. The captured images were analyzed using ImageJ software to
extract mean gray values or staining intensity as quantitative
indicators. During analysis, unified thresholding and background
correction were applied across all images. Statistical analysis was
performed using GraphPad Prism software, and group differences were
evaluated by one‐way ANOVA. A p‐value < 0.05 was considered
statistically significant.
qRT‐PCR Analysis of Gene Expression in Oxygen‐Releasing Microspheres
Quantitative real‐time PCR (qRT‐PCR) was performed to analyze the gene
expression levels of inflammation‐, angiogenesis‐, and
osteogenesis‐related markers in cells encapsulated within the
microspheres. Total RNA was extracted using TRIzol reagent and
reverse‐transcribed into cDNA using a reverse transcription kit. Gene
amplification was carried out on a real‐time PCR instrument using SYBR
Green dye. The relative expression levels of key markers, including
TNF‐α, iNOS, CD86, IL‐10, CD206, VEGF, CD31, BMP‐2, RUNX2, and COL‐1,
were normalized to the internal control CEL‐MIR‐39‐3P and calculated
using the 2^−ΔΔCt method. All experiments were conducted in triplicate.
The primer sequences required in the experiment are shown in Table
[152]S1 (Supporting Information).
Implantation of Oxygen‐Releasing Microspheres in a Mouse Calvarial 3 mm
Defect Model
The animal procedures adhered to the Guidelines for Care and Use of
Laboratory Animals of Shanghai University and received approval from
the University's Animal Ethics Committee (YS 2023–160). A
critical‐sized calvarial defect model was established using
4–5‐week‐old C57BL/6 male mice (n = 6 per group). Under general
anesthesia and sterile conditions, a circular defect with a diameter of
3 mm was created on the calvarial bone at the top of the skull.
Microspheres from the following groups were implanted into the defect
site: control group (Con, no treatment), Sil@B group, Sil@B/R group,
Sil@B/R/C group, and Sil@B/R/C‐H group. After implantation, the defect
area was sutured, and the mice were monitored postoperatively to ensure
their health. Mice were sacrificed at 4‐ and 8‐weeks postsurgery for
further analysis.
Micro‐CT Analysis of Oxygen‐Releasing Microspheres
At 4‐ and 8‐weeks postsurgery, bone regeneration in the defect area was
evaluated using micro‐computed tomography (micro‐CT). Samples were
scanned at a resolution of 10 µm, and 3D reconstructions were performed
to observe new bone formation at the defect site. Quantitative analysis
of BMD, BV/TV, and Tb. N was conducted using micro‐CT analysis
software.
Histological Analysis of Oxygen‐Releasing Microspheres
The excised calvarial samples were subjected to histological analysis.
Samples were fixed in 4% paraformaldehyde, decalcified, dehydrated,
embedded in paraffin, and sectioned at a thickness of 5 µm. Sections
were stained with hematoxylin and eosin (H&E) to observe tissue
structure and with Masson's trichrome stain to assess collagen
deposition. The stained sections were examined under a light microscope
to evaluate bone regeneration and ECM remodeling.
Immunohistochemical Analysis of Oxygen‐Releasing Microspheres
Immunohistochemical staining was performed to assess the expression of
osteogenesis‐related markers in the post‐surgery samples, including
OCN, OPN, and RUNX2. Paraffin sections were deparaffinized, rehydrated,
and subjected to antigen retrieval. The sections were incubated with
primary antibodies against OCN, OPN, and RUNX2, followed by incubation
with horseradish peroxidase (HRP)‐conjugated secondary antibodies.
Staining was visualized using a DAB substrate, and hematoxylin was used
for counterstaining. The stained sections were imaged using a light
microscope, and the percentage of positive staining areas was
quantitatively analyzed using ImageJ software.
Immunofluorescence Analysis of Oxygen‐Releasing Microspheres
Immunofluorescence staining was conducted to evaluate the role of
microspheres in promoting angiogenesis and immunomodulation. Key
markers included CD31 and CD206. Frozen tissue sections were prepared
and blocked, followed by incubation with primary antibodies against
CD31 and CD206. After incubation with fluorescence‐conjugated secondary
antibodies, the sections were counterstained with DAPI to label nuclei.
Fluorescent images were captured using a fluorescence microscope, and
ImageJ software was used to quantify the percentage of CD31‐ and
CD206‐positive staining areas.
In Vivo Biosafety Evaluation
To assess systemic biocompatibility, major organs—including the heart,
liver, spleen, lung, and kidney—were collected from mice in each
treatment group (Con, Sil@B, Sil@B/R, Sil@B/R/C, and Sil@B/R/C‐H) after
8 weeks of in vivo implantation. The harvested tissues were rinsed with
PBS, fixed in 4% paraformaldehyde for 24 h, dehydrated, embedded in
paraffin, and sectioned into 5 µm‐thick slices. The sections were
stained with H&E according to standard protocols. Histological
observations were performed using an optical microscope to assess
tissue architecture, inflammatory infiltration, necrosis, or other
pathological changes.
Subcutaneous Implantation of Oxygen‐Releasing Microspheres for Bone Organoid
Formation
Male BALB/c nude mice (4–5 weeks old) were used to evaluate the
formation of bone organoids through subcutaneous implantation of
oxygen‐releasing microspheres (n = 6). Under sterile conditions,
microspheres from different experimental groups (Con, Sil@B, Sil@B/R,
Sil@B/R/C, and Sil@B/R/C‐H) were implanted subcutaneously into the
dorsal region of each mouse. The mice were maintained under normal
conditions post‐surgery and sacrificed at 8 weeks to analyze the
morphological, histological, and immunological characteristics of the
bone organoids.
Histological, Immunohistochemical, and Immunofluorescence Analysis of Bone
Organoids
The retrieved bone organoid samples were fixed in 4% paraformaldehyde,
dehydrated, and embedded in paraffin. H&E staining was performed to
observe tissue structure, and Masson's trichrome staining was used to
assess collagen deposition. Microscopic images were captured to analyze
tissue structure and ECM remodeling in the organoids.
Immunohistochemical staining for OPN and OCN and immunofluorescence
staining for CD31and CD206 were performed to evaluate new bone
formation, vascularization, and inflammatory responses. The percentage
of stained areas was quantitatively analyzed using ImageJ software.
Long‐Term In Vitro Culture and Osteogenic Differentiation Validation of Bone
Organoid Units
To assess the role of oxygen‐releasing microspheres in constructing
bone organoid units, Sil@B and Sil@B/R/C‐H microspheres were cultured
long‐term under standard cell culture conditions (37 °C, 5% CO₂) using
ultra‐low adhesion culture plates to prevent substrate attachment. For
the first 3 days, microspheres were maintained in α‐MEM medium
supplemented with 10% fetal bovine serum and 1%
penicillin‐streptomycin. After three days, the culture was switched to
an osteogenic induction medium consisting of DMEM, 10% fetal bovine
serum, 1% penicillin‐streptomycin, 10 mmol L^−1 β‐glycerophosphate,
50 µg mL^−1 ascorbic acid, and 10 nmol L^−1 dexamethasone. Medium
replacement was performed every 2 days during the first 7 days, daily
from day 7 to 14, and twice daily from day 14 to 28.
The morphological changes and tissue formation of the microspheres were
periodically observed during the culture period, with their morphology
and thickening recorded under a microscope. To evaluate cell viability
and survival rates, live/dead cell fluorescence staining was performed
on microspheres at each time point. Microspheres were stained with
Calcein‐AM and PI solutions to label live and dead cells, respectively,
and observed under a fluorescence microscope to examine cell
distribution. The live cell area was quantitatively analyzed using
ImageJ software, and the cell activity was compared across different
time points.
To assess the osteogenic differentiation potential of the microspheres,
ALP staining was conducted on days 7, 14, 21, and 28. Microspheres were
treated with ALP staining solution, and the stained regions were
observed under a light microscope. The ALP‐stained area was
quantitatively analyzed using ImageJ software to compare osteogenic
differentiation between the two groups at different time points. To
evaluate the formation of a mineralized matrix within the microspheres,
ARS staining was performed at the end of the culture period.
Microspheres were stained with ARS solution, excess dye was removed,
and calcium deposition was observed under a microscope. The ARS‐stained
area was quantitatively analyzed using ImageJ software to evaluate the
mineralization capacity of microspheres at different time points.
In Vivo Cranial Bone Repair and Functional Evaluation of Bone Organoid Units
The repair ability of bone organoid units was evaluated using a cranial
critical‐sized defect model in male C57BL/6 mice (4‐5 weeks old), with
circular defects of 3 mm in diameter created at the parietal bone
region. Bone organoid units cultured in vitro for 28 days, including
Sil@B/R/C‐H and Sil@B groups, were injected and implanted into the
defect area, with a Con as a comparison. After implantation, the skin
over the defect area was directly sutured, and the mice were maintained
for 8 weeks. After harvesting, CT scans were performed to obtain images
of bone repair at the defect site, and BMD and BV/TV were analyzed to
assess the bone repair outcomes. Tissue sections were stained with
immunohistochemistry to detect osteogenic markers, including RUNX2,
OCN, and OPN, and the relative area of positive staining was quantified
using ImageJ software to evaluate osteogenic differentiation and
mineralization. Additionally, immunofluorescence staining was performed
to detect angiogenic marker CD31 and immunoregulatory marker CD206 in
the repair area, with quantitative analysis of the positive staining
area.
Statistical Analysis
All statistical analyses were performed using GraphPad Prism 9. Data
are presented as mean ± standard deviation (SD). For comparisons
between the two groups, unpaired two‐tailed Student's t‐tests were
used. For comparisons involving three or more groups, one‐way analysis
of variance (ANOVA) with Tukey's post‐hoc test was applied. Statistical
significance was defined as ^* p < 0.05, ^** p < 0.01, ^*** p < 0.001,
and ^**** p < 0.0001.
Conflict of Interest
The authors declare no conflict of interest.
Author Contributions
A.D., H.Z., Y.H., and J.L. contributed equally to this work. A.D.
wrote, reviewed, and edited the final draft and wrote the original
draft, methodology, and data curation. H.Z., J.W., and X.C. performed
the methodology. Y.H. wrote the original draft. J.L. and Y.L. performed
data curation. Z.G. and K.X. performed a formal analysis. J.W. and Y.J.
performed supervision. L.B. performed supervision and
conceptualization. J.S. performed supervision and funding acquisition.
Supporting information
Supporting Information
[153]ADVS-12-e01437-s001.docx^ (899.8KB, docx)
Acknowledgements