Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 220
Filtrar
1.
Bioorg Med Chem Lett ; 30(16): 127300, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32631520

RESUMEN

The transcription factor ΔFosB accumulates in response to chronic insults such as drugs of abuse, L-3,4-dihydroxyphenylalanine (l-DOPA) or stress in specific regions of the brain, triggering long lasting neural and behavioral changes that underlie aspects of drug addiction, dyskinesia, and depression. Thus, small molecule chemical probes are urgently needed to investigate biological functions of ΔFosB. Herein we describe the identification of a novel phenanthridine analogue ZL0220 (27) as an active and promising ΔFosB chemical probe with micromolar inhibitory activities against ΔFosB homodimers and ΔFosB/JunD heterodimers.


Asunto(s)
ADN/efectos de los fármacos , Descubrimiento de Drogas , Fenantridinas/farmacología , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Sitios de Unión/efectos de los fármacos , ADN/química , Relación Dosis-Respuesta a Droga , Humanos , Simulación del Acoplamiento Molecular , Estructura Molecular , Fenantridinas/química , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-jun/química , Relación Estructura-Actividad
2.
Mol Cell Biochem ; 467(1-2): 107-116, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32108279

RESUMEN

Hepatic stellate cells (HSCs) are known to play a key role in the progression of liver fibrosis by producing excessive extracellular matrix (ECM). Matrix metalloproteinases (MMPs) belong to a family of endopeptidases, which have a well-established role in the degradation of ECM. Our study suggests that, besides the degradation of the extracellular matrix, matrix metalloproteinase-8 (MMP-8) has a non-canonical role in activating the quiescent HSCs to myofibroblasts by regulating the expression of Col1A1 and αSMA. We have identified that MMP-8 secreted from macrophages as a response to LPS stimulation activates HSCs via ERK1/2-dependent pathway. In addition to this, we determined that MMP-8 may regulate the homodimerization of c-Jun in LX-2 cells, during the trans-differentiation process from quiescent HSC to activate myofibroblasts. Macrophage-released MMP-8 plays a master role in activating the dormant HSCs to activate myofibroblasts through the Erk-mediated pathway and Jun cellular translocation leading to liver fibrosis. Significance MMP-8 can be used as a therapeutic target against liver fibrosis.


Asunto(s)
Células Estrelladas Hepáticas/citología , Lipopolisacáridos/efectos adversos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 8 de la Matriz/metabolismo , Técnicas de Cultivo de Célula , Transdiferenciación Celular , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Humanos , Miofibroblastos/citología , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-jun/química , Proteína 1 para la Supervivencia de la Neurona Motora/metabolismo , Células THP-1
3.
EBioMedicine ; 42: 97-108, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30824386

RESUMEN

BACKGROUND: The latent HIV-1 reservoir in treated patients primarily consists of resting memory CD4+ T cells. Stimulating the T-cell receptor (TCR), which facilitates transition of resting into effector T cells, is the most effective strategy to purge these latently infected cells. Here we supply evidence that TCR-stimulated effector T cells still frequently harbor latent HIV-1. METHODS: Primary HIV-1 infected cells were used in a latency assay with or without dendritic cells (DCs) and reversion of HIV-1 latency was determined, in the presence or absence of specific pathway inhibitors. FINDINGS: Renewed TCR-stimulation or subsequent activation with latency reversing agents (LRAs) did not overcome latency. However, interaction of infected effector cells with DCs triggered further activation of latent HIV-1. When compared to TCR-stimulation only, CD4+ T cells from aviremic patients receiving TCR + DC-stimulation reversed latency more frequently. Such a "one-two punch" strategy seems ideal for purging the reservoir. We determined that DC contact activates the PI3K-Akt-mTOR pathway in CD4+ T cells. INTERPRETATION: This insight could facilitate the development of a novel class of potent LRAs that purge latent HIV beyond levels reached by T-cell activation.


Asunto(s)
Infecciones por VIH/metabolismo , Infecciones por VIH/virología , VIH-1/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Latencia del Virus , Adulto , Anciano , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , VIH-1/efectos de los fármacos , Humanos , Activación de Linfocitos/inmunología , Masculino , Persona de Mediana Edad , Modelos Biológicos , FN-kappa B/metabolismo , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Latencia del Virus/inmunología
4.
Biochemistry ; 57(42): 6108-6118, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30256622

RESUMEN

Basic leucine zipper (bZIP) proteins reside at the end of cell-signaling cascades and function to modulate transcription of specific gene targets. bZIPs are recognized as important regulators of cellular processes such as cell growth, apoptosis, and cell differentiation. One such validated transcriptional regulator, activator protein-1, is typically comprised of heterodimers of Jun and Fos family members and is key in the progression and development of a number of different diseases. The best described component, cJun, is upregulated in a variety of diseases such as cancer, osteoporosis, and psoriasis. Toward our goal of inhibiting bZIP proteins implicated in disease pathways, we here describe the first use of a novel in silico peptide library screening platform that facilitates the derivation of sequences exhibiting a high affinity for cJun while disfavoring homodimer formation or formation of heterodimers with other closely related Fos sequences. In particular, using Fos as a template, we have computationally screened a peptide library of more than 60 million members and ranked hypothetical on/off target complexes according to predicted stability. This resulted in the identification of a sequence that bound cJun but displayed little homomeric stability or preference for cFos. The computationally selected sequence maintains an interaction stability similar to that of a previous experimentally derived cJun antagonist while providing much improved specificity. Our study provides new insight into the use of tandem in silico screening/ in vitro validation and the ability to create a peptide that is capable of satisfying conflicting design requirements.


Asunto(s)
Simulación por Computador , Leucina Zippers , Biblioteca de Péptidos , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-jun/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-jun/química , Humanos , Proteínas Proto-Oncogénicas c-fos/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo
5.
Future Oncol ; 14(25): 2599-2613, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30073865

RESUMEN

AIM: The airway epithelium of smokers exhibits upregulated SPRR3, an indicator of pathogenic keratinization. The mechanisms underlying this phenomenon require investigation. PATIENTS & METHODS: Human bronchial epithelial (HBE) SPRR3 expression was analyzed by smoking status. Primary HBE cells were exposed to cigarette smoke (CS). SPRR3 expression, SPRR3 promoter activity, AP-1 factor binding and AP-1 factors' effects were analyzed. RESULTS: Current smokers display SPRR3 upregulation relative to never smokers. CS upregulates SPRR3 transcription in an exposure-dependent manner. CS promotes c-Jun and Fra1 binding to the SPRR3-AP-1/TRE site. Wild-type c-Jun and Fra1 upregulate, whereas c-Jun and Fra1, dominant-negative mutants, suppress SPRR3 promoter activity. CONCLUSION: CS induces SPRR3 upregulation in HBE cells by promoting aberrant c-Jun/Fra1 dimerization.


Asunto(s)
Bronquios/metabolismo , Proteínas Ricas en Prolina del Estrato Córneo/genética , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-jun/química , Fumar Tabaco/efectos adversos , Anciano , Anciano de 80 o más Años , Células Cultivadas , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proteína Quinasa 7 Activada por Mitógenos/fisiología , Proteína Quinasa 8 Activada por Mitógenos/fisiología , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/fisiología , Regulación hacia Arriba
6.
Elife ; 72018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29638215

RESUMEN

A key question in human genetics and evolutionary biology is how mutations in different genes combine to alter phenotypes. Efforts to systematically map genetic interactions have mostly made use of gene deletions. However, most genetic variation consists of point mutations of diverse and difficult to predict effects. Here, by developing a new sequencing-based protein interaction assay - deepPCA - we quantified the effects of >120,000 pairs of point mutations on the formation of the AP-1 transcription factor complex between the products of the FOS and JUN proto-oncogenes. Genetic interactions are abundant both in cis (within one protein) and trans (between the two molecules) and consist of two classes - interactions driven by thermodynamics that can be predicted using a three-parameter global model, and structural interactions between proximally located residues. These results reveal how physical interactions generate quantitatively predictable genetic interactions.


Asunto(s)
Modelos Genéticos , Mutación , Mapas de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Algoritmos , Evolución Biológica , Epistasis Genética , Humanos , Conformación Proteica , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/genética , Termodinámica
7.
Nucleic Acids Res ; 45(19): 11425-11436, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-28981703

RESUMEN

The transcription factor, activator protein-1 (AP-1), binds to cognate DNA under redox control; yet, the underlying mechanism has remained enigmatic. A series of crystal structures of the AP-1 FosB/JunD bZIP domains reveal ordered DNA-binding regions in both FosB and JunD even in absence DNA. However, while JunD is competent to bind DNA, the FosB bZIP domain must undergo a large conformational rearrangement that is controlled by a 'redox switch' centered on an inter-molecular disulfide bond. Solution studies confirm that FosB/JunD cannot undergo structural transition and bind DNA when the redox-switch is in the 'OFF' state, and show that the mid-point redox potential of the redox switch affords it sensitivity to cellular redox homeostasis. The molecular and structural studies presented here thus reveal the mechanism underlying redox-regulation of AP-1 Fos/Jun transcription factors and provide structural insight for therapeutic interventions targeting AP-1 proteins.


Asunto(s)
ADN/química , Dominios Proteicos , Proteínas Proto-Oncogénicas c-jun/química , Factor de Transcripción AP-1/química , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Homología de Secuencia de Aminoácido , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
8.
Nucleic Acids Res ; 45(5): 2503-2515, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28158710

RESUMEN

T: Activator protein 1 (AP-1) is a transcription factor that recognizes two versions of a 7-base pair response element, either 5΄- GAG CA-3΄ or 5΄- GAG CA-3΄ (where M = 5-methylcytosine). These two elements share the feature that 5-methylcytosine and thymine both have a methyl group in the same position, 5-carbon of the pyrimidine, so each of them has two methyl groups at nucleotide positions 1 and 5 from the 5΄ end, resulting in four methyl groups symmetrically positioned in duplex DNA. Epstein-Barr Virus Zta is a key transcriptional regulator of the viral lytic cycle that is homologous to AP-1. Zta recognizes several methylated Zta-response elements, including meZRE1 (5΄- GAG C A-3΄) and meZRE2 (5΄- GAG G A-3΄), where a methylated cytosine occupies one of the inner thymine residues corresponding to the AP-1 element, resulting in the four spatially equivalent methyl groups. Here, we study how AP-1 and Zta recognize these methyl groups within their cognate response elements. These methyl groups are in van der Waals contact with a conserved di-alanine in AP-1 dimer (Ala265 and Ala266 in Jun), or with the corresponding Zta residues Ala185 and Ser186 (via its side chain carbon Cß atom). Furthermore, the two ZRE elements differ at base pair 6 (C:G versus G:C), forming a pseudo-symmetric sequence (meZRE1) or an asymmetric sequence (meZRE2). In vitro DNA binding assays suggest that Zta has high affinity for all four sequences examined, whereas AP-1 has considerably reduced affinity for the asymmetric sequence (meZRE2). We ascribe this difference to Zta Ser186 (a unique residue for Zta) whose side chain hydroxyl oxygen atom interacts with the two half sites differently, whereas the corresponding Ala266 of AP-1 Jun protein lacks such flexibility. Our analyses demonstrate a novel mechanism of 5mC/T recognition in a methylation-dependent, spatial and sequence-specific approach by basic leucine-zipper transcriptional factors.


Asunto(s)
Metilación de ADN , Proteínas Proto-Oncogénicas c-jun/química , Elementos de Respuesta , Transactivadores/química , 5-Metilcitosina/química , Emparejamiento Base , Sitios de Unión , ADN/química , ADN/metabolismo , Humanos , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-jun/metabolismo , Timina/química , Transactivadores/metabolismo , Factor de Transcripción AP-1/química , Factor de Transcripción AP-1/metabolismo
9.
J Pept Sci ; 23(7-8): 644-649, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28185384

RESUMEN

Coiled coils, consisting of at least two α-helices, have important roles in the regulation of transcription, cell differentiation, and cell growth. Peptides composed of d-amino acids (d-peptides) have received great attention for their potential in biomedical applications, because they give large diversity for the design of peptidyl drug and are more resistant to proteolytic digestion than l-peptides. However, the interactions between l-peptides/l-protein and d-peptides in the formation of complex are poorly understood. In this study, stereoisomer-specific peptides were constructed corresponding to regions of the basic-leucine-zipper domains of Jun and Fos proteins. basic-leucine-zipper domains consist of an N-terminal basic domain, which is responsible for DNA binding, and a C-terminal domain that enables homodimerization or heterodimerization via formation of a coiled-coil. By combining peptides with different stereochemistries, the d-l heterochiral Jun-Fos heterodimer formation induced DNA binding by the basic domains of Jun-Fos. Our study provides new insight into the interaction between l-peptide and d-peptide enantiomers for developing d-peptide materials and drugs. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/química , Péptidos/química , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-jun/química , Secuencia de Aminoácidos , Dicroismo Circular , Dimerización , Ensayo de Cambio de Movilidad Electroforética , Conformación Proteica
10.
Fish Shellfish Immunol ; 61: 181-186, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28027988

RESUMEN

The transcription factor C-Jun widely exists in vertebrates and invertebrates and plays an important role in various kinds of stimulus response. In this study, PmC-jun gene was first cloned from Penaeus monodon. The full-length cDNA of PmC-jun was 1857 bp in length and included an 879 bp open reading frame (ORF), which encoded 293 amino acids. qRT-PCR analysis results showed that PmC-jun mRNAs were ubiquitously expressed in all the examined tissues. The highest expression level was observed in gill, followed by hepatopancreas. The expression patterns of PmC-jun after Vibrio harveyi and Streptococcus agalactiae injections were studied by qRT-PCR experiment. PmC-jun increased obviously in the gill and hepatopancreas. The expression pattern of PmC-jun in the hepatopancreas was further studied using in situ hybridization (ISH) method. The mRNA expression level of PmC-jun significantly increased in the hepatopancreas after bacterial infection. The expression sites of PmC-jun were almost unchanged. PmC-jun played a regulatory role in pathogen invasion.


Asunto(s)
Proteínas de Artrópodos/genética , Penaeidae/genética , Proteínas Proto-Oncogénicas c-jun/genética , Streptococcus agalactiae/fisiología , Vibrio/fisiología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/metabolismo , Secuencia de Bases , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Inmunidad Innata , Especificidad de Órganos , Penaeidae/inmunología , Penaeidae/metabolismo , Filogenia , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Alineación de Secuencia
11.
Structure ; 24(8): 1301-1310, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27427476

RESUMEN

Focal adhesion kinase (FAK) has emerged as a mediator of mechanotransduction in cardiomyocytes, regulating gene expression during hypertrophic remodeling. However, how FAK signaling is relayed onward to the nucleus is unclear. Here, we show that FAK interacts with and regulates myocyte enhancer factor 2 (MEF2), a master cardiac transcriptional regulator. In cardiomyocytes exposed to biomechanical stimulation, FAK accumulates in the nucleus, binds to and upregulates the transcriptional activity of MEF2 through an interaction with the FAK focal adhesion targeting (FAT) domain. In the crystal structure (2.9 Å resolution), FAT binds to a stably folded groove in the MEF2 dimer, known to interact with regulatory cofactors. FAK cooperates with MEF2 to enhance the expression of Jun in cardiomyocytes, an important component of hypertrophic response to mechanical stress. These findings underscore a connection between the mechanotransduction involving FAK and transcriptional regulation by MEF2, with potential relevance to the pathogenesis of cardiac disease.


Asunto(s)
Quinasa 1 de Adhesión Focal/química , Mecanotransducción Celular , Miocitos Cardíacos/metabolismo , Proteínas Proto-Oncogénicas c-jun/química , Transcripción Genética , Secuencias de Aminoácidos , Animales , Animales Recién Nacidos , Sitios de Unión , Línea Celular , Núcleo Celular/metabolismo , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Quinasa 1 de Adhesión Focal/genética , Quinasa 1 de Adhesión Focal/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Cinética , Factores de Transcripción MEF2/química , Factores de Transcripción MEF2/genética , Factores de Transcripción MEF2/metabolismo , Ratones , Modelos Moleculares , Miocitos Cardíacos/citología , Cultivo Primario de Células , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
PLoS One ; 10(4): e0123070, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25875593

RESUMEN

We collected mobility and interaction maps of c-Fos-eGFP and c-Jun-mRFP1 transcription factors within living cell nuclei. c-Fos dimerizes with c-Jun to form the transcription activator protein-1 (AP-1) which binds to the specific recognition site. To monitor this process, we used fluorescence cross-correlation spectroscopy on a single plane illumination microscope (SPIM-FCCS), which provides diffusion coefficient and protein-protein interaction data in the whole image plane simultaneously, instead of just one point on conventional confocal FCS. We find a strong correlation between diffusional mobility and interaction: regions of strong interaction show slow mobility. Controls containing either an eGFP-mRFP dimer, separately expressing eGFP and mRPF, or c-Fos-eGFP and c-Jun-mRFP1 mutants lacking dimerization and DNA-binding domains, showed no such correlation. These results extend our earlier findings from confocal FCCS to include spatial information.


Asunto(s)
Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Sitios de Unión , Núcleo Celular/metabolismo , ADN/química , ADN/metabolismo , Dimerización , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Mutagénesis , Dominios y Motivos de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Factor de Transcripción AP-1/química , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Proteína Fluorescente Roja
13.
Org Biomol Chem ; 13(19): 5385-90, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-25778494

RESUMEN

We describe a ds-oligonucleotide-peptide conjugate that is able to efficiently dismount preformed DNA complexes of the bZIP regions of oncoproteins c-Fos and c-Jun (AP-1), and therefore might be useful as disrupters of AP-1-mediated gene expression pathways.


Asunto(s)
ADN/metabolismo , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , ADN/química , Ensayo de Cambio de Movilidad Electroforética , Polarización de Fluorescencia , Datos de Secuencia Molecular , Oligonucleótidos , Péptidos/química , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun/química
14.
Fish Shellfish Immunol ; 43(1): 230-40, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25555808

RESUMEN

The nuclear phosphoprotein c-Jun is a member of the AP1 family of transcription activating complex, can be induced by various extracellular stimuli such as virus infection. In this study, the c-Jun gene (Ec-c-Jun) was cloned from orange-spotted grouper, Epinephelus coioides. The full-length Ec-c-Jun cDNA is composed of 2046 bp and encodes a polypeptide of 328 amino acids with 81% identity of zebrafish. Amino acid alignment analysis indicated that Ec-c-Jun contained three conserved domains including a transactivation domain (TAD), a DNA-binding domain (DBD) and leucine zipper domain (LZD). RT-PCR results showed that Ec-c-Jun transcript was most abundant in spleen, kidney, heart and gill. The expression of Ec-c-Jun was up-regulated after challenged with Singapore grouper iridovirus (SGIV). To investigate the roles of Ec-c-Jun during SGIV infection, we constructed its dominant-negative mutant (DN-Ec-c-Jun) by deleting the major TAD that lacks amino acids 3-122. Fluorescence microscopy observation revealed that Ec-c-Jun and DN-Ec-c-Jun were expressed predominantly in the nucleus in transfected cells. Interestingly, the green fluorescence of Ec-c-Jun was congregated and co-localized with virus assembly sites at the late stage of SGIV infection. However, in DN-Ec-c-Jun transfected cells, no virus assembly sites were observed, and the distribution of fluorescence remained unchanged. Moreover, overexpression of DN-Ec-c-Jun in vitro delayed the occurrence of CPE induced by SGIV infection and inhibited the virus gene transcription. In addition, ectopic expression of DN-Ec-c-Jun was able to inhibit SGIV induced c-Jun/AP1 promoter activity in GS cells. Thus, we proposed that c-Jun transcription factor was essential for SGIV replication in vitro. Our results will contribute to understanding the crucial roles of JNK signaling pathway in fish virus infection.


Asunto(s)
Lubina , Infecciones por Virus ADN/veterinaria , Enfermedades de los Peces/genética , Proteínas de Peces/genética , Iridovirus/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Infecciones por Virus ADN/genética , Infecciones por Virus ADN/virología , Enfermedades de los Peces/virología , Proteínas de Peces/química , Proteínas de Peces/metabolismo , Datos de Secuencia Molecular , Filogenia , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo
15.
Int J Mol Med ; 35(4): 1119-25, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25625784

RESUMEN

Endometriosis is a prevalent and complex gynecological disease which affects 10% of women of reproductive age. Certain studies have suggested that a substantial number of microRNAs (miRNAs or miRs) are aberrantly or differentially expressed in the ectopic endometrium. To date, to the best of our knowledge, there is no report available on the role of miR-29 in the endometrium. In this study, we investigated the expression of the miR-29 family in the endometrium samples from women without endometriosis, as well as in paired ectopic and eutopic endometrium samples by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). The results revealed that miR-29c was differentially expressed in the paired eutopic and ectopic endometrium samples. In addition, c-Jun was differentially expressed in the ectopic and eutopic endometrial tissues as determined by western blot analysis. Furthermore, the role of miR-29c in endometrial cell proliferation, invasion and apoptosis was examined in vitro. The results revealed that miR-29c exerted its effects on endometrial cells by suppressing cell proliferation and invasion, as well as promoting cell apoptosis. Furthermore, it was found that c-Jun was a novel target of miR-29c, and c-Jun reversed the effects of miR-29c on the proliferation, invasion and apoptosis of endometrial cells. To the best of our knowledge, this study is the first to identify miR-29c as a suppressor of endometriosis. Taken together, our results suggest that miR-29c exerts its effects on endometrial cell proliferation, apoptosis and invasion by inhibiting the expression of c-Jun. Our data may provide a novel potential therapeutic target for the treatment of endometriosis.


Asunto(s)
Apoptosis/genética , Endometriosis/genética , Endometrio/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas Proto-Oncogénicas c-jun/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular , Regulación hacia Abajo , Femenino , Expresión Génica , Humanos , MicroARNs/química , Proteínas Proto-Oncogénicas c-jun/química , Transfección
16.
Mol Cancer Ther ; 13(12): 3137-51, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25205654

RESUMEN

MicroRNA-200c (miR200c) recently emerged as an important regulator of tumorigenicity and cancer metastasis; however, its role in regulating multidrug resistance (MDR) remains unknown. In the current study, we found that the expression levels of miR200c in recurrent and metastatic colorectal cancers were significantly lower, whereas the JNK2 expression was higher compared with primary tumors. We showed that in MDR colorectal cancer cells, miR200c targeted the 3' untranslated region of the JNK2 gene. Overexpression of miR200c attenuated the levels of p-JNK, p-c-Jun, P-gp, and MMP-2/-9, the downstream factors of the JNK signaling pathway, resulting in increased sensitivity to chemotherapeutic drugs, which was accompanied by heightened apoptosis and decreased cell invasion and migration. Moreover, in an orthotopic MDR colorectal cancer mouse model, we demonstrated that overexpression of miR200c effectively inhibited the tumor growth and metastasis. At last, in the tumor samples from patients with locally advanced colorectal cancer with routine postsurgical chemotherapy, we observed an inverse correlation between the levels of mRNA expression of miR200c and JNK2, ABCB1, and MMP-9, thus predicting patient therapeutic outcomes. In summary, we found that miR200c negatively regulated the expression of JNK2 gene and increased the sensitivity of MDR colorectal cancer cells to chemotherapeutic drugs, via inhibiting the JNK2/p-JNK/p-c-Jun/ABCB1 signaling. Restoration of miR200c expression in MDR colorectal cancer may serve as a promising therapeutic approach in MDR-induced metastasis.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , MicroARNs/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Proteínas Proto-Oncogénicas c-jun/genética , Transducción de Señal , Regiones no Traducidas 3' , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , MicroARNs/química , Proteína Quinasa 9 Activada por Mitógenos/química , Proteína Quinasa 9 Activada por Mitógenos/metabolismo , Metástasis de la Neoplasia , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo , Interferencia de ARN , ARN Mensajero/genética , Ensayos Antitumor por Modelo de Xenoinjerto
17.
PLoS One ; 9(6): e101015, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24968026

RESUMEN

The AP-1 family of transcriptional activators plays pivotal roles in regulating a wide range of biological processes from the immune response to tumorigenesis. Determining the roles of specific AP-1 dimers in cells, however, has remained challenging because common molecular biology techniques are unable to distinguish between the role of, for example, cJun/cJun homodimers versus cJun/cFos heterodimers. Here we used SELEX (systematic evolution of ligands by exponential enrichment) to identify and characterize DNA aptamers that are >100-fold more specific for binding cJun/cJun compared to cJun/cFos, setting the foundation to investigate the biological functions of different AP-1 dimer compositions.


Asunto(s)
Aptámeros de Nucleótidos/genética , Aptámeros de Nucleótidos/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Animales , Aptámeros de Nucleótidos/química , Sitios de Unión , Línea Celular , Genes Reporteros , Interleucina-2/genética , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas , Unión Proteica , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/genética , Técnica SELEX de Producción de Aptámeros , Factor de Transcripción AP-1/metabolismo
18.
J Proteome Res ; 12(12): 5535-47, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24106799

RESUMEN

Conductometric monitoring of protein-protein and protein-sterol interactions is here proved feasible by coupling quartz crystal microbalance with dissipation monitoring (QCM_D) to nucleic acid programmable protein arrays (NAPPA). The conductance curves measured in NAPPA microarrays printed on quartz surface allowed the identification of binding events between the immobilized proteins and the query. NAPPA allows the immobilization on the quartz surface of a wide range of proteins and can be easily adapted to generate innumerous types of biosensors. Indeed multiple proteins on the same quartz crystal have been tested and envisaged proving the possibility of analyzing the same array for several distinct interactions. Two examples of NAPPA-based conductometer applications with clinical relevance are presented herein, the interaction between the transcription factors Jun and ATF2 and the interaction between Cytochrome P540scc and cholesterol.


Asunto(s)
Factor de Transcripción Activador 2/química , Técnicas Biosensibles , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/química , Proteínas Proto-Oncogénicas c-jun/química , Factor de Transcripción Activador 2/genética , Factor de Transcripción Activador 2/metabolismo , Anticuerpos/química , Anticuerpos/metabolismo , Colesterol/química , Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/genética , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Conductometría , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Proteínas Inmovilizadas/química , Proteínas Inmovilizadas/genética , Proteínas Inmovilizadas/metabolismo , Análisis por Matrices de Proteínas , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Tecnicas de Microbalanza del Cristal de Cuarzo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Propiedades de Superficie
19.
Cell Death Dis ; 4: e852, 2013 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-24113186

RESUMEN

Cerebellar granule cell (CGC) apoptosis by trophic/potassium (TK) deprivation is a model of election to study the interplay of pro-apoptotic and pro-survival signaling pathways in neuronal cell death. In this model, the c-Jun N-terminal kinase (JNK) induces pro-apoptotic genes through the c-Jun/activator protein 1 (AP-1) transcription factor. On the other side, a survival pathway initiated by lithium leads to repression of pro-apoptotic c-Jun/AP-1 target genes without interfering with JNK activity. Yet, the mechanism by which lithium inhibits c-Jun activity remains to be elucidated. Here, we used this model system to study the regulation and function of site-specific c-Jun phosphorylation at the S63 and T91/T93 JNK sites in neuronal cell death. We found that TK-deprivation led to c-Jun multiphosphorylation at all three JNK sites. However, immunofluorescence analysis of c-Jun phosphorylation at single cell level revealed that the S63 site was phosphorylated in all c-Jun-expressing cells, whereas the response of T91/T93 phosphorylation was more sensitive, mirroring the switch-like apoptotic response of CGCs. Conversely, lithium prevented T91T93 phosphorylation and cell death without affecting the S63 site, suggesting that T91T93 phosphorylation triggers c-Jun pro-apoptotic activity. Accordingly, a c-Jun mutant lacking the T95 priming site for T91/93 phosphorylation protected CGCs from apoptosis, whereas it was able to induce neurite outgrowth in PC12 cells. Vice versa, a c-Jun mutant bearing aspartate substitution of T95 overwhelmed lithium-mediate protection of CGCs from TK-deprivation, validating that inhibition of T91/T93/T95 phosphorylation underlies the effect of lithium on cell death. Mass spectrometry analysis confirmed multiphosphorylation of c-Jun at T91/T93/T95 in cells. Moreover, JNK phosphorylated recombinant c-Jun at T91/T93 in a T95-dependent manner. On the basis of our results, we propose that T91/T93/T95 multiphosphorylation of c-Jun functions as a sensitivity amplifier of the JNK cascade, setting the threshold for c-Jun pro-apoptotic activity in neuronal cells.


Asunto(s)
Apoptosis , Cerebelo/citología , Neuronas/citología , Neuronas/metabolismo , Fosfotreonina/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Litio/farmacología , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neuritas/efectos de los fármacos , Neuritas/metabolismo , Neuronas/efectos de los fármacos , Células PC12 , Fosfopéptidos/química , Fosfopéptidos/metabolismo , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun/química , Ratas , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
PLoS Pathog ; 9(9): e1003583, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068923

RESUMEN

Lytic gammaherpesvirus (GHV) replication facilitates the establishment of lifelong latent infection, which places the infected host at risk for numerous cancers. As obligate intracellular parasites, GHVs must control and usurp cellular signaling pathways in order to successfully replicate, disseminate to stable latency reservoirs in the host, and prevent immune-mediated clearance. To facilitate a systems-level understanding of phosphorylation-dependent signaling events directed by GHVs during lytic replication, we utilized label-free quantitative mass spectrometry to interrogate the lytic replication cycle of murine gammaherpesvirus-68 (MHV68). Compared to controls, MHV68 infection regulated by 2-fold or greater ca. 86% of identified phosphopeptides - a regulatory scale not previously observed in phosphoproteomic evaluations of discrete signal-inducing stimuli. Network analyses demonstrated that the infection-associated induction or repression of specific cellular proteins globally altered the flow of information through the host phosphoprotein network, yielding major changes to functional protein clusters and ontologically associated proteins. A series of orthogonal bioinformatics analyses revealed that MAPK and CDK-related signaling events were overrepresented in the infection-associated phosphoproteome and identified 155 host proteins, such as the transcription factor c-Jun, as putative downstream targets. Importantly, functional tests of bioinformatics-based predictions confirmed ERK1/2 and CDK1/2 as kinases that facilitate MHV68 replication and also demonstrated the importance of c-Jun. Finally, a transposon-mutant virus screen identified the MHV68 cyclin D ortholog as a viral protein that contributes to the prominent MAPK/CDK signature of the infection-associated phosphoproteome. Together, these analyses enhance an understanding of how GHVs reorganize and usurp intracellular signaling networks to facilitate infection and replication.


Asunto(s)
Gammaherpesvirinae/fisiología , Interacciones Huésped-Patógeno , Modelos Biológicos , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas Virales/metabolismo , Replicación Viral , Células 3T3 , Animales , Cromatografía Líquida de Alta Presión , Biología Computacional , Ciclina D/química , Ciclina D/genética , Ciclina D/metabolismo , Gammaherpesvirinae/genética , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Sistema de Señalización de MAP Quinasas , Ratones , Mutación , Fosfoproteínas/química , Fosfoproteínas/genética , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/metabolismo , Espectrometría de Masas en Tándem , Proteínas Virales/química , Proteínas Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...