Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 397
Filtrar
1.
Mol Biol Rep ; 51(1): 675, 2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38787484

RESUMEN

BACKGROUND: Bioscaffolds and cells are two main components in the regeneration of damaged tissues via cell therapy. Umbilical cord stem cells are among the most well-known cell types for this purpose. The main objective of the present study was to evaluate the effect of the pretreatment of the foreskin acellular matrix (FAM) by monophosphoryl lipid A (MPLA) and Lactobacillus casei supernatant (LCS) on the attraction of human umbilical cord mesenchymal stem cells (hucMSC). METHODS AND RESULTS: The expression of certain cell migration genes was studied using qRT-PCR. In addition to cell migration, transdifferentiation of these cells to the epidermal-like cells was evaluated via immunohistochemistry (IHC) and immunocytochemistry (ICC) of cytokeratin 19 (CK19). The hucMSC showed more tissue tropism in the presence of MPLA and LCS pretreated FAM compared to the untreated control group. We confirmed this result by scanning electron microscopy (SEM) analysis, glycosaminoglycan (GAG), collagen, and DNA content. Furthermore, IHC and ICC data demonstrated that both treatments increase the protein expression level of CK19. CONCLUSION: Pretreatment of acellular bioscaffolds by MPLA or LCS can increase the migration rate of cells and also transdifferentiation of hucMSC to epidermal-like cells without growth factors. This strategy suggests a new approach in regenerative medicine.


Asunto(s)
Lacticaseibacillus casei , Lípido A , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/metabolismo , Lacticaseibacillus casei/metabolismo , Lípido A/metabolismo , Lípido A/análogos & derivados , Movimiento Celular/efectos de los fármacos , Piel/metabolismo , Andamios del Tejido/química , Masculino , Cordón Umbilical/citología , Cordón Umbilical/metabolismo , Prepucio/citología , Transdiferenciación Celular/efectos de los fármacos , Ingeniería de Tejidos/métodos , Matriz Extracelular/metabolismo , Queratina-19/metabolismo , Queratina-19/genética
2.
PLoS One ; 16(11): e0256812, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34762649

RESUMEN

Transforming growth factor-beta 1 (TGF-ß1), a pro-fibrotic tumour-derived factor promotes fibroblast differentiation in the tumour microenvironment and is thought to contribute to the development of pro-tumourigenic cancer-associated fibroblasts (CAFs) by promoting myofibroblast differentiation. miRNA dysregulation has been demonstrated in myofibroblast transdifferentiation and CAF activation, however, their expression varies among cell types and with the method of fibroblast induction. Here, the expression profile of miRNA in human primary oral fibroblasts treated with TGF-ß1, to derive a myofibroblastic, CAF-like phenotype, was determined compared to untreated fibroblasts. Myofibroblast transdifferentiation was determined by the expression of alpha-smooth muscle actin (α-SMA) and fibronectin-1 extra domain A (FN-EDA1) using quantitative real-time PCR (qRT-PCR) and western blot. The formation of stress fibres was assessed by fluorescence microscopy, and associated changes in contractility were assessed using collagen contraction assays. Extracellular vesicles (EVs) were purified by using size exclusion chromatography and ultracentrifugation and their size and concentration were determined by nanoparticle tracking analysis. miRNA expression profiling in oral fibroblasts treated with TGF-ß1 and their extracellular vesicles was carried out using tiling low-density array cards. The Database for Annotation, Visualization, and Integrated Discovery (DAVID) was used to perform functional and pathway enrichment analysis of target genes. In this study, TGF-ß1 induced a myofibroblastic phenotype in normal oral fibroblasts as assessed by expression of molecular markers, the formation of stress fibres and increased contractility. TaqMan Low-Density Array (TLDA) analysis demonstrated that miR-503 and miR-708 were significantly upregulated, while miR-1276 was significantly downregulated in TGF-ß1-treated oral fibroblasts (henceforth termed experimentally-derived CAF, eCAF). The gene functional enrichment analysis showed that the candidate miRNAs have the potential to modulate various pathways; including the Ras associated protein 1 (Rap1), PI3K-Akt, and tumour necrosis factor (TNF) signalling pathways. In addition, altered levels of several miRNAs were detected in eCAF EV, including miR-142 and miR-222. No differences in size or abundance of EV were detected between eCAF and normal oral fibroblast (NOF). Little overlap was observed between changes in cellular and EV miRNA profiles, suggesting the possibility of selective loading of EV miRNA. The study reveals miRNA expression signature could be involved in myofibroblast transdifferentiation and the miRNA cargo of their EV, providing novel insight into the involvement of miRNA in CAF development and function.


Asunto(s)
Transdiferenciación Celular/fisiología , Vesículas Extracelulares/metabolismo , MicroARNs/metabolismo , Miofibroblastos/citología , Actinas/metabolismo , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Vesículas Extracelulares/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta1/farmacología
3.
Endocrinology ; 162(12)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34606582

RESUMEN

Endometriosis is characterized by inflammation and fibrotic changes. Our previous study using a mouse model showed that proinflammatory factors present in peritoneal hemorrhage exacerbated inflammation in endometriosis-like grafts, at least in part through the activation of prostaglandin (PG) E2 receptor and protease-activated receptor (PAR). In addition, menstruation-related factors, PGE2 and thrombin (P/T), a PAR1 agonist induced epithelial-mesenchymal transition (EMT) of endometrial cells under hypoxia. However, the molecular mechanisms by which P/T induce development of endometriosis have not been fully characterized. To investigate the effects of P/T, RNA extracted from endometrial stromal cells (ESCs) treated with P/T were subjected to RNA sequence analysis, and identified activin A, FOS, and GATA2 as upregulated genes. Activin A increased the expression of connective tissue growth factor (CTGF) and mesenchymal marker genes in ESCs. CTGF induced the expression of fibrosis marker type I collagen, fibronectin, and α-smooth muscle actin (αSMA), indicating fibroblast to myofibroblast transdifferentiation (FMT) of ESCs. In addition, activin A, FOS, GATA2, CTGF, and αSMA were localized in endometriosis lesions. Taken together, our data show that P/T induces changes resembling EMT and FMT in ectopic ESCs derived from retrograde menstruation, and that these are associated with fibrotic changes in the lesions. Pharmacological means that block P/T-induced activin A and CTGF signaling may be strategies to inhibit fibrosis in endometriotic lesions.


Asunto(s)
Transdiferenciación Celular/efectos de los fármacos , Dinoprostona/farmacología , Endometrio/efectos de los fármacos , Miofibroblastos/efectos de los fármacos , Trombina/farmacología , Activinas/genética , Activinas/metabolismo , Adulto , Transdiferenciación Celular/genética , Células Cultivadas , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Endometriosis/patología , Endometrio/citología , Endometrio/patología , Femenino , Humanos , Miofibroblastos/fisiología , Enfermedades Peritoneales/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/efectos de los fármacos , Células del Estroma/patología , Células del Estroma/fisiología
4.
Mediators Inflamm ; 2021: 3399357, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34690551

RESUMEN

Liver fibrosis, which results from chronic liver injury due to factors such as chronic alcohol consumption, hepatitis virus infections, and immune attacks, is marked by excessive deposition of extracellular matrix (ECM). Resveratrol (Res), a polyphenol phytoalexin, has been demonstrated to show anti-inflammatory, antioxidative, antiproliferative, and chemopreventive activities. In recent years, Res has been found to inhibit liver fibrosis. Enhanced Hippo pathway activation has also been reported to inhibit tumor progression and liver fibrosis. In the present study, the role of the Hippo pathway in mediating the effects of Res on hepatic stellate cells (HSCs) was examined. We found that Res significantly suppresses HSC proliferation, reducing the cell index. Res induced HSC inactivation, reducing collagen deposition and α-smooth muscle actin (α-SMA) expression. In addition, Res contributed to HSC apoptosis, upregulating Bax and downregulating Bcl-2 expression. Notably, the Hippo pathway was involved in the Res-mediated suppression of HSC activation. Res enhanced the activation of the Hippo pathway and reduced yes-associated protein (YAP) and transcriptional coactivator with the PDZ-binding motif (TAZ) expression. Interestingly, the YAP overexpression inhibited Res-induced HSC inactivation and apoptosis. In conclusion, these results demonstrate that Res inhibits HSC activation, at least in part, via the Hippo pathway. The present study indicates a new antifibrotic mechanism of Res and provides novel insights into Hippo-mediated HSC apoptosis and HSC activation in liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/efectos de los fármacos , Vía de Señalización Hippo/efectos de los fármacos , Resveratrol/farmacología , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Células Estrelladas Hepáticas/fisiología , Vía de Señalización Hippo/fisiología , Humanos , Cirrosis Hepática/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Resveratrol/uso terapéutico , Proteínas Señalizadoras YAP/fisiología
5.
Cell Rep ; 37(1): 109774, 2021 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-34610301

RESUMEN

While squamous transdifferentiation within subpopulations of adenocarcinomas represents an important drug resistance problem, its underlying mechanism remains poorly understood. Here, using surface markers of resistant basal cell carcinomas (BCCs) and patient single-cell and bulk transcriptomic data, we uncover the dynamic roadmap of basal to squamous cell carcinoma transition (BST). Experimentally induced BST identifies activator protein 1 (AP-1) family members in regulating tumor plasticity, and we show that c-FOS plays a central role in BST by regulating the accessibility of distinct AP-1 regulatory elements. Remarkably, despite prominent changes in cell morphology and BST marker expression, we show using inducible model systems that c-FOS-mediated BST demonstrates reversibility. Blocking EGFR pathway activation after c-FOS induction partially reverts BST in vitro and prevents BST features in both mouse models and human tumors. Thus, by identifying the molecular basis of BST, our work reveals a therapeutic opportunity targeting plasticity as a mechanism of tumor resistance.


Asunto(s)
Carcinoma Basocelular/patología , Carcinoma de Células Escamosas/patología , Transdiferenciación Celular , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/veterinaria , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/veterinaria , Transdiferenciación Celular/efectos de los fármacos , Ensamble y Desensamble de Cromatina , Resistencia a Antineoplásicos/genética , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mucina-1/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
6.
Sci Rep ; 11(1): 16096, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373467

RESUMEN

Benzalkonium chloride (BAC) is used as a preservative in eyedrops but induces subconjunctival fibrosis that can result in failure of glaucoma surgery. Tenon's capsule fibroblasts in subconjunctival tissue interact with the corneal epithelium through tear fluid. With the use of a coculture system, we have now investigated the effect of human corneal epithelial (HCE) cells on myofibroblastic transdifferentiation of human Tenon fibroblasts (HTFs) induced by BAC (5 × 10-6%). Immunofluorescence and immunoblot analyses revealed that the BAC-induced expression of α smooth muscle actin (αSMA) in HTFs was suppressed by coculture of these cells with HCE cells (p < 0.01). The concentration of interleukin-10 (IL-10) in culture supernatants of BAC-treated HTFs was increased by coculture with HCE cells (17.26-fold, vs. coculure, p < 0.001). Immunofluorescence and immunoblot analyses also showed that exogenous IL-10 (300 pg/ml) suppressed the BAC-induced expression of αSMA by 43.65% (p < 0.05) as well as the nuclear translocation of myocardin-related transcription factor-A (MRTF-A) by 39.32% (p < 0.01) in HTFs cultured alone. Our findings suggest that corneal epithelial cells may protect against subconjunctival fibrosis by maintaining IL-10 levels and preventing the MRTF-A-dependent transdifferentiation of HTFs into myofibroblasts.


Asunto(s)
Compuestos de Benzalconio/farmacología , Transdiferenciación Celular/efectos de los fármacos , Córnea/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Interleucina-10/metabolismo , Miofibroblastos/efectos de los fármacos , Cápsula de Tenon/efectos de los fármacos , Actinas/metabolismo , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Técnicas de Cocultivo/métodos , Córnea/metabolismo , Células Epiteliales/metabolismo , Fibroblastos/metabolismo , Fibrosis/tratamiento farmacológico , Fibrosis/metabolismo , Humanos , Miofibroblastos/metabolismo , Transducción de Señal/efectos de los fármacos , Cápsula de Tenon/metabolismo , Transactivadores/metabolismo
7.
PLoS One ; 16(8): e0255075, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34375370

RESUMEN

Induced endothelial cells (iECs) generated from neonatal fibroblasts via transdifferentiation have been shown to have pro-angiogenic properties and are a potential therapy for peripheral arterial disease (PAD). It is unknown if iECs can be generated from fibroblasts collected from PAD patients and whether these cells are pro-angiogenic. In this study fibroblasts were collected from four PAD patients undergoing carotid endarterectomies. These cells, and neonatal fibroblasts, were transdifferentiated into iECs using modified mRNA. Endothelial phenotype and pro-angiogenic cytokine secretion were investigated. NOD-SCID mice underwent surgery to induce hindlimb ischaemia in a murine model of PAD. Mice received intramuscular injections with either control vehicle, or 1 × 106 neonatal-derived or 1 × 106 patient-derived iECs. Recovery in perfusion to the affected limb was measured using laser Doppler scanning. Perfusion recovery was enhanced in mice treated with neonatal-derived iECs and in two of the three patient-derived iEC lines investigated in vivo. Patient-derived iECs can be successfully generated from PAD patients and for specific patients display comparable pro-angiogenic properties to neonatal-derived iECs.


Asunto(s)
Células Endoteliales/patología , Fibroblastos/patología , Neovascularización Fisiológica , Enfermedad Arterial Periférica/patología , Acetilación/efectos de los fármacos , Animales , Capilares/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Colágeno/farmacología , Medios de Cultivo Condicionados/farmacología , Citocinas/metabolismo , Combinación de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/trasplante , Fibroblastos/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intercelular/farmacología , Isquemia/patología , Isquemia/terapia , Laminina/farmacología , Lipoproteínas LDL/metabolismo , Masculino , Ratones Endogámicos NOD , Ratones SCID , Neovascularización Fisiológica/efectos de los fármacos , Perfusión , Lectinas de Plantas/metabolismo , Unión Proteica/efectos de los fármacos , Proteoglicanos/farmacología
8.
Cells ; 10(8)2021 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-34440909

RESUMEN

Human erythropoietin (EPO) is an N-linked glycoprotein consisting of 166 aa that is produced in the kidney during the adult life and acts both as a peptide hormone and hematopoietic growth factor (HGF), stimulating bone marrow erythropoiesis. EPO production is activated by hypoxia and is regulated via an oxygen-sensitive feedback loop. EPO acts via its homodimeric erythropoietin receptor (EPO-R) that increases cell survival and drives the terminal erythroid maturation of progenitors BFU-Es and CFU-Es to billions of mature RBCs. This pathway involves the activation of multiple erythroid transcription factors, such as GATA1, FOG1, TAL-1, EKLF and BCL11A, and leads to the overexpression of genes encoding enzymes involved in heme biosynthesis and the production of hemoglobin. The detection of a heterodimeric complex of EPO-R (consisting of one EPO-R chain and the CSF2RB ß-chain, CD131) in several tissues (brain, heart, skeletal muscle) explains the EPO pleotropic action as a protection factor for several cells, including the multipotent MSCs as well as cells modulating the innate and adaptive immunity arms. EPO induces the osteogenic and endothelial transdifferentiation of the multipotent MSCs via the activation of EPO-R signaling pathways, leading to bone remodeling, induction of angiogenesis and secretion of a large number of trophic factors (secretome). These diversely unique properties of EPO, taken together with its clinical use to treat anemias associated with chronic renal failure and other blood disorders, make it a valuable biologic agent in regenerative medicine for the treatment/cure of tissue de-regeneration disorders.


Asunto(s)
Remodelación Ósea/fisiología , Transdiferenciación Celular/fisiología , Células Endoteliales/citología , Eritropoyesis/fisiología , Eritropoyetina/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Remodelación Ósea/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Eritropoyetina/farmacología , Humanos , Receptores de Eritropoyetina/metabolismo , Medicina Regenerativa/métodos
9.
Biochem Biophys Res Commun ; 573: 55-61, 2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34388455

RESUMEN

Mammary epithelial cells are the only cells in the mammary glands that are capable of lactation and they are ideal for studying cellular and molecular biology mechanisms during growth, development and lactation of the mammary glands. The limiting factors in most of the currently available mammary epithelial cells are low cell viability, transgenerational efficiency and lactation function that renders them unsuitable for subsequent studies on mammary gland's cellular and lactation mechanisms and utilizing them as bioreactors. Hence, new methods are required to obtain mammary epithelial cells with high transgenerational efficiency and lactation function. In this study, transdifferentiation of goat ear fibroblasts (GEFs) into goat mammary epithelial cells (CiMECs) was induced in only eight days by five small molecule compounds, including 500 µg/mL VPA, 10 µM Tranylcypromine, 10 µM Forskolin, 1 µM TTNPB, 10 µM RepSox. Morphological observation, marker genes comparison, specific antigen expression and comparison of gene expression levels by transcriptome sequencing between the two types of cells that led to the primary deduction that CiMECs have similar biological properties to goat mammary epithelial cells (GMECs) and comparatively more lactation capacity. Therefore, we establish a novel reprogramming route to convert fibroblasts into CiMECs under fully chemically conditions. This study is expected to provide an in vitro platform for understanding cellular mechanisms such as mammary epithelial cells' fate determination and developmental differentiation, and also to find a new way to obtain a large number of functional mammary epithelial cells in vitro.


Asunto(s)
Benzoatos/farmacología , Colforsina/farmacología , Pirazoles/farmacología , Piridinas/farmacología , Retinoides/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Tranilcipromina/farmacología , Ácido Valproico/farmacología , Animales , Benzoatos/química , Transdiferenciación Celular/efectos de los fármacos , Colforsina/química , Relación Dosis-Respuesta a Droga , Oído , Células Epiteliales/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Cabras , Glándulas Mamarias Animales/efectos de los fármacos , Pirazoles/química , Piridinas/química , Retinoides/química , Bibliotecas de Moléculas Pequeñas/química , Tranilcipromina/química , Ácido Valproico/química
10.
Nutrients ; 13(7)2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34209455

RESUMEN

Glucose-based solutions remain the most used osmotic agents in peritoneal dialysis (PD), but unavoidably they contribute to the loss of peritoneal filtration capacity. Here, we evaluated at a molecular level the effects of XyloCore, a new PD solution with a low glucose content, in mesothelial and endothelial cells. Cell viability, integrity of mesothelial and endothelial cell membrane, activation of mesothelial and endothelial to mesenchymal transition programs, inflammation, and angiogenesis were evaluated by several techniques. Results showed that XyloCore preserves mesothelial and endothelial cell viability and membrane integrity. Moreover XyloCore, unlike glucose-based solutions, does not exert pro-fibrotic, -inflammatory, and -angiogenic effects. Overall, the in vitro evidence suggests that XyloCore could represent a potential biocompatible solution promising better outcomes in clinical practice.


Asunto(s)
Soluciones para Diálisis/farmacología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Glucosa/farmacología , Inflamación/patología , Mesodermo/metabolismo , Neovascularización Fisiológica , Diálisis Peritoneal , Biomarcadores/metabolismo , Línea Celular , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Impedancia Eléctrica , Células Epiteliales/efectos de los fármacos , Epitelio/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Mesodermo/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Permeabilidad , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
11.
J Exp Med ; 218(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34254999

RESUMEN

Astrocytes, a major glial cell type in the brain, play a critical role in supporting the progression of medulloblastoma (MB), the most common malignant pediatric brain tumor. Through lineage tracing analyses and single-cell RNA sequencing, we demonstrate that astrocytes are predominantly derived from the transdifferentiation of tumor cells in relapsed MB (but not in primary MB), although MB cells are generally believed to be neuronal-lineage committed. Such transdifferentiation of MB cells relies on Sox9, a transcription factor critical for gliogenesis. Our studies further reveal that bone morphogenetic proteins (BMPs) stimulate the transdifferentiation of MB cells by inducing the phosphorylation of Sox9. Pharmacological inhibition of BMP signaling represses MB cell transdifferentiation into astrocytes and suppresses tumor relapse. Our studies establish the distinct cellular sources of astrocytes in primary and relapsed MB and provide an avenue to prevent and treat MB relapse by targeting tumor cell transdifferentiation.


Asunto(s)
Astrocitos/patología , Neoplasias Cerebelosas/patología , Meduloblastoma/patología , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/farmacología , Transdiferenciación Celular/efectos de los fármacos , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/genética , Meduloblastoma/metabolismo , Ratones Transgénicos , Receptor Patched-1/genética , Receptor Patched-1/metabolismo , Fosforilación , Pirazoles/farmacología , Pirimidinas/farmacología , Factor de Transcripción SOX9/metabolismo , Análisis de la Célula Individual , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Toxicol In Vitro ; 76: 105220, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34271082

RESUMEN

Recently, more and more attention has been focused on silica nanoparticles (SiNPs) as they are increasingly used in various fields. Yet, their biological effects, especially on human beings, largely remain unknown. This study was implanted to assess the biological responses in vitro elicited by human macrophages exposed to the SiNPs and to explore its toxicity and fibrosis biomarker. We found that SiNPs suppressed the viability of THP-1 cells in a dose-dependent manner while they triggered apoptosis and promoted the secretion of inflammatory factors. Next, SiNPs-induced macrophage supernatant was used to act on fibroblast (MRC-5), indicating that the expression of hydroxyproline (Hyp), α-SMA, and collagonIin MRC-5 increased after SiNPs treatment. To further explore the biomarker of fibrosis, Liquid-mass spectrometry facilitated quantitative proteomics, identified 3247 proteins, of which 791 proteins were expressed differentially in human embryonic lung fibroblasts after treated with SiNPs. In conclusion, our observations suggest that SiNPs induced THP-1-derived macrophage damage and apoptosis. Moreover, SiNPs induced macrophages to secrete cytokines that promote fibroblasts' proliferation and differentiation and changed protein expression in MRC-5 cells, regulating biological processes such as apoptosis, protein synthesis, and cell growth. Among these results, our findings could provide a basis for determining fibrosis biomarkers of silica nanoparticle exposure.


Asunto(s)
Fibroblastos/efectos de los fármacos , Nanopartículas/toxicidad , Dióxido de Silicio/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Citocinas/metabolismo , Fibroblastos/metabolismo , Fibroblastos/fisiología , Humanos , Pulmón/citología , Pulmón/embriología , Macrófagos/efectos de los fármacos , Proteómica
13.
Anticancer Res ; 41(8): 3717-3729, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34281830

RESUMEN

BACKGROUND/AIM: Interleukin 6 (IL6) is increased in patients with progressive prostate cancer and induces its transdifferentiation to neuroendocrine prostate cancer. Neuroendocrine prostate cancer has become one of the greatest challenges in treating castration-resistant disease and is linked to poor prognosis. It is necessary to understand better the cellular events associated with IL6-mediated neuroendocrine differentiation to prevent it and identify potential new therapeutic targets. MATERIALS AND METHODS: In the present study, an IL6-inducible neuroendocrine differentiation model established specifically for this purpose was applied using LNCaP cells. Proteomics and western blot analyses were used to identify proteins involved in neuroendocrine differentiation. Subsequently, the role of gelsolin (GSN) in the neuroendocrine differentiation model was characterized (knock-down analyses, microscopic co-localization analyses, apoptosis assay) and GSN expression levels in patient material were investigated. RESULTS: This study revealed that GSN is a crucial factor in the neuroendocrine differentiation process. CONCLUSION: It was shown that siRNA-mediated knock-down of GSN can inhibit neuroendocrine differentiation, making it a valid target for preventing IL6-mediated neuroendocrine differentiation.


Asunto(s)
Gelsolina/metabolismo , Neoplasias de la Próstata/patología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/fisiología , Gelsolina/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucina-6/metabolismo , Interleucina-6/farmacología , Masculino , Células Neuroendocrinas/patología , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/mortalidad , Neoplasias de la Próstata/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo
14.
Aging (Albany NY) ; 13(12): 16749-16762, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34170851

RESUMEN

Our previous study showed that bone marrow mesenchymal stem cell derived exosomes (BMSC-Exos) suppress high phosphorus (Pi)-induced calcification of vascular smooth muscle cells (VSMCs). However, the mechanism had remained unclear. This study aimed to investigate the mechanism by which BMSC-Exos inhibit vascular calcification (VC). We found that BMSC-Exos reduced high Pi-induced Runx2, osteocalcin and BMP2 expression and inhibited the calcium deposition. Gene expression of human VSMCs stimulated by Pi or Pi plus BMSC-Exos (Pi + Exo) was systematically examined by microarray technology. NONHSAT 084969.2 and transcription factor p65 expression was significantly lower in the Pi + Exo group compared with the Pi group. This finding indicated that NONHSAT 084969.2 and the nuclear factor-κB pathway might play an important role in VC inhibition by BMSC-Exos. By silencing NONHSAT 084969.2 with small interfering RNA, Runx2, BMP2, and osteocalcin expression was decreased significantly. The calcified nodule content and alkaline phosphatase activity were reduced after NONHSAT 084969.2 inhibition and p65, p50, and IκB kinase-α expression was decreased significantly. These results indicated that BMSC-Exos inhibited Pi-induced transdifferentiation and calcification of VSMCs by regulating the NONHSAT 084969.2/nuclear factor-κB axis.


Asunto(s)
Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , FN-kappa B/metabolismo , Fósforo/toxicidad , ARN Largo no Codificante/metabolismo , Calcificación Vascular/genética , Línea Celular , Transdiferenciación Celular/efectos de los fármacos , Análisis por Conglomerados , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Exosomas/ultraestructura , Perfilación de la Expresión Génica , Humanos , Minerales/metabolismo , Miocitos del Músculo Liso/efectos de los fármacos , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/patología
15.
Pharmacol Res ; 170: 105739, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34171492

RESUMEN

Fibrosis is characterized by an abnormal accumulation of extracellular matrix (ECM) constituents in the liver parenchyma that lead to hepatic cirrhosis. After liver injury, the hepatic stellate cells (HSCs) undergo a response called "activation", transforming the cells into proliferative, fibrogenic and contractile myofibroblasts, representing the main collagen-producing cells in the injured tissue. Activated HSCs are considered as pro-inflammatory cells producing cytokines and several hepatomatogens; they are additionally involved in the recruitment of Kupffer cells, circulating monocytes and macrophages through the production of chemokines. Moreover, HSC have been proposed as being involved in the development of insulin resistance mainly mediated by their inflammatory properties, which undeniably links their activation to the development of diabetes and Non-alcoholic fatty liver disease. In addition, when the liver is injured, a complex interaction between hepatocytes and HSCs occurs, inducing mitochondrial dysfunction, which contributes to the accumulation of fats in hepatocytes that trigger to liver lipotoxicity. These mechanisms underlying the activation of HSC suggest their major role in the development of metabolic disorders. It turns out that several molecules including MicroRNAs and proteins have the ability to inhibit the activation and the proliferation of HSCs, which makes them interesting therapeutic targets for the subsequent management of metabolic conditions. This review focuses on the mechanisms and molecular pathways underlying the initiation and onset of metabolic disorders following HSCs activation, as well as on molecular therapeutic targets, which could limit their fibrogenic transdifferentiation and therefore improve the liver condition in the course of metabolic imbalance.


Asunto(s)
Proliferación Celular , Transdiferenciación Celular , Células Estrelladas Hepáticas/metabolismo , Resistencia a la Insulina , Cirrosis Hepática/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/metabolismo , Animales , Antifibróticos/uso terapéutico , Proliferación Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos , Hígado/efectos de los fármacos , Hígado/patología , Cirrosis Hepática/tratamiento farmacológico , Cirrosis Hepática/patología , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/patología , Transducción de Señal
16.
Neurotox Res ; 39(4): 1251-1273, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33945101

RESUMEN

Considering the consequences on human health, in general population and workplace, associated with the use of new psychoactive substances and their continuous placing on the market, novel in vitro models for neurotoxicology research, applying human-derived CNS cells, may provide a means to understand the mechanistic basis of molecular and cellular alterations in brain. Cytotoxic effects of MAM-2201, a potent-naphthoyl indole derivative-synthetic cannabinoid, have been evaluated applying a panel of human cell-based models of neurons and astrocytes, testing different concentrations (1-30 µM) and exposure times (3-24-48 h). MAM-2201 induced toxicity in primary neuron-like cells (hNLCs), obtained from transdifferentiation of mesenchymal stem cells derived from human umbilical cord. Effects occurred in a concentration- and time-dependent manner. The lowest concentration affecting cell viability, metabolic function, apoptosis, morphology, and neuronal markers (MAP-2, NSE) was 5 µM, and even 1 µM induced apoptosis. Effects appeared early (3 h) and persisted after 24 and 48 h. Similar behavior was evidenced for human D384-astrocytes treated with MAM-2201. Differently, human SH-SY5Y-neurons, both differentiated and undifferentiated, were not sensitive to MAM-2201. On D384, the different altered endpoints were reversed, attenuated, or not antagonized by AM251 indicating that CB1 receptors may partially mediate MAM-2201-induced cytotoxicity. While in hNLCs, all toxic effects caused by MAM-2201 were apparently unrelated to CB-receptors since they were not evidenced by immunofluorescence. The present in vitro findings demonstrate the cytotoxicity of MAM-2201 on human primary neurons (hNLCs) and astrocytes cell line (D384), and support the use of these cellular models as species-specific in vitro tools suitable to clarify the neurotoxicity mechanisms of synthetic cannabinoids.


Asunto(s)
Astrocitos/efectos de los fármacos , Cannabinoides/toxicidad , Indoles/toxicidad , Naftalenos/toxicidad , Neuronas/efectos de los fármacos , Astrocitos/patología , Línea Celular Tumoral , Transdiferenciación Celular/efectos de los fármacos , Transdiferenciación Celular/fisiología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Neuronas/patología
17.
J Cell Physiol ; 236(10): 6836-6851, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-33855709

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a disease of progressive scarring caused by excessive extracellular matrix (ECM) deposition and activation of α-SMA-expressing myofibroblasts. Human antigen R (HuR) is an RNA binding protein that promotes protein translation. Upon translocation from the nucleus to the cytoplasm, HuR functions to stabilize messenger RNA (mRNA) to increase protein levels. However, the role of HuR in promoting ECM production, myofibroblast differentiation, and lung fibrosis is unknown. Human lung fibroblasts (HLFs) treated with transforming growth factor ß1 (TGF-ß1) showed a significant increase in translocation of HuR from the nucleus to the cytoplasm. TGF-ß-treated HLFs that were transfected with HuR small interfering RNA had a significant reduction in α-SMA protein as well as the ECM proteins COL1A1, COL3A, and FN1. HuR was also bound to mRNA for ACTA2, COL1A1, COL3A1, and FN. HuR knockdown affected the mRNA stability of ACTA2 but not that of the ECM genes COL1A1, COL3A1, or FN. In mouse models of pulmonary fibrosis, there was higher cytoplasmic HuR in lung structural cells compared to control mice. In human IPF lungs, there was also more cytoplasmic HuR. This study is the first to show that HuR in lung fibroblasts controls their differentiation to myofibroblasts and consequent ECM production. Further research on HuR could assist in establishing the basis for the development of new target therapy for fibrotic diseases, such as IPF.


Asunto(s)
Transdiferenciación Celular , Proteína 1 Similar a ELAV/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Miofibroblastos/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Proteína 1 Similar a ELAV/genética , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Miofibroblastos/patología , Factor de Crecimiento Transformador beta1/farmacología
18.
Pharmacol Res ; 167: 105414, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33524540

RESUMEN

Cardiac fibrosis is characterized by excessive deposition of extracellular matrix proteins and myofibroblast differentiation. Our previous findings have implicated resistin in cardiac fibrosis; however, the molecular mechanisms underlying this process are still unclear. Here we investigated the role of resistin in fibroblast-to-myofibroblast differentiation and elucidated the pathways involved in this process. Fibroblast-to-myofibroblast transdifferentiation was induced with resistin or TGFß1 in NIH-3T3 and adult cardiac fibroblasts. mRNA and protein expression of fibrotic markers were analyzed by qPCR and immunoblotting. Resistin-knockout mice, challenged with a high-fat diet (HFD) for 20 weeks to stimulate cardiac impairment, were analyzed for cardiac function and fibrosis using histologic and molecular methods. Cardiac fibroblasts stimulated with resistin displayed increased fibroblast-to-myofibroblast conversion, with increased levels of αSma, col1a1, Fn, Ccn2 and Mmp9, with remarkable differences in the actin network appearance. Mechanistically, resistin promotes fibroblast-to-myofibroblast transdifferentiation and fibrogenesis via JAK2/STAT3 and JNK/c-Jun signaling pathways, independent of TGFß1. Resistin-null mice challenged with HFD showed an improvement in cardiac function and a decrease in tissue fibrosis and reduced mRNA levels of fibrogenic markers. These findings are the first to delineate the role of resistin in the process of cardiac fibroblast-to-myofibroblast differentiation via JAK/STAT3 and JNK/c-Jun pathways, potentially leading to stimulation of cardiac fibrosis.


Asunto(s)
Transdiferenciación Celular/fisiología , Fibroblastos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Miocitos Cardíacos/metabolismo , Resistina/farmacología , Factor de Transcripción STAT3/metabolismo , Animales , Transdiferenciación Celular/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Quinasas Janus/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/efectos de los fármacos , Células 3T3 NIH , Resistina/deficiencia
19.
Glycoconj J ; 38(1): 77-87, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33608773

RESUMEN

Liver fibrosis is a continuous wound healing response caused by chronic liver injury, and the activation of hepatic stellate cells (HSCs) is considered as the main event for it. Core fucosylation catalyzed by FUT8 refers to adding the fucosyl moiety to the innermost GlcNAc residue of N-linked oligosaccharides and is involved in many biological processes such as cell differentiation, migration, and signaling transduction. Aberrant core fucosylation is associated with a variety of diseases including cardiovascular disease, tumors and neuroinflammation, but much less is understood in liver fibrosis. Herein, we reported FUT8 mRNA level was increased in patients with liver fibrosis from GEO database and positively correlated with fibrosis progression. FUT8 expression and the core fucosylation were also elevated in TAA-induced mouse liver fibrosis model, and were mainly distributed in the fibrous septum of mouse liver. TGF-ß1, as the most pro-fibrogenic cytokine, could promote the expression of FUT8 and total core fucosylation levels in HSCs in vitro. However, up-regulation of FUT8 in turn inhibited TGF-ß1-induced trans-differentiation, migration and pro-fibrogenic signaling pathways in HSCs. In conclusion, our results suggest that the up-regulation of FUT8 inhibits TGF-ß1-induced HSC activation in a negative feedback loop, and provide potential new therapeutic strategy for liver fibrosis by targeting FUT8.


Asunto(s)
Fucosiltransferasas/genética , Células Estrelladas Hepáticas/metabolismo , Cirrosis Hepática/patología , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Transdiferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fucosiltransferasas/metabolismo , Expresión Génica , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/patología , Humanos , Cirrosis Hepática/inducido químicamente , Cirrosis Hepática/genética , Masculino , Ratones Endogámicos C57BL , Ratas , Transducción de Señal/efectos de los fármacos , Tioacetamida/toxicidad , Factor de Crecimiento Transformador beta1/farmacología , Regulación hacia Arriba
20.
Int J Hematol ; 113(3): 441-455, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33392972

RESUMEN

Adverse vascular events have become a serious clinical problem in chronic myeloid leukemia (CML) patients who receive certain BCR/ABL1 tyrosine kinase inhibitors (TKIs). Studies have shown that endothelial-to-mesenchymal transition (EndMT) can contribute to various vascular diseases. We investigated the effects of TKIs on the development of EndMT in human vascular-endothelial cells (VECs). Exposure of VECs to dasatinib, but not to other TKIs, produced a significant increase in the formation of spindle-shaped cells. This effect was accompanied by a significant increase in expression of the EndMT inducer transforming growth factor-ß (TGF-ß) and mesenchymal markers vimentin, smooth muscle alpha-actin, and fibronectin, as well as a significant decrease in expression of vascular-endothelial markers CD31 and VE-cadherin attributable at least in part to activation of ERK signaling. Inhibitors of TGF-ß and ERK partially attenuated dasatinib-induced EndMT. Interestingly, bosutinib efficiently counteracted dasatinib-induced EndMT and attenuated dasatinib-induced phosphorylation of ERK. Taken together, these results show that dasatinib induces EndMT, which might contribute to the development of vascular toxicity, such as the pulmonary hypertension observed in CML patients receiving dasatinib. Bosutinib could play a distinct role in protecting VECs from EndMT.


Asunto(s)
Compuestos de Anilina/farmacología , Transdiferenciación Celular/efectos de los fármacos , Dasatinib/farmacología , Células Endoteliales/efectos de los fármacos , Nitrilos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Quinolinas/farmacología , Células A549 , Animales , Biomarcadores , Forma de la Célula/efectos de los fármacos , Dasatinib/antagonistas & inhibidores , Células Endoteliales/citología , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células Híbridas , Mesilato de Imatinib/farmacología , Imidazoles/farmacología , Mesodermo , Ratones , Ratones Endogámicos C57BL , Piridazinas/farmacología , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/genética , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA