Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 8 de 8
1.
Biotechnol J ; 19(4): e2300714, 2024 Apr.
Article En | MEDLINE | ID: mdl-38622793

Natural bone tissue features a complex mechanical environment, with cells responding to diverse mechanical stimuli, including fluid shear stress (FSS) and hydrostatic pressure (HP). However, current in vitro experiments commonly employ a singular mechanical stimulus to simulate the mechanical environment in vivo. The understanding of the combined effects and mechanisms of multiple mechanical stimuli remains limited. Hence, this study constructed a mechanical stimulation device capable of simultaneously applying FSS and HP to cells. This study investigated the impact of FSS and HP on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and examined the distinctions and interactions between the two mechanisms. The results demonstrated that both FSS and HP individually enhanced the osteogenic differentiation of BMSCs, with a more pronounced effect observed through their combined application. BMSCs responded to external FSS and HP stimulation through the integrin-cytoskeleton and Piezo1 ion channel respectively. This led to the activation of downstream biochemical signals, resulting in the dephosphorylation and nuclear translocation of the intracellular transcription factors Yes Associated Protein 1 (YAP1) and nuclear factor of activated T cells 2 (NFAT2). Activated YAP1 could bind to NFAT2 to enhance transcriptional activity, thereby promoting osteogenic differentiation of BMSCs more effectively. This study highlights the significance of composite mechanical stimulation in BMSCs' osteogenic differentiation, offering guidance for establishing a complex mechanical environment for in vitro functional bone tissue construction.


Mesenchymal Stem Cells , Osteogenesis , Osteogenesis/physiology , Hydrostatic Pressure , Cell Differentiation/physiology , Transcription Factors/metabolism , Cells, Cultured , Bone Marrow Cells
2.
Anal Chem ; 95(17): 7036-7044, 2023 May 02.
Article En | MEDLINE | ID: mdl-37088925

The development of superior probes is highly desirable and valuable for viscosity measurement. Herein, we designed and reported a series of diphenylbenzofulvene (DPBF)-based organic luminophores according to the molecular regulation strategy. There are two free-rotating phenyl groups attached to the rigid fluorene skeleton in the DPBF, enabling its unique propeller-like noncoplanar chemical structure. Benefiting from this, DPBFs could feature outstanding PL and ECL emissions with intriguing aggregation-induced characteristics. Experimental and theoretical investigations revealed that substituent, spatial structure, and molecular orbital energy profoundly affected their luminescent behaviors. It was disclosed that fluoro-substituted DPBF(F)2 with a smaller LUMO-HOMO band gap demonstrated the strongest ECL emission and was selected as the optimal ECL emitter. Finally, DPBF(F)2 featured a linear response to the viscosity and VC content with lower limits of detection (LOD) of 5.69 µcP and 38.2 nM, respectively. This study represents the first example of the ECL probe toward viscosity and will be of great significance for both ECL application and viscosity measurement.

3.
Mol Biol Rep ; 49(4): 2723-2733, 2022 Apr.
Article En | MEDLINE | ID: mdl-35037196

BACKGROUND: Mesenchymal stem cells have been widely used in the treatment of diabetes mellitus. However, hyperglycemia associated with DM promotes cell apoptosis and affects osteogenic differentiation of MSCs in varying degrees, leading to osteoporosis in DM patients. Therefore, in this paper, the effect of high glucose on apoptosis and osteogenesis of MSCs was investigated and underlying mechanism was further determined. METHODS AND RESULTS: Intracellular ROS levels were determined using probe DCFH-DA. MMP was detected using JC-1 staining. Cell apoptosis was detected using Annexin V-FITC/PI and Flow Cytometer. The expression of genes and protein was detected by qRT-PCR and Western blot respectively. The results showed high glucose induced MSC apoptosis but promoted its osteogenesis. Western blot analysis revealed that high glucose downregulated AKT-Sirt1-TWIST pathway. Activation of Sirt1 via SRT1720 increased TWIST expression, alleviated MSC apoptosis and promoted osteogenesis of MSCs. TWIST knockdown studies demonstrated that inhibition of TWIST intensified high glucose-induced apoptosis but promoted osteogenesis differentiation of MSCs. TWIST is likely to be a new regulator for cross talk between Sirt1 and its downstream targets. CONCLUSION: Our data demonstrates that high glucose induces MSC apoptosis and enhances osteogenesis differentiation via downregulation of AKT-Sirt1-TWIST.


Osteogenesis , Sirtuin 1 , Apoptosis , Cell Differentiation , Down-Regulation , Glucose/pharmacology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
4.
Biotechnol Appl Biochem ; 69(5): 2138-2150, 2022 Oct.
Article En | MEDLINE | ID: mdl-34694656

The absence of blood vessels in tissue engineered bone often leads to necrosis of internal cells after implantation, ultimately affecting the process of bone repair. Herein, mesenchymal stem cells (MSCs) and human umbilical vein endothelial cells (HUVECs) were cocultured to induce osteogenesis and angiogenesis. Based on the findings, the number of HUVECs in the coculture system increased in the growth medium group, but decreased in the osteogenic induction medium (OIM) group. Considering that the paracrine effects of MSCs had changed, we tested the genes expression of osteogenically differentiated MSCs. The expression of osteogenic genes in MSCs increased during osteogenesis. Further, the expression levels of pigment epithelial-derived factor (PEDF) gene and protein, an antivascular factor, were also increased. To verify whether MSCs promote HUVECs apoptosis via PEDF, PEDF was silenced via siRNA. The conditioned medium of differentiated MSCs with PEDF silencing significantly improved the proliferation and apoptosis of HUVECs. Based on further experiments, PEDF mediated the apoptosis and proliferation of HUVECs through p53, BAX/BCL-2, FAS, and c-Caspase-3. However, when PEDF was silenced with siRNA, the osteogenic potential of MSCs was affected. The results of this study provide a theoretical basis for the construction of prevascularized bone tissues in vitro.


Mesenchymal Stem Cells , Humans , Human Umbilical Vein Endothelial Cells , RNA, Small Interfering/metabolism , Mesenchymal Stem Cells/metabolism , Cell Differentiation , Coculture Techniques , Apoptosis , Cells, Cultured , Neovascularization, Physiologic
5.
Biotechnol J ; 17(1): e2100096, 2022 Jan.
Article En | MEDLINE | ID: mdl-34378873

BACKGROUND: Bioartificial livers (BALs) are emerging as a potential supportive therapy for liver diseases. However, the maintenance of hepatocyte function and viability in vitro is a major challenge. Mesenchymal stem cells (MSCs) have attracted extensive attention for providing trophic support to hepatocytes, but only few studies have explored the interaction between human MSCs and human hepatocytes, and very little is known about the underlying molecular mechanisms whereby MSCs affect hepatocyte function, especially in serum-free medium (SFM). METHOD AND RESULTS: This study aims to explore the effects of human umbilical cord-derived MSCs (hUMSCs) on human-induced hepatocytes (hiHeps) function and viability, and know about the underlying molecular mechanism of interaction in SFM. The liver-specific function of hiHeps was evaluated by analysis of albumin secretion, urea synthesis, and metabolic enzyme activity. hiHeps apoptosis was mainly characterized by live/dead staining assay, JC-1 mitochondrial membrane potential assay, reactive oxygen species (ROS) generation, and cell apoptosis detection. The expression of related genes and proteins were measured by qRT-PCR and western blotting. The results indicate that co-culture with hUMSCs improved hiHep urea synthesis and reduced cell apoptosis compared to monoculture in SFM, and this effect was found to be mediated by secreted interleukin-6 (IL-6). Further, studies revealed that IL-6 reduced hiHep apoptosis via the activation of the JAK-Stat3-Ref-1 and JAK-Stat3-Bcl-2/Bax-Caspase3 pathways by binding to the IL-6 receptor. IL-6 also enhanced hiHep urea synthesis through the JAK-Akt-P53-ARG1 pathway. Finally, hiHep-specific functions were further prolonged and increased when co-cultured with hUMSCs on 3D polyethylene terephthalate (PET) fibrous scaffolds. CONCLUSION: The SFM co-culture strategy showed major advantages in maintaining hiHep function and viability in vitro, which is of great significance for the clinical application of hiHeps in BALs.


Interleukin-6 , Mesenchymal Stem Cells , Apoptosis , Coculture Techniques , Hepatocytes , Humans , Umbilical Cord , Urea
6.
Turk J Biol ; 45(6): 683-694, 2021.
Article En | MEDLINE | ID: mdl-35068949

Prevascularization and mechanical stimulation have been reported as effective methods for the construction of functional bone tissue. However, their combined effects on osteogenic differentiation and its mechanism remain to be explored. Here, the effects of fluid shear stress (FSS) on osteogenic differentiation of rat bone-marrow-derived mesenchymal stem cells (BMSCs) when cocultured with human umbilical vein endothelial cells (HUVECs) were investigated, and underlying signaling mechanisms were further explored. FSS stimulation for 1-4 h/day increased alkaline phosphatase (ALP) activity and calcium deposition in coculture systems and promoted the proliferation of cocultured cells. FSS stimulation for 2 h/day was selected as the optimized protocol according to osteogenesis in the coculture. In this situation, the mRNA levels of ALP, runt-related transcriptional factor 2 (Runx2) and osteocalcin (OCN), and protein levels of OCN and osteopontin (OPN) in BMSCs were upregulated. Furthermore, FSS and coculture with HUVECs synergistically increased integrin ß1 expression in BMSCs and further activated focal adhesion kinases (FAKs) and downstream extracellular signal-related kinase (ERK), leading to the enhancement of Runx2 expression. Blocking the phosphorylation of FAK abrogated FSS-induced ERK phosphorylation and inhibited osteogenesis of cocultured BMSCs. These results revealed that FSS and coculture with HUVECs synergistically promotes the osteogenesis of BMSCs, which was mediated by the integrin ß1-FAK-ERK signaling pathway.

7.
Stem Cell Res Ther ; 11(1): 343, 2020 08 06.
Article En | MEDLINE | ID: mdl-32762747

BACKGROUND AND AIM: Inadequate vascularization is a challenge in bone tissue engineering because internal cells are prone to necrosis due to a lack of nutrient supply. Rat bone marrow-derived mesenchymal stem cells (rBMSCs) and human umbilical vein endothelial cells (HUVECs) were cocultured to construct prevascularized bone tissue in osteogenic induction medium (OIM) in vitro. The angiogenic capacity of HUVECs was limited in the coculture system. In this study, the effects of the components in the medium on HUVEC angiogenesis were analyzed. METHODS: The coculture system was established in OIM. Alizarin red staining and alkaline phosphatase staining were used to assess the osteogenic ability of MSCs. A Matrigel tube assay was used to assess the angiogenic ability of HUVECs in vitro. The proliferation of HUVECs was evaluated by cell counting and CCK-8 assays, and migration was evaluated by the streaked plate assay. The expression levels of angiogenesis-associated genes and proteins in HUVECs were measured by qRT-PCR and Western blotting, respectively. RESULTS: Dexamethasone in the OIM suppressed the proliferation and migration of HUVECs, inhibiting the formation of capillary-like structures. Our research showed that dexamethasone stimulated HUVECs to secrete tissue inhibitor of metalloproteinase (TIMP-3), which competed with vascular endothelial growth factor (VEGF-A) to bind to vascular endothelial growth factor receptor 2 (VEGFR2, KDR). This effect was related to inhibiting the phosphorylation of ERK and AKT, which are two downstream targets of KDR. However, under hypoxia, the enhanced expression of hypoxia-inducible factor-1α (HIF-1α) decreased the expression of TIMP-3 and promoted the phosphorylation of KDR, improving HUVEC angiogenesis in the coculture system. CONCLUSION: Coculture of hypoxia-preconditioned HUVECs and MSCs showed robust angiogenesis and osteogenesis in OIM, which has important implications for prevascularization in bone tissue engineering in the future.


Human Umbilical Vein Endothelial Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Hypoxia , Vascular Endothelial Growth Factor A , Animals , Coculture Techniques , Dexamethasone/pharmacology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neovascularization, Pathologic , Neovascularization, Physiologic , Rats , Vascular Endothelial Growth Factor A/genetics
8.
Dig Dis Sci ; 65(7): 2009-2023, 2020 07.
Article En | MEDLINE | ID: mdl-31722057

BACKGROUND AND AIMS: Bioartificial livers (BALs) have attracted much attention as potential supportive therapies for liver diseases. A serum-free microcarrier culture strategy for the in vitro high-density expansion of human-induced hepatocyte-like cells (hiHeps) suitable for BALs was studied in this article. METHODS: hiHeps were transdifferentiated from human fibroblasts by the lentiviral overexpression of FOXA3, HNF1A, and HNF4A. Cells were cultured on microcarriers, their proliferation was evaluated by cell count and CCK-8 assays, and their function was evaluated by detecting liver function parameters in the supernatant, including urea secretion, albumin synthesis, and lactate dehydrogenase levels. The expressions of hepatocyte function-associated genes of hiHeps were measured by qRT-PCR in 2D and 3D conditions. The expression of related proteins during fibronectin promotes cell adhesion, and proliferation on microcarrier was detected by western blotting. RESULTS: During microcarrier culture, the optimal culture conditions during the adherence period were the use of half-volume high-density inoculation, Cytodex 3 at a concentration of 3 mg/mL, a cell seeding density of 2.0 × 105 cells/mL, and a stirring speed of 45 rpm. The final cell density in self-developed, chemically defined serum-free medium (SFM) reached 2.53 × 106 cells/mL, and the maximum increase in expansion was 12.61-fold. In addition, we found that fibronectin (FN) can promote hiHep attachment and proliferation on Cytodex 3 microcarriers and that this pro-proliferative effect was mediated by the integrin-ß1/FAK/ERK/CyclinD1 signaling pathway. Finally, the growth and function of hiHeps on Cytodex 3 in SFM were close to those of hiHeps on Cytodex 3 in hepatocyte maintenance medium (HMM), and cells maintained their morphology and function after harvest on microcarriers. CONCLUSIONS: Serum-free microcarrier culture has important implications for the expansion of a sufficient number of hiHeps prior to the clinical application of BALs.


Cell Culture Techniques/methods , Cell Proliferation , Cell Transdifferentiation , Hepatocytes/cytology , Liver, Artificial , Albumins/biosynthesis , Cell Adhesion , Cellular Reprogramming Techniques/methods , Culture Media, Serum-Free , Cyclin D1/metabolism , Dextrans , Fibroblasts/cytology , Fibroblasts/metabolism , Fibronectins/metabolism , Focal Adhesion Kinase 1/metabolism , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 3-gamma/genetics , Hepatocyte Nuclear Factor 4/genetics , Hepatocytes/metabolism , Hepatocytes/physiology , Humans , Integrin beta1/metabolism , L-Lactate Dehydrogenase/metabolism , MAP Kinase Signaling System , Reverse Transcriptase Polymerase Chain Reaction , Urea/metabolism
...