Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Rapid Commun Mass Spectrom ; 38(18): e9867, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-38973066

RESUMO

RATIONALE: mRNA technology has begun to play a significant role in the areas of therapeutic intervention and vaccine development. However, optimizing the mRNA sequence that influences protein expression levels is a resource-intensive and time-consuming process. This study introduces a new method to accelerate the selection of sequences of mRNA for optimal protein expression. METHODS: We designed the mRNA sequences in such a way that a unique peptide barcode, corresponding to each mRNA sequence, is attached to the expressed protein. These barcodes, cleaved off by a protease and simultaneously quantified by mass spectrometry, reflect the protein expression, enabling a parallel analysis. We validated this method using two mRNAs, each with different untranslated regions (UTRs) but encoding enhanced green fluorescence protein (eGFP), and investigated whether the peptide barcodes could analyze the differential eGFP expression levels. RESULTS: The fluorescence intensity of eGFP, a marker of its expression level, has shown noticeable changes between the two UTR sequences in mRNA-transfected cells when measured using flow cytometry. This suggests alterations in the expression level of eGFP due to the influence of different UTR sequences. Furthermore, the quantified amount of peptide barcodes that were released from eGFP showed consistent patterns with these changes. CONCLUSIONS: The experimental findings suggest that peptide barcodes serve as a valuable tool for assessing protein expression levels. The process of mRNA sequence selection, aimed at maximizing protein expression, can be enhanced by the parallel analysis of peptide barcodes using mass spectrometry.


Assuntos
Proteínas de Fluorescência Verde , Peptídeos , RNA Mensageiro , RNA Mensageiro/genética , RNA Mensageiro/análise , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/química , Proteínas de Fluorescência Verde/metabolismo , Peptídeos/química , Peptídeos/análise , Peptídeos/genética , Peptídeos/metabolismo , Humanos , Espectrometria de Massas/métodos , Perfilação da Expressão Gênica/métodos
2.
Genes (Basel) ; 14(11)2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-38003019

RESUMO

Translation initiation in eukaryotes is regulated at several steps, one of which involves the availability of the cap binding protein to participate in cap-dependent protein synthesis. Binding of eIF4E to translational repressors (eIF4E-binding proteins [4E-BPs]) suppresses translation and is used by cells to link extra- and intracellular cues to protein synthetic rates. The best studied of these interactions involves repression of translation by 4E-BP1 upon inhibition of the PI3K/mTOR signaling pathway. Herein, we characterize a novel 4E-BP, C8ORF88, whose expression is predominantly restricted to early spermatids. C8ORF88:eIF4E interaction is dependent on the canonical eIF4E binding motif (4E-BM) present in other 4E-BPs. Whereas 4E-BP1:eIF4E interaction is dependent on the phosphorylation of 4E-BP1, these sites are not conserved in C8ORF88 indicating a different mode of regulation.


Assuntos
Proteínas de Transporte , Fator de Iniciação 4E em Eucariotos , Proteínas de Transporte/metabolismo , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Fosforilação
3.
Proc Natl Acad Sci U S A ; 120(25): e2300008120, 2023 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-37307456

RESUMO

mRNA translation initiation plays a critical role in learning and memory. The eIF4F complex, composed of the cap-binding protein eIF4E, ATP-dependent RNA helicase eIF4A, and scaffolding protein eIF4G, is a pivotal factor in the mRNA translation initiation process. eIF4G1, the major paralogue of the three eIF4G family members, is indispensable for development, but its function in learning and memory is unknown. To study the role of eIF4G1 in cognition, we used an eIF4G1 haploinsufficient (eIF4G1-1D) mouse model. The axonal arborization of eIF4G1-1D primary hippocampal neurons was significantly disrupted, and the mice displayed impairment in hippocampus-dependent learning and memory. Translatome analysis showed that the translation of mRNAs encoding proteins of the mitochondrial oxidative phosphorylation (OXPHOS) system was decreased in the eIF4G1-1D brain, and OXPHOS was decreased in eIF4G1-silenced cells. Thus, eIF4G1-mediated mRNA translation is crucial for optimal cognitive function, which is dependent on OXPHOS and neuronal morphogenesis.


Assuntos
Fator de Iniciação Eucariótico 4G , Fosforilação Oxidativa , Animais , Camundongos , RNA Mensageiro , Iniciação Traducional da Cadeia Peptídica , Morfogênese , DNA Helicases
4.
J Mol Biol ; 434(9): 167523, 2022 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-35248544

RESUMO

The CCR4-NOT deadenylase complex is a major post-transcriptional regulator of eukaryotic gene expression. CNOT7 and CNOT8 are both vertebrate homologs of the yeast CCR4-NOT catalytic subunit Caf1. They are highly similar and are sometimes considered redundant, but Cnot7 and Cnot8 knockout mice exhibit different phenotypes, implying distinct physiological functions. In this study, we reveal a non-reciprocal effect of CNOT7 on CNOT8, in which CNOT8 protein is increased in the depletion of CNOT7 without corresponding changes in mRNA levels whereas CNOT7 is not affected by the loss of CNOT8. Cnot8 mRNA may be bound by the CCR4-NOT complex, suggesting that CCR4-NOT might directly regulate CNOT8 expression. Cnot8 mRNA is relatively unstable, but Cnot7 knockdown did not stabilize Cnot8 mRNA, nor did it increase translation. CNOT8 protein was also less stable than CNOT7. CNOT7 showed greater affinity than CNOT8 for the CCR4-NOT scaffold protein CNOT1 and was able to block CNOT8 from binding to CNOT1. Depletion of CNOT7 increased CNOT8 incorporation into the CCR4-NOT complex and stabilized CNOT8. These data suggest that CNOT7 is the dominant paralog in CCR4-NOT and that CNOT7 and CNOT8 protein stability is regulated in distinct ways.


Assuntos
Exorribonucleases , Proteínas Repressoras , Fatores de Transcrição , Animais , Exorribonucleases/genética , Exorribonucleases/metabolismo , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Ribonucleases/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
iScience ; 24(10): 103151, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34646989

RESUMO

Control of mRNA stability and degradation is essential for appropriate gene expression, and its dysregulation causes various disorders, including cancer, neurodegenerative diseases, diabetes, and obesity. The 5'-3' exoribonuclease XRN1 executes the last step of RNA decay, but its physiological impact is not well understood. To address this, forebrain-specific Xrn1 conditional knockout mice (Xrn1-cKO) were generated, as Xrn1 null mice were embryonic lethal. Xrn1-cKO mice exhibited obesity with leptin resistance, hyperglycemia, hyperphagia, and decreased energy expenditure. Obesity resulted from dysregulated communication between the central nervous system and peripheral tissues. Moreover, expression of mRNAs encoding proteins that regulate appetite and energy expenditure was dysregulated in the hypothalamus of Xrn1-cKO mice. Therefore, we propose that XRN1 function in the hypothalamus is critical for maintenance of metabolic homeostasis.

6.
Cell Mol Life Sci ; 78(19-20): 6709-6719, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34559254

RESUMO

Eukaryotic initiation factor (eIF) 4F plays a central role in the ribosome recruitment phase of cap-dependent translation. This heterotrimeric complex consists of a cap binding subunit (eIF4E), a DEAD-box RNA helicase (eIF4A), and a large bridging protein (eIF4G). In mammalian cells, there are two genes encoding eIF4A (eIF4A1 and eIF4A2) and eIF4G (eIF4G1 and eIF4G3) paralogs that can assemble into eIF4F complexes. To query the essential nature of the eIF4F subunits in normal development, we used CRISPR/Cas9 to generate mouse strains with targeted ablation of each gene encoding the different eIF4F subunits. We find that Eif4e, Eif4g1, and Eif4a1 are essential for viability in the mouse, whereas Eif4g3 and Eif4a2 are not. However, Eif4g3 and Eif4a2 do play essential roles in spermatogenesis. Crossing of these strains to the lymphoma-prone Eµ-Myc mouse model revealed that heterozygosity at the Eif4e or Eif4a1 loci significantly delayed tumor onset. Lastly, tumors derived from Eif4e∆38 fs/+/Eµ-Myc or Eif4a1∆5 fs/+/Eµ-Myc mice show increased sensitivity to the chemotherapeutic agent doxorubicin, in vivo. Our study reveals that eIF4A2 and eIF4G3 play non-essential roles in gene expression regulation during embryogenesis; whereas reductions in eIF4E or eIF4A1 levels are protective against tumor development in a murine Myc-driven lymphoma setting.


Assuntos
Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Fator de Iniciação 4F em Eucariotos/genética , Animais , Feminino , Regulação da Expressão Gênica/genética , Heterozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Subunidades Proteicas/genética , Espermatogênese/genética
7.
Commun Biol ; 3(1): 476, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859966

RESUMO

Pancreatic ß-cells are responsible for production and secretion of insulin in response to increasing blood glucose levels. Defects in ß-cell function lead to hyperglycemia and diabetes mellitus. Here, we show that CNOT3, a CCR4-NOT deadenylase complex subunit, is dysregulated in islets in diabetic db/db mice, and that it is essential for murine ß cell maturation and identity. Mice with ß cell-specific Cnot3 deletion (Cnot3ßKO) exhibit impaired glucose tolerance, decreased ß cell mass, and they gradually develop diabetes. Cnot3ßKO islets display decreased expression of key regulators of ß cell maturation and function. Moreover, they show an increase of progenitor cell markers, ß cell-disallowed genes, and genes relevant to altered ß cell function. Cnot3ßKO islets exhibit altered deadenylation and increased mRNA stability, partly accounting for the increased expression of those genes. Together, these data reveal that CNOT3-mediated mRNA deadenylation and decay constitute previously unsuspected post-transcriptional mechanisms essential for ß cell identity.


Assuntos
Diabetes Mellitus Experimental/patologia , Células Secretoras de Insulina/patologia , Fatores de Transcrição/metabolismo , Animais , Contagem de Células , Diferenciação Celular/genética , Diabetes Mellitus Experimental/genética , Modelos Animais de Doenças , Glucose/toxicidade , Teste de Tolerância a Glucose , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Lipídeos/toxicidade , Masculino , Camundongos Knockout , Modelos Biológicos , Obesidade/patologia , Fenótipo , Proteoma/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma/genética
8.
Life Sci Alliance ; 3(5)2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32238456

RESUMO

The biological significance of deadenylation in global gene expression is not fully understood. Here, we show that the CCR4-NOT deadenylase complex maintains expression of mRNAs, such as those encoding transcription factors, cell cycle regulators, DNA damage response-related proteins, and metabolic enzymes, at appropriate levels in the liver. Liver-specific disruption of Cnot1, encoding a scaffold subunit of the CCR4-NOT complex, leads to increased levels of mRNAs for transcription factors, cell cycle regulators, and DNA damage response-related proteins because of reduced deadenylation and stabilization of these mRNAs. CNOT1 suppression also results in an increase of immature, unspliced mRNAs (pre-mRNAs) for apoptosis-related and inflammation-related genes and promotes RNA polymerase II loading on their promoter regions. In contrast, mRNAs encoding metabolic enzymes become less abundant, concomitant with decreased levels of these pre-mRNAs. Lethal hepatitis develops concomitantly with abnormal mRNA expression. Mechanistically, the CCR4-NOT complex targets and destabilizes mRNAs mainly through its association with Argonaute 2 (AGO2) and butyrate response factor 1 (BRF1) in the liver. Therefore, the CCR4-NOT complex contributes to liver homeostasis by modulating the liver transcriptome through mRNA deadenylation.


Assuntos
Proteínas de Homeodomínio/metabolismo , Fígado/metabolismo , Receptores CCR4/metabolismo , Animais , Citoplasma/metabolismo , Feminino , Proteínas de Homeodomínio/genética , Homeostase , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Poli A/genética , Estabilidade de RNA , RNA Mensageiro/genética , Receptores CCR4/genética , Ribonucleases/genética , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fatores de Transcrição/genética
9.
RNA Biol ; 17(3): 403-416, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31924127

RESUMO

Shortening of mRNA poly(A) tails (deadenylation) to trigger their decay is mediated mainly by the CCR4-NOT deadenylase complex. While four catalytic subunits (CNOT6, 6L 7, and 8) have been identified in the mammalian CCR4-NOT complex, their individual biological roles are not fully understood. In this study, we addressed the contribution of CNOT7/8 to viability of primary mouse embryonic fibroblasts (MEFs). We found that MEFs lacking CNOT7/8 expression [Cnot7/8-double knockout (dKO) MEFs] undergo cell death, whereas MEFs lacking CNOT6/6L expression (Cnot6/6l-dKO MEFs) remain viable. Co-immunoprecipitation analyses showed that CNOT6/6L are also absent from the CCR4-NOT complex in Cnot7/8-dKO MEFs. In contrast, either CNOT7 or CNOT8 still interacts with other subunits in the CCR4-NOT complex in Cnot6/6l-dKO MEFs. Exogenous expression of a CNOT7 mutant lacking catalytic activity in Cnot7/8-dKO MEFs cannot recover cell viability, even though CNOT6/6L exists to some extent in the CCR4-NOT complex, confirming that CNOT7/8 is essential for viability. Bulk poly(A) tail analysis revealed that mRNAs with longer poly(A) tails are more numerous in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Consistent with elongated poly(A) tails, more mRNAs are upregulated and stabilized in Cnot7/8-dKO MEFs than in Cnot6/6l-dKO MEFs. Importantly, Cnot6/6l-dKO mice are viable and grow normally to adulthood. Taken together, the CNOT7/8 catalytic subunits are essential for deadenylation, which is necessary to maintain cell viability, whereas CNOT6/6L are not.


Assuntos
Exorribonucleases/metabolismo , RNA Mensageiro/metabolismo , Receptores CCR4/metabolismo , Proteínas Repressoras/metabolismo , Animais , Sobrevivência Celular/genética , Exorribonucleases/genética , Feminino , Fibroblastos/citologia , Fibroblastos/fisiologia , Masculino , Camundongos Knockout , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Poli A/genética , Poli A/metabolismo , Subunidades Proteicas , Estabilidade de RNA , RNA Mensageiro/genética , Receptores CCR4/genética , Proteínas Repressoras/genética
10.
PLoS Pathog ; 14(8): e1007264, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30138450

RESUMO

Herpes Simplex Virus 1 (HSV1) is amongst the most clinically advanced oncolytic virus platforms. However, efficient and sustained viral replication within tumours is limiting. Rapamycin can stimulate HSV1 replication in cancer cells, but active-site dual mTORC1 and mTORC2 (mammalian target of rapamycin complex 1 and 2) inhibitors (asTORi) were shown to suppress the virus in normal cells. Surprisingly, using the infected cell protein 0 (ICP0)-deleted HSV1 (HSV1-dICP0), we found that asTORi markedly augment infection in cancer cells and a mouse mammary cancer xenograft. Mechanistically, asTORi repressed mRNA translation in normal cells, resulting in defective antiviral response but also inhibition of HSV1-dICP0 replication. asTORi also reduced antiviral response in cancer cells, however in contrast to normal cells, transformed cells and cells transduced to elevate the expression of eukaryotic initiation factor 4E (eIF4E) or to silence the repressors eIF4E binding proteins (4E-BPs), selectively maintained HSV1-dICP0 protein synthesis during asTORi treatment, ultimately supporting increased viral replication. Our data show that altered eIF4E/4E-BPs expression can act to promote HSV1-dICP0 infection under prolonged mTOR inhibition. Thus, pharmacoviral combination of asTORi and HSV1 can target cancer cells displaying dysregulated eIF4E/4E-BPs axis.


Assuntos
Herpes Simples/patologia , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/genética , Proteínas Imediatamente Precoces/genética , Neoplasias/virologia , Inibidores de Proteínas Quinases/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Domínio Catalítico/efeitos dos fármacos , Proteínas de Ciclo Celular , Células Cultivadas , Chlorocebus aethiops , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Herpes Simples/complicações , Herpes Simples/genética , Humanos , Proteínas Imediatamente Precoces/deficiência , Camundongos , Neoplasias/complicações , Neoplasias/genética , Neoplasias/patologia , Organismos Geneticamente Modificados , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/química , Ubiquitina-Proteína Ligases/deficiência , Células Vero
11.
Oncotarget ; 8(31): 50542-50556, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28881582

RESUMO

Metformin inhibits the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway, which is frequently upregulated in hepatocellular carcinoma (HCC). Metformin has also been shown to induce apoptosis in this cancer. Here, we investigate whether metformin-induced apoptosis in HCC is mediated by the downstream mTORC1 effectors eukaryotic initiation factor 4E and (eIF4E)-binding proteins (4E-BPs). Further, we ask whether changes in 4E-BPs activity during metformin treatment negatively regulate translation of the anti-apoptotic myeloid cell leukemia 1 (Mcl-1) mRNA. A genetic HCC mouse model was employed to assess the ability of metformin to reduce tumor formation, induce apoptosis, and control 4E-BP1 activation and Mcl-1 protein expression. In parallel, the HCC cell line Huh7 was transduced with scrambled shRNA (control) or shRNAs targeting 4E-BP1 and 4E-BP2 (4E-BP knock-down (KD)) to measure differences in mRNA translation, apoptosis, and Mcl-1 protein expression after metformin treatment. In addition, immunohistochemical staining of eIF4E and 4E-BP1 protein levels was addressed in a HCC patient tissue microarray. We found that metformin decreased HCC tumor burden, and tumor tissues showed elevated apoptosis with reduced Mcl-1 and phosphorylated 4E-BP1 protein levels. In control but not 4E-BP KD Huh7 cells, metformin induced apoptosis and repressed Mcl-1 mRNA translation and protein levels. Immunostaining of HCC patient tumor tissues revealed a varying ratio of eIF4E/4E-BP1 expression. Our results propose that metformin induces apoptosis in mouse and cellular models of HCC through activation of 4E-BPs, thus tumors with elevated expression of 4E-BPs may display improved clinical chemopreventive benefit of metformin.

12.
Proc Natl Acad Sci U S A ; 113(44): 12360-12367, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27791185

RESUMO

Translational control of gene expression plays a key role during the early phases of embryonic development. Here we describe a transcriptional regulator of mouse embryonic stem cells (mESCs), Yin-yang 2 (YY2), that is controlled by the translation inhibitors, Eukaryotic initiation factor 4E-binding proteins (4E-BPs). YY2 plays a critical role in regulating mESC functions through control of key pluripotency factors, including Octamer-binding protein 4 (Oct4) and Estrogen-related receptor-ß (Esrrb). Importantly, overexpression of YY2 directs the differentiation of mESCs into cardiovascular lineages. We show that the splicing regulator Polypyrimidine tract-binding protein 1 (PTBP1) promotes the retention of an intron in the 5'-UTR of Yy2 mRNA that confers sensitivity to 4E-BP-mediated translational suppression. Thus, we conclude that YY2 is a major regulator of mESC self-renewal and lineage commitment and document a multilayer regulatory mechanism that controls its expression.


Assuntos
Processamento Alternativo/fisiologia , Diferenciação Celular , Autorrenovação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição/metabolismo , Animais , Blastocisto/metabolismo , Proteínas de Transporte/metabolismo , Linhagem da Célula , Autorrenovação Celular/genética , Ribonucleoproteínas Nucleares Heterogêneas/genética , Íntrons , Camundongos , Camundongos Knockout , Modelos Biológicos , Fator 3 de Transcrição de Octâmero/metabolismo , Fosfoproteínas , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Biossíntese de Proteínas/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Estrogênio/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia , Fator de Transcrição YY1/metabolismo
13.
Nat Commun ; 7: 12188, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27432119

RESUMO

Parkinson's disease gene leucine-rich repeat kinase 2 (LRRK2) has been implicated in a number of processes including the regulation of mitochondrial function, autophagy and endocytic dynamics; nevertheless, we know little about its potential role in the regulation of synaptic plasticity. Here we demonstrate that postsynaptic knockdown of the fly homologue of LRRK2 thwarts retrograde, homeostatic synaptic compensation at the larval neuromuscular junction. Conversely, postsynaptic overexpression of either the fly or human LRRK2 transgene induces a retrograde enhancement of presynaptic neurotransmitter release by increasing the size of the release ready pool of vesicles. We show that LRRK2 promotes cap-dependent translation and identify Furin 1 as its translational target, which is required for the synaptic function of LRRK2. As the regulation of synaptic homeostasis plays a fundamental role in ensuring normal and stable synaptic function, our findings suggest that aberrant function of LRRK2 may lead to destabilization of neural circuits.


Assuntos
Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/metabolismo , Junção Neuromuscular/metabolismo , Sinapses/metabolismo , Animais , Furina/metabolismo , Células HEK293 , Homeostase , Humanos , Larva/metabolismo , Proteínas Mutantes/metabolismo , Mutação/genética , Neurotransmissores/metabolismo , Biossíntese de Proteínas , Capuzes de RNA/metabolismo , Transdução de Sinais , Transmissão Sináptica , Vesículas Sinápticas/metabolismo
14.
Diabetes ; 65(8): 2235-48, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27217487

RESUMO

The mammalian target of rapamycin complex 1 (mTORC1) regulates several biological processes, although the key downstream mechanisms responsible for these effects are poorly defined. Using mice with deletion of eukaryotic translation initiation factor 4E-binding protein 2 (4E-BP2), we determine that this downstream target is a major regulator of glucose homeostasis and ß-cell mass, proliferation, and survival by increasing insulin receptor substrate 2 (IRS2) levels and identify a novel feedback mechanism by which mTORC1 signaling increases IRS2 levels. In this feedback loop, we show that 4E-BP2 deletion induces translation of the adaptor protein SH2B1 and promotes the formation of a complex with IRS2 and Janus kinase 2, preventing IRS2 ubiquitination. The changes in IRS2 levels result in increases in cell cycle progression, cell survival, and ß-cell mass by increasing Akt signaling and reducing p27 levels. Importantly, 4E-BP2 deletion confers resistance to cytokine treatment in vitro. Our data identify SH2B1 as a major regulator of IRS2 stability, demonstrate a novel feedback mechanism linking mTORC1 signaling with IRS2, and identify 4E-BP2 as a major regulator of proliferation and survival of ß-cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Ciclo Celular/genética , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Proliferação de Células/fisiologia , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Fatores de Iniciação em Eucariotos/genética , Proteínas Substratos do Receptor de Insulina/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Estabilidade Proteica , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
15.
Mol Cell ; 61(5): 760-773, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26942679

RESUMO

MicroRNAs predominantly decrease gene expression; however, specific mRNAs are translationally upregulated in quiescent (G0) mammalian cells and immature Xenopus laevis oocytes by an FXR1a-associated microRNA-protein complex (microRNP) that lacks the microRNP repressor, GW182. Their mechanism in these conditions of decreased mTOR signaling, and therefore reduced canonical (cap-and-poly(A)-tail-mediated) translation, remains undiscovered. Our data reveal that mTOR inhibition in human THP1 cells enables microRNA-mediated activation. Activation requires shortened/no poly(A)-tail targets; polyadenylated mRNAs are partially activated upon PAIP2 overexpression, which interferes with poly(A)-bound PABP, precluding PABP-enhanced microRNA-mediated inhibition and canonical translation. Consistently, inhibition of PARN deadenylase prevents activation. P97/DAP5, a homolog of canonical translation factor, eIF4G, which lacks PABP- and cap binding complex-interacting domains, is required for activation, and thereby for the oocyte immature state. P97 interacts with 3' UTR-binding FXR1a-associated microRNPs and with PARN, which binds mRNA 5' caps, forming a specialized complex to translate recruited mRNAs in these altered canonical translation conditions.


Assuntos
Senescência Celular , MicroRNAs/metabolismo , Oócitos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ribonucleoproteínas/metabolismo , Regiões 3' não Traduzidas , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sítios de Ligação , Linhagem Celular , Fator de Iniciação Eucariótico 4G/genética , Fator de Iniciação Eucariótico 4G/metabolismo , Exorribonucleases/genética , Exorribonucleases/metabolismo , Perfilação da Expressão Gênica/métodos , Humanos , MicroRNAs/genética , Proteômica/métodos , Capuzes de RNA/genética , Capuzes de RNA/metabolismo , Interferência de RNA , RNA Mensageiro/genética , Proteínas de Ligação a RNA/genética , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Ribonucleoproteínas/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Transfecção , Xenopus laevis
16.
J Biol Chem ; 290(26): 15996-6020, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25940091

RESUMO

The mammalian target of rapamycin complex 1 (mTORC1) is a critical regulator of protein synthesis. The best studied targets of mTORC1 in translation are the eukaryotic initiation factor-binding protein 1 (4E-BP1) and ribosomal protein S6 kinase 1 (S6K1). In this study, we identify the La-related protein 1 (LARP1) as a key novel target of mTORC1 with a fundamental role in terminal oligopyrimidine (TOP) mRNA translation. Recent genome-wide studies indicate that TOP and TOP-like mRNAs compose a large portion of the mTORC1 translatome, but the mechanism by which mTORC1 controls TOP mRNA translation is incompletely understood. Here, we report that LARP1 functions as a key repressor of TOP mRNA translation downstream of mTORC1. Our data show the following: (i) LARP1 associates with mTORC1 via RAPTOR; (ii) LARP1 interacts with TOP mRNAs in an mTORC1-dependent manner; (iii) LARP1 binds the 5'TOP motif to repress TOP mRNA translation; and (iv) LARP1 competes with the eukaryotic initiation factor (eIF) 4G for TOP mRNA binding. Importantly, from a drug resistance standpoint, our data also show that reducing LARP1 protein levels by RNA interference attenuates the inhibitory effect of rapamycin, Torin1, and amino acid deprivation on TOP mRNA translation. Collectively, our findings demonstrate that LARP1 functions as an important repressor of TOP mRNA translation downstream of mTORC1.


Assuntos
Autoantígenos/metabolismo , Regulação para Baixo , Glicoproteínas de Membrana/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/genética , Ribonucleoproteínas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autoantígenos/genética , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Glicoproteínas de Membrana/genética , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Ligação Proteica , RNA Longo não Codificante , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Proteína Regulatória Associada a mTOR , Ribonucleoproteínas/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Antígeno SS-B
17.
Nat Neurosci ; 18(6): 855-62, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25915475

RESUMO

The circadian (∼24 h) clock is continuously entrained (reset) by ambient light so that endogenous rhythms are synchronized with daily changes in the environment. Light-induced gene expression is thought to be the molecular mechanism underlying clock entrainment. mRNA translation is a key step of gene expression, but the manner in which clock entrainment is controlled at the level of mRNA translation is not well understood. We found that a light- and circadian clock-regulated MAPK/MNK pathway led to phosphorylation of the cap-binding protein eIF4E in the mouse suprachiasmatic nucleus of the hypothalamus, the locus of the master circadian clock in mammals. Phosphorylation of eIF4E specifically promoted translation of Period 1 (Per1) and Period 2 (Per2) mRNAs and increased the abundance of basal and inducible PER proteins, which facilitated circadian clock resetting and precise timekeeping. Together, these results highlight a critical role for light-regulated translational control in the physiology of the circadian clock.


Assuntos
Comportamento Animal/fisiologia , Ritmo Circadiano/fisiologia , Fator de Iniciação 4E em Eucariotos/fisiologia , Animais , Comportamento Animal/efeitos da radiação , Química Encefálica/genética , Química Encefálica/fisiologia , Ritmo Circadiano/efeitos da radiação , Regulação da Expressão Gênica/efeitos da radiação , Luz , Sistema de Sinalização das MAP Quinases/efeitos da radiação , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiologia , Fosforilação , Núcleo Supraquiasmático/metabolismo , Núcleo Supraquiasmático/fisiologia
18.
Neuron ; 79(4): 712-24, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23972597

RESUMO

Protein synthesis is critical for circadian clock function, but little is known of how translational regulation controls the master pacemaker in mammals, the suprachiasmatic nucleus (SCN). Here we demonstrate that the pivotal translational repressor, the eukaryotic translational initiation factor 4E binding protein 1 (4E-BP1), is rhythmically regulated via the mechanistic target of rapamycin (mTOR) signaling in the SCN and preferentially represses vasoactive intestinal peptide (Vip) mRNA translation. Knockout (KO) of Eif4ebp1 (gene encoding 4E-BP1) leads to upregulation of VIP and higher amplitude of molecular rhythms in the SCN. Consequently, the 4E-BP1 null mice exhibit accelerated re-entrainment to a shifted light/dark cycle and are more resistant to the rhythm-disruptive effects of constant light. Conversely, in Mtor(+/-) mice VIP expression is decreased and susceptibility to the effects of constant light is increased. These results reveal a key role for mTOR/4E-BP1-mediated translational control in regulating entrainment and synchrony of the master clock.


Assuntos
Proteínas de Transporte/fisiologia , Ritmo Circadiano/fisiologia , Regulação da Expressão Gênica/fisiologia , Fosfoproteínas/fisiologia , Transdução de Sinais/fisiologia , Núcleo Supraquiasmático/metabolismo , Serina-Treonina Quinases TOR/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antraquinonas/farmacologia , Butadienos/farmacologia , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Inibidores Enzimáticos/farmacologia , Fatores de Iniciação em Eucariotos , Regulação da Expressão Gênica/efeitos dos fármacos , Indóis/farmacologia , Luz , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nitrilas/farmacologia , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fosfoproteínas/deficiência , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Purinas/farmacologia , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Peptídeo Intestinal Vasoativo/genética , Peptídeo Intestinal Vasoativo/metabolismo
19.
PLoS Biol ; 11(5): e1001564, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23700384

RESUMO

Translation control often operates via remodeling of messenger ribonucleoprotein particles. The poly(A) binding protein (PABP) simultaneously interacts with the 3' poly(A) tail of the mRNA and the eukaryotic translation initiation factor 4G (eIF4G) to stimulate translation. PABP also promotes miRNA-dependent deadenylation and translational repression of target mRNAs. We demonstrate that isoform 2 of the mouse heterogeneous nuclear protein Q (hnRNP-Q2/SYNCRIP) binds poly(A) by default when PABP binding is inhibited. In addition, hnRNP-Q2 competes with PABP for binding to poly(A) in vitro. Depleting hnRNP-Q2 from translation extracts stimulates cap-dependent and IRES-mediated translation that is dependent on the PABP/poly(A) complex. Adding recombinant hnRNP-Q2 to the extracts inhibited translation in a poly(A) tail-dependent manner. The displacement of PABP from the poly(A) tail by hnRNP-Q2 impaired the association of eIF4E with the 5' m(7)G cap structure of mRNA, resulting in the inhibition of 48S and 80S ribosome initiation complex formation. In mouse fibroblasts, silencing of hnRNP-Q2 stimulated translation. In addition, hnRNP-Q2 impeded let-7a miRNA-mediated deadenylation and repression of target mRNAs, which require PABP. Thus, by competing with PABP, hnRNP-Q2 plays important roles in the regulation of global translation and miRNA-mediated repression of specific mRNAs.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas/genética , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , MicroRNAs/metabolismo , Animais , Sítios de Ligação , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fibroblastos/metabolismo , Células HeLa , Humanos , Camundongos , MicroRNAs/genética
20.
Neuron ; 78(2): 298-311, 2013 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-23622065

RESUMO

Control of protein synthesis is critical for synaptic plasticity and memory formation. However, the molecular mechanisms linking neuronal activity to activation of mRNA translation are not fully understood. Here, we report that the translational repressor poly(A)-binding protein (PABP)-interacting protein 2A (PAIP2A), an inhibitor of PABP, is rapidly proteolyzed by calpains in stimulated neurons and following training for contextual memory. Paip2a knockout mice exhibit a lowered threshold for the induction of sustained long-term potentiation and an enhancement of long-term memory after weak training. Translation of CaMKIIα mRNA is enhanced in Paip2a⁻/⁻ slices upon tetanic stimulation and in the hippocampus of Paip2a⁻/⁻ mice following contextual fear learning. We demonstrate that activity-dependent degradation of PAIP2A relieves translational inhibition of memory-related genes through PABP reactivation and conclude that PAIP2A is a pivotal translational regulator of synaptic plasticity and memory.


Assuntos
Potenciação de Longa Duração/genética , Memória/fisiologia , Neurônios/fisiologia , Sinapses/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Adenosina Trifosfatases/farmacologia , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Calpaína/farmacologia , Células Cultivadas , Condicionamento Psicológico/efeitos dos fármacos , Condicionamento Psicológico/fisiologia , Dactinomicina/farmacologia , Inibidores Enzimáticos/farmacologia , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Hipocampo/citologia , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Oligodesoxirribonucleotídeos/farmacologia , Proteínas de Ligação a Poli(A) , Inibidores da Síntese de Proteínas/farmacologia , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA , Tempo de Reação/efeitos dos fármacos , Tempo de Reação/genética , Proteínas Repressoras , Proteínas Supressoras de Tumor/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA