Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 81(2): 226-238.e5, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33378644

RESUMO

Currently, either highly multiplexed genetic manipulations can be delivered to mammalian cells all at once or extensive engineering of gene regulatory sequences can be used to conditionally activate a few manipulations. Here, we provide proof of principle for a new system enabling multiple genetic manipulations to be executed as a preprogrammed cascade of events. The system leverages the programmability of the S. pyogenes Cas9 and is based on flexible arrangements of individual modules of activity. The basic module consists of an inactive single-guide RNA (sgRNA)-like component that is converted to an active state through the effects of another sgRNA. Modules can be arranged to bring about an algorithmic program of sequential genetic manipulations without the need for engineering cell-type-specific promoters or gene regulatory sequences. With the expanding diversity of available tools that use spCas9, this sgRNA-based system provides multiple levels of interfacing with mammalian cell biology.


Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Edição de Genes/métodos , RNA Guia de Cinetoplastídeos/genética , Animais , Pareamento de Bases , Sequência de Bases , Proteína 9 Associada à CRISPR/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Conformação de Ácido Nucleico , Plasmídeos/química , Plasmídeos/metabolismo , Regiões Promotoras Genéticas , RNA Guia de Cinetoplastídeos/química , RNA Guia de Cinetoplastídeos/metabolismo , Streptococcus pyogenes/química , Streptococcus pyogenes/enzimologia
2.
Mol Cell ; 71(1): 42-55.e8, 2018 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-29979968

RESUMO

The ability to target the Cas9 nuclease to DNA sequences via Watson-Crick base pairing with a single guide RNA (sgRNA) has provided a dynamic tool for genome editing and an essential component of adaptive immune systems in bacteria. After generating a double-stranded break (DSB), Cas9 remains stably bound to DNA. Here, we show persistent Cas9 binding blocks access to the DSB by repair enzymes, reducing genome editing efficiency. Cas9 can be dislodged by translocating RNA polymerases, but only if the polymerase approaches from one direction toward the Cas9-DSB complex. By exploiting these RNA-polymerase/Cas9 interactions, Cas9 can be conditionally converted into a multi-turnover nuclease, mediating increased mutagenesis frequencies in mammalian cells and enhancing bacterial immunity to bacteriophages. These consequences of a stable Cas9-DSB complex provide insights into the evolution of protospacer adjacent motif (PAM) sequences and a simple method of improving selection of highly active sgRNAs for genome editing.


Assuntos
Proteína 9 Associada à CRISPR , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Edição de Genes , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Bactérias/genética , Bactérias/metabolismo , Bactérias/virologia , Bacteriófagos/genética , Bacteriófagos/metabolismo , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Linhagem Celular , Camundongos
3.
Development ; 145(4)2018 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-29361574

RESUMO

Human embryonic stem cells (hESCs) are exquisitely sensitive to WNT ligands, which rapidly cause differentiation. Therefore, hESC self-renewal requires robust mechanisms to keep the cells in a WNT inactive but responsive state. How they achieve this is largely unknown. We explored the role of transcriptional regulators of WNT signaling, the TCF/LEFs. As in mouse ESCs, TCF7L1 is the predominant family member expressed in hESCs. Genome-wide, it binds a gene cohort involved in primitive streak formation at gastrulation, including NODAL, BMP4 and WNT3 Comparing TCF7L1-bound sites with those bound by the WNT signaling effector ß-catenin indicates that TCF7L1 acts largely on the WNT signaling pathway. TCF7L1 overlaps less with the pluripotency regulators OCT4 and NANOG than in mouse ESCs. Gain- and loss-of-function studies indicate that TCF7L1 suppresses gene cohorts expressed in the primitive streak. Interestingly, we find that BMP4, another driver of hESC differentiation, downregulates TCF7L1, providing a mechanism of BMP and WNT pathway intersection. Together, our studies indicate that TCF7L1 plays a major role in maintaining hESC pluripotency, which has implications for human development during gastrulation.


Assuntos
Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Linha Primitiva/metabolismo , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Via de Sinalização Wnt/genética , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Linhagem da Célula , Eletroforese em Gel de Poliacrilamida , Expressão Gênica , Humanos , Imuno-Histoquímica , Análise em Microsséries , Reação em Cadeia da Polimerase
4.
Proc Natl Acad Sci U S A ; 113(5): E548-57, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26764381

RESUMO

Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/ß-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo-pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke's pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with ß-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.


Assuntos
Sistema Hipotálamo-Hipofisário , Proteína 1 Semelhante ao Fator 7 de Transcrição/fisiologia , Animais , Estudos de Coortes , Humanos , Camundongos , Hipófise/anormalidades , Hipófise/metabolismo , Hipófise/fisiopatologia , Prosencéfalo/anormalidades , Prosencéfalo/metabolismo
5.
Nucleic Acids Res ; 44(16): 7997-8010, 2016 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-27484482

RESUMO

CRISPR/Cas9 nucleases have enabled powerful, new genome editing capabilities; however, the preponderance of non-homologous end joining (NHEJ) mediated repair events over homology directed repair (HDR) in most cell types limits the ability to engineer precise changes in mammalian genomes. Here, we increase the efficiency of isolating precise HDR-mediated events in mouse embryonic stem (ES) cells by more than 20-fold through the use of co-incidental insertion (COIN) of independent donor DNA sequences. Analysis of on:off-target frequencies at the Lef1 gene revealed that bi-allelic insertion of a PGK-Neo cassette occurred more frequently than expected. Using various selection cassettes targeting multiple loci, we show that the insertion of a selectable marker at one control site frequently coincided with an insertion at an unlinked, independently targeted site, suggesting enrichment of a sub-population of HDR-proficient cells. When individual cell events were tracked using flow cytometry and fluorescent protein markers, individual cells frequently performed either a homology-dependent insertion event or a homology-independent event, but rarely both types of insertions in a single cell. Thus, when HDR-dependent selection donors are used, COIN enriches for HDR-proficient cells among heterogeneous cell populations. When combined with a self-excising selection cassette, COIN provides highly efficient and scarless genome editing.


Assuntos
Engenharia Genética/métodos , Genoma , Células-Tronco Embrionárias Murinas/metabolismo , Mutagênese Insercional/genética , Animais , Sequência de Bases , Proteínas Associadas a CRISPR/metabolismo , DNA/genética , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades/genética , Edição de Genes , Recombinação Homóloga/genética , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase , Reparo de DNA por Recombinação
6.
Development ; 140(8): 1665-75, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23487311

RESUMO

The core gene regulatory network (GRN) in embryonic stem cells (ESCs) integrates activities of the pro-self-renewal factors Oct4 (Pou5f1), Sox2 and Nanog with that of an inhibitor of self-renewal, Tcf7l1 (Tcf3). The inhibitor function of Tcf7l1 causes dependence on extracellular Wnt/ß-catenin signaling activity, making its embryonic role within the ESC GRN unclear. By analyzing intact mouse embryos, we demonstrate that the function of Tcf7l1 is necessary for specification of cell lineages to occur concomitantly with the elaboration of a three-dimensional body plan during gastrulation. In Tcf7l1(-/-) embryos, specification of mesoderm is delayed, effectively uncoupling it from the induction of the primitive streak. Tcf7l1 repressor activity is necessary for a rapid switch in the response of pluripotent cells to Wnt/ß-catenin stimulation, from one of self-renewal to a mesoderm specification response. These results identify Tcf7l1 as a unique factor that is necessary in pluripotent cells to prepare them for lineage specification. We suggest that the role of Tcf7l1 in mammals is to inhibit the GRN to ensure the coordination of lineage specification with the dynamic cellular events occurring during gastrulation.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Células-Tronco Embrionárias/fisiologia , Gástrula/citologia , Redes Reguladoras de Genes/fisiologia , Camadas Germinativas/citologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Imunofluorescência , Gástrula/metabolismo , Camadas Germinativas/metabolismo , Camadas Germinativas/fisiologia , Técnicas Histológicas , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética
7.
PLoS Genet ; 9(5): e1003424, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23658527

RESUMO

Canonical Wnt signaling plays a rate-limiting role in regulating self-renewal and differentiation in mouse embryonic stem cells (ESCs). We have previously shown that mutation in the Apc (adenomatous polyposis coli) tumor suppressor gene constitutively activates Wnt signaling in ESCs and inhibits their capacity to differentiate towards ecto-, meso-, and endodermal lineages. However, the underlying molecular and cellular mechanisms through which Wnt regulates lineage differentiation in mouse ESCs remain to date largely unknown. To this aim, we have derived and studied the gene expression profiles of several Apc-mutant ESC lines encoding for different levels of Wnt signaling activation. We found that down-regulation of Tcf3, a member of the Tcf/Lef family and a key player in the control of self-renewal and pluripotency, represents a specific and primary response to Wnt activation in ESCs. Accordingly, rescuing Tcf3 expression partially restored the neural defects observed in Apc-mutant ESCs, suggesting that Tcf3 down-regulation is a necessary step towards Wnt-mediated suppression of neural differentiation. We found that Tcf3 down-regulation in the context of constitutively active Wnt signaling does not result from promoter DNA methylation but is likely to be caused by a plethora of mechanisms at both the RNA and protein level as shown by the observed decrease in activating histone marks (H3K4me3 and H3-acetylation) and the upregulation of miR-211, a novel Wnt-regulated microRNA that targets Tcf3 and attenuates early neural differentiation in mouse ESCs. Our data show for the first time that Wnt signaling down-regulates Tcf3 expression, possibly at both the transcriptional and post-transcriptional levels, and thus highlight a novel mechanism through which Wnt signaling inhibits neuro-ectodermal lineage differentiation in mouse embryonic stem cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Via de Sinalização Wnt , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Metilação de DNA , Regulação para Baixo , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Mutação , Transcrição Gênica
8.
Development ; 139(12): 2118-29, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22573616

RESUMO

The canonical Wnt/ß-catenin signaling pathway classically functions through the activation of target genes by Tcf/Lef-ß-catenin complexes. In contrast to ß-catenin-dependent functions described for Tcf1, Tcf4 and Lef1, the known embryonic functions for Tcf3 in mice, frogs and fish are consistent with ß-catenin-independent repressor activity. In this study, we genetically define Tcf3-ß-catenin functions in mice by generating a Tcf3ΔN knock-in mutation that specifically ablates Tcf3-ß-catenin. Mouse embryos homozygous for the knock-in mutation (Tcf3(ΔN/ΔN)) progress through gastrulation without apparent defects, thus genetically proving that Tcf3 function during gastrulation is independent of ß-catenin interaction. Tcf3(ΔN/ΔN) mice were not viable, and several post-gastrulation defects revealed the first in vivo functions of Tcf3-ß-catenin interaction affecting limb development, vascular integrity, neural tube closure and eyelid closure. Interestingly, the etiology of defects indicated an indirect role for Tcf3-ß-catenin in the activation of target genes. Tcf3 directly represses transcription of Lef1, which is stimulated by Wnt/ß-catenin activity. These genetic data indicate that Tcf3-ß-catenin is not necessary to activate target genes directly. Instead, our findings support the existence of a regulatory circuit whereby Wnt/ß-catenin counteracts Tcf3 repression of Lef1, which subsequently activates target gene expression via Lef1-ß-catenin complexes. We propose that the Tcf/Lef circuit model provides a mechanism downstream of ß-catenin stability for controlling the strength of Wnt signaling activity during embryonic development.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas Repressoras/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo , Animais , Padronização Corporal/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Extremidades/embriologia , Pálpebras/metabolismo , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Introdução de Genes , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Ligação Proteica , Análise de Sobrevida , Via de Sinalização Wnt/genética
9.
Proc Natl Acad Sci U S A ; 108(29): 11912-7, 2011 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-21730189

RESUMO

The heterochromatin barrier must be overcome to generate induced pluripotent stem cells and cell fusion-mediated reprogrammed hybrids. Here, we show that the absence of T-cell factor 3 (Tcf3), a repressor of ß-catenin target genes, strikingly and rapidly enhances the efficiency of neural precursor cell (NPC) reprogramming. Remarkably, Tcf3(-/-) ES cells showed a genome-wide increase in AcH3 and decrease in H3K9me3 and can reprogram NPCs after fusion greatly. In addition, during reprogramming of NPCs into induced pluripotent stem cells, the silencing of Tcf3 increased AcH3 and decreased the number of H3K9me3-positive heterochromatin foci early and long before reactivation of the endogenous stem cell genes. In conclusion, our data suggest that Tcf3 functions as a repressor of the reprogramming potential of somatic cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Reprogramação Celular/fisiologia , Epigênese Genética/fisiologia , Deleção de Genes , Células-Tronco Pluripotentes Induzidas/fisiologia , Neurônios/citologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular , Reprogramação Celular/genética , Imunoprecipitação da Cromatina , Epigênese Genética/genética , Citometria de Fluxo , Imunofluorescência , Vetores Genéticos/genética , Immunoblotting , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Retroviridae , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
bioRxiv ; 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39345516

RESUMO

Although genomes encode instructions for mammalian cell differentiation with rich syntactic relationships, existing methods for genetically programming cells have modest capabilities for stepwise regulation of genes. Here, we developed a sequential genetic system that enables transcriptional activation of endogenous genes in a preprogrammed, stepwise manner. The system relies on the removal of an RNA polymerase III termination signal to induce both the transcriptional activation and the DNA endonuclease activities of a Cas9-VPR protein to effect stepwise progression through cascades of gene activation events. The efficiency of the cascading system enables a new dimension for cellular programming by allowing the manipulation of the sequential order of gene activation for directing the differentiation of human stem cells. One-Sentence Summary: Development of a synthetic biology system for preprogrammed, stepwise activation of endogenous genes.

11.
Proc Natl Acad Sci U S A ; 107(23): 10514-9, 2010 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-20498046

RESUMO

Two major goals of regenerative medicine are to reproducibly transform adult somatic cells into a pluripotent state and to control their differentiation into specific cell fates. Progress toward these goals would be greatly helped by obtaining a complete picture of the RNA isoforms produced by these cells due to alternative splicing (AS) and alternative promoter selection (APS). To investigate the roles of AS and APS, reciprocal exon-exon junctions were interrogated on a genome-wide scale in differentiating mouse embryonic stem (ES) cells with a prototype Affymetrix microarray. Using a recently released open-source software package named AltAnalyze, we identified 144 genes for 170 putative isoform variants, the majority (67%) of which were predicted to alter protein sequence and domain composition. Verified alternative exons were largely associated with pathways of Wnt signaling and cell-cycle control, and most were conserved between mouse and human. To examine the functional impact of AS, we characterized isoforms for two genes. As predicted by AltAnalyze, we found that alternative isoforms of the gene Serca2 were targeted by distinct microRNAs (miRNA-200b, miRNA-214), suggesting a critical role for AS in cardiac development. Analysis of the Wnt transcription factor Tcf3, using selective knockdown of an ES cell-enriched and characterized isoform, revealed several distinct targets for transcriptional repression (Stmn2, Ccnd2, Atf3, Klf4, Nodal, and Jun) as well as distinct differentiation outcomes in ES cells. The findings herein illustrate a critical role for AS in the specification of ES cells with differentiation, and highlight the utility of global functional analyses of AS.


Assuntos
Processamento Alternativo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Animais , Éxons , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , MicroRNAs/genética , Regiões Promotoras Genéticas , Seleção Genética , Transdução de Sinais , Transcrição Gênica , Proteínas Wnt/metabolismo
12.
Oncogene ; 42(31): 2374-2385, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37386128

RESUMO

Tuft cells are chemosensory epithelial cells that increase in number following infection or injury to robustly activate the innate immune response to alleviate or promote disease. Recent studies of castration resistant prostate cancer and its subtype, neuroendocrine prostate cancer, revealed Pou2f3+ populations in mouse models. The transcription factor Pou2f3 is a master regulator of the tuft cell lineage. We show that tuft cells are upregulated early during prostate cancer development, and their numbers increase with progression. Cancer-associated tuft cells in the mouse prostate express DCLK1, COX1, COX2, while human tuft cells express COX1. Mouse and human tuft cells exhibit strong activation of signaling pathways including EGFR and SRC-family kinases. While DCLK1 is a mouse tuft cell marker, it is not present in human prostate tuft cells. Tuft cells that appear in mouse models of prostate cancer display genotype-specific tuft cell gene expression signatures. Using bioinformatic analysis tools and publicly available datasets, we characterized prostate tuft cells in aggressive disease and highlighted differences between tuft cell populations. Our findings indicate that tuft cells contribute to the prostate cancer microenvironment and may promote development of more advanced disease. Further research is needed to understand contributions of tuft cells to prostate cancer progression.


Assuntos
Próstata , Neoplasias da Próstata , Masculino , Camundongos , Humanos , Animais , Próstata/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais/genética , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Células Epiteliais/metabolismo , Microambiente Tumoral , Quinases Semelhantes a Duplacortina
13.
Cancer Res ; 82(13): 2458-2471, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35583996

RESUMO

The transcription factor Forkhead box M1 (FoxM1) is overexpressed in breast cancers and correlates with poor prognosis. Mechanistically, FoxM1 associates with CBP to activate transcription and with Rb to repress transcription. Although the activating function of FoxM1 in breast cancer has been well documented, the significance of its repressive activity is poorly understood. Using CRISPR-Cas9 engineering, we generated a mouse model that expresses FoxM1-harboring point mutations that block binding to Rb while retaining its ability to bind CBP. Unlike FoxM1-null mice, mice harboring Rb-binding mutant FoxM1 did not exhibit significant developmental defects. The mutant mouse line developed PyMT-driven mammary tumors that were deficient in lung metastasis, which was tumor cell-intrinsic. Single-cell RNA-seq of the tumors revealed a deficiency in prometastatic tumor cells and an expansion of differentiated alveolar type tumor cells, and further investigation identified that loss of the FoxM1/Rb interaction caused enhancement of the mammary alveolar differentiation program. The FoxM1 mutant tumors also showed increased Pten expression, and FoxM1/Rb was found to activate Akt signaling by repressing Pten. In human breast cancers, expression of FoxM1 negatively correlated with Pten mRNA. Furthermore, the lack of tumor-infiltrating cells in FoxM1 mutant tumors appeared related to decreases in pro-metastatic tumor cells that express factors required for infiltration. These observations demonstrate that the FoxM1/Rb-regulated transcriptome is critical for the plasticity of breast cancer cells that drive metastasis, identifying a prometastatic role of Rb when bound to FoxM1. SIGNIFICANCE: This work provides new insights into how the interaction between FoxM1 and Rb facilitates the evolution of metastatic breast cancer cells by altering the transcriptome.


Assuntos
Neoplasias da Mama , Proteína Forkhead Box M1/metabolismo , Fatores de Transcrição Forkhead , Animais , Neoplasias da Mama/patologia , Diferenciação Celular/genética , Linhagem Celular Tumoral , Feminino , Proteína Forkhead Box M1/genética , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Transdução de Sinais , Transcrição Gênica
14.
Nat Cell Biol ; 24(1): 35-50, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35027734

RESUMO

Vascular endothelial growth factor receptor type 2 (VEGFR2, also known as KDR and FLK1) signalling in endothelial cells (ECs) is essential for developmental and reparative angiogenesis. Reactive oxygen species and copper (Cu) are also involved in these processes. However, their inter-relationship is poorly understood. Evidence of the role of the endothelial Cu importer CTR1 (also known as SLC31A1) in VEGFR2 signalling and angiogenesis in vivo is lacking. Here, we show that CTR1 functions as a redox sensor to promote angiogenesis in ECs. CTR1-depleted ECs showed reduced VEGF-induced VEGFR2 signalling and angiogenic responses. Mechanistically, CTR1 was rapidly sulfenylated at Cys189 at its cytosolic C terminus after stimulation with VEGF, which induced CTR1-VEGFR2 disulfide bond formation and their co-internalization to early endosomes, driving sustained VEGFR2 signalling. In vivo, EC-specific Ctr1-deficient mice or CRISPR-Cas9-generated redox-dead Ctr1(C187A)-knockin mutant mice had impaired developmental and reparative angiogenesis. Thus, oxidation of CTR1 at Cys189 promotes VEGFR2 internalization and signalling to enhance angiogenesis. Our study uncovers an important mechanism for sensing reactive oxygen species through CTR1 to drive neovascularization.


Assuntos
Transportador de Cobre 1/metabolismo , Cobre/metabolismo , Neovascularização Fisiológica/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Bovinos , Linhagem Celular , Transportador de Cobre 1/genética , Cisteína/metabolismo , Feminino , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Transdução de Sinais/fisiologia
15.
Stem Cells ; 28(10): 1794-804, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20734354

RESUMO

Delineating the signaling pathways that underlie ESC pluripotency is paramount for development of ESC applications in both the research and clinical settings. In culture pluripotency is maintained by leukemia inhibitory factor (LIF) stimulation of two separate signaling axes: Stat3/Klf4/Sox2 and PI3K/Tbx3/Nanog, which converge in the regulation of Oct4 expression. However, LIF signaling is not required in vivo for self-renewal, thus alternate signaling axes likely mediate these pathways. Additional factors that promote pluripotency gene expression have been identified, including the direct regulation of Oct4 by liver receptor homolog-1 (Lrh-1) and ß-catenin regulation of Nanog. Here, we present genetic, molecular, and pharmacological studies identifying a signaling axis in which ß-catenin promotes pluripotency gene expression in an Lrh-1-dependent manner. Furthermore, Lrh-1 was identified as a novel ß-catenin target gene, and Lrh-1 regulation is required for maintaining proper levels of Oct4, Nanog, and Tbx3. Elucidation of this pathway provides an alternate mechanism by which the primary pluripotency axis may be regulated in vivo and may pave the way for small molecule applications to manipulate pluripotency or improve the efficiency of somatic cell reprogramming.


Assuntos
Células-Tronco Embrionárias/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , beta Catenina/metabolismo , Animais , Western Blotting , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Imunoprecipitação da Cromatina , Feminino , Imunofluorescência , Fator 4 Semelhante a Kruppel , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Masculino , Camundongos , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , beta Catenina/genética
16.
Exp Cell Res ; 316(6): 1050-60, 2010 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-20006604

RESUMO

A combination of cell intrinsic factors and extracellular signals determine whether mouse embryonic stem cells (ESC) divide, self-renew, and differentiate. Here, we report a new interaction between cell intrinsic aspects of the canonical Wnt/Tcf/beta-catenin signaling pathway and extracellular Lif/Jak/Stat3 stimulation that combines to promote self-renewal and proliferation of ESC. Mutant ESC lacking the Tcf3 transcriptional repressor continue to self-renew in the absence of exogenous Lif and through pharmacological inhibition of Lif/Jak/Stat3 signaling; however, proliferation rates of TCF3-/- ESC were significantly decreased by inhibiting Jak/Stat3 activity. Cell mixing experiments showed that stimulation of Stat3 phosphorylation in TCF3-/- ESC was mediated through secretion of paracrine acting factors, but did not involve elevated Lif or LifR transcription. The new interaction between Wnt and Lif/Jak/Stat3 signaling pathways has potential for new insights into the growth of tumors caused by aberrant activity of Wnt/Tcf/beta-catenin signaling.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proliferação de Células , Células-Tronco Embrionárias/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Benzimidazóis/farmacologia , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Janus Quinases/antagonistas & inibidores , Janus Quinases/genética , Janus Quinases/metabolismo , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Piridonas/farmacologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
17.
Nat Cell Biol ; 22(4): 389-400, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32231305

RESUMO

In mouse embryonic stem cells (mESCs), chemical blockade of Gsk3α/ß and Mek1/2 (2i) instructs a self-renewing ground state whose endogenous inducers are unknown. Here we show that the axon guidance cue Netrin-1 promotes naive pluripotency by triggering profound signalling, transcriptomic and epigenetic changes in mESCs. Furthermore, we demonstrate that Netrin-1 can substitute for blockade of Gsk3α/ß and Mek1/2 to sustain self-renewal of mESCs in combination with leukaemia inhibitory factor and regulates the formation of the mouse pluripotent blastocyst. Mechanistically, we reveal how Netrin-1 and the balance of its receptors Neo1 and Unc5B co-regulate Wnt and MAPK pathways in both mouse and human ESCs. Netrin-1 induces Fak kinase to inactivate Gsk3α/ß and stabilize ß-catenin while increasing the phosphatase activity of a Ppp2r2c-containing Pp2a complex to reduce Erk1/2 activity. Collectively, this work identifies Netrin-1 as a regulator of pluripotency and reveals that it mediates different effects in mESCs depending on its receptor dosage, opening perspectives for balancing self-renewal and lineage commitment.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas do Tecido Nervoso/genética , Receptores de Netrina/genética , Netrina-1/genética , Receptores de Superfície Celular/genética , Via de Sinalização Wnt/genética , Animais , Linhagem Celular , Embrião de Mamíferos , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Receptores de Netrina/metabolismo , Netrina-1/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Proteína Fosfatase 2/genética , Proteína Fosfatase 2/metabolismo , Receptores de Superfície Celular/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
18.
Mol Cell Biol ; 26(20): 7479-91, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16894029

RESUMO

The dual function of stem cells requires them not only to form new stem cells through self-renewal but also to form lineage-committed cells through differentiation. Embryonic stem cells (ESC), which are derived from the blastocyst inner cell mass, retain properties of self-renewal and the potential for lineage commitment. To balance self-renewal and differentiation, ESC must carefully control the levels of several transcription factors, including Nanog, Sox2, and Oct4. While molecular mechanisms promoting transcription of these genes have been described, mechanisms preventing excessive levels in self-renewing ESC remain unknown. By examining the function of the TCF family of transcription factors in ESC, we have found that Tcf3 is necessary to limit the steady-state levels of Nanog mRNA, protein, and promoter activity in self-renewing ESC. Chromatin immunoprecipitation and promoter reporter assays showed that Tcf3 bound to a promoter regulatory region of the Nanog gene and repressed its transcriptional activity in ESC through a Groucho interaction domain-dependent process. The absence of Tcf3 caused delayed differentiation of ESC in vitro as elevated Nanog levels persisted through 5 days of embryoid body formation. These new data support a model wherein Tcf3-mediated control of Nanog levels allows stem cells to balance the creation of lineage-committed and undifferentiated cells.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Homeodomínio/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Fatores de Transcrição TCF/metabolismo , Transcrição Gênica/genética , Animais , Sequência de Bases , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Embrião de Mamíferos/citologia , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog , Fatores de Transcrição TCF/deficiência , Fatores de Transcrição TCF/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição
19.
Cell Stem Cell ; 25(2): 210-224.e6, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31104942

RESUMO

Progression through states of pluripotency is required for cells in early mammalian embryos to transition away from heightened self-renewal and toward competency for lineage specification. Here, we use a CRISPR mutagenesis screen in mouse embryonic stem cells (ESCs) to identify unexpected roles for nuclear export and intracellular Ca2+ homeostasis during the exit out of the naive state of pluripotency. Mutation of a plasma membrane Ca2+ pump encoded by Atp2b1 increased intracellular Ca2+ such that it overcame effects of intracellular Ca2+ reduction, which is required for naive exit. Persistent self-renewal of ESCs was supported both in Atp2b1-/-Tcf7l1-/- double-knockout ESCs passaged in defined media alone (no LIF or inhibitors) and in wild-type cells passaged in media containing only calcitonin and a GSK3 inhibitor. These new findings suggest a central role for intracellular Ca2+ in safeguarding naive pluripotency.


Assuntos
Sinalização do Cálcio/fisiologia , Espaço Intracelular/metabolismo , Células-Tronco Embrionárias Murinas/fisiologia , ATPases Transportadoras de Cálcio da Membrana Plasmática/metabolismo , Células-Tronco Pluripotentes/fisiologia , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Linhagem da Célula , Autorrenovação Celular/genética , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Quinase 3 da Glicogênio Sintase/metabolismo , Homeostase , Camundongos , Camundongos Knockout , Proteínas Nucleares/metabolismo , ATPases Transportadoras de Cálcio da Membrana Plasmática/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética
20.
Nat Struct Mol Biol ; 26(8): 679-685, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31285607

RESUMO

The RNA-guided Cas9 endonuclease from Streptococcus pyogenes is a single-turnover enzyme that displays a stable product state after double-stranded-DNA cleavage. Here, we present cryo-EM structures of precatalytic, postcatalytic and product states of the active Cas9-sgRNA-DNA complex in the presence of Mg2+. In the precatalytic state, Cas9 adopts the 'checkpoint' conformation with the HNH nuclease domain positioned far away from the DNA. Transition to the postcatalytic state involves a dramatic ~34-Å swing of the HNH domain and disorder of the REC2 recognition domain. The postcatalytic state captures the cleaved substrate bound to the catalytically competent HNH active site. In the product state, the HNH domain is disordered, REC2 returns to the precatalytic conformation, and additional interactions of REC3 and RuvC with nucleic acids are formed. The coupled domain motions and interactions between the enzyme and the RNA-DNA hybrid provide new insights into the mechanism of genome editing by Cas9.


Assuntos
Proteína 9 Associada à CRISPR/ultraestrutura , Sistemas CRISPR-Cas , DNA/metabolismo , Proteína 9 Associada à CRISPR/química , Proteína 9 Associada à CRISPR/metabolismo , Microscopia Crioeletrônica , DNA/ultraestrutura , Substâncias Macromoleculares/ultraestrutura , Modelos Moleculares , Movimento (Física) , Conformação Proteica , Domínios Proteicos , Edição de RNA , RNA Guia de Cinetoplastídeos/metabolismo , Streptococcus pyogenes/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA