Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
CRISPR J ; 3(5): 388-397, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33095043

RESUMO

CRISPR-based synthetic gene drives have the potential to deliver a more effective and humane method of invasive vertebrate pest control than current strategies. Relatively efficient CRISPR gene drive systems have been developed in insects and yeast but not in mammals. Here, we investigated the efficiency of CRISPR-Cas9-based gene drives in Mus musculus by constructing "split drive" systems where gRNA expression occurs on a separate chromosome to Cas9, which is under the control of either a zygotic (CAG) or germline (Vasa) promoter. While both systems generated double-strand breaks at their intended target site in vivo, no homology-directed repair between chromosomes ("homing") was detectable. Our data indicate that robust and specific Cas9 expression during meiosis is a critical requirement for the generation of efficient CRISPR-based synthetic gene drives in rodents.


Assuntos
Proteína 9 Associada à CRISPR/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Tecnologia de Impulso Genético , Genes Sintéticos , Meiose , Zigoto , Animais , Proteína 9 Associada à CRISPR/genética , Feminino , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Transgênicos , Modelos Animais , Regiões Promotoras Genéticas , RNA Guia de Cinetoplastídeos/metabolismo , Reparo de DNA por Recombinação
2.
Sci Rep ; 10(1): 6751, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317665

RESUMO

SOX3 is a transcription factor expressed within the developing and adult nervous system where it mostly functions to help maintain neural precursors. Sox3 is also expressed in other locations, notably within the spermatogonial stem/progenitor cell population in postnatal testis. Independent studies have shown that Sox3 null mice exhibit a spermatogenic block as young adults, the mechanism of which remains poorly understood. Using a panel of spermatogonial cell marker genes, we demonstrate that Sox3 is expressed within the committed progenitor fraction of the undifferentiated spermatogonial pool. Additionally, we use a Sox3 null mouse model to define a potential role for this factor in progenitor cell function. We demonstrate that Sox3 expression is required for transition of undifferentiated cells from a GFRα1+ self-renewing state to the NGN3 + transit-amplifying compartment. Critically, using chromatin immunoprecipitation, we demonstrate that SOX3 binds to a highly conserved region in the Ngn3 promoter region in vivo, indicating that Ngn3 is a direct target of SOX3. Together these studies indicate that SOX3 functions as a pro-commitment factor in spermatogonial stem/progenitor cells.


Assuntos
Células-Tronco Germinativas Adultas/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Regiões Promotoras Genéticas , Fatores de Transcrição SOXB1/genética , Espermatogônias/metabolismo , Testículo/metabolismo , Células-Tronco Germinativas Adultas/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial , Masculino , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , Ligação Proteica , Fatores de Transcrição SOXB1/deficiência , Transdução de Sinais , Espermatogênese/genética , Espermatogônias/citologia , Espermatogônias/crescimento & desenvolvimento , Testículo/citologia , Testículo/crescimento & desenvolvimento , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Neurobiol Dis ; 134: 104640, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31639411

RESUMO

Mutations in the GAP activity toward RAGs 1 (GATOR1) complex genes (DEPDC5, NPRL2 and NPRL3) have been associated with focal epilepsy and focal cortical dysplasia (FCD). GATOR1 functions as an inhibitor of the mTORC1 signalling pathway, indicating that the downstream effects of mTORC1 deregulation underpin the disease. However, the vast majority of putative disease-causing variants have not been functionally assessed for mTORC1 repression activity. Here, we develop a novel in vitro functional assay that enables rapid assessment of GATOR1-gene variants. Surprisingly, of the 17 variants tested, we show that only six showed significantly impaired mTORC1 inhibition. To further investigate variant function in vivo, we generated a conditional Depdc5 mouse which modelled a 'second-hit' mechanism of disease. Generation of Depdc5 null 'clones' in the embryonic brain resulted in mTORC1 hyperactivity and modelled epilepsy and FCD symptoms including large dysmorphic neurons, defective migration and lower seizure thresholds. Using this model, we validated DEPDC5 variant F164del to be loss-of-function. We also show that Q542P is not functionally compromised in vivo, consistent with our in vitro findings. Overall, our data show that mTORC1 deregulation is the central pathological mechanism for GATOR1 variants and also indicates that a significant proportion of putative disease variants are pathologically inert, highlighting the importance of GATOR1 variant functional assessment.


Assuntos
Epilepsias Parciais/metabolismo , Epilepsia/metabolismo , Proteínas Ativadoras de GTPase/genética , Malformações do Desenvolvimento Cortical do Grupo I/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Animais , Epilepsias Parciais/genética , Epilepsia/genética , Proteínas Ativadoras de GTPase/metabolismo , Técnicas Genéticas , Células HEK293 , Humanos , Malformações do Desenvolvimento Cortical do Grupo I/genética , Camundongos , Camundongos Knockout , Mutação
4.
Neuron ; 97(1): 59-66.e5, 2018 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-29301106

RESUMO

X-linked diseases typically exhibit more severe phenotypes in males than females. In contrast, protocadherin 19 (PCDH19) mutations cause epilepsy in heterozygous females but spare hemizygous males. The cellular mechanism responsible for this unique pattern of X-linked inheritance is unknown. We show that PCDH19 contributes to adhesion specificity in a combinatorial manner such that mosaic expression of Pcdh19 in heterozygous female mice leads to striking sorting between cells expressing wild-type (WT) PCDH19 and null PCDH19 in the developing cortex, correlating with altered network activity. Complete deletion of PCDH19 in heterozygous mice abolishes abnormal cell sorting and restores normal network activity. Furthermore, we identify variable cortical malformations in PCDH19 epilepsy patients. Our results highlight the role of PCDH19 in determining cell adhesion affinities during cortical development and the way segregation of WT and null PCDH19 cells is associated with the unique X-linked inheritance of PCDH19 epilepsy.


Assuntos
Caderinas/genética , Movimento Celular/genética , Córtex Cerebral/anormalidades , Epilepsia/genética , Animais , Córtex Cerebral/embriologia , Epilepsia/embriologia , Feminino , Genes Ligados ao Cromossomo X , Humanos , Masculino , Camundongos , Células-Tronco Neurais/metabolismo , Protocaderinas
5.
N Engl J Med ; 377(6): 544-552, 2017 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-28792876

RESUMO

BACKGROUND: Congenital malformations can be manifested as combinations of phenotypes that co-occur more often than expected by chance. In many such cases, it has proved difficult to identify a genetic cause. We sought the genetic cause of cardiac, vertebral, and renal defects, among others, in unrelated patients. METHODS: We used genomic sequencing to identify potentially pathogenic gene variants in families in which a person had multiple congenital malformations. We tested the function of the variant by using assays of in vitro enzyme activity and by quantifying metabolites in patient plasma. We engineered mouse models with similar variants using the CRISPR (clustered regularly interspaced short palindromic repeats)-Cas9 system. RESULTS: Variants were identified in two genes that encode enzymes of the kynurenine pathway, 3-hydroxyanthranilic acid 3,4-dioxygenase (HAAO) and kynureninase (KYNU). Three patients carried homozygous variants predicting loss-of-function changes in the HAAO or KYNU proteins (HAAO p.D162*, HAAO p.W186*, or KYNU p.V57Efs*21). Another patient carried heterozygous KYNU variants (p.Y156* and p.F349Kfs*4). The mutant enzymes had greatly reduced activity in vitro. Nicotinamide adenine dinucleotide (NAD) is synthesized de novo from tryptophan through the kynurenine pathway. The patients had reduced levels of circulating NAD. Defects similar to those in the patients developed in the embryos of Haao-null or Kynu-null mice owing to NAD deficiency. In null mice, the prevention of NAD deficiency during gestation averted defects. CONCLUSIONS: Disruption of NAD synthesis caused a deficiency of NAD and congenital malformations in humans and mice. Niacin supplementation during gestation prevented the malformations in mice. (Funded by the National Health and Medical Research Council of Australia and others.).


Assuntos
3-Hidroxiantranilato 3,4-Dioxigenase/genética , Anormalidades Congênitas/genética , Suplementos Nutricionais , Hidrolases/genética , NAD/deficiência , Niacina/uso terapêutico , 3-Hidroxiantranilato 3,4-Dioxigenase/metabolismo , Canal Anal/anormalidades , Animais , Anormalidades Congênitas/prevenção & controle , Modelos Animais de Doenças , Esôfago/anormalidades , Feminino , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/prevenção & controle , Humanos , Hidrolases/metabolismo , Rim/anormalidades , Deformidades Congênitas dos Membros/genética , Deformidades Congênitas dos Membros/prevenção & controle , Masculino , Camundongos , Camundongos Knockout , Mutação , NAD/biossíntese , NAD/genética , Análise de Sequência de DNA , Coluna Vertebral/anormalidades , Traqueia/anormalidades
6.
Metallomics ; 8(8): 762-73, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27460862

RESUMO

NAMI-A and KP1019 are Ru(III)-based anti-metastatic and cytotoxic anti-cancer drugs, respectively, and have been proposed to be activated by reduction to Ru(II). The potential reduction of NAMI-A and KP1019 in the hypoxic environment of a tumour model of neuroblastoma was examined. Normoxic, hypoxic and necrotic tumour tissues were modelled by multicellular spheroids of SH-SY5Y human neuroblastoma cells of various diameters (50-800 µm). The variation in spheroid environment was confirmed with pimonidazole staining. Laser-ablation inductively-coupled plasma mass spectrometry showed KP1019 and NAMI-A penetration into the spheroid hypoxic region. XANES showed that the speciation of NAMI-A biotransformation products did not change significantly as hypoxia levels increased. KP1019 metabolites showed a correlation between the degree of spheroid hypoxia and the Ru K-edge energy consistent with either partial reduction of Ru(III) to Ru(II) in tumour microenvironments, increased S/Cl coordination or a reduced fraction of polynuclear Ru species. EXAFS spectroscopy was undertaken in an attempt to distinguish between these scenarios but was inconclusive.


Assuntos
Antineoplásicos/farmacologia , Dimetil Sulfóxido/análogos & derivados , Hipóxia/fisiopatologia , Indazóis/farmacologia , Neuroblastoma/patologia , Compostos Organometálicos/farmacologia , Rutênio/farmacologia , Esferoides Celulares/patologia , Antineoplásicos/química , Dimetil Sulfóxido/química , Dimetil Sulfóxido/farmacologia , Humanos , Indazóis/química , Neuroblastoma/tratamento farmacológico , Compostos Organometálicos/química , Rutênio/química , Compostos de Rutênio , Esferoides Celulares/efeitos dos fármacos , Células Tumorais Cultivadas , Microambiente Tumoral/efeitos dos fármacos , Espectroscopia por Absorção de Raios X
7.
Sci Rep ; 6: 26765, 2016 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-27240640

RESUMO

Protocadherin 19 (Pcdh19) is an X-linked gene belonging to the protocadherin superfamily, whose members are predominantly expressed in the central nervous system and have been implicated in cell-cell adhesion, axon guidance and dendrite self-avoidance. Heterozygous loss-of-function mutations in humans result in the childhood epilepsy disorder PCDH19 Girls Clustering Epilepsy (PCDH19 GCE) indicating that PCDH19 is required for brain development. However, understanding PCDH19 function in vivo has proven challenging and has not been studied in mammalian models. Here, we validate a murine Pcdh19 null allele in which a ß-Geo reporter cassette is expressed under the control of the endogenous promoter. Analysis of ß-Geo reporter activity revealed widespread but restricted expression of PCDH19 in embryonic, postnatal and adult brains. No gross morphological defects were identified in Pcdh19(+/ß-Geo) and Pcdh19(Y/ß-Geo) brains and the location of Pcdh19 null cells was normal. However, in vitro migration assays revealed that the motility of Pcdh19 null neurons was significantly elevated, potentially contributing to pathogenesis in patients with PCDH19 mutations. Overall our initial characterization of Pcdh19(+/ß-Geo), Pcdh19(ß-Geo/ß-Geo) and Pcdh19(Y/ß-Geo)mice reveals that despite widespread expression of Pcdh19 in the CNS, and its role in human epilepsy, its function in mice is not essential for brain development.


Assuntos
Encéfalo/crescimento & desenvolvimento , Caderinas/fisiologia , Movimento Celular , Neurônios/fisiologia , Animais , Encéfalo/metabolismo , Caderinas/genética , Células Cultivadas , Epilepsia/genética , Feminino , Genótipo , Hipocampo/crescimento & desenvolvimento , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Células-Tronco Neurais , Neurônios/metabolismo , Fenótipo , Protocaderinas , Sinapses/metabolismo
8.
Clin Endocrinol (Oxf) ; 85(4): 609-15, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27146357

RESUMO

OBJECTIVES: IGSF1 deficiency syndrome (IDS) is a recently described X-linked congenital central hypothyroidism disorder characterized by loss-of-function mutations in the immunoglobulin superfamily member 1 (IGSF1) gene. The phenotypic spectrum and intrafamilial variability associated with IDS remain unclear due to a paucity of large, well-characterized pedigrees. Here, we present phenotypic analysis and molecular characterization of a five-generation pedigree with IGSF1 deficiency containing 10 affected males. PATIENTS AND METHODS: Pituitary function was assessed in all available family members (n = 8 affected males and n = 5 carrier females). Molecular characterization of the family was performed by Sanger sequencing of PCR products amplified from the IGSF1 locus and by array comparative genomic hybridization. RESULTS: A 42-kb IGSF1 deletion spanning the entire coding sequence was identified in all affected males. TSH deficiency, although subclinical in one case, was identified in all affected males (n = 8). PRL and GH deficiency were also present in 5 of 6 and 4 of 8 affected males, respectively. In contrast to previous reports, macroorchidism was not detected in any of the four affected males who were examined for this feature. Only 1 of 5 carrier females had pituitary dysfunction (TSH and GH deficiency). CONCLUSION: Individuals with identical IGSF1 deletions can exhibit variable pituitary hormone deficiencies, of which overt TSH deficiency is the most consistent feature. We also show that macroorchidism is not obligatory in males with IDS. Mutations of IGSF1 should therefore be considered in males with isolated hypopituitarism that includes TSH deficiency.


Assuntos
Hipotireoidismo Congênito/genética , Doenças Genéticas Ligadas ao Cromossomo X , Imunoglobulinas/genética , Proteínas de Membrana/genética , Deleção de Sequência , Hibridização Genômica Comparativa , Feminino , Humanos , Hipopituitarismo/genética , Masculino , Mutação , Linhagem
9.
Nat Struct Mol Biol ; 22(12): 1008-15, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26571108

RESUMO

Polyglutamine (polyGln) expansions in nine human proteins result in neurological diseases and induce the proteins' tendency to form ß-rich amyloid fibrils and intracellular deposits. Less well known are at least nine other human diseases caused by polyalanine (polyAla)-expansion mutations in different proteins. The mechanisms of how polyAla aggregates under physiological conditions remain unclear and controversial. We show here that aggregation of polyAla is mechanistically dissimilar to that of polyGln and hence does not exhibit amyloid kinetics. PolyAla assembled spontaneously into α-helical clusters with diverse oligomeric states. Such clustering was pervasive in cells irrespective of visible aggregate formation, and it disrupted the normal physiological oligomeric state of two human proteins natively containing polyAla: ARX and SOX3. This self-assembly pattern indicates that polyAla expansions chronically disrupt protein behavior by imposing a deranged oligomeric status.


Assuntos
Amiloide/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Agregação Patológica de Proteínas , Multimerização Proteica , Humanos , Estrutura Secundária de Proteína
10.
Methods Mol Biol ; 1017: 135-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23719913

RESUMO

Disease-causing polyalanine (PA) expansion mutations have been identified in nine genes, eight of which encode transcription factors (TFs) with important roles in development. In vitro and cell overexpression studies have shown that expanded PA tracts result in protein misfolding and the formation of aggregates. This feature of PA proteins is reminiscent of the related polyglutamine (PQ) disease proteins, which have been shown to cause disease via a gain-of-function (GOF) mechanism. However, in sharp contrast to PQ disorders, the disease phenotypes associated with PA mutations are more consistent with a LOF and/or mild GOF mechanism, suggesting that their molecular pathology is inherently different to PQ disorders. Elucidating the cellular impact of PA mutations in vivo has been difficult to address as, unlike the late-onset polyglutamine disorders, all PA disorders associated with TF gene mutations are congenital. However, in recent years, significant advances have been made through the analysis of engineered (knock-in) and spontaneous PA mouse models. Here we review these recent findings and propose an updated model of the molecular and cellular mechanism of PA disorders that incorporates both LOF and GOF features.


Assuntos
Peptídeos , Deficiências na Proteostase , Fatores de Transcrição , Expansão das Repetições de Trinucleotídeos , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Peptídeos/genética , Peptídeos/metabolismo , Deficiências na Proteostase/genética , Deficiências na Proteostase/metabolismo , Deficiências na Proteostase/patologia , Deficiências na Proteostase/fisiopatologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
11.
Nat Genet ; 45(5): 546-51, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23542697

RESUMO

The majority of epilepsies are focal in origin, with seizures emanating from one brain region. Although focal epilepsies often arise from structural brain lesions, many affected individuals have normal brain imaging. The etiology is unknown in the majority of individuals, although genetic factors are increasingly recognized. Autosomal dominant familial focal epilepsy with variable foci (FFEVF) is notable because family members have seizures originating from different cortical regions. Using exome sequencing, we detected DEPDC5 mutations in two affected families. We subsequently identified mutations in five of six additional published large families with FFEVF. Study of families with focal epilepsy that were too small for conventional clinical diagnosis with FFEVF identified DEPDC5 mutations in approximately 12% of families (10/82). This high frequency establishes DEPDC5 mutations as a common cause of familial focal epilepsies. Shared homology with G protein signaling molecules and localization in human neurons suggest a role of DEPDC5 in neuronal signal transduction.


Assuntos
Epilepsias Parciais/genética , Exoma/genética , Predisposição Genética para Doença/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Mutação/genética , Proteínas Repressoras/genética , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Células Cultivadas , Criança , Pré-Escolar , Estudos de Coortes , Biologia Computacional , Epilepsias Parciais/diagnóstico , Feminino , Imunofluorescência , Proteínas Ativadoras de GTPase , Ligação Genética , Genótipo , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Neurônios/citologia , Neurônios/metabolismo , Linhagem , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Adulto Jovem
12.
PLoS One ; 7(6): e38645, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22701686

RESUMO

Two lineages of endoderm develop during mammalian embryogenesis, the primitive endoderm in the pre-implantation blastocyst and the definitive endoderm at gastrulation. This complexity of endoderm cell populations is mirrored during pluripotent cell differentiation in vitro and has hindered the identification and purification of the definitive endoderm for use as a substrate for further differentiation. The aggregation and differentiation of early primitive ectoderm-like (EPL) cells, resulting in the formation of EPL-cell derived embryoid bodies (EPLEBs), is a model of gastrulation that progresses through the sequential formation of primitive streak-like intermediates to nascent mesoderm and more differentiated mesoderm populations. EPL cell-derived EBs have been further analysed for the formation of definitive endoderm by detailed morphological studies, gene expression and a protein uptake assay. In comparison to embryoid bodies derived from ES cells, which form primitive and definitive endoderm, the endoderm compartment of embryoid bodies formed from EPL cells was comprised almost exclusively of definitive endoderm. Definitive endoderm was defined as a population of squamous cells that expressed Sox17, CXCR4 and Trh, which formed without the prior formation of primitive endoderm and was unable to endocytose horseradish peroxidase from the medium. Definitive endoderm formed in EPLEBs provides a substrate for further differentiation into specific endoderm lineages; these lineages can be used as research tools for understanding the mechanisms controlling lineage establishment and the nature of the transient intermediates formed. The similarity between mouse EPL cells and human ES cells suggests EPLEBs can be used as a model system for the development of technologies to enrich for the formation of human ES cell-derived definitive endoderm in the future.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Corpos Embrioides/ultraestrutura , Endoderma/ultraestrutura , Mesoderma/ultraestrutura , Células-Tronco Pluripotentes/ultraestrutura , Linha Primitiva/ultraestrutura , Animais , Primers do DNA/genética , Citometria de Fluxo , Perfilação da Expressão Gênica , Peroxidase do Rábano Silvestre/farmacocinética , Camundongos , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Células-Tronco Pluripotentes/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
13.
Am J Hum Genet ; 90(1): 152-60, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22243967

RESUMO

Benign familial infantile epilepsy (BFIE) is a self-limited seizure disorder that occurs in infancy and has autosomal-dominant inheritance. We have identified heterozygous mutations in PRRT2, which encodes proline-rich transmembrane protein 2, in 14 of 17 families (82%) affected by BFIE, indicating that PRRT2 mutations are the most frequent cause of this disorder. We also report PRRT2 mutations in five of six (83%) families affected by infantile convulsions and choreoathetosis (ICCA) syndrome, a familial syndrome in which infantile seizures and an adolescent-onset movement disorder, paroxysmal kinesigenic choreoathetosis (PKC), co-occur. These findings show that mutations in PRRT2 cause both epilepsy and a movement disorder. Furthermore, PRRT2 mutations elicit pleiotropy in terms of both age of expression (infancy versus later childhood) and anatomical substrate (cortex versus basal ganglia).


Assuntos
Atetose/genética , Coreia/genética , Epilepsia Neonatal Benigna/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Convulsões/genética , Idade de Início , Animais , Sequência de Bases , Encéfalo/patologia , Pré-Escolar , Cromossomos Humanos Par 16/genética , Humanos , Lactente , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Linhagem
14.
J Cell Sci ; 123(Pt 10): 1796-804, 2010 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-20427322

RESUMO

The formation and differentiation of multipotent precursors underlies the generation of cell diversity during mammalian development. Recognition and analysis of these transient cell populations has been hampered by technical difficulties in accessing them in vivo. In vitro model systems, based on the differentiation of embryonic stem (ES) cells, provide an alternative means of identifying and characterizing these populations. Using a previously established mouse ES-cell-based system that recapitulates the development of the ectoderm lineage we have identified a transient population that is consistent with definitive ectoderm. This previously unidentified progenitor occurs as a temporally discrete population during ES cell differentiation, and differs from the preceding and succeeding populations in gene expression and differentiation potential, with the unique ability to form surface ectoderm in response to BMP4 signalling.


Assuntos
Antígenos de Diferenciação/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Ectoderma/embriologia , Neurogênese , Animais , Antígenos de Diferenciação/genética , Proteína Morfogenética Óssea 4/genética , Linhagem Celular , Linhagem da Célula , Embrião de Mamíferos , Células-Tronco Embrionárias , Imunofluorescência , Perfilação da Expressão Gênica , Camundongos , Transdução de Sinais/genética , Proteínas Smad/metabolismo
15.
Stem Cells ; 27(12): 2941-51, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19750540

RESUMO

gamma-Secretase is a membrane-associated protease with multiple intracellular targets, a number of which have been shown to influence embryonic development and embryonic stem (ES) cell differentiation. This paper describes the use of the gamma-secretase inhibitor N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester (DAPT) to evaluate the role of gamma-secretase in the differentiation of pluripotent stem cells to the germ lineages. The addition of DAPT did not prevent the formation of primitive ectoderm-like cells from ES cells in culture. In contrast, the addition of DAPT during primitive ectoderm-like cell differentiation interfered with the ability of both serum and BMP4 to induce a primitive streak-like intermediate and resulted in the preferential formation of neurectoderm. Similarly, DAPT reduced the formation of primitive streak-like intermediates from differentiating human ES cells; the culture conditions used resulted in a population enriched in human surface ectoderm. These data suggest that gamma-secretase may form part of the general pathway by which mesoderm is specified within the primitive streak. The addition of an E-cadherin neutralizing antibody was able to partially reverse the effect of DAPT, suggesting that DAPT may be preventing the formation of primitive streak-like intermediates and promoting neurectoderm differentiation by stabilizing E-cadherin and preventing its proteolysis.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/enzimologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Proteína Morfogenética Óssea 4/metabolismo , Caderinas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Dipeptídeos/farmacologia , Células-Tronco Embrionárias/efeitos dos fármacos , Humanos , Camundongos , Células-Tronco Pluripotentes/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Transdução de Sinais
16.
PLoS One ; 4(5): e5579, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19440553

RESUMO

In the mammal, the pluripotent cells of embryo differentiate and commit to either the mesoderm/endoderm lineages or the ectoderm lineage during gastrulation. In culture, the ability to direct lineage choice from pluripotent cells into the mesoderm/endoderm or ectoderm lineages will enable the development of technologies for the formation of highly enriched or homogenous populations of cells. Here we show that manipulation of cell:cell contact and a mesoderm suppressing activity in culture affects the outcome of pluripotent cell differentiation and when both variables are manipulated appropriately they can direct differentiation to either the mesoderm or ectoderm lineage. The disruption of cell:cell contacts and removal of a mesoderm suppressor activity results in the differentiation of pluripotent, primitive ectoderm-like cells to the mesoderm lineage, while maintenance of cell:cell contacts and inclusion, within the culture medium, of a mesoderm suppressing activity results in the formation of near homogenous populations of ectoderm. Understanding the contribution of these variables in lineage choice provides a framework for the development of directed differentiation protocols that result in the formation of specific cell populations from pluripotent cells in culture.


Assuntos
Comunicação Celular/fisiologia , Linhagem da Célula , Ectoderma/citologia , Mesoderma/citologia , Células-Tronco/citologia , Animais , Comunicação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Meios de Cultura/farmacologia , Ectoderma/metabolismo , Citometria de Fluxo , Mesoderma/metabolismo , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ultrafiltração
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...