Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 791
Filter
1.
PLoS One ; 17(2): e0263839, 2022.
Article in English | MEDLINE | ID: mdl-35213543

ABSTRACT

The ubiquitin/proteasome system controls the stability of Runx2 and JunB, proteins essential for differentiation of mesenchymal progenitor/stem cells (MPCs) to osteoblasts. Local administration of proteasome inhibitor enhances bone fracture healing by accelerating endochondral ossification. However, if a short-term administration of proteasome inhibitor enhances fracture repair and potential mechanisms involved have yet to be exploited. We hypothesize that injury activates the ubiquitin/proteasome system in callus, leading to elevated protein ubiquitination and degradation, decreased MPCs, and impaired fracture healing, which can be prevented by a short-term of proteasome inhibition. We used a tibial fracture model in Nestin-GFP reporter mice, in which a subgroup of MPCs are labeled by Nestin-GFP, to test our hypothesis. We found increased expression of ubiquitin E3 ligases and ubiquitinated proteins in callus tissues at the early phase of fracture repair. Proteasome inhibitor Bortezomib, given soon after fracture, enhanced fracture repair, which is accompanied by increased callus Nestin-GFP+ cells and their proliferation, and the expression of osteoblast-associated genes and Runx2 and JunB proteins. Thus, early treatment of fractures with Bortezomib could enhance the fracture repair by increasing the number and proliferation of MPCs.


Subject(s)
Bortezomib/pharmacology , Cell Proliferation/drug effects , Fracture Healing/drug effects , Mesenchymal Stem Cells/enzymology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Tibial Fractures/enzymology , Animals , Cell Proliferation/genetics , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Disease Models, Animal , Fracture Healing/genetics , Male , Mice , Mice, Transgenic , Osteoblasts/enzymology , Proteasome Endopeptidase Complex/genetics , Tibial Fractures/drug therapy , Tibial Fractures/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
2.
J Biol Chem ; 298(3): 101639, 2022 03.
Article in English | MEDLINE | ID: mdl-35090892

ABSTRACT

Phosphatidylinositol-4-phosphate 5-kinase type-1 gamma (Pip5k1c) is a lipid kinase that plays a pivotal role in the regulation of receptor-mediated calcium signaling in multiple tissues; however, its role in the skeleton is not clear. Here, we show that while deleting Pip5k1c expression in the mesenchymal stem cells using Prx1-Cre transgenic mice does not impair the intramembranous and endochondral ossification during skeletal development, it does cause osteopenia in adult mice, but not rapidly growing young mice. We found Pip5k1c loss dramatically decreases osteoblast formation and osteoid and mineral deposition, leading to reduced bone formation. Furthermore, Pip5k1c loss inhibits osteoblastic, but promotes adipogenic, differentiation of bone marrow stromal cells. Pip5k1c deficiency also impairs cytoplasmic calcium influx and inactivates the calcium/calmodulin-dependent protein kinase, which regulates levels of transcription factor Runx2 by modulating its stability and subsequent osteoblast and bone formation. In addition, Pip5k1c loss reduces levels of the receptor activator of nuclear factor-κB ligand, but not that of osteoprotegerin, its decoy receptor, in osteoblasts in bone and in sera. Finally, we found Pip5k1c loss impairs the ability of bone marrow stromal cells to support osteoclast formation of bone marrow monocytes and reduces the osteoclast precursor population in bone marrow, resulting in reduced osteoclast formation and bone resorption. We conclude Pip5k1c deficiency causes a low-turnover osteopenia in mice, with impairment of bone formation being greater than that of bone resorption. Collectively, we uncover a novel function and mechanism of Pip5k1c in the control of bone mass and identify a potential therapeutic target for osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Bone Resorption , Mesenchymal Stem Cells , Phosphotransferases (Alcohol Group Acceptor) , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Remodeling/physiology , Bone Resorption/enzymology , Bone Resorption/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Osteoclasts/metabolism , Osteogenesis , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RANK Ligand/metabolism
3.
Int J Mol Sci ; 22(22)2021 Nov 11.
Article in English | MEDLINE | ID: mdl-34830075

ABSTRACT

This study aimed to investigate the effects of the human macrophage (MP) secretome in cellular xenograft rejection. The role of human nucleoside diphosphate kinase A (hNME1), from the secretome of MPs involved in the neuronal differentiation of miniature pig adipose tissue-derived mesenchymal stem cells (mp AD-MSCs), was evaluated by proteomic analysis. Herein, we first demonstrate that hNME1 strongly binds to porcine ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyltransferase 1 (pST8SIA1), which is a ganglioside GD3 synthase. When hNME1 binds with pST8SIA1, it induces degradation of pST8SIA1 in mp AD-MSCs, thereby inhibiting the expression of ganglioside GD3 followed by decreased neuronal differentiation of mp AD-MSCs. Therefore, we produced nanobodies (NBs) named NB-hNME1 that bind to hNME1 specifically, and the inhibitory effect of NB-hNME1 was evaluated for blocking the binding between hNME1 and pST8SIA1. Consequently, NB-hNME1 effectively blocked the binding of hNME1 to pST8SIA1, thereby recovering the expression of ganglioside GD3 and neuronal differentiation of mp AD-MSCs. Our findings suggest that mp AD-MSCs could be a potential candidate for use as an additive, such as an immunosuppressant, in stem cell transplantation.


Subject(s)
Cell Differentiation/drug effects , Gangliosides/biosynthesis , Mesenchymal Stem Cells/enzymology , NM23 Nucleoside Diphosphate Kinases/pharmacology , Neurons/enzymology , Sialyltransferases/antagonists & inhibitors , Animals , Humans , Sialyltransferases/metabolism , Swine , Swine, Miniature
4.
Commun Biol ; 4(1): 1315, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799676

ABSTRACT

Telomere maintenance and tumor cell differentiation have been separately implicated in neuroblastoma malignancy. Their mechanistic connection is unclear. We analyzed neuroblastoma cell lines and morphologic subclones representing the adrenergic (ADRN) and mesenchymal (MES) differentiation states and uncovered sharp differences in their telomere protein and telomerase activity levels. Pharmacologic conversion of ADRN into MES cells elicited consistent and robust changes in the expression of telomere-related proteins. Conversely, stringent down-regulation of telomerase activity triggers the differentiation of ADRN into MES cells, which was reversible upon telomerase up-regulation. Interestingly, the MES differentiation state is associated with elevated levels of innate immunity factors, including key components of the DNA-sensing pathway. Accordingly, MES but not ADRN cells can mount a robust response to viral infections in vitro. A gene expression signature based on telomere and cell lineage-related factors can cluster neuroblastoma tumor samples into predominantly ADRN or MES-like groups, with distinct clinical outcomes. Our findings establish a strong mechanistic connection between telomere and differentiation and suggest that manipulating telomeres may suppress malignancy not only by limiting the tumor growth potential but also by inducing tumor cell differentiation and altering its immunogenicity.


Subject(s)
Cell Differentiation , Neuroblastoma/enzymology , Telomerase/metabolism , Cell Line, Tumor , Humans , Mesenchymal Stem Cells/enzymology
5.
Biosci Rep ; 41(8)2021 08 27.
Article in English | MEDLINE | ID: mdl-34350461

ABSTRACT

Periodontitis is a series of inflammatory processes caused by bacterial infection. Parathyroid hormone (PTH) plays a critical role in bone remodeling. The present study aimed to investigate the influences of PTH on human bone marrow mesenchymal stem cells (HBMSCs) pretreated with lipopolysaccharide (LPS). The proliferative ability was measured using cell counting kit-8 (CCK-8) and flow cytometry. The optimal concentrations of PTH and LPS were determined using alkaline phosphatase (ALP) activity assay, ALP staining, and Alizarin Red staining. Osteogenic differentiation was further assessed by quantitative reverse-transcription polymerase chain reaction (RT-qPCR), Western blot analysis, and immunofluorescence staining. PTH had no effects on the proliferation of HBMSCs. Also, 100 ng/ml LPS significantly inhibited HBMSC osteogenesis, while 10-9 mol/l PTH was considered as the optimal concentration to reverse the adverse effects. Mechanistically, c-Jun N-terminal kinase (JNK) phosphorylation was activated by PTH in LPS-induced HBMSCs. SP600125, a selective inhibitor targeting JNK mitogen-activated protein kinase (MAPK) signaling, weakened the effects of PTH. Taken together, the findings revealed the role and mechanism of PTH and JNK pathway in promoting the osteogenic differentiation of LPS-induced HBMSCs, which offered an alternative for treating periodontal diseases.


Subject(s)
Cell Differentiation/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Parathyroid Hormone/pharmacology , Periodontitis/drug therapy , Adult , Cells, Cultured , Humans , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Periodontitis/enzymology , Periodontitis/pathology , Phosphorylation , Signal Transduction , Young Adult
6.
Cells ; 10(6)2021 06 11.
Article in English | MEDLINE | ID: mdl-34207965

ABSTRACT

This study aims to obtain sufficient corneal endothelial cells for regenerative application. We examined the combinatory effects of Rho-associated kinase (ROCK) inhibitor Y-27632 and mesenchymal stem cell-derived conditioned medium (MSC-CM) on the proliferation and senescence of rabbit corneal endothelial cells (rCECs). rCECs were cultured in a control medium, a control medium mixed with either Y-27632 or MSC-CM, and a combinatory medium containing Y-27632 and MSC-CM. Cells were analyzed for morphology, cell size, nuclei/cytoplasmic ratio, proliferation capacity and gene expression. rCECs cultured in a combinatory culture medium showed a higher passage number, cell proliferation, and low senescence. rCECs on collagen type I film showed high expression of tight junction. The cell proliferation marker Ki-67 was positively stained either in Y-27632 or MSC-CM-containing media. Genes related to cell proliferation resulted in negligible changes in MKI67, CIP2A, and PCNA in the combinatory medium, suggesting proliferative capacity was maintained. In contrast, all of these genes were significantly downregulated in the other groups. Senescence marker ß-galactosidase-positive cells significantly decreased in either MSC-CM and/or Y-27632 mixed media. Senescence-related genes downregulated LMNB1 and MAP2K6, and upregulated MMP2. Cell cycle checkpoint genes such as CDC25C, CDCA2, and CIP2A did not vary in the combinatory medium but were significantly downregulated in either ROCK inhibitor or MSC-CM alone. These results imply the synergistic effect of combinatory culture medium on corneal endothelial cell proliferation and high cell number. This study supports high potential for translation to the development of human corneal endothelial tissue regeneration.


Subject(s)
Cell Proliferation , Cellular Senescence , Culture Media, Conditioned/pharmacology , Endothelium, Corneal/cytology , Mesenchymal Stem Cells/cytology , Protein Kinase Inhibitors/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Cell Differentiation , Cell Movement , Cells, Cultured , Endothelium, Corneal/drug effects , Endothelium, Corneal/enzymology , Enzyme Inhibitors/pharmacology , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Pyridines/pharmacology , Rabbits
7.
Int J Mol Sci ; 22(12)2021 Jun 15.
Article in English | MEDLINE | ID: mdl-34203758

ABSTRACT

Synovial fluid contains cytokines, growth factors and resident mesenchymal stem cells (MSCs). The present study aimed to (1) determine the effects of autologous and allogeneic synovial fluid on viability, proliferation and chondrogenesis of equine bone marrow MSCs (BMMSCs) and (2) compare the immunomodulatory properties of equine synovial fluid MSCs (SFMSCs) and BMMSCs after stimulation with interferon gamma (INF-γ). To meet the first aim of the study, the proliferation and viability of MSCs were evaluated by MTS and calcein AM staining assays. To induce chondrogenesis, MSCs were cultured in a medium containing TGF-ß1 or different concentrations of synovial fluid. To meet the second aim, SFMSCs and BMMSCs were stimulated with IFN-γ. The concentration of indoleamine-2,3-dioxygenase (IDO) and nitric oxide (NO) were examined. Our results show that MSCs cultured in autologous or allogeneic synovial fluid could maintain proliferation and viability activities. Synovial fluid affected chondrocyte differentiation significantly, as indicated by increased glycosaminoglycan contents, compared to the chondrogenic medium containing 5 ng/mL TGF-ß1. After culturing with IFN-γ, the conditioned media of both BMMSCs and SFMSCs showed increased concentrations of IDO, but not NO. Stimulating MSCs with synovial fluid or IFN-γ could enhance chondrogenesis and anti-inflammatory activity, respectively, suggesting that the joint environment is suitable for chondrogenesis.


Subject(s)
Chondrogenesis/drug effects , Immunomodulation/drug effects , Interferon-gamma/pharmacology , Mesenchymal Stem Cells/immunology , Synovial Fluid/metabolism , Animals , Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Colony-Forming Units Assay , Horses , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Nitric Oxide/metabolism
8.
Int J Mol Sci ; 22(6)2021 Mar 18.
Article in English | MEDLINE | ID: mdl-33803805

ABSTRACT

During biomineralization, the cells generating the biominerals must be able to sense the external physical stimuli exerted by the growing mineralized tissue and change their intracellular protein composition according to these stimuli. In molluscan shell, the myosin-chitin synthases have been suggested to be the link for this communication between cells and the biomaterial. Hyaluronan synthases (HAS) belong to the same enzyme family as chitin synthases. Their product hyaluronan (HA) occurs in the bone and is supposed to have a regulatory function during bone regeneration. We hypothesize that HASes' expression and activity are controlled by fluid-induced mechanotransduction as it is known for molluscan chitin synthases. In this study, bone marrow-derived human mesenchymal stem cells (hMSCs) were exposed to fluid shear stress of 10 Pa. The RNA transcriptome was analyzed by RNA sequencing (RNAseq). HA concentrations in the supernatants were measured by ELISA. The cellular structure of hMSCs and HAS2-overexpressing hMSCs was investigated after treatment with shear stress using confocal microscopy. Fluid shear stress upregulated the expression of genes that encode proteins belonging to the HA biosynthesis and bone mineralization pathways. The HAS activity appeared to be induced. Knowledge about the regulation mechanism governing HAS expression, trafficking, enzymatic activation and quality of the HA product in hMSCs is essential to understand the biological role of HA in the bone microenvironment.


Subject(s)
Hyaluronan Synthases/metabolism , Mesenchymal Stem Cells/enzymology , Rheology , Stress, Mechanical , Aged , Aged, 80 and over , Cell Shape , Cells, Cultured , Humans , Hyaluronic Acid/biosynthesis , Male , Mesenchymal Stem Cells/cytology , Middle Aged , Transcription, Genetic , Up-Regulation/genetics
9.
Cells ; 10(5)2021 04 27.
Article in English | MEDLINE | ID: mdl-33925659

ABSTRACT

The application of physiological oxygen (physoxia) concentrations is becoming increasingly commonplace within a mammalian stem cell culture. Human mesenchymal stem cells (hMSCs) attract widespread interest for clinical application due to their unique immunomodulatory, multi-lineage potential, and regenerative capacities. Descriptions of the impact of physoxia on global DNA methylation patterns in hMSCs and the activity of enzymatic machinery responsible for its regulation remain limited. Human bone marrow-derived mesenchymal stem cells (BM-hMSCs, passage 1) isolated in reduced oxygen conditions displayed an upregulation of SOX2 in reduced oxygen conditions vs. air oxygen (21% O2, AO), while no change was noted for either OCT-4 or NANOG. DNA methylation marks 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC) showed decreases in 2% O2 environment (workstation) (2% WKS). DNMT3B (DNA methyltransferase 3B) and TET1 (Ten-eleven translocation enzyme 1) displayed reduced transcription in physoxia. Consistent with transcriptional downregulation, we noted increased promoter methylation levels of DNMT3B in 2% WKS accompanied by reduced DNMT3B and TET1 protein expression. Finally, a decrease in HIF1A (Hypoxia-inducible factor 1A) gene expression in 2% WKS environment correlated with protein levels, while HIF2A was significantly higher in physoxia correlated with protein expression levels vs. AO. Together, these data have demonstrated, for the first time, that global 5mC, 5hmC, and DNMT3B are oxygen-sensitive in hMSCs. Further insights into the appropriate epigenetic regulation within hMSCs may enable increased safety and efficacy development within the therapeutic ambitions.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Mesenchymal Stem Cells/enzymology , Oxygen/metabolism , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Adult , Air , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunophenotyping , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Mixed Function Oxygenases/metabolism , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Up-Regulation , DNA Methyltransferase 3B
10.
Cell Mol Life Sci ; 78(10): 4639-4651, 2021 May.
Article in English | MEDLINE | ID: mdl-33787980

ABSTRACT

Hematopoietic system transports all necessary nutrients to the whole organism and provides the immunological protection. Blood cells have high turnover, therefore, this system must be dynamically controlled and must have broad regeneration potential. In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. First, we discuss how HO-1 influences hematopoietic stem cells (HSC) self-renewal, aging and differentiation. We also describe a critical role of HO-1 in endothelial cells and mesenchymal stromal cells that constitute the specialized bone marrow niche of HSC. We further discuss the molecular and cellular mechanisms by which HO-1 modulates innate and adaptive immune responses. Finally, we highlight how modulation of HO-1 activity regulates the mobilization of bone marrow hematopoietic cells to peripheral blood. We critically discuss the issue of metalloporphyrins, commonly used pharmacological modulators of HO-1 activity, and raise the issue of their important HO-1-independent activities.


Subject(s)
Aging , Cell Differentiation , Cell Self Renewal , Cellular Microenvironment , Hematopoiesis , Heme Oxygenase-1/metabolism , Mesenchymal Stem Cells/cytology , Animals , Humans , Mesenchymal Stem Cells/enzymology
11.
Biochem Biophys Res Commun ; 549: 221-228, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33706192

ABSTRACT

Human bone marrow mesenchymal stem cell (hBMSC) viability and osteogenic differentiation play a critical role in bone disorders such as osteoporosis. In the present study, we identified the aberrant PLK4 upregulation in osteoporosis and downregulation in BMSCs during osteogenic differentiation. In isolated hBMSCs, PLK4 overexpression significantly inhibited, whereas PLK4 knockdown promoted cell viability and hBMSC osteogenic differentiation. For molecular mechanism, PLK4 overexpression decreased, whereas PLK4 knockdown increased WNT1 and ß-catenin protein levels and the phosphorylation of Smad1/5/8. The Wnt/ß-catenin signaling antagonist Dickkopf 1 (DKK1) or the BMP-Smads antagonist LDN193189 dramatically suppressed hBMSC osteoblast differentiation, and partially attenuated the promotive effects of PLK4 knockdown on hBMSC osteogenic differentiation. Altogether, PLK4 overexpression impairs hBMSC viability and osteogenic differentiation potential, possibly through the Wnt/ß-catenin signaling and BMP/Smads signaling.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Osteogenesis , Protein Serine-Threonine Kinases/metabolism , Bone Morphogenetic Proteins/metabolism , Cell Survival , Down-Regulation , Humans , Osteogenesis/drug effects , Smad Proteins/metabolism , Wnt Signaling Pathway
12.
Cell Death Dis ; 12(3): 240, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664231

ABSTRACT

Liver diseases with different pathogenesis share common pathways of immune-mediated injury. Chitinase-3-like protein 1 (CHI3L1) was induced in both acute and chronic liver injuries, and recent studies reported that it possesses an immunosuppressive ability. CHI3L1 was also expressed in mesenchymal stem cells (MSCs), thus we investigates the role of CHI3L1 in MSC-based therapy for immune-mediated liver injury here. We found that CHI3L1 was highly expressed in human umbilical cord MSCs (hUC-MSCs). Downregulating CHI3L1 mitigated the ability of hUC-MSCs to inhibit T cell activation, proliferation and inflammatory cytokine secretion in vitro. Using Concanavalin A (Con A)-induced liver injury mouse model, we found that silencing CHI3L1 significantly abrogated the hUC-MSCs-mediated alleviation of liver injury, accompanying by weakened suppressive effects on infiltration and activation of hepatic T cells, and secretion of pro-inflammatory cytokines. In addition, recombinant CHI3L1 (rCHI3L1) administration inhibited the proliferation and function of activated T cells, and alleviated the Con A-induced liver injury in mice. Mechanistically, gene set enrichment analysis showed that JAK/STAT signalling pathway was one of the most significantly enriched gene pathways in T cells co-cultured with hUC-MSCs with CHI3L1 knockdown, and further study revealed that CHI3L1 secreted by hUC-MSCs inhibited the STAT1/3 signalling in T cells by upregulating peroxisome proliferator-activated receptor δ (PPARδ). Collectively, our data showed that CHI3L1 was a novel MSC-secreted immunosuppressive factor and provided new insights into therapeutic treatment of immune-mediated liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Chitinase-3-Like Protein 1/metabolism , Liver/enzymology , Lymphocyte Activation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/enzymology , Paracrine Communication , T-Lymphocytes/enzymology , Animals , Cell Proliferation , Cells, Cultured , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Coculture Techniques , Concanavalin A , Cytokines/metabolism , Disease Models, Animal , Female , Humans , Liver/immunology , Liver/pathology , Mice, Inbred C57BL , Phosphorylation , Receptors, Cytoplasmic and Nuclear/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Umbilical Cord/cytology
13.
Cell Death Dis ; 12(3): 238, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664230

ABSTRACT

The ubiquitin protease pathway plays important role in human bone marrow-derived mesenchymal stem cell (hBMSC) differentiation, including osteogenesis. However, the function of deubiquitinating enzymes in osteogenic differentiation of hBMSCs remains poorly understood. In this study, we aimed to investigate the role of ubiquitin-specific protease 53 (USP53) in the osteogenic differentiation of hBMSCs. Based on re-analysis of the Gene Expression Omnibus database, USP53 was selected as a positive regulator of osteogenic differentiation in hBMSCs. Overexpression of USP53 by lentivirus enhanced osteogenesis in hBMSCs, whereas knockdown of USP53 by lentivirus inhibited osteogenesis in hBMSCs. In addition, USP53 overexpression increased the level of active ß-catenin and enhanced the osteogenic differentiation of hBMSCs. This effect was reversed by the Wnt/ß-catenin inhibitor DKK1. Mass spectrometry showed that USP53 interacted with F-box only protein 31 (FBXO31) to promote proteasomal degradation of ß-catenin. Inhibition of the osteogenic differentiation of hBMSCs by FBXO31 was partially rescued by USP53 overexpression. Animal studies showed that hBMSCs with USP53 overexpression significantly promoted bone regeneration in mice with calvarial defects. These results suggested that USP53 may be a target for gene therapy for bone regeneration.


Subject(s)
Bone Marrow Cells/enzymology , Mesenchymal Stem Cells/enzymology , Osteogenesis , Ubiquitin-Specific Proteases/metabolism , Adult , Animals , Bone Regeneration , Case-Control Studies , Cells, Cultured , Dependovirus/genetics , F-Box Proteins/metabolism , Genetic Vectors , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mesenchymal Stem Cell Transplantation , Mice, Inbred ICR , Osteoporosis/metabolism , Osteoporosis/pathology , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Skull/metabolism , Skull/pathology , Skull/surgery , Tumor Suppressor Proteins/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism
14.
Int J Nanomedicine ; 16: 725-740, 2021.
Article in English | MEDLINE | ID: mdl-33542627

ABSTRACT

PURPOSE: As a dental material, polyetheretherketone (PEEK) is bioinert that does not induce cellular response and bone/gingival tissues regeneration. This study was to develop bioactive coating on PEEK and investigate the effects of coating on cellular response. MATERIALS AND METHODS: Tantalum pentoxide (TP) coating was fabricated on PEEK surface by vacuum evaporation and responses of rat bone marrow mesenchymal stem (RBMS) cells/human gingival epithelial (HGE) were studied. RESULTS: A dense coating (around 400 nm in thickness) of TP was closely combined with PEEK (PKTP). Moreover, the coating was non-crystalline TP, which contained many small humps (around 10 nm in size), exhibiting a nanostructured surface. In addition, the roughness, hydrophilicity, surface energy, and protein adsorption of PKTP were remarkably higher than that of PEEK. Furthermore, the responses (adhesion, proliferation, and osteogenic gene expression) of RBMS cells, and responses (adhesion and proliferation) of HGE cells to PKTP were remarkably improved in comparison with PEEK. It could be suggested that the nanostructured coating of TP on PEEK played crucial roles in inducing the responses of RBMS/HGE cells. CONCLUSION: PKTP with elevated surface performances and outstanding cytocompatibility might have enormous potential for dental implant application.


Subject(s)
Epithelial Cells/cytology , Gingiva/cytology , Ketones/pharmacology , Mesenchymal Stem Cells/cytology , Nanostructures/chemistry , Oxides/pharmacology , Polyethylene Glycols/pharmacology , Tantalum/pharmacology , Adsorption , Alkaline Phosphatase/metabolism , Animals , Benzophenones , Cell Adhesion/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Epithelial Cells/drug effects , Epithelial Cells/ultrastructure , Gene Expression Regulation/drug effects , Humans , Hydrophobic and Hydrophilic Interactions , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Nanostructures/ultrastructure , Osteogenesis/drug effects , Osteogenesis/genetics , Polymers , Rats, Sprague-Dawley , Spectroscopy, Fourier Transform Infrared , Surface Properties , X-Ray Diffraction
15.
Cell Cycle ; 20(4): 392-405, 2021 02.
Article in English | MEDLINE | ID: mdl-33487075

ABSTRACT

As an important histone acetylase, the transcriptional coactivator P300/CBP affects target gene expression and plays a role in the maintenance of stem cell characteristics and differentiation potential. In this study, we explored the action of a highly effective selective histone acetylase inhibitor, C646, on goat adipose-derived stem cells (gADSCs), and investigated the impact of histone acetylation on the growth characteristics and the differentiation potential of ADSCs. We found that C646 blocked the cell proliferation, arrested the cell cycle, and triggered apoptosis. Notably, immunocytochemistry and western blot analyses showed that the acetylation level of histone H3K9 was increased. Moreover, although real-time quantitative PCR and western blot confirmed that P300 expression was inhibited under these conditions, the expression level of two other histone acetylases, TIP60 and PCAF, was significantly increased. Furthermore, C646 clearly promoted the differentiation of gADSCs into adipocytes and had an impact on their differentiation into neuronal cells. This study provides new insights into the epigenetic regulation of stem cell differentiation and may represent an experimental basis for the comprehension of stem cell characteristics and function. Furthermore, it is of great relevance for the application of adult stem cells to somatic cell cloning, which may improve the efficiency of large livestock cloning and foster the production of transgenic animals.


Subject(s)
Benzoates/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Mesenchymal Stem Cells/drug effects , Nitrobenzenes/pharmacology , Pyrazolones/pharmacology , Animals , Cell Differentiation/physiology , Cell Proliferation/physiology , Cells, Cultured , Goats , Histone Acetyltransferases/metabolism , Mesenchymal Stem Cells/enzymology
16.
J Cell Physiol ; 236(6): 4273-4289, 2021 06.
Article in English | MEDLINE | ID: mdl-33452710

ABSTRACT

While mesenchymal stem cells (MSCs) have been widely used to repair radiation-induced bone damage, the molecular mechanism underlying the effects of MSCs in the maintenance of bone homeostasis under radiation stress remains largely unknown. In this study, the role and mechanisms of R-spondin 1 (Rspo1)-leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis on the initiation of self-defense of bone mesenchymal stem cells (BMSCs) and maintenance of bone homeostasis under radiation stress were investigated. Interestingly, radiation increased levels of Rspo1 and LGR4 in BMSCs. siRNA knockdown of Rspo1 or LGR4 aggravated radiation-induced impairment of self-renewal ability and osteogenic differentiation potential of BMSCs. However, exogenous Rspo1 significantly attenuated radiation-induced depletion of BMSCs, and promoted the lineage shift towards osteoblasts. This alteration was associated with the reversal of mammalian target of rapamycin (mTOR) activation and autophagy decrement. Pharmacological and genetic blockade of autophagy attenuated the radio-protective effects of Rspo1, rendering BMSCs more vulnerable to radiation-induced injury. Then bone radiation injury was induced in C57BL6J mice to further determine the radio-protective effects of Rspo1. In mice, administration of Rspo1 recombinant protein alleviated radiation-induced bone loss. Our results uncover that Rspo1-LGR4-mTOR-autophagy axis are key mechanisms by which BMSCs initiate self-defense against radiation and maintain bone homeostasis. Targeting Rspo1-LGR4 may provide a novel strategy for the intervention of radiation-induced bone damage.


Subject(s)
Autophagy , Bone Diseases/prevention & control , Mesenchymal Stem Cells/enzymology , Radiation Injuries/prevention & control , Receptors, G-Protein-Coupled/metabolism , TOR Serine-Threonine Kinases/metabolism , Thrombospondins/metabolism , Animals , Autophagy/radiation effects , Bone Diseases/enzymology , Bone Diseases/genetics , Bone Diseases/pathology , Cell Differentiation , Cell Proliferation , Cell Self Renewal , Cells, Cultured , DNA Damage , Disease Models, Animal , Mesenchymal Stem Cells/pathology , Mesenchymal Stem Cells/radiation effects , Mice, Inbred C57BL , Osteogenesis , Radiation Injuries/enzymology , Radiation Injuries/genetics , Radiation Injuries/pathology , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Thrombospondins/genetics
17.
Int J Mol Med ; 47(1): 161-170, 2021 01.
Article in English | MEDLINE | ID: mdl-33416107

ABSTRACT

The mitochondria have been proven to be involved in processes of aging; however, the mechansims through which mitoepigenetics affect the cytological behaviors of cardiomyocytes during the aging process are not yet fully understood. In the present study, two senescence models were constructed, replicative senescence (RS) and stress­induced premature senescence (SIPS), using human heart mesenchymal stem cells (HMSCs). First, the differences in age­related gene expression levels and telomere length were compared between the HMSCs in the RS and SIPS models by PCR. Subsequently, protein expression and the mitochondrial DNA (mtDNA) methylation status of cytochrome c oxidase subunit II (COX2) was measured by western blot analysis and bisulfite genomic sequencing (BSP). Finally, the value of the DNA methyltransferase (Dnmt) inhibitor, 5­aza­2'­deoxycytidine (AdC), in delaying the senescence of HMSCs was evaluated. It was found that the p16, p27 and p53 mRNA expression levels increased in the senescent cells, whereas p21 mRNA expression did not. It was also found that telomere shortening only occurred in the RS model, but not in the SIPS model. Along with the senescence of HMSCs, COX2 gene methylation increased and its protein expression level significantly decreased. It was demonstrated that AdC inhibited COX2 methylation and downregulated COX2 expression. The addition of exogenous COX2 or the administration of AdC promoted cell proliferation and delayed cell aging. On the whole, the present study demonstrates that COX2 methylation and downregulation are biomarkers of HMSC senescence. Thus, COX2 may have potential for use as a therapeutic target of cardiovascular diseases and this warrants further investigation.


Subject(s)
Cellular Senescence , DNA Methylation , DNA, Mitochondrial/metabolism , Down-Regulation , Electron Transport Complex IV/biosynthesis , Gene Expression Regulation, Enzymologic , Mesenchymal Stem Cells/enzymology , Mitochondria, Heart/enzymology , Myocardium/enzymology , DNA, Mitochondrial/genetics , Electron Transport Complex IV/genetics , Humans , Mitochondria, Heart/genetics
18.
Chem Commun (Camb) ; 57(9): 1145-1148, 2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33411866

ABSTRACT

The ability to custom-modify cell surface glycans holds great promise for treatment of a variety of diseases. We propose a glycomimetic of l-fucose that markedly inhibits the creation of sLeX by FTVI and FTVII, but has no effect on creation of LeX by FTIX. Our findings thus indicate that selective suppression of sLex display can be achieved, and STD-NMR studies surprisingly reveal that the mimetic does not compete with GDP-fucose at the enzymatic binding site.


Subject(s)
Fucose/analogs & derivatives , Fucose/pharmacology , Fucosyltransferases/antagonists & inhibitors , Cell Line, Tumor , Fucose/chemistry , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma
19.
Life Sci ; 267: 118929, 2021 Feb 15.
Article in English | MEDLINE | ID: mdl-33359244

ABSTRACT

OBJECTIVE: To investigate if the modification of human adipose-derived mesenchymal stem cells (hADSCs) by the antioxidants superoxide dismutase 2 (Sod2) and catalase (Cat) can attenuate the pathological conditions of intervertebral disc degeneration (IVD). METHODS: In vitro, MTT assay and qRT-PCR was used to detect cell proliferation and gene expressions in hADSCs transduced with Ad-null (an adenovirus vector containing no transgene expression cassette), Ad-Sod2 (recombinant adenovirus Sod2) and Ad-Cat. IVD mouse models were generated by needle puncture and treated with hADSCs with/without Ad-null/Ad-Sod2/Ad-Cat. X-ray evaluation, magnetic resonance imaging (MRI) analysis, histological analysis, immunohistochemistry, Western blots, ELISAs and qRT-PCR were performed. RESULTS: hADSCs transduced with Ad-Sod2 and Ad-Cat showed enhanced cell proliferation with the upregulation of SOX9, ACAN, and COL2. In vivo, IVD mice injected with hADSCs showed increased disc height index, MRI index and mean T2 intensities, as well as the attenuated histologic grading of the annulus fibrosus (AF) and NP accompanied by the upregulation of GAG and COL2, which were further improved in the Ad-Sod2 hADSC + IVD and Ad-Cat hADSC + IVD groups. Furthermore, the increased expression of IL-1ß, IL-6 and TNF-α was reduced in IVD mice injected with hADSCs. Compared with the hADSC + IVD group, the Ad-Sod2 hADSC/Ad-Cat hADSC + IVD groups had lower expression of pro-inflammatory factors. CONCLUSION: Modification of hADSCs by the antioxidants Sod2 and Cat improved the pathological condition of intervertebral disc tissues with increased GAG and COL2 expression, as well as reduced inflammation, thereby demonstrating a therapeutic effect in IVD.


Subject(s)
Catalase/metabolism , Intervertebral Disc Degeneration/therapy , Superoxide Dismutase/metabolism , Animals , Catalase/physiology , Cell Proliferation/physiology , Disease Models, Animal , Humans , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/enzymology , Intervertebral Disc Degeneration/pathology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/pathology , Mice , Random Allocation , Superoxide Dismutase/physiology , Tumor Necrosis Factor-alpha/metabolism
20.
Biomed Pharmacother ; 133: 111048, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378955

ABSTRACT

Subarachnoid hemorrhage (SAH) is an acute and severe disease with high disability and mortality. Inflammatory reactions have been proven to occur throughout SAH. Extracellular vesicles derived from mesenchymal stem cells (MSCs-EVs) have shown broad potential for the treatment of brain dysfunction and neuroprotective effects through neurogenesis and angiogenesis after stroke. However, the mechanisms of EVs in neuroinflammation during the acute phase of SAH are not well known. Our present study was designed to investigate the effects of MSCs-EVs on neuroinflammation and the polarization regulation of microglia to the M2 phenotype and related signaling pathways after SAH in rats. The SAH model was induced by an improved method of intravascular perforation, and MSCs-EVs were injected via the tail vein. Post-SAH assessments included neurobehavioral tests as well as brain water content, immunohistochemistry, PCR and Western blot analyses. Our results showed that MSCs-EVs alleviated the expression of inflammatory cytokines in the parietal cortex and hippocampus 24 h and 48 h after SAH and that MSCs-EVs inhibited NF-κB and activated AMPK to reduce inflammation after SAH. Furthermore, MSC-EVs regulated the polarization of microglia toward the M2 phenotype by downregulating interleukin-1ß, cluster of differentiation 16, cluster of differentiation 11b, and inducible nitric oxide synthase and upregulating the expression of cluster of differentiation 206 and arginase-1. Additionally, MSCs-EVs inhibited the neuroinflammatory response and had neuroprotective effects in the brain tissues of rats after SAH. This study may support their use as a potential treatment strategy for early SAH in the future.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Brain/enzymology , Extracellular Vesicles/transplantation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Microglia/enzymology , NF-kappa B/metabolism , Subarachnoid Hemorrhage/surgery , Animals , Brain/pathology , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Extracellular Vesicles/enzymology , Male , Mesenchymal Stem Cells/enzymology , Microglia/pathology , Phenotype , Phosphorylation , Rats, Sprague-Dawley , Signal Transduction , Subarachnoid Hemorrhage/enzymology , Subarachnoid Hemorrhage/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...