Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Tissue Cell ; 85: 102223, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37776785

ABSTRACT

Periodontitis represents a severe inflammatory illness in tooth supporting tissue. It has been supported that cAMP response element binding protein 1 (CREB1), a common transcription factor, extensively participates in osteogenic differentiation. Here, the current study was to look into the impacts of CREB1 on the process of periodontitis and its possible action mechanism. After human periodontal ligament stem cells (PDLSCs) were challenged with zoledronic acid (ZA), CREB1 expression was examined with RT-qPCR and western blotting. CCK-8 assay appraised cell activity. Following CREB1 elevation or/and vascular endothelial growth factor (VEGF) silencing in ZA-treated PDLSCs, CCK-8 and TUNEL assays separately estimated cell viability and apoptosis. Western blotting tested the expression of apoptosis- and osteogenic differentiation-associated proteins. ALP staining measured PDLSCs osteogenic ability and ARS staining estimated mineralized nodule formation. JASPAR predicted the potential binding of CREB1 with VEGF promoter, which was then testified by ChIP and luciferase reporter assays. RT-qPCR and western blotting tested VEGF expression. CREB1 expression was declined in ZA-exposed PDLSCs and CREB1 elevation exacerbated the viability and osteogenic differentiation while obstructed the apoptosis of PDLSCs. Additionally, CREB1 bond to VEGF promoter and transcriptionally activated VEGF expression. Further, VEGF absence partially stimulated the apoptosis while suppressed the osteogenic differentiation of CREB1-overexpressing PDLSCs treated by ZA. To be concluded, CREB1 might activate VEGF transcription to obstruct the apoptosis while contribute to the osteogenic differentiation of ZA-treated PDLSCs.


Subject(s)
Osteogenesis , Periodontitis , Humans , Apoptosis/genetics , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Osteogenesis/physiology , Periodontal Ligament , Periodontitis/metabolism , Signal Transduction/physiology , Sincalide/metabolism , Stem Cells , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Zoledronic Acid/pharmacology , Zoledronic Acid/metabolism
2.
Connect Tissue Res ; 64(6): 532-542, 2023 11.
Article in English | MEDLINE | ID: mdl-37427853

ABSTRACT

AIM OF THE STUDY: To investigate the role of MetaLnc9 in the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs). MATERIALS AND METHODS: We used lentiviruses to knockdown or overexpress MetaLnc9 in hBMSCs. qRT-PCR was employed to determine the mRNA levels of osteogenic-related genes in transfected cells. ALP staining and activity assay, ARS staining and quantification were used to identify the degree of osteogenic differentiation. Ectopic bone formation was conducted to examine the osteogenesis of transfected cells in vivo. AKT pathway activator SC-79 and inhibitor LY294002 were used to validate the relationship between MetaLnc9 and AKT signaling pathway. RESULTS: The expression of MetaLnc9 was significantly upregulated in the osteogenic differentiation of hBMSCs. MetaLnc9 knockdown inhibited the osteogenesis of hBMSCs, whereas overexpression of it promoted the osteogenic differentiation both in vitro and in vivo. Taking a deeper insight, we found that MetaLnc9 enhanced the osteogenic differentiation by activating AKT signaling. The inhibitor of AKT signaling LY294002 could reverse the positive effect on osteogenesis brought by MetaLnc9 overexpression, whereas the activator of AKT signaling SC-79 could reverse the negative effect caused by MetaLnc9 knockdown. CONCLUSION: Our works uncovered a vital role of MetaLnc9 in osteogenesis via regulating the AKT signaling pathway. [Figure: see text].


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Humans , Proto-Oncogene Proteins c-akt/metabolism , Cells, Cultured , Signal Transduction , Cell Differentiation , Mesenchymal Stem Cells/metabolism , Bone Marrow Cells
3.
Nano Res ; 16(2): 1992-2002, 2023.
Article in English | MEDLINE | ID: mdl-36405985

ABSTRACT

Single-atom nanozymes (SANs) are the new emerging catalytic nanomaterials with enzyme-mimetic activities, which have many extraordinary merits, such as low-cost preparation, maximum atom utilization, ideal catalytic activity, and optimized selectivity. With these advantages, SANs have received extensive research attention in the fields of chemistry, energy conversion, and environmental purification. Recently, a growing number of studies have shown the great promise of SANs in biological applications. In this article, we present the most recent developments of SANs in anti-infective treatment, cancer diagnosis and therapy, biosensing, and antioxidative therapy. This text is expected to better guide the readers to understand the current state and future clinical possibilities of SANs in medical applications.

4.
FASEB J ; 36(11): e22590, 2022 11.
Article in English | MEDLINE | ID: mdl-36208289

ABSTRACT

Many circular RNAs (circRNAs) involved in the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs) have recently been discovered. The role of circHIPK3 in osteogenesis has yet to be determined. Cell transfection was conducted using small-interfering RNAs (siRNAs). Expression of osteogenic markers were detected by quantitative reverse transcription-polymerase chain reaction, western blotting analysis, and immunofluorescence staining. Ectopic bone formation models in nude mice were used to examined the bone formation ability in vivo. The autophagy flux was examined via western blotting analysis, immunofluorescence staining and transmission electron microscopy analysis. RNA immunoprecipitation (RIP) analysis was carried out to analyze the binding between human antigen R (HUR) and circHIPK3 or autophagy-related 16-like 1 (ATG16L1). Actinomycin D was used to determine the mRNA stability. Our results demonstrated that silencing circHIPK3 promoted the osteogenesis of hBMSCs while silencing the linear mHIPK3 did not affect osteogenic differentiation, both in vivo and in vitro. Moreover, we found that knockdown of circHIPK3 activated autophagy flux. Activation of autophagy enhanced the osteogenesis of hBMSCs and inhibition of autophagy reduced the osteogenesis through using autophagy regulators chloroquine and rapamycin. We also discovered that circHIPK3 and ATG16L1 both bound to HUR. Knockdown of circHIPK3 released the binding sites of HUR to ATG16L1, which stabilized the mRNA expression of ATG16L1, resulting in the upregulation of ATG16L1 and autophagy activation. CircHIPK3 functions as an osteogenesis and autophagy regulator and has the potential for clinical application in the future.


Subject(s)
Mesenchymal Stem Cells , Osteogenesis , Animals , Autophagy/genetics , Bone Marrow Cells , Cell Differentiation/physiology , Cells, Cultured , Chloroquine , Dactinomycin , Humans , Mesenchymal Stem Cells/metabolism , Mice , Mice, Nude , Osteogenesis/genetics , RNA, Circular/genetics , RNA, Messenger/metabolism , Sirolimus/metabolism
5.
Stem Cell Res Ther ; 11(1): 450, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33097082

ABSTRACT

BACKGROUND: Heat shock protein B7 (HSPB7), which belongs to small heat shock protein family, has been reported to be involved in diverse biological processes and diseases. However, whether HSPB7 regulates osteogenic differentiation of human adipose derived stem cells (hASCs) remains unexplored. METHODS: The expression level of HSPB7 during the osteogenesis of hASCs was examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot analysis. Lentivirus transfection was used to knock down or overexpress HSPB7, which enabled us to investigate the effect of HSPB7 on osteogenic differentiation of hASCs. U0126 and extracellular signal-regulated protein kinase 1/2 (ERK1/2) siRNA were used to identify the mechanism of the HSPB7/ERK1/2 axis in regulating osteogenic differentiation of hASCs. Moreover, ectopic bone formation in nude mice and osteoporosis mice model was used to investigate the effect of HSPB7 on osteogenesis in vivo. RESULTS: In this study, we found the expression of HSPB7 was significantly downregulated during the osteogenic differentiation of hASCs. HSPB7 knockdown remarkably promoted osteogenic differentiation of hASCs, while HSPB7 overexpression suppressed osteogenic differentiation of hASCs both in vitro and in vivo. Moreover, we discovered that the enhancing effect of HSPB7 knockdown on osteogenic differentiation was related to the activation of extracellular signal-regulated protein kinase (ERK) signaling pathway. Inhibition of ERK signaling pathway with U0126 or silencing ERK1/2 effectively blocked the stimulation of osteogenic differentiation induced by HSPB7 knockdown. Additionally, we found that HSPB7 expression was markedly increased in mouse bone marrow mesenchymal stem cells (mBMSCs) from the osteoporotic mice which suggested that HSPB7 might be utilized as a potential target in the development of effective therapeutic strategies to treat osteoporosis and other bone diseases. CONCLUSION: Taken together, these findings uncover a previously unrecognized function of HSPB7 in regulating osteogenic differentiation of hASCs, partly via the ERK signaling pathway.


Subject(s)
Cell Differentiation , HSP27 Heat-Shock Proteins , Osteogenesis , Adipose Tissue/metabolism , Animals , Cells, Cultured , HSP27 Heat-Shock Proteins/genetics , HSP27 Heat-Shock Proteins/metabolism , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Osteogenesis/genetics , Signal Transduction , Stem Cells
6.
Cell Death Dis ; 11(7): 601, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32732881

ABSTRACT

Osteoporosis is a prevalent metabolic bone disease characterized by low bone mineral density and degenerative disorders of bone tissues. Previous studies showed the abnormal osteogenic differentiation of endogenous bone marrow mesenchymal stem cells (BMSCs) contributes to the development of osteoporosis. However, the underlying mechanisms by which BMSCs undergo osteogenic differentiation remain largely unexplored. Recently, long non-coding RNAs have been discovered to play important roles in regulating BMSC osteogenesis. In this study, we first showed MIR22HG, which has been demonstrated to be involved in the progression of several cancer types, played an important role in regulating BMSC osteogenesis. We found the expression of MIR22HG was significantly decreased in mouse BMSCs from the osteoporotic mice and it was upregulated during the osteogenic differentiation of human BMSCs. Overexpression of MIR22HG in human BMSCs enhanced osteogenic differentiation, whereas MIR22HG knockdown inhibited osteogenic differentiation both in vitro and in vivo. Mechanistically, MIR22HG promoted osteogenic differentiation by downregulating phosphatase and tensin homolog (PTEN) and therefore activating AKT signaling. Moreover, we found MIR22HG overexpression promoted osteoclastogenesis of RAW264.7 cells, which indicated that MIR22HG played a significant role in bone metabolism and could be a therapeutic target for osteoporosis and other bone-related diseases.


Subject(s)
Cell Differentiation/genetics , Mesenchymal Stem Cells/metabolism , Osteogenesis/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/metabolism , Animals , Enzyme Activation , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Mice , Osteoclasts/metabolism , Ovariectomy , RAW 264.7 Cells , RNA, Long Noncoding/genetics , Signal Transduction , X-Ray Microtomography
7.
Biochem Biophys Res Commun ; 524(2): 516-522, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32014253

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs), which have multipotential differentiation and self-renewal ability, have been becoming an attractive source of seed cells for bone tissue engineering. Nonetheless, the precise underlying mechanisms of osteogenesis of BMSCs have not been fully understood. Retinoic acid-induced gene 3 (RAI3) has been found to play important roles in mesenchymal stem cells (MSCs) adipogenesis in our previous study. However, its function in the osteogenic differentiation of BMSCs remains unknown. In this study, we found that RAI3 was significantly reduced in osteogenically differentiated BMSCs; RAI3 knockdown promoted osteogenesis of BMSCs both in vitro and in vivo. Moreover, we found RAI3 knockdown significantly upregulated the expression level of phosphorylated signal transducer and activator of transcription 3 (p-STAT3), and AG-490 which can inhibit the STAT3 signaling reversed the enhancing effect of RAI3 knockdown on the osteogenic differentiation of BMSCs. These results suggest that RAI3 plays important roles in BMSCs osteogenesis with an involvement of the STAT3 signaling, which might open a new avenue to explore BMSCs osteogenesis for the application of BMSCs in bone regeneration.


Subject(s)
Mesenchymal Stem Cells/cytology , Osteogenesis , Receptors, G-Protein-Coupled/genetics , STAT3 Transcription Factor/metabolism , Cell Differentiation , Cells, Cultured , Down-Regulation , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
8.
Biochem Biophys Res Commun ; 493(1): 618-624, 2017 11 04.
Article in English | MEDLINE | ID: mdl-28870805

ABSTRACT

Retinoic acid-induced protein 3 (RAI3) has been found to play significant roles in embryonic development, cellular proliferation and differentiation, but its role in adipogenesis has not been explored. In this study, we discovered RAI3 was downregulated during the adipogenic differentiation of human adipose derived stem cells (hASCs). Moreover, we demonstrated that knockdown of RAI3 promoted adipogenic differentiation of hASCs both in vitro and in vivo. Mechanistically, our findings showed that inhibition of RAI3 in hASCs reduced the expression of ß-catenin, and lithium chloride which can activate the ß-catenin pathway abolished the effect of RAI3 knockdown on the adipogenesis. These results suggest RAI3 plays an important role in adipogenesis of hASCs and may have a potential use in the future application.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis/physiology , Receptors, G-Protein-Coupled/metabolism , Stem Cells/cytology , Stem Cells/metabolism , beta Catenin/metabolism , Cell Differentiation/physiology , Cells, Cultured , Down-Regulation/physiology , Gene Knockdown Techniques , Humans
9.
Sci Rep ; 7(1): 8080, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28808264

ABSTRACT

Adipogenesis plays an important role in the regulation of whole-body energy homeostasis and is inextricably related to obesity. Several studies have highlighted the relevance of microRNAs in adipocyte differentiation, but the contributions of long non-coding RNAs (lncRNAs) are still largely uncharacterized. Here, we determined that lncRNA MIR31HG is related to adipocyte lineage commitment. We demonstrated that knockdown of MIR31HG inhibited adipocyte differentiation, whereas overexpression of MIR31HG promoted adipogenesis in vitro and in vivo. Furthermore, inhibition of MIR31HG reduced the enrichment of active histone markers, histone H3 lysine 4 trimethylation (H3K4me3) and acetylation (AcH3), in the promoter of the adipogenic-related gene, fatty acid binding protein 4 (FABP4), leading to suppression of its expression and adipogenesis. These results provide new insights into the molecular mechanisms of MIR31HG in terms of adipogenesis and may have implications for obesity and associated disorders.


Subject(s)
Adipocytes/physiology , Cell Differentiation/genetics , Fatty Acid-Binding Proteins/genetics , RNA, Long Noncoding/genetics , Stem Cells/physiology , Acetylation , Adipogenesis/genetics , Animals , Cell Line , Histones/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Obesity/genetics , Promoter Regions, Genetic/genetics
10.
Mol Cell Biochem ; 433(1-2): 51-60, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28382492

ABSTRACT

lncRNAs are an emerging class of regulators involved in multiple biological processes. MEG3, an lncRNA, acts as a tumor suppressor, has been reported to be linked with osteogenic differentiation of MSCs. However, limited knowledge is available concerning the roles of MEG3 in the multilineage differentiation of hASCs. The current study demonstrated that MEG3 was downregulated during adipogenesis and upregulated during osteogenesis of hASCs. Further functional analysis showed that knockdown of MEG3 promoted adipogenic differentiation, whereas inhibited osteogenic differentiation of hASCs. Mechanically, MEG3 may execute its role via regulating miR-140-5p. Moreover, miR-140-5p was upregulated during adipogenesis and downregulated during osteogenesis in hASCs, which was negatively correlated with MEG3. In conclusion, MEG3 participated in the balance of adipogenic and osteogenic differentiation of hASCs, and the mechanism may be through regulating miR-140-5p.


Subject(s)
Adipogenesis , Adipose Tissue/metabolism , Cell Differentiation , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Osteogenesis , RNA, Long Noncoding/metabolism , Adipose Tissue/cytology , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , RNA, Long Noncoding/genetics
11.
J Bone Miner Res ; 32(3): 508-521, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27696501

ABSTRACT

An imbalance in osteogenesis and adipogenesis is a crucial pathological factor in the development of osteoporosis. Many attempts have been made to develop drugs to prevent and treat this disease. In the present study, we investigated the phenomenon whereby downregulation of SLC7A11 significantly enhanced the osteogenic differentiation of mesenchymal stem cells (MSCs) in vitro, and promoted the bone formation in vivo. Sulfasalazine (SAS), an inhibitor of SLC7A11, increased the osteogenic potential effectively. Mechanistically, inhibition of SLC7A11 by SAS treatment or knockdown of SLC7A11 increased BMP2/4 expression dramatically. In addition, we detected increased Slc7a11 expression in bone marrow MSCs of ovariectomized (OVX) mice. Remarkably, SAS treatment attenuated bone loss in ovariectomized mice. Together, our data suggested that SAS could be used to treat osteoporosis by enhancing osteogenic differentiation of MSCs. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Amino Acid Transport System y+/antagonists & inhibitors , Bone Morphogenetic Proteins/metabolism , Bone Resorption/drug therapy , Cell Differentiation/drug effects , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Ovariectomy , Sulfasalazine/pharmacology , Amino Acid Transport System y+/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Resorption/pathology , Down-Regulation/drug effects , Enzyme Inhibitors/pharmacology , Female , Gene Knockdown Techniques , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred BALB C , Osteoporosis/enzymology , Osteoporosis/pathology , Sulfasalazine/therapeutic use
12.
Cell Biol Int ; 41(1): 33-41, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27797128

ABSTRACT

Recently, long non-coding RNAs (lncRNAs) have emerged as critical players in gene regulation for multiple biological processes. However, their roles and functions in human adipose-derived stem cells (hASCs) differentiation remain unclear. In the present study, we investigated the role of lncRNA myocardial infarction-associated transcript (MIAT) in the osteogenic differentiation of hASCs. We found that the expression of MIAT was downregulated in a time-dependent manner during hASCs osteoinduction. MIAT knockdown promoted osteogenic differentiation of hASCs both in vitro and in vivo. Moreover, MIAT expression was increased upon tumor necrosis factor-α treatment and MIAT knockdown reversed the negative effects of inflammation on osteoblastic differentiation. This study improves our knowledge of lncRNAs in governing the osteogenic differentiation of hASCs and may provide novel therapeutic strategies for treating bone diseases.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/genetics , Gene Knockdown Techniques , Osteogenesis/genetics , RNA, Long Noncoding/genetics , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Choristoma/pathology , Down-Regulation/drug effects , Down-Regulation/genetics , Humans , Mice, Inbred BALB C , Mice, Nude , Osteogenesis/drug effects , RNA, Long Noncoding/metabolism , Stem Cells/drug effects , Time Factors , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics
13.
Stem Cell Reports ; 7(2): 236-48, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27453008

ABSTRACT

MiR-34a was demonstrated to be upregulated during the osteogenic differentiation of human adipose-derived stem cells (hASCs). Overexpression of miR-34a significantly increased alkaline phosphatase activity, mineralization capacity, and the expression of osteogenesis-associated genes in hASCs in vitro. Enhanced heterotopic bone formation in vivo was also observed upon overexpression of miR-34a in hASCs. Mechanistic investigations revealed that miR-34a inhibited the expression of retinoblastoma binding protein 2 (RBP2) and reduced the luciferase activity of reporter gene construct comprising putative miR-34a binding sites in the 3' UTR of RBP2. Moreover, miR-34a downregulated the expression of NOTCH1 and CYCLIN D1 and upregulated the expression of RUNX2 by targeting RBP2, NOTCH1, and CYCLIN D1. Taken together, our results suggested that miR-34a promotes the osteogenic differentiation of hASCs via the RBP2/NOTCH1/CYCLIN D1 coregulatory network, indicating that miR-34a-targeted therapy could be a valuable approach to promote bone regeneration.


Subject(s)
Adipose Tissue/cytology , Cell Differentiation/genetics , Cyclin D1/metabolism , MicroRNAs/metabolism , Osteogenesis/genetics , Receptors, Notch/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Stem Cells/cytology , Core Binding Factor Alpha 1 Subunit/metabolism , Cyclin D1/genetics , Gene Expression Regulation , Gene Regulatory Networks , Humans , MicroRNAs/genetics , Receptors, Notch/genetics , Retinol-Binding Proteins, Cellular/genetics , Stem Cells/metabolism
14.
Stem Cells ; 34(9): 2332-41, 2016 09.
Article in English | MEDLINE | ID: mdl-27300495

ABSTRACT

Human mesenchymal stem cells (MSCs) are multipotent progenitor cells that can differentiate into osteoblasts, chondrocytes, and adipocytes. The importance of epigenetic regulation for osteogenic differentiation of MSCs is widely accepted. However, the molecular mechanisms are poorly understood. Here, we show that histone H3K9 acetyltransferase PCAF plays a critical role in osteogenic differentiation of MSCs. Knockdown of PCAF significantly reduced the bone formation both in vitro and in vivo. Mechanistically, PCAF controls BMP signaling genes expression by increasing H3K9 acetylation. Most importantly, PCAF expression is significantly decreased in bone sections of ovariectomized or aged mice. Histone modification enzyme is chemically modifiable; therefore, PCAF may represent a novel therapeutic target for stem cell-mediated regenerative medicine and the treatment of osteoporosis. Stem Cells 2016;34:2332-2341.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation , Histones/metabolism , Osteogenesis , Osteoporosis/metabolism , Signal Transduction , p300-CBP Transcription Factors/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage , Gene Expression Regulation , Gene Knockdown Techniques , HEK293 Cells , Humans , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Nude , Osteogenesis/genetics , Osteoporosis/pathology , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Smad Proteins/metabolism
15.
Stem Cells ; 34(11): 2707-2720, 2016 11.
Article in English | MEDLINE | ID: mdl-27334046

ABSTRACT

Osteogenic differentiation and bone formation is suppressed under condition of inflammation induced by proinflammation cytokines. A number of studies indicate miRNAs play a significant role in tumor necrosis factor-α-induced inhibition of bone formation, but whether long non-coding RNAs are also involved in this process remains unknown. In this study, we evaluated the role of MIR31HG in osteogenesis of human adipose-derived stem cells (hASCs) in vitro and in vivo. The results suggested that knockdown of MIR31HG not only significantly promoted osteogenic differentiation, but also dramatically overcame the inflammation-induced inhibition of osteogenesis in hASCs. Mechanistically, we found MIR31HG regulated bone formation and inflammation via interacting with NF-κB. The p65 subunit bound to the MIR31HG promoter and promoted MIR31HG expression. In turn, MIR31HG directly interacted with IκBα and participated in NF-κB activation, which builds a regulatory circuitry with NF-κB. Targeting this MIR31HG-NF-κB regulatory loop may be helpful to improve the osteogenic capacity of hASCs under inflammatory microenvironment in bone tissue engineering. Stem Cells 2016;34:2707-2720.


Subject(s)
Feedback, Physiological , NF-KappaB Inhibitor alpha/genetics , NF-kappa B p50 Subunit/genetics , Osteoblasts/metabolism , RNA, Long Noncoding/genetics , Stem Cells/metabolism , Transcription Factor RelA/genetics , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Humans , Lipopolysaccharides/pharmacology , Mice, Inbred BALB C , Mice, Nude , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B p50 Subunit/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Osteogenesis/genetics , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects , Tissue Engineering , Transcription Factor RelA/metabolism , Transplantation, Heterologous
16.
Sci Rep ; 6: 28897, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27349231

ABSTRACT

Bone marrow mesenchymal stem cells (BMSCs) exhibit an increased propensity toward adipocyte differentiation accompanied by a reduction in osteogenesis in osteoporotic bone marrow. However, limited knowledge is available concerning the role of long non-coding RNAs (lncRNAs) in the differentiation of BMSCs into adipocytes. In this study, we demonstrated that lncRNA H19 and microRNA-675 (miR-675) derived from H19 were significantly downregulated in BMSCs that were differentiating into adipocytes. Overexpression of H19 and miR-675 inhibited adipogenesis, while knockdown of their endogenous expression accelerated adipogenic differentiation. Mechanistically, we found that miR-675 targeted the 3' untranslated regions of the histone deacetylase (HDAC) 4-6 transcripts and resulted in deregulation of HDACs 4-6, essential molecules in adipogenesis. In turn, trichostatin A, an HDAC inhibitor, significantly reduced CCCTC-binding factor (CTCF) occupancy in the imprinting control region upstream of the H19 gene locus and subsequently downregulated the expression of H19. These results show that the CTCF/H19/miR-675/HDAC regulatory pathway plays an important role in the commitment of BMSCs into adipocytes.


Subject(s)
Adipocytes/metabolism , Cell Differentiation/genetics , Histone Deacetylases/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Adipocytes/cytology , Adipogenesis/genetics , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Epigenesis, Genetic , Gene Expression , HEK293 Cells , Histone Deacetylases/metabolism , Humans , Mesenchymal Stem Cells/cytology , RNA Interference , Sequence Homology, Nucleic Acid
17.
J Bone Miner Res ; 31(2): 391-402, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26420353

ABSTRACT

As the most well-studied histone acetyltransferase (HAT) in yeast and mammals, general control nonderepressible 5 (GCN5) was documented to play essential roles in various developmental processes. However, little is known about its role in osteogenic differentiation of mesenchymal stem cells (MSCs). Here, we detected the critical function of GCN5 in osteogenic commitment of MSCs. In this role, the HAT activity of GCN5 was not required. Mechanistically, GCN5 repressed nuclear factor kappa B (NF-κB)-dependent transcription and inhibited the NF-κB signaling pathway. The impaired osteogenic differentiation by GCN5 knockdown was blocked by inhibition of NF-κB. Most importantly, the expression of GCN5 was decreased significantly in the bone tissue sections of ovariectomized mice or aged mice. Collectively, these results may point to the GCN5-NF-κB pathway as a novel potential molecular target for stem cell mediated regenerative medicine and the treatment of metabolic bone diseases such as osteoporosis.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/metabolism , NF-kappa B/metabolism , Osteogenesis , Signal Transduction , p300-CBP Transcription Factors/metabolism , Animals , Female , HEK293 Cells , Humans , Mice , Osteoporosis/metabolism
18.
Biomaterials ; 35(23): 6015-25, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24794925

ABSTRACT

Human adipose-derived stem cells (hASCs) are a highly attractive source in bone tissue engineering. It has become increasingly clear that chromatin regulators play an important role in cell fate determination. However, how osteogenic differentiation of hASCs is controlled by epigenetic mechanisms is not fully understood. Here we use genetic tools and chemical inhibitors to modify the epigenetic program of hASCs and identify lysine-specific demethylase 1 (LSD1), a histone demethylase that specifically catalyzes demethylation of di- and mono- methyl histone H3 lysine 4 (H3K4me2/1), as a key regulator in osteogenic differentiation of hASCs. Specifically, we demonstrated that genetic depletion of LSD1 with lentiviral strategy for gene knockdown promoted osteogenic differentiation of hASCs by cell studies and xenograft assays. At the molecular level, we found that LSD1 regulates osteogenesis-associated genes expression through its histone demethylase activity. Significantly, we demonstrated LSD1 demethylase inhibitors could efficiently block its catalytic activity and epigenetically boost osteogenic differentiation of hASCs. Altogether, our study defined the functional and biological roles of LSD1 and extensively explored the effects of its enzymatic activity in osteogenic differentiation of hASCs. A better understanding of how LSD1 influences on osteogenesis associated epigenetic events will provide new insights into the modulation of hASCs based cell therapy and improve the development of bone tissue engineering with epigenetic intervention.


Subject(s)
Adipocytes/cytology , Histone Demethylases/chemistry , Histone Demethylases/genetics , Osteoblasts/cytology , Osteoblasts/physiology , Stem Cells/cytology , Stem Cells/physiology , Adipocytes/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Epigenesis, Genetic/physiology , Genetic Enhancement/methods , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Osteogenesis/genetics , Tissue Scaffolds
19.
Biomaterials ; 35(15): 4489-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24589359

ABSTRACT

The purpose of this study was to investigate the cooperative effects of simvastatin (SIM) and stromal cell-derived factor-1α (SDF-1α) on the osteogenic and migration capabilities of mesenchymal stem cells (MSCs), and construct a cell-free bone tissue engineering system comprising SIM, SDF-1α and scaffold. We found that 0.2 µm SIM significantly increased alkaline phosphatase activity (P < 0.05) of mouse bone marrow MSCs with no inhibition of cell proliferation, and enhanced the chemotactic capability of SDF-1α (P < 0.05). Next, we constructed a novel cell-free bone tissue engineering system using PLGA loaded with SIM and SDF-1α, and applied it in critical-sized calvarial defects in mice. New bone formation in the defect was evaluated by micro-CT, HE staining and immunohistochemistry. The results showed that PLGA loaded with SIM and SDF-1α promoted bone regeneration significantly more than controls. We investigated possible mechanisms, and showed that SDF-1α combined with SIM increased MSC migration and homing in vivo, promoted angiogenesis and enhanced the expression of BMP-2 in newly-formed bone tissue. In conclusion, SIM enhanced the chemotactic capability of SDF-1α and the cell-free bone tissue engineering system composed of SIM, SDF-1α and scaffold promoted bone regeneration in mouse critical-sized calvarial defects.


Subject(s)
Bone Regeneration/drug effects , Chemokine CXCL12/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Simvastatin/therapeutic use , Skull/injuries , Tissue Engineering/methods , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL12/administration & dosage , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Simvastatin/administration & dosage , Skull/drug effects , Skull/pathology
20.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 30(5): 526-9, 534, 2012 Oct.
Article in Chinese | MEDLINE | ID: mdl-23173321

ABSTRACT

OBJECTIVE: To synthesize a novel nano-antibacterial inorganic filler and provide a new way to give dental composite resin antibacterial property. METHODS: Quaternary ammonium iodide N,N,N-trimethyl-3-(trimethoxysilyl) propan-1-aminium iodide were organically synthesized firstly and then the N,N,N-trimethyl-3-(trimethoxysilyl) propan-1-aminium iodide was grafted to the nano-silica particle to synthesize the antihacterial inorganic fillers nano-silica particle grafted with quaternary ammonium salt. All the products were analyzed and identified by infrared spectrum analysis. Then Streptococcus mutans were chosen as experimental object to analysis the antibacterial property of nanoantibacterial inorganic filler. RESULTS: Quaternary ammonium salt was grafted to the surface of nano-silica particles successfully by infrared spectrum analysis. Compared with the control group, the nano-silica particle grafted with quaternary ammonium salt had a strong bactericidal effect on Streptococcus mutons (P < 0.01). CONCLUSION: The nano-silica particle grafted with quaternary ammonium salt has a strong antibacterial property and could be used to improve dental composite resin antibacterial property.


Subject(s)
Anti-Bacterial Agents , Streptococcus mutans , Acrylic Resins , Ammonium Compounds , Composite Resins , Polyurethanes , Quaternary Ammonium Compounds , Silicon Dioxide
SELECTION OF CITATIONS
SEARCH DETAIL
...