Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.550
Filter
1.
J Environ Sci (China) ; 147: 294-309, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39003048

ABSTRACT

Endocrine-disrupting chemicals (EDCs) are compounds, either natural or man-made, that interfere with the normal functioning of the endocrine system. There is increasing evidence that exposure to EDCs can have profound adverse effects on reproduction, metabolic disorders, neurological alterations, and increased risk of hormone-dependent cancer. Stem cells (SCs) are integral to these pathological processes, and it is therefore crucial to understand how EDCs may influence SC functionality. This review examines the literature on different types of EDCs and their effects on various types of SCs, including embryonic, adult, and cancer SCs. Possible molecular mechanisms through which EDCs may influence the phenotype of SCs are also evaluated. Finally, the possible implications of these effects on human health are discussed. The available literature demonstrates that EDCs can influence the biology of SCs in a variety of ways, including by altering hormonal pathways, DNA damage, epigenetic changes, reactive oxygen species production and alterations in the gene expression patterns. These disruptions may lead to a variety of cell fates and diseases later in adulthood including increased risk of endocrine disorders, obesity, infertility, reproductive abnormalities, and cancer. Therefore, the review emphasizes the importance of raising broader awareness regarding the intricate impact of EDCs on human health.


Subject(s)
Endocrine Disruptors , Stem Cells , Endocrine Disruptors/toxicity , Humans , Stem Cells/drug effects , Environmental Pollutants/toxicity , Environmental Exposure
2.
Mol Med Rep ; 30(5)2024 Nov.
Article in English | MEDLINE | ID: mdl-39301638

ABSTRACT

Periodontitis, a common oral disease characterized by the progressive infiltration of bacteria, is a leading cause of adult tooth loss. Periodontal stem cells (PDLSCs) possess good self­renewal and multi­potential differentiation abilities to maintain the integrity of periodontal support structure and repair defects. The present study aimed to analyze the roles of Wnt7B and frizzled4 (FZD4) in the osteogenic differentiation and macrophage polarization during periodontitis using an in vitro cell model. First, Wnt7B expression in the periodontitis­affected gingival tissue of patients and lipopolysaccharide (LPS)­stimulated PDLSCs was assessed using the GSE23586 dataset and western blot analysis, respectively. In Wnt7B­overexpressing PDLSCs exposed to LPS, the capacity of osteogenic differentiation was evaluated by detecting alkaline phosphatase activity, the level of Alizarin Red S staining and the expression of genes related to osteogenic differentiation. Subsequently, conditioned medium from PDLSCs overexpressing Wnt7B was used for M0 macrophage culture. The expression of CD86 and INOS was examined using immunofluorescence staining and western blot analysis. In addition, reverse transcription­quantitative PCR was employed to examine the expression of TNF­α, IL­6 and IL­1ß in macrophages. The binding between Wnt7B and FZD4 was estimated using co­immunoprecipitation. In addition, FZD4 was silenced to perform the rescue experiments to elucidate the regulatory mechanism between Wnt7B and FZD4. The results demonstrated a decreased expression of Wnt7B in periodontitis­affected gingival tissue and in LPS­exposed PDLSCs. Wnt7B overexpression promoted the osteogenic differentiation of LPS­exposed PDLSCs and suppressed the M1 polarization of macrophages. Additionally, Wnt7B bound to FZD4 and upregulated FZD4 expression. FZD4 silencing reversed the effects of Wnt7B overexpression on the osteogenic differentiation in LPS­exposed PDLSCs and the M1 polarization of macrophages. In summary, Wnt7B plays an anti­periodontitis role by binding FZD4 to strengthen the osteogenic differentiation of LPS­stimulated PDLSCs and suppress the M1 polarization of macrophages.


Subject(s)
Cell Differentiation , Frizzled Receptors , Lipopolysaccharides , Macrophages , Osteogenesis , Periodontal Ligament , Stem Cells , Wnt Proteins , Humans , Frizzled Receptors/metabolism , Frizzled Receptors/genetics , Osteogenesis/drug effects , Macrophages/metabolism , Macrophages/drug effects , Cell Differentiation/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Wnt Proteins/metabolism , Wnt Proteins/genetics , Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Periodontitis/metabolism , Periodontitis/pathology , Cells, Cultured , Adult , Protein Binding
3.
J Hazard Mater ; 479: 135734, 2024 Nov 05.
Article in English | MEDLINE | ID: mdl-39244982

ABSTRACT

Intestinal epithelium has the largest surface of human body, contributes dramatically to defense of toxicant-associated intestinal injury. Triclosan (TCS) and triclocarban (TCC), extensively employed as antibacterial agents in personal care products (PCPs) and healthcare facilities, caused serious damage to human intestine. However, the role of the intestinal epithelium in TCS/TCC-induced intestinal toxicity and its underlying toxic mechanisms remain incompletely understood. In this study, a novel 3D intestinal organoid model was utilized to investigate that exposure to TCS/TCC led to a compromised self-renewal and differentiation of intestinal stem cells (ISCs). Consequently, this disrupted intestinal epithelial homeostasis ultimately caused a reduction in nutrient absorption and deficient of epithelial defense to exogenous and endogenous pathogens stimulation. The inhibition of the Wnt signaling pathway in intestinal stem cell was contributed to the intestinal toxicity of TCS/TCC. These results were further confirmed in vivo with mice exposed to TCS/TCC. The findings of this study provide evidence that TCS/TCC possess the capacity to disturb the homeostasis of the intestinal epithelium, and emphasize the credibility of organoids as a valuable model for toxicological studies, as they could faithfully recapitulate in vivo phenomena.


Subject(s)
Carbanilides , Homeostasis , Intestinal Mucosa , Intestine, Small , Organoids , Stem Cells , Triclosan , Triclosan/toxicity , Carbanilides/toxicity , Organoids/drug effects , Animals , Homeostasis/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Stem Cells/drug effects , Wnt Signaling Pathway/drug effects , Anti-Infective Agents, Local/toxicity , Mice , Mice, Inbred C57BL , Humans , Male , Cell Differentiation/drug effects
4.
Nat Commun ; 15(1): 7957, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261467

ABSTRACT

Postprandial IL-1ß surges are predominant in the white adipose tissue (WAT), but its consequences are unknown. Here, we investigate the role of IL-1ß in WAT energy storage and show that adipocyte-specific deletion of IL-1 receptor 1 (IL1R1) has no metabolic consequences, whereas ubiquitous lack of IL1R1 reduces body weight, WAT mass, and adipocyte formation in mice. Among all major WAT-resident cell types, progenitors express the highest IL1R1 levels. In vitro, IL-1ß potently promotes adipogenesis in murine and human adipose-derived stem cells. This effect is exclusive to early-differentiation-stage cells, in which the adipogenic transcription factors C/EBPδ and C/EBPß are rapidly upregulated by IL-1ß and enriched near important adipogenic genes. The pro-adipogenic, but not pro-inflammatory effect of IL-1ß is potentiated by acute treatment and blocked by chronic exposure. Thus, we propose that transient postprandial IL-1ß surges regulate WAT remodeling by promoting adipogenesis, whereas chronically elevated IL-1ß levels in obesity blunts this physiological function.


Subject(s)
Adipocytes , Adipogenesis , Adipose Tissue, White , CCAAT-Enhancer-Binding Protein-beta , Interleukin-1beta , Receptors, Interleukin-1 Type I , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Interleukin-1beta/metabolism , Humans , Adipocytes/metabolism , Adipocytes/cytology , Receptors, Interleukin-1 Type I/metabolism , Receptors, Interleukin-1 Type I/genetics , Mice , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Adipose Tissue, White/metabolism , Adipose Tissue, White/cytology , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Protein-delta/genetics , Male , Mice, Knockout , Stem Cells/metabolism , Stem Cells/drug effects , Mice, Inbred C57BL , Cell Differentiation/drug effects
5.
Cells ; 13(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273035

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory disorder in the intestines without a cure. Current therapies suppress inflammation to prevent further intestinal damage. However, healing already damaged intestinal epithelia is still an unmet medical need. Under physiological conditions, Lgr5+ intestinal stem cells (ISCs) in the intestinal crypts replenish the epithelia every 3-5 days. Therefore, understanding the regulation of Lgr5+ ISCs is essential. Previous data suggest vitamin D signaling is essential to maintain normal Lgr5+ ISC function in vivo. Our recent data indicate that to execute its functions in the intestines optimally, 1,25(OH)2D requires high concentrations that, if present systemically, can cause hypercalcemia (i.e., blood calcium levels significantly higher than physiological levels), leading to severe consequences. Using 5-bromo-2'-deoxyuridine (BrdU) to label the actively proliferating ISCs, our previous data suggested that de novo synthesized locally high 1,25(OH)2D concentrations effectively enhanced the migration and differentiation of ISCs without causing hypercalcemia. However, although sparse in the crypts, other proliferating cells other than Lgr5+ ISCs could also be labeled with BrdU. This current study used high-purity Lgr5+ ISC lines and a mouse strain, in which Lgr5+ ISCs and their progeny could be specifically tracked, to investigate the effects of de novo synthesized locally high 1,25(OH)2D concentrations on Lgr5+ ISC function. Our data showed that 1,25(OH)2D at concentrations significantly higher than physiological levels augmented Lgr5+ ISC differentiation in vitro. In vivo, de novo synthesized locally high 1,25(OH)2D concentrations significantly elevated local 1α-hydroxylase expression, robustly suppressed experimental colitis, and promoted Lgr5+ ISC differentiation. For the first time, this study definitively demonstrated 1,25(OH)2D's role in Lgr5+ ISCs, underpinning 1,25(OH)2D's promise in IBD therapy.


Subject(s)
Receptors, G-Protein-Coupled , Stem Cells , Vitamin D , Animals , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology , Vitamin D/pharmacology , Vitamin D/analogs & derivatives , Vitamin D/metabolism , Mice , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Regeneration/drug effects , Mice, Inbred C57BL , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Intestines/drug effects
6.
Braz Oral Res ; 38: e079, 2024.
Article in English | MEDLINE | ID: mdl-39258632

ABSTRACT

Periodontal regeneration is a challenge, and tissue engineering based on periodontal ligament stem cells (PDLSCs) has been shown to be a promising alternative to this process. However, the need for scaffolds has limited the therapeutic use of PDLSCs. In this context, scaffold-free tissue engineering using the cell sheet (CS) technique has been developed as an alternative approach to improve tissue regeneration. Previously, we showed that Protease-activated receptor-1 (PAR1) can regulate PDLSCs. Herein, we evaluate whether PAR1 influences osteogenesis in CSs produced from PDLSCs, without the use of scaffolds. PDLSCs were isolated and immunophenotyped. Then, CSs were obtained by supplementing the culture medium with ascorbic acid (50 µg/mL), and PAR1 was activated through its agonist peptide (100 nM). Scaffold-free 3D CSs were successfully produced from PDLSCs, and they showed higher proliferation potential than isolated PDLSCs. Also, PAR1 activation decreased senescence and improved osteogenic differentiation of CSs by increasing mineralized nodule deposition and alkaline phosphatase concentration; PAR1 also modulated osteogenic markers at the gene and protein levels. We further demonstrated that this effect was regulated by Wnt, TGF-ßI, MEK, p38 MAPK, and FGF/VEGF signaling pathways in PDLSCs (p < 0.05%). Overall, PAR1 activation increased osteogenic activity in CSs, emerging as a promising scaffold-free therapeutic approach for periodontal regeneration.


Subject(s)
Cell Differentiation , Cell Proliferation , Osteogenesis , Periodontal Ligament , Receptor, PAR-1 , Stem Cells , Tissue Engineering , Periodontal Ligament/cytology , Osteogenesis/drug effects , Osteogenesis/physiology , Humans , Cell Differentiation/drug effects , Stem Cells/physiology , Stem Cells/drug effects , Cells, Cultured , Cell Proliferation/drug effects , Tissue Engineering/methods , Alkaline Phosphatase/analysis , Alkaline Phosphatase/metabolism , Reproducibility of Results , Adolescent , Time Factors , Real-Time Polymerase Chain Reaction , Immunophenotyping , Analysis of Variance
7.
Nihon Yakurigaku Zasshi ; 159(5): 295-299, 2024.
Article in Japanese | MEDLINE | ID: mdl-39218673

ABSTRACT

Prediction of intestinal drug absorption and drug-induced intestinal toxicity is critical for the development of orally-administered drugs. However, it is difficult to accurately predict these events because of large species differences and a lack of appropriate in vitro assay. Then, we proposed the use of human crypt-derived intestinal cells for the prediction of intestinal absorption and the risk of intestinal toxicity. 3D human intestinal spheroids were established from fresh surgical specimens of proximal jejunum and terminal ileum using the conditioned media containing Wnt3a, R-spondin 3, and noggin. To generate 2D monolayer, spheroids were enzymatically dissociated into single cells and plated onto Matrigel-precoated culture plates/inserts. We have confirmed the activities of typical drug-metabolizing enzymes and uptake/efflux transporters in human jejunal spheroid-derived differentiated cells. Intestinal availability (Fg) estimated from the apical-to-basal permeation clearance across the jejunal monolayer showed a good correlation with in vivo human Fg values for five CYP3A substrate drugs. As for the prediction of intestinal toxicity, we found that the degree of ATP decreases in intestinal spheroids incubated with different EGFR-TKIs varied greatly depending on the drugs and the rank order of the extent of ATP decrease corresponded with that of frequency of clinically-observed diarrhea. We also constructed enterochromaffin (EC) cell-rich spheroids and quantified serotonin release from EC cells upon exposure to drugs for the prediction of drug-induced nausea and vomiting. As a result, we found that the serotonin release was related to the high/low risk of nausea and vomiting of each ALK/ROS1 kinase inhibitors.


Subject(s)
Intestinal Absorption , Stem Cells , Humans , Animals , Stem Cells/metabolism , Stem Cells/drug effects , Cells, Cultured , Intestinal Mucosa/metabolism , Intestines/drug effects
8.
Stem Cell Res Ther ; 15(1): 247, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113140

ABSTRACT

BACKGROUND: The role of periodontal ligament stem cells (PDLSCs) in repairing periodontal destruction is crucial, but their functions can be impaired by excessive oxidative stress (OS). Nocardamine (NOCA), a cyclic siderophore, has been shown to possess anti-cancer and anti-bacterial properties. This study aimed to investigate the protective mechanisms of NOCA against OS-induced cellular dysfunction in PDLSCs. METHODS: The cytotoxicity of NOCA on PDLSCs was assessed using a CCK-8 assay. PDLSCs were then treated with hydrogen peroxide (H2O2) to induce OS. ROS levels, cell viability, and antioxidant factor expression were analyzed using relevant kits after treatment. Small molecule inhibitors U0126 and XAV-939 were employed to block ERK signaling and Wnt pathways respectively. Osteogenic differentiation was assessed using alkaline phosphatase (ALP) activity staining and Alizarin Red S (ARS) staining of mineralized nodules. Expression levels of osteogenic gene markers and ERK pathway were determined via real-time quantitative polymerase chain reaction (RT-qPCR) or western blot (WB) analysis. ß-catenin nuclear localization was examined by western blotting and confocal microscopy. RESULTS: NOCA exhibited no significant cytotoxicity at concentrations below 20 µM and effectively inhibited H2O2-induced OS in PDLSCs. NOCA also restored ALP activity, mineralized nodule formation, and the expression of osteogenic markers in H2O2-stimulated PDLSCs. Mechanistically, NOCA increased p-ERK level and promoted ß-catenin translocation into the nucleus; however, blocking ERK pathway disrupted the osteogenic protection provided by NOCA and impaired its ability to induce ß-catenin nuclear translocation under OS conditions in PDLSCs. CONCLUSIONS: NOCA protected PDLSCs against H2O2-induced OS and effectively restored impaired osteogenic differentiation in PDLSCs by modulating the ERK/Wnt signaling pathway.


Subject(s)
Cell Differentiation , Hydrogen Peroxide , Osteogenesis , Oxidative Stress , Periodontal Ligament , Stem Cells , Periodontal Ligament/cytology , Periodontal Ligament/metabolism , Periodontal Ligament/drug effects , Humans , Oxidative Stress/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/toxicity , Osteogenesis/drug effects , Cell Differentiation/drug effects , beta Catenin/metabolism , Cell Survival/drug effects , Wnt Signaling Pathway/drug effects , MAP Kinase Signaling System/drug effects , Cells, Cultured , Reactive Oxygen Species/metabolism
9.
J Appl Oral Sci ; 32: e20230462, 2024.
Article in English | MEDLINE | ID: mdl-39140577

ABSTRACT

OBJECTIVE: Several materials have been developed to preserve pulp vitality. They should have ideal cytocompatibility characteristics to promote the activity of stem cells of human exfoliated deciduous teeth (SHED) and thus heal pulp tissue. OBJECTIVE: To evaluate the cytotoxicity of different dilutions of bioceramic material extracts in SHED. METHODOLOGY: SHED were immersed in αMEM + the material extract according to the following experimental groups: Group 1 (G1) -BBio membrane, Group 2 (G2) - Bio-C Repair, Group 3 (G3) - MTA Repair HP, Group 4 (G4) - TheraCal LC, and Group 5 (G5) - Biodentine. Positive and negative control groups were maintained respectively in αMEM + 10% FBS and Milli-Q Water. The methods to analyze cell viability and proliferation involved MTT and Alamar Blue assays at 24, 48, and 72H after the contact of the SHED with bioceramic extracts at 1:1 and 1:2 dilutions. Data were analyzed by the three-way ANOVA, followed by Tukey's test (p<0.05). RESULTS: At 1:1 dilution, SHED in contact with the MTA HP Repair extract showed statistically higher cell viability than the other experimental groups and the negative control (p<0.05), except for TheraCal LC (p> 0.05). At 1:2 dilution, BBio Membrane and Bio-C showed statistically higher values in intra- and intergroup comparisons (p<0.05). BBio Membrane, Bio-C Repair, and Biodentine extracts at 1:1 dilution showed greater cytotoxicity than 1:2 dilution in all periods (p<0.05). CONCLUSION: MTA HP Repair showed the lowest cytotoxicity even at a 1:1 dilution. At a 1:2 dilution, the SHED in contact with the BBio membrane extract showed high cell viability. Thus, the BBio membrane would be a new non-cytotoxic biomaterial for SHED. Results offer possibilities of biomaterials that can be indicated for use in clinical regenerative procedures of the dentin-pulp complex.


Subject(s)
Aluminum Compounds , Biocompatible Materials , Calcium Compounds , Cell Proliferation , Cell Survival , Ceramics , Dental Pulp , Drug Combinations , Materials Testing , Oxides , Silicates , Stem Cells , Tooth, Deciduous , Humans , Tooth, Deciduous/drug effects , Silicates/chemistry , Silicates/toxicity , Silicates/pharmacology , Cell Survival/drug effects , Calcium Compounds/chemistry , Calcium Compounds/pharmacology , Calcium Compounds/toxicity , Stem Cells/drug effects , Time Factors , Oxides/chemistry , Oxides/toxicity , Cell Proliferation/drug effects , Dental Pulp/drug effects , Dental Pulp/cytology , Ceramics/chemistry , Ceramics/toxicity , Aluminum Compounds/chemistry , Aluminum Compounds/toxicity , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Analysis of Variance , Reproducibility of Results , Bismuth/chemistry , Bismuth/toxicity , Bismuth/pharmacology , Cells, Cultured , Reference Values , Tetrazolium Salts , Xanthenes/chemistry , Oxazines
10.
Int J Nanomedicine ; 19: 8285-8308, 2024.
Article in English | MEDLINE | ID: mdl-39161362

ABSTRACT

The endometrium is an extremely important component of the uterus and is crucial for individual health and human reproduction. However, traditional methods still struggle to ideally repair the structure and function of damaged endometrium and restore fertility. Therefore, seeking and developing innovative technologies and materials has the potential to repair and regenerate damaged or diseased endometrium. The emergence and functionalization of various nanomedicine and biomaterials, as well as the proposal and development of regenerative medicine and tissue engineering techniques, have brought great hope for solving these problems. In this review, we will summarize various nanomedicine, biomaterials, and innovative technologies that contribute to endometrial regeneration, including nanoscale exosomes, nanomaterials, stem cell-based materials, naturally sourced biomaterials, chemically synthesized biomaterials, approaches and methods for functionalizing biomaterials, as well as the application of revolutionary new technologies such as organoids, organ-on-chips, artificial intelligence, etc. The diverse design and modification of new biomaterials endow them with new functionalities, such as microstructure or nanostructure, mechanical properties, biological functions, and cellular microenvironment regulation. It will provide new options for the regeneration of endometrium, bring new hope for the reconstruction and recovery of patients' reproductive abilities.


Subject(s)
Biocompatible Materials , Endometrium , Nanomedicine , Regeneration , Regenerative Medicine , Tissue Engineering , Humans , Endometrium/drug effects , Endometrium/physiology , Nanomedicine/methods , Female , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Tissue Engineering/methods , Regeneration/drug effects , Regenerative Medicine/methods , Nanostructures/chemistry , Animals , Exosomes/chemistry , Stem Cells/drug effects , Stem Cells/cytology
11.
Sci Rep ; 14(1): 19354, 2024 08 21.
Article in English | MEDLINE | ID: mdl-39169066

ABSTRACT

Fibroblast growth factor 2 (FGF2) is a crucial factor in odontoblast differentiation and dentin matrix deposition, which facilitates pulpodentin repair and regeneration. Nevertheless, the specific biological function of FGF2 in odontoblastic differentiation remains unclear because it is controlled by complex signalling pathways. This study aimed to investigate the mechanism underlying the effect of FGF2 on osteo/odontogenic differentiation of stem cells from the apical papilla (SCAP). SCAP were pretreated with conditioned media containing FGF2 for 1 week, followed by culturing in induced differentiation medium for another week. RNA sequencing (RNA-seq) combined with quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to evaluate the pathways affected by FGF2 in SCAP. Osteo/odontogenic differentiation of SCAP was determined using Alizarin red S staining, alkaline phosphatase staining, RT-qPCR, and western blotting. Pretreatment with FGF2 for 1 week increased the osteo/odontogenic differentiation ability of SCAP. RNA-seq and Kyoto Encyclopedia of Genes and Genomes pathway analyses revealed that phosphatidylinositol 3-kinase (PI3K)/AKT signalling is involved in the osteogenic function of FGF2. RT-qPCR results indicated that SCAP expressed FGF receptors, and western blotting showed that p-AKT was reduced in FGF2-pretreated SCAP. The activation of the PI3K/AKT pathway partially reversed the stimulatory effect of FGF2 on osteo/odontogenic differentiation of SCAP. Our findings suggest that pretreatment with FGF2 enhances the osteo/odontogenic differentiation ability of SCAP by inhibiting the PI3K/AKT pathway.


Subject(s)
Cell Differentiation , Dental Papilla , Fibroblast Growth Factor 2 , Odontogenesis , Osteogenesis , Proto-Oncogene Proteins c-akt , Signal Transduction , Stem Cells , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Differentiation/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , Dental Papilla/cytology , Dental Papilla/metabolism , Humans , Odontogenesis/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Cells, Cultured
12.
BMC Oral Health ; 24(1): 1023, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39215266

ABSTRACT

BACKGROUND: Calcium silicate-based bioceramics have been applied in endodontics as advantageous materials for years, many chemical components and new synthesizing methods were used to improve the base formulation of the materials for positively affecting the sealers properties. Recently, a novel biomaterial formulation, grounded in strontium silicate, has been introduced to the market, offering potential advancements in the field. OBJECTIVE: To comparatively analyze the cytotoxicity and cell migration effects of a novel strontium silicate-based bioceramic material (CRoot SP) and those of calcium silicate-based (iRoot SP) and epoxide amine resin (AH Plus) sealers on stem cells derived from rat apical papilla(rSCAPs). METHODS: rSCAPs were isolated and characterized in vitro and subsequently cultured in the presence of various concentrations of CRoot SP, iRoot SP and AH Plus extracts. Cytotoxicity was assessed by CCK-8 assay, and cell-migration capacity was assessed by using wound healing assays . RESULTS: No significant differences in cell viability were observed in the 0.02 mg/mL and 0.2 mg/mL sealer groups. The cell viability of CRoot SP was consistently greater than that of iRoot SP at concentrations of 5 mg/mL and 10 mg/mL across all time points. Maximum cytotoxic effect was noted on day 5 with 10 mg/mL AH Plus.The scratch was partly healed by cell migration in all groups at 24 h, and the 0.02 mg/mL, and 0.2 mg/mL CRoot SP exerted beneficial effects on rSCAPs migration. CONCLUSIONS: CRoot SP exhibited less cytotoxic than the iRoot SP and AH Plus extracts after setting. A lower concentration of CRoot SP thus promotes the cell migration capacity of rSCAPs, and it may achieve better tissue repair during root canal treatment.


Subject(s)
Calcium Compounds , Cell Movement , Cell Survival , Epoxy Resins , Root Canal Filling Materials , Silicates , Stem Cells , Animals , Silicates/pharmacology , Cell Movement/drug effects , Root Canal Filling Materials/pharmacology , Root Canal Filling Materials/toxicity , Rats , Calcium Compounds/pharmacology , Epoxy Resins/pharmacology , Epoxy Resins/toxicity , Cell Survival/drug effects , Stem Cells/drug effects , In Vitro Techniques , Materials Testing , Cells, Cultured , Ceramics/pharmacology , Strontium/pharmacology , Dental Papilla/cytology , Dental Papilla/drug effects , Tooth Apex/drug effects , Tooth Apex/cytology
13.
Stem Cell Res Ther ; 15(1): 269, 2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39183353

ABSTRACT

BACKGROUND: Liver disease imposes a significant medical burden that persists due to a shortage of liver donors and an incomplete understanding of liver disease progression. Hepatobiliary organoids (HBOs) could provide an in vitro mini-organ model to increase the understanding of the liver and may benefit the development of regenerative medicine. METHODS: In this study, we aimed to establish HBOs with bile duct (BD) structures and mature hepatocytes (MHs) using human chemically induced liver progenitor cells (hCLiPs). hCLiPs were induced in mature cryo-hepatocytes using a small-molecule cocktail of TGF-ß inhibitor (A-83-01, A), GSK3 inhibitor (CHIR99021, C), and 10% FBS (FAC). HBOs were then formed by seeding hCLiPs into ultralow attachment plates and culturing them with a combination of small molecules of Rock-inhibitor (Y-27632) and AC (YAC). RESULTS: These HBOs exhibited bile canaliculi of MHs connected to BD structures, mimicking bile secretion and transportation functions of the liver. The organoids showed gene expression patterns consistent with both MHs and BD structures, and functional assays confirmed their ability to transport the bile analogs of rhodamine-123 and CLF. Functional patient-specific HBOs were also successfully created from hCLiPs sourced from cirrhotic liver tissues. CONCLUSIONS: This study demonstrated the potential of human HBOs as an efficient model for studying hepatobiliary diseases, drug discovery, and personalized medicine.


Subject(s)
Bile Ducts , Liver , Organoids , Pyridines , Stem Cells , Humans , Organoids/metabolism , Organoids/drug effects , Bile Ducts/metabolism , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology , Pyridines/pharmacology , Liver/drug effects , Liver/metabolism , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/cytology , Pyrimidines/pharmacology , Amides/pharmacology , Cell Differentiation/drug effects , Pyrazoles , Thiosemicarbazones
14.
PeerJ ; 12: e17913, 2024.
Article in English | MEDLINE | ID: mdl-39193517

ABSTRACT

Background: Dental pulp stem cells (DPSCs) possess mesenchymal stem cell characteristics and have potential for cell-based therapy. Cell expansion is essential to achieve sufficient cell numbers. However, continuous cell replication causes cell aging in vitro, which usually accompanies and potentially affect DPSC characteristics and activities. Continuous passaging could alter susceptibility to external factors such as drug treatment. Therefore, this study sought to investigate potential outcome of in vitro passaging on DPSC morphology and activities in the absence or presence of external factor. Methods: Human DPSCs were subcultured until reaching early passages (P5), extended passages (P10), and late passages (P15). Cells were evaluated and compared for cell and nuclear morphologies, cell adhesion, proliferative capacity, alkaline phosphatase (ALP) activity, and gene expressions in the absence or presence of external factor. Alendronate (ALN) drug treatment was used as an external factor. Results: Continuous passaging of DPSCs gradually lost their normal spindle shape and increased in cell and nuclear sizes. DPSCs were vulnerable to ALN. The size and shape were altered, leading to morphological abnormality and inhomogeneity. Long-term culture and ALN interfered with cell adhesion. DPSCs were able to proliferate irrespective of cell passages but the rate of cell proliferation in late passages was slower. ALN at moderate dose inhibited cell growth. ALN caused reduction of ALP activity in early passage. In contrast, extended passage responded differently to ALN by increasing ALP activity. Late passage showed higher collagen but lower osteocalcin gene expressions compared with early passage in the presence of ALN. Conclusion: An increase in passage number played critical role in cell morphology and activities as well as responses to the addition of an external factor. The effects of cell passage should be considered when used in basic science research and clinical applications.


Subject(s)
Alendronate , Cell Adhesion , Cell Proliferation , Dental Pulp , Humans , Dental Pulp/cytology , Dental Pulp/drug effects , Cell Proliferation/drug effects , Alendronate/pharmacology , Cell Adhesion/drug effects , Alkaline Phosphatase/metabolism , Cells, Cultured , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Cell Culture Techniques/methods , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Cell Differentiation/drug effects
15.
Inflammopharmacology ; 32(5): 3443-3459, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39207637

ABSTRACT

BACKGROUND: The nucleus pulposus (NP) degradation is a primary factor in intervertebral disk degeneration (IVD) and a major contributor to low back pain. Intervertebral disk-derived stem cell (IVDSC) therapy presents a promising solution, yet identifying suitable cell carriers for NP transplantation remains challenging. The present study investigates this issue by developing smart injectable hydrogels incorporating vanillin (V) and hyaluronic acid (HA) encapsulated with IVDSCs to facilitate IVD regeneration. MATERIALS AND METHODS: The hydrogel was cross linked by carbodiimide-succinimide (EDC-NHS) method. Enhanced mechanical properties were achieved by integrating collagen and HA into the hydrogel. The rheological analysis revealed the pre-gel viscoelastic and shear-thinning characteristics. RESULTS: In vitro, cell viability was maintained up to 500 µg/mL, with a high proliferation rate observed over 14 days. The hydrogels supported multilineage differentiation, as confirmed by osteogenic and adipogenic induction. Anti-inflammatory effects were demonstrated by reduced cytokine release (TNF-α, IL-6, IL-1ß) after 24 h of treatment. Gene expression studies indicated elevated levels of chondrocyte markers (Acan, Sox9, Col2). In vivo, hydrogel injection into the NP was monitored via X-ray imaging, showing a significant increase in disk height index (DHI%) after 8 weeks, alongside improved histologic scores. Biomechanical testing revealed that the hydrogel effectively mimicked NP properties, enhancing compressive stiffness and reducing neutral zone stiffness post-denucleation. CONCLUSION: The results suggest that the synthesized VCHA-NP hydrogel can be used as an alternative to NPs, offering a promising path for IVD regeneration.


Subject(s)
Benzaldehydes , Cell Differentiation , Hydrogels , Intervertebral Disc Degeneration , Rats, Sprague-Dawley , Animals , Hydrogels/pharmacology , Hydrogels/administration & dosage , Rats , Benzaldehydes/pharmacology , Benzaldehydes/administration & dosage , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/drug therapy , Cell Differentiation/drug effects , Cell Differentiation/physiology , Stem Cells/drug effects , Nucleus Pulposus/drug effects , Disease Models, Animal , Cell Survival/drug effects , Cell Survival/physiology , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Intervertebral Disc/drug effects , Intervertebral Disc/pathology , Male , Cells, Cultured , Cell Proliferation/drug effects , Cytokines/metabolism
16.
Eur Endod J ; 9(4): 260-265, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39213455

ABSTRACT

OBJECTIVE: L-arginine is a semi-essential amino acid produced by the body which has an important role in the process of stem cell regeneration. However, under inflammatory conditions, denaturation of pulp amino acids and proteins occurr resulting in a decrease in the ability of stem cells to self-renew. Therefore, in this study, L-arginine was added in vitro to the culture media Dulbecco's Modified Eagle Medium - (DMEM) of human dental pulp stem cells (hDPSCs) to analyse the potential of L-arginine on migration and proliferation by comparing between 3 concentrations, namely 300, 400, 500 µmol/L and control group (DMEM), to obtain the most optimal concentration for proliferation and migration. METHODS: Serum-starved hDPSCs were divided into four groups: control: hDPSCs in DMEM; hDPSCs in 300 µmol/L of the L-Arginine based culture media group; hDPSCs in 400 µmol/L of the L-Arginine based culture media group; and hDPSCs in 500 µmol/L of the L-Arginine based culture media group, which were added in two separate 24-well-plates (5×104 cell/well) for proliferation and migration evaluation. The proliferation of all groups was measured by using a cell count test (haemacytometer and manual checker) after 24 h. The migratory speed rate of all groups was measured by using cell migration assay (scratch wound assay) after 24 h. Cell characteristics were evaluated under microscope that was then evaluated using image-J® interpretation. This image J represented the measurement of migratory speed rate (nm/h) data. Statistical analysis was conducted using one-way ANOVA and post hoc Bonferroni (p<0.05) for proliferation and post hoc LSD (p<0.05) for migration. RESULTS: There was a statistically significant difference in hDPSCs proliferation among various concentration groups of the L-Arginine based solution (300, 400 and 500 µmol/L) compared to the control group (p<0.05). There was a statistically significant difference in the migratory speed rate of hDPSCs at 500 µmol/L of the L-Arginine based solution group compared to lower concentrations and control group (p<0.05). CONCLUSION: All three concentrations of L-arginine can induce proliferation of hDPSCs. L-arginine at 500 µmol/L can induce higher hDPSCs proliferation and faster migration at 24 hours compared to lower concen-trations and control.


Subject(s)
Arginine , Cell Movement , Cell Proliferation , Dental Pulp , Stem Cells , Dental Pulp/cytology , Humans , Arginine/pharmacology , Cell Proliferation/drug effects , Cell Movement/drug effects , Stem Cells/drug effects , Cells, Cultured
17.
Discov Med ; 36(187): 1657-1671, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39190381

ABSTRACT

BACKGROUND: Periodontitis is the leading cause of tooth loss and can exacerbate various systemic inflammatory conditions. Periodontal ligament stem cells (PDLSCs) stand out as prominent and favorable candidates for promoting periodontal tissue regeneration. This study aimed to investigate whether the protease-activated receptor type 1 (PAR1) can mitigate the sodium butyrate (NaB)-induced PDLSCs osteogenesis inhibition and unravel the underlying mechanism. METHODS: Public datasets from the Gene Expression Omnibus (GEO) were utilized to analyze differentially expressed genes (DEGs) in periodontitis and subsequent Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. PDLSCs were cultured normally in control medium (CM) as the negative control or in osteogenic medium (OM) to induce osteogenesis. PAR1 was either activated or suppressed using a selective agonist or antagonist (OM+agonist and OM+antagonist). The evaluation of PDLSCs osteogenesis was based on the levels of osteogenesis-related markers, including runt-related transcription factor 2 (RUNX2), osterix (OSX), osteocalcin (OCN), and osteopontin (OPN), alkaline phosphatase (ALP) activity, and calcium concentration. Additionally, cell proliferation and osteogenic differentiation were measured through the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and Alizarin Red Staining. To determine the PAR1 targeting the limb development membrane protein 1 (LMBR1)/bone morphogenetic protein (BMP) pathway, LMBR1 was upregulated through cell transfection and BMP2 was inhibited using the selective inhibitor Noggin protein. Finally, NaB was introduced into PDLSCs to investigate the effect on NaB-induced inhibition of PDLSCs osteogenesis. RESULTS: PAR1, RUNX2, OSX, OCN, OPN, proliferation, ALP activity, calcium concentration, osteogenic differentiation, BMP2, and BMP4 exhibited significant increases in PDLSCs cultured in OM (p < 0.01). These parameters were further elevated by PAR1 agonist and conversely reduced by PAR1 antagonist (p < 0.01). Conversely, LMBR1 was decreased in PDLSCs cultured in OM (p < 0.001), with further reduction induced by PAR1 agonist and a reverse increase observed with PAR1 antagonist (p < 0.001). OE-LMBR1 transfection successfully elevated LMBR1 levels, subsequently inhibiting BMP2 and BMP4 (p < 0.001). Meanwhile, the Noggin protein effectively suppressed BMP2 and BMP4 (p < 0.001). All observed osteogenesis-related changes were reversed by the increased LMBR1 or inhibition of the BMP pathway (p < 0.001). Furthermore, NaB suppressed osteogenesis-related changes in OM-cultured PDLSCs (p < 0.001), and these effects were entirely reversed by PAR1 agonist (p < 0.001). Conversely, the increased LMBR1 or inhibited BMP pathway disrupted the osteogenesis reversion induced by PAR1 agonist (p < 0.001). CONCLUSION: The activation of PAR1, through suppressing LMBR1 signaling and activating BMP pathway, demonstrates the ability to enhance the osteogenesis of PDLSCs and mitigate the inhibitory effects on PDLSCs osteogenesis caused by NaB.


Subject(s)
Osteogenesis , Periodontal Ligament , Receptor, PAR-1 , Stem Cells , Humans , Bone Morphogenetic Protein 2/metabolism , Butyric Acid/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Osteogenesis/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Periodontitis/metabolism , Periodontitis/pathology , Receptor, PAR-1/metabolism , Receptor, PAR-1/genetics , Receptor, PAR-1/antagonists & inhibitors , Signal Transduction/drug effects , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology
18.
Exp Parasitol ; 265: 108823, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39187057

ABSTRACT

Trichinosis is a common parasitic disease that affects the striated skeletal muscles, causing apoptotic and degenerative changes associated with myogenin expression in the affected myocytes. Hence, this study aimed to assess the ameliorative effects of stem cells and atorvastatin added to ivermectin on the infected myocytes during the muscular phase of murine trichinosis. 120 laboratory Swiss albino male mice were divided into 10 groups, and each group was subdivided into intestinal and muscular phases (each n = 6); uninfected control; untreated infected control; infected received ivermectin monotherapy; infected received atorvastatin monotherapy; infected received stem cells monotherapy; infected received ivermectin and atorvastatin dual therapy; infected received ivermectin and stem cells dual therapy; infected received atorvastatin and stem cells dual therapy; infected received ivermectin 0.2, atorvastatin 40, and stem cells triple therapy; and infected received ivermectin 0.1, atorvastatin 20, and stem cells triple therapy. Intestinal phase mice were sacrificed on the 5th day post-infection, while those of the muscular phase were sacrificed on the 35th day post-infection. Parasitological, histopathological, ultrastructural, histochemical, biochemical, and myogenin gene expression assessments were performed. The results revealed that mice that received ivermectin, atorvastatin, and stem cell triple therapies showed the maximum reduction in the adult worm and larvae burden, marked improvement in the underlying muscular degenerative changes (as was noticed by histopathological, ultrastructural, and histochemical Feulgen stain assessment), lower biochemical levels of serum NK-κB and tissue NO, and lower myogenin expression. Accordingly, the combination of stem cells, atorvastatin, and ivermectin affords a potential synergistic activity against trichinosis with considerable healing of the underlying degenerative sequel.


Subject(s)
Apoptosis , Atorvastatin , Ivermectin , Myogenin , Trichinellosis , Animals , Atorvastatin/pharmacology , Atorvastatin/therapeutic use , Male , Mice , Ivermectin/pharmacology , Ivermectin/therapeutic use , Trichinellosis/drug therapy , Trichinellosis/parasitology , Apoptosis/drug effects , Myogenin/genetics , Myogenin/metabolism , Muscle, Skeletal/parasitology , Muscle, Skeletal/pathology , Muscle, Skeletal/drug effects , Gene Expression/drug effects , Microscopy, Electron, Transmission , Stem Cell Transplantation , Trichinella spiralis/genetics , Trichinella spiralis/drug effects , Stem Cells/drug effects
19.
Clin Oral Investig ; 28(9): 476, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39120764

ABSTRACT

OBJECTIVES: To synthesize casein enzymatic hydrolysate (CEH)-laden gelatin methacryloyl (GelMA) fibrous scaffolds and evaluate the cytocompatibility and anti-inflammatory effects on dental pulp stem cells (DPSCs). MATERIALS AND METHODS: GelMA fibrous scaffolds with 10%, 20%, and 30% CEH (w/w) and without CEH (control) were obtained via electrospinning. Chemo-morphological, degradation, and mechanical analyses were conducted to evaluate the morphology and composition of the fibers, mass loss, and mechanical properties, respectively. Adhesion/spreading and viability of DPSCs seeded on the scaffolds were also assessed. The anti-inflammatory potential on DPSCs was tested after the chronic challenge of cells with lipopolysaccharides (LPS), followed by treatment with extracts obtained after immersing the scaffolds in α-MEM. The synthesis of the pro-inflammatory cytokines IL-6, IL-1α, and TNF-α was measured by ELISA. Data were analyzed by ANOVA/post-hoc tests (α = 5%). RESULTS: CEH-laden electrospun fibers had a larger diameter than pure GelMA (p ≤ 0.036). GelMA scaffolds laden with 20% and 30% CEH had a greater mass loss. Tensile strength was reduced for the 10% CEH fibers (p = 0.0052), whereas no difference was observed for the 20% and 30% fibers (p ≥ 0.6736) compared to the control. Young's modulus decreased with CEH (p < 0.0001). Elongation at break increased for the 20% and 30% CEH scaffolds (p ≤ 0.0038). Over time, DPSCs viability increased across all groups, indicating cytocompatibility, with CEH-laden scaffolds exhibiting greater cell viability after seven days (p ≤ 0.0166). Also, 10% CEH-GelMA scaffolds decreased the IL-6, IL-1α, and TNF-α synthesis (p ≤ 0.035). CONCLUSION: CEH-laden GelMA scaffolds facilitated both adhesion and proliferation of DPSCs, and 10% CEH provided anti-inflammatory potential after chronic LPS challenge. CLINICAL RELEVANCE: CEH incorporated in GelMA fibrous scaffolds demonstrated the potential to be used as a cytocompatible and anti-inflammatory biomaterial for vital pulp therapy.


Subject(s)
Anti-Inflammatory Agents , Caseins , Cell Survival , Dental Pulp , Gelatin , Tissue Scaffolds , Gelatin/chemistry , Dental Pulp/cytology , Dental Pulp/drug effects , Tissue Scaffolds/chemistry , Humans , Anti-Inflammatory Agents/pharmacology , Cell Survival/drug effects , Methacrylates/chemistry , Materials Testing , Enzyme-Linked Immunosorbent Assay , Tensile Strength , Cells, Cultured , Stem Cells/drug effects , Cell Adhesion/drug effects , Biocompatible Materials/pharmacology , Biocompatible Materials/chemistry , Cytokines/metabolism , Surface Properties
20.
Tissue Cell ; 90: 102508, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128193

ABSTRACT

BACKGROUND: The present study aimed to evaluate the viability of human dental pulp stem cells (hDPSCs) exposed to boric acid (BA) and injectable platelet-rich fibrin (I-PRF). MATERIALS AND METHODS: hDPSCs were isolated from impacted third molars. Nine milliliters of whole blood was transferred to I-PRF tubes and centrifuged at 700 rpm for 3 minutes. A BA solution was prepared by dissolving BA in a 0.1 g/ml stock solution. The cells were divided into four groups: control, I-PRF, BA, and BA + I-PRF. Cell viability was evaluated using flow cytometry. Mineralized calcium nodules were observed using Alizarin Red staining. The data were analyzed using two-way analysis of variance and Tukey's HSD test (p<0.05). RESULTS: The highest percentage of viable cells was in the I-PRF group, and the lowest percentage of viable cells was in the BA group at all times. Larger calcium nodules were observed in the BA group compared to the other groups. CONCLUSION: The use of I-PRF with or without BA had a positive effect on cell viability. BA and I-PRF affected the formation of mineralized calcium nodules. I-PRF and BA may be used in combination because these substances minimally reduce cell viability and promote mineralized nodule formation.


Subject(s)
Boric Acids , Cell Survival , Dental Pulp , Platelet-Rich Fibrin , Stem Cells , Humans , Dental Pulp/cytology , Dental Pulp/drug effects , Boric Acids/pharmacology , Cell Survival/drug effects , Stem Cells/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Calcification, Physiologic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL