Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 93(18)2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31243130

RESUMEN

Previously, the cyclophilin inhibitors cyclosporine (CsA) and alisporivir (ALV) were shown to inhibit the replication of diverse RNA viruses, including arteriviruses and coronaviruses, which both belong to the order Nidovirales In this study, we aimed to identify arterivirus proteins involved in the mode of action of cyclophilin inhibitors and to investigate how these compounds inhibit arterivirus RNA synthesis in the infected cell. Repeated passaging of the arterivirus prototype equine arteritis virus (EAV) in the presence of CsA revealed that reduced drug sensitivity is associated with the emergence of adaptive mutations in nonstructural protein 5 (nsp5), one of the transmembrane subunits of the arterivirus replicase polyprotein. Introduction of singular nsp5 mutations (nsp5 Q21R, Y113H, or A134V) led to an ∼2-fold decrease in sensitivity to CsA treatment, whereas combinations of mutations further increased EAV's CsA resistance. The detailed experimental characterization of engineered EAV mutants harboring CsA resistance mutations implicated nsp5 in arterivirus RNA synthesis. Particularly, in an in vitro assay, EAV RNA synthesis was far less sensitive to CsA treatment when nsp5 contained the adaptive mutations mentioned above. Interestingly, for increased sensitivity to the closely related drug ALV, CsA-resistant nsp5 mutants required the incorporation of an additional adaptive mutation, which resided in nsp2 (H114R), another transmembrane subunit of the arterivirus replicase. Our study provides the first evidence for the involvement of nsp2 and nsp5 in the mechanism underlying the inhibition of arterivirus replication by cyclophilin inhibitors.IMPORTANCE Currently, no approved treatments are available to combat infections with nidoviruses, a group of positive-stranded RNA viruses, including important zoonotic and veterinary pathogens. Previously, the cyclophilin inhibitors cyclosporine (CsA) and alisporivir (ALV) were shown to inhibit the replication of diverse nidoviruses (both arteriviruses and coronaviruses), and they may thus represent a class of pan-nidovirus inhibitors. In this study, using the arterivirus prototype equine arteritis virus, we have established that resistance to CsA and ALV treatment is associated with adaptive mutations in two transmembrane subunits of the viral replication machinery, nonstructural proteins 2 and 5. This is the first evidence for the involvement of specific replicase subunits of arteriviruses in the mechanism underlying the inhibition of their replication by cyclophilin inhibitors. Understanding this mechanism of action is of major importance to guide future drug design, both for nidoviruses and for other RNA viruses inhibited by these compounds.


Asunto(s)
Equartevirus/genética , ARN Polimerasa Dependiente del ARN/genética , Proteínas no Estructurales Virales/metabolismo , Arterivirus/genética , Línea Celular , Ciclofilinas/metabolismo , Ciclosporina/antagonistas & inhibidores , Equartevirus/metabolismo , Células HEK293 , Humanos , Mutación , Nidovirales/genética , Nidovirales/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas no Estructurales Virales/genética , Replicación Viral
2.
Biochemistry ; 55(7): 1168-77, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26836009

RESUMEN

We examined the impact of two clinically approved anti-herpes drugs, acyclovir and Forscarnet (phosphonoformate), on the exonuclease activity of the herpes simplex virus-1 DNA polymerase, UL30. Acyclovir triphosphate and Foscarnet, along with the closely related phosphonoacetic acid, did not affect exonuclease activity on single-stranded DNA. Furthermore, blocking the polymerase active site due to either binding of Foscarnet or phosphonoacetic acid to the E-DNA complex or polymerization of acyclovir onto the DNA also had a minimal effect on exonuclease activity. The inability of the exonuclease to excise acyclovir from the primer 3'-terminus results from the altered sugar structure directly impeding phosphodiester bond hydrolysis as opposed to inhibiting binding, unwinding of the DNA by the exonuclease, or transfer of the DNA from the polymerase to the exonuclease. Removing the 3'-hydroxyl or the 2'-carbon from the nucleotide at the 3'-terminus of the primer strongly inhibited exonuclease activity, although addition of a 2'-hydroxyl did not affect exonuclease activity. The biological consequences of these results are twofold. First, the ability of acyclovir and Foscarnet to block dNTP polymerization without impacting exonuclease activity raises the possibility that their effects on herpes replication may involve both direct inhibition of dNTP polymerization and exonuclease-mediated destruction of herpes DNA. Second, the ability of the exonuclease to rapidly remove a ribonucleotide at the primer 3'-terminus in combination with the polymerase not efficiently adding dNTPs onto this primer provides a novel mechanism by which the herpes replication machinery can prevent incorporation of ribonucleotides into newly synthesized DNA.


Asunto(s)
Aciclovir/farmacología , Antivirales/farmacología , Exodesoxirribonucleasas/antagonistas & inhibidores , Foscarnet/farmacología , Herpesvirus Humano 1/enzimología , Modelos Moleculares , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Proteínas Virales/antagonistas & inhibidores , Aciclovir/química , Aciclovir/metabolismo , Antivirales/química , Antivirales/metabolismo , Dominio Catalítico , ADN de Cadena Simple/química , ADN de Cadena Simple/metabolismo , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Exodesoxirribonucleasas/química , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Foscarnet/química , Foscarnet/metabolismo , Herpesvirus Humano 1/efectos de los fármacos , Hidrólisis/efectos de los fármacos , Cinética , Estructura Molecular , Inhibidores de la Síntesis del Ácido Nucleico/química , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ribonucleótidos/química , Ribonucleótidos/metabolismo , Especificidad por Sustrato , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/metabolismo
3.
Appl Microbiol Biotechnol ; 100(21): 9201-9215, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27448399

RESUMEN

Lactic acid bacteria (LAB) have many applications in food and industrial fermentations. Prophage induction and generation of new virulent phages is a risk for the dairy industry. We identified three complete prophages (PLE1, PLE2, and PLE3) in the genome of the well-studied probiotic strain Lactobacillus casei BL23. All of them have mosaic architectures with homologous sequences to Streptococcus, Lactococcus, Lactobacillus, and Listeria phages or strains. Using a combination of quantitative real-time PCR, genomics, and proteomics, we showed that PLE2 and PLE3 can be induced-but with different kinetics-in the presence of mitomycin C, although PLE1 remains as a prophage. A structural analysis of the distal tail (Dit) and tail associated lysin (Tal) baseplate proteins of these prophages and other L. casei/paracasei phages and prophages provides evidence that carbohydrate-binding modules (CBM) located within these "evolved" proteins may replace receptor binding proteins (RBPs) present in other well-studied LAB phages. The detailed study of prophage induction in this prototype strain in combination with characterization of the proteins involved in host recognition will facilitate the design of new strategies for avoiding phage propagation in the dairy industry.


Asunto(s)
Lacticaseibacillus casei/genética , Lacticaseibacillus casei/virología , Profagos/genética , Profagos/fisiología , Activación Viral , Microbiología de Alimentos , Mitomicina/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Proteínas de la Cola de los Virus/genética
4.
J Am Chem Soc ; 137(14): 4728-34, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25786104

RESUMEN

Human DNA polymerase η (hPol η) contributes to anticancer drug resistance by catalyzing the replicative bypass of DNA adducts formed by the widely used chemotherapeutic agent cis-diamminedichloroplatinum (cisplatin). A chemical basis for overcoming bypass-associated resistance requires greater knowledge of how small molecules influence the hPol η-catalyzed bypass of DNA adducts. In this study, we demonstrated how synthetic nucleoside triphosphates act as hPol η substrates and characterized their influence on hPol η-mediated DNA synthesis over unmodified and platinated DNA. The single nucleotide incorporation efficiency of the altered nucleotides varied by more than 10-fold and the higher incorporation rates appeared to be attributable to the presence of an additional hydrogen bond between incoming dNTP and templating base. Finally, full-length DNA synthesis in the presence of increasing concentrations of synthetic nucleotides reduced the amount of DNA product independent of the template, representing the first example of hPol η inhibition in the presence of a platinated DNA template.


Asunto(s)
Aductos de ADN/biosíntesis , ADN Polimerasa Dirigida por ADN/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/química , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Nucleótidos/química , Nucleótidos/farmacología , Secuencia de Bases , Línea Celular Tumoral , Cisplatino/química , Cisplatino/metabolismo , Aductos de ADN/química , Aductos de ADN/genética , Aductos de ADN/metabolismo , ADN Polimerasa Dirigida por ADN/química , Relación Dosis-Respuesta a Droga , Humanos , Enlace de Hidrógeno , Cinética , Modelos Moleculares , Conformación de Ácido Nucleico , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Nucleótidos/metabolismo , Conformación Proteica
5.
Bioorg Med Chem Lett ; 25(2): 210-5, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25515558

RESUMEN

A prodrug approach was developed to address the low oral bioavailability of a poorly soluble (<0.1µg/mL in pH 6.8 buffer) but highly permeable thumb pocket 1 HCV NS5B polymerase inhibitor. Bioconversion rates of structurally diverse prodrug derivatives were evaluated in a panel of in vitro assays using microsomes, from either liver or intestinal tissues, simulated intestinal fluids, simulated gastric fluids or plasma. In vivo bioconversion of promising candidates was evaluated following oral administration to rats. The most successful strategy involved modification of the parent drug carboxylic acid moiety to glycolic amide esters which improved solubility in lipid-based self-emulsifying drug delivery systems (SEDDS). Crystalline prodrug analog 36 (mp 161°C) showed good solubility in individual SEDDS components (up to 80mg/mL) compared to parent 2 (<3mg/mL; mp 267°C) and cross-species bioconversions which correlated with in vitro stability in liver microsomes.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Emulsiones/administración & dosificación , Inhibidores de la Síntesis del Ácido Nucleico/administración & dosificación , Profármacos/administración & dosificación , Proteínas no Estructurales Virales/antagonistas & inhibidores , Administración Oral , Animales , Emulsiones/química , Emulsiones/metabolismo , Microsomas/efectos de los fármacos , Microsomas/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/química , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Profármacos/química , Profármacos/metabolismo , Ratas , Solubilidad , Proteínas no Estructurales Virales/metabolismo
6.
J Bacteriol ; 196(15): 2842-50, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24858186

RESUMEN

We previously reported that the presence of dideoxythymidine (ddT) in the growth medium selectively inhibits the ability of bacteriophage T7 to infect Escherichia coli by inhibiting phage DNA synthese (N. Q. Tran, L. F. Rezende, U. Qimron, C. C. Richardson, and S. Tabor, Proc. Natl. Acad. Sci. U. S. A. 105:9373-9378, 2008, doi:10.1073/pnas.0804164105). In the presence of T7 gene 1.7 protein, ddT is taken up into the E. coli cell and converted to ddTTP. ddTTP is incorporated into DNA as ddTMP by the T7 DNA polymerase, resulting in chain termination. We have identified the pathway by which exogenous ddT is converted to ddTTP. The pathway consists of ddT transport by host nucleoside permeases and phosphorylation to ddTMP by the host thymidine kinase. T7 gene 1.7 protein phosphorylates ddTMP and ddTDP, resulting in ddTTP. A 74-residue peptide of the gene 1.7 protein confers ddT sensitivity to the same extent as the 196-residue wild-type gene 1.7 protein. We also show that cleavage of thymidine to thymine and deoxyribose-1-phosphate by the host thymidine phosphorylase greatly increases the sensitivity of phage T7 to ddT. Finally, a mutation in T7 DNA polymerase that leads to discrimination against the incorporation of ddTMP eliminates ddT sensitivity.


Asunto(s)
Bacteriófago T7/genética , Didesoxinucleótidos/farmacología , Escherichia coli/enzimología , Inhibidores de la Síntesis del Ácido Nucleico , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Nucleótidos de Timina/farmacología , Bacteriófago T7/efectos de los fármacos , Bacteriófago T7/enzimología , Bacteriófago T7/crecimiento & desarrollo , ADN Viral/biosíntesis , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Didesoxinucleótidos/metabolismo , Escherichia coli/virología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Técnicas de Inactivación de Genes , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Fosforilación , Pirimidina Fosforilasas/genética , Pirimidina Fosforilasas/metabolismo , Eliminación de Secuencia , Timidina/metabolismo , Timidina Quinasa/genética , Timidina Quinasa/metabolismo , Nucleótidos de Timina/metabolismo , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética , Proteínas Virales/metabolismo
7.
Biochemistry (Mosc) ; 78(10): 1137-45, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24237148

RESUMEN

Bivalent metal cations are key components in the reaction of DNA synthesis. They are necessary for all DNA polymerases, being involved as cofactors in catalytic mechanisms of nucleotide polymerization. It is also known that in the presence of Mn2+ the accuracy of DNA synthesis is considerably decreased. The findings of this work show that Cd2+ and Zn2+ selectively inhibit the Mn2+-induced error-prone DNA polymerase activity in extracts of cells from human and mouse tissues. Moreover, these cations in low concentrations also can efficiently inhibit the activity of homogeneous preparations of DNA polymerase iota (Pol ι), which is mainly responsible for the Mn2+-induced error-prone DNA polymerase activity in cell extracts. Using a primary culture of granular cells from postnatal rat cerebellum, we show that low concentrations of Cd2+ significantly increase cell survival in the presence of toxic Mn2+ doses. Thus, we have shown that in some cases low concentrations of Cd2+ can display a positive influence on cells, whereas it is widely acknowledged that this metal is not a necessary microelement and is toxic for organisms.


Asunto(s)
Cadmio/farmacología , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , ADN/biosíntesis , Manganeso/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Zinc/farmacología , Animales , Biocatálisis/efectos de los fármacos , Encéfalo/enzimología , Cadmio/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Hígado/enzimología , Manganeso/metabolismo , Melanoma/enzimología , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Ratas , Ratas Wistar , Relación Estructura-Actividad , Neoplasias de la Úvea/enzimología , Zinc/metabolismo , ADN Polimerasa iota
8.
Traffic ; 11(8): 1067-78, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20525016

RESUMEN

The molecular mechanisms underlying cytoskeleton-dependent Golgi positioning are poorly understood. In mammalian cells, the Golgi apparatus is localized near the juxtanuclear centrosome via dynein-mediated motility along microtubules. Previous studies implicate Cdc42 in regulating dynein-dependent motility. Here we show that reduced expression of the Cdc42-specific GTPase-activating protein, ARHGAP21, inhibits the ability of dispersed Golgi membranes to reposition at the centrosome following nocodazole treatment and washout. Cdc42 regulation of Golgi positioning appears to involve ARF1 and a binding interaction with the vesicle-coat protein coatomer. We tested whether Cdc42 directly affects motility, as opposed to the formation of a trafficking intermediate, using a Golgi capture and motility assay in permeabilized cells. Disrupting Cdc42 activation or the coatomer/Cdc42 binding interaction stimulated Golgi motility. The coatomer/Cdc42-sensitive motility was blocked by the addition of an inhibitory dynein antibody. Together, our results reveal that dynein and microtubule-dependent Golgi positioning is regulated by ARF1-, coatomer-, and ARHGAP21-dependent Cdc42 signaling.


Asunto(s)
Citoesqueleto/metabolismo , Aparato de Golgi/metabolismo , Microtúbulos/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Animales , Brefeldino A/metabolismo , Chlorocebus aethiops , Proteína Coatómero/genética , Proteína Coatómero/metabolismo , Citocalasina D/metabolismo , Citoesqueleto/efectos de los fármacos , Dineínas/metabolismo , Endocitosis/fisiología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Nocodazol/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Inhibidores de la Síntesis de la Proteína/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Moduladores de Tubulina/farmacología , Células Vero , Proteína de Unión al GTP cdc42/genética , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
9.
EMBO J ; 27(22): 3011-23, 2008 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-18923417

RESUMEN

The Smc5/6 holocomplex executes key functions in genome maintenance that include ensuring the faithful segregation of chromosomes at mitosis and facilitating critical DNA repair pathways. Smc5/6 is essential for viability and therefore, dissecting its chromosome segregation and DNA repair roles has been challenging. We have identified distinct epigenetic and post-translational modifications that delineate roles for fission yeast Smc5/6 in centromere function, versus replication fork-associated DNA repair. We monitored Smc5/6 subnuclear and genomic localization in response to different replicative stresses, using fluorescence microscopy and chromatin immunoprecipitation (ChIP)-on-chip methods. Following hydroxyurea treatment, and during an unperturbed S phase, Smc5/6 is transiently enriched at the heterochromatic outer repeats of centromeres in an H3-K9 methylation-dependent manner. In contrast, methyl methanesulphonate treatment induces the accumulation of Smc5/6 at subtelomeres, in an Nse2 SUMO ligase-dependent, but H3-K9 methylation-independent manner. Finally, we determine that Smc5/6 loads at all genomic tDNAs, a phenomenon that requires intact consensus TFIIIC-binding sites in the tDNAs.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Heterocromatina/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Telómero/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Proteínas Cromosómicas no Histona/genética , Replicación del ADN , ADN de Hongos/química , ADN de Hongos/metabolismo , Hidroxiurea/metabolismo , Metilmetanosulfonato/metabolismo , Mutágenos/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Schizosaccharomyces/citología , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética
10.
EMBO J ; 26(24): 5061-70, 2007 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-18007598

RESUMEN

Obligatory homologous recombination (HR) is required for chiasma formation and chromosome segregation in meiosis I. Meiotic HR is initiated by DNA double-strand breaks (DSBs), generated by Spo11, a homologue of the archaebacterial topoisomerase subunit Top6A. In Saccharomyces cerevisiae, Rad50, Mre11 and Com1/Sae2 are essential to process an intermediate of the cleavage reaction consisting of Spo11 covalently linked to the 5' termini of DNA. While Rad50 and Mre11 also confer genome stability to vegetative cells and are well conserved in evolution, Com1/Sae2 was believed to be fungal-specific. Here, we identify COM1/SAE2 homologues in all eukaryotic kingdoms. Arabidopsis thaliana Com1/Sae2 mutants are sterile, accumulate AtSPO11-1 during meiotic prophase and fail to form AtRAd51 foci despite the presence of unrepaired DSBs. Furthermore, DNA fragmentation in AtCom1 is suppressed by eliminating AtSPO11-1. In addition, AtCOM1 is specifically required for mitomycin C resistance. Interestingly, we identified CtIP, an essential protein interacting with the DNA repair machinery, as the mammalian homologue of Com1/Sae2, with important implications for the molecular role of CtIP.


Asunto(s)
Proteínas de Arabidopsis/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Genes de Plantas , Meiosis/fisiología , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Animales , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Daño del ADN , Fragmentación del ADN , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas , Endonucleasas , Humanos , Mitomicina/metabolismo , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Fenotipo , Recombinación Genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia
11.
Nat Med ; 10(3): 310-5, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14770178

RESUMEN

The TAT protein transduction domain (PTD) has been used to deliver a wide variety of biologically active cargo for the treatment of multiple preclinical disease models, including cancer and stroke. However, the mechanism of transduction remains unknown. Because of the TAT PTD's strong cell-surface binding, early assumptions regarding cellular uptake suggested a direct penetration mechanism across the lipid bilayer by a temperature- and energy-independent process. Here we show, using a transducible TAT-Cre recombinase reporter assay on live cells, that after an initial ionic cell-surface interaction, TAT-fusion proteins are rapidly internalized by lipid raft-dependent macropinocytosis. Transduction was independent of interleukin-2 receptor/raft-, caveolar- and clathrin-mediated endocytosis and phagocytosis. Using this information, we developed a transducible, pH-sensitive, fusogenic dTAT-HA2 peptide that markedly enhanced TAT-Cre escape from macropinosomes. Taken together, these observations provide a scientific basis for the development of new, biologically active, transducible therapeutic molecules.


Asunto(s)
Productos del Gen tat/metabolismo , Microdominios de Membrana/metabolismo , Péptidos/metabolismo , Pinocitosis/fisiología , Proteínas Virales de Fusión/metabolismo , Amilorida/metabolismo , Animales , Transporte Biológico/fisiología , Caveolina 1 , Caveolinas/metabolismo , Línea Celular , Clatrina/metabolismo , Citocalasina D/metabolismo , Endosomas/metabolismo , Productos del Gen tat/genética , Humanos , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Péptidos/genética , Linfocitos T/fisiología , Transducción Genética , Proteínas Virales de Fusión/genética
12.
Proc Natl Acad Sci U S A ; 105(48): 18730-4, 2008 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19028869

RESUMEN

The Saccharomyces cerevisiae Mec1-Ddc2 checkpoint kinase complex (the ortholog to human ATR-ATRIP) is an essential regulator of genomic integrity. The S. cerevisiae BRCT repeat protein Dpb11 functions in the initiation of both DNA replication and cell cycle checkpoints. Here, we report a genetic and physical interaction between Dpb11 and Mec1-Ddc2. A C-terminal domain of Dpb11 is sufficient to associate with Mec1-Ddc2 and strongly stimulates the kinase activity of Mec1 in a Ddc2-dependent manner. Furthermore, Mec1 phosphorylates Dpb11 and thereby amplifies the stimulating effect of Dpb11 on Mec1-Ddc2 kinase activity. Thus, Dpb11 is a functional ortholog of human TopBP1, and the Mec1/ATR activation mechanism is conserved from yeast to humans.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Fosfoproteínas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas de Ciclo Celular/genética , Daño del ADN , Reparación del ADN , Activación Enzimática , Humanos , Hidroxiurea/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
13.
BMC Biol ; 7: 35, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19558638

RESUMEN

BACKGROUND: The p53 tumor suppressor and its related protein, p73, share a homologous DNA binding domain, and mouse genetics studies have suggested that they have overlapping as well as distinct biological functions. Both p53 and p73 are activated by genotoxic stress to regulate an array of cellular responses. Previous studies have suggested that p53 and p73 independently activate the cellular apoptotic program in response to cytotoxic drugs. The goal of this study was to compare the promoter-binding activity of p53 and p73 at steady state and after genotoxic stress induced by hydroxyurea. RESULTS: We employed chromatin immunoprecipitation, the NimbleGen promoter arrays and a model-based algorithm for promoter arrays to identify promoter sequences enriched in anti-p53 or anti-p73 immunoprecipitates, either before or after treatment with hydroxyurea, which increased the expression of both p53 and p73 in the human colon cancer cell line HCT116-3(6). We calculated a model-based algorithm for promoter array score for each promoter and found a significant correlation between the promoter occupancy profiles of p53 and p73. We also found that after hydroxyurea treatment, the p53-bound promoters were still bound by p73, but p73 became associated with additional promoters that that did not bind p53. In particular, we showed that hydroxyurea induces the binding of p73 but not p53 to the promoter of MLH3, which encodes a mismatch repair protein, and causes an up-regulation of the MLH3 mRNA. CONCLUSION: These results suggest that hydroxyurea exerts differential effects on the promoter-binding functions of p53 and p73 and illustrate the power of model-based algorithm for promoter array in the analyses of promoter occupancy profiles of highly homologous transcription factors.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Hidroxiurea/toxicidad , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/toxicidad , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Algoritmos , Proteínas Portadoras/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Células HCT116 , Humanos , Hidroxiurea/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Proteínas MutL , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Terminación de Péptidos/genética , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
14.
Biochim Biophys Acta ; 1781(5): 239-44, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18374665

RESUMEN

The peroxisome proliferator-activated receptor alpha (PPARalpha) belongs to the nuclear receptor family and plays a central role in the regulation of lipid metabolism, glucose homeostasis and inflammatory processes. In addition to its ligand-induced activation, PPARalpha is regulated by phosphorylation via ERK-MAPK, PKA and PKC. In this study we examined the effect of p38-MAPK on PPARalpha transcriptional activity. In COS-7 cells, anisomycin, a p38 activator, induced a dose-dependent phosphorylation of PPARalpha and a 50% inhibition of its transcriptional activity. In H4IIE hepatoma cells, anisomycin-induced p38 phosphorylation decreased both endogenous and PPARalpha ligand-enhanced L-CPTI and ACO gene expression. Interestingly, PPARalpha/p38 interaction required the molecular adapter ZIP/p62. Reducing ZIP/p62 expression by siRNA, partially reversed the inhibitory effect of anisomycin on L-CPTI gene expression. In conclusion, we showed that p38 activation induced PPARalpha phosphorylation and inhibition of its transcriptional activity through a trimeric interaction between p38-MAPK, ZIP/p62 and PPARalpha.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , PPAR alfa/metabolismo , Transcripción Genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anisomicina/metabolismo , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Choque Térmico/genética , Sistema de Señalización de MAP Quinasas/fisiología , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , PPAR alfa/genética , Proteína Quinasa C/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Sequestosoma-1 , Proteínas Quinasas p38 Activadas por Mitógenos/genética
15.
J Cell Biol ; 160(4): 505-16, 2003 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-12578909

RESUMEN

The spliceosomal small nuclear RNAs (snRNAs) are distributed throughout the nucleoplasm and concentrated in nuclear inclusions termed Cajal bodies (CBs). A role for CBs in the metabolism of snRNPs has been proposed but is not well understood. The SART3/p110 protein interacts transiently with the U6 and U4/U6 snRNPs and promotes the reassembly of U4/U6 snRNPs after splicing in vitro. Here we report that SART3/p110 is enriched in CBs but not in gems or residual CBs lacking coilin. The U6 snRNP Sm-like (LSm) proteins, also involved in U4/U6 snRNP assembly, were localized to CBs as well. The levels of SART3/p110 and LSm proteins in CBs were reduced upon treatment with the transcription inhibitor alpha-amanitin, suggesting that CB localization reflects active processes dependent on transcription/splicing. The NH2-terminal HAT domain of SART3/p110 was necessary and sufficient for specific protein targeting to CBs. Overexpression of truncation mutants containing the HAT domain had dominant negative effects on U6 snRNP localization to CBs, indicating that endogenous SART3/p110 plays a role in targeting the U6 snRNP to CBs. We propose that U4 and U6 snRNPs accumulate in CBs for the purpose of assembly into U4/U6 snRNPs by SART3/p110.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Cuerpos Enrollados/metabolismo , Proteínas de Unión al ARN/metabolismo , Ribonucleoproteína Nuclear Pequeña U4-U6/metabolismo , Amanitinas/metabolismo , Animales , Antígenos de Neoplasias/genética , Núcleo Celular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Células HeLa , Humanos , Inmunohistoquímica , Ratones , Señales de Localización Nuclear , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética
16.
J Cell Biol ; 165(2): 191-202, 2004 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-15117966

RESUMEN

Many of the protein factors that play a role in nuclear export of mRNAs have been identified, but still little is known about how mRNAs are transported through the cell nucleus and which nuclear compartments are involved in mRNA transport. Using fluorescent 2'O-methyl oligoribonucleotide probes, we investigated the mobility of poly(A)+ RNA in the nucleoplasm and in nuclear speckles of U2OS cells. Quantitative analysis of diffusion using photobleaching techniques revealed that the majority of poly(A)+ RNA move throughout the nucleus, including in and out of speckles (also called SC-35 domains), which are enriched for splicing factors. Interestingly, in the presence of the transcription inhibitor 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole, the association of poly(A)+ RNA with speckles remained dynamic. Our results show that RNA movement is energy dependent and that the proportion of nuclear poly(A)+ RNA that resides in speckles is a dynamic population that transiently interacts with speckles independent of the transcriptional status of the cell. Rather than the poly(A)+ RNA within speckles serving a stable structural role, our findings support the suggestion of a more active role of these regions in nuclear RNA metabolism and/or transport.


Asunto(s)
Núcleo Celular/metabolismo , Colorantes Fluorescentes/metabolismo , ARN Mensajero/metabolismo , Transcripción Genética , Transporte Activo de Núcleo Celular/fisiología , Línea Celular , Línea Celular Tumoral , Desoxiadenosinas/metabolismo , Diclororribofuranosil Benzoimidazol/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Humanos , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Oligorribonucleótidos/química , Oligorribonucleótidos/metabolismo , Proteína II de Unión a Poli(A)/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Rodaminas/química , Rodaminas/metabolismo , Factores de Empalme Serina-Arginina
17.
J Cell Biol ; 159(4): 637-48, 2002 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-12438416

RESUMEN

In mammalian cells, the Golgi apparatus undergoes extensive fragmentation during apoptosis. p115 is a key vesicle tethering protein required for maintaining the structural organization of the Golgi apparatus. Here, we demonstrate that p115 was cleaved during apoptosis by caspases 3 and 8. Compared with control cells expressing native p115, those expressing a cleavage-resistant form of p115 delayed Golgi fragmentation during apoptosis. Expression of cDNAs encoding full-length or an NH2-terminal caspase cleavage fragment of p115 had no effect on Golgi morphology. In contrast, expression of the COOH-terminal caspase cleavage product of p115 itself caused Golgi fragmentation. Furthermore, this fragment translocated to the nucleus and its expression was sufficient to induce apoptosis. Most significantly, in vivo expression of the COOH-terminal fragment in the presence of caspase inhibitors, or upon coexpression with a cleavage-resistant mutant of p115, showed that p115 degradation plays a key role in amplifying the apoptotic response independently of Golgi fragmentation.


Asunto(s)
Apoptosis/fisiología , Proteínas Portadoras/metabolismo , Caspasas/metabolismo , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas de Transporte Vesicular , Clorometilcetonas de Aminoácidos/metabolismo , Animales , Autoantígenos , Proteínas Portadoras/genética , Inhibidores de Caspasas , Línea Celular , Tamaño de la Célula , Inhibidores Enzimáticos/metabolismo , Etopósido/metabolismo , Colorantes Fluorescentes/metabolismo , Proteínas de la Matriz de Golgi , Humanos , Proteínas de la Membrana/genética , Mutagénesis Sitio-Dirigida , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Fosforilación , Ratas , Estaurosporina/metabolismo
18.
J Cell Biol ; 163(5): 1045-55, 2003 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-14657236

RESUMEN

The espin actin-bundling proteins, which are the target of the jerker deafness mutation, caused a dramatic, concentration-dependent lengthening of LLC-PK1-CL4 cell microvilli and their parallel actin bundles. Espin level was also positively correlated with stereocilium length in hair cells. Villin, but not fascin or fimbrin, also produced noticeable lengthening. The espin COOH-terminal peptide, which contains the actin-bundling module, was necessary and sufficient for lengthening. Lengthening was blocked by 100 nM cytochalasin D. Espin cross-links slowed actin depolymerization in vitro less than twofold. Elimination of an actin monomer-binding WASP homology 2 domain and a profilin-binding proline-rich domain from espin did not decrease lengthening, but made it possible to demonstrate that actin incorporation was restricted to the microvillar tip and that bundles continued to undergo actin treadmilling at approximately 1.5 s-1 during and after lengthening. Thus, through relatively subtle effects on actin polymerization/depolymerization reactions in a treadmilling parallel actin bundle, espin cross-links cause pronounced barbed-end elongation and, thereby, make a longer bundle without joining shorter modules.


Asunto(s)
Actinas/metabolismo , Proteínas Contráctiles , Proteínas de Microfilamentos/metabolismo , Microvellosidades/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Citocalasina D/metabolismo , Recuperación de Fluorescencia tras Fotoblanqueo , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/genética , Microvellosidades/ultraestructura , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Profilinas , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Porcinos
19.
J Cell Biol ; 164(6): 887-97, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15024034

RESUMEN

We have previously shown that the seemingly static paracrystalline actin core of hair cell stereocilia undergoes continuous turnover. Here, we used the same approach of transfecting hair cells with actin-green fluorescent protein (GFP) and espin-GFP to characterize the turnover process. Actin and espin are incorporated at the paracrystal tip and flow rearwards at the same rate. The flux rates (approximately 0.002-0.04 actin subunits s(-1)) were proportional to the stereocilia length so that the entire staircase stereocilia bundle was turned over synchronously. Cytochalasin D caused stereocilia to shorten at rates matching paracrystal turnover. Myosins VI and VIIa were localized alongside the actin paracrystal, whereas myosin XVa was observed at the tips at levels proportional to stereocilia lengths. Electron microscopy analysis of the abnormally short stereocilia in the shaker 2 mice did not show the characteristic tip density. We argue that actin renewal in the paracrystal follows a treadmill mechanism, which, together with the myosins, dynamically shapes the functional architecture of the stereocilia bundle.


Asunto(s)
Actinas/metabolismo , Cilios/metabolismo , Células Ciliadas Auditivas/citología , Miosinas/metabolismo , Isoformas de Proteínas/metabolismo , Regeneración/fisiología , Actinas/genética , Animales , Transporte Biológico/fisiología , Células Cultivadas , Cilios/ultraestructura , Citocalasina D/metabolismo , Citoesqueleto/metabolismo , Células Ciliadas Auditivas/metabolismo , Inmunohistoquímica , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Proteínas Motoras Moleculares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Órgano Espiral/citología , Subunidades de Proteína/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
20.
Acta Med Okayama ; 63(2): 79-85, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19404339

RESUMEN

ADAMTS1 (a disintegrin and metalloproteinase with thrombospondin motifs 1) is an inflammatory-induced gene. We have previously reported that ADAMTS1 was strongly but transiently expressed in the infarcted heart. In this study, we investigated whether a 3'-untranslated region (UTR) affects the mRNA stability of this gene. When stimulated with tissue necrosis factor (TNF)-alpha, the expression level of ADAMTS1 mRNA rapidly increased, but the induction of ADAMTS1 mRNA peaked at 6h after stimulation, after which the expression levels of ADAMTS1 mRNA decreased. The 3'-UTR ADAMTS1 mRNA contains multiple adenine and uridine-rich elements, suggesting that the 3'-UTR may regulate gene stability. The addition of actinomycin D, an RNA synthesis inhibitor, demonstrated the decay of induced ADAMTS1 mRNA by TNF-alpha. Furthermore, a region containing multiple AUUUA motifs within the ADAMTS1 3'-UTR destabilized transfected Enhanced Green Fluorescence Protein (EGFP) mRNA expression. These results demonstrated that the ADAMTS1 3'-UTR may regulate the expression of ADAMTS1 mRNA.


Asunto(s)
Regiones no Traducidas 3'/genética , Proteínas ADAM , Estabilidad del ARN/genética , ARN Mensajero , Proteínas ADAM/genética , Proteínas ADAM/metabolismo , Proteína ADAMTS1 , Animales , Secuencia de Bases , Células COS , Chlorocebus aethiops , Dactinomicina/metabolismo , Humanos , Ratones , Datos de Secuencia Molecular , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA