Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 161(5): 1058-1073, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-26000482

RESUMEN

Regnase-1 and Roquin are RNA binding proteins essential for degradation of inflammation-related mRNAs and maintenance of immune homeostasis. However, their mechanistic relationship has yet to be clarified. Here, we show that, although Regnase-1 and Roquin regulate an overlapping set of mRNAs via a common stem-loop structure, they function in distinct subcellular locations: ribosome/endoplasmic reticulum and processing-body/stress granules, respectively. Moreover, Regnase-1 specifically cleaves and degrades translationally active mRNAs and requires the helicase activity of UPF1, similar to the decay mechanisms of nonsense mRNAs. In contrast, Roquin controls translationally inactive mRNAs, independent of UPF1. Defects in both Regnase-1 and Roquin lead to large increases in their target mRNAs, although Regnase-1 tends to control the early phase of inflammation when mRNAs are more actively translated. Our findings reveal that differential regulation of mRNAs by Regnase-1 and Roquin depends on their translation status and enables elaborate control of inflammation.


Asunto(s)
Inflamación/metabolismo , Estabilidad del ARN , ARN Mensajero/metabolismo , Ribonucleasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Secuencia de Bases , Codón de Terminación , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Conformación de Ácido Nucleico , Polirribosomas/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/química , Proteínas Ribosómicas/metabolismo , Transactivadores/metabolismo
2.
J Cell Sci ; 134(2)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33277379

RESUMEN

Protein kinase C (PKC) signaling is a highly conserved signaling module that plays a central role in a myriad of physiological processes, ranging from cell proliferation to cell death, via various signaling pathways, including MAPK signaling. Stress granules (SGs) are non-membranous cytoplasmic foci that aggregate in cells exposed to environmental stresses. Here, we explored the role of SGs in PKC/MAPK signaling activation in fission yeast. High-heat stress (HHS) induced Pmk1 MAPK activation and Pck2 translocation from the cell tips into poly(A)-binding protein (Pabp)-positive SGs. Pck2 dispersal from the cell tips required Pck2 kinase activity, and constitutively active Pck2 exhibited increased translocation to SGs. Importantly, Pmk1 deletion impaired Pck2 recruitment to SGs, indicating that MAPK activation stimulates Pck2 SG translocation. Consistently, HHS-induced SGs delayed Pck2 relocalization at the cell tips, thereby blocking subsequent Pmk1 reactivation after recovery from HHS. HHS partitioned Pck2 into the Pabp-positive SG-containing fraction, which resulted in reduced Pck2 abundance and kinase activity in the soluble fraction. Taken together, these results indicate that MAPK-dependent Pck2 SG recruitment serves as a feedback mechanism to intercept PKC/MAPK activation induced by HHS, which might underlie PKC-related diseases.


Asunto(s)
Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Gránulos Citoplasmáticos/metabolismo , Retroalimentación , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Estrés Fisiológico
3.
Biol Pharm Bull ; 46(2): 158-162, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36724943

RESUMEN

Translation initiation is the rate-limiting step of protein synthesis and is the main target of translation regulation. RNA-binding proteins (RBPs) are key mediators of the spatiotemporal control of translation and are critical for cell proliferation, development, and differentiation. We have previously shown that HuD, one of the neuronal RBPs, enhances cap-dependent translation through the direct interaction with eukaryotic initiation factor 4A (eIF4A) and poly(A) tail using a HeLa-derived in vitro translation system. We have also found that translation stimulation of HuD is essential for HuD-induced neurite outgrowth in PC12 cells. However, it remains unclear how HuD is involved in the regulation of translation initiation. Here, we report that HuD binds to eukaryotic initiation factor 3 (eIF3) via the eIF3b subunit, which belongs to the functional core of mammalian eIF3. eIF3 plays an essential role in recruiting the 40S ribosomal subunit onto mRNA in translation initiation. We hypothesize that the interaction between HuD and eIF3 stabilizes the translation initiation complex and increases translation efficiency. We also showed that the linker region of HuD is required for the interaction with eIF3b. Moreover, we found that eIF3b-binding region of HuD is conserved in all Hu proteins (HuB, HuC, HuD, and HuR). These data might also help to explain how Hu proteins stimulate translation in a cap- and poly(A)-dependent way.


Asunto(s)
Factor 3 de Iniciación Eucariótica , Factores Eucarióticos de Iniciación , Animales , Humanos , Ratas , Factor 3 de Iniciación Eucariótica/genética , Factor 3 de Iniciación Eucariótica/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Neuronas/metabolismo , Factor 3 Procariótico de Iniciación/genética , Factor 3 Procariótico de Iniciación/metabolismo , Unión Proteica , Biosíntesis de Proteínas , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Células HeLa
4.
RNA Biol ; 19(1): 234-246, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35129087

RESUMEN

CCR4-NOT complex-mediated mRNA deadenylation serves critical functions in multiple biological processes, yet how this activity is regulated is not fully understood. Here, we show that osmotic stress induces MAPKAPK-2 (MK2)-mediated phosphorylation of CNOT2. Programmed cell death is greatly enhanced by osmotic stress in CNOT2-depleted cells, indicating that CNOT2 is responsible for stress resistance of cells. Although wild-type (WT) and non-phosphorylatable CNOT2 mutants reverse this sensitivity, a phosphomimetic form of CNOT2, in which serine at the phosphorylation site is replaced with glutamate, does not have this function. We also show that mRNAs have elongated poly(A) tails in CNOT2-depleted cells and that introduction of CNOT2 WT or a non-phosphorylatable mutant, but not phosphomimetic CNOT2, renders their poly(A) tail lengths comparable to those in control HeLa cells. Consistent with this, the CCR4-NOT complex containing phosphomimetic CNOT2 exhibits less deadenylase activity than that containing CNOT2 WT. These data suggest that CCR4-NOT complex deadenylase activity is regulated by post-translational modification, yielding dynamic control of mRNA deadenylation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Complejos Multiproteicos/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores CCR4/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Activación Enzimática , Humanos , Presión Osmótica , Fosforilación , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Estrés Fisiológico/genética
5.
Mol Cell ; 56(1): 79-89, 2014 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-25280105

RESUMEN

In animals, key functions of microRNA-induced silencing complex (miRISC) are translational repression and deadenylation followed by mRNA decay. While miRISC represses translation initiation, it is poorly understood how miRISC exerts this function. Here we assessed the effect of miRISC on synergistic recruitment of translation initiation factors to target mRNAs by using direct biochemical assays. We show that miRISC promotes eIF4AI and eIF4AII release from target mRNAs prior to dissociation of eIF4E and eIF4G in a deadenylation-independent manner. Strikingly, miRISC-induced release of eIF4AI and eIF4AII from target mRNAs and miRISC-induced inhibition of cap-dependent translation can both be counteracted by the RNA-binding protein HuD via a direct interaction of HuD with eIF4A. Furthermore, the pharmacological eIF4A inhibitor silvestrol, which locks eIF4A on mRNAs, conferred resistance to miRNA-mediated translational repression. In summary, we propose that both eIF4AI and eIF4AII are functionally important targets in miRISC-mediated translation control.


Asunto(s)
Factor 4A Eucariótico de Iniciación/metabolismo , MicroARNs/fisiología , Modelos Genéticos , ARN Mensajero/metabolismo , Factor 4A Eucariótico de Iniciación/antagonistas & inhibidores , Factor 4A Eucariótico de Iniciación/genética , Células HEK293 , Humanos , Complejo Silenciador Inducido por ARN/fisiología , Iniciación de la Transcripción Genética , Triterpenos/farmacología
6.
Proc Natl Acad Sci U S A ; 116(35): 17450-17459, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31399545

RESUMEN

Although KRAS and TP53 mutations are major drivers of pancreatic ductal adenocarcinoma (PDAC), the incurable nature of this cancer still remains largely elusive. ARF6 and its effector AMAP1 are often overexpressed in different cancers and regulate the intracellular dynamics of integrins and E-cadherin, thus promoting tumor invasion and metastasis when ARF6 is activated. Here we show that the ARF6-AMAP1 pathway is a major target by which KRAS and TP53 cooperatively promote malignancy. KRAS was identified to promote eIF4A-dependent ARF6 mRNA translation, which contains a quadruplex structure at its 5'-untranslated region, by inducing TEAD3 and ETV4 to suppress PDCD4; and also eIF4E-dependent AMAP1 mRNA translation, which contains a 5'-terminal oligopyrimidine-like sequence, via up-regulating mTORC1. TP53 facilitated ARF6 activation by platelet-derived growth factor (PDGF), via its known function to promote the expression of PDGF receptor ß (PDGFRß) and enzymes of the mevalonate pathway (MVP). The ARF6-AMAP1 pathway was moreover essential for PDGF-driven recycling of PD-L1, in which KRAS, TP53, eIF4A/4E-dependent translation, mTOR, and MVP were all integral. We moreover demonstrated that the mouse PDAC model KPC cells, bearing KRAS/TP53 mutations, express ARF6 and AMAP1 at high levels and that the ARF6-based pathway is closely associated with immune evasion of KPC cells. Expression of ARF6 pathway components statistically correlated with poor patient outcomes. Thus, the cooperation among eIF4A/4E-dependent mRNA translation and MVP has emerged as a link by which pancreatic driver mutations may promote tumor cell motility, PD-L1 dynamics, and immune evasion, via empowering the ARF6-based pathway and its activation by external ligands.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Antígeno B7-H1/metabolismo , Evasión Inmune/genética , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Factor 6 de Ribosilación del ADP , Sitios de Unión , Biomarcadores de Tumor , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Modelos Moleculares , Mutación , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Unión Proteica , ARN Mensajero/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
7.
EMBO Rep ; 20(11): e48220, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31482640

RESUMEN

Codon bias has been implicated as one of the major factors contributing to mRNA stability in several model organisms. However, the molecular mechanisms of codon bias on mRNA stability remain unclear in humans. Here, we show that human cells possess a mechanism to modulate RNA stability through a unique codon bias. Bioinformatics analysis showed that codons could be clustered into two distinct groups-codons with G or C at the third base position (GC3) and codons with either A or T at the third base position (AT3): the former stabilizing while the latter destabilizing mRNA. Quantification of codon bias showed that increased GC3-content entails proportionately higher GC-content. Through bioinformatics, ribosome profiling, and in vitro analysis, we show that decoupling the effects of codon bias reveals two modes of mRNA regulation, one GC3- and one GC-content dependent. Employing an immunoprecipitation-based strategy, we identify ILF2 and ILF3 as RNA-binding proteins that differentially regulate global mRNA abundances based on codon bias. Our results demonstrate that codon bias is a two-pronged system that governs mRNA abundance.


Asunto(s)
Uso de Codones , Codón , ARN Mensajero/genética , Biología Computacional/métodos , Guanilato Ciclasa/genética , Humanos , Proteína del Factor Nuclear 45/metabolismo , Estabilidad del ARN , Ribosomas/genética , Ribosomas/metabolismo , Transcripción Genética
8.
Mol Microbiol ; 104(3): 428-448, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28142187

RESUMEN

RNA-binding proteins (RBPs) play important roles in the posttranscriptional regulation of gene expression, including mRNA stability, transport and translation. Fission yeast rnc1+ encodes a K Homology (KH)-type RBP, which binds and stabilizes the Pmp1 MAPK phosphatase mRNA thereby suppressing the Cl- hypersensitivity of calcineurin deletion and MAPK signaling mutants. Here, we analyzed the spatial regulation of Rnc1 and discovered a putative nuclear export signal (NES)Rnc1 , which dictates the cytoplasmic localization of Rnc1 in a Crm1-independent manner. Notably, mutations in the NESRnc1 altered nucleocytoplasmic distribution of Rnc1 and abolished its function to suppress calcineurin deletion, although the Rnc1 NES mutant maintains the ability to bind Pmp1 mRNA. Intriguingly, the Rnc1 NES mutant destabilized Pmp1 mRNA, suggesting the functional importance of the Rnc1 cytoplasmic localization. Mutation in Rae1, but not Mex67 deletion or overproduction, induced Rnc1 accumulation in the nucleus, suggesting that Rnc1 is exported from the nucleus to the cytoplasm via the mRNA export pathway involving Rae1. Importantly, mutations in the Rnc1 KH-domains abolished the mRNA-binding ability and induced nuclear localization, suggesting that Rnc1 may be exported from the nucleus together with its target mRNAs. Collectively, the functional Rae1-dependent mRNA export system may influence the cytoplasmic localization and function of Rnc1.


Asunto(s)
Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/metabolismo , Desoxirribonucleasas/metabolismo , Carioferinas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Schizosaccharomyces/metabolismo , ARNt Metiltransferasas/metabolismo , Citoplasma/metabolismo , Desoxirribonucleasas/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Dominios Proteicos , Estabilidad del ARN , ARN Mensajero/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Schizosaccharomyces/genética , Análisis Espacial , ARNt Metiltransferasas/genética , Proteína Exportina 1
9.
Biochem Biophys Res Commun ; 497(2): 713-718, 2018 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-29462623

RESUMEN

Staphylococcal superantigen like 5 (SSL5) is an exotoxin produced by S. aureus and has a strong inhibitory effect on MMP-9 enzymatic activity. However, the mechanism of inhibition remains unclear. We sought to identify the responsible regions of SSL5 for the interaction with MMP-9 by comparing a series of domain swap and deletion mutants of SSL5. Binding analyses revealed that SSL5 had two regions for binding to MMP-9 catalytic domain, ß1-3 region (25SKELKNVTGY RYSKGGKHYL IFDKNRKFTR VQIFGK60) in N-terminal half and α4ß9 region (138KELDFKLRQY LIQNFDLYKK FPKDSKIKVI MKD170) in C-terminal half. The collagen binding domain and zinc-chelating histidine residues of MMP-9 were not essential for the specific binding to SSL5. The domain swap mutants of SSL5 that conserved ß1-3 but not α4ß9 region inhibited the gelatinolysis by MMP-9, and the mutant of SSL7 that substituted ß1-3 region to that of SSL5 acquired the binding and inhibitory activity. Furthermore, the polypeptide that harbored ß1-3 region of SSL5 inhibited gelatinolysis by MMP-9. Taken together, SSL5 inhibits the MMP9 activity through binding to the catalytic domain, and the ß1-3 region is responsible for the inhibition of proteolytic activity of MMP-9.


Asunto(s)
Proteínas Bacterianas/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Infecciones Estafilocócicas/metabolismo , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Línea Celular , Humanos , Metaloproteinasa 9 de la Matriz/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/química
10.
Mol Cell ; 36(6): 1007-17, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-20064466

RESUMEN

The RNA-binding protein HuD promotes neuronal differentiation by an unknown mechanism. Here we identify an enhancer function of HuD in translation. Translation stimulation by HuD requires both a 3' poly(A) tail and a 5' m(7)G cap structure. We also show that HuD directly interacts with eIF4A. This interaction and the poly(A)-binding activity of HuD are critical for its translational enhancer function because HuD-eIF4A- and HuD-poly(A)-binding mutants fail to stimulate translation. We show that translation of HCV IRES mRNA, which is eIF4A independent, is not stimulated by HuD. We also find that the eIF4A and poly(A)-binding activities of HuD are not only important for stimulating translation but also are essential for HuD-induced neurite outgrowth in PC12 cells. This example of cap-dependent translational regulation might explain at least in part how HuD triggers the induction of neuronal differentiation.


Asunto(s)
Proteínas ELAV/metabolismo , Factor 4A Eucariótico de Iniciación/metabolismo , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas ELAV/genética , Proteína 4 Similar a ELAV , Elementos de Facilitación Genéticos , Factor 4A Eucariótico de Iniciación/genética , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/fisiología , Células PC12 , Caperuzas de ARN/química , Caperuzas de ARN/genética , ARN Mensajero/genética , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
11.
RNA Biol ; 12(9): 922-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26274611

RESUMEN

MicroRNAs (miRNAs) are evolutionarily conserved small noncoding RNAs found in most plants and animals. The miRNA pathway regulates posttranscriptional gene expression through the deadenylation and translation repression of target mRNAs. Recent studies revealed that the early step of translation initiation is the target of "pure" translation repression by the miRNA pathway. Moreover, particularly in animals, the miRNA pathway is required for neuronal development, differentiation, and plasticity. In addition, some functions of miRNAs are regulated by RNA-binding proteins (RBPs) in neuronal cells. This review summarizes new insights about the molecular mechanisms of pure translation repression by miRNA pathway and the communication between the miRNA pathway and RBPs in neuronal local translation.


Asunto(s)
Regulación de la Expresión Génica , MicroARNs/genética , MicroARNs/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Regulación hacia Abajo , Silenciador del Gen , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Humanos , Neuronas/metabolismo , Complejo Silenciador Inducido por ARN/metabolismo
12.
Proc Natl Acad Sci U S A ; 109(4): 1104-9, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22232654

RESUMEN

MicroRNA (miRNA) is a class of small noncoding RNA approximately 22 nt in length. Animal miRNA silences complementary mRNAs via translational inhibition, deadenylation, and mRNA degradation. However, the underlying molecular mechanisms remain unclear. A key question is whether these three outputs are independently induced by miRNA through distinct mechanisms or sequentially induced within a single molecular pathway. Here, we successfully dissected these intricate outputs of miRNA-mediated repression using zebrafish embryos as a model system. Our results indicate that translational inhibition and deadenylation are independent outputs mediated by distinct domains of TNRC6A, which is an effector protein in the miRNA pathway. Translational inhibition by TNRC6A is divided into two mechanisms: PAM2 motif-mediated interference of poly(A)-binding protein (PABP), and inhibition of 5' cap- and poly(A) tail-independent step(s) by a previously undescribed P-GL motif. Consistent with these observations, we show that, in zebrafish embryos, miRNA inhibits translation of the target mRNA in a deadenylation- and PABP-independent manner at early time points. These results indicate that miRNA exerts multiple posttranscriptional outputs via physically and functionally independent mechanisms and that direct translational inhibition is central to miRNA-mediated repression.


Asunto(s)
Autoantígenos/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Silenciador del Gen/fisiología , MicroARNs/fisiología , Modelos Biológicos , Estabilidad del ARN/fisiología , Proteínas de Pez Cebra/genética , Pez Cebra/embriología , Secuencias de Aminoácidos/genética , Animales , Autoantígenos/metabolismo , Western Blotting , Proteínas Fluorescentes Verdes/metabolismo , Hibridación in Situ , Luciferasas , MicroARNs/metabolismo , Mutagénesis Sitio-Dirigida , Reacción en Cadena en Tiempo Real de la Polimerasa , Pez Cebra/metabolismo , Proteínas de Pez Cebra/metabolismo
13.
J Biol Chem ; 288(30): 21569-80, 2013 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-23754290

RESUMEN

The staphylococcal superantigen-like protein (SSL) family is composed of 14 exoproteins sharing structural similarity with superantigens but no superantigenic activity. Target proteins of four SSLs have been identified to be involved in host immune responses. However, the counterparts of other SSLs have been functionally uncharacterized. In this study, we have identified porcine plasma prothrombin as SSL10-binding protein by affinity purification using SSL10-conjugated Sepharose. The resin recovered the prodomain of prothrombin (fragment 1 + 2) as well as factor Xa in pull-down analysis. The equilibrium dissociation constant between SSL10 and prothrombin was 1.36 × 10(-7) M in surface plasmon resonance analysis. On the other hand, the resin failed to recover γ-carboxyglutamic acid (Gla) domain-less coagulation factors and prothrombin from warfarin-treated mice, suggesting that the Gla domain of the coagulation factors is essential for the interaction. SSL10 prolonged plasma clotting induced by the addition of Ca(2+) and factor Xa. SSL10 did not affect the protease activity of thrombin but inhibited the generation of thrombin activity in recalcified plasma. S. aureus produces coagulase that non-enzymatically activates prothrombin. SSL10 attenuated clotting induced by coagulase, but the inhibitory effect was weaker than that on physiological clotting, and SSL10 did not inhibit protease activity of staphylothrombin, the complex of prothrombin with coagulase. These results indicate that SSL10 inhibits blood coagulation by interfering with activation of coagulation cascade via binding to the Gla domain of coagulation factor but not by directly inhibiting thrombin activity. This is the first finding that the bacterial protein inhibits blood coagulation via targeting the Gla domain of coagulation factors.


Asunto(s)
Ácido 1-Carboxiglutámico/inmunología , Proteínas Bacterianas/inmunología , Factor Xa/inmunología , Protrombina/inmunología , Staphylococcus aureus/inmunología , Superantígenos/inmunología , Ácido 1-Carboxiglutámico/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/metabolismo , Sitios de Unión/inmunología , Unión Competitiva/inmunología , Coagulación Sanguínea/inmunología , Calcio/inmunología , Calcio/metabolismo , Coagulasa/inmunología , Coagulasa/metabolismo , Electroforesis en Gel de Poliacrilamida , Factor Xa/metabolismo , Humanos , Sueros Inmunes/inmunología , Sueros Inmunes/metabolismo , Ratones , Datos de Secuencia Molecular , Unión Proteica/inmunología , Protrombina/metabolismo , Staphylococcus aureus/metabolismo , Superantígenos/metabolismo , Resonancia por Plasmón de Superficie , Porcinos , Trombina/inmunología , Trombina/metabolismo
14.
Anal Biochem ; 446: 37-43, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24141076

RESUMEN

Neurotensin receptor type-1 (NTSR1) is a member of the G-protein-coupled receptor (GPCR) family. The natural ligand of NTSR1 is neurotensin (NT), a neuromodulator of the central nervous system. Because NT is also involved in many oncogenic actions, the signaling mediator NTSR1 is a significant molecular target in medicinal and therapeutic fields. In the current study, we constructed a fluorescence-based microbial yeast biosensor that can monitor the activation of human NTSR1 signaling responding to its agonist. To increase the sensitivity of the biosensor, a yeast strain with the green fluorescent protein (GFP) reporter gene was genetically engineered to enhance binding with human NTSR1 expressed on the membrane. Following previous reports, the 5 carboxy-terminal amino acid residues of the guanine nucleotide binding protein α-subunit (Gα) in yeast Gpa1p were substituted with the equivalent human Gα(q) sequences (Gpa1/Gα(q) transplant). After optimizing the assay conditions, the Gα-engineered yeast demonstrated significantly improved sensing for NTSR1 signaling. Because detection using a GFP fluorescence reporter considerably simplifies the measurement procedure, this microbial fluorescence sensor holds promise for use in the diagnosis of NTSR1-associated diseases and the development of agonists.


Asunto(s)
Técnicas Biosensibles/métodos , Subunidades alfa de la Proteína de Unión al GTP/genética , Ingeniería Genética , Receptores de Neurotensina/metabolismo , Saccharomyces cerevisiae/citología , Humanos , Concentración de Iones de Hidrógeno , Espectrometría de Fluorescencia , Factores de Tiempo
15.
Nucleic Acids Res ; 40(5): 1944-53, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22075994

RESUMEN

The RNA binding protein HuD plays essential roles in neuronal development and plasticity. We have previously shown that HuD stimulates translation. Key for this enhancer function is the linker region and the poly(A) binding domain of HuD that are also critical for its function in neurite outgrowth. Here, we further explored the underlying molecular interactions and found that HuD but not the ubiquitously expressed HuR interacts directly with active Akt1. We identify that the linker region of HuD is required for this interaction. We also show by using chimeric mutants of HuD and HuR, which contain the reciprocal linker between RNA-binding domain 2 (RBD2) and RBD3, respectively, and by overexpressing a dominant negative mutant of Akt1 that the HuD-Akt1 interaction is functionally important, as it is required for the induction of neurite outgrowth in PC12 cells. These results suggest the model whereby RNA-bound HuD functions as an adapter to recruit Akt1 to trigger neurite outgrowth. These data might also help to explain how HuD enhances translation of mRNAs that encode proteins involved in neuronal development.


Asunto(s)
Proteínas ELAV/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Proteínas ELAV/química , Células HeLa , Humanos , Neuritas/enzimología , Células PC12 , Dominios y Motivos de Interacción de Proteínas , Ratas
16.
Biochimie ; 221: 20-26, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38244852

RESUMEN

The RNA-binding protein HuD/ELAVL4 is essential for neuronal development and synaptic plasticity by governing various post-transcriptional processes of target mRNAs, including stability, translation, and localization. We previously showed that the linker region and poly(A)-binding domain of HuD play a pivotal role in promoting translation and inducing neurite outgrowth. In addition, we found that HuD interacts exclusively with the active form of Akt1, through the linker region. Although this interaction is essential for neurite outgrowth, HuD is not a substrate for Akt1, raising questions about the dynamics between HuD-mediated translational stimulation and its association with active Akt1. Here, we demonstrate that active Akt1 interacts with the cap-binding complex via HuD. We identify key amino acids in linker region of HuD responsible for Akt1 interaction, leading to the generation of two point-mutated HuD variants: one that is incapable of binding to Akt1 and another that can interact with Akt1 regardless of its phosphorylation status. In vitro translation assays using these mutants reveal that HuD-mediated translation stimulation is independent of its binding to Akt1. In addition, it is evident that the interaction between HuD and active Akt1 is essential for HuD-induced neurite outgrowth, whereas a HuD mutant capable of binding to any form of Akt1 leads to aberrant neurite development. Collectively, our results revisit the understanding of the HuD-Akt1 interaction in translation and suggest that this interaction contributes to HuD-mediated neurite outgrowth via a unique molecular mechanism distinct from translation regulation.


Asunto(s)
Proteína 4 Similar a ELAV , Neuronas , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína 4 Similar a ELAV/metabolismo , Proteína 4 Similar a ELAV/genética , Humanos , Animales , Neuronas/metabolismo , Neuronas/citología , Diferenciación Celular , Células HEK293 , Unión Proteica , Fosforilación , Ratones , Neurogénesis , Ratas , Neuritas/metabolismo
17.
J Biol Chem ; 287(15): 11924-33, 2012 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-22334708

RESUMEN

We have previously shown that DPF2 (requiem/REQ) functions as a linker protein between the SWI/SNF complex and RelB/p52 NF-κB heterodimer and plays important roles in NF-κB transactivation via its noncanonical pathway. Using sensitive 293FT reporter cell clones that had integrated a SWI/SNF-dependent NF-κB reporter gene, we find in this study that the overexpression of DPF1, DPF2, DPF3a, DPF3b, and PHF10 significantly potentiates the transactivating activity of typical NF-κB dimers. Knockdown analysis using 293FT reporter cells that endogenously express these five proteins at low levels clearly showed that DPF3a and DPF3b, which are produced from the DPF3 gene by alternative splicing, are the most critical for the RelA/p50 NF-κB heterodimer transactivation induced by TNF-α stimulation. Our data further show that this transactivation requires the SWI/SNF complex. DPF3a and DPF3b are additionally shown to interact directly with RelA, p50, and several subunits of the SWI/SNF complex in vitro and to be co-immunoprecipitated with RelA/p50 and the SWI/SNF complex from the nuclear fractions of cells treated with TNF-α. In ChIP experiments, we further found that endogenous DPF3a/b and the SWI/SNF complex are continuously present on HIV-1 LTR, whereas the kinetics of RelA/p50 recruitment after TNF-α treatment correlate well with the viral transcriptional activation levels. Additionally, re-ChIP experiments showed DPF3a/b and the SWI/SNF complex associate with RelA on the endogenous IL-6 promoter after TNF-α treatment. In conclusion, our present data indicate that by linking RelA/p50 to the SWI/SNF complex, DPF3a/b induces the transactivation of NF-κB target gene promoters in relatively inactive chromatin contexts.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Regulación de la Expresión Génica , Subunidad p50 de NF-kappa B/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/fisiología , Activación Transcripcional , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Genes Reporteros , Humanos , Cinética , Luciferasas/biosíntesis , Luciferasas/genética , Complejos Multiproteicos/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Estructura Terciaria de Proteína , Transporte de Proteínas , Elementos de Respuesta , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/fisiología
18.
Biochem Biophys Res Commun ; 437(1): 12-7, 2013 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-23770370

RESUMEN

Negative regulator of differentiation 1 (Nrd1) is known as a negative regulator of sexual differentiation in fission yeast. Recently, it has been revealed that Nrd1 also regulates cytokinesis, in which physical separation of the cell is achieved by a contractile ring comprising many proteins including actin and myosin. Cdc4, a myosin II light chain, is known to be required for cytokinesis. Nrd1 binds and stabilizes Cdc4 mRNA, and thereby suppressing the cytokinesis defects of the cdc4 mutants. Interestingly, Pmk1 MAPK phosphorylates Nrd1, resulting in markedly reduced RNA binding activity. Furthermore, Nrd1 localizes to stress granules in response to various stresses, and Pmk1 phosphorylation enhances the localization. Nrd1 consists of four RRM domains, although the mechanism by which Pmk1 regulates the RNA binding activity of Nrd1 is unknown. In an effort to delineate the relationship between Nrd1 structure and function, we prepared each RNA binding domain of Nrd1 and examined RNA binding to chemically synthesized oligo RNA using NMR. The structure of the second RRM domain of Nrd1 was determined and the RNA binding site on the second RRM domain was mapped by NMR. A plausible mechanism pertaining to the regulation of RNA binding activity by phosphorylation is also discussed.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , ARN de Hongos/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteínas/química , Ribonucleoproteínas/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Sitios de Unión , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Unión Proteica , Estructura Terciaria de Proteína , Soluciones , Homología Estructural de Proteína , Relación Estructura-Actividad
19.
J Nanobiotechnology ; 11: 19, 2013 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-23800313

RESUMEN

BACKGROUND: Small interfering RNA (siRNA) has attracted attention in the field of nucleic acid medicine as a RNA interference (RNAi) application that leads to gene silencing due to specific messenger RNA (mRNA) destruction. However, since siRNA is unstable in blood and unable to cross the cell membrane, encapsulation of siRNA into a carrier is required. RESULTS: In this study, we used a carrier that combined ZHER2-displaying bio-nanocapsule (derived from hepatitis B virus surface antigen) and liposomes in a complex in order to investigate the feasibility of effective and target-cell-specific RNAi applications. As a result, by observing RNAi only in HER2-expressing breast cancer cells, using our proposed methodology, we successfully demonstrated target-cell-specific delivery and effective function expression of siRNA. CONCLUSIONS: These findings show that, in the field of nucleic acid medicine, ZHER2-BNC/LP can be a useful carrier for siRNA delivery, and could also become a useful tool for gene silencing and to accomplish protein knock-down.


Asunto(s)
Técnicas de Transferencia de Gen , Liposomas/química , Nanocápsulas/química , Neoplasias/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Electroforesis en Gel de Poliacrilamida , Fluorescencia , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Luz , Tamaño de la Partícula , Receptor ErbB-2/metabolismo , Reproducibilidad de los Resultados , Dispersión de Radiación , Electricidad Estática
20.
Nucleic Acids Res ; 39(19): 8314-28, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21745818

RESUMEN

The translation factor eIF5 is an important partner of eIF2, directly modulating its function in several critical steps. First, eIF5 binds eIF2/GTP/Met-tRNA(i)(Met) ternary complex (TC), promoting its recruitment to 40S ribosomal subunits. Secondly, its GTPase activating function promotes eIF2 dissociation for ribosomal subunit joining. Finally, eIF5 GDP dissociation inhibition (GDI) activity can antagonize eIF2 reactivation by competing with the eIF2 guanine exchange factor (GEF), eIF2B. The C-terminal domain (CTD) of eIF5, a W2-type HEAT domain, mediates its interaction with eIF2. Here, we characterize a related human protein containing MA3- and W2-type HEAT domains, previously termed BZW2 and renamed here as eIF5-mimic protein 1 (5MP1). Human 5MP1 interacts with eIF2 and eIF3 and inhibits general and gene-specific translation in mammalian systems. We further test whether 5MP1 is a mimic or competitor of the GEF catalytic subunit eIF2Bε or eIF5, using yeast as a model. Our results suggest that 5MP1 interacts with yeast eIF2 and promotes TC formation, but inhibits TC binding to the ribosome. Moreover, 5MP1 is not a GEF but a weak GDI for yeast eIF2. We propose that 5MP1 is a partial mimic and competitor of eIF5, interfering with the key steps by which eIF5 regulates eIF2 function.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor 2 Eucariótico de Iniciación/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Regulación de la Expresión Génica , Biosíntesis de Proteínas , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Animales , Línea Celular , Factor 2 Eucariótico de Iniciación/análisis , Factor 2B Eucariótico de Iniciación/metabolismo , Factor 3 de Iniciación Eucariótica/metabolismo , Guanosina Difosfato/metabolismo , Células HeLa , Humanos , Ratones , Imitación Molecular , Factores de Iniciación de Péptidos/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Factor 5A Eucariótico de Iniciación de Traducción
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA