Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Cell ; 154(1): 118-33, 2013 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-23827678

RESUMO

The CACNA1A gene, encoding the voltage-gated calcium channel subunit α1A, is involved in pre- and postsynaptic Ca(2+) signaling, gene expression, and several genetic neurological disorders. We found that CACNA1A coordinates gene expression using a bicistronic mRNA bearing a cryptic internal ribosomal entry site (IRES). The first cistron encodes the well-characterized α1A subunit. The second expresses a transcription factor, α1ACT, which coordinates expression of a program of genes involved in neural and Purkinje cell development. α1ACT also contains the polyglutamine (polyQ) tract that, when expanded, causes spinocerebellar ataxia type 6 (SCA6). When expressed as an independent polypeptide, α1ACT-bearing an expanded polyQ tract-lacks transcription factor function and neurite outgrowth properties, causes cell death in culture, and leads to ataxia and cerebellar atrophy in transgenic mice. Suppression of CACNA1A IRES function in SCA6 may be a potential therapeutic strategy.


Assuntos
Canais de Cálcio/genética , Ataxias Espinocerebelares/genética , Fatores de Transcrição/genética , Animais , Canais de Cálcio/metabolismo , Morte Celular , Linhagem Celular Tumoral , Cerebelo/embriologia , Cerebelo/fisiopatologia , Regulação da Expressão Gênica , Humanos , Camundongos , Neuritos/metabolismo , Peptídeos/genética , Células de Purkinje/metabolismo , Ratos , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/fisiopatologia , Fatores de Transcrição/metabolismo , Transcrição Gênica
2.
PLoS Biol ; 19(6): e3001265, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34143766

RESUMO

The search for potential antibody-based diagnostics, vaccines, and therapeutics for pandemic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has focused almost exclusively on the spike (S) and nucleocapsid (N) proteins. Coronavirus membrane (M), ORF3a, and ORF8 proteins are humoral immunogens in other coronaviruses (CoVs) but remain largely uninvestigated for SARS-CoV-2. Here, we use ultradense peptide microarray mapping to show that SARS-CoV-2 infection induces robust antibody responses to epitopes throughout the SARS-CoV-2 proteome, particularly in M, in which 1 epitope achieved excellent diagnostic accuracy. We map 79 B cell epitopes throughout the SARS-CoV-2 proteome and demonstrate that antibodies that develop in response to SARS-CoV-2 infection bind homologous peptide sequences in the 6 other known human CoVs. We also confirm reactivity against 4 of our top-ranking epitopes by enzyme-linked immunosorbent assay (ELISA). Illness severity correlated with increased reactivity to 9 SARS-CoV-2 epitopes in S, M, N, and ORF3a in our population. Our results demonstrate previously unknown, highly reactive B cell epitopes throughout the full proteome of SARS-CoV-2 and other CoV proteins.


Assuntos
Anticorpos Antivirais/imunologia , COVID-19/imunologia , SARS-CoV-2/imunologia , Proteínas Virais/imunologia , Anticorpos Antivirais/sangue , COVID-19/patologia , Coronavirus/imunologia , Reações Cruzadas , Epitopos de Linfócito B , Humanos , Epitopos Imunodominantes , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Proteoma/imunologia , Índice de Gravidade de Doença
3.
Proc Natl Acad Sci U S A ; 117(12): 6784-6791, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32152109

RESUMO

Infection by Rhinovirus-C (RV-C), a species of Picornaviridae Enterovirus, is strongly associated with childhood asthma exacerbations. Cellular binding and entry by all RV-C, which trigger these episodes, is mediated by the first extracellular domain (EC1) of cadherin-related protein 3 (CDHR3), a surface cadherin-like protein expressed primarily on the apical surfaces of ciliated airway epithelial cells. Although recombinant EC1 is a potent inhibitor of viral infection, there is no molecular description of this protein or its binding site on RV-C. Here we present cryo-electron microscopy (EM) data resolving the EC1 and EC1+2 domains of human CDHR3 complexed with viral isolate C15a. Structure-suggested residues contributing to required interfaces on both EC1 and C15a were probed and identified by mutagenesis studies with four different RV-C genotypes. In contrast to most other rhinoviruses, which bind intercellular adhesion molecule 1 receptors via a capsid protein VP1-specific fivefold canyon feature, the CDHR3 EC1 contacts C15a, and presumably all RV-Cs, in a unique cohesive footprint near the threefold vertex, encompassing residues primarily from viral protein VP3, but also from VP1 and VP2. The EC1+2 footprint on C15a is similar to that of EC1 alone but shows that steric hindrance imposed by EC2 would likely prevent multiprotein binding by the native receptor at any singular threefold vertex. Definition of the molecular interface between the RV-Cs and their receptors provides new avenues that can be explored for potential antiviral therapies.


Assuntos
Caderinas/química , Caderinas/metabolismo , Microscopia Crioeletrônica/métodos , Enterovirus/química , Enterovirus/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Virais/metabolismo , Proteínas Relacionadas a Caderinas , Enterovirus/classificação , Infecções por Enterovirus/virologia , Células HeLa , Humanos , Modelos Moleculares , Conformação Proteica
4.
Hum Mol Genet ; 29(5): 736-744, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-31841129

RESUMO

Selective pressures imposed by pathogens have varied among human populations throughout their evolution, leading to marked inter-population differences at some genes mediating susceptibility to infectious and immune-related diseases. Here, we investigated the evolutionary history of a common polymorphism resulting in a Y529 versus C529 change in the cadherin related family member 3 (CDHR3) receptor which underlies variable susceptibility to rhinovirus-C infection and is associated with severe childhood asthma. The protective variant is the derived allele and is found at high frequency worldwide (69-95%). We detected genome-wide significant signatures of natural selection consistent with a rapid increase of the haplotypes carrying the allele, suggesting that non-neutral processes have acted on this locus across all human populations. However, the allele has not fixed in any population despite multiple lines of evidence suggesting that the mutation predates human migrations out of Africa. Using an approximate Bayesian computation method, we estimate the age of the mutation while explicitly accounting for past demography and positive or frequency-dependent balancing selection. Our analyses indicate a single emergence of the mutation in anatomically modern humans ~150 000 years ago and indicate that balancing selection has maintained the beneficial allele at high equilibrium frequencies worldwide. Apart from the well-known cases of the MHC and ABO genes, this study provides the first evidence that negative frequency-dependent selection plausibly acted on a human disease susceptibility locus, a form of balancing selection compatible with typical transmission dynamics of communicable respiratory viruses that might exploit CDHR3.


Assuntos
Asma/patologia , Caderinas/genética , Enterovirus/fisiologia , Predisposição Genética para Doença , Haplótipos , Proteínas de Membrana/genética , Polimorfismo de Nucleotídeo Único , Seleção Genética , Asma/etiologia , Asma/história , Teorema de Bayes , Proteínas Relacionadas a Caderinas , Criança , Genoma Humano , História Antiga , Humanos
5.
J Virol ; 95(11)2021 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-33762415

RESUMO

Historically, men rather than women have been selected for invited speaking positions at the four prominent virology conference series we have followed since the 1980s. However, the low ratio of female representation in most earlier years (20%, 1982-2010) has shown encouraging improvement (37%, 2011-2017), particularly over the last few years (48% from 2018-2020). We describe this promising rise in inclusion and discuss factors that may help perpetuate and extend this overdue transformation towards gender parity.

6.
PLoS Pathog ; 14(12): e1007477, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30532249

RESUMO

Viruses in the rhinovirus C species (RV-C) are more likely to cause severe wheezing illnesses and asthma exacerbations in children than related isolates of the RV-A or RV-B. The RV-C capsid is structurally distinct from other rhinoviruses and does not bind ICAM-1 or LDL receptors. The RV-C receptor is instead, human cadherin-related family member 3 (CDHR3), a protein unique to the airway epithelium. A single nucleotide polymorphism (rs6967330, encoding C529Y) in CDHR3 regulates the display density of CDHR3 on cell surfaces and is among the strongest known genetic correlates for childhood virus-induced asthma susceptibility. CDHR3 immunoprecipitations from transfected or transduced cell lysates were used to characterize the RV-C interaction requirements. The C529 and Y529 variations in extracellular repeat domain 5 (EC5), bound equivalently to virus. Glycosylase treatment followed by mass spectrometry mapped 3 extracellular N-linked modification sites, and further detected surface-dependent, α2-6 sialyation unique to the Y529 format. None of these modifications were required for RV-C recognition, but removal or even dilution of structurally stabilizing calcium ions from the EC junctions irreversibly abrogated virus binding. CDHR3 deletions expressed in HeLa cells or as bacterial recombinant proteins, mapped the amino-terminal EC1 unit as the required virus contact. Derivatives containing the EC1 domain, could not only recapitulate virus:receptor interactions in vitro, but also directly inhibit RV-C infection of susceptible cells for several virus genotypes (C02, C15, C41, and C45). We propose that all RV-C use the same EC1 landing pad, interacting with putative EC3-mediated multimerization formats of CDHR3.


Assuntos
Caderinas/química , Infecções por Enterovirus , Enterovirus/patogenicidade , Proteínas de Membrana/química , Ligação Viral , Proteínas Relacionadas a Caderinas , Linhagem Celular , Humanos , Imunoprecipitação/métodos , Conformação Proteica
7.
Am J Respir Cell Mol Biol ; 61(4): 450-458, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30916989

RESUMO

CDHR3 (cadherin-related family member 3) is a transmembrane protein that is highly expressed in airway epithelia and the only known receptor for rhinovirus C (RV-C). A CDHR3 SNP (rs6967330) with G to A base change has been linked to severe exacerbations of asthma and increased susceptibility to RV-C infections in young children. The goals of this study were to determine the subcellular localization of CDHR3 and to test the hypothesis that CDHR3 asthma-risk genotype affects epithelial cell function and susceptibility to RV-C infections of the airway epithelia. We used immunofluorescence imaging, Western blot analysis, and transmission electron microscopy to show CDHR3 subcellular localization in apical cells, including expression in the cilia of airway epithelia. Polymorphisms in CDHR3 rs6967330 locus (G→A) that were previously associated with childhood asthma were related to differences in CDHR3 expression and epithelial cell function. The rs6967330 A allele was associated with higher overall protein expression and RV-C binding and replication compared with the rs6967330 G allele. Furthermore, the rs6967330 A allele was associated with earlier ciliogenesis and higher FOXJ1 expression. Finally, CDHR3 genotype had no significant effects on membrane integrity or ciliary beat function. These findings provide information on the subcellular localization and possible functions of CDHR3 in the airways and link CDHR3 asthma-risk genotype to increased RV-C binding and replication.


Assuntos
Caderinas/genética , Células Epiteliais/virologia , Proteínas de Membrana/genética , Infecções por Picornaviridae/genética , Polimorfismo de Nucleotídeo Único , Mucosa Respiratória/patologia , Rhinovirus/fisiologia , Alelos , Asma/complicações , Asma/genética , Brônquios/patologia , Proteínas Relacionadas a Caderinas , Caderinas/fisiologia , Cílios/química , Células Epiteliais/química , Células Epiteliais/ultraestrutura , Predisposição Genética para Doença , Genótipo , Humanos , Proteínas de Membrana/fisiologia , Infecções por Picornaviridae/virologia , Mucosa Respiratória/virologia , Frações Subcelulares/química
8.
Proc Natl Acad Sci U S A ; 113(32): 8997-9002, 2016 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-27511920

RESUMO

Isolates of rhinovirus C (RV-C), a recently identified Enterovirus (EV) species, are the causative agents of severe respiratory infections among children and are linked to childhood asthma exacerbations. The RV-C have been refractory to structure determination because they are difficult to propagate in vitro. Here, we report the cryo-EM atomic structures of the full virion and native empty particle (NEP) of RV-C15a. The virus has 60 "fingers" on the virus outer surface that probably function as dominant immunogens. Because the NEPs also display these fingers, they may have utility as vaccine candidates. A sequence-conserved surface depression adjacent to each finger forms a likely binding site for the sialic acid on its receptor. The RV-C, unlike other EVs, are resistant to capsid-binding antiviral compounds because the hydrophobic pocket in VP1 is filled with multiple bulky residues. These results define potential molecular determinants for designing antiviral therapeutics and vaccines.


Assuntos
Asma/etiologia , Enterovirus/ultraestrutura , Vírion/ultraestrutura , Asma/virologia , Sequência Conservada , Microscopia Crioeletrônica , Glicosilação , Humanos , Receptores Virais/química
9.
Emerg Infect Dis ; 24(2): 267-274, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29350142

RESUMO

We describe a lethal respiratory outbreak among wild chimpanzees in Uganda in 2013 for which molecular and epidemiologic analyses implicate human rhinovirus C as the cause. Postmortem samples from an infant chimpanzee yielded near-complete genome sequences throughout the respiratory tract; other pathogens were absent. Epidemiologic modeling estimated the basic reproductive number (R0) for the epidemic as 1.83, consistent with the common cold in humans. Genotyping of 41 chimpanzees and examination of 24 published chimpanzee genomes from subspecies across Africa showed universal homozygosity for the cadherin-related family member 3 CDHR3-Y529 allele, which increases risk for rhinovirus C infection and asthma in human children. These results indicate that chimpanzees exhibit a species-wide genetic susceptibility to rhinovirus C and that this virus, heretofore considered a uniquely human pathogen, can cross primate species barriers and threatens wild apes. We advocate engineering interventions and prevention strategies for rhinovirus infections for both humans and wild apes.


Assuntos
Doenças dos Símios Antropoides/virologia , Enterovirus , Pan troglodytes , Infecções por Picornaviridae/veterinária , Animais , Doenças dos Símios Antropoides/epidemiologia , Surtos de Doenças , Predisposição Genética para Doença , Genótipo , Modelos Biológicos , Pan troglodytes/genética , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/mortalidade , Infecções por Picornaviridae/virologia , Uganda
10.
J Virol ; 91(7)2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28100615

RESUMO

Human rhinoviruses (RVs) of the A, B, and C species are defined agents of the common cold. But more than that, RV-A and RV-C are the dominant causes of hospitalization category infections in young children, especially those with asthma. The use of cadherin-related family member 3 (CDHR3) by RV-C as its cellular receptor creates a direct phenotypic link between human genetics (G versus A alleles cause Cys529 versus Tyr529 protein variants) and the efficiency with which RV-C can infect cells. With a lower cell surface display density, the human-specific Cys529 variant apparently confers partial protection from the severest virus-induced asthma episodes. Selective pressure favoring the Cys529 codon may have coemerged with the evolution of RV-C and helped shape modern human genomes against the virus-susceptible, albeit ancestral Tyr529.


Assuntos
Caderinas/metabolismo , Resfriado Comum/virologia , Proteínas de Membrana/metabolismo , Rhinovirus/genética , Animais , Proteínas Relacionadas a Caderinas , Caderinas/genética , Evolução Molecular , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Proteínas de Membrana/genética , Rhinovirus/metabolismo , Ligação Viral
11.
J Virol ; 91(16)2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28592542

RESUMO

Scientific conferences are most beneficial to participants when they showcase significant new experimental developments, accurately summarize the current state of the field, and provide strong opportunities for collaborative networking. A top-notch slate of invited speakers, assembled by conference organizers or committees, is key to achieving these goals. The perceived underrepresentation of female speakers at prominent scientific meetings is currently a popular topic for discussion, but one that often lacks supportive data. We compiled the full rosters of invited speakers over the last 35 years for four prominent international virology conferences, the American Society for Virology Annual Meeting (ASV), the International Herpesvirus Workshop (IHW), the Positive-Strand RNA Virus Symposium (PSR), and the Gordon Research Conference on Viruses & Cells (GRC). The rosters were cross-indexed by unique names, gender, year, and repeat invitations. When plotted as gender-dependent trends over time, all four conferences showed a clear proclivity for male-dominated invited speaker lists. Encouragingly, shifts toward parity are emerging within all units, but at different rates. Not surprisingly, both selection of a larger percentage of first-time participants and the presence of a woman on the speaker selection committee correlated with improved parity. Session chair information was also collected for the IHW and GRC. These visible positions also displayed a strong male dominance over time that is eroding slowly. We offer our personal interpretation of these data to aid future organizers achieve improved equity among the limited number of available positions for session moderators and invited speakers.IMPORTANCE Politicians and media members have a tendency to cite anecdotes as conclusions without any supporting data. This happens so frequently now, that a name for it has emerged: fake news. Good science proceeds otherwise. The underrepresentation of women as invited speakers at international scientific conferences exemplifies a present-day discussion topic usually occurring without facts to support or refute the arguments. We now provide records profiling four prominent virology conferences over the years 1982 to 2017 with the intention that the trends and accompanying analyses of the gender parity of invited speakers may allow the ongoing discussions to be informed.

12.
J Virol ; 91(8)2017 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-28179529

RESUMO

The RNA rhinoviruses (RV) encode 2A proteases (2Apro) that contribute essential polyprotein processing and host cell shutoff functions during infection, including the cleavage of Phe/Gly-containing nucleoporin proteins (Nups) within nuclear pore complexes (NPC). Within the 3 RV species, multiple divergent genotypes encode diverse 2Apro sequences that act differentially on specific Nups. Since only subsets of Phe/Gly motifs, particularly those within Nup62, Nup98, and Nup153, are recognized by transport receptors (karyopherins) when trafficking large molecular cargos through the NPC, the processing preferences of individual 2Apro predict RV genotype-specific targeting of NPC pathways and cargos. To test this idea, transformed HeLa cell lines were created with fluorescent cargos (mCherry) for the importin α/ß, transportin 1, and transportin 3 import pathways and the Crm1-mediated export pathway. Live-cell imaging of single cells expressing recombinant RV 2Apro (A16, A45, B04, B14, B52, C02, and C15) showed disruption of each pathway with measurably different efficiencies and reaction rates. The B04 and B52 proteases preferentially targeted Nups in the import pathways, while B04 and C15 proteases were more effective against the export pathway. Virus-type-specific trends were also observed during infection of cells with A16, B04, B14, and B52 viruses or their chimeras, as measured by NF-κB (p65/Rel) translocation into the nucleus and the rates of virus-associated cytopathic effects. This study provides new tools for evaluating the host cell response to RV infections in real time and suggests that differential 2Apro activities explain, in part, strain-dependent host responses and diverse RV disease phenotypes.IMPORTANCE Genetic variation among human rhinovirus types includes unexpected diversity in the genes encoding viral proteases (2Apro) that help these viruses achieve antihost responses. When the enzyme activities of 7 different 2Apro were measured comparatively in transformed cells programed with fluorescent reporter systems and by quantitative cell imaging, the cellular substrates, particularly in the nuclear pore complex, used by these proteases were indeed attacked at different rates and with different affinities. The importance of this finding is that it provides a mechanistic explanation for how different types (strains) of rhinoviruses may elicit different cell responses that directly or indirectly lead to distinct disease phenotypes.


Assuntos
Cisteína Endopeptidases/metabolismo , Interações Hospedeiro-Patógeno , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Rhinovirus/enzimologia , Rhinovirus/patogenicidade , Proteínas Virais/metabolismo , Células HeLa , Humanos , Microscopia de Fluorescência , Transporte Proteico
13.
Proc Natl Acad Sci U S A ; 112(17): 5485-90, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25848009

RESUMO

Members of rhinovirus C (RV-C) species are more likely to cause wheezing illnesses and asthma exacerbations compared with other rhinoviruses. The cellular receptor for these viruses was heretofore unknown. We report here that expression of human cadherin-related family member 3 (CDHR3) enables the cells normally unsusceptible to RV-C infection to support both virus binding and replication. A coding single nucleotide polymorphism (rs6967330, C529Y) was previously linked to greater cell-surface expression of CDHR3 protein, and an increased risk of wheezing illnesses and hospitalizations for childhood asthma. Compared with wild-type CDHR3, cells transfected with the CDHR3-Y529 variant had about 10-fold increases in RV-C binding and progeny yields. We developed a transduced HeLa cell line (HeLa-E8) stably expressing CDHR3-Y529 that supports RV-C propagation in vitro. Modeling of CDHR3 structure identified potential binding sites that could impact the virus surface in regions that are highly conserved among all RV-C types. Our findings identify that the asthma susceptibility gene product CDHR3 mediates RV-C entry into host cells, and suggest that rs6967330 mutation could be a risk factor for RV-C wheezing illnesses.


Assuntos
Caderinas , Proteínas de Membrana , Modelos Moleculares , Mutação Puntual , Polimorfismo de Nucleotídeo Único , Rhinovirus/fisiologia , Replicação Viral/fisiologia , Asma , Proteínas Relacionadas a Caderinas , Caderinas/química , Caderinas/genética , Caderinas/metabolismo , Células Cultivadas , Predisposição Genética para Doença , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Estrutura Terciária de Proteína , Fatores de Risco , Internalização do Vírus
14.
Respir Res ; 18(1): 84, 2017 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-28472984

RESUMO

BACKGROUND: The Rhinovirus C (RV-C), first identified in 2006, produce high symptom burdens in children and asthmatics, however, their primary target host cell in the airways remains unknown. Our primary hypotheses were that RV-C target ciliated airway epithelial cells (AECs), and that cell specificity is determined by restricted and high expression of the only known RV-C cell-entry factor, cadherin related family member 3 (CDHR3). METHODS: RV-C15 (C15) infection in differentiated human bronchial epithelial cell (HBEC) cultures was assessed using immunofluorescent and time-lapse epifluorescent imaging. Morphology of C15-infected differentiated AECs was assessed by immunohistochemistry. RESULTS: C15 produced a scattered pattern of infection, and infected cells were shed from the epithelium. The percentage of cells infected with C15 varied from 1.4 to 14.7% depending on cell culture conditions. Infected cells had increased staining for markers of ciliated cells (acetylated-alpha-tubulin [aat], p < 0.001) but not markers of goblet cells (wheat germ agglutinin or Muc5AC, p = ns). CDHR3 expression was increased on ciliated epithelial cells, but not other epithelial cells (p < 0.01). C15 infection caused a 27.4% reduction of ciliated cells expressing CDHR3 (p < 0.01). During differentiation of AECs, CDHR3 expression progressively increased and correlated with both RV-C binding and replication. CONCLUSIONS: The RV-C only replicate in ciliated AECs in vitro, leading to infected cell shedding. CDHR3 expression positively correlates with RV-C binding and replication, and is largely confined to ciliated AECs. Our data imply that factors regulating differentiation and CDHR3 production may be important determinants of RV-C illness severity.


Assuntos
Brônquios/citologia , Brônquios/virologia , Enterovirus/fisiologia , Células Epiteliais/citologia , Células Epiteliais/virologia , Internalização do Vírus , Replicação Viral/fisiologia , Células Cultivadas , Cílios/fisiologia , Cílios/ultraestrutura , Cílios/virologia , Enterovirus/ultraestrutura , Humanos , Eliminação de Partículas Virais/fisiologia
15.
Proc Natl Acad Sci U S A ; 111(44): 15792-7, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25331866

RESUMO

Cardiovirus Leader (L) proteins induce potent antihost inhibition of active cellular nucleocytoplasmic trafficking by triggering aberrant hyperphosphorylation of nuclear pore proteins (Nup). To achieve this, L binds protein RanGTPase (Ran), a key trafficking regulator, and diverts it into tertiary or quaternary complexes with required kinases. The activity of L is regulated by two phosphorylation events not required for Ran binding. Matched NMR studies on the unphosphorylated, singly, and doubly phosphorylated variants of Mengovirus L (L(M)) show both modifications act together to partially stabilize a short internal α-helix comprising L(M) residues 43-46. This motif implies that ionic and Van der Waals forces contributed by phosphorylation help organize downstream residues 48-67 into a new interface. The full structure of L(M) as bound to Ran (unlabeled) and Ran (216 aa) as bound by L(M) (unlabeled) places L(M) into the BP1 binding site of Ran, wrapped by the conformational flexible COOH tail. The arrangement explains the tight KD for this complex and places the LM zinc finger and phosphorylation interface as surface exposed and available for subsequent reactions. The core structure of Ran, outside the COOH tail, is not altered by L(M) binding and remains accessible for canonical RanGTP partner interactions. Pull-down assays identify at least one putative Ran:L(M) partner as an exportin, Crm1, or CAS. A model of Ran:L(M):Crm1, based on the new structures suggests LM phosphorylation status may mediate Ran's selection of exportin(s) and cargo(s), perverting these native trafficking elements into the lethal antihost Nup phosphorylation pathways.


Assuntos
Mengovirus/química , Complexos Multiproteicos/química , Proteínas Virais/química , Proteína ran de Ligação ao GTP/química , Sítios de Ligação , Mengovirus/genética , Mengovirus/metabolismo , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fosforilação , Estrutura Quaternária de Proteína , Proteínas Virais/genética , Proteínas Virais/metabolismo , Dedos de Zinco , Proteína ran de Ligação ao GTP/genética , Proteína ran de Ligação ao GTP/metabolismo
16.
J Virol ; 88(4): 2219-26, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24335301

RESUMO

Encephalomyocarditis virus and Theilovirus are species in the Cardiovirus genus of the Picornaviridae family. For all cardioviruses, the viral polyprotein is initiated with a short Leader (L) protein unique to this genus. The nuclear magnetic resonance (NMR) structure of LE from encephalomyocarditis virus (EMCV) has been determined. The protein has an NH2-proximal CHCC zinc finger, a central linker, and a contiguous, highly acidic motif. The theiloviruses encode the same domains, with one or two additional, COOH-proximal domains, characteristic of the human Saffold viruses (SafV) and Theiler's murine encephalomyelitis viruses (TMEV), respectively. The expression of a cardiovirus L, in recombinant form, or during infection/transfection, triggers an extensive, cell-dependent, antihost phosphorylation cascade, targeting nucleoporins (Nups) that form the hydrophobic core of nuclear pore complexes (NPC). The consequent inhibition of active nucleocytoplasmic trafficking is potent and prevents the host from mounting an effective antiviral response. For this inhibition, the L proteins themselves must be phosphorylated. In cells (extracts or recombinant form), LE was shown to be phosphorylated at Thr47 and Tyr41. The first reaction (Thr47), catalyzed by casein kinase 2 (CK2), is an obligatory precedent to the second event (Tyr41), catalyzed by spleen tyrosine kinase (Syk). Site mutations in LE, or kinase-specific inhibitors, prevented LE phosphorylation and subsequent Nup phosphorylation. Parallel experiments with LS (SafV-2) and LT (TMEV BeAn) proteins confirmed the general cardiovirus requirement for L phosphorylation, but CK2 was not the culpable kinase. It is likely that LS and LT are both activated by alternative kinases in different cell types, probably reactive within the Theilo-specific domains. IMPORTANCE An understanding of the diverse methods used by viruses to interfere with cellular processes is important because they can teach us how to control virus infections. This report shows how viruses in the same genus use different cellular enzymes to phosphorylate their proteins. If these processes are interfered with, the viruses are severely disabled.


Assuntos
Caseína Quinase II/metabolismo , Vírus da Encefalomiocardite/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Theilovirus/genética , Proteínas Virais/metabolismo , Sequência de Bases , Western Blotting , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fosforilação , Plasmídeos/genética , Alinhamento de Sequência , Análise de Sequência de DNA , Quinase Syk , Proteínas Virais/genética
17.
J Virol ; 88(22): 13503-9, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25210192

RESUMO

UNLABELLED: The leader (L) and 2A proteins of cardioviruses are the primary antihost agents produced during infection. For encephalomyocarditis virus (EMCV), the prototype of the genus Cardiovirus, these proteins interact independently with key cellular partners to bring about inhibition of active nucleocytoplasmic trafficking and cap-dependent translation, respectively. L and 2A also bind each other and require this cooperation to achieve their effects during infection. Recombinant L and 2A interact with 1:1 stoichiometry at a KD (equilibrium dissociation constant) of 1.5 µM. The mapped contact domains include the amino-proximal third of 2A (first 50 amino acids) and the central hinge region of L. This contact partially overlaps the L segment that makes subsequent contact with Ran GTPase in the nucleus, and Ran can displace 2A from L. The equivalent proteins from Theiler's murine encephalomyelitis virus (TMEV; BeAn) and Saffold virus interact similarly in any subtype combination, with various affinities. The data suggest a mechanism whereby L takes advantage of the nuclear localization signal in the COOH region of 2A to enhance its trafficking to the nucleus. Once there, it exchanges partners in favor of Ran. This required cooperation during infection explains many observed codependent phenotypes of L and 2A mutations. IMPORTANCE: Cardiovirus pathogenesis phenotypes vary dramatically, from asymptomatic, to mild gastrointestinal (GI) distress, to persistent demyelination and even encephalitic death. Leader and 2A are the primary viral determinants of pathogenesis, so understanding how these proteins cooperate to induce such a wide variety of outcomes for the host is of great important and interest to the field of virology, especially to those who use TMEV as a murine model for multiple sclerosis.


Assuntos
Vírus da Encefalomiocardite/fisiologia , Mapeamento de Interação de Proteínas , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Células HeLa , Humanos , Dados de Sequência Molecular , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Alinhamento de Sequência , Replicação Viral
18.
J Virol ; 87(11): 6517-20, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536659

RESUMO

The leader (L) protein of encephalomyocarditis virus (EMCV) shuts off host cell nucleocytoplasmic trafficking (NCT) by inducing hyperphosphorylation of nuclear pore proteins. This dramatic effect by a nonenzymatic protein of 6 kDa is not well understood but clearly involves L binding to cellular Ran GTPase, a critical factor of active NCT. Exogenous GDP and GTP are inhibitory to L-Ran binding, but the guanine-nucleotide exchange factor RCC1 can relieve this inhibition. In the presence of RCC1, L binds Ran with a KD (equilibrium dissociation constant) of ≈ 3 nM and reaches saturation within 20 min. The results of fluorescently tagged nucleotide experiments suggest that L-Ran interactions affect the nucleotide-binding pocket of Ran.


Assuntos
Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Proteínas de Ciclo Celular/metabolismo , Vírus da Encefalomiocardite/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Virais/metabolismo , Proteína ran de Ligação ao GTP/metabolismo , Infecções por Cardiovirus/genética , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Vírus da Encefalomiocardite/genética , Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/genética , Interações Hospedeiro-Patógeno , Humanos , Cinética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica , Proteínas Virais/química , Proteínas Virais/genética , Proteína ran de Ligação ao GTP/química , Proteína ran de Ligação ao GTP/genética
19.
J Virol ; 86(19): 10686-94, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22837200

RESUMO

Although picornavirus RNA genomes contain a 3'-terminal poly(A) tract that is critical for their replication, the impact of encephalomyocarditis virus (EMCV) infection on the host poly(A)-binding protein (PABP) remains unknown. Here, we establish that EMCV infection stimulates site-specific PABP proteolysis, resulting in accumulation of a 45-kDa N-terminal PABP fragment in virus-infected cells. Expression of a functional EMCV 3C proteinase was necessary and sufficient to stimulate PABP cleavage in uninfected cells, and bacterially expressed 3C cleaved recombinant PABP in vitro in the absence of any virus-encoded or eukaryotic cellular cofactors. N-terminal sequencing of the resulting C-terminal PABP fragment identified a 3C(pro) cleavage site on PABP between amino acids Q437 and G438, severing the C-terminal protein-interacting domain from the N-terminal RNA binding fragment. Single amino acid substitution mutants with changes at Q437 were resistant to 3C(pro) cleavage in vitro and in vivo, validating that this is the sole detectable PABP cleavage site. Finally, while ongoing protein synthesis was not detectably altered in EMCV-infected cells expressing a cleavage-resistant PABP variant, viral RNA synthesis and infectious virus production were both reduced. Together, these results establish that the EMCV 3C proteinase mediates site-specific PABP cleavage and demonstrate that PABP cleavage by 3C regulates EMCV replication.


Assuntos
Cisteína Endopeptidases/química , Vírus da Encefalomiocardite/enzimologia , Proteínas de Ligação a Poli(A)/metabolismo , Proteínas Virais/química , Proteases Virais 3C , Linhagem Celular , Escherichia coli/metabolismo , Fibroblastos/metabolismo , Regulação Viral da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Plasmídeos/metabolismo , Proteínas de Ligação a Poli(A)/química , Polirribossomos/metabolismo , Estrutura Terciária de Proteína , RNA Viral/metabolismo , Proteínas Recombinantes/metabolismo , Replicação Viral
20.
J Virol ; 85(20): 10874-83, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21835805

RESUMO

Human rhinoviruses (HRVs) from the HRV-A, HRV-B, and HRV-C species use encoded proteases, 2A(pro) and 3C(pro), to process their polyproteins and shut off host cell activities detrimental to virus replication. Reactions attributed to 2A(pro) include cleavage of eIF4G-I and -II to inhibit cellular mRNA translation and cleavage of select nucleoporin proteins (Nups) within nuclear pore complexes (NPCs) to disrupt karyopherin-dependent nuclear-cytoplasmic transport and signaling. Sequence diversity among 2A(pro) proteases from different HRV clades, even within species, suggested individual viruses might carry out these processes with unique mechanistic signatures. Six different recombinant 2A(pro) proteases (A16, A89, B04, B14, Cw12, and Cw24) were compared for their relative substrate preferences and cleavage kinetics using eIF4G from cellular extracts and Nups presented in native (NPC) or recombinant formats. The enzyme panel attacked these substrates with different rates or processing profiles, mimicking the preferences observed during natural infection (A16 and B14). For eIF4G, all 2A(pro) proteases cleaved at similar sites, but the comparative rates were species specific (HRV-A > HRV-C ≫ HRV-B). For Nup substrates, 5 of the 6 enzymes had unique product profiles (order of Nup selection) or reacted at different sites within Nup62, Nup98, and Nup153. Only A16 and A89 behaved similarly in most assays. Since each type of karyopherin receptor prefers particular Nups or uses a limited cohort of binding motifs within those Nups, the consequences of individual 2A(pro) avidities could profoundly affect relative viral replication levels, intracellular signaling, or extracellular signaling, all of which are underlying triggers for different host immune responses.


Assuntos
Cisteína Endopeptidases/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Rhinovirus/fisiologia , Proteínas Virais/metabolismo , Fator de Iniciação Eucariótico 4G/metabolismo , Células HeLa , Humanos , Cinética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA