osteoblast apoptosis
Recently Published Documents


TOTAL DOCUMENTS

120
(FIVE YEARS 33)

H-INDEX

33
(FIVE YEARS 3)

Author(s):  
Chen Yue ◽  
Hongting Jin ◽  
Xue Zhang ◽  
Wuyin Li ◽  
Deli Wang ◽  
...  

2021 ◽  
Vol 22 (5) ◽  
Author(s):  
Keda Yang ◽  
Lei Pei ◽  
Siming Zhou ◽  
Lin Tao ◽  
Yue Zhu

2021 ◽  
Author(s):  
Carley V. Cook ◽  
Mohammad Aminul Islam ◽  
Brenda J. Smith ◽  
Ashlee N. Ford Versypt

Bone health is determined by many factors including bone metabolism or remodeling. Wnt-10b has been shown to alter osteoblastogenesis through pre-osteoblast proliferation and differentiation as well as the osteoblast apoptosis rate, which collectively lead to the increase of bone density. To model this change, we adapted a previously published model of bone remodeling. The resulting model is a single compartment system that includes ordinary differential equations for active osteoclasts, pre-osteoblasts, osteoblasts, and osteocytes and a differential equation that tracks the amount of bone present at the remodeling site. Our alterations to the original model consist of extending it past a single remodeling cycle and implementing a direct relationship to Wnt-10b. Four new parameters were estimated and validated using normalized data from mice. The model connects Wnt-10b to bone metabolism and predicts the change in bone volume caused by a change in Wnt-10b. We find that this model predicts the expected increase in pre-osteoblasts and osteoblasts while also pointing to a decrease in osteoclasts when Wnt-10b is increased.


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 1353.2-1353
Author(s):  
J. Sun ◽  
W. Zheng ◽  
Y. Zhang ◽  
Z. Gu

Background:Osteonecrosis of the femoral head (ONFH) is a common disease caused by many trauma factors and un-trauma factors. Among those un-trauma factors, steroid-induced osteonecrosis of the femoral head (SNFH) accounted for a large proportion and mainly concentrated in young people. SNFH has been reported as an irreversible disease and associated with the damage of blood vessels and the loss balance of bone homeostasis. Circulating endothelial progenitor cells (CEPCs), one part of circulating endothelial cells (CECs), are immature precursor cells with proliferative potential. The damage of vascular endothelial cells in SNFH has been confirmed by many studies, but the changes of CECs and CEPCs in the peripheral blood of patients with SNFH have not been studied yet.Objectives:The objective of the study is to explore the number of CECs and CEPCs in SNFH patients and normal people and then investigate whether EC-secreted exosomes (EC-exos) could prevent the progression of SNFH in rat model and its mechanism of action.Methods:We collect peripheral blood of 3 SNFH patients and 3 heathy people and detected the levels of CECs and CEPCs by Flow cytometer. TEM, NTA and western blot was used to characterize the isolated EC-exos. Annexin V-FITC/PI double staining with flow cytometric analysis and western blot were used to evaluate MC3T3-E1 cells apoptosis. CCK-8, scratching experiment and transwell were used to evaluate MC3T3-E1 cells viability and migration ability. Micro-CT and morphological staining were used to evaluate the progress of SNFH in rat model.Results:Firstly, we found that the number of CECs and CEPCs in the peripheral blood was decreased in SNFH patients than normal people. Then our results indicated that EC-exos could improve the migration, viability and prevent apoptosis of osteoblasts under dexamethasone by activating the PI3K/AKT/Bcl-2 signal pathway in vitro. Finally, our Micro-CT and morphological staining results in SNFH rat model revealed that EC-exos prevented the progression of SNFH.Conclusion:EC-exos could enhance the cell viability and migration ability of osteoblasts under dexamethasone and play an anti-apoptosis role against steroids-induced osteoblast apoptosis by activating the PI3K/AKT/Bcl-2 signal pathway. EC-exos prevented the progression of SNFH in rat model.References:[1]Zalavras CG, Lieberman JR. Osteonecrosis of the femoral head: evaluation and treatment. J Am Acad Orthop Surg. 2014;22(7):455-64.[2]Microsurgery Department of the Orthopedics Branch of the Chinese Medical Doctor A, Group from the O, Bone Defect Branch of the Chinese Association of R, Reconstructive S, Microsurgery, Reconstructive Surgery Group of the Orthopedics Branch of the Chinese Medical A. Chinese Guideline for the Diagnosis and Treatment of Osteonecrosis of the Femoral Head in Adults. Orthop Surg. 2017;9(1):3-12.[3]Mont MA, Jones LC, Hungerford DS. Nontraumatic osteonecrosis of the femoral head: ten years later. J Bone Joint Surg Am. 2006;88(5):1117-32.[4]Yuan HF, Zhang J, Guo CA, Yan ZQ. Clinical outcomes of osteonecrosis of the femoral head after autologous bone marrow stem cell implantation: a meta-analysis of seven case-control studies. Clinics (Sao Paulo). 2016;71(2):110-3.[5]Houdek MT, Wyles CC, Packard BD, Terzic A, Behfar A, Sierra RJ. Decreased Osteogenic Activity of Mesenchymal Stem Cells in Patients With Corticosteroid-Induced Osteonecrosis of the Femoral Head. J Arthroplasty. 2016;31(4):893-8.Disclosure of Interests:None declared.


2021 ◽  
Vol 16 (1) ◽  
Author(s):  
Haiping Zhang ◽  
Ziliang Yu ◽  
Farui Sun ◽  
Jin Jin

Abstract Background The purpose of the current study was to explore the role and underlying mechanism of cellular retinoic acid binding protein 2 (CRABP2) in dexamethasone (DEX)-induced apoptosis in human osteoblast cells. Methods GSE10311 was downloaded from the Gene Expression Omnibus (GEO) database to identify the differentially expressed genes (DEGs) by the limma/R package. Primary human osteoblast was isolated and treated with different concentration of DEX (0, 10-8, 10-7, 10-6, 10-5, and 10-4 mol/L), and cell viability and flow cytometry were used to detect cell proliferation and apoptosis. A CRABP2 overexpression plasmid (oe-CRABP2) was used to overexpress CRABP2, and western blotting was conducted to detect protein expression. Results We found that CRABP2 was downregulated in the DEX-treated group. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses indicated that DEGs were associated with PI3K/Akt signaling pathway. DEX downregulated CRABP2 gene and protein expression, inhibited viability, and induced human osteoblast apoptosis. Overexpression of CRABP2 reversed DEX-induced apoptosis in human osteoblast. Moreover, overexpression of CRABP2 delayed the progression of DEX-induced osteonecrosis of the femoral head (ONFH) animal model. Conclusion In conclusion, CRABP2 is effective at inhibiting DEX-induced human osteoblast apoptosis and delayed ONFH progression.


IUBMB Life ◽  
2020 ◽  
Author(s):  
Hu Liu ◽  
Yu‐Wu Wang ◽  
Wei‐Dong Chen ◽  
Hong‐Hua Dong ◽  
You‐Jia Xu

2020 ◽  
Vol 75 ◽  
pp. 109740
Author(s):  
Jiajie Guo ◽  
Ziyi Wang ◽  
Yao Weng ◽  
Haoze Yuan ◽  
Kaya Yoshida ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-14
Author(s):  
Liang Chen ◽  
Sun-Li Hu ◽  
Jun Xie ◽  
De-Yi Yan ◽  
She-Ji Weng ◽  
...  

The widespread use of therapeutic glucocorticoids has increased the frequency of glucocorticoid-induced osteoporosis (GIOP). One of the potential pathological processes of GIOP is an increased level of oxidative stress and mitochondrial dysfunction, which eventually leads to osteoblast apoptosis. Proanthocyanidins (PAC) are plant-derived antioxidants that have therapeutic potential against GIOP. In our study, a low dose of PAC was nontoxic to healthy osteoblasts and restored osteogenic function in dexamethasone- (Dex-) treated osteoblasts by suppressing oxidative stress, mitochondrial dysfunction, and apoptosis. Mechanistically, PAC neutralized Dex-induced damage in the osteoblasts by activating the Nrf2 pathway, since silencing Nrf2 partly eliminated the protective effects of PAC. Furthermore, PAC injection restored bone mass and promoted the expression of Nrf2 in the distal femur of Dex-treated osteoporotic rats. In summary, PAC protect osteoblasts against Dex-induced oxidative stress and mitochondrial dysfunction via the Nrf2 pathway activation and may be a promising drug for treating GIOP.


Sign in / Sign up

Export Citation Format

Share Document