Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cell Prolif ; 57(6): e13598, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38196265

RESUMEN

Ecto-mesenchymal cells of mammalian tooth germ develops from cranial neural crest cells. These cells are recognised as a promising source for tooth development and regeneration. Despite the high heterogeneity of the neural crest, the cellular landscape of in vitro cultured cranial neural crest cells (CNCCs) for odontogenesis remains unclear. In this study, we used large-scale single-cell RNA sequencing to analyse the cellular landscape of in vitro cultured mouse CNCCs for odontogenesis. We revealed distinct cell trajectories from primary cells to passage 5 and identified a rare Alx3+/Barx1+ sub-population in primary CNCCs that differentiated into two odontogenic clusters characterised by the up-regulation of Pax9/Bmp3 and Lhx6/Dmp1. We successfully induced whole tooth-like structures containing enamel, dentin, and pulp under the mouse renal capsule using in vitro cultured cells from both cranial and trunk neural crests with induction rates of 26.7% and 22.1%, respectively. Importantly, we confirmed only cells sorted from odontogenic path can induce tooth-like structures. Cell cycle and DNA replication genes were concomitantly upregulated in the cultured NCCs of the tooth induction groups. Our data provide valuable insights into the cell heterogeneity of in vitro cultured CNCCs and their potential as a source for tooth regeneration.


Asunto(s)
Diferenciación Celular , Cresta Neural , Odontogénesis , RNA-Seq , Análisis de la Célula Individual , Animales , Cresta Neural/citología , Cresta Neural/metabolismo , Ratones , Odontogénesis/genética , Análisis de la Célula Individual/métodos , Células Cultivadas , Germen Dentario/metabolismo , Germen Dentario/citología , Análisis de Expresión Génica de una Sola Célula
2.
Sci Bull (Beijing) ; 67(11): 1154-1169, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-36545982

RESUMEN

The spatiotemporal relationships in high-resolution during odontogenesis remain poorly understood. We report a cell lineage and atlas of developing mouse teeth. We performed a large-scale (92,688 cells) single cell RNA sequencing, tracing the cell trajectories during odontogenesis from embryonic days 10.5 to 16.5. Combined with an assay for transposase-accessible chromatin with high-throughput sequencing, our results suggest that mesenchymal cells show the specific transcriptome profiles to distinguish the tooth types. Subsequently, we identified key gene regulatory networks in teeth and bone formation and uncovered spatiotemporal patterns of odontogenic mesenchymal cells. CD24+ and Plac8+ cells from the mesenchyme at the bell stage were distributed in the upper half and preodontoblast layer of the dental papilla, respectively, which could individually induce nonodontogenic epithelia to form tooth-like structures. Specifically, the Plac8+ tissue we discovered is the smallest piece with the most homogenous cells that could induce tooth regeneration to date. Our work reveals previously unknown heterogeneity and spatiotemporal patterns of tooth germs that may lead to tooth regeneration for regenerative dentistry.


Asunto(s)
Células Madre Mesenquimatosas , Diente , Ratones , Animales , Odontogénesis/genética , Germen Dentario , Epitelio
3.
Sci China Life Sci ; 64(12): 2100-2113, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33740188

RESUMEN

A stable, rapid and effective neural differentiation method is essential for the clinical applications of human embryonic stem cells (ESCs) or induced pluripotent stem cells (iPSCs) in treating neurological disorders and diseases. Herein, we established a novel and robust monolayer differentiation method to produce functional neural progenitor cells (NPCs) from human ESC/iPSCs on Type I Collagen. The derived cells not only displayed the requisite markers, but also behaved similarly to classic NPCs both in vitro and in vivo. Upon transplantation into traumatic brain injury model, the derived NPCs facilitated recovery from injury. We also found that SMAD signaling stayed down throughout the differentiation process on Type I Collagen, and the pluripotent signals were rapidly downregulated along with raising up of neural early markers on the third day. Meanwhile, ATAC-seq data showed the related mediation of distinct transcriptome and global chromatin dynamics during NPC induction. Totally, our results thus provide a convenient way to generate NPCs from human ESC/iPSCs for neural diseases' treatment.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias Humanas/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Células-Madre Neurales , Lesiones Traumáticas del Encéfalo/terapia , Técnicas de Cultivo de Célula , Colágeno Tipo I , Humanos , Células-Madre Neurales/trasplante , Análisis de Secuencia de ARN
4.
Life Sci ; 212: 145-149, 2018 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-30290185

RESUMEN

The development of induced pluripotent stem cells (iPS cells) has raised the prospect of patient-specific treatments for various diseases. Theoretically, iPS cell technology avoids the limitations of human embryonic stem cells (ES cells), including poor establishment, ethical issues, and immune rejection of allogeneic transplantation. However, the immunogenicity of iPS cells has attracted the attention of researchers, and it remains unclear whether iPS cells and their derivatives will be recognized as a patient's own cells. Even though iPS-derived functional cells have been used in the treatment of some diseases, the process of somatic cell reprogramming and iPS cell differentiation is time-consuming, making it difficult to use iPS cells in acute illness or injury. In recent years, it has been suggested that iPS cells may be used as allografts by establishing an iPS cell bank and HLA matching, providing a novel strategy for the clinical application of iPS cells. This article provides a concise overview of iPS cell immunogenicity, and summarizes published data regarding the application of iPS cells in both autologous and allogeneic transplantation in order to help develop more reliable biotechnical strategies utilizing iPS cells.


Asunto(s)
Diferenciación Celular , Enfermedad , Células Madre Pluripotentes Inducidas/citología , Trasplante de Células Madre , Animales , Humanos , Trasplante Autólogo , Trasplante Homólogo
5.
Stem Cell Res Ther ; 8(1): 141, 2017 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-28595634

RESUMEN

BACKGROUND: A number of studies have shown that tooth-like structures can be regenerated using induced pluripotent stem cells and mouse embryonic stem (mES) cells. However, few studies have reported the regeneration of tooth-periodontium complex structures, which are more suitable for clinical tooth transplantation. We established an optimized approach to induce high-odontogenic potential dental epithelium derived from mES cells by temporally controlling bone morphogenic protein 4 (BMP4) function and regenerated tooth-periodontium complex structures in vivo. METHODS: First, immunofluorescence and quantitative reverse transcription-polymerase chain reaction were used to identify the watershed of skin and the oral ectoderm. LDN193189 was then used to inhibit the BMP4 receptor around the watershed, followed by the addition of exogenous BMP4 to promote BMP4 function. The generated dental epithelium was confirmed by western blot analysis and immunofluorescence. The generated epithelium was ultimately combined with embryonic day 14.5 mouse mesenchyme and transplanted into the renal capsules of nude mice. After 4 weeks, the tooth-periodontium complex structure was examined by micro-computed tomography (CT) and hematoxylin and eosin (H&E) staining. RESULTS: Our study found that the turning point of oral ectoderm differentiation occurred around day 3 after the embryoid body was transferred to a common culture plate. Ameloblastin-positive dental epithelial cells were detected following the temporal regulation of BMP4. Tooth-periodontium complex structures, which included teeth, a periodontal membrane, and alveolar bone, were formed when this epithelium was combined with mouse dental mesenchyme and transplanted into the renal capsules of nude mice. Micro-CT and H&E staining revealed that the generated tooth-periodontium complex structures shared a similar histological structure with normal mouse teeth. CONCLUSIONS: An optimized induction method was established to promote the differentiation of mES cells into dental epithelium by temporally controlling the function of BMP4. A novel tooth-periodontium complex structure was generated using the epithelium.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células Madre Embrionarias de Ratones , Odontogénesis/genética , Periodoncio/citología , Animales , Diferenciación Celular/genética , Epitelio/crecimiento & desarrollo , Células Madre Pluripotentes Inducidas/clasificación , Mesodermo/citología , Ratones , Regeneración/genética , Diente/citología
6.
Mater Sci Eng C Mater Biol Appl ; 79: 661-670, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28629066

RESUMEN

Glucosamine (GA) is an important cartilage matrix precursor for the glycosaminoglycan biochemical synthesis, and has positive effects on cartilage regeneration, particularly in osteoarthritis therapy. However, it has not been used as a bioactive group in scaffolds for cartilage repair widely. In this study, we synthesized modified polyethylene glycol (PEG) hydrogel with glucosamine and then encapsulated human bone mesenchymal stem cells (hBMSCs) in the hydrogel to induce the differentiation of hBMSCs into chondrocytes in three-dimensional culture. The GA-modified PEG hydrogels promoted the chondrogenesis of hBMSCs, particularly in the concentration of 5mM and 10mM. The subcutaneous transplantation of 10mM GA-modified hydrogels with hBMSCs formed cartilage-like blocks in vivo for 8weeks. Importantly, with glucosamine increase, the modified hydrogels down-regulated the fibrosis and hypertrophic cartilage markers in protein level. Therefore, glucosamine modified PEG hydrogels facilitated the chondrogenesis of hBMSCs, which might represent a new method for cartilage repair using a tissue-engineering approach.


Asunto(s)
Diferenciación Celular , Condrogénesis , Glucosamina , Humanos , Hidrogeles , Células Madre Mesenquimatosas , Polietilenglicoles
7.
Life Sci ; 164: 9-14, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27633838

RESUMEN

AIMS: The aim of this study was to improve a method that induce cartilage differentiation of human embryoid stem cells (hESCs) in vitro, and test the effect of in vivo environments on the further maturation of hESCs derived cells. MAIN METHODS: Embryoid bodies (EBs) formed from hESCs, with serum-free KSR-based medium and mesodermal specification related factors, CHIR, and Noggin for first 8days. Then cells were digested and cultured as micropellets in serum-free KSR-based chondrogenic medium that was supplemented with PDGF-BB, TGF ß3, BMP4 in sequence for 24days. The morphology, FACS, histological staining as well as the expression of chondrogenic specific genes were detected in each stage, and further in vivo experiments, cell injections and tissue transplantations, further verified the formation of chondrocytes. KEY FINDINGS: We were able to obtain chondrocyte/cartilage from hESCs using serum-free KSR-based conditioned medium. qPCR analysis showed that expression of the chondroprogenitor genes and the chondrocyte/cartilage matrix genes. Morphology analysis demonstrated we got PG+COL2+COL1-particles. It indicated we obtained hyaline cartilage-like particles. 32-Day differential cells were injected subcutaneous. Staining results showed grafts developed further mature in vivo. But when transplanted in subrenal capsule, their effect was not good as in subcutaneous. Microenvironment might affect the cartilage formation. SIGNIFICANCE: The results of this study provide an absolute serum-free and efficient approach for generation of hESC-derived chondrocytes, and cells will become further maturation in vivo. It provides evidence and technology for the hypothesis that hESCs may be a promising therapy for the treatment of cartilage disease.


Asunto(s)
Cartílago/fisiología , Técnicas de Cultivo de Célula , Medio de Cultivo Libre de Suero/farmacología , Células Madre Embrionarias Humanas/citología , Regeneración/efectos de los fármacos , Inductores de la Angiogénesis/farmacología , Animales , Becaplermina , Proteína Morfogenética Ósea 4/farmacología , Cartílago/citología , Proteína de la Matriz Oligomérica del Cartílago/genética , Diferenciación Celular/efectos de los fármacos , Medio de Cultivo Libre de Suero/química , Citometría de Flujo , Células Madre Embrionarias Humanas/trasplante , Humanos , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-sis/farmacología , Factor de Crecimiento Transformador beta3/farmacología
8.
PLoS One ; 11(4): e0152893, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27050091

RESUMEN

During embryonic organogenesis, the odontogenic potential resides in dental mesenchyme from the bud stage until birth. Mouse dental mesenchymal cells (mDMCs) isolated from the inductive dental mesenchyme of developing molars are frequently used in the context of tooth development and regeneration. We wondered if and how the odontogenic potential could be retained when mDMCs were cultured in vitro. In the present study, we undertook to test the odontogenic potential of cultured mDMCs and attempted to maintain the potential during culturing. We found that cultured mDMCs could retain the odontogenic potential for 24 h with a ratio of 60% for tooth formation, but mDMCs were incapable of supporting tooth formation after more than 24 h in culture. This loss of odontogenic potential was accompanied by widespread transcriptomic alteration and, specifically, the downregulation of some dental mesenchyme-specific genes, such as Pax9, Msx1, and Pdgfrα. To prolong the odontogenic potential of mDMCs in vitro, we then cultured mDMCs in a serum-free medium with Knockout Serum Replacement (KSR) and growth factors (fibroblastic growth factor 2 and epidermal growth factor). In this new micromilieu, mDMCs could maintain the odontogenic potential for 48 h with tooth formation ratio of 50%. Moreover, mDMCs cultured in KSR-supplemented medium gave rise to tooth-like structures when recombined with non-dental second-arch epithelium. Among the supplements, KSR is essential for the survival and adhesion of mDMCs, and both Egf and Fgf2 induced the expression of certain dental mesenchyme-related genes. Taken together, our results demonstrated that the transcriptomic changes responded to the alteration of odontogenic potential in cultured mDMCs and a new micromilieu partly retained this potential in vitro, providing insight into the long-term maintenance of odontogenic potential in mDMCs.


Asunto(s)
Diente/crecimiento & desarrollo , Animales , Proliferación Celular , Células Cultivadas , Medio de Cultivo Libre de Suero , Ratones , Regeneración , Diente/citología , Diente/fisiología , Transcriptoma
9.
PeerJ ; 4: e1684, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26925321

RESUMEN

Background. Mouse dental mesenchymal cells (mDMCs) from tooth germs of cap or later stages are frequently used in the context of developmental biology or whole-tooth regeneration due to their odontogenic potential. In vitro-expanded mDMCs serve as an alternative cell source considering the difficulty in obtaining primary mDMCs; however, cultured mDMCs fail to support tooth development as a result of functional failures of specific genes or pathways. The goal of this study was to identify the genes that maintain the odontogenic potential of mDMCs in culture. Methods. We examined the odontogenic potential of freshly isolated versus cultured mDMCs from the lower first molars of embryonic day 14.5 mice. The transcriptome of mDMCs was detected using RNA sequencing and the data were validated by qRT-PCR. Differential expression analysis and pathway analysis were conducted to identify the genes that contribute to the loss of odontogenic potential. Results. Cultured mDMCs failed to develop into well-structured tooth when they were recombined with dental epithelium. Compared with freshly isolated mDMCs, we found that 1,004 genes were upregulated and 948 were downregulated in cultured mDMCs. The differentially expressed genes were clustered in the biological processes and signaling pathways associated with tooth development. Following in vitro culture, genes encoding a wide array of components of MAPK, TGF-ß/BMP, and Wnt pathways were significantly downregulated. Moreover, the activities of Bdnf, Vegfα, Bmp2, and Bmp7 were significantly inhibited in cultured mDMCs. Supplementation of VEGFα, BMP2, and BMP7 restored the expression of a subset of downregulated genes and induced mDMCs to form dentin-like structures in vivo. Conclusions. Vegfα, Bmp2, and Bmp7 play a role in the maintenance of odontogenic potential in mDMCs.

10.
PLoS One ; 10(10): e0141346, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26496354

RESUMEN

As a key factor for cell pluripotent and self-renewing phenotypes, SOX2 has attracted scientists' attention gradually in recent years. However, its exact effects in dental pulp stem cells (DPSCs) are still unclear. In this study, we mainly investigated whether SOX2 could affect some biological functions of DPSCs. DPSCs were isolated from the dental pulp of human impacted third molar. SOX2 overexpressing DPSCs (DPSCs-SOX2) were established through retroviral infection. The effect of SOX2 on cell proliferation, migration and adhesion ability was evaluated with CCK-8, trans-well system and fibronectin-induced cell attachment experiment respectively. Whole genome expression of DPSCs-SOX2 was analyzed with RNA microarray. Furthermore, a rescue experiment was performed with SOX2-siRNA in DPSC-SOX2 to confirm the effect of SOX2 overexpression in DPSCs. We found that SOX2 overexpression could result in the enhancement of cell proliferation, migration, and adhesion in DPSCs obviously. RNA microarray analysis indicated that some key genes in the signal pathways associated with cell cycle, migration and adhesion were upregulated in different degree, and the results were further confirmed with qPCR and western-blot. Finally, DPSC-SOX2 transfected with SOX2-siRNA showed a decrease of cell proliferation, migration and adhesion ability, which further confirmed the biological effect of SOX2 in human DPSCs. This study indicated that SOX2 could improve the cell proliferation, migration and adhesion ability of DPSCs through regulating gene expression about cell cycle, migration and adhesion, and provided a novel strategy to develop seed cells with strong proliferation, migration and adhesion ability for tissue engineering.


Asunto(s)
Células Madre Adultas/fisiología , Pulpa Dental/citología , Factores de Transcripción SOXB1/metabolismo , Adhesión Celular , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Células Cultivadas , Expresión Génica , Humanos , Factores de Transcripción SOXB1/genética
11.
Cell Regen ; 4(1): 4, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26246892

RESUMEN

BACKGROUND: Hematopoiesis is a progressive process collectively controlled by an elaborate network of transcription factors (TFs). Among these TFs, GATA2 has been implicated to be critical for regulating multiple steps of hematopoiesis in mouse models. However, whether similar function of GATA2 is conserved in human hematopoiesis, especially during early embryonic development stage, is largely unknown. RESULTS: To examine the role of GATA2 in human background, we generated homozygous GATA2 knockout human embryonic stem cells (GATA2 (-/-) hESCs) and analyzed their blood differentiation potential. Our results demonstrated that GATA2 (-/-) hESCs displayed attenuated generation of CD34(+)CD43(+) hematopoietic progenitor cells (HPCs), due to the impairment of endothelial to hematopoietic transition (EHT). Interestingly, GATA2 (-/-) hESCs retained the potential to generate erythroblasts and macrophages, but never granulocytes. We further identified that SPI1 downregulation was partially responsible for the defects of GATA2 (-/-) hESCs in generation of CD34(+)CD43(+) HPCs and granulocytes. Furthermore, we found that GATA2 (-/-) hESCs restored the granulocyte potential in the presence of Notch signaling. CONCLUSION: Our findings revealed the essential roles of GATA2 in EHT and granulocyte development through regulating SPI1, and uncovered a role of Notch signaling in granulocyte generation during hematopoiesis modeled by human ESCs.

12.
Stem Cell Rev Rep ; 10(5): 663-70, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24917330

RESUMEN

The stem-cell-based tissue-engineering approaches are widely applied in establishing functional organs and tissues for regenerative medicine. Successful generation of induced pluripotent stem cells (iPS cells) and rapid progress of related technical platform provide great promise in the development of regenerative medicine, including organ regeneration. We have previously reported that iPS cells could be an appealing stem cells source contributing to tooth regeneration. In the present paper, we mainly review the application of iPS technology in dental bioengineering and discuss the challenges for iPS cells in the whole tooth regeneration.


Asunto(s)
Bioingeniería , Células Madre Pluripotentes Inducidas/citología , Diente/citología , Animales , Humanos
13.
Life Sci ; 108(1): 22-9, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24834837

RESUMEN

AIMS: Hemophilia A (HA) is a severe, congenital bleeding disorder caused by the deficiency of clotting factor VIII (FVIII). For years, traditional laboratory animals have been used to study HA and its therapies, although animal models may not entirely mirror the human pathophysiology. Human induced pluripotent stem cells (iPSCs) can undergo unlimited self-renewal and differentiate into all cell types. This study aims to generate hemophilia A (HA) patient-specific iPSCs that differentiate into disease-affected hepatocyte cells. These hepatocytes are potentially useful for in vitro disease modeling and provide an applicable cell source for autologous cell therapy after genetic correction. MAIN METHODS: In this study, we mainly generated iPSCs from urine collected from HA patients with integration-free episomal vectors PEP4-EO2S-ET2K containing human genes OCT4, SOX2, SV40LT and KLF4, and differentiated these iPSCs into hepatocyte-like cells. We further identified the genetic phenotype of the FVIII genes and the FVIII activity in the patient-specific iPSC derived hepatic cells. KEY FINDINGS: HA patient-specific iPSCs (HA-iPSCs) exhibited typical pluripotent properties evident by immunostaining, in vitro assays and in vivo assays. Importantly, we showed that HA-iPSCs could differentiate into functional hepatocyte-like cells and the HA-iPSC-derived hepatocytes failed to produce FVIII, but otherwise functioned normally, recapitulating the phenotype of HA disease in vitro. SIGNIFICANCE: HA-iPSCs, particular those generated from the urine using a non-viral approach, provide an efficient way for modeling HA in vitro. Furthermore, HA-iPSCs and their derivatives serve as an invaluable cell source that can be used for gene and cell therapy in regenerative medicine.


Asunto(s)
Diferenciación Celular , Hemofilia A/patología , Hepatocitos/patología , Células Madre Pluripotentes Inducidas/citología , Modelos Biológicos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Factor VIII/biosíntesis , Factor VIII/genética , Terapia Genética/métodos , Hemofilia A/genética , Hepatocitos/citología , Humanos , Factor 4 Similar a Kruppel , Masculino , Fenotipo , Medicina Regenerativa/métodos
14.
Sci China Life Sci ; 57(2): 162-70, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24443177

RESUMEN

The breakthrough development of induced pluripotent stem cells (iPSCs) raises the prospect of patient-specific treatment for many diseases through the replacement of affected cells. However, whether iPSC-derived functional cell lineages generate a deleterious immune response upon auto-transplantation remains unclear. In this study, we differentiated five human iPSC lines from skin fibroblasts and urine cells into neural progenitor cells (NPCs) and analyzed their immunogenicity. Through co-culture with autogenous peripheral blood mononuclear cells (PBMCs), we showed that both somatic cells and iPSC-derived NPCs do not stimulate significant autogenous PBMC proliferation. However, a significant immune reaction was detected when these cells were co-cultured with allogenous PBMCs. Furthermore, no significant expression of perforin or granzyme B was detected following stimulation of autogenous immune effector cells (CD3(+)CD8(-) T cells, CD3(+)CD8(+) T cells or CD3(-)CD56(+) NK cells) by NPCs in both PBMC and T cell co-culture systems. These results suggest that human iPSC-derived NPCs may not initiate an immune response in autogenous transplants, and thus set a base for further preclinical evaluation of human iPSCs.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Antígenos CD/inmunología , Secuencia de Bases , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Cartilla de ADN , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Granzimas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células-Madre Neurales/inmunología , Perforina/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Trasplante de Células Madre
15.
PLoS One ; 8(7): e69617, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23922758

RESUMEN

The groundbreaking discovery of induced pluripotent stem cells (iPS cells) provides a new source for cell therapy. However, whether the iPS derived functional lineages from different cell origins have different immunogenicity remains unknown. It had been known that the cells isolated from extra-embryonic tissues, such as umbilical cord mesenchymal cells (UMCs), are less immunogenic than other adult lineages such as skin fibroblasts (SFs). In this report, we differentiated iPS cells from human UMCs and SFs into neural progenitor cells (NPCs) and analyzed their immunogenicity. Through co-culture with allologous peripheral blood mononuclear cells (PBMCs), we showed that UMCs were indeed less immunogenic than skin cells to simulate proliferation of PBMCs. Surprisingly, we found that the NPCs differentiated from UMC-iPS cells retained low immunogenicity as the parental UMCs based on the PBMC proliferation assay. In cytotoxic expression assay, reactions in most kinds of immune effector cells showed more perforin and granzyme B expression with SF-NPCs stimulation than that with UMC-NPCs stimulation in PBMC co-culture system, in T cell co-culture system as well. Furthermore, through whole genome expression microarray analysis, we showed that over 70 immune genes, including all members of HLA-I, were expressed at lower levels in NPCs derived from UMC-iPS cells than that from SF-iPS cells. Our results demonstrated a phenomenon that the low immunogenicity of the less immunogenic cells could be retained after cell reprogramming and further differentiation, thus provide a new concept to generate functional lineages with lower immunogenicity for regenerative medicine.


Asunto(s)
Diferenciación Celular/inmunología , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Células-Madre Neurales/inmunología , Adulto , Proliferación Celular , Técnicas de Cocultivo , Células Madre Embrionarias/citología , Granzimas/metabolismo , Antígenos HLA/metabolismo , Humanos , Cinética , Leucocitos Mononucleares/citología , Mesodermo/citología , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Perforina/metabolismo , Piel/citología , Cordón Umbilical/citología
16.
Stem Cell Res ; 11(1): 529-39, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23578695

RESUMEN

Induced pluripotent stem cells (iPSCs) hold promise for the treatment of motoneuron diseases because of their distinct features including pluripotency, self-derivation and potential ability to differentiate into motoneurons. However, it is still unknown whether human iPSC-derived motoneurons can functionally innervate target muscles in vivo, which is the definitive sign of successful cell therapy for motoneuron diseases. In the present study, we demonstrated that human iPSCs derived from mesenchymal cells of the umbilical cord possessed a high yield in neural differentiation. Using a chemically-defined in vitro system, human iPSCs efficiently differentiated into motoneurons which displayed typical morphology, expressed specific molecules, and generated repetitive trains of action potentials. When transplanted into the injured musculocutaneous nerve of rats, they survived robustly, extended axons along the nerve, and formed functional connections with the target muscle (biceps brachii), thereby protecting the muscle from atrophy. Our study provides evidence for the first time that human iPSC-derived motoneurons are truly functional not only in vitro but also in vivo, and they have potential for stem cell-based therapies for motoneuron diseases.


Asunto(s)
Enfermedad de la Neurona Motora/terapia , Neuronas Motoras/trasplante , Músculo Esquelético/inervación , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Neuronas Motoras/citología , Neuronas Motoras/fisiología , Ratas , Ratas Sprague-Dawley
17.
Stem Cell Res ; 10(3): 338-48, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23416351

RESUMEN

Induced pluripotent stem cells (iPSCs) exhibit reduced efficiency and higher variability in neural differentiation compared to embryonic stem cells (ESCs). In this study, we showed that mouse iPSCs failed to efficiently give rise to neuronal cells using conventional methods previously established for driving mouse ESC differentiation. We reported a novel approach which remarkably increases neural differentiation of mouse iPSCs. This novel approach initiated embryoid body (EB) formation directly from the whole cell clones isolated from the top of feeder cells. Compared to conventional neural induction methods such as single cell suspension or monolayer culture, the cell clone-derived EB method led to a pronounced increase in directed generation of various types of neural cells including neural stem cells, motoneurons and dopaminergic neurons in response to different inducers. Through gene expression microarray analysis, we identified 14 genes that were highly expressed in the cell clone-derived EBs. Among them, we found that Cdh2, also known as N-cadherin, played important roles in controlling the neural differentiation efficiency of mouse iPSCs. Forced expression of Cdh2 in iPSCs substantially enhanced the differentiation efficiency while knocking-down of Cdh2 by shRNA blocked the neural differentiation. Our results revealed a critical role of Cdh2 in the process of efficient neural differentiation of mouse iPS cells.


Asunto(s)
Cadherinas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/metabolismo , Animales , Cadherinas/antagonistas & inhibidores , Cadherinas/genética , Diferenciación Celular , Línea Celular , Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Trasplante Homólogo
18.
Cell Regen ; 2(1): 6, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-25408878

RESUMEN

BACKGROUND: Tooth is vital not only for a good smile, but also good health. Yet, we lose tooth regularly due to accidents or diseases. An ideal solution to this problem is to regenerate tooth with patients' own cells. Here we describe the generation of tooth-like structures from integration-free human urine induced pluripotent stem cells (ifhU-iPSCs). RESULTS: We first differentiated ifhU-iPSCs to epithelial sheets, which were then recombined with E14.5 mouse dental mesenchymes. Tooth-like structures were recovered from these recombinants in 3 weeks with success rate up to 30% for 8 different iPSC lines, comparable to H1 hESC. We further detected that ifhU-iPSC derived epithelial sheets differentiated into enamel-secreting ameloblasts in the tooth-like structures, possessing physical properties such as elastic modulus and hardness found in the regular human tooth. CONCLUSION: Our results demonstrate that ifhU-iPSCs can be used to regenerate patient specific dental tissues or even tooth for further drug screening or regenerative therapies.

19.
Stem Cells Int ; 2012: 721538, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23093978

RESUMEN

The amniotic membrane (AM) and amniotic fluid (AF) have a long history of use in surgical and prenatal diagnostic applications, respectively. In addition, the discovery of cell populations in AM and AF which are widely accessible, nontumorigenic and capable of differentiating into a variety of cell types has stimulated a flurry of research aimed at characterizing the cells and evaluating their potential utility in regenerative medicine. While a major focus of research has been the use of amniotic membrane and fluid in tissue engineering and cell replacement, AM- and AF-derived cells may also have capabilities in protecting and stimulating the repair of injured tissues via paracrine actions, and acting as vectors for biodelivery of exogenous factors to treat injury and diseases. Much progress has been made since the discovery of AM and AF cells with stem cell characteristics nearly a decade ago, but there remain a number of problematic issues stemming from the inherent heterogeneity of these cells as well as inconsistencies in isolation and culturing methods which must be addressed to advance the field towards the development of cell-based therapies. Here, we provide an overview of the recent progress and future perspectives in the use of AM- and AF-derived cells for therapeutic applications.

20.
Hum Mol Genet ; 21(1): 32-45, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21949351

RESUMEN

Many human diseases share a developmental origin that manifests during childhood or maturity. Aneuploid syndromes are caused by supernumerary or reduced number of chromosomes and represent an extreme example of developmental disease, as they have devastating consequences before and after birth. Investigating how alterations in gene dosage drive these conditions is relevant because it might help treat some clinical aspects. It may also provide explanations as to how quantitative differences in gene expression determine phenotypic diversity and disease susceptibility among natural populations. Here, we aimed to produce induced pluripotent stem cell (iPSC) lines that can be used to improve our understanding of aneuploid syndromes. We have generated iPSCs from monosomy X [Turner syndrome (TS)], trisomy 8 (Warkany syndrome 2), trisomy 13 (Patau syndrome) and partial trisomy 11;22 (Emanuel syndrome), using either skin fibroblasts from affected individuals or amniocytes from antenatal diagnostic tests. These cell lines stably maintain the karyotype of the donors and behave like embryonic stem cells in all tested assays. TS iPSCs were used for further studies including global gene expression analysis and tissue-specific directed differentiation. Multiple clones displayed lower levels of the pseudoautosomal genes ASMTL and PPP2R3B than the controls. Moreover, they could be transformed into neural-like, hepatocyte-like and heart-like cells, but displayed insufficient up-regulation of the pseudoautosomal placental gene CSF2RA during embryoid body formation. These data support that abnormal organogenesis and early lethality in TS are not caused by a tissue-specific differentiation blockade, but rather involves other abnormalities including impaired placentation.


Asunto(s)
Aneuploidia , Trastornos de los Cromosomas/genética , Células Madre Pluripotentes Inducidas/citología , Diferenciación Celular , Células Cultivadas , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/fisiopatología , Femenino , Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Lactante , Masculino , Modelos Genéticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...