Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Dev Biol ; 60(10-11-12): 327-336, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28000904

RESUMO

In 2016, a symposium was convened in Leroy C. Stevens' honor, in association with a meeting of the International Stem Cell Initiative (ISCI). ISCI, funded internationally, is composed of a group of ~100 scientists from many countries, under the leadership of Peter Andrews, who have worked together to characterize a significant number of human pluripotent stem cell lines, to monitor their genetic stability and their differentiation into mature cell types and tissues in vitro and in vivo. Those at the ISCI meeting puzzled through one of the thorniest problems in the therapeutic use of the differentiated derivatives of pluripotent stem cells for human therapy; namely, pluripotent stem cells can differentiate into any cell type in the adult organism, but they also have the capacity for unlimited self-renewal, hence if mutated they may have tumorigenic potential. The meeting considered how these cells might become genetically or epigenetically abnormal and how the safety of these cells for human therapeutic uses could be assessed and assured. The symposium was an opportunity to pay tribute to Leroy Stevens and to the basic science origins of this newest aspect of regenerative medicine. It was a time to reflect on the past and on how it can influence the future of our field.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes , Medicina Regenerativa , História do Século XX , Humanos , Estados Unidos
2.
Stem Cells Dev ; 24(22): 2634-48, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26154167

RESUMO

Primordial germ cells (PGCs) share many properties with embryonic stem cells (ESCs) and innately express several key pluripotency-controlling factors, including OCT4, NANOG, and LIN28. Therefore, PGCs may provide a simple and efficient model for studying somatic cell reprogramming to induced pluripotent stem cells (iPSCs), especially in determining the regulatory mechanisms that fundamentally define pluripotency. Here, we report a novel model of PGC reprogramming to generate iPSCs via transfection with SOX2 and OCT4 using integrative lentiviral. We also show the feasibility of using nonintegrative approaches for generating iPSC from PGCs using only these two factors. We show that human PGCs express endogenous levels of KLF4 and C-MYC protein at levels similar to embryonic germ cells (EGCs) but lower levels of SOX2 and OCT4. Transfection with both SOX2 and OCT4 together was required to induce PGCs to a pluripotent state at an efficiency of 1.71%, and the further addition of C-MYC increased the efficiency to 2.33%. Immunohistochemical analyses of the SO-derived PGC-iPSCs revealed that these cells were more similar to ESCs than EGCs regarding both colony morphology and molecular characterization. Although leukemia inhibitory factor (LIF) was not required for the generation of PGC-iPSCs like EGCs, the presence of LIF combined with ectopic exposure to C-MYC yielded higher efficiencies. Additionally, the SO-derived PGC-iPSCs exhibited differentiation into representative cell types from all three germ layers in vitro and successfully formed teratomas in vivo. Several lines were generated that were karyotypically stable for up to 24 subcultures. Their derivation efficiency and survival in culture significantly supersedes that of EGCs, demonstrating their utility as a powerful model for studying factors regulating pluripotency in future studies.


Assuntos
Reprogramação Celular , Células-Tronco Embrionárias/citologia , Células Germinativas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Células Germinativas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo
3.
PLoS One ; 10(1): e0116933, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25635918

RESUMO

Induced pluripotent stem (iPS) cells are at the forefront of research in regenerative medicine and are envisaged as a source for personalized tissue repair and cell replacement therapy. Here, we demonstrate for the first time that oligodendrocyte progenitors (OPs) can be derived from iPS cells generated using either an episomal, non-integrating plasmid approach or standard integrating retroviruses that survive and differentiate into mature oligodendrocytes after early transplantation into the injured spinal cord. The efficiency of OP differentiation in all 3 lines tested ranged from 40% to 60% of total cells, comparable to those derived from human embryonic stem cells. iPS cell lines derived using episomal vectors or retroviruses generated a similar number of early neural progenitors and glial progenitors while the episomal plasmid-derived iPS line generated more OPs expressing late markers O1 and RIP. Moreover, we discovered that iPS-derived OPs (iPS-OPs) engrafted 24 hours following a moderate contusive spinal cord injury (SCI) in rats survived for approximately two months and that more than 70% of the transplanted cells differentiated into mature oligodendrocytes that expressed myelin associated proteins. Transplanted OPs resulted in a significant increase in the number of myelinated axons in animals that received a transplantation 24 h after injury. In addition, nearly a 5-fold reduction in cavity size and reduced glial scarring was seen in iPS-treated groups compared to the control group, which was injected with heat-killed iPS-OPs. Although further investigation is needed to understand the mechanisms involved, these results provide evidence that patient-specific, iPS-derived OPs can survive for three months and improve behavioral assessment (BBB) after acute transplantation into SCI. This is significant as determining the time in which stem cells are injected after SCI may influence their survival and differentiation capacity.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Animais , Axônios/fisiologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Intervenção Médica Precoce , Feminino , Humanos , Atividade Motora , Bainha de Mielina/fisiologia , Regeneração Nervosa , Oligodendroglia/fisiologia , Ratos Endogâmicos Lew , Recuperação de Função Fisiológica , Medula Espinal/patologia , Medula Espinal/fisiopatologia , Resultado do Tratamento
4.
PLoS One ; 7(6): e39088, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737227

RESUMO

Human embryonic germ cells (EGCs) provide a powerful model for identifying molecules involved in the pluripotent state when compared to their progenitors, primordial germ cells (PGCs), and other pluripotent stem cells. Microarray and Principal Component Analysis (PCA) reveals for the first time that human EGCs possess a transcription profile distinct from PGCs and other pluripotent stem cells. Validation with qRT-PCR confirms that human EGCs and PGCs express many pluripotency-associated genes but with quantifiable differences compared to pluripotent embryonic stem cells (ESCs), induced pluripotent stem cells (IPSCs), and embryonal carcinoma cells (ECCs). Analyses also identified a number of target genes that may be potentially associated with their unique pluripotent states. These include IPO7, MED7, RBM26, HSPD1, and KRAS which were upregulated in EGCs along with other pluripotent stem cells when compared to PGCs. Other potential target genes were also found which may contribute toward a primed ESC-like state. These genes were exclusively up-regulated in ESCs, IPSCs and ECCs including PARP1, CCNE1, CDK6, AURKA, MAD2L1, CCNG1, and CCNB1 which are involved in cell cycle regulation, cellular metabolism and DNA repair and replication. Gene classification analysis also confirmed that the distinguishing feature of EGCs compared to ESCs, ECCs, and IPSCs lies primarily in their genetic contribution to cellular metabolism, cell cycle, and cell adhesion. In contrast, several genes were found upregulated in PGCs which may help distinguish their unipotent state including HBA1, DMRT1, SPANXA1, and EHD2. Together, these findings provide the first glimpse into a unique genomic signature of human germ cells and pluripotent stem cells and provide genes potentially involved in defining different states of germ-line pluripotency.


Assuntos
Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica/métodos , Estudo de Associação Genômica Ampla , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Genoma , Humanos , Camundongos , Microscopia de Contraste de Fase/métodos , Modelos Genéticos , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , Transcrição Gênica
5.
Stem Cells Dev ; 20(2): 351-61, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20486775

RESUMO

Human primordial germ cells (PGCs) have proven to be a source of pluripotent stem cells called embryonic germ cells (EGCs). Unlike embryonic stem cells, virtually little is known regarding the factors that regulate EGC survival and maintenance. In mice, the growth factor bone morphogenetic protein 4 (BMP4) has been shown to be required for maintaining mouse embryonic stem cells, and disruptions in this gene lead to defects in mouse PGC specification. Here, we sought to determine whether recombinant human BMP4 could influence EGC derivation and/or human PGC survival. We found that the addition of recombinant BMP4 increased the number of human PGCs after 1 week of culture in a dose-responsive manner. The efficiency of EGC derivation and maintenance in culture was also enhanced by the presence of recombinant BMP4 based on alkaline phosphatase and OCT4 staining. In addition, an antagonist of the BMP4 pathway, Noggin, decreased PGC proliferation and led to an increase in cystic embryoid body formation. Quantitative real-time (qRT)-polymerase chain reaction analyses and immunostaining confirmed that the constituents of the BMP4 pathway were upregulated in EGCs versus PGCs. Downstream activators of the BMP4 pathway such as ID1 and phosphorylated SMADs 1 and 5 were also expressed, suggesting a role of this growth factor in EGC pluripotency.


Assuntos
Proteína Morfogenética Óssea 4/farmacologia , Células Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Proteínas Recombinantes/farmacologia , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Proteína Morfogenética Óssea 4/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas/genética , Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Corpos Embrioides/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Células Germinativas/efeitos dos fármacos , Idade Gestacional , Humanos , Células-Tronco Pluripotentes/efeitos dos fármacos , Gravidez , Proteínas Smad/genética , Proteínas Smad/metabolismo , Regulação para Cima
6.
PLoS One ; 5(7): e11536, 2010 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-20634944

RESUMO

Stem cell transplantation holds great promise for the treatment of myocardial infarction injury. We recently described the embryonic stem cell-derived cardiac progenitor cells (CPCs) capable of differentiating into cardiomyocytes, vascular endothelium, and smooth muscle. In this study, we hypothesized that transplanted CPCs will preserve function of the infarcted heart by participating in both muscle replacement and neovascularization. Differentiated CPCs formed functional electromechanical junctions with cardiomyocytes in vitro and conducted action potentials over cm-scale distances. When transplanted into infarcted mouse hearts, CPCs engrafted long-term in the infarct zone and surrounding myocardium without causing teratomas or arrhythmias. The grafted cells differentiated into cross-striated cardiomyocytes forming gap junctions with the host cells, while also contributing to neovascularization. Serial echocardiography and pressure-volume catheterization demonstrated attenuated ventricular dilatation and preserved left ventricular fractional shortening, systolic and diastolic function. Our results demonstrate that CPCs can engraft, differentiate, and preserve the functional output of the infarcted heart.


Assuntos
Células-Tronco Embrionárias/transplante , Infarto do Miocárdio/terapia , Miócitos Cardíacos/transplante , Animais , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Feminino , Estimativa de Kaplan-Meier , Camundongos , Infarto do Miocárdio/mortalidade , Miócitos Cardíacos/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos
7.
Int J Neurosci ; 120(4): 305-13, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20374080

RESUMO

This study utilized a contusion model of spinal cord injury (SCI) in rats using the standardized NYU-MASCIS impactor, after which oligodendrocyte progenitor cells (OPCs) derived from human embryonic stem cell (ESC) were transplanted into the spinal cord to study their survival and migration route toward the areas of injury. One critical aspect of successful cell-based SCI therapy is the time of injection following injury. OPCs were injected at two clinically relevant times when most damage occurs to the surrounding tissue, 3 and 24 hours following injury. Migration and survivability after eight days was measured postmortem. In-vitro immunofluorescence revealed that most ESC-derived OPCs expressed oligodendrocyte markers, including CNPase, GalC, Olig1, O4, and O1. Results showed that OPCs survived when injected at the center of injury and migrated away from the injection sites after one week. Histological sections revealed integration of ESC-derived OPCs into the spinal cord with contusion injury without disruption to the parenchyma. Cells survived for a minimum of eight days after injury, without tumor or cyst formation. The extent of injury and effect of early cell transplant was measured using behavioral and electrophysiological assessments which demonstrated increased neurological responses in rats transplanted with OPCs compared to controls.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Oligodendroglia/fisiologia , Traumatismos da Medula Espinal/cirurgia , Animais , Antígenos/metabolismo , Modelos Animais de Doenças , Potenciais Somatossensoriais Evocados/fisiologia , Feminino , Gangliosídeos/metabolismo , Humanos , Proteínas do Tecido Nervoso/metabolismo , Antígenos O/metabolismo , Proteoglicanas/metabolismo , Ratos , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Fatores de Transcrição SOXE/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Transplante de Células-Tronco/métodos
8.
Proteomics ; 10(7): 1359-73, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20104618

RESUMO

Pluripotent human embryonic stem cells (ESCs) can be differentiated in vitro into a variety of cells which hold promise for transplantation therapy. Human embryonal carcinoma cells (ECCs), stem cells of human teratocarcinomas, are considered a close but malignant counterpart to human ESCs. In this study, a comprehensive quantitative proteomic analysis of ESCs and ECCs was carried out using the iTRAQ method. Using two-dimensional LC and MS/MS analyses, we identified and quantitated approximately 1800 proteins. Among these are proteins associated with pluripotency and development as well as tight junction signaling and TGFbeta receptor pathway. Nearly approximately 200 proteins exhibit more than twofold difference in abundance between ESCs and ECCs. Examples of early developmental markers high in ESCs include beta-galactoside-binding lectin, undifferentiated embryonic cell transcription factor-1, DNA cytosine methyltransferase 3beta isoform-B, melanoma antigen family-A4, and interferon-induced transmembrane protein-1. In contrast, CD99-antigen (CD99), growth differentiation factor-3, cellular retinoic acid binding protein-2, and developmental pluripotency associated-4 were among the highly expressed proteins in ECCs. Several proteins that were highly expressed in ECCs such as heat shock 27 kDa protein-1, mitogen-activated protein kinase kinase-1, nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor like-2, and S100 calcium-binding protein-A4 have also been attributed to malignancy in other systems. Importantly, immunocytochemistry was used to validate the proteomic analyses for a subset of the proteins. In summary, this is the first large-scale quantitative proteomic study of human ESCs and ECCs, which provides critical information about the regulators of these two closely related, but developmentally distinct, stem cells.


Assuntos
Células-Tronco de Carcinoma Embrionário/metabolismo , Células-Tronco Embrionárias/metabolismo , Proteômica/métodos , Cromatografia Líquida , Humanos , Imuno-Histoquímica , Marcação por Isótopo , Microscopia de Fluorescência , Fragmentos de Peptídeos/metabolismo , Proteoma/metabolismo , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
9.
J Proteome Res ; 8(3): 1315-26, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19173612

RESUMO

The ability to derive neural progenitors, differentiated neurons and glial cells from human embryonic stem cells (hESCs) with high efficiency holds promise for a number of clinical applications. However, investigating the temporal events is crucial for defining the underlying mechanisms that drive this process of differentiation along different lineages. We carried out quantitative proteomic profiling using a multiplexed approach capable of analyzing eight different samples simultaneously to monitor the temporal dynamics of protein abundance as human embryonic stem cells differentiate into motor neurons or astrocytes. With this approach, a catalog of approximately 1200 proteins along with their relative quantitative expression patterns was generated. The differential expression of the large majority of these proteins has not previously been reported or studied in the context of neural differentiation. As expected, two of the widely used markers of pluripotency, alkaline phosphatase (ALPL) and LIN28, were found to be downregulated during differentiation, while S-100 and tenascin C were upregulated in astrocytes. Neurofilament 3 protein, doublecortin and CAM kinase-like 1 and nestin proteins were upregulated during motor neuron differentiation. We identified a number of proteins whose expression was largely confined to specific cell types, embryonic stem cells, embryoid bodies and differentiating motor neurons. For example, glycogen phosphorylase (PYGL) and fatty acid binding protein 5 (FABP5) were enriched in ESCs, while beta spectrin (SPTBN5) was highly expressed in embryoid bodies. Karyopherin, heat shock 27 kDa protein 1 and cellular retinoic acid binding protein 2 (CRABP2) were upregulated in differentiating motor neurons but were downregulated in mature motor neurons. We validated some of the novel markers of the differentiation process using immunoblotting and immunocytochemical labeling. To our knowledge, this is the first large-scale temporal proteomic profiling of human stem cell differentiation into neural cell types highlighting proteins with limited or undefined roles in neural fate.


Assuntos
Antígenos de Diferenciação/metabolismo , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Neurogênese/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/fisiologia , Humanos , Proteoma/metabolismo
11.
Physiol Genomics ; 34(3): 277-84, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18523156

RESUMO

SOX17 is a SRY-related high-mobility group (HMG) box transcription factor that is necessary for endoderm formation in multiple species. Despite its essential function during endoderm formation and differentiation, few direct targets of SOX17 are known. To identify targets of SOX17, we isolated SOX17 binding sites with a chromatin immunoprecipitation (ChIP)-cloning screen. SOX17-ChIP identified zinc finger protein 202 (Zfp202) as a direct target of SOX17 during endoderm differentiation of F9 embryonal carcinoma cells. A sequence in the first intron of Zfp202 activated transcription in differentiated F9 cells, and overexpression of Sox17 increased the transcriptional activity of this sequence. SOX17 binds to a site within this sequence in electrophoretic mobility shift assays, and mutation of this site decreases the transcriptional activation. Zfp202 is induced concomitantly with Sox17 during endoderm differentiation of F9 cells. We also show that ZFP202 represses Hnf4a, which has been reported for the human ortholog ZNF202. Identifying targets of SOX17 will help to elucidate the molecular basis of endoderm differentiation and may provide a better understanding of the role of endoderm in patterning the other germ layers.


Assuntos
Diferenciação Celular , Endoderma/citologia , Proteínas Repressoras/genética , Fatores de Transcrição SOXF/metabolismo , Transcrição Gênica , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Células Clonais , Clonagem Molecular , Ensaio de Desvio de Mobilidade Eletroforética , Desenvolvimento Embrionário/genética , Regulação da Expressão Gênica no Desenvolvimento , Fator 4 Nuclear de Hepatócito/genética , Humanos , Camundongos , Regiões Promotoras Genéticas/genética , Ligação Proteica , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXF/genética
12.
Hum Reprod ; 23(3): 589-99, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18203707

RESUMO

BACKGROUND: Human primordial germ cells (PGCs) can give rise to pluripotent stem cells such as embryonal carcinoma cells (ECCs) and embryonic germ cells (EGCs). METHODS: In order to determine whether PGCs express markers associated with pluripotency in EGCs and ECCs, the following study cross examines the expression patterns of multiple pluripotent markers in the human fetal ovary, 5.5-15 weeks post-fertilizaton (pF) and relates this expression with the ability to derive pluripotent EGCs in vitro. RESULTS: Specific subpopulations were identified which included OCT4(+)/Nanog(+)/cKIT(+)/VASA(+) PGCs and oogonia. Interestingly, these cells also expressed SSEA1 and alkaline phosphatase (AP) and SSEA4 expression occurred throughout the entire gonad. Isolation of SSEA1(+) cells from the gonad resulted in AP(+) EGC colony formation. The number of OCT4(+) or Nanog(+) expressing cells peaked by week 8 and then diminished after week 9 pF, as oogonia enter meiosis. In addition, the efficiency of EGC derivation was associated with the number of OCT4(+) cells. TRA-1-60 and TRA-1-81 were only detected in the lining of the mesonephric ducts and occasionally in the gonad. CONCLUSIONS: These results demonstrate that PGCs, a unipotent cell, express most, but not all, of the markers associated with pluripotent cells in the human fetal ovary.


Assuntos
Ovário/citologia , Ovário/embriologia , Células-Tronco Pluripotentes/metabolismo , Fosfatase Alcalina/biossíntese , Antígenos de Superfície/biossíntese , Biomarcadores/metabolismo , Feminino , Feto/citologia , Glicoesfingolipídeos/biossíntese , Humanos , Hibridização in Situ Fluorescente , Antígenos CD15/biossíntese , Fator 3 de Transcrição de Octâmero/biossíntese , Gravidez , Proteoglicanas/biossíntese , Proteínas Proto-Oncogênicas c-kit/biossíntese , Antígenos Embrionários Estágio-Específicos
13.
Stem Cells ; 26(2): 412-21, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18024420

RESUMO

Human primordial germ cells (PGCs) have proven to be a source of pluripotent stem cells called embryonic germ cells (EGCs). However, the developmental potency of these cells in the fetal gonad still remains elusive. Thus, this study provides a comprehensive analysis of pluripotent and germ cell marker expression in human fetal testis 7-15 weeks postfertilization (pF) and compares this expression to their ability to derive EGCs. Although the majority of germ cells expressed stem cell markers stage-specific embryonic antigen (SSEA) 1, SSEA4, EMA-1, and alkaline phosphatase, only a small percentage of those (<1%) expressed OCT4, CKIT, and NANOG. Specifically, the number of OCT4(+)/CKIT(+)/NANOG(+) cells significantly increased in the developing cords during weeks 7-9, followed by a gradual decline into week 15 pF. By week 15 pF, the remaining OCT4(+)/CKIT(+)/NANOG(+) cells were found in the cords surrounding the periphery of the testis, and the predominant germ cells, CKIT(+) cells, no longer expressed OCT4 or NANOG. Based on morphology and early germ cell marker expression, including VASA, PUM2, and DAZL, we suggest these cells are mitotically active gonocytes or prespermatogonia. Importantly, the number of OCT4(+) cells correlated with an increase in the number of EGC colonies derived in culture. Interestingly, two pluripotent markers, Tra-1-60 and Tra-1-81, although highly expressed in EGCs, were not expressed by PGCs in the gonad. Together, these results suggest that PGCs maintain expression of pluripotent stem cell markers during and after sexual differentiation of the gonad, albeit in very low numbers.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Testículo/embriologia , Biomarcadores/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Técnica Indireta de Fluorescência para Anticorpo , Idade Gestacional , Glicoesfingolipídeos/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Antígenos CD15/metabolismo , Masculino , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Diferenciação Sexual , Espermatozoides/citologia , Espermatozoides/metabolismo , Antígenos Embrionários Estágio-Específicos , Testículo/citologia , Testículo/metabolismo
14.
Prog Cardiovasc Dis ; 49(6): 396-413, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17498520

RESUMO

Stem cells are potential agents for the treatment of myocardial infarcts among other heart diseases. Over the past decade, the scientific community has extensively used a wide variety of cells and examined their capacity to both regenerate the infarcted myocardium and improve functionally the diseased hearts. Some of the cells used include skeletal myoblasts, bone marrow-derived cells, adult cardiac resident stem cells, mesenchymal stem cells, and both mouse and human embryonic stem cells (Nat Biotechnol 2005;23:845-856). The reported cardiogenic capacity of the utilitized stem cells is assayed both in vitro through the use of differentiation paradigms and in vivo through transplantation into a variety of animal models of cardiac disease. The purpose of this review article is to summarize recent stem cell applications in cell-based cardiac therapies and their outcomes.


Assuntos
Procedimentos Cirúrgicos Cardíacos , Cardiopatias/cirurgia , Coração/fisiopatologia , Miocárdio/patologia , Regeneração , Transplante de Células-Tronco , Células-Tronco/fisiologia , Células-Tronco Adultas/fisiologia , Células-Tronco Adultas/transplante , Animais , Células da Medula Óssea/fisiologia , Transplante de Medula Óssea , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/transplante , Cardiopatias/patologia , Cardiopatias/fisiopatologia , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/fisiologia , Mioblastos Cardíacos/fisiologia , Mioblastos Cardíacos/transplante , Mioblastos Esqueléticos/fisiologia , Mioblastos Esqueléticos/transplante , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Resultado do Tratamento
15.
Methods Enzymol ; 419: 400-26, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17141064

RESUMO

To date, stem cells have been derived from three sources of germ cells. These include embryonic germ cells (EGCs), embryonal carcinoma cells (ECCs), and multipotent germ line stem cells (GSCs). EGCs are derived from primordial germ cells that arise in the late embryonic and early fetal period of development. ECCs are derived from adult testicular tumors whereas GSCs have been derived by culturing spermatogonial stem cells from mouse neonates and adults. For each of these lines, their pluripotency has been demonstrated by their ability to differentiate into cell types derived from the three germ layers in vitro and in vivo and in chimeric animals, including germ line transmission. These germ line-derived stem cells have been generated from many species including human, mice, porcine, and chicken albeit with only slight modifications. This chapter describes general considerations regarding critical aspects of their derivation compared with their counterpart, embryonic stem cells (ESCs). Detailed protocols for EGC derivation and maintenance from human and mouse primordial germ cells (PGCs) will be presented.


Assuntos
Células-Tronco Adultas , Células Germinativas , Células-Tronco Pluripotentes , Animais , Técnicas de Cultura de Células/métodos , Humanos
16.
Semin Reprod Med ; 24(5): 304-13, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17123225

RESUMO

Embryonic germ cells (EGCs) are pluripotent stem cells derived from primordial germ cells (PGCs). PGCs are progenitors of adult gametes, which diverge from the somatic lineage between late embryonic to early fetal development. First derived in the mouse, EGCs have also been derived from human, chicken, and pig. As pluripotent stem cells, EGCs demonstrate long-term self-renewal via clonal expansion in an undifferentiated state, and differentiate in vitro to form embryoid bodies containing cells that represent all three germ layers as well as mixed cell populations of less differentiated progenitors and precursors. This is also demonstrated in vivo by their formation into experimentally induced teratocarcinomas following transplantation. Furthermore, mice, pig, and chicken EGCs have also been shown to contribute to experimentally produced chimeric animals, including germline transmission. Importantly, EGCs demonstrate normal and stable karyotypes as well as normal patterns of genomic imprinting, including X-inactivation. Transplantation studies have begun in a variety of models in hopes of defining their potential use to treat a wide variety of human conditions, including diabetes and urological and neurological disorders.


Assuntos
Embrião de Mamíferos/citologia , Células Germinativas/citologia , Células-Tronco/citologia , Animais , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/metabolismo , Células Germinativas/transplante , Humanos , Células-Tronco/metabolismo
18.
J Neurosci Res ; 82(5): 592-608, 2005 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16247803

RESUMO

Stem cell therapy is a hope for the treatment of some childhood neurological disorders. We examined whether human neural stem cells (hNSCs) replace lost cells in a newborn mouse model of brain damage. Excitotoxic lesions were made in neonatal mouse forebrain with the N-methyl-D-aspartate (NMDA) receptor agonist quinolinic acid (QA). QA induced apoptosis in neocortex, hippocampus, striatum, white matter, and subventricular zone. This degeneration was associated with production of cleaved caspase-3. Cells immunopositive for inducible nitric oxide synthase were present in damaged white matter and subventricular zone. Three days after injury, mice received brain parenchymal or intraventricular injections of hNSCs derived from embryonic germ (EG) cells. Human cells were prelabeled in vitro with DiD for in vivo tracking. The locations of hNSCs within the mouse brain were determined through DiD fluorescence and immunodetection of human-specific nestin and nuclear antigen 7 days after transplantation. hNSCs survived transplantation into the lesioned mouse brain, as evidenced by human cell markers and DiD fluorescence. The cells migrated away from the injection site and were found at sites of injury within the striatum, hippocampus, thalamus, and white matter tracts and at remote locations in the brain. Subsets of grafted cells expressed neuronal and glial cell markers. hNSCs restored partially the complement of striatal neurons in brain-damaged mice. We conclude that human EG cell-derived NSCs can engraft successfully into injured newborn brain, where they can survive and disseminate into the lesioned areas, differentiate into neuronal and glial cells, and replace lost neurons. (c) 2005 Wiley-Liss, Inc.


Assuntos
Dano Encefálico Crônico/terapia , Células Germinativas/transplante , Neurônios/fisiologia , Oligodendroglia/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco/fisiologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Dano Encefálico Crônico/induzido quimicamente , Dano Encefálico Crônico/fisiopatologia , Caspase 3 , Caspases/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Células Germinativas/citologia , Células Germinativas/fisiologia , Sobrevivência de Enxerto/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Degeneração Neural/induzido quimicamente , Degeneração Neural/fisiopatologia , Degeneração Neural/terapia , Neurônios/citologia , Neurotoxinas , Óxido Nítrico Sintase/metabolismo , Oligodendroglia/citologia , Prosencéfalo/citologia , Prosencéfalo/fisiologia , Prosencéfalo/cirurgia , Transplante de Células-Tronco/tendências , Células-Tronco/citologia , Resultado do Tratamento
19.
Cloning Stem Cells ; 7(1): 27-34, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15996115

RESUMO

Mouse embryoid bodies (EBs) differentiate into dorsal spinal cord neural progenitors in response to retinoic acid (RA). Our data demonstrate that the addition of Sonic Hedgehog (Shh) directs towards a ventral spinal cord neural tube fate, but only at extremely high concentrations. One possible explanation is the presence of dorsal directing factors. Bone morphogenetic proteins (BMPs), known to direct dorsal spinal cord neural differentiation, were expressed in RA-treated EBs. Shh more potently directed ventral differentiation when combined with the BMP inhibitor Noggin. Further, when BMP7 was added, the ability of Shh to direct ventral differentiation was further mitigated.


Assuntos
Proteínas Morfogenéticas Ósseas/biossíntese , Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Transativadores/metabolismo , Animais , Padronização Corporal , Proteína Morfogenética Óssea 7 , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Transporte/metabolismo , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Técnicas de Cultura , Indução Embrionária , Proteínas Hedgehog , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodomínio/metabolismo , Camundongos , Mitose , Neurônios/citologia , Estrutura Terciária de Proteína , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/embriologia , Células-Tronco/citologia , Fatores de Tempo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Fator de Crescimento Transformador beta/metabolismo , Tretinoína/metabolismo , Tretinoína/farmacologia , Proteínas de Peixe-Zebra
20.
Urology ; 65(4): 827-32, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15833555

RESUMO

OBJECTIVES: To evaluate the capability of a human embryonic germ (hEG) cell-derived cell line (SDEC), previously characterized in our laboratory, seeded on porcine small intestinal submucosa (SIS) to regenerate the injured rat bladder. METHODS: Fluorescent-labeled SDEC cells seeded on SIS for 8 days in vitro were used as bladder grafts in rats. A total of 30 congenitally athymic rats (six groups of 5 rats each), underwent partial cystectomy and replacement with plain SIS (groups 1 to 3) or cell-seeded SIS (groups 4 to 6). The rats were sacrificed after 7 (groups 1 and 4), 14 (groups 2 and 5), and 28 (groups 3 and 6) days. The bladders were analyzed by histopathologic examination and fluorescence microscopy. RESULTS: No graft rejection or diminution in bladder capacity occurred. Plain SIS implants had multiple calcareous deposits, not seen with the cell-seeded implants. Macroscopically, at 7 days, the grafts were healed with a cellular lining on the luminal aspect in groups 4 to 6. Microscopically, the rat bladder was completely regenerated 28 days after stem cell-seeded SIS implantation. Labeled stem cells were identified throughout the graft and contributed significantly to bladder regeneration. CONCLUSIONS: The results of this study have demonstrated the successful replacement of a bladder defect in a rat model using hEG cell-derived cells seeded on SIS grafts. Longer term analysis of these bladder grafts will allow evaluation of function, cell migration, and differentiation processes of human stem cells.


Assuntos
Regeneração , Células-Tronco/fisiologia , Bexiga Urinária/fisiologia , Animais , Linhagem Celular , Modelos Animais , Ratos , Ratos Nus , Transplante de Células-Tronco , Técnicas de Cultura de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA