scholarly journals Ligustrum Japonicum fructus Induces Anti-adipogenetic and Pro-osteoblastogenic Activities in Human Bone Marrow-derived Mensencymal Stem Cells (P06-016-19)

2019 ◽  
Vol 3 (Supplement_1) ◽  
Author(s):  
JungHwan Oh ◽  
Fatih Karadeniz ◽  
JungIm Lee ◽  
Youngwan Seo ◽  
Chang-Suk Kong

Abstract Objectives Masenchymal stem cells (MSCs) have pluripotent differentiation properties that confirmed to differentiate into myocyte, adipocyte, osteoblast, neuron and chondrocyte when specific culture conditions and stimuli are applied. In bone, both adipocytes and osteoblasts are derived from bone marrow MSCs (BMSCs) and production of these cells has been reported as reciprocal processes. The bone mass disequilibrium causes osteoporosis, as a result of elevated adipogenic differentiation accompanied by reducing bone formation. Therefore, in this study the effect of Ligustrum japonicum fructus (Waxleaf privet) on the adipogenesis and osteoblast differentiation was investigated in BMSCs. Methods The fruits of L. japonicum were extracted with dichloromethane and methanol, and the combined extracts were concentrated. Differentiation of BMSCs was performed by changing the medium into adipocyte and osteoblast differentiation supplied by Promocell GmbH. The cellular lipid was stained with Oil Red O and the alkaline phosphatase (ALP) activity was measured using a colormetric assay kit (Biovision, Inc.). The relative protein levels were measured by immunoblotting assay. Results Presense of L. japonicum fructus extract (LJE) inhibited the cellular lipid accumulation in a dose-dependent manner. Consistent with the effects on lipid accumulation, the adipocyte specific genes including PPARγ, C/EBPα and SREBP1c was down regulated by treatment with LJE. Moreover, treatment with LJE enhanced osteoblast differentiation observed as increased ALP activity and upregulated the proteALP, BMP-2 and osteocalcine protein levels. Conclusions The results indicated that LJE may prevent bone loss by inhibiting adipogenesis while activating the osteogenic differentiation in BMSCs. Therefore, LJE possess the potential to be utilized as a source of nutraceutical agents against osteoporosis. Funding Sources This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIP).

2015 ◽  
Vol 4 (1) ◽  
pp. 69-73
Author(s):  
Thomas J. Rhodes ◽  
Michelle Pazienza ◽  
Jodi F. Evans

ACTH is a major hormone of the stress axis or hypothalamic-pituitary-adrenal (HPA) axis.  It is derived from pro-opiomelanocortin (POMC) the precursor to the melanocortin family of peptides. POMC produces the biologically active melanocortin peptides via a series of enzymatic steps in a tissue-specific manner, yielding the melanocyte-stimulating hormones (MSHs), corticotrophin (ACTH) and ?-endorphin. The melanocortin system plays an imperative role in energy expenditure, insulin release and insulin sensitivity.  Bone marrow derived mesenchymal stem cells circulate in the blood stream and as progenitor cells have the potential to differentiate into many cell types such as osteoblasts, chondrocytes and adipocytes. Here we examine the effects of ACTH on the mouse D1 bone-marrow derived MSC.  ACTH significantly increased lipid accumulation during the adipogenic differentiation of D1 cells in a concentration- dependent manner. ACTH also shifts the temporal pattern of D1 adipogenic differentiation to the left i.e. differentiation occurs earlier with ACTH treatment. No significant differences in protein expression of peroxisome proliferator-activated receptor gamma (PPAR-?2), a regulating transcription factor of adipogenesis were found.  Therefore the effects of ACTH are suggested to be mediated by an alternative pathway. Overall the results indicate a connection between increased adipose deposition and the elevated circulating ACTH associated with stress.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Qiting He ◽  
Ruixi Qin ◽  
Julie Glowacki ◽  
Shuanhu Zhou ◽  
Jie Shi ◽  
...  

Abstract Background Vitamin D is important for the mineralization of bones by stimulating osteoblast differentiation of bone marrow mesenchymal stem cells (BMMSCs). BMMSCs are a target of vitamin D action, and the metabolism of 25(OH)D3 to biologically active 1α,25(OH)2D3 in BMMSCs promotes osteoblastogenesis in an autocrine/paracrine manner. Our previous study with human BMMSCs showed that megalin is required for the 25(OH)D3-DBP complex to enter cells and for 25(OH)D3 to stimulate osteoblast differentiation in BMMSCs. Furthermore, we reported that leptin up-regulates megalin in those cells. Leptin is a known inhibitor of PI3K/AKT-dependent chaperone-mediated autophagy (CMA). In this study, we tested the hypothesis that leptin acts synergistically with 25(OH)D3 to promote osteoblastogenesis in rat BMMSCs by a mechanism that entails inhibition of PI3K/AKT-dependent CMA. Methods BMMSCs were isolated from rat bone marrow (4-week-old male SD rats); qRT-PCR and western immunoblots or immunofluorescence were used to evaluate the expression of megalin, ALP, COL1A1, RUNX2, OSX, OSP, and CMA in rBMMSCs. The osteoblast differentiation was evaluated by ALP activity, ALP staining, and calcium deposition. The viability of rBMMSCs was assessed with the CCK-8 kit. Biosynthesis of 1α,25(OH)2D3 was measured by a Rat 1α,25(OH)2D3 ELISA Kit. Results The combination of leptin and 25(OH)D3 treatment significantly enhanced osteoblast differentiation as shown by ALP activity, ALP staining, and calcium deposition, the expression of osteogenic genes ALP, COL1A1, RUNX2, OSX, and OSP by qRT-PCR and western immunoblots in rBMMSCs. Leptin enhanced the expression of megalin and synthesis of 1α,25(OH)2D3 in rBMMSCs. Our data showed that leptin inhibited CMA activity of rBMMSCs by activating PI3K/AKT signal pathway; the ability of leptin to enhance 25(OH)D3 promoted osteoblast differentiation of rBMMSCs was weakened by the PI3K/AKT signal pathway inhibitor. Conclusions Our data reveal the mechanism by which leptin and 25(OH)D3 promote osteoblast differentiation in rBMMSCs. Leptin promoted the expression of megalin by inhibiting CMA, increased the utilization of 25(OH)D3 by rBMMSCs, and enhanced the ability of 25(OH)D3 to induce osteoblast differentiation of rBMMSCs. PI3K/AKT is at least partially involved in the regulation of CMA. These data indicate the importance of megalin in BMMSCs for vitamin D’s role in skeletal health.


2021 ◽  
Vol 2021 ◽  
pp. 1-8
Author(s):  
Meng Yang ◽  
Xin Yan ◽  
Fu-Zhen Yuan ◽  
Jing Ye ◽  
Ming-Ze Du ◽  
...  

Cartilage injury of the knee joint is very common. Due to the limited self-healing ability of articular cartilage, osteoarthritis is very likely to occur if left untreated. Bone marrow mesenchymal stem cells (BMMSCs) are widely used in the study of cartilage injury due to their low immunity and good amplification ability, but they still have disadvantages, such as heterogeneous undifferentiated cells. MicroRNAs can regulate the chondrogenic differentiation ability of MSCs by inhibiting or promoting mRNA translation and degradation. In this research, we primarily investigated the effect of microRNA-210-3p (miR-210-3p) on chondrogenic and adipogenic differentiation of BMMSCs in vitro. Our results demonstrate that miR-210-3p promoted chondrogenic differentiation and inhibited adipogenic differentiation of rat BMMSCs, which was related to the HIF-3α signalling pathway. Additionally, miR-210-3p promotes mRNA and protein levels of the chondrogenic expression genes COLII and SOX9 and inhibits mRNA and protein levels of the adipogenic expression genes PPARγ and LPL. Thus, miR-210-3p combined with BMMSCs is a candidate for future clinical applications in cartilage regeneration and could represent a promising new therapeutic target for OA.


2022 ◽  
Vol 13 (1) ◽  
Author(s):  
Francesco Da Ros ◽  
Luca Persano ◽  
Dario Bizzotto ◽  
Mariagrazia Michieli ◽  
Paola Braghetta ◽  
...  

Abstract Background Dissection of mechanisms involved in the regulation of bone marrow microenvironment through cell–cell and cell–matrix contacts is essential for the detailed understanding of processes underlying bone marrow activities both under physiological conditions and in hematologic malignancies. Here we describe Emilin-2 as an abundant extracellular matrix component of bone marrow stroma. Methods Immunodetection of Emilin-2 was performed in bone marrow sections of mice from 30 days to 6 months of age. Emilin-2 expression was monitored in vitro in primary and mesenchymal stem cell lines under undifferentiated and adipogenic conditions. Hematopoietic stem cells and progenitors in bone marrow of 3- to 10-month-old wild-type and Emilin-2 null mice were analyzed by flow cytometry. Results Emilin-2 is deposited in bone marrow extracellular matrix in an age-dependent manner, forming a meshwork that extends from compact bone boundaries to the central trabecular regions. Emilin-2 is expressed and secreted by both primary and immortalized bone marrow mesenchymal stem cells, exerting an inhibitory action in adipogenic differentiation. In vivo Emilin-2 deficiency impairs the frequency of hematopoietic stem/progenitor cells in bone marrow during aging. Conclusion Our data provide new insights in the contribution of bone marrow extracellular matrix microenvironment in the regulation of stem cell niches and hematopoietic progenitor differentiation.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 1454-1454 ◽  
Author(s):  
Weijie Cao ◽  
Lizhen Liu ◽  
Xiaoyu Lai ◽  
Xiaohong Yu ◽  
He Huang

Abstract Abstract 1454 Poster Board I-477 Introduction Mycophenolate mofetil is now widely used in transplantation as a potent immunosuppressant, whose active metabolite is mycophenolic acid (MPA). MPA inhibits de novo purine biosynthesis by reversible, noncompetitive inhibition of inosine monophosphate dehydrogenase (IMPDH). The inhibition of IMPDH in lymphocytes reduces intracellular guanine nucleotide pools, thus arrests lymphocytes proliferation. Recently investigators reported the antiproliferative effects of MPA on fibroblasts, smooth muscle cells and endothelial cells, but there is no reports of the effects of MPA on human bone marrow-derived mesenchymal stem cells (MSCs). Here we examined the effects of MPA on the proliferation and differentiation of human bone marrow-derived mesenchymal stem cells. Methods Bone marrow aspirates were obtained from healthy volunteers after informed consent, and MSCs were expanded from bone marrow mononuclear cells by discarding non-adherent cells. For proliferation and survival assays, MSCs were treated with MPA at the concentration of 1μM, 10μM, 50μM, and 100μM. Cell proliferation was analyzed using CCK-8 method (Dojindo). Cell viability was assessed by trypan blue exclusion. Apoptosis was detected by PI/Annexin V assay kit (Invitrogen). To assess the effects of MPA on MSCs differentiation, osteogenic differentiation and adipogenic differentiation were induced in the presence of MPA. For the detection of osteogenic differentiation, the deposited minerals was stained with silver by the method of von Kossa and Ca2+ contents was quantified with calcium colorimetric assay kit (Biovision). Adipogenic differentiation was analyzed by Oil Red O staining and Oil Red O staining extraction. Results In the range of 1μM to 100μM, MPA caused a significant subdued proliferation rate of MSCs in a concentration- and time-dependent manner. After 7d of incubation with MPA at the concentration of 1μM, 10μM, 50μM, and 100μM, the proliferation rate was reduced to 65.33±11.03%, 24±3.74%, 15.33±4.03%, and 15.33±6.94% respectively (P<0.01). Adding guanosine (100μM) to the culture restored the proliferation rate (P<0.01) indicating that MPA exerted antiproliferative effects by guanosine depletion. Trypan blue staining showed that there was no statistically significant difference in the ratio of living cells between MPA treated cells and the control group (P>0.05), and PI/Annexin V staining showed no apoptosis induce by MPA. Von Kossa stainnging indicated that treatment with MPA reduced Ca2+ deposition during osteogenic differentiation of MSCs, and Ca2+ quantification further confirmed that MPA inhibited osteogenic differentiation in a concentration-dependent manner. Ca2+ quantification was 78.43±12.79 μg/well and 22.8±6.58 μg/well respectively at the concentration of 10μM and 100μM of MPA, which were significantly lower than the control group(118.33±12.50ug/well, P<0.05). Oil Red O staining and Quantification of lipid contents showed that MPA had no effect on lipid production during adipogenic differentiation. Conclusion Our study demonstrated that MPA inhibited the proliferation of MSCs by guanosine depletion, and also inhibited the osteogenic differentiation in a concentration-dependent manner. However, MPA had no impact on adipogenic differentiation in vitro. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Qiting He ◽  
Ruixi Qin ◽  
Julie Glowacki ◽  
Shuanhu Zhou ◽  
Jie Shi ◽  
...  

Abstract Background 25(OH)D3 is important for the osteoblast differentiation of bone marrow mesenchymual stem cells (BMMSCs), BMMSCs can directly hydroxylate 25(OH)D3 to 1α,25(OH)2D3 to induce osteoblast differentiation. Our previous research with human BMMSCs showed that the cell membrane receptor megalin is required for the 25(OH)D3-DBP complex to enter cells and thereby to stimulate osteoblast differentiation. Furthermore, leptin was shown to upregulate megalin in those cells. Leptin is a known inhibitor of PI3K/AKT-dependent chaperone-mediated autophagy (CMA). In this study, we tested the hypothesis that leptin acts synergistically with 25(OH)D3 to promote osteoblastogenesis in rat BMMSCs by a mechanism that entails inhibition of PI3K/AKT-dependent CMA. Methods The BMMSCs were isolated from rat bone marrow, qRT-PCR and western immunoblots were used to evaluate the expression of megalin, ALP, COL1A1, RUNX2 and CMA activity. The osteoblast differentiation ability was evaluated by ALP activity, ALP staining, and calcium deposition. Results After rBMMSCs were exposed to a combination of leptin and 25(OH)D3, osteoblast differentiation was significantly enhanced, the expression of osteoblastogenic genes ALP, COL1A1, and RUNX2 by qRT-PCR were up-regulated, and ALP activity, ALP staining, and calcium deposition were also significantly increased. The quantity of 25(OH)D3 entering rBMMSCs was increased through increased megalin receptors, and ELISA confirmed that the synthesis of 1α,25(OH)2D3 was increased. Addition of a PI3K/AKT inhibitor reduced the synergistic effect of osteoblast differentiation of rBMMSCs induced by combination leptin with 25(OH)D3; the inhibited CMA activity was partially rescued by a PI3K/AKT inhibitor, and the expression of megalin was down-regulated. Up-regulation of megalin expression by leptin played a synergistic role in osteoblast differentiation of rBMMSCs induced by 25(OH)D3; leptin promoted the expression of megalin by inhibiting the activity of CMA activity. Conclusions These studies indicate that leptin enhanced 25(OH)D3 stimulation of osteoblast differentiation of rBMMSCs by inhibiting CMA activity to increase megalin expression, and that PI3K/AKT signaling pathway is at least partially involved in the regulation of CMA activity and megalin expression.


2020 ◽  
Author(s):  
Shuting Jiang ◽  
Hongyan Liu ◽  
Weiyan Zhu ◽  
Hui Yan ◽  
Beizhan Yan

Abstract Background Mesenchymal stem cells transplantation gradually become a potential treatment for bone defect in clinic practice. This study aimed to investigate the molecular mechanism of PRP and autophagy for osteogenic differentiation in bone marrow mesenchymal stem cells (BMSCs). Methods Thrombin activated PRP was prepared and the BMSCs were treated with activated PRP with different concentration and transfected with miR-140-3p vector (mimics or inhibitor), si-SPRED2 or co-transfected with miR-140-3p inhibitor and si-SPRED2, respectively. qRT-PCR and Western blotting were used to determine the mRNA expression and protein expression. A luciferase reporter assay was conducted to identified the targeting relationship between iR-140-3p and SPRED2 Subsequently, cell proliferation was detected by MTT and ALP activity was also determined. Alizarin red staining was used for the evaluating the formation of calcium nodules. Results MiR-140-3p expression was found to be inhibited by PRP in a dose-dependent manner, besides, cell proliferation, ALP activity, the expression of COL-I, OPN, Runx2 and OCN, and the formation of calcium nodules related to osteogenic differentiation were enhanced by PRP. Subsequently, we found that PRP activated autophagy and up-regulated SPRED2 expression in BMSCs through suppressing miR-140-3p expression. Moreover, we confirmed that miR-140-3p targeted SPRED2 and negatively regulation its expression. Finally, the findings showed that inhibition of miR-140-3p enhanced cell proliferation, osteogenic differentiation and autophagy of BMSCs by negatively regulating SPRED2 expression. Conclusion Thrombin activated PRP accelerated osteogenic differentiation of BMSCs by activing autophagy through miR-140-3p/SPRED2 axis.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 2412-2412
Author(s):  
Fernando Fierro ◽  
David Poitz ◽  
Jan A. Nolta ◽  
Martin Bornhaeuser ◽  
Gerhard Ehninger ◽  
...  

Abstract MicroRNAs (miRNAs) can regulate hematopoietic stem/progenitor cells (HSPC) by modulation of intrinsic cell components such as transcription factors and receptors. In addition, miRNAs could play a role in the microenvironment were HSPC host and consequently affect HSPC via extrinsic factors, which to our knowledge is an hypothesis that has not been tested. Since the chemokine stromal derived factor 1alpha (SDF-1alpha) is essential for both homing and retention of HSPC in the bone marrow, we tested if miRNAs expressed by human bone marrow-derived mesenchymal stem cells (MSC; a key source of SDF-1alpha), could potentially inhibit SDF-1alpha expression. In deed, using luciferase reporter-systems we show specific binding of miR-23a to the 3′UTR of SDF-1alpha. Consequently, transfection of MSCs with a precursor miR-23a (pre-miR-23a) leads to a 30% reduction of SDF-1alpha at both mRNA and protein levels. In contrast, inhibition of endogenous miR-23a with anti-sense oligonucleotides (anti-miR-23a), leads to a significant increase of SDF-1alpha also at both mRNA (30%) and protein (10%) levels as compared to controls (scramble pre-/anti-miR). As a result, migration of CD34+ HSPC in transwell assays is strongly affected upon overexpression or inhibition of miR-23a in MSCs (with pre-miR-23a 35% less migration; with anti-miR-23a 20% more migration, as compared to respective controls). Interestingly, transforming growth factor beta 1 (TGF-beta1) inhibits SDF-1alpha expression in MSCs in a concentration dependent manner, while miR-23a is increased under same experimental settings. Even more, silencing endogenous miR-23a significantly reduces the effect of TGF-beta1 on SDF-1alpha mRNA and protein levels, suggesting that at least in part TGF-beta1 inhibits SDF-1alpha expression via increasing miR-23a levels. This is to our knowledge the first established connection between miRNA biology and HSPC-niche related factors.


2021 ◽  
Vol 11 (8) ◽  
pp. 1636-1642
Author(s):  
Yonghuan Zhou ◽  
Guotang Lan ◽  
Yan Zhou ◽  
Tianhao Qu ◽  
Qing Xiong

Oxidative stress affects bone marrow mesenchymal stem cells (BMSCs). YAP is an effector in Hippo signaling, but its’ role in BMSCs osteogenesis/adipogenesis under oxidative stress has not been reported. Mice BMSCs were isolated and assigned into 3 groups, normal control group; oxidative stress group; and YAP group (transfected with YAP plasmid) followed by analysis of YAP expression by Real time PCR. After 14 days of osteogenesis or adipogenic induction, RUNX2, OPN, FABP4 and PPARγ2 mRNA level was measured along with ROS and SOD activities, ALP activity and Wnt5 expression by western blot. Under oxidative stress, YAP expression significantly decreased, RUNX2 and OPN mRNA expression decreased, ROS expression increased, SOD activity decreased, FABP4 and PPARγ2 protein expression increased, ALP activity and Wnt5 expression decreased (P <0.05). YAP plasmid transfection could significantly up-regulate YAP, RUNX2 and OPN mRNA level, decrease ROS, increase SOD and ALP activity, reduce FABP4 and PPARγ2 mRNA expression and increase Wnt5 expression (P <0.05). YAP level in BMSCs is decreased under oxidative stress. Up-regulating YAP can improve the redox balance, promote BMSCs osteogenic differentiation under oxidative stress and inhibit their differentiation to adipocytes.


2019 ◽  
Vol 20 (6) ◽  
pp. 1519 ◽  
Author(s):  
Lu Wu ◽  
Qinzhi Wei ◽  
Yingjian Lv ◽  
Junchao Xue ◽  
Bo Zhang ◽  
...  

Cadmium is a common environmental pollutant that causes bone damage. However, the effects of cadmium on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs) and its mechanism of action in this process are unclear. Here, we determined the effects of cadmium chloride (CdCl2) on the osteogenic differentiation of BMMSCs and the potential mechanism involved in this process. As determined in the present investigation, CdCl2, in a concentration-dependent manner, affected the viability of BMMSCs and their cytoskeletons. Exposure to 0.1 or 0.2 µM CdCl2 inhibited osteogenic differentiation of BMMSCs, which was reflected in the down-regulation of osteoblast-related genes (ALP, OCN, Runx2, OSX, and OPN); in suppression of the protein expression of alkaline phosphatase (ALP) and runt-related transcription factor 2 (Runx2); and in decreased ALP activity and capacity for mineralization. Moreover, mRNA microarray was performed to determine the roles of these factors in BMMSCs treated with CdCl2 in comparison to control BMMSCs. As determined with the microarrays, the Wingless-type (Wnt), mothers against decapentaplegic and the C. elegans gene Sam (SMAD), and Janus kinase-Signal Transducers and Activators of Transcription (JAK-STAT) signaling pathways were involved in the effects caused by CdCl2. Moreover, during differentiation, the protein levels of Wnt3a, β-catenin, lymphoid enhancer factor 1 (LEF1), and T-cell factor 1 (TCF1) were reduced by CdCl2. The current research shows that CdCl2 suppresses the osteogenesis of BMMSCs via inhibiting the Wnt/β-catenin pathway. The results establish a previously unknown mechanism for bone injury induced by CdCl2.


Sign in / Sign up

Export Citation Format

Share Document