Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 72.795
Filtrer
2.
J Cell Biol ; 223(10)2024 Oct 07.
Article de Anglais | MEDLINE | ID: mdl-38958606

RÉSUMÉ

Calorie restriction increases lifespan. Among the tissue-specific protective effects of calorie restriction, the impact on the gastrointestinal tract remains unclear. We report increased numbers of chromogranin A-positive (+), including orexigenic ghrelin+ cells, in the stomach of calorie-restricted mice. This effect was accompanied by increased Notch target Hes1 and Notch ligand Jag1 and was reversed by blocking Notch with DAPT, a gamma-secretase inhibitor. Primary cultures and genetically modified reporter mice show that increased endocrine cell abundance is due to altered Lgr5+ stem and Neurog3+ endocrine progenitor cell proliferation. Different from the intestine, calorie restriction decreased gastric Lgr5+ stem cells, while increasing a FOXO1/Neurog3+ subpopulation of endocrine progenitors in a Notch-dependent manner. Further, activation of FOXO1 was sufficient to promote endocrine cell differentiation independent of Notch. The Notch inhibitor PF-03084014 or ghrelin receptor antagonist GHRP-6 reversed the phenotypic effects of calorie restriction in mice. Tirzepatide additionally expanded ghrelin+ cells in mice. In summary, calorie restriction promotes Notch-dependent, FOXO1-regulated gastric endocrine cell differentiation.


Sujet(s)
Restriction calorique , Protéine O1 à motif en tête de fourche , Ghréline , Récepteurs Notch , Transduction du signal , Animaux , Ghréline/métabolisme , Protéine O1 à motif en tête de fourche/métabolisme , Protéine O1 à motif en tête de fourche/génétique , Récepteurs Notch/métabolisme , Récepteurs Notch/génétique , Souris , Différenciation cellulaire , Souris de lignée C57BL , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Prolifération cellulaire , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Cellules souches/métabolisme , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Muqueuse gastrique/métabolisme , Facteur de transcription HES-1/métabolisme , Facteur de transcription HES-1/génétique , Mâle , Estomac
3.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 696-705, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949138

RÉSUMÉ

Objective: To investigate the presence of a distinct stem cell populations different from mesenchymal stem cells in the mandibular periosteum of both human and non-human primates (macaca mulatta), to explore its properties during intramembranous osteogenesis and to establish standard protocols for the isolation, culturing and expanding of mandibular periosteal stem cells (PSC) distinguished from other PSCs in other anatomical regions. Methods: Periosteum was harvested from the bone surface during flap bone removal in patients aged 18-24 years undergoing third molar extraction and from the buccal side of the mandibular premolar region of 6-year-old macaca mulatta respectively, and then subjected to single-cell sequencing using the Illumina platform Novaseq 6000 sequencer. Cross-species single-cell transcriptome sequencing results were compared using homologous gene matching. PSC were isolated from primary tissues using two digestion methods with body temperature and low temperature, and their surface markers (CD200, CD31, CD45 and CD90) were identified by cell flow cytometry. The ability of cell proliferation and three-lineage differentiation of PSC expanded to the third generation in vitro in different species were evaluated. Finally, the similarities and differences in osteogenic properties of PSC and bone marrow mesenchymal stem cells (BMSC) were compared. Results: The single-cell sequencing results indicated that 18 clusters of cell populations were identified after homologous gene matching for dimensionality reduction, and manual cellular annotation was conducted for each cluster based on cell marker databases. The comparison of different digestion protocols proved that the low-temperature overnight digestion protocol can stably isolate PSC from the human and m. mulatta mandibular periosteum and the cells exhibited a fibroblast-like morphology. This research confirmed that PSC of human and m. mulatta had similar proliferation capabilities through the cell counting kit-8 assay. Flow cytometry analysis was then used to identify the cells isolated from the periosteum expressed CD200(+), CD31(-), CD45(-), CD90(-). Then, human and m. mulatta PSC were induced into osteogenesis, adipogenesis, and chondrogenesis to demonstrate their corresponding multi-lineage differentiation capabilities. Finally, comparison with BMSC further clarified the oesteogenesis characteristics of PSC. The above experiments proved that the cells isolated from the periosteum were peiosteal cells with characteristics of stem cells evidenced by their cell morphology, proliferation ability, surface markers, and differentiation ability, and that this group of PSC possessed characteristics different from traditional mesenchymal stem cells. Conclusions: In this study, normal mandibular PSC from humans and m. mulatta were stably isolated and identified for the first time, providing a cellular foundation for investigating the mechanism of mandibular intramembranous osteogenesis, exploring ideal non-human primate models and establishing innovative strategies for clinically mandibular injury repair.


Sujet(s)
Différenciation cellulaire , Macaca mulatta , Mandibule , Périoste , Analyse sur cellule unique , Animaux , Humains , Périoste/cytologie , Mandibule/cytologie , Ostéogenèse , Cellules souches/cytologie , Cellules souches mésenchymateuses/cytologie , Cytométrie en flux , Jeune adulte , Adolescent , Séparation cellulaire/méthodes
4.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 663-671, 2024 Jul 09.
Article de Chinois | MEDLINE | ID: mdl-38949134

RÉSUMÉ

Objective: To investigate the characteristics of extracellular matrix vesicle mimetics prepared by mechanical extrusion and their effects on the cell viability and osteogenic differentiation potential of human periodontal ligament stem cells (PDLSC). Methods: PDLSC derived extracellular matrix vesicles were prepared by collagenase digestion, while the cell derived vesicle mimetics were simulated by mechanical extrusion. The obtained extracellular matrix vesicles and parental cell derived vesicle mimetics were divided into 4 groups: matrix vesicles derived from PDLSC cultured in basic medium for 7 days (PDLSC matrix vesicles, MVs), vesicle mimetics derived from PDLSC cultured in basic medium for 7 days (PDLSC vesicle mimetics, CVMs), matrix vesicles derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC matrix vesicles, O-MVs) and vesicle mimetics derived from PDLSC cultured in osteogenic inducing medium for 7 days (osteogenic-induced PDLSC vesicle mimetics, O-CVMs). Vesicles morphologies and sizes were observed by transmission electron microscopy and nanoparticle tracking analysis. Vesicles uptake was detected by immunofluorescence. With PDLSC as the control group, the effects of vesicles on the viability of PDLSC were detected by cell activity assay (cell counting kit-8), and the effects of vesicles on the osteogenic differentiation potential of PDLSC were detected by alizarin red staining and Western blotting. Results: Vesicles in MVs, O-MVs, CVMs and O-CVMs were all observed with a round structure (size 50-250 nm), and could be taken up by PDLSC without affecting the cell viability. Under osteogenic inducing conditions, PDLSC incubated with O-MVs or O-CVMs could produce more mineralized nodules than those in the control group (PDLSC). MVs, O-MVs, CVMs and O-CVMs could promote the expression of osteogenic-related proteins in PDLSC. PDLSC in group O-CVMs showed significant higher expressions of osteogenic-related proteins, including alkaline phosphatase (ALP) (1.571±0.348), osteopontin (OPN) (1.827±0.627) and osteocalcin (OCN) (1.798±0.537) compared to MVs (ALP: 1.156±0.170, OPN: 1.260±0.293, OCN: 1.286±0.302) (P<0.05). Compared to CMVs-incubated PDLSC, O-CVMs-incubated PDLSC expressed more Runt-related transcription factor 2 (1.632±0.455 vs 1.176±0.128) and OPN (1.827±0.627 vs 1.428±0.427) (P<0.05). Moreover, there was no significant difference in the expression levels of osteoblast-related proteins in PDLSC cultured with MVs, O-MVs and CVMs (P>0.05). Conclusions: The vesicle mimetics prepared by mechanical extrusion method are similar in shape and size to the extracellular matrix vesicles. MVs, O-MVs, CVMs and O-CVMs do not affect the cell viability of PDLSC, and can promote the osteogenic differentiation potential of PDLSC to a certain extent.


Sujet(s)
Différenciation cellulaire , Matrice extracellulaire , Vésicules extracellulaires , Ostéogenèse , Humains , Matrice extracellulaire/métabolisme , Vésicules extracellulaires/métabolisme , Cellules souches/cytologie , Phosphatase alcaline/métabolisme , Desmodonte/cytologie , Desmodonte/métabolisme , Ostéocalcine/métabolisme , Ostéopontine/métabolisme , Sous-unité alpha 1 du facteur CBF/métabolisme
5.
Int J Mol Sci ; 25(13)2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-39000134

RÉSUMÉ

Stem cells possess the ability to differentiate into different lineages and the ability to self-renew, thus representing an excellent tool for regenerative medicine. They can be isolated from different tissues, including the adipose tissue. Adipose tissue and human adipose-derived stem cells (hADSCs) are privileged candidates for regenerative medicine procedures or other plastic reconstructive surgeries. The cellular environment is able to influence the fate of stem cells residing in the tissue. In a previous study, we exposed hADSCs to an exhausted medium of a breast cancer cell line (MCF-7) recovered at different days (4, 7, and 10 days). In the same paper, we inferred that the medium was able to influence the behaviour of stem cells. Considering these results, in the present study, we evaluated the expression of the major genes related to adipogenic and osteogenic differentiation. To confirm the gene expression data, oil red and alizarin red colorimetric assays were performed. Lastly, we evaluated the expression of miRNAs influencing the differentiation process and the proliferation rate, maintaining a proliferative state. The data obtained confirmed that cells exposed to the medium maintained a stem and proliferative state that could lead to a risky proliferative phenotype.


Sujet(s)
Tissu adipeux , Différenciation cellulaire , Prolifération cellulaire , Ostéogenèse , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tissu adipeux/cytologie , Tissu adipeux/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Ostéogenèse/génétique , Femelle , microARN/génétique , microARN/métabolisme , Adipogenèse/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Cellules souches/effets des médicaments et des substances chimiques , Milieux de culture/pharmacologie , Milieux de culture/composition chimique
6.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000523

RÉSUMÉ

The dental implant surface plays a crucial role in osseointegration. The topography and physicochemical properties will affect the cellular functions. In this research, four distinct titanium surfaces have been studied: machined acting (MACH), acid etched (AE), grit blasting (GBLAST), and a combination of grit blasting and subsequent acid etching (GBLAST + AE). Human amniotic mesenchymal (hAMSCs) and epithelial stem cells (hAECs) isolated from the amniotic membrane have attractive stem-cell properties. They were cultured on titanium surfaces to analyze their impact on biological behavior. The surface roughness, microhardness, wettability, and surface energy were analyzed using interferometric microscopy, Vickers indentation, and drop-sessile techniques. The GBLAST and GBLAST + AE surfaces showed higher roughness, reduced hydrophilicity, and lower surface energy with significant differences. Increased microhardness values for GBLAST and GBLAST + AE implants were attributed to surface compression. Cell viability was higher for hAMSCs, particularly on GBLAST and GBLAST + AE surfaces. Alkaline phosphatase activity enhanced in hAMSCs cultured on GBLAST and GBLAST + AE surfaces, while hAECs showed no mineralization signals. Osteogenic gene expression was upregulated in hAMSCs on GBLAST surfaces. Moreover, α2 and ß1 integrin expression enhanced in hAMSCs, suggesting a surface-integrin interaction. Consequently, hAMSCs would tend toward osteoblastic differentiation on grit-blasted surfaces conducive to osseointegration, a phenomenon not observed in hAECs.


Sujet(s)
Amnios , Implants dentaires , Propriétés de surface , Titane , Humains , Titane/composition chimique , Amnios/cytologie , Amnios/métabolisme , Ostéogenèse , Différenciation cellulaire , Cellules cultivées , Ostéo-intégration , Cellules souches/cytologie , Cellules souches/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Survie cellulaire , Phosphatase alcaline/métabolisme
7.
Methods Mol Biol ; 2805: 3-18, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008171

RÉSUMÉ

Three-dimensional (3D) organoid cultures retain self-renewing stem cells that differentiate into multiple cell types that display spatial organization and functional key features, providing a highly physiological relevant system. Here we describe a strategy for the generation of 3D murine lung organoids derived from freshly isolated primary tracheal and distal lung epithelial stem cells. Isolated tracheas are subjected to enzymatic digestion to release the epithelial layer that is then dissociated into a single cell suspension for organoid culture. Lung epithelial cells are obtained from dissected lobes, which are applied to mechanical and enzymatic dissociation. After flow sorting, organoids are established from tracheal basal, secretory club, and alveolar type 2 cells in the defined conditioned medium that is required to sustain organoid growth and generate the differentiated cells. Multi-cell-type organoid co-culture replicates niches for distal epithelial stem cells to differentiate into bronchiolar and alveolar cell types. Established organoids can be fixed for wholemount staining and paraffin embedding, or passaged for further culture. Taken together, this protocol provides an efficient and validated approach to generate murine lung organoids, as well as a platform for further analysis.


Sujet(s)
Différenciation cellulaire , Poumon , Organoïdes , Animaux , Organoïdes/cytologie , Souris , Poumon/cytologie , Techniques de culture cellulaire/méthodes , Séparation cellulaire/méthodes , Cellules épithéliales/cytologie , Cellules souches/cytologie , Cellules souches/métabolisme , Phénotype , Trachée/cytologie , Techniques de coculture/méthodes
8.
Methods Mol Biol ; 2805: 203-212, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008184

RÉSUMÉ

Planarians are flatworms that have the remarkable ability to regenerate entirely new animals. This regenerative ability requires abundant adult stem cells called neoblasts, which are relatively small in size, sensitive to irradiation and the only proliferative cells in the animal. Despite the lack of cell surface markers, fluorescence-activated cell sorting (FACS) protocols have been developed to discriminate and isolate neoblasts, based on DNA content. Here, we describe a protocol that combines staining of far-red DNA dye Draq5, Calcein-AM and DAPI, along with a shortened processing time. This profiling strategy can be used to functionally characterize the neoblast population in pharmacologically-treated or gene knockdown animals. Highly purified neoblasts can be analyzed with downstream assays, such as in situ hybridization and RNA sequencing.


Sujet(s)
Cytométrie en flux , Planaires , Cellules souches , Animaux , Planaires/cytologie , Planaires/génétique , Cytométrie en flux/méthodes , Cellules souches/cytologie , Cellules souches/métabolisme , Régénération , Séparation cellulaire/méthodes , Colorants fluorescents/composition chimique
9.
Shanghai Kou Qiang Yi Xue ; 33(2): 135-140, 2024 Apr.
Article de Chinois | MEDLINE | ID: mdl-39005088

RÉSUMÉ

PURPOSE: To investigate the effect of TNF-α on osteogenic differentiation of stem cells from human exfoliated deciduous teeth (SHED), and to analyze the changes of ERK1/2-Runx2 signaling pathway in the regulation process. METHODS: SHED cells were isolated and cultured from normal deciduous permanent teeth of healthy children aged 6-8 years old, and the third passage of SHED cells were taken and divided into control group (osteogenic inducer culture), observation group (osteogenic inducer and TNF-α co-culture) and agonist group (osteogenic inducer, TNF-α and ERK pathway agonist co-culture). The osteogenic differentiation was determined by alizarin red staining. The protein expression levels of Osterix, OPN, ERK1/2, pERK1/2 and Runx2 in SHED cells were determined by Western blot. The expressions of Osterix, OPN, ERK1/2, pERK1/2 and Runx2 mRNA were detected by qRT-PCR. Statistical analysis was performed with SPSS 26.0 software package. RESULTS: Comparison of osteogenic differentiation ability of the three groups of cells showed that red-brown mineralized nodules were observed in the three groups of cells. Compared among the three groups, the control group had the most mineralized nodules, followed by the activation group, and the observation group had the least mineralized nodules. Compared with the control group, the expression levels of Osterix and OPN protein and mRNA in the observation group and the agonist group were significantly decreased, while the expression levels of Osterix and OPN protein and mRNA in the agonist group were significantly higher than those in the observation group. There was no significant difference in the expression levels of ERK1/2 protein and mRNA among the three groups, while the expression levels of pERK1/2 and Runx2 protein and mRNA in the observation group and the agonist group were significantly higher than those in the control group, and the expression levels of pERK1/2 and Runx2 protein and mRNA in the agonist group were significantly higher than those in the observation group. CONCLUSIONS: TNF-α can inhibit osteogenic differentiation of SHED cells, which may be related to the inhibition of ERK1/2-Runx2 signaling pathway.


Sujet(s)
Différenciation cellulaire , Sous-unité alpha 1 du facteur CBF , Système de signalisation des MAP kinases , Ostéogenèse , Dent de lait , Facteur de nécrose tumorale alpha , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Sous-unité alpha 1 du facteur CBF/métabolisme , Sous-unité alpha 1 du facteur CBF/génétique , Ostéogenèse/effets des médicaments et des substances chimiques , Facteur de nécrose tumorale alpha/métabolisme , Enfant , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Dent de lait/cytologie , Dent de lait/métabolisme , Facteur de transcription Sp7/métabolisme , Facteur de transcription Sp7/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Cellules souches/métabolisme , Cellules souches/cytologie , Cellules cultivées
10.
Development ; 151(20)2024 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-38950937

RÉSUMÉ

The capacity to regenerate lost tissues varies significantly among animals. Some phyla, such as the annelids, display substantial regenerating abilities, although little is known about the cellular mechanisms underlying the process. To precisely determine the origin, plasticity and fate of the cells participating in blastema formation and posterior end regeneration after amputation in the annelid Platynereis dumerilii, we developed specific tools to track different cell populations. Using these tools, we find that regeneration is partly promoted by a population of proliferative gut cells whose regenerative potential varies as a function of their position along the antero-posterior axis of the worm. Gut progenitors from anterior differentiated tissues are lineage restricted, whereas gut progenitors from the less differentiated and more proliferative posterior tissues are much more plastic. However, they are unable to regenerate the stem cells responsible for the growth of the worms. Those stem cells are of local origin, deriving from the cells present in the segment abutting the amputation plane, as are most of the blastema cells. Our results favour a hybrid and flexible cellular model for posterior regeneration in Platynereis relying on different degrees of cell plasticity.


Sujet(s)
Plasticité cellulaire , Prolifération cellulaire , Polychaeta , Régénération , Animaux , Régénération/physiologie , Polychaeta/physiologie , Polychaeta/cytologie , Plasticité cellulaire/physiologie , Cellules souches/cytologie , Différenciation cellulaire/physiologie , Annelida/physiologie
11.
Int J Biol Sci ; 20(9): 3544-3556, 2024.
Article de Anglais | MEDLINE | ID: mdl-38993564

RÉSUMÉ

Hepatic progenitor cells (HPCs) have a bidirectional potential to differentiate into hepatocytes and bile duct epithelial cells and constitute a second barrier to liver regeneration in the adult liver. They are usually located in the Hering duct in the portal vein region where various cells, extracellular matrix, cytokines, and communication signals together constitute the niche of HPCs in homeostasis to maintain cellular plasticity. In various types of liver injury, different cellular signaling streams crosstalk with each other and point to the inducible transcription factor set, including FoxA1/2/3, YB-1, Foxl1, Sox9, HNF4α, HNF1α, and HNF1ß. These transcription factors exert different functions by binding to specific target genes, and their products often interact with each other, with diverse cascades of regulation in different molecular events that are essential for homeostatic regulation, self-renewal, proliferation, and selective differentiation of HPCs. Furthermore, the tumor predisposition of adult HPCs is found to be significantly increased under transcriptional factor dysregulation in transcriptional analysis, and the altered initial commitment of the differentiation pathway of HPCs may be one of the sources of intrahepatic tumors. Related transcription factors such as HNF4α and HNF1 are expected to be future targets for tumor treatment.


Sujet(s)
Différenciation cellulaire , Humains , Animaux , Cellules souches/métabolisme , Cellules souches/cytologie , Foie/métabolisme , Foie/cytologie , Hépatocytes/métabolisme , Hépatocytes/cytologie , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Transcription génétique
12.
Cells ; 13(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38994934

RÉSUMÉ

The luminal surface of the intestinal epithelium is protected by a vital mucus layer, which is essential for lubrication, hydration, and fostering symbiotic bacterial relationships. Replicating and studying this complex mucus structure in vitro presents considerable challenges. To address this, we developed a hydrogel-integrated millifluidic tissue chamber capable of applying precise apical shear stress to intestinal models cultured on flat or 3D structured hydrogel scaffolds with adjustable stiffness. The chamber is designed to accommodate nine hydrogel scaffolds, 3D-printed as flat disks with a storage modulus matching the physiological range of intestinal tissue stiffness (~3.7 kPa) from bioactive decellularized and methacrylated small intestinal submucosa (dSIS-MA). Computational fluid dynamics simulations were conducted to confirm a laminar flow profile for both flat and 3D villi-comprising scaffolds in the physiologically relevant regime. The system was initially validated with HT29-MTX seeded hydrogel scaffolds, demonstrating accelerated differentiation, increased mucus production, and enhanced 3D organization under shear stress. These characteristic intestinal tissue features are essential for advanced in vitro models as they critically contribute to a functional barrier. Subsequently, the chamber was challenged with human intestinal stem cells (ISCs) from the terminal ileum. Our findings indicate that biomimicking hydrogel scaffolds, in combination with physiological shear stress, promote multi-lineage differentiation, as evidenced by a gene and protein expression analysis of basic markers and the 3D structural organization of ISCs in the absence of chemical differentiation triggers. The quantitative analysis of the alkaline phosphatase (ALP) activity and secreted mucus demonstrates the functional differentiation of the cells into enterocyte and goblet cell lineages. The millifluidic system, which has been developed and optimized for performance and cost efficiency, enables the creation and modulation of advanced intestinal models under biomimicking conditions, including tunable matrix stiffness and varying fluid shear stresses. Moreover, the readily accessible and scalable mucus-producing cellular tissue models permit comprehensive mucus analysis and the investigation of pathogen interactions and penetration, thereby offering the potential to advance our understanding of intestinal mucus in health and disease.


Sujet(s)
Hydrogels , Mucus , Humains , Mucus/métabolisme , Hydrogels/composition chimique , Structures d'échafaudage tissulaires/composition chimique , Muqueuse intestinale/métabolisme , Cellules HT29 , Modèles biologiques , Cellules souches/métabolisme , Cellules souches/cytologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Impression tridimensionnelle , Ingénierie tissulaire/méthodes
13.
Cells ; 13(13)2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38994993

RÉSUMÉ

The reparative and regenerative capabilities of dental pulp stem cells (DPSCs) are crucial for responding to pulp injuries, with protein phosphatase 1 (PP1) playing a significant role in regulating cellular functions pertinent to tissue healing. Accordingly, this study aimed to explore the effects of a novel cell-penetrating peptide Modified Sperm Stop 1-MSS1, that disrupts PP1, on the proliferation and odontogenic differentiation of DPSCs. Employing MSS1 as a bioportide, DPSCs were cultured and characterized for metabolic activity, cell proliferation, and cell morphology alongside the odontogenic differentiation through gene expression and alkaline phosphatase (ALP) activity analysis. MSS1 exposure induced early DPSC proliferation, upregulated genes related to odontogenic differentiation, and increased ALP activity. Markers associated with early differentiation events were induced at early culture time points and those associated with matrix mineralization were upregulated at mid-culture stages. This investigation is the first to document the potential of a PP1-disrupting bioportide in modulating DPSC functionality, suggesting a promising avenue for enhancing dental tissue regeneration and repair.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Pulpe dentaire , Odontogenèse , Protein Phosphatase 1 , Cellules souches , Pulpe dentaire/cytologie , Pulpe dentaire/effets des médicaments et des substances chimiques , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Cellules souches/métabolisme , Humains , Protein Phosphatase 1/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Odontogenèse/effets des médicaments et des substances chimiques , Peptides/pharmacologie , Peptides/métabolisme , Cellules cultivées , Phosphatase alcaline/métabolisme
14.
Int J Nanomedicine ; 19: 6811-6828, 2024.
Article de Anglais | MEDLINE | ID: mdl-39005959

RÉSUMÉ

Purpose: Angiogenesis is a tightly controlled process that initiates the formation of new vessels and its dysfunction can lead to life-threatening diseases. Apoptotic extracellular vesicles (ApoEVs) have emerged as a proangiogenic agent with high safety and isolation efficiency profile, and ApoEVs from supernumerary tooth-derived pulp stem cells (SNTSC-ApoEVs) have their unique advantages with an easily accessible parental cell source and non-invasive cell harvesting. However, the detailed characteristics of SNTSC-ApoEVs are largely unknown. This study aimed to investigate the proangiogenic capacity and function molecule of SNTSC-ApoEVs. Methods: SNTSC-ApoEVs were isolated and characterized. In vitro effects of SNTSC-ApoEVs on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) were evaluated by CCK-8, wound healing, transwell, and tube formation assays. The mRNA and protein levels of proangiogenic genes were quantified by qRT-PCR, Western blot, and immunofluorescence analysis. A Matrigel plug model was established in 6-week-old male nu/nu mice for one week, and the in vivo impact of SNTSC-ApoEVs on micro-vessel formation was assessed by histological analysis. Proteomic analysis and RNA sequencing were performed to explore the active ingredients and underlying mechanisms. Results: SNTSC-ApoEVs enhanced the proliferation, migration, and angiogenesis of HUVECs in vitro. In the Matrigel plug model in vivo, SNTSC-ApoEVs promoted CD31-positive luminal structure formation. Apart from expressing general ApoEV markers, SNTSC-ApoEVs were enriched with multiple proteins related to extracellular matrix-cell interactions. Mechanistically, SNTSC-ApoEVs transferred COL1A1 to HUVECs and promoted endothelial functions by activating the PI3K/Akt/VEGF cascade. Conclusion: SNTSC-ApoEVs can promote angiogenesis by transferring the functional molecule COL1A1 and activating the PI3K/Akt/VEGF pathway, making SNTSC-ApoEVs a promising strategy for the treatment of angiogenesis-related diseases.


Sujet(s)
Apoptose , Collagène de type I , Pulpe dentaire , Vésicules extracellulaires , Cellules endothéliales de la veine ombilicale humaine , Néovascularisation physiologique , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Dent surnuméraire , Facteur de croissance endothéliale vasculaire de type A , Vésicules extracellulaires/composition chimique , Humains , Pulpe dentaire/cytologie , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Néovascularisation physiologique/physiologie , Mâle , Phosphatidylinositol 3-kinases/métabolisme , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Souris , Collagène de type I/métabolisme , Prolifération cellulaire , Cellules souches/cytologie , Cellules souches/métabolisme , Transduction du signal , Souris nude , Mouvement cellulaire ,
15.
Cell Mol Life Sci ; 81(1): 305, 2024 Jul 16.
Article de Anglais | MEDLINE | ID: mdl-39012348

RÉSUMÉ

Lentiviral vectors have markedly enhanced gene therapy efficiency in treating congenital diseases, but their long-term safety remains controversial. Most gene therapies for congenital eye diseases need to be carried out at early ages, yet the assessment of related risks to ocular development posed by lentiviral vectors is challenging. Utilizing single-cell transcriptomic profiling on human retinal organoids, this study explored the impact of lentiviral vectors on the retinal development and found that lentiviral vectors can cause retinal precursor cells to shift toward photoreceptor fate through the up-regulation of key fate-determining genes such as PRDM1. Further investigation demonstrated that the intron and intergenic region of PRDM1 was bound by PHLDA1, which was also up-regulated by lentiviral vectors exposure. Importantly, knockdown of PHLDA1 successfully suppressed the lentivirus-induced differentiation bias of photoreceptor cells. The findings also suggest that while lentiviral vectors may disrupt the fate determination of retinal precursor cells, posing risks in early-stage retinal gene therapy, these risks could potentially be reduced by inhibiting the PHLDA1-PRDM1 axis.


Sujet(s)
Différenciation cellulaire , Vecteurs génétiques , Lentivirus , Rétine , Cellules souches , Facteurs de transcription , Humains , Rétine/métabolisme , Rétine/cytologie , Lentivirus/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Vecteurs génétiques/métabolisme , Vecteurs génétiques/génétique , Différenciation cellulaire/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Facteur-1 liant le domaine de régulation positive I/génétique , Facteur-1 liant le domaine de régulation positive I/métabolisme , Organoïdes/métabolisme , Organoïdes/cytologie , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Thérapie génétique/méthodes
16.
Biomed Phys Eng Express ; 10(5)2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38959871

RÉSUMÉ

Biocompatible polymer-based scaffolds hold great promise for neural repair, especially when they are coupled with electrostimulation to induce neural differentiation. In this study, a combination of polyacrylonitrile/polyaniline (PAN/PANI) and Carbon Nanotubes (CNTs) were used to fabricate three different biomimetic electrospun scaffolds (samples 1, 2 and 3 containing 0.26 wt%, 1 wt% and 2 wt% of CNTs, respectively). These scaffolds underwent thorough characterization for assessing electroconductivity, tensile strength, wettability, degradability, swelling, XRD, and FTIR data. Notably, scanning electron microscopy (SEM) images revealed a three-dimensional scaffold morphology with aligned fibers ranging from 60 nm to 292 nm in diameter. To comprehensively investigate the impact of electrical stimulation on the nervous differentiation of the stem cells seeded on these scaffolds, cell morphology and adhesion were assessed based on SEM images. Additionally, scaffold biocompatibility was studied through MTT assay. Importantly, Real-Time PCR results indicated the expression of neural markers-Nestin,ß-tubulin III, and MAP2-by the cells cultured on these samples. In comparison with the control group, samples 1 and 2 exhibited significant increases in Nestin marker expression, indicating early stages of neuronal differentiation, whileß-tubulin III expression was significantly reduced and MAP2 expression remained statistically unchanged. In contrast, sample 3 did not display a statistically significant upturn in Nestin maker expression, while showcasing remarkable increases in the expression of both MAP2 andß-tubulin III, as markers of the end stages of differentiation, leading to postmitotic neurons. These results could be attributed to the higher electroconductivity of S3 compared to other samples. Our findings highlight the biomimetic potential of the prepared scaffolds for neural repair, illustrating their effectiveness in guiding stem cell differentiation toward a neural lineage.


Sujet(s)
Résines acryliques , Dérivés de l'aniline , Différenciation cellulaire , Nanotubes de carbone , Régénération nerveuse , Ingénierie tissulaire , Structures d'échafaudage tissulaires , Structures d'échafaudage tissulaires/composition chimique , Nanotubes de carbone/composition chimique , Dérivés de l'aniline/composition chimique , Résines acryliques/composition chimique , Ingénierie tissulaire/méthodes , Matériaux biocompatibles/composition chimique , Stimulation électrique , Humains , Adhérence cellulaire , Microscopie électronique à balayage , Cellules souches/cytologie , Résistance à la traction , Neurones/métabolisme , Neurones/cytologie , Animaux , Nestine/métabolisme
17.
Cells ; 13(13)2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38994981

RÉSUMÉ

Aging is the main risk factor for chronic lung diseases (CLDs) including idiopathic pulmonary fibrosis (IPF) and chronic obstructive pulmonary disease (COPD). Accordingly, hallmarks of aging like cellular senescence are increased in these patients in different lung cell types including fibroblasts. However, little is known about the different triggers that induce a senescence phenotype in different disease backgrounds and its role in CLD pathogenesis. Therefore, we characterized senescence in primary human lung fibroblasts (phLF) from control, IPF, or COPD patients at baseline and after exposure to disease-relevant insults (H2O2, bleomycin, TGF-ß1) and studied their capacity to support progenitor cell potential in a lung organoid model. Bulk-RNA sequencing revealed that phLF from IPF and COPD activate different transcriptional programs but share a similar senescence phenotype at baseline. Moreover, H2O2 and bleomycin but not TGF-ß1 induced senescence in phLF from different disease origins. Exposure to different triggers resulted in distinct senescence programs in phLF characterized by different SASP profiles. Finally, co-culture with bleomycin- and H2O2-treated phLF reduced the progenitor cell potential of alveolar epithelial progenitor cells. In conclusion, phLF from COPD and IPF share a conserved senescence response that varies depending on the insult and impairs alveolar epithelial progenitor capacity ex vivo.


Sujet(s)
Bléomycine , Vieillissement de la cellule , Fibroblastes , Peroxyde d'hydrogène , Fibrose pulmonaire idiopathique , Poumon , Cellules souches , Humains , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Fibroblastes/effets des médicaments et des substances chimiques , Fibrose pulmonaire idiopathique/anatomopathologie , Fibrose pulmonaire idiopathique/métabolisme , Poumon/cytologie , Poumon/anatomopathologie , Bléomycine/pharmacologie , Cellules souches/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/cytologie , Peroxyde d'hydrogène/pharmacologie , Broncho-pneumopathie chronique obstructive/anatomopathologie , Broncho-pneumopathie chronique obstructive/métabolisme , Facteur de croissance transformant bêta-1/pharmacologie , Facteur de croissance transformant bêta-1/métabolisme , Pneumocytes/métabolisme , Pneumocytes/effets des médicaments et des substances chimiques , Cellules cultivées
18.
Plant Cell Rep ; 43(7): 188, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38960994

RÉSUMÉ

KEY MESSAGE: BIG regulates ground tissue formative divisions by bridging the auxin gradient with SHR abundance in Arabidopsis roots. The formative divisions of cortex/endodermis initials (CEIs) and CEI daughter cells (CEIDs) in Arabidopsis roots are coordinately controlled by the longitudinal auxin gradient and the radial SHORT ROOT (SHR) abundance. However, the mechanism underlying this coordination remains poorly understood. In this study, we demonstrate that BIG regulates ground tissue formative divisions by bridging the auxin gradient with SHR abundance. Mutations in BIG gene repressed cell cycle progression, delaying the formative divisions within the ground tissues and impairing the establishment of endodermal and cortical identities. In addition, we uncovered auxin's suppressive effect on BIG expression, triggering CYCLIND6;1 (CYCD6;1) activation in an SHR-dependent fashion. Moreover, the degradation of RETINOBLASTOMA-RELATED (RBR) is jointly regulated by BIG and CYCD6;1. The loss of BIG function led to RBR protein accumulation, detrimentally impacting the SHR/SCARECROW (SCR) protein complex and the CEI/CEID formative divisions. Collectively, these findings shed light on a fundamental mechanism wherein BIG intricately coordinates the interplay between SHR/SCR and auxin, steering ground tissue patterning within Arabidopsis root tissue.


Sujet(s)
Protéines d'Arabidopsis , Arabidopsis , Régulation de l'expression des gènes végétaux , Acides indolacétiques , Racines de plante , Arabidopsis/génétique , Arabidopsis/métabolisme , Arabidopsis/cytologie , Acides indolacétiques/métabolisme , Protéines d'Arabidopsis/métabolisme , Protéines d'Arabidopsis/génétique , Racines de plante/cytologie , Racines de plante/croissance et développement , Racines de plante/métabolisme , Racines de plante/génétique , Division cellulaire asymétrique , Mutation/génétique , Cellules souches/métabolisme , Cellules souches/cytologie , Cyclines/métabolisme , Cyclines/génétique , Protéines de liaison à la calmoduline , Facteurs de transcription
19.
BMC Musculoskelet Disord ; 25(1): 537, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997667

RÉSUMÉ

BACKGROUND: Human intervertebral disk degeneration (IVDD) is a sophisticated degenerative pathological process. A key cause of IVDD progression is nucleus pulposus cell (NPC) degeneration, which contributes to excessive endoplasmic reticulum stress in the intervertebral disk. However, the mechanisms underlying IVDD and NPC degeneration remain unclear. METHODS: We used interleukin (IL)-1ß stimulation to establish an NPC-degenerated IVDD model and investigated whether human urine-derived stem cell (USC) exosomes could prevent IL-1ß-induced NPC degeneration using western blotting, quantitative real-time polymerase chain reaction, flow cytometry, and transcriptome sequencing techniques. RESULTS: We successfully extracted and identified USCs and exosomes from human urine. IL-1ß substantially downregulated NPC viability and induced NPC degeneration while modulating the expression of SOX-9, collagen II, and aggrecan. Exosomes from USCs could rescue IL-1ß-induced NPC degeneration and restore the expression levels of SOX-9, collagen II, and aggrecan. CONCLUSIONS: USC-derived exosomes can prevent NPCs from degeneration following IL-1ß stimulation. This finding can aid the development of a potential treatment strategy for IVDD.


Sujet(s)
Exosomes , Interleukine-1 bêta , Dégénérescence de disque intervertébral , Nucleus pulposus , Facteur de transcription SOX-9 , Humains , Interleukine-1 bêta/métabolisme , Exosomes/métabolisme , Dégénérescence de disque intervertébral/anatomopathologie , Dégénérescence de disque intervertébral/métabolisme , Dégénérescence de disque intervertébral/thérapie , Nucleus pulposus/métabolisme , Nucleus pulposus/anatomopathologie , Nucleus pulposus/cytologie , Nucleus pulposus/effets des médicaments et des substances chimiques , Facteur de transcription SOX-9/métabolisme , Facteur de transcription SOX-9/génétique , Animaux , Cellules souches/métabolisme , Cellules cultivées , Agrécanes/métabolisme , Agrécanes/génétique , Mâle , Urine/cytologie , Urine/composition chimique , Femelle , Collagène de type II/métabolisme
20.
Chem Pharm Bull (Tokyo) ; 72(7): 669-675, 2024.
Article de Anglais | MEDLINE | ID: mdl-39010213

RÉSUMÉ

Tendon injury is a prevalent orthopedic disease that currently lacks effective treatment. Galangin (GLN) is a vital flavonoid found abundantly in galangal and is known for its natural activity. This study aimed to investigate the GLN-mediated molecular mechanism of tendon-derived stem cells (TDSCs) in tendon repair. The TDSCs were characterized using alkaline phosphatase staining, alizarin red S staining, oil red O staining, and flow cytometry. The effect of GLN treatment on collagen deposition was evaluated using Sirius red staining and quantitative (q)PCR, while a Western bot was used to assess protein levels and analyze pathways. Results showed that GLN treatment not only increased the collagen deposition but also elevated the mRNA expression and protein levels of multiple tendon markers like collagen type I alpha 1 (COL1A1), decorin (DCN) and tenomodulin (TNMD) in TDSCs. Moreover, GLN was also found to upregulate the protein levels of transforming growth factor ß1 (TGF-ß1) and p-Smad3 to activate the TGF-ß1/Smad3 signaling pathway, while GLN mediated collagen deposition in TDSCs was reversed by LY3200882, a TGF-ß receptor inhibitor. The study concluded that GLN-mediated TDSCs enhanced tendon repair by activating the TGF-ß1/Smad3 signaling pathway, suggesting a novel therapeutic option in treating tendon repair.


Sujet(s)
Flavonoïdes , Transduction du signal , Protéine Smad-3 , Cellules souches , Tendons , Facteur de croissance transformant bêta-1 , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Facteur de croissance transformant bêta-1/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Animaux , Protéine Smad-3/métabolisme , Protéine Smad-3/antagonistes et inhibiteurs , Cellules souches/effets des médicaments et des substances chimiques , Cellules souches/métabolisme , Cellules souches/cytologie , Tendons/cytologie , Tendons/métabolisme , Tendons/effets des médicaments et des substances chimiques , Rats , Cellules cultivées , Rat Sprague-Dawley , Traumatismes des tendons/traitement médicamenteux , Traumatismes des tendons/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...