Your browser doesn't support javascript.
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 65
Filtre
1.
Front Immunol ; 14: 1196031, 2023.
Article Dans Anglais | MEDLINE | ID: covidwho-20236991

Résumé

Swine acute diarrhoea syndrome coronavirus (SADS-CoV), which is a recently discovered enteric coronavirus, is the major aetiological agent that causes severe clinical diarrhoea and intestinal pathological damage in pigs, and it has caused significant economic losses to the swine industry. Nonstructural protein 5, also called 3C-like protease, cleaves viral polypeptides and host immune-related molecules to facilitate viral replication and immune evasion. Here, we demonstrated that SADS-CoV nsp5 significantly inhibits the Sendai virus (SEV)-induced production of IFN-ß and inflammatory cytokines. SADS-CoV nsp5 targets and cleaves mRNA-decapping enzyme 1a (DCP1A) via its protease activity to inhibit the IRF3 and NF-κB signaling pathways in order to decrease IFN-ß and inflammatory cytokine production. We found that the histidine 41 and cystine 144 residues of SADS-CoV nsp5 are critical for its cleavage activity. Additionally, a form of DCP1A with a mutation in the glutamine 343 residue is resistant to nsp5-mediated cleavage and has a stronger ability to inhibit SADS-CoV infection than wild-type DCP1A. In conclusion, our findings reveal that SADS-CoV nsp5 is an important interferon antagonist and enhance the understanding of immune evasion by alpha coronaviruses.


Sujets)
Alphacoronavirus , Coronavirus , Interféron de type I , Animaux , Suidae , Alphacoronavirus/génétique , Alphacoronavirus/métabolisme , Coronavirus/métabolisme , Endopeptidases , Interféron de type I/métabolisme
2.
EMBO J ; 42(13): e112542, 2023 07 03.
Article Dans Anglais | MEDLINE | ID: covidwho-2327293

Résumé

Lipid droplets (LDs) form inter-organelle contacts with the endoplasmic reticulum (ER) that promote their biogenesis, while LD contacts with mitochondria enhance ß-oxidation of contained fatty acids. Viruses have been shown to take advantage of lipid droplets to promote viral production, but it remains unclear whether they also modulate the interactions between LDs and other organelles. Here, we showed that coronavirus ORF6 protein targets LDs and is localized to the mitochondria-LD and ER-LD contact sites, where it regulates LD biogenesis and lipolysis. At the molecular level, we find that ORF6 inserts into the LD lipid monolayer via its two amphipathic helices. ORF6 further interacts with ER membrane proteins BAP31 and USE1 to mediate ER-LDs contact formation. Additionally, ORF6 interacts with the SAM complex in the mitochondrial outer membrane to link mitochondria to LDs. In doing so, ORF6 promotes cellular lipolysis and LD biogenesis to reprogram host cell lipid flux and facilitate viral production.


Sujets)
Coronavirus , Coronavirus/métabolisme , Réticulum endoplasmique/métabolisme , Gouttelettes lipidiques/métabolisme , Lipolyse , Acides gras/métabolisme
3.
Proc Natl Acad Sci U S A ; 120(20): e2221324120, 2023 05 16.
Article Dans Anglais | MEDLINE | ID: covidwho-2320604

Résumé

The frameshifting RNA element (FSE) in coronaviruses (CoVs) regulates the programmed -1 ribosomal frameshift (-1 PRF) mechanism common to many viruses. The FSE is of particular interest as a promising drug candidate. Its associated pseudoknot or stem loop structure is thought to play a large role in frameshifting and thus viral protein production. To investigate the FSE structural evolution, we use our graph theory-based methods for representing RNA secondary structures in the RNA-As-Graphs (RAG) framework to calculate conformational landscapes of viral FSEs with increasing sequence lengths for representative 10 Alpha and 13 Beta-CoVs. By following length-dependent conformational changes, we show that FSE sequences encode many possible competing stems which in turn favor certain FSE topologies, including a variety of pseudoknots, stem loops, and junctions. We explain alternative competing stems and topological FSE changes by recurring patterns of mutations. At the same time, FSE topology robustness can be understood by shifted stems within different sequence contexts and base pair coevolution. We further propose that the topology changes reflected by length-dependent conformations contribute to tuning the frameshifting efficiency. Our work provides tools to analyze virus sequence/structure correlations, explains how sequence and FSE structure have evolved for CoVs, and provides insights into potential mutations for therapeutic applications against a broad spectrum of CoV FSEs by targeting key sequence/structural transitions.


Sujets)
Infections à coronavirus , Coronavirus , Humains , ARN viral/métabolisme , Coronavirus/génétique , Coronavirus/métabolisme , Séquence nucléotidique , Conformation d'acide nucléique , Décalage ribosomique/génétique , Infections à coronavirus/génétique
4.
Phytochemistry ; 212: 113713, 2023 Aug.
Article Dans Anglais | MEDLINE | ID: covidwho-2318116

Résumé

The potential antiviral effects of indole-3-carbinol (I3C), a phytochemical found in Cruciferous vegetables, were investigated. Fibroblasts and epithelial cells were co-cultured on Alvetex® scaffolds, to obtain ad hoc 3D in vitro platforms able to mimic the trachea and intestinal mucosae, which represent the primary structures involved in the coronavirus pathogenesis. The two barriers generated in vitro were treated with various concentrations of I3C for different incubation periods. A protective effect of I3C on both intestinal and trachea models was demonstrated. A significant reduction in the transcription of the two main genes belonging to the Homologous to E6AP C-terminus (HECT)-E3 ligase family members, namely NEDD4 E3 Ubiquitin Protein Ligase (NEDD4) and WW Domain Containing E3 Ubiquitin Protein Ligase 1 (WWP1), which promote virus matrix protein ubiquitination and inhibit viral egression, were detected. These findings indicate I3C potential effect in preventing coronavirus cell egression processes that inhibit viral production. Although further studies are needed to clarify the molecular mechanisms whereby HECT family members control virus life cycle, this work paves the way to the possible therapeutic use of new natural compounds that may reduce the clinical severity of future pandemics.


Sujets)
Brassicaceae , Coronavirus , Légumes/composition chimique , Coronavirus/métabolisme , Trachée/métabolisme , Ubiquitin-protein ligases/composition chimique , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme , Brassicaceae/métabolisme
5.
J Virol ; 97(5): e0037523, 2023 05 31.
Article Dans Anglais | MEDLINE | ID: covidwho-2316566

Résumé

Porcine deltacoronavirus (PDCoV) is an emerging swine enteropathogenic coronavirus that has the potential to infect humans. Histone deacetylase 6 (HDAC6) is a unique type IIb cytoplasmic deacetylase with both deacetylase activity and ubiquitin E3 ligase activity, which mediates a variety of cellular processes by deacetylating histone and nonhistone substrates. In this study, we found that ectopic expression of HDAC6 significantly inhibited PDCoV replication, while the reverse effects could be observed after treatment with an HDAC6-specific inhibitor (tubacin) or knockdown of HDAC6 expression by specific small interfering RNA. Furthermore, we demonstrated that HDAC6 interacted with viral nonstructural protein 8 (nsp8) in the context of PDCoV infection, resulting in its proteasomal degradation, which was dependent on the deacetylation activity of HDAC6. We further identified the key amino acid residues lysine 46 (K46) and K58 of nsp8 as acetylation and ubiquitination sites, respectively, which were required for HDAC6-mediated degradation. Through a PDCoV reverse genetics system, we confirmed that recombinant PDCoV with a mutation at either K46 or K58 exhibited resistance to the antiviral activity of HDAC6, thereby exhibiting higher replication compared with wild-type PDCoV. Collectively, these findings contribute to a better understanding of the function of HDAC6 in regulating PDCoV infection and provide new strategies for the development of anti-PDCoV drugs. IMPORTANCE As an emerging enteropathogenic coronavirus with zoonotic potential, porcine deltacoronavirus (PDCoV) has sparked tremendous attention. Histone deacetylase 6 (HDAC6) is a critical deacetylase with both deacetylase activity and ubiquitin E3 ligase activity and is extensively involved in many important physiological processes. However, little is known about the role of HDAC6 in the infection and pathogenesis of coronaviruses. Our present study demonstrates that HDAC6 targets PDCoV-encoded nonstructural protein 8 (nsp8) for proteasomal degradation through the deacetylation at the lysine 46 (K46) and the ubiquitination at K58, suppressing viral replication. Recombinant PDCoV with a mutation at K46 and/or K58 of nsp8 displayed resistance to the antiviral activity of HDAC6. Our work provides significant insights into the role of HDAC6 in regulating PDCoV infection, opening avenues for the development of novel anti-PDCoV drugs.


Sujets)
Infections à coronavirus , Coronavirus , Maladies des porcs , Animaux , Antiviraux/pharmacologie , Antiviraux/métabolisme , Coronavirus/métabolisme , Histone deacetylase 6/génétique , Histone deacetylase 6/métabolisme , Lysine/métabolisme , Suidae , Ubiquitine/métabolisme , Ubiquitin-protein ligases/métabolisme , Ubiquitination , Réplication virale
6.
Viruses ; 15(4)2023 03 31.
Article Dans Anglais | MEDLINE | ID: covidwho-2292459

Résumé

The fusion of viral and cell membranes is one of the basic processes in the life cycles of viruses. A number of enveloped viruses confer fusion of the viral envelope and the cell membrane using surface viral fusion proteins. Their conformational rearrangements lead to the unification of lipid bilayers of cell membranes and viral envelopes and the formation of fusion pores through which the viral genome enters the cytoplasm of the cell. A deep understanding of all the stages of conformational transitions preceding the fusion of viral and cell membranes is necessary for the development of specific inhibitors of viral reproduction. This review systematizes knowledge about the results of molecular modeling aimed at finding and explaining the mechanisms of antiviral activity of entry inhibitors. The first section of this review describes types of viral fusion proteins and is followed by a comparison of the structural features of class I fusion proteins, namely influenza virus hemagglutinin and the S-protein of the human coronavirus.


Sujets)
Infections à coronavirus , Coronavirus , Orthomyxoviridae , Humains , Protéines de fusion virale/métabolisme , Coronavirus/métabolisme , Hémagglutinines/métabolisme , Glycoprotéine de spicule des coronavirus/génétique , Fusion membranaire , Orthomyxoviridae/métabolisme , Pénétration virale
7.
J Virol ; 96(22): e0147322, 2022 11 23.
Article Dans Anglais | MEDLINE | ID: covidwho-2274306

Résumé

Transmissible gastroenteritis virus (TGEV) is member of the family Coronaviridae and mainly causes acute diarrhea. TGEV infection is characterized by vomiting, watery diarrhea, and severe dehydration, resulting in high mortality rates in neonatal piglets. TGEV infection symptoms are related to an imbalance of sodium absorption in small intestinal epithelial cells; however, the etiology of sodium imbalance diarrhea caused by TGEV remains unclear. In this study, we performed transcriptomic analysis of intestinal tissues from infected and healthy piglets and observed that the expression of NHE3, encoding Na+/H+ exchanger 3 (NHE3), the main exchanger of electroneutral sodium in intestinal epithelial cells, was significantly reduced upon TGEV infection. We also showed that specific inhibition of intestinal NHE3 activity could lead to the development of diarrhea in piglets. Furthermore, we revealed an interaction between TGEV N protein and NHE3 near the nucleus. The binding of TGEV N to NHE3 directly affected the expression and activity of NHE3 on the cell surface and affected cellular electrolyte absorption, leading to diarrhea. Molecular docking and computer-aided screening techniques were used to screen for the blocker of the interaction between TGEV N and NHE3, which identified irinotecan. We then demonstrated that irinotecan was effective in relieving TGEV-induced diarrhea in piglets. These findings provide new insights into the mechanism of TGEV-induced sodium imbalance diarrhea and could lead to the design of novel antiviral strategies against TGEV. IMPORTANCE A variety of coronaviruses have been found to cause severe diarrhea in hosts, including TGEV; however, the pathogenic mechanism is not clear. Therefore, prompt determination of the mechanism and identification of efficient therapeutic agents are required, both for public health reasons and for economic development. In this study, we demonstrated that NHE3 is the major expressed protein of NHEs in the intestine, and its expression decreased by nearly 70% after TGEV infection. Also, specific inhibition of intestinal NHE3 resulted in severe diarrhea in piglets. This demonstrated that NHE3 plays an important role in TGEV-induced diarrhea. In addition, we found that TGEV N directly regulates NHE3 expression and activity through protein-protein interaction, which is essential to promote diarrhea. Molecular docking and other techniques demonstrated that irinotecan could block the interaction and diarrhea caused by TGEV. Thus, our results provide a basis for the development of novel therapeutic agents against TGEV and guidance for the development of drugs for other diarrhea-causing coronaviruses.


Sujets)
Infections à coronavirus , Coronavirus , Virus de la gastroentérite transmissible , Animaux , Suidae , Virus de la gastroentérite transmissible/physiologie , Échangeur-3 de sodium-hydrogène/génétique , Échangeur-3 de sodium-hydrogène/métabolisme , Protéines nucléocapside/métabolisme , Irinotécan , Simulation de docking moléculaire , Diarrhée/médecine vétérinaire , Antiport des ions sodium-hydrogène/métabolisme , Coronavirus/métabolisme , Sodium/métabolisme
8.
Environ Sci Pollut Res Int ; 29(57): 85569-85573, 2022 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-2148925

Résumé

Pentraxin 3 (PTX3) and ficolin are the plasma phase of pattern recognition receptors (PRRs) and can activate complement through classical and lectin pathways, respectively, which may contribute to disease severity. This study aimed to investigate the association between PTX3 and ficolin with disease severity in patients with coronavirus disease-2019 (COVID-19). Seventy-three COVID-19 patients and 25 healthy controls were enrolled in this study. The participants were divided into three groups as follows: 14 patients as the intensive care unit (ICU) group, 59 patients as the non-ICU group, and 25 subjects as the healthy control group. The serum levels of PTX3 and ficolin were measured by enzyme-linked immunosorbent assay (ELISA) kits. Patients in ICU and non-ICU groups had significantly higher levels of PTX3 compared to the healthy control group (p = 0.0002 and p = 0.0072, respectively). Patients in the ICU group also had an increased amount of PTX3 (1957 ± 1769 pg/ml) compared to non-ICU patients (1220 ± 1784 pg/ml). However, this difference was not significant. On the other hand, serum levels of ficolin were not different among the three groups. PTX3, as an acute phase protein, may contribute to disease severity. Its probable inflammatory role could result from the high activation of the complement system. On the other hand, it could be suggested that ficolin has no crucial role in the disease severity of COVID-19 patients.


Sujets)
COVID-19 , Coronavirus , Humains , Protéine C-réactive/analyse , Coronavirus/métabolisme , COVID-19/sang , COVID-19/génétique , COVID-19/métabolisme , Composant sérique amyloïde P/analyse , Composant sérique amyloïde P/métabolisme
10.
Environ Res ; 215(Pt 2): 114288, 2022 Dec.
Article Dans Anglais | MEDLINE | ID: covidwho-2041735

Résumé

There is abundant epidemiological data indicating that the incidence of severe cases of coronavirus disease (COVID-19) is significantly higher in males than females worldwide. Moreover, genetic variation at the X-chromosome linked TLR7 gene has been associated with COVID-19 severity. It has been suggested that the sex-biased incidence of COVID-19 might be related to the fact that TLR7 escapes X-chromosome inactivation during early embryogenesis in females, thus encoding a doble dose of its gene product compared to males. We analyzed TLR7 expression in two acute phase cohorts of COVID-19 patients that used two different technological platforms, one of them in a multi-tissue context including saliva, nasal, and blood samples, and a third cohort that included different post-infection timepoints of long-COVID-19 patients. We additionally explored methylation patterns of TLR7 using epigenomic data from an independent cohort of COVID-19 patients stratified by severity and sex. In line with genome-wide association studies, we provide supportive evidence indicating that TLR7 has altered CpG methylation patterns and it is consistently downregulated in males compared to females in the most severe cases of COVID-19.


Sujets)
COVID-19 , Infections à coronavirus , Coronavirus , COVID-19/complications , COVID-19/épidémiologie , COVID-19/génétique , Coronavirus/génétique , Coronavirus/métabolisme , Méthylation de l'ADN , Épigénomique , Femelle , Étude d'association pangénomique , Humains , Mâle , Récepteur de type Toll-7/génétique , Transcriptome ,
11.
Front Immunol ; 13: 921613, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-2009864

Résumé

Increasing evidence supports the ability of eugenol to maintain intestinal barrier integrity and anti-inflammatory in vitro and in vivo; however, whether eugenol alleviates virus-mediated intestinal barrier damage and inflammation remains a mystery. Transmissible gastroenteritis virus (TGEV), a coronavirus, is one of the main causative agents of diarrhea in piglets and significantly impacts the global swine industry. Here, we found that eugenol could alleviate TGEV-induced intestinal functional impairment and inflammatory responses in piglets. Our results indicated that eugenol improved feed efficiency in TGEV-infected piglets. Eugenol not only increased serum immunoglobulin concentration (IgG) but also significantly decreased serum inflammatory cytokine concentration (TNF-α) in TGEV-infected piglets. In addition, eugenol also significantly decreased the expression of NF-κB mRNA and the phosphorylation level of NF-κB P65 protein in the jejunum mucosa of TGEV-infected piglets. Eugenol increased villus height and the ratio of villus height to crypt depth in the jejunum and ileum, and decreased serum D-lactic acid levels. Importantly, eugenol increased tight junction protein (ZO-1) and mRNA expression levels of nutrient transporter-related genes (GluT-2 and CaT-1) in the jejunum mucosa of TGEV-infected piglets. Meanwhile, compared with TGEV-infected IPEC-J2 cells, treatment with eugenol reduced the cell cytopathic effect, attenuated the inflammatory response. Interestingly, eugenol did not increase the expression of ZO-1 and Occludin in IPEC-J2 cells. However, western blot and immunofluorescence results showed that eugenol restored TGEV-induced down-regulation of ZO-1 and Occludin, while BAY11-7082 (The NF-κB specific inhibitor) enhanced the regulatory ability of eugenol. Our findings demonstrated that eugenol attenuated TGEV-induced intestinal injury by increasing the expression of ZO-1 and Occludin, which may be related to the inhibition of NF-κB signaling pathway. Eugenol may offer some therapeutic opportunities for coronavirus-related diseases.


Sujets)
Coronavirus , Virus de la gastroentérite transmissible , Animaux , Lignée cellulaire , Coronavirus/métabolisme , Eugénol/pharmacologie , Eugénol/usage thérapeutique , Facteur de transcription NF-kappa B/métabolisme , Occludine , ARN messager , Transduction du signal , Suidae , Virus de la gastroentérite transmissible/physiologie
12.
Front Immunol ; 13: 912717, 2022.
Article Dans Anglais | MEDLINE | ID: covidwho-2005867

Résumé

We present evidence suggesting that the severe acute respiratory syndrome (SARS) coronavirus non-structural protein 13 (Nsp13) modulates the Z-RNA dependent regulated cell death pathways . We show that Z-prone sequences [called flipons] exist in coronavirus and provide a signature (Z-sig) that enables identification of the animal viruses from which the human pathogens arose. We also identify a potential RIP Homology Interaction Motif (RHIM) in the helicase Nsp13 that resembles those present in proteins that initiate Z-RNA-dependent cell death through interactions with the Z-RNA sensor protein ZBP1. These two observations allow us to suggest a model in which Nsp13 down regulates Z-RNA activated innate immunity by two distinct mechanisms. The first involves a novel ATP-independent Z-flipon helicase (flipase) activity in Nsp13 that differs from that of canonical A-RNA helicases. This flipase prevents formation of Z-RNAs that would otherwise activate cell death pathways. The second mechanism likely inhibits the interactions between ZBP1 and the Receptor Interacting Proteins Kinases RIPK1 and RIPK3 by targeting their RHIM domains. Together the described Nsp13 RHIM and flipase activities have the potential to alter the host response to coronaviruses and impact the design of drugs targeting the Nsp13 protein. The Z-sig and RHIM domains may provide a way of identifying previously uncharacterized viruses that are potentially pathogenic for humans.


Sujets)
Infections à coronavirus , Coronavirus , Syndrome respiratoire aigu sévère , Virus du SRAS , Animaux , Coronavirus/métabolisme , Helicase/métabolisme , ARN , Virus du SRAS/métabolisme
13.
Viruses ; 14(8)2022 08 09.
Article Dans Anglais | MEDLINE | ID: covidwho-1979415

Résumé

Porcine epidemic diarrhea virus (PEDV) has been endemic in most parts of the world since its emergence in the 1970s. It infects the small intestine and intestinal villous cells, spreads rapidly, and causes infectious intestinal disease characterized by vomiting, diarrhea, and dehydration, leading to high mortality in newborn piglets and causing massive economic losses to the pig industry. The entry of PEDV into cells is mediated by the binding of its spike protein (S protein) to a host cell receptor. Here, we review the structure of PEDV, its strains, and the structure and function of the S protein shared by coronaviruses, and summarize the progress of research on possible host cell receptors since the discovery of PEDV.


Sujets)
Infections à coronavirus , Coronavirus , Virus de la diarrhée porcine épidémique , Maladies des porcs , Animaux , Coronavirus/métabolisme , Infections à coronavirus/médecine vétérinaire , Virus de la diarrhée porcine épidémique/physiologie , Glycoprotéine de spicule des coronavirus/métabolisme , Suidae
14.
ACS Sens ; 7(7): 2084-2092, 2022 07 22.
Article Dans Anglais | MEDLINE | ID: covidwho-1900429

Résumé

With the rapid spread and multigeneration variation of coronavirus, rapid drug development has become imperative. A major obstacle to addressing this issue is adequately constructing the cell membrane at the molecular level, which enables in vitro observation of the cell response to virus and drug molecules quantitatively, shortening the drug experiment cycle. Herein, we propose a rapid and label-free supported lipid bilayer-based lab-on-a-chip biosensor for the screening of effective inhibition drugs. An extended gate electrode was prepared and functionalized by an angiotensin-converting enzyme II (ACE2) receptor-incorporated supported lipid bilayer (SLB). Such an integrated system can convert the interactions of targets and membrane receptors into real-time charge signals. The platform can simulate the cell membrane microenvironment in vitro and accurately capture the interaction signal between the target and the cell membrane with minimized interference, thus observing the drug action pathway quantitatively and realizing drug screening effectively. Due to these label-free, low-cost, convenient, and integrated advantages, it is a suitable candidate method for the rapid drug screening for the early treatment and prevention of worldwide spread of coronavirus.


Sujets)
Techniques de biocapteur , Coronavirus , Membrane cellulaire/métabolisme , Coronavirus/métabolisme , Laboratoires sur puces , Double couche lipidique/métabolisme
15.
Cell ; 185(13): 2279-2291.e17, 2022 06 23.
Article Dans Anglais | MEDLINE | ID: covidwho-1866951

Résumé

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.


Sujets)
Coronavirus humain 229E , Infections à coronavirus , Coronavirus , Animaux , Antigènes CD13/composition chimique , Antigènes CD13/métabolisme , Chats , Lignée cellulaire , Coronavirus/métabolisme , Coronavirus humain 229E/métabolisme , Chiens , Humains , Récepteurs viraux/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Suidae
16.
Histochem Cell Biol ; 158(3): 241-251, 2022 Sep.
Article Dans Anglais | MEDLINE | ID: covidwho-1858993

Résumé

After their assembly by budding into the lumen of the intermediate compartment (IC) at the endoplasmic reticulum (ER)-Golgi interface, coronaviruses (CoVs) are released from their host cells following a pathway that remains poorly understood. The traditional view that CoV exit occurs via the constitutive secretory route has recently been questioned by studies suggesting that this process involves unconventional secretion. Here, using the avian infectious bronchitis virus (IBV) as a well-established model virus, we have applied confocal microscopy to investigate the pathway of CoV egress from epithelial Vero cells. We report a novel effect of IBV infection on cellular endomembranes, namely, the compaction of the pericentrosomal endocytic recycling compartment (ERC) defined by the GTPase Rab11, which coincides with the previously described Golgi fragmentation, as well as virus release. Despite Golgi disassembly, the IC elements containing the major IBV membrane protein (M)-which mostly associates with newly formed virus particles-maintain their close spatial connection with the Rab11-positive endocytic recycling system. Moreover, partial colocalization of the M protein with Rab11 was observed, whereas M displayed negligible overlap with LAMP-1, indicating that IBV egress does not occur via late endosomes or lysosomes. Synchronization of virus release using temperature-shift protocols was accompanied by increased colocalization of M and Rab11 in vesicular and vacuolar structures in the pericentrosomal region and at the cell periphery, most likely representing IBV-containing transport carriers. In conclusion, these results add CoVs to the growing list of viruses exploiting the endocytic recycling apparatus defined by Rab11 for their assembly and/or release.


Sujets)
Coronavirus , Animaux , Chlorocebus aethiops , Coronavirus/métabolisme , Endosomes/métabolisme , Appareil de Golgi/métabolisme , Cellules Vero , Protéines G rab/métabolisme
17.
Virol Sin ; 37(3): 437-444, 2022 Jun.
Article Dans Anglais | MEDLINE | ID: covidwho-1815255

Résumé

The coronavirus 3C-like (3CL) protease, a cysteine protease, plays an important role in viral infection and immune escape. However, there is still a lack of effective tools for determining the cleavage sites of the 3CL protease. This study systematically investigated the diversity of the cleavage sites of the coronavirus 3CL protease on the viral polyprotein, and found that the cleavage motif were highly conserved for viruses in the genera of Alphacoronavirus, Betacoronavirus and Gammacoronavirus. Strong residue preferences were observed at the neighboring positions of the cleavage sites. A random forest (RF) model was built to predict the cleavage sites of the coronavirus 3CL protease based on the representation of residues in cleavage motifs by amino acid indexes, and the model achieved an AUC of 0.96 in cross-validations. The RF model was further tested on an independent test dataset which were composed of cleavage sites on 99 proteins from multiple coronavirus hosts. It achieved an AUC of 0.95 and predicted correctly 80% of the cleavage sites. Then, 1,352 human proteins were predicted to be cleaved by the 3CL protease by the RF model. These proteins were enriched in several GO terms related to the cytoskeleton, such as the microtubule, actin and tubulin. Finally, a webserver named 3CLP was built to predict the cleavage sites of the coronavirus 3CL protease based on the RF model. Overall, the study provides an effective tool for identifying cleavage sites of the 3CL protease and provides insights into the molecular mechanism underlying the pathogenicity of coronaviruses.


Sujets)
Infections à coronavirus , Coronavirus , Algorithmes , Coronavirus/métabolisme , Cysteine endopeptidases/composition chimique , Cysteine endopeptidases/génétique , Cysteine endopeptidases/métabolisme , Humains , Apprentissage machine , Peptide hydrolases/métabolisme , Inhibiteurs de protéases , Protéines virales/métabolisme
18.
Cells ; 11(4)2022 02 11.
Article Dans Anglais | MEDLINE | ID: covidwho-1688673

Résumé

Transmembrane proteins of adherens and tight junctions are known targets for viruses and bacterial toxins. The coronavirus receptor ACE2 has been localized at the apical surface of epithelial cells, but it is not clear whether ACE2 is localized at apical Cell-Cell junctions and whether it associates with junctional proteins. Here we explored the expression and localization of ACE2 and its association with transmembrane and tight junction proteins in epithelial tissues and cultured cells by data mining, immunoblotting, immunofluorescence microscopy, and co-immunoprecipitation experiments. ACE2 mRNA is abundant in epithelial tissues, where its expression correlates with the expression of the tight junction proteins cingulin and occludin. In cultured epithelial cells ACE2 mRNA is upregulated upon differentiation and ACE2 protein is widely expressed and co-immunoprecipitates with the transmembrane proteins ADAM17 and CD9. We show by immunofluorescence microscopy that ACE2 colocalizes with ADAM17 and CD9 and the tight junction protein cingulin at apical junctions of intestinal (Caco-2), mammary (Eph4) and kidney (mCCD) epithelial cells. These observations identify ACE2, ADAM17 and CD9 as new epithelial junctional transmembrane proteins and suggest that the cytokine-enhanced endocytic internalization of junction-associated protein complexes comprising ACE2 may promote coronavirus entry.


Sujets)
Angiotensin-converting enzyme 2/métabolisme , Jonctions intercellulaires/métabolisme , Jonctions intercellulaires/virologie , Protéine ADAM17/métabolisme , Jonctions adhérentes/métabolisme , Angiotensin-converting enzyme 2/génétique , Cadhérines/métabolisme , Protéines de transport/métabolisme , Lignée cellulaire , Perméabilité des membranes cellulaires , Coronavirus/métabolisme , Cellules épithéliales/métabolisme , Cellules épithéliales/virologie , Expression des gènes/génétique , Antigène CD9/métabolisme , Protéines de la jonction serrée/métabolisme , Jonctions serrées/métabolisme , Transcriptome/génétique
19.
Med Sci Monit ; 28: e934102, 2022 Jan 25.
Article Dans Anglais | MEDLINE | ID: covidwho-1651076

Résumé

BACKGROUND Heat-clearing and detoxifying herbs (HDHs) play an important role in the prevention and treatment of coronavirus infection. However, their mechanism of action needs further study. This study aimed to explore the anti-coronavirus basis and mechanism of HDHs. MATERIAL AND METHODS Database mining was performed on 7 HDHs. Core ingredients and targets were screened according to ADME rules combined with Neighborhood, Co-occurrence, Co-expression, and other algorithms. GO enrichment and KEGG pathway analyses were performed using the R language. Finally, high-throughput molecular docking was used for verification. RESULTS HDHs mainly acts on NOS3, EGFR, IL-6, MAPK8, PTGS2, MAPK14, NFKB1, and CASP3 through quercetin, luteolin, wogonin, indirubin alkaloids, ß-sitosterol, and isolariciresinol. These targets are mainly involved in the regulation of biological processes such as inflammation, activation of MAPK activity, and positive regulation of NF-kappaB transcription factor activity. Pathway analysis further revealed that the pathways regulated by these targets mainly include: signaling pathways related to viral and bacterial infections such as tuberculosis, influenza A, Ras signaling pathways; inflammation-related pathways such as the TLR, TNF, MAPK, and HIF-1 signaling pathways; and immune-related pathways such as NOD receptor signaling pathways. These pathways play a synergistic role in inhibiting lung inflammation and regulating immunity and antiviral activity. CONCLUSIONS HDHs play a role in the treatment of coronavirus infection by regulating the body's immunity, fighting inflammation, and antiviral activities, suggesting a molecular basis and new strategies for the treatment of COVID-19 and a foundation for the screening of new antiviral drugs.


Sujets)
, Coronavirus/effets des médicaments et des substances chimiques , Médicaments issus de plantes chinoises/pharmacologie , SARS-CoV-2/effets des médicaments et des substances chimiques , Alcaloïdes/composition chimique , Alcaloïdes/pharmacologie , Caspase-3/effets des médicaments et des substances chimiques , Caspase-3/génétique , Coronavirus/métabolisme , Infections à coronavirus/traitement médicamenteux , Cyclooxygenase 2/effets des médicaments et des substances chimiques , Cyclooxygenase 2/génétique , Bases de données pharmaceutiques , Médicaments issus de plantes chinoises/composition chimique , Médicaments issus de plantes chinoises/usage thérapeutique , Flavanones/composition chimique , Flavanones/pharmacologie , Humains , Indoles/composition chimique , Indoles/pharmacologie , Interleukine-6/génétique , Lignine/composition chimique , Lignine/pharmacologie , Lutéoline/composition chimique , Lutéoline/pharmacologie , Mitogen-Activated Protein Kinase 14/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 14/génétique , Mitogen-Activated Protein Kinase 8/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinase 8/génétique , Simulation de docking moléculaire , Sous-unité p50 de NF-kappa B/effets des médicaments et des substances chimiques , Sous-unité p50 de NF-kappa B/génétique , Naphtols/composition chimique , Naphtols/pharmacologie , Nitric oxide synthase type III/effets des médicaments et des substances chimiques , Nitric oxide synthase type III/génétique , Cartes d'interactions protéiques , Quercétine/composition chimique , Quercétine/pharmacologie , SARS-CoV-2/métabolisme , Transduction du signal , Sitostérol/composition chimique , Sitostérol/pharmacologie , Transcriptome/effets des médicaments et des substances chimiques , Transcriptome/génétique
20.
Viruses ; 13(12)2021 12 17.
Article Dans Anglais | MEDLINE | ID: covidwho-1580424

Résumé

Infectious bronchitis virus (IBV), a gammacoronavirus, is an economically important virus to the poultry industry, as well as a significant welfare issue for chickens. As for all positive strand RNA viruses, IBV infection causes rearrangements of the host cell intracellular membranes to form replication organelles. Replication organelle formation is a highly conserved and vital step in the viral life cycle. Here, we investigate the localization of viral RNA synthesis and the link with replication organelles in host cells. We have shown that sites of viral RNA synthesis and virus-related dsRNA are associated with one another and, significantly, that they are located within a membrane-bound compartment within the cell. We have also shown that some viral RNA produced early in infection remains within these membranes throughout infection, while a proportion is trafficked to the cytoplasm. Importantly, we demonstrate conservation across all four coronavirus genera, including SARS-CoV-2. Understanding more about the replication of these viruses is imperative in order to effectively find ways to control them.


Sujets)
Coronavirus/métabolisme , Membranes intracellulaires/métabolisme , ARN viral/biosynthèse , Animaux , Lignée cellulaire , Coronavirus/classification , Coronavirus/croissance et développement , Cytoplasme/métabolisme , Humains , Virus de la bronchite infectieuse/croissance et développement , Virus de la bronchite infectieuse/métabolisme , ARN double brin/métabolisme , Compartiments de réplication virale/métabolisme
SÉLECTION CITATIONS
Détails de la recherche