Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artigo em Inglês | MEDLINE | ID: covidwho-2287228

RESUMO

Porcine epidemic diarrhea (PED) is an acute and severe atrophic enteritis caused by porcine epidemic diarrhea virus (PEDV) that infects pigs and makes huge economic losses to the global swine industry. Previously, researchers have believed that porcine aminopeptidase-N (pAPN) was the primary receptor for PEDV, but it has been found that PEDV can infect pAPN knockout pigs. Currently, the functional receptor for PEDV remains unspecified. In the present study, we performed virus overlay protein binding assay (VOPBA), found that ATP1A1 was the highest scoring protein in the mass spectrometry results, and confirmed that the CT structural domain of ATP1A1 interacts with PEDV S1. First, we investigated the effect of ATP1A1 on PEDV replication. Inhibition of hosts ATP1A1 protein expression using small interfering RNA (siRNAs) significantly reduced the cells susceptibility to PEDV. The ATP1A1-specific inhibitors Ouabain (a cardiac steroid) and PST2238 (a digitalis toxin derivative), which specifically bind ATP1A1, could block the ATP1A1 protein internalization and degradation, and consequently reduce the infection rate of host cells by PEDV significantly. Additionally, as expected, overexpression of ATP1A1 notably enhanced PEDV infection. Next, we observed that PEDV infection of target cells resulted in upregulation of ATP1A1 at the mRNA and protein levels. Furthermore, we found that the host protein ATP1A1 was involved in PEDV attachment and co-localized with PEDV S1 protein in the early stage of infection. In addition, pretreatment of IPEC-J2 and Vero-E6 cells with ATP1A1 mAb significantly reduced PEDV attachment. Our observations provided a perspective on identifying key factors in PEDV infection, and may provide valuable targets for PEDV infection, PEDV functional receptor, related pathogenesis, and the development of new antiviral drugs.


Assuntos
Infecções por Coronavirus , Interações Hospedeiro-Patógeno , Vírus da Diarreia Epidêmica Suína , ATPase Trocadora de Sódio-Potássio , Doenças dos Suínos , Animais , Antígenos CD13/metabolismo , Chlorocebus aethiops , Vírus da Diarreia Epidêmica Suína/fisiologia , Receptores Virais/metabolismo , RNA de Cadeia Dupla , RNA Interferente Pequeno , Suínos , Doenças dos Suínos/metabolismo , Células Vero , Ligação Viral , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , ATPase Trocadora de Sódio-Potássio/metabolismo
2.
J Virol ; 97(2): e0194722, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: covidwho-2193457

RESUMO

Members of deltacoronavirus (DCoV) have mostly been identified in diverse avian species as natural reservoirs, though the porcine DCoV (PDCoV) is a major swine enteropathogenic virus with global spread. The important role of aminopeptidase N (APN) orthologues from various mammalian and avian species in PDCoV cellular entry and interspecies transmission has been revealed recently. In this study, comparative analysis indicated that three avian DCoVs, bulbul DCoV HKU11, munia DCoV HKU13, and sparrow DCoV HKU17 (Chinese strain), and PDCoV in the subgenera Buldecovirus are grouped together at whole-genome levels; however, the spike (S) glycoprotein and its S1 subunit of HKU17 are more closely related to night heron DCoV HKU19 in Herdecovirus. Nevertheless, the S1 protein of HKU11, HKU13, or HKU17 bound to or interacted with chicken APN (chAPN) or porcine APN (pAPN) by flow cytometry analysis of cell surface expression of APN and by coimmunoprecipitation in APN-overexpressing cells. Expression of chAPN or pAPN allowed entry of pseudotyped lentiviruses with the S proteins from HKU11, HKU13 and HKU17 into nonsusceptible cells and natural avian and porcine cells, which could be inhibited by the antibody against APN or anti-PDCoV-S1. APN knockdown by siRNA or knockout by CRISPR/Cas9 in chicken or swine cell lines significantly or almost completely blocked infection of these pseudoviruses. Hence, we demonstrate that HKU11, HKU13, and HKU17 with divergent S genes likely engage chAPN or pAPN to enter the cells, suggesting a potential interspecies transmission from wild birds to poultry and from birds to mammals by certain avian DCoVs. IMPORTANCE The receptor usage of avian deltacoronaviruses (DCoVs) has not been investigated thus far, though porcine deltacoronavirus (PDCoV) has been shown to utilize aminopeptidase N (APN) as a cell receptor. We report here that chicken or porcine APN also mediates cellular entry by three avian DCoV (HKU11, HKU13, and HKU17) spike pseudoviruses, and the S1 subunit of three avian DCoVs binds to APN in vitro and in the surface of avian and porcine cells. The results fill the gaps in knowledge about the avian DCoV receptor and elucidate important insights for the monitoring and prevention of potential interspecies transmission of certain avian DCoVs. In view of the diversity of DCoVs, whether this coronavirus genus will cause novel virus to emerge in other mammals from birds, are worthy of further surveillance and investigation.


Assuntos
Antígenos CD13 , Deltacoronavirus , Glicoproteína da Espícula de Coronavírus , Internalização do Vírus , Animais , Antígenos CD13/genética , Antígenos CD13/metabolismo , Galinhas/metabolismo , Infecções por Coronavirus , Deltacoronavirus/metabolismo , Suínos , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Lentivirus/genética , Lentivirus/metabolismo
3.
J Med Virol ; 94(12): 5723-5738, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: covidwho-1971295

RESUMO

Porcine deltacoronavirus (PDCoV) is a novel coronavirus that causes diarrhea in suckling piglets and has the potential for cross-species transmission, posing a threat to animal and human health. However, the susceptibility profile of different species of mice to PDCoV infection and its evolutionary characteristics are still unclear. In the current study, we found that BALB/c and Kunming mice are susceptible to PDCoV. Our results showed that there were obvious lesions in intestinal and lung tissues from the infected mice. PDCoV RNAs were detected in the lung, kidney, and intestinal tissues from the infected mice of both strains, and there existed wider tissue tropism in the PDCoV-infected BALB/c mice. The RNA and protein levels of aminopeptidase N from mice were relatively high in the kidney and intestinal tissues and obviously increased after PDCoV infection. The viral-specific IgG and neutralizing antibodies against PDCoV were detected in the serum of infected mice. An interesting finding was that two key amino acid mutations, D138H and Q641K, in the S protein were identified in the PDCoV-infected mice. The essential roles of these two mutations for PDCoV-adaptive evolution were confirmed by cryo-electron microscope structure model analysis. The evolutionary characteristics of PDCoV among Deltacoronaviruses (δ-CoVs) were further analyzed. δ-CoVs from multiple mammals are closely related based on the phylogenetic analysis. The codon usage analysis demonstrated that similar codon usage patterns were used by most of the mammalian δ-CoVs at the global codon, synonymous codon, and amino acid usage levels. These results may provide more insights into the evolution, host ranges, and cross-species potential of PDCoV.


Assuntos
COVID-19 , Doenças dos Suínos , Aminoácidos , Animais , Anticorpos Neutralizantes , Antígenos CD13/genética , Antígenos CD13/metabolismo , Deltacoronavirus , Humanos , Imunoglobulina G , Mamíferos/metabolismo , Camundongos , Filogenia , RNA , Suínos
4.
Front Immunol ; 13: 925922, 2022.
Artigo em Inglês | MEDLINE | ID: covidwho-1933699

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
5.
Cell ; 185(13): 2279-2291.e17, 2022 06 23.
Artigo em Inglês | MEDLINE | ID: covidwho-1866951

RESUMO

The isolation of CCoV-HuPn-2018 from a child respiratory swab indicates that more coronaviruses are spilling over to humans than previously appreciated. We determined the structures of the CCoV-HuPn-2018 spike glycoprotein trimer in two distinct conformational states and showed that its domain 0 recognizes sialosides. We identified that the CCoV-HuPn-2018 spike binds canine, feline, and porcine aminopeptidase N (APN) orthologs, which serve as entry receptors, and determined the structure of the receptor-binding B domain in complex with canine APN. The introduction of an oligosaccharide at position N739 of human APN renders cells susceptible to CCoV-HuPn-2018 spike-mediated entry, suggesting that single-nucleotide polymorphisms might account for viral detection in some individuals. Human polyclonal plasma antibodies elicited by HCoV-229E infection and a porcine coronavirus monoclonal antibody inhibit CCoV-HuPn-2018 spike-mediated entry, underscoring the cross-neutralizing activity among ɑ-coronaviruses. These data pave the way for vaccine and therapeutic development targeting this zoonotic pathogen representing the eighth human-infecting coronavirus.


Assuntos
Coronavirus Humano 229E , Infecções por Coronavirus , Coronavirus , Animais , Antígenos CD13/química , Antígenos CD13/metabolismo , Gatos , Linhagem Celular , Coronavirus/metabolismo , Coronavirus Humano 229E/metabolismo , Cães , Humanos , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Suínos
6.
Biosci Rep ; 41(12)2021 12 22.
Artigo em Inglês | MEDLINE | ID: covidwho-1592575

RESUMO

Parasporin-2Aa1 (PS2Aa1) is a toxic protein of 37 KDa (30 kDa, activated form produced by proteolysis) that was shown to be cytotoxic against specific human cancer cells, although its mechanism of action has not been elucidated yet. In order to study the role of some native peptide fragments of proteins on anticancer activity, here we investigated the cytotoxic effect of peptide fragments from domain-1 of PS2Aa1 and one of the loops present in the binding region of the virus spike protein from Alphacoronavirus (HCoV-229E), the latter according to scientific reports, who showed interaction with the human APN (h-APN) receptor, evidence corroborated through computational simulations, and thus being possible active against colon cancer cells. Peptides namely P264-G274, Loop1-PS2Aa, and Loop2-PS2Aa were synthesized using the Fmoc solid-phase synthesis and characterized by mass spectrometry (MS). Additionally, one region from loop 1 of HCoV-229E, Loop1-HCoV-229E, was also synthesized and characterized. The A4W-GGN5 anticancer peptide and 5-fluorouracil (5-FU) were taken as a control in all experiments. Circular dichroism revealed an α-helix structure for the peptides derived from PS2Aa1 (P264-G274, Loop1-PS2Aa, and Loop2-PS2Aa) and ß-laminar structure for the peptide derived from Alphacoronavirus spike protein Loop1-HCoV-229E. Peptides showed a hemolysis percentage of less than 20% at 100 µM concentration. Besides, peptides exhibited stronger anticancer activity against SW480 and SW620 cells after exposure for 48 h. Likewise, these compounds showed significantly lower toxicity against normal cells CHO-K1. The results suggest that native peptide fragments from Ps2Aa1 may be optimized as a novel potential cancer-therapeutic agents.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Endotoxinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Glicoproteína da Espícula de Coronavírus/farmacologia , Alphacoronavirus , Animais , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Antígenos CD13/metabolismo , Células CHO , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Cricetulus , Endotoxinas/toxicidade , Hemólise/efeitos dos fármacos , Humanos , Simulação de Acoplamento Molecular , Fragmentos de Peptídeos/síntese química , Fragmentos de Peptídeos/toxicidade , Conformação Proteica em alfa-Hélice , Carneiro Doméstico , Glicoproteína da Espícula de Coronavírus/toxicidade , Relação Estrutura-Atividade
7.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188641, 2021 12.
Artigo em Inglês | MEDLINE | ID: covidwho-1474353

RESUMO

Aminopeptidase N (APN/CD13) is a multifunctional glycoprotein that acts as a peptidase, receptor, and signalling molecule in a tissue-dependent manner. The activities of APN have been implicated in the progression of many cancers, pointing toward significant therapeutic potential for cancer treatment. However, despite the tumour-specific functions of this protein that have been uncovered, the ubiquitous nature of its expression in normal tissues as generally reported remains a limitation to the potential utility of APN as a target for cancer therapeutics and drug discovery. With this in mind, we have extensively explored the literature, and present a comprehensive review that for the first-time provides evidence to support the suggestion that tumour-expressed APN may in fact be unique in structure, function, substrate specificity and activity, contrary to its nature in normal tissues. The review also focuses on the biology of APN, and its "moonlighting" functional roles in both normal physiology and cancer development. Several APN-targeting approaches that have been explored over recent decades as therapeutic strategies in cancer treatment, including APN-targeting agents reported both in preclinical and clinical studies, are also extensively discussed. This review concludes by posing critical questions about APN that remain unanswered and unexplored, hence providing opportunities for further research.


Assuntos
Antígenos CD13/metabolismo , Neoplasias/fisiopatologia , Peptídeo Hidrolases/metabolismo , Humanos
8.
Nat Commun ; 12(1): 141, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: covidwho-1387322

RESUMO

Coronaviruses spike (S) glycoproteins mediate viral entry into host cells by binding to host receptors. However, how the S1 subunit undergoes conformational changes for receptor recognition has not been elucidated in Alphacoronavirus. Here, we report the cryo-EM structures of the HCoV-229E S trimer in prefusion state with two conformations. The activated conformation may pose the potential exposure of the S1-RBDs by decreasing of the interaction area between the S1-RBDs and the surrounding S1-NTDs and S1-RBDs compared to the closed conformation. Furthermore, structural comparison of our structures with the previously reported HCoV-229E S structure showed that the S trimers trended to open the S2 subunit from the closed conformation to open conformation, which could promote the transition from pre- to postfusion. Our results provide insights into the mechanisms involved in S glycoprotein-mediated Alphacoronavirus entry and have implications for vaccine and therapeutic antibody design.


Assuntos
Antígenos CD13/metabolismo , Coronavirus Humano 229E/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Linhagem Celular Tumoral , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/virologia , Microscopia Crioeletrônica , Humanos , Modelos Moleculares , Conformação Proteica em alfa-Hélice , Multimerização Proteica , Estrutura Quaternária de Proteína , Subunidades Proteicas/metabolismo , Glicoproteína da Espícula de Coronavírus/ultraestrutura
9.
Virulence ; 12(1): 1111-1121, 2021 12.
Artigo em Inglês | MEDLINE | ID: covidwho-1243446

RESUMO

Coronaviruses and influenza viruses are circulating in humans and animals all over the world. Co-infection with these two viruses may aggravate clinical signs. However, the molecular mechanisms of co-infections by these two viruses are incompletely understood. In this study, we applied air-liquid interface (ALI) cultures of well-differentiated porcine tracheal epithelial cells (PTECs) to analyze the co-infection by a swine influenza virus (SIV, H3N2 subtype) and porcine respiratory coronavirus (PRCoV) at different time intervals. Our results revealed that in short-term intervals, prior infection by influenza virus caused complete inhibition of coronavirus infection, while in long-term intervals, some coronavirus replication was detectable. The influenza virus infection resulted in (i) an upregulation of porcine aminopeptidase N, the cellular receptor for PRCoV and (ii) in the induction of an innate immune response which was responsible for the inhibition of PRCoV replication. By contrast, prior infection by coronavirus only caused a slight inhibition of influenza virus replication. Taken together, the timing and the order of virus infection are important determinants in co-infections. This study is the first to show the impact of SIV and PRCoV co- and super-infection on the cellular level. Our results have implications also for human viruses, including potential co-infections by SARS-CoV-2 and seasonal influenza viruses.


Assuntos
Células Epiteliais/virologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Coronavirus Respiratório Porcino/fisiologia , Interferência Viral , Animais , Antígenos CD13/metabolismo , Células Cultivadas , Coinfecção/virologia , Infecções por Coronavirus/virologia , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Imunidade Inata , Infecções por Orthomyxoviridae/virologia , Suínos , Traqueia/citologia , Replicação Viral
10.
Viruses ; 12(11)2020 10 23.
Artigo em Inglês | MEDLINE | ID: covidwho-895404

RESUMO

Porcine respiratory coronavirus (PRCoV) infects the epithelial cells in the respiratory tract of pigs, causing a mild respiratory disease. We applied air-liquid interface (ALI) cultures of well-differentiated porcine airway cells to mimic the respiratory tract epithelium in vitro and use it for analyzing the infection by PRCoV. As reported for most coronaviruses, virus entry and virus release occurred mainly via the apical membrane domain. A novel finding was that PRCoV preferentially targets non-ciliated and among them the non-mucus-producing cells. Aminopeptidase N (APN), the cellular receptor for PRCoV was also more abundantly expressed on this type of cell suggesting that APN is a determinant of the cell tropism. Interestingly, differentiation-dependent differences were found both in the expression of pAPN and the susceptibility to PRCoV infection. Cells in an early differentiation stage express higher levels of pAPN and are more susceptible to infection by PRCoV than are well-differentiated cells. A difference in the susceptibility to infection was also detected when tracheal and bronchial cells were compared. The increased susceptibility to infection of bronchial epithelial cells was, however, not due to an increased abundance of APN on the cell surface. Our data reveal a complex pattern of infection in porcine differentiated airway epithelial cells that could not be elucidated with immortalized cell lines. The results are expected to have relevance also for the analysis of other respiratory viruses.


Assuntos
Antígenos CD13/metabolismo , Células Epiteliais/metabolismo , Coronavirus Respiratório Porcino/fisiologia , Receptores Virais/metabolismo , Mucosa Respiratória/virologia , Tropismo Viral , Animais , Brônquios/metabolismo , Brônquios/virologia , Diferenciação Celular , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/virologia , Suínos , Traqueia/metabolismo , Traqueia/virologia , Internalização do Vírus , Liberação de Vírus , Replicação Viral
12.
Respir Investig ; 58(3): 155-168, 2020 May.
Artigo em Inglês | MEDLINE | ID: covidwho-154704

RESUMO

BACKGROUND: Coronavirus 229E (HCoV-229E), one of the causes of the common cold, exacerbates chronic obstructive pulmonary disease (COPD) and bronchial asthma. Long-acting muscarinic antagonists and ß2-agonists and inhaled corticosteroids inhibit the exacerbation of COPD and bronchial asthma caused by infection with viruses, including HCoV-229E. However, the effects of these drugs on HCoV-229E replication and infection-induced inflammation in the human airway are unknown. METHODS: Primary human nasal (HNE) and tracheal (HTE) epithelial cell cultures were infected with HCoV-229E. RESULTS: Pretreatment of HNE and HTE cells with glycopyrronium or formoterol decreased viral RNA levels and/or titers, the expression of the HCoV-229E receptor CD13, the number and fluorescence intensity of acidic endosomes where HCoV-229E RNA enters the cytoplasm, and the infection-induced production of cytokines, including IL-6, IL-8, and IFN-ß. Treatment of the cells with the CD13 inhibitor 2'2'-dipyridyl decreased viral titers. Pretreatment of the cells with a combination of three drugs (glycopyrronium, formoterol, and budesonide) exerted additive inhibitory effects on viral titers and cytokine production. Pretreatment of HNE cells with glycopyrronium or formoterol reduced the susceptibility to infection, and pretreatment with the three drugs inhibited activation of nuclear factor-kappa B p50 and p65 proteins. Pretreatment with formoterol increased cAMP levels and treatment with cAMP decreased viral titers, CD13 expression, and the fluorescence intensity of acidic endosomes. CONCLUSIONS: These findings suggest that glycopyrronium, formoterol, and a combination of glycopyrronium, formoterol, and budesonide inhibit HCoV-229E replication partly by inhibiting receptor expression and/or endosomal function and that these drugs modulate infection-induced inflammation in the airway.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Budesonida/farmacologia , Coronavirus/fisiologia , Citocinas/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Fumarato de Formoterol/farmacologia , Glicopirrolato/farmacologia , Antagonistas Muscarínicos/farmacologia , Mucosa Nasal/citologia , Traqueia/citologia , Replicação Viral/efeitos dos fármacos , Antígenos CD13/metabolismo , Células Cultivadas , Humanos
13.
Viruses ; 12(4)2020 04 05.
Artigo em Inglês | MEDLINE | ID: covidwho-31709

RESUMO

Porcine epidemic diarrhea virus (PEDV) and transmissible gastroenteritis virus (TGEV) have been reported to use aminopeptidase N (APN) as a cellular receptor. Recently, the role of APN as a receptor for PEDV has been questioned. In our study, the role of APN in PEDV and TGEV infections was studied in primary porcine enterocytes. After seven days of cultivation, 89% of enterocytes presented microvilli and showed a two- to five-fold higher susceptibility to PEDV and TGEV. A significant increase of PEDV and TGEV infection was correlated with a higher expression of APN, which was indicative that APN plays an important role in porcine coronavirus infections. However, PEDV and TGEV infected both APN positive and negative enterocytes. PEDV and TGEV Miller showed a higher infectivity in APN positive cells than in APN negative cells. In contrast, TGEV Purdue replicated better in APN negative cells. These results show that an additional receptor exists, different from APN for porcine coronaviruses. Subsequently, treatment of enterocytes with neuraminidase (NA) had no effect on infection efficiency of TGEV, implying that terminal cellular sialic acids (SAs) are no receptor determinants for TGEV. Treatment of TGEV with NA significantly enhanced the infection which shows that TGEV is masked by SAs.


Assuntos
Antígenos CD13/metabolismo , Gastroenterite Suína Transmissível/patologia , Vírus da Diarreia Epidêmica Suína/metabolismo , Receptores Virais/metabolismo , Ácidos Siálicos/metabolismo , Vírus da Gastroenterite Transmissível/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Infecções por Coronavirus/patologia , Infecções por Coronavirus/veterinária , Enterócitos/virologia , Hidrocortisona/farmacologia , Insulina/farmacologia , Mucosa Respiratória/virologia , Espermidina/farmacologia , Suínos , Células Vero , Ligação Viral , Replicação Viral/efeitos dos fármacos
14.
Nat Microbiol ; 5(4): 562-569, 2020 04.
Artigo em Inglês | MEDLINE | ID: covidwho-1966

RESUMO

Over the past 20 years, several coronaviruses have crossed the species barrier into humans, causing outbreaks of severe, and often fatal, respiratory illness. Since SARS-CoV was first identified in animal markets, global viromics projects have discovered thousands of coronavirus sequences in diverse animals and geographic regions. Unfortunately, there are few tools available to functionally test these viruses for their ability to infect humans, which has severely hampered efforts to predict the next zoonotic viral outbreak. Here, we developed an approach to rapidly screen lineage B betacoronaviruses, such as SARS-CoV and the recent SARS-CoV-2, for receptor usage and their ability to infect cell types from different species. We show that host protease processing during viral entry is a significant barrier for several lineage B viruses and that bypassing this barrier allows several lineage B viruses to enter human cells through an unknown receptor. We also demonstrate how different lineage B viruses can recombine to gain entry into human cells, and confirm that human ACE2 is the receptor for the recently emerging SARS-CoV-2.


Assuntos
Betacoronavirus/fisiologia , Peptidil Dipeptidase A/metabolismo , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Enzima de Conversão de Angiotensina 2 , Animais , Betacoronavirus/química , Betacoronavirus/classificação , Antígenos CD13/metabolismo , COVID-19 , Linhagem Celular , Infecções por Coronavirus/metabolismo , Infecções por Coronavirus/virologia , Dipeptidil Peptidase 4/metabolismo , Humanos , Mutação , Pandemias , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/genética , Pneumonia Viral/metabolismo , Pneumonia Viral/virologia , Domínios Proteicos , Receptores de Coronavírus , Receptores Virais/química , Receptores Virais/genética , Proteínas Recombinantes de Fusão/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/química , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Tripsina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA