Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Protein & Cell ; (12): 717-733, 2021.
Artículo en Inglés | WPRIM | ID: wpr-888715

RESUMEN

The coronavirus disease 2019 (COVID-19) pandemic is caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is spread primary via respiratory droplets and infects the lungs. Currently widely used cell lines and animals are unable to accurately mimic human physiological conditions because of the abnormal status of cell lines (transformed or cancer cells) and species differences between animals and humans. Organoids are stem cell-derived self-organized three-dimensional culture in vitro and model the physiological conditions of natural organs. Here we showed that SARS-CoV-2 infected and extensively replicated in human embryonic stem cells (hESCs)-derived lung organoids, including airway and alveolar organoids which covered the complete infection and spread route for SARS-CoV-2 within lungs. The infected cells were ciliated, club, and alveolar type 2 (AT2) cells, which were sequentially located from the proximal to the distal airway and terminal alveoli, respectively. Additionally, RNA-seq revealed early cell response to virus infection including an unexpected downregulation of the metabolic processes, especially lipid metabolism, in addition to the well-known upregulation of immune response. Further, Remdesivir and a human neutralizing antibody potently inhibited SARS-CoV-2 replication in lung organoids. Therefore, human lung organoids can serve as a pathophysiological model to investigate the underlying mechanism of SARS-CoV-2 infection and to discover and test therapeutic drugs for COVID-19.


Asunto(s)
Humanos , Adenosina Monofosfato/uso terapéutico , Alanina/uso terapéutico , Células Epiteliales Alveolares/virología , Anticuerpos Neutralizantes/uso terapéutico , COVID-19/virología , Regulación hacia Abajo , Descubrimiento de Drogas , Células Madre Embrionarias Humanas/metabolismo , Inmunidad , Metabolismo de los Lípidos , Pulmón/virología , ARN Viral/metabolismo , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
2.
International Journal of Stem Cells ; : 484-496, 2019.
Artículo en Inglés | WPRIM | ID: wpr-785825

RESUMEN

Previously, the majority of human embryonic stem cells and human induced pluripotent stem cells have been derived on feeder layers and chemically undefined medium. Those media components related to feeder cells, or animal products, often greatly affect the consistency of the cell culture. There are clear advantages of a defined, xeno-free, and feeder-free culture system for human pluripotent stem cells (hPSCs) cultures, since consistency in the formulations prevents lot-to-lot variability. Eliminating all non-human components reduces health risks for downstream applications, and those environments reduce potential immunological reactions from stem cells. Therefore, development of feeder-free hPSCs culture systems has been an important focus of hPSCs research. Recently, researchers have established a variety of culture systems in a defined combination, xeno-free matrix and medium that supports the growth and differentiation of hPSCs. Here we described detailed hPSCs culture methods under feeder-free and chemically defined conditions using vitronetin and TeSR-E8 medium including supplement bioactive lysophospholipid for promoting hPSCs proliferation and maintaining stemness.


Asunto(s)
Animales , Humanos , Técnicas de Cultivo de Célula , Células Madre Embrionarias , Matriz Extracelular , Células Nutrientes , Células Madre Embrionarias Humanas , Células Madre Pluripotentes Inducidas , Células Madre Pluripotentes , Células Madre
4.
Protein & Cell ; (12): 333-350, 2018.
Artículo en Inglés | WPRIM | ID: wpr-757991

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) and Werner syndrome (WS) are two of the best characterized human progeroid syndromes. HGPS is caused by a point mutation in lamin A (LMNA) gene, resulting in the production of a truncated protein product-progerin. WS is caused by mutations in WRN gene, encoding a loss-of-function RecQ DNA helicase. Here, by gene editing we created isogenic human embryonic stem cells (ESCs) with heterozygous (G608G/+) or homozygous (G608G/G608G) LMNA mutation and biallelic WRN knockout, for modeling HGPS and WS pathogenesis, respectively. While ESCs and endothelial cells (ECs) did not present any features of premature senescence, HGPS- and WS-mesenchymal stem cells (MSCs) showed aging-associated phenotypes with different kinetics. WS-MSCs had early-onset mild premature aging phenotypes while HGPS-MSCs exhibited late-onset acute premature aging characterisitcs. Taken together, our study compares and contrasts the distinct pathologies underpinning the two premature aging disorders, and provides reliable stem-cell based models to identify new therapeutic strategies for pathological and physiological aging.


Asunto(s)
Humanos , Envejecimiento , Genética , Fisiología , ADN Helicasas , Genética , Células Madre Embrionarias Humanas , Metabolismo , Fisiología , Cinética , Lamina Tipo A , Genética , Células Madre Mesenquimatosas , Metabolismo , Fisiología , Mutación , Progeria , Genética , Síndrome de Werner , Genética
5.
International Neurourology Journal ; : S34-S45, 2018.
Artículo en Inglés | WPRIM | ID: wpr-740030

RESUMEN

PURPOSE: To evaluate the therapeutic effect of human embryonic stem cell (hESC)-derived multipotent mesenchymal stem cells (M-MSCs) on ketamine-induced cystitis (KC) in rats. METHODS: To induce KC, 10-week-old female rats were injected with 25-mg/kg ketamine hydrochloride twice weekly for 12 weeks. In the sham group, phosphate buffered saline (PBS) was injected instead of ketamine. One week after the final injection of ketamine, the indicated doses (0.25, 0.5, and 1×106 cells) of M-MSCs (KC+M-MSC group) or PBS vehicle (KC group) were directly injected into the bladder wall. One week after M-MSC injection, the therapeutic outcomes were evaluated via cystometry, histological analyses, and measurement of gene expression. Next, we compared the efficacy of M-MSCs at a low dose (1×105 cells) to that of an identical dose of adult bone marrow (BM)-derived MSCs. RESULTS: Rats in the KC group exhibited increased voiding frequency and reduced bladder capacity compared to rats of the sham group. However, these parameters recovered after transplantation of M-MSCs at all doses tested. KC bladders exhibited markedly increased mast cell infiltration, apoptosis, and tissue fibrosis. Administration of M-MSCs significantly reversed these characteristic histological alterations. Gene expression analyses indicated that several genes associated with tissue fibrosis were markedly upregulated in KC bladders. However the expression of these genes was significantly suppressed by the administration of M-MSCs. Importantly, M-MSCs ameliorated bladder deterioration in KC rats after injection of a low dose (1×105) of cells, at which point BM-derived MSCs did not substantially improve bladder function. CONCLUSIONS: This study demonstrates for the first time the therapeutic efficacy of hESC-derived M-MSCs on KC in rats. M-MSCs restored bladder function more effectively than did BM-derived MSCs, protecting against abnormal changes including mast cell infiltration, apoptosis and fibrotic damage.


Asunto(s)
Adulto , Animales , Femenino , Humanos , Ratas , Apoptosis , Médula Ósea , Cistitis , Fibrosis , Expresión Génica , Células Madre Embrionarias Humanas , Ketamina , Mastocitos , Células Madre Mesenquimatosas , Células Madre Multipotentes , Dolor Pélvico , Vejiga Urinaria
6.
International Journal of Stem Cells ; : 105-110, 2018.
Artículo en Inglés | WPRIM | ID: wpr-739916

RESUMEN

BACKGROUND: Human embryonic stem cells (hESCs) have the potential to treat various human disorders currently labeled as incurable and/or terminal illness. However, the fear that the patients' immune system would recognize them as non self and lead to an immune rejection has hampered their use. The main cause for immune rejection is usually the incompatibility of both donor and recipient's major histocompatibility complex (MHC). METHODS: We describe a hESC line developed through a patented technology that does not lead to immune reaction upon transplantation. We have transplanted these cells in >1,400 patients with chronic/terminal conditions and did not observe any immune reaction. No immunosuppressant were administered to these patients. We analyzed the expression levels of MHC-I and MHC-II on the surface of these hESCs using microarray technology. The gene targets for miRNA were analyzed using Gene ontology and DAVID database and pathways for these genes were determined using Reactome and Panther databases. RESULTS: Our results showed that the levels of expression of MHC-I and MHC-II on hESCs is almost negligible and thus the hESCs are less susceptible to an immune rejection. CONCLUSIONS: The hESCs cultured at our facility expresses low levels of MHC-I and do not produce an immune reaction. These can be administered universally and need no cross matching before transplantation.


Asunto(s)
Humanos , Línea Celular , Ontología de Genes , Células Madre Embrionarias Humanas , Sistema Inmunológico , Complejo Mayor de Histocompatibilidad , MicroARNs , Neuronas , Donantes de Tejidos , Cigoto
7.
Journal of Experimental Hematology ; (6): 1186-1193, 2018.
Artículo en Chino | WPRIM | ID: wpr-689508

RESUMEN

<p><b>OBJECTIVE</b>To explore the role of bromodomain and extra terminal (BET) bromodomain in hematopoietic differentiation from human enbryonic stem cells (hESC).</p><p><b>METHODS</b>The effect of BET hematopoietic inhibitor I-BET151 on hematopoietic differentiation from hESC was detected by using a monolayer hematopoietic defferentiation model, immunofluorescence, flow cytometry and real-time PCR; moreover the role of I-BET151 in process of hematopoietic differentiation was explored by adding I-BET151 in different differentiation stages.</p><p><b>RESULTS</b>The analysis results of immunofluorescence, flow cytometry and real-time PCR showed that I-BET 151 significantly inhibited the generation of CD43 positive hematopoietic stem and progenitor cells (HSPCs). It was found that the addition of I-BET 151 in different stages, including APLNR lateral plate mesoderm production, CD34CD31 hemogenic endothelium (HEP) generation and endothelial-to-hematopoietic transition, significantly suppressed the generation of CD43 positive hematopoietic progenitor cells.</p><p><b>CONCLUSION</b>I-BET 151 inhibites hematopoietic differentiation from hESCs at several stages, suggesting that the BET bromodomain plays important roles in multiple stages of hematopoietic differentiation from hESCs.</p>


Asunto(s)
Humanos , Receptores de Apelina , Diferenciación Celular , Citometría de Flujo , Hemangioblastos , Células Madre Hematopoyéticas , Células Madre Embrionarias Humanas
8.
Journal of Experimental Hematology ; (6): 389-394, 2018.
Artículo en Chino | WPRIM | ID: wpr-690979

RESUMEN

<p><b>OBJECTIVE</b>To investigate the effect of embryonic stem cells on the proliferation and apoptosis in human acute myeloid leukemia cell line KG-1a and to explore its potential mechanism.</p><p><b>METHODS</b>The direct co-culture system between human embryonic stem cells H9 and human acute myeloid leukemia cell line KG-1a was established, and CCK8 assay was used to detect the proliferation of KG-1a cells. The changes of cell cycle and apoptosis were detected by flow cytometry (FCM). The mRNA expressions of BCL-2, BAX, Caspase-3 were assessed by RT-PCR. Meanwhile, the protein-expressions of BCL-2, BAX, Caspase-3 were detected by Western blot.</p><p><b>RESULTS</b>The proliferation level of KG-1a cells was significantly inhibited by H9, and the apoptotic rate increased, and the cell cycle was blocked at G/M phase. The mRNA-expression and the protein-expression of BAX and Caspase-3 increased, the mRNA and protein-expression of BCL-2 decreased.</p><p><b>CONCLUSION</b>Embryonic stem cells can inhibit the proliferation of KG-1a and induce the apoptosis that maybe relate with the down-regulation of BCL-2 expression and up-regulation of BAX and caspase-3 expression.</p>


Asunto(s)
Humanos , Apoptosis , Caspasa 3 , Línea Celular Tumoral , Proliferación Celular , Células Madre Embrionarias Humanas , Leucemia Mieloide Aguda , Proteínas Proto-Oncogénicas c-bcl-2 , Proteína X Asociada a bcl-2
9.
Protein & Cell ; (12): 379-393, 2017.
Artículo en Inglés | WPRIM | ID: wpr-757327

RESUMEN

Human pluripotent stem cells (hPSCs) are an important system to study early human development, model human diseases, and develop cell replacement therapies. However, genetic manipulation of hPSCs is challenging and a method to simultaneously activate multiple genomic sites in a controllable manner is sorely needed. Here, we constructed a CRISPR-ON system to efficiently upregulate endogenous genes in hPSCs. A doxycycline (Dox) inducible dCas9-VP64-p65-Rta (dCas9-VPR) transcription activator and a reverse Tet transactivator (rtTA) expression cassette were knocked into the two alleles of the AAVS1 locus to generate an iVPR hESC line. We showed that the dCas9-VPR level could be precisely and reversibly controlled by the addition and withdrawal of Dox. Upon transfection of multiplexed gRNA plasmid targeting the NANOG promoter and Dox induction, we were able to control NANOG gene expression from its endogenous locus. Interestingly, an elevated NANOG level promoted naïve pluripotent gene expression, enhanced cell survival and clonogenicity, and enabled hESCs to integrate with the inner cell mass (ICM) of mouse blastocysts in vitro. Thus, iVPR cells provide a convenient platform for gene function studies as well as high-throughput screens in hPSCs.


Asunto(s)
Animales , Humanos , Ratones , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Doxiciclina , Farmacología , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas , Metabolismo , Proteína Homeótica Nanog , Genética , Células Madre Pluripotentes , Metabolismo
10.
Tissue Engineering and Regenerative Medicine ; (6): 453-464, 2017.
Artículo en Inglés | WPRIM | ID: wpr-655769

RESUMEN

Human embryonic stem cell (hESC) culture system has been changing culture conditions from conventional to xeno-free for therapeutic cell applications, and N-glycolylneuraminic acid (Neu5Gc) could be a useful indicator of xenogeneic contaminations in hESCs because human cells can no longer produce it genetically. We set up the humanized culture condition using commercially available humanized materials and two different adaptation methods: sequential or direct. SNUhES4 and H1 hESC lines, previously established in conventional culture conditions, were maintained using the humanized culture condition and were examined for the presence of Neu5Gc. The hESCs showed the same morphology and character as those of the conventional culture condition. Moreover, they were negative for Neu5Gc within two passages without loss of pluripotency. This study suggested that this method can effectively cleanse previously established hESC lines, bringing them one step closer to being clinical-grade hESCs.


Asunto(s)
Humanos , Células Madre Embrionarias Humanas , Métodos
11.
IBJ-Iranian Biomedical Journal. 2017; 21 (1): 24-31
en Inglés | IMEMR | ID: emr-185664

RESUMEN

Background: Mesenchymal stem cells [MSCs] are important candidates for MSC-based cellular therapy. Current paradigm states that MSCs support local progenitor cells in damaged tissue through paracrine signaling. Therefore, the study of paracrine effects and secretome of MSCs could lead to the appreciation of mechanisms and molecules associated with the therapeutic effects of these cells. This study analyzed anti-inflammatory and immune-modulatory effects of MSC secretomes derived from embryonic stem cells [ESCs] and bone marrow cells after hypoxia and normoxia preconditioning


Methods: ESCs differentiated into MSCs and characterized by flow cytometry as well as by differentiation into adipocytes and osteoblasts. The experimental groups were consisted of individual groups of ESC-MSCs and BM-MSCs [bone marrow-derived mesenchymal stromal cells], which were preconditioned with either hypoxia or normoxia for 24, 48 and 72 h. After collecting the cell-free medium from each treatment, secretomes were concentrated by centrifugal filters. Using a peripheral blood mononuclear cell [PBMC] assay and ELISA, IL-10 concentration in PBMCs was evaluated after their incubation with different secretomes from preconditioned and non-preconditioned MSCs


Results: A significant difference was observed between ESC-MSC normoxia and ESC-MSC hypoxia in IL-10 concentration, and normoxia secretomes increased IL-10 secretion from PBMCs. Moreover, the strongest IL-10 secretion from PBMCs could be detected after the stimulation by ESC-MSC conditioned secretomes, but not BM-MSC conditioned medium


Conclusions: Human hypoxia preconditioned ESC-MSC secretome indicated stronger immune-modulatory effects compared to BMMSC conditioned medium. It could be suggested that induced MSCs confer less immune-modulatory effects but produce more inflammatory molecules such as tumor necrosis factor alpha, which needs further investigation


Asunto(s)
Humanos , Células Madre Embrionarias Humanas , Medios de Cultivo Condicionados/farmacología , Hipoxia de la Célula/fisiología , Células Sanguíneas , Leucocitos Mononucleares/fisiología , Interleucina-10/metabolismo
12.
International Journal of Stem Cells ; : 28-37, 2017.
Artículo en Inglés | WPRIM | ID: wpr-29543

RESUMEN

Although microRNAs have emerged as key regulators in diverse cellular processes, the roles of microRNAs are poorly understood in human embryonic stem cells (hESCs) during differentiation into specialized cell types. In this study, we used a microRNA array with 799 human microRNA probes to examine the expression profiles of microRNAs in hESCs during differentiation into endodermal and mesodermal lineages in vitro. Among the microRNAs analyzed, 7 and 20 microRNAs were enriched in the developmental process of hESCs into mesodermal and endodermal lineages, respectively. In particular, the expression levels of miR-200 family, which is known to regulate the epithelial to mesenchymal transition (EMT), gradually increased in hESCs during differentiation into hepatocytes while they gradually decreased during differentiation into vascular endothelial cells. Downregulation of ZEB1, a direct target of miR-200 family, and E-CADHERIN, a target protein of ZEB1, was observed in hESCs during differentiation into endodermal and mesodermal lineages, respectively. These results indicate that miR-200 family has an important role in determining the cell fate between endodermal and mesodermal lineages from the pluripotent state.


Asunto(s)
Humanos , Humanos , Cadherinas , Regulación hacia Abajo , Endodermo , Células Endoteliales , Hepatocitos , Células Madre Embrionarias Humanas , Técnicas In Vitro , Mesodermo , MicroARNs
13.
Tissue Engineering and Regenerative Medicine ; (6): 163-169, 2017.
Artículo en Inglés | WPRIM | ID: wpr-649847

RESUMEN

MicroRNAs (miRNAs) are small non-coding RNA molecules that participate in transcriptional and post-transcriptional regulation of gene expression. miRNAs have numerous roles in cellular function including embryonic development. Human embryonic stem cells (hESCs) are capable of self-renewal and can differentiate into most of cell types including cardiomyocytes (CMs). These characteristics of hESCs make them considered as an important model for studying human embryonic development and tissue specific differentiation. In this study, we tried to demonstrate the profile of miRNA expression in cardiac differentiation from hESCs. To induce differentiation, we differentiated hESCs into CMs by direct differentiation method and characterized differentiated cells. To analyze the expression of miRNAs, we distinguished (days 4, 8, 12, 16, 20, 24, 28) and isolated RNAs from each differentiation stage. miRNA specific RT-qPCR was performed and the expression profile of miR-1, -30d, -133a, -143, -145, -378a, -499a was evaluated. The expression of all miRs was up-regulated at day 8. miR-143 and -145 expression was also up-regulated at the later stage of differentiation. Only miR-378a expression returned to undifferentiated hESC levels at the other stages of differentiation. In conclusion, we elucidated the expression profile of miRNAs during differentiation into cardiomyocytes from hESCs. Our findings demonstrate the expression of miRNAs was stage-dependent during differentiation and suggest that the differentiation into CMs can be regulated by miRNAs through direct or indirect pathway.


Asunto(s)
Femenino , Humanos , Embarazo , Desarrollo Embrionario , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas , Métodos , MicroARNs , Miocitos Cardíacos , ARN , ARN Pequeño no Traducido
14.
Blood Research ; : 37-43, 2017.
Artículo en Inglés | WPRIM | ID: wpr-226884

RESUMEN

BACKGROUND: Embryonic stem cells (ESCs) can be expanded infinitely in vitro and have the potential to differentiate into hematopoietic stem cells (HSCs); thus, they are considered a useful source of cells for HSC production. Although several technical in vitro methods for engineering HSCs from pluripotent stem cells have been developed, clinical application of HSCs engineered from pluripotent stem cells is restricted because of the possibility of xenogeneic contamination resulting from the use of murine materials. METHODS: Human ESCs (CHA-hES15) were cultured on growth factor-reduced Matrigel-coated dishes in the mTeSR1 serum-free medium. When the cells were 70% confluent, we initiated HSC differentiation by three methods involving (1) knockout serum replacement (KSR), cytokines, TGFb1, EPO, and FLT3L; (2) KSR, cytokines, and bFGF; or (3) cytokines and bFGF. RESULTS: Among the three differentiation methods, the minimal number of cytokines without KSR resulted in the greatest production of HSCs. The optimized method resulted in a higher proportion of CD34⁺CD43⁺ hematopoietic progenitor cells (HPCs) and CD34⁺CD45⁺ HPCs compared to the other methods. In addition, the HSCs showed the potential to differentiate into multiple lineages of hematopoietic cells in vitro. CONCLUSION: In this study, we optimized a two-step, serum-free, animal protein-free, KSR-free, feeder-free, chemically defined monolayer culture method for generation of HSCs and hematopoietic stem and progenitor cells (HSPCs) from human ESCs.


Asunto(s)
Animales , Humanos , Citocinas , Células Madre Embrionarias , Células Madre Hematopoyéticas , Células Madre Embrionarias Humanas , Técnicas In Vitro , Métodos , Células Madre Pluripotentes , Células Madre
15.
Protein & Cell ; (12): 175-186, 2016.
Artículo en Inglés | WPRIM | ID: wpr-757145

RESUMEN

The mammalian brain is heterogeneous, containing billions of neurons and trillions of synapses forming various neural circuitries, through which sense, movement, thought, and emotion arise. The cellular heterogeneity of the brain has made it difficult to study the molecular logic of neural circuitry wiring, pruning, activation, and plasticity, until recently, transcriptome analyses with single cell resolution makes decoding of gene regulatory networks underlying aforementioned circuitry properties possible. Here we report success in performing both electrophysiological and whole-genome transcriptome analyses on single human neurons in culture. Using Weighted Gene Coexpression Network Analyses (WGCNA), we identified gene clusters highly correlated with neuronal maturation judged by electrophysiological characteristics. A tight link between neuronal maturation and genes involved in ubiquitination and mitochondrial function was revealed. Moreover, we identified a list of candidate genes, which could potentially serve as biomarkers for neuronal maturation. Coupled electrophysiological recording and single cell transcriptome analysis will serve as powerful tools in the future to unveil molecular logics for neural circuitry functions.


Asunto(s)
Humanos , Antígenos de Diferenciación , Fenómenos Electrofisiológicos , Fisiología , Regulación de la Expresión Génica , Fisiología , Estudio de Asociación del Genoma Completo , Células Madre Embrionarias Humanas , Biología Celular , Metabolismo , Células Madre Pluripotentes Inducidas , Biología Celular , Metabolismo , Familia de Multigenes , Fisiología , Neuronas , Biología Celular , Metabolismo , Transcriptoma , Fisiología
16.
Tissue Engineering and Regenerative Medicine ; (6): 740-749, 2016.
Artículo en Inglés | WPRIM | ID: wpr-647606

RESUMEN

Pluripotent stem cells can differentiate into many cell types including mature hepatocytes, and can be used in the development of new drugs, treatment of diseases, and in basic research. In this study, we established a protocol leading to efficient hepatic differentiation, and compared the capacity to differentiate into the hepatocyte lineage of human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Optimal combinations of cytokines and growth factors were added to embryoid bodies produced by both types of cell. Differentiation of the cells was assessed with optical and electron microscopes, and hepatic-specific transcripts and proteins were detected by quantitative reverse transcription polymerase chain reaction and immunocytochemistry, respectively. Both types of embryoid body produced polygonal hepatocyte-like cells accompanied by time-dependent up regulation of genes for α-fetoprotein, albumin (ALB), asialoglycoprotein1, CK8, CK18, CK19, CYP1A2, and CYP3A4, which are expressed in fetal and adult hepatocytes. Both types of cell displayed functions characteristic of mature hepatocytes such as accumulation of glycogen, secretion of ALB, and uptake of indocyanine green. And these cells are transplanted into mouse model. Our findings indicate that hESCs and hiPSCs have similar abilities to differentiate into hepatocyte in vitro using the protocol developed here, and these cells are transplantable into damaged liver.


Asunto(s)
Adulto , Animales , Humanos , Ratones , Citocromo P-450 CYP1A2 , Citocromo P-450 CYP3A , Citocinas , Cuerpos Embrioides , Glucógeno , Hepatocitos , Células Madre Embrionarias Humanas , Inmunohistoquímica , Técnicas In Vitro , Verde de Indocianina , Células Madre Pluripotentes Inducidas , Péptidos y Proteínas de Señalización Intercelular , Hígado , Células Madre Pluripotentes , Reacción en Cadena de la Polimerasa , Transcripción Reversa , Regulación hacia Arriba
17.
Korean Journal of Blood Transfusion ; : 209-219, 2016.
Artículo en Coreano | WPRIM | ID: wpr-201804

RESUMEN

Blood transfusion is a well-established cell therapy. However, blood available for transfusion is a limited resource and is available only through donations by healthy volunteers. Moreover, the perpetual and widespread shortage of blood products, problems related to transfusion transmitted infections, and new emerging pathogens have elicited an increase in demand for artificial blood. Therefore, research for alternative RBC substitutes has begun in the 1960s. Hemoglobin-based oxygen carriers (HBOC) and perfluorocarbon-based oxygen carrier (PBOC) were two popular study subjects; however, research on these substitute candidates was halted due to unsatisfactory results and safety issues, including death, in the 1990s. Since then, worldwide efforts to produce RBC have shifted over to stem cell-derived RBC production using cord blood and G-CSF-mobilized peripheral blood stem cells, and some progress has been made. In terms of practical usefulness, however, large-scale production and cost effectiveness are still problematic. Recently, human embryonic stem cells (hESC) and human-induced pluripotent stem cells (hiPSC) have shown the potential to produce RBCs as unlimited cell sources. These two methods using hESCs and hiPSCs are also cost-effective since autologous and O, D negative blood RBCs will be used for alloimmunized patients with multiple alloantibodies or rare blood types (high incidence antigens) as well as universal blood production. We will review the current research on in vitro RBC production from hematopoietic stem cells and pluripotent stem cells and assess future directions in this field.


Asunto(s)
Humanos , Sustitutos Sanguíneos , Transfusión Sanguínea , Tratamiento Basado en Trasplante de Células y Tejidos , Análisis Costo-Beneficio , Eritrocitos , Sangre Fetal , Voluntarios Sanos , Células Madre Hematopoyéticas , Células Madre Embrionarias Humanas , Técnicas In Vitro , Incidencia , Células Madre Pluripotentes Inducidas , Isoanticuerpos , Oxígeno , Células Madre Pluripotentes , Células Madre
18.
International Journal of Stem Cells ; : 264-270, 2016.
Artículo en Inglés | WPRIM | ID: wpr-88588

RESUMEN

BACKGROUND: The normal cells derived from human embryonic stem cells (hESCs) are regarded as substitutes for damaged or dysfunctional adult cells. However, tumorigenicity of hESCs remains a major challenge in clinical application of hESC-derived cell transplantation. Previously, we generated monoclonal antibody (MAb) 57-C11 specific to the surface molecule on undifferentiated hESCs. The aim of this study is to prove whether 57-C11-positive hESCs are pluripotent and tumorigenic in immunodeficient mice. METHODS: Undifferentiated hESCs were mixed with retinoic acid (RA)-differentiated hESCs at different ratios prior to 57-C11-mediated separation. To isolate 57-C11-positive hESCs from the mixture, biotinylated 57-C11 and streptavidin-coated magnetic beads were added to the mixture. Unbound 57-C11-negative hESCs were first isolated after applying magnet to the cell mixture, and 57-C11-bound hESCs were then released from the magnetic beads. In order to measure the efficiency of separation, 57-C11-positive or -negative hESCs were counted after isolation. To evaluate the efficiency of teratoma formation in vivo, 57-C11-positive or negative cells were further injected into left and right, respectively, testes of nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice. RESULTS: Approximately 77~100% of undifferentiated hESCs were isolated after applying 57-C11-coated magnetic beads to the mixed cell populations. Importantly, teratomas were not observed in NOD/SCID mice after the injection of isolated 57-C11-negative hESCs, whereas teratomas were observed with 57-C11-positive hESCs. CONCLUSION: 57-C11-positive hESCs are pluripotent and tumorigenic. The combination of 57-C11 and magnetic beads will be useful to eliminate remaining undifferentiated hESCs for the safe cell transplantation.


Asunto(s)
Adulto , Animales , Humanos , Ratones , Trasplante de Células , Células Madre Embrionarias Humanas , Teratoma , Testículo , Trasplantes , Tretinoina
19.
Tissue Engineering and Regenerative Medicine ; (6): 274-283, 2016.
Artículo en Inglés | WPRIM | ID: wpr-649677

RESUMEN

Hemangioblasts or blood islands only arise in early development thereby the sources to obtain these bi-potential cells are limited. While previous studies have isolated both lineages in vitro through the hemangioblast, derivation efficiency was rather low due to cellular damage attributed by enzyme usage and fluorescent activated cell sorting (FACS). This study focused on avoiding the use of damaging factors in the derivation of endothelial cells (ECs). Single cell H9-human embryonic stem cells (hESCs) were obtained by using a mild dissociation protocol then human embryoid body (hEB) formation was performed under hemangioblast differentiation conditions. The hEBs were subjected to a two-stage cytokine treatment procedure. Subsequent culture of the adhesive cells in day 4 hEBs gave arise to a seemingly pure population of ECs. The hESC-derived ECs were characterized by identifying signature endothelial gene and protein markers as well as testing for in vitro functionality. Furthermore, in vivo functionality was also confirmed by transplanting the cells in hindlimb ischemic murine models. We demonstrate that the genetic change required for EC derivation precedes blast colony formation. Furthermore, cell damage was prevented by abating enzyme usage and FACS, resulting in a high yield of ECs upon adhesion. Under this method, confluent cultures of ECs were obtainable 4 days after hEB formation which is significantly faster than previous protocols.


Asunto(s)
Animales , Humanos , Adhesivos , Cuerpos Embrioides , Células Madre Embrionarias , Células Endoteliales , Hemangioblastos , Miembro Posterior , Células Madre Embrionarias Humanas , Técnicas In Vitro , Islas , Métodos
20.
Journal of Zhejiang University. Medical sciences ; (6): 105-111, 2016.
Artículo en Chino | WPRIM | ID: wpr-239614

RESUMEN

Embryonic stem cells have unlimited proliferative capacity, which may provide a source of tendon stem/progenitor cells for tissue engineering. Experts of International Science and Technology Collaborative Program of Ministry of Science and Technology have developed a protocol consensus on differentiation of human embryonic stem cells into the tendon cells. The consensus recommends a protocol of two-step generation of human embryonic stem cells into tendon cells: the human embryonic stem cells are first differentiated into mesenchymal stem cells on different material surfaces; then with the scaffold-free tissue engineering tendon formed by high-density planting, the mesenchymal stem cells are induced into tendon cells under static or dynamic mechanical stimulation in vivo and in vitro. Tissue engineering tendon established in vitro by the protocol can be used as a model in toxicological analysis and safety evaluation of tendon-relevant small molecule compounds, medical materials and drugs.


Asunto(s)
Humanos , Diferenciación Celular , Consenso , Células Madre Embrionarias Humanas , Biología Celular , Células Madre Mesenquimatosas , Biología Celular , Tendones , Biología Celular , Ingeniería de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA