Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 36
Filtrer
1.
Reprod Sci ; 28(10): 2906-2915, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-33959891

RÉSUMÉ

The self-renewal of spermatogonial cells (SCs) provides the foundation for life-long spermatogenesis. To date, only a few growth factors have been used for the culture of SCs in vitro, and how to enhance proliferation capacity of SCs in vitro needs further research. This study aimed to explore the effects of periostin (POSTN) on the proliferation of human SCs. GC-1 spg cells were cultured in a medium with POSTN, cell proliferation was evaluated by MTS analysis and EdU assay, and the Wnt/ß-catenin signaling pathway was examined. Thereafter, the proliferations of human SC were detected using immunofluorescence and RT-PCR. In this study, we found that CM secreted by human amniotic mesenchymal stem cells (hAMSCs) could enhance the proliferation capacity of mouse GC-1 spg cells. Label-free mass spectrometry and ELISA analysis demonstrated that high level of POSTN was secreted by hAMSCs. MTS and EdU staining showed that POSTN increased GC-1 spg cell proliferation, whereas CM from POSTN-silenced hAMSCs suppressed cell proliferation capacity. Then POSTN was found to activate the Wnt/ß-catenin signaling pathway to regulate the proliferation of GC-1 spg cells. XAV-939, a Wnt/ß-catenin inhibitor, partially reversed the effects of POSTN on GC-1 spg cell proliferation. We then analyzed human SCs and found that POSTN promoted human SC proliferation in vitro. These findings provide insights regarding the role of POSTN in regulating SC proliferation via the Wnt/ß-catenin signaling pathway and suggest that POSTN may serve as a cytokine for male infertility therapy.


Sujet(s)
Molécules d'adhérence cellulaire/pharmacologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Spermatogonies/effets des médicaments et des substances chimiques , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire , Prolifération cellulaire/physiologie , Relation dose-effet des médicaments , Humains , Mâle , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Souris , Spermatogonies/métabolisme , Voie de signalisation Wnt/physiologie
2.
Aging (Albany NY) ; 12(13): 12960-12986, 2020 07 04.
Article de Anglais | MEDLINE | ID: mdl-32621591

RÉSUMÉ

In this study, we identified wound healing-related proteins secreted by human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stem cells (hAMSCs). We observed increased migration and reduced proliferation and differentiation when keratinocytes were co-cultured in media conditioned by hAECs (hAECs-CM) and hAMSCs (hAMSCs-CM). Label-free mass spectrometry and bioinformatic analyses of the hAECs-CM and hAMSCs-CM proteome revealed several proteins associated with wound healing, angiogenesis, cellular differentiation, immune response and cell motility. The levels of the proteins related to wound healing, including CTHRC1, LOXL2 and LGALS1, were significantly higher in hAMSCs-CM than hAECs-CM. LOXL2 significantly enhanced in vitro keratinocyte migration and differentiation compared to CTHRC1 and LGALS1. Moreover, LOXL2 enhanced keratinocyte migration and differentiation by activating the JNK signaling pathway. We observed significant reduction in the in vitro migration and differentiation of keratinocytes when co-cultured with medium conditioned by LOXL2-silenced hAMSCs and when treated with 10 µM SP600125, a specific JNK inhibitor. Treatment with hAMSCs-CM and LOXL2 significantly accelerated wound healing in the murine skin wound model. These findings show that LOXL2 promotes wound healing by inducing keratinocyte migration and differentiation via a JNK signaling pathway.


Sujet(s)
Amino-acid oxidoreductases/métabolisme , Différenciation cellulaire/physiologie , Kératinocytes/physiologie , Cellules souches mésenchymateuses/cytologie , Réépithélialisation/physiologie , Amnios/cytologie , Animaux , Mouvement cellulaire/physiologie , Humains , Kératinocytes/cytologie , Système de signalisation des MAP kinases/physiologie , Mâle , Souris
3.
J Biomed Mater Res A ; 108(9): 1824-1833, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32388898

RÉSUMÉ

Titanium (Ti) and Ti-based alloys are widely used in the manufacture of dental and orthopedic implants. However, how to improve their osteogenic differentiation ability is still a key issue to be resolved. In this study, gradient nanostructured surface (GNS) samples were prepared by surface mechanical grinding treatment, and coarse-grained (CG) samples were obtained by recrystallization annealing, making sure that the two kinds of specimens had similar roughness. Then, human amniotic mesenchymal stem cells (hAMSCs) were cocultured with the two kinds of Ti to investigate the material effects on the cellular functions. The results demonstrated that the grains with size ~56 nm were formed on the surface of the GNS Ti, and the grain size gradually increases from the sample surface to interior. Compared to the CG samples, the GNS ones could make the adhesion effect of the hAMSCs better, and promote the cell proliferation and osteogenic differentiation more significantly, the preliminary mechanism of which might be due to their specific nanostructure, the thicker oxide layer formed on their surface and the enhanced hardness. Our results indicated that the gradient nanostructured Ti materials could enhance both osteogenic differentiation and mechanical properties, which may possess broader applications in bone tissue engineering and clinical implanting.


Sujet(s)
Matériaux biocompatibles/composition chimique , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Nanostructures/composition chimique , Ostéogenèse/effets des médicaments et des substances chimiques , Titane/composition chimique , Amnios/cytologie , Matériaux biocompatibles/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Humains , Cellules souches mésenchymateuses/cytologie , Propriétés de surface , Titane/pharmacologie
4.
Biomed Res Int ; 2020: 8150780, 2020.
Article de Anglais | MEDLINE | ID: mdl-32190682

RÉSUMÉ

Mesenchymal stem cells (MSCs) have been a potential strategy in the pretreatment of pulmonary diseases, while the mechanisms of MSCs-conditioned medium (MSCs-CM) involved with microRNAs on the regulation of lung ion transport are seldom reported. We investigated the role of miR-124-5p in lipopolysaccharide-involved epithelial sodium channel (ENaC) dysfunction and explored the potential target of miR-124-5p. We observed the lower expression of miR-124-5p after the administration of MSCs-CM, and the overexpression or inhibition of miR-124-5p regulated epithelial sodium channel α-subunit (α-ENaC) expression at protein levels in mouse alveolar type 2 epithelial (AT2) cells. We confirmed that α-ENaC is one of the target genes of miR-124-5p through dual luciferase assay and Ussing chamber assay revealed that miR-124-5p inhibited amiloride-sensitive currents associated with ENaC activity in intact H441 monolayers. Our results demonstrate that miR-124-5p can decrease the expression and function of α-ENaC in alveolar epithelial cells by targeting the 3'-UTR. The involvement of MSCs-CM in lipopolysaccharide-induced acute lung injury cell model could be related to the downregulation of miR-124-5p on α-ENaC, which may provide a new target for the treatment of acute lung injury.


Sujet(s)
Pneumocytes/effets des médicaments et des substances chimiques , Pneumocytes/métabolisme , Bloqueurs de canaux sodiques épithéliaux/pharmacologie , Canaux sodium épithéliaux/biosynthèse , Lipopolysaccharides/pharmacologie , microARN/métabolisme , Régions 3' non traduites , Lésion pulmonaire aigüe/métabolisme , Amiloride/pharmacologie , Animaux , Milieux de culture conditionnés , Canaux sodium épithéliaux/génétique , Canaux sodium épithéliaux/métabolisme , Transport des ions , Mâle , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Souris
5.
Biomaterials ; 240: 119832, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-32113114

RÉSUMÉ

The small intestine (SI) is difficult to regenerate or reconstruct due to its complex structure and functions. Recent developments in stem cell research, advanced engineering technologies, and regenerative medicine strategies bring new hope of solving clinical problems of the SI. This review will first summarize the structure, function, development, cell types, and matrix components of the SI. Then, the major cell sources for SI regeneration are introduced, and state-of-the-art biofabrication technologies for generating engineered SI tissues or models are overviewed. Furthermore, in vitro models and in vivo transplantation, based on intestinal organoids and tissue engineering, are highlighted. Finally, current challenges and future perspectives are discussed to help direct future applications for SI repair and regeneration.


Sujet(s)
Bio-impression , Intestin grêle , Régénération , Ingénierie tissulaire , Structures d'échafaudage tissulaires
6.
Mol Ther Nucleic Acids ; 19: 1110-1122, 2020 Mar 06.
Article de Anglais | MEDLINE | ID: mdl-32059337

RÉSUMÉ

MicroRNAs (miRNAs) that play key roles in the generation of insulin-producing cells from stem cells provide a cell-based approach for insulin replacement therapy. In this study, we used next-generation sequencing to detect the miRNA expression profile of normal mouse pancreatic ß cells, non-ß cells, bone marrow mesenchymal stem cells (BM-MSCs), and adipose-derived stem cells (ADSCs) and determined relative miRNA expression levels in mouse pancreatic ß cells. After the novel mouse miRNA candidates were identified using miRDeep 2.0, we found that Chr13_novelMiR7354-5p, a novel miRNA candidate, significantly promoted the differentiation of BM-MSCs into insulin-producing cells in vitro. Furthermore, Chr13_novelMiR7354-5p-transfected BM-MSCs reversed hyperglycemia in streptozotocin (STZ)-treated diabetic mice. In addition, bioinformatics analyses, a luciferase reporter assay, and western blotting demonstrated that Chr13_novelMiR7354-5p targeted Notch1 and Rbpj. Our results provide compelling evidence of the existence of 65 novel mouse miRNA candidates and present a new treatment strategy to generate insulin-producing cells from stem cells.

7.
Int J Mol Med ; 44(5): 1629-1640, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31545472

RÉSUMÉ

Stem cells derived from human amniotic membrane (hAM) are promising targets in regenerative medicine. A previous study focused on human amniotic stem cells in skin wound and scar­free healing. The present study aimed to investigate whether hydrogen peroxide (H2O2)­induced senescence of human dermal fibroblasts (hDFs) was influenced by the anti­aging effect of conditioned medium (CdM) derived from human amniotic stem cells. First, the biological function of two types of amniotic stem cells, namely human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stem cells (hAMSCs), on hDFs was compared. The results of cell proliferation and wound healing assays showed that CdM promoted cell proliferation and migration. In addition, CdM from hAECs and hAMSCs significantly promoted proliferation of senescent hDFs induced by H2O2. These results indicated that CdM protects cells from damage caused by H2O2. Treatment with CdM decreased senescence­associated ß­galactosidase activity and improved the entry of proliferating cells into the S phase. Simultaneously, it was found that CdM increased the activity of superoxide dismutase and catalase and decreased malondialdehyde by reducing H2O2­induced intracellular reactive oxygen species production. It was found that CdM downregulated H2O2­stimulated 8­hydroxydeoxyguanosine and γ­H2AX levels and decreased the expression of the senescence­associated proteins p21 and p16. In conclusion, the findings indicated that the paracrine effects derived from human amniotic stem cells aided delaying oxidative stress­induced premature senescence.


Sujet(s)
Amnios/métabolisme , Vieillissement de la cellule/physiologie , Milieux de culture conditionnés/métabolisme , Derme/métabolisme , Fibroblastes/métabolisme , Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Cellules cultivées , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Derme/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Humains , Peroxyde d'hydrogène/pharmacologie , Malonaldéhyde/métabolisme , Cellules souches mésenchymateuses/métabolisme , Espèces réactives de l'oxygène/métabolisme , Phase S/physiologie , Peau/effets des médicaments et des substances chimiques , Peau/métabolisme , Superoxide dismutase/métabolisme , Cicatrisation de plaie/physiologie
8.
Int J Mol Med ; 44(4): 1425-1435, 2019 Oct.
Article de Anglais | MEDLINE | ID: mdl-31364743

RÉSUMÉ

Mesenchymal stem cells (MSCs) show immunosuppressive activities and alleviate atherosclerosis (AS) formation in apolipoprotein E­knockout (apoE­KO) mice. Human amnion mesenchymal stem cells (hAMSCs), a particular population of mesenchymal stem cells, have been shown to have immunomodulatory abilities. The present study investigated the effects of hAMSCs treatment on early atherosclerotic plaque formation and the progression of established lesion in apoE­KO mice. In total, 36 mice were fed with a high­fat diet. Mice were subjected to hAMSCs­injection treatment simultaneously with high­fat diet (early treatment) or after 8 weeks of high­fat diet (delayed treatment). In each treatment, mice were divided into three groups: i) hAMSCs group with hAMSCs treatment; ii) PBS group injected with PBS; and iii) control group without injection. Histological results showed that the plaque area in the aortic arch of mice was significantly reduced after hAMSCs treatment in the early and delayed treatment groups. In addition, immunohistochemical analysis suggested that the accumulation of macrophages was significantly decreased after hAMSCs treatment. Similarly, the release of the pro­inflammatory cytokine tumor necrosis factor­α was also decreased, whereas the release of the anti­inflammatory cytokine interleukin­10 was increased. In addition, hAMSCs treatment suppressed the phosphorylation of p65 and inhibitor of κB­α, suggesting that NF­κB pathway was involved in the hAMSCs­mediated suppression of immune response. In conclusion, hAMSCs treatment was effective in reducing immune response, which is the one of the major causes of AS, eventually leading to a significant reduction in size of atherosclerotic lesions.


Sujet(s)
Amnios/cytologie , Athérosclérose/métabolisme , Communication cellulaire , Immunomodulation , Macrophages/immunologie , Macrophages/métabolisme , Cellules souches mésenchymateuses/métabolisme , Animaux , Athérosclérose/étiologie , Athérosclérose/anatomopathologie , Athérosclérose/thérapie , Marqueurs biologiques , Différenciation cellulaire , Cellules cultivées , Cytokines/génétique , Cytokines/métabolisme , Modèles animaux de maladie humaine , Expression des gènes , Humains , Immunophénotypage , Lipides/sang , Mâle , Transplantation de cellules souches mésenchymateuses , Cellules souches mésenchymateuses/cytologie , Souris , Plaque d'athérosclérose/métabolisme , Plaque d'athérosclérose/anatomopathologie , Facteur de transcription RelA/métabolisme
9.
Ann Clin Lab Sci ; 49(1): 63-71, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30814079

RÉSUMÉ

This study aimed to evaluate the use of Insulin-Transferrin-Selenium (ITS) medium in place of fetal bovine serum (FBS) to culture human amnion mesenchymal stem cells (hAMSCs). Cell morphology, ultrastructure, proliferation, migration and MSC related markers were assessed accordingly. The hAMSCs were induced to osteocyte, chondrocyte, adipocyte and keratinocyte by culturing in appropriate induction medium. hAMSCs mRNA expression was detected for the matrix metalloproteinases 2 (MMP2), keratinocyte growth factor (KGF), vascular endothelial growth factor (VEGF), insulin-like growth factor-I (IGF-I), Platelet-derived Growth Factor (PDGF), and transforming growth factor beta 1 (TGF-ß) by real-time quantitative RT-PCR. Our results showed that hAMSCs cultured in ITS medium exhibited similar proliferation rates, demonstrated a statistically significant increased migration and expressed similar levels of MSC markers(CD73+, CD90+, CD105+, CD45-, CD34-) compared with those cultured in FBS. Osteoblasts, chondrocytes, adipocytes and keratinocytes were differentiated. Results of transmission electron microscope (TEM) revealed that hAMSCs cultured in ITS medium underwent active metabolism. The mRNA expression of MMP2, VEGF, KGF, TGF-ß, IGF-I and PDGF upregulated in ITS medium. In conclusion, ITS medium has the potential to be used for the expansion of hAMSCs before clinical application.


Sujet(s)
Adipocytes/cytologie , Amnios/cytologie , Chondrocytes/cytologie , Milieux de culture/pharmacologie , Kératinocytes/cytologie , Cellules souches mésenchymateuses/cytologie , Ostéocytes/cytologie , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Amnios/effets des médicaments et des substances chimiques , Amnios/métabolisme , Antioxydants/pharmacologie , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Chondrocytes/effets des médicaments et des substances chimiques , Chondrocytes/métabolisme , Humains , Hypoglycémiants/pharmacologie , Insuline/pharmacologie , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/métabolisme , Ostéocytes/effets des médicaments et des substances chimiques , Ostéocytes/métabolisme , Sélénium/pharmacologie , Transduction du signal , Transferrine/pharmacologie
10.
Mol Ther Nucleic Acids ; 14: 410-423, 2019 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-30731322

RÉSUMÉ

Early in gestation, wounds in fetal skin heal by regeneration, in which microRNAs play key roles. Seq-915_x4024 is a novel microRNA candidate confirmed by deep sequencing and mirTools 2.0. It is highly expressed in fetal keratinocytes during early gestation. Using an in vitro wound-healing assay, Transwell cell migration assay, and MTS proliferation assay, we demonstrated that keratinocytes overexpressing seq-915_x4024 exhibited higher proliferative activity and the ability to promote fibroblast migration and fibroblast proliferation. These characteristics of keratinocytes are the same biological behaviors as those of fetal keratinocytes, which contribute to skin regeneration. In addition, seq-915_x4024 suppressed the expression of the pro-inflammatory markers TNF-α, IL-6, and IL-8 and the pro-inflammatory chemokines CXCL1 and CXCL5. We also demonstrated that seq-915_x4024 regulates TGF-ß isoforms and the extracellular matrix. Moreover, using an in vivo wound-healing model, we demonstrated that overexpression of seq-915_x4024 in keratinocytes suppresses inflammatory cell infiltration and scar formation. Using bioinformatics analyses, luciferase reporter assays, and western blotting, we further demonstrated that Sar1A, Smad2, TNF-α, and IL-8 are direct targets of seq-915_x4024. Furthermore, the expression of phosphorylated Smad2 and Smad3 was reduced by seq-915_x4024. Seq-915_x4024 could be used as an anti-fibrotic factor for the treatment of wound healing.

11.
Int J Mol Med ; 42(5): 2831-2838, 2018 Nov.
Article de Anglais | MEDLINE | ID: mdl-30132574

RÉSUMÉ

Type 1 diabetes involves the immunologically mediated destruction of insulin­producing cells (IPCs) in the pancreatic islet. Mesenchymal stem cells (MSCs) have the ability to differentiate into IPCs and have become the most promising means for diabetes therapy. The present study demonstrated that human adipose­derived stem cells (hADSCs) and human amniotic MSCs (hAMSCs) are able to differentiate into functional IPCs by knocking down neuronal restrictive silencing factor (NRSF) and Sonic hedgehog (SHH). In the current study, PEI@Fe3O4 nanoparticles (NPs) were used to deliver NRSF small interfering (si)RNA and SHH siRNA to hADSCs and hAMSCs. Following infection with PEI@Fe3O4 NPs containing NRSF siRNA and SHH siRNA, the MSCs were induced to differentiate into IPCs. Four specific genes for islet cells were expressed in the differentiated cells. These cells also produced and released insulin in a glucose­responsive manner. These findings indicated that hADSCs and hAMSCs may be induced to differentiate into IPCs via PEI@Fe3O4 NP­mediated NRSF and SHH silencing.


Sujet(s)
Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Survie cellulaire/physiologie , Cellules souches mésenchymateuses/métabolisme , Technique de Western , Différenciation cellulaire/génétique , Différenciation cellulaire/physiologie , Survie cellulaire/génétique , Cellules cultivées , Cytométrie en flux , Technique d'immunofluorescence , Extinction de l'expression des gènes/physiologie , Protéines Hedgehog/métabolisme , Humains , Insuline/métabolisme , Nanoparticules
12.
Int J Mol Sci ; 19(5)2018 May 04.
Article de Anglais | MEDLINE | ID: mdl-29734654

RÉSUMÉ

Human amnion mesenchymal stem cells (hAMSCs) are promising sources of stem cells in regenerative medicine. The migration stimulated by cytokines is critical for mesenchymal stem cells (MSCs)-based cytotherapy, while the regulatory mechanisms of EGF (epidermal growth factor)-induced hAMSC migration are largely unclear. Here, a novel miRNA N-72 (GenBank accession number: MH269369) has been discovered, and its function on EGF-induced migration in hAMSCs was investigated. High-purity hAMSCs were isolated and cultured in vitro, which were characterized by flow cytometry and trilineage differentiation. The N-72 located on chromosome three was conserved, and pri-N-72 owned the ability to form a stem-loop secondary structure, which was predicated by bioinformatic programs. The expression of mature N-72 was verified in several human cells including hAMSC by real-time PCR. In EGF-stimulated hAMSC, N-72 showed a significant reduction in a PI3K and p38 MAPK-dependent manner, and N-72 mimics transfection-inhibited EGF-induced migration, which was verified by scratch assay and transwell assay. Further, the predicated target gene MMP2 was proved to be a direct target of N-72 via luciferase reporter assay, real-time PCR, and Western blotting. The results that MMP2 silencing repressed hAMSC migration suggested MMP2 as a functional downstream target of N-72. In summary, we have discovered the novel N-72, and it was crucial for EGF-induced migration by targeting MMP2 in hAMSCs.


Sujet(s)
Facteur de croissance épidermique/génétique , Matrix metalloproteinase 2/génétique , Cellules souches mésenchymateuses/métabolisme , microARN/génétique , Amnios/cytologie , Différenciation cellulaire/génétique , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Cellules cultivées , Cytométrie en flux , Régulation de l'expression des gènes au cours du développement , Humains , Phosphatidylinositol 3-kinases/génétique , p38 Mitogen-Activated Protein Kinases/génétique
13.
Int J Mol Med ; 41(2): 791-799, 2018 Feb.
Article de Anglais | MEDLINE | ID: mdl-29207015

RÉSUMÉ

Since the beginning of the use of stem cells in tissue regenerative medicine, there has been a search for optimal sources of stem cells. Human amniotic epithelial cells (hAECs) are derived from human amnions, which are typically discarded as medical waste, but were recently found to include cells with trilineage differentiation potential in vitro. Previous study has focused on the osteogenic differentiation ability of hAECs as seed cells in bone regeneration; however, their paracrine effects on osteoblasts (OBs) are yet to be elucidated. In the present study, conditioned medium (CM) derived from hAECs was used to determine their paracrine effects on the human fetal OB cell line (hFOB1.19), and the potential bioactive factors involved in this process were investigated. The results suggested that hAEC-CM markedly promoted the proliferation, migration and osteogenic differentiation of hFOB1.19 cells. Expression of transforming growth factor ß1 (TGFß1) and microRNA 34a-5p (miR-34a-5p) were detected in hAECs. Furthermore, it was demonstrated that TGFß1 and miR-34a-5p stimulated the differentiation of hFOB1.19 cells, and that TGFß1 promoted cell migration. Moreover, the effects of hAEC-CM were downregulated following the depletion of either TGFß1 or miR-34a-5p. These results demonstrated that hAECs promote OB differentiation through the secretion of TGFß1 and miR-34a-5p, and that hAECs may be an optimal cell source in bone regenerative medicine.


Sujet(s)
Régénération osseuse/effets des médicaments et des substances chimiques , Différenciation cellulaire/génétique , microARN/génétique , Facteur de croissance transformant bêta-1/génétique , Amnios/cytologie , Amnios/croissance et développement , Différenciation cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Milieux de culture conditionnés/pharmacologie , Cellules épithéliales/cytologie , Cellules épithéliales/métabolisme , Humains , Ostéoblastes/cytologie , Ostéoblastes/métabolisme
14.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 39(5): 611-614, 2017 Oct 30.
Article de Anglais | MEDLINE | ID: mdl-29125101

RÉSUMÉ

Objective To label human amniotic epithelial cells(hAECs) by using PEI-Fe3O4 nanoparticles. Methods The PEI-Fe3O4 nanoparticles were characterized by using transmission electron microscopy and dynamic light scattering. The primary cultured hAECs were labeled with the nanoparticles,and the labeling efficiency was evaluated by Prussian blue staining. The cell survival rate and viability were tested by using placenta blue staining and CCK-8 assay,respectively. Results The PEI-Fe3O4 nanoparticles were compact spheres with an average particle size of 13 nm,a hydrodynamic radius of 17.56 nm,and a zeta potential of+34.5 mV. The labeling efficiency of the nanoparticles on hAECs reached 91% when the concentrations were greater than 20 µg/ml. When the concentrations of nanoparticles were at 50 µg/ml(t=16.37,P<0.0001;t=10.39,P<0.0001) and 100 µg/ml(t=29.89,P<0.0001;t=16.86,P<0.0001),the cell survival rates and cell viabilities were significantly reduced versus controls. Conclusion The PEI-Fe3O4 nanoparticles can be used for labeling hAECs without obvious cytotoxicity at its working concentration.


Sujet(s)
Amnios/cytologie , Cellules épithéliales/cytologie , Composés du fer III/composition chimique , Nanoparticules/composition chimique , Polyéthylèneimine/composition chimique , Coloration et marquage , Survie cellulaire , Cellules cultivées , Humains , Taille de particule
15.
Sci Rep ; 7(1): 15321, 2017 11 10.
Article de Anglais | MEDLINE | ID: mdl-29127399

RÉSUMÉ

Neural stem cell (NSC) proliferation and differentiation play a pivotal role in the repair of brain function in central nervous system (CNS) diseases. Radial extracorporeal shock wave therapy (rESWT) is a non-invasive and innovative treatment for many conditions, yet little is known about the effects of this treatment on NSCs. Mouse NSCs (NE-4C) were exposed to rESWT with 1.0, 1.5, 2.0, 2.5, 3.0, and 3.5 bar (500 impulses, and 2 Hz) in vitro. Cell viability test results indicated that rESWT, at a dose of 2.5 bar, 500 impulses, and 2 Hz, increased NE-4C viability within 72 h, and that the PI3K/AKT pathway was involved in its mechanisms. Exposure to rESWT also affected proliferation and differentiation of NE-4C after 8 weeks, which may be associated with Wnt/ß-catenin and Notch pathways. This assessment is corroborated by the ability of inhibitors of Wnt/ß-catenin [Dickkopf-1 (Dkk-1)] and the Notch pathway (DAPT) to weaken proliferation and differentiation of NSCs. In summary, a proper dose of rESWT enhanced NSCs augment via the PI3K/AKT pathway initially. Also, Wnt/ß-catenin and the Notch pathway play important roles in regulation of the long-term efficacy of rESWT. This study reveals a novel approach to culture NSCs in vitro and support neurogenesis.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Traitement par ondes de choc extracorporelles , Cellules souches neurales/métabolisme , Voie de signalisation Wnt , Animaux , Peptide relié au gène de la calcitonine/métabolisme , Humains , Souris , Cellules souches neurales/cytologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs Notch/métabolisme
16.
Mol Ther Nucleic Acids ; 8: 220-231, 2017 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-28918023

RÉSUMÉ

Identifying molecular mechanisms that regulate insulin expression in bone marrow-derived mesenchymal stem cells (bmMSCs) can provide clues on how to stimulate the differentiation of bmMSCs into insulin-producing cells (IPCs), which can be used as a therapeutic approach against type 1 diabetes (T1D). As repression factors may inhibit differentiation, the efficiency of this process is insufficient for cell transplantation. In this study, we used the mouse insulin 2 (Ins2) promoter sequence and performed a DNA affinity precipitation assay combined with liquid chromatography-mass spectrometry to identify the transcription factor, chicken ovalbumin upstream promoter transcriptional factor I (COUP-TFI). Functionally, bmMSCs were reprogrammed into IPCs via COUP-TFI suppression and MafA overexpression. The differentiated cells expressed higher levels of genes specific for islet endocrine cells, and they released C-peptide and insulin in response to glucose stimulation. Transplantation of IPCs into streptozotocin-induced diabetic mice caused a reduction in hyperglycemia. Mechanistically, COUP-TFI bound to the DR1 (direct repeats with 1 spacer) element in the Ins2 promoter, thereby negatively regulating promoter activity. Taken together, the data provide a novel mechanism by which COUP-TFI acts as a negative regulator in the Ins2 promoter. The differentiation of bmMSCs into IPCs could be improved by knockdown of COUP-TFI, which may provide a novel stem cell-based therapy for T1D.

17.
Mol Med Rep ; 16(2): 2156-2162, 2017 Aug.
Article de Anglais | MEDLINE | ID: mdl-28627609

RÉSUMÉ

A fibrotic or pathological scar is an undesired consequence of skin wound healing and may trigger a series of problems. An attenuated inflammatory response is a significant characteristic of fetal skin wound healing, which can contribute to the scarless healing of fetal skin. According to deep sequencing data, microRNA­149 (miR­149) expression was increased in mid-gestational compared with that in late­gestational fetal skin keratinocytes. It was demonstrated that overexpression of miR­149 in HaCaT cells can downregulate the expression of pro­inflammatory cytokines interleukin (IL)­1α, IL­1ß, and IL­6 at basal levels and in inflammatory conditions. Furthermore, miR­149 was revealed to indirectly accelerate transforming growth factor­ß3 and collagen type III expression in fibroblasts, which are essential cells in extracellular matrix remodeling. In a rat skin wound model, miR­149 improved the quality of the arrangement of collagen bundles and reduced inflammatory cell infiltration during skin wound healing. These results indicate that miR­149 may be a potential regulator in improving the quality of skin wound healing.


Sujet(s)
Cicatrice/anatomopathologie , microARN/métabolisme , Cicatrisation de plaie/physiologie , Régions 3' non traduites , Animaux , Antagomirs/métabolisme , Antagomirs/pharmacologie , Lignée cellulaire , Mouvement cellulaire/effets des médicaments et des substances chimiques , Techniques de coculture , Collagène de type III/génétique , Collagène de type III/métabolisme , Modèles animaux de maladie humaine , Fibroblastes/cytologie , Fibroblastes/métabolisme , Humains , Interleukine-1 alpha/analyse , Interleukine-1 alpha/génétique , Interleukine-1 alpha/métabolisme , Interleukine-1 bêta/analyse , Interleukine-6/analyse , Mâle , microARN/antagonistes et inhibiteurs , microARN/génétique , Rats , Rat Wistar , Peau/anatomopathologie , Facteur de croissance transformant bêta/génétique , Facteur de croissance transformant bêta/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Cicatrisation de plaie/effets des médicaments et des substances chimiques
18.
Exp Ther Med ; 13(4): 1447-1455, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28413491

RÉSUMÉ

Well-controlled trophoblast invasion at the maternal-fetal interface is crucial for normal placentation and successful pregnancy, otherwise pathological conditions of pregnancy occur, such as preeclampsia. In previous studies, it has been demonstrated that unique long 16 binding protein (ULBP)1, a ligand for the natural-killer group (NKG)2D receptor on uterine natural killer (uNK) cells, is upregulated in the placenta in patients with preeclampsia. As they are present on the majority of the decidua, uNK have an important role in pregnancy. The aim of the present study was to determine the role of ULBP1 in trophoblast cell invasion, which is closely associated with the occurrence of preeclampsia. In the present study, ULBP1 expression levels in placentas collected after cesarean section from women with preeclampsia and normal pregnant women were determined by immunohistochemistry, reverse transcription-quantitative polymerase chain reaction and western blotting. The effects of ULBP1 on extravillous trophoblast cell line (HTR-8/SVneo) invasion mediated via uNK cells and the underlying mechanisms were investigated. mRNA and protein expression levels of ULBP1 were significantly upregulated (P<0.05) in preeclamptic placentas compared with normal controls. ULBP1 inhibited HTR-8/SVneo cells via the regulation of biological functions of uNK cells, including the downregulation of NKG2D expression on uNK cells and the stimulation of production of cytokines and chemokines that affect extravillous cytotrophoblast invasion by uNK cells. ULBP1 may have an important role in the pathophysiology of preeclampsia through the modification of biological functions of uNK cells, which may affect trophoblast invasion.

19.
Int J Mol Med ; 39(4): 918-926, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28259958

RÉSUMÉ

In vivo studies have shown that amnion-produced growth factors participate in many diseases that involve angiogenesis, re-epithelialization and immunomodulation. Although human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stem cells (hAMSCs) can be obtained from amniotic membranes, there is little information regarding their biological differences. The aim of the present study was to isolate and characterize cells from human amnions, to investigate the biological potential and behavior of these cells on the function of endothelial cells in vivo and in vitro and to examine variations in the expression profile of growth factors in different human amnion-derived cell types. Amnion fragments were enzymatically digested into two cell fractions, which were analyzed by mesenchymal and epithelial cell markers. Human aortic endothelial cells (hAoECs) were cultured with conditioned medium (CdM) collected from hAECs or hAMSCs. We used scratch and Transwell assays to evaluate migration ability; Cell Counting Kit-8 (CCK-8) and cell cycle analysis to evaluate proliferation ability; and a Matrigel tube formation assay to evaluate angiogenesis ability. To detect expression of angiogenesis-related genes, qPCR and enzyme-linked immunosorbent assay (ELISA) analyses were conducted. As stem cells, hAECs and hAMSCs all expressed the stem cell markers SSEA-4, OCT-4 and SOX-2. CdM obtained from hAECs promoted cell migration; CdM obtained from hAMSCs promoted cell proliferation; CdM obtained from hAECs and hAMSCs both promoted angiogenesis in hAoECs. Amnion-derived cells secreted significant amounts of angiogenic factors including HGF, IGF-1, VEGF, EGF, HB-EGF and bFGF, although differences in the cellular expression profile of these soluble factors were observed. Our results highlight that human amniotic epithelial and mesenchymal stem cells, which showed differences in their soluble factor secretion and angiogenic functions, could be ideal cell sources for regenerative medicine.


Sujet(s)
Amnios/métabolisme , Mouvement cellulaire , Prolifération cellulaire , Cellules endothéliales/métabolisme , Cellules épithéliales/métabolisme , Cellules souches mésenchymateuses/métabolisme , Amnios/cytologie , Agents angiogéniques/métabolisme , Animaux , Antigènes de différenciation/biosynthèse , Cellules cultivées , Cellules endothéliales/cytologie , Cellules épithéliales/cytologie , Femelle , Régulation de l'expression des gènes , Humains , Cellules souches mésenchymateuses/cytologie , Souris
20.
In Vitro Cell Dev Biol Anim ; 53(2): 157-166, 2017 Feb.
Article de Anglais | MEDLINE | ID: mdl-27699654

RÉSUMÉ

Islet transplantation provides curative treatments to patients with type 1 diabetes, but donor shortage restricts the broad use of this therapy. Thus, generation of alternative transplantable cell sources is intensively investigated worldwide. We previously showed that bone marrow-derived mesenchymal stem cells (bmMSCs) can be reprogrammed to pancreatic-like cells through simultaneously forced suppression of Rest/Nrsf (repressor element-1 silencing transcription factor/neuronal restrictive silencing factor) and Shh (sonic hedgehog) and activation of Pdx1 (pancreas and duodenal transcription factor 1). We here aimed to reprogram bmMSCs further along the developmental pathway towards the islet lineages by improving our previous strategy and by overexpression of Ngn3 (neurogenin 3) and NeuroD1 (neurogenic differentiation 1), critical regulators of the development of endocrine pancreas. We showed that compared to the previous protocol, the overexpression of only Pdx1 and Ngn3 reprogrammed bmMSCs into cells with more characteristics of islet endocrine lineages verified with bioinformatic analyses of our RNA-Seq datasets. These analyses indicated 2325 differentially expressed genes including those involved in the pancreas and islet development. We validated with qRT-PCR analysis selective genes identified from the RNA-Seq datasets. Thus, we reprogrammed bmMSCs into islet endocrine-like cells and advanced the endeavor to generate surrogate functional insulin-secreting cells.


Sujet(s)
Cellules de la moelle osseuse/cytologie , Reprogrammation cellulaire , Ilots pancréatiques/cytologie , Cellules souches mésenchymateuses/cytologie , Animaux , Technique d'immunofluorescence , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes , Gene Ontology , Insuline/métabolisme , Sécrétion d'insuline , Rats , Réaction de polymérisation en chaine en temps réel , Reproductibilité des résultats , Analyse de séquence d'ARN , Facteurs de transcription/métabolisme , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE