Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 121(21): e2316006121, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38748577

RESUMO

Blood-brain barrier (BBB) models derived from human stem cells are powerful tools to improve our understanding of cerebrovascular diseases and to facilitate drug development for the human brain. Yet providing stem cell-derived endothelial cells with the right signaling cues to acquire BBB characteristics while also retaining their vascular identity remains challenging. Here, we show that the simultaneous activation of cyclic AMP and Wnt/ß-catenin signaling and inhibition of the TGF-ß pathway in endothelial cells robustly induce BBB properties in vitro. To target this interaction, we present a small-molecule cocktail named cARLA, which synergistically enhances barrier tightness in a range of BBB models across species. Mechanistically, we reveal that the three pathways converge on Wnt/ß-catenin signaling to mediate the effect of cARLA via the tight junction protein claudin-5. We demonstrate that cARLA shifts the gene expressional profile of human stem cell-derived endothelial cells toward the in vivo brain endothelial signature, with a higher glycocalyx density and efflux pump activity, lower rates of endocytosis, and a characteristic endothelial response to proinflammatory cytokines. Finally, we illustrate how cARLA can improve the predictive value of human BBB models regarding the brain penetration of drugs and targeted nanoparticles. Due to its synergistic effect, high reproducibility, and ease of use, cARLA has the potential to advance drug development for the human brain by improving BBB models across laboratories.


Assuntos
Barreira Hematoencefálica , Células Endoteliais , Barreira Hematoencefálica/metabolismo , Humanos , Células Endoteliais/metabolismo , Animais , Via de Sinalização Wnt , Claudina-5/metabolismo , Claudina-5/genética , AMP Cíclico/metabolismo , Camundongos , Células-Tronco/metabolismo , Células-Tronco/citologia , Junções Íntimas/metabolismo , beta Catenina/metabolismo
2.
Genes (Basel) ; 10(11)2019 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-31752312

RESUMO

Separation of germline cells from somatic lineages is one of the earliest decisions of embryogenesis. Genes expressed in germline cells include apoptotic and meiotic factors, which are not transcribed in the soma normally, but a number of testis-specific genes are active in numerous cancer types. During germ cell development, germ-cell-specific genes can be regulated by specific transcription factors, retinoic acid signaling and multimeric protein complexes. Non-canonical polycomb repressive complexes, like ncPRC1.6, play a critical role in the regulation of the activity of germ-cell-specific genes. RING1 and YY1 binding protein (RYBP) is one of the core members of the ncPRC1.6. Surprisingly, the role of Rybp in germ cell differentiation has not been defined yet. This review is focusing on the possible role of Rybp in this process. By analyzing whole-genome transcriptome alterations of the Rybp-/- embryonic stem (ES) cells and correlating this data with experimentally identified binding sites of ncPRC1.6 subunits and retinoic acid receptors in ES cells, we propose a model how germ-cell-specific transcription can be governed by an RYBP centered regulatory network, underlining the possible role of RYBP in germ cell differentiation and tumorigenesis.


Assuntos
Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Modelos Genéticos , Proteínas Repressoras/metabolismo , Transcrição Gênica , Animais , Sítios de Ligação , Carcinogênese/genética , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Células-Tronco Embrionárias/patologia , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Redes Reguladoras de Genes , Humanos , Neoplasias Embrionárias de Células Germinativas/genética , Neoplasias Embrionárias de Células Germinativas/patologia , Complexo Repressor Polycomb 1/metabolismo , RNA-Seq , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/genética
3.
Stem Cells Int ; 2016: 4034620, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26788067

RESUMO

Rybp (Ring1 and Yy1 Binding Protein) is a transcriptional regulator and member of the noncanonical polycomb repressive complex 1 with essential role in early embryonic development. We have previously described that alteration of Rybp dosage in mouse models induced striking neural tube defects (NTDs), exencephaly, and disorganized neurocortex. In this study we further investigated the role of Rybp in neural differentiation by utilising wild type (rybp (+/+)) and rybp null mutant (rybp (-/-)) embryonic stem cells (ESCs) and tried to uncover underlying molecular events that are responsible for the observed phenotypic changes. We found that rybp null mutant ESCs formed less matured neurons, astrocytes, and oligodendrocytes from existing progenitors than wild type cells. Furthermore, lack of rybp coincided with altered gene expression of key neural markers including Pax6 and Plagl1 pinpointing a possible transcriptional circuit among these genes.

4.
Exp Neurol ; 269: 188-201, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25889458

RESUMO

Human plexus injuries often include the avulsion of one or more ventral roots, resulting in debilitating conditions. In this study the effects of undifferentiated murine iPSCs on damaged motoneurons were investigated following avulsion of the lumbar 4 (L4) ventral root, an injury known to induce the death of the majority of the affected motoneurons. Avulsion and reimplantation of the L4 ventral root (AR procedure) were accompanied by the transplantation of murine iPSCs into the injured spinal cord segment in rats. Control animals underwent ventral root avulsion and reimplantation, but did not receive iPSCs. The grafted iPSCs induced an improved reinnervation of the reimplanted ventral root by the host motoneurons as compared with the controls (number of retrogradely labeled motoneurons: 503 ± 38 [AR+iPSCs group] vs 48 ± 6 [controls, AR group]). Morphological reinnervation resulted in a functional recovery, i.e. the grafted animals exhibited more motor units in their reinnervated hind limb muscles, which produced a greater force than that in the controls (50 ± 2.1% vs 11.9 ± 4.2% maximal tetanic tension [% ratio of operated/intact side]). Grafting of undifferentiated iPSCs downregulated the astroglial activation within the L4 segment. The grafted cells differentiated into neurons and astrocytes in the injured cord. The grafted iPSCs, host neurons and glia were found to produce the cytokines and neurotrophic factors MIP-1a, IL-10, GDNF and NT-4. These findings suggest that, following ventral root avulsion injury, iPSCs are able to induce motoneuron survival and regeneration through combined neurotrophic and cytokine modulatory effects.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Neurônios Motores/citologia , Regeneração Nervosa/fisiologia , Raízes Nervosas Espinhais/lesões , Animais , Morte Celular , Sobrevivência Celular/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Camundongos , Fatores de Crescimento Neural/metabolismo , Ratos , Recuperação de Função Fisiológica/fisiologia , Medula Espinal/citologia
5.
Mol Genet Metab ; 114(4): 547-56, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25669595

RESUMO

POLYCOMB group (PCG) proteins belong to the family of epigenetic regulators of genes playing important roles in differentiation and development. Mutants of PcG genes were isolated first in the fruit fly, Drosophila melanogaster, resulting in spectacular segmental transformations due to the ectopic expression of homeotic genes. Homologs of Drosophila PcG genes were also identified in plants and in vertebrates and subsequent experiments revealed the general role of PCG proteins in the maintenance of the repressed state of chromatin through cell divisions. The past decades of gene targeting experiments have allowed us to make significant strides towards understanding how the network of PCG proteins influences multiple aspects of cellular fate determination during development. Being involved in the transmission of specific expression profiles of different cell lineages, PCG proteins were found to control wide spectra of unrelated epigenetic processes in vertebrates, such as stem cell plasticity and renewal, genomic imprinting and inactivation of X-chromosome. PCG proteins also affect regulation of metabolic genes being important for switching programs between pluripotency and differentiation. Insight into the precise roles of PCG proteins in normal physiological processes has emerged from studies employing cell culture-based systems and genetically modified animals. Here we summarize the findings obtained from PcG mutant fruit flies and mice generated to date with a focus on PRC1 and PRC2 members altered by nucleotide substitutions resulting in specific alleles. We also include a compilation of lessons learned from these models about the in vivo functions of this complex protein family. With multiple knockout lines, sophisticated approaches to study the consequences of peculiar missense point mutations, and insights from complementary gain-of-function systems in hand, we are now in a unique position to significantly advance our understanding of the molecular basis of in vivo functions of PcG proteins.


Assuntos
Drosophila melanogaster/genética , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/fisiologia , Alelos , Animais , Diferenciação Celular , Cromatina/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Epigênese Genética , Regulação da Expressão Gênica , Genes Homeobox , Camundongos , Nucleotídeos/genética , Mutação Puntual
6.
Tissue Eng Part C Methods ; 20(5): 383-92, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24006854

RESUMO

Somatic cell reprogramming has generated enormous interest after the first report by Yamanaka and his coworkers in 2006 on the generation of induced pluripotent stem cells (iPSCs) from mouse fibroblasts. Here we report the generation of stable iPSCs from mouse fibroblasts by recombinant protein transduction (Klf4, Oct4, Sox2, and c-Myc), a procedure designed to circumvent the risks caused by integration of exogenous sequences in the target cell genome associated with gene delivery systems. The recombinant proteins were fused in the frame to the glutathione-S-transferase tag for affinity purification and to the transactivator transcription-nuclear localization signal polypeptide to facilitate membrane penetration and nuclear localization. We performed the reprogramming procedure on embryonic fibroblasts from inbred (C57BL6) and outbred (ICR) mouse strains. The cells were treated with purified proteins four times, at 48-h intervals, and cultured on mitomycin C treated mouse embryonic fibroblast (MEF) cells in complete embryonic stem cell (ESC) medium until colonies formed. The iPSCs generated from the outbred fibroblasts exhibited similar morphology and growth properties to ESCs and were sustained in an undifferentiated state for more than 20 passages. The cells were checked for pluripotency-related markers (Oct4, Sox2, Klf4, cMyc, Nanog) by immunocytochemistry and by reverse transcription-polymerase chain reaction. The protein iPSCs (piPSCs) formed embryoid bodies and subsequently differentiated towards all three germ layer lineages. Importantly, the piPSCs could incorporate into the blastocyst and led to variable degrees of chimerism in newborn mice. These data show that recombinant purified cell-penetrating proteins are capable of reprogramming MEFs to iPSCs. We also demonstrated that the cells of the generated cell line satisfied all the requirements of bona fide mouse ESCs: form round colonies with defined boundaries; have a tendency to attach together with high nuclear/cytoplasmic ratio; express key pluripotency markers; and are capable of in vitro differentiation into ecto-, endo-, and mesoderm, and in vivo chimera formation.


Assuntos
Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Recombinantes/metabolismo , Transdução Genética/métodos , Animais , Diferenciação Celular/genética , Quimera/metabolismo , Regulação da Expressão Gênica , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Camundongos , Proteínas Recombinantes/isolamento & purificação , Reprodutibilidade dos Testes , Fatores de Transcrição/metabolismo
7.
Cell Reprogram ; 15(5): 443-58, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24020697

RESUMO

Embryonic stem cells (ESCs) have the ability to form aggregates, which are called embryoid bodies (EBs). EBs mimic early embryonic development and are commonly produced for cardiomyogenesis. Here, we describe a method of EB formation in hydrodynamic conditions using a slow-turning lateral vessel (STLV) bioreactor and the subsequent differentiation of EBs into cardiomyocytes. EBs formed in the STLV were compared with conventional techniques, such as hanging drop (HD) or static suspension cell culture (SSC), for homogeneity of EB size, shape, proliferation, apoptosis, and in vitro cardiac differentiation. After 3 days of culture, a four-fold improvement in the yield of EB formation/mL, a six-fold enhancement in total yield of EB/mL, and a nearly 10-fold reduction of cells that failed to incorporate into EBs were achieved in STLV versus SSC. During cardiac differentiation, a 1.5- to 4.2-fold increase in the area of cardiac troponin T (cTnT) per single EB in STLV versus SSC and HD was achieved. These results demonstrate that the STLV method improves the quality and quantity of ES cells to form EBs and enhances the efficiency of cardiac differentiation. We have demonstrated that the mechanical method of cell differentiation creates different microenvironments for the cells and thus influences their lineage commitments, even when genetic origin and the culture medium are the same. Ascorbic acid (ASC) improved further cardiac commitment in differentiation assays. Hence, this culture system is suitable for the production of large numbers of cells for clinical cell replacement therapies and industrial drug testing applications.


Assuntos
Reatores Biológicos , Diferenciação Celular , Corpos Embrioides/citologia , Células-Tronco Embrionárias/citologia , Coração/embriologia , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
BMC Dev Biol ; 13: 33, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23971992

RESUMO

BACKGROUND: Velo-cardio-facial syndrome/DiGeorge syndrome (VCFS/DGS) is caused by a 1.5-3 Mb microdeletion of chromosome 22q11.2, frequently referred to as 22q11.2 deletion syndrome (22q11DS). This region includes TBX1, a T-box transcription factor gene that contributes to the etiology of 22q11DS. The requirement for TBX1 in mammalian development is dosage-sensitive, such that loss-of-function (LOF) and gain-of-function (GOF) of TBX1 in both mice and humans results in disease relevant congenital malformations. RESULTS: To further gain insight into the role of Tbx1 in development, we have targeted the Rosa26 locus to generate a new GOF mouse model in which a Tbx1-GFP fusion protein is expressed conditionally using the Cre/LoxP system. Tbx1-GFP expression is driven by the endogenous Rosa26 promoter resulting in ectopic and persistent expression. Tbx1 is pivotal for proper ear and heart development; ectopic activation of Tbx1-GFP in the otic vesicle by Pax2-Cre and Foxg1-Cre represses neurogenesis and produces morphological defects of the inner ear. Overexpression of a single copy of Tbx1-GFP using Tbx1Cre/+ was viable, while overexpression of both copies resulted in neonatal lethality with cardiac outflow tract defects. We have partially rescued inner ear and heart anomalies in Tbx1Cre/- null embryos by expression of Tbx1-GFP. CONCLUSIONS: We have generated a new mouse model to conditionally overexpress a GFP-tagged Tbx1 protein in vivo. This provides a useful tool to investigate in vivo direct downstream targets and protein binding partners of Tbx1.


Assuntos
Orelha/embriologia , Coração/embriologia , Modelos Animais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Animais , Orelha/patologia , Embrião de Mamíferos , Dosagem de Genes , Proteínas de Fluorescência Verde/metabolismo , Camundongos , RNA não Traduzido/genética , Proteínas Recombinantes de Fusão/metabolismo
9.
Exp Cell Res ; 318(19): 2482-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22846649

RESUMO

Induced pluripotent stem (iPS) cell technology involves reprogramming somatic cells to a pluripotent state. The original technology used to produce these cells requires viral gene transduction and results in the permanent integration of exogenous genes into the genome. This can lead to the development of abnormalities in the derived iPS cells. Here, we report that non-viral transfection of a Sleeping Beauty (SB) transposon containing the coding sequences Oct3/4 (Pouf1), Sox-2, Klf-4 and c-Myc (OSKM) linked with 2A peptides, can reprogram mouse fibroblasts. We have established reprogrammed mouse cell lines from three different genetic backgrounds: (1) ICR-outbred, (2) C57BL/6-inbred and (3) F1-hybrid (C57BL/6 x DBA/2J), with parallel robust expression of all exogenous (Oct3/4, Sox-2, Klf-4, and c-Myc) and endogenous (e.g. Oct3/4 and Nanog) pluripotency genes. The iPS cell lines exhibited characteristics typical for undifferentiated embryonic stem (ES) cell lines: ES cell-like morphology, alkaline phosphatase (ALP) positivity and gene expression pattern (shown by reverse transcription PCR, and immunofluorescence of ES cell markers-e.g. Oct3/4, SSEA1, Nanog). Furthermore, cells were able to form embryoid bodies (EBs), to beat rhythmically, and express cardiac (assayed by immunofluorescence, e.g. cardiac Troponin T, desmin) and neuronal (assayed by immunofluorescence e.g. nestin, Tuj1) markers. The in vitro differentiation potential was found to be the highest in the ICR-derived iPS lines (ICR-iPS). Interestingly, the ICR-iPS lines had even higher differentiation potential than the ICR-ES cell lines: the rate of EBs forming rhythmically beating cardiomyocytes was 4% in ICR-ES and 79% in ICR-iPS cells, respectively. In vivo, the ICR and F1 hybrid iPS cells formed chimeras and one of the iPS cells from the F1 hybrid background transmitted to the germline. Our results suggest that iPS technology may be useful for generating pluripotent stem cells from genetic backgrounds of which good quality ES cell generation is difficult. These studies provide insights into viral-free iPS technology and may contribute towards defining future cell-based therapies, drug-screening methods and production of transgenic animals using genetically modified iPS cells.


Assuntos
Elementos de DNA Transponíveis/genética , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Transfecção/métodos , Fosfatase Alcalina/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos ICR , Miócitos Cardíacos/metabolismo
10.
Cell Reprogram ; 14(5): 390-7, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22917491

RESUMO

Mouse embryonic stem cells (ESCs) and induced pluripotent stem (iPS) cells can be used as models of neuronal differentiation for the investigation of mammalian neurogenesis, pharmacological testing, and development of cell-based therapies. Recently, mouse iPS cell lines have been generated by Sleeping Beauty (SB) transposon-mediated transgenesis (SB-iPS). In this study, we determined for the first time the differentiation potential of mouse SB-iPS cells to form neuronal progenitor cells (NPCs) and neurons. Undifferentiated SB-iPS and ES cells were aggregated into embryoid bodies (EBs) and cultured in neuronal differentiation medium supplemented with 5 µM all-trans retinoic acid. Thereafter, EBs were dissociated and plated to observe further neuronal differentiation. Samples were fixed on days 10 and 14 for immunocytochemistry staining using the NPC markers Pax6 and Nestin and the neuron marker ßIII-tubulin/Tuj1. Nestin-labeled cells were analyzed further by flow cytometry. Our results demonstrated that SB-iPS cells can generate NPCs and differentiate further into neurons in culture, although SB-iPS cells produced less nestin-positive cells than ESCs (6.12 ± 1.61 vs. 74.36 ± 1.65, respectively). In conclusion, the efficiency of generating SB-iPS cells-derived NPCs needs to be improved. However, given the considerable potential of SB-iPS cells for drug testing and as therapeutic models in neurological disorders, continuing investigation of their neuronal differentiation ability is required.


Assuntos
Elementos de DNA Transponíveis , Neurônios/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco/citologia , Animais , Diferenciação Celular , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Neurogênese
11.
Biotechnol Lett ; 33(8): 1565-73, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21476093

RESUMO

Embryoid body (EB) formation is a common intermediate during in vitro differentiation of pluripotent stem cells into specialized cell types. We have optimized the slow-turning, lateral vessel (STLV) for large scale and homogenous EB production from mouse embryonic stem cells. The effects of inoculating different cell numbers, time of EB adherence to gelatin-coated dishes, and rotation speed for optimal EB formation and cardiac differentiation were investigated. Using 3 × 10(5) cells/ml, 10 rpm rotary speed and plating of EBs onto gelatin-coated surfaces three days after culture, were the best parameters for optimal size and EB quality on consequent cardiac differentiation. These optimized parameters enrich cardiac differentiation in ES cells when using the STLV method.


Assuntos
Reatores Biológicos , Técnicas de Cultura de Células/instrumentação , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Miócitos Cardíacos/citologia , Animais , Adesão Celular/fisiologia , Técnicas de Cultura de Células/métodos , Diferenciação Celular/genética , Tamanho Celular , Corpos Embrioides/citologia , Células-Tronco Embrionárias/metabolismo , Perfilação da Expressão Gênica , Marcadores Genéticos , Camundongos , Miócitos Cardíacos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Epigenetics Chromatin ; 3(1): 21, 2010 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-21118511

RESUMO

BACKGROUND: Brahma-related gene 1 (Brg1, also known as Smarca4 and Snf2ß) encodes an adenosine-5'-triphosphate (ATP)-dependent catalytical subunit of the (switch/sucrose nonfermentable) (SWI/SNF) chromatin remodeling complexes. SWI/SNF complexes are recruited to chromatin through multiple mechanisms, including specific DNA-binding factors (for example, heat shock transcription factor 4 (Hsf4) and paired box gene 6 (Pax6)), chromatin structural proteins (for example, high-mobility group A1 (HMGA1)) and/or acetylated core histones. Previous studies have shown that a single amino acid substitution (K798R) in the Brg1 ATPase domain acts via a dominant-negative (dn) mechanism. Genetic studies have demonstrated that Brg1 is an essential gene for early (that is, prior implantation) mouse embryonic development. Brg1 also controls neural stem cell maintenance, terminal differentiation of multiple cell lineages and organs including the T-cells, glial cells and limbs. RESULTS: To examine the roles of Brg1 in mouse lens development, a dnBrg1 transgenic construct was expressed using the lens-specific αA-crystallin promoter in postmitotic lens fiber cells. Morphological studies revealed abnormal lens fiber cell differentiation in transgenic lenses resulting in cataract. Electron microscopic studies showed abnormal lens suture formation and incomplete karyolysis (that is, denucleation) of lens fiber cells. To identify genes regulated by Brg1, RNA expression profiling was performed in embryonic day 15.5 (E15.5) wild-type and dnBrg1 transgenic lenses. In addition, comparisons between differentially expressed genes in dnBrg1 transgenic, Pax6 heterozygous and Hsf4 homozygous lenses identified multiple genes coregulated by Brg1, Hsf4 and Pax6. DNase IIß, a key enzyme required for lens fiber cell denucleation, was found to be downregulated in each of the Pax6, Brg1 and Hsf4 model systems. Lens-specific deletion of Brg1 using conditional gene targeting demonstrated that Brg1 was required for lens fiber cell differentiation, for expression of DNase IIß, for lens fiber cell denucleation and indirectly for retinal development. CONCLUSIONS: These studies demonstrate a cell-autonomous role for Brg1 in lens fiber cell terminal differentiation and identified DNase IIß as a potential direct target of SWI/SNF complexes. Brg1 is directly or indirectly involved in processes that degrade lens fiber cell chromatin. The presence of nuclei and other organelles generates scattered light incompatible with the optical requirements for the lens.

13.
Stem Cell Res Ther ; 1(2): 12, 2010 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-20624271

RESUMO

Induced pluripotent stem cells (iPSCs) are novel tools for biomedical research, with a promise for future regenerative medicine applications. Recently, Han and colleagues reported in Nature that T box gene 3 (Tbx3) can improve the quality of mouse iPSCs and increase their germline transmission efficacy. This observation contributes greatly to the improvement of iPSC technology and might be a step towards 'designer' reprogramming strategies by generating high quality iPSCs. Further studies comparing pluripotency regulation in different species, including that in human, will be necessary to verify the universal role of Tbx3 and the medical relevance of the observation.


Assuntos
Reprogramação Celular/genética , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo
14.
BMC Mol Biol ; 10: 88, 2009 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-19732419

RESUMO

BACKGROUND: The POU5F1 gene encodes the octamer-binding transcription factor-4 (Oct4). It is crucial in the regulation of pluripotency during embryonic development and widely used as molecular marker of embryonic stem cells (ESCs). The objective of this study was to identify and to analyse the promoter region of rabbit POU5F1 gene; furthermore to examine its expression pattern in preimplantation stage rabbit embryos. RESULTS: The upstream region of rabbit POU5F1 was subcloned sequenced and four highly conserved promoter regions (CR1-4) were identified. The highest degree of similarity on sequence level was found among the conserved domains between rabbit and human. Among the enhancers the proximal enhancer region (PE-1A) exhibited the highest degree of homology (96.4%). Furthermore, the CR4 regulator domain containing the distal enhancer (DE-2A) was responsible for stem cell-specific expression. Also, BAC library screen revealed the existence of a processed pseudogene of rabbit POU5F1. The results of quantitative real-time PCR experiments showed that POU5F1 mRNA was abundantly present in oocytes and zygotes, but it was gradually reduced until the activation of the embryonic genome, thereafter a continuous increase in POU5F1 mRNA level was observed until blastocyst stage. By using the XYClone laser system the inner cell mass (ICM) and trophoblast portions of embryos were microdissected and examined separately and POU5F1 mRNA was detected in both cell types. CONCLUSION: In this study we provide a comparative sequence analysis of the regulatory region of rabbit POU5F1 gene. Our data suggest that the POU5F1 gene is strictly regulated during early mammalian development. We proposed that the well conserved CR4 region containing the DE-2A enhancer is responsible for the highly conserved ESC specific gene expression. Notably, we are the first to report that the rabbit POU5F1 is not restricted to ICM cells only, but it is expressed in trophoblast cells as well. This information may be well applicable to investigate further the possible phylogenetic role and the regulation of POU5F1 gene.


Assuntos
Blastocisto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fator 3 de Transcrição de Octâmero/genética , Regiões Promotoras Genéticas , Coelhos/genética , Animais , Sequência de Bases , Desenvolvimento Embrionário , Feminino , Humanos , Dados de Sequência Molecular , Fator 3 de Transcrição de Octâmero/metabolismo , Coelhos/embriologia , Coelhos/metabolismo , Homologia de Sequência do Ácido Nucleico
15.
World J Stem Cells ; 1(1): 11-21, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21607103

RESUMO

Embryonic stem (ES) cells have the ability to differentiate into all germ layers, holding great promise not only for a model of early embryonic development but also for a robust cell source for cell-replacement therapies and for drug screening. Embryoid body (EB) formation from ES cells is a common method for producing different cell lineages for further applications. However, conventional techniques such as hanging drop or static suspension culture are either inherently incapable of large scale production or exhibit limited control over cell aggregation during EB formation and subsequent EB aggregation. For standardized mass EB production, a well defined scale-up platform is necessary. Recently, novel scenario methods of EB formation in hydrodynamic conditions created by bioreactor culture systems using stirred suspension systems (spinner flasks), rotating cell culture system and rotary orbital culture have allowed large-scale EB formation. Their use allows for continuous monitoring and control of the physical and chemical environment which is difficult to achieve by traditional methods. This review summarizes the current state of production of EBs derived from pluripotent cells in various culture systems. Furthermore, an overview of high quality EB formation strategies coupled with systems for in vitro differentiation into various cell types to be applied in cell replacement therapy is provided in this review. Recently, new insights in induced pluripotent stem (iPS) cell technology showed that differentiation and lineage commitment are not irreversible processes and this has opened new avenues in stem cell research. These cells are equivalent to ES cells in terms of both self-renewal and differentiation capacity. Hence, culture systems for expansion and differentiation of iPS cells can also apply methodologies developed with ES cells, although direct evidence of their use for iPS cells is still limited.

16.
World J Stem Cells ; 1(1): 22-9, 2009 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-21607104

RESUMO

In mice, gene targeting by homologous recombination continues to play an essential role in the understanding of functional genomics. This strategy allows precise location of the site of transgene integration and is most commonly used to ablate gene expression ("knock-out"), or to introduce mutant or modified alleles at the locus of interest ("knock-in"). The efficacy of producing live, transgenic mice challenges our understanding of this complex process, and of the factors which influence germline competence of embryonic stem cell lines. Increasingly, evidence indicates that culture conditions and in vitro manipulation can affect the germline-competence of Embryonic Stem cell (ES cell) lines by accumulation of chromosome abnormalities and/or epigenetic alterations of the ES cell genome. The effectiveness of ES cell derivation is greatly strain-dependent and it may also influence the germline transmission capability. Recent technical improvements in the production of germline chimeras have been focused on means of generating ES cells lines with a higher germline potential. There are a number of options for generating chimeras from ES cells (ES chimera mice); however, each method has its advantages and disadvantages. Recent developments in induced pluripotent stem (iPS) cell technology have opened new avenues for generation of animals from genetically modified somatic cells by means of chimera technologies. The aim of this review is to give a brief account of how the factors mentioned above are influencing the germline transmission capacity and the developmental potential of mouse pluripotent stem cell lines. The most recent methods for generating specifically ES and iPS chimera mice, including the advantages and disadvantages of each method are also discussed.

17.
BMC Dev Biol ; 7: 39, 2007 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-17470285

RESUMO

BACKGROUND: Rybp (Ring1 and YY1 binding protein) is a zinc finger protein which interacts with the members of the mammalian polycomb complexes. Previously we have shown that Rybp is critical for early embryogenesis and that haploinsufficiency of Rybp in a subset of embryos causes failure of neural tube closure. Here we investigated the requirement for Rybp in ocular development using four in vivo mouse models which resulted in either the ablation or overexpression of Rybp. RESULTS: Our results demonstrate that loss of a single Rybp allele in conventional knockout mice often resulted in retinal coloboma, an incomplete closure of the optic fissure, characterized by perturbed localization of Pax6 but not of Pax2. In addition, about one half of Rybp-/- <-> Rybp+/+ chimeric embryos also developed retinal colobomas and malformed lenses. Tissue-specific transgenic overexpression of Rybp in the lens resulted in abnormal fiber cell differentiation and severe lens opacification with increased levels of AP-2alpha and Sox2, and reduced levels of betaA4-crystallin gene expression. Ubiquitous transgenic overexpression of Rybp in the entire eye caused abnormal retinal folds, corneal neovascularization, and lens opacification. Additional changes included defects in anterior eye development. CONCLUSION: These studies establish Rybp as a novel gene that has been associated with coloboma. Other genes linked to coloboma encode various classes of transcription factors such as BCOR, CBP, Chx10, Pax2, Pax6, Six3, Ski, Vax1 and Vax2. We propose that the multiple functions for Rybp in regulating mouse retinal and lens development are mediated by genetic, epigenetic and physical interactions between these genes and proteins.


Assuntos
Olho/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Repressoras/genética , Dedos de Zinco/genética , Animais , Western Blotting , Coloboma/embriologia , Coloboma/genética , Córnea/anormalidades , Córnea/embriologia , Córnea/ultraestrutura , Embrião de Mamíferos , Feminino , Cristalino/anormalidades , Cristalino/embriologia , Cristalino/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica , Gravidez , Retina/anormalidades , Retina/embriologia , Retina/ultraestrutura , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Genesis ; 44(4): 202-18, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16604528

RESUMO

The visualization of live cell behaviors operating in situ combined with the power of mouse genetics represents a major step toward understanding the mechanisms regulating embryonic development, homeostasis, and disease progression in mammals. The availability of genetically encoded fluorescent protein reporters, combined with improved optical imaging modalities, have led to advances in our ability to examine cells in vivo. We developed a series of lipid-modified fluorescent protein fusions that are targeted to and label the secretory pathway and the plasma membrane, and that are amenable for use in mice. Here we report the generation of two strains of mice, each expressing a spectrally distinct lipid-modified GFP-variant fluorescent protein fusion. The CAG::GFP-GPI strain exhibited widespread expression of a glycosylphosphatidylinositol-tagged green fluorescent protein (GFP) fusion, while the CAG::myr-Venus strain exhibited widespread expression of a myristoyl-Venus yellow fluorescent protein fusion. Imaging of live transgenic embryonic stem (ES) cells, either live or fixed embryos and postnatal tissues demonstrated that glycosylphosphatidyl inositol- and myristoyl-tagged GFP-variant fusion proteins are targeted to and serve as markers of the plasma membrane. Moreover, our data suggest that these two lipid-modified protein fusions are dynamically targeted both to overlapping as well as distinct lipid-enriched compartments within cells. These transgenic strains not only represent high-contrast reporters of cell morphology and plasma membrane dynamics, but also may be used as in vivo sensors of lipid localization. Furthermore, combining these reporters with the study of mouse mutants will be a step forward in understanding the inter- and intracellular behaviors underlying morphogenesis in both normal and mutant contexts.


Assuntos
Embrião de Mamíferos/citologia , Técnicas Genéticas , Lipídeos/química , Proteínas Luminescentes/química , Proteínas Luminescentes/metabolismo , Células-Tronco/metabolismo , Animais , Animais não Endogâmicos , Blastocisto/citologia , Células COS , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Cruzamentos Genéticos , Variação Genética , Vetores Genéticos , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Processamento de Imagem Assistida por Computador , Proteínas Luminescentes/genética , Camundongos , Camundongos Endogâmicos ICR , Camundongos Endogâmicos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Células-Tronco/citologia , Transgenes
19.
Mol Cell Biol ; 25(16): 7193-202, 2005 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16055728

RESUMO

The Rybp/DEDAF protein has been implicated in both transcriptional regulation and apoptotic signaling, but its precise molecular function is unclear. To determine the physiological role of Rybp, we analyzed its expression during mouse development and generated mice carrying a targeted deletion of Rybp using homologous recombination in embryonic stem cells. Rybp was found to be broadly expressed during embryogenesis and was particularly abundant in extraembryonic tissues, including trophoblast giant cells. Consistent with this result, rybp homozygous null embryos exhibited lethality at the early postimplantation stage. At this time, Rybp was essential for survival of the embryo, for the establishment of functional extraembryonic structures, and for the execution of full decidualization. Through the use of a chimeric approach, the embryonic lethal phenotype was circumvented and a role for Rybp in central nervous system development was uncovered. Specifically, the presence of Rybp-deficient cells resulted in marked forebrain overgrowth and in localized regions of disrupted neural tube closure. Functions for Rybp in the brain also were supported by the finding of exencephaly in about 15% of rybp heterozygous mutant embryos, and by Rybp's distinct neural expression pattern. Together, these findings support critical roles for Rybp at multiple stages of mouse embryogenesis.


Assuntos
Sistema Nervoso Central/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Repressoras/fisiologia , Animais , Blastocisto/metabolismo , Encéfalo/metabolismo , Morte Celular , Sobrevivência Celular , DNA Complementar/metabolismo , Implantação do Embrião , Deleção de Genes , Vetores Genéticos , Genótipo , Heterozigoto , Homozigoto , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Mutação , Neurônios/metabolismo , Fenótipo , Prosencéfalo/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA