Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Blood ; 143(10): 895-911, 2024 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-37890146

RESUMO

ABSTRACT: A major hurdle in adoptive T-cell therapy is cell exhaustion and failure to maintain antitumor responses. Here, we introduce an induced pluripotent stem cell (iPSC) strategy for reprogramming and revitalizing precursor exhausted B-cell maturation antigen (BCMA)-specific T cells to effectively target multiple myeloma (MM). Heteroclitic BCMA72-80 (YLMFLLRKI)-specific CD8+ memory cytotoxic T lymphocytes (CTL) were epigenetically reprogrammed to a pluripotent state, developed into hematopoietic progenitor cells (CD34+ CD43+/CD14- CD235a-), differentiated into the T-cell lineage and evaluated for their polyfunctional activities against MM. The final T-cell products demonstrated (1) mature CD8αß+ memory phenotype, (2) high expression of activation or costimulatory molecules (CD38, CD28, and 41BB), (3) no expression of immune checkpoint and senescence markers (CTLA4, PD1, LAG3, and TIM3; CD57), and (4) robust proliferation and polyfunctional immune responses to MM. The BCMA-specific iPSC-T cells possessed a single T-cell receptor clonotype with cognate BCMA peptide recognition and specificity for targeting MM. RNA sequencing analyses revealed distinct genome-wide shifts and a distinctive transcriptional profile in selected iPSC clones, which can develop CD8αß+ memory T cells. This includes a repertoire of gene regulators promoting T-cell lineage development, memory CTL activation, and immune response regulation (LCK, IL7R, 4-1BB, TRAIL, GZMB, FOXF1, and ITGA1). This study highlights the potential application of iPSC technology to an adaptive T-cell therapy protocol and identifies specific transcriptional patterns that could serve as a biomarker for selection of suitable iPSC clones for the successful development of antigen-specific CD8αß+ memory T cells to improve the outcome in patients with MM.


Assuntos
Antineoplásicos , Antígenos CD8 , Células-Tronco Pluripotentes Induzidas , Mieloma Múltiplo , Humanos , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Células-Tronco Pluripotentes Induzidas/metabolismo , Antígeno de Maturação de Linfócitos B/metabolismo , Linfócitos T Citotóxicos , Antineoplásicos/metabolismo
2.
Commun Biol ; 5(1): 1246, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36380073

RESUMO

Stromal cells interact with immune cells during initiation and resolution of immune responses, though the precise underlying mechanisms remain to be resolved. Lessons learned from stromal cell-based therapies indicate that environmental signals instruct their immunomodulatory action contributing to immune response control. Here, to the best of our knowledge, we show a novel function for the guanine-exchange factor DOCK2 in regulating immunosuppressive function in three human stromal cell models and by siRNA-mediated DOCK2 knockdown. To identify immune function-related stromal cell molecular signatures, we first reprogrammed mesenchymal stem/progenitor cells (MSPCs) into induced pluripotent stem cells (iPSCs) before differentiating these iPSCs in a back-loop into MSPCs. The iPSCs and immature iPS-MSPCs lacked immunosuppressive potential. Successive maturation facilitated immunomodulation, while maintaining clonogenicity, comparable to their parental MSPCs. Sequential transcriptomics and methylomics displayed time-dependent immune-related gene expression trajectories, including DOCK2, eventually resembling parental MSPCs. Severe combined immunodeficiency (SCID) patient-derived fibroblasts harboring bi-allelic DOCK2 mutations showed significantly reduced immunomodulatory capacity compared to non-mutated fibroblasts. Conditional DOCK2 siRNA knockdown in iPS-MSPCs and fibroblasts also immediately reduced immunomodulatory capacity. Conclusively, CRISPR/Cas9-mediated DOCK2 knockout in iPS-MSPCs also resulted in significantly reduced immunomodulation, reduced CDC42 Rho family GTPase activation and blunted filopodia formation. These data identify G protein signaling as key element devising stromal cell immunomodulation.


Assuntos
Proteínas Ativadoras de GTPase , Guanina , Humanos , Proteínas Ativadoras de GTPase/genética , RNA Interferente Pequeno , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Imunidade , Imunomodulação
3.
Nat Biotechnol ; 33(11): 1182-92, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26501952

RESUMO

Research on human pluripotent stem cells has been hampered by the lack of a standardized, quantitative, scalable assay of pluripotency. We previously described an assay called ScoreCard that used gene expression signatures to quantify differentiation efficiency. Here we report an improved version of the assay based on qPCR that enables faster, more quantitative assessment of functional pluripotency. We provide an in-depth characterization of the revised signature panel (commercially available as the TaqMan hPSC Scorecard Assay) through embryoid body and directed differentiation experiments as well as a detailed comparison to the teratoma assay. We further show that the improved ScoreCard enables a wider range of applications, such as screening of small molecules, genetic perturbations and assessment of culture conditions. Our approach can be extended beyond stem cell applications to characterize and assess the utility of other cell types and lineages.


Assuntos
Diferenciação Celular/genética , Biologia Computacional/métodos , Corpos Embrioides/fisiologia , Células-Tronco Pluripotentes/fisiologia , Reação em Cadeia da Polimerase/métodos , Animais , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Humanos , Camundongos , Neoplasias Experimentais/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Teratoma/patologia
4.
Cell Stem Cell ; 16(1): 13-7, 2015 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-25575079

RESUMO

Induced pluripotent stem cells (iPSCs) have the potential to transform drug discovery and healthcare in the 21(st) century. However, successful commercialization will require standardized manufacturing platforms. Here we highlight the need to define standardized practices for iPSC generation and processing and discuss current challenges to the robust manufacture of iPSC products.


Assuntos
Técnicas de Cultura de Células/métodos , Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Humanos , Reprodutibilidade dos Testes , Transplante de Células-Tronco
5.
Proc Natl Acad Sci U S A ; 110(31): 12774-9, 2013 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-23861493

RESUMO

Efficient generation of competent vasculogenic cells is a critical challenge of human induced pluripotent stem (hiPS) cell-based regenerative medicine. Biologically relevant systems to assess functionality of the engineered vessels in vivo are equally important for such development. Here, we report a unique approach for the derivation of endothelial precursor cells from hiPS cells using a triple combination of selection markers--CD34, neuropilin 1, and human kinase insert domain-containing receptor--and an efficient 2D culture system for hiPS cell-derived endothelial precursor cell expansion. With these methods, we successfully generated endothelial cells (ECs) from hiPS cells obtained from healthy donors and formed stable functional blood vessels in vivo, lasting for 280 d in mice. In addition, we developed an approach to generate mesenchymal precursor cells (MPCs) from hiPS cells in parallel. Moreover, we successfully generated functional blood vessels in vivo using these ECs and MPCs derived from the same hiPS cell line. These data provide proof of the principle that autologous hiPS cell-derived vascular precursors can be used for in vivo applications, once safety and immunological issues of hiPS-based cellular therapy have been resolved. Additionally, the durability of hiPS-derived blood vessels in vivo demonstrates a potential translation of this approach in long-term vascularization for tissue engineering and treatment of vascular diseases. Of note, we have also successfully generated ECs and MPCs from type 1 diabetic patient-derived hiPS cell lines and use them to generate blood vessels in vivo, which is an important milestone toward clinical translation of this approach.


Assuntos
Prótese Vascular , Células Endoteliais/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Neovascularização Fisiológica , Engenharia Tecidual , Animais , Células Endoteliais/transplante , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Camundongos , Camundongos SCID , Transplante Heterólogo , Doenças Vasculares/terapia
6.
Mol Cell Biol ; 33(16): 3274-85, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23775117

RESUMO

Polycomb group (PcG)-mediated repression is an evolutionarily conserved process critical for cell fate determination and maintenance of gene expression during embryonic development. However, the mechanisms underlying PcG recruitment in mammals remain unclear since few regulatory sites have been identified. We report two novel prospective PcG-dependent regulatory elements within the human HOXB and HOXC clusters and compare their repressive activities to a previously identified element in the HOXD cluster. These regions recruited the PcG proteins BMI1 and SUZ12 to a reporter construct in mesenchymal stem cells and conferred repression that was dependent upon PcG expression. Furthermore, we examined the potential of two DNA-binding proteins, JARID2 and YY1, to regulate PcG activity at these three elements. JARID2 has differential requirements, whereas YY1 appears to be required for repressive activity at all 3 sites. We conclude that distinct elements of the mammalian HOX clusters can recruit components of the PcG complexes and confer repression, similar to what has been seen in Drosophila. These elements, however, have diverse requirements for binding factors, which, combined with previous data on other loci, speaks to the complexity of PcG targeting in mammals.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Células-Tronco Mesenquimais/metabolismo , Família Multigênica , Proteínas do Grupo Polycomb/metabolismo , Animais , Células Cultivadas , Proteínas de Ligação a DNA/genética , Genes Homeobox , Humanos , Complexo Repressor Polycomb 2/genética , Complexo Repressor Polycomb 2/metabolismo , Proteínas do Grupo Polycomb/genética , Elementos Reguladores de Transcrição , Transcrição Gênica
7.
Stem Cell Res Ther ; 2(1): 10, 2011 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-21338517

RESUMO

INTRODUCTION: Pluripotent, human stem cells hold tremendous promise as a source of progenitor and terminally differentiated cells for application in future regenerative therapies. However, such therapies will be dependent upon the development of novel approaches that can best assess tissue outcomes of pluripotent stem cell-derived cells and will be essential to better predict their safety and stability following in vivo transplantation. METHODS: In this study we used engineered, human skin equivalents (HSEs) as a platform to characterize fibroblasts that have been derived from human embryonic stem (hES) cell. We characterized the phenotype and the secretion profile of two distinct hES-derived cell lines with properties of mesenchymal cells (EDK and H9-MSC) and compared their biological potential upon induction of differentiation to bone and fat and following their incorporation into the stromal compartment of engineered, HSEs. RESULTS: While both EDK and H9-MSC cell lines exhibited similar morphology and mesenchymal cell marker expression, they demonstrated distinct functional properties when incorporated into the stromal compartment of HSEs. EDK cells displayed characteristics of dermal fibroblasts that could support epithelial tissue development and enable re-epithelialization of wounds generated using a 3D tissue model of cutaneous wound healing, which was linked to elevated production of hepatocyte growth factor (HGF). Lentiviral shRNA-mediated knockdown of HGF resulted in a dramatic decrease of HGF secretion from EDK cells that led to a marked reduction in their ability to promote keratinocyte proliferation and re-epithelialization of cutaneous wounds. In contrast, H9-MSCs demonstrated features of mesenchymal stem cells (MSC) but not those of dermal fibroblasts, as they underwent multilineage differentiation in monolayer culture, but were unable to support epithelial tissue development and repair and produced significantly lower levels of HGF. CONCLUSIONS: Our findings demonstrate that hES-derived cells could be directed to specified and alternative mesenchymal cell fates whose function could be distinguished in engineered HSEs. Characterization of hES-derived mesenchymal cells in 3D, engineered HSEs demonstrates the utility of this tissue platform to predict the functional properties of hES-derived fibroblasts before their therapeutic transplantation.


Assuntos
Células-Tronco Embrionárias/citologia , Fibroblastos/citologia , Técnicas de Cultura de Células , Linhagem da Célula , Proliferação de Células , Fibroblastos/transplante , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/genética , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Queratinócitos/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Engenharia Tecidual , Cicatrização
8.
Endocrinology ; 152(3): 1080-9, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21209016

RESUMO

The human endometrium is a tissue with remarkable plasticity and regenerative capacity. Additionally, endometrial cells can be retrieved using minimally invasive procedures, which makes them an ideal source for reprogramming into a pluripotent state. Endometrial cells were obtained from donors in their fifth decade and reprogrammed into induced pluripotent stem (iPS) cells using retroviral transduction with SOX2, OCT4, KLF4, and MYC. The human endometrial cells displayed accelerated expression of endogenous NANOG and OCT4 during reprogramming compared with neonatal skin fibroblasts. As a result, iPS cell colonies that could be subcultured and propagated were established as early as 12 d after transduction rather than the usually reported 3-4 wk for other cell types. After 3 wk of reprogramming, the human endometrial cells also yielded significantly higher numbers of iPS colonies in comparison with the neonatal skin fibroblasts. Although the efficiency of iPS colony formation varied depending on the donor, the basal level of endogenous expression of the defined factors was positively correlated with reprogramming efficiency. The reprogramming resulted in an average colony-forming efficiency of 0.49 ± 0.10%, with a range from 0.31-0.66%, compared with the neonatal skin fibroblasts, resulting in an average efficiency of 0.03 ± 0.00% per transduction, with a range from 0.02-0.03%. Our studies show that the human endometrium expresses elevated levels of pluripotent factors, which with additional defined factors, results in significantly more efficient and accelerated generation of induced pluripotent stem cells compared with conventional somatic cells.


Assuntos
Endométrio/citologia , Regulação da Expressão Gênica/fisiologia , Células-Tronco Pluripotentes/metabolismo , Células Cultivadas , Feminino , Humanos , Fator 4 Semelhante a Kruppel
9.
Cell Stem Cell ; 6(6): 535-46, 2010 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-20569691

RESUMO

Murine pluripotent stem cells can exist in two functionally distinct states, LIF-dependent embryonic stem cells (ESCs) and bFGF-dependent epiblast stem cells (EpiSCs). However, human pluripotent cells so far seemed to assume only an epiblast-like state. Here we demonstrate that human iPSC reprogramming in the presence of LIF yields human stem cells that display morphological, molecular, and functional properties of murine ESCs. We termed these hLR5 iPSCs because they require the expression of five ectopic reprogramming factors, Oct4, Sox2, Klf4, cMyc, and Nanog, to maintain this more naive state. The cells are "metastable" and upon ectopic factor withdrawal they revert to standard human iPSCs. Finally, we demonstrate that the hLR5 state facilitates gene targeting, and as such provides a powerful tool for the generation of recombinant human pluripotent stem cell lines.


Assuntos
Células-Tronco Embrionárias/metabolismo , Técnicas de Transferência de Genes , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator Inibidor de Leucemia/farmacologia , Fatores de Transcrição/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Desdiferenciação Celular/efeitos dos fármacos , Desdiferenciação Celular/genética , Linhagem Celular , Células-Tronco Embrionárias/patologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Terapia Genética/métodos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/patologia , Fator 4 Semelhante a Kruppel , Camundongos , Recombinação Genética/genética , Homologia de Sequência , Fatores de Transcrição/genética
10.
Proc Natl Acad Sci U S A ; 107(8): 3311-6, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20133604

RESUMO

Clinical protocols utilize bone marrow to seed synthetic and decellularized allogeneic bone grafts for enhancement of scaffold remodeling and fusion. Marrow-derived cytokines induce host neovascularization at the graft surface, but hypoxic conditions cause cell death at the core. Addition of cellular components that generate an extensive primitive plexus-like vascular network that would perfuse the entire scaffold upon anastomosis could potentially yield significantly higher-quality grafts. We used a mouse model to develop a two-stage protocol for generating vascularized bone grafts using mesenchymal stem cells (hMSCs) from human bone marrow and umbilical cord-derived endothelial cells. The endothelial cells formed tube-like structures and subsequently networks throughout the bone scaffold 4-7 days after implantation. hMSCs were essential for stable vasculature both in vitro and in vivo; however, contrary to expectations, vasculature derived from hMSCs briefly cultured in medium designed to maintain a proliferative, nondifferentiated state was more extensive and stable than that with hMSCs with a TGF-beta-induced smooth muscle cell phenotype. Anastomosis occurred by day 11, with most hMSCs associating closely with the network. Although initially immature and highly permeable, at 4 weeks the network was mature. Initiation of scaffold mineralization had also occurred by this period. Some human-derived vessels were still present at 5 months, but the majority of the graft vasculature had been functionally remodeled with host cells. In conclusion, clinically relevant progenitor sources for pericytes and endothelial cells can serve to generate highly functional microvascular networks for tissue engineered bone grafts.


Assuntos
Vasos Sanguíneos/fisiologia , Osso e Ossos/irrigação sanguínea , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica , Pericitos/citologia , Engenharia Tecidual/métodos , Transplantes , Animais , Vasos Sanguíneos/citologia , Transplante Ósseo , Osso e Ossos/citologia , Linhagem da Célula , Humanos , Camundongos , Camundongos Endogâmicos , Modelos Animais , Osteogênese , Alicerces Teciduais
11.
Cell ; 140(1): 99-110, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20085705

RESUMO

Polycomb group (PcG) proteins are essential for accurate axial body patterning during embryonic development. PcG-mediated repression is conserved in metazoans and is targeted in Drosophila by Polycomb response elements (PREs). However, targeting sequences in humans have not been described. While analyzing chromatin architecture in the context of human embryonic stem cell (hESC) differentiation, we discovered a 1.8kb region between HOXD11 and HOXD12 (D11.12) that is associated with PcG proteins, becomes nuclease hypersensitive, and then shows alteration in nuclease sensitivity as hESCs differentiate. The D11.12 element repressed luciferase expression from a reporter construct and full repression required a highly conserved region and YY1 binding sites. Furthermore, repression was dependent on the PcG proteins BMI1 and EED and a YY1-interacting partner, RYBP. We conclude that D11.12 is a Polycomb-dependent regulatory region with similarities to Drosophila PREs, indicating conservation in the mechanisms that target PcG function in mammals and flies.


Assuntos
Células-Tronco Embrionárias/metabolismo , Genes Homeobox/genética , Proteínas de Homeodomínio/genética , Elementos Reguladores de Transcrição , Proteínas Repressoras/metabolismo , Diferenciação Celular , Cromatina/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteínas Nucleares/metabolismo , Complexo Repressor Polycomb 1 , Proteínas do Grupo Polycomb , Proteínas Proto-Oncogênicas/metabolismo
12.
J Tissue Eng Regen Med ; 3(5): 348-60, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19382119

RESUMO

Cartilage tissue can be engineered by starting from a diversity of cell sources, including stem-cell based and primary cell-based platforms. Selecting an appropriate cell source for the process of cartilage tissue engineering or repair is critical and challenging, due to the variety of cell options available. In this study, cellular responses of isolated human chondrocytes, human embryonic stem cells and mesenchymal stem cells (MSCs) derived from three sources, human embryonic stem cells, bone marrow and adipose tissue, were assessed for chondrogenic potential in 3D culture. All cell sources were characterized by FACS analysis to compare expression of some surface markers. The cells were differentiated in two different biomaterial matrices, silk and chitosan scaffolds, in the presence and absence of bone morphogenetic protein 6 (BMP6), along with the standard chondrogenic differentiating factors. Embryonic stem cells-derived MSCs showed unique characteristics, with preserved chondrogenic phenotype in both scaffolds with regard to chondrogenesis, as determined by real time RT-PCR, histological and microscopical analyses. After 4 weeks of cultivation, embryonic stem cells-derived MSCs were promising for chondrogenesis, particularly in the silk scaffolds with BMP6. The results suggest that cell source differences are important to consider with regard to chondrogenic outcomes, and among the variables addressed here the human embryonic stem cells-derived MSCs were the preferred cell source.


Assuntos
Cartilagem/citologia , Condrócitos/citologia , Células-Tronco Mesenquimais/citologia , Engenharia Tecidual/métodos , Tecido Adiposo/citologia , Medula Óssea/metabolismo , Proteína Morfogenética Óssea 6/metabolismo , Separação Celular , Quitosana/química , Condrócitos/metabolismo , Fibroínas/química , Citometria de Fluxo , Humanos , Imageamento Tridimensional , Fenótipo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Blood ; 111(3): 1302-5, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17993613

RESUMO

Tissue engineering requires formation of a de novo stable vascular network. Because of their ability to proliferate, differentiate into endothelial cells, and form new vessels, blood-derived endothelial progenitor cells (EPCs) are attractive source of cells for use in engineering blood vessels. However, the durability and function of EPC-derived vessels implanted in vivo are unclear. To this end, we directly compared formation and functions of tissue-engineered blood vessels generated by peripheral blood- and umbilical cord blood-derived EPCs in a model of in vivo vasculogenesis. We found that adult peripheral blood EPCs form blood vessels that are unstable and regress within 3 weeks. In contrast, umbilical cord blood EPCs form normal-functioning blood vessels that last for more than 4 months. These vessels exhibit normal blood flow, perm-selectivity to macromolecules, and induction of leukocyte-endothelial interactions in response to cytokine activation similar to normal vessels. Thus, umbilical cord blood EPCs hold great therapeutic potential, and their use should be pursued for vascular engineering.


Assuntos
Vasos Sanguíneos/citologia , Células Endoteliais/citologia , Sangue Fetal/citologia , Células-Tronco Hematopoéticas/citologia , Adulto , Linhagem Celular , Permeabilidade da Membrana Celular , Proliferação de Células , Separação Celular , Humanos , Fatores de Tempo
14.
Nat Biotechnol ; 25(3): 317-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17322871

RESUMO

We describe the differentiation of human embryonic stem (hES) cells into endothelial cells using a scalable two-dimensional method that avoids an embryoid-body intermediate. After transplantation into severe combined immunodeficient (SCID) mice, the differentiated cells contributed to arborized blood vessels that integrated into the host circulatory system and served as blood conduits for 150 d.


Assuntos
Vasos Sanguíneos/citologia , Vasos Sanguíneos/crescimento & desenvolvimento , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células Endoteliais/citologia , Animais , Células Endoteliais/fisiologia , Humanos , Camundongos , Camundongos SCID , Gravação em Vídeo
15.
Stem Cell Rev ; 2(3): 185-201, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17625255

RESUMO

The ability of stem cells to self-renew and to replace mature cells is fundamental to ontogeny and tissue regeneration. Stem cells of the adult organism can be categorized as mono-, bi-, or multipotent, based on the number of mature cell types to which they can give rise. In contrast, pluripotent stem cells of the early embryo have the ability to form every cell type of the adult body. Permanent lines of pluripotent stem cells have been derived from preimplantation embryos (embryonic stem cells), fetal primordial germ cells (embryonic germ cells), and malignant teratocarcinomas (embryonal carcinoma cells). Cultured pluripotent stem cells can easily be manipulated genetically, and they can be matured into adult-type stem cells and terminally differentiated cell types in vitro, thereby, providing powerful model systems for the study of mammalian embryogenesis and disease processes. In addition, human embryonic stem cell lines hold great promise for the development of novel regenerative therapies. To fully utilize the potential of these cells, we must first understand the mechanisms that control pluripotent stem cell fate and function. In recent decades, the microenvironment or niche has emerged as particularly critical for stem cell regulation. In this article, we review how pluripotent stem cell signal transduction mechanisms and transcription factor circuitries integrate information provided by the microenvironment. In addition, we consider the potential existence and location of adult pluripotent stem cell niches, based on the notion that a revealing feature indicating the presence of stem cells in a given tissue is the occurrence of tumors whose characteristics reflect the normal developmental potential of the cognate stem cells.


Assuntos
Células-Tronco Pluripotentes/citologia , Animais , Células-Tronco Embrionárias/citologia , Matriz Extracelular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Neoplasias Embrionárias de Células Germinativas/patologia , Fatores de Transcrição/metabolismo
16.
Stem Cells ; 23(3): 299-305, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15749924

RESUMO

RNA interference methodology suppresses gene expression, thus mimicking loss-of-function mutation and enabling in vitro and in vivo gene function analysis. In this study, we used retroviral and lentiviral vectors to deliver small interfering RNAs and report high-efficiency silencing of a green fluorescent protein (GFP) trans gene and the stem cell-specific transcription factors Oct4/POU5F1 and Nanog in human embryonic stem cells. Gene knockdown of Oct4 and Nanog promotes differentiation, thereby demonstrating a role for these factors in human embryonic stem cell self-renewal.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco Pluripotentes/fisiologia , Interferência de RNA , Fatores de Transcrição/fisiologia , Antígenos de Superfície , Diferenciação Celular/fisiologia , Linhagem Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento , Vetores Genéticos/genética , Subunidade alfa de Hormônios Glicoproteicos/genética , Glicoproteínas/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Humanos , Lentivirus/genética , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas , RNA Interferente Pequeno/genética , Retroviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Albumina Sérica/genética , Fatores de Transcrição/genética , Transfecção
17.
Stem Cells ; 22(5): 770-8, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15342941

RESUMO

Murine embryonic stem (mES) cells remain undifferentiated in the presence of leukemia inhibitory factor (LIF), and activation of signal transducer and activator of transcription 3 (STAT3) via LIF receptor (LIFR) signaling appears sufficient for maintenance of mES cell pluripotency. Anecdotal and contradictory accounts exist for the action of LIF in the culture of human embryonic stem cells, and the nature of LIF signaling and whether the LIF-STAT3 pathway is conserved in human embryonic stem cells (hESCs) has not been systematically explored. In this study, we show that the LIFRbeta and the signaling subunit gp130 are expressed in hESCs and that human LIF can induce STAT3 phosphorylation and nuclear translocation in hESCs. Nevertheless, despite the functional activation of the LIF-STAT3 signaling pathway, human LIF is unable to maintain the pluripotent state of hESCs. Feeder-free culture conditions that maintain hESCs in an undifferentiated state do not show activation of STAT3, suggesting that distinct signaling mechanisms govern the self-renewal of hESCs.


Assuntos
Divisão Celular/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas/metabolismo , Transplante de Células-Tronco/métodos , Transativadores/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/genética , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Receptor gp130 de Citocina , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Interleucina-6 , Fator Inibidor de Leucemia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/imunologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas/genética , Proteínas/farmacologia , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores de OSM-LIF , Fator de Transcrição STAT3 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transativadores/genética
18.
Hematol J ; 3(3): 153-6, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12111652

RESUMO

INTRODUCTION: Disseminated intravascular coagulation (DIC) is a rare event in acute lymphoblastic leukemia (ALL). However, it has been described in a few cases of pre-B ALL with translocation t(17;19)(q22;p13) which results in the fusion of E2A gene with sequences of HLF gene. Here, we report a case of pre-B ALL with DIC and an apparently normal karyotype by R banding. MATERIALS AND METHODS: Fluorescent in situ hybridization (FISH) studies were performed on bone marrow cells from the patient at presentation and after three months of therapy. RT-PCR was used to detect the E2A-HLF transcript. The type of rearrangement was characterized by sequencing. RESULTS: The t(17;19)(q22;p13) was detected by FISH analysis. The fusion E2A-HLF was amplified by RT-PCR and sequenced, giving a type I rearrangement with a long insertion (146 nucleotides) between E2A exon 13 and HLF exon 4. CONCLUSION: While translocation t(17;19) is undetectable by R-banding technique, it can be detected with FISH and amplified with RT-PCR. Therefore, systematic molecular investigations should be conducted for all patients with pre-B ALL associated with DIC, in order to appreciate the incidence and the prognostic value of this rare abnormality.


Assuntos
Proteínas de Ligação a DNA/genética , Coagulação Intravascular Disseminada/genética , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicações , Criança , Cromossomos Humanos Par 17 , Cromossomos Humanos Par 19 , Coagulação Intravascular Disseminada/etiologia , Evolução Fatal , Feminino , Humanos , Cariotipagem , Leucemia-Linfoma Linfoblástico de Células Precursoras B/complicações , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Fatores de Transcrição , Translocação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA