Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Tissue Eng ; 15: 20417314241248753, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725732

RESUMEN

Solar ultraviolet (sUV) exposure is known to cause skin damage. However, the pathological mechanisms of sUV on hair follicles have not been extensively explored. Here, we established a model of sUV-exposed skin and its appendages using human induced pluripotent stem cell-derived skin organoids with planar morphology containing hair follicles. Our model closely recapitulated several symptoms of photodamage, including skin barrier disruption, extracellular matrix degradation, and inflammatory response. Specifically, sUV induced structural damage and catagenic transition in hair follicles. As a potential therapeutic agent for hair follicles, we applied exosomes isolated from human umbilical cord blood-derived mesenchymal stem cells to sUV-exposed organoids. As a result, exosomes effectively alleviated inflammatory responses by inhibiting NF-κB activation, thereby suppressing structural damage and promoting hair follicle regeneration. Ultimately, our model provided a valuable platform to mimic skin diseases, particularly those involving hair follicles, and to evaluate the efficacy and underlying mechanisms of potential therapeutics.

2.
Ann Dermatol ; 35(3): 165-172, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37290950

RESUMEN

BACKGROUND: The exact definition of sensitive skin is not established yet. Since its high prevalence and significant influence on quality of life, it has become an important topic of research. Among various ingredients, conditioned media from umbilical cord blood-derived mesenchymal stem cells (UCB-MSC-CM) can be a promising source for the treatment of sensitive skin. OBJECTIVE: We evaluated the efficacy and safety of UCB-MSC-CM on patients with sensitive skin. METHODS: We designed a randomized, single blinded, prospective, split-face comparison study and enrolled thirty patients. All patients underwent nonablative fractional laser over the entire face before UCB-MSC-CM or normal saline was applied. Each facial area was randomly assigned to undergo treatment with either UCB-MSC-CM or normal saline. We performed three sessions at two-week intervals, and final results were assessed on six weeks after the last session. As an outcome measure, we evaluated a five-point global assessment scale, transepidermal water loss (TEWL), erythema index (EI) and Sensitive Scale-10. Twenty seven subjects were included in final analysis. RESULTS: The treated side exhibited greater improvement compared to the untreated side based on a five-point global assessment scale. TEWL, EI of the treated side were significantly lower than those of the untreated side throughout study period. Sensitive Scale-10 was significantly improved after treatment. CONCLUSION: The application of UCB-MSC-CM resulted in improved skin barrier function and reduced inflammatory responsiveness, which could provide beneficial effect on sensitive skin.

3.
Biomed Pharmacother ; 131: 110789, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33152947

RESUMEN

Atopic dermatitis (AD) is an inflammatory skin disease in which type 2 allergic inflammation plays a critical role. In this study, the anti-inflammatory effect of conditioned media from human umbilical cord blood-derived mesenchymal stem cells (USC-CM) was investigated in order to apply it as an effective treatment with a low risk of side effects that can overcome the limitations of AD treatment which is currently in use. We found that USC-CM has various growth factors and cytokines associated with anti-inflammatory effect. RT-PCR and ELISA analysis showed that USC-CM inhibited the levels of type 2 cytokine and chemokine Thymus and activation-regulated chemokine (TARC), TNF-α and IL-6 in TNF-α/IFN-γ-stimulated HaCaT cells. In addition, USC-CM inhibited IL-4 and IL-13 levels in Th2 cells. Therefore, the results of our study demonstrated that USC-CM has anti-inflammatory effect in TNF-α/IFN-γ-stimulated HaCaT cells which associated with the inhibition of the immunoglobulin (IgE) secretion by activating B cell line. Our In vivo results showed that when the USC-CM was applied to lesions of patients with the mild AD for 4 weeks, the skin barrier was strengthened by increasing the level of Corneometer and decreasing the value of transepidermal water loss (TEWL). In conclusion, the results suggest that USC-CM may have therapeutic effect for AD as cosmetics and drug materials.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Dermatitis Atópica/terapia , Células Madre Mesenquimatosas/citología , Piel/patología , Línea Celular , Quimiocinas/inmunología , Citocinas/inmunología , Dermatitis Atópica/inmunología , Femenino , Sangre Fetal/citología , Humanos , Inmunidad/inmunología , Masculino , Piel/inmunología , Resultado del Tratamiento , Pérdida Insensible de Agua/fisiología
4.
Stem Cell Rev Rep ; 16(4): 792-808, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32712868

RESUMEN

Mesenchymal stem cells (MSC) are an important type of cell that are highly recognized for their safety and efficacy as a cell therapy agent. In order to obtain MSC, primary tissues (adipose tissue, bone marrow, and umbilical cord blood) must be used; however, these tissues, especially umbilical cord blood, are difficult to obtain due to various reasons, such as the low birth rate trend. In addition, to maximize the safety and efficacy of MSC as allogenic cell therapeutic agents, it is desirable to minimize the possibility of an immune rejection reaction after in vivo transplantation. This study tried to establish a novel method for producing induced pluripotent stem cells (iPSC)-derived MSC in which the human leukocyte antigen (HLA)-class I gene is knocked out. To do so, dermal fibroblast originated iPSC generation using Yamanaka 4-factor, HLA class I gene edited iPSC generation using CRISPR/Cas9, and differentiation from iPSC to MSC using MSC culture medium was utilized. Through this, HLA-A, B, and C pseudo-homozygous iPSC-derived MSC (KO iMSC) were produced by monoallelically knocking out the polymorphic HLA-A, B, and C genes, which are the major causes of immune rejection during allogenic cell transplantation. Produced KO iMSC possesses multipotency and it was safe in vivo to be able to be differentiated to cartilage. In addition, it was not attacked by natural killer cells unlike HLA class I null cells. In conclusion, KO iMSC that do not induce immune rejection during allogenic cell transplantation can be produced. In the future, KO iMSC can be successfully utilized as allogenic cell therapeutic agents for many recipients through HLA screening.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Mesenquimatosas/citología , Animales , Secuencia de Bases , Diferenciación Celular , Homocigoto , Humanos , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Modelos Biológicos , Reproducibilidad de los Resultados
5.
Acta Biomater ; 86: 269-279, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30599245

RESUMEN

With increasing demand for treatment of glottal insufficiency, several injection materials have been examined. However, biological resorption, degradation of injected materials, and the subsequent need to perform multiple injections still remain major clinical problems. In this study, we fabricated two different growth factor (GF) [single basic fibroblast growth factor (bFGF), single hepatocyte growth factor (HGF), or dual bFGF/HGF]-immobilized polycaprolactone (PCL)/Pluronic F127 microspheres. These materials were investigated for their potential use as bioactive injection laryngoplasty agents. HGF was found to be continuously released over 20 days and the bFGF was found to be continuously released over 25 days, as demonstrated by ELISA assay. Human vocal fold fibroblasts (hVFFs) showed significantly higher proliferative ability on dual GF-immobilized microspheres. GF-immobilized microspheres (bFGF, HGF, and dual GF) were injected into paralyzed vocal folds of New Zealand white rabbits. Through endoscopic observation and H&E staining, we identified that the microspheres remained localized at the injection site, resulting in constant volume augmentation of the paralyzed vocal fold without significant loss of the initial volume after 4 weeks. The expression of genes related to the extracellular matrix (ECM) in the vocal fold was upregulated by dual GF-immobilized microspheres. Furthermore, dual GF-immobilized microspheres inhibited muscle degeneration and upregulation of myogenic-related genes. In conclusion, dual GF-immobilized microspheres passively augmented the volume of the paralyzed vocal fold while actively inducing ECM synthesis at the injected vocal fold and preserving muscle tissue. Dual GF-immobilized microspheres could be a new and promising injection material for paralyzed vocal folds. STATEMENT OF SIGNIFICANCE: Limitation of prolonged augmentation of vocal fold and degeneration of vocal fold tissue still remain as major clinical problems in the treatment of vocal fold paralysis. Herein, we fabricated the polycaprolactone (PCL)/Pluronic F127 microspheres to augment volume of paralyzed vocal folds. On top of that, we additionally immobilized the growth factors (bFGF, HGF, or dual bFGF/HGF) on the surface of these microspheres. We highlight the efficacy of the dual GF-immobilized microspheres which augmented the volume of the paralyzed vocal fold passively, induced ECM synthesis actively at the injected vocal fold and preserved laryngeal muscle tissue. Our results suggest that the dual GF-immobilized microsphere could be a new promising injection material for injection laryngoplasty to treat paralyzed vocal fold.


Asunto(s)
Materiales Biocompatibles/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Factor de Crecimiento de Hepatocito/farmacología , Inyecciones , Pliegues Vocales/patología , Animales , Proliferación Celular/efectos de los fármacos , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Heparina/farmacología , Humanos , Laringe/efectos de los fármacos , Microesferas , Desarrollo de Músculos/efectos de los fármacos , Desarrollo de Músculos/genética , Cadenas Pesadas de Miosina/metabolismo , Poloxámero/química , Poliésteres/química , Conejos , Pliegues Vocales/diagnóstico por imagen , Pliegues Vocales/efectos de los fármacos
6.
ACS Biomater Sci Eng ; 5(3): 1497-1508, 2019 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-33405624

RESUMEN

Various growth factor delivery systems were used in the treatment of glottal insufficiency; however, relatively little attention has been paid to a gene delivery system for aspects of active vocal fold (VF) regeneration. Herein, we present a plasmid DNA (pDNA; bFGF gene encoding) complex-loaded alginate (ALG)/hyaluronic acid (HA) mixture hydrogel dispersed with polycaprolactone (PCL) microspheres that can enhance simultaneous regeneration of VF muscle and lamina propria, as well as have a bulking effect on atrophied VF. We have demonstrated long-term efficacy of bFGF synthesized from pDNA complex-transfected cells in vitro. PCL microspheres-dispersed ALG/HA hydrogel (with or without pDNA complex loading) are injected into rabbit VFs with recurrent laryngeal nerve denervation. The PCL microspheres dispersed in the hydrogel bulking agents remain stable at the applied site, leading to constant medialization of the paralyzed VF without significant initial volume loss even after 24 weeks. A regenerative effect for collagen deposition and HA synthesis around the injected site, which are major components of VF tissue, is well confirmed in the pDNA-complex-loaded hydrogel group. Moreover, the compensation of atrophied VFs also leads to the contact of bilateral VF and the remarkable recovery of voice function in the pDNA-complex-loaded group. Based on the results, pDNA (bFGF encoding) complex-loaded hydrogel dispersed with PCL microspheres may be employed as a bioactive bulking agent for the treatment of glottal insufficiency.

7.
Biochem Biophys Rep ; 16: 96-102, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30417126

RESUMEN

Developing treatments that inhibit skin aging is an important research project. Rejuvenation, which focuses on prevention of skin aging, is one of the major issues. Recent studies suggested that mesenchymal stem cells (MSCs) secrete many cytokines, which are important in wound healing. In this study, we investigated the effect of human umbilical cord blood-derived mesenchymal stem cells conditioned media (USC-CM) in cutaneous wound healing and collagen synthesis. We found that USC-CM has many useful growth factors associated with skin rejuvenation, such as Epithelial Growth Factor (EGF), basic Fibroblast Growth Factor (bFGF), Platelet Derived Growth Factor (PDGF), Hepatocyte Growth Factor (HGF), Collagen type 1, and especially, one of the rejuvenation factors, the growth differentiation factor-11 (GDF-11). Our in vitro results showed that USC-CM stimulate growth and extracellular matrix (ECM) production of Human Dermal Fibroblasts (HDFs) compared to those of other MSCs conditioned media (CM) from different origins. Moreover, we evaluated the roles of GDF-11. The results showed that GDF-11 accelerates growth, migration and ECM production of HDFs. Our In vivo results showed that topical treatment of USC-CM showed anti-wrinkle effect and significantly increased dermal density in women. In conclusion, USC-CM has various useful growth factors including GDF-11 that can stimulate skin rejuvenation by increasing growth and ECM production of HDFs.

8.
Results Probl Cell Differ ; 66: 103-121, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30209656

RESUMEN

Human neural stem cells (hNSC) are multipotent adult stem cells. Various studies are underway worldwide to identify new methods for treatment of neurological diseases using hNSC. This chapter summarizes the latest research trends in and fields for application of patient-specific hNSC using direct phenotypic conversion technology. The aim of the study was to analyze the advantages and disadvantages of current technology and to suggest relevant directions for future hNSC research.


Asunto(s)
Diferenciación Celular , Células-Madre Neurales/citología , Fenotipo , Investigación con Células Madre , Humanos
9.
Int J Mol Sci ; 18(10)2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28946669

RESUMEN

Anti-osteoporotic activity of a blocker of the ubiquitin-proteasome system, bortezomib, has known to be achieved by directly opposed action in increased bone formation by osteoblasts and in decreased bone destruction by osteoclasts. However, the mechanisms underlying the proteasome blocker inhibition of osteoclast differentiation and function are not fully understood. Here, we observed that proteasome inhibitors, such as MG132 and bortezomib, in osteoclasts accelerated the degradation of c-Fms, a cognate receptor of macrophage colony-stimulating factor (M-CSF), and did not affect the amount of receptor activator of nuclear factor kappa-B (RANK), a receptor of receptor activator of nuclear factor kappa-B ligand (RANKL). c-Fms degradation induced by proteasome inhibitors was controlled by the activation of p38/tumor necrosis factor-alpha converting enzyme (TACE)-mediated regulated intramembrane proteolysis (RIPping). This was validated through the restoration of c-Fms using specific inhibitors of p38 and TACE, and a stimulation of p38-dependent TACE. In addition, c-Fms degradation by proteasome inhibition completely blocked M-CSF-mediated intrinsic signalling and led to the suppression of osteoclast differentiation and bone resorption. In a mouse model with intraperitoneal administration of lipopolysaccharide (LPS) that stimulates osteoclast formation and leads to bone loss, proteasome blockers prevented LPS-induced inflammatory bone resorption due to a decrease in the number of c-Fms-positive osteoclasts. Our study showed that accelerating c-Fms proteolysis by proteasome inhibitors may be a therapeutic option for inflammation-induced bone loss.


Asunto(s)
Resorción Ósea/etiología , Resorción Ósea/metabolismo , Inflamación/complicaciones , Osteoclastos/citología , Osteoclastos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Ubiquitina/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Resorción Ósea/patología , Resorción Ósea/prevención & control , Bortezomib/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Inhibidores de Proteasoma/farmacología , Proteolisis , Receptor de Factor Estimulante de Colonias de Macrófagos/genética
10.
Tissue Eng Regen Med ; 14(5): 631-639, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30603516

RESUMEN

Tracheal restenosis is a major obstacle to successful tracheal replacement, and remains the greatest challenge in tracheal regeneration. However, there have been no detailed investigations of restenosis. The present study was performed to analyze the serial changes in recruited inflammatory cells and associated histological changes after tracheal scaffold implantation. Asymmetrically porous scaffolds, which successfully prevented tracheal stenosis in a partial trachea defect model, designed with a tubular shape by electrospinning and reinforced by 3D-printing to reconstruct 2-cm circumferential tracheal defect. Serial rigid bronchoscopy, micro-computed tomography, and histology [H&E, Masson's Trichrome, IHC against α-smooth muscle actin (α-SMA)] were performed 1, 4, and 8 weeks after transplantation. Progressive stenosis developed especially at the site of anastomosis. Neutrophils were the main inflammatory cells recruited in the early stage, while macrophage infiltration increased with time. Recruitment of fibroblasts peaked at 4 weeks and deposition of α-SMA increased from 4 weeks and was maintained through 8 weeks. During the first 8 weeks post-transplantation, neutrophils and macrophages played significant roles in restenosis of the trachea. Antagonists to these would be ideal targets to reduce restenosis and thus play a pivotal role in successful tracheal regeneration.

11.
PLoS One ; 11(4): e0153886, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27077737

RESUMEN

Osteoclasts seem to be metabolic active during their differentiation and bone-resorptive activation. However, the functional role of lactate dehydrogenase (LDH), a tetrameric enzyme consisting of an A and/or B subunit that catalyzes interconversion of pyruvate to lactate, in RANKL-induced osteoclast differentiation is not known. In this study, RANKL treatment induced gradual gene expression and activation of the LDH A2B2 isotype during osteoclast differentiation as well as the LDH A1B3 and B4 isotypes during osteoclast maturation after pre-osteoclast formation. Glucose consumption and lactate production in growth media were accelerated during osteoclast differentiation, together with enhanced expression of H+-lactate co-transporter and increased extracellular acidification, demonstrating that glycolytic metabolism was stimulated during differentiation. Further, oxygen consumption via mitochondria was stimulated during osteoclast differentiation. On the contrary, depletion of LDH-A or LDH-B subunit suppressed both glycolytic and mitochondrial metabolism, resulting in reduced mature osteoclast formation via decreased osteoclast precursor fusion and down-regulation of the osteoclastogenic critical transcription factor NFATc1 and its target genes. Collectively, our findings suggest that RANKL-induced LDH activation stimulates glycolytic and mitochondrial respiratory metabolism, facilitating mature osteoclast formation via osteoclast precursor fusion and NFATc1 signaling.


Asunto(s)
Diferenciación Celular/genética , L-Lactato Deshidrogenasa/genética , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Transducción de Señal/genética , Animales , Células Cultivadas , Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Glucólisis/efectos de los fármacos , Glucólisis/genética , Immunoblotting , Isoenzimas/genética , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Lactatos/metabolismo , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Consumo de Oxígeno/genética , Ligando RANK/farmacología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Int J Biol Sci ; 12(2): 235-45, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26884720

RESUMEN

Different stimuli often activate the same intracellular signaling molecules but trigger distinct cell responses. We explored whether or not MAPK signaling induced by macrophage colony-stimulating factor (M-CSF), which is responsible for osteoclast proliferation, differs from that induced by receptor activator of NF-κB ligand (RANKL), which is essential for inducing osteoclast differentiation. The activation of MAPKs by M-CSF or RANKL differed in terms of the extent and duration of ERK, p38, and JNK phosphorylation as well as the isoform specificity of JNK phosphorylation. In particular, RANKL induced a second wave of MAPK activation coincident with the onset of osteoclast differentiation, whereas M-CSF triggered only a monophasic response. M-CSF was also able to trigger a full MAPK response on restimulation of cells earlier than was RANKL, representing that MAPK resensitization by M-CSF differs from that by RANKL. Furthermore, the adapter protein TRAF6 recruitment to the cytoplasmic tail of RANK in a submembrane compartment is specifically required for RANKL-induced activation of p38 MAPK, expression of osteoclastogenic transcription factors, and osteoclast differentiation, indicating that the switch from proliferation to differentiation in osteoclast precursors is dependent on p38 activation via the RANKL-RANK-TRAF6 axis. Our results suggest that selective control of MAPK signaling induced by M-CSF and by RANKL mediates the proliferation versus differentiation decision in osteoclast precursors.


Asunto(s)
Diferenciación Celular , Sistema de Señalización de MAP Quinasas , Osteoclastos/citología , Animales , Proliferación Celular , Regulación de la Expresión Génica , Factor Estimulante de Colonias de Macrófagos/farmacología , Masculino , Ratones Endogámicos C57BL , Fosforilación , Isoformas de Proteínas/metabolismo , Ligando RANK/metabolismo , Ligando RANK/fisiología , Factor 6 Asociado a Receptor de TNF/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
13.
Proc Natl Acad Sci U S A ; 112(50): 15426-31, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26621717

RESUMEN

In this study, we present a method for assembling biofunctionalized paper into a multiform structured scaffold system for reliable tissue regeneration using an origami-based approach. The surface of a paper was conformally modified with a poly(styrene-co-maleic anhydride) layer via initiated chemical vapor deposition followed by the immobilization of poly-l-lysine (PLL) and deposition of Ca(2+). This procedure ensures the formation of alginate hydrogel on the paper due to Ca(2+) diffusion. Furthermore, strong adhesion of the alginate hydrogel on the paper onto the paper substrate was achieved due to an electrostatic interaction between the alginate and PLL. The developed scaffold system was versatile and allowed area-selective cell seeding. Also, the hydrogel-laden paper could be folded freely into 3D tissue-like structures using a simple origami-based method. The cylindrically constructed paper scaffold system with chondrocytes was applied into a three-ring defect trachea in rabbits. The transplanted engineered tissues replaced the native trachea without stenosis after 4 wks. As for the custom-built scaffold system, the hydrogel-laden paper system will provide a robust and facile method for the formation of tissues mimicking native tissue constructs.


Asunto(s)
Hidrogel de Polietilenoglicol-Dimetacrilato/farmacología , Papel , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Alginatos/química , Animales , Cartílago/efectos de los fármacos , Cartílago/fisiología , Condrocitos/citología , Condrocitos/efectos de los fármacos , Condrocitos/trasplante , Fuerza Compresiva , Ácido Glucurónico/química , Células HeLa , Ácidos Hexurónicos/química , Humanos , Maleatos/química , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Rastreo , Peso Molecular , Neovascularización Fisiológica/efectos de los fármacos , Poliestirenos/química , Conejos , Espectrometría por Rayos X , Tráquea/efectos de los fármacos , Tráquea/fisiología
14.
Oncotarget ; 6(42): 44523-37, 2015 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-26540344

RESUMEN

The tumor suppressor protein p53 is unstable in quiescent cells and undergoes proteosomal degradation. Under conditions of cellular stress, p53 is rapidly stabilized by post-translational modification, thereby escaping degradation and translocating to the nucleus where it activates genes related to cell cycle arrest or apoptosis. Here, we report that the transcription elongation factor Ell3 sensitizes luminal type-cancer cell line, MCF7, which have wild-type p53, to the chemotherapeutic agent cis-diamminedichloroplatinum(II) (CDDP) by stabilizing p53. Overexpression of Ell3 in MCF7 cells suppressed the MDM2-mediated ubiquitin-dependent degradation pathway. In addition, Ell3 promoted binding of p53 to NADH quinone oxidoreductase 1, which is linked to the ubiquitin-independent degradation of p53. We found that Ell3 activates interleukin-20 (IL20) expression, which is linked to the ERK1/2 signaling pathway. Chemical inhibition of ERK1/2 signaling or molecular suppression of IL20 revealed that the ERK1/2 signaling pathway and IL20 are the main causes of p53 stabilization in Ell3-overexpressing MCF7 cells. These findings suggest that the ERK1/2 pathway can be targeted in the rational development of therapies to induce chemosensitization of breast cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Cisplatino/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Factores de Elongación Transcripcional/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Interleucinas/genética , Interleucinas/metabolismo , Células MCF-7 , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Estabilidad Proteica , Proteolisis , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Factores de Elongación Transcripcional/genética , Transfección , Proteína p53 Supresora de Tumor/genética , Ubiquitinación , Regulación hacia Arriba
15.
J Nanosci Nanotechnol ; 15(3): 2342-5, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26413664

RESUMEN

We investigated the structural, morphological, and electrical properties of cuprous oxide (Cu2O) film dependency on substrate type. Thin films grown using RF magnetron sputtering were characterized by scanning electron microscopy, X-ray diffraction (XRD), and Hall effect measurements. Cu2O thin films were deposited onto sapphire (0001), Si (100), and MgO (110) substrates, and showed Cu2O single phase only, which was confirmed by XRD measurement. Relatively larger compressive strain existed in Cu2O film grown on sapphire and Si, while a smaller tensile strain appeared in Cu2O film grown on MgO. Cu2O thin film crystallite sizes showed a linear dependence on strain. Moreover, film carrier concentration and mobility increased with increasing strain, while resistivity decreased with decreasing strain. Cu2O film strain due to induced strain opens the possibility of controlling structural and electrical properties in device applications.

16.
Biomaterials ; 44: 134-42, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25617133

RESUMEN

Although several studies have been successfully undertaken of tracheal reconstruction in terms of the maintaining the framework of the graft, most cases of reconstruction failure have resulted from delayed mucosal regeneration. The purposes of this study were to evaluate whether laminin-coated asymmetrically porous membrane (APM) scaffold enhances mucosal regeneration, to compare the mucosalization capability with mesenchymal stem cell (MSC) seeded APM, and to determine whether laminin coating and MSC seeding has a synergistic effect on mucosal regeneration. We reconstructed the full-thickness anterior tracheal defect of 36 New Zealand White rabbits with the APM scaffold. MSCs were isolated from the rabbit's inguinal fat. The animals were divided into 4 groups by the presence of laminin coating on APM and application of MSC [Group I, -/- (laminin/MSC); Group II, -/+; Group III, +/-; Group IV, +/+]. Endoscopy and histologic evaluation were performed and the results were compared among the groups. The results showed that ciliated columnar epithelium was regenerated earlier in groups II and III than in group I. Furthermore, the application of laminin and MSC had synergistic effects on tracheal epithelial regeneration. These results demonstrate that tracheal reconstruction by laminin-coated APM seeded with MSCs is most effective in enhancing tracheal mucosalization, and appears to be promising strategy in the regenerative treatment of tracheal defects.


Asunto(s)
Laminina/farmacología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Membrana Mucosa/fisiología , Regeneración/efectos de los fármacos , Tráquea/fisiología , Animales , Rastreo Celular , Cilios/ultraestructura , Endoscopía , Técnica del Anticuerpo Fluorescente , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Membrana Mucosa/citología , Conejos , Coloración y Etiquetado , Tráquea/efectos de los fármacos
17.
Stem Cells Dev ; 23(5): 457-66, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24147624

RESUMEN

Lefty expression has been recognized as a stemness marker because Lefty is enriched both in undifferentiated embryonic stem cells (ESCs) and in blastocysts. Here, we examined the function of Lefty1 and Lefty2 in the maintenance of self-renewal and pluripotency of mouse ESCs (mESCs). Suppression of Lefty1 or Lefty2 expression in mESCs did not alter the self-renewal properties of mESCs under nondifferentiating conditions, but suppression of these genes did affect Smad2 phosphorylation and differentiation. Lefty1 knockdown mESCs showed enhanced phosphorylation of Smad2 and increased differentiation potential, whereas Lefty2 knockdown mESCs exhibited reduced phosphorylation of Smad2 and enhanced self-renewal in the presence of a differentiation signal. In vivo, teratomas developed from Lefty2 knockdown mESCs contained massive expansions of immature neuroepithelium, a marker of malignant teratomas. Taken together, these results suggest that optimal expression of Lefty1 and Lefty2 is critical for the balanced differentiation of mESCs into three germ layers.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/citología , Factores de Determinación Derecha-Izquierda/biosíntesis , Células Madre Pluripotentes/citología , Animales , Células Madre Embrionarias/metabolismo , Estratos Germinativos , Factores de Determinación Derecha-Izquierda/genética , Ratones , Células Madre Pluripotentes/metabolismo , Transducción de Señal , Proteína Smad2/genética
18.
Biochem Biophys Res Commun ; 437(4): 557-64, 2013 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-23850691

RESUMEN

Ell3 is a RNA polymerase II transcription elongation factor that is enriched in testis. The C-terminal domain of Ell3 shows strong similarities to that of Ell (eleven-nineteen lysine-rich leukemia gene), which acts as a negative regulator of p53 and regulates cell proliferation and survival. Recent studies in our laboratory showed that Ell3 induces the differentiation of mouse embryonic stem cells by protecting differentiating cells from apoptosis via the promotion of p53 degradation. In this study, we evaluated the function of Ell3 in breast cancer cell lines. MCF-7 cell lines overexpressing Ell3 were used to examine cell proliferation and cancer stem cell properties. Ectopic expression of Ell3 in breast cancer cell lines induces proliferation and 5-FU resistance. In addition, Ell3 expression increases the cancer stem cell population, which is characterized by CD44 (+) or ALDH1 (+) cells. Mammosphere-forming potential and migration ability were also increased upon Ell3 expression in breast cancer cell lines. Through biochemical and molecular biological analyses, we showed that Ell3 regulates proliferation, cancer stem cell properties and drug resistance in breast cancer cell lines partly through the MEK-extracellular signal-regulated kinase signaling pathway. Murine xenograft experiments showed that Ell3 expression promotes tumorigenesis in vivo. These results suggest that Ell3 may play a critical role in promoting oncogenesis in breast cancer by regulating cell proliferation and cancer stem cell properties via the ERK1/2 signaling pathway.


Asunto(s)
Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Factores de Elongación Transcripcional/metabolismo , Familia de Aldehído Deshidrogenasa 1 , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Femenino , Fluorouracilo/farmacología , Perfilación de la Expresión Génica , Humanos , Receptores de Hialuranos/metabolismo , Isoenzimas/metabolismo , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Células Madre Neoplásicas/citología , ARN Interferente Pequeño/metabolismo , Retinal-Deshidrogenasa/metabolismo
19.
Stem Cells ; 31(2): 282-92, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23169579

RESUMEN

Self-renewal and pluripotency are hallmark properties of pluripotent stem cells, including embryonic stem cells (ESCs) and iPS cells. Previous studies revealed the ESC-specific core transcription circuitry and showed that these core factors (e.g., Oct3/4, Sox2, and Nanog) regulate not only self-renewal but also pluripotent differentiation. However, it remains elusive how these two cell states are regulated and balanced during in vitro replication and differentiation. Here, we report that the transcription elongation factor Tcea3 is highly enriched in mouse ESCs (mESCs) and plays important roles in regulating the differentiation. Strikingly, altering Tcea3 expression in mESCs did not affect self-renewal under nondifferentiating condition; however, upon exposure to differentiating cues, its overexpression impaired in vitro differentiation capacity, and its knockdown biased differentiation toward mesodermal and endodermal fates. Furthermore, we identified Lefty1 as a downstream target of Tcea3 and showed that the Tcea3-Lefty1-Nodal-Smad2 pathway is an innate program critically regulating cell fate choices between self-replication and differentiation commitment. Together, we propose that Tcea3 critically regulates pluripotent differentiation of mESCs as a molecular rheostat of Nodal-Smad2/3 signaling.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Pluripotentes/metabolismo , Transducción de Señal/genética , Factores de Elongación Transcripcional/genética , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Endodermo/citología , Endodermo/crecimiento & desarrollo , Endodermo/metabolismo , Perfilación de la Expresión Génica , Factores de Determinación Derecha-Izquierda/genética , Factores de Determinación Derecha-Izquierda/metabolismo , Mesodermo/citología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Proteína Nodal/genética , Proteína Nodal/metabolismo , Células Madre Pluripotentes/citología , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factores de Elongación Transcripcional/metabolismo
20.
Gene Expr ; 16(1): 25-30, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24397209

RESUMEN

Tcea3 is present in high concentrations in mouse embryonic stem cells (mESCs) and functions to activate Lefly1, a negative regulator of Nodal signaling. The Nodal pathway has numerous biological activities, including mesoderm induction and patterning in early embryogenesis. Here, we demonstrate that the suppression of Tcea3 in mESCs shifts the cells from pluripotency into enhanced mesoderm development. Vascular endothelial growth factor A (VEGFA) and VEGFC, major transcription factors that regulate vasculogenesis, are activated in Tcea3 knocked down (Tcea3 KD) mESCs. Moreover, differentiating Tcea3 KD mESCs have perturbed gene expression profiles with suppressed ectoderm and activated mesoderm lineage markers. Most early differentiating Tcea3 KD cells expressed Brachyury-T, a mesoderm marker, whereas control cells did not express the gene. Finally, development of chimeric embryos that included Tcea3 KD mESCs was perturbed.


Asunto(s)
Vasos Sanguíneos/citología , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Factores de Elongación Transcripcional/fisiología , Animales , Vasos Sanguíneos/metabolismo , Células Cultivadas , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Elongación Transcripcional/genética , Factor A de Crecimiento Endotelial Vascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...