Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Lab Anim (NY) ; 52(12): 324-331, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38017180

RESUMO

Adoptive cell transfer between genetically identical hosts relies on the use of a congenic marker to distinguish the donor cells from the host cells. CD45, a glycoprotein expressed by all hematopoietic cells, is one of the main congenic markers used because its two isoforms, CD45.1 and CD45.2, can be discriminated by flow cytometry. As a consequence, C57BL/6J (B6; CD45.2) and B6.SJL-Ptprca Pepcb/BoyJ (B6.SJL; CD45.1) mice are widely used in adoptive cell transfer experiments, under the presumption that they differ only at the CD45 (Ptprc) locus. However, recent studies have identified genetic variations between these congenic strains and have notably highlighted a differential expression of cathepsin E (CTSE). The B6.SJL mouse presents a number of functional differences in hematopoietic stem cell engraftment potential and immune cell numbers compared with the B6 mouse. In this study, we showed that B6 and B6.SJL mice also differ in their CD8+ T cell compartment and CD8+ T cell responses to viral infection. We identified Ctse as the most differentially expressed gene between CD8+ T cells of B6 and B6.SJL and demonstrated that the differences reported between these two mouse strains are not due to CTSE. Finally, using CRISPR-Cas9 genome editing, we generated a CD45.1-expressing B6 mouse by inserting one nucleotide mutation (A904G) leading to an amino acid change (K302E) in the Ptprc gene of the B6 mouse. We showed that this new B6-Ptprcem(K302E)Jmar/J mouse resolves the experimental biases reported between the B6 and B6.SJL mouse lines and should thus represent the new gold standard for adoptive cell transfer experiments in B6.


Assuntos
Linfócitos T CD8-Positivos , Células-Tronco Hematopoéticas , Camundongos , Animais , Camundongos Endogâmicos C57BL , Epitopos , Camundongos Endogâmicos , Transferência Adotiva
2.
Neurobiol Dis ; 174: 105896, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36243247

RESUMO

Inactivating mutations in the specific thyroid hormone transporter monocarboxylate transporter 8 (MCT8) lead to an X-linked rare disease named MCT8 deficiency or Allan-Herndon-Dudley Syndrome. Patients exhibit a plethora of severe endocrine and neurological alterations, with no effective treatment for the neurological symptoms. An optimal mammalian model is essential to explore the pathological mechanisms and potential therapeutic approaches. Here we have generated by CRISPR/Cas9 an avatar mouse model for MCT8 deficiency with a point mutation found in two MCT8-deficient patients (P253L mice). We have predicted by in silico studies that this mutation alters the substrate binding pocket being the probable cause for impairing thyroid hormone transport. We have characterized the phenotype of MCT8-P253L mice and found endocrine alterations similar to those described in patients and in MCT8-deficient mice. Importantly, we detected brain hypothyroidism, structural and functional neurological alterations resembling the patient's neurological impairments. Thus, the P253L mouse provides a valuable model for studying the pathophysiology of MCT8 deficiency and in the future will allow to test therapeutic alternatives such as in vivo gene therapy and pharmacological chaperone therapy to improve the neurological impairments in MCT8 deficiency.


Assuntos
Transportadores de Ácidos Monocarboxílicos , Simportadores , Animais , Camundongos , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/genética , Simportadores/metabolismo , Sistemas CRISPR-Cas , Hormônios Tireóideos/metabolismo , Modelos Animais de Doenças , Mamíferos/metabolismo
3.
Sci Signal ; 15(738): eabj4583, 2022 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-35700264

RESUMO

Resistance to thyroid hormone due to mutations in THRA, which encodes the thyroid hormone receptor α (TRα1), shows variable clinical presentation. Mutations affecting TRß1 and TRß2 cause deafness in mice and have been associated with deafness in humans. To test whether TRα1 also affects hearing function, we used mice heterozygous for a frameshift mutation in Thra that is similar to human THRA mutations (ThraS1/+ mice) and reduces tissue sensitivity to thyroid hormone. Compared to wild-type littermates, ThraS1/+ mice showed moderate high-frequency sensorineural hearing loss as juveniles and increased age-related hearing loss. Ultrastructural examination revealed aberrant orientation of ~20% of sensory outer hair cells (OHCs), as well as increased numbers of mitochondria with fragmented morphology and autophagic vacuoles in both OHCs and auditory nerve fibers. Molecular dissection of the OHC lateral wall components revealed that the potassium ion channel Kcnq4 was aberrantly targeted to the cytoplasm of mutant OHCs. In addition, mutant cochleae showed increased oxidative stress, autophagy, and mitophagy associated with greater age-related cochlear cell damage, demonstrating that TRα1 is required for proper development of OHCs and for maintenance of OHC function. These findings suggest that patients with THRA mutations may present underdiagnosed, mild hearing loss and may be more susceptible to age-related hearing loss.


Assuntos
Surdez , Perda Auditiva , Receptores alfa dos Hormônios Tireóideos , Animais , Perda Auditiva/genética , Camundongos , Mutação , Receptores alfa dos Hormônios Tireóideos/genética , Hormônios Tireóideos
4.
Chemosphere ; 287(Pt 3): 132253, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34543901

RESUMO

Nanopesticides are innovative pesticides involving engineered nanomaterials in their formulation to increase the efficiency of plant protection products, while mitigating their environmental impact. Despite the predicted growth of the nanopesticide use, no data is available on their inhalation toxicity and the potential cocktail effects between their components. In particular, the neurodevelopmental toxicity caused by prenatal exposures might have long lasting consequences. In the present study, we repeatedly exposed gestating mice in a whole-body exposure chamber to three aerosols, involving the paraquat herbicide, nanoscaled titanium dioxide particles (nTiO2), or a mixture of both. Particle number concentrations and total mass concentrations were followed to enable a metrological follow-up of the exposure sessions. Based on the aerosols characteristics, the alveolar deposited dose in mice was then estimated. RNA-seq was used to highlight dysregulations in the striatum of pups in response to the in utero exposure. Modifications in gene expression were identified at post-natal day 14, which might reflect neurodevelopmental alterations in this key brain area. The data suggest an alteration in the mitochondrial function following paraquat exposure, which is reminiscent of the pathological process leading to Parkinson disease. Markers of different cell lineages were dysregulated, showing effects, which were not limited to dopaminergic neurons. Exposure to the nTiO2 aerosol modulated the regulation of cytokines and neurotransmitters pathways, perhaps reflecting a minor neuroinflammation. No synergy was found between paraquat and nTiO2. Instead, the neurodevelopmental effects were surprisingly lower than the one measured for each substance separately.


Assuntos
Paraquat , Efeitos Tardios da Exposição Pré-Natal , Aerossóis , Animais , Encéfalo , Feminino , Expressão Gênica , Exposição por Inalação , Camundongos , Paraquat/toxicidade , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Titânio/toxicidade
5.
iScience ; 23(3): 100899, 2020 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-32092701

RESUMO

Mammalian brain development critically depends on proper thyroid hormone signaling, via the TRα1 nuclear receptor. The downstream mechanisms by which TRα1 impacts brain development are currently unknown. In order to investigate these mechanisms, we used mouse genetics to induce the expression of a dominant-negative mutation of TRα1 specifically in GABAergic neurons, the main inhibitory neurons in the brain. This triggered post-natal epileptic seizures and a profound impairment of GABAergic neuron maturation in several brain regions. Analysis of the transcriptome and TRα1 cistrome in the striatum allowed us to identify a small set of genes, the transcription of which is upregulated by TRα1 in GABAergic neurons and which probably plays an important role during post-natal maturation of the brain. Thus, our results point to GABAergic neurons as direct targets of thyroid hormone during brain development and suggest that many defects seen in hypothyroid brains may be secondary to GABAergic neuron malfunction.

6.
Thyroid ; 29(9): 1327-1335, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31298651

RESUMO

Background: Resistance to thyroid hormone alpha (RTHα) is a rare genetic disease due to mutations in the THRA gene, which encodes thyroid hormone receptor alpha 1 (TRα1). Since its first description in 2012, 46 cases of RTHα have been reported worldwide, corresponding to 26 different mutations of TRα1. RTHα patients share some common symptoms with hypothyroid patients, without significant reduction in thyroid hormone level. The high variability of clinical features and the absence of reliable biochemical markers make the diagnosis of this disease difficult. Some of these mutations have been recently modeled in mice. Methods: In our study, we used four different mouse models heterozygous for frameshift mutations in the Thra gene. Two of them are very close to human mutations, while the two others have not yet been found in patients. We characterized the metabolic phenotypes of urine and plasma samples collected from these four animal models using an untargeted nuclear magnetic resonance (NMR)-based metabolomic approach. Results: Multivariate statistical analysis of the metabolomic profiles shows that biofluids of mice that carry human-like mutations can be discriminated from controls. Metabolic signatures associated with Thra mutations in urine and plasma are stable over time and clearly differ from the metabolic fingerprint of hypothyroidism in the mouse. Conclusion: Our results provide a proof-of-principle that easily accessible NMR-based metabolic fingerprints of biofluids could be used to diagnose RTHα in humans.


Assuntos
Líquidos Corporais/metabolismo , Espectroscopia de Ressonância Magnética/métodos , Metabolômica/métodos , Mutação , Receptores alfa dos Hormônios Tireóideos/genética , Animais , Genes erbA , Humanos , Hipotireoidismo/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL
7.
Sci Rep ; 8(1): 4679, 2018 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-29535400

RESUMO

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

8.
Sci Rep ; 8(1): 474, 2018 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-29323173

RESUMO

In this report, we present an improved protocol for CRISPR/Cas9 genome editing in mice. The procedure consists in the electroporation of intact mouse zygotes with ribonucleoprotein complexes prepared in vitro from recombinant Cas9 nuclease and synthetic dual guide RNA. This simple cloning-free method proves to be extremely efficient for the generation of indels and small deletions by non-homologous end joining, and for the generation of specific point mutations by homology-directed repair. The procedure, which avoids DNA construction, in vitro transcription and oocyte microinjection, greatly simplifies genome editing in mice.


Assuntos
Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , RNA Guia de Cinetoplastídeos/genética , Zigoto/metabolismo , Animais , Reparo do DNA por Junção de Extremidades , Enzimas Desubiquitinantes , Eletroporação , Endopeptidases/química , Endopeptidases/genética , Feminino , Loci Gênicos , Técnicas de Genotipagem , Mutação INDEL , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto
9.
Thyroid ; 28(1): 139-150, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29205102

RESUMO

BACKGROUND: Resistance to thyroid hormone due to THRA mutations (RTHα) is a recently discovered genetic disease, displaying important variability in its clinical presentation. The mutations alter the function of TRα1, one of the two nuclear receptors for thyroid hormone. METHODS: The aim of this study was to understand the relationship between specific THRA mutations and phenotype. CRISPR/Cas9 genome editing was used to generate five new mouse models of RTHα, with frameshift or missense mutations. RESULTS: Like human patients, mutant mice displayed a hypothyroid-like phenotype, with altered development. Phenotype severity varied between the different mouse models, mainly depending on the ability of the mutant receptor to interact with transcription corepressor in the presence of thyroid hormone. CONCLUSION: The present mutant mice represent highly relevant models for the human genetic disease which will be useful for future investigations.


Assuntos
Genes erbA/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Animais , Sistemas CRISPR-Cas , Camundongos , Mutação , Fenótipo
10.
J Mol Endocrinol ; 57(2): R81-92, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27272521

RESUMO

CRISPR/Cas9 is a recent development in genome editing which is becoming an indispensable element of the genetic toolbox in mice. It provides outstanding possibilities for targeted modification of the genome, and is often extremely efficient. There are currently two main limitations to in ovo genome editing in mice: the first is mosaicism, which is frequent in founder mice. The second is the difficulty to evaluate the advent of off-target mutations, which often imposes to wait for germline transmission to ensure genetic segregation between wanted and unwanted genetic mutations. However rapid progresses are made, suggesting that these difficulties can be overcome in the near future.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Camundongos Transgênicos , Animais , Reparo do DNA por Junção de Extremidades , Modelos Animais de Doenças , Endocrinologistas , Endocrinologia/métodos , Edição de Genes/métodos , Marcação de Genes/efeitos adversos , Marcação de Genes/métodos , Camundongos , Mosaicismo , Oócitos/metabolismo , RNA Guia de Cinetoplastídeos/genética , Reparo de DNA por Recombinação
11.
Nat Commun ; 6: 7398, 2015 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-26154507

RESUMO

The generation of induced pluripotent stem (iPS) cells holds great promise in regenerative medicine. The use of the transcription factors Oct4, Sox2, Klf4 and c-Myc for reprogramming is extensively documented, but comparatively little is known about soluble molecules promoting reprogramming. Here we identify the secreted cue Netrin-1 and its receptor DCC, described for their respective survival/death functions in normal and oncogenic contexts, as reprogramming modulators. In various somatic cells, we found that reprogramming is accompanied by a transient transcriptional repression of Netrin-1 mediated by an Mbd3/Mta1/Chd4-containing NuRD complex. Mechanistically, Netrin-1 imbalance induces apoptosis mediated by the receptor DCC in a p53-independent manner. Correction of the Netrin-1/DCC equilibrium constrains apoptosis and improves reprogramming efficiency. Our work also sheds light on Netrin-1's function in protecting embryonic stem cells from apoptosis mediated by its receptor UNC5b, and shows that the treatment with recombinant Netrin-1 improves the generation of mouse and human iPS cells.


Assuntos
Reprogramação Celular/fisiologia , Fatores de Crescimento Neural/metabolismo , Fatores de Crescimento Neural/farmacologia , Células-Tronco Pluripotentes/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/farmacologia , Animais , Células Cultivadas , Fibroblastos , Regulação da Expressão Gênica/fisiologia , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Fatores de Crescimento Neural/genética , Receptores de Netrina , Netrina-1 , Regiões Promotoras Genéticas , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
12.
Biol Open ; 2(6): 613-28, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23789112

RESUMO

Not much is known about the molecular and functional features of pluripotent stem cells (PSCs) in rabbits. To address this, we derived and characterized 2 types of rabbit PSCs from the same breed of New Zealand White rabbits: 4 lines of embryonic stem cells (rbESCs), and 3 lines of induced PSCs (rbiPSCs) that were obtained by reprogramming adult skin fibroblasts. All cell lines required fibroblast growth factor 2 for their growth and proliferation. All rbESC lines showed molecular and functional properties typically associated with primed pluripotency. The cell cycle of rbESCs had a prolonged G1 phase and a DNA damage checkpoint before entry into the S phase, which are the 2 features typically associated with the somatic cell cycle. In contrast, the rbiPSC lines exhibited some characteristics of naïve pluripotency, including resistance to single-cell dissociation by trypsin, robust activity of the distal enhancer of the mouse Oct4 gene, and expression of naïve pluripotency-specific genes, as defined in rodents. According to gene expression profiles, rbiPSCs were closer to the rabbit inner cell mass (ICM) than rbESCs. Furthermore, rbiPSCs were capable of colonizing the ICM after aggregation with morulas. Therefore, we propose that rbiPSCs self-renew in an intermediate state between naïve and primed pluripotency, which represents a key step toward the generation of bona fide naïve PSC lines in rabbits.

13.
Stem Cell Res ; 10(1): 118-31, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23178806

RESUMO

A short G1 phase is a characteristic feature of mouse embryonic stem cells (ESCs). To determine if there is a causal relationship between G1 phase restriction and pluripotency, we made use of the Fluorescence Ubiquitination Cell Cycle Indicator (FUCCI) reporter system to FACS-sort ESCs in the different cell cycle phases. Hence, the G1 phase cells appeared to be more susceptible to differentiation, particularly when ESCs self-renewed in the naïve state of pluripotency. Transitions from ground to naïve, then from naïve to primed states of pluripotency were associated with increased durations of the G1 phase, and cyclin E-mediated alteration of the G1/S transition altered the balance between self-renewal and differentiation. LIF withdrawal resulted in a lengthening of the G1 phase in naïve ESCs, which occurred prior to the appearance of early lineage-specific markers, and could be reversed upon LIF supplementation. We concluded that the short G1 phase observed in murine ESCs was a determinant of naïve pluripotency and was partially under the control of LIF signaling.


Assuntos
Células-Tronco Embrionárias/citologia , Fase G1 , Animais , Diferenciação Celular , Ciclina E/antagonistas & inibidores , Ciclina E/genética , Ciclina E/metabolismo , Fase G1/efeitos dos fármacos , Humanos , Fator Inibidor de Leucemia/farmacologia , Camundongos , Microscopia Confocal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Transdução de Sinais , Imagem com Lapso de Tempo , Ubiquitinação
14.
Stem Cells Dev ; 20(2): 301-11, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20649486

RESUMO

The LIM homeodomain transcription factor 1b (Lmx1b) is a key factor in the specification of the serotonergic neurotransmitter phenotype. Here, we explored the capacity of Lmx1b to direct differentiation of mouse embryonic stem (mES) cells into serotonergic neurons. mES cells stably expressing human Lmx1b were generated by lentiviral vector infection. Clones expressing Lmx1b at a low level showed increased neurogenesis and elevated production of neurons expressing serotonin, serotonin transporter, tryptophan hydroxylase 2, and transcription factor Pet1, the landmarks of serotonergic differentiation. To explore the role of Lmx1b in the specification of the serotonin neurotransmission phenotype further, a conditional system making use of a floxed inducible vector targeted into the ROSA26 locus and a hormone-dependent Cre recombinase was engineered. This novel strategy was tested with the reporter gene encoding human placental alkaline phosphatase, and demonstrated its capacity to drive transgene expression in nestin(+) neural progenitors (NPs) and in Tuj1(+) neurons. When it was applied to inducible expression of human Lmx1b, it resulted in elevated expression of serotonergic markers. Treatment of neural precursors with the floor plate signal Sonic hedgehog further enhanced differentiation of Lmx1b-overexpressing NPs into neurons expressing 5-HT, serotonin transporter, tryptophan hydroxylase 2, and Pet1, when compared with Lmx1b-nonexpressing progenitors. Together, our results demonstrate the capacity of Lmx1b to specify a serotonin neurotransmitter phenotype when overexpressed in mES cell-derived NPs.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/biossíntese , Neurônios/citologia , Serotonina/metabolismo , Fatores de Transcrição/biossíntese , Fosfatase Alcalina/biossíntese , Animais , Células Cultivadas , Técnicas de Cocultura , Células-Tronco Embrionárias/metabolismo , Fator 4 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos/metabolismo , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Proteínas Ligadas por GPI/biossíntese , Vetores Genéticos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Isoenzimas/biossíntese , Proteínas com Homeodomínio LIM , Lentivirus/genética , Camundongos , Neurônios/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas da Membrana Plasmática de Transporte de Serotonina/genética , Proteínas da Membrana Plasmática de Transporte de Serotonina/metabolismo , Fatores de Transcrição/genética , Ativação Transcricional , Tirosina 3-Mono-Oxigenase/genética , Tirosina 3-Mono-Oxigenase/metabolismo , Regulação para Cima
15.
Int J Dev Biol ; 53(4): 569-78, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378248

RESUMO

Embryonic stem (ES) cells which constitutively express the Pdx-1, Ngn-3, NeuroD1, Nkx2.2, and Nkx6.1 transcription factors were engineered by means of lentiviral vectors, following a multi-step infection procedure to successively generate ES cell lines expressing one, two, and three factors, respectively. Each ES cell line was allowed to differentiate into nestin+/Isl-1+ endocrine precursors, then into more mature pancreatic cells, and subsequently analysed for expression of Glc, Ins, and Sst, markers of alpha, beta and delta cells, respectively. Each ES cell line generated displayed a unique pattern of gene expression. The ES cell line expressing NeuroD1 displayed vastly elevated levels of Glc, Ins-1, Ins-2 and Sst, and showed an increase in Pdx-1, Pax-4, Nkx6.1, Isl-1, Glut-2 and GK transcript levels. Furthermore, immunofluorescence analysis revealed that differentiation of NeuroD1-expressing ES cells in nestin+/Isl-1+ multilineage progenitors, followed by the formation of C-peptide+/insulin+ clusters, was accelerated. Together, these results indicate that stable expression of NeuroD1 in ES cells facilitates differentiation into endocrine and insulin-producing cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Linhagem da Célula , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Sistema Endócrino/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Biomarcadores/metabolismo , Sistema Endócrino/citologia , Sistema Endócrino/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteína Homeobox Nkx-2.2 , Insulina/biossíntese , Células Secretoras de Insulina/citologia , Camundongos , Fatores de Transcrição/metabolismo
16.
Stem Cells ; 26(6): 1444-53, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18356572

RESUMO

Embryonic stem cells (ESC) have the ability of indefinite self-renewal and multilineage differentiation, and they carry great potential in cell-based therapies. The rhesus macaque is the most relevant preclinical model for assessing the benefit, safety, and efficacy of ESC-based transplantations in the treatment of neurodegenerative diseases. In the case of neural cell grafting, tracing both the neurons and their axonal projections in vivo is essential for studying the integration of the grafted cells in the host brain. Tau-Green fluorescent protein (tau-GFP) is a powerful viable lineage tracer, allowing visualization of cell bodies, dendrites, and axons in exquisite detail. Here, we report the first rhesus monkey ESC line that ubiquitously and stably expresses tau-GFP. First, we derived a new line of rhesus monkey ESC (LYON-ES1) that show marker expression and cell cycle characteristics typical of primate ESCs. LYON-ES1 cells are pluripotent, giving rise to derivatives of the three germ layers in vitro and in vivo through teratoma formation. They retain all their undifferentiated characteristics and a normal karyotype after prolonged culture. Using lentiviral infection, we then generated a monkey ESC line stably expressing tau-GFP that retains all the characteristics of the parental wild-type line and is clonogenic. We show that neural precursors derived from the tau-GFP ESC line are multipotent and that their fate can be precisely mapped in vivo after grafting in the adult rat brain. Disclosure of potential conflicts of interest is found at the end of this article.


Assuntos
Células-Tronco Embrionárias/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas tau/genética , Fosfatase Alcalina/metabolismo , Animais , Blastocisto/citologia , Diferenciação Celular , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Células-Tronco Embrionárias/virologia , Genes Reporter , Lentivirus , Macaca mulatta , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante de Células-Tronco/métodos , Teratoma/genética , Teratoma/patologia , Transfecção , Zona Pelúcida/fisiologia
17.
Oncogene ; 21(36): 5515-28, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12165850

RESUMO

Mouse embryonic stem (ES) cells are known to express D-type cyclins at very low levels and these levels increase dramatically during in vitro and in vivo differentiation. Here, we investigate some of the signalling pathways regulating expression of cyclin D1 and progression to S phase, the Ras/Extracellular signal-regulated protein kinase (ERK) pathway and the phosphatidylinositol 3-kinase (PI3-kinase) pathway. We demonstrate that ERK phosphorylation is fully dispensable for the regulation of cyclin D1 level and for the progression from G1 to S phase in ES cells. By contrast, PI3-kinase activity is required for both. Differentiation induced by retinoic acid results in the gain of ERK-dependent control of cyclin D1 expression and of S phase progression. Differentiation is also paralleled by an increase in PI3-kinase activity. This leads (a) to an increase in the p70 S6 kinase-dependent regulation of the steady-state level of cyclin D1, and (b) to a concomitant decrease in the GSK3beta-dependent rate of cyclin D1 degradation. Altogether, these multiple pathways account for the dramatic increase in the level of cyclin D1 protein which parallels ES cell differentiation. Our studies suggest that PI3-kinase is an important regulator of the ES cell cycle and that its activity is not regulated by mitogen stimulation.


Assuntos
Ciclina D1/biossíntese , Fase G1/fisiologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fase S/fisiologia , Células-Tronco/metabolismo , Animais , Antineoplásicos/farmacologia , Northern Blotting , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular/efeitos dos fármacos , Embrião de Mamíferos/citologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase , Imunossupressores/farmacologia , Camundongos , Fosforilação , RNA Mensageiro/metabolismo , Sirolimo/farmacologia , Tretinoína/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA