Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros











Intervalo de año de publicación
1.
Sheng Wu Gong Cheng Xue Bao ; 39(7): 2669-2683, 2023 Jul 25.
Artículo en Chino | MEDLINE | ID: mdl-37584123

RESUMEN

The goal of this study was to investigate the regulatory effect of angiotensin converting enzyme 2 (ACE2) on cellular inflammation caused by avian infectious bronchitis virus (IBV) and the underlying mechanism of such effect. Vero and DF-1 cells were used as test target to be exposed to recombinant IBV virus (IBV-3ab-Luc). Four different groups were tested: the control group, the infection group[IBV-3ab-Luc, MOI (multiplicity of infection)=1], the ACE2 overexpression group[IBV-3ab Luc+pcDNA3.1(+)-ACE2], and the ACE2-depleted group (IBV-3ab-Luc+siRNA-ACE2). After the cells in the infection group started to show cytopathic indicators, the overall protein and RNA in cell of each group were extracted. real-time quantitative polymerase chain reaction (RT-qPCR) was used to determine the mRNA expression level of the IBV nucleoprotein (IBV-N), glycoprotein 130 (gp130) and cellular interleukin-6 (IL-6). Enzyme linked immunosorbent assay (ELISA) was used to determine the level of IL-6 in cell supernatant. Western blotting was performed to determine the level of ACE2 phosphorylation of janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). We found that ACE2 was successfully overexpressed and depleted in both Vero and DF-1 cells. Secondly, cytopathic indicators were observed in infected Vero cells including rounding, detaching, clumping, and formation of syncytia. These indicators were alleviated in ACE2 overexpression group but exacerbated when ACE2 was depleted. Thirdly, in the infection group, capering with the control group, the expression level of IBV-N, gp130, IL-6 mRNA and increased significantly (P < 0.05), the IL-6 level was significant or extremely significant elevated in cell supernatant (P < 0.05 or P < 0.01); the expression of ACE2 decreased significantly (P < 0.05); protein phosphorylation level of JAK2 and STAT3 increased significantly (P < 0.05). Fourthly, comparing with the infected group, the level of IBV-N mRNA expression in the ACE2 overexpression group had no notable change (P > 0.05), but the expression of gp130 mRNA, IL-6 level and expression of mRNA were elevated (P < 0.05) and the protein phosphorylation level of JAK2 and STAT3 decreased significantly (P < 0.05). In the ACE2-depleted group, there was no notable change in IBV-N (P > 0.05), but the IL-6 level and expression of mRNA increased significantly (P < 0.05) and the phosphorylation level of JAK2 and STAT3 protein decreased slightly (P > 0.05). The results demonstrated for the first time that ACE2 did not affect the replication of IBV in DF-1 cell, but it did contribute to the prevention of the activation of the IL-6/JAK2/STAT3 signaling pathway, resulting in an alleviation of IBV-induced cellular inflammation in Vero and DF-1 cells.


Asunto(s)
Virus de la Bronquitis Infecciosa , Interleucina-6 , Animales , Chlorocebus aethiops , Humanos , Interleucina-6/genética , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Janus Quinasa 2/farmacología , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/farmacología , Receptor gp130 de Citocinas/metabolismo , Células Vero , Transducción de Señal , Inflamación , ARN Mensajero
2.
Int J Mol Sci ; 23(19)2022 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-36232993

RESUMEN

Coronavirus nonstructural protein 3 (nsp3) is a multi-functional protein, playing a critical role in viral replication and in regulating host antiviral innate immunity. In this study, we demonstrate that nsp3 from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and avian coronavirus infectious bronchitis virus (IBV) directly interacts with melanoma differentiation-associated gene 5 (MDA5), rendering an inhibitory effect on the MDA5-mediated type I interferon (IFN) response. By the co-expression of MDA5 with wild-type and truncated nsp3 constructs, at least three interacting regions mapped to the papain-like protease (PLpro) domain and two other domains located at the N- and C-terminal regions were identified in SARS-CoV-2 nsp3. Furthermore, by introducing point mutations to the catalytic triad, the deubiquitylation activity of the PLpro domain from both SARS-CoV-2 and IBV nsp3 was shown to be responsible for the suppression of the MDA5-mediated type I IFN response. It was also demonstrated that both MDA5 and nsp3 were able to interact with ubiquitin and ubiquitinated proteins, contributing to the interaction between the two proteins. This study confirms the antagonistic role of nsp3 in the MDA5-mediated type I IFN signaling, highlighting the complex interaction between a multi-functional viral protein and the innate immune response.


Asunto(s)
Infecciones por Coronavirus , Virus de la Bronquitis Infecciosa , Interferón Tipo I , Helicasa Inducida por Interferón IFIH1 , SARS-CoV-2 , Proteínas no Estructurales Virales , COVID-19 , Infecciones por Coronavirus/inmunología , Humanos , Virus de la Bronquitis Infecciosa/metabolismo , Interferón Tipo I/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , SARS-CoV-2/metabolismo , Ubiquitina/metabolismo , Proteínas Ubiquitinadas , Proteínas no Estructurales Virales/metabolismo
3.
J Virol ; 96(5): e0208621, 2022 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-34985993

RESUMEN

Coronavirus infections induce the expression of multiple proinflammatory cytokines and chemokines. We have previously shown that in cells infected with gammacoronavirus infectious bronchitis virus (IBV), interleukin 6 (IL-6), and IL-8 were drastically upregulated, and the MAP kinase p38 and the integrated stress response pathways were implicated in this process. In this study, we report that coronavirus infection activates a negative regulatory loop that restricts the upregulation of a number of proinflammatory genes. As revealed by the initial transcriptomic and subsequent validation analyses, the anti-inflammatory adenine-uridine (AU)-rich element (ARE)-binding protein, zinc finger protein 36 (ZFP36), and its related family members were upregulated in cells infected with IBV and three other coronaviruses, alphacoronaviruses porcine epidemic diarrhea virus (PEDV), human coronavirus 229E (HCoV-229E), and betacoronavirus HCoV-OC43, respectively. Characterization of the functional roles of ZFP36 during IBV infection demonstrated that ZFP36 promoted the degradation of transcripts coding for IL-6, IL-8, dual-specificity phosphatase 1 (DUSP1), prostaglandin-endoperoxide synthase 2 (PTGS2) and TNF-α-induced protein 3 (TNFAIP3), through binding to AREs in these transcripts. Consistently, knockdown and inhibition of JNK and p38 kinase activities reduced the expression of ZFP36, as well as the expression of IL-6 and IL-8. On the contrary, overexpression of mitogen-activated protein kinase kinase 3 (MKK3) and MAPKAP kinase-2 (MK2), the upstream and downstream kinases of p38, respectively, increased the expression of ZFP36 and decreased the expression of IL-8. Taken together, this study reveals an important regulatory role of the MKK3-p38-MK2-ZFP36 axis in coronavirus infection-induced proinflammatory response. IMPORTANCE Excessive and uncontrolled induction and release of proinflammatory cytokines and chemokines, the so-called cytokine release syndrome (CRS), would cause life-threatening complications and multiple organ failure in severe coronavirus infections, including severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and COVID-19. This study reveals that coronavirus infection also induces the expression of ZFP36, an anti-inflammatory ARE-binding protein, promoting the degradation of ARE-containing transcripts coding for IL-6 and IL-8 as well as a number of other proteins related to inflammatory response. Furthermore, the p38 MAP kinase, its upstream kinase MKK3 and downstream kinase MK2 were shown to play a regulatory role in upregulation of ZFP36 during coronavirus infection cycles. This MKK3-p38-MK2-ZFP36 axis would constitute a potential therapeutic target for severe coronavirus infections.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Tristetraprolina/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Adenina/metabolismo , Animales , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/genética , Regulación de la Expresión Génica , Humanos , Virus de la Bronquitis Infecciosa/metabolismo , Virus de la Bronquitis Infecciosa/patogenicidad , Interleucina-6/genética , Interleucina-8/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Activación Transcripcional , Regulación hacia Arriba , Uridina/metabolismo , Células Vero
4.
Int J Mol Sci ; 22(11)2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34073283

RESUMEN

Infection induces the production of proinflammatory cytokines and chemokines such as interleukin-8 (IL-8) and IL-6. Although they facilitate local antiviral immunity, their excessive release leads to life-threatening cytokine release syndrome, exemplified by the severe cases of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In this study, we investigated the roles of the integrated stress response (ISR) and activator protein-1 (AP-1) family proteins in regulating coronavirus-induced IL-8 and IL-6 upregulation. The mRNA expression of IL-8 and IL-6 was significantly induced in cells infected with infectious bronchitis virus (IBV), a gammacoronavirus, and porcine epidemic diarrhea virus, an alphacoronavirus. Overexpression of a constitutively active phosphomimetic mutant of eukaryotic translation initiation factor 2α (eIF2α), chemical inhibition of its dephosphorylation, or overexpression of its upstream double-stranded RNA-dependent protein kinase (PKR) significantly enhanced IL-8 mRNA expression in IBV-infected cells. Overexpression of the AP-1 protein cJUN or its upstream kinase also increased the IBV-induced IL-8 mRNA expression, which was synergistically enhanced by overexpression of cFOS. Taken together, this study demonstrated the important regulatory roles of ISR and AP-1 proteins in IL-8 production during coronavirus infection, highlighting the complex interactions between cellular stress pathways and the innate immune response.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Estrés del Retículo Endoplásmico/genética , Factor 2 Eucariótico de Iniciación/metabolismo , Interleucina-8/metabolismo , Respuesta de Proteína Desplegada/genética , Alphacoronavirus/metabolismo , Alphacoronavirus/patogenicidad , Animales , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/genética , Gammacoronavirus/metabolismo , Gammacoronavirus/patogenicidad , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Virus de la Bronquitis Infecciosa/metabolismo , Virus de la Bronquitis Infecciosa/patogenicidad , Interleucina-8/genética , Fosforilación , Virus de la Diarrea Epidémica Porcina/metabolismo , Virus de la Diarrea Epidémica Porcina/patogenicidad , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/genética , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Regulación hacia Arriba , Células Vero , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo
5.
J Vet Sci ; 22(4): e49, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34170090

RESUMEN

The S1 protein of the infectious bronchitis virus (IBV) is a major structural protein that induces the production of the virus-neutralization antibodies. The monoclonal antibody against the IBV M41 S1 protein was used as a target for biopanning. After three rounds of biopanning, randomly selected phages bound to the monoclonal antibody. Sequence analysis showed that the dominant sequence was SFYDFEMQGFFI. Indirect competitive enzyme-linked immunosorbent assay showed that SFYDFEMQGFFI is a mimotope of the S1 protein that was predicted by PepSurf. The mimotope may provide information for further structural and functional analyses of the S1 protein.


Asunto(s)
Virus de la Bronquitis Infecciosa/metabolismo , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/metabolismo , Anticuerpos Monoclonales , Bacteriófagos , Bioprospección
6.
Viruses ; 12(10)2020 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003350

RESUMEN

The Gammacoronavirus infectious bronchitis virus (IBV) causes a highly contagious and economically important respiratory disease in poultry. In the laboratory, most IBV strains are restricted to replication in ex vivo organ cultures or in ovo and do not replicate in cell culture, making the study of their basic virology difficult. Entry of IBV into cells is facilitated by the large glycoprotein on the surface of the virion, the spike (S) protein, comprised of S1 and S2 subunits. Previous research showed that the S2' cleavage site is responsible for the extended tropism of the IBV Beaudette strain. This study aims to investigate whether protease treatment can extend the tropism of other IBV strains. Here we demonstrate that the addition of exogenous trypsin during IBV propagation in cell culture results in significantly increased viral titres. Using a panel of IBV strains, exhibiting varied tropisms, the effects of spike cleavage on entry and replication were assessed by serial passage cell culture in the presence of trypsin. Replication could be maintained over serial passages, indicating that the addition of exogenous protease is sufficient to overcome the barrier to infection. Mutations were identified in both S1 and S2 subunits following serial passage in cell culture. This work provides a proof of concept that exogenous proteases can remove the barrier to IBV replication in otherwise non-permissive cells, providing a platform for further study of elusive field strains and enabling sustainable vaccine production in vitro.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Virus de la Bronquitis Infecciosa/efectos de los fármacos , Virus de la Bronquitis Infecciosa/fisiología , Tripsina/uso terapéutico , Tropismo Viral/efectos de los fármacos , Animales , Línea Celular , Chlorocebus aethiops , Gammacoronavirus/efectos de los fármacos , Virus de la Bronquitis Infecciosa/metabolismo , Cinética , Pase Seriado , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Virión/efectos de los fármacos , Virión/metabolismo , Replicación Viral/efectos de los fármacos
7.
J Gen Virol ; 99(5): 619-630, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29557770

RESUMEN

Enveloped viruses gain entry into host cells by fusing with cellular membranes, a step that is required for virus replication. Coronaviruses, including the severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) and infectious bronchitis virus (IBV), fuse at the plasma membrane or use receptor-mediated endocytosis and fuse with endosomes, depending on the cell or tissue type. The virus spike (S) protein mediates fusion with the host cell membrane. We have shown previously that an Abelson (Abl) kinase inhibitor, imatinib, significantly reduces SARS-CoV and MERS-CoV viral titres and prevents endosomal entry by HIV SARS S and MERS S pseudotyped virions. SARS-CoV and MERS-CoV are classified as BSL-3 viruses, which makes experimentation into the cellular mechanisms involved in infection more challenging. Here, we use IBV, a BSL-2 virus, as a model for studying the role of Abl kinase activity during coronavirus infection. We found that imatinib and two specific Abl kinase inhibitors, GNF2 and GNF5, reduce IBV titres by blocking the first round of virus infection. Additionally, all three drugs prevented IBV S-induced syncytia formation prior to the hemifusion step. Our results indicate that membrane fusion (both virus-cell and cell-cell) is blocked in the presence of Abl kinase inhibitors. Studying the effects of Abl kinase inhibitors on IBV will be useful in identifying the host cell pathways required for coronavirus infection. This will provide an insight into possible therapeutic targets to treat infections by current as well as newly emerging coronaviruses.


Asunto(s)
Endosomas/virología , Virus de la Bronquitis Infecciosa/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus/metabolismo , Internalización del Virus , Animales , Antivirales/farmacología , Benzamidas/farmacología , Membrana Celular , Chlorocebus aethiops , Mesilato de Imatinib/farmacología , Virus de la Bronquitis Infecciosa/metabolismo , Pirimidinas/farmacología , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Replicación Viral
8.
Protein Sci ; 26(5): 1037-1048, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28257598

RESUMEN

The potential for infection by coronaviruses (CoVs) has become a serious concern with the recent emergence of Middle East respiratory syndrome and severe acute respiratory syndrome (SARS) in the human population. CoVs encode two large polyproteins, which are then processed into 15-16 nonstructural proteins (nsps) that make significant contributions to viral replication and transcription by assembling the RNA replicase complex. Among them, nsp9 plays an essential role in viral replication by forming a homodimer that binds single-stranded RNA. Thus, disrupting nsp9 dimerization is a potential anti-CoV therapy. However, different nsp9 dimer forms have been reported for alpha- and beta-CoVs, and no structural information is available for gamma-CoVs. Here we determined the crystal structure of nsp9 from the avian infectious bronchitis virus (IBV), a representative gamma-CoV that affects the economy of the poultry industry because it can infect domestic fowl. IBV nsp9 forms a homodimer via interactions across a hydrophobic interface, which consists of two parallel alpha helices near the carboxy terminus of the protein. The IBV nsp9 dimer resembles that of SARS-CoV nsp9, indicating that this type of dimerization is conserved among all CoVs. This makes disruption of the dimeric interface an excellent strategy for developing anti-CoV therapies. To facilitate this effort, we characterized the roles of six conserved residues on this interface using site-directed mutagenesis and a multitude of biochemical and biophysical methods. We found that three residues are critical for nsp9 dimerization and its abitlity to bind RNA.


Asunto(s)
Virus de la Bronquitis Infecciosa/química , Multimerización de Proteína , ARN Viral/química , Proteínas de Unión al ARN/química , Proteínas Virales/química , Virus de la Bronquitis Infecciosa/metabolismo , ARN Viral/metabolismo , Proteínas de Unión al ARN/metabolismo , Relación Estructura-Actividad , Proteínas Virales/metabolismo
9.
Arch Virol ; 162(7): 1943-1950, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28316013

RESUMEN

Coronavirus papain-like proteases (PLPs) can act as proteases that process virus-encoded large replicase polyproteins and also as deubiquitinating (DUB) enzymes. Like the PLPs of other coronaviruses (CoVs), the avian infectious bronchitis virus (IBV) PLP catalyzes proteolysis of Gly-Gly dipeptide bonds to release mature cleavage products. However, the other functions of the IBV PLP are not well understood. In this study, we found that IBV exhibits strong global DUB activity with significant reductions of the levels of ubiquitin (Ub)-, K48-, and K63-conjugated proteins. The DUB activity exhibited a clear time dependence, with stronger DUB activity in the early stage of viral infection. Furthermore, the IBV replicase-encoded PLP, including the downstream transmembrane (TM) domain, is a DUB enzyme and dramatically reduced the level of Ub-conjugated proteins, while processing both K48- and K63-linked polyubiquitin chains. By contrast, PLP did not cause any reduction of haemagglutinin (HA)-Ub-conjugated proteins. In addition, mutations of the catalytic residues of PLP-TM, Cys1274Ser and His1437Lys, reduced DUB activity against Ub-, K48- and K63- conjugated proteins, indicating that the DUB activity of the PLP-TM wild-type protein is not completely dependent on its catalytic activity. Overall, these results demonstrate that the IBV-encoded PLP-TM functions as a DUB enzyme and suggest that IBV may interfere with the activation of host antiviral signaling pathway by degrading polyubiquitin-associated proteins.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Regulación Viral de la Expresión Génica/fisiología , Virus de la Bronquitis Infecciosa/enzimología , Péptido Hidrolasas/metabolismo , Proteínas Virales/metabolismo , Animales , Células Cultivadas , Embrión de Pollo , Clonación Molecular , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/metabolismo , Riñón/citología , Mutagénesis Sitio-Dirigida , Péptido Hidrolasas/genética , Ubiquitinación , Proteínas Virales/genética
10.
Virol Sin ; 31(1): 57-68, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26920710

RESUMEN

Avian infectious bronchitis virus (IBV) is a Gammacoronavirus in the family Coronaviridae and causes highly contagious respiratory disease in chickens. Innate immunity plays significant roles in host defense against IBV. Here, we explored the interaction between IBV and the host innate immune system. Severe histopathological lesions were observed in the tracheal mucosa at 3-5 days post inoculation (dpi) and in the kidney at 8 dpi, with heavy viral loads at 1-11 and 1-28 dpi, respectively. The expression of mRNAs encoding Toll-like receptor (TLR) 3 and TLR7 were upregulated at 3-8 dpi, and that of TIR-domain-containing adapter-inducing interferon (IFN) ß (TRIF) was upregulated at 21 dpi in the trachea and kidney. Myeloid differentiation primary response protein 88 (MyD88) was upregulated in the trachea during early infection. Tumor necrosis factor receptor-associated factor (TRAF) 3 and TRAF6 were upregulated expression in both tissues. Moreover, melanoma differentiation-associated protein 5 (MDA5), laboratory of genetics and physiology 2 (LGP2), stimulator of IFN genes (STING), and mitochondrial antiviral signaling protein (MAVS), as well as TANK binding kinase 1 (TBK1), inhibitor of kappaB kinase (IKK) ε, IKKα, IKKß, IFN regulatory factor (IRF) 7, nuclear factor of kappaB (NF-ĸB), IFN-α, IFN-ß, various interleukins(ILs), and macrophage inflammatory protein-1ß (MIP-1ß) were significantly upregulated in the trachea and downregulated in the kidney. These results suggested that the TLR and MDA5 signaling pathways and innate immune cytokine were induced after IBV infection. Additionally, consistent responses to IBV infection were observed during early infection, with differential and complicated responses in the kidney.


Asunto(s)
Pollos/virología , Infecciones por Coronavirus/veterinaria , Virus de la Bronquitis Infecciosa/metabolismo , Helicasa Inducida por Interferón IFIH1/metabolismo , Enfermedades de las Aves de Corral/metabolismo , Enfermedades de las Aves de Corral/virología , Receptores Toll-Like/metabolismo , Animales , Diferenciación Celular/fisiología , Quimiocina CCL4/metabolismo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/patología , Citocinas/análisis , Citocinas/biosíntesis , ARN Helicasas DEAD-box/metabolismo , Interacciones Huésped-Patógeno , Quinasa I-kappa B/metabolismo , Inmunidad Innata , Inmunoglobulina G/sangre , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/inmunología , Interferón beta/biosíntesis , Factor 88 de Diferenciación Mieloide/metabolismo , Enfermedades de las Aves de Corral/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Mensajero/biosíntesis , Transducción de Señal , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/inmunología , Activación Transcripcional , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/biosíntesis , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo
11.
Biotechnol Lett ; 38(2): 299-304, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26463372

RESUMEN

OBJECTIVE: To assemble infectious bronchitis virus (IBV)-like particles bearing the recombinant spike protein and investigate the humoral immune responses in chickens. RESULTS: IBV virus-like particles (VLPs) were generated through the co-infection with three recombinant baculoviruses separately encoding M, E or the recombinant S genes. The recombinant S protein was sufficiently flexible to retain the ability to self-assemble into VLPs. The size and morphology of the VLPs were similar to authentic IBV particles. In addition, the immunogenicity of IBV VLPs had been investigated. The results demonstrated that the efficiency of the newly generated VLPs was comparable to that of the inactivated M41 viruses in eliciting IBV-specific antibodies and neutralizing antibodies in chickens via subcutaneous inoculation. CONCLUSIONS: This work provides basic information for the mechanism of IBV VLP formation and develops a platform for further designing IBV VLP-based vaccines against IBV or other viruses.


Asunto(s)
Virus de la Bronquitis Infecciosa/metabolismo , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/metabolismo , Virosomas/metabolismo , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Baculoviridae , Pollos , Vectores Genéticos , Virus de la Bronquitis Infecciosa/genética , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Vacunas de Partículas Similares a Virus/genética , Vacunas de Partículas Similares a Virus/ultraestructura , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Virosomas/genética
12.
J Biol Chem ; 290(52): 31138-50, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26546678

RESUMEN

Cystine knot α-amylase inhibitors are cysteine-rich, proline-rich peptides found in the Amaranthaceae and Apocynaceae plant species. They are characterized by a pseudocyclic backbone with two to four prolines and three disulfides arranged in a knotted motif. Similar to other knottins, cystine knot α-amylase inhibitors are highly resistant to degradation by heat and protease treatments. Thus far, only the α-amylase inhibition activity has been described for members of this family. Here, we show that cystine knot α-amylase inhibitors named alstotides discovered from the Alstonia scholaris plant of the Apocynaceae family display antiviral activity. The alstotides (As1-As4) were characterized by both proteomic and genomic methods. All four alsotides are novel, heat-stable and enzyme-stable and contain 30 residues. NMR determination of As1 and As4 structures reveals their conserved structural fold and the presence of one or more cis-proline bonds, characteristics shared by other cystine knot α-amylase inhibitors. Genomic analysis showed that they contain a three-domain precursor, an arrangement common to other knottins. We also showed that alstotides are antiviral and cell-permeable to inhibit the early phase of infectious bronchitis virus and Dengue infection, in addition to their ability to inhibit α-amylase. Taken together, our results expand membership of cystine knot α-amylase inhibitors in the Apocynaceae family and their bioactivity, functional promiscuity that could be exploited as leads in developing therapeutics.


Asunto(s)
Alstonia/química , Antivirales , Infecciones por Coronavirus/tratamiento farmacológico , Virus del Dengue , Dengue , Inhibidores de Glicósido Hidrolasas , Virus de la Bronquitis Infecciosa/efectos de los fármacos , Proteínas de Plantas , Alstonia/genética , Animales , Antivirales/química , Antivirales/aislamiento & purificación , Antivirales/farmacología , Chlorocebus aethiops , Infecciones por Coronavirus/metabolismo , Inhibidores de Glicósido Hidrolasas/química , Inhibidores de Glicósido Hidrolasas/aislamiento & purificación , Inhibidores de Glicósido Hidrolasas/farmacología , Virus de la Bronquitis Infecciosa/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/genética , Proteínas de Plantas/aislamiento & purificación , Proteínas de Plantas/farmacología , Estructura Terciaria de Proteína , Células Vero
13.
PLoS Pathog ; 8(5): e1002674, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22570613

RESUMEN

The coronavirus E protein is a small membrane protein with a single predicted hydrophobic domain (HD), and has a poorly defined role in infection. The E protein is thought to promote virion assembly, which occurs in the Golgi region of infected cells. It has also been implicated in the release of infectious particles after budding. The E protein has ion channel activity in vitro, although a role for channel activity in infection has not been established. Furthermore, the membrane topology of the E protein is of considerable debate, and the protein may adopt more than one topology during infection. We previously showed that the HD of the infectious bronchitis virus (IBV) E protein is required for the efficient release of infectious virus, an activity that correlated with disruption of the secretory pathway. Here we report that a single residue within the hydrophobic domain, Thr16, is required for secretory pathway disruption. Substitutions of other residues for Thr16 were not tolerated. Mutations of Thr16 did not impact virus assembly as judged by virus-like particle production, suggesting that alteration of secretory pathway and assembly are independent activities. We also examined how the membrane topology of IBV E affected its function by generating mutant versions that adopted either a transmembrane or membrane hairpin topology. We found that a transmembrane topology was required for disrupting the secretory pathway, but was less efficient for virus-like particle production. The hairpin version of E was unable to disrupt the secretory pathway or produce particles. The findings reported here identify properties of the E protein that are important for its function, and provide insight into how the E protein may perform multiple roles during infection.


Asunto(s)
Infecciones por Coronavirus/virología , Virus de la Bronquitis Infecciosa/química , Virus de la Bronquitis Infecciosa/metabolismo , Vías Secretoras , Proteínas del Envoltorio Viral/metabolismo , Secuencia de Aminoácidos , Transporte Biológico , Línea Celular Tumoral , Infecciones por Coronavirus/metabolismo , Aparato de Golgi/ultraestructura , Células HeLa , Humanos , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/patogenicidad , Mutación , Estructura Terciaria de Proteína , Transporte de Proteínas , Alineación de Secuencia , Proteínas del Envoltorio Viral/química , Ensamble de Virus
14.
Viral Immunol ; 25(1): 55-62, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22225471

RESUMEN

Infectious bronchitis virus (IBV) is prevented primarily by the use of live attenuated vaccines, which are known to have a limited strain range of protection. Alternative vaccines against the emerging new virus strains can improve control of the disease. The aim of this study was to evaluate the immunogenic potential of two recombinant viral proteins, when administered by eyedrop, without the assistance of a vector. The recombinant S1 (rS1) and N (rN) proteins of the M41 strain expressed in E. coli were tested, and the live attenuated vaccine H120 was used as a positive control. Protection was evaluated by re-isolation of virus from tracheas of vaccinated chickens after challenge with strain M41. After three immunizations, rS1 glycoprotein induced 40% protection, while vaccination with rN provided no protection. Vaccination with rS1, rN, or H120 induced a cellular immune response as demonstrated by in vitro ChIFN-γ production by splenocytes of vaccinated birds. Vaccination with H120, and to a lesser extent rS1, induced HI and virus-specific IgG antibody production. These findings indicate that recombinant viral proteins administered through the mucosal route can evoke an immune response without the assistance of a vector.


Asunto(s)
Virus de la Bronquitis Infecciosa/inmunología , Glicoproteínas de Membrana/inmunología , Proteínas de la Nucleocápside/inmunología , Enfermedades de las Aves de Corral/inmunología , Proteínas Recombinantes/inmunología , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología , Administración a través de la Mucosa , Animales , Anticuerpos Antivirales/sangre , Pollos , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Proteínas de la Nucleocápside de Coronavirus , Virus de la Bronquitis Infecciosa/metabolismo , Interferón gamma/biosíntesis , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/genética , Proteínas de la Nucleocápside/administración & dosificación , Proteínas de la Nucleocápside/genética , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Glicoproteína de la Espiga del Coronavirus , Vacunación , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
15.
Virol J ; 8: 435, 2011 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-21910859

RESUMEN

BACKGROUND: Transmissible gastroenteritis virus (TGEV) has a sialic acid binding activity that is believed to be important for enteropathogenicity, but that has so far appeared to be dispensable for infection of cultured cells. The aims of this study were to determine the effect of sialic acid binding for the infection of cultured cells under unfavorable conditions, and comparison of TGEV strains and mutants, as well as the avian coronavirus IBV concerning their dependence on the sialic acid binding activity. METHODS: The infectivity of different viruses was analyzed by a plaque assay after adsorption times of 5, 20, and 60 min. Prior to infection, cultured cells were either treated with neuraminidase to deplete sialic acids from the cell surface, or mock-treated. In a second approach, pre-treatment of the virus with porcine intestinal mucin was performed, followed by the plaque assay after a 5 min adsorption time. A student's t-test was used to verify the significance of the results. RESULTS: Desialylation of cells only had a minor effect on the infection by TGEV strain Purdue 46 when an adsorption period of 60 min was allowed for initiation of infection. However, when the adsorption time was reduced to 5 min the infectivity on desialylated cells decreased by more than 60%. A TGEV PUR46 mutant (HAD3) deficient in sialic acid binding showed a 77% lower titer than the parental virus after a 5 min adsorption time. After an adsorption time of 60 min the titer of HAD3 was 58% lower than that of TGEV PUR46. Another TGEV strain, TGEV Miller, and IBV Beaudette showed a reduction in infectivity after neuraminidase treatment of the cultured cells irrespective of the virion adsorption time. CONCLUSIONS: Our results suggest that the sialic acid binding activity facilitates the infection by TGEV under unfavorable environmental conditions. The dependence on the sialic acid binding activity for an efficient infection differs in the analyzed TGEV strains.


Asunto(s)
Membrana Celular/metabolismo , Gastroenteritis Porcina Transmisible/virología , Glicoproteínas de Membrana/metabolismo , Neuraminidasa/farmacología , Ácidos Siálicos/metabolismo , Testículo/metabolismo , Virus de la Gastroenteritis Transmisible/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Aves , Membrana Celular/efectos de los fármacos , Membrana Celular/virología , Células Cultivadas , Chlorocebus aethiops , Gastroenteritis Porcina Transmisible/metabolismo , Virus de la Bronquitis Infecciosa/metabolismo , Masculino , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/química , Mucinas/farmacología , Mutación , Neuraminidasa/metabolismo , Unión Proteica/efectos de los fármacos , Ácidos Siálicos/antagonistas & inhibidores , Glicoproteína de la Espiga del Coronavirus , Porcinos , Testículo/citología , Testículo/efectos de los fármacos , Testículo/virología , Virus de la Gastroenteritis Transmisible/efectos de los fármacos , Virus de la Gastroenteritis Transmisible/genética , Virus de la Gastroenteritis Transmisible/patogenicidad , Células Vero , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/química , Ensayo de Placa Viral , Virión/efectos de los fármacos , Virión/metabolismo , Acoplamiento Viral/efectos de los fármacos
16.
Autophagy ; 7(11): 1335-47, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21799305

RESUMEN

Autophagy is a cellular response to starvation which generates autophagosomes to carry cellular organelles and long-lived proteins to lysosomes for degradation. Degradation through autophagy can provide an innate defence against virus infection, or conversely autophagosomes can promote infection by facilitating assembly of replicase proteins. We demonstrate that the avian coronavirus, Infectious Bronchitis Virus (IBV) activates autophagy. A screen of individual IBV non-structural proteins (nsps) showed that autophagy was activated by IBV nsp6. This property was shared with nsp6 of mammalian coronaviruses Mouse Hepatitis Virus, and Severe Acute Respiratory Syndrome Virus, and the equivalent nsp5-7 of the arterivirus Porcine Reproductive and Respiratory Syndrome Virus. These multiple-spanning transmembrane proteins located to the endoplasmic reticulum (ER) where they generated Atg5 and LC3II-positive vesicles, and vesicle formation was dependent on Atg5 and class III PI3 kinase. The vesicles recruited double FYVE-domain containing protein (DFCP) indicating localised concentration of phosphatidylinositol 3 phosphate, and therefore shared many features with omegasomes formed from the ER in response to starvation. Omegasomes induced by viral nsp6 matured into autophagosomes that delivered LC3 to lysosomes and therefore recruited and recycled the proteins needed for autophagosome nucleation, expansion, cellular trafficking and delivery of cargo to lysosomes. The coronavirus nsp6 proteins activated omegasome and autophagosome formation independently of starvation, but activation did not involve direct inhibition of mTOR signalling, activation of sirtuin1 or induction of ER stress.


Asunto(s)
Autofagia , Retículo Endoplásmico/metabolismo , Virus de la Bronquitis Infecciosa/metabolismo , Fagosomas/metabolismo , Proteínas no Estructurales Virales/metabolismo , Androstadienos/farmacología , Animales , Arterivirus/efectos de los fármacos , Autofagia/efectos de los fármacos , Proteína 5 Relacionada con la Autofagia , Línea Celular , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/virología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Genoma Viral/genética , Humanos , Virus de la Bronquitis Infecciosa/genética , Fusión de Membrana/efectos de los fármacos , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Fosfatos de Fosfatidilinositol/farmacología , Estructura Terciaria de Proteína , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factor de Transcripción CHOP/metabolismo , Proteínas no Estructurales Virales/química , Wortmanina
17.
Protein Sci ; 18(1): 6-16, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19177346

RESUMEN

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.


Asunto(s)
Adenosina Difosfato Ribosa/metabolismo , Coronavirus Humano 229E/química , Virus de la Bronquitis Infecciosa/química , Proteínas no Estructurales Virales/química , Adenosina Difosfato Ribosa/análogos & derivados , Adenosina Difosfato Ribosa/química , Secuencia de Aminoácidos , Coronavirus Humano 229E/genética , Coronavirus Humano 229E/metabolismo , Cristalografía por Rayos X , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Unión Proteica/fisiología , Conformación Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/metabolismo , Alineación de Secuencia , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo
18.
Virology ; 379(2): 175-80, 2008 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-18678384

RESUMEN

Coronavirus 3C-like proteinase (3CLpro) plays important roles in viral life cycle through extensive processing of the polyproteins 1a and 1ab into 12 mature, non-structural proteins (nsp5-nsp16). Structural and biochemical studies have revealed that all confirmed 3CLpro cleavage sites have a conserved Gln residue at the P1 position, which is thought to be absolutely required for efficient cleavage. Recent studies on murine hepatitis virus (MHV) showed that processing of the 1a polyprotein at the position between nsp10-nsp11 is essential for viral replication. In this report, we investigated the requirement of processing at the equivalent position for replication of avian coronavirus infectious bronchitis virus (IBV), using an infectious cloning system. The results showed that mutation of the P1 Gln to Pro or deletion of the Gln residue in the nsp10-nsp11/12 site completely abolished the 3CLpro-mediated processing, but allowed production of infectious recombinant viruses with variable degrees of growth defect, suggesting that cleavage at the nsp10-nsp11/12 site of IBV is dispensable for viral replication in cultured cells. This study would pave a way for potential vaccine development by generation of attenuated IBV from field isolates through manipulation of the nsp10-nsp11/12 cleavage site. Similar approaches would be also applicable to other human and animal coronaviruses.


Asunto(s)
Virus de la Bronquitis Infecciosa/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas Virales/metabolismo , Sustitución de Aminoácidos , Animales , Dominio Catalítico/genética , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Humanos , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/fisiología , Mutagénesis Sitio-Dirigida , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eliminación de Secuencia , Células Vero , Replicación Viral
19.
J Virol ; 82(6): 2765-71, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18094153

RESUMEN

We have analyzed the intracellular transport of the spike (S) protein of infectious bronchitis virus (IBV), an avian coronavirus. Surface expression was analyzed by immunofluorescence microscopy, by surface biotinylation, and by syncytium formation by S-expressing cells. By applying these methods, the S protein was found to be retained intracellularly. Tyr1143 in the cytoplasmic tail was shown to be a crucial component of the retention signal. Deletion of a dilysine motif that has previously been suggested to function as a retrieval signal did not abolish intracellular retention. Treatment of the S proteins with endoglycosidases did not reveal any differences between the parental and the mutant proteins. Furthermore, all S proteins analyzed were posttranslationally cleaved into the subunits S1 and S2. In coexpression experiments, the S protein was found to colocalize with a Golgi marker. Taken together, these results indicate that the S protein of IBV is retained at a late Golgi compartment. Therefore, this viral surface protein differs from the S proteins of transmissible gastroenteritis virus and severe acute respiratory syndrome coronavirus, which are retained at a pre-Golgi compartment or transported to the cell surface, respectively. The implications of these differences are discussed.


Asunto(s)
Virus de la Bronquitis Infecciosa/metabolismo , Glicoproteínas de Membrana/metabolismo , Tirosina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Animales , Células Cultivadas , Cricetinae , Endocitosis , Técnica del Anticuerpo Fluorescente , Inmunoprecipitación , Glicoproteínas de Membrana/química , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA