Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Cell Physiol ; 231(9): 2040-7, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-26773436

RESUMO

The lineage relationship between prostate adenocarcinoma and small cell carcinoma was studied by using the LuCaP family of xenografts established from primary neoplasm to metastasis. Expression of four stem cell transcription factor (TF) genes, LIN28A, NANOG, POU5F1, SOX2, were analyzed in the LuCaP lines. These genes, when force expressed in differentiated cells, can reprogram the recipients into stem-like induced pluripotent stem (iPS) cells. Most LuCaP lines expressed POU5F1, while LuCaP 145.1, representative of small cell carcinoma, expressed all four. Through transcriptome database query, many small cell carcinoma genes were also found in stem cells. To test the hypothesis that prostate cancer progression from "differentiated" adenocarcinoma to "undifferentiated" small cell carcinoma could involve re-expression of stem cell genes, the four TF genes were transduced via lentiviral vectors into five adenocarcinoma LuCaP lines-70CR, 73CR, 86.2, 92, 105CR-as done in iPS cell reprogramming. The resultant cells from these five transductions displayed a morphology of small size and dark appearing unlike the parentals. Transcriptome analysis of LuCaP 70CR* ("*" to denote transfected progeny) revealed a unique gene expression close to that of LuCaP 145.1. In a prostate principal components analysis space based on cell-type transcriptomes, the different LuCaP transcriptome datapoints were aligned to suggest a possible ordered sequence of expression changes from the differentiated luminal-like adenocarcinoma cell types to the less differentiated, more stem-like small cell carcinoma types, and LuCaP 70CR*. Prostate cancer progression can thus be molecularly characterized by loss of differentiation with re-expression of stem cell genes. J. Cell. Physiol. 231: 2040-2047, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Adenocarcinoma/metabolismo , Carcinoma de Células Pequenas/metabolismo , Diferenciação Celular/genética , Células-Tronco Pluripotentes Induzidas/citologia , Próstata/metabolismo , Neoplasias da Próstata/metabolismo , Diferenciação Celular/fisiologia , Linhagem Celular Tumoral , Reprogramação Celular , Perfilação da Expressão Gênica/métodos , Genes Homeobox/genética , Humanos , Masculino , Próstata/patologia , Neoplasias da Próstata/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
2.
Nat Cell Biol ; 17(12): 1523-35, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571212

RESUMO

For nearly a century developmental biologists have recognized that cells from embryos can differ in their potential to differentiate into distinct cell types. Recently, it has been recognized that embryonic stem cells derived from both mice and humans exhibit two stable yet epigenetically distinct states of pluripotency: naive and primed. We now show that nicotinamide N-methyltransferase (NNMT) and the metabolic state regulate pluripotency in human embryonic stem cells (hESCs).  Specifically, in naive hESCs, NNMT and its enzymatic product 1-methylnicotinamide are highly upregulated, and NNMT is required for low S-adenosyl methionine (SAM) levels and the H3K27me3 repressive state. NNMT consumes SAM in naive cells, making it unavailable for histone methylation that represses Wnt and activates the HIF pathway in primed hESCs. These data support the hypothesis that the metabolome regulates the epigenetic landscape of the earliest steps in human development.


Assuntos
Diferenciação Celular , Epigênese Genética/genética , Células-Tronco Embrionárias Humanas/metabolismo , Metaboloma , Animais , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Perfilação da Expressão Gênica/métodos , Técnicas de Silenciamento de Genes , Histonas/metabolismo , Humanos , Lisina/metabolismo , Espectrometria de Massas , Metabolômica/métodos , Metilação , Camundongos , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Nicotinamida N-Metiltransferase/genética , Nicotinamida N-Metiltransferase/metabolismo , Proteômica/métodos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , S-Adenosilmetionina/metabolismo , Transdução de Sinais
3.
Stem Cells ; 33(9): 2674-85, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26013465

RESUMO

During early patterning of the neural plate, a single region of the embryonic forebrain, the eye field, becomes competent for eye development. The hallmark of eye field specification is the expression of the eye field transcription factors (EFTFs). Experiments in fish, amphibians, birds, and mammals have demonstrated largely conserved roles for the EFTFs. Although some of the key signaling events that direct the synchronized expression of these factors to the eye field have been elucidated in fish and frogs, it has been more difficult to study these mechanisms in mammalian embryos. In this study, we have used two different methods for directed differentiation of mouse embryonic stem cells (mESCs) to generate eye field cells and retina in vitro to test for a role of the PDZ domain-containing protein GIPC1 in the specification of the mammalian eye primordia. We find that the overexpression of a dominant-negative form of GIPC1 (dnGIPC1), as well as the downregulation of endogenous GIPC1, is sufficient to inhibit the development of eye field cells from mESCs. GIPC1 interacts directly with IGFR and participates in Akt1 activation, and pharmacological inhibition of Akt1 phosphorylation mimics the dnGIPC1 phenotype. Our data, together with previous studies in Xenopus, support the hypothesis that the GIPC1-PI3K-Akt1 pathway plays a key role in eye field specification in vertebrates.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas do Olho/biossíntese , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Proto-Oncogênicas c-akt/biossíntese , Retina/metabolismo , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Células HEK293 , Humanos , Camundongos , Retina/citologia , Xenopus laevis
4.
Nature ; 522(7555): 221-5, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25896322

RESUMO

Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections, and comprise nearly 8% of the human genome. The most recently acquired human ERV is HERVK(HML-2), which repeatedly infected the primate lineage both before and after the divergence of the human and chimpanzee common ancestor. Unlike most other human ERVs, HERVK retained multiple copies of intact open reading frames encoding retroviral proteins. However, HERVK is transcriptionally silenced by the host, with the exception of in certain pathological contexts such as germ-cell tumours, melanoma or human immunodeficiency virus (HIV) infection. Here we demonstrate that DNA hypomethylation at long terminal repeat elements representing the most recent genomic integrations, together with transactivation by OCT4 (also known as POU5F1), synergistically facilitate HERVK expression. Consequently, HERVK is transcribed during normal human embryogenesis, beginning with embryonic genome activation at the eight-cell stage, continuing through the emergence of epiblast cells in preimplantation blastocysts, and ceasing during human embryonic stem cell derivation from blastocyst outgrowths. Remarkably, we detected HERVK viral-like particles and Gag proteins in human blastocysts, indicating that early human development proceeds in the presence of retroviral products. We further show that overexpression of one such product, the HERVK accessory protein Rec, in a pluripotent cell line is sufficient to increase IFITM1 levels on the cell surface and inhibit viral infection, suggesting at least one mechanism through which HERVK can induce viral restriction pathways in early embryonic cells. Moreover, Rec directly binds a subset of cellular RNAs and modulates their ribosome occupancy, indicating that complex interactions between retroviral proteins and host factors can fine-tune pathways of early human development.


Assuntos
Blastocisto/virologia , Retrovirus Endógenos/metabolismo , Células-Tronco Pluripotentes/virologia , Ativação Viral , Antígenos de Diferenciação/metabolismo , Blastocisto/citologia , Blastocisto/metabolismo , Linhagem Celular , Metilação de DNA , Retrovirus Endógenos/genética , Feminino , Produtos do Gene gag/metabolismo , Humanos , Masculino , Fator 3 de Transcrição de Octâmero/metabolismo , Fases de Leitura Aberta/genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ribossomos/genética , Ribossomos/metabolismo , Sequências Repetidas Terminais/genética , Transcrição Gênica/genética , Ativação Transcricional , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
5.
Curr Urol Rep ; 16(1): 468, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25404182

RESUMO

Prostate cancer progression is characterized by tumor dedifferentiation. Cancer cells of less differentiated tumors have a gene expression/transcriptome more similar to that of stem cells. In dedifferentiation, cancer cells may follow a specific program of gene expression changes to a stem-like state. In order to treat cancer effectively, the stem-like cancer cells and cancer differentiation pathway need to be identified and studied. Due to the very low abundance of stem-like cancer cells, their isolation from fresh human tumors is technically challenging. Induced pluripotent stem cell technology can reprogram differentiated cells into stem-like, and this may be a tool to generate sufficient stem-like cancer cells.


Assuntos
Desdiferenciação Celular , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas , Células-Tronco Neoplásicas/citologia , Próstata/citologia , Neoplasias da Próstata , Linhagem Celular Tumoral , Humanos , Masculino , Análise de Componente Principal
6.
Proc Natl Acad Sci U S A ; 111(12): 4484-9, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24623855

RESUMO

The naïve pluripotent state has been shown in mice to lead to broad and more robust developmental potential relative to primed mouse epiblast cells. The human naïve ES cell state has eluded derivation without the use of transgenes, and forced expression of OCT4, KLF4, and KLF2 allows maintenance of human cells in a naïve state [Hanna J, et al. (2010) Proc Natl Acad Sci USA 107(20):9222-9227]. We describe two routes to generate nontransgenic naïve human ES cells (hESCs). The first is by reverse toggling of preexisting primed hESC lines by preculture in the histone deacetylase inhibitors butyrate and suberoylanilide hydroxamic acid, followed by culture in MEK/ERK and GSK3 inhibitors (2i) with FGF2. The second route is by direct derivation from a human embryo in 2i with FGF2. We show that human naïve cells meet mouse criteria for the naïve state by growth characteristics, antibody labeling profile, gene expression, X-inactivation profile, mitochondrial morphology, microRNA profile and development in the context of teratomas. hESCs can exist in a naïve state without the need for transgenes. Direct derivation is an elusive, but attainable, process, leading to cells at the earliest stage of in vitro pluripotency described for humans. Reverse toggling of primed cells to naïve is efficient and reproducible.


Assuntos
Células-Tronco Embrionárias/citologia , Animais , Linhagem da Célula , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Inibidores de Histona Desacetilases/farmacologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Inibidores de Proteínas Quinases/farmacologia , Transgenes , Inativação do Cromossomo X
7.
Stem Cells ; 31(9): 1737-48, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23765801

RESUMO

Adult stem cells reside in hypoxic niches, and embryonic stem cells (ESCs) are derived from a low oxygen environment. However, it is not clear whether hypoxia is critical for stem cell fate since for example human ESCs (hESCs) are able to self-renew in atmospheric oxygen concentrations as well. We now show that hypoxia can govern cell fate decisions since hypoxia alone can revert hESC- or iPSC-derived differentiated cells back to a stem cell-like state, as evidenced by re-activation of an Oct4-promoter reporter. Hypoxia-induced "de-differentiated" cells also mimic hESCs in their morphology, long-term self-renewal capacity, genome-wide mRNA and miRNA profiles, Oct4 promoter methylation state, cell surface markers TRA1-60 and SSEA4 expression, and capacity to form teratomas. These data demonstrate that hypoxia can influence cell fate decisions and could elucidate hypoxic niche function.


Assuntos
Linhagem da Célula , Células-Tronco Pluripotentes/citologia , Adulto , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Biomarcadores/metabolismo , Desdiferenciação Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Histona Desacetilases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Modelos Biológicos , Fator 3 de Transcrição de Octâmero/metabolismo , Oxigênio/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo
8.
Inflamm Bowel Dis ; 19(1): 174-84, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22619032

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) pathogenesis involves an inadequately controlled immune reaction to intestinal microbiota, and CD4(+) T cells, dependent on MHC class II (MHC-II) processing and presentation by antigen-presenting cells (APC), play important roles. The role of professional APC (macrophages and dendritic cells [DCs]) and nonprofessional APC (intestinal epithelial cells [IECs]) in microbial-driven intestinal inflammation remains controversial. METHODS: We generated transgenic animals on an MHC-II(-/-) genetic background in which MHC-II is expressed on 1) DC via the CD11c promoter (CD11cTg) or 2) IEC via the fatty acid binding protein (liver) promoter (EpithTg). These mice were crossed with Rag2(-/-) mice to eliminate T and B cells (CD11cTg/Rag2(-/-) and EpithTg/Rag2(-/-)). Helicobacter bilis (Hb) infection and adoptive transfer (AT) of naïve CD4 T cells were used to trigger IBD. RESULTS: CD11cTg/Rag2(-/-) mice infected with Hb+AT developed severe colitis within 3 weeks post-AT, similar to disease in positive control Rag2(-/-) mice infected with Hb+AT. CD11cTg/Rag2(-/-) mice given AT alone or Hb alone had significantly less severe colitis. In contrast, EpithTg/Rag2(-/-) mice infected with Hb+AT developed mild colitis by 3 weeks and even after 16 weeks post-AT had only mild lesions. CONCLUSIONS: MHC-II expression restricted to DCs is sufficient to induce severe colitis in the presence of T cells and a microorganism such as Hb within 3 weeks of AT. Expression of MHC-II solely on IEC in the presence of a microbial trigger and T cells was insufficient to trigger severe colitis.


Assuntos
Colite/etiologia , Células Dendríticas/imunologia , Células Epiteliais/imunologia , Infecções por Helicobacter/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Inflamação/imunologia , Transferência Adotiva , Animais , Western Blotting , Antígeno CD11c/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Colite/patologia , Proteínas de Ligação a DNA/fisiologia , Células Dendríticas/microbiologia , Células Epiteliais/microbiologia , Feminino , Citometria de Fluxo , Helicobacter , Infecções por Helicobacter/genética , Infecções por Helicobacter/microbiologia , Interações Hospedeiro-Patógeno , Imunidade Inata/imunologia , Técnicas Imunoenzimáticas , Imunofenotipagem , Inflamação/genética , Inflamação/microbiologia , Camundongos , Camundongos Transgênicos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/microbiologia
9.
EMBO J ; 31(9): 2103-16, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22446391

RESUMO

The function of metabolic state in stemness is poorly understood. Mouse embryonic stem cells (ESC) and epiblast stem cells (EpiSC) are at distinct pluripotent states representing the inner cell mass (ICM) and epiblast embryos. Human embryonic stem cells (hESC) are similar to EpiSC stage. We now show a dramatic metabolic difference between these two stages. EpiSC/hESC are highly glycolytic, while ESC are bivalent in their energy production, dynamically switching from glycolysis to mitochondrial respiration on demand. Despite having a more developed and expanding mitochondrial content, EpiSC/hESC have low mitochondrial respiratory capacity due to low cytochrome c oxidase (COX) expression. Similarly, in vivo epiblasts suppress COX levels. These data reveal EpiSC/hESC functional similarity to the glycolytic phenotype in cancer (Warburg effect). We further show that hypoxia-inducible factor 1α (HIF1α) is sufficient to drive ESC to a glycolytic Activin/Nodal-dependent EpiSC-like stage. This metabolic switch during early stem-cell development may be deterministic.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Glicólise , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Ativinas/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , DNA Mitocondrial/análise , Feminino , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos C57BL , Prostaglandina-Endoperóxido Sintases/metabolismo
10.
Prostate ; 72(13): 1453-63, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22314551

RESUMO

BACKGROUND: CD90(+) prostate cancer-associated (CP) stromal cells represent a diseased cell type found only in tumor tissue. They differ from their normal counterpart in gene expression and inductive signaling. Genetic reprogramming by induced pluripotent stem (iPS) cell technology can effectively change adult cells into stem-like cells through wholesale alteration of the gene expression program. This technology might be used to 'erase' the abnormal gene expression of diseased cells. The resultant iPS cells would no longer express the disease phenotype, and behave like stem cells. METHODS: CP stromal cells, isolated from tumor tissue of a surgically resected prostate by anti-CD90-mediated sorting and cultured in vitro, were transfected with in vitro packaged lentiviral expression vectors containing stem cell transcription factor genes POU5F1, LIN28, NANOG, and SOX2. RESULTS: Alkaline phosphatase-positive iPS cells were obtained in about 3 weeks post-transfection at a frequency of 10(-4) . Their colony morphology was indistinguishable from that of human embryonic stem (ES) cells. Transcriptome analysis showed a virtually complete match in gene expression between the iPS and ES cells. CONCLUSIONS: Genes of CP stromal cells could be fully inactivated by genetic reprogramming. As a consequence, the disease phenotype was 'cured'.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Neoplasias da Próstata/patologia , Células Estromais/patologia , Células-Tronco Embrionárias/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células Estromais/metabolismo , Transcriptoma
11.
Cancer Res ; 71(13): 4640-52, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21712410

RESUMO

Low oxygen levels have been shown to promote self-renewal in many stem cells. In tumors, hypoxia is associated with aggressive disease course and poor clinical outcomes. Furthermore, many aggressive tumors have been shown to display gene expression signatures characteristic of human embryonic stem cells (hESC). We now tested whether hypoxia might be responsible for the hESC signature observed in aggressive tumors. We show that hypoxia, through hypoxia-inducible factor (HIF), can induce an hESC-like transcriptional program, including the induced pluripotent stem cell (iPSC) inducers, OCT4, NANOG, SOX2, KLF4, cMYC, and microRNA-302 in 11 cancer cell lines (from prostate, brain, kidney, cervix, lung, colon, liver, and breast tumors). Furthermore, nondegradable forms of HIFα, combined with the traditional iPSC inducers, are highly efficient in generating A549 iPSC-like colonies that have high tumorigenic capacity. To test potential correlation between iPSC inducers and HIF expression in primary tumors, we analyzed primary prostate tumors and found a significant correlation between NANOG-, OCT4-, and HIF1α-positive regions. Furthermore, NANOG and OCT4 expressions positively correlated with increased prostate tumor Gleason score. In primary glioma-derived CD133 negative cells, hypoxia was able to induce neurospheres and hESC markers. Together, these findings suggest that HIF targets may act as key inducers of a dynamic state of stemness in pathologic conditions.


Assuntos
Biomarcadores Tumorais/biossíntese , Células-Tronco Embrionárias/metabolismo , Fator 1 Induzível por Hipóxia/metabolismo , Células-Tronco Neoplásicas/metabolismo , Biomarcadores Tumorais/genética , Hipóxia Celular/genética , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Células-Tronco Embrionárias/fisiologia , Glioma/genética , Glioma/metabolismo , Glioma/patologia , Células HCT116 , Células HT29 , Células HeLa , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Fator 1 Induzível por Hipóxia/biossíntese , Fator 4 Semelhante a Kruppel , Masculino , MicroRNAs/biossíntese , Proteína Homeobox Nanog , Células-Tronco Neoplásicas/fisiologia , Fator 3 de Transcrição de Octâmero/biossíntese , Fator 3 de Transcrição de Octâmero/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Regulação para Cima
12.
BMC Med Genomics ; 4: 46, 2011 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-21605402

RESUMO

BACKGROUND: Prostate tumor heterogeneity is a major factor in disease management. Heterogeneity could be due to multiple cancer cell types with distinct gene expression. Of clinical importance is the so-called cancer stem cell type. Cell type-specific transcriptomes are used to examine lineage relationship among cancer cell types and their expression similarity to normal cell types including stem/progenitor cells. METHODS: Transcriptomes were determined by Affymetrix DNA array analysis for the following cell types. Putative prostate progenitor cell populations were characterized and isolated by expression of the membrane transporter ABCG2. Stem cells were represented by embryonic stem and embryonal carcinoma cells. The cancer cell types were Gleason pattern 3 (glandular histomorphology) and pattern 4 (aglandular) sorted from primary tumors, cultured prostate cancer cell lines originally established from metastatic lesions, xenografts LuCaP 35 (adenocarcinoma phenotype) and LuCaP 49 (neuroendocrine/small cell carcinoma) grown in mice. No detectable gene expression differences were detected among serial passages of the LuCaP xenografts. RESULTS: Based on transcriptomes, the different cancer cell types could be clustered into a luminal-like grouping and a non-luminal-like (also not basal-like) grouping. The non-luminal-like types showed expression more similar to that of stem/progenitor cells than the luminal-like types. However, none showed expression of stem cell genes known to maintain stemness. CONCLUSIONS: Non-luminal-like types are all representatives of aggressive disease, and this could be attributed to the similarity in overall gene expression to stem and progenitor cell types.


Assuntos
Linhagem da Célula/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Antígeno AC133 , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Masculino , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/genética , Peptídeos/metabolismo , Fenótipo , Análise de Componente Principal , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Ther ; 18(6): 1192-9, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20407427

RESUMO

Precise genetic manipulation of human pluripotent stem cells will be required to realize their scientific and therapeutic potential. Here, we show that adeno-associated virus (AAV) gene targeting vectors can be used to genetically engineer human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). Different types of sequence-specific changes, including the creation and correction of mutations, were introduced into the human HPRT1 and HMGA1 genes (HPRT1 mutations being responsible for Lesch-Nyhan syndrome). Gene targeting occurred at high frequencies in both ESCs and iPSCs, with over 1% of all colony-forming units (CFUs) undergoing targeting in some experiments. AAV vectors could also be used to target genes in human fibroblasts that were subsequently used to derive iPSCs. Accurate and efficient targeting took place with minimal or no cytotoxicity, and most of the gene-targeted stem cells produced were euploid and pluripotent.


Assuntos
Dependovirus/genética , Marcação de Genes , Engenharia Genética , Células-Tronco Pluripotentes , Humanos , Hipoxantina Fosforribosiltransferase/genética
14.
Cell Cycle ; 8(22): 3729-41, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19823043

RESUMO

microRNAs (miRNAs) regulate numerous physiological processes such as cell division and differentiation in many tissue types including stem cells. To probe the role that miRNAs play in regulating processes relevant to embryonic stem cell biology, we used RNA interference to silence DICER and DROSHA, the two main miRNA processing enzymes. Consistent with a role for miRNAs in maintaining normal stem cell division and renewal, we found that perturbation of miRNA pathway function in human embryonic stem cells (hESCs) attenuates cell proliferation. Normal cell growth can be partially restored by introduction of the mature miRNAs miR-195 and miR-372. These miRNAs regulate two tumor suppressor genes, respectively: WEE1, which encodes a negative G2/M kinase modulator of the CycB/CDK complex and CDKN1A, which encodes p21, a CycE/CDK cyclin dependent kinase inhibitor that regulates the G1/S transition. We show that in wild-type hESCs, WEE 1 levels control the rate of hESC division, whereas p21 levels must be maintained at a low level for hESC division to proceed. These data support a model for hESC cell cycle control in which miRNAs regulate negative cell cycle modulators at two phases of the cell cycle to ensure proper replenishment of the stem cell population.


Assuntos
Divisão Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , MicroRNAs/metabolismo , Modelos Biológicos , Sequência de Bases , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células-Tronco Embrionárias/metabolismo , Humanos , MicroRNAs/genética , Análise em Microsséries , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Oligonucleotídeos/genética , Proteínas Tirosina Quinases/metabolismo , Interferência de RNA , Ribonuclease III/genética
15.
Cell Stem Cell ; 4(4): 359-69, 2009 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-19341625

RESUMO

Recent evidence indicates that mouse and human embryonic stem cells (ESCs) are fixed at different developmental stages, with the former positioned earlier. We show that a narrow concentration of the naturally occurring short-chain fatty acid, sodium butyrate, supports the extensive self-renewal of mouse and human ESCs, while promoting their convergence toward an intermediate stem cell state. In response to butyrate, human ESCs regress to an earlier developmental stage characterized by a gene expression profile resembling that of mouse ESCs, preventing precocious Xist expression while retaining the ability to form complex teratomas in vivo. Other histone deacetylase inhibitors (HDACi) also support human ESC self-renewal. Our results indicate that HDACi can promote ESC self-renewal across species, and demonstrate that ESCs can toggle between alternative states in response to environmental factors.


Assuntos
Butiratos/farmacologia , Diferenciação Celular , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Inibidores de Histona Desacetilases , Animais , Células-Tronco Embrionárias/enzimologia , Inibidores Enzimáticos/farmacologia , Perfilação da Expressão Gênica , Histona Desacetilases/metabolismo , Humanos , Camundongos , RNA Longo não Codificante , RNA não Traduzido/metabolismo
16.
Exp Cell Res ; 314(16): 2930-40, 2008 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-18692044

RESUMO

Despite a growing body of literature concerning the hematopoietic differentiation of human embryonic stem cells (hESCs), the full hematopoietic potential of the majority of existing hESC lines remains unknown. In this study, the hematopoietic response of five NIH-approved hESC lines (H1, hSF6, BG01, BG02, and BG03) was compared. Our data show that despite expressing similar hESC markers under self-renewing conditions and initiating mesodermal differentiation under spontaneous differentiation conditions, marked differences in subsequent hematopoietic differentiation potential among these lines existed. A high degree of hematopoietic differentiation was attained only by H1 and BG02, whereas this process appeared to be abortive in nature for hSF6, BG01, and BG03. This difference in hematopoietic differentiation predisposition was readily apparent during spontaneous differentiation, and further augmented under hematopoietic-inducing conditions. This predisposition appeared to be intrinsic to the specific hESC line and independent of passage number or gender karyotype. Interestingly, H1 and BG02 displayed remarkable similarities in their kinetics of hematopoietic marker expression, hematopoietic colony formation, erythroid differentiation, and globin expression, suggesting that a similar, predetermined differentiation sequence is followed. The identification of intrinsic and extrinsic factors governing the hematopoietic differentiation potential of hESCs will be of great importance for the putative clinical utility of hESC lines.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Biomarcadores/metabolismo , Linhagem Celular , Linhagem da Célula , Técnicas de Cocultura , Células-Tronco Embrionárias/citologia , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/metabolismo , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Fenótipo
17.
Blood ; 110(12): 4111-9, 2007 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-17761519

RESUMO

Despite progress in developing defined conditions for human embryonic stem cell (hESC) cultures, little is known about the cell-surface receptors that are activated under conditions supportive of hESC self-renewal. A simultaneous interrogation of 42 receptor tyrosine kinases (RTKs) in hESCs following stimulation with mouse embryonic fibroblast (MEF) conditioned medium (CM) revealed rapid and prominent tyrosine phosphorylation of insulin receptor (IR) and insulin-like growth factor-1 receptor (IGF1R); less prominent tyrosine phosphorylation of epidermal growth factor receptor (EGFR) family members, including ERBB2 and ERBB3; and trace phosphorylation of fibroblast growth factor receptors. Intense IGF1R and IR phosphorylation occurred in the absence of MEF conditioning (NCM) and was attributable to high concentrations of insulin in the proprietary KnockOut Serum Replacer (KSR). Inhibition of IGF1R using a blocking antibody or lentivirus-delivered shRNA reduced hESC self-renewal and promoted differentiation, while disruption of ERBB2 signaling with the selective inhibitor AG825 severely inhibited hESC proliferation and promoted apoptosis. A simple defined medium containing an IGF1 analog, heregulin-1beta (a ligand for ERBB2/ERBB3), fibroblast growth factor-2 (FGF2), and activin A supported long-term growth of multiple hESC lines. These studies identify previously unappreciated RTKs that support hESC proliferation and self-renewal, and provide a rationally designed medium for the growth and maintenance of pluripotent hESCs.


Assuntos
Proliferação de Células , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Receptor ErbB-2/metabolismo , Receptor IGF Tipo 2/metabolismo , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Benzotiazóis/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados , Células-Tronco Embrionárias/citologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Neuregulina-1/farmacologia , Fosforilação/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-3/antagonistas & inibidores , Receptor ErbB-3/metabolismo , Receptor IGF Tipo 2/antagonistas & inibidores , Receptor de Insulina/antagonistas & inibidores , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tirfostinas/farmacologia
18.
Mol Ther ; 15(10): 1827-33, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17622243

RESUMO

Human embryonic stem cells (hESCs) are important tools for the study of stem cell biology and may ultimately be used in cellular therapies and regenerative medicine. For hESCs to achieve their potential, stable genetic modification of the hESC genome will be required. Here we have studied the transduction of hESCs by vectors based on foamy virus (FV), an integrating retrovirus with no known pathogenicity. We find that hESCs and also ESCs derived from rhesus monkeys can be efficiently transduced by FV vectors at frequencies of 14-48%. Integration of FV vector DNA was demonstrated by Southern blot analysis, and stable expression was observed from a single integrated provirus in several clones. Transduced hESCs expressed markers characteristic of undifferentiated cells, differentiated and expressed markers from all three germ layers after serum exposure, and formed teratomas with persistent transgene expression in differentiated cells. Thus, FV vectors are promising tools for the genetic modification of hESCs.


Assuntos
Células-Tronco Embrionárias/virologia , Vetores Genéticos , Spumavirus/genética , Transdução Genética , Sequência de Bases , Southern Blotting , Linhagem Celular , Primers do DNA , Imunofluorescência , Técnicas de Transferência de Genes , Humanos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Toxicol Sci ; 99(2): 628-36, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17584759

RESUMO

The analgesic and antipyretic drug acetaminophen (APAP) is bioactivated to the reactive intermediate N-acetyl-p-benzoquinoneimine, which is scavenged by glutathione (GSH). APAP overdose can deplete GSH leading to the accumulation of APAP-protein adducts and centrilobular necrosis in the liver. N-acetylcysteine (NAC), a cysteine prodrug and GSH precursor, is often given as a treatment for APAP overdose. The rate-limiting step in GSH biosynthesis is catalyzed by glutamate cysteine ligase (GCL) a heterodimer composed of catalytic and modifier (GCLM) subunits. Previous studies have indicated that GCL activity is likely to be an important determinant of APAP toxicity. In this study, we investigated APAP toxicity, and NAC or GSH ethyl ester (GSHee)-mediated rescue in mice with normal or compromised GCLM expression. Gclm wild-type, heterozygous, and null mice were administered APAP (500 mg/kg) alone, or immediately following NAC (800 mg/kg) or GSHee (168 mg/kg), and assessed for hepatotoxicity 6 h later. APAP caused GSH depletion in all mice. Gclm null and heterozygous mice exhibited more extensive hepatic damage compared to wild-type mice as assessed by serum alanine aminotransferase activity and histopathology. Additionally, male Gclm wild-type mice demonstrated greater APAP-induced hepatotoxicity than female wild-type mice. Cotreatment with either NAC or GSHee mitigated the effects of APAP in Gclm wild-type and heterozygous mice, but not in Gclm null mice. Collectively, these data reassert the importance of GSH in protection against APAP-induced hepatotoxicity, and indicate critical roles for GCL activity and gender in APAP-induced liver damage in mice.


Assuntos
Acetaminofen/toxicidade , Analgésicos não Narcóticos/toxicidade , Glutamato-Cisteína Ligase/fisiologia , Fígado/efeitos dos fármacos , Alanina Transaminase/sangue , Animais , Feminino , Glutamato-Cisteína Ligase/deficiência , Glutationa/análise , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Subunidades Proteicas , Caracteres Sexuais
20.
FASEB J ; 21(7): 1345-57, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17284483

RESUMO

Embryonic stem (ES) cells are promising for cardiac repair, but directing their differentiation toward cardiomyocytes remains challenging. We investigated whether the heart guides ES cells toward cardiomyocytes in vivo and whether allogeneic ES cells were immunologically tolerated. Undifferentiated mouse ES cells consistently formed cardiac teratomas in nude or immunocompetent syngeneic mice. Cardiac teratomas contained no more cardiomyocytes than hind-limb teratomas, suggesting lack of guided differentiation. ES cells also formed teratomas in infarcted hearts, indicating injury-related signals did not direct cardiac differentiation. Allogeneic ES cells also caused cardiac teratomas, but these were immunologically rejected after several weeks, in association with increased inflammation and up-regulation of class I and II histocompatibility antigens. Fusion between ES cells and cardiomyocytes occurred in vivo, but was rare. Infarct autofluorescence was identified as an artifact that might be mistaken for enhanced GFP expression and true regeneration. Hence, undifferentiated ES cells were not guided toward a cardiomyocyte fate in either normal or infarcted hearts, and there was no evidence for allogeneic immune tolerance of ES cell derivatives. Successful cardiac repair strategies involving ES cells will need to control cardiac differentiation, avoid introducing undifferentiated cells, and will likely require immune modulation to avoid rejection.


Assuntos
Neoplasias Cardíacas/patologia , Miocárdio/citologia , Transplante de Células-Tronco , Teratoma/patologia , Animais , Diferenciação Celular , Linhagem Celular , Feminino , Neoplasias Cardíacas/imunologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Teratoma/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA