Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 25(2): 427-442, 2017 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-28153093

RESUMO

Restoring pluripotency using chemical compounds alone would be a major step forward in developing clinical-grade pluripotent stem cells, but this has not yet been reported in human cells. We previously demonstrated that VPA_AFS cells, human amniocytes cultivated with valproic acid (VPA) acquired functional pluripotency while remaining distinct from human embryonic stem cells (hESCs), questioning the relationship between the modulation of cell fate and molecular regulation of the pluripotency network. Here, we used single-cell analysis and functional assays to reveal that VPA treatment resulted in a homogeneous population of self-renewing non-transformed cells that fulfill the hallmarks of pluripotency, i.e., a short G1 phase, a dependence on glycolytic metabolism, expression of epigenetic modifications on histones 3 and 4, and reactivation of endogenous OCT4 and downstream targets at a lower level than that observed in hESCs. Mechanistic insights into the process of VPA-induced reprogramming revealed that it was dependent on OCT4 promoter activation, which was achieved independently of the PI3K (phosphatidylinositol 3-kinase)/AKT/mTOR (mammalian target of rapamycin) pathway or GSK3ß inhibition but was concomitant with the presence of acetylated histones H3K9 and H3K56, which promote pluripotency. Our data identify, for the first time, the pluripotent transcriptional and molecular signature and metabolic status of human chemically induced pluripotent stem cells.


Assuntos
Âmnio/citologia , Transdiferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Biomarcadores , Ciclo Celular/genética , Transdiferenciação Celular/genética , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Metabolismo Energético , Epigênese Genética , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Genes Reporter , Glicólise , Histonas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteína Homeobox Nanog/genética , Fator 3 de Transcrição de Octâmero/genética , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes de Fusão , Serina-Treonina Quinases TOR/metabolismo , Ativação Transcricional
2.
Sci Rep ; 6: 39656, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27995994

RESUMO

The impaired maturation of bone-forming osteoblasts results in reduced bone formation and subsequent bone weakening, which leads to a number of conditions such as osteogenesis imperfecta (OI). Transplantation of human fetal mesenchymal stem cells has been proposed as skeletal anabolic therapy to enhance bone formation, but the mechanisms underlying the contribution of the donor cells to bone health are poorly understood and require further elucidation. Here, we show that intraperitoneal injection of human amniotic mesenchymal stem cells (AFSCs) into a mouse model of OI (oim mice) reduced fracture susceptibility, increased bone strength, improved bone quality and micro-architecture, normalised bone remodelling and reduced TNFα and TGFß sigalling. Donor cells engrafted into bones and differentiated into osteoblasts but importantly, also promoted endogenous osteogenesis and the maturation of resident osteoblasts. Together, these findings identify AFSC transplantation as a countermeasure to bone fragility. These data have wider implications for bone health and fracture reduction.


Assuntos
Âmnio/citologia , Fraturas Ósseas/prevenção & controle , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Osteogênese Imperfeita/prevenção & controle , Animais , Remodelação Óssea , Osso e Ossos/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Perfilação da Expressão Gênica , Marcadores Genéticos , Humanos , Masculino , Camundongos , Osteoblastos/metabolismo , Osteogênese , Estresse Mecânico , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Microtomografia por Raio-X
3.
Stem Cells Dev ; 25(5): 395-404, 2016 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-26728561

RESUMO

Alport syndrome (AS) is a hereditary glomerulopathy caused by a mutation in type IV collagen genes, which disrupts glomerular basement membrane, leading to progressive glomerulosclerosis and end-stage renal failure. There is at present no cure for AS, and cell-based therapies offer promise to improve renal function. In this study, we found that human first trimester fetal chorionic stem cells (CSC) are able to migrate to glomeruli and differentiate down the podocyte lineage in vitro and in vivo. When transplanted into 7-week-old Alport 129Sv-Col4α3(tm1Dec)/J (-/-) mice, a single intraperitoneal injection of CSC significantly lowered blood urea and urine proteinuria levels over the ensuing 2 weeks. In addition, nearly two-thirds of transplanted -/- mice maintained their weight above the 80% welfare threshold, with both males and females weighing more than age-matched nontransplanted -/- mice. This was associated with less renal cortical fibrosis and interstitial inflammation compared to nontransplanted mice as shown by reduction in murine CD4, CD68, and CD45.2 cells. Transplanted CSC homed to glomeruli, where they expressed CR1, VEGFA, SYNAPTOPODIN, CD2AP, and PODOCIN at the RNA level and produced PODOCIN, CD2AP, and COLIVα3 proteins in nontransplanted -/- mice, indicating that CSC have adopted a podocyte phenotype. Together, these data indicate that CSC may be used to delay progression of renal pathology by a combination of anti-inflammatory effects and replacement of the defective resident podocytes.


Assuntos
Diferenciação Celular , Córion/citologia , Nefrite Hereditária/terapia , Podócitos/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Córion/transplante , Técnicas de Cocultura , Colágeno Tipo IV/farmacologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Fibrose , Humanos , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Córtex Renal/patologia , Glomérulos Renais/efeitos dos fármacos , Glomérulos Renais/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Mutação/genética , Nefrite Hereditária/patologia , Fenótipo , Podócitos/efeitos dos fármacos , Podócitos/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
4.
Stem Cells Dev ; 23(3): 262-76, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24028330

RESUMO

Osteogenesis imperfecta (OI) is a genetic bone pathology with prenatal onset, characterized by brittle bones in response to abnormal collagen composition. There is presently no cure for OI. We previously showed that human first trimester fetal blood mesenchymal stem cells (MSCs) transplanted into a murine OI model (oim mice) improved the phenotype. However, the clinical use of fetal MSC is constrained by their limited number and low availability. In contrast, human fetal early chorionic stem cells (e-CSC) can be used without ethical restrictions and isolated in high numbers from the placenta during ongoing pregnancy. Here, we show that intraperitoneal injection of e-CSC in oim neonates reduced fractures, increased bone ductility and bone volume (BV), increased the numbers of hypertrophic chondrocytes, and upregulated endogenous genes involved in endochondral and intramembranous ossification. Exogenous cells preferentially homed to long bone epiphyses, expressed osteoblast genes, and produced collagen COL1A2. Together, our data suggest that exogenous cells decrease bone brittleness and BV by directly differentiating to osteoblasts and indirectly stimulating host chondrogenesis and osteogenesis. In conclusion, the placenta is a practical source of stem cells for the treatment of OI.


Assuntos
Córion/citologia , Células-Tronco Fetais/citologia , Células-Tronco Fetais/transplante , Fraturas Ósseas/terapia , Osteogênese Imperfeita/terapia , Placenta/citologia , Animais , Osso e Ossos/anormalidades , Osso e Ossos/metabolismo , Terapia Baseada em Transplante de Células e Tecidos , Condrócitos/citologia , Condrócitos/metabolismo , Condrogênese , Córion/metabolismo , Colágeno Tipo I/agonistas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Feminino , Células-Tronco Fetais/metabolismo , Feto , Fraturas Ósseas/genética , Fraturas Ósseas/metabolismo , Fraturas Ósseas/patologia , Expressão Gênica , Humanos , Injeções Intraperitoneais , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Placenta/metabolismo , Gravidez , Transplante de Células-Tronco , Transplante Heterólogo
5.
Curr Stem Cell Res Ther ; 8(1): 73-81, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23270629

RESUMO

Mid-gestation c-KIT(+) amniotic fluid stem cells (AFSC) have an intermediate phenotype between embryonic and adult stem cells and are easy to reprogram to pluripotency. We previously showed that 1st trimester AFSC can be reprogrammed to functional pluripotency in a transgene-free approach. Despite both parental populations sharing a common phenotype, expressing CD29, CD44, CD73, CD90, CD105, SSEA4 and OCT4, 2nd trimester AFSC, contrary to 1st trimester cells, do not express NANOG, SSEA3, TRA-1-60 and TRA-1-81, and have slower growth kinetics. Here, we used the Illumina Beadstudio microarray platform to analyse the transcriptome of 1st and 2nd trimester AFSC and show a unique 1st trimester AFSC-specific gene expression signature consisting of 366 genes and a larger set of 603 genes common with hESC compared to 496 genes overlapping between 2nd trimester AFSC and hESC. We conclude that both populations are related but distinct to each other as well as to hESC.


Assuntos
Líquido Amniótico/citologia , Biomarcadores/metabolismo , Perfilação da Expressão Gênica , Primeiro Trimestre da Gravidez/genética , Segundo Trimestre da Gravidez/genética , Células-Tronco/citologia , Adulto , Líquido Amniótico/metabolismo , Diferenciação Celular , Células Cultivadas , Feminino , Desenvolvimento Fetal , Citometria de Fluxo , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Gravidez , Primeiro Trimestre da Gravidez/metabolismo , Segundo Trimestre da Gravidez/metabolismo , Células-Tronco/metabolismo
6.
Stem Cells Dev ; 22(3): 444-58, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23050522

RESUMO

Human mid-trimester amniotic fluid stem cells (AFSC) have promising applications in regenerative medicine, being broadly multipotent with an intermediate phenotype between embryonic (ES) and mesenchymal stem cells (MSC). Despite this propluripotent phenotype, AFSC are usually cultured in adherence in a serum-based expansion medium, and how expansion in conditions sustaining pluripotency might affect their phenotype remains unknown. We recently showed that early AFSC from first trimester amniotic fluid, which endogenously express Sox2 and Klf4, can be reprogrammed to pluripotency without viral vectors using the histone deacetylase inhibitor valproic acid (VPA). Here, we show that mid-trimester AFSC cultured under MSC conditions contained a subset of cells endogenously expressing telomerase, CD24, OCT4, C-MYC, and SSEA4, but low/null levels of SOX2, NANOG, KLF4, SSEA3, TRA-1-60, and TRA-1-81, with cells unable to form embryoid bodies (EBs) or teratomas. In contrast, AFSC cultured under human ESC conditions were smaller in size, grew faster, formed colonies, upregulated OCT4 and C-MYC, and expressed KLF4 and SOX2, but not NANOG, SSEA3, TRA-1-60, and TRA-1-81. Supplementation with VPA for 5 days further upregulated OCT4, KLF4, and SOX2, and induced expression of NANOG, SSEA3, TRA-1-60, and TRA-1-81, with cells now able to form EBs and teratomas. We conclude that human mid-trimester AFSC, which may be isolated autologously during pregnancy without ethics restriction, can acquire pluripotent characteristics without the use of ectopic factors. Our data suggest that this medium-dependant approach to pluripotent mid-trimester AFSC reflects true reprogramming and not the selection of prepluripotent cells.


Assuntos
Líquido Amniótico/citologia , Antígenos de Diferenciação/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Pluripotentes/metabolismo , Ácido Valproico/farmacologia , Animais , Antígenos de Diferenciação/genética , Proliferação de Células , Forma Celular , Células Cultivadas , Meios de Cultura , Células-Tronco Embrionárias/metabolismo , Feminino , Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Neoplasias Experimentais/patologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante , Gravidez , Segundo Trimestre da Gravidez , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Telômero/metabolismo , Teratoma/patologia , Regulação para Cima/efeitos dos fármacos
7.
Stem Cells Transl Med ; 1(1): 70-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23197643

RESUMO

Stem cells have considerable potential to repair damaged organs and tissues. We previously showed that prenatal transplantation of human first trimester fetal blood mesenchymal stem cells (hfMSCs) in a mouse model of osteogenesis imperfecta (oim mice) led to a phenotypic improvement, with a marked decrease in fracture rate. Donor cells differentiated into mature osteoblasts, producing bone proteins and minerals, including collagen type Iα2, which is absent in nontransplanted mice. This led to modifications of the bone matrix and subsequent decrease of bone brittleness, indicating that grafted cells directly contribute to improvement of bone mechanical properties. Nevertheless, the therapeutic effect was incomplete, attributing to the limited level of engraftment in bone. In this study, we show that although migration of hfMSCs to bone and bone marrow is CXCR4-SDF1 (SDF1 is stromal-derived factor) dependent, only a small number of cells present CXCR4 on the cell surface despite high levels of internal CXCR4. Priming with SDF1, however, upregulates CXCR4 to increase the CXCR4(+) cell fraction, improving chemotaxis in vitro and enhancing engraftment in vivo at least threefold in both oim and wild-type bone and bone marrow. Higher engraftment in oim bones was associated with decreased bone brittleness. This strategy represents a step to improve the therapeutic benefits of fetal cell therapy toward being curative.


Assuntos
Fraturas Ósseas/prevenção & controle , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Osteoblastos/transplante , Osteogênese Imperfeita/cirurgia , Receptores CXCR4/metabolismo , Animais , Matriz Óssea/metabolismo , Diferenciação Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Quimiotaxia , Colágeno Tipo I/deficiência , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Modelos Animais de Doenças , Feminino , Fraturas Ósseas/metabolismo , Fraturas Ósseas/patologia , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/imunologia , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteogênese Imperfeita/genética , Osteogênese Imperfeita/imunologia , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Fatores de Tempo , Regulação para Cima
8.
PLoS One ; 7(9): e43395, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22962584

RESUMO

Human mesenchymal stromal/stem cells (MSC) isolated from fetal tissues hold promise for use in tissue engineering applications and cell-based therapies, but their collection is restricted ethically and technically. In contrast, the placenta is a potential source of readily-obtainable stem cells throughout pregnancy. In fetal tissues, early gestational stem cells are known to have advantageous characteristics over neonatal and adult stem cells. Accordingly, we investigated whether early fetal placental chorionic stem cells (e-CSC) were physiologically superior to their late gestation fetal chorionic counterparts (l-CSC). We showed that e-CSC shared a common phenotype with l-CSC, differentiating down the osteogenic, adipogenic and neurogenic pathways, and containing a subset of cells endogenously expressing NANOG, SOX2, c-MYC, and KLF4, as well as an array of genes expressed in pluripotent stem cells and primordial germ cells, including CD24, NANOG, SSEA4, SSEA3, TRA-1-60, TRA-1-81, STELLA, FRAGILIS, NANOS3, DAZL and SSEA1. However, we showed that e-CSC have characteristics of an earlier state of stemness compared to l-CSC, such as smaller size, faster kinetics, uniquely expressing OCT4A variant 1 and showing higher levels of expression of NANOG, SOX2, c-MYC and KLF4 than l-CSC. Furthermore e-CSC, but not l-CSC, formed embryoid bodies containing cells from the three germ layer lineages. Finally, we showed that e-CSC demonstrate higher tissue repair in vivo; when transplanted in the osteogenesis imperfecta mice, e-CSC, but not l-CSC increased bone quality and plasticity; and when applied to a skin wound, e-CSC, but not l-CSC, accelerated healing compared to controls. Our results provide insight into the ontogeny of the stemness phenotype during fetal development and suggest that the more primitive characteristics of early compared to late gestation fetal chorionic stem cells may be translationally advantageous.


Assuntos
Córion/citologia , Células-Tronco Fetais/citologia , Células-Tronco Mesenquimais/citologia , Placenta/citologia , Antígenos Embrionários Estágio-Específicos/metabolismo , Adulto , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Córion/metabolismo , Feminino , Células-Tronco Fetais/metabolismo , Células-Tronco Fetais/transplante , Humanos , Fator 4 Semelhante a Kruppel , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Placenta/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , Pele/lesões , Antígenos Embrionários Estágio-Específicos/genética , Transplante de Células-Tronco , Fatores de Tempo , Transplante Heterólogo , Cicatrização
9.
Mol Ther ; 20(10): 1953-67, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22760542

RESUMO

Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling.


Assuntos
Líquido Amniótico/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Ácido Valproico/farmacologia , Líquido Amniótico/citologia , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Genoma Humano , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Cariotipagem , Cinética , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Masculino , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Fenótipo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Análise de Sequência de DNA , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transcriptoma , Transgenes , Inativação do Cromossomo X/efeitos dos fármacos
10.
Epigenetics ; 6(1): 52-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20864803

RESUMO

Human embryonic stem (hES) cells and fetal mesenchymal stem cells (fMSC) offer great potential for regenerative therapy strategies. It is therefore important to characterise the properties of these cells in vitro. One major way the environment impacts on cellular physiology is through changes to epigenetic mechanisms. Genes subject to epigenetic regulation via genomic imprinting have been characterised extensively. The integrity of imprinted gene expression therefore provides a measurable index for epigenetic stability. Allelic expression of 26 imprinted genes and DNA methylation at associated differentially methylated regions (DMRs) was measured in fMSC and hES cell lines. Both cell types exhibited monoallelic expression of 13 imprinted genes, biallelic expression of six imprinted genes, and there were seven genes that differed in allelic expression between cell lines. fMSCs exhibited the differential DNA methylation patterns associated with imprinted expression. This was unexpected given that gene expression of several imprinted genes was biallelic. However, in hES cells, differential methylation was perturbed. These atypical methylation patterns did not correlate with allelic expression. Our results suggest that regardless of stem cell origin, in vitro culture affects the integrity of imprinted gene expression in human cells. We identify biallelic and variably expressed genes that may inform on overall epigenetic stability. As differential methylation did not correlate with imprinted expression changes we propose that other epigenetic effectors are adversely influenced by the in vitro environment. Since DMR integrity was maintained in fMSC but not hES cells, we postulate that specific hES cell derivation and culturing practices result in changes in methylation at DMRs.


Assuntos
Metilação de DNA/fisiologia , Células-Tronco Embrionárias/metabolismo , Impressão Genômica/fisiologia , Células-Tronco Mesenquimais/metabolismo , Animais , Células-Tronco Embrionárias/citologia , Feto , Perfilação da Expressão Gênica/métodos , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Medicina Regenerativa/métodos
11.
J R Soc Interface ; 7 Suppl 6: S689-706, 2010 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-20739312

RESUMO

Foetal stem cells (FSCs) can be isolated during gestation from many different tissues such as blood, liver and bone marrow as well as from a variety of extraembryonic tissues such as amniotic fluid and placenta. Strong evidence suggests that these cells differ on many biological aspects such as growth kinetics, morphology, immunophenotype, differentiation potential and engraftment capacity in vivo. Despite these differences, FSCs appear to be more primitive and have greater multi-potentiality than their adult counterparts. For example, foetal blood haemopoietic stem cells proliferate more rapidly than those found in cord blood or adult bone marrow. These features have led to FSCs being investigated for pre- and post-natal cell therapy and regenerative medicine applications. The cells have been used in pre-clinical studies to treat a wide range of diseases such as skeletal dysplasia, diaphragmatic hernia and respiratory failure, white matter damage, renal pathologies as well as cancers. Their intermediate state between adult and embryonic stem cells also makes them an ideal candidate for reprogramming to the pluripotent status.


Assuntos
Sangue Fetal/citologia , Células-Tronco Fetais/citologia , Feto/citologia , Medicina Regenerativa , Âmnio/citologia , Líquido Amniótico/citologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Feminino , Células-Tronco Fetais/transplante , Humanos , Camundongos , Fenótipo , Placenta/citologia , Gravidez , Terapêutica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...