type H vessels

  • 文章类型: Journal Article
    Endomucin(MUC14),由EMCN基因编码,是一种O-糖基化的跨膜粘蛋白,主要存在于静脉内皮细胞(EC)中,并在骨组织的H型血管中高度表达。其主要生物学功能包括通过血管内皮生长因子(VEGF)信号通路促进内皮生成和迁移,抑制炎症细胞与ECs的粘附。此外,它诱导血管生成并促进骨形成。由于Endomucin在上述方面的优异功能,为血管炎症相关疾病和骨疾病的治疗提供了新的研究靶点。根据目前对其功能的理解,子宫内膜蛋白的研究主要集中在上述两种疾病上。众所周知,癌症的进展与血管生成密切相关。最近发现子宫内膜蛋白在多种肿瘤中差异表达,并与生存率相关。子宫内膜蛋白在癌症中的生物学作用是不清楚的。本文介绍了Endomucin在血管炎症相关疾病和骨疾病中的研究进展,探讨了其在治疗中的应用价值和前景,并收集了子宫内膜蛋白在肿瘤中的最新研究情况,以期为拓展Endomucin的研究领域提供有意义的证据。
    Endomucin (MUC14), encoded by EMCN gene, is an O-glycosylated transmembrane mucin that is mainly found in venous endothelial cells (ECs) and highly expressed in type H vessels of bone tissue. Its main biological functions include promoting endothelial generation and migration through the vascular endothelial growth factor (VEGF) signaling pathway and inhibiting the adhesion of inflammatory cells to ECs. In addition, it induces angiogenesis and promotes bone formation. Due to the excellent functions of Endomucin in the above aspects, it provides a new research target for the treatment of vascular inflammatory-related diseases and bone diseases. Based on the current understanding of its function, the research of Endomucin mainly focuses on the above two diseases. As it is known, the progression of cancer is closely related to angiogenesis. Endomucin recently is found to be differentially expressed in a variety of tumors and correlated with survival rate. The biological role of Endomucin in cancer is opaque. This article introduces the research progress of Endomucin in vascular inflammatory-related diseases and bone diseases, discusses its application value and prospect in the treatment, and collects the latest research situation of Endomucin in tumors, to provide meaningful evidence for expanding the research field of Endomucin.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨质疏松(osteoporosis,OP)是一种以骨量减少为特征的慢性系统性骨代谢疾病,微建筑恶化,和脆性骨折。随着长寿和人口老龄化导致的人口变化,OP是一个日益严重的健康问题。miRNA在OP发病机制中的作用近年来也引起了学者的广泛关注。H型血管是骨的独特微血管,由于它们在成骨-血管生成偶联中起着重要作用,因此已成为OP发病机制的新焦点。先前的研究发现一些miRNA通过调节因子调节H型血管的形成,包括血小板衍生生长因子-BB(PDGF-BB),缺氧诱导因子1α(HIF-1α),血管内皮生长因子(VEGF),等等。这些发现有助于我们更深入地了解miRNAs之间的关系,H型血管,寻找治疗OP的新视角。在目前的小型审查中,我们将介绍H型血管在OP发病机制中的作用,以及通过影响调节因子来调节miRNAs对H型血管形成的影响,为今后OP治疗的研究提供一些有价值的见解。
    Osteoporosis (OP) is a chronic systemic bone metabolism disease characterized by decreased bone mass, microarchitectural deterioration, and fragility fractures. With the demographic change caused by long lifespans and population aging, OP is a growing health problem. The role of miRNA in the pathogenesis of OP has also attracted widespread attention from scholars in recent years. Type H vessels are unique microvessels of the bone and have become a new focus in the pathogenesis of OP because they play an essential role in osteogenesis-angiogenesis coupling. Previous studies found some miRNAs regulate type H vessel formation through the regulatory factors, including platelet-derived growth factor-BB (PDGF-BB), hypoxia-inducible factor 1α (HIF-1α), vascular endothelial growth factor (VEGF), and so on. These findings help us gain a more in-depth understanding of the relationship among miRNAs, type H vessels, and OP to find a new perspective on treating OP. In the present mini-review, we will introduce the role of type H vessels in the pathogenesis of OP and the regulation of miRNAs on type H vessel formation by affecting regulatory factors to provide some valuable insights for future studies of OP treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    延迟的萎缩性愈合和不愈合所带来的挑战是骨质疏松性骨折治疗中的巨大障碍。H型血管生成和成骨过程是骨再生过程中的关键机制。值得注意的是,低氧条件下脂肪干细胞(ADSC)外泌体的预处理因其增加外泌体分泌和功能的潜力而备受关注.在目前的调查中,我们着手全面阐明骨质疏松性骨再生环境中hypo-ADSC-Exos的潜在机制。我们的发现揭示了hypo-ADSC-Exos拥有一个卓越的miRNA,即,miR-21-5p,成为血管生成效应的主要协调者。通过体外实验,我们证明了hypo-ADSC-Exos刺激增殖的能力,迁移,以及通过miR-21-5p介导人脐静脉内皮细胞(HUVECs)的血管生成潜力。miR-21-5p的抑制有效地减弱了由hypo-ADSC-Exos介导的促血管生成作用。机械上,我们的研究表明,来自hypo-ADSCs的外泌体miR-21-5p通过靶向HUVECs内的sprouly1(SPRY1)发挥其调控作用,从而促进PI3K/AKT信号通路的激活。值得注意的是,发现HUVEC中SPRY1的敲低可增强PI3K/AKT的激活,同时,HUVEC增殖,迁移,和血管生成。我们研究的高潮阶段涉及令人信服的体内演示,其中验证了装载有hypo-ADSC-Exos的GelMA可显著增强局部H型血管生成和伴随的骨再生。这种增强明确归因于SPRY1的外泌体调节。总之,我们的调查为hypo-ADSC-Exos在骨质疏松性骨折治疗中的潜在效用提供了一个开创性的观点.
    The challenges posed by delayed atrophic healing and nonunion stand as formidable obstacles in osteoporotic fracture treatment. The processes of type H angiogenesis and osteogenesis emerge as pivotal mechanisms during bone regeneration. Notably, the preconditioning of adipose-derived stem cell (ADSC) exosomes under hypoxic conditions has garnered attention for its potential to augment the secretion and functionality of these exosomes. In the present investigation, we embarked upon a comprehensive elucidation of the underlying mechanisms of hypo-ADSC-Exos within the milieu of osteoporotic bone regeneration. Our findings revealed that hypo-ADSC-Exos harboured a preeminent miRNA, namely, miR-21-5p, which emerged as the principal orchestrator of angiogenic effects. Through in vitro experiments, we demonstrated the capacity of hypo-ADSC-Exos to stimulate the proliferation, migration, and angiogenic potential of human umbilical vein endothelial cells (HUVECs) via the mediation of miR-21-5p. The inhibition of miR-21-5p effectively attenuated the proangiogenic effects mediated by hypo-ADSC-Exos. Mechanistically, our investigation revealed that exosomal miR-21-5p emanating from hypo-ADSCs exerts its regulatory influence by targeting sprouly1 (SPRY1) within HUVECs, thereby facilitating the activation of the PI3K/AKT signalling pathway. Notably, knockdown of SPRY1 in HUVECs was found to potentiate PI3K/AKT activation and, concomitantly, HUVEC proliferation, migration, and angiogenesis. The culminating stage of our study involved a compelling in vivo demonstration wherein GelMA loaded with hypo-ADSC-Exos was validated to substantially enhance local type H angiogenesis and concomitant bone regeneration. This enhancement was unequivocally attributed to the exosomal modulation of SPRY1. In summary, our investigation offers a pioneering perspective on the potential utility of hypo-ADSC-Exos as readily available for osteoporotic fracture treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    血管是骨骼发育和新陈代谢所必需的。骨H型血管,以CD31和Endomucin(Emcn)的高表达而得名,最近有报道称主要位于干phy端,表现出不同的分子特性,并耦合成骨和血管生成。H型血管与骨代谢之间的强相关性现已得到公认。H型血管与骨祖细胞之间的串扰也参与骨代谢相关疾病,如骨质疏松症,骨关节炎,骨折愈合和骨缺损。以H型血管形成为目标可能成为管理多种骨骼疾病的新方法。本文综述了H型血管在骨相关疾病中的作用,并总结了有针对性的干预措施的研究尝试。这将有助于我们更好地了解它们在临床应用中的潜在价值。
    Blood vessels are essential for bone development and metabolism. Type H vessels in bone, named after their high expression of CD31 and Endomucin (Emcn), have recently been reported to locate mainly in the metaphysis, exhibit different molecular properties and couple osteogenesis and angiogenesis. A strong correlation between type H vessels and bone metabolism is now well-recognized. The crosstalk between type H vessels and osteoprogenitor cells is also involved in bone metabolism-related diseases such as osteoporosis, osteoarthritis, fracture healing and bone defects. Targeting the type H vessel formation may become a new approach for managing a variety of bone diseases. This review highlighted the roles of type H vessels in bone-related diseases and summarized the research attempts to develop targeted intervention, which will help us gain a better understanding of their potential value in clinical application.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    设计可以调节先天免疫反应并促进血管化骨再生的支架为骨组织工程提供了希望。在这里,通过将修复性M2巨噬细胞衍生的外泌体锚定到拓扑孔结构的纳米纤维支架上,制造了结合物理和生物线索的电纺支架。纤维的拓扑孔结构和外泌体的固定增加了纤维支架的纳米级粗糙度和亲水性。体外细胞实验表明,外泌体可以被靶细胞内化,促进细胞迁移,管形成,成骨分化,和抗炎巨噬细胞极化。纤维化的激活,血管生成,在小鼠皮下植入外泌体功能化纤维支架介导的异物反应(FBR)过程中,巨噬细胞得到了阐明。外泌体功能化的纳米纤维支架还增强了临界大小的大鼠颅骨缺损模型中的血管化骨形成。重要的是,组织学分析表明,生物功能支架调节血管生成的偶联作用,破骨细胞生成,和通过刺激H型血管形成成骨。本研究阐述了纤维支架介导的FBR和血管化骨再生过程中细胞微环境生态位内的复杂过程,以指导骨科和颌面外科植入物或器械的设计。重要声明:如何设计能够调节局部免疫生态位并真正实现功能性血管化骨再生的支架材料仍然是一个悬而未决的问题。这里,结合物理和生物线索,我们提出了无细胞和无生长因子治疗的新见解,将修复性M2巨噬细胞衍生的外泌体锚定到拓扑孔结构的纳米纤维支架上。外泌体功能化支架系统减轻了异物反应,包括过度的纤维化,肿瘤样血管化,和巨噬细胞激活。重要的是,生物功能支架调节血管生成的偶联效应,破骨细胞生成,和通过刺激H型血管形成成骨。
    Designing scaffolds that can regulate the innate immune response and promote vascularized bone regeneration holds promise for bone tissue engineering. Herein, electrospun scaffolds that combined physical and biological cues were fabricated by anchoring reparative M2 macrophage-derived exosomes onto topological pore structured nanofibrous scaffolds. The topological pore structure of the fiber and the immobilization of exosomes increased the nanoscale roughness and hydrophilicity of the fibrous scaffold. In vitro cell experiments showed that exosomes could be internalized by target cells to promote cell migration, tube formation, osteogenic differentiation, and anti-inflammatory macrophage polarization. The activation of fibrosis, angiogenesis, and macrophage was elucidated during the exosome-functionalized fibrous scaffold-mediated foreign body response (FBR) in subcutaneous implantation in mice. The exosome-functionalized nanofibrous scaffolds also enhanced vascularized bone formation in a critical-sized rat cranial bone defect model. Importantly, histological analysis revealed that the biofunctional scaffolds regulated the coupling effect of angiogenesis, osteoclastogenesis, and osteogenesis by stimulating type H vessel formation. This study elaborated on the complex processes within the cell microenvironment niche during fibrous scaffold-mediated FBR and vascularized bone regeneration to guide the design of implants or devices used in orthopedics and maxillofacial surgery. STATEMENT OF SIGNIFICANCE: How to design scaffold materials that can regulate the local immune niche and truly achieve functional vascularized bone regeneration still remain an open question. Here, combining physical and biological cues, we proposed new insight to cell-free and growth factor-free therapy, anchoring reparative M2 macrophage-derived exosomes onto topological pore structured nanofibrous scaffolds. The exosomes functionalized-scaffold system mitigated foreign body response, including excessive fibrosis, tumor-like vascularization, and macrophage activation. Importantly, the biofunctional scaffolds regulated the coupling effect of angiogenesis, osteoclastogenesis, and osteogenesis by stimulating type H vessel formation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    血管系统在骨组织中起着至关重要的作用。血管生成和成骨过程通过时空连接耦合。最近的研究已经确定了骨骼系统中的三种类型的毛细血管。与L型和E型血管相比,H型血管表达高水平的CD31和内粘蛋白,和功能耦合血管生成和成骨。H型血管中的内皮细胞与骨细胞相互作用(例如,成骨细胞,破骨细胞,和骨细胞)通过细胞因子或信号通路维持骨骼生长和稳态。在不平衡的骨骼稳态中,比如骨质疏松症和骨关节炎,调节H型血管内皮细胞活性修复失衡可能是一种新的治疗策略。这里,本文综述了血管生成与成骨耦合相关因子或信号通路的最新进展。这篇综述将有助于进一步了解H型血管在耦合血管生成和成骨过程中的作用和机制。此外,它将通过靶向H型血管促进骨疾病治疗方法的发展。
    The vascular system plays a crucial role in bone tissue. Angiogenic and osteogenic processes are coupled through a spatial-temporal connection. Recent studies have identified three types of capillaries in the skeletal system. Compared with type L and E vessels, type H vessels express high levels of CD31 and endomucin, and function to couple angiogenesis and osteogenesis. Endothelial cells in type H vessels interact with osteolineage cells (e.g., osteoblasts, osteoclasts, and osteocytes) through cytokines or signaling pathways to maintain bone growth and homeostasis. In imbalanced bone homeostases, such as osteoporosis and osteoarthritis, it may be a new therapeutic strategy to regulate the endothelial cell activity in type H vessels to repair the imbalance. Here, we reviewed the latest progress in relevant factors or signaling pathways in coupling angiogenesis and osteogenesis. This review would contribute to further understanding the role and mechanisms of type H vessels in coupling angiogenic and osteogenic processes. Furthermore, it will facilitate the development of therapeutic approaches for bone disorders by targeting type H vessels.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    肌腱-骨界面(TBI)整合不良是导致前交叉韧带(ACL)重建后患者愈合质量不佳的主要原因之一。最近发现H型血管通过调节骨-血管生成串扰密切调节骨形成。因此,有利于H型血管形成的策略可能是改善移植物骨整合的有希望的治疗方法。在这项研究中,我们首次报道了狭缝引导配体3(slit3)的治疗结果,一种有利于H型血管形成的新型促血管生成因子,在ACL重建小鼠的TBI愈合中。将小鼠(n=87)分为三组进行各种治疗:水凝胶微粒(HMP,对照组),slit3@HMP,和slit3中和抗体@HMP(slit3-AB@HMP)。组织学分析,步态表现,射线照相测量,并进行生物力学测试以评估TBI愈合质量。与HMP组相比,在Slit3@HMP组中,在TBI处形成了骨向内生长增加和纤维瘢痕组织减少。同时,相对于HMP组,slit3-AB@HMP抑制骨向内生长并增加纤维瘢痕组织的形成。与HMP组相比,slit3@HMP有利于TBI的H型血管形成,而slit3-AB@HMP阻碍了它。根据Micro-CT评估,与HMP组相比,slit3@HMP显着增加了隧道周围的骨量,而slit3-AB@HMP显着减少了隧道周围的骨量。Slit3@HMP组中的小鼠在站立时间方面表现出最佳的步态表现,步幅长度,爪印区域,和立场压力。动态松弛度测量和拉伸测试显示,相对于其他两组,slit3@HMP组表现出显着降低的松弛位移和改善的破坏载荷和刚度。总的来说,注射Slit3可用于增强腱-骨整合,这可能归因于H型血管耦合的血管生成-成骨串扰的调节。
    Poor tendon-bone interface (TBI) integration is one of the major causes contributing to unsatisfactory healing quality in patients after anterior cruciate ligament (ACL) reconstruction. Type H vessels have been recently found to closely modulate bone formation via regulation of the osteo-angiogenic crosstalk, so the strategies favoring type H vessel formation may be promising therapeutic approaches for improved graft osteointegration. In this study, we reported for the first time the treatment outcome of slit guidance ligand 3 (slit3), a novel proangiogenic factor favoring type H vessel formation, in TBI healing in mice with ACL reconstruction. The mice (n = 87) were divided into three groups for various treatments: hydrogel microparticles (HMP, control group), slit3@HMP, and slit3 neutralizing antibody@HMP (slit3-AB@HMP). Histological analysis, gait performance, radiographic measurement, and biomechanical testing were performed to assess the TBI healing quality. Increased bony ingrowth and reduced fibrous scar tissue was formed at the TBI in the slit3@HMP group when compared to the HMP group. Meanwhile, the slit3-AB@HMP inhibited the osseous ingrowth and increased fibrous scar tissue formation relative to the HMP group. Compared to the HMP group, the slit3@HMP favored type H vessel formation at the TBI while the slit3-AB@HMP impeded it. According to micro-CT assessment, compared to the HMP group, the slit3@HMP significantly increased the peri-tunnel bone mass while the slit3-AB@HMP significantly reduced the peri-tunnel bone mass. The mice in the slit3@HMP group showed the best gait performance in terms of stance time, stride length, paw print area, and stance pressure. Dynamic laxity measurement and tensile testing showed the slit3@HMP group exhibited significantly reduced laxity displacement and improved failure load and stiffness relative to the other two groups. Collectively, the injection of slit3 could be used to enhance tendon-bone integration, which may be ascribed to modulation of angiogenesis-osteogenesis crosstalk coupled by type H vessels.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    UNASSIGNED: To summarize the regulatory effect of non-coding RNA (ncRNA) on type H vessels angiogenesis of bone.
    UNASSIGNED: Recent domestic and foreign related literature about the regulation of ncRNA in type H vessels angiogenesis was widely reviewed and summarized.
    UNASSIGNED: Type H vessels is a special subtype of bone vessels with the ability to couple bone formation. At present, the research on ncRNA regulating type H vessels angiogenesis in bone diseases mainly focuses on microRNA, long ncRNA, and small interfering RNA, which can affect the expressions of hypoxia inducible factor 1α, platelet derived growth factor BB, slit guidance ligand 3, and other factors through their own unique ways of action, thus regulating type H vessels angiogenesis and participating in the occurrence and development of bone diseases.
    UNASSIGNED: At present, the mechanism of ncRNA regulating bone type H vessels angiogenesis has been preliminarily explored. With the deepening of research, ncRNA is expected to be a new target for the diagnosis and treatment of vascular related bone diseases.
    UNASSIGNED: 对非编码RNA(non-coding RNA,ncRNA)调控骨H型血管生成的相关研究进行综述。.
    UNASSIGNED: 广泛查阅近年来国内外有关ncRNA 调控骨H型血管生成的研究报道,并进行归纳总结。.
    UNASSIGNED: H型血管是一种特殊的骨血管亚型,具有偶联骨形成的能力。目前对于ncRNA在骨骼疾病中调控H型血管生成的研究主要集中于微小RNA、长链非编码RNA以及小干扰RNA,其可通过各自特有的作用方式影响缺氧诱导因子1α、PDGF-BB、神经轴突导向因子3等因子的表达,从而调控H型血管生成,参与骨骼疾病的发生发展。.
    UNASSIGNED: 目前对于ncRNA调控H型血管生成的作用机制已有初步探索,随着研究深入,ncRNA有望作为诊断与治疗血管相关性骨骼疾病的新靶点。.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    由于界面骨结合不足,骨科植入物的故障率很高,尤其是在骨质疏松的情况下。H型血管是CD31+EMCN+毛细血管,在介导新骨形成中起关键作用,但是它们在骨质疏松性骨折部位的丰度是非常有限的。在这里,我们报道了一种纳米工程复合涂层,以改善用于骨质疏松性骨折修复的钛植入物的原位骨整合,这是通过抑制内皮细胞(EC)中干扰素基因(STING)的刺激物来刺激H型血管形成来实现的。通过STINGmRNA降解DNA酶的滚环扩增(RCA)制备了自主催化DNA酶-ZnO纳米花(DNFzns),然后将其整合在Ti表面上,并进一步与硫代酮桥连的聚多巴胺和柚皮素(Ti/DNFzn/PDA-Nar)依次络合。ECs和间充质干细胞(MSCs)可以通过电流驱动被募集到植入物表面,考虑DNFzn/PDA-Nar的负电荷,随后在活性氧(ROS)刺激下释放Nar,以上调募集的ECs中的内皮型一氧化氮合酶(eNOS),导致局部血管生成增强。同时,协调释放的DNFzns将取消ECs中的STING表达,将新形成的血管转化为H型血管,从而大大促进钛植入物的骨整合。本研究为改善种植体骨整合治疗骨质疏松性骨折提供了应用前景。
    Orthopedic implants have a high failure rate due to insufficient interfacial osseointegration, especially under osteoporotic conditions. Type H vessels are CD31+EMCN+ capillaries with crucial roles in mediating new bone formation, but their abundance in osteoporotic fracture site is highly limited. Herein, we report a nanoengineered composite coating to improve the in situ osseointegration of a Ti implant for osteoporotic fracture repair, which is realized through inhibiting the stimulator of interferon genes (STING) in endothelial cells (ECs) to stimulate type H vessel formation. Autonomously catalytic DNAzyme-ZnO nanoflowers (DNFzns) were prepared through rolling circle amplification (RCA) of STING mRNA-degrading DNAzymes, which were then integrated on the Ti surface and further sequentially complexed with thioketal-bridged polydopamine and naringenin (Ti/DNFzn/PDA-Nar). ECs and mesenchymal stem cells (MSCs) can be recruited to the implant surface by galvanotaxis, accounting for the negative charges of DNFzn/PDA-Nar, subsequently released Nar under reactive oxygen species (ROS) stimulation to upregulate endothelial nitric oxide synthase (eNOS) in recruited ECs, leading to enhanced local angiogenesis. Meanwhile, the coordinately released DNFzns would abolish STING expression in ECs to transform the newly formed vessels into Type H vessels, thus substantially promoting the osseointegration of Ti implants. This study provides application prospects for improving implant osteointegration for osteoporotic fracture treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    在骨修复过程中,钙化软骨骨痂(CACC)的形成至关重要。CACC可以刺激H型血管侵入愈伤组织,使血管生成和成骨,诱导破骨细胞再吸收钙化基质,促进破骨细胞分泌因子,增强成骨,最终实现骨软骨的置换。在这项研究中,使用3D打印开发了多孔聚己内酯/羟基磷灰石-亚氨基二乙酸-去铁胺(PCL/HA-SF-DFO)3D仿生CACC。多孔结构可以模拟软骨基质金属蛋白酶降解形成的孔隙,含HA的PCL可以模拟钙化的软骨基质,和SF锚定DFO到HA的DFO的缓慢释放。体外实验结果表明该支架能显著增强血管生成,促进破骨细胞生成和吸收,并通过促进破骨细胞表达含胶原三螺旋重复序列1来增强骨髓基质干细胞的成骨分化。体内实验结果表明,该支架能显著促进H型血管的形成和偶联因子的表达,从而促进成骨,最终促进大鼠大段骨缺损的再生,防止内固定螺钉脱落。总之,受生物骨修复过程启发的支架有效促进骨再生。
    The formation of a calcified cartilaginous callus (CACC) is crucial during bone repair. CACC can stimulate the invasion of type H vessels into the callus to couple angiogenesis and osteogenesis, induce osteoclastogenesis to resorb the calcified matrix, and promote osteoclast secretion of factors to enhance osteogenesis, ultimately achieving the replacement of cartilage with bone. In this study, a porous polycaprolactone/hydroxyapatite-iminodiacetic acid-deferoxamine (PCL/HA-SF-DFO) 3D biomimetic CACC is developed using 3D printing. The porous structure can mimic the pores formed by the matrix metalloproteinase degradation of the cartilaginous matrix, HA-containing PCL can mimic the calcified cartilaginous matrix, and SF anchors DFO onto HA for the slow release of DFO. The in vitro results show that the scaffold significantly enhances angiogenesis, promotes osteoclastogenesis and resorption by osteoclasts, and enhances the osteogenic differentiation of bone marrow stromal stem cells by promoting collagen triple helix repeat-containing 1 expression by osteoclasts. The in vivo results show that the scaffold significantly promotes type H vessels formation and the expression of coupling factors to promote osteogenesis, ultimately enhancing the regeneration of large-segment bone defects in rats and preventing dislodging of the internal fixation screw. In conclusion, the scaffold inspired by biological bone repair processes effectively promotes bone regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号