Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 30(12): 3570-3586, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36348622

RESUMO

Recombinant adeno-associated virus (rAAV) vectors are often produced in HEK293 or Spodoptera frugiperda (Sf)-based cell lines. We compared expression profiles of "oversized" (∼5,000 bp) and "standard-sized" (4,600 bp) rAAV5-human α1-antitrypsin (rAAV5-hA1AT) vectors manufactured in HEK293 or Sf cells and investigated molecular mechanisms mediating expression decline. C57BL/6 mice received 6 × 1013 vg/kg of vector, and blood and liver samples were collected through week 57. For all vectors, peak expression (weeks 12-24) declined by 50% to week 57. For Sf- and HEK293-produced oversized vectors, serum hA1AT was initially comparable, but in weeks 12-57, Sf vectors provided significantly higher expression. For HEK293 oversized vectors, liver genomes decreased continuously through week 57 and significantly correlated with A1AT protein. In RNA-sequencing analysis, HEK293 vector-treated mice had significantly higher inflammatory responses in liver at 12 weeks compared with Sf vector- and vehicle-treated mice. Thus, HEK293 vector genome loss led to decreased transgene protein. For Sf-produced vectors, genomes did not decrease from peak expression. Instead, vector genome accessibility significantly decreased from peak to week 57 and correlated with transgene RNA. Vector DNA interactions with active histone marks (H3K27ac/H3K4me3) were significantly reduced from peak to week 57, suggesting that epigenetic regulation impacts transgene expression of Sf-produced vectors.


Assuntos
Epigênese Genética , Insetos , Humanos , Camundongos , Animais , Células HEK293 , Camundongos Endogâmicos C57BL , RNA , Mamíferos
2.
Sci Rep ; 7(1): 14285, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-29079788

RESUMO

The Melanoma-associated Antigen gene family (MAGE) generally encodes for tumour antigens. We had identified that one of the MAGE gene members, Mageb16 was highly expressed in undifferentiated murine embryonic stem cells (ESCs). While the role of Mageb16 in stemness and differentiation of pluripotent stem cells is completely unknown, here, in our current study, we have demonstrated that Mageb16 (41 kDa) is distributed in cytosol and/or in surface membrane in undifferentiated ESCs. A transcriptome study performed at  differentiated short hairpin RNA (shRNA)-mediated Mageb16 knockdown (KD) ESCs and scrambled control (SCR) ESCs until a period of 22 days, revealed that Mageb16 KD ESCs mainly differentiated towards cells expressing mesodermal and cardiovascular lineage - gene markers. Gene markers of other mesoderm-oriented biological processes such as adipogenesis, osteogenesis, limb morphogenesis and spermatogenesis were also significantly enriched in the differentiated Mageb16 KD ESCs. The expression levels of contractile genes were higher in differentiated Mageb16 KD ESCs when compared to differentiated SCR and wild ESCs, suggesting a higher cardiomyogenic potential of Mageb16 depleted ESCs. Further analysis indicates  that regulative epigenetic networks and nucleocytoplasmic modifications induced by the depletion of Mageb16, may play a probable role in differentiation.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Neoplasias/deficiência , Células-Tronco Pluripotentes/metabolismo , Animais , Antígenos de Neoplasias/genética , Membrana Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Proteínas de Neoplasias/genética , Células-Tronco Pluripotentes/citologia , RNA Interferente Pequeno/metabolismo , Transcriptoma
3.
Mol Ther Methods Clin Dev ; 5: 116-129, 2017 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-28480311

RESUMO

The role of striatin interacting protein 2 (Strip2) in differentiation of embryonic stem cells (ESCs) is still under debate. Strip2-silenced murine (KD) ESCs were differentiated for 4, 8, 12, and 16 days. We show that Strip2 is distributed in the perinucleus or nuclei of wild-type (WT) undifferentiated ESCs, but is localized in high-density nuclear bodies in differentiated cells. CellNet analysis of microarray gene expression data for the KD and scrambled control (SCR) embryoid bodies (EBs), as well as immunostainings of key pluripotent factors, demonstrated that differentiation of KD ESCs is repressed. This occurs even in 16-day-old EBs, which possessed a high tumorigenic potential. Correlated with very high expression levels of epigenetic regulator genes, Hat1 and Dnmt3, enzymatic activities of the histone acetyltransferase type B (Hat1) and DNA (cytosine-5)-methyltransferase 3 beta (Dnmt3b) were higher in differentiated 16-day-old KD EBs than in SCR or WT EBs. The expression levels of let-7, 290, and 302 microRNA families were opposed in KD ESCs, while KD EBs had levels comparable to WT and SCR ESCs during differentiation. Strip2 is critical for the regular differentiation of ESCs. Moreover, Strip2 deficient ESCs showed a dysregulation of epigenetic regulators and microRNAs regulating pluripotency.

4.
Stem Cell Rev Rep ; 8(1): 116-27, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21617963

RESUMO

A potential application of embryonic and inducible pluripotent stem cells for the therapy of degenerative diseases involves pure somatic cells, free of tumorigenic undifferentiated embryonic and inducible pluripotent stem cells. In complex collections of chemicals with pharmacological potential we expect to find molecules able to induce specific pluripotent stem cell death, which could be used in some cell therapy settings to eliminate undifferentiated cells. Therefore, we have screened a chemical library of 1120 small chemicals to identify compounds that induce specifically apoptotic cell death in undifferentiated mouse embryonic stem cells (ESCs). Interestingly, three compounds currently used as clinically approved drugs, nortriptyline, benzethonium chloride and methylbenzethonium chloride, induced differential effects in cell viability in ESCs versus mouse embryonic fibroblasts (MEFs). Nortriptyline induced apoptotic cell death in MEFs but not in ESCs, whereas benzethonium and methylbenzethonium chloride showed the opposite effect. Nortriptyline, a tricyclic antidepressant, has also been described as a potent inhibitor of mitochondrial permeability transition, one of two major mechanisms involved in mitochondrial membrane permeabilization during apoptosis. Benzethonium chloride and methylbenzethonium chloride are quaternary ammonium salts used as antimicrobial agents with broad spectrum and have also been described as anticancer agents. A similar effect of benzethonium chloride was observed in human induced pluripotent stem cells (hiPSCs) when compared to both primary human skin fibroblasts and an established human fibroblast cell line. Human fibroblasts and hiPSCs were similarly resistant to nortriptyline, although with a different behavior. Our results indicate differential sensitivity of ESCs, hiPSCs and fibroblasts to certain chemical compounds, which might have important applications in the stem cell-based therapy by eliminating undifferentiated pluripotent stem cells from stem cell-derived somatic cells to prevent tumor formation after transplantation for therapy of degenerative diseases.


Assuntos
Apoptose/efeitos dos fármacos , Citotoxinas/farmacologia , Células-Tronco Pluripotentes/fisiologia , Animais , Benzetônio/análogos & derivados , Benzetônio/farmacologia , Caspase 3/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/fisiologia , Humanos , Dose Letal Mediana , Camundongos , Nortriptilina/farmacologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Bibliotecas de Moléculas Pequenas
5.
Stem Cell Rev Rep ; 8(1): 43-54, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21519850

RESUMO

The vertebrate early stage embryo is consisting of the three primary germ layers ectoderm, mesoderm and endoderm, from which all organ tissues are developed. During early embryonic development, mesodermal cells become sequentially determined to more precisely defined cell types including muscle, heart, vasculature, blood, kidney, gonads, dermis and cartilage. How the prospective mesodermal cells integrate the various signals they receive and how they resolve this information to regulate their morphogenetic behavior and cell fate decisions is largely unknown. Understanding of this complex phenomenon is essential to induce selective differentiation of pluripotent stem cells into clinically relevant, physiologically functional cells such as cardiomyocytes or for transdifferentiation of easily accessible cell types such as fibroblasts into other clinically relevant cell types for applications such as cell replacement therapy, accelerated drug discovery and drug toxicological testing. This demands an in-depth analysis of the mesodermal endogenous signaling cascades and transcription factor networks. Emerging results from isolation and transcriptome characterization of pure mesodermal cells derived from murine embryonic stem cells define the genetic and cellular identity of mesodermal cells and allows a comprehensive analysis of the very dynamic process of mesodermal patterning which would not be technically feasible with conventional embryology methods.This review focuses on defining the transcriptomic signatures of mesodermal cells and their lineages with special reference to the molecular and signaling pathways associated with the complex process of mesodermal patterning.


Assuntos
Células-Tronco Embrionárias/fisiologia , Mesoderma/citologia , Actinas/genética , Actinas/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Humanos , Cadeias Pesadas de Miosina/genética , Cadeias Pesadas de Miosina/metabolismo , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Transdução de Sinais , Transplante de Células-Tronco , Transcriptoma
6.
PLoS One ; 5(10): e13446, 2010 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-20975998

RESUMO

BACKGROUND: Initial specification of cardiomyocytes in the mouse results from interactions between the extraembryonic anterior visceral endoderm (AVE) and the nascent mesoderm. However the mechanism by which AVE activates cardiogenesis is not well understood, and the identity of specific cardiogenic factors in the endoderm remains elusive. Most mammalian studies of the cardiogenic potential of the endoderm have relied on the use of cell lines that are similar to the heart-inducing AVE. These include the embryonal-carcinoma-derived cell lines, END2 and PYS2. The recent development of protocols to isolate eXtraembryonic ENdoderm (XEN) stem cells, representing the extraembryonic endoderm lineage, from blastocyst stage mouse embryos offers new tools for the genetic dissection of cardiogenesis. METHODOLOGY/PRINCIPAL FINDINGS: Here, we demonstrate that XEN cell-conditioned media (CM) enhances cardiogenesis during Embryoid Body (EB) differentiation of mouse embryonic stem (ES) cells in a manner comparable to PYS2-CM and END2-CM. Addition of CM from each of these three cell lines enhanced the percentage of EBs that formed beating areas, but ultimately, only XEN-CM and PYS2-CM increased the total number of cardiomyocytes that formed. Furthermore, our observations revealed that both contact-independent and contact-dependent factors are required to mediate the full cardiogenic potential of the endoderm. Finally, we used gene array comparison to identify factors in these cell lines that could mediate their cardiogenic potential. CONCLUSIONS/SIGNIFICANCE: These studies represent the first step in the use of XEN cells as a molecular genetic tool to study cardiomyocyte differentiation. Not only are XEN cells functionally similar to the heart-inducing AVE, but also can be used for the genetic dissection of the cardiogenic potential of AVE, since they can be isolated from both wild type and mutant blastocysts. These studies further demonstrate the importance of both contact-dependent and contact-independent factors in cardiogenesis and identify potential heart-inducing proteins in the endoderm.


Assuntos
Células-Tronco Embrionárias/citologia , Endoderma/citologia , Coração/embriologia , Animais , Diferenciação Celular , Linhagem Celular , Meios de Cultivo Condicionados , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
7.
PLoS One ; 5(8): e12016, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20711519

RESUMO

Prior to gastrulation in the mouse, all endodermal cells arise from the primitive endoderm of the blastocyst stage embryo. Primitive endoderm and its derivatives are generally referred to as extra-embryonic endoderm (ExEn) because the majority of these cells contribute to extra-embryonic lineages encompassing the visceral endoderm (VE) and the parietal endoderm (PE). During gastrulation, the definitive endoderm (DE) forms by ingression of cells from the epiblast. The DE comprises most of the cells of the gut and its accessory organs. Despite their different origins and fates, there is a surprising amount of overlap in marker expression between the ExEn and DE, making it difficult to distinguish between these cell types by marker analysis. This is significant for two main reasons. First, because endodermal organs, such as the liver and pancreas, play important physiological roles in adult animals, much experimental effort has been directed in recent years toward the establishment of protocols for the efficient derivation of endodermal cell types in vitro. Conversely, factors secreted by the VE play pivotal roles that cannot be attributed to the DE in early axis formation, heart formation and the patterning of the anterior nervous system. Thus, efforts in both of these areas have been hampered by a lack of markers that clearly distinguish between ExEn and DE. To further understand the ExEn we have undertaken a comparative analysis of three ExEn-like cell lines (END2, PYS2 and XEN). PYS2 cells are derived from embryonal carcinomas (EC) of 129 strain mice and have been characterized as parietal endoderm-like [1], END2 cells are derived from P19 ECs and described as visceral endoderm-like, while XEN cells are derived from blastocyst stage embryos and are described as primitive endoderm-like. Our analysis suggests that none of these cell lines represent a bona fide single in vivo lineage. Both PYS2 and XEN cells represent mixed populations expressing markers for several ExEn lineages. Conversely END2 cells, which were previously characterized as VE-like, fail to express many markers that are widely expressed in the VE, but instead express markers for only a subset of the VE, the anterior visceral endoderm. In addition END2 cells also express markers for the PE. We extended these observations with microarray analysis which was used to probe and refine previously published data sets of genes proposed to distinguish between DE and VE. Finally, genome-wide pathway analysis revealed that SMAD-independent TGFbeta signaling through a TAK1/p38/JNK or TAK1/NLK pathway may represent one mode of intracellular signaling shared by all three of these lines, and suggests that factors downstream of these pathways may mediate some functions of the ExEn. These studies represent the first step in the development of XEN cells as a powerful molecular genetic tool to study the endodermal signals that mediate the important developmental functions of the extra-embryonic endoderm. Our data refine our current knowledge of markers that distinguish various subtypes of endoderm. In addition, pathway analysis suggests that the ExEn may mediate some of its functions through a non-classical MAP Kinase signaling pathway downstream of TAK1.


Assuntos
Endoderma/citologia , Animais , Biomarcadores/metabolismo , Linhagem Celular , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Perfilação da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos
8.
BMC Genomics ; 10: 73, 2009 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-19203379

RESUMO

BACKGROUND: Mouse embryonic stem (ES) cells remain pluripotent in vitro when grown in the presence of the cytokine Leukaemia Inhibitory Factor (LIF). Identification of LIF targets and of genes regulating the transition between pluripotent and early differentiated cells is a critical step for understanding the control of ES cell pluripotency. RESULTS: By gene profiling studies carried out with mRNAs from ES cells and their early derivatives treated or not with LIF, we have identified i) LIF-dependent genes, highly expressed in pluripotent cells, whose expression level decreases sharply upon LIF withdrawal [Pluri genes], ii) LIF induced genes [Lifind genes] whose expression is differentially regulated depending upon cell context and iii) genes specific to the reversible or irreversible committed states. In addition, by hierarchical gene clustering, we have identified, among eight independent gene clusters, two atypical groups of genes, whose expression level was highly modulated in committed cells only. Computer based analyses led to the characterization of different sub-types of Pluri and Lifind genes, and revealed their differential modulation by Oct4 or Nanog master genes. Individual knock down of a selection of Pluri and Lifind genes leads to weak changes in the expression of early differentiation markers, in cell growth conditions in which these master genes are still expressed. CONCLUSION: We have identified different sets of LIF-regulated genes depending upon the cell state (reversible or irreversible commitment), which allowed us to present a novel global view of LIF responses. We are also reporting on the identification of genes whose expression is strictly regulated during the commitment step. Furthermore, our studies identify sub-networks of genes with a restricted expression in pluripotent ES cells, whose down regulation occurs while the master knot (composed of OCT4, SOX2 and NANOG) is still expressed and which might be down-regulated together for driving cells towards differentiation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Fator Inibidor de Leucemia/metabolismo , Camundongos/genética , Animais , Diferenciação Celular/genética , Linhagem Celular , Análise por Conglomerados , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco Pluripotentes/metabolismo
9.
Chin J Physiol ; 51(4): 226-9, 2008 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-19112880

RESUMO

Cardiovascular diseases are the leading cause of death globally. The pluripotency and indefinite proliferative capacity of embryonic stem (ES) cells make them a promising candidate for the cell replacement therapy where the damaged cells are replaced by the functional cells derived from stem cells in vitro. Emerging results with human ES cells for the myocardial repair are encouraging, but this approach is still in its infancy and is under extensive investigation. The daily upcoming experimental observations are reinforcing the solid hope that ES cells will be the potential source for use in cell replacement therapy. Although this demands serious considerations ethically and on practical applicability, the newly upcoming discoveries show that the adult human fibroblasts can be reprogrammed to embryonic stem cell like cells (called induced pluripotent stem cells) which can be used for cell replacement therapies. Remarkably, this obviates the need for the embryo destruction and overcomes related immunological problems which are the long time hurdles for the ES cell based cell replacement therapy. This review weighs the actual stand off in the human ES cells based cell replacement therapy for the treatment of cardiovascular degenerative diseases with a special emphasis on the hurdles and challenges to be resolved before the onset of clinical trials and the potential of the recently reported "induced pluripotent stem cells" for their use in cell replacement therapy.


Assuntos
Células-Tronco Embrionárias/citologia , Cardiopatias/terapia , Miócitos Cardíacos/citologia , Transplante de Células-Tronco/tendências , Diferenciação Celular/fisiologia , Humanos
10.
Curr Med Chem ; 13(13): 1481-9, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16787199

RESUMO

Epidemiological studies have repeatedly demonstrated a correlation between nutrition, development and the severity of malignant and non-malignant proliferative diseases such as cancer and atherosclerosis. Therefore, the prevention of chronic proliferative diseases through dietary intervention is currently receiving considerable attention. Until now, much of the research is being focused on the cellular and molecular action mechanisms of dietary small molecules explaining their beneficial effects. Dietary chemicals may affect gene expression in several human diseases. However, significant progress has been made and several molecular action mechanisms have been proposed. Alteration of genetical pathways by nutrition, also called "Nutrigenomics", may offer a new approach for understanding the beneficial effects of dietary compounds on the development of severe polygenic diseases, such as cardiovascular disease, diabetes and hypertension. This review focuses on the nutritional genomics of dietary chemicals with a special emphasis on catechins. Catechins belong to the flavonoid family, which are polyphenolic compounds available in foods of plant origin. Several epidemiological studies have reported that consumption of flavonoids, and especially catechins might function as chemopreventive agents against cancer and cardiovascular diseases.


Assuntos
Regulação da Expressão Gênica , Genômica/métodos , Neoplasias/prevenção & controle , Fenômenos Fisiológicos da Nutrição , Animais , Arildialquilfosfatase/efeitos dos fármacos , Arildialquilfosfatase/genética , Catequina/análogos & derivados , Catequina/farmacologia , Suplementos Nutricionais , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias/tratamento farmacológico , Proteômica/métodos , Transdução de Sinais
11.
J Nutr Biochem ; 16(5): 259-66, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15866224

RESUMO

The prevention of cancer through dietary intervention is currently receiving considerable attention. Several epidemiological studies substantiate that green tea has a protective effect against a variety of malignant proliferative disorders such as lung cancer, breast cancer and prostate cancer. This preventive potential of green tea against cancer is attributed to the biologically active flavonoids called catechins. Epigallocatechin 3-o-gallate, the major catechin found in green tea, mediates diverse physiological and pharmacological actions in bringing about the regression of the tumors and also lowers the risk of nonmalignant cardiovascular proliferative diseases. Much of the current research is being focused on how these catechins specifically bring about the regression of the experimentally induced tumors both in vitro and in vivo. These catechins exert diverse physiological effects against proliferative diseases by several mechanisms, most of which are not completely characterized. This review summarizes the mechanisms by which these catechins play an essential role in regulating the process of carcinogenesis, with a special emphasis on how these catechins antagonize the growth factor-induced proliferative disorders.


Assuntos
Catequina/análogos & derivados , Catequina/uso terapêutico , Substâncias de Crescimento/metabolismo , Neoplasias/prevenção & controle , Animais , Catequina/metabolismo , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/efeitos dos fármacos , Receptores ErbB/fisiologia , Fator de Crescimento Derivado de Plaquetas/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/fisiologia
12.
J Cell Mol Med ; 8(4): 465-73, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15601575

RESUMO

Embryonic stem (ES) cells are revolutionizing the field of developmental biology as a potential tool to understand the molecular mechanisms occurring during the process of differentiation from the embryonic stage to the adult phenotype. ES cells harvested from the inner cell mass (ICM) of the early embryo can proliferate indefinitely in vitro while retaining the ability to differentiate into all somatic cells. Emerging results from mice models with ES cells are promising and raising tremendous hope among the scientific community for the ES-cell based cell replacement therapy (CRT) of various severe diseases. ES cells could potentially revolutionize medicine by providing an unlimited renewable source of cells capable of replacing or repairing tissues that have been damaged in almost all degenerative diseases such as diabetes, myocardial infarction and Parkinson's disease. This review updates the progress of ES cell research in CRT, discusses about the problems encountered in the practical utility of ES cells in CRT and evaluates how far this approach is successful experimentally.


Assuntos
Embrião de Mamíferos/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Linhagem da Célula , Transplante de Células , Diabetes Mellitus/terapia , Humanos , Fígado/patologia , Modelos Biológicos , Infarto do Miocárdio/patologia , Miocárdio/patologia , Doença de Parkinson/terapia , Fenótipo , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA