Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Stem Cells ; 39(6): 673-696, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33586253

RESUMO

Hearing loss (HL) is a major global health problem of pandemic proportions. The most common type of HL is sensorineural hearing loss (SNHL) which typically occurs when cells within the inner ear are damaged. Human induced pluripotent stem cells (hiPSCs) can be generated from any individual including those who suffer from different types of HL. The development of new differentiation protocols to obtain cells of the inner ear including hair cells (HCs) and spiral ganglion neurons (SGNs) promises to expedite cell-based therapy and screening of potential pharmacologic and genetic therapies using human models. Considering age-related, acoustic, ototoxic, and genetic insults which are the most frequent causes of irreversible damage of HCs and SGNs, new methods of genome editing (GE), especially the CRISPR/Cas9 technology, could bring additional opportunities to understand the pathogenesis of human SNHL and identify novel therapies. However, important challenges associated with both hiPSCs and GE need to be overcome before scientific discoveries are correctly translated to effective and patient-safe applications. The purpose of the present review is (a) to summarize the findings from published reports utilizing hiPSCs for studies of SNHL, hence complementing recent reviews focused on animal studies, and (b) to outline promising future directions for deciphering SNHL using disruptive molecular and genomic technologies.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Edição de Genes , Perda Auditiva Neurossensorial/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Terapia Genética/métodos , Perda Auditiva Neurossensorial/genética , Humanos
2.
Stem Cells ; 38(10): 1321-1325, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32614127

RESUMO

Increased pollution by plastics has become a serious global environmental problem, but the concerns for human health have been raised after reported presence of microplastics (MPs) and nanoplastics (NPs) in food and beverages. Unfortunately, few studies have investigate the potentially harmful effects of MPs/NPs on early human development and human health. Therefore, we used a new platform to study possible effects of polystyrene NPs (PSNPs) on the transcription profile of preimplantation human embryos and human induced pluripotent stem cells (hiPSCs). Two pluripotency genes, LEFTY1 and LEFTY2, which encode secreted ligands of the transforming growth factor-beta, were downregulated, while CA4 and OCLM, which are related to eye development, were upregulated in both samples. The gene set enrichment analysis showed that the development of atrioventricular heart valves and the dysfunction of cellular components, including extracellular matrix, were significantly affected after exposure of hiPSCs to PSNPs. Finally, using the HiPathia method, which uncovers disease mechanisms and predicts clinical outcomes, we determined the APOC3 circuit, which is responsible for increased risk for ischemic cardiovascular disease. These results clearly demonstrate that better understanding of NPs bioactivities and its implications for human health is of extreme importance. Thus, the presented platform opens further aspects to study interactions between different environmental and intracellular pollutions with the aim to decipher the mechanism and origin of human diseases.


Assuntos
Poluição Ambiental/análise , Nanopartículas/química , Plásticos/análise , Poliestirenos/química , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Espaço Intracelular , Transcriptoma/genética , Resultado do Tratamento
3.
Environ Pollut ; 320: 120849, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36509347

RESUMO

Plastic pollution is increasing at an alarming rate yet the impact of this pollution on human health is poorly understood. Because human induced pluripotent stem cells (hiPSC) are frequently derived from dermal fibroblasts, these cells offer a powerful platform for the identification of molecular biomarkers of environmental pollution in human cells. Here, we describe a novel proof-of-concept for deriving hiPSC from human dermal fibroblasts deliberately exposed to polystyrene (PS) nanoplastic particles; unexposed hiPSC served as controls. In parallel, unexposed hiPSC were exposed to low and high concentrations of PS nanoparticles. Transcriptomic and epigenomic signatures of all fibroblasts and hiPSCs were defined using RNA-seq and whole genome methyl-seq, respectively. Both PS-treated fibroblasts and derived hiPSC showed alterations in expression of ESRRB and HNF1A genes and circuits involved in the pluripotency of stem cells, as well as in pathways involved in cancer, inflammatory disorders, gluconeogenesis, carbohydrate metabolism, innate immunity, and dopaminergic synapse. Similarly, the expression levels of identified key transcriptional and DNA methylation changes (DNMT3A, ESSRB, FAM133CP, HNF1A, SEPTIN7P8, and TTC34) were significantly affected in both PS-exposed fibroblasts and hiPSC. This study illustrates the power of human cellular models of environmental pollution to narrow down and prioritize the list of candidate molecular biomarkers of environmental pollution. This knowledge will facilitate the deciphering of the origins of environmental diseases.


Assuntos
Células-Tronco Pluripotentes Induzidas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Poliestirenos/metabolismo , Microplásticos/metabolismo , Transcriptoma , Diferenciação Celular/genética , Epigenômica , Fibroblastos , Biomarcadores/metabolismo
4.
Environ Pollut ; 305: 119206, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35405220

RESUMO

Commercially manufactured or generated through environmental degradation, microplastics (MPs) and nanoplastics (NPs) considerably contribute to environmental pollution. There is a knowledge gap in how exposure to MPs/NPs changes cellular function and affects animal and human health. Here, we demonstrate that after oral uptake, fluorescent polystyrene (PS) nanoparticles pass through the mouse digestive system, accumulate and aggregate in different organs, and induce functional changes in cells and organs. Using cochlear explant as a novel in vitro system, we confirmed the consequences of PS-MP/NP interaction with inner ear cells by detecting aggregates and hetero-aggregates of PS particles in hair cells. The testes of treated males accumulated MPs/NPs in the interstitial compartment surrounding the seminiferous tubules, which was associated with a statistically significant decrease in testosterone levels. Male mice showed increased secretion of interleukins (IL-12p35 and IL-23) by splenocytes while cyto- and genotoxicity tests indicated impaired cell viability and increased DNA damage in spleen tissue. Males also showed a broad range of anxiogenic responses to PS nanoparticles while hippocampal samples from treated females showed an increased expression of Bax and Nlrp3 genes, indicating a pro-apoptotic/proinflammatory effect of PS treatment. Taken together, induced PS effects are also gender-dependent, and therefore, strongly motivate future research to mitigate the deleterious effects of nanosized plastic particles.


Assuntos
Nanopartículas , Poluentes Químicos da Água , Animais , Sobrevivência Celular , Corantes , Feminino , Masculino , Camundongos , Microplásticos , Nanopartículas/toxicidade , Plásticos , Poliestirenos/toxicidade
5.
Br Med Bull ; 99: 155-68, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21669982

RESUMO

INTRODUCTION: Stem cells are cells with the ability to grow and differentiate into more than 200 cell types. SOURCES OF DATA: We review here the characteristics and potential of human embryonic stem cells (hESCs), induced pluripotent stem cells (iPSCs) and adult stem cells (ASCs). AREAS OF AGREEMENT: The differentiation ability of all stem cell types could be stimulated to obtain specialized cells that represent renewable sources of functional cells useful for cell-based therapy. AREAS OF CONTROVERSY: The proof of functional differentiated cells needs to be investigated in more detail using both in vitro and in vivo assays including animal disease models and clinical studies. GROWING POINTS: Much progress has been made in the ASCs-based therapies. Meanwhile hESCs and iPSCs have dramatically emerged as novel approaches to understand pathogenesis of different diseases. AREAS TIMELY FOR DEVELOPING RESEARCH: A number of new strategies become very important in regenerative medicine. However, we discuss the limitations of stem cells and latest development in the reprogramming research.


Assuntos
Células-Tronco Adultas , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Células-Tronco Pluripotentes Induzidas , Medicina Regenerativa , Pesquisa com Células-Tronco , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Previsões , Humanos , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Pluripotentes Induzidas/transplante , Regeneração/fisiologia , Medicina Regenerativa/métodos , Medicina Regenerativa/tendências , Projetos de Pesquisa , Transplante de Células-Tronco/métodos , Transplante de Células-Tronco/tendências
6.
Environ Pollut ; 269: 116144, 2021 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-33288300

RESUMO

Increase in plastic pollution causes irreparable harm to the environment lasting for decades. While current data of plastic pollution include marine and terrestrial ecology, the impacts of degraded or intentionally produced microscopic-sized plastics on human health remain unknown. Here, we are proposing the usage of pluripotent stem cells, modern transcriptomics, and bioinformatics as a unique scientific tool to define the link between environmental and intracellular pollution, its outcome on early human development and origin of diseases. This commentary is an urgent appeal to the scientific and policy communities to invest more time and resources to establish reliable standards and methods to define and address the consequences of plastic pollution on human health.


Assuntos
Plásticos , Células-Tronco Pluripotentes , Monitoramento Ambiental , Poluição Ambiental , Humanos
7.
Hum Reprod ; 25(9): 2387-95, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20634187

RESUMO

BACKGROUND: It has long been appreciated that environmental cues may trigger adaptive responses. Many of these responses are a result of changes in the epigenetic landscape influencing transcriptional states and consequently altering phenotypes. In the context of human reproductive health, the procedures necessary for assisted reproduction may result in altered phenotypes by primarily influencing DNA methylation. Among the well-documented effects of assisted reproduction technologies (ART), imprinted genes appear to be frequently altered, likely owing to their reliance on DNA methylation to impose parent-specific monoallelic expression. However, the generality of the potential deregulation of DNA methylation in ART-derived human embryos has not been evaluated. METHODS: In this study, we evaluate the genome-wide DNA methylation together with chromatin organisation in human embryos derived by either IVF (n = 89) or ICSI (n = 76). DNA methylation was assessed using an antibody against 5-methyl-cytidine, and chromatin organisation by DNA staining. RESULTS: Irrespective of the ART procedure, similar errors were observed in both groups and abnormal chromatin was positively correlated (P < 0.001) with inappropriate DNA methylation. Development up to the blastocyst stage was consistent with normal DNA methylation and chromatin organisation, reinforcing the importance of epigenetic regulation to form the early lineages of the blastocyst. Most importantly, we found no evidence that ICSI blastocysts were more severely affected than those derived by IVF. CONCLUSIONS: We conclude that ICSI does not lead to an increased incidence of epigenetic errors (DNA methylation and chromatin) compared with IVF.


Assuntos
Metilação de DNA , Embrião de Mamíferos/química , Epigênese Genética , Fertilização in vitro/efeitos adversos , Injeções de Esperma Intracitoplásmicas/efeitos adversos , Biomarcadores , Blastocisto/química , Blastocisto/patologia , Montagem e Desmontagem da Cromatina , Aberrações Cromossômicas/embriologia , Aberrações Cromossômicas/estatística & dados numéricos , Embrião de Mamíferos/patologia , Desenvolvimento Embrionário , Feminino , Humanos
8.
Stem Cells ; 25(12): 3045-57, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17872502

RESUMO

Human embryonic stem cells (hESC) promise tremendous potential as a developmental and cell therapeutic tool. The combined effort of stimulatory and inhibitory signals regulating gene expression, which drives the tissue differentiation and morphogenetic processes during early embryogenesis, is still very poorly understood. With the scarcity of availability of human embryos for research, hESC can be used as an alternative source to study the early human embryogenesis. Hyaluronan (HA), a simple hydrating sugar, is present abundantly in the female reproductive tract during fertilization, embryo growth, and implantation and plays an important role in early development of the mammalian embryo. HA and its binding protein RHAMM regulate various cellular and hydrodynamic processes from cell migration, proliferation, and signaling to regulation of gene expression, cell differentiation, morphogenesis, and metastasis via both extracellular and intracellular pathways. In this study, we show for the first time that HA synthase gene HAS2 and its binding receptor RHAMM are differentially expressed during all stages of preimplantation human embryos and hESC. RHAMM expression is significantly downregulated during differentiation of hESC, in contrast to HAS2, which is significantly upregulated. Most importantly, RHAMM knockdown results in downregulation of several pluripotency markers in hESC, induction of early extraembryonic lineages, loss of cell viability, and changes in hESC cycle. These data therefore highlight an important role for RHAMM in maintenance of hESC pluripotency, viability, and cell cycle control. Interestingly, HAS2 knockdown results in suppression of hESC differentiation without affecting hESC pluripotency. This suggests an intrinsic role for HAS2 in hESC differentiation process. In accordance with this, addition of exogenous HA to the differentiation medium enhances hESC differentiation to mesodermal and cardiac lineages. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Blastocisto/metabolismo , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/fisiologia , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Glucuronosiltransferase/genética , Receptores de Hialuronatos/genética , Ácido Hialurônico/fisiologia , Blastocisto/citologia , Blastocisto/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas da Matriz Extracelular/fisiologia , Glucuronosiltransferase/fisiologia , Humanos , Receptores de Hialuronatos/fisiologia , Hialuronan Sintases , Ácido Hialurônico/biossíntese , Ácido Hialurônico/genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/fisiologia
9.
Stem Cells Transl Med ; 6(4): 1217-1226, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28213969

RESUMO

Neural differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs) can produce a valuable and robust source of human neural cell subtypes, holding great promise for the study of neurogenesis and development, and for treating neurological diseases. However, current hESCs and hiPSCs neural differentiation protocols require either animal factors or embryoid body formation, which decreases efficiency and yield, and strongly limits medical applications. Here we develop a simple, animal-free protocol for neural conversion of both hESCs and hiPSCs in adherent culture conditions. A simple medium formula including insulin induces the direct conversion of >98% of hESCs and hiPSCs into expandable, transplantable, and functional neural progenitors with neural rosette characteristics. Further differentiation of neural progenitors into dopaminergic and spinal motoneurons as well as astrocytes and oligodendrocytes indicates that these neural progenitors retain responsiveness to instructive cues revealing the robust applicability of the protocol in the treatment of different neurodegenerative diseases. The fact that this protocol includes animal-free medium and human extracellular matrix components avoiding embryoid bodies makes this protocol suitable for the use in clinic. Stem Cells Translational Medicine 2017;6:1217-1226.


Assuntos
Células-Tronco Pluripotentes/citologia , Diferenciação Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia
10.
Cell Transplant ; 22(8): 1309-23, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23043847

RESUMO

Stem cells (SCs) represent a new therapeutic approach for spinal cord injury (SCI) by enabling improved sensory and motor functions in animal models. The main goal of SC-based therapy for SCI is the replacement of neurons and glial cells that undergo cell death soon after injury. Stem cells are able to promote remyelination via oligodendroglia cell replacement to produce trophic factors enhancing neurite outgrowth, axonal elongation, and fiber density and to activate resident or transplanted progenitor cells across the lesion cavity. While several SC transplantation strategies have shown promising yet partial efficacy, mechanistic proof is generally lacking and is arguably the largest impediment toward faster progress and clinical application. The main challenge ahead is to spur on cooperation between clinicians, researchers, and patients in order to define and optimize the mechanisms of SC function and to establish the ideal source/s of SCs that produce efficient and also safe therapeutic approaches.


Assuntos
Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Humanos , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia
11.
PLoS One ; 3(5): e2122, 2008 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-18461168

RESUMO

BACKGROUND: Human embryonic stem cells (hESC) provide a unique model to study early events in human development. The hESC-derived cells can potentially be used to replace or restore different tissues including neuronal that have been damaged by disease or injury. METHODOLOGY AND PRINCIPAL FINDINGS: The cells of two different hESC lines were converted to neural rosettes using adherent and chemically defined conditions. The progenitor cells were exposed to retinoic acid (RA) or to human recombinant basic fibroblast growth factor (bFGF) in the late phase of the rosette formation. Exposing the progenitor cells to RA suppressed differentiation to rostral forebrain dopamine neural lineage and promoted that of spinal neural tissue including motor neurons. The functional characteristics of these differentiated neuronal precursors under both, rostral (bFGF) and caudalizing (RA) signals were confirmed by patch clamp analysis. CONCLUSIONS/SIGNIFICANCE: These findings suggest that our differentiation protocol has the capacity to generate region-specific and electrophysiologically active neurons under in vitro conditions without embryoid body formation, co-culture with stromal cells and without presence of cells of mesodermal or endodermal lineages.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Embrionário/fisiologia , Células-Tronco Embrionárias/citologia , Neurônios/citologia , Neurônios/fisiologia , Técnicas de Cultura de Células/métodos , Linhagem Celular , Meios de Cultura , Eletrofisiologia , Humanos , Imuno-Histoquímica , Especificidade de Órgãos , Técnicas de Patch-Clamp , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Tissue Eng Part C Methods ; 14(4): 311-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19055357

RESUMO

The most valuable property of stem cells (SCs) is their potential to differentiate into many or all cell types of the body. So far, monitoring SC differentiation has only been possible after cells were fixed or destroyed during sample preparation. It is, however, important to develop nondestructive methods of monitoring SCs. Scanning ion conductance microscopy (SICM) is a unique imaging technique that uses similar principles to the atomic force microscope, but with a pipette for the probe. This allows scanning of the surface of living cells noninvasively and enables measurement of cellular activities under more physiological conditions than is possible with other high-resolution microscopy techniques. We report here the novel use of the SICM for studying SCs to assess and monitor the status of SCs and various cell types differentiated from SCs.


Assuntos
Microscopia Eletrônica de Varredura/métodos , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Desenho de Equipamento , Humanos , Camundongos , Microscopia Confocal/métodos , Crista Neural/patologia , Neurônios/citologia , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células-Tronco/ultraestrutura , Propriedades de Superfície
13.
Stem Cells ; 24(7): 1628-37, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16556706

RESUMO

The development and transplantation of autologous cells derived from nuclear transfer embryonic stem cell (NT-ESC) lines to treat patients suffering from disease has been termed therapeutic cloning. Human NT is still a developing field, with further research required to improve somatic cell NT and human embryonic stem cell differentiation to deliver safe and effective cell replacement therapies. Furthermore, the implications of transferring mitochondrial heteroplasmic cells, which may harbor aberrant epigenetic gene expression profiles, are of concern. The production of human NT-ESC lines also remains plagued by ethical dilemmas, societal concerns, and controversies. Recently, a number of alternate therapeutic strategies have been proposed to circumvent the moral implications surrounding human nuclear transfer. It will be critical to overcome these biological, legislative, and moral restraints to maximize the potential of this therapeutic strategy and to alleviate human disease.


Assuntos
Clonagem de Organismos/estatística & dados numéricos , Transplante de Células-Tronco/estatística & dados numéricos , Animais , Clonagem de Organismos/ética , Clonagem de Organismos/legislação & jurisprudência , Clonagem de Organismos/tendências , DNA Mitocondrial/metabolismo , Epigênese Genética , Genes Mitocondriais , Rejeição de Enxerto/etiologia , Rejeição de Enxerto/genética , Humanos , Modelos Biológicos , Mutação , Doação de Oócitos/ética , Doação de Oócitos/legislação & jurisprudência , Doação de Oócitos/psicologia , Transplante de Células-Tronco/ética , Transplante de Células-Tronco/tendências
14.
Stem Cells ; 24(12): 2669-76, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16990582

RESUMO

Human embryonic stem cells (hESC) hold huge promise in modern regenerative medicine, drug discovery, and as a model for studying early human development. However, usage of embryos and derivation of hESC for research and potential medical application has resulted in polarized ethical debates since the process involves destruction of viable developing human embryos. Here we describe that not only developing embryos (morulae and blastocysts) of both good and poor quality but also arrested embryos could be used for the derivation of hESC. Analysis of arrested embryos demonstrated that these embryos express pluripotency marker genes such OCT4, NANOG, and REX1. Derived hESC lines also expressed specific pluripotency markers (TRA-1-60, TRA-1-81, SSEA4, alkaline phosphatase, OCT4, NANOG, TERT, and REX1) and differentiated under in vitro and in vivo conditions into derivates of all three germ layers. All of the new lines, including lines derived from late arrested embryos, have normal karyotypes. These results demonstrate that arrested embryos are additional valuable resources to surplus and donated developing embryos and should be used to study early human development or derive pluripotent hESC.


Assuntos
Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Células-Tronco Embrionárias/citologia , Embrião de Mamíferos/citologia , Humanos
15.
Hum Mol Genet ; 15(11): 1894-913, 2006 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-16644866

RESUMO

Understanding the molecular mechanism by which pluripotency is maintained in human embryonic stem cells (hESC) is important for the development of improved methods to derive, culture and differentiate these into cells of potential therapeutic use. Large-scale transcriptional comparison of the hES-NCL1 line derived from a day 8 embryo with H1 line derived from a day 5 embryo (WiCell Inc.) showed that only 0.52% of the transcripts analysed varied significantly between the two cell lines. This is within the variability range that has been reported when hESC derived from days 5-6 embryos have been compared with each other. This implies that transcriptional differences between the cell lines are likely to reflect their genetic profile rather than the embryonic stage from which they were derived. Bioinformatic analysis of expression changes observed when these cells were induced to differentiate as embryoid bodies suggested that quite a few of the downregulated genes were components of signal transduction networks. Subsequent analysis using western blotting, flow cytometry and antibody arrays implicated components of the PI3K/AKT kinase, MAPK/ERK and NFkappabeta pathways and confirmed that these components are decreased upon differentiation. Disruption of these pathways in isolation using specific inhibitors resulted in loss of pluripotency and/or loss of viability suggesting the importance of such signalling pathways in embryonic stem cell maintenance.


Assuntos
Embrião de Mamíferos/citologia , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Células-Tronco/citologia , Sobrevivência Celular , Biologia Computacional , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Humanos , Cariotipagem , Masculino , Modelos Biológicos , Transdução de Sinais , Transcrição Gênica
16.
Stem Cells ; 23(3): 306-14, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15749925

RESUMO

Human embryonic stem cells (hESCs) have great potential as a source of cells for therapeutic uses, but their culture requires the support of mouse or human cells, either directly as a feeder cell layer or indirectly as a source of conditioned medium in feeder-free culture systems. Unfortunately, the risks of cross-transfer of pathogens from xenogeneic or allogeneic feeders or cell by-products limit their medical applications. In addition, not all human feeders support the growth of hESCs equally well, and ethical concerns have been raised regarding the derivation of feeder cells from aborted human fetuses. We report here the culture of hESCs on a novel feeder cell system, comprising fibroblast-like cells derived from the spontaneous differentiation of hESCs. Isogenicity of the hESCs and hESC-derived fibroblasts was confirmed by micro satellite analysis. The nature of the hESC-derived fibroblasts was identified by the expression of specific markers. This feeder system permits continuous growth of undifferentiated and pluripotent hESCs, as demonstrated by the expression of specific hESC markers, by the formation of teratomas after injection of hESCs into severely combined immunodeficient mice, and by in vitro differentiation of hESCs into differentiated cells of ectodermal, endodermal, and mesodermal origin. Feeder cells derived from hESCs offers a potentially more secure autogeneic and genotypically homogenous system for the growth of undifferentiated hESCs.


Assuntos
Proliferação de Células , Embrião de Mamíferos/citologia , Fibroblastos/citologia , Células-Tronco Pluripotentes/citologia , Animais , Antígenos CD/metabolismo , Antígenos de Superfície/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Técnicas de Cocultura/métodos , Criopreservação , Meios de Cultivo Condicionados/farmacologia , Citometria de Fluxo , Expressão Gênica/genética , Humanos , Cariotipagem , Camundongos , Camundongos SCID , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Teratoma/patologia , Fatores de Transcrição/genética
17.
Stem Cells ; 23(7): 895-902, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15888688

RESUMO

One of the most frequently used matrices for feeder-free growth of undifferentiated human embryonic stem cells (hESCs) is Matrigel, which supports attachment and growth of undifferentiated hESCs in the presence of mouse embryonic fibroblast-conditioned medium. Unfortunately, application of Matrigel or medium conditioned by mouse embryonic feeder cells is not ideal for potential medical application of hESCs because xenogeneic pathogens can be transmitted through culture conditions. We demonstrate here that human serum as matrix and medium conditioned by differentiated hESCs reduce exposure of hESCs to animal ingredients and provide a safer direction toward completely animal-free conditions for application, handling, and understanding of hESC biology. At the same time, hESCs grown under these conditions maintain all hESC features after prolonged culture, including the developmental potential to differentiate into representative tissues of all three embryonic germ layers, unlimited and undifferentiated proliferative ability, and maintenance of normal karyotype.


Assuntos
Técnicas de Cultura de Células/métodos , Embrião de Mamíferos/citologia , Soro/metabolismo , Células-Tronco/citologia , Animais , Técnicas de Cultura de Células/instrumentação , Diferenciação Celular , Proliferação de Células , Colágeno/farmacologia , Meios de Cultivo Condicionados/farmacologia , Primers do DNA/química , Combinação de Medicamentos , Humanos , Cariotipagem/métodos , Laminina/farmacologia , Masculino , Camundongos , Camundongos SCID , Microscopia Eletrônica de Varredura , Proteoglicanas/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Testículo/metabolismo
18.
Reprod Biomed Online ; 11(2): 226-31, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16168222

RESUMO

This paper describes the derivation of a blastocyst following heterologous nuclear transfer (NT) into a human oocyte. It also demonstrates that a major obstacle to continuing research in human NT is the availability of suitable human oocytes. In this study, 36 oocytes were donated by 11 women undergoing four different treatments and their developmental potential was evaluated after NT. The time from oocyte collection to NT seems to be crucial, and only oocytes that were enucleated within 1 h proved successful. After enucleation of oocytes, fusion with undifferentiated human embryonic stem cells and in-vitro culture, early cleavage and blastocyst development of fused complexes was observed. The DNA fingerprinting comparison of the donor cells and derived blastocyst revealed successful heterologous NT, since both oocytes and donor cells were recovered from different patients. It has therefore been demonstrated that NT can be achieved in humans, using heterologous donor nuclei and surplus and donated oocytes. However, if the promise of this new science is to achieve its potential in the foreseeable future, it will be necessary to identify new sources of oocytes that can be used immediately after retrieval.


Assuntos
Blastocisto , Transferência Embrionária , Técnicas de Transferência Nuclear , Doação de Oócitos , Oócitos , Animais , Impressões Digitais de DNA , Feminino , Humanos , Masculino , Camundongos , Células-Tronco
19.
Stem Cells ; 23(8): 1035-43, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15983365

RESUMO

The homeobox transcription factor Nanog has been proposed to play a crucial role in the maintenance of the undifferentiated state of murine embryonic stem cells. A human counterpart, NANOG, has been identified, but its function and localization have not hitherto been described. We have used a combination of RNA interference and quantitative real-time polymerase chain reaction to study NANOG in human embryonic stem and embryonic carcinoma cells. Transfection of NANOG-specific small interfering RNAs reduced levels of NANOG transcript and protein and induced activation of the extraembryonic endoderm-associated genes GATA4, GATA6, LAMININ B1, and AFP as well as upregulation of trophectoderm-associated genes CDX2, GATA2, hCG-alpha, and hCG-beta. Immunostaining of preimplantation human embryos showed that NANOG was expressed in the inner cell mass of expanded blastocysts but not in earlier-stage embryos, consistent with a role in the maintenance of pluripotency. Taken together, our findings suggest that NANOG acts as a gatekeeper of pluripotency in human embryonic stem and carcinoma cells by preventing their differentiation to extraembryonic endoderm and trophectoderm lineages.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Proteínas de Homeodomínio/fisiologia , Células-Tronco Pluripotentes/citologia , Blastocisto/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula/fisiologia , Proteínas de Ligação a DNA/genética , Regulação para Baixo , Embrião de Mamíferos/citologia , Células-Tronco de Carcinoma Embrionário , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA6/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Proteínas de Fluorescência Verde , Proteínas de Homeodomínio/genética , Humanos , Proteína Homeobox Nanog , Células-Tronco Neoplásicas , Células-Tronco Pluripotentes/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Transfecção , alfa-Fetoproteínas/metabolismo
20.
Stem Cells ; 22(7): 1142-51, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15579635

RESUMO

There are several different technical approaches to the isolation of hematopoietic stem cells (HSCs) with long-term repopulating ability, but these have problems in terms of yield, complexity, or cell viability. Simpler strategies for HSC isolation are needed. We have enriched primitive hematopoietic progenitors from murine bone marrow of mice from different genetic backgrounds by lineage depletion followed by selection of cells with high aldehyde dehydrogenase activity using the Aldefluor reagent (BD Biosciences, Oxford, U.K.). Lin- ALDH(bright) cells comprised 26.8 +/- 1.0% of the total Lin- population of C57BL6 mice, and 23.5 +/- 1.0% of the Lin- population of BALB/c mice expressed certain cell-surface markers typical of primitive hematopoietic progenitors. In vitro hematopoietic progenitor function was substantially higher in the Lin- ALDH(bright) population compared with the Lin- ALDH(low) cells. These cells have higher telomerase activity and the lowest percentage of cells in S phase. These data strongly suggest that progenitor enrichment from Lin- cells on the basis of ALDH is a valid method whose simplicity of application makes it advantageous over conventional separations.


Assuntos
Técnicas de Cultura de Células/métodos , Separação Celular/métodos , Células-Tronco Hematopoéticas/citologia , Animais , Células da Medula Óssea/citologia , Ciclo Celular , Linhagem da Célula , Sobrevivência Celular , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fenótipo , Fase S , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA