Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Viruses ; 16(6)2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38932190

RESUMO

Human coronavirus 229E (HCoV-229E) is associated with upper respiratory tract infections and generally causes mild respiratory symptoms. HCoV-229E infection can cause cell death, but the molecular pathways that lead to virus-induced cell death as well as the interplay between viral proteins and cellular cell death effectors remain poorly characterized for HCoV-229E. Studying how HCoV-229E and other common cold coronaviruses interact with and affect cell death pathways may help to understand its pathogenesis and compare it to that of highly pathogenic coronaviruses. Here, we report that the main protease (Mpro) of HCoV-229E can cleave gasdermin D (GSDMD) at two different sites (Q29 and Q193) within its active N-terminal domain to generate fragments that are now unable to cause pyroptosis, a form of lytic cell death normally executed by this protein. Despite GSDMD cleavage by HCoV-229E Mpro, we show that HCoV-229E infection still leads to lytic cell death. We demonstrate that during virus infection caspase-3 cleaves and activates gasdermin E (GSDME), another key executioner of pyroptosis. Accordingly, GSDME knockout cells show a significant decrease in lytic cell death upon virus infection. Finally, we show that HCoV-229E infection leads to increased lytic cell death levels in cells expressing a GSDMD mutant uncleavable by Mpro (GSDMD Q29A+Q193A). We conclude that GSDMD is inactivated by Mpro during HCoV-229E infection, preventing GSDMD-mediated cell death, and point to the caspase-3/GSDME axis as an important player in the execution of virus-induced cell death. In the context of similar reported findings for highly pathogenic coronaviruses, our results suggest that these mechanisms do not contribute to differences in pathogenicity among coronaviruses. Nonetheless, understanding the interactions of common cold-associated coronaviruses and their proteins with the programmed cell death machineries may lead to new clues for coronavirus control strategies.


Assuntos
Morte Celular , Coronavirus Humano 229E , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Ligação a Fosfato , Piroptose , Humanos , Proteínas de Ligação a Fosfato/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Coronavirus Humano 229E/fisiologia , Coronavirus Humano 229E/genética , Infecções por Coronavirus/virologia , Infecções por Coronavirus/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Linhagem Celular , Interações Hospedeiro-Patógeno , Células HEK293 , Gasderminas
2.
Environ Pollut ; 347: 123700, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38452839

RESUMO

Emerging bio-contaminants (airborne viruses) exploits and manipulate host (human) metabolism to produce new viral particles, evading the host's immune defences and leading to infections. Non-thermal plasma, operating at atmospheric pressure and ambient temperature, is explored for virus inactivation, generating RONS that interact and denatures viral proteins. However, various factors affecting virus survival influence the efficacy of non-thermal plasma. Glucose analogue 2-DG, a metabolic modifier used in this study, disrupts the glycolysis pathway viruses rely on, creating an unfavourable environment for replication. Here, airborne HCoV-229E bio-contaminant was treated with plasma for inactivation, and the presence of RONS was analysed. Metabolically altered lung cells were subsequently exposed to the treated airborne viruses. Cytopathic effect, spike protein, and cell death were evaluated via flow cytometry and confocal microscopy, and CPRRs mediated antiviral gene expression was evaluated using PCR. Gas plasma-treated viruses led to reduced virus proliferation in unaltered lung cells, although few virus particles survived the exposure, as confirmed by biological assessment (cytopathic effects and live/dead staining). A combination approach of gas plasma-treated viruses and altered lung cells displayed drastic virus reduction compared to the control group, established through confocal microscopy and flow cytometry. Furthermore, altered lung cell enhances gene transcription responsible for innate immunity when exposed to the gas plasma-treated virus, thereby impeding airborne virus propagation. This study demonstrates the significance of a surface air gas plasma and metabolic alteration approach in enhancing genes targeted towards antiviral innate immunity and tackling outbreaks of emerging bio-contaminants of concerns (airborne viruses).


Assuntos
Coronavirus Humano 229E , Humanos , Coronavirus Humano 229E/genética , Inativação de Vírus , Pulmão , Imunidade Inata , Antivirais
3.
J Med Virol ; 95(1): e28101, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36031726

RESUMO

In 2019, an outbreak of pharyngoconjunctival fever (PCF) occurred at a swimming center in Zhejiang Province, China. A total of 97 (13.55%) of the 716 amateur swimmers had illnesses, with 24 patients (24.74%) hospitalized in the pediatric ward. Human adenovirus serotype 7 (HAdV-7) was isolated from one concentrated water from the swimming pool, and 20 of 97 positive cases without liver damage. This outbreak led to a nosocomial outbreak in the pediatric ward, in which 1 nurse had a fever and was confirmed to be adenovirus positive. The hexon, fiber, and penton genes from 20 outbreak cases, 1 water sample, and 1 nurse had 100% homology. Furthermore, 2 cases admitted to the pediatric ward, 2 parents, and 1 doctor were confirmed to be human coronaviruses (HCoV-229E) positive. Finally, all outbreak cases had fully recovered, regardless of a single infection (adenovirus or HCoV-229E) or coinfection of these two viruses simultaneously. Thus, PCF and acute respiratory disease outbreaks in Zhejiang were caused by the completely homologous type 7 adenovirus and HCoV-229E, respectively. The swimming pool water contaminated with HAdV-7 was most likely the source of the PCF outbreak, whereas nosocomial transmission might be the source of HCoV-229E outbreak.


Assuntos
Infecções por Adenovirus Humanos , Adenovírus Humanos , Coronavirus Humano 229E , Infecção Hospitalar , Infecções Respiratórias , Humanos , Criança , Coronavirus Humano 229E/genética , Adenovírus Humanos/genética , Infecções Respiratórias/epidemiologia , China/epidemiologia , Infecções por Adenovirus Humanos/epidemiologia , Água , Surtos de Doenças , Infecção Hospitalar/epidemiologia
4.
Cell Rep ; 41(4): 111540, 2022 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-36243002

RESUMO

The cellular fate after infection with human coronaviruses (HCoVs) is typically death. Previous data suggest, however, that the transcriptional state of an individual cell may sometimes allow additional outcomes of infection. Here, to probe the range of interactions a permissive cell type can have with a HCoV, we perform a CRISPR activation screen with HCoV-229E. The screen identified the transcription factor ZBTB7A, which strongly promotes cell survival after infection. Rather than suppressing viral infection, ZBTB7A upregulation allows the virus to induce a persistent infection and homeostatic state with the cell. We also find that control of oxidative stress is a primary driver of cellular survival during HCoV-229E infection. These data illustrate that, in addition to the nature of the infecting virus and the type of cell that it encounters, the cellular gene expression profile prior to infection can affect the eventual fate.


Assuntos
Coronavirus Humano 229E , Humanos , Coronavirus Humano 229E/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Fatores de Transcrição/genética , Homeostase
5.
Front Cell Infect Microbiol ; 12: 958634, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36211973

RESUMO

Rationale: Human coronaviruses (HCoVs) seriously affect human health by causing respiratory diseases ranging from common colds to severe acute respiratory diseases. Immunophilins, including peptidyl-prolyl isomerases of the FK506-binding protein (FKBP) and the cyclophilin family, are promising targets for pharmaceutical inhibition of coronavirus replication, but cell-type specific effects have not been elucidated. FKBPs and cyclophilins bind the immunosuppressive drugs FK506 and cyclosporine A (CsA), respectively. Methods: Primary human bronchial epithelial cells (phBECs) were treated with CsA, Alisporivir (ALV), FK506, and FK506-derived non-immunosuppressive analogs and infected with HCoV-229E. RNA and protein were assessed by RT-qPCR and immunoblot analysis. Treatment with the same compounds was performed in hepatoma cells (Huh-7.5) infected with HCoV-229E expressing Renilla luciferase (HCoV-229E-RLuc) and the kidney cell line HEK293 transfected with a SARS-CoV-1 replicon expressing Renilla luciferase (SARS-CoV-1-RLuc), followed by quantification of luminescence as a measure of viral replication. Results: Both CsA and ALV robustly inhibited viral replication in all models; both compounds decreased HCoV-229E RNA in phBECs and reduced luminescence in HCoV-229E-RLuc-infected Huh7.5 and SARS-CoV-1-RLuc replicon-transfected HEK293. In contrast, FK506 showed inconsistent and less pronounced effects in phBECs while strongly affecting coronavirus replication in Huh-7.5 and HEK293. Two non-immunosuppressive FK506 analogs had no antiviral effect in any infection model. Conclusion: The immunophilin inhibitors CsA and ALV display robust anti-coronaviral properties in multiple infection models, including phBECs, reflecting a primary site of HCoV infection. In contrast, FK506 displayed cell-type specific effects, strongly affecting CoV replication in Huh7.5 and HEK293, but inconsistently and less pronounced in phBECs.


Assuntos
Coronavirus Humano 229E , Infecções por Coronavirus , Coronavirus , Coronavirus/genética , Coronavirus Humano 229E/genética , Infecções por Coronavirus/genética , Ciclofilinas , Ciclosporina/química , Ciclosporina/farmacologia , Ciclosporina/uso terapêutico , Células HEK293 , Humanos , Imunossupressores/farmacologia , Luciferases de Renilla , Preparações Farmacêuticas , RNA , Tacrolimo/química , Tacrolimo/farmacologia , Tacrolimo/uso terapêutico , Proteínas de Ligação a Tacrolimo/farmacologia , Proteínas de Ligação a Tacrolimo/uso terapêutico
6.
EBioMedicine ; 81: 104132, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35779493

RESUMO

BACKGROUND: Human seasonal coronaviruses usually cause mild upper-respiratory tract infection, but severe complications can occur in specific populations. Research into seasonal coronaviruses is limited and robust experimental models are largely lacking. This study aims to establish human airway organoids (hAOs)-based systems for seasonal coronavirus infection and to demonstrate their applications in studying virus-host interactions and therapeutic development. METHODS: The infections of seasonal coronaviruses 229E, OC43 and NL63 in 3D cultured hAOs with undifferentiated or differentiated phenotypes were tested. The kinetics of virus replication and production was profiled at 33 °C and 37 °C. Genome-wide transcriptome analysis by RNA sequencing was performed in hAOs under various conditions. The antiviral activity of molnupiravir and remdesivir, two approved medications for treating COVID19, was tested. FINDINGS: HAOs efficiently support the replication and infectious virus production of seasonal coronaviruses 229E, OC43 and NL63. Interestingly, seasonal coronaviruses replicate much more efficiently at 33 °C compared to 37 °C, resulting in over 10-fold higher levels of viral replication. Genome-wide transcriptomic analyses revealed distinct patterns of infection-triggered host responses at 33 °C compared to 37 °C temperature. Treatment of molnupiravir and remdesivir dose-dependently inhibited the replication of 229E, OC43 and NL63 in hAOs. INTERPRETATION: HAOs are capable of modeling 229E, OC43 and NL63 infections. The intriguing finding that lower temperature resembling that in the upper respiratory tract favors viral replication may help to better understand the pathogenesis and transmissibility of seasonal coronaviruses. HAOs-based innovative models shall facilitate the research and therapeutic development against seasonal coronavirus infections. FUNDING: This research is supported by funding of a VIDI grant (No. 91719300) from the Netherlands Organization for Scientific Research and the Dutch Cancer Society Young Investigator Grant (10140) to Q.P., and the ZonMw COVID project (114025011) from the Netherlands Organization for Health Research and Development to R.R.


Assuntos
Tratamento Farmacológico da COVID-19 , Coronavirus Humano 229E , Infecções Respiratórias , Antivirais/farmacologia , Antivirais/uso terapêutico , Coronavirus Humano 229E/genética , Humanos , Organoides/patologia , Sistema Respiratório/patologia , Infecções Respiratórias/patologia , Estações do Ano
7.
Viruses ; 13(6)2021 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-34198852

RESUMO

Epithelial characteristics underlying the differential susceptibility of chronic asthma to SARS-CoV-2 (COVID-19) and other viral infections are currently unclear. By revisiting transcriptomic data from patients with Th2 low versus Th2 high asthma, as well as mild, moderate, and severe asthmatics, we characterized the changes in expression of human coronavirus and influenza viral entry genes relative to sex, airway location, and disease endotype. We found sexual dimorphism in the expression of SARS-CoV-2-related genes ACE2, TMPRSS2, TMPRSS4, and SLC6A19. ACE2 receptor downregulation occurred specifically in females in Th2 high asthma, while proteases broadly assisting coronavirus and influenza viral entry, TMPRSS2, and TMPRSS4, were highly upregulated in both sexes. Overall, changes in SARS-CoV-2-related gene expression were specific to the Th2 high molecular endotype of asthma and different by asthma severity and airway location. The downregulation of ACE2 (COVID-19, SARS) and ANPEP (HCoV-229E) viral receptors wascorrelated with loss of club and ciliated cells in Th2 high asthma. Meanwhile, the increase in DPP4 (MERS-CoV), ST3GAL4, and ST6GAL1 (influenza) was associated with increased goblet and basal activated cells. Overall, this study elucidates sex, airway location, disease endotype, and changes in epithelial heterogeneity as potential factors underlying asthmatic susceptibility, or lack thereof, to SARS-CoV-2.


Assuntos
Asma/imunologia , COVID-19/imunologia , Infecções por Coronavirus/imunologia , Células Epiteliais/virologia , Expressão Gênica , Interações entre Hospedeiro e Microrganismos , Influenza Humana/imunologia , Índice de Gravidade de Doença , Asma/genética , Asma/virologia , COVID-19/genética , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/imunologia , Infecções por Coronavirus/genética , Células Epiteliais/classificação , Feminino , Perfilação da Expressão Gênica , Interações entre Hospedeiro e Microrganismos/genética , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Influenza Humana/genética , Masculino , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Orthomyxoviridae/genética , Orthomyxoviridae/imunologia , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Caracteres Sexuais
8.
Nat Genet ; 53(4): 435-444, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33686287

RESUMO

The ongoing COVID-19 pandemic has caused a global economic and health crisis. To identify host factors essential for coronavirus infection, we performed genome-wide functional genetic screens with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human coronavirus 229E. These screens uncovered virus-specific as well as shared host factors, including TMEM41B and PI3K type 3. We discovered that SARS-CoV-2 requires the lysosomal protein TMEM106B to infect human cell lines and primary lung cells. TMEM106B overexpression enhanced SARS-CoV-2 infection as well as pseudovirus infection, suggesting a role in viral entry. Furthermore, single-cell RNA-sequencing of airway cells from patients with COVID-19 demonstrated that TMEM106B expression correlates with SARS-CoV-2 infection. The present study uncovered a collection of coronavirus host factors that may be exploited to develop drugs against SARS-CoV-2 infection or future zoonotic coronavirus outbreaks.


Assuntos
COVID-19/genética , Sistemas CRISPR-Cas , Genoma Humano/genética , Estudo de Associação Genômica Ampla/métodos , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Líquido da Lavagem Broncoalveolar/citologia , COVID-19/epidemiologia , COVID-19/virologia , Linhagem Celular Tumoral , Células Cultivadas , Coronavirus Humano 229E/genética , Epidemias , Células Epiteliais/virologia , Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Provírus/fisiologia , SARS-CoV-2/fisiologia , Internalização do Vírus
9.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33472860

RESUMO

RNA-dependent RNA polymerases (RdRps) of the Nidovirales (Coronaviridae, Arteriviridae, and 12 other families) are linked to an amino-terminal (N-terminal) domain, called NiRAN, in a nonstructural protein (nsp) that is released from polyprotein 1ab by the viral main protease (Mpro). Previously, self-GMPylation/UMPylation activities were reported for an arterivirus NiRAN-RdRp nsp and suggested to generate a transient state primed for transferring nucleoside monophosphate (NMP) to (currently unknown) viral and/or cellular biopolymers. Here, we show that the coronavirus (human coronavirus [HCoV]-229E and severe acute respiratory syndrome coronavirus 2) nsp12 (NiRAN-RdRp) has Mn2+-dependent NMPylation activity that catalyzes the transfer of a single NMP to the cognate nsp9 by forming a phosphoramidate bond with the primary amine at the nsp9 N terminus (N3825) following Mpro-mediated proteolytic release of nsp9 from N-terminally flanking nsps. Uridine triphosphate was the preferred nucleotide in this reaction, but also adenosine triphosphate, guanosine triphosphate, and cytidine triphosphate were suitable cosubstrates. Mutational studies using recombinant coronavirus nsp9 and nsp12 proteins and genetically engineered HCoV-229E mutants identified residues essential for NiRAN-mediated nsp9 NMPylation and virus replication in cell culture. The data corroborate predictions on NiRAN active-site residues and establish an essential role for the nsp9 N3826 residue in both nsp9 NMPylation in vitro and virus replication. This residue is part of a conserved N-terminal NNE tripeptide sequence and shown to be the only invariant residue in nsp9 and its homologs in viruses of the family Coronaviridae The study provides a solid basis for functional studies of other nidovirus NMPylation activities and suggests a possible target for antiviral drug development.


Assuntos
Coronavirus Humano 229E/genética , Proteínas de Ligação a RNA/metabolismo , SARS-CoV-2/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Sequência de Aminoácidos , Substituição de Aminoácidos , Asparagina/genética , Linhagem Celular , Sequência Conservada , Coronavirus Humano 229E/fisiologia , RNA-Polimerase RNA-Dependente de Coronavírus/genética , RNA-Polimerase RNA-Dependente de Coronavírus/metabolismo , Humanos , Manganês/metabolismo , Domínios Proteicos , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transcrição Gênica , Proteínas não Estruturais Virais/genética
10.
Infect Genet Evol ; 84: 104440, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32622082

RESUMO

SARS-CoV-2, a new coronavirus strain responsible for COVID-19, has emerged in Wuhan City, China, and continuing its global pandemic nature. The availability of the complete gene sequences of the virus helps to know about the origin and molecular characteristics of this virus. In the present study, we performed bioinformatic analysis of the available gene sequence data of SARS-CoV-2 for the understanding of evolution and molecular characteristics and immunogenic resemblance of the circulating viruses. Phylogenetic analysis was performed for four types of representative viral proteins (spike, membrane, envelope and nucleoprotein) of SARS-CoV-2, HCoV-229E, HCoV-OC43, SARS-CoV, HCoV-NL63, HKU1, MERS-CoV, HKU4, HKU5 and BufCoV-HKU26. The findings demonstrated that SARS-CoV-2 exhibited convergent evolutionary relation with previously reported SARS-CoV. It was also depicted that SARS-CoV-2 proteins were highly similar and identical to SARS-CoV proteins, though proteins from other coronaviruses showed a lower level of resemblance. The cross-checked conservancy analysis of SARS-CoV-2 antigenic epitopes showed significant conservancy with antigenic epitopes derived from SARS-CoV. Descriptive epidemiological analysis on several epidemiological indices was performed on available epidemiological outbreak information from several open databases on COVID-19 (SARS-CoV-2). Satellite-derived imaging data have been employed to understand the role of temperature in the environmental persistence of the virus. Findings of the descriptive analysis were used to describe the global impact of newly emerged SARS-CoV-2, and the risk of an epidemic in Bangladesh.


Assuntos
Antígenos Virais/genética , Betacoronavirus/genética , Infecções por Coronavirus/epidemiologia , Genoma Viral , Pandemias , Pneumonia Viral/epidemiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Glicoproteína da Espícula de Coronavírus/química , Alphacoronavirus/classificação , Alphacoronavirus/genética , Alphacoronavirus/metabolismo , Sequência de Aminoácidos , Animais , Antígenos Virais/química , Antígenos Virais/metabolismo , Bangladesh/epidemiologia , Sequência de Bases , Betacoronavirus/classificação , Betacoronavirus/metabolismo , Sítios de Ligação , COVID-19 , Quirópteros/virologia , Biologia Computacional , Coronavirus Humano 229E/classificação , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/metabolismo , Infecções por Coronavirus/virologia , Coronavirus Humano NL63/classificação , Coronavirus Humano NL63/genética , Coronavirus Humano NL63/metabolismo , Coronavirus Humano OC43/classificação , Coronavirus Humano OC43/genética , Coronavirus Humano OC43/metabolismo , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/classificação , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/metabolismo , Modelos Moleculares , Mutação , Nucleoproteínas/química , Nucleoproteínas/genética , Nucleoproteínas/metabolismo , Filogenia , Pneumonia Viral/virologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , SARS-CoV-2 , Alinhamento de Sequência , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo
11.
Antiviral Res ; 173: 104620, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31634494

RESUMO

The well-known immunosuppressive drug cyclosporin A inhibits replication of various viruses including coronaviruses by binding to cellular cyclophilins thus inactivating their cis-trans peptidyl-prolyl isomerase function. Viral nucleocapsid proteins are inevitable for genome encapsidation and replication. Here we demonstrate the interaction between the N protein of HCoV-229E and cyclophilin A, not cyclophilin B. Cyclophilin inhibitors abolish this interaction. Upon infection, cyclophilin A stays evenly distributed throughout the cell, whereas cyclophilin B concentrates at ER-bleb-like structures. We further show the inhibitory potential of non-immunosuppressive CsA derivatives Alisporivir, NIM811, compound 3 on HCoV-229E-GFP and -Luciferase replication in human Huh-7.5 hepatoma cells at 18 and 48 h time points post infection with EC50 s at low micromolar ranges. Thus, non-immunosuppressive CsA derivatives effectively inhibit HCoV-229E replication suggesting them as possible candidates for the treatment of HCoV infection. The interruption of interaction between CypA and N protein by CsA and its derivatives suggest a mechanism how CypA inhibitors suppress viral replication.


Assuntos
Coronavirus Humano 229E/fisiologia , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Ciclofilina A/metabolismo , Ciclofilinas/metabolismo , Ciclosporina/farmacologia , Proteínas do Nucleocapsídeo/metabolismo , Coronavirus Humano 229E/efeitos dos fármacos , Coronavirus Humano 229E/genética , Infecções por Coronavirus/genética , Proteínas do Nucleocapsídeo de Coronavírus , Ciclofilina A/genética , Ciclofilinas/genética , Ciclosporina/química , Interações Hospedeiro-Patógeno , Humanos , Proteínas do Nucleocapsídeo/genética , Ligação Proteica/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
Viruses ; 11(8)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31412613

RESUMO

Primary human airway epithelial cell (hAEC) cultures represent a universal platform to propagate respiratory viruses and characterize their host interactions in authentic target cells. To further elucidate specific interactions between human respiratory viruses and important host factors in the airway epithelium, it is important to make hAEC cultures amenable to genetic modification. However, the short and finite lifespan of primary cells in cell culture creates a bottleneck for the genetic modification of these cultures. In the current study, we show that the incorporation of the Rho-associated protein kinase (ROCK) inhibitor (Y-27632) during cell propagation extends the life span of primary human cells in vitro and thereby facilitates the incorporation of lentivirus-based expression systems. Using fluorescent reporters for fluorescence-activated cell sorting (FACS)-based sorting, we generated homogenously fluorescent hAEC cultures that differentiate normally after lentiviral transduction. As a proof-of-principle, we demonstrate that host gene expression can be modulated post-differentiation via inducible short hairpin (sh)RNA-mediated knockdown. Importantly, functional characterization of these transgenic hAEC cultures with exogenous poly (I:C), as a proxy for virus infection, demonstrates that such modifications do not influence the host innate immune response. Moreover, the propagation kinetics of both human coronavirus 229E (HCoV-229E) and human respiratory syncytial virus (hRSV) were not affected. Combined, these results validate our newly established protocol for the genetic modification of hAEC cultures, thereby unlocking a unique potential for detailed molecular characterization of virus-host interactions in human respiratory epithelium.


Assuntos
Coronavirus Humano 229E/fisiologia , Infecções por Coronavirus/virologia , Células Epiteliais/virologia , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sincicial Respiratório Humano/fisiologia , Linhagem Celular , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/crescimento & desenvolvimento , Interações Hospedeiro-Patógeno , Humanos , Cultura Primária de Células , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/crescimento & desenvolvimento , Cultura de Vírus
13.
J Gen Virol ; 99(7): 908-912, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29786498

RESUMO

Human coronavirus 229E (HCoV-229E) is responsible for common colds. Like other coronaviruses, HCoV-229E exploits cellular proteases to activate fusion mediated by the spike protein. We analysed the proteolytic processing of the HCoV-229E spike protein by trypsin-like serine proteases leading to activation of the fusion process. Unlike in other coronaviruses, HCoV-229E fusion activation appears to be a one-step process. Indeed, cleavage of the S1/S2 interface does not seem to be a prerequisite, and the fusion activation is highly reliant on the S2' region, with arginine residue 683 acting as the recognition site.


Assuntos
Coronavirus Humano 229E/genética , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Proteínas do Core Viral/genética , Proteínas Virais de Fusão/química , Coronavirus Humano 229E/química , Infecções por Coronavirus , Células HEK293 , Humanos , Glicoproteína da Espícula de Coronavírus/genética , Proteínas Virais de Fusão/genética
14.
Biochem Biophys Res Commun ; 497(2): 705-712, 2018 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-29458023

RESUMO

HCoV-229E spike (S) protein mediates virion attachment to cells and subsequent fusion of the viral and cellular membranes. This protein is composed of an N-terminal receptor-binding domain (S1) and a C-terminal trans-membrane fusion domain (S2). S2 contains a highly conserved heptad repeat 1 and 2 (HR1 and HR2). In this study, the HRs sequences were designed and connected with a flexible linker. The recombinant fusion core protein was crystallized and its structure was solved at a resolution of 2.45 Å. Then we characterized the binding of HR1s and HR2s via both sequence alignment and structural analysis. The overall structures, especially the residues in some positions of HR2 are highly conserved. Fourteen hydrophobic and three polar residues from each HR1 peptide are packed in layers at the coiled-coil interface. These core amino acids can be grouped into seven heptad repeats. Analysis of hydrophobic and hydrophilic interactions between HR2 helix and HR1 helices, shows that the HR1 and HR2 polypeptides are highly complementary in both shape and chemical properties. Furthermore, the available knowledge concerning HCoV-229E fusion core may make it possible to design small molecule or polypeptide drugs targeting membrane fusion, a crucial step of HCoV-229E infection.


Assuntos
Coronavirus Humano 229E/química , Infecções por Coronavirus/virologia , Glicoproteína da Espícula de Coronavírus/química , Sequência de Aminoácidos , Clonagem Molecular , Coronavirus Humano 229E/genética , Cristalografia por Raios X , Humanos , Modelos Moleculares , Conformação Proteica , Alinhamento de Sequência , Glicoproteína da Espícula de Coronavírus/genética
15.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27733646

RESUMO

Human coronavirus 229E (HCoV-229E), a causative agent of the common cold, enters host cells via two distinct pathways: one is mediated by cell surface proteases, particularly transmembrane protease serine 2 (TMPRSS2), and the other by endosomal cathepsin L. Thus, specific inhibitors of these proteases block virus infection. However, it is unclear which of these pathways is actually utilized by HCoV-229E in the human respiratory tract. Here, we examined the mechanism of cell entry used by a pseudotyped virus bearing the HCoV-229E spike (S) protein in the presence or absence of protease inhibitors. We found that, compared with a laboratory strain isolated in 1966 and passaged for a half century, clinical isolates of HCoV-229E were less likely to utilize cathepsin L; rather, they showed a preference for TMPRSS2. Two amino acid substitutions (R642M and N714K) in the S protein of HCoV-229E clinical isolates altered their sensitivity to a cathepsin L inhibitor, suggesting that these amino acids were responsible for cathepsin L use. After 20 passages in HeLa cells, the ability of the isolate to use cathepsin increased so that it was equal to that of the laboratory strain; this increase was caused by an amino acid substitution (I577S) in the S protein. The passaged virus showed a reduced ability to replicate in differentiated airway epithelial cells cultured at an air-liquid interface. These results suggest that the endosomal pathway is disadvantageous for HCoV-229E infection of human airway epithelial cells; therefore, clinical isolates are less able to use cathepsin. IMPORTANCE: Many enveloped viruses enter cells through endocytosis. Viral spike proteins drive the fusion of viral and endosomal membranes to facilitate insertion of the viral genome into the cytoplasm. Human coronavirus 229E (HCoV-229E) utilizes endosomal cathepsin L to activate the spike protein after receptor binding. Here, we found that clinical isolates of HCoV-229E preferentially utilize the cell surface protease TMPRSS2 rather than endosomal cathepsin L. The endosome is a main site of Toll-like receptor recognition, which then triggers an innate immune response; therefore, HCoV-229E presumably evolved to bypass the endosome by entering the cell via TMPRSS2. Thus, the virus uses a simple mechanism to evade the host innate immune system. Therefore, therapeutic agents for coronavirus-mediated diseases, such as severe acute respiratory syndrome (SARS) and Middle East respiratory syndrome (MERS), should target cell surface TMPRSS2 rather than endosomal cathepsin.


Assuntos
Catepsina L/genética , Membrana Celular/virologia , Coronavirus Humano 229E/genética , Evasão da Resposta Imune , Serina Endopeptidases/genética , Glicoproteína da Espícula de Coronavírus/genética , Internalização do Vírus , Sequência de Aminoácidos , Substituição de Aminoácidos , Evolução Biológica , Catepsina L/antagonistas & inibidores , Catepsina L/imunologia , Membrana Celular/imunologia , Resfriado Comum/imunologia , Resfriado Comum/virologia , Coronavirus Humano 229E/metabolismo , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Endocitose , Endossomos/efeitos dos fármacos , Endossomos/imunologia , Endossomos/virologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Células HeLa , Humanos , Mutação , Inibidores de Proteases/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Alinhamento de Sequência , Serina Endopeptidases/imunologia , Glicoproteína da Espícula de Coronavírus/metabolismo
16.
Nucleic Acids Res ; 44(6): e52, 2016 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-26657632

RESUMO

Stable isotope labeling is central to NMR studies of nucleic acids. Development of methods that incorporate labels at specific atomic positions within each nucleotide promises to expand the size range of RNAs that can be studied by NMR. Using recombinantly expressed enzymes and chemically synthesized ribose and nucleobase, we have developed an inexpensive, rapid chemo-enzymatic method to label ATP and GTP site specifically and in high yields of up to 90%. We incorporated these nucleotides into RNAs with sizes ranging from 27 to 59 nucleotides using in vitro transcription: A-Site (27 nt), the iron responsive elements (29 nt), a fluoride riboswitch from Bacillus anthracis(48 nt), and a frame-shifting element from a human corona virus (59 nt). Finally, we showcase the improvement in spectral quality arising from reduced crowding and narrowed linewidths, and accurate analysis of NMR relaxation dispersion (CPMG) and TROSY-based CEST experiments to measure µs-ms time scale motions, and an improved NOESY strategy for resonance assignment. Applications of this selective labeling technology promises to reduce difficulties associated with chemical shift overlap and rapid signal decay that have made it challenging to study the structure and dynamics of large RNAs beyond the 50 nt median size found in the PDB.


Assuntos
Trifosfato de Adenosina/síntese química , Guanosina Trifosfato/síntese química , Marcação por Isótopo/métodos , Nucleotídeos/síntese química , Bacillus anthracis/química , Bacillus anthracis/genética , Isótopos de Carbono , Coronavirus Humano 229E/química , Coronavirus Humano 229E/genética , Creatina Quinase/química , Creatina Quinase/genética , Espectroscopia de Ressonância Magnética , Pentosiltransferases/química , Pentosiltransferases/genética , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Elementos de Resposta , Ribose/química , Ribose-Fosfato Pirofosfoquinase/química , Ribose-Fosfato Pirofosfoquinase/genética , Riboswitch , Transcrição Gênica
17.
Euro Surveill ; 17(39)2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-23041020

RESUMO

We present two real-time reverse-transcription polymerase chain reaction assays for a novel human coronavirus (CoV), targeting regions upstream of the E gene (upE) or within open reading frame (ORF)1b, respectively. Sensitivity for upE is 3.4 copies per reaction (95% confidence interval (CI): 2.5­6.9 copies) or 291 copies/mL of sample. No cross-reactivity was observed with coronaviruses OC43, NL63, 229E, SARS-CoV, nor with 92 clinical specimens containing common human respiratory viruses. We recommend using upE for screening and ORF1b for confirmation.


Assuntos
Infecções por Coronavirus/virologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/isolamento & purificação , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/genética , Coronavirus Humano NL63/genética , Coronavirus Humano NL63/isolamento & purificação , Coronavirus Humano OC43/genética , Coronavirus Humano OC43/isolamento & purificação , Humanos , Fases de Leitura Aberta , Arábia Saudita , Sensibilidade e Especificidade , Viagem , Proteínas do Envelope Viral , Proteínas Viroporinas
18.
Virus Genes ; 45(3): 433-9, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22926811

RESUMO

Human coronavirus 229E has been identified in the mid-1960s, yet still only one full-genome sequence is available. This full-length sequence has been determined from the cDNA-clone Inf-1 that is based on the lab-adapted strain VR-740. Lab-adaptation might have resulted in genomic changes, due to insufficient pressure to maintain gene integrity of non-essential genes. We present here the first full-length genome sequence of two clinical isolates. Each encoded gene was compared to Inf-1. In general, little sequence changes were noted, most could be attributed to genetic drift, since the clinical isolates originate from 2009 to 2010 and VR740 from 1962. Hot spots of substitutions were situated in the S1 region of the Spike, the nucleocapsid gene, and the non-structural protein 3 gene, whereas several deletions were detected in the 3'UTR. Most notable was the difference in genome organization: instead of an ORF4A and ORF4B, an intact ORF4 was present in clinical isolates.


Assuntos
Coronavirus Humano 229E/genética , Evolução Molecular , Genoma Viral , Glicoproteínas de Membrana/genética , Proteínas do Envelope Viral/genética , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Sequência de Bases , Células Cultivadas , Coronavirus Humano 229E/classificação , Coronavirus Humano 229E/isolamento & purificação , Infecções por Coronavirus/virologia , Proteínas do Nucleocapsídeo de Coronavírus , Deriva Genética , Humanos , Proteínas do Nucleocapsídeo/genética , Fases de Leitura Aberta , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Glicoproteína da Espícula de Coronavírus
19.
J Gen Virol ; 93(Pt 9): 1908-1917, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22673931

RESUMO

Human coronavirus (HCoV) is a causative agent of the common cold. Although HCoV is highly prevalent in the world, studies of the genomic and antigenic details of circulating HCoV strains have been limited. In this study, we compared four Japanese isolates with the standard HCoV-229E strain obtained from ATCC (ATCC-VR740) by focusing on the spike (S) protein, a major determinant of neutralizing antigen and pathogenicity. The isolates were found to have nucleotide deletions and a number of sequence differences in the S1 region of the S protein. We compared two of the Japanese isolates with the ATCC-VR740 strain by using virus-neutralizing assays consisting of infectious HCoV-229E particles and vesicular stomatitis virus (VSV)-pseudotyped virus carrying the HCoV-229E S protein. The two clinical isolates (Sendai-H/1121/04 and Niigata/01/08) did not react with antiserum to the ATCC-VR740 strain via the neutralizing test. We then constructed a pseudotype VSV-harboured chimeric S protein with the ATCC S1 and Sendai S2 regions or that with Sendai S1 and ATCC S2 regions and compared them by a neutralization test. The results revealed that the difference in the neutralizing antigenicity depends on the S1 region. This different antigenic phenotype was also confirmed by a neutralizing test with clinically isolated human sera. These results suggest that the HCoV-229E viruses prevalent in Japan are quite different from the laboratory strain ATCC-VR740 in terms of the S sequence and neutralization antigenicity, which is attributed to the difference in the S1 region.


Assuntos
Coronavirus Humano 229E/classificação , Coronavirus Humano 229E/genética , Infecções por Coronavirus/virologia , Glicoproteínas de Membrana/genética , Proteínas do Envelope Viral/genética , Adulto , Motivos de Aminoácidos , Anticorpos Antivirais/imunologia , Linhagem Celular , Coronavirus Humano 229E/imunologia , Coronavirus Humano 229E/isolamento & purificação , Infecções por Coronavirus/imunologia , Feminino , Humanos , Japão , Masculino , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/imunologia , Pessoa de Meia-Idade , Testes de Neutralização , Filogenia , Deleção de Sequência , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Adulto Jovem
20.
Protein Sci ; 18(1): 6-16, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19177346

RESUMO

The polyproteins of coronaviruses are cleaved by viral proteases into at least 15 nonstructural proteins (Nsps). Consisting of five domains, Nsp3 is the largest of these (180-210 kDa). Among these domains, the so-called X-domain is believed to act as ADP-ribose-1''-phosphate phosphatase or to bind poly(ADP-ribose). However, here we show that the X-domain of Infectious Bronchitis Virus (strain Beaudette), a Group-3 coronavirus, fails to bind ADP-ribose. This is explained on the basis of the crystal structure of the protein, determined at two different pH values. For comparison, we also describe the crystal structure of the homologous X-domain from Human Coronavirus 229E, a Group-1 coronavirus, which does bind ADP-ribose.


Assuntos
Adenosina Difosfato Ribose/metabolismo , Coronavirus Humano 229E/química , Vírus da Bronquite Infecciosa/química , Proteínas não Estruturais Virais/química , Adenosina Difosfato Ribose/análogos & derivados , Adenosina Difosfato Ribose/química , Sequência de Aminoácidos , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/metabolismo , Cristalografia por Raios X , Vírus da Bronquite Infecciosa/genética , Vírus da Bronquite Infecciosa/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Monoéster Fosfórico Hidrolases/química , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Ligação Proteica/fisiologia , Conformação Proteica , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Alinhamento de Sequência , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA