Dynamic protein corona influences immune-modulating osteogenesis in magnetic nanoparticle (MNP)-infiltrated bone regeneration scaffoldsin vivo

Nanoscale ◽  
2019 ◽  
Vol 11 (14) ◽  
pp. 6817-6827 ◽  
Author(s):  
Yue Zhu ◽  
Peipei Jiang ◽  
Bin Luo ◽  
Fang Lan ◽  
Jing He ◽  
...  

An inflammatory reaction initiates fracture healing and directly influences the osteoinductive effect of the magnetic hydroxyapatite (MHA) scaffold, but the underlying mechanism is yet to be elucidated.

Author(s):  
Yanping Gong ◽  
Yang Wang ◽  
Yiqing Zhang ◽  
Liangchen Wang ◽  
Lijuan Wan ◽  
...  

Bone regeneration is a delicate physiological process. Non-union and delayed fracture healing remains a great challenge in clinical practice nowadays. Bone and fat hold a close relationship to remain balanced through hormones and cytokines. Adiponectin is a well-known protein to maintain the hemostasis, which may be an interesting target for fracture healing. Herein, we provided a facile and efficient method to obtain high-purity and high-yield recombinant human adiponectin (ADPN). The biocompatibility and the pharmaceutical behaviors were evaluated in Sprague–Dawley rats. The paracrine effects of adiponectin on bone fracture healing were investigated with a rat tibia fracture model via intrabone injection. Significantly accelerated bone healing was observed in the medulla injection group, indicating the paracrine effects of adiponectin could be potentially utilized for clinical treatments. The underlying mechanism was primarily assessed, and the expression of osteogenic markers, including bone morphogenic protein 2, alkaline phosphatase, and osteocalcin, along with adiponectin receptor 1 (AdipoR1), was markedly increased at the fracture site. The increased bone healing of ADPN treatment may result from both enhanced osteogenic proliferation as well as differentiation. Cell experiments confirmed that the expression of osteogenesis markers increased significantly in ADPN treatment groups, while it decreased when the expression of AdipoR1 was knocked down by siRNA. Our study provided a feasible and efficacious way for bone fracture treatment with local administration of ADPN, which could be rapidly translated into the clinics.


2021 ◽  
Author(s):  
Yanghua Tang ◽  
Yafeng Mo ◽  
Dawei Xin ◽  
Zhenfei Xiong ◽  
Linru Zeng ◽  
...  

Purpose: To investigate the effects of β-ecdysterone on fracture healing and the underlying mechanism. Methods: MTT assay was used to detect the cell viability and alkaline phosphatase (ALP) activity was measured using a commercial kit. AO/PI and flow cytometry assays were used to determine the state of apoptosis of osteoblasts. The expression level of RunX2, ATG7 and LC3 was evaluated by qRT-PCR and Western blot assays. X-ray and HE staining were conducted on the fractured femur to evaluate the pathological state. Immunohistochemical assay was used to detect the expression level of Beclin-1 and immunofluorescence assay was used to measure the expression level of LC3 in the fractured femurs. Western blot was utilized to determine the expression level of PI3K, p-AKT1, AKT1, p-mTOR, mTOR, p-p70S6K, and p70S6K. Results: The ALP activity and expression of RunX2 in fractured osteoblasts were significantly suppressed by 3-methyladenine and elevated by rapamycin, 60, and 80 μM β-ecdysterone. The apoptotic state of fractured osteoblasts was enhanced by 3-methyladenine and alleviated by rapamycin, 60, and 80 μM β-ecdysterone. The state of autophagy both in fractured osteoblasts and femurs was inhibited by 3-methyladenine and facilitated by rapamycin and β-ecdysterone. Compared to control, Garrett score in 3-methyladenine group was significantly decreased and promoted in rapamycin and β-ecdysterone groups, accompanied by ameliorated pathological state. Lastly, the PI3K/AKT/mTOR pathway both in fractured osteoblasts and femurs was activated by 3-methyladenine and inhibited by rapamycin and β-ecdysterone. Conclusion: β-ecdysterone might facilitate fracture healing by activating autophagy through suppressing PI3K/AKT/mTOR signal pathway.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Nina Schmitz ◽  
Melanie Timmen ◽  
Katharina Kostka ◽  
Verena Hoerr ◽  
Christian Schwarz ◽  
...  

Abstract Over the last years, murine in vivo magnetic resonance imaging (MRI) contributed to a new understanding of tissue composition, regeneration and diseases. Due to artefacts generated by the currently used metal implants, MRI is limited in fracture healing research so far. In this study, we investigated a novel MRI-compatible, ceramic intramedullary fracture implant during bone regeneration in mice. Three-point-bending revealed a higher stiffness of the ceramic material compared to the metal implants. Electron microscopy displayed a rough surface of the ceramic implant that was comparable to standard metal devices and allowed cell attachment and growth of osteoblastic cells. MicroCT-imaging illustrated the development of the callus around the fracture site indicating a regular progressing healing process when using the novel implant. In MRI, different callus tissues and the implant could clearly be distinguished from each other without any artefacts. Monitoring fracture healing using MRI-compatible implants will improve our knowledge of callus tissue regeneration by 3D insights longitudinal in the same living organism, which might also help to reduce the consumption of animals for future fracture healing studies, significantly. Finally, this study may be translated into clinical application to improve our knowledge about human bone regeneration.


2018 ◽  
Vol 20 (1) ◽  
pp. 83 ◽  
Author(s):  
Gabriele Russow ◽  
Denise Jahn ◽  
Jessika Appelt ◽  
Sven Märdian ◽  
Serafeim Tsitsilonis ◽  
...  

Osteoporosis represents the most common bone disease worldwide and results in a significantly increased fracture risk. Extrinsic and intrinsic factors implicated in the development of osteoporosis are also associated with delayed fracture healing and impaired bone regeneration. Based on a steadily increasing life expectancy in modern societies, the global implications of osteoporosis and impaired bone healing are substantial. Research in the last decades has revealed several molecular pathways that stimulate bone formation and could be targeted to treat both osteoporosis and impaired fracture healing. The identification and development of therapeutic approaches modulating bone formation, rather than bone resorption, fulfils an essential clinical need, as treatment options for reversing bone loss and promoting bone regeneration are limited. This review focuses on currently available and future approaches that may have the potential to achieve these aims.


JCI Insight ◽  
2020 ◽  
Vol 5 (9) ◽  
Author(s):  
Nadine Schall ◽  
Julian J. Garcia ◽  
Hema Kalyanaraman ◽  
Shyamsundar Pal China ◽  
Jenna J. Lee ◽  
...  

2021 ◽  
Author(s):  
Kai Hang ◽  
Li Ying ◽  
Jinwu Bai ◽  
Yibo Wang ◽  
Zhihui Kuang ◽  
...  

Abstract Background Managing healing of impaired bone fracture has long been a subject of concern. Prolonged or uncontrolled inflammation exerts detrimental effects on bone healing. Tumor necrosis factor (TNF)-α is a critical inflammatory factor, whose absence impairs normal bone formation. However, TNF-α exposure for longer duration inhibits bone regeneration. In this study, we aim to find a new therapeutic target for the impaired osteogenesis induced by long exposure of TNF-α.MethodsIn vitro, mRNA microarray analysis was used to identify differentially expressed genes. Cell proliferation assay was used to assess the proliferation of cells. qPCR and Western blotting analysis were applied to detect the expression of target genes and proteins respectively. ALP staining and Alizarin Red staining (ARS) were used to evaluate ALP activity and mineral deposition respectively. Co-immunoprecipitation was used to detect the interaction of proteins. In vivo, a murie tibial fracture model was established, histological evaluation and radiographic analysis was used to confirm bone regeneration in fracture healing. statistical significance between two groups was determined by Student’s t test, one-way ANOVA or Bonferroni’s post-hoc test according to the distribution of the tested population.ResultsIn this study, we show that heat shock protein family B (small) member 7 (HSPB7) mitigates negative impact of long-term TNF-α exposure on bone formation by binding heat shock protein family H (small) member 1 (HSPH1). HSPH1, in turn, inhibits the ATPase of heat shock cognate A8 (HSCA8), one of the key components involved in chaperone-mediated autophagy (CMA). Deletion of genes encoding for either HSCA8 or lysosome-associated membrane protein 2A (LAMP2A), another component of CMA, failed to reverse the osteogenic differentiation of hBMSCs induced by TNF-α by deleting HSPH1. Moreover, LAMP2A overexpression reverse the impaired osteogenesis induced by TNF-α, and this effect was attenuated by DKK1, a specific Wnt/β-catenin signaling pathway inhibitors. Thus, a heat shock protein family network composed by HAPB7, HSPH1 and HSCA8 rescued the impairment of bone healing by TNF-α through the CMA/β-catenin pathway, making it a potential therapeutic agent for bone regeneration in cases of prolonged or severe inflammation in the clinical settings. ConclusionsTaken together, these findings indicate that a heat shock protein family network including HSPB7, HSPH1 and HSCA8 protects the impaired osteogenesis induced by TNF-α via the CMA/β-catenin pathway. And in vivo, HSPB7 overexpression lentiviral particles effectively protects the impaired fracture healing induced by TNF-α in a mouse tibia fracture healing model.


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Dong Zhang ◽  
Yifan Wu ◽  
Zonghuan Li ◽  
Hairen Chen ◽  
Siyuan Huang ◽  
...  

Abstract Background Patients with diabetes have an increased risk of nonunion and delayed union of fractures. Macrophages have been shown as a key player in diabetic complications. However, it remains obscure how diabetic milieu affects macrophage-derived exosomes and its implications on osteogenic differentiation of BMSCs. In this study, we aim to define the impact of diabetic milieu on macrophage-derived exosomes, role of extracellular vesicles in intercellular communication with BMSCs, and subsequent effects on osteogenic differentiation and fracture repair. Results The osteogenic potential and the ability of fracture repair of exosomes derived from diabetic bone marrow-derived macrophages (dBMDM-exos) were revealed to be lower, as compared with non-diabetic bone marrow-derived macrophages (nBMDM-exos) in vitro and in vivo. Interestingly, miR-144-5p levels were sharply elevated in dBMDM-exos and it could be transferred into BMSCs to regulate bone regeneration by targeting Smad1. In addition, the adverse effects of dBMDM-exos on the osteogenic potential and the ability of fracture repair were reversed through the suppression of miR-144-5p inhibition in vitro and vivo. Conclusions The results demonstrated an important role of exosomal miR-144-5p in bone regeneration, offering insight into developing new strategy for the improvement of fracture healing in patients with diabetes mellitus. Graphic Abstract


2019 ◽  
Vol 20 (22) ◽  
pp. 5805 ◽  
Author(s):  
Carsten W. Schlickewei ◽  
Holger Kleinertz ◽  
Darius M. Thiesen ◽  
Konrad Mader ◽  
Matthias Priemel ◽  
...  

Bone regeneration represents a complex process, of which basic biologic principles have been evolutionarily conserved over a broad range of different species. Bone represents one of few tissues that can heal without forming a fibrous scar and, as such, resembles a unique form of tissue regeneration. Despite a tremendous improvement in surgical techniques in the past decades, impaired bone regeneration including non-unions still affect a significant number of patients with fractures. As impaired bone regeneration is associated with high socio-economic implications, it is an essential clinical need to gain a full understanding of the pathophysiology and identify novel treatment approaches. This review focuses on the clinical implications of impaired bone regeneration, including currently available treatment options. Moreover, recent advances in the understanding of fracture healing are discussed, which have resulted in the identification and development of novel therapeutic approaches for affected patients.


Sign in / Sign up

Export Citation Format

Share Document