Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.554
Filter
1.
Nat Commun ; 15(1): 8708, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379362

ABSTRACT

The precise cellular mechanisms underlying heightened proinflammatory cytokine production during coronavirus infection remain incompletely understood. Here we identify the envelope (E) protein in severe coronaviruses (SARS-CoV-2, SARS, or MERS) as a potent inducer of interleukin-1 release, intensifying lung inflammation through the activation of TMED10-mediated unconventional protein secretion (UcPS). In contrast, the E protein of mild coronaviruses (229E, HKU1, or OC43) demonstrates a less pronounced effect. The E protein of severe coronaviruses contains an SS/DS motif, which is not present in milder strains and facilitates interaction with TMED10. This interaction enhances TMED10-oligomerization, facilitating UcPS cargo translocation into the ER-Golgi intermediate compartment (ERGIC)-a pivotal step in interleukin-1 UcPS. Progesterone analogues were identified as compounds inhibiting E-enhanced release of proinflammatory factors and lung inflammation in a Mouse Hepatitis Virus (MHV) infection model. These findings elucidate a molecular mechanism driving coronavirus-induced hyperinflammation, proposing the E-TMED10 interaction as a potential therapeutic target to counteract the adverse effects of coronavirus-induced inflammation.


Subject(s)
Murine hepatitis virus , SARS-CoV-2 , Animals , Humans , Mice , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Coronavirus Envelope Proteins/metabolism , COVID-19/virology , COVID-19/immunology , COVID-19/metabolism , Golgi Apparatus/metabolism , Coronavirus Infections/virology , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus Infections/drug therapy , HEK293 Cells , Middle East Respiratory Syndrome Coronavirus/immunology , Inflammation/metabolism , Lung/virology , Lung/metabolism , Lung/immunology
2.
BMC Med ; 22(1): 445, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39380062

ABSTRACT

BACKGROUND: Long COVID, also known as post-acute sequelae of COVID-19 (PASC), is a poorly understood condition with symptoms across a range of biological domains that often have debilitating consequences. Some have recently suggested that lingering SARS-CoV-2 virus particles in the gut may impede serotonin production and that low serotonin may drive many Long COVID symptoms across a range of biological systems. Therefore, selective serotonin reuptake inhibitors (SSRIs), which increase synaptic serotonin availability, may be used to prevent or treat Long COVID. SSRIs are commonly prescribed for depression, therefore restricting a study sample to only include patients with depression can reduce the concern of confounding by indication. METHODS: In an observational sample of electronic health records from patients in the National COVID Cohort Collaborative (N3C) with a COVID-19 diagnosis between September 1, 2021, and December 1, 2022, and a comorbid depressive disorder, the leading indication for SSRI use, we evaluated the relationship between SSRI use during acute COVID-19 and subsequent 12-month risk of Long COVID (defined by ICD-10 code U09.9). We defined SSRI use as a prescription for SSRI medication beginning at least 30 days before acute COVID-19 and not ending before SARS-CoV-2 infection. To minimize bias, we estimated relationships using nonparametric targeted maximum likelihood estimation to aggressively adjust for high-dimensional covariates. RESULTS: We analyzed a sample (n = 302,626) of patients with a diagnosis of a depressive condition before COVID-19 diagnosis, where 100,803 (33%) were using an SSRI. We found that SSRI users had a significantly lower risk of Long COVID compared to nonusers (adjusted causal relative risk 0.92, 95% CI (0.86, 0.99)) and we found a similar relationship comparing new SSRI users (first SSRI prescription 1 to 4 months before acute COVID-19 with no prior history of SSRI use) to nonusers (adjusted causal relative risk 0.89, 95% CI (0.80, 0.98)). CONCLUSIONS: These findings suggest that SSRI use during acute COVID-19 may be protective against Long COVID, supporting the hypothesis that serotonin may be a key mechanistic biomarker of Long COVID.


Subject(s)
COVID-19 , SARS-CoV-2 , Selective Serotonin Reuptake Inhibitors , Humans , COVID-19/epidemiology , COVID-19/complications , Selective Serotonin Reuptake Inhibitors/therapeutic use , Female , Male , Middle Aged , SARS-CoV-2/drug effects , Adult , Aged , Depression/drug therapy , Pandemics , Post-Acute COVID-19 Syndrome , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Coronavirus Infections/complications , Betacoronavirus/drug effects , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Risk Factors
3.
Int J Biol Macromol ; 279(Pt 3): 135375, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39244115

ABSTRACT

Porcine deltacoronavirus (PDCoV) poses a significant threat to both the pig industry and public safety, and has recently been identified in humans. Currently, there are no commercially available vaccines or antiviral treatments for PDCoV. In this study, recombinant porcine interferon δ8 (rINF-δ8) expressed by the HEK 293F expression system was used to evaluated its antiviral activity against PDCoV both in vitro and in vivo. Results demonstrated that rIFN-δ8 displayed non-toxic to ST cells and primary PAMs, and effectively inhibited PDCoV replication in a dose-dependent manner in vitro, with complete suppression of virus replication at a concentration of 2 µg/ml. Treatment of piglets with two doses of 25 µg/kg of rIFN-δ8 reduced clinical symptoms, decreased virus shedding, alleviated intestinal damage, and lowered the viral load in the jejunum and ileum. Furthermore, the levels of interferon-stimulated genes (ISGs) such as Viper, Mx1, ISG15, IFIT1, OSA, and IFITM1 were significantly increased both in vitro and in vivo, with elevated ISG levels sustained for at least 3 days in vivo. These findings suggest that rIFN-δ8 has the potential to serve as an effective antiviral agent for preventing PDCoV in pigs in the future.


Subject(s)
Antiviral Agents , Coronavirus Infections , Deltacoronavirus , Recombinant Proteins , Swine Diseases , Virus Replication , Animals , Swine , Recombinant Proteins/pharmacology , Humans , Deltacoronavirus/drug effects , Virus Replication/drug effects , Antiviral Agents/pharmacology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , HEK293 Cells , Swine Diseases/virology , Swine Diseases/drug therapy , Interferons/pharmacology , Viral Load/drug effects
4.
JAMA Netw Open ; 7(9): e2431309, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39240567

ABSTRACT

Importance: Colchicine has many drug-drug interactions with commonly prescribed medications. Only pharmacokinetic studies have provided data on colchicine drug-drug interactions. Objective: To evaluate the clinical tolerability of colchicine according to the presence or absence of a drug-drug interaction. Design, Setting, and Participants: A secondary analysis of the COLCORONA trial was performed. The COLCORONA trial was a randomized, double-blind, placebo-controlled trial conducted in Brazil, Canada, Greece, South Africa, Spain, and the US between March 23, 2020, and January 20, 2021. The COLCORONA trial included ambulatory patients with COVID-19 with at least 1 high-risk characteristic and compared the effects of colchicine (0.5 mg twice daily for 3 days, then 0.5 mg daily thereafter) with placebo for 27 days. Data analysis was performed from February 24, 2023, to June 20, 2024. Exposure: In this secondary analysis, baseline medications that had interactions with colchicine were identified using a previously published expert classification. Main Outcomes and Measures: The primary outcome for this analysis was the composite of serious and nonserious treatment-related and treatment-unrelated gastrointestinal adverse events. The secondary outcomes were other adverse events and the composite of death or hospital admission due to COVID-19 infection. Logistic regression models adjusted for age, sex, estimated glomerular filtration rate, diabetes, heart failure, and myocardial infarction were assessed for effect modification of the association between the randomization arm and the outcomes of interest by drug-drug interaction status. Results: The cohort included 2205 participants in the colchicine arm and 2227 in the placebo arm (median age, 54 [IQR, 47-61] years; 2389 [54%] women). The most common colchicine drug-drug interactions were rosuvastatin (12%) and atorvastatin (10%). In fully adjusted models, the odds of any gastrointestinal adverse event were 1.80 (95% CI, 1.51-2.15) times higher in the colchicine arm than the placebo arm among people without a drug-drug interaction and 1.68 (95% CI, 1.24-2.26) times higher in the colchicine arm than the placebo arm among people with a drug-drug interaction (P = .69 for interaction). Drug-drug interaction status did not significantly modify the effect of colchicine on the composite of COVID-19 hospitalization or death (odds ratio, 0.91; 95% CI, 0.59-1.40 for drug-drug interaction and 0.84; 95% CI, 0.60-1.19 for no drug-drug interaction; P = .80 for interaction). Conclusions and Relevance: In this secondary analysis of the COLCORONA trial, operational classification of drug interactions system class 3 or 4 drug-drug interactions did not appear to significantly increase the risk of colchicine-related adverse effects. Trial Registration: ClinicalTrials.gov Identifier: NCT04322682.


Subject(s)
COVID-19 , Colchicine , Drug Interactions , SARS-CoV-2 , Humans , Colchicine/adverse effects , Colchicine/therapeutic use , Colchicine/pharmacokinetics , Female , Male , Middle Aged , Double-Blind Method , Aged , COVID-19 Drug Treatment , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Pandemics
5.
Influenza Other Respir Viruses ; 18(9): e70006, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39284764

ABSTRACT

INTRODUCTION: There is still a lack of clinical evidence comprehensively evaluating the effectiveness of antiviral treatments for COVID-19 hospitalized patients. METHODS: A retrospective cohort study was conducted at Beijing You'An Hospital, focusing on patients treated with nirmatrelvir/ritonavir or azvudine. The study employed a tripartite analysis-viral dynamics, survival curve analysis, and AI-based radiological analysis of pulmonary CT images-aiming to assess the severity of pneumonia. RESULTS: Of 370 patients treated with either nirmatrelvir/ritonavir or azvudine as monotherapy, those in the nirmatrelvir/ritonavir group experienced faster viral clearance than those treated with azvudine (5.4 days vs. 8.4 days, p < 0.001). No significant differences were observed in the survival curves between the two drug groups. AI-based radiological analysis revealed that patients in the nirmatrelvir group had more severe pneumonia conditions (infection ratio is 11.1 vs. 5.35, p = 0.007). Patients with an infection ratio higher than 9.2 had nearly three times the mortality rate compared to those with an infection ratio lower than 9.2. CONCLUSIONS: Our study suggests that in real-world studies regarding hospitalized patients with COVID-19 pneumonia, the antiviral effect of nirmatrelvir/ritonavir is significantly superior to azvudine, but the choice of antiviral agents is not necessarily linked to clinical outcomes; the severity of pneumonia at admission is the most important factor to determine prognosis. Additionally, our findings indicate that pulmonary AI imaging analysis can be a powerful tool for predicting patient prognosis and guiding clinical decision-making.


Subject(s)
Antiviral Agents , Artificial Intelligence , COVID-19 Drug Treatment , COVID-19 , Ritonavir , SARS-CoV-2 , Humans , Antiviral Agents/therapeutic use , Male , Retrospective Studies , Female , Middle Aged , Ritonavir/therapeutic use , COVID-19/diagnostic imaging , COVID-19/mortality , SARS-CoV-2/drug effects , Aged , Treatment Outcome , Tomography, X-Ray Computed , Hospitalization , Pneumonia, Viral/drug therapy , Pneumonia, Viral/diagnostic imaging , Pneumonia, Viral/mortality , Adult , Pandemics , Coronavirus Infections/drug therapy , Coronavirus Infections/diagnostic imaging , Coronavirus Infections/mortality , Betacoronavirus/drug effects , Drug Combinations , Lung/diagnostic imaging , Lung/drug effects , Lung/virology
6.
Int J Nanomedicine ; 19: 9009-9033, 2024.
Article in English | MEDLINE | ID: mdl-39246425

ABSTRACT

Background: The high infectivity of coronaviruses has led to increased interest in developing new strategies to prevent virus spread. Silver nanoparticles (AgNPs) and graphene oxide (GO) have attracted much attention in the antiviral field. We investigated the potential antiviral activity of GO and AgNPs combined in the nanocomposite GO-Ag against murine betacoronavirus MHV using an in vitro model. Methods: GO, AgNPs, and GO-Ag characterization (size distribution, zeta potential, TEM visualization, FT-IR, and EDX analysis) and XTT assay were performed. The antiviral activity of GO-Ag nanocomposites was evaluated by RT-qPCR and TCID50 assays. The results were compared with free AgNPs and pure GO. Cell growth and morphology of MHV-infected hepatocytes treated with GO-Ag composites were analyzed by JuLI™Br. Immunofluorescence was used to visualize the cell receptor used by MHV. Ultrastructural SEM analysis was performed to examine cell morphology after MHV infection and GO-Ag composite treatment. Results: A significant reduction in virus titer was observed for all nanocomposites tested, ranging from 3.2 to 7.3 log10 TCID50. The highest titer reduction was obtained for GO 5 µg/mL - Ag 25 µg/mL in the post-treatment method. These results were confirmed by RT-qPCR analysis. The results indicate that GO-Ag nanocomposites exhibited better antiviral activity compared to AgNPs and GO. Moreover, the attachment of AgNPs to the GO flake platform reduced their cytotoxicity. In addition, the GO-Ag composite modulates the distribution of the Ceacam1 cell receptor and can modulate cell morphology. Conclusion: Graphene oxide sheets act as a stabilizing agent, inhibiting the accumulation of AgNPs and reducing their cellular toxicity. The GO-Ag composite can physically bind and inhibit murine betacoronavirus from entering cells. Furthermore, the constant presence of GO-Ag can inhibit MHV replication and significantly limit its extracellular release. In conclusion, GO-Ag shows promise as an antiviral coating on solid surfaces to minimize virus transmission and spread.


Subject(s)
Antiviral Agents , Graphite , Metal Nanoparticles , Nanocomposites , Silver , Graphite/pharmacology , Graphite/chemistry , Silver/chemistry , Silver/pharmacology , Animals , Nanocomposites/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Mice , Metal Nanoparticles/chemistry , Murine hepatitis virus/drug effects , Hepatocytes/drug effects , Hepatocytes/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Cell Line
7.
Virus Res ; 349: 199458, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39187047

ABSTRACT

Coronaviruses (CoVs) are significant animal and human pathogens, characterized by being enveloped RNA viruses with positive-sense single-stranded RNA. The Coronaviridae family encompasses four genera, among which gammacoronaviruses pose a major threat to the poultry industry, which infectious bronchitis virus (IBV) being the most prominent of these threats. Particularly, IBV adversely affects broiler growth and egg production, causing substantial losses. The IBV strains currently circulating in Taiwan include the IBV Taiwan-I (TW-I) serotype, IBV Taiwan-II (TW-II) serotype, and vaccine strains. Therefore, ongoing efforts have focused on developing novel vaccines and discovering antiviral agents. The envelope (E) proteins of CoVs accumulate in the endoplasmic reticulum-Golgi intermediate compartment prior to virus budding. These E proteins assemble into viroporins, exhibiting ion channel activity that leads to cell membrane disruption, making them attractive targets for antiviral therapy. In this study, we investigated the E proteins of IBV H-120, as well as IBV serotypes TW-I and TW-II. E protein expression resulted in inhibited bacteria growth, increased permeability of bacteria to ß-galactosidase substrates, and blocked protein synthesis of bacteria by hygromycin B (HygB). Furthermore, in the presence of E proteins, HygB also impeded protein translation in DF-1 cells and damaged their membrane integrity. Collectively, these findings confirm the viroporin activity of the E proteins from IBV H-120, IBV serotype TW-I, and IBV serotype TW-II. Next, the viroporin inhibitors, 5-(N,N-hexamethylene) amiloride (HMA) and 4,4'-diisothiocyano stilbene-2,2'-disulphonic acid (DIDS) were used to inhibit the viroporin activities of the E proteins of IBV H-120, IBV serotype TW-I, and IBV serotype TW-II. In chicken embryos and chickens infected with IBV serotypes TW-I and IBV TW-II, no survivors were observed at 6 and 11 days post-infection (dpi), respectively. However, treatments with both DIDS and HMA increased the survival rates in infected chicken embryos and chickens and mitigated histopathological lesions in the trachea and kidney. Additionally, a 3D pentameric structure of the IBV E protein was constructed via homology modeling. As expected, both inhibitors were found to bind to the lipid-facing surface within the transmembrane domain of the E protein, inhibiting ion conduction. Taken together, our findings provide comprehensive evidence supporting the use of viroporin inhibitors as promising antiviral agents against IBV Taiwan isolates.


Subject(s)
Antiviral Agents , Infectious bronchitis virus , Infectious bronchitis virus/drug effects , Infectious bronchitis virus/genetics , Antiviral Agents/pharmacology , Taiwan , Animals , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Chickens , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics , Poultry Diseases/virology , Poultry Diseases/drug therapy , Viroporin Proteins/antagonists & inhibitors
8.
Int J Biol Macromol ; 278(Pt 2): 134571, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147344

ABSTRACT

Interstitial nephritis is the primary cause of mortality in IBV-infected chickens. Our previous research has demonstrated that Radix Isatidis polysaccharide (RIP) could alleviate this form of interstitial nephritis. To explore the mechanism, SPF chickens and chicken embryonic kidney cells (CEKs) were pre-treated with RIP and subsequently infected with QX-genotype IBV strain. Kidneys were sampled for transcriptomic and metabolomic analyses, and the cecum contents were collected for 16S rRNA gene sequencing. Results showed that pre-treatment with RIP led to a 50 % morbidity reduction in infected-chickens, along with decreased tissue lesion and viral load in the kidneys. Multi-omics analysis indicated three possible pathways (including antioxidant, anti-inflammatory and anti-apoptosis) which associated with RIP's efficacy against interstitial nephritis. Following further validation both in vivo and in vitro, the results showed that pre-treatment with RIP could activate the antioxidant transcription factor Nrf2, stimulate antioxidant enzyme expression, and consequently inhibit oxidative stress. Pre-treatment with RIP could also significantly reduce the expression of NLRP3 inflammasome and apoptosis-associated proteins (including Bax, Caspase-3, and Caspase-9). Additionally, RIP was also observed to promote the growth of beneficial bacteria in the intestine. Overall, pretreatment with RIP can alleviate QX-genotype IBV-induced interstitial nephritis via the Nrf2/NLRP3/Caspase-3 signaling pathway. This study lays the groundwork for the potential use of RIP in controlling avian infectious bronchitis (IB).


Subject(s)
Caspase 3 , Chickens , Infectious bronchitis virus , NF-E2-Related Factor 2 , NLR Family, Pyrin Domain-Containing 3 Protein , Nephritis, Interstitial , Polysaccharides , Signal Transduction , Animals , Infectious bronchitis virus/drug effects , Infectious bronchitis virus/pathogenicity , Signal Transduction/drug effects , Polysaccharides/pharmacology , Polysaccharides/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nephritis, Interstitial/drug therapy , Nephritis, Interstitial/virology , Caspase 3/metabolism , Caspase 3/genetics , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Poultry Diseases/drug therapy , Poultry Diseases/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Coronavirus Infections/veterinary , Genotype , Kidney/drug effects , Kidney/pathology
9.
FASEB J ; 38(16): e23882, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39143727

ABSTRACT

Phillygenin (PHI) is an active ingredient derived from the leaf of Forsythia suspensa that has been found to alleviate inflammation and peroxidation response. Avian infectious bronchitis (IB) is a major threat to poultry industry viral respiratory tract disease that infected with infectious bronchitis virus (IBV). This study investigated the protection of PHI to CEK cell and broiler's tracheal injury triggered by avian infectious bronchitis virus (IBV). The results showed that IBV infection did not cause serious clinical symptoms and slowing-body weight in PHI-treated broilers. The expression of virus loads, pro-inflammation factors (IL-6, TNF-α, and IL-1ß) in CEK cell, and tracheas were decreased compared to the IBV group, exhibiting its potent anti-inflammation. Mechanistically, the study demonstrated that the inhibition of TLR7/MyD88/NF-κB pathway was mainly involved in the protection effect of PHI to inflammation injury. Interestingly, a higher abundance of Firmicutes and Lactobacillus in respiratory tract was observed in PHI-treated broilers than in the IBV group. Significant differences were observed between the IBV group and PHI-treated group in the Ferroptosis, Tryptophan metabolism, and Glutathione metabolism pathways. PHI exhibited potent protection effect on IBV infection and alleviated inflammation injury, mainly through inhibiting TLR7/MyD88/NF-κB pathway. The study encourages further development of PHI, paving the way to its clinical use as a new candidate drug to relieve IBV-induced respiratory symptoms.


Subject(s)
Chickens , Coronavirus Infections , Infectious bronchitis virus , Myeloid Differentiation Factor 88 , NF-kappa B , Poultry Diseases , Toll-Like Receptor 7 , Animals , NF-kappa B/metabolism , Myeloid Differentiation Factor 88/metabolism , Poultry Diseases/drug therapy , Poultry Diseases/microbiology , Poultry Diseases/metabolism , Toll-Like Receptor 7/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Microbiota/drug effects , Signal Transduction/drug effects
10.
Int J Mol Sci ; 25(15)2024 Jul 27.
Article in English | MEDLINE | ID: mdl-39125764

ABSTRACT

NIBV is an acute and highly contagious virus that has a major impact on the poultry industry. Wogonin, as a flavonoid drug, has antiviral effects, but there have been no reports indicating its role in renal injury caused by NIBV infection. The aim of this study is to investigate the antiviral effect of wogonin against NIBV. Renal tubular epithelial cells were isolated and cultured, and divided into four groups: Con, Con+Wog, NIBV and NIBV+Wog. We found that wogonin significantly inhibited the copy number of NIBV and significantly alleviated NIBV-induced cell apoptosis and necrosis. Moreover, wogonin inhibited the reduction in mitochondrial membrane potential and the aberrant opening of mPTP caused by NIBV. In conclusion, wogonin can protect renal tubular epithelial cells from damage by inhibiting the replication of NIBV and preventing mitochondrial apoptosis and necroptosis induced by NIBV.


Subject(s)
Apoptosis , Chickens , Epithelial Cells , Flavanones , Kidney Tubules , Necroptosis , Animals , Flavanones/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , Epithelial Cells/metabolism , Necroptosis/drug effects , Apoptosis/drug effects , Kidney Tubules/virology , Kidney Tubules/drug effects , Kidney Tubules/cytology , Kidney Tubules/pathology , Membrane Potential, Mitochondrial/drug effects , Antiviral Agents/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Poultry Diseases/virology , Poultry Diseases/drug therapy , Virus Replication/drug effects , Cells, Cultured
11.
Sci Rep ; 14(1): 18929, 2024 08 15.
Article in English | MEDLINE | ID: mdl-39147857

ABSTRACT

Porcine Epidemic Diarrhea Virus (PEDV) poses a significant threat to neonatal piglets, particularly due to the limited efficacy of existing vaccines and the scarcity of efficacious therapeutic drugs. Gegen Qinlian Decoction (GQD) has been employed for over two millennia in treating infectious diarrhea. Nonetheless, further scrutiny is required to improve the drug's efficacy and elucidate its underlying mechanisms of action. In this study, a modified GQD (MGQD) was developed and demonstrated its capacity to inhibit the replication of PEDV. Animal trials indicated that MGQD effectively alleviated pathological damage in immune tissues and modulated T-lymphocyte subsets. The integration of network analysis with UHPLC-MS/MS facilitated the identification of active ingredients within MGQD and elucidated the molecular mechanisms underlying its therapeutic effects against PEDV infections. In vitro studies revealed that MGQD significantly impeded PEDV proliferation in IPEC-J2 cells, promoting cellular growth via virucidal activity, inhibition of viral attachment, and disruption of viral biosynthesis. Furthermore, MGQD treatment led to increased expression levels of IFN-α, IFN-ß, and IFN-λ3, while concurrently decreasing the expression of TNF-α, thereby enhancing resistance to PEDV infection in IPEC-J2 cells. In conclusion, our findings suggest that MGQD holds promise as a novel antiviral agent for the treatment of PEDV infections.


Subject(s)
Coronavirus Infections , Drugs, Chinese Herbal , Network Pharmacology , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/drug effects , Swine , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Swine Diseases/drug therapy , Swine Diseases/virology , Antiviral Agents/pharmacology , Virus Replication/drug effects , Cell Line , Tandem Mass Spectrometry , Diarrhea/drug therapy , Diarrhea/virology , Diarrhea/veterinary , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology
12.
Viruses ; 16(8)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39205203

ABSTRACT

The objective of this study was to elucidate the mechanism of action of the active components of Coptidis rhizoma against porcine epidemic diarrhea and to provide a theoretical foundation for further development of novel anti-PED therapeutic agents based on Coptidis rhizoma. The potential targets of Coptidis rhizoma against PEDV were identified through a comprehensive literature review and analysis using the TCMSP pharmacological database, SwissDrugDesign database, GeneCards database, and UniProt database. Subsequently, the STRING database and Cytoscape 3.7.1 software were employed to construct a protein-protein interaction (PPI) network and screen key targets. Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were conducted on the identified targets. Molecular docking studies were performed using AutoDock 1.5.7 software to analyze the binding energy and modes of interaction between the active components of Coptidis rhizoma and the target proteins. The PyMOL 2.5.0a0 software was employed to visualize the docking results. Through comprehensive analysis, 74 specific targets of active components of Coptidis rhizoma against PEDV were identified. The core gene targets were screened, and an interaction network diagram was subsequently generated. Ultimately, 14 core targets were identified, with STAT3, ESR1, CASP3, and SRC exhibiting the most significant interactions. GO enrichment analysis revealed a total of 215 molecular items, including 48 biological function items, 139 biological process items, and 28 cellular component items. KEGG enrichment analysis identified 140 signaling pathways. Molecular docking analysis demonstrated that epiberberine and palmatine exhibited high binding affinity with STAT3 protein, worenine showed high binding affinity with ESR1 protein, obacunone exhibited high binding affinity with CASP3 protein, and epiberberine, obacunone, berberine, and berberruine exhibited high binding affinity with SRC protein. A network pharmacology and molecular docking technology approach was employed to screen six important active components of Coptidis rhizoma and four important potential targets against PEDV infection. The findings indicated that the active components of Coptidis rhizoma could serve as promising pharmaceutical agents for the prevention and control of PEDV, with significant potential for clinical application.


Subject(s)
Drugs, Chinese Herbal , Molecular Docking Simulation , Network Pharmacology , Porcine epidemic diarrhea virus , Protein Interaction Maps , Animals , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/chemistry , Swine , Porcine epidemic diarrhea virus/drug effects , Protein Interaction Maps/drug effects , Swine Diseases/drug therapy , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Coptis chinensis , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Gene Ontology
13.
Virol J ; 21(1): 157, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992629

ABSTRACT

Newborn piglets' health is seriously threatened by the porcine epidemic diarrhea virus (PEDV), which also has a significant effect on the pig industry. The gut microbiota produces butyrate, an abundant metabolite that modulates intestinal function through many methods to improve immunological and intestinal barrier function. The objective of this investigation was to ascertain how elevated butyrate concentrations impacted the host transcriptional profile of PEDV CV777 strain infection. Our findings showed that higher concentrations of butyrate have a stronger inhibitory effect on PEDV CV777 strain infection. According to RNA-seq data, higher concentrations of butyrate induced more significant transcriptional changes in IPEC-J2 cells, and signaling pathways such as PI3K-AKT may play a role in the inhibition of PEDV CV777 strain by high concentrations of butyrate. Ultimately, we offer a theoretical and experimental framework for future research and development of novel approaches to harness butyrate's antiviral infection properties.


Subject(s)
Butyrates , Epithelial Cells , Porcine epidemic diarrhea virus , Animals , Porcine epidemic diarrhea virus/drug effects , Porcine epidemic diarrhea virus/physiology , Swine , Butyrates/pharmacology , Butyrates/metabolism , Epithelial Cells/virology , Epithelial Cells/drug effects , Cell Line , Swine Diseases/virology , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/veterinary , Antiviral Agents/pharmacology , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/virology , Intestinal Mucosa/drug effects , Virus Replication/drug effects , Intestines/virology
14.
EMBO Rep ; 25(8): 3547-3573, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39009832

ABSTRACT

The COVID-19 pandemic reminded us of the urgent need for new antivirals to control emerging infectious diseases and potential future pandemics. Immunotherapy has revolutionized oncology and could complement the use of antivirals, but its application to infectious diseases remains largely unexplored. Nucleoside analogs are a class of agents widely used as antiviral and anti-neoplastic drugs. Their antiviral activity is generally based on interference with viral nucleic acid replication or transcription. Based on our previous work and computer modeling, we hypothesize that antiviral adenosine analogs, like remdesivir, have previously unrecognized immunomodulatory properties which contribute to their therapeutic activity. In the case of remdesivir, we here show that these properties are due to its metabolite, GS-441524, acting as an Adenosine A2A Receptor antagonist. Our findings support a new rationale for the design of next-generation antiviral agents with dual - immunomodulatory and intrinsic - antiviral properties. These compounds could represent game-changing therapies to control emerging viral diseases and future pandemics.


Subject(s)
Adenosine Monophosphate , Adenosine , Alanine , Antiviral Agents , COVID-19 , SARS-CoV-2 , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine/chemistry , Humans , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Alanine/analogs & derivatives , Alanine/pharmacology , Alanine/chemistry , COVID-19/immunology , COVID-19/virology , Animals , Immunomodulating Agents/pharmacology , Immunomodulating Agents/chemistry , Adenosine A2 Receptor Antagonists/pharmacology , Adenosine A2 Receptor Antagonists/chemistry , Adenosine A2 Receptor Antagonists/therapeutic use , Pandemics , COVID-19 Drug Treatment , Chlorocebus aethiops , Virus Replication/drug effects , Vero Cells , Betacoronavirus/drug effects , Betacoronavirus/immunology , Receptor, Adenosine A2A/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/virology
15.
Expert Opin Drug Discov ; 19(9): 1023-1041, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39078037

ABSTRACT

INTRODUCTION: Highly pathogenic coronaviruses (CoVs), such as severe acute respiratory syndrome CoV (SARS-CoV), Middle East respiratory syndrome CoV (MERS-CoV), and the most recent SARS-CoV-2 responsible for the COVID-19 pandemic, pose significant threats to human populations over the past two decades. These CoVs have caused a broad spectrum of clinical manifestations ranging from asymptomatic to severe distress syndromes (ARDS), resulting in high morbidity and mortality. AREAS COVERED: The accelerated advancements in antiviral drug discovery, spurred by the COVID-19 pandemic, have shed new light on the imperative to develop treatments effective against a broad spectrum of CoVs. This perspective discusses strategies and lessons learnt in targeting viral non-structural proteins, structural proteins, drug repurposing, and combinational approaches for the development of antivirals against CoVs. EXPERT OPINION: Drawing lessons from the pandemic, it becomes evident that the absence of efficient broad-spectrum antiviral drugs increases the vulnerability of public health systems to the potential onslaught by highly pathogenic CoVs. The rapid and sustained spread of novel CoVs can have devastating consequences without effective and specifically targeted treatments. Prioritizing the effective development of broad-spectrum antivirals is imperative for bolstering the resilience of public health systems and mitigating the potential impact of future highly pathogenic CoVs.


Subject(s)
Antiviral Agents , COVID-19 Drug Treatment , Coronavirus Infections , Drug Discovery , Drug Repositioning , Animals , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Coronavirus Infections/complications , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , COVID-19/complications , COVID-19/epidemiology , COVID-19/virology , Drug Development/methods , Drug Discovery/methods , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Pandemics/prevention & control
16.
Int J Biol Macromol ; 276(Pt 1): 133706, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38981557

ABSTRACT

Main proteases (Mpros) are a class of conserved cysteine hydrolases among coronaviruses and play a crucial role in viral replication. Therefore, Mpros are ideal targets for the development of pan-coronavirus drugs. X77, previously developed against SARS-CoV Mpro, was repurposed as a non-covalent tight binder inhibitor against SARS-CoV-2 Mpro during COVID-19 pandemic. Many novel inhibitors with favorable efficacy have been discovered using X77 as a reference, suggesting that X77 could be a valuable scaffold for drug design. However, the broad-spectrum performance of X77 and underlying mechanism remain less understood. Here, we reported the crystal structures of Mpros from SARS-CoV-2, SARS-CoV, and MERS-CoV, and several Mpro mutants from SARS-CoV-2 variants bound to X77. A detailed analysis of these structures revealed key structural determinants essential for interaction and elucidated the binding modes of X77 with different coronaviral Mpros. The potencies of X77 against these investigated Mpros were further evaluated through molecular dynamic simulation and binding free energy calculation. These data provide molecular insights into broad-spectrum inhibition against coronaviral Mpros by X77 and the similarities and differences of X77 when bound to various Mpros, which will promote X77-based design of novel antivirals with broad-spectrum efficacy against different coronaviruses and SARS-CoV-2 variants.


Subject(s)
Coronavirus 3C Proteases , Molecular Dynamics Simulation , SARS-CoV-2 , SARS-CoV-2/enzymology , SARS-CoV-2/drug effects , Crystallography, X-Ray , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Humans , Protein Binding , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , COVID-19/virology , Severe acute respiratory syndrome-related coronavirus/enzymology , Betacoronavirus/enzymology , Betacoronavirus/drug effects , Middle East Respiratory Syndrome Coronavirus/enzymology , Middle East Respiratory Syndrome Coronavirus/drug effects , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , Binding Sites , Coronavirus Infections/virology , Coronavirus Infections/drug therapy , Pandemics , Pneumonia, Viral/virology , Pneumonia, Viral/drug therapy
17.
Int J Antimicrob Agents ; 64(3): 107281, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39047913

ABSTRACT

OBJECTIVES: Immunocompromised patients may experience prolonged shedding of influenza virus potentially leading to severe infections. Alternatives to monotherapy with neuraminidase inhibitors should be evaluated to entirely suppress viral replication and prevent drug-resistant mutations. METHODS: We investigated the clinical and virological evolution in a case of persistent influenza A and human coronavirus OC43 (HCoV-OC43) coinfection in a hematopoietic stem cell transplant recipient after different therapeutic strategies. RESULTS: Successive oseltamivir and zanamivir monotherapies failed to control both infections, with positive results persisting for over 110 days each. This led to the emergence of highly resistant oseltamivir strains due to neuraminidase mutations (E119V and R292K) followed by a deletion (del245-248), while maintaining sensitivity to zanamivir. The intra-host viral diversity data showed that the treatments impacted viral diversity of influenza virus, but not of HCoV-OC43. Considering the patient's underlying condition and the impact of prolonged viral shedding on pulmonary function, eradicating the influenza virus was necessary. A 10-day regimen combining zanamivir and baloxavir-marboxil effectively controlled influenza virus replication and was associated with the clearance of HCoV-OC43, finally resulting in comprehensive respiratory recovery. CONCLUSION: These observations underscore the importance of further investigating combination treatments as the primary approach to achieve influenza eradication in immunocompromised patients.


Subject(s)
Antiviral Agents , Dibenzothiepins , Hematopoietic Stem Cell Transplantation , Influenza, Human , Morpholines , Pyridones , Triazines , Zanamivir , Humans , Zanamivir/therapeutic use , Zanamivir/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Antiviral Agents/therapeutic use , Antiviral Agents/pharmacology , Influenza, Human/drug therapy , Influenza, Human/virology , Pyridones/therapeutic use , Dibenzothiepins/therapeutic use , Morpholines/therapeutic use , Triazines/therapeutic use , Triazines/pharmacology , Coronavirus OC43, Human/drug effects , Coronavirus OC43, Human/genetics , Drug Resistance, Viral/genetics , Influenza A virus/drug effects , Influenza A virus/genetics , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Immunocompromised Host , Male , Drug Therapy, Combination , Middle Aged , Virus Shedding/drug effects , Virus Replication/drug effects , Female
18.
Antimicrob Agents Chemother ; 68(8): e0057324, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39016593

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has triggered a serious global health crisis, resulting in millions of reported deaths since its initial identification in China in November 2019. The global disparities in immunization access emphasize the urgent need for ongoing research into therapeutic interventions. This study focuses on the potential use of molecular dihydrogen (H2) inhalation as an adjunctive treatment for COVID-19. H2 therapy shows promise in inhibiting intracellular signaling pathways associated with inflammation, particularly when administered early in conjunction with nasal oxygen therapy. This phase I study, characterized by an open-label, prospective, monocentric, and single ascending-dose design, seeks to assess the safety and tolerability of the procedure in individuals with confirmed SARS-CoV-2 infection. Employing a 3 + 3 design, the study includes three exposure durations (target durations): 1 day (D1), 3 days (D2), and 6 days (D3). We concluded that the maximum tolerated duration is at least 3 days. Every patient showed clinical improvement and excellent tolerance to H2 therapy. To the best of our knowledge, this phase I clinical trial is the first to establish the safety of inhaling a mixture of H2 (3.6%) and N2 (96.4%) in hospitalized COVID-19 patients. The original device and method employed ensure the absence of explosion risk. The encouraging outcomes observed in the 12 patients included in the study justify further exploration through larger, controlled clinical trials. CLINICAL TRIALS: This study is registered with ClinicalTrials.gov as NCT04633980.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , COVID-19/therapy , Male , Middle Aged , Administration, Inhalation , Prospective Studies , Female , Adult , Pandemics , Aged , COVID-19 Drug Treatment , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Betacoronavirus
19.
Viruses ; 16(7)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39066320

ABSTRACT

Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) continues to be a global threat due to its ability to evolve and generate new subvariants, leading to new waves of infection. Additionally, other coronaviruses like Middle East respiratory syndrome coronavirus (MERS-CoV, formerly known as hCoV-EMC), which first emerged in 2012, persist and continue to present a threat of severe illness to humans. The continued identification of novel coronaviruses, coupled with the potential for genetic recombination between different strains, raises the possibility of new coronavirus clades of global concern emerging. As a result, there is a pressing need for pan-CoV therapeutic drugs and vaccines. After the extensive optimization of an HCV protease inhibitor screening hit, a novel 3CLPro inhibitor (MK-7845) was discovered and subsequently profiled. MK-7845 exhibited nanomolar in vitro potency with broad spectrum activity against a panel of clinical SARS-CoV-2 subvariants and MERS-CoV. Furthermore, when administered orally, MK-7845 demonstrated a notable reduction in viral burdens by >6 log orders in the lungs of transgenic mice infected with SARS-CoV-2 (K18-hACE2 mice) and MERS-CoV (K18-hDDP4 mice).


Subject(s)
Antiviral Agents , SARS-CoV-2 , Animals , Mice , SARS-CoV-2/drug effects , Humans , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Middle East Respiratory Syndrome Coronavirus/drug effects , Middle East Respiratory Syndrome Coronavirus/genetics , COVID-19 Drug Treatment , Protease Inhibitors/pharmacology , COVID-19/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/virology
20.
Vet Res ; 55(1): 92, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39049059

ABSTRACT

Swine acute diarrhoea syndrome coronavirus (SADS-CoV), which originates from zoonotic transmission of bat coronaviruses in the HKU2 lineage, causes severe illness in pigs and carries a high risk of spreading to humans. At present, there are no licenced therapeutics for the treatment of SADS-CoV. In this study, we examined the effectiveness of recombinant porcine interferon delta 8 (IFN-δ8) against SADS-CoV both in vitro and in vivo. In vitro experiments showed that IFN-δ8 inhibited SADS-CoV proliferation in a concentration-dependent manner, with complete inhibition occurring at a concentration of 5 µg/mL. In vivo experiments demonstrated that two 50 µg/kg doses of IFN-δ8 injected intraperitoneally protected piglets against lethal challenge, blocked viral shedding, attenuated intestinal damage, and decreased the viral load in the jejunum and ileum. Further findings suggested that IFN-δ8 inhibited SADS-CoV infection by increasing the expression of IFN-stimulated genes. These results indicate that IFN-δ8 shows promise as a biological macromolecule drug against SADS-CoV infection.


Subject(s)
Coronavirus Infections , Recombinant Proteins , Swine Diseases , Animals , Swine , Swine Diseases/virology , Swine Diseases/drug therapy , Coronavirus Infections/veterinary , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Interferons , Coronavirus/drug effects , Coronavirus/physiology , Antiviral Agents/pharmacology , Alphacoronavirus
SELECTION OF CITATIONS
SEARCH DETAIL