Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Immunol ; 201(9): 2683-2699, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30249809

RESUMO

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.


Assuntos
Macrófagos , Microglia , Organogênese/genética , Ratos/crescimento & desenvolvimento , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/deficiência , Animais , Modelos Animais , Mutação , Ratos/genética
3.
Sci Rep ; 6: 25592, 2016 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-27185277

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Modelos Animais de Doenças , Dopamina/metabolismo , Síndrome de Lesch-Nyhan/metabolismo , Animais , Humanos , Hipoxantina Fosforribosiltransferase/deficiência , Hipoxantina Fosforribosiltransferase/genética , Síndrome de Lesch-Nyhan/genética , Masculino , Metabolômica/métodos , Camundongos Knockout , Mutação , Nucleotídeos de Purina/metabolismo , Ratos Transgênicos , Roedores , Espectrometria de Massas em Tandem
4.
Stem Cells ; 31(10): 2104-15, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23843312

RESUMO

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.


Assuntos
Células-Tronco Embrionárias/fisiologia , beta Catenina/metabolismo , Animais , Benzamidas/farmacologia , Fator de Transcrição CDX2 , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultura , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Proteínas Fetais/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas de Homeodomínio/metabolismo , Laminina/metabolismo , Camundongos , Piridinas/farmacologia , Pirimidinas/farmacologia , Ratos , Proteínas com Domínio T/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima , Via de Sinalização Wnt
5.
Cell Reprogram ; 14(2): 112-22, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22339199

RESUMO

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.


Assuntos
Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Fibroblastos/metabolismo , Fator Inibidor de Leucemia/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Reprogramação Celular/efeitos dos fármacos , Reprogramação Celular/genética , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Feto/citologia , Feto/metabolismo , Feto/fisiologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Células HEK293 , Humanos , Fator 4 Semelhante a Kruppel , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/farmacologia , Fator Inibidor de Leucemia/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Análise em Microsséries , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Suínos , Transfecção
6.
Cell Reprogram ; 14(1): 8-19, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22217199

RESUMO

Pluripotential stem cells from livestock offer an exciting prospect for the biotechnology industry. Applying strategies established for the derivation of murine induced pluripotential stem cells (iPSCs), we have isolated ovine iPSCs that can give rise to cells characteristic of all three germ cell layers both in vitro from embryoid bodies and in teratomas in vivo. Furthermore, although at a low level, these ovine iPS cells can contribute to live-born chimeric lambs. Colonies derived from ovine embryonic fibroblasts transfected with murine cMyc, Klf4, Oct4, and Sox2 displayed smooth domes with sharp edges when grown in human embryonic stem cell (ESC) medium but not in mouse ESC medium. These ovine iPSCs were alkaline phosphatase positive, expressed Nanog, and had a normal karyotype. These cells represent an important step in the understanding of mechanistic nature of pluripotency in ungulates.


Assuntos
Diferenciação Celular , Quimera/embriologia , Desenvolvimento Embrionário , Células-Tronco Pluripotentes/citologia , Ovinos/embriologia , Fosfatase Alcalina/metabolismo , Animais , Linhagem Celular , Proteínas de Homeodomínio/metabolismo , Humanos , Técnicas In Vitro , Cariótipo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Transfecção
7.
Nat Commun ; 2: 440, 2011 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-21863007

RESUMO

A major barrier to research on Parkinson's disease is inaccessibility of diseased tissue for study. One solution is to derive induced pluripotent stem cells from patients and differentiate them into neurons affected by disease. Triplication of SNCA, encoding α-synuclein, causes a fully penetrant, aggressive form of Parkinson's disease with dementia. α-Synuclein dysfunction is the critical pathogenic event in Parkinson's disease, multiple system atrophy and dementia with Lewy bodies. Here we produce multiple induced pluripotent stem cell lines from an SNCA triplication patient and an unaffected first-degree relative. When these cells are differentiated into midbrain dopaminergic neurons, those from the patient produce double the amount of α-synuclein protein as neurons from the unaffected relative, precisely recapitulating the cause of Parkinson's disease in these individuals. This model represents a new experimental system to identify compounds that reduce levels of α-synuclein, and to investigate the mechanistic basis of neurodegeneration caused by α-synuclein dysfunction.


Assuntos
Dosagem de Genes , Células-Tronco Pluripotentes Induzidas/metabolismo , Doença de Parkinson/genética , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Neurônios/metabolismo , Doença de Parkinson/metabolismo
8.
Cloning Stem Cells ; 10(2): 217-30, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18386992

RESUMO

Genetic modification of human embryonic stem cells (hESCs) will be an essential tool to allow full exploitation of these cells in regenerative medicine and in the study of hESC biology. Here we report multiple sequential modifications of an endogenous gene (hprt) in hESCs. A selectable marker flanked by heterospecific lox sites was first introduced by homologous recombination (HR) into the hprt gene. In a subsequent step, exchange of the selectable marker with another cassette was achieved by recombinase-mediated cassette exchange (RMCE). We show that 100% of the recovered clones were the result of RMCE using a promoter trap strategy at the hprt locus. hprt-targeted H1 cells maintained a diploid karyotype and expressed hESC surface markers before and after RMCE. Finally, we report a double replacement strategy using two sequential gene targeting steps resulting in the targeted correction of an hprt-mutated hESC line.


Assuntos
Células-Tronco Embrionárias/metabolismo , Marcação de Genes/métodos , Engenharia Genética/métodos , Hipoxantina Fosforribosiltransferase/genética , Recombinases/metabolismo , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Humanos , Hipoxantina Fosforribosiltransferase/metabolismo , Recombinação Genética , Transfecção
9.
Stem Cells ; 25(1): 10-8, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16960131

RESUMO

Although undifferentiated human embryonic stem cells (hESCs) are tumorigenic, this capacity is lost after differentiation, and hESCs are being widely investigated for applications in regenerative medicine. To engineer protection against the unintentional transplantation of undifferentiated cells, we generated hESCs carrying a construct in which the alpha1,3-galactosyltransferase (GalT) open reading frame was transcribed from the hTERT promoter (pmGT). Because the endogenous GalT gene is inactive, GalT expression was limited to undifferentiated cells. A second chimeric construct (pmfGT) differed by replacement of the GalT leader sequence for that of the fucosyltransferase gene. Two subclones containing stable integrations of pmGT and pmfGT (M2 and F11, respectively) were assessed for their response to human serum containing antibodies to the Galalpha1-3Galbeta1-4GlcNAc-R (alpha-gal) epitope. The low-variegation line, M2, and to a lesser extent the more variegated line F11, were sensitive to human serum when exposed in the undifferentiated state. However, M2 cells were largely insensitive after differentiation and retained both a normal karyotype and the ability to differentiate into derivatives of the three germ layers in severe combined immunodeficient mice. These data exemplify a method of protection against residual, undifferentiated hESCs prior to engraftment and may provide ongoing immune surveillance after engraftment against dedifferentiation or against de novo tumorigenesis involving hTERT reactivation. Untransfected H9 cells were not sensitive to the human serum used in this study. Hence, in our system, interactions of hESCs with other circulating antibodies, such as anti-Neu5Gc, were not observed.


Assuntos
Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/imunologia , Galactosiltransferases/genética , Oligossacarídeos/imunologia , Sistema ABO de Grupos Sanguíneos , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Clonagem Molecular , Primers do DNA , Humanos , Imunidade Inata , Cariotipagem , Camundongos , Camundongos SCID , Fases de Leitura Aberta , Valores de Referência , Transcrição Gênica , Transfecção
10.
J Biotechnol ; 125(2): 185-93, 2006 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-16621085

RESUMO

Gene targeting in livestock fibroblasts has proven difficult to achieve, particularly if the target gene is silent. We first tested whether efficient gene targeting at the transcriptionally active ovine alpha1(I) procollagen (COL1A1) locus required the use of a promoter trap vector. We compared gene targeting frequencies at the ovine COL1A1 locus using both a promoter trap and a non-promoter trap selection strategy. We demonstrated that targeted cells could be isolated regardless of whether an enrichment step (promoter trap) was used. Next, we used our optimised protocol to target a non-expressed gene, ovine beta-casein. We obtained clones that were scored positive by PCR for the targeting event, but were negative after cell expansion and Southern analysis. We propose that targeted cells were initially generated but that they were at a selective growth disadvantage during culture. We suggest modifications to the conventional targeting protocol that would prevent such loss of targeted cells.


Assuntos
Fibroblastos/metabolismo , Marcação de Genes/métodos , Animais , Southern Blotting , Células Cultivadas , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/citologia , Vetores Genéticos/genética , Modelos Genéticos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas/genética , Ovinos
11.
Mol Biotechnol ; 27(3): 231-44, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15247496

RESUMO

Additive transgenesis by pronuclear injection of the mouse zygote has been in use for more than 20 yr and gene targeting in mouse embryonic stem cells for almost as long. Together, these techniques have revolutionized animal biology by helping to unravel much of what we now know about gene function. Both additive transgenics and targeting can also be performed in livestock species but the impact has not yet been substantial. In part, this has been the result of the inefficiency of the techniques but-at least in agriculture-also to a lack of obvious practicality. This review assesses the extent to which this situation is changing, with particular reference to applications in biopharming, xenotransplantation, and large animal models.


Assuntos
Agricultura/métodos , Animais Domésticos/genética , Animais Geneticamente Modificados , Biofarmácia/métodos , Engenharia Genética/métodos , Engenharia Genética/veterinária , Transplante Heterólogo/métodos , Agricultura/tendências , Animais , Engenharia Biomédica/métodos , Engenharia Biomédica/tendências , Biofarmácia/tendências , Modelos Animais de Doenças , Engenharia Genética/tendências , Terapia Genética/métodos , Terapia Genética/tendências , Terapia Genética/veterinária , Humanos , Transplante Heterólogo/tendências , Transplante Heterólogo/veterinária
13.
Cloning Stem Cells ; 5(1): 51-62, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12713701

RESUMO

Human and mouse embryonic stem (ES) cells have the capacity to differentiate into derivatives of all three germ layers, suggesting novel therapies for degenerative, metabolic, and traumatic disorders. ES-based regenerative medicine will be further advanced by the development of reliable methods for transgene introduction and expression. Here, we show infection of human and mouse embryonic stem (ES) cells with two of the most popular vectors in gene transfer, adenovirus type 5 (Ad5) and adeno-associated virus (AAV; serotypes 2, 4, and 5). All vectors express the nuclear-localized marker gene beta-galactosidase expressed from the Rous Sarcoma Virus long terminal repeat (RSV-LTR). Both Ad5 and AAV2 infected human and mouse ES cells and gave rise to beta-galactosidase expression. AAV4 and 5 did not yield detectable levels of beta-galactosidase expression. Quantitative PCR analysis of virally infected human and mouse ES cells revealed that only Ad5 and AAV2 are capable of transducing both cell-types. No viral DNA was detected in cells infected with either AAV4 or AAV5. Infection and subsequent differentiation of mouse and human ES cells with Ad5 showed that beta-galactosidase-expressing cells were restricted to cells in the interior of the embryoid body mass. No beta-galactosidase expression was observed in AAV-infected cells following differentiation. There was no difference in morphology or differentiation patterns between infected and noninfected differentiating mouse and human ES cells. Differentiation of hES cells prior to infection led to transduction of neuronally differentiated cells with good efficiency using all vectors. These data show that Ad5- and AAV2-based vectors are capable of infecting both human and mouse ES cells, in both their undifferentiated and differentiated states, whereas AAV4 and AAV5 can infect human and mouse ES cells only following differentiation.


Assuntos
Adenoviridae/genética , Dependovirus/genética , Embrião de Mamíferos/citologia , Técnicas Genéticas , Células-Tronco/citologia , Animais , Diferenciação Celular , Vetores Genéticos , Humanos , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase , beta-Galactosidase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...