Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell ; 153(2): 335-47, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23582324

RESUMO

Factors that sustain self-renewal of mouse embryonic stem cells (ESCs) are well described. In contrast, the machinery regulating exit from pluripotency is ill defined. In a large-scale small interfering RNA (siRNA) screen, we found that knockdown of the tumor suppressors Folliculin (Flcn) and Tsc2 prevent ESC commitment. Tsc2 lies upstream of mammalian target of rapamycin (mTOR), whereas Flcn acts downstream and in parallel. Flcn with its interaction partners Fnip1 and Fnip2 drives differentiation by restricting nuclear localization and activity of the bHLH transcription factor Tfe3. Conversely, enforced nuclear Tfe3 enables ESCs to withstand differentiation conditions. Genome-wide location and functional analyses showed that Tfe3 directly integrates into the pluripotency circuitry through transcriptional regulation of Esrrb. These findings identify a cell-intrinsic rheostat for destabilizing ground-state pluripotency to allow lineage commitment. Congruently, stage-specific subcellular relocalization of Tfe3 suggests that Flcn-Fnip1/2 contributes to developmental progression of the pluripotent epiblast in vivo.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Redes Reguladoras de Genes , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Transporte/metabolismo , Células-Tronco Embrionárias/metabolismo , Estrona/genética , Estrona/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Serina-Treonina Quinases TOR/metabolismo
2.
EMBO J ; 40(12): e106818, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-33909924

RESUMO

Mouse embryonic stem cells (mESCs) are biased toward producing embryonic rather than extraembryonic endoderm fates. Here, we identify the mechanism of this barrier and report that the histone deacetylase Hdac3 and the transcriptional corepressor Dax1 cooperatively limit the lineage repertoire of mESCs by silencing an enhancer of the extraembryonic endoderm-specifying transcription factor Gata6. This restriction is opposed by the pluripotency transcription factors Nr5a2 and Esrrb, which promote cell type conversion. Perturbation of the barrier extends mESC potency and allows formation of 3D spheroids that mimic the spatial segregation of embryonic epiblast and extraembryonic endoderm in early embryos. Overall, this study shows that transcriptional repressors stabilize pluripotency by biasing the equilibrium between embryonic and extraembryonic lineages that is hardwired into the mESC transcriptional network.


Assuntos
Receptor Nuclear Órfão DAX-1 , Histona Desacetilases , Células-Tronco Embrionárias Murinas/citologia , Animais , Diferenciação Celular , Células Cultivadas , Receptor Nuclear Órfão DAX-1/genética , Receptor Nuclear Órfão DAX-1/metabolismo , Feminino , Fator de Transcrição GATA6/genética , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Masculino , Camundongos , RNA Interferente Pequeno/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo
3.
EMBO J ; 39(2): e102591, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31782544

RESUMO

Developmental cell fate specification is a unidirectional process that can be reverted in response to injury or experimental reprogramming. Whether differentiation and de-differentiation trajectories intersect mechanistically is unclear. Here, we performed comparative screening in lineage-related mouse naïve embryonic stem cells (ESCs) and primed epiblast stem cells (EpiSCs), and identified the constitutively expressed zinc finger transcription factor (TF) Zfp281 as a bidirectional regulator of cell state interconversion. We showed that subtle chromatin binding changes in differentiated cells translate into activation of the histone H3 lysine 9 (H3K9) methyltransferase Ehmt1 and stabilization of the zinc finger TF Zic2 at enhancers and promoters. Genetic gain-of-function and loss-of-function experiments confirmed a critical role of Ehmt1 and Zic2 downstream of Zfp281 both in driving exit from the ESC state and in restricting reprogramming of EpiSCs. Our study reveals that cell type-invariant chromatin association of Zfp281 provides an interaction platform for remodeling the cis-regulatory network underlying cellular plasticity.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Histona-Lisina N-Metiltransferase/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Cromatina/química , Cromatina/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/metabolismo
4.
Nature ; 557(7707): 739-743, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29795351

RESUMO

De novo mutations in ADNP, which encodes activity-dependent neuroprotective protein (ADNP), have recently been found to underlie Helsmoortel-Van der Aa syndrome, a complex neurological developmental disorder that also affects several other organ functions 1 . ADNP is a putative transcription factor that is essential for embryonic development 2 . However, its precise roles in transcriptional regulation and development are not understood. Here we show that ADNP interacts with the chromatin remodeller CHD4 and the chromatin architectural protein HP1 to form a stable complex, which we refer to as ChAHP. Besides mediating complex assembly, ADNP recognizes DNA motifs that specify binding of ChAHP to euchromatin. Genetic ablation of ChAHP components in mouse embryonic stem cells results in spontaneous differentiation concomitant with premature activation of lineage-specific genes and in a failure to differentiate towards the neuronal lineage. Molecularly, ChAHP-mediated repression is fundamentally different from canonical HP1-mediated silencing: HP1 proteins, in conjunction with histone H3 lysine 9 trimethylation (H3K9me3), are thought to assemble broad heterochromatin domains that are refractory to transcription. ChAHP-mediated repression, however, acts in a locally restricted manner by establishing inaccessible chromatin around its DNA-binding sites and does not depend on H3K9me3-modified nucleosomes. Together, our results reveal that ADNP, via the recruitment of HP1 and CHD4, regulates the expression of genes that are crucial for maintaining distinct cellular states and assures accurate cell fate decisions upon external cues. Such a general role of ChAHP in governing cell fate plasticity may explain why ADNP mutations affect several organs and body functions and contribute to cancer progression1,3,4. Notably, we found that the integrity of the ChAHP complex is disrupted by nonsense mutations identified in patients with Helsmoortel-Van der Aa syndrome, and this could be rescued by aminoglycosides that suppress translation termination 5 . Therefore, patients might benefit from therapeutic agents that are being developed to promote ribosomal read-through of premature stop codons6,7.


Assuntos
Linhagem da Célula/genética , Proteínas Cromossômicas não Histona/metabolismo , DNA Helicases/metabolismo , Proteínas de Homeodomínio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Eucromatina/genética , Eucromatina/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Nucleossomos/metabolismo , Ligação Proteica , Proteínas Repressoras/metabolismo , Transcrição Gênica
5.
J Biol Chem ; 297(2): 100947, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34270961

RESUMO

Transcription factors (TFs) harboring broad-complex, tramtrack, and bric-a-brac (BTB) domains play important roles in development and disease. These BTB domains are thought to recruit transcriptional modulators to target DNA regions. However, a systematic molecular understanding of the mechanism of action of this TF family is lacking. Here, we identify the zinc finger BTB-TF Zbtb2 from a genetic screen for regulators of exit from pluripotency and demonstrate that its absence perturbs embryonic stem cell differentiation and the gene expression dynamics underlying peri-implantation development. We show that ZBTB2 binds the chromatin remodeler Ep400 to mediate downstream transcription. Independently, the BTB domain directly interacts with nucleosome remodeling and deacetylase and histone chaperone histone regulator A. Nucleosome remodeling and deacetylase recruitment is a common feature of BTB TFs, and based on phylogenetic analysis, we propose that this is a conserved evolutionary property. Binding to UBN2, in contrast, is specific to ZBTB2 and requires a C-terminal extension of the BTB domain. Taken together, this study identifies a BTB-domain TF that recruits chromatin modifiers and a histone chaperone during a developmental cell state transition and defines unique and shared molecular functions of the BTB-domain TF family.


Assuntos
Proteínas Repressoras , Fatores de Transcrição , Domínio BTB-POZ , Chaperonas de Histonas , Humanos , Filogenia , Dedos de Zinco
6.
J Med Genet ; 57(12): 808-819, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32409512

RESUMO

INTRODUCTION: Pigmentary mosaicism (PM) manifests by pigmentation anomalies along Blaschko's lines and represents a clue toward the molecular diagnosis of syndromic intellectual disability (ID). Together with new insights on the role for lysosomal signalling in embryonic stem cell differentiation, mutations in the X-linked transcription factor 3 (TFE3) have recently been reported in five patients. Functional analysis suggested these mutations to result in ectopic nuclear gain of functions. MATERIALS AND METHODS: Subsequent data sharing allowed the clustering of de novo TFE3 variants identified by exome sequencing on DNA extracted from leucocytes in patients referred for syndromic ID with or without PM. RESULTS: We describe the detailed clinical and molecular data of 17 individuals harbouring a de novo TFE3 variant, including the patients that initially allowed reporting TFE3 as a new disease-causing gene. The 12 females and 5 males presented with pigmentation anomalies on Blaschko's lines, severe ID, epilepsy, storage disorder-like features, growth retardation and recognisable facial dysmorphism. The variant was at a mosaic state in at least two male patients. All variants were missense except one splice variant. Eleven of the 13 variants were localised in exon 4, 2 in exon 3, and 3 were recurrent variants. CONCLUSION: This series further delineates the specific storage disorder-like phenotype with PM ascribed to de novo TFE3 mutation in exons 3 and 4. It confirms the identification of a novel X-linked human condition associated with mosaicism and dysregulation within the mechanistic target of rapamycin (mTOR) pathway, as well as a link between lysosomal signalling and human development.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Epilepsia/genética , Deficiência Intelectual/genética , Transtornos da Pigmentação/genética , Adolescente , Adulto , Criança , Pré-Escolar , Epilepsia/complicações , Epilepsia/patologia , Feminino , Genes Ligados ao Cromossomo X/genética , Humanos , Lactente , Deficiência Intelectual/complicações , Deficiência Intelectual/patologia , Masculino , Mosaicismo , Patologia Molecular/normas , Transtornos da Pigmentação/complicações , Transtornos da Pigmentação/patologia , Sequenciamento do Exoma , Adulto Jovem
7.
Nature ; 454(7201): 241-5, 2008 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-18528333

RESUMO

Drosophila neuroblasts and ovarian stem cells are well characterized models for stem cell biology. In both cell types, one daughter cell self-renews continuously while the other undergoes a limited number of divisions, stops to proliferate mitotically and differentiates. Whereas neuroblasts segregate the Trim-NHL (tripartite motif and Ncl-1, HT2A and Lin-41 domain)-containing protein Brain tumour (Brat) into one of the two daughter cells, ovarian stem cells are regulated by an extracellular signal from the surrounding stem cell niche. After division, one daughter cell looses niche contact. It undergoes 4 transit-amplifying divisions to form a cyst of 16 interconnected cells that reduce their rate of growth and stop to proliferate mitotically. Here we show that the Trim-NHL protein Mei-P26 (refs 7, 8) restricts growth and proliferation in the ovarian stem cell lineage. Mei-P26 expression is low in stem cells but is strongly induced in 16-cell cysts. In mei-P26 mutants, transit-amplifying cells are larger and proliferate indefinitely leading to the formation of an ovarian tumour. Like brat, mei-P26 regulates nucleolar size and can induce differentiation in Drosophila neuroblasts, suggesting that these genes act through the same pathway. We identify Argonaute-1, a component of the RISC complex, as a common binding partner of Brat and Mei-P26, and show that Mei-P26 acts by inhibiting the microRNA pathway. Mei-P26 and Brat have a similar domain composition that is also found in other tumour suppressors and might be a defining property of a new family of microRNA regulators that act specifically in stem cell lineages.


Assuntos
Linhagem da Célula , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , MicroRNAs/metabolismo , Ovário/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proteínas Argonautas , Ciclo Celular , Diferenciação Celular , Crescimento Celular , Linhagem Celular , Nucléolo Celular/metabolismo , Tamanho Celular , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/classificação , Drosophila melanogaster/genética , Fatores de Iniciação em Eucariotos , Feminino , MicroRNAs/genética , Mutação , Neurônios/citologia , Neurônios/metabolismo , Ovário/metabolismo
8.
Stem Cell Reports ; 16(9): 2107-2117, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34450039

RESUMO

Wilms tumor is the most widespread kidney cancer in children and frequently associated with homozygous loss of the tumor suppressor WT1. Pediatric tumorigenesis is largely inaccessible in humans. Here, we develop a human kidney organoid model for Wilms tumor formation and show that deletion of WT1 during organoid development induces overgrowth of kidney progenitor cells at the expense of differentiating glomeruli and tubules. Functional and gene expression analyses demonstrate that absence of WT1 halts progenitor cell progression at a pre-epithelialized cell state and recapitulates the transcriptional changes detected in a subgroup of Wilms tumor patients with ectopic myogenesis. By "transplanting" WT1 mutant cells into wild-type kidney organoids, we find that their propagation requires an untransformed microenvironment. This work defines the role of WT1 in kidney progenitor cell progression and tumor suppression, and establishes human kidney organoids as a phenotypic model for pediatric tumorigenesis.


Assuntos
Transformação Celular Neoplásica/genética , Genes Supressores de Tumor , Neoplasias Renais/etiologia , Células-Tronco Neoplásicas/metabolismo , Proteínas WT1/genética , Tumor de Wilms/etiologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/metabolismo , Biologia Computacional/métodos , Deleção de Genes , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Hiperplasia , Imunofenotipagem , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Anotação de Sequência Molecular , Células-Tronco Neoplásicas/patologia , Organoides/metabolismo , Organoides/patologia , Proteínas WT1/metabolismo , Tumor de Wilms/metabolismo , Tumor de Wilms/patologia
9.
Dev Cell ; 8(1): 43-52, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15621528

RESUMO

Fragile X syndrome, the most common form of inherited mental retardation, is caused by loss of function for the Fragile X Mental Retardation 1 gene (FMR1). FMR1 protein (FMRP) has specific mRNA targets and is thought to be involved in their transport to subsynaptic sites as well as translation regulation. We report a saturating genetic screen of the Drosophila autosomal genome to identify functional partners of dFmr1. We recovered 19 mutations in the tumor suppressor lethal (2) giant larvae (dlgl) gene and 90 mutations at other loci. dlgl encodes a cytoskeletal protein involved in cellular polarity and cytoplasmic transport and is regulated by the PAR complex through phosphorylation. We provide direct evidence for a Fmrp/Lgl/mRNA complex, which functions in neural development in flies and is developmentally regulated in mice. Our data suggest that Lgl may regulate Fmrp/mRNA sorting, transport, and anchoring via the PAR complex.


Assuntos
Oxirredutases do Álcool/metabolismo , Proteínas de Drosophila/metabolismo , Genes Supressores de Tumor/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Western Blotting/métodos , Fracionamento Celular/métodos , Células Cultivadas , Clonagem Molecular/métodos , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Drosophila , Olho/patologia , Olho/ultraestrutura , Proteína do X Frágil da Deficiência Intelectual , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imuno-Histoquímica/métodos , Camundongos , Microscopia Eletrônica de Varredura/métodos , Mutagênese , Mutação , Junção Neuromuscular/genética , Junção Neuromuscular/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , RNA Mensageiro/metabolismo , Retina/patologia , Retina/ultraestrutura , Frações Subcelulares/metabolismo , Sinapses/metabolismo , Fatores de Tempo
10.
Dev Cell ; 6(6): 845-54, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15177032

RESUMO

How epithelial cells subdivide their plasma membrane into an apical and a basolateral domain is largely unclear. In Drosophila embryos, epithelial cells are generated from a syncytium during cellularization. We show here that polarity is established shortly after cellularization when Par-6 and the atypical protein kinase C concentrate on the apical side of the newly formed cells. Apical localization of Par-6 requires its interaction with activated Cdc42 and dominant-active or dominant-negative Cdc42 disrupt epithelial polarity, suggesting that activation of this GTPase is crucial for the establishment of epithelial polarity. Maintenance of Par-6 localization requires the cytoskeletal protein Lgl. Genetic and biochemical experiments suggest that phosphorylation by aPKC inactivates Lgl on the apical side. On the basolateral side, Lgl is active and excludes Par-6 from the cell cortex, suggesting that complementary cortical domains are maintained by mutual inhibition of aPKC and Lgl on opposite sides of an epithelial cell.


Assuntos
Polaridade Celular/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/embriologia , Células Epiteliais/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteína Quinase C/metabolismo , Proteínas/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Membrana Celular/genética , Membrana Celular/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Células Epiteliais/citologia , Proteínas de Ligação ao GTP/genética , Ligação Proteica/genética , Proteína Quinase C/genética , Proteínas/genética , Proteínas Supressoras de Tumor/genética
11.
Cell Stem Cell ; 24(2): 257-270.e8, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30595499

RESUMO

Self-renewal and differentiation of pluripotent murine embryonic stem cells (ESCs) is regulated by extrinsic signaling pathways. It is less clear whether cellular metabolism instructs developmental progression. In an unbiased genome-wide CRISPR/Cas9 screen, we identified components of a conserved amino-acid-sensing pathway as critical drivers of ESC differentiation. Functional analysis revealed that lysosome activity, the Ragulator protein complex, and the tumor-suppressor protein Folliculin enable the Rag GTPases C and D to bind and seclude the bHLH transcription factor Tfe3 in the cytoplasm. In contrast, ectopic nuclear Tfe3 represses specific developmental and metabolic transcriptional programs that are associated with peri-implantation development. We show differentiation-specific and non-canonical regulation of Rag GTPase in ESCs and, importantly, identify point mutations in a Tfe3 domain required for cytoplasmic inactivation as potentially causal for a human developmental disorder. Our work reveals an instructive and biomedically relevant role of metabolic signaling in licensing embryonic cell fate transitions.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Diferenciação Celular , Lisossomos/metabolismo , Transdução de Sinais , Alelos , Animais , Autorrenovação Celular , Feminino , GTP Fosfo-Hidrolases/metabolismo , Genoma , Humanos , Masculino , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fosforilação , Mutação Puntual/genética , Ligação Proteica , Transcrição Gênica
12.
Curr Biol ; 15(3): 276-82, 2005 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-15694314

RESUMO

During asymmetric cell division, cell fate determinants localize asymmetrically and segregate into one of the two daughter cells. In Drosophila neuroblasts, the asymmetric localization of cell fate determinants to the basal cell cortex requires aPKC. aPKC localizes to the apical cell cortex and phosphorylates the cytoskeletal protein Lethal (2) giant larvae (Lgl). Upon phosphorylation, Lgl dissociates from the cytoskeleton and becomes inactive. Here, we show that phosphorylation regulates Lgl by allowing an autoinhibitory interaction of the N terminus with the C terminus of the protein. We demonstrate that interaction with the cytoskeleton is mediated by a C-terminal domain while the N terminus is not required. Instead, the N terminus can bind to the C terminus and can compete for binding to the cytoskeleton. Interaction between the N- and C-terminal domains requires phosphorylation of Lgl by aPKC. Our results suggest that unphosphorylated, active Lgl exists in an open conformation that interacts with the cytoskeleton while phosphorylation changes the protein to an autoinhibited state.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Drosophila/metabolismo , Drosophila/embriologia , Modelos Moleculares , Neurônios/fisiologia , Proteína Quinase C/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Animais Geneticamente Modificados , Divisão Celular/fisiologia , Células Cultivadas , Citoesqueleto/metabolismo , Imunoprecipitação , Neurônios/metabolismo , Fosforilação , Proteína Quinase C/fisiologia , Estrutura Terciária de Proteína
13.
Curr Biol ; 13(13): 1086-95, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12842007

RESUMO

BACKGROUND: Shot (previously named Kakapo), is a Drosophila Plakin family member containing both Actin binding and microtubule binding domains. In Drosophila, it is required for a wide range of processes, including axon extension, dendrite formation, axonal terminal arborization at the neuromuscular junction, tendon cell development, and adhesion of wing epithelium. RESULTS: To address how Shot exerts its activity at the molecular level, we investigated the molecular interactions of Shot with candidate proteins in mature larval tendon cells. We show that Shot colocalizes with EB1/APC1 and with a compact microtubule array extending between the muscle-tendon junction and the cuticle. Shot forms a protein complex with EB1 via its C-terminal EF-hands and GAS2-containing domains. In tendon cells with reduced Shot activity, EB1/APC1 dissociate from the muscle-tendon junction, and the microtubule array elongates. The resulting tendon cell, although associated with the muscle and the cuticle ends, loses its stress resistance and elongates. CONCLUSIONS: Our results suggest that Shot mediates tendon stress resistance by the organization of a compact microtubule network at the muscle-tendon junction. This is achieved by Shot association with the cytoplasmic faces of the basal hemiadherens junction and with the EB1/APC1 complex.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Drosophila , Proteínas dos Microfilamentos , Microtúbulos/metabolismo , Tendões/crescimento & desenvolvimento , Animais , Western Blotting , Drosophila , Imunofluorescência , Junções Intercelulares/fisiologia , Larva/fisiologia , Músculos/fisiologia , Testes de Precipitina , Transfecção
14.
J Mol Biol ; 429(10): 1441-1458, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28013029

RESUMO

The formation of tissues and organs during metazoan development begs fundamental questions of cellular plasticity: How can the very same genome program have diverse cell types? How do cell identity programs unfold during development in space and time? How can defects in these mechanisms cause disease and also provide opportunities for therapeutic intervention? And ultimately, can developmental programs be exploited for bioengineering tissues and organs? Understanding principle designs of cellular identity and developmental progression is crucial for providing answers. Here, I will discuss how the capture of embryonic pluripotency in murine embryonic stem cells (ESCs) in vitro has allowed fundamental insights into the molecular underpinnings of a developmental cell state and how its ordered disassembly during differentiation prepares for lineage specification.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Murinas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Regulação da Expressão Gênica , Camundongos
15.
Dev Cell ; 14(4): 535-46, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18342578

RESUMO

In both vertebrates and insects, neurons typically arise from neural stem cells or terminally dividing intermediate progenitors. Here, we describe another mode of neurogenesis where neural stem cells generate secondary precursors that undergo multiple rounds of self-renewing transit-amplifying divisions. We identify the Posterior Asense-Negative (PAN) neuroblasts, which do not express the transcription factors Asense or Prospero. PAN neuroblasts rely on the segregating determinants Numb and Brat to generate smaller, secondary neuroblasts that in turn give rise to ganglion mother cells (GMCs) and neurons throughout larval development. In brat or numb mutants, misspecified secondary neuroblasts are unable to produce differentiated progeny and initiate tumor-like overgrowth. In prospero mutants, however, tumors arise from GMCs while secondary neuroblasts are correctly specified. Our data describe a transit-amplifying lineage in the Drosophila nervous system and suggest that different vulnerabilities in intermediate cell types can affect the outcome of tumor suppressor loss in stem cell lineages.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Genes Supressores de Tumor , Hormônios Juvenis/metabolismo , Neurônios/fisiologia , Células-Tronco/fisiologia , Animais , Encéfalo/citologia , Encéfalo/embriologia , Encéfalo/metabolismo , Ciclo Celular/fisiologia , Diferenciação Celular , Linhagem da Célula , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/embriologia , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hormônios Juvenis/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
Cell ; 124(6): 1241-53, 2006 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16564014

RESUMO

How stem cells generate both differentiating and self-renewing daughter cells is unclear. Here, we show that Drosophila larval neuroblasts-stem cell-like precursors of the adult brain-regulate proliferation by segregating the growth inhibitor Brat and the transcription factor Prospero into only one daughter cell. Like Prospero, Brat binds and cosegregates with the adaptor protein Miranda. In larval neuroblasts, both Brat and Prospero are required to inhibit self-renewal in one of the two daughter cells. While Prospero regulates cell-cycle gene transcription, Brat acts as a posttranscriptional inhibitor of dMyc. In brat or prospero mutants, both daughter cells grow and behave like neuroblasts leading to the formation of larval brain tumors. Similar defects are seen in lethal giant larvae (lgl) mutants where Brat and Prospero are not asymmetric. We have identified a molecular mechanism that may control self-renewal and prevent tumor formation in other stem cells as well.


Assuntos
Divisão Celular , Neoplasias/patologia , Neurônios/metabolismo , Células-Tronco/metabolismo , Animais , Encéfalo/citologia , Proteínas de Ciclo Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/farmacologia , Proteínas de Ligação a DNA/fisiologia , Drosophila/embriologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/farmacologia , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica , Genes Supressores de Tumor , Genes myc/fisiologia , Humanos , Larva/citologia , Mutação , Proteínas do Tecido Nervoso/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neurônios/citologia , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiologia , Células-Tronco/citologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA