Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 491(7424): 454-7, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23064229

RESUMO

Most of the mammalian genome is transcribed. This generates a vast repertoire of transcripts that includes protein-coding messenger RNAs, long non-coding RNAs (lncRNAs) and repetitive sequences, such as SINEs (short interspersed nuclear elements). A large percentage of ncRNAs are nuclear-enriched with unknown function. Antisense lncRNAs may form sense-antisense pairs by pairing with a protein-coding gene on the opposite strand to regulate epigenetic silencing, transcription and mRNA stability. Here we identify a nuclear-enriched lncRNA antisense to mouse ubiquitin carboxy-terminal hydrolase L1 (Uchl1), a gene involved in brain function and neurodegenerative diseases. Antisense Uchl1 increases UCHL1 protein synthesis at a post-transcriptional level, hereby identifying a new functional class of lncRNAs. Antisense Uchl1 activity depends on the presence of a 5' overlapping sequence and an embedded inverted SINEB2 element. These features are shared by other natural antisense transcripts and can confer regulatory activity to an artificial antisense to green fluorescent protein. Antisense Uchl1 function is under the control of stress signalling pathways, as mTORC1 inhibition by rapamycin causes an increase in UCHL1 protein that is associated to the shuttling of antisense Uchl1 RNA from the nucleus to the cytoplasm. Antisense Uchl1 RNA is then required for the association of the overlapping sense protein-coding mRNA to active polysomes for translation. These data reveal another layer of gene expression control at the post-transcriptional level.


Assuntos
Biossíntese de Proteínas/genética , RNA Antissenso/metabolismo , Elementos Nucleotídeos Curtos e Dispersos/genética , Ubiquitina Tiolesterase/genética , Ubiquitina Tiolesterase/metabolismo , Animais , Antibacterianos/farmacologia , Linhagem Celular , Humanos , Masculino , Camundongos , Biossíntese de Proteínas/efeitos dos fármacos , RNA Antissenso/genética , Inversão de Sequência , Sirolimo/farmacologia
2.
Biochem Biophys Res Commun ; 493(4): 1567-1572, 2017 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-29017919

RESUMO

Antibodies are essential reagents that are increasingly used in diagnostics and therapy. Their specificity and capacity to recognize their native antigen are critical characteristics for their in vivo application. Follicle-stimulating hormone receptor is a GPCR protein regulating ovarian follicular maturation and spermatogenesis. Recently, its potentiality as a cancer biomarker has been demonstrated but no antibody suitable for in vivo tumor targeting and treatment has been characterized so far. In this paper we describe the first successful attempt to recover recombinant antibodies against the FSHR and that: i) are directly panned from a pre-immune library using whole cells expressing the target receptor at their surface; ii) show inhibitory activity towards the FSH-induced cAMP accumulation; iii) do not share the same epitope with the natural binder FSH; iv) can be produced inexpensively as mono- or bivalent functional molecules in the bacterial cytoplasm. We expect that the proposed biopanning strategy will be profitable to identify useful functional antibodies for further members of the GPCR class.


Assuntos
Biblioteca de Peptídeos , Receptores do FSH/antagonistas & inibidores , Receptores do FSH/imunologia , Anticorpos de Domínio Único/genética , Anticorpos de Domínio Único/imunologia , Animais , Especificidade de Anticorpos , AMP Cíclico/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Células HEK293 , Humanos , Imunização , Células L , Masculino , Camundongos , Domínios Proteicos , Receptores do FSH/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Transdução de Sinais , Solubilidade
3.
Microb Cell Fact ; 13: 140, 2014 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-25223348

RESUMO

BACKGROUND: The isolation of recombinant antibody fragments from displayed libraries represents a powerful alternative to the generation of IgGs using hybridoma technology. The selected antibody fragments can then be easily engineered into (multi)-tagged constructs of variable mass and complexity as well as reconstituted into Camelidae IgG-like molecules when expressed fused to Fc domains. Nevertheless, all antibody constructs depend on an oxidizing environment for correct folding and consequently still belong to the proteins difficult to express in bacteria. In such organisms they are mostly produced at low yields in the periplasmic space. RESULTS: We demonstrate that fusion constructs of recombinant antibodies in combination with multiple tags can be produced at high yields and totally functional in the cytoplasm of bacteria expressing sulfhydryl oxidase. The method was applied to structurally demanding molecules such as VHHs fused to SNAP and Fc domains and was validated using the antibody-derived reagents in a variety of immune techniques (FACS, ELISA, WB, IP, SPR, and IF). CONCLUSIONS: The collected data demonstrate the feasibility of a method that establishes a totally new approach for producing rapidly and inexpensively functional Camelidae IgG-like monoclonal antibodies and antibody-based reagents containing multiple disulfide bonds and suitable for both basic research and clinical applications.


Assuntos
Camelus/metabolismo , Compartimento Celular , Citoplasma/metabolismo , Escherichia coli/metabolismo , Imunoglobulina G/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Anticorpos de Cadeia Única/biossíntese , Animais , Afinidade de Anticorpos , Linhagem Celular Tumoral , Imunofluorescência , Humanos , Camundongos , Periplasma/metabolismo , Receptor ErbB-2/metabolismo , Reprodutibilidade dos Testes , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Nature ; 455(7213): 684-8, 2008 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-18784653

RESUMO

Cell growth and proliferation require coordinated ribosomal biogenesis and translation. Eukaryotic initiation factors (eIFs) control translation at the rate-limiting step of initiation. So far, only two eIFs connect extracellular stimuli to global translation rates: eIF4E acts in the eIF4F complex and regulates binding of capped messenger RNA to 40S subunits, downstream of growth factors, and eIF2 controls loading of the ternary complex on the 40S subunit and is inhibited on stress stimuli. No eIFs have been found to link extracellular stimuli to the activity of the large 60S ribosomal subunit. eIF6 binds 60S ribosomes precluding ribosome joining in vitro. However, studies in yeasts showed that eIF6 is required for ribosome biogenesis rather than translation. Here we show that mammalian eIF6 is required for efficient initiation of translation, in vivo. eIF6 null embryos are lethal at preimplantation. Heterozygous mice have 50% reduction of eIF6 levels in all tissues, and show reduced mass of hepatic and adipose tissues due to a lower number of cells and to impaired G1/S cell cycle progression. eIF6(+/-) cells retain sufficient nucleolar eIF6 and normal ribosome biogenesis. The liver of eIF6(+/-) mice displays an increase of 80S in polysomal profiles, indicating a defect in initiation of translation. Consistently, isolated hepatocytes have impaired insulin-stimulated translation. Heterozygous mouse embryonic fibroblasts recapitulate the organism phenotype and have normal ribosome biogenesis, reduced insulin-stimulated translation, and delayed G1/S phase progression. Furthermore, eIF6(+/-) cells are resistant to oncogene-induced transformation. Thus, eIF6 is the first eIF associated with the large 60S subunit that regulates translation in response to extracellular signals.


Assuntos
Transformação Celular Neoplásica , Iniciação Traducional da Cadeia Peptídica , Fatores de Iniciação de Peptídeos/metabolismo , Fase S , Tecido Adiposo/citologia , Animais , Peso Corporal , Divisão Celular/efeitos dos fármacos , Nucléolo Celular/metabolismo , Células Cultivadas , Citoplasma/metabolismo , Fibroblastos , Fase G1/efeitos dos fármacos , Heterozigoto , Insulina/farmacologia , Fígado/citologia , Fígado/crescimento & desenvolvimento , Camundongos , Células NIH 3T3 , Oncogenes/genética , Iniciação Traducional da Cadeia Peptídica/efeitos dos fármacos , Fatores de Iniciação de Peptídeos/deficiência , Fatores de Iniciação de Peptídeos/genética , Ribossomos/química , Ribossomos/metabolismo , Fase S/efeitos dos fármacos
5.
Cell Mol Life Sci ; 70(8): 1439-50, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23212600

RESUMO

The receptor for activated C-kinase 1 (RACK1) is a conserved structural protein of 40S ribosomes. Strikingly, deletion of RACK1 in yeast homolog Asc1 is not lethal. Mammalian RACK1 also interacts with many nonribosomal proteins, hinting at several extraribosomal functions. A knockout mouse for RACK1 has not previously been described. We produced the first RACK1 mutant mouse, in which both alleles of RACK1 gene are defective in RACK1 expression (ΔF/ΔF), in a pure C57 Black/6 background. In a sample of 287 pups, we observed no ΔF/ΔF mice (72 expected). Dissection and genotyping of embryos at various stages showed that lethality occurs at gastrulation. Heterozygotes (ΔF/+) have skin pigmentation defects with a white belly spot and hypopigmented tail and paws. ΔF/+ have a transient growth deficit (shown by measuring pup size at P11). The pigmentation deficit is partly reverted by p53 deletion, whereas the lethality is not. ΔF/+ livers have mild accumulation of inactive 80S ribosomal subunits by polysomal profile analysis. In ΔF/+ fibroblasts, protein synthesis response to extracellular and pharmacological stimuli is reduced. These results highlight the role of RACK1 as a ribosomal protein converging signaling to the translational apparatus.


Assuntos
Neuropeptídeos/genética , Pigmentação , Biossíntese de Proteínas , Animais , Células Cultivadas , Perda do Embrião/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/ultraestrutura , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neuropeptídeos/metabolismo , Fenótipo , Interferência de RNA , RNA Interferente Pequeno/genética , Receptores de Quinase C Ativada , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
EMBO Rep ; 10(5): 459-65, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19373251

RESUMO

Eukaryotic ribosome biogenesis and translation are linked processes that limit the rate of cell growth. Although ribosome biogenesis and translation are mainly controlled by distinct factors, eukaryotic initiation factor 6 (eIF6) has been found to regulate both processes. eIF6 is a necessary protein with a unique anti-association activity, which prevents the interaction of 40S ribosomal subunits with 60S subunits through its binding to 60S ribosomes. In the nucleolus, eIF6 is a component of the pre-ribosomal particles and is required for the biogenesis of 60S subunits, whereas in the cytoplasm it mediates translation downstream from growth factors. The translational activity of eIF6 could be due to its anti-association properties, which are regulated by post-translational modifications; whether this anti-association activity is required for the biogenesis and nuclear export of ribosomes is unknown. eIF6 is necessary for tissue-specific growth and oncogene-driven transformation, and could be a new rate-limiting step for the initiation of translation.


Assuntos
Fatores de Iniciação de Peptídeos/fisiologia , Biossíntese de Proteínas/fisiologia , Subunidades Ribossômicas Maiores de Eucariotos/metabolismo , Animais , Humanos , Modelos Biológicos , Fatores de Iniciação de Peptídeos/química , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas/genética
7.
iScience ; 23(4): 100987, 2020 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-32224433

RESUMO

Human mononuclear phagocytes comprise several subsets of dendritic cells (DCs), monocytes, and macrophages. Distinguishing one population from another is challenging, especially in inflamed tissues, owing to the promiscuous expression of phenotypic markers. Using a synthetic library of humanized llama single domain antibodies, we identified a novel surface marker for human naturally occurring monocyte-derived DCs. Our antibody targets an extra-cellular domain of LSP-1, specifically on monocyte-derived DCs, but not on other leukocytes, in particular monocytes, macrophages, classical DCs, or the recently described blood DC3 population. Our findings will pave the way for a better characterization of human mononuclear phagocytes in pathological settings.

8.
Mol Cell Biol ; 25(7): 2558-72, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15767663

RESUMO

Signaling through the mammalian target of rapamycin (mTOR) controls cell size and growth as well as other functions, and it is a potential therapeutic target for graft rejection, certain cancers, and disorders characterized by inappropriate cell or tissue growth. mTOR signaling is positively regulated by hormones or growth factors and amino acids. mTOR signaling regulates the phosphorylation of several proteins, the best characterized being ones that control mRNA translation. Eukaryotic initiation factor 4E-binding protein 1 (4E-BP1) undergoes phosphorylation at multiple sites. Here we show that amino acids regulate the N-terminal phosphorylation sites in 4E-BP1 through the RAIP motif in a rapamycin-insensitive manner. Several criteria indicate this reflects a rapamycin-insensitive output from mTOR. In contrast, the insulin-stimulated phosphorylation of the C-terminal site Ser64/65 is generally sensitive to rapamycin, as is phosphorylation of another well-characterized target for mTOR signaling, S6K1. Our data imply that it is unlikely that mTOR directly phosphorylates Thr69/70 in 4E-BP1. Although 4E-BP1 and S6K1 bind the mTOR partner, raptor, our data indicate that the outputs from mTOR to 4E-BP1 and S6K1 are distinct. In cells, efficient phosphorylation of 4E-BP1 requires it to be able to bind to eIF4E, whereas phosphorylation of 4E-BP1 by mTOR in vitro shows no such preference. These data have important implications for understanding signaling downstream of mTOR and the development of new strategies to impair mTOR signaling.


Assuntos
Aminoácidos/farmacologia , Proteínas de Transporte/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Insulina/farmacologia , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Ciclo Celular , Linhagem Celular , Cromonas/farmacologia , Cricetinae , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Morfolinas/farmacologia , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Quinases/genética , Ratos , Proteínas Quinases S6 Ribossômicas/metabolismo , Alinhamento de Sequência , Serina/genética , Serina/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Treonina/genética , Treonina/metabolismo
9.
Mol Cell Biol ; 23(5): 1546-57, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12588975

RESUMO

Eukaryotic initiation factor 4E (eIF4E) binds the mRNA cap structure and forms eIF4F complexes that recruit 40S subunits to the mRNA. Formation of eIF4F is blocked by eIF4E-binding proteins such as 4E-BP1, which interacts with eIF4E via a motif in the center of its 118-residue sequence. 4E-BP1 plays key roles in cell proliferation, growth, and survival. Binding of 4E-BP1 to eIF4E is regulated by hierarchical multisite phosphorylation. Here we demonstrate that three different features in the C terminus of 4E-BP1 play distinct roles in regulating its phosphorylation and function. Firstly, we identify a new phosphorylation site in its C terminus (S101). A serine or glutamate at this position is required for efficient phosphorylation at Ser65. A second C-terminal site, S112, directly affects binding of 4E-BP1 to eIF4E without influencing phosphorylation of other sites. Thirdly, a conserved C-terminal motif influences phosphorylation of multiple residues, including rapamycin-insensitive sites. These relatively long-range effects are surprising given the reportedly unstructured nature of 4E-BP1 and may imply that phosphorylation of 4E-BP1 and/or binding to eIF4E induces a more-ordered structure. 4E-BP2 and -3 lack phosphorylatable residues corresponding to both S101 and S112. However, in 4E-BP3, replacement of the alanine at the position corresponding to S112 by serine or glutamate did not confer the ability to be released from eIF4E in response to insulin.


Assuntos
Proteínas de Transporte/química , Fosfoproteínas/química , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Sequência de Aminoácidos , Sítios de Ligação , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Divisão Celular , Linhagem Celular , Sobrevivência Celular , Inibidores Enzimáticos/farmacologia , Fator de Iniciação 4E em Eucariotos/metabolismo , Vetores Genéticos , Ácido Glutâmico/química , Glutamina/metabolismo , Humanos , Immunoblotting , Insulina/farmacologia , Modelos Genéticos , Dados de Sequência Molecular , Mutação , Peptídeos/química , Fosfoproteínas/metabolismo , Fosforilação , Ligação Proteica , Isoformas de Proteínas , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Serina/química , Serina/metabolismo , Fatores de Tempo , Transfecção
10.
J Biotechnol ; 214: 147-55, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26433047

RESUMO

We prepared and characterized polymersomes functionalized with nanobodies (VHHs) on the basis of biocompatible, biodegradable and FDA-approved poly(ethylene glycol)-block-poly(ϵ-caprolactone) (PEG-b-PCL). Fluorescein isothiocyanate (FITC) and N-beta-maleimidopropyl-oxysuccinimide ester were allowed reacting with H2N-PEG-b-PCL to produce FITC and maleimide (Mal) functionalized copolymers, Mal-PEG-b-PCL and FITC-PEG-b-PCL. A mixture of MeO-PEG-b-PCL, Mal-PEG-b-PCL and FITC-PEG-b-PCL was used to prepare polymersomes by thin film hydration and nanoprecipitation methods. Morphological studies by cryogenic transmission electron microscopy (Cryo-TEM) showed that the nanoparticles exhibited predominantly vesicular structures (polymersomes). Their mean diameters measured by dynamic light scattering were around 150 nm and the zeta-potentials around -1 mV at pH 7.4. The nanoparticles were functionalized with either anti-HER2 (VHH1) or anti-GFP (VHH2) nanobodies using maleimide-cysteine chemistry. Their particle size and zeta-potential increased slightly after nanobody-functionalization. The specific binding of VHH-functionalized polymersomes and control nanoparticles towards HER2 positive breast cancer cells was analyzed by flow cytometry and confocal microscopy. The collected results represent the first report which experimentally demonstrates that VHH1-functionalized PEO-b-PCL polymersomes can target specifically breast cancer cells expressing HER2 receptors. The detailed morphological and cell-binding studies described herein pave the way for future in vivo studies to evaluate the feasibility to use such nanoparticles for targeted drug delivery.


Assuntos
Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Lactonas/química , Polietilenoglicóis/química , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Linhagem Celular Tumoral , Microscopia Crioeletrônica , Humanos , Tamanho da Partícula , Receptor ErbB-2/metabolismo
11.
In Vitro Cell Dev Biol Anim ; 47(2): 132-8, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21082278

RESUMO

Ribosomal proteins and ribosomal associated proteins are complicated subjects to target and study because of their high conservation through evolution which led to highly structured and regulated proteins. Tagging of ribosomal proteins may allow following of protein synthesis in vivo and isolating translated mRNAs. HaloTag® is a new technology which allows detection in living cells, biochemical purification, and localization studies. In the present work, we tested HaloTag®-based ribosomal tagging. We focused on eIF6 (eukaryotic Initiation Factor 6 free 60S ribosomal marker), RACK1 (Receptor for Activated C Kinase 1; 40S and polysomes, not nuclear), and rpS9 (40S ribosomes, both in the nucleus and in the cytoplasm). Experiments performed on HEK293 cells included ribosomal profiles and Western blot on the fractions, purification of HaloTag® proteins, and fluorescence with time-lapse microscopy. We show that tagged proteins can be incorporated on ribosomes and followed by time-lapse microscopy. eIF6 properly accumulates in the nucleolus, and it is redistributed upon actinomycin D treatment. RACK1 shows a specific cytoplasmic localization, whereas rpS9 is both nucleolar and cytoplasmic. However, efficiency of purification varies due to steric hindrances. In addition, the level of overexpression and degradation may vary upon different constructs. In summary, HaloTag® technology is highly suitable to ribosome tagging, but requires prior characterization for each construct.


Assuntos
Western Blotting/métodos , Imunofluorescência/métodos , Proteínas Ribossômicas/análise , Ribossomos/química , Imagem com Lapso de Tempo/métodos , Nucléolo Celular/química , Nucléolo Celular/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Células HEK293 , Células HeLa , Humanos , Fatores de Iniciação de Peptídeos/análise , Fatores de Iniciação de Peptídeos/isolamento & purificação , Fatores de Iniciação de Peptídeos/metabolismo , Biossíntese de Proteínas , Proteínas Ribossômicas/isolamento & purificação , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo
12.
PLoS One ; 6(12): e29136, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22216185

RESUMO

Initiation is the rate-limiting phase of protein synthesis, controlled by signaling pathways regulating the phosphorylation of translation factors. Initiation has three steps, 43S, 48S and 80S formation. 43S formation is repressed by eIF2α phosphorylation. The subsequent steps, 48S and 80S formation are enabled by growth factors. 48S relies on eIF4E-mediated assembly of eIF4F complex; 4E-BPs competitively displace eIF4E from eIF4F. Two pathways control eIF4F: 1) mTORc1 phosphorylates and inactivates 4E-BPs, leading to eIF4F formation; 2) the Ras-Mnk cascade phosphorylates eIF4E. We show that REN and NCI-H28 mesothelioma cells have constitutive activation of both pathways and maximal translation rate, in the absence of exogenous growth factors. Translation is rapidly abrogated by phosphorylation of eIF2α. Surprisingly, pharmacological inhibition of mTORc1 leads to the complete dephosphorylation of downstream targets, without changes in methionine incorporation. In addition, the combined administration of mTORc1 and MAPK/Mnk inhibitors has no additive effect. The inhibition of both mTORc1 and mTORc2 does not affect the metabolic rate. In spite of this, mTORc1 inhibition reduces eIF4F complex formation, and depresses translocation of TOP mRNAs on polysomes. Downregulation of eIF4E and overexpression of 4E-BP1 induce rapamycin sensitivity, suggesting that disruption of eIF4F complex, due to eIF4E modulation, competes with its recycling to ribosomes. These data suggest the existence of a dynamic equilibrium in which eIF4F is not essential for all mRNAs and is not displaced from translated mRNAs, before recycling to the next.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fator de Iniciação 4E em Eucariotos/metabolismo , Fosfoproteínas/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Humanos , Metionina/metabolismo , Fosforilação , Ligação Proteica , RNA Mensageiro/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Transcrição Gênica
13.
Cancer Cell ; 19(6): 765-75, 2011 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-21665150

RESUMO

Eukaryotic Initiation Factor 6 (eIF6) controls translation by regulating 80S subunit formation. eIF6 is overexpressed in tumors. Here, we demonstrate that eIF6 inactivation delays tumorigenesis and reduces tumor growth in vivo. eIF6(+/-) mice resist to Myc-induced lymphomagenesis and have prolonged tumor-free survival and reduced tumor growth. eIF6(+/-) mice are also protected by p53 loss. Myc-driven lymphomas contain PKCßII and phosphorylated eIF6; eIF6 is phosphorylated by tumor-derived PKCßII, but not by the eIF4F activator mTORC1. Mutation of PKCßII phosphosite of eIF6 reduces tumor growth. Thus, eIF6 is a rate-limiting controller of initiation of translation, able to affect tumorigenesis and tumor growth. Modulation of eIF6 activity, independent from eIF4F complex, may lead to a therapeutical avenue in tumor therapy.


Assuntos
Transformação Celular Neoplásica/metabolismo , Linfoma/etiologia , Fatores de Iniciação de Peptídeos/fisiologia , Animais , Apoptose , Ciclo Celular , Citoplasma/metabolismo , Progressão da Doença , Genes myc , Linfoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Biossíntese de Proteínas , Proteína Quinase C/fisiologia , Proteína Quinase C beta , Proteína Supressora de Tumor p53/fisiologia
15.
J Biol Chem ; 278(42): 40717-22, 2003 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-12912989

RESUMO

The translational repressor protein eIF4E-binding protein 1 (4E-BP1, also termed PHAS-I) is regulated by phosphorylation through the rapamycin-sensitive mTOR (mammalian target of rapamycin) pathway. Recent studies have identified two regulatory motifs in 4E-BP1, an mTOR-signaling (TOS) motif in the C terminus of 4E-BP1 and an RAIP motif (named after its sequence) in the N terminus. Other recent work has shown that the protein raptor binds to mTOR and 4E-BP1. We show that raptor binds to full-length 4E-BP1 or a C-terminal fragment containing the TOS motif but not to an N-terminal fragment containing the RAIP motif. Mutation of several residues within the TOS motif abrogates binding to raptor, indicating that the TOS motif is required for this interaction. 4E-BP1 undergoes phosphorylation at multiple sites in intact cells. The effects of removal or mutation of the RAIP and TOS motifs differ. The RAIP motif is absolutely required for phosphorylation of sites in the N and C termini of 4E-BP1, whereas the TOS motif primarily affects phosphorylation of Ser-64/65, Thr-69/70, and also the rapamycin-insensitive site Ser-101. Phosphorylation of N-terminal sites that are dependent upon the RAIP motif is sensitive to rapamycin. The RAIP motif thus promotes the mTOR-dependent phosphorylation of multiple sites in 4E-BP1 independently of the 4E-BP1/raptor interaction.


Assuntos
Proteínas de Transporte/metabolismo , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Motivos de Aminoácidos , Animais , Sítios de Ligação , Western Blotting , Proteínas de Ciclo Celular , Linhagem Celular , DNA Complementar/metabolismo , Vetores Genéticos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mutação , Fosforilação , Ligação Proteica , Proteínas Quinases/genética , Estrutura Terciária de Proteína , Ratos , Serina/química , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Transfecção
16.
Biochem J ; 372(Pt 2): 555-66, 2003 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-12611592

RESUMO

In mammalian cells, amino acids affect the phosphorylation state and function of several proteins involved in mRNA translation that are regulated via the rapamycin-sensitive mTOR (mammalian target of rapamycin) pathway. These include ribosomal protein S6 kinase, S6K1, and eukaryotic initiation factor 4E-binding protein, 4E-BP1. Amino acids, especially branched-chain amino acids, such as leucine, promote phosphorylation of 4E-BP1 and S6K1, and permit insulin to further increase their phosphorylation. However, it is not clear whether these effects are exerted by extracellular or intracellular amino acids. Inhibition of protein synthesis is expected to increase the intracellular level of amino acids, whereas inhibiting proteolysis has the opposite effect. We show in the present study that inhibition of protein synthesis by any of several protein synthesis inhibitors tested allows insulin to regulate 4E-BP1 or S6K1 in amino-acid-deprived cells, as does the addition of amino acids to the medium. In particular, insulin activates S6K1 and promotes initiation factor complex assembly in amino-acid-deprived cells treated with protein synthesis inhibitors, but cannot do so in the absence of these compounds. Their effects occur at concentrations commensurate with their inhibition of protein synthesis and are not due to activation of stress-activated kinase cascades. Inhibition of protein breakdown (autophagy) impairs the ability of insulin to regulate 4E-BP1 or S6K1 under such conditions. These and other data presented in the current study are consistent with the idea that it is intracellular amino acid levels that regulate mTOR signalling.


Assuntos
Adenina/análogos & derivados , Aminoácidos/metabolismo , Proteínas de Transporte/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno , Fosfoproteínas/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Adenina/farmacologia , Aminoácidos/química , Animais , Anisomicina/farmacologia , Western Blotting , Células CHO/efeitos dos fármacos , Células CHO/metabolismo , Proteínas de Transporte/genética , Cricetinae , Cicloeximida/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Imunossupressores/farmacologia , Insulina/farmacologia , MAP Quinase Quinase 4 , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Fosfoproteínas/genética , Fosforilação/efeitos dos fármacos , Proteínas Quinases/genética , Inibidores da Síntese de Proteínas/farmacologia , Proteínas Quinases S6 Ribossômicas/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Proteínas Quinases p38 Ativadas por Mitógeno
17.
Biochem J ; 367(Pt 2): 475-81, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12133000

RESUMO

Eukaryotic initiation factor (eIF) 2B is a guanine-nucleotide exchange factor that plays a key role in the regulation of protein synthesis. It is activated by insulin, serum and other agents that stimulate general protein synthesis. The largest (epsilon) subunit of eIF2B is a substrate for glycogen synthase kinase (GSK)-3 in vitro, and phosphorylation by GSK3 inhibits the activity of eIF2B. The site of phosphorylation has previously been identified as Ser(535). GSK3 is inactivated by phosphorylation in response to insulin or serum. In Chinese-hamster ovary cells, insulin and serum bring about the dephosphorylation of Ser(535) in vivo, concomitantly with the phosphorylation of GSK3, and these effects are mediated through signalling via phosphoinositide 3-kinase. We have made use of inhibitors of GSK3 to determine whether GSK3 is responsible for phosphorylation of Ser(535) in vivo and to explore the role of phosphorylation of Ser(535) in the regulation of eIF2B. Treatment of cells with LiCl or with either of two recently developed GSK3 inhibitors, SB-415286 and SB-216763, brought about the dephosphorylation of Ser(535), which strongly indicates that this site is indeed a target for GSK3 in vivo. However, these compounds did not elicit significant activation of eIF2B, indicating, consistent with conclusions from one of our previous studies, that additional inputs are required for the activation of eIF2B. Our results also show that each of the inhibitors used affects overall protein synthesis and have additional effects on translation factors or signalling pathways apparently unrelated to their effects on GSK3, indicating that caution must be exercised when interpreting data obtained using these compounds.


Assuntos
Fator de Iniciação 2B em Eucariotos/metabolismo , Insulina/metabolismo , Serina/metabolismo , Aminofenóis/farmacologia , Animais , Butadienos/farmacologia , Células CHO , Cromonas/farmacologia , Cricetinae , Inibidores Enzimáticos/farmacologia , Fator de Iniciação 2B em Eucariotos/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/metabolismo , Indóis/farmacologia , Insulina/farmacologia , Cloreto de Lítio/farmacologia , Maleimidas/farmacologia , Morfolinas/farmacologia , Nitrilas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/metabolismo , Subunidades Proteicas , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA