Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Emerg Microbes Infect ; 12(1): 2207688, 2023 Dec.
Article in English | MEDLINE | ID: covidwho-2320534

ABSTRACT

ABSTRACTPorcine deltacoronavirus (PDCoV) is an emerging enteric coronavirus that has been reported to infect a variety of animals and even humans. Cell-cell fusion has been identified as an alternative pathway for the cell-to-cell transmission of certain viruses, but the ability of PDCoV to exploit this transmission model, and the relevant mechanisms, have not been fully elucidated. Herein, we provide evidence that cell-to-cell transmission is the main mechanism supporting PDCoV spread in cell culture and that this efficient spread model is mediated by spike glycoprotein-driven cell-cell fusion. We found that PDCoV efficiently spread to non-susceptible cells via cell-to-cell transmission, and demonstrated that functional receptor porcine aminopeptidase N and cathepsins in endosomes are involved in the cell-to-cell transmission of PDCoV. Most importantly, compared with non-cell-to-cell infection, the cell-to-cell transmission of PDCoV was resistant to neutralizing antibodies and immune sera that potently neutralized free viruses. Taken together, our study revealed key characteristics of the cell-to-cell transmission of PDCoV and provided new insights into the mechanism of PDCoV infection.


Subject(s)
Coronavirus Infections , Coronavirus , Swine Diseases , Humans , Animals , Swine , Deltacoronavirus , Coronavirus/physiology , Antibodies, Neutralizing , Coronavirus Infections/veterinary
2.
Springer Proceedings in Materials ; 21:103-109, 2023.
Article in English | Scopus | ID: covidwho-2250059

ABSTRACT

Coronaviruses (CoVs) display prevalence and great cross-transmissibility due to diverse receptor recognition and high mutability of their spike proteins. In this study, spike protein interactions that influence coronavirus evolution and complex virology were determined. To establish coronavirus classification, phylogenetic analysis of spike proteins based on maximum likelihood was performed using MEGA X. To identify the most suitable interactions between spike proteins and vertebrate cell receptors, molecular docking between full spike proteins and angiotensin-converting enzyme 2 (ACE2), aminopeptidase N (APN), and dipeptidyl peptidase 4 (DPP4) receptors from vertebrates was performed using Hex, a 6D spherical polar Fourier (SPF) transform-based software. Results supported the current coronavirus taxonomy, and molecular docking showed that highly different classes recognized similar receptors. Specifically, spike protein of Munia CoV with cat ACE2 (E = -478.3 kcal/mol) and for SARS-CoV-2 variants, spike protein of alpha with chicken ACE2 (E = -349.6 kcal/mol) formed the most suitable interactions. SARS-CoV-2 variants showed additional affinity toward chicken and cat receptors. Therefore, preferred cell receptor and animal hosts were predicted for all coronaviruses using a sequence-based approach which may serve as future guide for further studies. © 2023, The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd.

3.
J Virol ; 97(2): e0194722, 2023 02 28.
Article in English | MEDLINE | ID: covidwho-2193457

ABSTRACT

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.


Subject(s)
CD13 Antigens , Deltacoronavirus , Spike Glycoprotein, Coronavirus , Virus Internalization , Animals , CD13 Antigens/genetics , CD13 Antigens/metabolism , Chickens/metabolism , Coronavirus Infections , Deltacoronavirus/metabolism , Swine , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Lentivirus/genetics , Lentivirus/metabolism
4.
Acta Veterinaria et Zootechnica Sinica ; 53(5):1587-1597, 2022.
Article in Chinese | CAB Abstracts | ID: covidwho-1994513

ABSTRACT

HEK293 cells were used as the cell model to investigate the role of human aminopeptidase N (hAPN) in the invasion of porcine deltacoronavirus (PDCoV) into human cells. The proliferation of PDCoV on HEK293 cells was firstly identified by RT-qPCR/RT-PCR. And then, hAPN knockout cell line was constructed by CRISPR/Cas9 technology and cell viability of HEK293 hAPN knockout and wild-type cells was verified by CCK-8 assay. Effect of hAPN knockout and overexpression on PDCoV replication was detected by RT-qPCR and Western blot. Meanwhile, interaction of PDCoV S protein and hAPN protein was analyzed by homology modeling and molecular docking. Results showed that PDCoV virus copies rapidly increased at 12-36 h and reached peak level at 36 h, it could propagate at least for passage 2 on HEK293 cells. There was no significant difference in cell viability between hAPN knockout cells and wild-type cells. Knockout of hAPN inhibit PDCoV replication and overexpression of hAPN enhance PDCoV replication. Homology modeling and molecular docking analysis showed S1 protein could bind hAPN domain II. Residues TYR92, THR51, THR48, PHE16 and MET14of S1 protein receptor binding motif 1 (RBM1) can form hydrogen bonds with residues PHE490, GLN531, ARG528 and SER529 of hAPN. This study indicates that hAPN plays a critical role in HEK293 cells during PDCoV infection, which provides new theoretical evidence for further studies on the mechanism of PDCoV entry into host cells and cross-species transmission.

5.
Biochim Biophys Acta Rev Cancer ; 1876(2): 188641, 2021 12.
Article in English | MEDLINE | ID: covidwho-1474353

ABSTRACT

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.


Subject(s)
CD13 Antigens/metabolism , Neoplasms/physiopathology , Peptide Hydrolases/metabolism , Humans
6.
Virulence ; 12(1): 1111-1121, 2021 12.
Article in English | MEDLINE | ID: covidwho-1243446

ABSTRACT

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.


Subject(s)
Epithelial Cells/virology , Influenza A Virus, H3N2 Subtype/physiology , Porcine Respiratory Coronavirus/physiology , Viral Interference , Animals , CD13 Antigens/metabolism , Cells, Cultured , Coinfection/virology , Coronavirus Infections/virology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Immunity, Innate , Orthomyxoviridae Infections/virology , Swine , Trachea/cytology , Virus Replication
7.
Biotechnol Appl Biochem ; 69(1): 124-128, 2022 Feb.
Article in English | MEDLINE | ID: covidwho-985952

ABSTRACT

Coronaviruses recognize a variety of host receptors to infect many humans and animals. Newly emerged severe acute respiratory syndrome coronavirus2 (SARS-CoV-2) recognizes angiotensin-converting enzyme 2 (ACE2) to gain entry into different cells. Interestingly, besides SARS-CoV2, four other human coronaviruses (HCoVs) use three different ectopeptidases (ACE2, dipeptidyl peptidase 4, and aminopeptidase N) as receptors independent of their common peptidase activity. This issue has led to the important question "why do several HCoVs rely on peptidases as their receptors?." In this paper, we discussed to answer this question. Mostly, it seems that the use of peptidases by HCoVs may be more related to their widespread presence on target cells and also viruses prefer to take advantage of molecules with relatively low affinity for their natural ligands through evolving a stronger binding affinity to the surface receptors for entry and endocytosis. Meanwhile evolutionary conservation of these receptors may allow HCoVs to switch between different host species. Finally, the choice of peptidases by HCoVs may reflect the "trial and error" nature of evolution. In conclusion, substantial efforts are needed to get a strong picture of this fascinating question and poorly explored area. Detailed understanding of the entry mechanisms offers opportunities for the development of refined strategies to stop viruses.


Subject(s)
SARS-CoV-2 , COVID-19 , Host Specificity , Humans , Peptide Hydrolases
8.
Viruses ; 12(11)2020 10 23.
Article in English | MEDLINE | ID: covidwho-895404

ABSTRACT

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.


Subject(s)
CD13 Antigens/metabolism , Epithelial Cells/metabolism , Porcine Respiratory Coronavirus/physiology , Receptors, Virus/metabolism , Respiratory Mucosa/virology , Viral Tropism , Animals , Bronchi/metabolism , Bronchi/virology , Cell Differentiation , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/virology , Swine , Trachea/metabolism , Trachea/virology , Virus Internalization , Virus Release , Virus Replication
9.
Viruses ; 12(4)2020 04 05.
Article in English | MEDLINE | ID: covidwho-31709

ABSTRACT

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.


Subject(s)
CD13 Antigens/metabolism , Gastroenteritis, Transmissible, of Swine/pathology , Porcine epidemic diarrhea virus/metabolism , Receptors, Virus/metabolism , Sialic Acids/metabolism , Transmissible gastroenteritis virus/metabolism , Animals , Cells, Cultured , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/veterinary , Enterocytes/virology , Hydrocortisone/pharmacology , Insulin/pharmacology , Respiratory Mucosa/virology , Spermidine/pharmacology , Swine , Vero Cells , Virus Attachment , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL