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










Base de dados
Intervalo de ano de publicação
1.
Mol Metab ; 69: 101677, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36693621

RESUMO

OBJECTIVE: Mitochondria fuel most animal cells with ATP, ensuring proper energetic metabolism of organs. Early and extensive mitochondrial dysfunction often leads to severe disorders through multiorgan failure. Hacd2 gene encodes an enzyme involved in very long chain fatty acid (C ≥ 18) synthesis, yet its roles in vivo remain poorly understood. Since mitochondria function relies on specific properties of their membranes conferred by a particular phospholipid composition, we investigated if Hacd2 gene participates to mitochondrial integrity. METHODS: We generated two mouse models, the first one leading to a partial knockdown of Hacd2 expression and the second one, to a complete knockout of Hacd2 expression. We performed an in-depth analysis of the associated phenotypes, from whole organism to molecular scale. RESULTS: Thanks to these models, we show that Hacd2 displays an early and broad expression, and that its deficiency in mice is lethal. Specifically, partial knockdown of Hacd2 expression leads to death within one to four weeks after birth, from a sudden growth arrest followed by cachexia and lethargy. The total knockout of Hacd2 is even more severe, characterized by embryonic lethality around E9.5 following developmental arrest and pronounced cardiovascular malformations. In-depth mechanistic analysis revealed that Hacd2 deficiency causes altered mitochondrial efficiency and ultrastructure, as well as accumulation of oxidized cardiolipin. CONCLUSIONS: Altogether, these data indicate that the Hacd2 gene is essential for energetic metabolism during embryonic and postnatal development, acting through the control of proper mitochondrial organization and function.


Assuntos
Mitocôndrias , Doenças Mitocondriais , Animais , Camundongos , Cardiolipinas , Ácidos Graxos não Esterificados/metabolismo , Hidroliases/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Fosfolipídeos/metabolismo
2.
EMBO Rep ; 23(9): e54762, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35899551

RESUMO

MicroRNA (miRNA) loaded Argonaute (AGO) complexes regulate gene expression via direct base pairing with their mRNA targets. Previous works suggest that up to 60% of mammalian transcripts might be subject to miRNA-mediated regulation, but it remains largely unknown which fraction of these interactions are functional in a specific cellular context. Here, we integrate transcriptome data from a set of miRNA-depleted mouse embryonic stem cell (mESC) lines with published miRNA interaction predictions and AGO-binding profiles. Using this integrative approach, combined with molecular validation data, we present evidence that < 10% of expressed genes are functionally and directly regulated by miRNAs in mESCs. In addition, analyses of the stem cell-specific miR-290-295 cluster target genes identify TFAP4 as an important transcription factor for early development. The extensive datasets developed in this study will support the development of improved predictive models for miRNA-mRNA functional interactions.


Assuntos
MicroRNAs , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Mamíferos/genética , Mamíferos/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
3.
EMBO Rep ; 23(9): e54458, 2022 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-35856394

RESUMO

LINE-1 (L1) retroelements have retained their ability to mobilize. Mechanisms regulating L1 mobility include DNA methylation in somatic cells and the piRNA pathway in the germline. During preimplantation stages of mouse embryonic development, however, both pathways are inactivated leading to a window necessitating alternate means of L1 regulation. We previously reported an increase in L1 levels in Dicer_KO mouse embryonic stem cells (mESCs), which was accompanied by only a marginal increase in retrotransposition, suggesting additional mechanisms suppressing L1 mobility. Here, we demonstrate that L1 ribonucleoprotein complexes (L1 RNP) accumulate as aggregates in the cytoplasm of Dicer_KO mESCs along with the RNA helicase MOV10. The combined overexpression of L1 ORF1p and MOV10 is sufficient to create L1 RNP aggregates. In Dicer_KO mESCs, MOV10 is upregulated due to the loss of its direct regulation by miRNAs. The newly discovered posttranscriptional regulation of Mov10, and its role in preventing L1 retrotransposition by driving cytosolic aggregation, provides routes to explore for therapy in disease conditions where L1s are upregulated.


Assuntos
Desenvolvimento Embrionário , MicroRNAs , Animais , Elementos Nucleotídeos Longos e Dispersos , Camundongos , MicroRNAs/metabolismo , RNA Interferente Pequeno/metabolismo , Retroelementos/genética
4.
Stem Cell Reports ; 10(2): 461-476, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29396181

RESUMO

In mouse, although four Argonaute (AGO) proteins with partly overlapping functions in small-RNA pathways exist, only Ago2 deficiency causes embryonic lethality. To investigate the role of AGO2 during mouse early development, we generated Ago2-deficient mouse embryonic stem cells (mESCs) and performed a detailed characterization of their differentiation potential. Ago2 disruption caused a global reduction of microRNAs, which resulted in the misregulation of only a limited number of transcripts. We demonstrated, both in vivo and in vitro, that AGO2 is dispensable for the embryonic germ-layer formation. However, Ago2-deficient mESCs showed a specific defect during conversion into extra-embryonic endoderm cells. We proved that this defect is cell autonomous and can be rescued by both a catalytically active and an inactive Ago2, but not by Ago2 deprived of its RNA binding capacity or by Ago1 overexpression. Overall, our results suggest a role for AGO2 in stem cell differentiation.


Assuntos
Proteínas Argonautas/genética , Diferenciação Celular/genética , Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias Murinas/citologia , Animais , Linhagem Celular , Endoderma/citologia , Endoderma/crescimento & desenvolvimento , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camadas Germinativas/citologia , Camadas Germinativas/crescimento & desenvolvimento , Camundongos , MicroRNAs/genética
6.
FEBS Open Bio ; 7(2): 204-220, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28174687

RESUMO

A gene regulation network orchestrates processes ensuring the maintenance of cellular identity and genome integrity. Small RNAs generated by the RNAse III DICER have emerged as central players in this network. Moreover, deletion of Dicer in mice leads to early embryonic lethality. To better understand the underlying mechanisms leading to this phenotype, we generated Dicer-deficient mouse embryonic stem cells (mESCs). Their detailed characterization revealed an impaired differentiation potential, and incapacity to exit from the pluripotency state. We also observed a strong accumulation of LINE-1 (L1s) transcripts, which was translated at protein level and led to an increased L1s retrotransposition. Our findings reveal Dicer as a new essential player that sustains mESCs self-renewal and genome integrity by controlling L1s regulation.

7.
J Cell Biol ; 216(2): 355-366, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28100686

RESUMO

Mouse embryonic stem cells (mESCs) deficient for DGCR8, a key component of the microprocessor complex, present strong differentiation defects. However, the exact reasons impairing their commitment remain elusive. The analysis of newly generated mutant mESCs revealed that DGCR8 is essential for the exit from the pluripotency state. To dissociate canonical versus noncanonical functions of DGCR8, we complemented the mutant mESCs with a phosphomutant DGCR8, which restored microRNA levels but did not rescue the exit from pluripotency defect. Integration of omics data and RNA immunoprecipitation experiments established DGCR8 as a direct interactor of Tcf7l1 mRNA, a core component of the pluripotency network. Finally, we found that DGCR8 facilitated the splicing of Tcf7l1, an event necessary for the differentiation of mESCs. Our data reveal a new noncanonical function of DGCR8 in the modulation of the alternative splicing of Tcf7l1 mRNA in addition to its established function in microRNA biogenesis.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas de Ligação a RNA/metabolismo , Processamento Alternativo , Animais , Ciclo Celular , Linhagem Celular , Proliferação de Células , Técnicas de Silenciamento de Genes , Genótipo , Camundongos , MicroRNAs/biossíntese , MicroRNAs/genética , Mutação , Fenótipo , Fosforilação , Ligação Proteica , Interferência de RNA , Precursores de RNA/genética , Precursores de RNA/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Transdução de Sinais , Fatores de Tempo , Proteína 1 Semelhante ao Fator 7 de Transcrição/genética , Proteína 1 Semelhante ao Fator 7 de Transcrição/metabolismo , Transfecção
8.
J Mol Biol ; 429(10): 1532-1543, 2017 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-28118980

RESUMO

Complex gene regulation systems ensure the maintenance of cellular identity during early development in mammals. Eukaryotic small RNAs have emerged as critical players in RNA interference (RNAi) by mediating gene silencing during embryonic stem cell self-renewal. Most of the proteins involved in the biogenesis of small RNAs are essential for proliferation and differentiation into the three germ layers of mouse embryonic stem cells. In the last decade, new functions for some RNAi proteins, independent of their roles in RNAi pathways, have been demonstrated in different biological systems. In parallel, new concepts in stem cell biology have emerged. Here, we review and integrate the current understanding of how RNAi proteins regulate stem cell identity with the new advances in the stem cell field and the recent non-canonical functions of the RNAi proteins. Finally, we propose a reevaluation of all RNAi mutant phenotypes, as non-canonical (small non-coding RNA independent) functions may contribute to the molecular mechanisms governing mouse embryonic stem cells commitment.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica , Interferência de RNA , Animais , Diferenciação Celular , Proliferação de Células , Camundongos
9.
Methods Mol Biol ; 1400: 237-59, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26895058

RESUMO

Long Interspersed Elements-1 (LINE-1 or L1) are a class of transposable elements which account for almost 19 % of the mouse genome. This represents around 600,000 L1 fragments, among which it is estimated that 3000 intact copies still remain capable to retrotranspose and to generate deleterious mutation by insertion into genomic coding region. In differentiated cells, full length L1 are transcriptionally repressed by DNA methylation. However at the blastocyst stage, L1 elements are subject to a demethylation wave and able to be expressed and to be inserted into new genomic locations. Mouse Embryonic Stem Cells (mESCs) are pluripotent stem cells derived from the inner cell mass of blastocysts. Mouse ESCs can be maintained undifferentiated under controlled culture conditions or induced into the three primary germ layers, therefore they represent a suitable model to follow mechanisms involved in L1 repression during the process of differentiation of mESCs. This protocol presents how to maintain culture of undifferentiated mESCs, induce their differentiation, and monitor L1 expression at the transcriptional and translational levels. L1 transcriptional levels are assessed by real-time qRT-PCR performed on total RNA extracts using specific L1 primers and translation levels are measured by Western blot analysis of L1 protein ORF1 using a specific L1 antibody.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica , Elementos Nucleotídeos Longos e Dispersos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Western Blotting/métodos , Linhagem Celular , Proliferação de Células , Células Cultivadas , Imuno-Histoquímica/métodos , Camundongos , Reação em Cadeia da Polimerase em Tempo Real/métodos
10.
Methods Mol Biol ; 1341: 321-43, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25762293

RESUMO

CRISPR/Cas9, originally discovered as a bacterial immune system, has recently been engineered into the latest tool to successfully introduce site-specific mutations in a variety of different organisms. Composed only of the Cas9 protein as well as one engineered guide RNA for its functionality, this system is much less complex in its setup and easier to handle than other guided nucleases such as Zinc-finger nucleases or TALENs.Here, we describe the simultaneous transfection of two paired CRISPR sgRNAs-Cas9 plasmids, in mouse embryonic stem cells (mESCs), resulting in the knockout of the selected target gene. Together with a four primer-evaluation system, it poses an efficient way to generate new independent knockout mouse embryonic stem cell lines.


Assuntos
Sistemas CRISPR-Cas , Técnicas de Inativação de Genes/métodos , Células-Tronco Embrionárias Murinas/metabolismo , Transfecção/métodos , Animais , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Genoma , Camundongos , Camundongos Knockout , Plasmídeos/genética , RNA Guia de Cinetoplastídeos/genética
11.
EMBO Rep ; 16(3): 341-50, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25608529

RESUMO

RNA interference (RNAi) is a widespread and widely exploited phenomenon. Here, we show that changing inositol 1,4,5-trisphosphate (IP3) signalling alters RNAi sensitivity in Caenorhabditis elegans. Reducing IP3 signalling enhances sensitivity to RNAi in a broad range of genes and tissues. Conversely up-regulating IP3 signalling decreases sensitivity. Tissue-specific rescue experiments suggest IP3 functions in the intestine. We also exploit IP3 signalling mutants to further enhance the sensitivity of RNAi hypersensitive strains. These results demonstrate that conserved cell signalling pathways can modify RNAi responses, implying that RNAi responses may be influenced by an animal's physiology or environment.


Assuntos
Caenorhabditis elegans/fisiologia , Inositol 1,4,5-Trifosfato/metabolismo , Interferência de RNA/fisiologia , Transdução de Sinais/fisiologia , Animais , Caenorhabditis elegans/genética , Processamento de Imagem Assistida por Computador , Mucosa Intestinal/metabolismo , Microscopia de Fluorescência , Modelos Biológicos , RNA de Cadeia Dupla , Transdução de Sinais/genética
12.
Biomol Concepts ; 5(5): 409-28, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25367621

RESUMO

Transposable elements (TEs) are mobile DNA elements that represent almost half of the human genome. Transposition of TEs has been implicated as a source of genome evolution and acquisition of new traits but also as an origin of diseases. The activity of these elements is therefore tightly regulated during the life cycle of each individual, and many recent discoveries involved the genetic and epigenetic mechanisms in their control. In this review, we present recent findings in this field of research, focusing on the case of one specific family of TEs: the long-interspersed nuclear elements-1 (LINE-1 or L1). LINE-1 elements are the most representative class of retrotransposons in mammalian genomes. We illustrate how these elements are conserved between mice and humans, and how they are regulated during the life cycle. Additionally, recent advances in genome-wide sequencing approaches allow us not only to better understand the regulation of LINE-1 but also highlight new issues specifically at the bioinformatics level. Therefore, we discuss the state of the art in analyzing such bioinformatics datasets to identify epigenetic regulators of repeated elements in the human genomes.


Assuntos
Regulação da Expressão Gênica , Elementos Nucleotídeos Longos e Dispersos , Mamíferos/crescimento & desenvolvimento , Mamíferos/genética , Animais , Epigênese Genética , Evolução Molecular , Genoma Humano , Genômica , Humanos , Camundongos , Filogenia
13.
EMBO Rep ; 14(8): 718-25, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23797875

RESUMO

Wnt/ß-catenin signalling is central to development and its regulation is essential in preventing cancer. Using phosphorylation of Dishevelled as readout of pathway activation, we identified Drosophila Wnk kinase as a new regulator of canonical Wnt/ß-catenin signalling. WNK kinases are known for regulating ion co-transporters associated with hypertension disorders. We demonstrate that wnk loss-of-function phenotypes resemble canonical Wnt pathway mutants, while Wnk overexpression causes gain-of-function canonical Wnt-signalling phenotypes. Importantly, knockdown of human WNK1 and WNK2 also results in decreased Wnt signalling in mammalian cell culture, suggesting that Wnk kinases have a conserved function in ensuring peak levels of canonical Wnt signalling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinases/genética , Via de Sinalização Wnt/genética , beta Catenina/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Proteínas Desgrenhadas , Proteínas de Drosophila , Drosophila melanogaster , Regulação da Expressão Gênica , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lentivirus/genética , Antígenos de Histocompatibilidade Menor , Fosfoproteínas/metabolismo , Fosforilação , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Quinase 1 Deficiente de Lisina WNK , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...