-
Photobiomodulation therapy at 650 nm enhances osteogenic differentiation of osteoporotic bone marrow mesenchymal stem cells through modulating autophagy.
Photobiomodulatiom therapy (PBMT) has biostimulatory effects on bone marrow mesenchymal stem cells (BMSCs), which takes a pivotal role in maintaining bone mass and avoiding osteoporosis (OP). Autophagy is an important regulator for cell survival and homeostasis. Previous researchers found that BMSCs derived from osteoporotic rats (OP-BMSCs) were with the feature of reduced osteogenic differentiation and autophagy dysfunction. However, the potential regulation of PBMT in osteogenic differentiation of OP-BMSCs and its underling relationship with autophagy remain unclear.
650 nm red light-emitting diode (LED) was selected to initiate PBMT effects. The isolation and culture of OP-BMSCs were implemented after the establishment of the OP rat model. Firstly, the optimal dose of LED was screened on OP-BMSCs by CCK-8. Meanwhile, the osteogenic and mineralization activities were studied through the detection of Alkaline phosphatase (ALP) and alizarin red S (ARS). Then, the levels of osteogenesis and autophagy were investigated via western blot and immunofluorescence staining. Finally, the autophagy inhibitor 3-MA was applied to illustrate the underlying mechanism of the osteogenic effect of PBMT on OP-BMSCs.
Firstly, the optimal dose of 6 J/cm2 LED was selected in the subsequent experiments according to CCK-8. Then, the ALP activity and the mineralization ability of OP-BMSCs were obviously increased by PBMT. Meanwhile, Runx-2, OCN and OPN were significantly upregulated in LED group. Furthermore, the expressions of autophagic proteins increased significantly in LED group by immunofluorescence staining and western blot assay. At last, the promoted effects of PBMT on osteogenic differentiation in OP-BMSCs were distinctly reversed via inhibiting autophagy.
Our research illustrated that 650 nm LED could improve osteogenic differentiation of OP-BMSCs, suggesting a potential correlation between PBMT-mediated activation of autophagy and promotion of osteogenic differentiation.
Li H
,Yang W
,Zhu B
,Li M
,Zhang X
... -
《-》
-
Single-cell RNA transcriptomics reveals Du-Zhong-Wan promotes osteoporotic fracture healing via YAP/β-catenin/VEGF axis in BMSCs.
Our previous study demonstrated that Du-Zhong-Wan (DZW) promoted osteoporotic fracture (OPF) healing by enhancing osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and angiogenesis of endothelial cells (ECs). However, the heterogeneity of BMSCs and ECs, as well as the specific molecular mechanism underlying these effects, still require further evaluation.
The primary objective of this study was to elucidate the heterogeneity of BMSCs and ECs, as well as the cellular-level mechanism of DZW against OPF through single-cell RNA sequencing.
In this study, we presented a single-cell atlas of mouse femoral callus, comparing samples with and without DZW treatment, utilizing single-cell RNA sequencing. Variable genes were identified using the FindVariableGenes (FVG) and principal component analysis (PCA) analysis. Additionally, uniform manifold approximation and projection (U-MAP) was employed to reduce and visualize the distinct subclusters. The CellPhoneDB2 method was employed to analyze intercellular communication and quantify the interaction between ligands and receptors within distinct cell clusters. The osteogenic differentiation capacity of BMSCs was assessed by micro-CT, alkaline phosphatase (ALP), and alizarin red S (ARS) assay. The scratch wound assay and tube formation assay were utilized to assess the angiogenic capabilities of ECs in vitro. Additionally, western blot and immunofluorescence experiments were utilized to elucidate the related protein expression.
Consistent with our previous studies, DZW obviously promoted osteoporotic fracture healing. Moreover, this study discovered 14 cell clusters at the femoral fracture callus, where the BMSCs most actively interacted with ECs, through single-cell sequencing. Notably, DZW significantly elevated the proportion of Lepr+ BMSCs and Podxl+ ECs subgroup, which were respectively considered essential cells for osteoblastogenesis and angiogenesis of arteriolar vessels. The increased proportion of Podxl+ ECs was partially attributed to vascular endothelial growth factor (VEGF), secreted by BMSCs, which were able to be reversed by YAP pharmacological inhibitor verteporfin. Furthermore, the western blot assay revealed elevated expression levels of YAP/β-catenin, VEGF, RUNX2, and OCN in BMSCs treated with DZW, which were counteracted by verteporfin.
The data above indicates that DZW elevates the proportion of LEPR+ BMSCs and Podxl+ ECs, therefore contributing for the osteogenic ability of BMSCs and BMSCs-mediated angiogenesis via activation of the YAP/β-catenin/VEGF axis, which provides novel potential targets and mechanism for DZW in treating OPF in sub-clusters and molecular level.
Dong R
,Wei J
,Tian S
,Wang J
,Ma Y
,Li Y
,Liu RX
,Liu YQ
... -
《-》
-
Osteoking inhibits apoptosis of BMSCs in osteoporotic rats via PI3K/AKT signaling pathway.
In China, Osteoking is a commonly used treatment and preventive measure for osteoporosis. The pathophysiology of osteoporosis is closely associated with apoptosis; however, it remains unclear whether the role of Osteoking in promoting bone formation is linked to apoptosis.
This study aims to investigate whether Osteoking inhibits apoptosis of BMSCs in osteoporotic rats via the PI3K/AKT signaling pathway and to conduct a detailed exploration of this mechanism. The goal is to provide a theoretical basis for the clinical application of Osteoking in osteoporosis treatment.
A rat model of osteoporosis was established through bilateral ovariectomy (OVX), followed by treatment with Osteoking. After ten weeks of therapy, BMD was evaluated. The biomechanics of the left tibia were measured, the left femur was sequenced, and the right tibia was stained using histomorphometric and Masson's staining methods. Peripheral serum was collected to measure bone-related markers, including E2, PINP, and CTX. RNA-Seq results were verified using the remaining bone samples. Comparative analysis demonstrated the efficacy of Osteoking in treating osteoporosis and provided preliminary insights into the underlying mechanisms. Primary BMSCs were cultured using bone marrow apposition. CCK8 assays were conducted to screen the intervention conditions of Osteoking and LY294002. Various concentrations of Osteoking-containing serum and LY294002 were tested separately to determine the optimal intervention concentration for drug delivery. The impact of Osteoking on lipid formation was also evaluated. Following treatment of BMSCs from OVX rats with Sham serum, OVX serum, OVX + LY294002 serum, and Osteoking + LY294002 serum, the expression of PI3K/AKT/mTOR, osteogenesis-related regulatory factors, and apoptosis-related regulatory factors was assessed. Flow cytometry was employed to evaluate apoptosis in BMSCs.
Osteoking significantly improved whole-body BMD and bone biomechanical indices in OVX rats. It also significantly elevated the serum levels of E2 and PINP while reducing the level of CTX, which significantly improved bone microstructure and promoted new bone formation. RNA-seq analysis indicated that the therapeutic mechanism involved the PI3K/AKT signaling pathway. Osteoking increased the expression of RUNX2 and decreased the expression of PPAR-γ, a marker of lipogenesis, in OVX rats. Extraction of BMSCs for subsequent studies revealed a significant reduction in proliferation and osteogenic differentiation, along with an increase in lipogenic differentiation, in the OVX group. Osteoking treatment inhibited the expression of PPAR-γ and increased the expression of RUNX2 in BMSCs. Additionally, Osteoking reversed the LY294002-mediated inhibition of PI3K/AKT/mTOR signaling pathway activation, increased the expression of the apoptosis-protecting protein Bcl2, and decreased the expression of apoptosis-associated proteins Caspase3 and Bax.
Osteoking markedly improved bone microstructure, biomechanics, and bone density in OVX rats. Osteoking-containing serum reversed the imbalance in lineage differentiation in OVX rats, characterized by reduced osteogenic differentiation and increased lipid differentiation of BMSCs. Furthermore, Osteoking-containing serum significantly increased BMSC proliferation and prevented apoptosis in OVX rats through the PI3K/AKT signaling pathway.
Huang G
,Yin W
,Zhao X
,Xu M
,Wang P
,Li R
,Zhou L
,Tang W
,Jiao J
... -
《-》
-
Studies on the Role of MAP4K2, SPI1, and CTSD in Osteoporosis.
Osteoporosis (OP) is a prevalent skeletal disorder characterized by an imbalance between bone resorption and bone formation, resulting in a significant global burden. Previous research utilizing bioinformatics analysis has identified MAP4K2, SPI1, and CTSD as hub genes associated with OP. In this current investigation, we have successfully established a differential expression system of MAP4K2, SPI1, and CTSD in rat bone marrow mesenchymal stem cells (BMSCs) through transfection techniques. Additionally, the CCK-8 assay was employed to assess cell proliferation, while the alkaline phosphatase (ALP) activity assay and ALP staining assay were utilized to evaluate osteogenic differentiation. Alizarin red staining was employed to detect mineralization of BMSCs. Furthermore, the expression of relevant genes and molecules associated with the MAPK signaling pathway, autophagy, and apoptosis in the sera of rat BMSCs were examined using quantitative real-time polymerase chain reaction (qRT-PCR). The purpose of this study was to preliminarily investigate whether MAP4K2, SPI1, and CTSD have an effect on the osteogenic capacity of rat BMSCs and whether these genes, when differentially expressed, affect the expression of related genes in the MAPK, autophagy, and apoptosis signaling pathways and thus the osteogenic function of BMSCs. In summary, the findings of this study indicate that MAP4K2 and CTSD exert significant influence on the proliferation, osteogenic differentiation, and mineralization processes of rat BMSCs cells. Furthermore, these proteins may contribute to the development of OP through their involvement in the regulation of autophagy and apoptosis. Conversely, our investigation did not reveal any discernible impact of SPI1 on OP-related phenotypes. Consequently, this research serves as a fundamental basis for further exploration of potential therapeutic targets for the treatment of OP.
Sun C
,He W
,Wang L
,Hao T
,Yang X
,Feng W
,Wu Y
,Meng C
,Wang Z
,Chen X
,Sun M
,Zheng F
,Zhang B
... -
《-》
-
Naringin promotes osteogenic potential in bone marrow-derived mesenchymal stem cells via mediation of miR-26a/Ski axis.
Osteonecrosis of the femoral head (ONFH) is a common orthopedic disease, which seriously affects the quality of life of patients. Naringin has protective effect on ONFH. In present study, we aimed to investigate the mechanism of Naringin regulating miR-26a in ONFH.
Two sequencing datasets (GSE89587 for micro-RNA, GSE123568 for mRNA) related to ONFH were obtained from the GEO database for bioinformatics analysis. Bone marrow-derived mesenchymal stem cells (BMSCs) were treated with adipogenic medium (AM) or osteogenic medium (OM), and then treated with 10 μM, 100 μM or 1000 μM Naringin. Gene and protein levels were detected by RT-qPCR and Western blotting. ALP activity and alizarin red staining (ARS) were applied to investigate the osteogenic differentiation of BMSCs. Oil red O staining was performed to test adipogenic differentiation. The content of triglycerides (TG) in BMSCs was detected by TG detection kit. Double luciferase reporter gene measured the interaction between miR-26a and Ski.
Bioinfomatic analyses indicated a significant downregulation of miR-26a and a significant upregulation of Ski in the peripheral blood of patients with ONFH. Naringin significantly promoted the osteogenic differentiation, and increased the ALP activity and ARS. Meanwhile, it decreased the adipogenic differentiation and inhibited TG levels. In addition, miR-26a was downregulated and Ski was increased in AM-treated BMSCs, while miR-26a was upregulated and Ski was decreased in OM-treated BMSCs. Furthermore, miR-26a promoted the osteogenic differentiation and suppressed the adipogenic differentiation in BMSCs. Moreover, Naringin enhanced osteogenic potential in BMSCs was reversed by knockdown of miR-26a or overexpression of Ski.
Naringin could promote osteogenic differentiation of BMSCs via mediation of miR-26a/Ski axis. Thereby, Naringin might be a new agent for ONFH treatment.
Zou J
,Zhou L
,Liu G
,Zhang Y
,Zeng L
... -
《-》