Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Infect Dis ; 211(6): 889-97, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25057042

RESUMO

Middle East respiratory syndrome coronavirus (MERS-CoV) infection is associated with a high case-fatality rate, and the potential pandemic spread of the virus is a public health concern. The spike protein of MERS-CoV (MERS-S) facilitates viral entry into host cells, which depends on activation of MERS-S by cellular proteases. Proteolytic activation of MERS-S during viral uptake into target cells has been demonstrated. However, it is unclear whether MERS-S is also cleaved during S protein synthesis in infected cells and whether cleavage is required for MERS-CoV infectivity. Here, we show that MERS-S is processed by proprotein convertases in MERS-S-transfected and MERS-CoV-infected cells and that several RXXR motifs located at the border between the surface and transmembrane subunit of MERS-S are required for efficient proteolysis. However, blockade of proprotein convertases did not impact MERS-S-dependent transduction of target cells expressing high amounts of the viral receptor, DPP4, and did not modulate MERS-CoV infectivity. These results show that MERS-S is a substrate for proprotein convertases and demonstrate that processing by these enzymes is dispensable for S protein activation. Efforts to inhibit MERS-CoV infection by targeting host cell proteases should therefore focus on enzymes that process MERS-S during viral uptake into target cells.


Assuntos
Coronavirus/fisiologia , Pró-Proteína Convertases/antagonistas & inibidores , Glicoproteína da Espícula de Coronavírus/metabolismo , Sequência de Aminoácidos , Antivirais/farmacologia , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Dados de Sequência Molecular , Inibidores de Proteases/farmacologia , Processamento de Proteína Pós-Traducional , Proteólise , Glicoproteína da Espícula de Coronavírus/química , Internalização do Vírus
2.
J Virol ; 88(2): 1293-307, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24227843

RESUMO

The type II transmembrane serine proteases TMPRSS2 and HAT can cleave and activate the spike protein (S) of the severe acute respiratory syndrome coronavirus (SARS-CoV) for membrane fusion. In addition, these proteases cleave the viral receptor, the carboxypeptidase angiotensin-converting enzyme 2 (ACE2), and it was proposed that ACE2 cleavage augments viral infectivity. However, no mechanistic insights into this process were obtained and the relevance of ACE2 cleavage for SARS-CoV S protein (SARS-S) activation has not been determined. Here, we show that arginine and lysine residues within ACE2 amino acids 697 to 716 are essential for cleavage by TMPRSS2 and HAT and that ACE2 processing is required for augmentation of SARS-S-driven entry by these proteases. In contrast, ACE2 cleavage was dispensable for activation of the viral S protein. Expression of TMPRSS2 increased cellular uptake of soluble SARS-S, suggesting that protease-dependent augmentation of viral entry might be due to increased uptake of virions into target cells. Finally, TMPRSS2 was found to compete with the metalloprotease ADAM17 for ACE2 processing, but only cleavage by TMPRSS2 resulted in augmented SARS-S-driven entry. Collectively, our results in conjunction with those of previous studies indicate that TMPRSS2 and potentially related proteases promote SARS-CoV entry by two separate mechanisms: ACE2 cleavage, which might promote viral uptake, and SARS-S cleavage, which activates the S protein for membrane fusion. These observations have interesting implications for the development of novel therapeutics. In addition, they should spur efforts to determine whether receptor cleavage promotes entry of other coronaviruses, which use peptidases as entry receptors.


Assuntos
Proteínas ADAM/metabolismo , Peptidil Dipeptidase A/metabolismo , Serina Endopeptidases/metabolismo , Síndrome Respiratória Aguda Grave/enzimologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Internalização do Vírus , Proteínas ADAM/genética , Proteína ADAM17 , Motivos de Aminoácidos , Enzima de Conversão de Angiotensina 2 , Linhagem Celular , Humanos , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/genética , Processamento de Proteína Pós-Traducional , Proteólise , Receptores Virais/genética , Receptores Virais/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Serina Endopeptidases/genética , Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/metabolismo , Síndrome Respiratória Aguda Grave/virologia , Glicoproteína da Espícula de Coronavírus/genética
3.
J Virol ; 87(11): 6150-60, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23536651

RESUMO

Infection with human coronavirus 229E (HCoV-229E) is associated with the common cold and may result in pneumonia in immunocompromised patients. The viral spike (S) protein is incorporated into the viral envelope and mediates infectious entry of HCoV-229E into host cells, a process that depends on the activation of the S-protein by host cell proteases. However, the proteases responsible for HCoV-229E activation are incompletely defined. Here we show that the type II transmembrane serine proteases TMPRSS2 and HAT cleave the HCoV-229E S-protein (229E-S) and augment 229E-S-driven cell-cell fusion, suggesting that TMPRSS2 and HAT can activate 229E-S. Indeed, engineered expression of TMPRSS2 and HAT rendered 229E-S-driven virus-cell fusion insensitive to an inhibitor of cathepsin L, a protease previously shown to facilitate HCoV-229E infection. Inhibition of endogenous cathepsin L or TMPRSS2 demonstrated that both proteases can activate 229E-S for entry into cells that are naturally susceptible to infection. In addition, evidence was obtained that activation by TMPRSS2 rescues 229E-S-dependent cell entry from inhibition by IFITM proteins. Finally, immunohistochemistry revealed that TMPRSS2 is coexpressed with CD13, the HCoV-229E receptor, in human airway epithelial (HAE) cells, and that CD13(+) TMPRSS2(+) cells are preferentially targeted by HCoV-229E, suggesting that TMPRSS2 can activate HCoV-229E in infected humans. In sum, our results indicate that HCoV-229E can employ redundant proteolytic pathways to ensure its activation in host cells. In addition, our observations and previous work suggest that diverse human respiratory viruses are activated by TMPRSS2, which may constitute a target for antiviral intervention.


Assuntos
Catepsinas/metabolismo , Coronavirus Humano 229E/fisiologia , Infecções por Coronavirus/enzimologia , Mucosa Respiratória/enzimologia , Serina Endopeptidases/metabolismo , Internalização do Vírus , Catepsinas/genética , Linhagem Celular , Coronavirus Humano 229E/genética , Infecções por Coronavirus/genética , Infecções por Coronavirus/virologia , Expressão Gênica , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Mucosa Respiratória/virologia , Serina Endopeptidases/genética
4.
J Virol ; 87(10): 5502-11, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23468491

RESUMO

The novel human coronavirus EMC (hCoV-EMC), which recently emerged in Saudi Arabia, is highly pathogenic and could pose a significant threat to public health. The elucidation of hCoV-EMC interactions with host cells is critical to our understanding of the pathogenesis of this virus and to the identification of targets for antiviral intervention. Here we investigated the viral and cellular determinants governing hCoV-EMC entry into host cells. We found that the spike protein of hCoV-EMC (EMC-S) is incorporated into lentiviral particles and mediates transduction of human cell lines derived from different organs, including the lungs, kidneys, and colon, as well as primary human macrophages. Expression of the known coronavirus receptors ACE2, CD13, and CEACAM1 did not facilitate EMC-S-driven transduction, suggesting that hCoV-EMC uses a novel receptor for entry. Directed protease expression and inhibition analyses revealed that TMPRSS2 and endosomal cathepsins activate EMC-S for virus-cell fusion and constitute potential targets for antiviral intervention. Finally, EMC-S-driven transduction was abrogated by serum from an hCoV-EMC-infected patient, indicating that EMC-S-specific neutralizing antibodies can be generated in patients. Collectively, our results indicate that hCoV-EMC uses a novel receptor for protease-activated entry into human cells and might be capable of extrapulmonary spread. In addition, they define TMPRSS2 and cathepsins B and L as potential targets for intervention and suggest that neutralizing antibodies contribute to the control of hCoV-EMC infection.


Assuntos
Anticorpos Neutralizantes/sangue , Coronavirus/fisiologia , Interações Hospedeiro-Patógeno , Glicoproteínas de Membrana/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Internalização do Vírus , Anticorpos Antivirais/sangue , Catepsinas/metabolismo , Coronavirus/isolamento & purificação , Coronavirus/patogenicidade , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Humanos , Glicoproteínas de Membrana/imunologia , Receptores de Coronavírus , Arábia Saudita , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus , Transdução Genética , Proteínas do Envelope Viral/imunologia , Tropismo Viral
5.
Emerg Infect Dis ; 19(12): 2034-6, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24274664

RESUMO

We used a lentiviral vector bearing the viral spike protein to detect neutralizing antibodies against Middle East respiratory syndrome coronavirus (MERS-CoV) in persons from the Eastern Province of Saudi Arabia. None of the 268 samples tested displayed neutralizing activity, which suggests that MERS-CoV infections in humans are infrequent in this province.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/imunologia , Coronavirus/imunologia , Adulto , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Infecções por Coronavirus/transmissão , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Arábia Saudita/epidemiologia , Estudos Soroepidemiológicos , Adulto Jovem
6.
PLoS One ; 12(6): e0179177, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28636671

RESUMO

The spike (S) protein of severe acute respiratory syndrome coronavirus (SARS-CoV) mediates viral entry into target cells. Cleavage and activation of SARS S by a host cell protease is essential for infectious viral entry and the responsible enzymes are potential targets for antiviral intervention. The type II transmembrane serine protease TMPRSS2 cleaves and activates SARS S in cell culture and potentially also in the infected host. Here, we investigated which determinants in SARS S control cleavage and activation by TMPRSS2. We found that SARS S residue R667, a previously identified trypsin cleavage site, is also required for S protein cleavage by TMPRSS2. The cleavage fragments produced by trypsin and TMPRSS2 differed in their decoration with N-glycans, suggesting that these proteases cleave different SARS S glycoforms. Although R667 was required for SARS S cleavage by TMPRSS2, this residue was dispensable for TMPRSS2-mediated S protein activation. Conversely, residue R797, previously reported to be required for SARS S activation by trypsin, was dispensable for S protein cleavage but required for S protein activation by TMPRSS2. Collectively, these results show that different residues in SARS S control cleavage and activation by TMPRSS2, suggesting that these processes are more complex than initially appreciated.


Assuntos
Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Mutação/genética , Transporte Proteico , Serina Endopeptidases/genética , Glicoproteína da Espícula de Coronavírus/genética , Internalização do Vírus
7.
Antiviral Res ; 100(3): 605-14, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24121034

RESUMO

The severe acute respiratory syndrome (SARS) pandemic revealed that zoonotic transmission of animal coronaviruses (CoV) to humans poses a significant threat to public health and warrants surveillance and the development of countermeasures. The activity of host cell proteases, which cleave and activate the SARS-CoV spike (S) protein, is essential for viral infectivity and constitutes a target for intervention. However, the identities of the proteases involved have been unclear. Pioneer studies identified cathepsins and type II transmembrane serine proteases as cellular activators of SARS-CoV and demonstrated that several emerging viruses might exploit these enzymes to promote their spread. Here, we will review the proteolytic systems hijacked by SARS-CoV for S protein activation, we will discuss their contribution to viral spread in the host and we will outline antiviral strategies targeting these enzymes. This paper forms part of a series of invited articles in Antiviral Research on "From SARS to MERS: 10years of research on highly pathogenic human coronaviruses.''


Assuntos
Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Enzima de Conversão de Angiotensina 2 , Catepsinas/metabolismo , Doenças Transmissíveis Emergentes , Infecções por Coronavirus/virologia , Endossomos/metabolismo , Humanos , Pulmão/enzimologia , Pulmão/patologia , Pulmão/virologia , Proteínas de Membrana/metabolismo , Modelos Biológicos , Peptidil Dipeptidase A/metabolismo , Proteólise , Serina Endopeptidases/metabolismo , Serina Proteases/metabolismo , Síndrome Respiratória Aguda Grave/patologia , Internalização do Vírus , Replicação Viral
8.
PLoS One ; 7(4): e35876, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22558251

RESUMO

The type II transmembrane serine proteases TMPRSS2 and HAT activate influenza viruses and the SARS-coronavirus (TMPRSS2) in cell culture and may play an important role in viral spread and pathogenesis in the infected host. However, it is at present largely unclear to what extent these proteases are expressed in viral target cells in human tissues. Here, we show that both HAT and TMPRSS2 are coexpressed with 2,6-linked sialic acids, the major receptor determinant of human influenza viruses, throughout the human respiratory tract. Similarly, coexpression of ACE2, the SARS-coronavirus receptor, and TMPRSS2 was frequently found in the upper and lower aerodigestive tract, with the exception of the vocal folds, epiglottis and trachea. Finally, activation of influenza virus was conserved between human, avian and porcine TMPRSS2, suggesting that this protease might activate influenza virus in reservoir-, intermediate- and human hosts. In sum, our results show that TMPRSS2 and HAT are expressed by important influenza and SARS-coronavirus target cells and could thus support viral spread in the human host.


Assuntos
Reservatórios de Doenças/veterinária , Trato Gastrointestinal/enzimologia , Influenza Humana/enzimologia , Sistema Respiratório/enzimologia , Serina Endopeptidases/genética , Síndrome Respiratória Aguda Grave/enzimologia , Enzima de Conversão de Angiotensina 2 , Animais , Aves , Linhagem Celular , Reservatórios de Doenças/virologia , Ativação Enzimática , Trato Gastrointestinal/virologia , Expressão Gênica , Humanos , Influenza Humana/genética , Influenza Humana/transmissão , Influenza Humana/virologia , Orthomyxoviridae/fisiologia , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Receptores Virais/genética , Receptores Virais/metabolismo , Sistema Respiratório/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , Serina Endopeptidases/metabolismo , Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/virologia , Ácidos Siálicos/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA