Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Methods Mol Biol ; 2768: 135-151, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38502392

RESUMO

The receptor binding domain (RBD) of SARS-CoV-2 (SCoV2) has been used recently to identify the RBD sequences of feline coronavirus serotypes 1 (FCoV1) and 2 (FCoV2). Cats naturally infected with FCoV1 have been shown to possess serum reactivities with FCoV1 and SCoV2 RBDs but not with FCoV2 RBD. In the current study, COVID-19-vaccinated humans and FCoV1-infected laboratory cats were evaluated for interferon-gamma (IFNγ) and interleukin-2 (IL-2 ELISpot responses by their peripheral blood mononuclear cells (PBMC) to SCoV2, FCoV1, and FCoV2 RBDs. Remarkably, the PBMC from COVID-19-vaccinated subjects developed IFNγ responses to SCoV2, FCoV1, and FCoV2 RBDs. The most vaccinated subject (five vaccinations over 2 years) appeared to produce hyperreactive IFNγ responses to all three RBDs, including the PBS media control. This subject lost IFNγ responses to all RBDs at 9 months (9 mo) post-last vaccination. However, her IL-2 responses to FCoV1 and FCoV2 RBDs were low but detectable at 10 mo post-last vaccination. This observation suggests that initially robust IFNγ responses to SCoV2 RBD may be an outcome of robust inflammatory IFNγ responses to SCoV2 RBD. Hence, the T-cell responses of vaccine immunity should be monitored by vaccine immunogen-specific IL-2 production. The PBMC from chronically FCoV1-infected cats developed robust IFNγ responses to SCoV2 and FCoV2 RBDs but had the lowest IFNγ responses to FCoV1 RBD. The constant exposure to FCoV1 reinfection may cause the IFNγ responses to be downregulated to the infecting virus FCoV1 but not to the cross-reacting epitopes on the SCoV2 and FCoV2 RBDs.


Assuntos
COVID-19 , Coronavirus Felino , Vacinas , Humanos , Feminino , Gatos , Animais , Interferon gama , Interleucina-2 , Coronavirus Felino/metabolismo , Leucócitos Mononucleares/metabolismo , RNA Viral , Linfócitos T , RNA Mensageiro , Sorogrupo , SARS-CoV-2/metabolismo , Anticorpos Antivirais/metabolismo
2.
Front Immunol ; 13: 925922, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35837396

RESUMO

Although feline coronavirus (FCoV) infection is extremely common in cats, there are currently few effective treatments. A peptide derived from the heptad repeat 2 (HR2) domain of the coronavirus (CoV) spike protein has shown effective for inhibition of various human and animal CoVs in vitro, but further use of FCoV-HR2 in vivo has been limited by lack of practical delivery vectors and small animal infection model. To overcome these technical challenges, we first constructed a recombinant Bacillus subtilis (rBSCotB-HR2P) expressing spore coat protein B (CotB) fused to an HR2-derived peptide (HR2P) from a serotype II feline enteric CoV (FECV). Immunogenic capacity was evaluated in mice after intragastric or intranasal administration, showing that recombinant spores could trigger strong specific cellular and humoral immune responses. Furthermore, we developed a novel mouse model for FECV infection by transduction with its primary receptor (feline aminopeptidase N) using an E1/E3-deleted adenovirus type 5 vector. This model can be used to study the antiviral immune response and evaluate vaccines or drugs, and is an applicable choice to replace cats for the study of FECV. Oral administration of rBSCotB-HR2P in this mouse model effectively protected against FECV challenge and significantly reduced pathology in the digestive tract. Owing to its safety, low cost, and probiotic features, rBSCotB-HR2P is a promising oral vaccine candidate for use against FECV/FCoV infection in cats.


Assuntos
Infecções por Coronavirus , Coronavirus Felino , Animais , Bacillus subtilis/genética , Antígenos CD13/metabolismo , Gatos , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Modelos Animais de Doenças , Humanos , Imunidade , Camundongos , Peptídeos/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Esporos Bacterianos/genética
3.
J Vet Sci ; 23(2): e27, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35363438

RESUMO

BACKGROUND: The role of Toll-like receptors (TLRs) in a feline infectious peritonitis virus (FIPV) infection is not completely understood. OBJECTIVES: This study examined the expression of TLR3, TLR7, TLR9, tumor necrosis factor-alpha (TNF-α), interferon (IFN)-ß, and interleukin (IL)-10 upon an FIPV infection in Crandell-Reese feline kidney (CRFK) cells and feline monocytes. METHODS: CRFK cells and monocytes from feline coronavirus (FCoV)-seronegative cats and FCoV-seropositive cats were infected with type II FIPV-79-1146. At four, 12, and 24 hours post-infection (hpi), the expression of TLR3, TLR7, TLR9, TNF-α, IFN-ß, and IL-10, and the viral load were measured using reverse transcription quantitative polymerase chain reaction. Viral protein production was confirmed using immunofluorescence. RESULTS: FIPV-infected CRFK showed the upregulation of TLR9, TNF-α, and IFN-ß expression between 4 and 24 hpi. Uninfected monocytes from FCoV-seropositive cats showed lower TLR3 and TLR9 expression but higher TLR7 expression compared to uninfected monocytes from FCoV-seronegative cats. FIPV-infected monocytes from FCoV-seropositive cats downregulated TLR7 and TNF-α expression between 4 and 24 hpi, and 4 and 12 hpi, respectively. IFN-ß was upregulated early in FIPV-infected monocytes from FCoV-seropositive cats, with a significant difference observed at 12 hpi compared to FCoV-seronegative cats. The viral load in the CRFK and FIPV-infected monocytes in both cohorts of cats was similar over time. CONCLUSION: TLR7 may be the key TLR involved in evading the innate response against inhibiting TNF-α production. Distinct TLR expression profiles between FCoV-seronegative and FCoV-seropositive cats were observed. The associated TLR that plays a role in the induction of IFN-ß needs to be explored further.


Assuntos
Doenças do Gato , Coronavirus Felino , Peritonite Infecciosa Felina , Animais , Gatos , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Citocinas/genética , Citocinas/metabolismo , Rim/metabolismo , Monócitos/metabolismo , Receptor 3 Toll-Like
4.
Gene ; 825: 146443, 2022 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-35337854

RESUMO

Both feline coronavirus (FCoV) and SARS-CoV-2 are coronaviruses that infect cats and humans, respectively. However, cats have been shown to be susceptible to SARS-CoV-2, and FCoV also had been shown to infect human. To elucidate the relationship between FCoV and SARS-CoV-2, we highlight the main characteristics of the genome, the receptor usage, and the correlation of the receptor-binding domain (RBD) of spike proteins in FCoV and SARS-CoV-2. It is demonstrated that FCoV and SARS-CoV-2 are closely related to the main characteristics of the genome, receptor usage, and RBD of spike proteins with similar furin cleavage sites. In particular, the affinity of the conserved feline angiotensin-converting enzyme 2 (fACE2) receptor to the RBD of SARS-CoV-2 suggests that cats are susceptible to SARS-CoV-2. In addition, cross-species of coronaviruses between cats and humans or other domesticated animals are also discussed. This review sheds light on cats as potential intermediate hosts for SARS-CoV-2 transmission, and cross-species transmission or zoonotic infection of FCoV and SARS-CoV-2 between cats and humans was identified.


Assuntos
COVID-19 , Coronavirus Felino , Animais , COVID-19/veterinária , Gatos , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Ligação Proteica , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química
5.
Viruses ; 12(1)2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31936749

RESUMO

Feline coronavirus (FCoV) is a complex viral agent that causes a variety of clinical manifestations in cats, commonly known as feline infectious peritonitis (FIP). It is recognized that FCoV can occur in two different serotypes. However, differences in the S protein are much more than serological or antigenic variants, resulting in the effective presence of two distinct viruses. Here, we review the distinct differences in the S proteins of these viruses, which are likely to translate into distinct biological outcomes. We introduce a new concept related to the non-taxonomical classification and differentiation among FCoVs by analyzing and comparing the genetic, structural, and functional characteristics of FCoV and the FCoV S protein among the two serotypes and FCoV biotypes. Based on our analysis, we suggest that our understanding of FIP needs to consider whether the presence of these two distinct viruses has implications in clinical settings.


Assuntos
Coronavirus Felino/genética , Peritonite Infecciosa Felina/virologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Gatos , Coronavirus Felino/metabolismo , Coronavirus Felino/patogenicidade , Peritonite Infecciosa Felina/metabolismo , Fusão de Membrana , Modelos Moleculares , Receptores Virais/metabolismo , Sorogrupo , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus/genética
6.
J Gen Virol ; 100(10): 1417-1430, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31483243

RESUMO

Feline coronavirus (FCoV) has been identified as the aetiological agent of feline infectious peritonitis (FIP), a highly fatal systemic disease in cats. FCoV open reading frame 3 (ORF3) encodes accessory proteins 3a, 3b and 3 c. The FCoV 3b accessory protein consists of 72 amino acid residues and localizes to nucleoli and mitochondria. The present work focused on peptide domains within FCoV 3b that drive its intracellular trafficking. Transfection of different cell types with FCoV 3b fused to enhanced green fluorescent protein (EGFP) or 3×FLAG confirmed localization of FCoV 3b in the mitochondria and nucleoli. Using serial truncated mutants, we showed that nucleolar accumulation is controlled by a joint nucleolar and nuclear localization signal (NoLS/NLS) in which the identified overlapping pat4 motifs (residues 53-57) play a critical role. Mutational analysis also revealed that mitochondrial translocation is mediated by N-terminal residues 10-35, in which a Tom20 recognition motif (residues 13-17) and two other overlapping hexamers (residues 24-30) associated with mitochondrial targeting were identified. In addition, a second Tom20 recognition motif was identified further downstream (residues 61-65), although the mitochondrial translocation evoked by these residues seemed less efficient as a diffuse cytoplasmic distribution was also observed. Assessing the spatiotemporal distribution of FCoV 3b did not provide convincing evidence of dynamic shuttling behaviour between the nucleoli and the mitochondria.


Assuntos
Coronavirus Felino/metabolismo , Peritonite Infecciosa Felina/virologia , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/metabolismo , Animais , Gatos , Nucléolo Celular/virologia , Coronavirus Felino/química , Coronavirus Felino/genética , Mitocôndrias/virologia , Sinais de Localização Nuclear , Fases de Leitura Aberta , Domínios Proteicos , Transporte Proteico , Proteínas não Estruturais Virais/genética
7.
Viruses ; 12(1)2019 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-31905881

RESUMO

Feline infectious peritonitis (FIP), caused by virulent feline coronavirus, is the leading infectious cause of death in cats. The type I interferon (type I IFN)-mediated immune responses provide host protection from infectious diseases. Several coronaviruses have been reported to evolve diverse strategies to evade host IFN response. However, whether feline infectious peritonitis virus (FIPV) antagonizes the type I IFN signaling remains unclear. In this study, we demonstrated that FIPV strain DF2 infection not only failed to induce interferon-ß (IFN-ß) and interferon-stimulated gene (ISG) production, but also inhibited Sendai virus (SEV) or polyinosinic-polycytidylic acid (poly(I:C))-induced IFN-ß production. Subsequently, we found that one of the non-structural proteins encoded by the FIPV genome, nsp5, interrupted type I IFN signaling in a protease-dependent manner by cleaving the nuclear factor κB (NF-κB) essential modulator (NEMO) at three sites-glutamine132 (Q132), Q205, and Q231. Further investigation revealed that the cleavage products of NEMO lost the ability to activate the IFN-ß promoter. Mechanistically, the nsp5-mediated NEMO cleavage disrupted the recruitment of the TRAF family member-associated NF-κB activator (TANK) to NEMO, which reduced the phosphorylation of interferon regulatory factor 3 (IRF3), leading to the inhibition of type I IFN production. Our research provides new insights into the mechanism for FIPV to counteract host innate immune response.


Assuntos
Infecções por Coronavirus/imunologia , Coronavirus Felino/fisiologia , Cisteína Endopeptidases/metabolismo , Quinase I-kappa B/metabolismo , Interferon Tipo I/antagonistas & inibidores , Proteínas não Estruturais Virais/metabolismo , Animais , Gatos , Linhagem Celular , Proteases 3C de Coronavírus , Coronavirus Felino/metabolismo , Cisteína Endopeptidases/genética , Quinase I-kappa B/genética , Evasão da Resposta Imune , Imunidade Inata , Fator Regulador 3 de Interferon , Interferon Tipo I/metabolismo , Interferon beta/antagonistas & inibidores , Interferon beta/genética , Interferon beta/metabolismo , Mutação , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas não Estruturais Virais/genética
8.
Acta Vet Hung ; 66(3): 493-508, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30264619

RESUMO

Feline enteric coronaviruses have three open reading frames (ORFs) in region 3 (3a, 3b, and 3c). All three ORFs were expressed with C-terminal eGFP and 3xFLAG tags in different cell lines and their localisation was determined. ORF 3a is predicted to contain DNA-binding and transcription activator domains, and it is localised in the nucleus and in the cytoplasm. ORF 3b is also predicted to contain DNA-binding and activator domains, and was found to localise in the mitochondrion. Besides that, in some of the non-infected and FIPV-infected cells nucleolar, perinuclear or nuclear membrane accumulation of the eGFP-tagged 3b was observed. The exact compartmental localisation of ORF 3c is yet to be determined. However, based on our co-localisation studies 3c does not seem to be localised in the ER-Golgi network, ERGIC or peroxisomes. The expression of 3c-eGFP is clearly cell type dependent, it is more stable in MARC 145 cells than in Fcwf-4 or CrFK cells, which might reflect in vivo stability differences of 3c in natural target cells (enterocytes vs. monocytes/macrophages).


Assuntos
Coronavirus Felino/metabolismo , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Animais , Gatos , Linhagem Celular , Coronavirus Felino/genética , Regulação Viral da Expressão Gênica , Transporte Proteico , Proteínas Virais/química
9.
Virology ; 517: 108-121, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29329682

RESUMO

Feline coronavirus (FCoV) is an etiological agent that causes a benign enteric illness and the fatal systemic disease feline infectious peritonitis (FIP). The FCoV spike (S) protein is considered the viral regulator for binding and entry to the cell. This protein is also involved in FCoV tropism and virulence, as well as in the switch from enteric disease to FIP. This regulation is carried out by spike's major functions: receptor binding and virus-cell membrane fusion. In this review, we address important aspects in FCoV genetics, replication and pathogenesis, focusing on the role of S. To better understand this, FCoV S protein models were constructed, based on the human coronavirus NL63 (HCoV-NL63) S structure. We describe the specific structural characteristics of the FCoV S, in comparison with other coronavirus spikes. We also revise the biochemical events needed for FCoV S activation and its relation to the structural features of the protein.


Assuntos
Coronavirus Felino/patogenicidade , Peritonite Infecciosa Felina/virologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , Gatos , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Peritonite Infecciosa Felina/patologia , Regulação Viral da Expressão Gênica/fisiologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Replicação Viral
10.
J Gen Virol ; 98(8): 2017-2029, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28758629

RESUMO

Feline coronaviruses encode five accessory proteins with largely elusive functions. Here, one of these proteins, called 7b (206 residues), was investigated using a reverse genetic approach established for feline infectious peritonitis virus (FIPV) strain 79-1146. Recombinant FIPVs (rFPIVs) expressing mutant and/or FLAG-tagged forms of 7b were generated and used to investigate the expression, processing, glycosylation, localization and trafficking of the 7b protein in rFIPV-infected cells, focusing on a previously predicted ER retention signal, KTEL, at the C-terminus of 7b. The study revealed that 7b is N-terminally processed by a cellular signalase. The cleavage site, 17-Ala|Thr-18, was unambiguously identified by N-terminal sequence analysis of a 7b processing product purified from rFIPV-infected cells. Based on this information, rFIPVs expressing FLAG-tagged 7b proteins were generated and the effects of substitutions in the C-terminal 202KTEL206 sequence were investigated. The data show that (i) 7b localizes to and is retained in the medial- and/or trans-Golgi compartment, (ii) the C-terminal KTEL sequence acts as a Golgi [rather than an endoplasmic reticulum (ER)] retention signal, (iii) minor changes in the KTEL motif (such as KTE, KTEV, or the addition of a C-terminal tag) abolish Golgi retention, resulting in relocalization and secretion of 7b, and (iv) a KTEL-to-KDEL replacement causes retention of 7b in the ER of rFIPV-infected feline cells. Taken together, this study provides interesting new insights into an efficient Golgi retention signal that controls the cellular localization and trafficking of the FIPV 7b protein in virus-infected feline cells.


Assuntos
Coronavirus Felino/metabolismo , Peritonite Infecciosa Felina/virologia , Complexo de Golgi/virologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Gatos , Coronavirus Felino/química , Coronavirus Felino/genética , Glicosilação , Complexo de Golgi/ultraestrutura , Dados de Sequência Molecular , Sinais Direcionadores de Proteínas , Transporte Proteico , Proteínas Virais Reguladoras e Acessórias/química , Proteínas Virais Reguladoras e Acessórias/genética
11.
Microbiol Immunol ; 61(8): 318-327, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28675506

RESUMO

Unlike for serotype II feline coronaviruses (FCoV II), the cellular receptor for serotype I FCoV (FCoV I), the most prevalent FCoV serotype, is unknown. To provide a platform for assessing the pattern by which FCoV I attaches to its host receptor(s), HEK293 cell lines that stably express the ectodomains of the spike (S) proteins derived from a FCoV I feline enteric coronavirus strain UU7 (FECV UU7) and a feline infectious peritonitis virus strain UU4 (FIPV UU4) were established. Using the recombinant S proteins as probes to perform S protein affinity histochemistry in paraffin-embedded tissues, although no tissue or enteric binding of FECV UU7 S protein was detected, it was found that by immunohistochemistry that the tissue distribution of FIPV UU4 S protein-bound cells correlated with that of FIPV antigen-positive cells and lesions associated with FIP and that the affinity binding of FIPV UU4 S protein on macrophages was not affected by enzymatic removal of host cell-surface sialic acid with neuraminidase. These findings suggest that a factor(s) other than sialic acid contribute(s) to the macrophage tropism of FIPV strain UU4. This approach allowed obtaining more information about both virus-host cell interactions and the biological characteristics of the unidentified cellular receptor for FCoV I.


Assuntos
Coronavirus Felino/metabolismo , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Tropismo Viral/fisiologia , Ligação Viral , Animais , Gatos , Linhagem Celular , Células HEK293 , Interações Hospedeiro-Patógeno/fisiologia , Humanos , Macrófagos/virologia , Ácido N-Acetilneuramínico/química , Ligação Proteica/genética , Ligação Proteica/fisiologia , Sorogrupo , Glicoproteína da Espícula de Coronavírus/genética
12.
Vet Res ; 44: 57, 2013 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-23865689

RESUMO

Feline infectious peritonitis (FIP) is a fatal disease caused by feline coronavirus (FCoV) infection. FCoV can be divided into serotypes I and II. The virus that causes FIP (FIPV) is believed to occur sporadically and spread infrequently from cat to cat. Recently, an FIP outbreak from an animal shelter was confirmed in Taiwan. FCoV from all the cats in this shelter were analyzed to determine the epidemiology of this outbreak. Thirteen of 46 (28.2%) cats with typical signs of FIP were identified. Among them, seven cats were confirmed by necropsy and/or histopathological examinations. Despite the fact that more than one FCoV was identified in this multi-cat environment, the eight FIP cats were invariably found to be infected with a type II FCoV. Sequence analysis revealed that the type II FIPV detected from fecal samples, body effusions and granulomatous tissue homogenates from the cats that succumbed to FIP all harbored an identical recombination site in their S gene. Two of the cats that succumbed to FIP were found to harbor an identical nonsense mutation in the 3c gene. Fecal shedding of this type II virus in the effusive form of FIP can be detected up to six days before death. Taken together, our data demonstrate that horizontal transmission of FIPV is possible and that FIP cats can pose a potential risk to other cats living in the same environment.


Assuntos
Coronavirus Felino/classificação , Coronavirus Felino/isolamento & purificação , Surtos de Doenças/veterinária , Peritonite Infecciosa Felina/transmissão , Peritonite Infecciosa Felina/virologia , Genes Virais , Animais , Sequência de Bases , Gatos , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Fezes/virologia , Epidemiologia Molecular , Dados de Sequência Molecular , Mutação , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/veterinária , Alinhamento de Sequência/veterinária , Taiwan , Eliminação de Partículas Virais
13.
Biomaterials ; 34(32): 7895-904, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23886734

RESUMO

Total internal reflection microscopy combined with microfluidics and supported bilayers is a powerful, single particle tracking (SPT) platform for host-pathogen membrane fusion studies. But one major inadequacy of this platform has been capturing the complexity of the cell membrane, including membrane proteins. Because of this, viruses requiring proteinaceous receptors, or other unknown cellular co-factors, have been precluded from study. Here we describe a general method to integrate proteinaceous receptors and cellular components into supported bilayers for SPT fusion studies. This method is general to any enveloped virus-host cell pair, but demonstrated here for feline coronavirus (FCoV). Supported bilayers are formed from mammalian cell membrane vesicles that express feline aminopeptidase N (the viral receptor) using a cell blebbing technique. SPT is then used to identify fusion intermediates and measure membrane fusion kinetics for FCoV. Overall, the fusion results recapitulate what is observed in vivo, that coronavirus entry requires binding to specific receptors, a low-pH environment, and that membrane fusion is receptor- and protease-dependent. But this method also provides quantitative kinetic rate parameters for intermediate steps in the coronavirus fusion pathway, which to our knowledge have not been obtained before. Moreover, the platform offers versatile, precise control over the sequence of triggers for fusion; these triggers may define the fusion pathway, tissue tropism, and pathogenicity of coronaviruses. Systematically varying these triggers in this platform provides a new route to study how viruses rapidly adapt to other hosts, and to identify factors that led to the emergence of zoonotic viruses, such as human SARS-CoV and the newly emerging human MERS-CoV.


Assuntos
Coronavirus Felino/metabolismo , Fusão de Membrana/fisiologia , Receptores Virais/metabolismo , Animais , Antígenos CD13/metabolismo , Gatos , Linhagem Celular , Membrana Celular/metabolismo , Membrana Celular/virologia , Cricetinae , Humanos , Processamento de Imagem Assistida por Computador , Proteínas de Membrana/metabolismo , Técnicas Analíticas Microfluídicas , Microfluídica , Proteínas do Envelope Viral/metabolismo
14.
Vet Microbiol ; 162(2-4): 447-455, 2013 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-23182908

RESUMO

The ability to productively infect monocytes/macrophages is the most important difference between the low virulent feline enteric coronavirus (FECV) and the lethal feline infectious peritonitis virus (FIPV). In vitro, the replication of FECV in peripheral blood monocytes always drops after 12h post inoculation, while FIPV sustains its replication in the monocytes from 45% of the cats. The accessory proteins of feline coronaviruses have been speculated to play a prominent role in virulence as deletions were found to be associated with attenuated viruses. Still, no functions have been ascribed to them. In order to investigate if the accessory proteins of FIPV are important for sustaining its replication in monocytes, replication kinetics were determined for FIPV 79-1146 and its deletion mutants, lacking either accessory protein open reading frame 3abc (FIPV-Δ3), 7ab (FIPV-Δ7) or both (FIPV-Δ3Δ7). Results showed that the deletion mutants FIPV-Δ7 and FIPV-Δ3Δ7 could not maintain their replication, which was in sharp contrast to wt-FIPV. FIPV-Δ3 could still sustain its replication, but the percentage of infected monocytes was always lower compared to wt-FIPV. In conclusion, this study showed that ORF7 is crucial for FIPV replication in monocytes/macrophages, giving an explanation for its importance in vivo, its role in the development of FIP and its conservation in field strains. The effect of an ORF3 deletion was less pronounced, indicating only a supportive role of ORF3 encoded proteins during the infection of the in vivo target cell by FIPVs.


Assuntos
Doenças do Gato/sangue , Doenças do Gato/virologia , Infecções por Coronavirus/veterinária , Coronavirus Felino/fisiologia , Leucócitos Mononucleares/virologia , Proteínas Virais Reguladoras e Acessórias/fisiologia , Replicação Viral/fisiologia , Animais , Gatos , Linhagem Celular , Infecções por Coronavirus/sangue , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Coronavirus Felino/patogenicidade , Mutação , Proteínas do Nucleocapsídeo/biossíntese , Fases de Leitura Aberta , Proteínas Virais Reguladoras e Acessórias/metabolismo , Virulência , Replicação Viral/genética
15.
Emerg Infect Dis ; 18(7): 1089-95, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22709821

RESUMO

Coronaviruses are well known for their potential to change their host or tissue tropism, resulting in unpredictable new diseases and changes in pathogenicity; severe acute respiratory syndrome and feline coronaviruses, respectively, are the most recognized examples. Feline coronaviruses occur as 2 pathotypes: nonvirulent feline enteric coronaviruses (FECVs), which replicate in intestinal epithelium cells, and lethal feline infectious peritonitis viruses (FIPVs), which replicate in macrophages. Evidence indicates that FIPV originates from FECV by mutation, but consistent distinguishing differences have not been established. We sequenced the full genome of 11 viruses of each pathotype and then focused on the single most distinctive site by additionally sequencing hundreds of viruses in that region. As a result, we identified 2 alternative amino acid differences in the putative fusion peptide of the spike protein that together distinguish FIPV from FECV in >95% of cases. By these and perhaps other mutations, the virus apparently acquires its macrophage tropism and spreads systemically.


Assuntos
Coronavirus Felino/patogenicidade , Glicoproteínas de Membrana/genética , Mutação , Peptídeos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Gatos , Infecções por Coronavirus/virologia , Coronavirus Felino/genética , Coronavirus Felino/metabolismo , Peritonite Infecciosa Felina/virologia , Genoma Viral , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência , Análise de Sequência de DNA , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/metabolismo , Virulência/genética
16.
J Gen Virol ; 93(Pt 9): 1930-1934, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718568

RESUMO

In this study, the Japanese strain of type I feline infectious peritonitis virus (FIPV), C3663, was found to have a large deletion of 735 bp within the gene encoding the spike (S) protein, with a deduced loss of 245 aa of the N-terminal region of the S protein. This deletion is similar to that observed in porcine respiratory coronavirus (PRCoV) when compared to transmissible gastroenteritis virus, which correlates with reduced virulence. By analogy to PRCoV, we expected that the pathogenicity of C3663 may be attenuated in cats. However, two of four cats inoculated with C3663 died of FIP, and a third C3663-inoculated cat showed FIP lesions at 91 days after challenge. These results indicate that the 5'-terminal region of the S gene is not essential for the development of FIP.


Assuntos
Coronavirus Felino/genética , Coronavirus Felino/patogenicidade , Peritonite Infecciosa Felina/virologia , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Deleção de Sequência , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Motivos de Aminoácidos , Animais , Gatos , Coronavirus Felino/química , Coronavirus Felino/metabolismo , Masculino , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/metabolismo , Virulência
17.
Vaccine ; 29(9): 1791-800, 2011 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-21216312

RESUMO

The antibody-dependent enhancement (ADE) of feline infectious peritonitis virus (FIPV) infection has been recognized in experimentally infected cats, and cellular immunity is considered to play an important role in preventing the onset of feline infectious peritonitis (FIP). In the present study, we synthesized eighty-one kinds of peptides derived from the spike (S)2 domain of type I FIPV KU-2 strain, the S2 domain of type II FIPV 79-1146 strain, and the nucleocapcid (N) protein of FIPV KU-2 strain. To detect the T helper (Th)1 epitope, peripheral blood mononuclear cells (PBMCs) obtained from FIPV-infected cats were cultured with each peptide, and Th1-type immune responses were measured using feline interferon (fIFN)-γ production as an index. To detect the linear immunodominant antibody-binding epitope, we investigated the reactivity of plasma collected from FIPV-infected cats against each peptide by ELISA. Four and 2 peptides containing Th1 epitopes were identified in the heptad repeat (HR)1 and inter-helical (IH) regions of the S2 domain of type I FIPV, respectively, and these were located on the N-terminal side of the regions. In the S2 domain of type II FIPV, 2, 3, and 2 peptides containing Th1 epitopes were identified in the HR1, IH, and HR2 regions, respectively, and these were mainly located on the C-terminal side of the regions. In the S2 domain of type I FIPV, 3 and 7 peptides containing linear immunodominant antibody-binding epitopes were identified in the IH and HR2 regions, respectively. In the S2 domain of type II FIPV, 4 peptides containing linear immunodominant antibody-binding epitopes were identified in the HR2 region. The Th1 epitopes in the S2 domain of type I and II FIPV were located in different regions, but the linear immunodominant antibody-binding epitopes were mostly located in the HR2 region. Eight peptides containing Th1 epitopes were identified in N protein, and 3 peptides derived from residues 81 to 100 and 137 to 164 showed strong inductivity of fIFN-γ production in PBMCs isolated from type I FIPV- and type II FIPV-infected non-FIP cats. In N protein, 4 peptides containing linear immunodominant antibody-binding epitopes were identified, and 2 peptides derived from residues 345 to 372 showed strong reactivity with plasma of type I FIPV- and type II FIPV-infected cats. The Th1 and linear immunodominant antibody-binding epitopes were located at different positions in both the S2 domain and N protein. Our results may provide important information for the development of peptide-based vaccine against FIPV infection.


Assuntos
Anticorpos Antivirais/imunologia , Sítios de Ligação de Anticorpos , Coronavirus Felino/imunologia , Epitopos de Linfócito T/imunologia , Testes Genéticos/métodos , Epitopos Imunodominantes/imunologia , Proteínas do Nucleocapsídeo/imunologia , Células Th1/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/metabolismo , Gatos , Coronavirus Felino/metabolismo , Epitopos/imunologia , Epitopos/metabolismo , Epitopos de Linfócito T/metabolismo , Peritonite Infecciosa Felina/imunologia , Peritonite Infecciosa Felina/metabolismo , Epitopos Imunodominantes/metabolismo , Dados de Sequência Molecular , Proteínas do Nucleocapsídeo/metabolismo , Estrutura Terciária de Proteína , Células Th1/metabolismo
18.
J Virol ; 82(16): 8071-84, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18417574

RESUMO

The coronavirus family of positive-strand RNA viruses includes important pathogens of livestock, companion animals, and humans, including the severe acute respiratory syndrome coronavirus that was responsible for a worldwide outbreak in 2003. The unusually complex coronavirus replicase/transcriptase is comprised of 15 or 16 virus-specific subunits that are autoproteolytically derived from two large polyproteins. In line with bioinformatics predictions, we now show that feline coronavirus (FCoV) nonstructural protein 16 (nsp16) possesses an S-adenosyl-L-methionine (AdoMet)-dependent RNA (nucleoside-2'O)-methyltransferase (2'O-MTase) activity that is capable of cap-1 formation. Purified recombinant FCoV nsp16 selectively binds to short capped RNAs. Remarkably, an N7-methyl guanosine cap ((7Me)GpppAC(3-6)) is a prerequisite for binding. High-performance liquid chromatography analysis demonstrated that nsp16 mediates methyl transfer from AdoMet to the 2'O position of the first transcribed nucleotide, thus converting (7Me)GpppAC(3-6) into (7Me)GpppA(2')(O)(Me)C(3-6). The characterization of 11 nsp16 mutants supported the previous identification of residues K45, D129, K169, and E202 as the putative K-D-K-E catalytic tetrad of the enzyme. Furthermore, residues Y29 and F173 of FCoV nsp16, which may be the functional counterparts of aromatic residues involved in substrate recognition by the vaccinia virus MTase VP39, were found to be essential for both substrate binding and 2'O-MTase activity. Finally, the weak inhibition profile of different AdoMet analogues indicates that nsp16 has evolved an atypical AdoMet binding site. Our results suggest that coronavirus mRNA carries a cap-1, onto which 2'O methylation follows an order of events in which 2'O-methyl transfer must be preceded by guanine N7 methylation, with the latter step being performed by a yet-unknown N7-specific MTase.


Assuntos
Biologia Computacional/métodos , Coronavirus Felino/metabolismo , Metiltransferases/química , Capuzes de RNA/química , Proteínas não Estruturais Virais/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Gatos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Nucleotídeos/química , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , Proteínas não Estruturais Virais/fisiologia
19.
J Virol ; 81(3): 1261-73, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17093189

RESUMO

Feline coronavirus (FCoV), porcine transmissible gastroenteritis coronavirus (TGEV), canine coronavirus (CCoV), and human coronavirus HCoV-229E, which belong to the group 1 coronavirus, use aminopeptidase N (APN) of their natural host and feline APN (fAPN) as receptors. Using mouse-feline APN chimeras, we identified three small, discontinuous regions, amino acids (aa) 288 to 290, aa 732 to 746 (called R1), and aa 764 to 788 (called R2) in fAPN that determined the host ranges of these coronaviruses. Blockade of infection with anti-fAPN monoclonal antibody RG4 suggested that these three regions lie close together on the fAPN surface. Different residues in fAPN were required for infection with each coronavirus. HCoV-229E infection was blocked by an N-glycosylation sequon present between aa 288 to 290 in murine APN. TGEV required R1 of fAPN, while FCoV and CCoV required both R1 and R2 for entry. N740 and T742 in fAPN and the homologous R741 in human APN (hAPN) were key determinants of host range for FCoV, TGEV, and CCoV. Residue N740 in fAPN was essential only for CCoV receptor activity. A conservative T742V substitution or a T742R substitution in fAPN destroyed receptor activity for the pig, dog, and cat coronaviruses, while a T742S substitution retained these receptor activities. Thus, the hydroxyl on T742 is required for the coronavirus receptor activity of fAPN. In hAPN an R741T substitution caused a gain of receptor activity for TGEV but not for FCoV or CCoV. Therefore, entry and host range of these group 1 coronaviruses depend on the ability of the viral spike glycoproteins to recognize small, species-specific amino acid differences in the APN proteins of different species.


Assuntos
Antígenos CD13/metabolismo , Coronavirus/patogenicidade , Receptores Virais/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Substituição de Aminoácidos , Animais , Sítios de Ligação , Antígenos CD13/química , Antígenos CD13/genética , Gatos , Linhagem Celular , Coronavirus/metabolismo , Coronavirus Felino/metabolismo , Mutação , Receptores de Coronavírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...