Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cell ; 162(2): 314-327, 2015 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-26144317

RESUMEN

The large (L) proteins of non-segmented, negative-strand RNA viruses, a group that includes Ebola and rabies viruses, catalyze RNA-dependent RNA polymerization with viral ribonucleoprotein as template, a non-canonical sequence of capping and methylation reactions, and polyadenylation of viral messages. We have determined by electron cryomicroscopy the structure of the vesicular stomatitis virus (VSV) L protein. The density map, at a resolution of 3.8 Å, has led to an atomic model for nearly all of the 2109-residue polypeptide chain, which comprises three enzymatic domains (RNA-dependent RNA polymerase [RdRp], polyribonucleotidyl transferase [PRNTase], and methyltransferase) and two structural domains. The RdRp resembles the corresponding enzymatic regions of dsRNA virus polymerases and influenza virus polymerase. A loop from the PRNTase (capping) domain projects into the catalytic site of the RdRp, where it appears to have the role of a priming loop and to couple product elongation to large-scale conformational changes in L.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , ARN Polimerasas Dirigidas por ADN/ultraestructura , Virus de la Estomatitis Vesicular Indiana/química , Proteínas Virales/química , Proteínas Virales/ultraestructura , Microscopía por Crioelectrón , Modelos Moleculares , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Transcripción Genética
2.
EMBO J ; 31(5): 1320-9, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22246179

RESUMEN

The minimal RNA synthesis machinery of non-segmented negative-strand RNA viruses comprises a genomic RNA encased within a nucleocapsid protein (N-RNA), and associated with the RNA-dependent RNA polymerase (RdRP). The RdRP is contained within a viral large (L) protein, which associates with N-RNA through a phosphoprotein (P). Here, we define that vesicular stomatitis virus L initiates synthesis via a de-novo mechanism that does not require N or P, but depends on a high concentration of the first two nucleotides and specific template requirements. Purified L copies a template devoid of N, and P stimulates L initiation and processivity. Full processivity of the polymerase requires the template-associated N protein. This work provides new mechanistic insights into the workings of a minimal RNA synthesis machine shared by a broad group of important human, animal and plant pathogens, and defines a mechanism by which specific inhibitors of RNA synthesis function.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Viral/metabolismo , Vesiculovirus/enzimología , Proteínas Virales/metabolismo , ARN Polimerasas Dirigidas por ADN/aislamiento & purificación , Modelos Biológicos , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Virales/aislamiento & purificación
3.
Proc Natl Acad Sci U S A ; 109(36): 14628-33, 2012 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-22908284

RESUMEN

The RNA-dependent RNA polymerase (RdRP) of nonsegmented negative-sense RNA viruses consists of a large catalytic protein (L) and a phosphoprotein cofactor (P). During infection, the RdRP replicates and transcribes the viral genome, which resides inside an oligomer of nucleocapsid protein (N-RNA). The classical view of P as a cofactor for L assigns a primary role of P as a bridge mediating the access of L to the RNA template, whereby its N-terminal domain (P(NTD)) binds L and its C-terminal domain (P(CTD)) binds N-RNA. Recent biochemical and structural studies of a prototype nonsegmented negative-sense RNA virus, vesicular stomatitis virus, suggest a role for P beyond that of a mere physical link: P induces a structural rearrangement in L and stimulates polymerase processivity. In this study, we investigated the critical requirements within P mediating the functional interaction with L to form a fully functional RdRP. We analyzed the correlation between the impact of P on the conformation of L and its activity in RNA synthesis and the consequences of these events on RdRP function. We identified three separable elements of the P(NTD) that are required for inducing the conformational rearrangement of L, stimulating polymerase processivity, and mediating transcription of the N-RNA. The functional interplay between these elements provides insight into the role of P as a dynamic player in the RNA synthesis machine, influencing essential aspects of polymerase structure and function.


Asunto(s)
Modelos Biológicos , Fosfoproteínas/metabolismo , Conformación Proteica , ARN Polimerasa Dependiente del ARN/metabolismo , Vesiculovirus/enzimología , Proteínas Estructurales Virales/metabolismo , Replicación Viral/fisiología , Western Blotting , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Espectrometría de Masas , Microscopía Electrónica , Proteínas de la Nucleocápside/metabolismo , Proteínas Virales/metabolismo
4.
Virus Evol ; 10(1): veae032, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779130

RESUMEN

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) can infect various human tissues and cell types, principally via interaction with its cognate receptor angiotensin-converting enzyme-2 (ACE2). However, how the virus evolves in different cellular environments is poorly understood. Here, we used experimental evolution to study the adaptation of the SARS-CoV-2 spike to four human cell lines expressing different levels of key entry factors. After twenty passages of a spike-expressing recombinant vesicular stomatitis virus (VSV), cell-type-specific phenotypic changes were observed and sequencing allowed the identification of sixteen adaptive spike mutations. We used VSV pseudotyping to measure the entry efficiency, ACE2 affinity, spike processing, TMPRSS2 usage, and entry pathway usage of all the mutants, alone or in combination. The fusogenicity of the mutant spikes was assessed with a cell-cell fusion assay. Finally, mutant recombinant VSVs were used to measure the fitness advantage associated with selected mutations. We found that the effects of these mutations varied across cell types, both in terms of viral entry and replicative fitness. Interestingly, two spike mutations (L48S and A372T) that emerged in cells expressing low ACE2 levels increased receptor affinity, syncytia induction, and entry efficiency under low-ACE2 conditions. Our results demonstrate specific adaptation of the SARS-CoV-2 spike to different cell types and have implications for understanding SARS-CoV-2 tissue tropism and evolution.

5.
J Virol ; 86(16): 8720-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22674999

RESUMEN

The vesicular stomatitis virus (VSV) nucleoprotein (N) associates tightly with the viral genomic RNA. This N-RNA complex constitutes the template for the RNA-dependent RNA polymerase L, which engages the nucleocapsid via its phosphoprotein cofactor P. While N and P proteins play important roles in regulating viral gene expression, the molecular basis of this regulation remains incompletely understood. Here we show that mutations in the extreme C terminus of N cause defects in viral gene expression. To determine the underlying cause of such defects, we examined the effects of the mutations separately on encapsidation and RNA synthesis. Expression of N together with P in Escherichia coli results predominantly in the formation of decameric N-RNA rings. In contrast, nucleocapsid complexes containing the substitution N(Y415A) or N(K417A) were more loosely coiled, as revealed by electron microscopy (EM). In addition, the N(EF419/420AA) mutant was unable to encapsidate RNA. To further characterize these mutants, we engineered an infectious cDNA clone of VSV and employed N-RNA templates from those viruses to reconstitute RNA synthesis in vitro. The transcription assays revealed specific defects in polymerase utilization of the template that result in overall decreased RNA quantities, including reduced amounts of leader RNA. Passage of the recombinant viruses in cell culture led to the accumulation of compensatory second-site mutations in close proximity to the original mutations, underscoring the critical role of structural features within the C terminus in regulating N function.


Asunto(s)
Nucleoproteínas/metabolismo , ARN Viral/metabolismo , Vesiculovirus/fisiología , Proteínas Virales/metabolismo , Ensamble de Virus , Replicación Viral , Cápside/ultraestructura , Análisis Mutacional de ADN , Escherichia coli/genética , Expresión Génica , Microscopía Electrónica , Unión Proteica , Multimerización de Proteína
6.
PLoS Pathog ; 7(6): e1002073, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21655110

RESUMEN

The RNA synthesis machinery of vesicular stomatitis virus (VSV) comprises the genomic RNA encapsidated by the viral nucleocapsid protein (N) and associated with the RNA dependent RNA polymerase, the viral components of which are a large protein (L) and an accessory phosphoprotein (P). The 241 kDa L protein contains all the enzymatic activities necessary for synthesis of the viral mRNAs, including capping, cap methylation and polyadenylation. Those RNA processing reactions are intimately coordinated with nucleotide polymerization such that failure to cap results in termination of transcription and failure to methylate can result in hyper polyadenylation. The mRNA processing reactions thus serve as a critical check point in viral RNA synthesis which may control the synthesis of incorrectly modified RNAs. Here, we report the length at which viral transcripts first gain access to the capping machinery during synthesis. By reconstitution of transcription in vitro with highly purified recombinant polymerase and engineered templates in which we omitted sites for incorporation of UTP, we found that transcripts that were 30-nucleotides in length were uncapped, whereas those that were 31-nucleotides in length contained a cap structure. The minimal RNA length required for mRNA cap addition was also sufficient for methylation since the 31-nucleotide long transcripts were methylated at both ribose-2'-O and guanine-N-7 positions. This work provides insights into the spatial relationship between the active sites for the RNA dependent RNA polymerase and polyribonucleotidyltransferase responsible for capping of the viral RNA. We combine the present findings with our recently described electron microscopic structure of the VSV polymerase and propose a model of how the spatial arrangement of the capping activities of L may influence nucleotide polymerization.


Asunto(s)
ARN Mensajero/metabolismo , ARN Viral/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Transcripción Genética , Vesiculovirus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Región de Flanqueo 5'/genética , Animales , Células Cultivadas , Guanina/metabolismo , Metilación , Nucleocápside/genética , Organismos Modificados Genéticamente , Caperuzas de ARN/genética , Caperuzas de ARN/metabolismo , ARN Mensajero/genética , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/genética , Ribosa/metabolismo , Spodoptera , Uridina Trifosfato/metabolismo , Estomatitis Vesicular/virología , Vesiculovirus/genética , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/genética
7.
Proc Natl Acad Sci U S A ; 107(46): 20075-80, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-21041632

RESUMEN

Nonsegmented negative-strand (NNS) RNA viruses initiate infection by delivering into the host cell a highly specialized RNA synthesis machine comprising the genomic RNA completely encapsidated by the viral nucleocapsid protein and associated with the viral polymerase. The catalytic core of this protein-RNA complex is a 250-kDa multifunctional large (L) polymerase protein that contains enzymatic activities for nucleotide polymerization as well as for each step of mRNA cap formation. Working with vesicular stomatitis virus (VSV), a prototype of NNS RNA viruses, we used negative stain electron microscopy (EM) to obtain a molecular view of L, alone and in complex with the viral phosphoprotein (P) cofactor. EM analysis, combined with proteolytic digestion and deletion mapping, revealed the organization of L into a ring domain containing the RNA polymerase and an appendage of three globular domains containing the cap-forming activities. The capping enzyme maps to a globular domain, which is juxtaposed to the ring, and the cap methyltransferase maps to a more distal and flexibly connected globule. Upon P binding, L undergoes a significant rearrangement that may reflect an optimal positioning of its functional domains for transcription. The structural map of L provides new insights into the interrelationship of its various domains, and their rearrangement on P binding that is likely important for RNA synthesis. Because the arrangement of conserved regions involved in catalysis is homologous, the structural insights obtained for VSV L likely extend to all NNS RNA viruses.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , Vesiculovirus/enzimología , ARN Polimerasas Dirigidas por ADN/metabolismo , ARN Polimerasas Dirigidas por ADN/ultraestructura , Modelos Moleculares , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Fosfoproteínas/ultraestructura , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Virales/química , Proteínas Virales/ultraestructura
8.
Proc Natl Acad Sci U S A ; 107(46): 20069-74, 2010 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-20978208

RESUMEN

Segmented negative-sense viruses of the family Arenaviridae encode a large polymerase (L) protein that contains all of the enzymatic activities required for RNA synthesis. These activities include an RNA-dependent RNA polymerase (RdRP) and an RNA endonuclease that cleaves capped primers from cellular mRNAs to prime transcription. Using purified catalytically active Machupo virus L, we provide a view of the overall architecture of this multifunctional polymerase and reconstitute complex formation with an RNA template in vitro. The L protein contains a central ring domain that is similar in appearance to the RdRP of dsRNA viruses and multiple accessory appendages that may be responsible for 5' cap formation. RNA template recognition by L requires a sequence-specific motif located at positions 2-5 in the 3' terminus of the viral genome. Moreover, L-RNA complex formation depends on single-stranded RNA, indicating that inter-termini dsRNA interactions must be partially broken for complex assembly to occur. Our results provide a model for arenavirus polymerase-template interactions and reveal the structural organization of a negative-strand RNA virus L protein.


Asunto(s)
Arenavirus del Nuevo Mundo/enzimología , ARN Polimerasa Dependiente del ARN/metabolismo , Secuencia de Bases , Biocatálisis , Microscopía Electrónica , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , ARN Viral/genética , ARN Polimerasa Dependiente del ARN/aislamiento & purificación , ARN Polimerasa Dependiente del ARN/ultraestructura , Moldes Genéticos , Proteínas Virales/aislamiento & purificación , Proteínas Virales/ultraestructura
9.
Biochemistry ; 51(1): 416-24, 2012 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-22148433

RESUMEN

DNA polymerase delta (Pol δ) is a central enzyme for eukaryotic DNA replication and repair. Pol δ is a complex of four subunits p125, p68, p50, and p12. The functional properties of Pol δ are largely determined by its interaction with its DNA sliding clamp PCNA (proliferating cellular nuclear antigen). The regulatory mechanisms that govern the association of Pol δ with PCNA are largely unknown. In this study, we identified S458, located in the PCNA-interacting protein (PIP-Box) motif of p68, as a phosphorylation site for PKA. Phosphomimetic mutation of S458 resulted in a decrease in p68 affinity for PCNA as well as the processivity of Pol δ. Our results suggest a role of phosphorylation of the PIP-motif of p68 as a molecular switch that dynamically regulates the functional properties of Pol δ.


Asunto(s)
ADN Polimerasa III/química , Antígeno Nuclear de Célula en Proliferación/química , Antígeno Nuclear de Célula en Proliferación/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos , Ácido Aspártico/genética , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , ADN Polimerasa III/antagonistas & inhibidores , ADN Polimerasa III/genética , Regulación hacia Abajo/genética , Células HeLa , Humanos , Imitación Molecular/genética , Datos de Secuencia Molecular , Mutación , Fosforilación/genética , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica/genética , Procesamiento Proteico-Postraduccional/genética , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/química , Subunidades de Proteína/genética , Serina/genética
10.
PLoS Pathog ; 6(6): e1000958, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20585632

RESUMEN

Positive-strand and double-strand RNA viruses typically compartmentalize their replication machinery in infected cells. This is thought to shield viral RNA from detection by innate immune sensors and favor RNA synthesis. The picture for the non-segmented negative-strand (NNS) RNA viruses, however, is less clear. Working with vesicular stomatitis virus (VSV), a prototype of the NNS RNA viruses, we examined the location of the viral replication machinery and RNA synthesis in cells. By short-term labeling of viral RNA with 5'-bromouridine 5'-triphosphate (BrUTP), we demonstrate that primary mRNA synthesis occurs throughout the host cell cytoplasm. Protein synthesis results in the formation of inclusions that contain the viral RNA synthesis machinery and become the predominant sites of mRNA synthesis in the cell. Disruption of the microtubule network by treatment of cells with nocodazole leads to the accumulation of viral mRNA in discrete structures that decorate the surface of the inclusions. By pulse-chase analysis of the mRNA, we find that viral transcripts synthesized at the inclusions are transported away from the inclusions in a microtubule-dependent manner. Metabolic labeling of viral proteins revealed that inhibiting this transport step diminished the rate of translation. Collectively those data suggest that microtubule-dependent transport of viral mRNAs from inclusions facilitates their translation. Our experiments also show that during a VSV infection, protein synthesis is required to redirect viral RNA synthesis to intracytoplasmic inclusions. As viral RNA synthesis is initially unrestricted, we speculate that its subsequent confinement to inclusions might reflect a cellular response to infection.


Asunto(s)
Cuerpos de Inclusión/fisiología , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Estomatitis Vesicular/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas Virales/metabolismo , Humanos , Polirribosomas , Biosíntesis de Proteínas , ARN Mensajero/genética , ARN Viral/genética , Transcripción Genética , Estomatitis Vesicular/genética , Proteínas Virales/genética , Replicación Viral
11.
Nat Commun ; 13(1): 5907, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207308

RESUMEN

The therapeutic effectiveness of oncolytic viruses (OVs) delivered intravenously is limited by the development of neutralizing antibody responses against the virus. To circumvent this limitation and to enable repeated systemic administration of OVs, here we develop Synthetic RNA viruses consisting of a viral RNA genome (vRNA) formulated within lipid nanoparticles. For two Synthetic RNA virus drug candidates, Seneca Valley virus (SVV) and Coxsackievirus A21, we demonstrate vRNA delivery and replication, virus assembly, spread and lysis of tumor cells leading to potent anti-tumor efficacy, even in the presence of OV neutralizing antibodies in the bloodstream. Synthetic-SVV replication in tumors promotes immune cell infiltration, remodeling of the tumor microenvironment, and enhances the activity of anti-PD-1 checkpoint inhibitor. In mouse and non-human primates, Synthetic-SVV is well tolerated reaching exposure well above the requirement for anti-tumor activity. Altogether, the Synthetic RNA virus platform provides an approach that enables repeat intravenous administration of viral immunotherapy.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Picornaviridae , Animales , Anticuerpos Neutralizantes , Inmunoterapia , Liposomas , Ratones , Nanopartículas , Neoplasias/terapia , Virus Oncolíticos/genética , ARN Viral/genética , Microambiente Tumoral
12.
Nat Commun ; 12(1): 7071, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34862378

RESUMEN

While multiple technologies for small allele genome editing exist, robust technologies for targeted integration of large DNA fragments in mammalian genomes are still missing. Here we develop a gene delivery tool (FiCAT) combining the precision of a CRISPR-Cas9 (find module), and the payload transfer efficiency of an engineered piggyBac transposase (cut-and-transfer module). FiCAT combines the functionality of Cas9 DNA scanning and targeting DNA, with piggyBac donor DNA processing and transfer capacity. PiggyBac functional domains are engineered providing increased on-target integration while reducing off-target events. We demonstrate efficient delivery and programmable insertion of small and large payloads in cellulo (human (Hek293T, K-562) and mouse (C2C12)) and in vivo in mouse liver. Finally, we evolve more efficient versions of FiCAT by generating a targeted diversity of 394,000 variants and undergoing 4 rounds of evolution. In this work, we develop a precise and efficient targeted insertion of multi kilobase DNA fragments in mammalian genomes.


Asunto(s)
Elementos Transponibles de ADN/genética , Edición Génica/métodos , Transposasas/genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Electroporación , Femenino , Humanos , Hígado , Masculino , Ratones , Ingeniería de Proteínas , Transposasas/metabolismo
13.
J Virol ; 83(4): 1930-40, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19073725

RESUMEN

The multifunctional large (L) polymerase protein of vesicular stomatitis virus (VSV) contains enzymatic activities essential for RNA synthesis, including mRNA cap addition and polyadenylation. We previously mapped amino acid residues G1154, T1157, H1227, and R1228, present within conserved region V (CRV) of L, as essential for mRNA cap addition. Here we show that alanine substitutions to these residues also affect 3'-end formation. Specifically, the cap-defective polymerases produced truncated transcripts that contained A-rich sequences at their 3' termini and predominantly terminated within the first 500 nucleotides (nt) of the N gene. To examine how the cap-defective polymerases respond to an authentic VSV termination and reinitiation signal present at each gene junction, we reconstituted RNA synthesis using templates that contained genes inserted (I) at the leader-N gene junction. The I genes ranged in size from 382 to 1,098 nt and were typically transcribed into full-length uncapped transcripts. In addition to lacking a cap structure, the full-length I transcripts synthesized by the cap-defective polymerases lacked an authentic polyadenylate tail and instead contained 0 to 24 A residues. Moreover, the cap-defective polymerases were also unable to copy efficiently the downstream gene. Thus, single amino acid substitutions in CRV of L protein that inhibit cap addition also inhibit polyadenylation and sequential transcription of the genome. In contrast, an amino acid substitution, K1651A, in CRVI of L protein that completely inhibits cap methylation results in the hyperpolyadenylation of mRNA. This work reveals that inhibiting cap addition and cap methylation have opposing effects on polyadenylation during VSV mRNA synthesis and provides evidence in support of a link between correct 5' cap formation and 3' polyadenylation.


Asunto(s)
Poliadenilación , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Vesiculovirus/fisiología , Proteínas no Estructurales Virales/metabolismo , Sustitución de Aminoácidos , Metilación , Mutagénesis Sitio-Dirigida , Vesiculovirus/genética , Proteínas no Estructurales Virales/genética
14.
J Virol ; 83(21): 11043-50, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19710136

RESUMEN

During conventional mRNA cap formation, two separate methyltransferases sequentially modify the cap structure, first at the guanine-N-7 (G-N-7) position and subsequently at the ribose 2'-O position. For vesicular stomatitis virus (VSV), a prototype of the nonsegmented negative-strand RNA viruses, the two methylase activities share a binding site for the methyl donor S-adenosyl-l-methionine and are inhibited by individual amino acid substitutions within the C-terminal domain of the large (L) polymerase protein. This led to the suggestion that a single methylase domain functions for both 2'-O and G-N-7 methylations. Here we report a trans-methylation assay that recapitulates both ribose 2'-O and G-N-7 modifications by using purified recombinant L and in vitro-synthesized RNA. Using this assay, we demonstrate that VSV L typically modifies the 2'-O position of the cap prior to the G-N-7 position and that G-N-7 methylation is diminished by pre-2'-O methylation of the substrate RNA. Amino acid substitutions in the C terminus of L that prevent all cap methylation in recombinant VSV (rVSV) partially retain the ability to G-N-7 methylate a pre-2'-O-methylated RNA, therefore uncoupling the effect of substitutions in the C terminus of the L protein on the two methylations. In addition, we show that the 2'-O and G-N-7 methylase activities act specifically on RNA substrates that contain the conserved elements of a VSV mRNA start at the 5' terminus. This study provides new mechanistic insights into the mRNA cap methylase activities of VSV L, demonstrates that 2'-O methylation precedes and facilitates subsequent G-N-7 methylation, and reveals an RNA sequence and length requirement for the two methylase activities. We propose a model of regulation of the activity of the C terminus of L protein in 2'-O and G-N-7 methylation of the cap structure.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/metabolismo , Guanina/metabolismo , Caperuzas de ARN/metabolismo , ARN Mensajero/metabolismo , Ribosa/metabolismo , Vesiculovirus/genética , Vesiculovirus/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Secuencia de Bases , Sitios de Unión , ARN Polimerasas Dirigidas por ADN/antagonistas & inhibidores , ARN Polimerasas Dirigidas por ADN/genética , Metilación , Metiltransferasas/antagonistas & inhibidores , Metiltransferasas/metabolismo , Caperuzas de ARN/genética , ARN Mensajero/genética , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/genética
15.
J Virol ; 82(2): 775-84, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18003731

RESUMEN

Nonsegmented negative-sense (NNS) RNA viruses cap their mRNA by an unconventional mechanism. Specifically, 5' monophosphate mRNA is transferred to GDP derived from GTP through a reaction that involves a covalent intermediate between the large polymerase protein L and mRNA. This polyribonucleotidyltransferase activity contrasts with all other capping reactions, which are catalyzed by an RNA triphosphatase and guanylyltransferase. In these reactions, a 5' diphosphate mRNA is capped by transfer of GMP via a covalent enzyme-GMP intermediate. RNA guanylyltransferases typically have a KxDG motif in which the lysine forms this covalent intermediate. Consistent with the distinct mechanism of capping employed by NNS RNA viruses, such a motif is absent from L. To determine the residues of L protein required for capping, we reconstituted the capping reaction of the prototype NNS RNA virus, vesicular stomatitis virus, from highly purified components. Using a panel of L proteins with single-amino-acid substitutions to residues universally conserved among NNS RNA virus L proteins, we define a new motif, GxxT[n]HR, present within conserved region V of L protein that is essential for this unconventional mechanism of mRNA cap formation.


Asunto(s)
Secuencia Conservada , Caperuzas de ARN/metabolismo , ARN Polimerasa Dependiente del ARN/metabolismo , Vesiculovirus/fisiología , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , ARN Polimerasa Dependiente del ARN/genética , Vesiculovirus/genética , Proteínas Virales/genética
16.
Biochemistry ; 47(43): 11367-76, 2008 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-18826257

RESUMEN

Protein phosphatase-1 (PP1) is a Ser/Thr protein phosphatase that participates in the phosphorylation/dephosphorylation regulation of a diverse range of cellular processes. The PP1 catalytic subunit (PP1) achieves this by its ability to interact with many targeting subunits such that PP1 activity is thereby specified against phosphoprotein substrates in the microvicinity of its targeting subunit. DNA polymerase delta (Pol delta) is a key enzyme in mammalian chromosomal replication. It consists of four subunits, p125, p50, p68, and p12. We identify p68 as a novel PP1 targeting subunit. PP1 was shown to associate with human DNA polymerase delta by affinity chromatography and coimmunoprecipitation assays from mammalian cell lysates and in vitro by pull-down assays. The binding domain for PP1 was identified as the sequence KRVAL, a variant of the canonical RVxF PP1 binding motif. These studies provide the first evidence for the targeting of PP1 to DNA polymerase delta. We also show that CK2 phosphorylates the Pol delta p125, p68, and p12 subunits and that these phosphorylated subunits are substrates for PP1. These findings identify a new role for p68 as a PP1 targeting subunit that implicates PP1 in the dephosphorylation of Pol delta. Our findings also show that CK2 is a strong candidate for the protein kinase involved in the in vivo phosphorylation of p68.


Asunto(s)
ARN Helicasas DEAD-box/química , ADN Polimerasa III/metabolismo , Proteína Fosfatasa 1/metabolismo , Subunidades de Proteína/metabolismo , Sitios de Unión/genética , Dominio Catalítico/genética , ARN Helicasas DEAD-box/genética , ADN Polimerasa III/química , ADN Polimerasa III/genética , Células HeLa , Humanos , Modelos Biológicos , Unión Proteica/genética , Proteína Fosfatasa 1/química , Proteína Fosfatasa 1/genética , Subunidades de Proteína/química , Subunidades de Proteína/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
Epilepsy Res ; 81(2-3): 161-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18603412

RESUMEN

Status epilepticus (SE) induces a number of events leading to programmed cell death (PCD). The aim of our work is to study the time sequence of activation of different factors in experimental SE (intraperitoneal kainic acid (KA) model). We studied ceramide, a known mediator of apoptosis in multiple models, sphingomyelinases (SMases), enzymes that break down sphingomyelin and increase ceramide thus leading to apoptosis in many models, Bcl(2), Bax, and caspase-3. SE induced a sustained ceramide increase starting 2h after kainic acid injection followed by an increase in Bax protein at 6 and 12h, and the appearance of caspase-3-activated fragment (caspase-3a) immunostaining and TUNEL positivity at 12h. Status epilepticus also induced an increase in acidic and neutral sphingomyelinases that preceded (acidic sphingomyelinase) and parallelled (acidic and neutral sphingomyelinase) the increases in ceramide. These data suggest that, in this model, Bax is activated early in the process and that its increase is sustained till 12h after kainic acid injection which is the time of first appearance of caspase-3 activation and TUNEL positivity, and that SMases contribute to increases in ceramide levels during and after status epilepticus.


Asunto(s)
Caspasa 3/metabolismo , Ceramidas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Estado Epiléptico/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Anticonvulsivantes/uso terapéutico , Muerte Celular/efectos de los fármacos , Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Etiquetado Corte-Fin in Situ/métodos , Ácido Kaínico , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/fisiopatología , Factores de Tiempo
18.
Brain Dev ; 30(8): 513-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18295995

RESUMEN

Ceramide is known to induce programmed cell death (PCD) in neural and non-neural tissues and to increase after kainic acid (KA) status epilepticus (SE). Ceramide increases have been shown to depend on NMDA receptor activation in the KA model, but these changes have not been studied in the lithium pilocarpine (LiPC) model. Thus, the purpose of this study was to determine if hippocampal ceramide levels increase after LiPC induced SE and if NMDA receptor blockade prevents PCD and any such ceramide increases. We found that LiPC induced SE resulted in ceramide increases and DNA fragmentation in the hippocampus of adult, P21, and P7 rats. The administration of MK-801, the NMDA receptor antagonist, in adults, 15min prior to pilocarpine, prevented ceramide increases, and DNA fragmentation.


Asunto(s)
Muerte Celular/fisiología , Ceramidas/metabolismo , Litio/farmacología , Pilocarpina/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones , Animales , Modelos Animales de Enfermedad , Maleato de Dizocilpina/metabolismo , Antagonistas de Aminoácidos Excitadores/metabolismo , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Ácido Kaínico/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/metabolismo
19.
ACS Infect Dis ; 4(9): 1395-1406, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30027735

RESUMEN

Dengue virus is a major human pathogen that infects over 390 million people annually leading to approximately 500 000 hospitalizations due to severe dengue. Since the only marketed vaccine, Dengvaxia, has recently been shown to increase disease severity in those lacking natural immunity, antivirals to prevent or treat dengue  infection represent a large, unmet medical need. Small molecules that target the dengue virus envelope protein, E, on the surface of the virion could act analogously to antibodies by engaging E extracellularly to block infection; however, a shortage of target-based assays suitable for screening and medicinal chemistry studies has limited efforts in this area. Here we demonstrate that the dengue E protein offers a tractable drug target for preventing dengue infection by developing a target-based assay using a recombinantly expressed dengue serotype 2 E protein. We performed a high-throughput screen of ∼20 000 compounds followed by secondary assays to confirm target-binding and antiviral activity and counter-screens to exclude compounds with nonspecific activities. These efforts yielded eight distinct chemical leads that inhibit dengue infection by binding to E and preventing E-mediated membrane fusion with potencies equal to or greater than previously described small molecule inhibitors of E. We show that a subset of these compounds inhibit viruses representative of the other three dengue serotypes and Zika virus. This work provides tools for discovery and optimization of direct-acting antivirals against dengue E and shows that this approach may be useful in developing antivirals with broad-spectrum activity against other flavivirus pathogens.


Asunto(s)
Antivirales/química , Antivirales/farmacología , Virus del Dengue/efectos de los fármacos , Descubrimiento de Drogas/métodos , Bibliotecas de Moléculas Pequeñas/farmacología , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Dengue/inmunología , Dengue/virología , Virus del Dengue/genética , Virus del Dengue/fisiología , Humanos , Bibliotecas de Moléculas Pequeñas/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos , Virus Zika/efectos de los fármacos , Virus Zika/fisiología
20.
FEBS J ; 273(13): 2984-3001, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16762037

RESUMEN

The contributions of human DNA polymerases (pols) alpha, delta and epsilon during S-phase progression were studied in order to elaborate how these enzymes co-ordinate their functions during nuclear DNA replication. Pol delta was three to four times more intensely UV cross-linked to nascent DNA in late compared with early S phase, whereas the cross-linking of pols alpha and epsilon remained nearly constant throughout the S phase. Consistently, the chromatin-bound fraction of pol delta, unlike pols alpha and epsilon, increased in the late S phase. Moreover, pol delta neutralizing antibodies inhibited replicative DNA synthesis most efficiently in late S-phase nuclei, whereas antibodies against pol epsilon were most potent in early S phase. Ultrastructural localization of the pols by immuno-electron microscopy revealed pol epsilon to localize predominantly to ring-shaped clusters at electron-dense regions of the nucleus, whereas pol delta was mainly dispersed on fibrous structures. Pol alpha and proliferating cell nuclear antigen displayed partial colocalization with pol delta and epsilon, despite the very limited colocalization of the latter two pols. These data are consistent with models where pols delta and epsilon pursue their functions at least partly independently during DNA replication.


Asunto(s)
Replicación del ADN , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/fisiología , Cromatina/química , ADN Polimerasa I/química , ADN Polimerasa II/química , ADN Polimerasa III/química , Fibroblastos/metabolismo , Células HeLa , Humanos , Microscopía Inmunoelectrónica , Mimosina/farmacología , Fase S , Rayos Ultravioleta
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA