1.Clematichinenoside AR protects bone marrow mesenchymal stem cells from hypoxia-induced apoptosis by maintaining mitochondrial homeostasis.
Zi-Tong ZHAO ; Peng-Cheng TU ; Xiao-Xian SUN ; Ya-Lan PAN ; Yang GUO ; Li-Ning WANG ; Yong MA
China Journal of Chinese Materia Medica 2025;50(5):1331-1339
This study aims to elucidate the role and mechanism of clematichinenoside AR(CAR) in protecting bone marrow mesenchymal stem cells(BMSCs) from hypoxia-induced apoptosis. BMSCs were isolated by the bone fragment method and identified by flow cytometry. Cells were cultured under normal conditions(37℃, 5% CO_2) and hypoxic conditions(37℃, 90% N_2, 5% CO_2) and treated with CAR. The BMSCs were classified into eight groups: control(normal conditions), CAR(normal conditions + CAR), hypoxia 24 h, hypoxia 24 h + CAR, hypoxia 48 h, hypoxia 48 h + CAR, hypoxia 72 h, and hypoxia 72 h + CAR. The cell counting kit-8(CCK-8) assay and terminal-deoxynucleoitidyl transferase mediated nick end labeling(TUNEL) were employed to measure cell proliferation and apoptosis, respectively. The number of mitochondria and mitochondrial membrane potential were measured by MitoTracker®Red CM-H2XRo staining and JC-1 staining, respectively. The level of reactive oxygen species(ROS) was measured with the DCFH-DA fluorescence probe. The protein levels of B-cell lymphoma-2 associated X protein(BAX), caspase-3, and optic atrophy 1(OPA1) were determined by Western blot. The results demonstrated that CAR significantly increased cell proliferation. Compared with the control group, the hypoxia groups showed increased apoptosis rates, reduced mitochondria, elevated ROS levels, decreased mitochondrial membrane potential, upregulated expression of BAX and caspase-3, and downregulated expression of OPA1. In comparison to the corresponding hypoxia groups, CAR intervention significantly decreased the apoptosis rate, increased mitochondria, reduced ROS levels, elevated mitochondrial membrane potential, downregulated the expression of BAX and caspase-3, and upregulated the expression of OPA1. Therefore, it can be concluded that CAR may exert an anti-apoptotic effect on BMSCs under hypoxic conditions by regulating OPA1 to maintain mitochondrial homeostasis.
Mesenchymal Stem Cells/metabolism*
;
Apoptosis/drug effects*
;
Mitochondria/metabolism*
;
Animals
;
Rats
;
Cell Hypoxia/drug effects*
;
Homeostasis/drug effects*
;
Reactive Oxygen Species/metabolism*
;
Rats, Sprague-Dawley
;
Membrane Potential, Mitochondrial/drug effects*
;
Saponins/pharmacology*
;
Caspase 3/genetics*
;
Male
;
bcl-2-Associated X Protein/genetics*
;
Bone Marrow Cells/metabolism*
;
Cell Proliferation/drug effects*
;
Protective Agents/pharmacology*
;
Cells, Cultured
2.Yougui Yin attenuates adipogenic differentiation of bone marrow mesenchymal stem cells by modulating PPARγ pathway to treat glucocorticoid-induced osteonecrosis.
Hong-Zhong XI ; Hao CHEN ; Shuai HE ; Wei SONG ; Jia-Hao FU ; Bin DU ; Xin LIU
China Journal of Chinese Materia Medica 2025;50(12):3356-3367
This study aims to investigate the pharmacological effects and mechanisms of Yougui Yin in treating glucocorticoid-induced osteonecrosis. A rat model of glucocorticoid-associated osteonecrosis of the femoral head(GA-ONFH) was established by intramuscular injection of dexamethasone at 20 mg·kg~(-1) every other day for 8 weeks. Rats were randomly allocated into control, model, and low-and high-dose(1.5 and 3.0 g·kg~(-1), respectively) Yougui Yin groups. After modeling, rats in Yougui Yin groups were administrated with Yougui Yin via gavage, which was followed by femoral specimen collection. Hematoxylin-eosin staining was employed to observe femoral head repair, and immunofluorescence was employed to assess adipogenic differentiation of bone marrow mesenchymal stem cells(BMSCs) within the femoral head. Cell experiments were carried out with dexamethasone(1 μmol·L~(-1))-treated BMSCs to evaluate the effects of Yougui Yin-medicated serum on adipogenic differentiation. Animal experiments demonstrated that compared with the model group, Yougui Yin at both high and low doses significantly improved bone mineral density(BMD), bone volume/total volume(BV/TV) ratio, and trabecular thickness(Tb.Th) in the femoral head. Additionally, Yougui Yin alleviated necrosis-like changes and adipocyte infiltration and significantly reduced the expression level of peroxisome proliferator-activated receptor γ(PPARγ) in the femoral head, thereby suppressing the adipogenic differentiation of BMSCs in GA-ONFH rats. The cell experiments revealed that Yougui Yin-medicated serum markedly inhibited dexamethasone-induced adipogenic differentiation of BMSCs and down-regulated the level of PPARγ. The overexpression of PPARγ attenuated the inhibitory effect of Yougui Yin-medicated serum on the adipogenic differentiation of BMSCs, indicating the critical role of PPARγ in Yougui Yin-mediated suppression of adipogenic differentiation of BMSCs. In conclusion, Yougui Yin exerts therapeutic effects on glucocorticoid-induced osteonecrosis by down-regulating PPARγ expression and inhibiting adipogenic differentiation of BMSCs.
Animals
;
Mesenchymal Stem Cells/metabolism*
;
PPAR gamma/genetics*
;
Rats
;
Drugs, Chinese Herbal/administration & dosage*
;
Male
;
Glucocorticoids/adverse effects*
;
Rats, Sprague-Dawley
;
Adipogenesis/drug effects*
;
Osteonecrosis/genetics*
;
Cell Differentiation/drug effects*
;
Bone Marrow Cells/metabolism*
;
Femur Head Necrosis/chemically induced*
;
Humans
3.Mechanism of sodium valproate in inhibiting ferroptosis of bone marrow mesenchymal stem cells via the adenosine monophosphate-activated protein kinase/Sirtuin 1 axis.
Qingsong GU ; Jianqiao LI ; Yuhu CHEN ; Linhui WANG ; Yiheng LI ; Ziru WANG ; Yicong WANG ; Min YANG
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(2):215-223
OBJECTIVE:
To investigate the effects of sodium valproate (VPA) in inhibiting Erastin-induced ferroptosis in bone marrow mesenchymal stem cells (BMSCs) and its underlying mechanisms.
METHODS:
BMSCs were isolated from bone marrow of 8-week-old Spragur Dawley rats and identified [cell surface antigens CD90, CD44, and CD45 were analyzed by flow cytometry, and osteogenic and adipogenic differentiation abilities were assessed by alizarin red S (ARS) and oil red O staining, respectively]. Cells of passage 3 were used for the Erastin-induced ferroptosis model, with different concentrations of VPA for intervention. The optimal drug concentration was determined using the cell counting kit 8 assay. The experiment was divided into 4 groups: group A, cells were cultured in osteogenic induction medium for 24 hours; group B, cells were cultured in osteogenic induction medium containing optimal concentration Erastin for 24 hours; group C, cells were cultured in osteogenic induction medium containing optimal concentration Erastin and VPA for 24 hours; group D, cells were cultured in osteogenic induction medium containing optimal concentration Erastin and VPA, and 8 μmol/L EX527 for 24 hours. The mitochondrial state of the cells was evaluated, including the levels of malondialdehyde (MDA), glutathione (GSH), and reactive oxygen species (ROS). Osteogenic capacity was assessed by alkaline phosphatase (ALP) activity and ARS staining. Western blot analysis was performed to detect the expressions of osteogenic-related proteins [Runt-related transcription factor 2 (RUNX2) and osteopontin (OPN)], ferroptosis-related proteins [glutathione peroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1), and solute carrier family 7 member 11 (SLC7A11)], and pathway-related proteins [adenosine monophosphate-activated protein kinase (AMPK) and Sirtuin 1 (SIRT1)].
RESULTS:
The cultured cells were identified as BMSCs. VPA inhibited Erastin-induced ferroptosis and the decline of osteogenic ability in BMSCs, acting through the activation of the AMPK/SIRT1 pathway. VPA significantly reduced the levels of ROS and MDA in Erastin-treated BMSCs and significantly increased GSH levels. Additionally, the expression levels of ferroptosis-related proteins (GPX4, FTH1, and SLC7A11) significantly decreased. VPA also upregulated the expressions of osteogenic-related proteins (RUNX2 and OPN), enhanced mineralization and osteogenic differentiation, and increased the expressions of pathway-related proteins (AMPK and SIRT1). These effects could be reversed by the SIRT1 inhibitor EX527.
CONCLUSION
VPA inhibits ferroptosis in BMSCs through the AMPK/SIRT1 axis and promotes osteogenesis.
Mesenchymal Stem Cells/metabolism*
;
Ferroptosis/drug effects*
;
Animals
;
Valproic Acid/pharmacology*
;
Rats
;
Rats, Sprague-Dawley
;
Sirtuin 1/metabolism*
;
Cell Differentiation/drug effects*
;
Cells, Cultured
;
AMP-Activated Protein Kinases/metabolism*
;
Osteogenesis/drug effects*
;
Piperazines/pharmacology*
;
Bone Marrow Cells/cytology*
;
Reactive Oxygen Species/metabolism*
;
Signal Transduction/drug effects*
4.Preparation of calcium phosphate nanoflowers and evaluation of their antioxidant and osteogenic induction capabilities in vitro.
Mingyu JIA ; Zhihong CHEN ; Huajian ZHOU ; Yukang ZHANG ; Min WU
Chinese Journal of Reparative and Reconstructive Surgery 2025;39(9):1203-1211
OBJECTIVE:
To investigate the antioxidant and osteogenic induction capabilities of calcium phosphate nanoflowers (hereinafter referred to as nanoflowers) in vitro at different concentrations.
METHODS:
Nanoflowers were prepared using gelatin, tripolyphosphate, and calcium chloride. Their morphology, microstructure, elemental composition and distribution, diameter, and molecular constitution were characterized using scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, and energy-dispersive spectroscopy. Femurs and tibias were harvested from twelve 4-week-old Sprague Dawley rats, and bone marrow mesenchymal stem cells (BMSCs) were isolated and cultured using the whole bone marrow adherent method, followed by passaging. The third passage cells were identified as stem cells by flow cytometry and then co-cultured with nanoflowers at concentrations of 0, 0.4, 0.8, 1.2, 1.6, 2.0, 2.4, 2.8, 3.2, and 3.6 mg/mL. Cell counting kit 8 (CCK-8) assay was performed to screen for the optimal concentration that demonstrated the best cell viability, which was subsequently used as the experimental concentration for further studies. After co-culturing BMSCs with the screened concentration of nanoflowers, the biocompatibility of the nanoflowers was verified through live/dead cell staining, scratch assay, and cytoskeleton staining. The antioxidant capacity was assessed by using reactive oxygen species (ROS) fluorescence staining. The in vitro osteoinductive ability was evaluated via alkaline phosphatase (ALP) staining, alizarin red staining, and immunofluorescence staining of osteocalcin (OCN) and Runt-related transcription factor 2 (RUNX2). All the above indicators were compared with the control group of normally cultured BMSCs without the addition of nanoflowers.
RESULTS:
Scanning electron microscopy revealed that the prepared nanoflowers exhibited a flower-like structure; transmission electron microscopy scans discovered that the nanoflowers possessed a multi-layered structure, and high-magnification images displayed continuous atomic arrangements, with the nanoflower diameter measuring (2.00±0.25) μm; energy-dispersive spectroscopy indicated that the nanoflowers contained elements such as C, N, O, P, and Ca, which were uniformly distributed across the flower region; Fourier transform infrared spectroscopy analyzed the absorption peaks of each component, demonstrating the successful preparation of the nanoflowers. Through CCK-8 screening, the concentrations of 0.8, 1.2, and 1.6 mg/mL were selected for subsequent experiments. The live/dead cell staining showed that nanoflowers at different concentrations exhibited good cell compatibility, with the 1.2 mg/mL concentration being the best (P<0.05). The scratch assay results indicated that the cell migration ability in the 1.2 mg/mL group was superior to the other groups (P<0.05). The cytoskeleton staining revealed that the cell morphology was well-extended in all concentration groups, with no significant difference compared to the control group. The ROS fluorescence staining demonstrated that the ROS fluorescence in all concentration groups decreased compared to the control group after lipopolysaccharide induction (P<0.05), with the 1.2 mg/mL group showing the weakest fluorescence. The ALP staining showed blue-purple nodular deposits around the cells in all groups, with the 1.2 mg/mL group being significantly more prominent. The alizarin red staining displayed orange-red mineralized nodules around the cells in all groups, with the 1.2 mg/mL group having more and denser nodules. The immunofluorescence staining revealed that the expressions of RUNX2 and OCN proteins in all concentration groups increased compared to the control group, with the 1.2 mg/mL group showing the strongest protein expression (P<0.05).
CONCLUSION
The study successfully prepares nanoflowers, among which the 1.2 mg/mL nanoflowers exhibits excellent cell compatibility, antioxidant properties, and osteogenic induction capability, demonstrating their potential as an artificial bone substitute material.
Animals
;
Osteogenesis/drug effects*
;
Mesenchymal Stem Cells/drug effects*
;
Calcium Phosphates/pharmacology*
;
Rats, Sprague-Dawley
;
Rats
;
Antioxidants/chemistry*
;
Cells, Cultured
;
Cell Differentiation/drug effects*
;
Nanostructures/chemistry*
;
Tissue Engineering/methods*
;
Bone Marrow Cells/cytology*
;
Coculture Techniques
;
Tissue Scaffolds/chemistry*
;
Male
;
Biocompatible Materials/chemistry*
;
Cell Survival
;
Core Binding Factor Alpha 1 Subunit/metabolism*
;
Cell Proliferation
5.The effect of gentiopicroside on osteogenic differentiation of human bone marrow mesenchymal stem cells by regulating the SDF-1/CXCR4 signaling pathway.
Ruifang WANG ; Yingchun YANG ; Haibing QIAO ; Ying YANG
Chinese Journal of Cellular and Molecular Immunology 2025;41(9):784-789
Objective To investigate the effect of gentiopicroside on osteogenic differentiation of human bone marrow mesenchymal stem cells (BMSCs), and to determine whether its mechanism involves the stromal cell-derived factor 1(SDF-1)/C-X-C chemokine receptor 4 (CXCR4) pathway. Methods BMSCs were divided into six groups: normal culture control group, osteogenic induction model group, low-dose gentiopicroside (L-gentiopicroside, 10 μmol/L) group, medium-dose gentiopicroside (M-gentiopicroside, 20 μmol/L) group, high-dose gentiopicroside (H-gentiopicroside, 40 μmol/L) group, and H-gentiopicroside+SDF-1/CXCR4 pathway inhibitor (AMD3100) group (H-gentiopicroside+AMD3100, 40 μmol/L gentiopicroside+10 μg/mL AMD3100). Cell viability, apoptosis, ALP activity, mineralized nodule formation, and protein levels of the SDF-1/CXCR4 pathway were assessed using the CCK-8 assay, flow cytometry, ALP staining, Alizarin Red S staining, and Western blotting, respectively. Results No mineralized nodules were observed in either the control and model group, although the color of the model group deepened. Compared with the control group, the model group showed significantly increased A value, ALP activity, expression levels of Runt related transcription factor 2 (RUNX2), osteopontin (OPN), SDF-1, CXCR4 proteins, along with a lower apoptosis rate. Compared with the model group, the L-gentiopicroside, M-gentiopicroside and H-gentiopicroside groups showed dose-dependently (L
6.Establishment and Application of an in Vitro Cellular Model of Adipogenic Differentiation of Bone Marrow Mesenchymal Stem Cells with Serum Injury in aGVHD Mouse.
Run-Xiang XU ; Pei-Lin LI ; Jia-Yi TIAN ; Jie TANG ; Bo-Feng YIN ; Fu-Hao YU ; Fei-Yan WANG ; Xiao-Tong LI ; Xiao-Yu ZHANG ; Wen-Rong XIA ; Heng ZHU ; Li DING
Journal of Experimental Hematology 2025;33(1):255-261
OBJECTIVE:
To establish an in vitro cell model simulating acute graft-versus-host disease (aGVHD) bone marrow microenvironment injury with the advantage of mouse serum of aGVHD model and explore the effect of serum of aGVHD mouse on the adipogenic differentiation ability of mesenchymal stem cells (MSCs).
METHODS:
The 6-8-week-old C57BL/6N female mice and BALB/c female mice were used as the donor and recipient mice of the aGVHD model, respectively. Bone marrow transplantation (BMT) mouse model (n=20) was established by being injected with bone marrow cells (1×107 per mouse) from donor mice within 4-6 hours after receiving a lethal dose (8.0 Gy, 72.76 cGy/min) of γ ray general irradiation. A mouse model of aGVHD (n=20) was established by infusing a total of 0.4 ml of a mixture of donor mouse-derived bone marrow cells (1×107 per mouse) and spleen lymphocytes (2×106 per mouse). The blood was removed from the eyeballs and the mouse serum was aspirated on the 7th day after modeling. Bone marrow-derived MSCs were isolated from 1-week-old C57BL/6N male mice and incubated with 2%, 5% and 10% BMT mouse serum and aGVHD mouse serum in the medium, respectively. The effect of serum in the two groups on the in vitro adipogenic differentiation ability of mouse MSCs was detected by Oil Red O staining. The expression levels of related proteins PPARγ and CEBPα were detected by Western blot. The expression differences of key adipogenic transcription factors including PPARγ, CEBPα, FABP4 and LPL were determined by real-time quantitative PCR (RT-qPCR).
RESULTS:
An in vitro cell model simulating the damage of bone marrow microenvironment in mice with aGVHD was successfully established. Oil Red O staining showed that the number of orange-red fatty droplets was significantly reduced and the adipogenic differentiation ability of MSC was impaired at aGVHD serum concentration of 10% compared with BMT serum. Western blot experiments showed that adipogenesis-related proteins PPARγ and CEBPα expressed in MSCs were down-regulated. Further RT-qPCR assay showed that the production of PPARγ, CEBPα, FABP4 and LPL, the key transcription factors for adipogenic differentiation of MSC, were significantly reduced.
CONCLUSION
The adipogenic differentiation capacity of MSCs is inhibited by aGVHD mouse serum.
Animals
;
Mesenchymal Stem Cells/cytology*
;
Mice
;
Mice, Inbred BALB C
;
Mice, Inbred C57BL
;
Adipogenesis
;
Female
;
Cell Differentiation
;
Graft vs Host Disease/blood*
;
Bone Marrow Cells/cytology*
;
PPAR gamma/metabolism*
;
Disease Models, Animal
;
CCAAT-Enhancer-Binding Protein-alpha/metabolism*
7.Knockdown of NPTX1 promotes osteogenic differentiation of human bone marrow mesenchymal stem cells.
Ting SHUAI ; Yanyan GUO ; Chunping LIN ; Xiaomei HOU ; Chanyuan JIN
Journal of Peking University(Health Sciences) 2025;57(1):7-12
OBJECTIVE:
To initially investigate the function of neuronal pentraxin 1 (NPTX1) gene on osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs).
METHODS:
hBMSCs were induced to undergo osteogenic differentiation, and then RNA was collected at different time points, namely 0, 3, 7, 10 and 14 d. The mRNA expression levels of key genes related with osteogenic differentiation, including runt-related transcription factor 2 (RUNX2), alkaline phosphatase (ALP), osteocalcin (OCN), and NPTX1, were detected on the basis of quantitative real-time polymerase chain reaction (qPCR) technology. In order to establish a stable NPTX1-knockdown hBMSCs cell line, NPTX1 shRNA lentivirus was constructed and used to infect hBMSCs. ALP staining, alizarin red (AR) staining, and qPCR were employed to assess the impact of NPTX1-knockdown on the osteogenic differentiation ability of hBMSCs.
RESULTS:
The results showed that during the osteogenic differentiation of hBMSCs in vitro, the mRNA expression levels of osteogenic genes RUNX2, ALP and OCN significantly increased compared with 0 d, while NPTX1 expression decreased markedly (P < 0.01) as the osteogenic induction period exten-ded. At 72 h post-infection with lentivirus, the result of qPCR indicated that the knockdown efficiency of NPTX1 was over 60%. After knocking down NPTX1 in hBMSCs, RNA was extracted from both the NPTX1-knockdown group (sh NPTX1 group) and the control group (shNC group) cultured in regular proliferation medium. The results of qPCR showed that the expression levels of osteogenic-related genes RUNX2 and osterix (OSX) were significantly higher in the sh NPTX1 group compared with the shNC group (P < 0.01). ALP staining revealed a significantly deeper coloration in the sh NPTX1 group than in the shNC group at the end of 7 d of osteogenic induction. AR staining demonstrated a marked increase in mineralized nodules in the sh NPTX1 group compared with the shNC group at the end of 14 d of osteogenic induction.
CONCLUSION
NPTX1 exerts a modulatory role in the osteogenic differentiation of hBMSCs, and its knockdown has been found to enhance the osteogenic differentiation of hBMSCs. This finding implies that NPTX1 could potentially serve as a therapeutic target for the treatment of osteogenic abnormalities, including osteoporosis.
Humans
;
Mesenchymal Stem Cells/cytology*
;
Osteogenesis/genetics*
;
Cell Differentiation/genetics*
;
Nerve Tissue Proteins/genetics*
;
Cells, Cultured
;
C-Reactive Protein/genetics*
;
RNA, Small Interfering/genetics*
;
Core Binding Factor Alpha 1 Subunit/metabolism*
;
Bone Marrow Cells/cytology*
;
Gene Knockdown Techniques
;
Osteocalcin/metabolism*
;
Alkaline Phosphatase/metabolism*
;
RNA, Messenger/metabolism*
8.Impact of lithocholic acid on the osteogenic and adipogenic differentiation balance of bone marrow mesenchymal stem cells.
Cui WANG ; Jiao LI ; Lingyun LU ; Lu LIU ; Xijie YU
Chinese Journal of Reparative and Reconstructive Surgery 2024;38(1):82-90
OBJECTIVE:
To Investigate the effects of lithocholic acid (LCA) on the balance between osteogenic and adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs).
METHODS:
Twelve 10-week-old SPF C57BL/6J female mice were randomly divided into an experimental group (undergoing bilateral ovariectomy) and a control group (only removing the same volume of adipose tissue around the ovaries), with 6 mice in each group. The body mass was measured every week after operation. After 4 weeks post-surgery, the weight of mouse uterus was measured, femur specimens of the mice were taken for micro-CT scanning and three-dimensional reconstruction to analyze changes in bone mass. Tibia specimens were taken for HE staining to calculate the number and area of bone marrow adipocytes in the marrow cavity area. ELISA was used to detect the expression of bone turnover markers in the serum. Liver samples were subjected to real-time fluorescence quantitative PCR (RT-qPCR) to detect the expression of key genes related to bile acid metabolism, including cyp7a1, cyp7b1, cyp8b1, and cyp27a1. BMSCs were isolated by centrifugation from 2 C57BL/6J female mice (10-week-old). The third-generation cells were exposed to 0, 1, 10, and 100 μmol/L LCA, following which cell viability was evaluated using the cell counting kit 8 assay. Subsequently, alkaline phosphatase (ALP) staining and oil red O staining were conducted after 7 days of osteogenic and adipogenic induction. RT-qPCR was employed to analyze the expressions of osteogenic-related genes, namely ALP, Runt-related transcription factor 2 (Runx2), and osteocalcin (OCN), as well as adipogenic-related genes including Adiponectin (Adipoq), fatty acid binding protein 4 (FABP4), and peroxisome proliferator-activated receptor γ (PPARγ).
RESULTS:
Compared with the control group, the body mass of the mice in the experimental group increased, the uterus atrophied, the bone mass decreased, the bone marrow fat expanded, and the bone metabolism showed a high bone turnover state. RT-qPCR showed that the expressions of cyp7a1, cyp8b1, and cyp27a1, which were related to the key enzymes of bile acid metabolism in the liver, decreased significantly ( P<0.05), while the expression of cyp7b1 had no significant difference ( P>0.05). Intervention with LCA at concentrations of 1, 10, and 100 μmol/L did not demonstrate any apparent toxic effects on BMSCs. Furthermore, LCA inhibited the expressions of osteogenic-related genes (ALP, Runx2, and OCN) in a dose-dependent manner, resulting in a reduction in ALP staining positive area. Concurrently, LCA promoted the expressions of adipogenic-related genes (Adipoq, FABP4, and PPARγ), and an increase in oil red O staining positive area.
CONCLUSION
After menopause, the metabolism of bile acids is altered, and secondary bile acid LCA interferes with the balance of osteogenic and adipogenic differentiation of BMSCs, thereby affecting bone remodelling.
Female
;
Mice
;
Animals
;
Core Binding Factor Alpha 1 Subunit/pharmacology*
;
PPAR gamma/metabolism*
;
Steroid 12-alpha-Hydroxylase/metabolism*
;
Mice, Inbred C57BL
;
Cell Differentiation
;
Osteogenesis
;
Mesenchymal Stem Cells
;
Bile Acids and Salts/pharmacology*
;
Bone Marrow Cells
;
Cells, Cultured
;
Azo Compounds
9.The impact of mitochondrial transfer on leukemia progression.
Wen-Jia FANG ; Biao ZHANG ; Tao CHENG ; Hui CHENG
Acta Physiologica Sinica 2024;76(6):943-952
The objective of the present study was to investigate the role and mechanism of bone marrow microenvironmental cells in regulating the mitochondrial mass of leukemia cells, and to uncover the mechanism of leukemia progression at the metabolic level. A mouse model of acute myeloid leukemia (AML) induced by the overexpression of the MLL-AF9 (MA9) fusion protein was established, and the bone marrow cells of AML mice were transplanted into mitochondrial fluorescence reporter mice expressing the Dendra2 protein (mito-Dendra2 mice). The proportion of Dendra2+ cells in bone marrow leukemia cells at different stages of AML was quantified by flow cytometry. The effects of transferred mitochondria on leukemia cells were studied by fluorescence-activated cell sorting (FACS), followed by functional experiments and bulk RNA sequencing. Finally, components within the bone marrow niche, such as mesenchymal stromal cells (MSCs) and endothelial cells (ECs), were co-cultured with leukemia cells in vitro. The proportion of leukemia cells that underwent mitochondrial transfer and the apoptosis level of leukemia cells were then detected by flow cytometry. The results showed that mitochondria from bone marrow cells were transferred to leukemia cells in the AML mouse model, and the proportion of mitochondrial transfer decreased with AML progression. The proportion of mitochondria transferred to leukemia stem cells (LSCs) was lower than that of mature AML cells. In AML cells receiving Dendra2+ mitochondria, there was a significant increase in the levels of intracellular reactive oxygen species (ROS) and apoptosis, while the levels of protein translation and their colony-forming capacities were decreased. The transplantation of Dendra2+ AML cells resulted in an extension of the survival of mice. RNA sequencing analysis demonstrated a significant downregulation of pathways related to translation, aerobic respiration and mitochondrial organization in AML cells that had received mitochondria. In vitro co-culture experiments indicated that MSCs within the bone marrow niche tended to transfer their mitochondria to leukemia cells and promoted the apoptosis of leukemia cells. These results indicate that in the MA9-induced AML mouse model, bone marrow niche cells can transfer mitochondria to leukemia cells, resulting in a reduction in the overall survival and function of the leukemia cells. Mitochondrial transfer in the bone marrow microenvironment may serve as a self-defensive mechanism of the host bone marrow niche cells, inhibiting the progression of AML.
Animals
;
Mice
;
Mitochondria/metabolism*
;
Disease Progression
;
Leukemia, Myeloid, Acute/genetics*
;
Apoptosis
;
Mesenchymal Stem Cells/metabolism*
;
Bone Marrow Cells/metabolism*
;
Humans
;
Tumor Microenvironment
;
Mice, Inbred C57BL
10.Role of R-spondin 2 on osteogenic differentiation of bone marrow mesenchymal stem cells and bone metabolism in ovariectomized mice.
Xin LIU ; Bowen SHI ; Chengkuo CAI ; Haotian WANG ; Peng JIA
Chinese Journal of Reparative and Reconstructive Surgery 2024;38(11):1399-1407
OBJECTIVE:
To investigate the effects of R-spondin 2 (Rspo2) on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and bone mineral content in ovariectomized mice.
METHODS:
BMSCs were extracted from the bone marrow of the long bones of 7 4-week-old female C57BL/6 mice using whole bone marrow culture and passaged. After the cell phenotype was identified by flow cytometry, the 3rd generation cells were co-cultured with 10, 20, 40, 80, and 100 nmol/L Rspo2. Then, the cell activity and proliferative capacity were determined by cell counting kit 8 (CCK-8), and the intervention concentration of Rspo2 was screened for the subsequent experiments. The osteogenic differentiation ability of BMSCs was detected by alkaline phosphatase (ALP) staining, and the mRNA levels of osteogenesis-related genes [RUNX family transcription factor 2 (Runx2), collagen type Ⅰ alpha 1 (Col1), osteocalcin (OCN)] were detected by real-time fluorescence quantitative PCR (RT-qPCR). In addition, 18 10-week-old female C57BL/6 mice were randomly divided into sham operation group (sham group), ovariectomy group (OVX group), and OVX+Rspo2-intervention group (OVX+Rspo2 group), with 6 mice in each group. The sham group only underwent bilateral back incision and suturing, while the other two groups established osteoporosis mouse models by bilateral ovarian castration. Then, the mice were given a weekly intraperitoneal Rspo2 (1 mg/kg) treatment in OVX+Rspo2 group and saline at the same dosage in sham group and OVX group. After 12 weeks of treatment, the body mass and uterus mass of the mice were weighed in the 3 groups to assess whether the OVX model was successfully prepared; the tibia bones were stained with HE and immunohistochemistry staining to observe the changes in tibial bone mass and the expression level of Runx2 protein in the bone tissues. Blood was collected to detect the expressions of bone metabolism markers [ALP, OCN, type Ⅰ procollagen amino-terminal peptide (PINP)] and bone resorption marker [β-collagen degradation product (β-CTX)] by ELISA assay. Micro-CT was used to detect the bone microstructure changes in the tibia, and three-dimensional histomorphometric analyses were performed to analyze the trabeculae thickness (Tb.Th), trabeculae number (Tb.N), trabeculae separation (Tb.Sp), and bone volume fraction (BV/TV).
RESULTS:
CCK-8 assay showed that Rspo2 concentrations below 80 nmol/L were not cytotoxic ( P>0.05), and the cell viability of 20 nmol/L Rspo2 group was significantly higher than that of the control group ( P<0.05). Based on the above results, 10, 20, and 40 nmol/L Rspo2 were selected for subsequent experiments. ALP staining showed that the positive cell area of each concentration of Rspo2 group was significantly larger than that of the control group ( P<0.05), with the highest showed in the 20 nmol/L Rspo2 group. The expression levels of the osteogenesis-related genes (Runx2, Col1, OCN) significantly increased, and the differences were significant between Rspo2 groups and control group ( P<0.05) except for Runx2 in the 40 nmol/L Rspo2 group. In animal experiments, all groups of mice survived until the completion of the experiment, and the results of the body mass and uterus mass after 12 weeks of treatment showed that the OVX model was successfully prepared. Histological and immunohistochemical staining showed that the sparseness and connectivity of bone trabecula and the expression of Runx2 in the OVX group were lower than those in the sham group, whereas they were reversed in the OVX+Rspo2 group after treatment with Rspo2, and the differences were significant ( P<0.05). ELISA assay showed that compared with the sham group, the serum bone metabolism markers in OVX group had an increase in ALP and a decrease in PINP ( P<0.05). After Rspo2 intervention, PINP expression significantly reversed and increased, with significant differences compared to the sham group and OVX group ( P<0.05). The bone resorption marker (β-CTX) was significantly higher in the OVX group than in the sham group ( P<0.05), and it was significantly decreased in the OVX+Rspo2 group when compared with the OVX group ( P<0.05). Compared with the sham group, Tb.Th, Tb.N, and BV/TV significantly decreased in the OVX group, while Tb.Sp significantly increased ( P<0.05); after Rspo2 intervention, all of the above indexes significantly improved in the OVX+Rspo2 group ( P<0.05) except Tb.Th.
CONCLUSION
Rspo2 promotes differentiation of BMSCs to osteoblasts, ameliorates osteoporosis due to estrogen deficiency, and promotes bone formation in mice.
Animals
;
Female
;
Ovariectomy
;
Mice
;
Cell Differentiation
;
Osteogenesis
;
Mice, Inbred C57BL
;
Mesenchymal Stem Cells/cytology*
;
Cells, Cultured
;
Thrombospondins/metabolism*
;
Bone Marrow Cells/metabolism*
;
Bone Density
;
Cell Proliferation
;
Intercellular Signaling Peptides and Proteins/metabolism*
;
Coculture Techniques

Result Analysis
Print
Save
E-mail