Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters










Publication year range
1.
J Virol ; 98(8): e0061124, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39078151

ABSTRACT

Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, is a serious threat to piglets and has zoonotic potential. Here, we aimed to further explore the role of aminopeptidase N (APN) as a receptor for PDCoV and test the inhibitory effect of a chimeric APN protein strategy on PDCoV infection. PK-15 cells and LLC-PK1 cells expressing chimeric APN were selected and infected with PDCoV. Viral replication was significantly decreased in these chimeric APN cells compared with that in control group cells. To further characterize the effect of the chimeric APN strategy on PDCoV infection in vitro, primary intestinal epithelial cells isolated from chimeric APN pigs were inoculated with PDCoV. Viral challenge of these cells led to decreased PDCoV infection. More importantly, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. Taken together, these findings deepen our understanding of the mechanism of PDCoV infection and provide a valuable model for the production of disease-resistant animals. IMPORTANCE: Porcine deltacoronavirus (PDCoV), an emerging enteropathogenic coronavirus, causes diarrhea in piglets and possesses the potential to infect humans. However, there are currently no effective measures for the prevention or control of PDCoV infection. Here, we have developed PK-15 cells, LLC-PK1 cells, and primary intestinal epithelial cells expressing chimeric APN, and viral challenge of these cells led to decreased PDCoV infection. Furthermore, virally challenged chimeric APN neonatal piglets displayed reduced viral load, significantly fewer microscopic lesions in the intestinal tissue, and no diarrhea. These data show that chimeric APN is a promising strategy to combat PDCoV infection.


Subject(s)
Animals, Newborn , CD13 Antigens , Coronavirus Infections , Deltacoronavirus , Swine Diseases , Virus Replication , Animals , Swine , CD13 Antigens/genetics , CD13 Antigens/metabolism , Swine Diseases/virology , Deltacoronavirus/genetics , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Coronavirus Infections/prevention & control , Viral Load , Gene Editing/methods , Cell Line , Epithelial Cells/virology , Diarrhea/virology
2.
J Virol ; 98(8): e0192923, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39078152

ABSTRACT

Hepatitis B virus (HBV) expresses co-terminal large (L), middle (M), and small (S) envelope proteins containing preS1/preS2/S, preS2/S, and S domain alone, respectively. S and preS1 domains mediate sequential virion attachment to heparan sulfate proteoglycans and sodium taurocholate cotransporting polypeptide (NTCP), respectively, which can be blocked by anti-S and anti-preS1 antibodies. How anti-preS2 antibodies neutralize HBV infectivity remains enigmatic. The late stage of chronic HBV infection often selects for mutated preS2 translation initiation codon to prevent M protein expression, or in-frame preS2 deletions to shorten both L and M proteins. When introduced to infectious clone of genotype C or D, both M-minus mutations and most 5' preS2 deletions sustained virion production. Such mutant progeny viral particles were infectious in NTCP-reconstituted HepG2 cells. Neutralization experiments were performed on the genotype D clone. Although remaining susceptible to anti-preS1 and anti-S neutralizing antibodies, M-minus mutants were only partially neutralized by two anti-preS2 antibodies tested while preS2 deletion mutants were resistant. By infection experiments using viral particles with lost versus increased M protein expression, or a neutralization escaping preS2 deletion only present on L or M protein, we found that both full-length L and M proteins contributed to virus neutralization by the two anti-preS2 antibodies. Thus, immune escape could be a driving force for the selection of M-minus mutations, and especially preS2 deletions. The fact that both L and M proteins could mediate neutralization by anti-preS2 antibodies may shed light on the underlying molecular mechanism.IMPORTANCEThe large (L), middle (M), and small (S) envelope proteins of hepatitis B virus (HBV) contain preS1/preS2/S, preS2/S, and S domain alone, respectively. The discovery of heparan sulfate proteoglycans and sodium taurocholate cotransporting polypeptide (NTCP) as the low- and high-affinity HBV receptors could explain neutralizing potential of anti-S and anti-preS1 antibodies, respectively, but how anti-preS2 neutralizing antibodies work remains enigmatic. In this study, we found two M-minus mutants in the context of genotype D partially escaped two anti-preS2 neutralizing antibodies in NTCP-reconstituted HepG2 cells, while several naturally occurring preS2 deletion mutants escaped both antibodies. By point mutations to eliminate or enhance M protein expression, and by introducing preS2 deletion selectively to L or M protein, we found binding of anti-preS2 antibodies to both L and M proteins contributed to neutralization of wild-type HBV infectivity. Our finding may shed light on the possible mechanism(s) whereby anti-preS2 antibodies neutralize HBV infectivity.


Subject(s)
Antibodies, Neutralizing , Hepatitis B Surface Antigens , Hepatitis B virus , Viral Envelope Proteins , Hepatitis B virus/immunology , Hepatitis B virus/genetics , Humans , Hepatitis B Surface Antigens/immunology , Hepatitis B Surface Antigens/genetics , Viral Envelope Proteins/immunology , Viral Envelope Proteins/genetics , Antibodies, Neutralizing/immunology , Hep G2 Cells , Sequence Deletion , Symporters/immunology , Symporters/genetics , Protein Precursors/immunology , Protein Precursors/genetics , Hepatitis B Antibodies/immunology , Hepatitis B/immunology , Hepatitis B/virology , Genotype , Immune Evasion , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/immunology , Organic Anion Transporters, Sodium-Dependent/metabolism , Virion/immunology
3.
Viruses ; 16(2)2024 02 02.
Article in English | MEDLINE | ID: mdl-38400014

ABSTRACT

This review presents comparative information corresponding to the progress in knowledge of some aspects of infection by the porcine epidemic diarrhea virus (PEDV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) coronaviruses. PEDV is an alphacoronavirus of great economic importance due to the million-dollar losses it generates in the pig industry. PEDV has many similarities to the SARS-CoV-2 betacoronavirus that causes COVID-19 disease. This review presents possible scenarios for SARS-CoV-2 based on the collected literature on PEDV and the tools or strategies currently developed for SARS-CoV-2 that would be useful in PEDV research. The speed of the study of SARS-CoV-2 and the generation of strategies to control the pandemic was possible due to the knowledge derived from infections caused by other human coronaviruses such as severe acute respiratory syndrome (SARS) and middle east respiratory syndrome (MERS). Therefore, from the information obtained from several coronaviruses, the current and future behavior of SARS-CoV-2 could be inferred and, with the large amount of information on the virus that causes COVID-19, the study of PEDV could be improved and probably that of new emerging and re-emerging coronaviruses.


Subject(s)
COVID-19 , Porcine epidemic diarrhea virus , Humans , Animals , Swine , SARS-CoV-2
4.
J Virol ; 97(11): e0090623, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37843369

ABSTRACT

IMPORTANCE: It is well known that influenza A viruses (IAV) initiate host cell infection by binding to sialic acid, a sugar molecule present at the ends of various sugar chains called glycoconjugates. These sugar chains can vary in chain length, structure, and composition. However, it remains unknown if IAV strains preferentially bind to sialic acid on specific glycoconjugate type(s) for host cell infection. Here, we utilized CRISPR gene editing to abolish sialic acid on different glycoconjugate types in human lung cells, and evaluated human versus avian IAV infections. Our studies show that both human and avian IAV strains can infect human lung cells by utilizing any of the three major sialic acid-containing glycoconjugate types, specifically N-glycans, O-glycans, and glycolipids. Interestingly, simultaneous elimination of sialic acid on all three major glycoconjugate types in human lung cells dramatically decreased human IAV infection, yet had little effect on avian IAV infection. These studies show that avian IAV strains effectively utilize other less prevalent glycoconjugates for infection, whereas human IAV strains rely on a limited repertoire of glycoconjugate types. The remarkable ability of avian IAV strains to utilize diverse glycoconjugate types may allow for easy transmission into new host species.


Subject(s)
Influenza A virus , Influenza, Human , Lung , Receptors, Cell Surface , Animals , Humans , Carrier Proteins/metabolism , Glycoconjugates/metabolism , Influenza A virus/metabolism , Lung/virology , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Sugars/metabolism , Influenza in Birds/metabolism , Receptors, Cell Surface/metabolism , Receptors, Virus/metabolism
5.
J Virol ; 97(7): e0016123, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37367301

ABSTRACT

Parvoviruses are among the smallest and superficially simplest animal viruses, infecting a broad range of hosts, including humans, and causing some deadly infections. In 1990, the first atomic structure of the canine parvovirus (CPV) capsid revealed a 26-nm-diameter T=1 particle made up of two or three versions of a single protein, and packaging about 5,100 nucleotides of single-stranded DNA. Our structural and functional understanding of parvovirus capsids and their ligands has increased as imaging and molecular techniques have advanced, and capsid structures for most groups within the Parvoviridae family have now been determined. Despite those advances, significant questions remain unanswered about the functioning of those viral capsids and their roles in release, transmission, or cellular infection. In addition, the interactions of capsids with host receptors, antibodies, or other biological components are also still incompletely understood. The parvovirus capsid's apparent simplicity likely conceals important functions carried out by small, transient, or asymmetric structures. Here, we highlight some remaining open questions that may need to be answered to provide a more thorough understanding of how these viruses carry out their various functions. The many different members of the family Parvoviridae share a capsid architecture, and while many functions are likely similar, others may differ in detail. Many of those parvoviruses have not been experimentally examined in detail (or at all in some cases), so we, therefore, focus this minireview on the widely studied protoparvoviruses, as well as the most thoroughly investigated examples of adeno-associated viruses.


Subject(s)
Parvoviridae , Animals , Humans , Capsid/ultrastructure , Capsid Proteins/chemistry , Capsid Proteins/metabolism , DNA, Viral/metabolism , Parvoviridae/genetics , Parvoviridae/ultrastructure , Parvoviridae Infections/metabolism , Parvoviridae Infections/virology , Dependovirus/genetics , Dependovirus/metabolism , Dependovirus/ultrastructure
6.
Biotechnol J ; 18(10): e2300130, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37300425

ABSTRACT

The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused considerable morbidity and mortality worldwide. Although authorized COVID-19 vaccines have been shown highly effective, their significantly lower efficacy against heterologous variants, and the rapid decrease of vaccine-elicited immunity raises serious concerns, calling for improved vaccine tactics. To this end, a pseudovirus nanoparticle (PVNP) displaying the receptor binding domains (RBDs) of SARS-CoV-2 spike, named S-RBD, was generated and shown it as a promising COVID-19 vaccine candidate. The S-RBD PVNP was produced using both prokaryotic and eukaryotic systems. A 3D structural model of the S-RBD PVNPs was built based on the known structures of the S60 particle and RBDs, revealing an S60 particle-based icosahedral symmetry with multiple surface-displayed RBDs that retain authentic conformations and receptor-binding functions. The PVNP is highly immunogenic, eliciting high titers of RBD-specific IgG and neutralizing antibodies in mice. The S-RBD PVNP demonstrated exceptional protective efficacy, and fully (100%) protected K18-hACE2 mice from mortality and weight loss after a lethal SARS-CoV-2 challenge, supporting the S-RBD PVNPs as a potent COVID-19 vaccine candidate. By contrast, a PVNP displaying the N-terminal domain (NTD) of SARS-CoV-2 spike exhibited only 50% protective efficacy. Since the RBD antigens of our PVNP vaccine are adjustable as needed to address the emergence of future variants, and various S-RBD PVNPs can be combined as a cocktail vaccine for broad efficacy, these non-replicating PVNPs offer a flexible platform for a safe, effective COVID-19 vaccine with minimal manufacturing cost and time.


Subject(s)
COVID-19 , Nanoparticles , Animals , Humans , Mice , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19 Vaccines/genetics , Pandemics , Weight Loss
7.
J Med Primatol ; 52(4): 213-222, 2023 08.
Article in English | MEDLINE | ID: mdl-37296521

ABSTRACT

BACKGROUND: Cell culture is the proliferation of a cell population in vitro by isolating from the original tissue or growing from existing ones. One essential source is the monkey kidney cell cultures which have an essential role in biomedical study. This is due to the significant homology between the human and macaque genomes making these useful for cultivating human viruses, especially enteroviruses, and growing vaccines. METHODS: This study developed cell cultures derived from the kidney of Macaca fascicularis (Mf) and validated its gene expression. RESULTS: The primary cultures were successfully subcultured up to six passages, grew as monolayers, and exhibited epithelial-like morphology. The cultured cells remained heterogeneous in phenotype and they expressed CD155 and CD46 as viral receptors, cell morphology (CD24, endosialin, and vWF), proliferation, also apoptosis markers (Ki67 and p53). CONCLUSIONS: These results indicated that the cell cultures can be used as in vitro model cells for vaccine development and bioactive compound.


Subject(s)
Cell Culture Techniques , Kidney , Humans , Animals , Macaca fascicularis , Cells, Cultured , Vaccine Development
8.
Viruses ; 14(4)2022 03 31.
Article in English | MEDLINE | ID: mdl-35458470

ABSTRACT

Human T-cell leukemia virus type 1 (HTLV-1) causes serious and intractable diseases in some carriers after infection. The elimination of infected cells is considered important to prevent this onset, but there are currently no means by which to accomplish this. We previously developed "virotherapy", a therapeutic method that targets and kills HTLV-1-infected cells using a cytolytic recombinant vesicular stomatitis virus (rVSV). Infection with rVSV expressing an HTLV-1 primary receptor elicits therapeutic effects on HTLV-1-infected envelope protein (Env)-expressing cells in vitro and in vivo. Simian T-cell leukemia virus type 1 (STLV-1) is closely related genetically to HTLV-1, and STLV-1-infected Japanese macaques (JMs) are considered a useful HTLV-1 surrogate, non-human primate model in vivo. Here, we performed an in vitro drug evaluation of rVSVs against STLV-1 as a preclinical study. We generated novel rVSVs encoding the STLV-1 primary receptor, simian glucose transporter 1 (JM GLUT1), with or without an AcGFP reporter gene. Our data demonstrate that these rVSVs specifically and efficiently infected/eliminated the STLV-1 Env-expressing cells in vitro. These results indicate that rVSVs carrying the STLV-1 receptor could be an excellent candidate for unique anti-STLV-1 virotherapy; therefore, such antivirals can now be applied to STLV-1-infected JMs to determine their therapeutic usefulness in vivo.


Subject(s)
Deltaretrovirus Infections , Human T-lymphotropic virus 1 , Leukemia, T-Cell , Simian T-lymphotropic virus 1 , Vesicular Stomatitis , Animals , Deltaretrovirus Infections/genetics , Human T-lymphotropic virus 1/genetics , Simian T-lymphotropic virus 1/genetics , Vesiculovirus
9.
Emerg Microbes Infect ; 11(1): 761-774, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35191820

ABSTRACT

Usutu virus (USUV) and West Nile virus (WNV) are phylogenetically close emerging arboviruses and constitute a global public health threat. Since USUV and WNV are transmitted by mosquitoes, the first immune cells they encounter are skin-resident dendritic cells, the most peripheral outpost of immune defense. This unique network is composed of Langerhans cells (LCs) and dermal DCs, which reside in the epidermis and the dermis, respectively. Using human skin explants, we show that while both viruses can replicate in keratinocytes, they can also infect resident DCs with distinct tropism: WNV preferentially infects DCs in the dermis, whereas USUV has a greater propensity to infect LCs. Using both purified human epidermal LCs (eLCs) and monocyte derived LCs (MoLCs), we confirm that LCs sustain a faster and more efficient replication of USUV than WNV and that this correlates with a more intense innate immune response to USUV compared with WNV. Next, we show that ectopic expression of the LC-specific C-type lectin receptor (CLR), langerin, in HEK293T cells allows WNV and USUV to bind and enter, but supports the subsequent replication of USUV only. Conversely, blocking or silencing langerin in MoLCs or eLCs made them resistant to USUV infection, thus demonstrating that USUV uses langerin to enter and replicate in LCs. Altogether, our results demonstrate that LCs constitute privileged target cells for USUV in human skin, because langerin favours its entry and replication. Intriguingly, this suggests that USUV efficiently escapes the antiviral functions of langerin, which normally safeguards LCs from most viral infections.


Subject(s)
Flavivirus Infections , West Nile Fever , West Nile virus , Animals , Flavivirus , HEK293 Cells , Humans , Langerhans Cells , West Nile virus/genetics
10.
Physiol Behav ; 247: 113707, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35063424

ABSTRACT

The lateral hypothalamic area (LHA) is essential for ingestive behavior but has primarily been studied in modulating feeding, with comparatively scant attention on drinking. This is partly because most LHA neurons simultaneously promote feeding and drinking, suggesting that ingestive behaviors track together. A notable exception are LHA neurons expressing neurotensin (LHANts neurons): activating these neurons promotes water intake but modestly restrains feeding. Here we investigated the connectivity of LHANts neurons, their necessity and sufficiency for drinking and feeding, and how timing and resource availability influence their modulation of these behaviors. LHANts neurons project broadly throughout the brain, including to the lateral preoptic area (LPO), a brain region implicated in modulating drinking behavior. LHANts neurons also receive inputs from brain regions implicated in sensing hydration and energy status. While activation of LHANts neurons is not required to maintain homeostatic water or food intake, it selectively promotes drinking during the light cycle, when ingestive drive is low. Activating LHANts neurons during this period also increases willingness to work for water or palatable fluids, regardless of their caloric content. By contrast, LHANts neuronal activation during the dark cycle does not promote drinking, but suppresses feeding during this time. Finally, we demonstrate that the activation of the LHANts â†’ LPO projection is sufficient to mediate drinking behavior, but does not suppress feeding as observed after generally activating all LHANts neurons. Overall, our work suggests how and when LHANts neurons oppositely modulate ingestive behaviors.


Subject(s)
Hypothalamic Area, Lateral , Neurotensin , Food , Hypothalamic Area, Lateral/metabolism , Neurons/metabolism , Neurotensin/metabolism , Water
11.
J Infect Public Health ; 15(1): 123-131, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34742639

ABSTRACT

More than 125 million confirmed cases of COVID-19 have been reported globally with rising cases in all countries since the first case was reported. A vaccine is the best measure for the effective prevention and control of COVID-19. There are more than 292 COVID-19 candidates' vaccines being developed as of July 2021 of which 184 are in human preclinical trials. A patent provides protection and a marketing monopoly to the inventor of an invention for a specified period. Therefore, vaccine developers, including Moderna, BioNTech, Janssen, Inovio, and Gamaleya also filed patent applications for the protection of their vaccines. This review aims to provide an insight into the patent literature of COVID-19 vaccines. The patent search was done using Patentscope and Espacenet databases. The results have revealed that most of the key players have patented their inventive COVID-19 vaccine. Many patent applications related to COVID-19 vaccines developed via different technologies (DNA, RNA, virus, bacteria, and protein subunit) have also been filed. The publication of a normal patent application takes place after 18 months of its filing. Therefore, many patents/patent applications related to the COVID-19 vaccine developed through different technology may come into the public domain in the coming days.


Subject(s)
COVID-19 , Vaccines , COVID-19 Vaccines , Humans , SARS-CoV-2
12.
Elife ; 102021 08 26.
Article in English | MEDLINE | ID: mdl-34435953

ABSTRACT

The interaction between the SARS-CoV-2 virus Spike protein receptor binding domain (RBD) and the ACE2 cell surface protein is required for viral infection of cells. Mutations in the RBD are present in SARS-CoV-2 variants of concern that have emerged independently worldwide. For example, the B.1.1.7 lineage has a mutation (N501Y) in its Spike RBD that enhances binding to ACE2. There are also ACE2 alleles in humans with mutations in the RBD binding site. Here we perform a detailed affinity and kinetics analysis of the effect of five common RBD mutations (K417N, K417T, N501Y, E484K, and S477N) and two common ACE2 mutations (S19P and K26R) on the RBD/ACE2 interaction. We analysed the effects of individual RBD mutations and combinations found in new SARS-CoV-2 Alpha (B.1.1.7), Beta (B.1.351), and Gamma (P1) variants. Most of these mutations increased the affinity of the RBD/ACE2 interaction. The exceptions were mutations K417N/T, which decreased the affinity. Taken together with other studies, our results suggest that the N501Y and S477N mutations enhance transmission primarily by enhancing binding, the K417N/T mutations facilitate immune escape, and the E484K mutation enhances binding and immune escape.


As the COVID-19 pandemic has progressed, new variants of the virus SARS-CoV-2 have emerged that are more infectious than the original form. The variants known as Alpha, Beta and Gamma have mutations in a protein on the virus's surface that is vital for attaching to cells and infecting them. This protein, called Spike, carries out its role by binding to ACE2, a protein on the surface of human cells. Mutations on Spike are found on the region where it binds to ACE2. The interaction between these two proteins appears to be important to the behaviour of SARS-CoV-2, but the impact of individual mutations in Spike is unknown. In addition, some people have different variants of ACE2 with mutations in the region that interacts with Spike, but it is not known whether this affects these people's risk of contracting COVID-19. To answer these questions, Barton et al. measured the precise effect of mutations in Spike and ACE2 on the strength of the interaction between the two proteins. The experiments showed that three of the five common Spike mutations in the Alpha, Beta and Gamma SARS-CoV-2 variants strengthened binding to ACE2. The two mutations that weakened binding were only found together with other mutations that strengthened binding. This meant that the Spike proteins in all three of these SARS-CoV-2 variants bind to ACE2 more strongly than the original form. The experiments also showed that two common variants of ACE2 also increased the strength of binding to Spike. Interestingly, one of these ACE2 variants reversed the effect of a specific SARS-CoV-2 mutation, suggesting that carriers would be resistant to SARS-CoV-2 variants with this mutation. Identifying the precise effects of Spike mutations on ACE2 binding helps understand why new variants of SARS-CoV-2 spread more rapidly. This could help to identify concerning new variants before they spread widely and inform the response by health authorities. The finding that two common ACE2 variants bind more strongly to Spike suggests that people with these mutations could be more susceptible to SARS-CoV-2.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , Mutation , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Binding Sites , COVID-19/virology , Humans , Kinetics , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs , SARS-CoV-2/classification , SARS-CoV-2/metabolism , Spike Glycoprotein, Coronavirus/chemistry
13.
Microorganisms ; 9(8)2021 Aug 09.
Article in English | MEDLINE | ID: mdl-34442770

ABSTRACT

Angiotensin-Converting Enzyme 2 (ACE2) has been proved to be the main host cell receptor for the binding of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the COVID-19 pandemic. The SARS-CoV-2 spike (S) protein binds to ACE2 to initiate the process of replication. This enzyme is widely present in human organ tissues, such as the heart and lung. The pathophysiology of ACE2 in SARS-CoV-2 infection is complex and may be associated with several factors and conditions that are more severe in COVID-19 patients, such as age, male gender, and comorbidities, namely, cardiovascular diseases, chronic respiratory diseases, obesity, and diabetes. Here we present a comprehensive review that aims to correlate the levels of expression of the ACE2 in patients with comorbidities and with a poor outcome in COVID-19 disease. Significantly higher levels of expression of ACE2 were observed in myocardial and lung tissues in heart failure and COPD patients, respectively. An age-dependent increase in SARS2-CoV-2 receptors in the respiratory epithelium may be also responsible for the increased severity of COVID-19 lung disease in elderly people. Although the role of ACE2 is highlighted regarding the damage that can arise upon the SARS-CoV-2 invasion, there was no association observed between renin-angiotensin-aldosterone system (RAAS) inhibitors and the severity of COVID-19.

14.
Adv Exp Med Biol ; 1288: 95-109, 2021.
Article in English | MEDLINE | ID: mdl-34453733

ABSTRACT

Coxsackievirus and adenovirus receptor (CXADR) belongs to immunoglobulin superfamily of cell adhesion molecules. It expresses in most tissues, but displays unique and indispensable functions in some tissues such as heart and testis. CXADR is a multifunctional protein that can serve as a viral receptor, a junction structural protein and a signalling molecule. Thus, it exerts a wide range of functions such as facilitating leukocyte transmigration, regulating barrier function and cell adhesion, promoting EMT transition, and mediating spermatogenesis. This review aims to provide an overview and highlights some recent findings on CXADR in the field with emphasis on studies in the testis, upon which future studies can be designed to delineate the roles and regulation of CXADR in spermatogenesis.


Subject(s)
Receptors, Virus , Spermatogenesis , Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Humans , Male , Receptors, Virus/genetics , Signal Transduction
15.
Theranostics ; 11(13): 6193-6213, 2021.
Article in English | MEDLINE | ID: mdl-33995653

ABSTRACT

Rationale: The pandemic caused by the novel coronavirus SARS-CoV-2 is advancing rapidly. In particular, the number of severe courses of the disease is still dramatically high. An efficient drug therapy that helps to improve significantly the fatal combination of damages in the airway epithelia, in the extensive pulmonary microvascularization and finally multiorgan failure, is missing. The physiological, inorganic polymer, polyphosphate (polyP) is a molecule which could prevent the initial phase of the virus life cycle, the attachment of the virus to the target cells, and improve the epithelial integrity as well as the mucus barrier. Results: Surprisingly, polyP matches perfectly with the cationic groove on the RBD. Subsequent binding studies disclosed that polyP, with a physiological chain length of 40 phosphate residues, abolishes the binding propensity of the RBD to the ACE2 receptor. In addition to this first mode of action of polyP, this polymer causes in epithelial cells an increased gene expression of the major mucins in the airways, of MUC5AC and MUC1, as well as a subsequent glycoprotein production. MUC5AC forms a gel-like mucus layer trapping inhaled particles which are then transported out of the airways, while MUC1 constitutes the periciliary liquid layer and supports ciliary beating. As a third mode of action, polyP undergoes enzymatic hydrolysis of the anhydride bonds in the airway system by alkaline phosphatase, releasing metabolic energy. Conclusions: This review summarizes the state of the art of the biotherapeutic potential of the polymer polyP and the findings from basic research and outlines future biomedical applications.


Subject(s)
COVID-19 Drug Treatment , Pandemics/prevention & control , Polyphosphates/pharmacology , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , COVID-19/epidemiology , COVID-19/transmission , COVID-19/virology , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Mice , Mucins/metabolism , Nanoparticles/chemistry , Polyphosphates/chemistry , Polyphosphates/therapeutic use , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Virus Attachment/drug effects
16.
Viruses ; 13(2)2021 02 13.
Article in English | MEDLINE | ID: mdl-33668417

ABSTRACT

Human adenoviruses cause disease at multiple mucosal sites, including the respiratory, gastrointestinal, and genitourinary tracts, and are common agents of conjunctivitis. One site of infection that has received sparse attention is the cornea, a transparent tissue and the window of the eye. While most adenovirus infections are self-limited, corneal inflammation (keratitis) due to adenovirus can persist or recur for months to years after infection, leading to reduced vision, discomfort, and light sensitivity. Topical corticosteroids effectively suppress late adenovirus keratitis but are associated with vision-threatening side effects. In this short review, we summarize current knowledge on infection of the cornea by adenoviruses, including corneal epithelial cell receptors and determinants of corneal tropism. We briefly discuss mechanisms of stromal keratitis due to adenovirus infection, and review an emerging therapy to mitigate adenovirus corneal infections based on evolving knowledge of corneal epithelial receptor usage.


Subject(s)
Adenoviridae Infections/virology , Adenoviruses, Human/physiology , Cornea/virology , Corneal Diseases/virology , Adenoviruses, Human/classification , Adenoviruses, Human/genetics , Adenoviruses, Human/isolation & purification , Animals , Humans
17.
Res Vet Sci ; 135: 78-84, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33453552

ABSTRACT

Porcine reproductive and respiratory syndrome virus (PRRSV) is an economically important pathogen affecting global swine industry. Our recent study has shown that the first four Ig-like domains of sialoadhesin (Sn4D) and the scavenger receptor cysteine-rich domains 5-9 (SRCR59) of CD163 can act as the soluble viral receptors (SVRs) of PRRSV. Co-injection with the two SVR-expressing recombinant adenovirus (rAd) vectors can protect pigs from the lethal challenge with three PRRSV strains. However, the in vivo expression of the two SVRs persists for only two weeks and thus their long-term anti-PRRSV effects remain to be improved. In this study, we fused the two SVRs with a flexible linker or self-cleaving peptide and expressed them with a single recombinant adeno-associated virus (rAAV) vector. The two rAAVs, namely rAAV-Sn4D-SRCR59-Fc and rAAV-SRCR59-Fc/Sn4D-Fc, were generated by using baculovirus-insect cell system. Western blotting analysis showed that the two SVR fusions were efficiently expressed in and secreted from the rAAV-transduced cells. Viral infection blocking assay showed that PRRSV titers in porcine alveolar macrophage (PAM) cells were reduced by 1.6-2.7 log10 after co-cultivation with rAAV-Sn4D-SRCR59-Fc-transduced cells or by 1.9-3.2 log10 after co-cultivation with rAAV-SRCR59-Fc/Sn4D-Fc-transduced cells. After single-dose injection of mice with the rAAV vectors, the expression of two SVR fusions persisted for at least 35 days, which was significantly longer than SRCR59-Fc expression in rAd-SRCR59-Fc-injected mice. Among the two SVR fusions expressed, both expression level and anti-PRRSV activity of SRCR59-Fc/Sn4D-Fc were higher than that of Sn4D-SRCR59-Fc. Therefore, rAAV-SRCR59-Fc/Sn4D-Fc generated can be developed as a novel anti-PRRSV reagent.


Subject(s)
Genetic Vectors , Porcine respiratory and reproductive syndrome virus , Receptors, Virus/chemistry , Recombinant Fusion Proteins/immunology , Viral Vaccines/immunology , Animals , Antigens, CD , Antigens, Differentiation, Myelomonocytic , Dependovirus , Gene Expression Regulation , Macrophages/metabolism , Macrophages, Alveolar/virology , Mice , Receptors, Cell Surface , Receptors, Virus/genetics , Receptors, Virus/metabolism , Sialic Acid Binding Ig-like Lectin 1/genetics , Sialic Acid Binding Ig-like Lectin 1/metabolism , Swine , Swine Diseases/metabolism , Swine Diseases/virology
18.
Viruses ; 13(1)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478139

ABSTRACT

The first step of cellular entry for the human immunodeficiency virus type-1 (HIV-1) occurs through the binding of its envelope protein (Env) with the plasma membrane receptor CD4 and co-receptor CCR5 or CXCR4 on susceptible cells, primarily CD4+ T cells and macrophages. Although there is considerable knowledge of the molecular interactions between Env and host cell receptors that lead to successful fusion, the precise way in which HIV-1 receptors redistribute to sites of virus binding at the nanoscale remains unknown. Here, we quantitatively examine changes in the nanoscale organisation of CD4 on the surface of CD4+ T cells following HIV-1 binding. Using single-molecule super-resolution imaging, we show that CD4 molecules are distributed mostly as either individual molecules or small clusters of up to 4 molecules. Following virus binding, we observe a local 3-to-10-fold increase in cluster diameter and molecule number for virus-associated CD4 clusters. Moreover, a similar but smaller magnitude reorganisation of CD4 was also observed with recombinant gp120. For one of the first times, our results quantify the nanoscale CD4 reorganisation triggered by HIV-1 on host CD4+ T cells. Our quantitative approach provides a robust methodology for characterising the nanoscale organisation of plasma membrane receptors in general with the potential to link spatial organisation to function.


Subject(s)
CD4 Antigens/metabolism , Cell Membrane/metabolism , Cell Membrane/virology , HIV-1/physiology , Single Molecule Imaging/methods , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Virus Attachment , Algorithms , Antibodies, Monoclonal , Cell Line , Data Interpretation, Statistical , HIV Envelope Protein gp120/metabolism , Host-Pathogen Interactions , Humans , Image Processing, Computer-Assisted , Protein Binding , Receptors, CCR5/metabolism , Receptors, HIV/metabolism
19.
Food Microbiol ; 95: 103709, 2021 May.
Article in English | MEDLINE | ID: mdl-33397626

ABSTRACT

The ongoing pandemic involving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has raised the question whether this virus, which is known to be spread primarily though respiratory droplets, could be spread through the fecal-oral route or via contaminated food. In this article, we present a critical review of the literature exploring the potential foodborne transmission of several respiratory viruses including human coronaviruses, avian influenza virus (AVI), parainfluenza viruses, human respiratory syncytial virus, adenoviruses, rhinoviruses, and Nipah virus. Multiple lines of evidence, including documented expression of receptor proteins on gastrointestinal epithelial cells, in vivo viral replication in gastrointestinal epithelial cell lines, extended fecal shedding of respiratory viruses, and the ability to remain infectious in food environments for extended periods of time raises the theoretical ability of some human respiratory viruses, particularly human coronaviruses and AVI, to spread via food. However, to date, neither epidemiological data nor case reports of clear foodborne transmission of either viruses exist. Thus, foodborne transmission of human respiratory viruses remains only a theoretical possibility.


Subject(s)
Foodborne Diseases/virology , Respiratory Tract Infections/transmission , Respiratory Tract Infections/virology , Animals , Birds , COVID-19/transmission , COVID-19/virology , Feces/virology , Humans , SARS-CoV-2/isolation & purification
20.
Int J Mol Sci ; 21(13)2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32630682

ABSTRACT

In previous research, voltage-dependent anion channel protein 2 (VDAC2) and the receptor of activated protein C kinase 1 (RACK1) in flounder (Paralichthys olivaceus) were confirmed as functional receptors for lymphocystis disease virus (LCDV) entry; however, the underlying mechanism of VDAC2- and RACK1-mediated LCDV entry remains unclear. In this study, we elucidated the endocytosis pathway of LCDV entry into flounder gill (FG) cells by treatment with specific inhibitory agents, siRNAs, and co-localization analysis. LCDV entry was significantly inhibited by the disruption of caveolae-mediated endocytosis, dynamin, and microtubules, and the knockdown of caveoline-1 and dynamin expression, but was not inhibited by the disruption of clathrin-mediated endocytosis, micropinocytosis, or low-pH conditions. The disruption of caveolae-mediated and clathrin-mediated endocytosis was verified by the internalization of cholera toxin subunit B (CTB) and transferrin, respectively. Confocal immunofluorescence assay demonstrated that LCDV was co-localized with VDAC2 and RACK1, CTB was co-localized with VDAC2 and RACK1 and partially with LCDV, but transferrin was not co-localized with LCDV, VDAC2, or RACK1, indicating that LCDV utilized the same pathway as CTB, i.e., caveolae-mediated endocytosis. This was different from the pathway of transferrin, which used clathrin-mediated endocytosis. Furthermore, caveolin-1 was co-localized with LCDV, VDAC2, and RACK1, suggesting that caveolin-1 was involved in LCDV entry. These results revealed for the first time that LCDV entered into FG cells via caveolae-mediated endocytosis facilitated by VDAC2 and RACK1 receptors, relying on dynamin and microtubules in a pH-independent manner, which provided new insight into the molecular mechanisms of LCDV entry and potential for the development of antiviral agents, expanding our understanding of iridovirus infection.


Subject(s)
Endocytosis/physiology , Iridoviridae/physiology , Receptors, Virus/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Caveolae/metabolism , DNA Virus Infections/virology , Fish Diseases/virology , Flounder/metabolism , Flounder/virology , GTP-Binding Proteins/metabolism , Gills/metabolism , Gills/virology , Iridoviridae/metabolism , Iridoviridae/pathogenicity , Saccharomyces cerevisiae Proteins/metabolism , Virus Replication/physiology , Voltage-Dependent Anion Channel 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL