Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72.963
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 Cancer ; 23(1): 153, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090713

ABSTRACT

The hallmarks of stem cells, such as proliferation, self-renewal, development, differentiation, and regeneration, are critical to maintain stem cell identity which is sustained by genetic and epigenetic factors. Super-enhancers (SEs), which consist of clusters of active enhancers, play a central role in maintaining stemness hallmarks by specifically transcriptional model. The SE-navigated transcriptional complex, including SEs, non-coding RNAs, master transcriptional factors, Mediators and other co-activators, forms phase-separated condensates, which offers a toggle for directing diverse stem cell fate. With the burgeoning technologies of multiple-omics applied to examine different aspects of SE, we firstly raise the concept of "super-enhancer omics", inextricably linking to Pan-omics. In the review, we discuss the spatiotemporal organization and concepts of SEs, and describe links between SE-navigated transcriptional complex and stem cell features, such as stem cell identity, self-renewal, pluripotency, differentiation and development. We also elucidate the mechanism of stemness and oncogenic SEs modulating cancer stem cells via genomic and epigenetic alterations hijack in cancer stem cell. Additionally, we discuss the potential of targeting components of the SE complex using small molecule compounds, genome editing, and antisense oligonucleotides to treat SE-associated organ dysfunction and diseases, including cancer. This review also provides insights into the future of stem cell research through the paradigm of SEs.


Subject(s)
Enhancer Elements, Genetic , Stem Cells , Humans , Animals , Stem Cells/metabolism , Stem Cells/cytology , Genomics/methods , Epigenesis, Genetic , Cell Differentiation/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology
3.
Int Ophthalmol ; 44(1): 337, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093517

ABSTRACT

PURPOSE: To evaluate limbal graft transplantation success in pediatric patients with chemical injury-induced limbal stem cell deficiency (LSCD) using the 'LSCD Working Group' staging system. METHODS: Medical records of 11 eyes of 11 children who underwent limbal graft transplantation (limbal autograft/limbal allograft) were included. Surgical success was defined as improvement in the post-operative 1st year LSCD stage. RESULTS: The mean age was 12 ± 5 (4-17) years. Causative agent was alkaline in 4(36.4%) and acid in 3(27.2%) patients. Limbal autograft was performed in 9 (81.8%) eyes with unilateral LSCD, and allograft transplantation was performed in 2 (18.2%) eyes with bilateral LSCD. The mean follow-up time was 33.89 ± 30.73 (12-102.33) months. The overall limbal graft transplantation success rate was 72.7%. Among 9 patients who receive limbal autograft, 8 had improvement in post-operative LSCD stage, 1 had stable LSCD stage. Of the 2 patients who receive limbal allograft, post-operative LSCD stage remained the same in 1 and worsened in 1 patient. The mean time between injury and the surgery was 30.47 ± 30.08 (7-108.47) months. Penetrating keratoplasty was performed in 3 (27.2%) of 11 patients following limbal graft transplantation. CONCLUSION: Management of LSCD in children is challenging and appears to be somewhat different from that of adults. Limited data in the literature indicate that cultivated or simple limbal epithelial transplantations (CLET/SLET) are primarily preferred in children. Although the tendency to take small tissue from the healthy eye is noteworthy, conventional limbal allograft and autograft transplantations also show promising results without any further complications in at least 1 year follow-up period.


Subject(s)
Limbal Stem Cell Deficiency , Limbus Corneae , Visual Acuity , Adolescent , Child , Child, Preschool , Female , Humans , Male , Burns, Chemical/surgery , Corneal Transplantation/methods , Eye Burns/surgery , Eye Burns/chemically induced , Eye Burns/diagnosis , Follow-Up Studies , Limbal Stem Cell Deficiency/chemically induced , Limbal Stem Cell Deficiency/diagnosis , Limbal Stem Cell Deficiency/surgery , Limbus Corneae/cytology , Retrospective Studies , Stem Cell Transplantation/methods , Stem Cells/cytology , Transplantation, Autologous , Treatment Outcome
5.
Sci Adv ; 10(33): eado0424, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39151009

ABSTRACT

In animals, stem cell populations of varying potency facilitate regeneration and tissue homeostasis. Notably, germline stem cells in both vertebrates and invertebrates express highly conserved RNA binding proteins, such as nanos, vasa, and piwi. In highly regenerative animals, these genes are also expressed in somatic stem cells, which led to the proposal that they had an ancestral role in all stem cells. In cnidarians, multi- and pluripotent interstitial stem cells have only been identified in hydrozoans. Therefore, it is currently unclear if cnidarian stem cell systems share a common evolutionary origin. We, therefore, aimed to characterize conserved stem cell marker genes in the sea anemone Nematostella vectensis. Through transgenic reporter genes and single-cell transcriptomics, we identify cell populations expressing the germline-associated markers piwi1 and nanos2 in the soma and germline, and gene knockout shows that Nanos2 is indispensable for germline formation. This suggests that nanos and piwi genes have a conserved role in somatic and germline stem cells in cnidarians.


Subject(s)
Germ Cells , RNA-Binding Proteins , Sea Anemones , Animals , Sea Anemones/genetics , Sea Anemones/metabolism , Germ Cells/metabolism , Germ Cells/cytology , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Lineage/genetics , Stem Cells/metabolism , Stem Cells/cytology , Argonaute Proteins/metabolism , Argonaute Proteins/genetics
6.
J Transl Med ; 22(1): 762, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143486

ABSTRACT

BACKGROUND: Personalized disease models are crucial for evaluating how diseased cells respond to treatments, especially in case of innovative biological therapeutics. Extracellular vesicles (EVs), nanosized vesicles released by cells for intercellular communication, have gained therapeutic interest due to their ability to reprogram target cells. We here utilized urinary podocytes obtained from children affected by steroid-resistant nephrotic syndrome with characterized genetic mutations as a model to test the therapeutic potential of EVs derived from kidney progenitor cells (nKPCs). METHODS: EVs were isolated from nKPCs derived from the urine of a preterm neonate. Three lines of urinary podocytes obtained from nephrotic patients' urine and a line of Alport syndrome patient podocytes were characterized and used to assess albumin permeability in response to nKPC-EVs or various drugs. RNA sequencing was conducted to identify commonly modulated pathways after nKPC-EV treatment. siRNA transfection was used to demonstrate the involvement of SUMO1 and SENP2 in the modulation of permeability. RESULTS: Treatment with the nKPC-EVs significantly reduced permeability across all the steroid-resistant patients-derived and Alport syndrome-derived podocytes. At variance, podocytes appeared unresponsive to standard pharmacological treatments, with the exception of one line, in alignment with the patient's clinical response at 48 months. By RNA sequencing, only two genes were commonly upregulated in nKPC-EV-treated genetically altered podocytes: small ubiquitin-related modifier 1 (SUMO1) and Sentrin-specific protease 2 (SENP2). SUMO1 and SENP2 downregulation increased podocyte permeability confirming the role of the SUMOylation pathway. CONCLUSIONS: nKPCs emerge as a promising non-invasive source of EVs with potential therapeutic effects on podocytes with genetic dysfunction, through modulation of SUMOylation, an important pathway for the stability of podocyte slit diaphragm proteins. Our findings also suggest the feasibility of developing a non-invasive in vitro model for screening regenerative compounds on patient-derived podocytes.


Subject(s)
Extracellular Vesicles , Nephrotic Syndrome , Podocytes , Podocytes/metabolism , Podocytes/drug effects , Podocytes/pathology , Humans , Nephrotic Syndrome/pathology , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/metabolism , Extracellular Vesicles/metabolism , Drug Evaluation, Preclinical , Models, Biological , Stem Cells/metabolism , Steroids/pharmacology , Kidney/pathology , Kidney/metabolism , Drug Resistance , Infant, Newborn , Male
7.
J Ovarian Res ; 17(1): 167, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39153978

ABSTRACT

BACKGROUND: Polycystic ovary syndrome (PCOS) presents a significant challenge in women's reproductive health, characterized by disrupted folliculogenesis and ovulatory dysfunction. Central to PCOS pathogenesis are granulosa cells, whose dysfunction contributes to aberrant steroid hormone production and oxidative stress. Mitochondrial dysfunction emerges as a key player, influencing cellular energetics, oxidative stress, and steroidogenesis. This study investigates the therapeutic potential of menstrual blood-derived stem cells (MenSCs) and their exosomes in mitigating mitochondrial dysfunction and oxidative stress in PCOS granulosa cells. METHODS: Using a rat model of PCOS induced by letrozole, granulosa cells were harvested and cultured. MenSCs and their exosomes were employed to assess their effects on mitochondrial biogenesis, oxidative stress, and estrogen production in PCOS granulosa cells. RESULTS: Results showed diminished mitochondrial biogenesis and increased oxidative stress in PCOS granulosa cells, alongside reduced estrogen production. Treatment with MenSCs and their exosomes demonstrated significant improvements in mitochondrial biogenesis, oxidative stress levels, and estrogen production in PCOS granulosa cells. Further analysis showed MenSCs' superior efficacy over exosomes, attributed to their sustained secretion of bioactive factors. Mechanistically, MenSCs and exosomes activated pathways related to mitochondrial biogenesis and antioxidative defense, highlighting their therapeutic potential for PCOS. CONCLUSIONS: This study offers insights into granulosa cells mitochondria's role in PCOS pathogenesis and proposes MenSCs and exosomes as a potential strategy for mitigating mitochondrial dysfunction and oxidative stress in PCOS. Further research is needed to understand underlying mechanisms and validate clinical efficacy, presenting promising avenues for addressing PCOS complexity.


Subject(s)
Exosomes , Granulosa Cells , Mitochondria , Oxidative Stress , Polycystic Ovary Syndrome , Polycystic Ovary Syndrome/metabolism , Female , Granulosa Cells/metabolism , Exosomes/metabolism , Mitochondria/metabolism , Rats , Animals , Humans , Menstruation , Stem Cells/metabolism , Letrozole/pharmacology , Disease Models, Animal
8.
Nat Commun ; 15(1): 7097, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154007

ABSTRACT

Converging evidence indicates that extra-embryonic yolk sac is the source of both macrophages and endothelial cells in adult mouse tissues. Prevailing views are that these embryonically derived cells are maintained after birth by proliferative self-renewal in their differentiated states. Here we identify clonogenic endothelial-macrophage (EndoMac) progenitor cells in the adventitia of embryonic and postnatal mouse aorta, that are independent of Flt3-mediated bone marrow hematopoiesis and derive from an early embryonic CX3CR1+ and CSF1R+ source. These bipotent progenitors are proliferative and vasculogenic, contributing to adventitial neovascularization and formation of perfused blood vessels after transfer into ischemic tissue. We establish a regulatory role for angiotensin II, which enhances their clonogenic and differentiation properties and rapidly stimulates their proliferative expansion in vivo. Our findings demonstrate that embryonically derived EndoMac progenitors participate in local vasculogenic responses in the aortic wall by contributing to the expansion of endothelial cells and macrophages postnatally.


Subject(s)
Aorta , Macrophages , Animals , Macrophages/cytology , Macrophages/metabolism , Aorta/cytology , Mice , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Cell Differentiation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Angiotensin II , Cell Proliferation , Stem Cells/cytology , Stem Cells/metabolism , Mice, Inbred C57BL , Female , Neovascularization, Physiologic , Receptors, Chemokine/metabolism , Receptors, Chemokine/genetics , Male , Hematopoiesis/physiology , fms-Like Tyrosine Kinase 3
9.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39158015

ABSTRACT

Clémentine Villeneuve is a Postdoctoral Researcher at the Max Planck Institute for Molecular Biomedicine, Germany, where her research focuses on the role of mechanobiology in driving stem cell fate specification and tissue patterning during development. Clémentine is one of the fellows of Development's Pathway to Independence Programme, and we caught up with her over Zoom to find out more about her research background, her hopes for the programme and her future research questions.


Subject(s)
Developmental Biology , Developmental Biology/history , History, 21st Century , Humans , History, 20th Century , Animals , Germany , Stem Cells , Cell Differentiation
10.
J Vis Exp ; (210)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39158276

ABSTRACT

Endometrial organoids offer valuable insights into the development and pathophysiology of endometrial diseases and serve as platforms for drug testing. While human and mouse endometrial organoids have been developed, research on rat endometrial organoids remains limited. Given that rats can better simulate certain endometrial pathologies, such as intrauterine adhesions, this study aimed to establish rat endometrial organoids. We present a detailed protocol for the isolation and culture of rat endometrial epithelial stem cells (reESCs) and the generation of rat endometrial organoids. Using a refined reESCs expansion medium, we successfully isolated and stably expanded reESCs, demonstrating their long-term culture potential. The reESC-generated organoids exhibited typical structural and functional characteristics of the endometrium, including hormone responsiveness. Our results showed that rat endometrial organoids could be cultured over a long term with stable proliferation, maintaining the glandular structure, cell polarity, and functional characteristics of the endometrial epithelium. This novel rat-derived endometrial organoid model provides a valuable platform for studying endometrial diseases and testing therapeutic interventions, with potential applications across various mammalian species.


Subject(s)
Endometrium , Epithelial Cells , Organoids , Animals , Female , Organoids/cytology , Rats , Endometrium/cytology , Epithelial Cells/cytology , Stem Cells/cytology , Uterus/cytology
11.
Invest Ophthalmol Vis Sci ; 65(10): 12, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39106056

ABSTRACT

Purpose: The role of specific extracellular matrix (ECM) molecules in lens cell development and regeneration is poorly understood, as appropriate cellular models are lacking. Here, a laminin-based lens cell in vitro induction system was developed to study the role of laminin in human lens epithelial stem/progenitor cell (LES/PC) development. Methods: The human embryonic stem cell-based lens induction system followed a three-stage protocol. The expression profile of laminins during lens induction was screened, and laminin-511 (LN511) was tested as a candidate substitute. LN511 induction system cellular and molecular features, including induction efficiency, transcription factor expression related to different lens development stages, ECM alterations, and Hippo/YAP signaling, were evaluated. Results: LAMA5, LAMB1, and LAMC1 were highly expressed around the time of LES/PC derivation. We chose LN511 (product of LAMA5, LAMB1, and LAMC1) and found that it considerably enhanced lens cell induction efficiency, compared to that in Matrigel-coated culture, as more and larger lentoid bodies were detected. Notably, LES/PC induction efficiency improved by promoting lens specification-related transcription factor expression and cell proliferation. Transcriptome analysis revealed that compared to those with Matrigel, ECM accumulation and cell adhesion were downregulated in the LN511 system. Hippo/YAP signaling was hypoactive during LES/P-like cell generation, and small molecule inhibitors of YAP/TAZ activity upregulated LES/PC marker expression and promoted the efficiency of LES/P-like cell derivation. Conclusions: The laminin isoform LN511 is a reliable substitute for the LES/P-like cell induction system, and LN511-YAP acted as efficient modulators of LES/PC derivation; this contributes to knowledge of the role of the ECM in human lens development.


Subject(s)
Cell Differentiation , Cell Proliferation , Epithelial Cells , Laminin , Lens, Crystalline , Humans , Laminin/metabolism , Lens, Crystalline/cytology , Lens, Crystalline/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Cells, Cultured , Signal Transduction , Stem Cells/metabolism , Stem Cells/cytology , Transcription Factors/metabolism , Transcription Factors/genetics , Extracellular Matrix/metabolism
12.
BMC Biol ; 22(1): 157, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39090637

ABSTRACT

BACKGROUND: The remarkable regenerative abilities observed in planarians and cnidarians are closely linked to the active proliferation of adult stem cells and the precise differentiation of their progeny, both of which typically deteriorate during aging in low regenerative animals. While regeneration-specific genes conserved in highly regenerative organisms may confer regenerative abilities and long-term maintenance of tissue homeostasis, it remains unclear whether introducing these regenerative genes into low regenerative animals can improve their regeneration and aging processes. RESULTS: Here, we ectopically express highly regenerative species-specific JmjC domain-encoding genes (HRJDs) in Drosophila, a widely used low regenerative model organism. Surprisingly, HRJD expression impedes tissue regeneration in the developing wing disc but extends organismal lifespan when expressed in the intestinal stem cell lineages of the adult midgut under non-regenerative conditions. Notably, HRJDs enhance the proliferative activity of intestinal stem cells while maintaining their differentiation fidelity, ameliorating age-related decline in gut barrier functions. CONCLUSIONS: These findings together suggest that the introduction of highly regenerative species-specific genes can improve stem cell functions and promote a healthy lifespan when expressed in aging animals.


Subject(s)
Regeneration , Animals , Regeneration/genetics , Regeneration/physiology , Aging/genetics , Aging/physiology , Species Specificity , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Drosophila melanogaster/growth & development , Stem Cells/metabolism , Intestines/physiology , Cell Differentiation/genetics , Cell Proliferation
13.
Sci Adv ; 10(32): eadl1584, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110797

ABSTRACT

Histone acetyltransferases KAT2A and KAT2B are paralogs highly expressed in the intestinal epithelium, but their functions are not well understood. In this study, double knockout of murine Kat2 genes in the intestinal epithelium was lethal, resulting in robust activation of interferon signaling and interferon-associated phenotypes including the loss of intestinal stem cells. Use of pharmacological agents and sterile organoid cultures indicated a cell-intrinsic double-stranded RNA trigger for interferon signaling. Acetyl-proteomics and sequencing of immunoprecipitated double-stranded RNA were used to interrogate the mechanism behind this response, which identified mitochondria-encoded double-stranded RNA as the source of intrinsic interferon signaling. Kat2a and Kat2b therefore play an essential role in regulating mitochondrial functions and maintaining intestinal health.


Subject(s)
Histone Acetyltransferases , Interferons , Mice, Knockout , RNA, Double-Stranded , Signal Transduction , Stem Cells , Animals , RNA, Double-Stranded/metabolism , Mice , Stem Cells/metabolism , Stem Cells/cytology , Interferons/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/cytology , Mitochondria/metabolism , Cell Self Renewal/genetics , Intestines/cytology
14.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125629

ABSTRACT

Photoreceptor degeneration is a major cause of untreatable blindness worldwide and has recently been targeted by emerging technologies, including cell- and gene-based therapies. Cell types of neural lineage have shown promise for replacing either photoreceptors or retinal pigment epithelial cells following delivery to the subretinal space, while cells of bone marrow lineage have been tested for retinal trophic effects following delivery to the vitreous cavity. Here we explore an alternate approach in which cells from the immature neural retinal are delivered to the vitreous cavity with the goal of providing trophic support for degenerating photoreceptors. Rat and human retinal progenitor cells were transplanted to the vitreous of rats with a well-studied photoreceptor dystrophy, resulting in substantial anatomical preservation and functional rescue of vision. This work provides scientific proof-of-principle for a novel therapeutic approach to photoreceptor degeneration that is currently being evaluated in clinical trials.


Subject(s)
Retina , Retinal Degeneration , Stem Cell Transplantation , Animals , Rats , Retinal Degeneration/therapy , Retinal Degeneration/pathology , Stem Cell Transplantation/methods , Humans , Retina/pathology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/transplantation , Disease Models, Animal
15.
Stem Cell Res Ther ; 15(1): 249, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113098

ABSTRACT

Inflammation is a key pathological feature of many diseases, disrupting normal tissue structure and resulting in irreversible damage. Despite the need for effective inflammation control, current treatments, including stem cell therapies, remain insufficient. Recently, extracellular vesicles secreted by adipose-derived stem cells (ADSC-EVs) have garnered attention for their significant anti-inflammatory properties. As carriers of bioactive substances, these vesicles have demonstrated potent capabilities in modulating inflammation and promoting tissue repair in conditions such as rheumatoid arthritis, osteoarthritis, diabetes, cardiovascular diseases, stroke, and wound healing. Consequently, ADSC-EVs are emerging as promising alternatives to conventional ADSC-based therapies, offering advantages such as reduced risk of immune rejection, enhanced stability, and ease of storage and handling. However, the specific mechanisms by which ADSC-EVs regulate inflammation under pathological conditions are not fully understood. This review discusses the role of ADSC-EVs in inflammation control, their impact on disease prognosis, and their potential to promote tissue repair. Additionally, it provides insights into future clinical research focused on ADSC-EV therapies for inflammatory diseases, which overcome some limitations associated with cell-based therapies.


Subject(s)
Adipose Tissue , Extracellular Vesicles , Inflammation , Humans , Extracellular Vesicles/metabolism , Inflammation/therapy , Inflammation/metabolism , Inflammation/pathology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Stem Cells/metabolism , Stem Cells/cytology , Wound Healing
16.
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
17.
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
18.
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
19.
Nat Commun ; 15(1): 7145, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39164270

ABSTRACT

The existence and function of Lgr5+ cells within the developing esophagus remains unknown. Here, we document multiple discrete Lgr5+ populations in the developing mouse esophagus, predominantly within nascent epithelial and external muscle layers. Lgr5 expression initially emerges in the developing proximal embryonic epithelium, but progressively extends distally and persists within the distal epithelium of neonates. Fate mapping and ablation analyses reveal a long-term contribution of epithelial Lgr5+ cells to esophageal organogenesis. Additionally, Lgr5-expressing cells are present in the developing external muscle layer, particularly during the development of the striated component. Fate mapping reveals a significant contribution of these embryonic Lgr5+ cells to the adult muscle layer. Embryonic Lgr5+ epithelial cells are also found to be important for regulating epithelial development, serving as a key source of Wnt6, among other ligands, to promote epithelial cell proliferation and formation of epithelial layers. These findings significantly enhance our understanding of esophageal development and shed light on the involvement of Lgr5+ stem/progenitor cells during organogenesis. Importantly, this study lays the foundation for investigating esophageal diseases related to the Lgr5+ stem/progenitor cell pool.


Subject(s)
Epithelial Cells , Esophagus , Muscle Development , Receptors, G-Protein-Coupled , Stem Cells , Animals , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Esophagus/cytology , Esophagus/metabolism , Esophagus/embryology , Mice , Stem Cells/metabolism , Stem Cells/cytology , Epithelial Cells/metabolism , Epithelial Cells/cytology , Cell Proliferation , Organogenesis , Epithelium/metabolism , Epithelium/embryology , Female , Gene Expression Regulation, Developmental , Cell Differentiation
20.
J Transl Med ; 22(1): 779, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39169334

ABSTRACT

The skin epidermis is continually influenced by a myriad of internal and external elements. At its basal layer reside epidermal stem cells, which fuels epidermal renovation and hair regeneration with powerful self-renewal ability, as well as keeping diverse signals that direct their activity under surveillance with quick response. The importance of epidermal stem cells in wound healing and immune-related skin conditions has been increasingly recognized, and their potential for clinical applications is attracting attention. In this review, we delve into recent advancements and the various physiological and psychological factors that govern distinct epidermal stem cell populations, including psychological stress, mechanical forces, chronic aging, and circadian rhythm, as well as providing an overview of current methodological approaches. Furthermore, we discuss the pathogenic role of epidermal stem cells in immune-related skin disorders and their potential clinical applications.


Subject(s)
Epidermal Cells , Stem Cells , Humans , Stem Cells/cytology , Animals , Epidermis , Skin/pathology , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL