scholarly journals MiR-27b Impairs Adipocyte Differentiation of Human Adipose Tissue-Derived Mesenchymal Stem Cells by Targeting LPL

2018 ◽  
Vol 47 (2) ◽  
pp. 545-555 ◽  
Author(s):  
Xumin Hu ◽  
Jianhua Tang ◽  
Xuyun Hu ◽  
Peng Bao ◽  
Jinxin Pan ◽  
...  

Background/Aims: In this study, the molecular mechanisms of miR-27b and lipoprotein lipase (LPL) that regulate human adipose-derived mesenchymal stem cells (hASCs) adipogenic differentiation were detected. Methods: Microarray analysis was applied to screen for differentially expressed miRNAs and mRNA during hASCs adipocyte differentiation induction. MiR-27b and LPL were found to have abnormal expression. Then, a dual luciferase reporter assay was employed to validate the targeting relationship between miR-27b and LPL. We also utilized qRT-PCR, western blot, cellular immunofluorescence and an oil red O staining assay to analyze the regulation of miR-27b and LPL during adipogenic differentiation. Results: The microarray analysis demonstrated that, during adipogenic differentiation, miR-27b was down-regulated, while LPL was up-regulated but tended to become stable 14 days after induction. A dual luciferase reporter assay confirmed the negative targeting regulatory relationship between miR-27b and LPL. After overexpressing and silencing miR-27b, LPL was found to be reversely regulated by miR-27b according to qRT-PCR and western blot. The fat-formation-related biomarkers CCAAT-enhancer binding protein α (c/EBPα) and peroxisome proliferator-activated receptors γ (PPARγ) had decreasing levels after over-expressing miR-27b or knockdown of LPL followed by adipogenic differentiation. Meanwhile, the oil red O staining assay revealed that the accumulation of lipid droplets decreased. There was no change in the expression of c/EBPα, PPARγ, or lipid droplet accumulation when overexpressing miR-27b and LPL. Conclusion: During the adipogenic differentiation of hASCs, miR-27b expression decreased, and LPL expression increased. The abnormal expression of miR-27b and LPL effectively regulated the adipogenic differentiation of hASCs.

2015 ◽  
Vol 35 (6) ◽  
pp. 2285-2298 ◽  
Author(s):  
Liang Wang ◽  
Lei Xu ◽  
Min Xu ◽  
Guoqiang Liu ◽  
Jian Xing ◽  
...  

Background/Aims: The elucidation of the molecular mechanism of adipocyte differentiation in mesenchymal stem cells is of essential importance for the development of treatments for metabolic diseases, such as obesity and diabetes. Methods: The expression levels of miR-342-3p and carboxy-terminal binding protein 2 (CtBP2) were regulated by oligonucleotide transfection. Adipogenic differentiation was induced by adipogenic medium containing indomethacin, dexamethasone and 3-isobutyl-1-methylxanthine on day 12, as determined by Oil Red O staining and triglyceride concentration assay to assess intracellular lipid accumulation. The induction of adipocyte-specific transcription factors and markers was detected by qRT-PCR and western blot. The regulation of CtBP2 expression by miR-342-3p was determined by western blot, qRT-PCR, luciferase reporter assay, ChIP assay and functional experiments. Results: We revealed that miR-342-3p was enriched in the adipose tissue of obese mice, and its expression was significantly elevated during adipogenic differentiation in both human mesenchymal stem cells (hMSCs) and 3T3L1 cells. Using gain- and loss-of-function assays, we demonstrated that the overexpression of miR-342-3p markedly promoted the differentiation of hMSCs into an adipogenic lineage. Adipogenesis was significantly blocked by miR-342-3p downregulation. We identified and validated that CtBP2 was a direct target of miR-342-3p in this process. The effects of the inhibition of CtBP2 were similar to those of miR-342-5p overexpression on adipogenic differentiation, promoting the release of C/EBPα from CtBP2 binding. Conclusion: miR-342-3p is a powerful enhancer of the adipogenesis of human adipose-derived MSCs that acts by inhibiting CtBP2 and releasing the key adipogenic regulator C/EBPα from CtBP2 binding, subsequently activating the expression of adipogenic transcription factors and markers.


2021 ◽  
Author(s):  
Yangge Du ◽  
Yunsong Liu ◽  
Yongsheng Zhou ◽  
Ping Zhang

Abstract Background: Bone is a rigid organ that provides support and physical protection to vital organs of the body. Several bone loss disorders are commonly associated with increased bone marrow adipose tissue. Bone marrow mesenchymal stromal/stem cells (BMSCs) are multipotent progenitors differentiating into osteoblasts, adipocytes, and chondrocytes. CDC20 is a co-activator of APC/C, required for full ubiquitin ligase activity. In our previous study, CDC20 promoted the osteogenic commitment of BMSCs and Cdc20 conditional knockout mice suggested a decline in bone mass. In this study, we investigated the function of CDC20 in the adipogenic differentiation of BMSCs and provided a new clue between adipogenesis and osteogenesis. Methods: Lentivirus containing CDC20 shRNA was used for CDC20 knockdown in hBMSCs. Primary mBMSCs were isolated from Cdc20f/f and Sp7-Cre;Cdc20f/f mice. Adipogenesis was examined by qRT-PCR and western blot analysis of adipogenic regulators, Oil Red O staining and transplantation into nude mice. The CDC20 knockout efficiency was determined through immunochemistry, qRT-PCR and western blot of bone marrow. Accumulation of adiposity was measured through histology and staining of bone sections. Results: CDC20 expression in hBMSCs was significantly decreased during adipogenic differentiation. Knockdown of CDC20 enhanced adipogenic differentiation of hBMSCs in vitro. CDC20-knockdown hBMSCs showed more adipose tissue–like constructs in H&E staining and Oil Red O staining. Sp7-Cre;Cdc20f/f mice presented increased adipocytes in bone marrow compared with control mice. mBMSCs from Sp7-Cre;Cdc20f/f mice exerted upregulated adipogenic differentiation. Conclusions: Our findings showed that knockdown of CDC20 enhanced adipogenesis of h(m)BMSCs in vitro and in vivo. Overall, CDC20 regulated both adipogenesis and osteogenesis of BMSCs, and may lead to the development of new therapeutic target for “fatty bone” and osteoporosis.


2020 ◽  
Author(s):  
Gang Luo ◽  
Shenqiang Hu ◽  
Tianfu Lai ◽  
Jie Wang ◽  
Li Wang ◽  
...  

Abstract Background: MicroRNAs are a class of small non-coding RNAs and participate in the regulation of cell differentiation programs. Previous studies have demonstrated that miR-9-5p play a key role during cancer cell research, but the mechanisms regulating of miR-9-5p in adipogenesis remain poorly understood. This study intended to investigate its significance in rabbits with high quality meat by observing the regulating effect of miR-9-5p in pre-adipocytes and finding related targets. Methods: In this study, a dual luciferase reporter assay was employed to validate the targeting relationship between miR-9-5p and leptin gene. We also utilized quantitative reverse transcription PCR (qRT-PCR), western blot, determination of triglyceride and oil red O staining assay to analyze the regulation of miR-9-5p and leptin gene during adipocyte differentiation. Results: The analysis demonstrated that during pre-adipocyte differentiation, miR-9-5p was up-regulated and the fat formation related biomarkers fatty acid-binding protein 4(FABP4), CCAAT-enhancer binding protein α(C/EBPα), and peroxisome proliferator activated receptorγ (PPARγ) were also up-regulated. Meanwhile, the oil red O staining assay revealed that the accumulation of lipid droplets increased. We explored the expression pattern and role of miR-9-5p in adipogenesis using white pre-adipocytes. The results showed that miR-9-5p was up-regulated during pre-adipocyte differentiation and overexpression of miR-9-5p enhanced lipid accumulation. Furthermore, we found overexpression of miR-9-5p significantly up regulated the mRNA levels of marker gene PPARγ, C/EBPα and FABP4, as well as the protein levels of PPARγ and content of triglyceride, the results suggested that miR-9-5p might be involved in the regulation of rabbit pre-adipocyte differentiation. We predicted leptin is a target of miR-9-5p by using bioinformatics tools and the conclusion was validated in a luciferase reporter assay. Finally, we verified inhibition of leptin by si-leptin promoted rabbits pre-adipocyte differentiation. Conclusion: In a word, these results indicate that miR-9-5p influences rabbits white pre-adipocyte differentiation by targeting leptin.


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.


2017 ◽  
Vol 41 (2) ◽  
pp. 530-542 ◽  
Author(s):  
Huaqing Wang ◽  
Zhao Xie ◽  
Tianyong Hou ◽  
Zhiqiang Li ◽  
Ke Huang ◽  
...  

Background/Aims: Osteogenic differentiation of mesenchymal stem cells (MSCs) plays a crucial role in bone regeneration and bone reparation. This complex process is regulated precisely and firmly by specific factors. Recent studies have demonstrated that miR-125b regulates osteogenic differentiation, but little is known about the molecular mechanisms of this regulation. Furthermore, how miR-125b regulates the osteogenic differentiation of MSCs still needs elucidation. Methods: In the present study, human bone marrow-derived mesenchymal stem cells (hBMSCs) were isolated and induced to osteoblasts with miR-125b inhibition or overexpression. qRT-PCR and western blot analysis were used to detect the expression of osteogenic marker genes and proteins. Alkaline phosphatase (ALP) and Alizarin Red (ARS) staining were performed to evaluate the osteoblast phenotype. TargetScan, PicTar and miRanda database were used to predict the target gene of miR-125b. Dual luciferase reporter assay and RNA interference were performed to verify the target gene. Micro-CT imaging and histochemical staining were used to investigate the bone defect repair capacity of miR-125b in vivo. Results: We observed that miR-125b was expressed at a low level during the osteogenic differentiation of hBMSCs. Then, we found that osteogenic marker genes were negatively regulated by miR-125b during the course of osteogenic differentiation, suggesting that miR-125b down regulation plays an important role in the process of osteogenic differentiation. Bioinformatics approaches using miRNA target prediction algorithms indicated that the bone morphogenetic protein type Ib receptor (BMPR1b) is a potential target of miR-125b. The results of the dual luciferase reporter assay indicated that miR-125b binds to the 3’-UTR of the BMPR1b gene. We observed that knockdown of BMPR1b by siRNA inhibited the osteogenic differentiation of hBMSCs. Furthermore, by co-transfecting cells with an miR-125b inhibitor and si-BMPR1b, we found that the osteogenic capacity of the cells transfected with miR-125b inhibitor was blocked upon knockdown of BMPR1b. In vivo, demineralized bone matrix (DBM) was composited with hBMSCs as a scaffold to repair segmental femoral defects. By inhibiting the expression of miR-125b, hBMSCs showed a better capacity to repair bone defects. Conclusions: Taken together, our study demonstrated that miR-125b regulated the osteogenic differentiation of hBMSCs by targeting BMPR1b and that inhibiting miR-125b expression could enhance the capacity of bone defect repair in vivo.


2021 ◽  
Vol 8 ◽  
Author(s):  
Qing Xu ◽  
Yong Wang ◽  
Xin Li ◽  
Yu Du ◽  
Yanyan Li ◽  
...  

Intramuscular fat contributes to the improvement of meat quality of goats. MicroRNAs (miRNAs) have been reported to regulate adipocyte differentiation and maturation. The aim of our study was to clarify whether miR-10a-5p regulates goat intramuscular preadipocyte (GIPC) differentiation and its direct downstream signaling pathway. GIPCs were isolated from longissimus dorsi, whose miR-10a-5p level was measured at different time point of differentiation induction. Adipogenic differentiation of the GIPCs was evaluated by Oil Red O and BODIPY staining, and the expression changes of adipogenic genes like ACC, ATGL, CEBPβ, PPARγ, etc. Related mechanisms were verified by qPCR, a bioinformatic analysis, a dual-luciferase reporter assay, overexpression, and siRNA transfection. Oil Red O and BODIPY staining both with adipogenic gene detection showed that miR-10a-5p suppressed the accumulation of lipid droplets in GIPCs and inhibited its differentiation. The dual-luciferase reporter assay experiment revealed that miR-10a-5p regulates GIPC differentiation by directly binding to KLF8 3’UTR to regulate its expression. Thus, the results indicated that miR-10a-5p inhibits GIPC differentiation by targeting KLF8 and supply a new target for fat deposition and meat quality improvement.


2020 ◽  
Author(s):  
Gang Luo ◽  
Shenqiang Hu ◽  
Tianfu Lai ◽  
Jie Wang ◽  
Li Wang ◽  
...  

Abstract Background: MicroRNAs are a class of small non-coding RNAs and participate in the regulation of cell differentiation programs. Previous studies have demonstrated that miR-9-5p play a key role during cancer cell research, but the mechanisms regulating of miR-9-5p in adipogenesis remain poorly understood. This study intended to investigate its significance in rabbits with high quality meat by observing the regulating effect of miR-9-5p in pre-adipocytes and finding related targets. Methods: In this study, a dual luciferase reporter assay was employed to validate the targeting relationship between miR-9-5p and leptin gene. We also utilized quantitative reverse transcription PCR (qRT-PCR), western blot, determination of triglyceride and oil red O staining assay to analyze the regulation of miR-9-5p and leptin gene during adipocyte differentiation. Results: The analysis demonstrated that during pre-adipocyte differentiation, miR-9-5p was up-regulated and the fat formation related biomarkers fatty acid-binding protein 4(FABP4), CCAAT-enhancer binding protein α(C/EBPα), and peroxisome proliferator activated receptorγ (PPARγ) were also up-regulated. Meanwhile, the oil red O staining assay revealed that the accumulation of lipid droplets increased. We explored the expression pattern and role of miR-9-5p in adipogenesis using white pre-adipocytes. The results showed that miR-9-5p was up-regulated during pre-adipocyte differentiation and overexpression of miR-9-5P enhanced lipid accumulation. Furthermore, we found overexpression of miR-9-5p significantly up regulated the mRNA levels of marker gene PPARγ, C/EBPα and FABP4, as well as the protein levels of PPARγ and content of triglyceride, the results suggested that miR-9-5p might be involved in the regulation of rabbit pre-adipocyte differentiation. We predicted leptin is a target of miR-9-5p by using bioinformatics tools and the conclusion was validated in a luciferase reporter assay. Finally, we verified inhibition of leptin by si-leptin promoted rabbits pre-adipocyte differentiation. Conclusion: In a word, these results indicate that miR-9-5p promotes rabbits white pre-adipocyte differentiation by targeting leptin .


Molecules ◽  
2021 ◽  
Vol 26 (7) ◽  
pp. 1831
Author(s):  
Thitianan Kulsirirat ◽  
Sittisak Honsawek ◽  
Mariko Takeda-Morishita ◽  
Nuttanan Sinchaipanid ◽  
Wanvisa Udomsinprasert ◽  
...  

Andrographolide is a labdane diterpenoid herb, which is isolated from the leaves of Andrographis paniculata, and widely used for its potential medical properties. However, there are no reports on the effects of andrographolide on the human suprapatellar fat pad of osteoarthritis patients. In the present study, our goal was to evaluate the innovative effects of andrographolide on viability and Tri-lineage differentiation of human mesenchymal stem cells from suprapatellar fat pad tissues. The results revealed that andrographolide had no cytotoxic effects when the concentration was less than 12.5 µM. Interestingly, andrographolide had significantly enhanced, dose dependent, osteogenesis and chondrogenesis as evidenced by a significantly intensified stain for Alizarin Red S, Toluidine Blue and Alcian Blue. Moreover, andrographolide can upregulate the expression of genes related to osteogenic and chondrogenic differentiation, including Runx2, OPN, Sox9, and Aggrecan in mesenchymal stem cells from human suprapatellar fat pad tissues. In contrast, andrographolide suppressed adipogenic differentiation as evidenced by significantly diminished Oil Red O staining and expression levels for adipogenic-specific genes for PPAR-γ2 and LPL. These findings confirm that andrographolide can specifically enhance osteogenesis and chondrogenesis of mesenchymal stem cells from human suprapatellar fat pad tissues. It has potential as a therapeutic agent derived from natural sources for regenerative medicine.


2020 ◽  
Author(s):  
Zhixi Li ◽  
Gang Wu ◽  
Jie Li ◽  
Youyu Wang ◽  
Xueming Ju ◽  
...  

Abstract Background This article focuses on the roles and mechanism of lncRNA CRNDE on the progression of HCC. Methods We used qRT-PCR to detect the expression of lncRNA CRNDE in HCC cells, normal cells and clinical tissues. MTT assay, FCM analysis, Transwell migration and invasion assay were used to detect the effects of lncRNA CRNDE on cell viability, apoptosis, migration and invasion of HCC cells. The expression of apoptosis-related proteins Bcl-2, Bax, Cleaved Caspase 3, Cleaved Caspase 9, EMT epithelial marker E-cadherin and mesothelial marker Vimentin were analyzed by Western blot. Online prediction software was used to predict the binding sites between lncRNA CRNDE and miR-539-5p, or miR-539-5p and POU2F1 3’UTR. Dual luciferase reporter assay, qRT-PCR and RNA pulldown were used to detect target-relationship between lncRNA CRNDE and miR-539-5p. Dual luciferase reporter assay, qRT-PCR, Western blot and Immunofluorescence were used to detect target-relationship between miR-539-5p and POU2F1. qRT-PCR was used to detect the expression of miR-539-5p and POU2F1 in clinical tissues. Rescue experiments was used to evaluate the association among lncRNA CRNDE, miR-539-5p and POU2F1. Finally, we used Western blot to detect the effects of lncRNA CRNDE, miR-539-5p and POU2F1 on NF-κB and AKT pathway. Results lncRNA CRNDE was highly expressed in HCC cells and HCC tissues compared with normal cells and the corresponding adjacent normal tissues. lncRNA CRNDE promoted the cell viability, migration and invasion of HCC cells, while inhibited the apoptosis and promoted the EMT process of HCC cells. lncRNA CRNDE adsorbed miR-539-5p acts as a competitive endogenous RNA to regulate POU2F1 expression indirectly. In HCC clinical tissues, miR-539-5p expression decreased and POU2F1 increased compared with the corresponding adjacent normal tissues. lncRNA CRNDE/miR-539-5p/POU2- F1 participated the NF-κB and AKT pathway in HCC. Conclusion lncRNA CRNDE promotes the expression of POU2F1 by adsorbing miR-539-5p, thus promoting the progression of HCC. Keywords: HCC, lncRNA CRNDE, miR-539-5p, POU2F1, ceRNA


2020 ◽  
Author(s):  
Yijing Chu ◽  
Yan Zhang ◽  
Guoqiang Gao ◽  
Jun Zhou ◽  
Yang Lv ◽  
...  

Abstract Background: Human chorionic villous mesenchymal stem cells (CV-MSCs) are found to be a promising and effective treatment for tissue injury. Trophoblast dysfunction during pregnancies is significantly involved in the pathogenesis of preeclampsia (PE). This work was to understand how CV-MSCs regulated trophoblast function. Methods: In this study, we treated trophoblasts with CV-MSC-derived exosomes and RNA-seq analysis was used to understand the changes in trophoblasts. We examined the levels of TXNIP and β-catenin in trophoblasts by immunohistochemistry, western blot and qRT-PCR assays. Luciferase reporter assays and qRT-PCR assays were used to understand the role of miR135b-5p in the effects of CV-MSC-derived exosomes. The growth and invasion of trophoblasts was evaluated with the CCK-8 and transwell assays. Results: The treatment markedly enhanced the trophoblast proliferation and invasion. Furthermore, a significant decrease of TXNIP expression and inactivation of the β-catenin pathway in CV-MSCs exosomes-treated trophoblasts was observed. Consistent with these findings, TXNIP inhibition exhibited the same effect of promoting trophoblast proliferation and invasion as induced by CV-MSC-derived exosomes, also with the accompaniment of inactivation of β-catenin pathway. In addition, overexpression of TXNIP activated the β-catenin pathway in trophoblasts, and reduced the proliferation and invasion of trophoblasts. Importantly, miR135b-5p was found to be highly expressed in CV-MSC exosomes and interact with TXNIP. The miR-135b-5p overexpression significantly elevated the proliferation and invasion of trophoblasts, which could be attenuated by TXNIP overexpression. Conclusion: Our results suggest that TXNIP-dependent β-catenin pathway inactivation mediated by miR135b-5p which is delivered by CV-MSC-derived exosomes could promote the proliferation and invasion of trophoblasts.


Sign in / Sign up

Export Citation Format

Share Document