Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3880, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719804

RESUMEN

Correlative evidence has suggested that the methyl-CpG-binding protein MeCP2 contributes to the formation of heterochromatin condensates via liquid-liquid phase separation. This interpretation has been reinforced by the observation that heterochromatin, DNA methylation and MeCP2 co-localise within prominent foci in mouse cells. The findings presented here revise this view. MeCP2 localisation is independent of heterochromatin as MeCP2 foci persist even when heterochromatin organisation is disrupted. Additionally, MeCP2 foci fail to show hallmarks of phase separation in live cells. Importantly, we find that mouse cellular models are highly atypical as MeCP2 distribution is diffuse in most mammalian species, including humans. Notably, MeCP2 foci are absent in Mus spretus which is a mouse subspecies lacking methylated satellite DNA repeats. We conclude that MeCP2 has no intrinsic tendency to form condensates and its localisation is independent of heterochromatin. Instead, the distribution of MeCP2 in the nucleus is primarily determined by global DNA methylation patterns.


Asunto(s)
Metilación de ADN , Heterocromatina , Proteína 2 de Unión a Metil-CpG , Proteína 2 de Unión a Metil-CpG/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Heterocromatina/metabolismo , Animales , Ratones , Humanos , Núcleo Celular/metabolismo , Unión Proteica , ADN/metabolismo , ADN Satélite/metabolismo , ADN Satélite/genética , Separación de Fases
2.
BMC Biol ; 20(1): 14, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-35027054

RESUMEN

BACKGROUND: Infectious diseases of farmed and wild animals pose a recurrent threat to food security and human health. The macrophage, a key component of the innate immune system, is the first line of defence against many infectious agents and plays a major role in shaping the adaptive immune response. However, this phagocyte is a target and host for many pathogens. Understanding the molecular basis of interactions between macrophages and pathogens is therefore crucial for the development of effective strategies to combat important infectious diseases. RESULTS: We explored how porcine pluripotent stem cells (PSCs) can provide a limitless in vitro supply of genetically and experimentally tractable macrophages. Porcine PSC-derived macrophages (PSCdMs) exhibited molecular and functional characteristics of ex vivo primary macrophages and were productively infected by pig pathogens, including porcine reproductive and respiratory syndrome virus (PRRSV) and African swine fever virus (ASFV), two of the most economically important and devastating viruses in pig farming. Moreover, porcine PSCdMs were readily amenable to genetic modification by CRISPR/Cas9 gene editing applied either in parental stem cells or directly in the macrophages by lentiviral vector transduction. CONCLUSIONS: We show that porcine PSCdMs exhibit key macrophage characteristics, including infection by a range of commercially relevant pig pathogens. In addition, genetic engineering of PSCs and PSCdMs affords new opportunities for functional analysis of macrophage biology in an important livestock species. PSCs and differentiated derivatives should therefore represent a useful and ethical experimental platform to investigate the genetic and molecular basis of host-pathogen interactions in pigs, and also have wider applications in livestock.


Asunto(s)
Virus de la Fiebre Porcina Africana , Enfermedades Transmisibles , Virus de la Fiebre Porcina Africana/genética , Animales , Interacciones Huésped-Patógeno/genética , Macrófagos , Células Madre , Porcinos
3.
Stem Cell Reports ; 16(4): 727-740, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33667414

RESUMEN

Apelin receptor (APLNR/AGTRLl1/APJ) marks a transient cell population during the differentiation of hematopoietic stem and progenitor cells (HSPCs) from pluripotent stem cells, but its function during the production and maintenance of hematopoietic stem cells is not clear. We generated an Aplnr-tdTomato reporter mouse embryonic stem cell (mESC) line and showed that HSPCs are generated exclusively from mesodermal cells that express Aplnr-tdTomato. HSPC production from mESCs was impaired when Aplnr was deleted, implying that this pathway is required for their production. To address the role of APLNR signaling in HSPC maintenance, we added APELIN ligands to ex vivo AGM cultures. Activation of the APLNR pathway in this system impaired the generation of long-term reconstituting HSPCs and appeared to drive myeloid differentiation. Our data suggest that the APLNR signaling is required for the generation of cells that give rise to HSCs, but that its subsequent downregulation is required for their maintenance.


Asunto(s)
Receptores de Apelina/metabolismo , Hematopoyesis , Transducción de Señal , Animales , Apelina/metabolismo , Receptores de Apelina/genética , Agregación Celular , Diferenciación Celular , Células Cultivadas , Eliminación de Gen , Regulación de la Expresión Génica , Genes Reporteros , Hemangioblastos/metabolismo , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ligandos , Mesodermo/citología , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Hormonas Peptídicas/metabolismo
4.
Mol Reprod Dev ; 87(9): 930-933, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32853477

RESUMEN

The purine hypoxanthine plays important role in regulating oocyte maturation and early embryonic development. The enzyme hypoxanthine phosphoribosyltransferase (HPRT) recycles hypoxanthine to generate substrates for nucleotide synthesis and key metabolites, and here we show that HPRT deficiency in the rat disrupts early embryonic development and causes infertility in females.


Asunto(s)
Infertilidad Femenina/etiología , Síndrome de Lesch-Nyhan/complicaciones , Animales , Desarrollo Embrionario/genética , Femenino , Fertilidad/genética , Viabilidad Fetal/genética , Hipoxantina/metabolismo , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/genética , Hipoxantina Fosforribosiltransferasa/metabolismo , Infertilidad Femenina/genética , Síndrome de Lesch-Nyhan/genética , Síndrome de Lesch-Nyhan/patología , Embarazo , Purinas/metabolismo , Ratas
5.
Stem Cell Reports ; 14(1): 154-166, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31902707

RESUMEN

Rat embryonic stem cells (rESCs) are capable of contributing to all differentiated tissues, including the germ line in chimeric animals, and represent a unique, authentic alternative to mouse embryonic stem cells for studying stem cell pluripotency and self-renewal. Here, we describe an EGFP reporter transgene that tracks expression of the benchmark naive pluripotency marker gene Rex1 (Zfp42) in the rat. Insertion of the EGFP reporter gene downstream of the Rex1 promoter disrupted Rex1 expression, but REX1-deficient rESCs and rats were viable and apparently normal, validating this targeted knockin transgene as a neutral reporter. The Rex1-EGFP gene responded to self-renewal/differentiation factors and validated the critical role of ß-catenin/LEF1 signaling. The stem cell reporter also allowed the identification of functionally distinct sub-populations of cells within rESC cultures, thus demonstrating its utility in discriminating between cell states in rat stem cell cultures, as well as providing a tool for tracking Rex1 expression in the rat.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Genes Reporteros , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Inmunofenotipificación , Ratas
6.
Anal Chem ; 91(16): 10713-10722, 2019 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-31393105

RESUMEN

The ultimate performance of flow-based measurements in microfluidic systems is currently limited by their accuracy at the nanoliter-per-minute scale. Improving such measurements (especially in contexts that require continuous monitoring) is challenging because of constraints associated with shrinking system geometries and limitations imposed by making precise measurements of smaller quantities in real time. A particularly interesting limit is the relative uncertainty as flow approaches zero, which diverges for most measurement methods. To address these problems, we have developed an optofluidic measurement system that can deliver and record light in a precise interrogation region of a microfluidic channel. The system utilizes photobleaching of fluorophore dyes in the bulk flow and can identify zero flow to better than 1 nL/min absolute accuracy. The technique also provides an independent method for determining nonzero flow rates based on a robust scaling relationship between the fluorescence emission and flow. Together, these two independent approaches enable precise measurement of flow to within 5% accuracy down to 10 nL/min and validation of flow control to within 5% uncertainty down to 2 nL/min. We also demonstrate that our technique can be used to extend a calibrated flow meter well below its specified range (e.g., 500 nL/min) and to make dynamic measurements of similar relative uncertainties to the calibrated meter, which would have otherwise expanded significantly in this regime.

8.
Sci Rep ; 9(1): 6068, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30988473

RESUMEN

X chromosome inactivation (XCI) is a mammalian specific, developmentally regulated process relying on several mechanisms including antisense transcription, non-coding RNA-mediated silencing, and recruitment of chromatin remodeling complexes. In vitro modeling of XCI, through differentiation of embryonic stem cells (ESCs), provides a powerful tool to study the dynamics of XCI, overcoming the need for embryos, and facilitating genetic modification of key regulatory players. However, to date, robust initiation of XCI in vitro has been mostly limited to mouse pluripotent stem cells. Here, we adapted existing protocols to establish a novel monolayer differentiation protocol for rat ESCs to study XCI. We show that differentiating rat ESCs properly downregulate pluripotency factor genes, and present female specific Xist RNA accumulation and silencing of X-linked genes. We also demonstrate that RNF12 seems to be an important player in regulation of initiation of XCI in rat, acting as an Xist activator. Our work provides the basis to investigate the mechanisms directing the XCI process in a model organism different from the mouse.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , ARN Largo no Codificante/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Inactivación del Cromosoma X/fisiología , Animales , Células Cultivadas , Embrión de Mamíferos , Femenino , Masculino , Modelos Animales , Cultivo Primario de Células , Ratas
9.
Soft Matter ; 14(44): 9020-9035, 2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30378623

RESUMEN

We investigate the unstable flow of wormlike micelle solutions in pressure driven capillary flow, with a focus on the effect of entrance geometry on the fluid fluctuations. The flow is measured at different points in the capillary using particle image velocimetry while simultaneously measuring the pressure drop across the entire capillary. The fluctuations are characterized by rapid flow rate jumps that correspond with a decrease in the pressure drop followed by a longer recovery period. Velocimetry measurements in the entrance region show a transition to unstable flow above a critical flow rate, where large flow circulations are observed in the tapered geometry and localized jets are observed in an abrupt contraction. The transition to this unstable flow is shown to occur at a similar dimensionless extension rate normalized by the micelle relaxation time. A rapid breakdown in micelle alignment is observed in polarized light microscopy at the onset of the flow rate jump, indicating the importance of rapid micelle structural changes on the fluctuations. We characterize the system by analyzing the power spectral densities and develop a dynamical systems model to describe the relationship between pressure and flow rate. These developments provide understanding to control flow fluctuations and motivation for more detailed study of the coupling of fluid microstructure transitions and flow fluctuations.

10.
J Immunol ; 201(9): 2683-2699, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30249809

RESUMEN

We have produced Csf1r-deficient rats by homologous recombination in embryonic stem cells. Consistent with the role of Csf1r in macrophage differentiation, there was a loss of peripheral blood monocytes, microglia in the brain, epidermal Langerhans cells, splenic marginal zone macrophages, bone-associated macrophages and osteoclasts, and peritoneal macrophages. Macrophages of splenic red pulp, liver, lung, and gut were less affected. The pleiotropic impacts of the loss of macrophages on development of multiple organ systems in rats were distinct from those reported in mice. Csf1r-/- rats survived well into adulthood with postnatal growth retardation, distinct skeletal and bone marrow abnormalities, infertility, and loss of visceral adipose tissue. Gene expression analysis in spleen revealed selective loss of transcripts associated with the marginal zone and, in brain regions, the loss of known and candidate novel microglia-associated transcripts. Despite the complete absence of microglia, there was little overt phenotype in brain, aside from reduced myelination and increased expression of dopamine receptor-associated transcripts in striatum. The results highlight the redundant and nonredundant functions of CSF1R signaling and of macrophages in development, organogenesis, and homeostasis.


Asunto(s)
Macrófagos , Microglía , Organogénesis/genética , Ratas/crecimiento & desarrollo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Animales , Modelos Animales , Mutación , Ratas/genética
11.
J Cell Sci ; 131(18)2018 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-30154213

RESUMEN

The cytokine leukaemia inhibitory factor (LIF) promotes self-renewal of mouse embryonic stem cells (ESCs) through activation of the transcription factor Stat3. However, the contribution of other ancillary pathways stimulated by LIF in ESCs, such as the MAPK and PI3K pathways, is less well understood. We show here that naive-type mouse ESCs express high levels of a novel effector of the MAPK and PI3K pathways. This effector is an isoform of the Gab1 (Grb2-associated binder protein 1) adaptor protein that lacks the N-terminal pleckstrin homology (PH) membrane-binding domain. Although not essential for rapid unrestricted growth of ESCs under optimal conditions, the novel Gab1 variant (Gab1ß) is required for LIF-mediated cell survival under conditions of limited nutrient availability. This enhanced survival is absolutely dependent upon a latent palmitoylation site that targets Gab1ß directly to ESC membranes. These results show that constitutive association of Gab1 with membranes through a novel mechanism promotes LIF-dependent survival of murine ESCs in nutrient-poor conditions.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Madre Embrionarias/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Animales , Células Cultivadas , Transducción de Señal
12.
Mamm Genome ; 28(7-8): 302-314, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28752194

RESUMEN

Since its domestication over 100 years ago, the laboratory rat has been the preferred experimental animal in many areas of biomedical research (Lindsey and Baker The laboratory rat. Academic, New York, pp 1-52, 2006). Its physiology, size, genetics, reproductive cycle, cognitive and behavioural characteristics have made it a particularly useful animal model for studying many human disorders and diseases. Indeed, through selective breeding programmes numerous strains have been derived that are now the mainstay of research on hypertension, obesity and neurobiology (Okamoto and Aoki Jpn Circ J 27:282-293, 1963; Zucker and Zucker J Hered 52(6):275-278, 1961). Despite this wealth of genetic and phenotypic diversity, the ability to manipulate and interrogate the genetic basis of existing phenotypes in rat strains and the methodology to generate new rat models has lagged significantly behind the advances made with its close cousin, the laboratory mouse. However, recent technical developments in stem cell biology and genetic engineering have again brought the rat to the forefront of biomedical studies and enabled researchers to exploit the increasingly accessible wealth of genome sequence information. In this review, we will describe how a breakthrough in understanding the molecular basis of self-renewal of the pluripotent founder cells of the mammalian embryo, embryonic stem (ES) cells, enabled the derivation of rat ES cells and their application in transgenesis. We will also describe the remarkable progress that has been made in the development of gene editing enzymes that enable the generation of transgenic rats directly through targeted genetic modifications in the genomes of zygotes. The simplicity, efficiency and cost-effectiveness of the CRISPR/Cas gene editing system, in particular, mean that the ability to engineer the rat genome is no longer a limiting factor. The selection of suitable targets and gene modifications will now become a priority: a challenge where ES culture and gene editing technologies can play complementary roles in generating accurate bespoke rat models for studying biological processes and modelling human disease.


Asunto(s)
Edición Génica , Ingeniería Genética , Genoma , Animales , Animales Modificados Genéticamente , Sistemas CRISPR-Cas , Diferenciación Celular , Embrión de Mamíferos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Reordenamiento Génico , Marcación de Gen/métodos , Ratones , Oligodesoxirribonucleótidos , Ratas , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Nucleasas con Dedos de Zinc/metabolismo
13.
Sci Rep ; 6: 25592, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27185277

RESUMEN

Lesch-Nyhan disease (LND) is a severe neurological disorder caused by loss-of-function mutations in the gene encoding hypoxanthine phosphoribosyltransferase (HPRT), an enzyme required for efficient recycling of purine nucleotides. Although this biochemical defect reconfigures purine metabolism and leads to elevated levels of the breakdown product urea, it remains unclear exactly how loss of HPRT activity disrupts brain function. As the rat is the preferred rodent experimental model for studying neurobiology and diseases of the brain, we used genetically-modified embryonic stem cells to generate an HPRT knock-out rat. Male HPRT-deficient rats were viable, fertile and displayed normal caged behaviour. However, metabolomic analysis revealed changes in brain biochemistry consistent with disruption of purine recycling and nucleotide metabolism. Broader changes in brain biochemistry were also indicated by increased levels of the core metabolite citrate and reduced levels of lipids and fatty acids. Targeted MS/MS analysis identified reduced levels of dopamine in the brains of HPRT-deficient animals, consistent with deficits noted previously in human LND patients and HPRT knock-out mice. The HPRT-deficient rat therefore provides a new experimental platform for future investigation of how HPRT activity and disruption of purine metabolism affects neural function and behaviour.


Asunto(s)
Encéfalo/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Animales , Humanos , Hipoxantina Fosforribosiltransferasa/deficiencia , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/genética , Masculino , Metabolómica/métodos , Ratones Noqueados , Mutación , Nucleótidos de Purina/metabolismo , Ratas Transgénicas , Roedores , Espectrometría de Masas en Tándem
14.
Methods Mol Biol ; 1212: 73-85, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25224161

RESUMEN

The rat is one of the most commonly used laboratory animals in biomedical research and the recent isolation of genuine pluripotent rat embryonic stem (ES) cell lines has provided new opportunities for applying contemporary genetic engineering techniques to the rat and enhancing the use of this rodent in scientific research. Technical refinements that improve the stability of the rat ES cell cultures will undoubtedly further strengthen and broaden the use of these stem cells in biomedical research. Here, we describe a relatively simple and robust protocol that supports the propagation of germ line competent rat ES cells, and outline how tuning stem cell signaling using small molecule inhibitors can be used to both stabilize self-renewal of rat ES cell cultures and aid evaluation of their differentiation potential in vitro.


Asunto(s)
Técnicas de Cultivo de Célula , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Transducción de Señal , Animales , Linaje de la Célula , Cuerpos Embrioides/citología , Inmunohistoquímica , Ratas
15.
Stem Cells ; 31(10): 2104-15, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23843312

RESUMEN

Stabilization of ß-catenin, through inhibition of glycogen synthase kinase 3 (GSK3) activity, in conjunction with inhibition of mitogen-activated protein kinase kinase 1/2 (MEK) promotes self-renewal of naïve-type mouse embryonic stem cells (ESC). In developmentally more advanced, primed-type, epiblast stem cells, however, ß-catenin activity induces differentiation. We investigated the response of rat ESCs to ß-catenin signaling and found that when maintained on feeder-support cells in the presence of a MEK inhibitor alone (1i culture), the derivation efficiency, growth, karyotypic stability, transcriptional profile, and differentiation potential of rat ESC cultures was similar to that of cell lines established using both MEK and GSK3 inhibitors (2i culture). Equivalent mouse ESCs, by comparison, differentiated in identical 1i conditions, consistent with insufficient ß-catenin activity. This interspecies difference in reliance on GSK3 inhibition corresponded with higher overall levels of ß-catenin activity in rat ESCs. Indeed, rat ESCs displayed widespread expression of the mesendoderm-associated ß-catenin targets, Brachyury and Cdx2 in 2i medium, and overt differentiation upon further increases in ß-catenin activity. In contrast, mouse ESCs were resistant to differentiation at similarly elevated doses of GSK3 inhibitor. Interestingly, without feeder support, moderate levels of GSK3 inhibition were necessary to support effective growth of rat ESC, confirming the conserved role for ß-catenin in ESC self-renewal. This work identifies ß-catenin signaling as a molecular rheostat in rat ESC, regulating self-renewal in a dose-dependent manner, and highlights the potential importance of controlling flux in this signaling pathway to achieve effective stabilization of naïve pluripotency.


Asunto(s)
Células Madre Embrionarias/fisiología , beta Catenina/metabolismo , Animales , Benzamidas/farmacología , Factor de Transcripción CDX2 , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Medios de Cultivo , Difenilamina/análogos & derivados , Difenilamina/farmacología , Proteínas Fetales/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Proteínas de Homeodominio/metabolismo , Laminina/metabolismo , Ratones , Piridinas/farmacología , Pirimidinas/farmacología , Ratas , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba , Vía de Señalización Wnt
16.
Cell Reprogram ; 14(2): 112-22, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22339199

RESUMEN

Distinct signaling pathways are reported to maintain pluripotency in embryo-derived stem cells. Mouse embryonic stem cells (ESCs) respond to leukemia inhibitory factor (LIF) and bone morphogenetic protein (BMP)-mediated activity, whereas human ESCs depend upon Fibroblast growth factor (FGF) and activin signaling. In the majority of mammals investigated, however, the signals that support stem cell pluripotency are not well defined, as is evident by the persistent difficulties in maintaining authentic stable ESC lines. Induction of pluripotency by transcription factor-mediated reprogramming could provide an alternative way to produce ESC-like cells from nonpermissive species, and facilitate identification of core ESC signaling requirements. To evaluate the effectiveness of this approach in pigs, we transduced porcine foetal fibroblasts with retroviruses expressing Oct4, Sox2, Klf4, and c-Myc, and maintained the resulting cultures in medium containing either LIF or FGF2. Alkaline phosphatase positive colonies with compact, mouse ESC-like morphology were preferentially recovered using serum-free medium supplemented with LIF. These cell lines expressed the endogenous stem cell transcription factors, OCT4, NANOG, and SOX2, and the cell surface marker SSEA-4, consistent with acquisition of an undifferentiated state. However, restricted differentiation potential, and persistent expression of retroviral transgenes indicated that reprogramming was incomplete. Interestingly, LIF activated both the transcription factor STAT3 and its target gene SOCS3, and stimulated cell growth, indicating functional coupling of the signaling pathway in these cells. This demonstration of LIF-dependence in reprogrammed pig cells supports the notion that the connection between LIF/STAT3 signaling and the core regulatory network of pluripotent stem cells is a conserved pathway in mammals.


Asunto(s)
Reprogramación Celular/fisiología , Células Madre Embrionarias/metabolismo , Fibroblastos/metabolismo , Factor Inhibidor de Leucemia/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/genética , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Feto/citología , Feto/metabolismo , Feto/fisiología , Fibroblastos/efectos de los fármacos , Fibroblastos/fisiología , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Factor 4 Similar a Kruppel , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/farmacología , Factor Inhibidor de Leucemia/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Análisis por Micromatrices , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Porcinos , Transfección
17.
EMBO J ; 31(2): 317-29, 2012 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-22056776

RESUMEN

The role of DNA sequence in determining chromatin state is incompletely understood. We have previously demonstrated that large chromosomal segments from human cells recapitulate their native chromatin state in mouse cells, but the relative contribution of local sequences versus their genomic context remains unknown. In this study, we compare orthologous chromosomal regions for which the human locus establishes prominent sites of Polycomb complex recruitment in pluripotent stem cells, whereas the corresponding mouse locus does not. Using recombination-mediated cassette exchange at the mouse locus, we establish the primacy of local sequences in the encoding of chromatin state. We show that the signal for chromatin bivalency is redundantly encoded across a bivalent domain and that this reflects competition between Polycomb complex recruitment and transcriptional activation. Furthermore, our results suggest that a high density of unmethylated CpG dinucleotides is sufficient for vertebrate Polycomb recruitment. This model is supported by analysis of DNA methyltransferase-deficient embryonic stem cells.


Asunto(s)
Islas de CpG/fisiología , Regulación de la Expresión Génica/genética , Proteínas Represoras/metabolismo , Globinas alfa/genética , Animales , Células Cultivadas/metabolismo , Cromatina/genética , Mapeo Cromosómico , Cromosomas Humanos Par 16 , Metilación de ADN , ADN Recombinante/genética , Células Madre Embrionarias/metabolismo , Humanos , Ratones , Ratones Transgénicos , Células Madre Pluripotentes/metabolismo , Proteínas del Grupo Polycomb , Recombinación Genética , Secuencias Reguladoras de Ácidos Nucleicos , Especificidad de la Especie , Transcripción Genética
18.
PLoS One ; 5(12): e14225, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21151976

RESUMEN

The rat is the preferred experimental animal in many biological studies. With the recent derivation of authentic rat embryonic stem (ES) cells it is now feasible to apply state-of-the art genetic engineering in this species using homologous recombination. To establish whether rat ES cells are amenable to in vivo recombination, we tested targeted disruption of the hypoxanthine phosphoribosyltransferase (hprt) locus in ES cells derived from both inbred and outbred strains of rats. Targeting vectors that replace exons 7 and 8 of the hprt gene with neomycinR/thymidine kinase selection cassettes were electroporated into male Fisher F344 and Sprague Dawley rat ES cells. Approximately 2% of the G418 resistant colonies also tolerated selection with 6-thioguanine, indicating inactivation of the hprt gene. PCR and Southern blot analysis confirmed correct site-specific targeting of the hprt locus in these clones. Embryoid body and monolayer differentiation of targeted cell lines established that they retained differentiation potential following targeting and selection. This report demonstrates that gene modification via homologous recombination in rat ES cells is efficient, and should facilitate implementation of targeted, genetic manipulation in the rat.


Asunto(s)
Células Madre Embrionarias/citología , Marcación de Gen/métodos , Técnicas Genéticas , Recombinación Genética , Animales , Animales Modificados Genéticamente , Blastocisto/citología , Células Cultivadas , Hipoxantina Fosforribosiltransferasa/genética , Masculino , Modelos Genéticos , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Especificidad de la Especie
19.
Cell ; 135(7): 1287-98, 2008 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-19109897

RESUMEN

Embryonic stem (ES) cells have been available from inbred mice since 1981 but have not been validated for other rodents. Failure to establish ES cells from a range of mammals challenges the identity of cultivated stem cells and our understanding of the pluripotent state. Here we investigated derivation of ES cells from the rat. We applied molecularly defined conditions designed to shield the ground state of authentic pluripotency from inductive differentiation stimuli. Undifferentiated cell lines developed that exhibited diagnostic features of ES cells including colonization of multiple tissues in viable chimeras. Definitive ES cell status was established by transmission of the cell line genome to offspring. Derivation of germline-competent ES cells from the rat paves the way to targeted genetic manipulation in this valuable biomedical model species. Rat ES cells will also provide a refined test-bed for functional evaluation of pluripotent stem cell-derived tissue repair and regeneration.


Asunto(s)
Masa Celular Interna del Blastocisto/citología , Células Madre Embrionarias/citología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Quimera , Femenino , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Glucógeno Sintasa Quinasas/antagonistas & inhibidores , Masculino , Ratones , Ratones SCID , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Ratas , Ratas Endogámicas F344 , Ratas Endogámicas
20.
Proc Natl Acad Sci U S A ; 105(24): 8292-7, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18544649

RESUMEN

The p110 isoforms of phosphoinositide 3-kinase (PI3K) are acutely regulated by extracellular stimuli. The class IA PI3K catalytic subunits (p110alpha, p110beta, and p110delta) occur in complex with a Src homology 2 (SH2) domain-containing p85 regulatory subunit, which has been shown to link p110alpha and p110delta to Tyr kinase signaling pathways. The p84/p101 regulatory subunits of the p110gamma class IB PI3K lack SH2 domains and instead couple p110gamma to G protein-coupled receptors (GPCRs). Here, we show, using small-molecule inhibitors with selectivity for p110beta and cells derived from a p110beta-deficient mouse line, that p110beta is not a major effector of Tyr kinase signaling but couples to GPCRs. In macrophages, both p110beta and p110gamma contributed to Akt activation induced by the GPCR agonist complement 5a, but not by the Tyr kinase ligand colony-stimulating factor-1. In fibroblasts, which express p110beta but not p110gamma, p110beta mediated Akt activation by the GPCR ligands stromal cell-derived factor, sphingosine-1-phosphate, and lysophosphatidic acid but not by the Tyr kinase ligands PDGF, insulin, and insulin-like growth factor 1. Introduction of p110gamma in these cells reduced the contribution of p110beta to GPCR signaling. Taken together, these data show that p110beta and p110gamma can couple redundantly to the same GPCR agonists. p110beta, which shows a much broader tissue distribution than the leukocyte-restricted p110gamma, could thus provide a conduit for GPCR-linked PI3K signaling in the many cell types where p110gamma expression is low or absent.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase I , Complemento C5a/farmacología , Fibroblastos/enzimología , Prueba de Complementación Genética , Isoenzimas/genética , Isoenzimas/metabolismo , Ligandos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/enzimología , Ratones , Ratones Mutantes , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...