Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 198
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ecotoxicol Environ Saf ; 272: 116108, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38364764

RESUMEN

The importance of evaluating the cardiotoxicity potential of common chemicals as well as new drugs is increasing as a result of the development of animal alternative test methods using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM). Bisphenol A (BPA), which is used as a main material in plastics, is known as an endocrine-disrupting chemical, and recently reported to cause cardiotoxicity through inhibition of ion channels in CMs even with acute exposure. Accordingly, the need for the development of alternatives to BPA has been highlighted, and structural analogues including bisphenol AF, C, E, F, and S have been developed. However, cardiotoxicity data for analogues of bisphenol are not well known. In this study, in order to evaluate the cardiotoxicity potential of analogues, including BPA, a survival test of hiPSC-CMs and a dual-cardiotoxicity evaluation based on a multi-electrode array were performed. Acute exposure to all bisphenol analogues did not affect survival rate, but spike amplitude, beat period, and field potential duration were decreased in a dose-dependent manner in most of the bisphenols except bisphenol S. In addition, bisphenols, except for bisphenol S, reduced the contractile force of hiPSC-CMs and resulted in beating arrest at high doses. Taken together, it can be suggested that the developed bisphenol analogues could cause cardiotoxicity even with acute exposure, and it is considered that the application of the MEA-based dual-cardiotoxicity evaluation method can be an effective help in the development of safe alternatives.


Asunto(s)
Compuestos de Bencidrilo , Células Madre Pluripotentes Inducidas , Miocitos Cardíacos , Animales , Humanos , Cardiotoxicidad/etiología , Células Madre Pluripotentes Inducidas/fisiología , Fenoles/toxicidad
2.
Biochem Biophys Res Commun ; 555: 67-73, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33813278

RESUMEN

A recent in vitro cardiovascular safety pharmacology test uses cardiomyocytes derived from human induced pluripotent stem cells (hiPSCs) to overcome the limitations of the classical test systems, such as species differences and local channel analysis. The Comprehensive in vitro Proarrhythmia Assay (CiPA) is a new proarrhythmia screening paradigm proposed by a CiPA steering expert group, which essentially requires iPSCs derived cardiomyocyte-based electrophysiological evaluation technology. Moreover, the measurement of the contractile force is also emerging as an important parameter to recapitulate non-proarrhythmic cardiotoxicity. Therefore, we constructed an multielectrode assay (MEA) evaluation method that can measure the electrophysiological changes with 6 reference drugs in hiPSC-derived cardiomyocytes. Subsequently, it was confirmed that the electrophysiological were changed in accordance with the mechanism of action of the drugs. Furthermore, based on the multi-probe impedance, we confirmed the decrease in contractile force due to treatment with drugs, and developed a platform to evaluate cardiotoxicity according to drugs along with field potential changes. Our excitation-contraction coupling cardiotoxicity assessment is considered to be more supportive in cardiac safety studies on pharmacologic sensitivity by complementing each assessment parameter.


Asunto(s)
Cardiotoxicidad/etiología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Pruebas de Toxicidad/métodos , Bloqueadores de los Canales de Calcio/toxicidad , Cardiotoxicidad/patología , Células Cultivadas , Electrodos , Humanos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/citología , Nifedipino/toxicidad , Quinidina/toxicidad , Pruebas de Toxicidad/instrumentación
3.
Stem Cells ; 37(5): 623-630, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30721559

RESUMEN

The derivation of human embryonic stem cells (hESCs) by somatic cell nuclear transfer (SCNT) has prompted a re-emerging interest in using such cells for therapeutic cloning. Despite recent advancements in derivation protocols, the functional potential of CHA-NT4 derived cells is yet to be elucidated. For this reason, this study sought to differentiate CHA-NT4 cells toward an endothelial lineage in order to evaluate in vitro and in vivo functionality. To initial differentiation, embryoid body formation of CHA-NT4 was mediated by concave microwell system which was optimized for hESC-endothelial cell (EC) differentiation. The isolated CD31+ cells exhibited hallmark endothelial characteristics in terms of morphology, tubule formation, and ac-LDL uptake. Furthermore, CHA-NT4-derived EC (human nuclear transfer [hNT]-ESC-EC) transplantation in hind limb ischemic mice rescued the hind limb and restored blood perfusion. These findings suggest that hNT-ESC-EC are functionally equivalent to hESC-ECs, warranting further study of CHA-NT4 derivatives in comparison to other well established pluripotent stem cell lines. This revival of human SCNT-ESC research may lead to interesting insights into cellular behavior in relation to donor profile, mitochondrial DNA, and oocyte quality. Stem Cells 2019;37:623-630.


Asunto(s)
Diferenciación Celular/genética , Células Endoteliales/trasplante , Células Madre Embrionarias Humanas/trasplante , Células Madre Pluripotentes Inducidas/trasplante , Animales , Miembro Posterior/patología , Miembro Posterior/trasplante , Humanos , Isquemia/terapia , Ratones , Técnicas de Transferencia Nuclear
4.
Microvasc Res ; 126: 103912, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31433972

RESUMEN

Critical limb ischemia is one of the most common types of peripheral arterial disease. Preclinical development of ischemia therapeutics relies on the availability of a relevant and reproducible in vivo disease model. Thus, establishing appropriate animal disease models is essential for the development of new therapeutic strategies. Currently, the most commonly employed model of hindlimb ischemia is the surgical induction method with ligation of the femoral artery and its branches after skin incision. However, the efficiency of the method is highly variable depending on the availability of skilled technicians. In addition, after surgical procedures, animals can quickly and spontaneously recover from damage, limiting observations of the therapeutic effect of potential agents. The aim of this study was to develop a hindlimb ischemia mouse model with similarities to human ischemic disease. To that end, a photochemical reaction was used to induce thrombosis in the hindlimb. After the photochemical reaction was induced by light irradiation, thrombotic plugs and adjacent red blood cell stasis were observed in hindlimb vessels in the light-irradiated zone. Additionally, the photochemically induced thrombosis maintained the ischemic condition and did not cause notable side effects in mice.


Asunto(s)
Eritrosina , Isquemia/fisiopatología , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica , Trombosis/fisiopatología , Animales , Velocidad del Flujo Sanguíneo , Modelos Animales de Enfermedad , Miembro Posterior , Isquemia/inducido químicamente , Luz , Masculino , Ratones Endogámicos ICR , Procesos Fotoquímicos , Flujo Sanguíneo Regional , Trombosis/inducido químicamente , Factores de Tiempo
5.
J Appl Toxicol ; 38(2): 259-273, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29027214

RESUMEN

An evaluation of intestinal toxicity is important because the mucosal lining of the gastrointestinal tract is the first barrier for oral xenobiotics. Until now, a rat model has been recommended as the standard intestinal toxicity model and the Caco-2 cell line, originated from a human colon adenocarcinoma, has been used as an alternative to this model, but there are limitations regarding cost-effectiveness and the need for mimicry of the human system. In this study, we investigated whether zebrafish could be a valid alternative to rats and Caco-2 cells as an intestinal toxicity model. We focused on intestinal gene expression of cytochrome P450 3A65, oxidative stress, apoptosis, inflammation, and intestinal function. Reverse transcription-quantitative polymerase chain reaction analysis was conducted using three models: zebrafish, Sprague-Dawley rats and Caco-2 cells, and the transcript levels and patterns of indicator genes were analyzed in conjunction with histopathological changes. Our results suggested that representative intestinal toxicants, indomethacin, diclofenac and methotrexate, induced significant transcript level changes in marker genes such as CYP3A, inducible nitric oxide synthase, heme oxygenase 1, superoxide dismutase 1, glutathione peroxidase 1, BCL2 associated X, B-cell lymphoma 2, caspase 9, tumor protein p53, nuclear factor-κB, interleukin-1ß, tumor necrosis factor-alphaα and toll-like receptor 2 in the zebrafish model as in the rat and Caco-2 cells models. These results suggest that zebrafish model is sufficiently worth developing as an intestinal toxicity model that can replace or compensate the rat model or Caco-2 cell model.


Asunto(s)
Alternativas a las Pruebas en Animales , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/etiología , Mucosa Intestinal/efectos de los fármacos , Pruebas de Toxicidad/métodos , Pez Cebra , Animales , Células CACO-2 , Diclofenaco/toxicidad , Humanos , Indometacina/toxicidad , Dosificación Letal Mediana , Metotrexato/toxicidad , Ratas Sprague-Dawley
6.
J Proteome Res ; 16(12): 4455-4467, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28960081

RESUMEN

One of the major goals of the Chromosome-Centric Human Proteome Project (C-HPP) is to fill the knowledge gaps between human genomic information and the corresponding proteomic information. These gaps are due to "missing" proteins (MPs)-predicted proteins with insufficient evidence from mass spectrometry (MS), biochemical, structural, or antibody analyses-that currently account for 2579 of the 19587 predicted human proteins (neXtProt, 2017-01). We address some of the lessons learned from the inconsistent annotations of missing proteins in databases (DB) and demonstrate a systematic proteogenomic approach designed to explore a potential new function of a known protein. To illustrate a cautious and strategic approach for characterization of novel function in vitro and in vivo, we present the case of Na(+)/H(+) exchange regulatory cofactor 1 (NHERF1/SLC9A3R1, located at chromosome 17q25.1; hereafter NHERF1), which was mistakenly labeled as an MP in one DB (Global Proteome Machine Database; GPMDB, 2011-09 release) but was well known in another public DB and in the literature. As a first step, NHERF1 was determined by MS and immunoblotting for its molecular identity. We next investigated the potential new function of NHERF1 by carrying out the quantitative MS profiling of placental trophoblasts (PXD004723) and functional study of cytotrophoblast JEG-3 cells. We found that NHERF1 was associated with trophoblast differentiation and motility. To validate this newly found cellular function of NHERF1, we used the Caenorhabditis elegans mutant of nrfl-1 (a nematode ortholog of NHERF1), which exhibits a protruding vulva (Pvl) and egg-laying-defective phenotype, and performed genetic complementation work. The nrfl-1 mutant was almost fully rescued by the transfection of the recombinant transgenic construct that contained human NHERF1. These results suggest that NHERF1 could have a previously unknown function in pregnancy and in the development of human embryos. Our study outlines a stepwise experimental platform to explore new functions of ambiguously denoted candidate proteins and scrutinizes the mandated DB search for the selection of MPs to study in the future.


Asunto(s)
Fosfoproteínas/fisiología , Proteogenómica/métodos , Intercambiadores de Sodio-Hidrógeno/fisiología , Animales , Caenorhabditis elegans/genética , Diferenciación Celular , Movimiento Celular , Bases de Datos de Proteínas , Femenino , Humanos , Immunoblotting , Espectrometría de Masas , Reproducción , Transgenes , Trofoblastos/citología
7.
J Am Chem Soc ; 139(31): 10880-10886, 2017 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-28708389

RESUMEN

Switchable phototheranostic nanomaterials are of particular interest for specific biosensing, high-quality imaging, and targeted therapy in the field of precision nanomedicine. Here, we develop a "one-for-all" nanomaterial that self-assembles from flexible and versatile phthalocyanine building blocks. The nanostructured phthalocyanine assemblies (NanoPcTBs) display intrinsically unique photothermal and photoacoustic properties. Fluorescence and reactive oxygen species generation can be triggered depending on a targeted, protein-induced, partial disassembly mechanism, which creates opportunities for low-background fluorescence imaging and activatable photodynamic therapy. In vitro evaluations indicate that NanoPcTB has a high selectivity for biotin receptor-positive cancer cells (e.g., A549) compared to biotin receptor-negative cells (e.g., WI38-VA13) and permits a combined photodynamic and photothermal therapeutic effect. Following systemic administration, the NanoPcTBs accumulate in A549 tumors of xenograft-bearing mice, and laser irradiation clearly induces the inhibition of tumor growth.


Asunto(s)
Indoles/química , Nanoestructuras , Fotoquimioterapia/métodos , Fotones , Fármacos Fotosensibilizantes/química , Animales , Línea Celular Tumoral , Fluorescencia , Xenoinjertos , Humanos , Indoles/uso terapéutico , Isoindoles , Ratones , Microscopía Electrónica de Transmisión , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Fármacos Fotosensibilizantes/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
8.
Biochim Biophys Acta ; 1849(6): 709-21, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25725482

RESUMEN

The ubiquitin-proteasome system (UPS) plays an important role in protein quality control, cellular signalings, and cell differentiation through the regulated turnover of key transcription factors in cardiac tissue. However, the molecular mechanism underlying Fbxo25-mediated ubiquitination of cardiac transcription factors remains elusive. We report that an Fbxo25-mediated SCF ubiquitination pathway regulates the protein levels and activities of Tbx5 and Nkx2-5 based on our studies using MG132, proteasome inhibitor, and the temperature sensitive ubiquitin system in ts20 cells. Our data indicate that Fbxo25 directly interacts with Tbx5 and Nkx2-5 in vitro and in vivo. In support of our findings, a dominant-negative mutant of Fbxo25, Fbxo251-236, prevents Tbx5 degradation and increases Tbx5 transcriptional activity in a Tbx5 responsive luciferase assay. Therefore, Fbxo25 facilitates Tbx5 degradation in an SCF-dependent manner. In addition, the silencing of endogenous Fbxo25 increases Tbx5 and Nkx2-5 mRNA levels and suppresses mESC-derived cardiomyocyte differentiation. Likewise, the exogenous expression of FBXO25 downregulates NKX2-5 level in human ESC-derived cardiomyocytes. In myocardial infarction model, Fbxo25 mRNA decreases, whereas the mRNA and protein levels of Tbx5 and Nkx2-5 increase. The protein levels of Tbx5 and Nkx2-5 are regulated negatively by Fbxo25-mediated SCF ubiquitination pathway. Thus, our findings reveal a novel mechanism for regulation of SCFFbox25-dependent Nkx2-5 and Tbx5 ubiquitination in cardiac development and provide a new insight into the regulatory mechanism of Nkx2-5 and Tbx5 transcriptional activity.


Asunto(s)
Diferenciación Celular/genética , Proteínas F-Box/genética , Proteínas de Homeodominio/genética , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/genética , Factores de Transcripción/genética , Animales , Células Madre Embrionarias , Proteínas F-Box/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/biosíntesis , Humanos , Leupeptinas/administración & dosificación , Ratones , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/genética , Proteolisis , Proteínas Ligasas SKP Cullina F-box , Proteínas de Dominio T Box/biosíntesis , Factores de Transcripción/biosíntesis , Activación Transcripcional/efectos de los fármacos
9.
J Cell Biochem ; 117(5): 1112-25, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26448537

RESUMEN

Various source-derived mesenchymal stem cells (MSCs) with multipotent capabilities were considered for cell therapeutics of incurable diseases. The applicability of MSCs depends on the cellular source and on their different in vivo functions, despite having similar phenotypic and cytological characteristics. We characterized MSCs from different sources, including human bone marrow (BM), placenta (PL), and adipose tissue (AT), in terms of the phenotype, surface antigen expression, differentiation ability, proteome reference map, and blood flow recovery in a hindlimb ischemic disease model. The MSCs exhibit different differentiation potentials depending on the cellular source despite having similar phenotypic and surface antigen expression. We identified approximately 90 differentially regulated proteins. Most up- or down-regulated proteins show cytoskeletal or oxidative stress, peroxiredoxin, and apoptosis roles according to their functional involvement. In addition, the PL-MSCs retained a higher therapeutic efficacy than the BM- and AT-MSCs in the hindlimb ischemic disease model. In summary, we examined differentially expressed key regulatory factors for MSCs that were obtained from several cellular sources and demonstrated their differentially expressed proteome profiles. Our results indicate that primitive PL-MSCs have biological advantages relative to those from other sources, making PL-MSCs a useful model for clinical applications of cell therapy.


Asunto(s)
Tejido Adiposo/citología , Células de la Médula Ósea/citología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Madre Mesenquimatosas/citología , Placenta/citología , Adipogénesis , Animales , Western Blotting , Diferenciación Celular , Células Cultivadas , Condrogénesis , Cromatografía Liquida , Electroforesis en Gel Bidimensional , Femenino , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/terapia , Células Madre Mesenquimatosas/metabolismo , Ratones Desnudos , Osteogénesis , Embarazo , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas en Tándem
10.
Wound Repair Regen ; 24(4): 686-94, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27237949

RESUMEN

As wound contraction in the cutaneous layer occurs rapidly in mice, mechanical means are typically used to deliberately expose the wound to properly investigate healing by secondary intention. Previously, silicon rings and splinting models were attempted to analyze histological recovery but prevention of surrounding epidermal cell migration and subsequent closure was minimal. Here, we developed an ideal chimney wound model to evaluate epidermal regeneration in murine under hESC-EC transplantation through histological analysis encompassing the three phases of regeneration: migration, proliferation, and remodeling. Human embryonic stem cell derived endothelial cells (hESC-EC) were transplanted due to possessing a well-known therapeutic effect in angiogenesis which also enhances epidermal repair to depict the process of regeneration. Following a standard 1 mm biopsy punch, a chimney manufactured by modifying a 1.7 mL microtube was simply inserted into the excisional wound to complete the modeling process. Under this model, the excisional wound remained fully exposed for 14 days and even after 4 weeks, only a thin transparent layer of epidermal tissue covered the wound site. This approach is able to more accurately depict epidermal repair in relation to histology while also being a user-friendly and cost-effective way to mimic human recovery in rodents and evaluate epithelial repair induced by a form of therapy.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Embrionarias Humanas/trasplante , Regeneración/fisiología , Trasplante de Células Madre/métodos , Cicatrización de Heridas/fisiología , Heridas Penetrantes/fisiopatología , Animales , Colágeno Tipo VIII/metabolismo , Análisis Costo-Beneficio , Modelos Animales de Enfermedad , Células Endoteliales/citología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Heridas Penetrantes/terapia
11.
Proc Natl Acad Sci U S A ; 110(35): E3281-90, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23918355

RESUMEN

The future of safe cell-based therapy rests on overcoming teratoma/tumor formation, in particular when using human pluripotent stem cells (hPSCs), such as human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Because the presence of a few remaining undifferentiated hPSCs can cause undesirable teratomas after transplantation, complete removal of these cells with no/minimal damage to differentiated cells is a prerequisite for clinical application of hPSC-based therapy. Having identified a unique hESC signature of pro- and antiapoptotic gene expression profile, we hypothesized that targeting hPSC-specific antiapoptotic factor(s) (i.e., survivin or Bcl10) represents an efficient strategy to selectively eliminate pluripotent cells with teratoma potential. Here we report the successful identification of small molecules that can effectively inhibit these antiapoptotic factors, leading to selective and efficient removal of pluripotent stem cells through apoptotic cell death. In particular, a single treatment of hESC-derived mixed population with chemical inhibitors of survivin (e.g., quercetin or YM155) induced selective and complete cell death of undifferentiated hPSCs. In contrast, differentiated cell types (e.g., dopamine neurons and smooth-muscle cells) derived from hPSCs survived well and maintained their functionality. We found that quercetin-induced selective cell death is caused by mitochondrial accumulation of p53 and is sufficient to prevent teratoma formation after transplantation of hESC- or hiPSC-derived cells. Taken together, these results provide the "proof of concept" that small-molecule targeting of hPSC-specific antiapoptotic pathway(s) is a viable strategy to prevent tumor formation by selectively eliminating remaining undifferentiated pluripotent cells for safe hPSC-based therapy.


Asunto(s)
Células Madre Pluripotentes/citología , Bibliotecas de Moléculas Pequeñas , Teratoma/patología , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Apoptosis , Proteína 10 de la LLC-Linfoma de Células B , Diferenciación Celular , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Imidazoles/farmacología , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Mitocondrias/metabolismo , Naftoquinonas/farmacología , Células Madre Pluripotentes/metabolismo , Trasplante de Células Madre , Survivin , Teratoma/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
J Cell Sci ; 126(Pt 11): 2516-24, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23525019

RESUMEN

Induced pluripotent stem cells (iPSCs), generated from somatic cells by overexpression of transcription factors Oct4, Sox2, Klf4 and c-Myc have the same characteristics as pluripotent embryonic stem cells (ESCs). iPSCs reprogrammed from differentiated cells undergo epigenetic modification during reprogramming, and ultimately acquire a similar epigenetic state to that of ESCs. In this study, these epigenetic changes were observed in reprogramming of uniparental parthenogenetic somatic cells. The parthenogenetic pattern of imprinted genes changes during the generation of parthenogenetic maternal iPSCs (miPSCs), a process referred to as pluripotent reprogramming. We determined whether altered imprinted genes are maintained or revert to the parthenogenetic state when the reprogrammed cells are redifferentiated into specialized cell types. To address this question, we redifferentiated miPSCs into neural stem cells (miPS-NSCs) and compared them with biparental female NSCs (fNSCs) and parthenogenetic NSCs (pNSCs). We found that pluripotent reprogramming of parthenogenetic somatic cells could reset parthenogenetic DNA methylation patterns in imprinted genes, and that alterations in DNA methylation were maintained even after miPSCs were redifferentiated into miPS-NSCs. Notably, maternally methylated imprinted genes (Peg1, Peg3, Igf2r, Snrpn and Ndn), whose differentially methylated regions were fully methylated in pNSCs, were demethylated and their expression levels were found to be close to the levels in normal biparental fNSCs after reprogramming and redifferentiation. Our findings suggest that pluripotent reprogramming of parthenogenetic somatic cells followed by redifferentiation leads to changes in DNA methylation of imprinted genes and the reestablishment of gene expression levels to those of normal biparental cells.


Asunto(s)
Desdiferenciación Celular , Metilación de ADN , Impresión Genómica , Células Madre Pluripotentes Inducidas/metabolismo , Células-Madre Neurales/metabolismo , Factores de Transcripción/biosíntesis , Animales , Femenino , Regulación de la Expresión Génica/genética , Células Madre Pluripotentes Inducidas/citología , Factor 4 Similar a Kruppel , Ratones , Ratones Mutantes , Células-Madre Neurales/citología , Factores de Transcripción/genética
13.
Liver Transpl ; 21(9): 1186-94, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25761987

RESUMEN

The study was designed (1) to examine the hypothesis that circulating progenitor cells play a role in the process of de novo regeneration in human liver transplants and that these cells arise from a cell population originating in, or associated with, the bone marrow and (2) to investigate whether the transplanted liver volume has an effect on the circulating recipient-derived progenitor cells that generate hepatocytes during this process. Clinical data and liver tissue characteristics were analyzed in male individuals who underwent sex-mismatched adult-to-adult living donor liver transplantation using dual left lobe grafts. Dual left lobe grafts were examined at the time of transplantation and 19 to 27 days after transplantation. All recipients showed recovery of normal liver function and a significant increase in the volume of the engrafted left lobes after transplantation. Double staining for a Y-chromosome probe and the CD31 antigen showed the presence of hybrid vessels composed of recipient-derived cells and donor cells within the transplanted liver tissues. Furthermore, CD34-expressing cells were observed commingling with Y-chromosome+ cells. The ratio of recipient-derived vessels and the number of Y+ CD34+ cells tended to be higher when smaller graft volumes underwent transplantation. These findings suggest that the recruitment of circulating bone marrow-derived progenitor cells could contribute to vessel formation and de novo regeneration in human liver transplants. Moreover, graft volume may be an important determinant for the active mobilization of circulating recipient-derived progenitor cells and their contribution to liver regeneration.


Asunto(s)
Células de la Médula Ósea/patología , Diferenciación Celular , Hepatocitos/patología , Regeneración Hepática , Trasplante de Hígado/métodos , Hígado/patología , Hígado/cirugía , Células Madre/patología , Adulto , Antígenos CD34/metabolismo , Células de la Médula Ósea/metabolismo , Cromosomas Humanos Y , Femenino , Marcadores Genéticos , Hepatocitos/metabolismo , Humanos , Hígado/metabolismo , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Células Madre/metabolismo , Factores de Tiempo , Resultado del Tratamiento
14.
Biotechnol Lett ; 37(6): 1315-22, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25700820

RESUMEN

Human hemangioblasts exist only during the early embryonic developmental stage thereby limiting the adult cellular source from which to obtain such cells for study. To overcome this, hemangioblast studies have focused on utilizing human embryonic stem cell (hESC) derivatives but current methods are cell-line dependent. Single cell dissociation of a hESC colony quickly led to cell death in most hESC lines due to enzyme treatment which, in turn, reduced induction potential and hemangioblast differentiation efficiency. Therefore, we sought to effectively improve the process of cell dissociation that is adaptable to various hESC lines and increase the initial induction potential of embryoid body (hEB). As a result, we determined an effective cell dissociation method through a comparison study involving various reagents which demonstrated successful dissociation regardless of cell line and enhanced hemangioblast differentiation efficiency.


Asunto(s)
Diferenciación Celular , Técnicas Citológicas/métodos , Cuerpos Embrioides , Hemangioblastos/fisiología , Células Madre Embrionarias Humanas/fisiología , Humanos
15.
Cell Biol Int ; 38(1): 32-40, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23956071

RESUMEN

Generation of reactive oxygen species (ROS) by NADPH oxidase 4 (Nox4) induces the proliferation and migration of adipose-derived stem cells (ASCs). However, the functional role of mitochondrial ROS (mtROS) generation in ASCs is unknown. Therefore, we have investigated whether hypoxia induces the differentiation of ASCs via ROS generation. We also have tried to identify the cellular mechanisms of ROS generation underlying adipocyte differentiation. Hypoxia (2%) and ROS generators, such as antimycin and rotenone, induced adipocyte differentiation, which was attenuated by an ROS scavenger. Although Nox4 generates ROS and regulates proliferation of ASCs, Nox4 inhibition or Nox4 silencing did not inhibit adipocyte differentiation; indeed fluorescence intensity of mito-SOX increased in hypoxia, and treatment with mito-CP, a mtROS scavenger, significantly reduced hypoxia-induced adipocyte differentiation. Phosphorylation of Akt and mTOR was induced by hypoxia, while inhibition of these molecules prevented adipocyte differentiation. Thus hypoxia induces adipocyte differentiation by mtROS generation, and the PI3K/Akt/mTOR pathway is involved.


Asunto(s)
Diferenciación Celular , Hipoxia de la Célula , Especies Reactivas de Oxígeno/metabolismo , Células Madre/citología , Tejido Adiposo/citología , Antimicina A/análogos & derivados , Antimicina A/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Óxidos N-Cíclicos/química , Óxidos N-Cíclicos/farmacología , Depuradores de Radicales Libres/farmacología , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , NADPH Oxidasa 4 , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Compuestos Organofosforados/química , Compuestos Organofosforados/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Rotenona/farmacología , Transducción de Señal , Células Madre/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 33(12): 2839-49, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24092748

RESUMEN

OBJECTIVE: Allogeneic transplantation of human embryonic stem cell (hESC) derivatives has the potential to elicit the patient's immune response and lead to graft rejection. Although hESCs and their derivatives have been shown to have advantageous immune properties in vitro, such observations could not be determined experimentally in vivo because of ethical and technical constraints. However, the generation of humanized mice (hu-mice) harboring a human immune system has provided a tool to perform in vivo immunologic studies of human cells and tissues. Using this model, we sought to examine the therapeutic potential of hESC-derived endothelial cells, human embryonic fibroblasts, and cord blood-derived endothelial progenitor cells in a human immune system environment. APPROACH AND RESULTS: All cell types transplanted in hu-mice showed significantly reduced cell survival during the first 14 days post-transplantation compared with that observed in immunodeficient mice. During this period, no observable therapeutic effects were detected in the hindlimb ischemic mouse models. After this point, the cells demonstrated improved survival and contributed to a long-term improvement in blood perfusion. All cell types showed reduced therapeutic efficacy in hu-mice compared with NOD scid IL2 receptor gamma chain knockout mice. Interestingly, the eventual improvement in blood flow caused by the hESC-derived endothelial cells in hu-mice was not much lower than that observed in NOD scid IL2 receptor gamma chain knockout mice. CONCLUSIONS: These findings suggest that hESC derivatives may be considered a good source for cell therapy and that hu-mice could be used as a preclinical in vivo animal model for the evaluation of therapeutic efficacy to predict the outcomes of human clinical trials.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Células Madre Embrionarias/trasplante , Células Endoteliales/trasplante , Sangre Fetal/inmunología , Isquemia/cirugía , Músculo Esquelético/irrigación sanguínea , Animales , Biomarcadores/sangre , Línea Celular , Supervivencia Celular , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Endoteliales/inmunología , Fibroblastos/inmunología , Fibroblastos/trasplante , Supervivencia de Injerto , Miembro Posterior , Humanos , Inmunocompetencia , Huésped Inmunocomprometido , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Isquemia/inmunología , Isquemia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Neovascularización Fisiológica , Recuperación de la Función , Flujo Sanguíneo Regional , Especificidad de la Especie , Factores de Tiempo , Trasplante Heterólogo
17.
Exp Cell Res ; 319(20): 3190-200, 2013 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-24095950

RESUMEN

Induced pluripotent stem cells (iPSCs) hold tremendous potential for the development of new regenerative medicine therapies and the study of molecular mechanisms of pluripotency and development. However, reactivation of c-Myc, which results in tumor formation in chimeric mice, is a major roadblock in the translation of iPSCs into therapies. Although ectopic expression of c-Myc is not absolutely required for somatic reprogramming, in the absence of c-Myc, the overall efficiency of reprogramming is drastically reduced and the reprogramming time is increased. Subtle, abnormal epigenetic modifications in iPSCs derived in the absence of c-Myc have also been documented. Therefore, we developed a reprogramming method without c-Myc to generate high-quality iPSCs, a prerequisite to harnessing the full potential of iPSCs. In this study, we determined that serum replacement (SR)-based culture conditions dramatically increased the transcription factor-mediated reprogramming of mouse embryonic fibroblast cells (MEFs). The process was shortened to approximately 8 days when Oct4/Sox2/Klf4 (3F)-transduced MEFs were first cultured for 3 days under low serum conditions (LS protocol). The 3F-derived iPSCs that were generated by this method resembled mouse ES cells (mESCs) in morphology, gene expression, and in vitro differentiation. Finally, we observed that 3F-derived iPSC colonies were able to reach definite pluripotency in terms of molecular signatures when the catalytic function of c-Myc was tolerated. The 3F induction of pluripotency described here should facilitate the use of iPSCs and may also facilitate the mechanistic dissection of somatic reprogramming.


Asunto(s)
Separación Celular/métodos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Proteínas Proto-Oncogénicas c-myc/deficiencia , Animales , Células Cultivadas , Factor 4 Similar a Kruppel , Ratones
18.
Biotechnol Lett ; 36(4): 859-68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24322769

RESUMEN

Evaluation of therapeutic effects of transplanted cells in ischemic heart failure models are important issues. However, traditional injection needles that are widely used in clinical practice tend to reduce the amount of functional cells relative to the injected amount. We now describe a cell transplantation technique using a screw needle. After inducing acute myocardial infarction in a rat model, human embryonic stem cell-derived endothelial cells were injected into the infarcted regions with a screw or straight-curved needle. When an equal volume of cells was transplanted, the screw group suffered minimal cell loss, showed improvement in LV wall thickness (74.5 ± 6.2 vs. 64.4 ± 7.8 %), epicardium scar length (19.3 ± 2.8 vs. 24.6 ± 6.4 %), and area of engraft. Thus, even a simple change in the structure of an instrument can have a large impact on transplantation efficiency.


Asunto(s)
Trasplante de Células/métodos , Inyecciones/métodos , Infarto del Miocardio/cirugía , Agujas , Animales , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Ratas , Resultado del Tratamiento
19.
Biotechnol J ; 19(1): e2300311, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37953523

RESUMEN

Beyond single cell two-dimensional (2D) culture, research on organoids that can mimic human organs is rapidly developing. However, there are still problems in commercialization and joint research using organoids due to the lack of technology to safely store organoids. Since organoids are 3D complex structures with a certain size (0.1-5 mm) beyond the size of cells, the conventional cell-level cryopreservation method using cryoprotectant (CPA) cannot overcome the damage caused by volume change due to osmotic pressure difference and ice nucleation. Herein, we attempted to solve such limitations by applying a nanowarming system using CPA with high cell permeability and Fe3 O4 nanoparticles. By performing beat rate measurement, histological analysis, contractility analysis, and multi-electrode array, it was verified that the developed method could significantly improve functional recovery and survival of heart organoids after freezing and thawing. In this study, we demonstrated a successful organoid cryopreservation method based on a Fe3 O4 nanowarming system. The developed technology will provide clues to the field of tissue cryopreservation and spur the application of organoids.


Asunto(s)
Criopreservación , Nanopartículas , Humanos , Criopreservación/métodos , Congelación , Crioprotectores/farmacología , Organoides
20.
Artículo en Inglés | MEDLINE | ID: mdl-38946019

RESUMEN

As research on in vitro cardiotoxicity assessment and cardiac disease modeling becomes more important, the demand for human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) is increasing. However, it has been reported that differentiated hPSC-CMs are in a physiologically immature state compared to in vivo adult CMs. Since immaturity of hPSC-CMs can lead to poor drug response and loss of acquired heart disease modeling, various approaches have been attempted to promote maturation of CMs. Here, we confirm that peroxisome proliferator-activated receptor alpha (PPARα), one of the representative mechanisms of CM metabolism and cardioprotective effect also affects maturation of CMs. To upregulate PPARα expression, we treated hPSC-CMs with fenofibrate (Feno), a PPARα agonist used in clinical hyperlipidemia treatment, and demonstrated that the structure, mitochondria-mediated metabolism, and electrophysiology-based functions of hPSC-CMs were all mature. Furthermore, as a result of multi electrode array (MEA)-based cardiotoxicity evaluation between control and Feno groups according to treatment with arrhythmia-inducing drugs, drug response was similar in a dose-dependent manner. However, main parameters such as field potential duration, beat period, and spike amplitude were different between the 2 groups. Overall, these results emphasize that applying matured hPSC-CMs to the field of preclinical cardiotoxicity evaluation, which has become an essential procedure for new drug development, is necessary.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA