Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Biomed Mater Res A ; 112(9): 1484-1493, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38487991

RESUMO

Mesenchymal stem cell-derived secretome represents an emerging acellular therapeutic which possess significant opportunity for clinical applications due to its anti-inflammatory, immunomodulatory, and wound healing properties. However, maintaining therapeutic efficacy and ensuring stability of cell-based products is challenging, requiring a robust delivery method. Therefore, we designed a hydrogel-based scaffold loaded with CK Cell Technologies' proprietary Mesenchymal stem cell-secretome for controlled release treatment of acute and chronic wounds. We incorporated both conditioned media (CM) and extracellular vesicles (EVs) into gelatin methacryloyl (GelMA) hydrogels and demonstrated how we can tune the diffusive release of the EVs from them. To demonstrate viability of the approach, we developed a wound healing scratch assay where we see in situ release of CM and EVs promote enhanced migration of human dermal fibroblasts (hDFs). We see the colocalization of these EVs in the fibroblasts using fluorescent microscopy. Finally, as a surrogate for in vivo neovascularization, we conducted an in vitro tube formation assay for the MSC-secretome using matrigel-embedded human microvascular endothelial cells. By adding CM and EVs, we observe an increase in tubulogenesis. Collectively, our data demonstrates by tuning the GelMA properties, we can influence the controlled release of the MSC-secretome for a wound dressing and bandage application for chronic and acute wounds.


Assuntos
Gelatina , Hidrogéis , Células-Tronco Mesenquimais , Cicatrização , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Humanos , Hidrogéis/química , Cicatrização/efeitos dos fármacos , Gelatina/química , Fibroblastos/citologia , Vesículas Extracelulares/metabolismo , Polimerização , Metacrilatos/química , Movimento Celular/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia
2.
Pancreas ; 41(1): 54-64, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22143343

RESUMO

OBJECTIVE: A challenge in using human embryonic stem cells (hESCs) as the source of surrogate ß cells is the establishment of methods that could effectively direct their differentiation into functional ß cells. The aim of this study was to assess the effect of NANOG gene suppression in differentiating hESCs as a mean of increasing the efficiency with which endoderm-derived pancreatic cells could be generated. METHODS: A homogenous cell population with stable suppression of NANOG was generated in hESC ENVY line using plasmid-based siRNA approach. Pancreatic differentiation was undertaken according to the ontology-based in vitro selection protocol and followed by transplantation into immunodeficiency mice to mature in vivo. RESULTS: We observed up-regulation of definitive endoderm genes, which expand the role of NANOG in blocking definitive endoderm differentiation. The ontology-based differentiation protocol resulted in increased expression of markers essential for pancreatic epithelium development. Transplantation of these cells further revealed a homogenous pancreatic exocrine-like morphology that stained positively for amylase. CONCLUSIONS: The suppression of NANOG displayed an effective differentiation toward endoderm and pancreatic progenitors. Investigation of the factors required for endocrine formation combined with a prolonged in vivo culturing could be further used to increase the ratio of endocrine-exocrine cells fate.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Endoderma/citologia , Proteínas de Homeodomínio/metabolismo , Pâncreas/citologia , Amilases/metabolismo , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia de Contraste de Fase , Proteína Homeobox Nanog , Pâncreas/metabolismo , Pâncreas Exócrino/citologia , Pâncreas Exócrino/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Células-Tronco/citologia , Células-Tronco/metabolismo
3.
Expert Opin Biol Ther ; 11(5): 569-79, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21314494

RESUMO

INTRODUCTION: The advent of induced pluripotent stem cell (iPSC) technology has opened up new vistas to generate patient-specific pluripotent stem cells from somatic cells. During the last 5 years, the iPSCs produced from a variety of somatic cell sources are found to be very similar, if not identical to embryonic stem cells. Invariably these cells are produced by viral transduction of four transcriptional factors that renders these cells unfit for therapeutic purposes. AREAS COVERED: This review discusses current developments emphasising on new and improved methods of generating iPSCs, including minimal or no genetic modifications via excisable lentiviral and transposon vectors or through repeated application of transient plasmid, episomal and adenovirus vectors. Recent use of small molecules, synthetic mRNA and microRNAs is also reviewed. EXPERT OPINION: iPSC technology is emerging as an unprecedented opportunity in biomedical research, disease modeling, drug discovery and regenerative medicine. However, to harness the full potential of this technology, a number of issues that need to be resolved pertaining to iPSC safety, stability, culture variability, their comparison with ES cells, the reprogramming mechanisms and better ways to direct a specific reprogramming process including lineage specifications.


Assuntos
Células-Tronco Pluripotentes/citologia , Animais , Diferenciação Celular , Vetores Genéticos , Humanos , Camundongos , Modelos Biológicos
4.
PLoS One ; 5(8): e12297, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20808872

RESUMO

Reprogramming of somatic cells to different extents has been reported using different methods. However, this is normally accompanied by the use of exogenous materials, and the overall reprogramming efficiency has been low. Chemicals and small molecules have been used to improve the reprogramming process during somatic cell nuclear transfer (SCNT) and induced pluripotent stem (iPS) cell generation. We report here the first application of a combined epigenetic and non-genetic approach for reprogramming somatic cells, i.e., DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors, and human embryonic stem cell (hESC) extracts. When somatic cells were pretreated with these inhibitors before exposure to hESC (MEL1) extracts, morphological analysis revealed a higher rate of hESC-like colony formation than without pretreatment. Quantitative PCR (qPCR) demonstrated that pluripotency genes were upregulated when compared to those of somatic cells or treated with hESC extracts alone. Overall changes in methylation and acetylation levels of pretreated somatic cells suggests that epigenetic states of the cells have an effect on reprogramming efficiency induced by hESC extracts. KnockOutserum replacement (KOSR) medium (KO-SR) played a positive role in inducing expression of the pluripotency genes. hESC extracts could be an alternative approach to reprogram somatic cells without introducing exogenous materials. The epigenetic pre-treatment of somatic cells could be used to improve the efficiency of reprogramming process. Under differentiation conditions, the reprogrammed cells exhibited differentiation ability into neurons suggesting that, although fully reprogramming was not achieved, the cells could be transdifferentiated after reprogramming.


Assuntos
Reprogramação Celular/genética , Epigênese Genética , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , Metilação de DNA/efeitos dos fármacos , Dopamina/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Epigênese Genética/efeitos dos fármacos , Fibroblastos/citologia , Perfilação da Expressão Gênica , Inibidores de Histona Desacetilases/farmacologia , Proteínas de Homeodomínio/genética , Humanos , Proteína Homeobox Nanog , Neurônios/citologia , Neurônios/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas/genética , Transdução de Sinais/efeitos dos fármacos , Transdução Genética , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
5.
Reprod Biomed Online ; 19(1): 99-105, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19573297

RESUMO

Mesenchymal stem cells derived from bone marrow are a well characterized population of adult stem cells that can be maintained and propagated in culture for a long time with the capacity to form a variety of cell types. Reports have shown that murine and human embryonic stem cells can differentiate into primordial germ cells and then to early gametes. Evidence has indicated that some adult stem cells also have the potential to differentiate into germ cells. Currently, there are no reports on directed differentiation of human mesenchymal stem cells into germ cells. This study investigated the ability of retinoic acid and testicular extracts to induce human bone marrow stem cells (hBMSC) to differentiate into male germ cells. It was found that a small population of hBMSC seem to transdifferentiate into male germ cell-like cells. These cells expressed early germ cell markers OCT4, STELLA, NANOG and VASA, and male germ-ceil-specific markers such as DAZL, TH2, c-kit, beta(1)-integrin, ACR, PRMl, FSHR, STRA8 and SCP3, as analysed by reverse transcription-polymerase chain reaction and immunohistochemistry. These results demonstrated that hBMSC may differentiate into male germ cells and the same could be used as a potential source of cells for reproductive toxicological studies.


Assuntos
Linhagem da Célula , Feto/citologia , Células-Tronco Hematopoéticas/citologia , Espermatozoides/citologia , Sequência de Bases , Biomarcadores , Diferenciação Celular , Primers do DNA , Imunofluorescência , Humanos , Masculino , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
Stem Cells Dev ; 17(6): 1087-94, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18513165

RESUMO

Recent work indicates that neural progenitors can be isolated from the skin of rodents and humans. The persistence of these cells in accessible adult tissue raises the possibility of their exploitation for research and therapeutic purposes. This study reports on the derivation, culture, and characterization of homogenous canine skin-derived neuroprecursor cells (SKiNPs) from mature animals. Canine tissue was used because naturalistic brain diseases in community-dwelling dogs are emerging as ecologically sound models for a range of neurological conditions. Adult SKiNPs were initially isolated as neurospheres and then cultured for 10-15 passages in an adherent monolayer assay. Serumfree expansion conditions contained B-27, 20 ng/mL EGF, and 40 ng/mL bFGF. Gene expressions by PCR indicated expression of nestin, CD133, NCAM, and FGF2R, but not GFAP. Highly uniform expression of nestin (76 +/- 8.3%), NCAM (84 +/- 3.3%), betaIII-tubulin (96 +/- 4.3%), and CD133 (68 +/- 13.5%) was also observed. Directed differentiation of SKiNPs in the presence of serum induced betaIIItubulin, NSE, NCAM, and MAP2 in >90% of differentiated cells by immunophenotype analysis. Our culture system rapidly induces canine skin cells into neural precursors, maintains nestin expression in more than 75% of proliferating cells, and generates an almost universal neuronal-like phenotype after 7 days of in vitro differentiation. Their biological characteristics are suggestive of transiently amplifying fate-restricted neuroprecursors rather than true neural stem cells. This system may be an effective alternative for autologous neurorestorative cell replacement in canine models for further translational research.


Assuntos
Células-Tronco Adultas/citologia , Diferenciação Celular , Neurônios/citologia , Transplante de Células-Tronco , Células-Tronco Adultas/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Encefalopatias/terapia , Células Cultivadas , Cães , Humanos , Neurônios/metabolismo , Pele , Transplante Autólogo
7.
Stem Cells Dev ; 17(1): 41-51, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18271699

RESUMO

Here we describe the derivation of a novel human embryonic stem (hES) cell line, Endeavour-1 (E1), its four new clonal lines (E1C1, E1C2, E1C3, E1C4), and their characterization. E1 and its clonal lines are propagated on human fetal fibroblasts (HFFs) derived and grown in a largely serum-free medium. Seven inner cell masses were isolated from 34 donated human embryos (27 survived), and one new hES cell line was obtained. E1 has been in culture for over 1 year and possesses all the typical features of stem cells, i.e., expression of stem cell surface markers (stage-specific embryonic antigens SSEA-3 and SSEA-4, and tumor recognition antigens TRA-1-60 and TRA-1-81), staining for alkaline phosphatase, and the presence of the pluripotent gene marker (nanog). This line shows pluripotency both under in vitro and in vivo conditions. E1 has a normal karyotype (46XX). Using our optimized procedure for cloning, four new clonal lines were derived from E1: E1C1, E1C2, E1C3, and E1C4. These clonal lines show normal characteristics: karyotype of that of the parent line (46XX) except for E1C3, which showed reciprocal translocation involving chromosomes 15 and 17; stem cell surface markers SSEA-4, TRA-1-60, and TRA-1-81; and gene expression for pluripotency (Nanog). All of these clonal lines formed embryoid bodies (EBs) in suspension cultures. After seeding, the EBs differentiated, forming cell lineages derived from all three germ layers as indicated by immunolocalization for the ectodermal marker beta-III tubulin, the mesodermal marker CD34, and the endodermal marker alpha-fetoprotein (AFP). There were subtle differences in the expression of these markers between clones. These clonal lines showed pluripotency in vivo. E1 and its clonal lines can differentiate to definitive endoderm after treatment with activin A, and, as indicated by expression of SOX17, FOXa2, and GATA-4 by RT-PCR, there are some subtle differences between these clonal lines. This may help in selecting clonal lines for specific lineage specification and for developing future cell therapy for various diseases.


Assuntos
Linhagem Celular , Células-Tronco Embrionárias/citologia , Biomarcadores/análise , Proliferação de Células , Células Clonais , Humanos , Técnicas de Cultura de Tecidos
8.
Curr Stem Cell Res Ther ; 3(1): 66-74, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18220925

RESUMO

Recently considerable interests have been roused in nuclear reprogramming by somatic cell nuclear transfer using an egg cytoplasm and/or by other means, such as fusion, cell extracts treatment and genes transfections. However, the very mechanism of reprogramming still remains elusive. Epigenetic modifications, which play a significant role in normal mammalian development in vivo is also involved in the process of reprogramming in vitro. The latter shares some of the other features observed in nuclear reprogramming in vivo. In this review, we discuss the main epigenetic changes involved in nuclear reprogramming and currently available approaches to achieve nuclear reprogramming in vitro and its future prospects.


Assuntos
Reprogramação Celular/fisiologia , Técnicas de Transferência Nuclear , Células-Tronco Pluripotentes/fisiologia , Animais , Fusão Celular/métodos , Clonagem de Organismos , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Epigênese Genética , Humanos , Modelos Biológicos
9.
Stem Cells Cloning ; 1: 11-21, 2008 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-24198501

RESUMO

Epigenetics is a growing field not only in the area of cancer research but recently in stem cells including human embryonic stem cell (hESC) research. The hallmark of profiling epigenetic changes in stem cells lies in maintaining pluripotency or multipotency and in attaining lineage specifications that are relevant for regenerative medicine. Epigenetic modifications including DNA methylation, histone acetylation and methylation, play important roles in regulating gene expressions. Other epigenetic modifications include X chromosome silencing, genomic stability and imprinting and mammalian development. This review attempts to elucidate the mechanism(s) behind epigenetic modifications and review techniques scientists use for identifying each modification. We also discuss some of the trends of epigenetic modifications in the fields of directed differentiation of embryonic stem cells and de-differentiation of somatic cells.

10.
Curr Neurovasc Res ; 3(4): 281-8, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17109623

RESUMO

Human embryonic stem cells (hESC) demonstrate a remarkable proliferative and developmental potential and thus have huge therapeutic potential. To direct the differentiation of hESC to a specific lineage of high purity for cell transplantation is highly desirable. Here we describe a modified in vitro procedure to direct differentiation of three clonal hESC lines, hES 3.1, hES 3.2 and hES 3.3 efficiently to spinal motor neurons by using various differentiation factors namely retinoic acid (RA), sonic hedgehog (Shh), bone morphogenetic protein-2 (BMP-2) and Wnt3A. The highest number of motor neurons (58.0 +/- 7.6%) were obtained by an early treatment of embryoid bodies with a combination of RA + Shh from all the clonal hESC lines combined. The hES 3.1 line, however, produced relatively more motor neurons (69.5 +/- 11.8%) compared to other two hES clones, 3.2 (52.4 +/- 13.1%) and 3.3 (52.3 +/- 15.5%). Immunolocalisation studies revealed the expression of neuronal specific marker, beta omega-tubulin and motor neuron specific marker, HB9/HLXB9 in all the three hESC clones after 45 days of differentiation. The RT-PCR analyses showed the presence of the neuron-specific genes. This modified differentiation protocol provides a mean of obtaining an enriched population of motor neurons from hESC for possible use in studies of lineage development, drug discovery and also as a potential cell therapy for motor neuron disease.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/metabolismo , Neurônios Motores/metabolismo , Fatores de Crescimento Neural/farmacologia , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/farmacologia , Contagem de Células , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células Clonais/efeitos dos fármacos , Células Clonais/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Hedgehog/farmacologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imuno-Histoquímica , Neurônios Motores/citologia , Neurônios Motores/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Tretinoína/farmacologia , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Proteínas Wnt/farmacologia , Proteína Wnt3 , Proteína Wnt3A
11.
Transplantation ; 82(9): 1175-84, 2006 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17102769

RESUMO

BACKGROUND: Embryonic stem cells (ESC) when transplanted into recipients with different major histocompatibility antigens may be rejected, especially as cells differentiate and expression of these antigens increases. One method to prevent rejection is to place the developing ESC in microcapsules. It is currently unknown what effect encapsulation has on the ability of ESC to differentiate. METHODS: Human ESC (hESC; hES03 line) and mouse ESC (mESC; R1 line) were encapsulated in 2.2% barium alginate and transplanted intraperitoneally in SCID and BALB/c mice respectively. Cell morphology, viability, and gene characterization were assessed after retrieving the capsules up to four weeks from SCID mice and three months from BALB/c mice. RESULTS: Encapsulation prevented hESC and mESC from forming teratomas up to four weeks and three months, respectively. mESC but not hESC formed aggregates within the capsules, which remained free of fibrosis. Some but not all the transplanted encapsulated hESC differentiated towards all three lineages, but more so towards an endodermal lineage as shown by increased expression of alpha fetoprotein. This was similar to what occurred when encapsulated and non-encapsulated hESC were cultured in vitro for two weeks. In contrast to the hESC, transplanted encapsulated mESC differentiated mostly towards an ectodermal lineage as shown by increased expression of nestin and glial fibrillary acidic protein. In vitro, encapsulated and nonencapsulated mESC also began to differentiate, but not down any specific lineage. CONCLUSIONS: Encapsulated ESC do differentiate, although along multiple pathways, both when transplanted and maintained in culture, just as nonencapsulated ESC do when removed from their feeder layer.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Transplante de Células-Tronco/métodos , Alginatos/administração & dosagem , Alginatos/química , Animais , Materiais Biocompatíveis/administração & dosagem , Materiais Biocompatíveis/química , Cápsulas/administração & dosagem , Cápsulas/química , Diferenciação Celular/genética , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Expressão Gênica , Perfilação da Expressão Gênica , Ácido Glucurônico/administração & dosagem , Ácido Glucurônico/química , Ácidos Hexurônicos/administração & dosagem , Ácidos Hexurônicos/química , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Stem Cells Dev ; 15(1): 61-9, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16522163

RESUMO

Here we describe the first report of three human embryonic stem cell (hESC) clones, hES 3.1, 3.2, and 3.3, derived from the parent line hES3 by sorting of single-cell preparations by flow cytometry. The viability of single-cell preparations before and after cell sorting remained >98%. The hESC were selected by size gating and forward-angle light scatter and were dispersed directly as single cell/ well into 96-well plates containing human fetal fibroblasts as feeder layers. Single stem cell dispersion into 96-well plates was confirmed by using cells from a hES3 line that constitutively expressed green fluorescence protein (eGFP) under similar conditions of flow cytometry. Three clones were obtained from the parent line hES3 -- hES3.1, 3.2, and 3.3 -- and they have been in continuous culture for more than 1 year. The cloning efficiency was less than <0.5%. These hESC clones show normal stem cell characteristics, such as undifferentiated growth, high nucleocytoplasmic ratio, the same karyotype as that of the parent line (46 XX), stem cell surface markers (i.e., SSEA3, SSEA4, OCT4, TRA-1-60, and TRA-1-81), and gene expression for pluripotency (Oct-4 and nanog). They all formed embryoid bodies in suspension cultures, and after seeding in culture plates they showed pluripotency in vitro by forming cell lineages derived from all three germ layers as indicated by expression of the ectodermal marker nestin, the mesodermal marker renin, and the endodermal markers alpha-fetoprotein and GATA6. All clones showed normal expression of alkaline phosphatase activity, a marker of in vitro pluripotency. When hESC clones (1-2 x 10(6) total) were injected into nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice under the kidney capsule, all formed teratomas within 6-8 weeks. Analysis of the stem cell surface marker TRA-1-160 by flow cytometry showed nonsignificant (p < 0.05) differences between the clones and the parent line. The clones also differed in their expression of genes, with only one, hES 3.2, expressing the endodermal markers, i.e., alpha-fetoprotein and GATA6. The ability to produce clones from a parent hESC line rapidly by FACS sorting will help provide a homogeneous population of cells for achieving uniformed lineage specifications for future transplantation therapies and biomedical research.


Assuntos
Embrião de Mamíferos/citologia , Citometria de Fluxo/métodos , Rim/patologia , Transplante de Células-Tronco , Células-Tronco/fisiologia , Teratoma/patologia , Animais , Biomarcadores/análise , Técnicas de Cultura de Células , Diferenciação Celular , Linhagem Celular , Separação Celular , Células Clonais/química , Células Clonais/fisiologia , Expressão Gênica , Humanos , Cariotipagem , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Curr Stem Cell Res Ther ; 1(3): 425-36, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18220886

RESUMO

Transplantation of insulin-producing cells offers a promising therapy to treat diabetes. However, due to the limited number of donor islet cells available, researchers are looking for different sources of pancreatic islet progenitor or stem cells. A stem cell with extensive proliferative ability may provide a valuable source of islet progenitor cells. Several studies have demonstrated that a progenitor/stem-cell population can be expanded in vitro to generate large numbers of islet progenitor cells. However, efficient and directed differentiation of these cells to an endocrine pancreatic lineage has been difficult to achieve. We discuss here various pancreatic islet stem cells that we and others have obtained from embryonic, fetal or adult human tissues. We review the progress that has been achieved with pancreatic islet progenitor cell differentiation in the last 2 decades and discuss how close we are to translate this research to the clinics.


Assuntos
Diabetes Mellitus/terapia , Células-Tronco Embrionárias/transplante , Transplante das Ilhotas Pancreáticas/métodos , Pâncreas/citologia , Pâncreas/fisiologia , Transplante de Células-Tronco/tendências , Adulto , Diabetes Mellitus/cirurgia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA