Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 295(35): 12426-12436, 2020 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-32641492

RESUMO

Many RNA viruses create specialized membranes for genome replication by manipulating host lipid metabolism and trafficking, but in most cases, we do not know the molecular mechanisms responsible or how specific lipids may impact the associated membrane and viral process. For example, hepatitis C virus (HCV) causes a specific, large-fold increase in the steady-state abundance of intracellular desmosterol, an immediate precursor of cholesterol, resulting in increased fluidity of the membrane where HCV RNA replication occurs. Here, we establish the mechanism responsible for HCV's effect on intracellular desmosterol, whereby the HCV NS3-4A protease controls activity of 24-dehydrocholesterol reductase (DHCR24), the enzyme that catalyzes conversion of desmosterol to cholesterol. Our cumulative evidence for the proposed mechanism includes immunofluorescence microscopy experiments showing co-occurrence of DHCR24 and HCV NS3-4A protease; formation of an additional, faster-migrating DHCR24 species (DHCR24*) in cells harboring a HCV subgenomic replicon RNA or ectopically expressing NS3-4A; and biochemical evidence that NS3-4A cleaves DHCR24 to produce DHCR24* in vitro and in vivo We further demonstrate that NS3-4A cleaves DHCR24 between residues Cys91 and Thr92 and show that this reduces the intracellular conversion of desmosterol to cholesterol. Together, these studies demonstrate that NS3-4A directly cleaves DHCR24 and that this results in the enrichment of desmosterol in the membranes where NS3-4A and DHCR24 co-occur. Overall, this suggests a model in which HCV directly regulates the lipid environment for RNA replication through direct effects on the host lipid metabolism.


Assuntos
Hepacivirus/enzimologia , Metabolismo dos Lipídeos , Lipídeos de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Proteólise , RNA Viral/biossíntese , Serina Proteases/metabolismo , Proteínas não Estruturais Virais/metabolismo , Linhagem Celular Tumoral , Hepacivirus/genética , Humanos , Lipídeos de Membrana/genética , Proteínas do Tecido Nervoso/genética , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/genética , RNA Viral/genética , Serina Proteases/genética , Proteínas não Estruturais Virais/genética
2.
J Biol Chem ; 295(9): 2749-2759, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-31915248

RESUMO

Adiponectin is a highly abundant protein hormone secreted by adipose tissue. It elicits diverse biological responses, including anti-diabetic, anti-inflammatory, anti-tumor, and anti-atherosclerotic effects. Adiponectin consists of a globular domain and a collagen-like domain, and it occurs in three major oligomeric forms that self-assemble: trimers, hexamers, and high-molecular-weight oligomers. Adiponectin has been reported to bind to two seven-transmembrane domain receptors, AdipoR1 and AdipoR2, as well as to the protein T-cadherin, which is highly expressed in the cardiovascular system and binds only the high-molecular-weight form of adiponectin. The molecular mechanisms underlying this specificity remain unclear. Here we used a combination of X-ray crystallography and protein engineering to define the details of adiponectin's interaction with T-cadherin. We found that T-cadherin binds to the globular domain of adiponectin, relying on structural stabilization of this domain by bound metal ions. Moreover, we show that the adiponectin globular domain can be engineered to enhance its binding affinity for T-cadherin. These results help to define the molecular basis for the interaction between adiponectin and T-cadherin, and our engineered globular domain variants may be useful tools for further investigating adiponectin's functions.


Assuntos
Adiponectina/metabolismo , Caderinas/metabolismo , Engenharia de Proteínas , Adiponectina/química , Animais , Sítios de Ligação , Cristalografia por Raios X , Humanos , Ligação Proteica , Multimerização Proteica
3.
J Biol Chem ; 295(6): 1694-1703, 2020 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-31914414

RESUMO

Small-molecule inhibitors of translation are critical tools to study the molecular mechanisms of protein synthesis. In this study, we sought to characterize how QL47, a host-targeted, small-molecule antiviral agent, inhibits steady-state viral protein expression. We demonstrate that this small molecule broadly inhibits both viral and host protein synthesis and targets a translation step specific to eukaryotic cells. We show that QL47 inhibits protein neosynthesis initiated by both canonical cap-driven and noncanonical initiation strategies, most likely by targeting an early step in translation elongation. Our findings thus establish QL47 as a new small-molecule inhibitor that can be utilized to probe the eukaryotic translation machinery and that can be further developed as a new therapeutic agent.


Assuntos
Antivirais/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Antivirais/química , Linhagem Celular , Células HEK293 , Humanos , Proteínas/metabolismo , Bibliotecas de Moléculas Pequenas/química , Proteínas Virais/metabolismo , Viroses/tratamento farmacológico , Viroses/metabolismo , Vírus/efeitos dos fármacos , Vírus/metabolismo
4.
J Virol ; 90(2): 705-14, 2016 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-26512085

RESUMO

UNLABELLED: Arenaviruses are emerging viruses including several causative agents of severe hemorrhagic fevers in humans. The advent of next-generation sequencing technology has greatly accelerated the discovery of novel arenavirus species. However, for many of these viruses, only genetic information is available, and their zoonotic disease potential remains unknown. During the arenavirus life cycle, processing of the viral envelope glycoprotein precursor (GPC) by the cellular subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P) is crucial for productive infection. The ability of newly emerging arenaviruses to hijack human SKI-1/S1P appears, therefore, to be a requirement for efficient zoonotic transmission and human disease potential. Here we implement a newly developed cell-based molecular sensor for SKI-1/S1P to characterize the processing of arenavirus GPC-derived target sequences by human SKI-1/S1P in a quantitative manner. We show that only nine amino acids flanking the putative cleavage site are necessary and sufficient to accurately recapitulate the efficiency and subcellular location of arenavirus GPC processing. In a proof of concept, our sensor correctly predicts efficient processing of the GPC of the newly emergent pathogenic Lujo virus by human SKI-1/S1P and defines the exact cleavage site. Lastly, we employed our sensor to show efficient GPC processing of a panel of pathogenic and nonpathogenic New World arenaviruses, suggesting that GPC cleavage represents no barrier for zoonotic transmission of these pathogens. Our SKI-1/S1P sensor thus represents a rapid and robust test system for assessment of the processing of putative cleavage sites derived from the GPCs of newly discovered arenavirus by the SKI-1/S1P of humans or any other species, based solely on sequence information. IMPORTANCE: Arenaviruses are important emerging human pathogens that can cause severe hemorrhagic fevers with high mortality in humans. A crucial step in productive arenavirus infection of human cells is the processing of the viral envelope glycoprotein by the cellular subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P). In order to break the species barrier during zoonotic transmission and cause severe disease in humans, newly emerging arenaviruses must be able to hijack human SKI-1/S1P efficiently. Here we implement a newly developed cell-based molecular sensor for human SKI-1/S1P to characterize the processing of arenavirus glycoproteins in a quantitative manner. We further use our sensor to correctly predict efficient processing of the glycoprotein of the newly emergent pathogenic Lujo virus by human SKI-1/S1P. Our sensor thus represents a rapid and robust test system with which to assess whether the glycoprotein of any newly emerging arenavirus can be efficiently processed by human SKI-1/S1P, based solely on sequence information.


Assuntos
Glicoproteínas/metabolismo , Lujo virus/fisiologia , Pró-Proteína Convertases/metabolismo , Processamento de Proteína Pós-Traducional , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Humanos , Técnicas de Sonda Molecular
5.
Antimicrob Agents Chemother ; 59(1): 85-95, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25313218

RESUMO

Dengue virus (DENV), a member of the Flaviviridae family, is a mosquito-borne pathogen and the cause of dengue fever. The increasing prevalence of DENV worldwide heightens the need for an effective vaccine and specific antivirals. Due to the dependence of DENV upon the lipid biosynthetic machinery of the host cell, lipid signaling and metabolism present unique opportunities for inhibiting viral replication. We screened a library of bioactive lipids and modulators of lipid metabolism and identified 4-hydroxyphenyl retinamide (4-HPR) (fenretinide) as an inhibitor of DENV in cell culture. 4-HPR inhibits the steady-state accumulation of viral genomic RNA and reduces viremia when orally administered in a murine model of DENV infection. The molecular target responsible for this antiviral activity is distinct from other known inhibitors of DENV but appears to affect other members of the Flaviviridae, including the West Nile, Modoc, and hepatitis C viruses. Although long-chain ceramides have been implicated in DENV replication, we demonstrate that DENV is insensitive to the perturbation of long-chain ceramides in mammalian cell culture and that the effect of 4-HPR on dihydroceramide homeostasis is separable from its antiviral activity. Likewise, the induction of reactive oxygen species by 4-HPR is not required for the inhibition of DENV. The inhibition of DENV in vivo by 4-HPR, combined with its well-established safety and tolerability in humans, suggests that it may be repurposed as a pan-Flaviviridae antiviral agent. This work also illustrates the utility of bioactive lipid screens for identifying critical interactions of DENV and other viral pathogens with host lipid biosynthesis, metabolism, and signal transduction.


Assuntos
Vírus da Dengue/crescimento & desenvolvimento , Dengue/tratamento farmacológico , Fenretinida/uso terapêutico , Viremia/tratamento farmacológico , Replicação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Feminino , Células HEK293 , Hepacivirus/crescimento & desenvolvimento , Humanos , Camundongos , Camundongos Transgênicos , Espécies Reativas de Oxigênio/metabolismo , Células Vero , Vírus do Nilo Ocidental/crescimento & desenvolvimento
6.
J Virol ; 87(11): 6406-14, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536681

RESUMO

The arenaviruses are an important family of emerging viruses that includes several causative agents of severe hemorrhagic fevers in humans that represent serious public health problems. A crucial step of the arenavirus life cycle is maturation of the envelope glycoprotein precursor (GPC) by the cellular subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P). Comparison of the currently known sequences of arenavirus GPCs revealed the presence of a highly conserved aromatic residue at position P7 relative to the SKI-1/S1P cleavage side in Old World and clade C New World arenaviruses but not in New World viruses of clades A and B or cellular substrates of SKI-1/S1P. Using a combination of molecular modeling and structure-function analysis, we found that residue Y285 of SKI-1/S1P, distal from the catalytic triad, is implicated in the molecular recognition of the aromatic "signature residue" at P7 in the GPC of Old World Lassa virus. Using a quantitative biochemical approach, we show that Y285 of SKI-1/S1P is crucial for the efficient processing of peptides derived from Old World and clade C New World arenavirus GPCs but not of those from clade A and B New World arenavirus GPCs. The data suggest that during coevolution with their mammalian hosts, GPCs of Old World and clade C New World viruses expanded the molecular contacts with SKI-1/S1P beyond the classical four-amino-acid recognition sequences and currently occupy an extended binding pocket.


Assuntos
Infecções por Arenaviridae/enzimologia , Arenavirus do Novo Mundo/metabolismo , Arenavirus do Velho Mundo/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Infecções por Arenaviridae/genética , Infecções por Arenaviridae/virologia , Arenavirus do Novo Mundo/classificação , Arenavirus do Novo Mundo/genética , Arenavirus do Velho Mundo/classificação , Arenavirus do Velho Mundo/genética , Células CHO , Cricetinae , Humanos , Dados de Sequência Molecular , Pró-Proteína Convertases/química , Pró-Proteína Convertases/genética , Processamento de Proteína Pós-Traducional , Alinhamento de Sequência , Serina Endopeptidases/química , Serina Endopeptidases/genética , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
7.
Virology ; 436(1): 127-33, 2013 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-23218200

RESUMO

Maturation of the arenavirus GP precursor (GPC) involves proteolytic processing by cellular signal peptidase and the proprotein convertase subtilisin kexin isozyme 1 (SKI-1)/site 1 protease (S1P), yielding a tripartite complex comprised of a stable signal peptide (SSP), the receptor-binding GP1, and the fusion-active transmembrane GP2. Here we investigated the roles of SKI-1/S1P processing and SSP in the biosynthesis of the recombinant GP ectodomains of lymphocytic choriomeningitis virus (LCMV) and Lassa virus (LASV). When expressed in mammalian cells, the LCMV and LASV GP ectodomains underwent processing by SKI-1/S1P, followed by dissociation of GP1 from GP2. The GP2 ectodomain spontaneously formed trimers as revealed by chemical cross-linking. The endogenous SSP, known to be crucial for maturation and transport of full-length arenavirus GPC was dispensable for processing and secretion of the soluble GP ectodomain, suggesting a specific role of SSP in the stable prefusion conformation and transport of full-length GPC.


Assuntos
Glicoproteínas/química , Glicoproteínas/metabolismo , Vírus Lassa/metabolismo , Vírus da Coriomeningite Linfocítica/metabolismo , Pró-Proteína Convertases/metabolismo , Sinais Direcionadores de Proteínas , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetinae , Glicoproteínas/biossíntese , Células HEK293 , Humanos , Proteólise , Proteínas do Envelope Viral/biossíntese
8.
Expert Rev Anti Infect Ther ; 10(11): 1297-309, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23241187

RESUMO

Arenaviruses are a large group of emerging viruses including several causative agents of severe hemorrhagic fevers with high mortality in man. Considering the number of people affected and the currently limited therapeutic options, novel efficacious therapeutics against arenaviruses are urgently needed. Over the past decade, significant advances in knowledge about the basic virology of arenaviruses have been accompanied by the development of novel therapeutics targeting different steps of the arenaviral life cycle. High-throughput, small-molecule screens identified potent and broadly active inhibitors of arenavirus entry that were instrumental for the dissection of unique features of arenavirus fusion. Novel inhibitors of arenavirus replication have been successfully tested in animal models and hold promise for application in humans. Late in the arenavirus life cycle, the proteolytic processing of the arenavirus envelope glycoprotein precursor and cellular factors critically involved virion assembly and budding provide further promising 'druggable' targets for novel therapeutics to combat human arenavirus infection.


Assuntos
Antivirais/farmacologia , Antivirais/uso terapêutico , Infecções por Arenaviridae/tratamento farmacológico , Arenaviridae/efeitos dos fármacos , Arenaviridae/fisiologia , Descoberta de Drogas , Humanos , Proteínas do Envelope Viral , Proteínas Virais de Fusão/antagonistas & inibidores , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
9.
Viruses ; 4(10): 2162-81, 2012 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23202458

RESUMO

Arenaviruses include lethal human pathogens which pose serious public health threats. So far, no FDA approved vaccines are available against arenavirus infections, and therapeutic options are limited, making the identification of novel drug targets for the development of efficacious therapeutics an urgent need. Arenaviruses are comprised of two RNA genome segments and four proteins, the polymerase L, the envelope glycoprotein GP, the matrix protein Z, and the nucleoprotein NP. A crucial step in the arenavirus life-cycle is the biosynthesis and maturation of the GP precursor (GPC) by cellular signal peptidases and the cellular enzyme Subtilisin Kexin Isozyme-1 (SKI-1)/Site-1 Protease (S1P) yielding a tripartite mature GP complex formed by GP1/GP2 and a stable signal peptide (SSP). GPC cleavage by SKI-1/S1P is crucial for fusion competence and incorporation of mature GP into nascent budding virion particles. In a first part of our review, we cover basic aspects and newer developments in the biosynthesis of arenavirus GP and its molecular interaction with SKI-1/S1P. A second part will then highlight the potential of SKI-1/S1P-mediated processing of arenavirus GPC as a novel target for therapeutic intervention to combat human pathogenic arenaviruses.


Assuntos
Arenavirus/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/biossíntese , Sequência de Aminoácidos , Antivirais/farmacologia , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/metabolismo , Infecções por Arenaviridae/virologia , Arenavirus/efeitos dos fármacos , Arenavirus/patogenicidade , Glicosilação , Complexo de Golgi/metabolismo , Complexo de Golgi/virologia , Humanos , Pró-Proteína Convertases/antagonistas & inibidores , Sinais Direcionadores de Proteínas , Proteólise , Pirrolidinas/farmacologia , Receptores de Superfície Celular/metabolismo , Proteínas do Envelope Viral/química , Montagem de Vírus , Ligação Viral
10.
J Virol ; 86(9): 4935-46, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22357276

RESUMO

A crucial step in the life cycle of arenaviruses is the biosynthesis of the mature fusion-active viral envelope glycoprotein (GP) that is essential for virus-host cell attachment and entry. The maturation of the arenavirus GP precursor (GPC) critically depends on proteolytic processing by the cellular proprotein convertase (PC) subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P). Here we undertook a molecular characterization of the SKI-1/S1P processing of the GPCs of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV) and the pathogenic Lassa virus (LASV). Previous studies showed that the GPC of LASV undergoes processing in the endoplasmic reticulum (ER)/cis-Golgi compartment, whereas the LCMV GPC is cleaved in a late Golgi compartment. Herein we confirm these findings and provide evidence that the SKI-1/S1P recognition site RRLL, present in the SKI-1/S1P prodomain and LASV GPC, but not in the LCMV GPC, is crucial for the processing of the LASV GPC in the ER/cis-Golgi compartment. Our structure-function analysis revealed that the cleavage of arenavirus GPCs, but not cellular substrates, critically depends on the autoprocessing of SKI-1/S1P, suggesting differences in the processing of cellular and viral substrates. Deletion mutagenesis showed that the transmembrane and intracellular domains of SKI-1/S1P are dispensable for arenavirus GPC processing. The expression of a soluble form of the protease in SKI-I/S1P-deficient cells resulted in the efficient processing of arenavirus GPCs and rescued productive virus infection. However, exogenous soluble SKI-1/S1P was unable to process LCMV and LASV GPCs displayed at the surface of SKI-I/S1P-deficient cells, indicating that GPC processing occurs in an intracellular compartment. In sum, our study reveals important differences in the SKI-1/S1P processing of viral and cellular substrates.


Assuntos
Arenavirus/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Motivos de Aminoácidos , Animais , Arenavirus/genética , Linhagem Celular , Membrana Celular/metabolismo , Cricetinae , Retículo Endoplasmático/metabolismo , Ordem dos Genes , Complexo de Golgi/metabolismo , Humanos , Pró-Proteína Convertases/química , Estrutura Terciária de Proteína , Serina Endopeptidases/química , Solubilidade , Especificidade por Substrato , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
11.
Virology ; 423(1): 14-22, 2012 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-22154237

RESUMO

The cellular protease subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P) is implicated in the proteolytic processing of the viral envelope glycoprotein precursor (GPC) of arenaviruses, a step strictly required for production of infectious progeny. The small molecule SKI-1/S1P inhibitor PF-429242 was shown to have anti-viral activity against Old World arenaviruses. Here we extended these studies and show that PF-429242 also inhibits GPC processing and productive infection of New World arenaviruses, making PF-429242 a broadly active anti-arenaviral drug. In combination therapy, PF-429242 potentiated the anti-viral activity of ribavirin, indicating a synergism between the two drugs. A hallmark of arenaviruses is their ability to establish persistent infection in vitro and in vivo. Notably, PF-429242 was able to efficiently and rapidly clear persistent infection by arenaviruses. Interruption of drug treatment did not result in re-emergence of infection, indicating that PF-429242 treatment leads to virus extinction.


Assuntos
Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/enzimologia , Arenavirus do Velho Mundo/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Pró-Proteína Convertases/antagonistas & inibidores , Pirrolidinas/farmacologia , Sequência de Aminoácidos , Infecções por Arenaviridae/virologia , Arenavirus do Velho Mundo/metabolismo , Sequência de Bases , Linhagem Celular , Humanos , Dados de Sequência Molecular , Pró-Proteína Convertases/genética , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
12.
Virology ; 417(1): 18-26, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21612810

RESUMO

A crucial step in the arenavirus life cycle is the proteolytic processing of the viral envelope glycoprotein precursor (GPC) by the cellular proprotein convertase (PC) subtilisin kexin isozyme-1 (SKI-1)/site-1 protease (S1P). Here we conducted a systematic and quantitative analysis of SKI-1/S1P processing of peptides derived from the recognition sites of GPCs of different Old World and New World arenaviruses. We found that SKI-1/S1P showed a strong preference for arenaviral sequences resembling its autoprocessing sites, which are recurrent motifs in arenaviral GPCs. The African arenaviruses Lassa, Mobala, and Mopeia resemble the SKI-1/S1P autoprocessing C-site, whereas sequences derived from Clade B New World viruses Junin and Tacaribe have similarities to the autoprocessing B-site. In contrast, analogous peptides derived from cellular SKI-1/S1P substrates were remarkably poor substrates. The data suggest that arenavirus GPCs evolved to mimic SKI-1/S1P autoprocessing sites, likely ensuring efficient cleavage and perhaps avoiding competition with SKI-1/S1P's cellular substrates.


Assuntos
Arenavirus/metabolismo , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Proteínas do Envelope Viral/metabolismo , Arenavirus/genética , Células HEK293 , Humanos , Pró-Proteína Convertases/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina Endopeptidases/genética , Proteínas do Envelope Viral/genética
13.
J Virol ; 85(4): 1662-70, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21106748

RESUMO

Arenaviruses are enveloped RNA viruses with a nonlytic life cycle that cause acute and persistent infections. Here, we investigated the role of the host cell's unfolded protein response (UPR) in infection of the prototypic arenavirus lymphocytic choriomeningitis virus (LCMV). In mammalian cells, the endoplasmic reticulum (ER) chaperone protein GRP78/BiP functions as the principal sensor for the induction of the UPR and interacts with three mediators: kinase/endonuclease inositol-requiring protein 1 (IRE1), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Acute infection with LCMV resulted in a selective induction of the ATF6-regulated branch of the UPR, whereas pathways controlled by PERK and IRE1 were neither activated nor blocked. Expression of individual LCMV proteins revealed that the viral glycoprotein precursor (GPC), but not that of other viral proteins, was responsible for the induction of ATF6. Rapid downregulation of the viral GPC during transition from acute to persistent LCMV infection restored basal levels of UPR signaling. To address a possible role of ATF6 signaling in LCMV infection, we used cells deficient in site 2 protease (S2P), a metalloprotease required for the activation of ATF6. Cells deficient in S2P showed significantly lower levels of production of infectious virus during acute but not persistent infection, indicating a requirement for ATF6-mediated signaling for optimal virus multiplication. In summary, acute LCMV infection seems to selectively induce the ATF6-regulated branch of the UPR that is likely beneficial for virus replication and cell viability, but it avoids induction of PERK and IRE1, whose activation may be detrimental for virus and the host cell.


Assuntos
Fator 6 Ativador da Transcrição/metabolismo , Células Epiteliais/virologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Precursores de Proteínas/farmacologia , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Proteínas do Envelope Viral/farmacologia , Fator 6 Ativador da Transcrição/farmacologia , Animais , Arenavirus/patogenicidade , Linhagem Celular , Linhagem Celular Tumoral , Cricetinae , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Regulação da Expressão Gênica , Glicoproteínas/metabolismo , Glicoproteínas/farmacologia , Proteínas de Choque Térmico/metabolismo , Humanos , Fígado/citologia , Fígado/virologia , Pulmão/citologia , Pulmão/virologia , Vírus da Coriomeningite Linfocítica/metabolismo , Dobramento de Proteína , Precursores de Proteínas/metabolismo , Transdução de Sinais , Resposta a Proteínas não Dobradas/fisiologia , Proteínas do Envelope Viral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA