Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69.938
Filtrar
1.
J Contemp Dent Pract ; 25(3): 267-275, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38690701

RESUMEN

AIM: To study the effect of glycyrrhizin (GA) on the viability and proliferation of dental pulp stem cells (DPSCs) compared with intracanal medicaments. MATERIALS AND METHODS: Third molars of an adult donor were used to obtain the DPSCs. Flow cytometry was utilized to conduct phenotypic analysis for DPSCs. The methyl-thiazol tetrazolium (MTT) test was used to detect the cell viability. Cell proliferation assay was conducted at distinct time intervals: 3, 5, and 7 days. RESULTS: The flow cytometry analysis verified the positive expression of mesenchymal cell surface antigen molecules (CD73, CD90, and CD105) and the absence of hematological markers (CD14, CD34, and CD45) in the DPSCs. The cells that treated with concentrations more than 0.5 mg/mL of Ca(OH2) and triple antibiotic paste (TAP) gave significant decrease in viability in comparison to the untreated cells (p < 0.05). Also, the cells treated with concentrations 50 and 25 µM of GA showed no significant difference compared with the untreated cells (p > 0.05), while concentrations 12.5 and 6.25 µM expressed a significant increase in viability compared with the untreated cells (p < 0.05). At 7 days, cells treated with the three different concentrations of GA (12.5, 25, and 50 µM) demonstrated a significant increase in cell density compared with Ca(OH)2 and TAP-treated cells (p < 0.05). CONCLUSION: Based upon the potential of GA on DPSCs proliferation compared with Ca(OH)2 and TAP, It is conceivable to acknowledge that GA could be used as an intracanal medicaments for revascularization process of necrotic immature teeth. CLINICAL SIGNIFICANCE: This study emphasizes the significance of assessing alternative root canal medicaments and their impact on the proliferation and viability of DPSCs. The results regarding GA, specifically its impact on the viability and growth of DPSCs, provide essential understanding for its potential application as an intracanal medicine. This study adds to the continuous endeavors in identifying safer and more efficient intracanal therapies, which are essential for improving patient outcomes in endodontic operations. How to cite this article: Alrashidi MA, Badawi MF, Elbeltagy MG, et al. The Effect of Glycyrrhizin on the Viability and Proliferation of Dental Pulp Stem Cells Compared to Intracanal Medicaments. J Contemp Dent Pract 2024;25(3):267-275.


Asunto(s)
Proliferación Celular , Supervivencia Celular , Pulpa Dental , Ácido Glicirrínico , Irrigantes del Conducto Radicular , Células Madre , Humanos , Pulpa Dental/citología , Pulpa Dental/efectos de los fármacos , Ácido Glicirrínico/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Irrigantes del Conducto Radicular/farmacología , Células Madre/efectos de los fármacos , Citometría de Flujo , Hidróxido de Calcio/farmacología , Células Cultivadas , Adulto
2.
Cell Stem Cell ; 31(5): 591-592, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38701755

RESUMEN

Recently in Cell Metabolism, Wei et al.1 unveiled a brain-to-gut pathway that conveys psychological stress to intestinal epithelial cells, leading to their dysfunction. This gut-brain axis involves a microbial metabolite, indole-3-acetate (IAA), as a niche signal that hampers mitochondrial respiration to skew intestinal stem cell (ISC) fate.


Asunto(s)
Células Madre , Células Madre/metabolismo , Células Madre/citología , Animales , Humanos , Intestinos/citología , Intestinos/microbiología , Estrés Fisiológico , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Diferenciación Celular , Mitocondrias/metabolismo
3.
J Nanobiotechnology ; 22(1): 215, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38693585

RESUMEN

Stem cells (SCs) have been used therapeutically for decades, yet their applications are limited by factors such as the risk of immune rejection and potential tumorigenicity. Extracellular vesicles (EVs), a key paracrine component of stem cell potency, overcome the drawbacks of stem cell applications as a cell-free therapeutic agent and play an important role in treating various diseases. However, EVs derived from two-dimensional (2D) planar culture of SCs have low yield and face challenges in large-scale production, which hinders the clinical translation of EVs. Three-dimensional (3D) culture, given its ability to more realistically simulate the in vivo environment, can not only expand SCs in large quantities, but also improve the yield and activity of EVs, changing the content of EVs and improving their therapeutic effects. In this review, we briefly describe the advantages of EVs and EV-related clinical applications, provide an overview of 3D cell culture, and finally focus on specific applications and future perspectives of EVs derived from 3D culture of different SCs.


Asunto(s)
Técnicas de Cultivo Tridimensional de Células , Vesículas Extracelulares , Células Madre , Vesículas Extracelulares/metabolismo , Humanos , Células Madre/citología , Células Madre/metabolismo , Animales , Técnicas de Cultivo Tridimensional de Células/métodos , Técnicas de Cultivo de Célula/métodos
4.
Int J Biol Sci ; 20(7): 2339-2355, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725853

RESUMEN

Chronic cholestatic damage is associated to both accumulation of cytotoxic levels of bile acids and expansion of adult hepatic progenitor cells (HPC) as part of the ductular reaction contributing to the regenerative response. Here, we report a bile acid-specific cytotoxic response in mouse HPC, which is partially impaired by EGF signaling. Additionally, we show that EGF synergizes with bile acids to trigger inflammatory signaling and NLRP3 inflammasome activation in HPC. Aiming at understanding the impact of this HPC specific response on the liver microenvironment we run a proteomic analysis of HPC secretome. Data show an enrichment in immune and TGF-ß regulators, ECM components and remodeling proteins in HPC secretome. Consistently, HPC-derived conditioned medium promotes hepatic stellate cell (HSC) activation and macrophage M1-like polarization. Strikingly, EGF and bile acids co-treatment leads to profound changes in the secretome composition, illustrated by an abolishment of HSC activating effect and by promoting macrophage M2-like polarization. Collectively, we provide new specific mechanisms behind HPC regulatory action during cholestatic liver injury, with an active role in cellular interactome and inflammatory response regulation. Moreover, findings prove a key contribution for EGFR signaling jointly with bile acids in HPC-mediated actions.


Asunto(s)
Ácidos y Sales Biliares , Receptores ErbB , Inflamación , Ratones Endogámicos C57BL , Transducción de Señal , Animales , Ácidos y Sales Biliares/metabolismo , Receptores ErbB/metabolismo , Ratones , Inflamación/metabolismo , Células Madre/metabolismo , Hígado/metabolismo , Hígado/patología , Masculino , Proteómica , Macrófagos/metabolismo , Células Estrelladas Hepáticas/metabolismo
5.
Commun Biol ; 7(1): 525, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702433

RESUMEN

Disabled 2 (Dab2), an adaptor protein, is up regulated in the hair follicle stem cells (HFSCs); however, its role in any tissue stem cells has not been studied. In the present study, we have reported that Dab2 conditional knockout (Dab2-cKO) mice exhibited a delay in the HF cycle due to perturbed activation of HFSCs. Further, Dab2-cKO mice showed a reduction in the number of HFSCs and reduced colony forming ability of HFSCs. Dab2-cKO mice showed extended quiescence of HFSCs concomitant with an increased expression of Nfatc1. Dab2-cKO mice showed a decreased expression of anti-aging genes such as Col17a1, decorin, Sirt2 and Sirt7. Dab2-cKO mice did not show full hair coat recovery in aged mice thereby suggesting an accelerated aging process. Overall, we unveil for the first time, the role of Dab2 that regulate activation and self-renewal of HFSCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Folículo Piloso , Ratones Noqueados , Células Madre , Animales , Folículo Piloso/metabolismo , Folículo Piloso/citología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Ratones , Células Madre/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Reguladoras de la Apoptosis/genética , Autorrenovación de las Células/genética , Ratones Endogámicos C57BL , Proliferación Celular
6.
Breast Cancer Res ; 26(1): 74, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702730

RESUMEN

The transcription factor TRPS1 is a context-dependent oncogene in breast cancer. In the mammary gland, TRPS1 activity is restricted to the luminal population and is critical during puberty and pregnancy. Its function in the resting state remains however unclear. To evaluate whether it could be a target for cancer therapy, we investigated TRPS1 function in the healthy adult mammary gland using a conditional ubiquitous depletion mouse model where long-term depletion does not affect fitness. Using transcriptomic approaches, flow cytometry and functional assays, we show that TRPS1 activity is essential to maintain a functional luminal progenitor compartment. This requires the repression of both YAP/TAZ and SRF/MRTF activities. TRPS1 represses SRF/MRTF activity indirectly by modulating RhoA activity. Our work uncovers a hitherto undisclosed function of TRPS1 in luminal progenitors intrinsically linked to mechanotransduction in the mammary gland. It may also provide new insights into the oncogenic functions of TRPS1 as luminal progenitors are likely the cells of origin of many breast cancers.


Asunto(s)
Glándulas Mamarias Animales , Proteínas Represoras , Factor de Respuesta Sérica , Células Madre , Factores de Transcripción , Animales , Femenino , Ratones , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Células Madre/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Factor de Respuesta Sérica/metabolismo , Factor de Respuesta Sérica/genética , Humanos , Transactivadores/metabolismo , Transactivadores/genética
7.
Int J Nanomedicine ; 19: 3773-3804, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38708181

RESUMEN

Geriatric diseases are a group of diseases with unique characteristics related to senility. With the rising trend of global aging, senile diseases now mainly include endocrine, cardiovascular, neurodegenerative, skeletal, and muscular diseases and cancer. Compared with younger populations, the structure and function of various cells, tissues and organs in the body of the elderly undergo a decline as they age, rendering them more susceptible to external factors and diseases, leading to serious tissue damage. Tissue damage presents a significant obstacle to the overall health and well-being of older adults, exerting a profound impact on their quality of life. Moreover, this phenomenon places an immense burden on families, society, and the healthcare system.In recent years, stem cell-derived exosomes have become a hot topic in tissue repair research. The combination of these exosomes with biomaterials allows for the preservation of their biological activity, leading to a significant improvement in their therapeutic efficacy. Among the numerous biomaterial options available, hydrogels stand out as promising candidates for loading exosomes, owing to their exceptional properties. Due to the lack of a comprehensive review on the subject matter, this review comprehensively summarizes the application and progress of combining stem cell-derived exosomes and hydrogels in promoting tissue damage repair in geriatric diseases. In addition, the challenges encountered in the field and potential prospects are presented for future advancements.


Asunto(s)
Exosomas , Hidrogeles , Células Madre , Exosomas/química , Humanos , Hidrogeles/química , Anciano , Envejecimiento/fisiología , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Geriatría
8.
J Nanobiotechnology ; 22(1): 219, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38698419

RESUMEN

BACKGROUND: Adipose-derived stem cells (ASCs) represent the most advantageous choice for soft tissue regeneration. Studies proved the recruitment of ASCs post tissue injury was mediated by chemokine CXCL12, but the mechanism by which CXCL12 is generated after tissue injury remains unclear. Migrasomes are newly discovered membrane-bound organelles that could deliver CXCL12 spatially and temporally in vivo. In this study, we sought to investigate whether migrasomes participate ASC-mediated tissue regeneration. METHODS: Discrepant and asymmetrical soft tissue regeneration mice model were established, in which HE staining, immunofluorescent staining, western blot and qPCR were conducted to confirm the role of CXCL12 and migrasomes in ASC-mediated tissue regeneration. Characterization of ASC-derived migrasomes were carried out by confocal microscopy, scanning electron microscopy, transmission electron microscopy as well as western blot analysis. The function and mechanism of migrasomes were further testified by assisting tissue regeneration with isolated migrasomes in vivo and by in vitro transwell combined with co-culture system. RESULTS: Here, we show for the first time that migrasomes participate in soft tissue regeneration. ASCs generate migrasomes enriched with CXCL12 to mediate tissue regeneration. Migrasomes from ASCs could promote stem cells migration by activating CXCR4/RhoA signaling in vivo and in vitro. Chemoattracted ASCs facilitate regeneration, as demonstrated by the upregulation of an adipogenesis-associated protein. This positive feed-back-loop creates a favorable microenvironment for soft tissue regeneration. Thus, migrasomes represent a new therapeutic target for ASC-mediated tissue regeneration. CONCLUSIONS: Our findings reveal a previously unknown function of ASCs in mediating tissue regeneration by generating migrasomes. The ASC-derived migrasomes can restore tissue regeneration by recruiting stem cells, which highlighting the potential application of ASC-derived migrasomes in regenerative medicine.


Asunto(s)
Tejido Adiposo , Quimiocina CXCL12 , Receptores CXCR4 , Regeneración , Células Madre , Proteína de Unión al GTP rhoA , Quimiocina CXCL12/metabolismo , Animales , Receptores CXCR4/metabolismo , Ratones , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Células Madre/metabolismo , Células Madre/citología , Ratones Endogámicos C57BL , Retroalimentación Fisiológica , Movimiento Celular , Células Cultivadas , Masculino , Transducción de Señal
9.
Ceska Gynekol ; 89(2): 151-155, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38704229

RESUMEN

The human placenta serves as a vital barrier between the mother and the developing fetus during pregnancy. A defect in the early development of the placenta is associated with severe pregnancy disorders. Despite its complex development, various molecular processes control placental development, and the specialization of trophoblast cells is still not fully understood. One primary obstacle is the lack of suitable cell model systems. Traditional two-dimensional (2D) cell cultures fail to mimic in vivo conditions and do not capture the intricate intercellular interactions vital for studying placental development. However, three-dimensional (3D) organoid models derived from stem cells that replicate natural cell organization and architecture have greatly improved our understanding of trophoblast behavior and its medicinal applications. Organoids with relevant phenotypes provide a valuable platform to model both placental physiology and pathology, including the modeling of placental disorders. They hold great promise for personalized medicine, improved diagnostics, and the evaluation of pharmaceutical drug efficacy and safety. This article provides a concise overview of trophoblast stem cells, trophoblast invasion, and the evolving role of organoids in gynecology.


Asunto(s)
Organoides , Células Madre , Trofoblastos , Humanos , Trofoblastos/fisiología , Organoides/fisiología , Femenino , Embarazo , Células Madre/fisiología , Placenta/citología , Placenta/fisiología , Placenta/patología , Placentación/fisiología
10.
Stem Cell Res Ther ; 15(1): 136, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38715083

RESUMEN

BACKGROUND: Alzheimer's disease (AD) is a prevalent form of dementia leading to memory loss, reduced cognitive and linguistic abilities, and decreased self-care. Current AD treatments aim to relieve symptoms and slow disease progression, but a cure is elusive due to limited understanding of the underlying disease mechanisms. MAIN CONTENT: Stem cell technology has the potential to revolutionize AD research. With the ability to self-renew and differentiate into various cell types, stem cells are valuable tools for disease modeling, drug screening, and cell therapy. Recent advances have broadened our understanding beyond the deposition of amyloidß (Aß) or tau proteins in AD to encompass risk genes, immune system disorders, and neuron-glia mis-communication, relying heavily on stem cell-derived disease models. These stem cell-based models (e.g., organoids and microfluidic chips) simulate in vivo pathological processes with extraordinary spatial and temporal resolution. Stem cell technologies have the potential to alleviate AD pathology through various pathways, including immunomodulation, replacement of damaged neurons, and neurotrophic support. In recent years, transplantation of glial cells like oligodendrocytes and the infusion of exosomes have become hot research topics. CONCLUSION: Although stem cell-based models and therapies for AD face several challenges, such as extended culture time and low differentiation efficiency, they still show considerable potential for AD treatment and are likely to become preferred tools for AD research.


Asunto(s)
Enfermedad de Alzheimer , Trasplante de Células Madre , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Humanos , Trasplante de Células Madre/métodos , Animales , Células Madre/metabolismo , Células Madre/citología
11.
Life Sci Alliance ; 7(7)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38719753

RESUMEN

We recently reported that growth/differentiation factor 15 (GDF15) and its receptor GDNF family receptor alpha-like (GFRAL) are expressed in the periventricular germinal epithelium thereby regulating apical progenitor proliferation. However, the mechanisms are unknown. We now found GFRAL in primary cilia and altered cilia morphology upon GDF15 ablation. Mutant progenitors also displayed increased histone deacetylase 6 (Hdac6) and ciliary adenylate cyclase 3 (Adcy3) transcript levels. Consistently, microtubule acetylation, endogenous sonic hedgehog (SHH) activation and ciliary ADCY3 were all affected in this group. Application of exogenous GDF15 or pharmacological antagonists of either HDAC6 or ADCY3 similarly normalized ciliary morphology, proliferation and SHH signalling. Notably, Gdf15 ablation affected Hdac6 expression and cilia length only in the mutant periventricular niche, in concomitance with ciliary localization of GFRAL. In contrast, in the hippocampus, where GFRAL was not expressed in the cilium, progenitors displayed altered Adcy3 expression and SHH signalling, but Hdac6 expression, cilia morphology and ciliary ADCY3 levels remained unchanged. Thus, ciliary signalling underlies the effect of GDF15 on primary cilia elongation and proliferation in apical progenitors.


Asunto(s)
Adenilil Ciclasas , Proliferación Celular , Cilios , Proteínas Hedgehog , Histona Desacetilasa 6 , Transducción de Señal , Cilios/metabolismo , Animales , Proliferación Celular/genética , Ratones , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Adenilil Ciclasas/metabolismo , Adenilil Ciclasas/genética , Histona Desacetilasa 6/metabolismo , Histona Desacetilasa 6/genética , Células Madre/metabolismo , Células Madre/citología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Ratones Noqueados , Acetilación
12.
Nat Commun ; 15(1): 3873, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719882

RESUMEN

Human glial progenitor cells (hGPCs) exhibit diminished expansion competence with age, as well as after recurrent demyelination. Using RNA-sequencing to compare the gene expression of fetal and adult hGPCs, we identify age-related changes in transcription consistent with the repression of genes enabling mitotic expansion, concurrent with the onset of aging-associated transcriptional programs. Adult hGPCs develop a repressive transcription factor network centered on MYC, and regulated by ZNF274, MAX, IKZF3, and E2F6. Individual over-expression of these factors in iPSC-derived hGPCs lead to a loss of proliferative gene expression and an induction of mitotic senescence, replicating the transcriptional changes incurred during glial aging. miRNA profiling identifies the appearance of an adult-selective miRNA signature, imposing further constraints on the expansion competence of aged GPCs. hGPC aging is thus associated with acquisition of a MYC-repressive environment, suggesting that suppression of these repressors of glial expansion may permit the rejuvenation of aged hGPCs.


Asunto(s)
Envejecimiento , MicroARNs , Neuroglía , Factores de Transcripción , Humanos , Neuroglía/metabolismo , Neuroglía/citología , Envejecimiento/genética , Envejecimiento/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , MicroARNs/genética , MicroARNs/metabolismo , Senescencia Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre/metabolismo , Células Madre/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Adulto , Redes Reguladoras de Genes , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Perfilación de la Expresión Génica
13.
Cells ; 13(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38727284

RESUMEN

Stem cells (SCs) undergo asymmetric division, producing transit-amplifying cells (TACs) with increased proliferative potential that move into tissues and ultimately differentiate into a specialized cell type. Thus, TACs represent an intermediary state between stem cells and differentiated cells. In the cornea, a population of stem cells resides in the limbal region, named the limbal epithelial stem cells (LESCs). As LESCs proliferate, they generate TACs that move centripetally into the cornea and differentiate into corneal epithelial cells. Upon limbal injury, research suggests a population of progenitor-like cells that exists within the cornea can move centrifugally into the limbus, where they dedifferentiate into LESCs. Herein, we summarize recent advances made in understanding the mechanism that governs the differentiation of LESCs into TACs, and thereafter, into corneal epithelial cells. We also outline the evidence in support of the existence of progenitor-like cells in the cornea and whether TACs could represent a population of cells with progenitor-like capabilities within the cornea. Furthermore, to gain further insights into the dynamics of TACs in the cornea, we outline the most recent findings in other organ systems that support the hypothesis that TACs can dedifferentiate into SCs.


Asunto(s)
Diferenciación Celular , Epitelio Corneal , Limbo de la Córnea , Células Madre , Humanos , Células Madre/citología , Células Madre/metabolismo , Limbo de la Córnea/citología , Epitelio Corneal/citología , Animales , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proliferación Celular
14.
Mol Biol Rep ; 51(1): 644, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38727958

RESUMEN

BACKGROUND: MicroRNAs are differentially expressed in periodontitis tissues. They are involved in cellular responses to inflammation and can be used as markers for diagnosing periodontitis. Microarray analysis showed that the expression level of microRNA-671-5p in periodontal tissues of patients with periodontitis was increased. In this study, we investigated the mechanism of action of microRNA-671-5p in human periodontal ligament stem cells (hPDLSCs) under inflammatory conditions. METHODS AND RESULTS: HPDLSCs were treated with lipopolysaccharide (LPS) to establish an inflammation model. The cell survival rate was determined using the cell counting kit-8 (CCK8). Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot analyses were used to detect the expression of microRNA-671-5p and dual-specificity phosphatase (DUSP) 8 proteins, respectively, Interleukin (IL)-6, IL-1ß, and tumor necrosis factor (TNF)-α were detected using qRT-PCR and Enzyme-linked immunosorbent assay (ELISA). A dual-luciferase reporter system was employed to determine the relationship between micoRNA-671-5p and DUSP8 expression. Activation of the p38 mitogen-activated protein kinase (MAPK) signaling pathway was confirmed using western blot analysis. Following the treatment of hPDLSCs with LPS, the expression levels of microRNA-671-5p in hPDLSCs were increased, cell viability decreased, and the expression of inflammatory factors displayed an increasing trend. MicroRNA-671-5p targets and binds to DUSP8. Silencing microRNA-671-5p or overexpressing DUSP8 can improve cell survival rate and reduce inflammatory responses. When DUSP8 was overexpressed, the expression of p-p38 was reduced. CONCLUSIONS: microRNA-671-5p targets DUSP8/p38 MAPK pathway to regulate LPS-induced proliferation and inflammation in hPDLSCs.


Asunto(s)
Fosfatasas de Especificidad Dual , Inflamación , Lipopolisacáridos , MicroARNs , Ligamento Periodontal , Células Madre , Proteínas Quinasas p38 Activadas por Mitógenos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/citología , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Células Madre/metabolismo , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lipopolisacáridos/farmacología , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Periodontitis/genética , Periodontitis/metabolismo , Periodontitis/patología , Supervivencia Celular/genética , Supervivencia Celular/efectos de los fármacos , Transducción de Señal/genética , Células Cultivadas
15.
Cell Mol Neurobiol ; 44(1): 44, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38713302

RESUMEN

Multiple Sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) characterized by inflammation and demyelination of CNS neurons. Up to now, there are many therapeutic strategies for MS but they are only being able to reduce progression of diseases and have not got any effect on repair and remyelination. Stem cell therapy is an appropriate method for regeneration but has limitations and problems. So recently, researches were used of exosomes that facilitate intercellular communication and transfer cell-to-cell biological information. MicroRNAs (miRNAs) are a class of short non-coding RNAs that we can used to their dysregulation in order to diseases diagnosis. The miRNAs of microvesicles obtained stem cells may change the fate of transplanted cells based on received signals of injured regions. The miRNAs existing in MSCs may be displayed the cell type and their biological activities. Current studies show also that the miRNAs create communication between stem cells and tissue-injured cells. In the present review, firstly we discuss the role of miRNAs dysregulation in MS patients and miRNAs expression by stem cells. Finally, in this study was confirmed the relationship of microRNAs involved in MS and miRNAs expressed by stem cells and interaction between them in order to find appropriate treatment methods in future for limit to disability progression.


Asunto(s)
Exosomas , MicroARNs , Esclerosis Múltiple , Células Madre , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Exosomas/metabolismo , Esclerosis Múltiple/terapia , Esclerosis Múltiple/genética , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Animales , Células Madre/metabolismo
16.
Cell Mol Life Sci ; 81(1): 208, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710919

RESUMEN

Trophoblast stem cells (TSCs) can be chemically converted from embryonic stem cells (ESCs) in vitro. Although several transcription factors (TFs) have been recognized as essential for TSC formation, it remains unclear how differentiation cues link elimination of stemness with the establishment of TSC identity. Here, we show that PRDM14, a critical pluripotent circuitry component, is reduced during the formation of TSCs. The reduction is further shown to be due to the activation of Wnt/ß-catenin signaling. The extinction of PRDM14 results in the erasure of H3K27me3 marks and chromatin opening in the gene loci of TSC TFs, including GATA3 and TFAP2C, which enables their expression and thus the initiation of the TSC formation process. Accordingly, PRDM14 reduction is proposed here as a critical event that couples elimination of stemness with the initiation of TSC formation. The present study provides novel insights into how induction signals initiate TSC formation.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN , Factores de Transcripción , Trofoblastos , Vía de Señalización Wnt , Trofoblastos/metabolismo , Trofoblastos/citología , Animales , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA3/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Células Madre/metabolismo , Células Madre/citología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Histonas/metabolismo , Histonas/genética
17.
Cell Biochem Funct ; 42(3): e4008, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38613198

RESUMEN

Temporal phases of wound healing and their corresponding healing factors are essential in wound regeneration. Mesenchymal stem cells (MSCs) accelerate wound healing via their paracrine secretions by enhancing cell migration, angiogenesis, and reducing inflammation. This study evaluated the local therapeutic effect of human umbilical cord MSCs (hUCMSCs) in the healing of cold-induced burn wounds. An in vitro wound (scratch) was developed in rat skin fibroblasts. The culture was maintained in the conditioned medium (CM) which was prepared by inducing an artificial wound in hUCMSCs in a separate experiment. Treated fibroblasts were analyzed for the gene expression profile of healing mediators involved in wound closure. Findings revealed enhanced cell migration and increased levels of healing mediators in the treated fibroblasts relative to the untreated group. Cold-induced burn wounds were developed in Wistar rats, followed by a single injection of hUCMSCs. Wound healing pattern was examined based on the healing phases: hemostasis/inflammation (Days 1, 3), cell proliferation (Day 7), and remodeling (Day 14). Findings exhibited enhanced wound closure in the treated wound. Gene expression, histological, and immunohistochemical analyses further confirmed enhanced wound regeneration after hUCMSC transplantation. Temporal gene expression profile revealed that the level of corresponding cytokines was substantially increased in the treated wound as compared with the control, indicating improvement in the processes of angiogenesis and remodeling, and a substantial reduction in inflammation. Histology revealed significant collagen formation along with regenerated skin layers and appendages, whereas immunohistochemistry exhibited increased neovascularization during remodeling. Leukocyte infiltration was also suppressed in the treated group. Overall findings demonstrate that a single dose of hUCMSCs enhances wound healing in vivo, and their secreted growth factors accelerate cell migration in vitro.


Asunto(s)
Quemaduras , Células Madre , Animales , Femenino , Humanos , Ratas , Quemaduras/terapia , Inflamación , Ratas Wistar , Cicatrización de Heridas
18.
Nanotheranostics ; 8(3): 298-311, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38577321

RESUMEN

Exosomes are nanosized extracellular vesicles secreted by all cell types, including canine adipose-derived stem cells (cADSCs). By mediating intercellular communication, exosomes modulate the biology of adjacent and distant cells by transferring their cargo. In the present work after isolation and characterization of exosomes derived from canine adipose tissue, we treated the same canine donors affected by hepatopathies with the previously isolated exosomes. We hypothesize that cADSC-sourced miRNAs are among the factors responsible for a regenerative and anti-inflammatory effect in the treatment of hepatopathies in dogs, providing the clinical veterinary field with an effective and innovative cell-free therapy. Exosomes were isolated and characterized for size, distribution, surface markers, and for their miRNomic cargo by microRNA sequencing. 295 dogs affected with hepatopathies were treated and followed up for 6 months to keep track of their biochemical marker levels. Results confirmed that exosomes derived from cADSCs exhibited an average diameter of 91 nm, and positivity to 8 known exosome markers. The administration of exosomes to dogs affected by liver-associated inflammatory pathologies resulted in the recovery of the animal alongside the normalization of biochemical parameters of kidney function. In conclusion, cADSCs-derived exosomes are a promising therapeutic tool for treating inflammatory disorders in animal companions.


Asunto(s)
Exosomas , Vesículas Extracelulares , MicroARNs , Perros , Animales , MicroARNs/genética , Exosomas/genética , Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Hepatitis Crónica/metabolismo , Células Madre/metabolismo
19.
Int J Mol Sci ; 25(7)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38612526

RESUMEN

Cellular senescence is a tightly regulated pathophysiologic process and is caused by replicative exhaustion or external stressors. Since naturally derived bioactive compounds with anti-ageing properties have recently captured scientific interest, we analysed the anti-ageing and antioxidant efficacy of Cryptomphalus aspersa egg extract (CAEE). Its effects on stemness, wound-healing properties, antioxidant defense mechanisms, and DNA damage repair ability of Human Wharton's jelly mesenchymal stem cells (WJ-MSCs) were analysed. Our results revealed that CAEE fortifies WJ-MSCs stemness, which possibly ameliorates their wound-healing ability. Additionally, we show that CAEE possesses a strong antioxidant capacity as demonstrated by the elevation of the levels of the basic antioxidant molecule, GSH, and the induction of the NRF2, a major antioxidant regulator. In addition, CAEE alleviated cells' oxidative stress and therefore prevented stress-induced premature senescence (SIPS). Furthermore, we demonstrated that the prevention of SIPS could be mediated via the extract's ability to induce autophagy, as indicated by the elevation of the protein levels of all basic autophagic molecules and the increase in formation of autophagolysosomes in CAEE-treated WJ-MSCs. Moreover, CAEE-treated cells exhibited decreased Caveolin-1 levels. We propose that Cryptomphalus aspersa egg extract comprises bioactive compounds that can demonstrate strong antioxidant/anti-ageing effects by regulating the Caveolin-1-autophagy-senescence molecular axis.


Asunto(s)
Antioxidantes , Caveolina 1 , Humanos , Antioxidantes/farmacología , Senescencia Celular , Células Madre , Envejecimiento
20.
Int J Mol Sci ; 25(7)2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38612687

RESUMEN

Dimethyloxalylglycine (DMOG) has been found to stimulate osteogenesis and angiogenesis of stem cells, promoting neo-angiogenesis in bone tissue regeneration. In this review, we conducted a comprehensive search of the literature to investigate the effects of DMOG on osteogenesis and bone regeneration. We screened the studies based on specific inclusion criteria and extracted relevant information from both in vitro and in vivo experiments. The risk of bias in animal studies was evaluated using the SYRCLE tool. Out of the 174 studies retrieved, 34 studies met the inclusion criteria (34 studies were analyzed in vitro and 20 studies were analyzed in vivo). The findings of the included studies revealed that DMOG stimulated stem cells' differentiation toward osteogenic, angiogenic, and chondrogenic lineages, leading to vascularized bone and cartilage regeneration. Addtionally, DMOG demonstrated therapeutic effects on bone loss caused by bone-related diseases. However, the culture environment in vitro is notably distinct from that in vivo, and the animal models used in vivo experiments differ significantly from humans. In summary, DMOG has the ability to enhance the osteogenic and angiogenic differentiation potential of stem cells, thereby improving bone regeneration in cases of bone defects. This highlights DMOG as a potential focus for research in the field of bone tissue regeneration engineering.


Asunto(s)
Aminoácidos Dicarboxílicos , Enfermedades Óseas Metabólicas , Osteogénesis , Animales , Humanos , Regeneración Ósea , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA