Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Proc Natl Acad Sci U S A ; 119(26): e2122897119, 2022 06 28.
Article in English | MEDLINE | ID: covidwho-1890411

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evolves rapidly under the pressure of host immunity, as evidenced by waves of emerging variants despite effective vaccinations, highlighting the need for complementing antivirals. We report that targeting a pyrimidine synthesis enzyme restores inflammatory response and depletes the nucleotide pool to impede SARS-CoV-2 infection. SARS-CoV-2 deploys Nsp9 to activate carbamoyl-phosphate synthetase, aspartate transcarbamoylase, and dihydroorotase (CAD) that catalyzes the rate-limiting steps of the de novo pyrimidine synthesis. Activated CAD not only fuels de novo nucleotide synthesis but also deamidates RelA. While RelA deamidation shuts down NF-κB activation and subsequent inflammatory response, it up-regulates key glycolytic enzymes to promote aerobic glycolysis that provides metabolites for de novo nucleotide synthesis. A newly synthesized small-molecule inhibitor of CAD restores antiviral inflammatory response and depletes the pyrimidine pool, thus effectively impeding SARS-CoV-2 replication. Targeting an essential cellular metabolic enzyme thus offers an antiviral strategy that would be more refractory to SARS-CoV-2 genetic changes.


Subject(s)
Antiviral Agents , Aspartate Carbamoyltransferase , COVID-19 Drug Treatment , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing) , Dihydroorotase , Enzyme Inhibitors , Pyrimidines , SARS-CoV-2 , Virus Replication , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Aspartate Carbamoyltransferase/antagonists & inhibitors , Carbamoyl-Phosphate Synthase (Glutamine-Hydrolyzing)/antagonists & inhibitors , Dihydroorotase/antagonists & inhibitors , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Inflammation/drug therapy , Mice , Pyrimidines/antagonists & inhibitors , Pyrimidines/biosynthesis , RNA-Binding Proteins/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Transcription Factor RelA/metabolism , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
2.
Int J Mol Sci ; 23(3)2022 Jan 19.
Article in English | MEDLINE | ID: covidwho-1625123

ABSTRACT

SARS-CoV-2 uses the human cell surface protein angiotensin converting enzyme 2 (ACE2) as the receptor by which it gains access into lung and other tissue. Early in the pandemic, there was speculation that a number of commonly used medications-including ibuprofen and other non-steroidal anti-inflammatory drugs (NSAIDs)-have the potential to upregulate ACE2, thereby possibly facilitating viral entry and increasing the severity of COVID-19. We investigated the influence of the NSAIDS with a range of cyclooxygenase (COX)1 and COX2 selectivity (ibuprofen, flurbiprofen, etoricoxib) and paracetamol on the level of ACE2 mRNA/protein expression and activity as well as their influence on SARS-CoV-2 infection levels in a Caco-2 cell model. We also analysed the ACE2 mRNA/protein levels and activity in lung, heart and aorta in ibuprofen treated mice. The drugs had no effect on ACE2 mRNA/protein expression and activity in the Caco-2 cell model. There was no up-regulation of ACE2 mRNA/protein expression and activity in lung, heart and aorta tissue in ibuprofen-treated mice in comparison to untreated mice. Viral load was significantly reduced by both flurbiprofen and ibuprofen at high concentrations. Ibuprofen, flurbiprofen, etoricoxib and paracetamol demonstrated no effects on ACE2 expression or activity in vitro or in vivo. Higher concentrations of ibuprofen and flurbiprofen reduced SARS-CoV-2 replication in vitro.


Subject(s)
Angiotensin-Converting Enzyme 2 , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , COVID-19/genetics , Acetaminophen/pharmacology , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/metabolism , COVID-19/pathology , Caco-2 Cells , Disease Progression , Enzyme Activation/drug effects , Etoricoxib/pharmacology , Flurbiprofen/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Ibuprofen/pharmacology , Male , Mice , Mice, Inbred C57BL , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Internalization/drug effects
3.
Front Immunol ; 12: 750969, 2021.
Article in English | MEDLINE | ID: covidwho-1551506

ABSTRACT

The COVID-19 is an infectious disease caused by SARS-CoV-2 infection. A large number of clinical studies found high-level expression of pro-inflammatory cytokines in patients infected with SARS-CoV-2, which fuels the rapid development of the disease. However, the specific molecular mechanism is still unclear. In this study, we found that SARS-CoV-2 Nsp5 can induce the expression of cytokines IL-1ß, IL-6, TNF-α, and IL-2 in Calu-3 and THP1 cells. Further research found that Nsp5 enhances cytokine expression through activating the NF-κB signaling pathway. Subsequently, we investigated the upstream effectors of the NF-κB signal pathway on Nsp5 overexpression and discovered that Nsp5 increases the protein level of MAVS. Moreover, Nsp5 can promote the SUMOylation of MAVS to increase its stability and lead to increasing levels of MAVS protein, finally triggering activation of NF-κB signaling. The knockdown of MAVS and the inhibitor of SUMOylation treatment can attenuate Nsp5-mediated NF-κB activation and cytokine induction. We identified a novel role of SARS-CoV-2 Nsp5 to enhance cytokine production by activating the NF-κB signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Coronavirus 3C Proteases/immunology , Cytokines/biosynthesis , NF-kappa B/metabolism , SARS-CoV-2/immunology , Sumoylation/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , COVID-19/immunology , Cell Line , Chlorocebus aethiops , Enzyme Activation/drug effects , HEK293 Cells , Humans , Immunity, Innate/immunology , Interleukin-1beta/biosynthesis , Interleukin-2/biosynthesis , Interleukin-6/biosynthesis , Signal Transduction/physiology , Sumoylation/drug effects , THP-1 Cells , Tumor Necrosis Factor-alpha/biosynthesis , Vero Cells
4.
Viruses ; 13(9)2021 09 14.
Article in English | MEDLINE | ID: covidwho-1411090

ABSTRACT

The porcine epidemic diarrhea virus (PEDV) is an Alphacoronavirus (α-CoV) that causes high mortality in infected piglets, resulting in serious economic losses in the farming industry. Hypericin is a dianthrone compound that has been shown as an antiviral activity on several viruses. Here, we first evaluated the antiviral effect of hypericin in PEDV and found the viral replication and egression were significantly reduced with hypericin post-treatment. As hypericin has been shown in SARS-CoV-2 that it is bound to viral 3CLpro, we thus established a molecular docking between hypericin and PEDV 3CLpro using different software and found hypericin bound to 3CLpro through two pockets. These binding pockets were further verified by another docking between hypericin and PEDV 3CLpro pocket mutants, and the fluorescence resonance energy transfer (FRET) assay confirmed that hypericin inhibits the PEDV 3CLpro activity. Moreover, the alignments of α-CoV 3CLpro sequences or crystal structure revealed that the pockets mediating hypericin and PEDV 3CLpro binding were highly conserved, especially in transmissible gastroenteritis virus (TGEV). We then validated the anti-TGEV effect of hypericin through viral replication and egression. Overall, our results push forward that hypericin was for the first time shown to have an inhibitory effect on PEDV and TGEV by targeting 3CLpro, and it deserves further attention as not only a pan-anti-α-CoV compound but potentially also as a compound of other coronaviral infections.


Subject(s)
Alphacoronavirus/drug effects , Alphacoronavirus/physiology , Anthracenes/pharmacology , Antiviral Agents/pharmacology , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus Infections/virology , Perylene/analogs & derivatives , Virus Replication/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Coronavirus 3C Proteases/chemistry , Enzyme Activation/drug effects , Models, Molecular , Perylene/pharmacology , Porcine epidemic diarrhea virus/drug effects , Recombinant Proteins , Structure-Activity Relationship , Swine , Swine Diseases/virology , Vero Cells
5.
Angew Chem Int Ed Engl ; 60(40): 21662-21667, 2021 09 27.
Article in English | MEDLINE | ID: covidwho-1363645

ABSTRACT

There is an urgent need to develop antiviral drugs and alleviate the current COVID-19 pandemic. Herein we report the design and construction of chimeric oligonucleotides comprising a 2'-OMe-modified antisense oligonucleotide and a 5'-phosphorylated 2'-5' poly(A)4 (4A2-5 ) to degrade envelope and spike RNAs of SARS-CoV-2. The oligonucleotide was used for searching and recognizing target viral RNA sequence, and the conjugated 4A2-5 was used for guided RNase L activation to sequence-specifically degrade viral RNAs. Since RNase L can potently cleave single-stranded RNA during innate antiviral response, degradation efficiencies with these chimeras were twice as much as those with only antisense oligonucleotides for both SARS-CoV-2 RNA targets. In pseudovirus infection models, chimera-S4 achieved potent and broad-spectrum inhibition of SARS-CoV-2 and its N501Y and/or ΔH69/ΔV70 mutants, indicating a promising antiviral agent based on the nucleic acid-hydrolysis targeting chimera (NATAC) strategy.


Subject(s)
Antiviral Agents/pharmacology , Endoribonucleases/metabolism , Enzyme Activation/drug effects , Oligonucleotides, Antisense/pharmacology , SARS-CoV-2/drug effects , Animals , Chlorocebus aethiops , Coronavirus Envelope Proteins/genetics , Drug Design , HEK293 Cells , Humans , Hydrolysis/drug effects , Microbial Sensitivity Tests , Mutation , RNA, Viral/metabolism , Spike Glycoprotein, Coronavirus/genetics , Vero Cells
6.
Sci Immunol ; 6(59)2021 05 18.
Article in English | MEDLINE | ID: covidwho-1234281

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global pandemic, resulting millions of infections and deaths with few effective interventions available. Here, we demonstrate that SARS-CoV-2 evades interferon (IFN) activation in respiratory epithelial cells, resulting in a delayed response in bystander cells. Since pretreatment with IFNs can block viral infection, we reasoned that pharmacological activation of innate immune pathways could control SARS-CoV-2 infection. To identify potent antiviral innate immune agonists, we screened a panel of 75 microbial ligands that activate diverse signaling pathways and identified cyclic dinucleotides (CDNs), canonical STING agonists, as antiviral. Since CDNs have poor bioavailability, we tested the small molecule STING agonist diABZI, and found that it potently inhibits SARS-CoV-2 infection of diverse strains including variants of concern (B.1.351) by transiently stimulating IFN signaling. Importantly, diABZI restricts viral replication in primary human bronchial epithelial cells and in mice in vivo. Our study provides evidence that activation of STING may represent a promising therapeutic strategy to control SARS-CoV-2.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , COVID-19/prevention & control , Interferons/immunology , Membrane Proteins/agonists , Animals , Cell Line , Chlorocebus aethiops , Enzyme Activation/drug effects , Epithelial Cells/virology , Humans , Immune Evasion/immunology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , SARS-CoV-2/growth & development , SARS-CoV-2/immunology , Vero Cells , Virus Replication/drug effects
7.
Sci Immunol ; 6(59)2021 05 18.
Article in English | MEDLINE | ID: covidwho-1234280

ABSTRACT

Coronaviruses are a family of RNA viruses that cause acute and chronic diseases of the upper and lower respiratory tract in humans and other animals. SARS-CoV-2 is a recently emerged coronavirus that has led to a global pandemic causing a severe respiratory disease known as COVID-19 with significant morbidity and mortality worldwide. The development of antiviral therapeutics are urgently needed while vaccine programs roll out worldwide. Here we describe a diamidobenzimidazole compound, diABZI-4, that activates STING and is highly effective in limiting SARS-CoV-2 replication in cells and animals. diABZI-4 inhibited SARS-CoV-2 replication in lung epithelial cells. Administration of diABZI-4 intranasally before or even after virus infection conferred complete protection from severe respiratory disease in K18-ACE2-transgenic mice infected with SARS-CoV-2. Intranasal delivery of diABZI-4 induced a rapid short-lived activation of STING, leading to transient proinflammatory cytokine production and lymphocyte activation in the lung associated with inhibition of viral replication. Our study supports the use of diABZI-4 as a host-directed therapy which mobilizes antiviral defenses for the treatment and prevention of COVID-19.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , COVID-19 Drug Treatment , COVID-19/prevention & control , Membrane Proteins/agonists , SARS-CoV-2/drug effects , A549 Cells , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Chlorocebus aethiops , Enzyme Activation/drug effects , Epithelial Cells/virology , Female , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Male , Membrane Proteins/metabolism , Mice , Mice, Knockout , SARS-CoV-2/growth & development , Vero Cells , Virus Replication/drug effects
8.
EMBO J ; 39(21): e106057, 2020 11 02.
Article in English | MEDLINE | ID: covidwho-846583

ABSTRACT

Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2 and has spread across the globe. SARS-CoV-2 is a highly infectious virus with no vaccine or antiviral therapy available to control the pandemic; therefore, it is crucial to understand the mechanisms of viral pathogenesis and the host immune responses to SARS-CoV-2. SARS-CoV-2 is a new member of the betacoronavirus genus like other closely related viruses including SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV). Both SARS-CoV and MERS-CoV have caused serious outbreaks and epidemics in the past eighteen years. Here, we report that one of the interferon-stimulated genes (ISGs), cholesterol 25-hydroxylase (CH25H), is induced by SARS-CoV-2 infection in vitro and in COVID-19-infected patients. CH25H converts cholesterol to 25-hydrocholesterol (25HC) and 25HC shows broad anti-coronavirus activity by blocking membrane fusion. Furthermore, 25HC inhibits USA-WA1/2020 SARS-CoV-2 infection in lung epithelial cells and viral entry in human lung organoids. Mechanistically, 25HC inhibits viral membrane fusion by activating the ER-localized acyl-CoA:cholesterol acyltransferase (ACAT) which leads to the depletion of accessible cholesterol from the plasma membrane. Altogether, our results shed light on a potentially broad antiviral mechanism by 25HC through depleting accessible cholesterol on the plasma membrane to suppress virus-cell fusion. Since 25HC is a natural product with no known toxicity at effective concentrations, it provides a potential therapeutic candidate for COVID-19 and emerging viral diseases in the future.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Cholesterol/metabolism , Coronavirus Infections/drug therapy , Pneumonia, Viral/drug therapy , Respiratory Mucosa/virology , Steroid Hydroxylases/pharmacology , Virus Internalization/drug effects , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , COVID-19 , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Chlorocebus aethiops , Enzyme Activation/drug effects , Humans , Middle East Respiratory Syndrome Coronavirus/drug effects , Organoids/virology , Pandemics , Respiratory Mucosa/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , SARS-CoV-2 , Vero Cells , COVID-19 Drug Treatment
9.
Med Hypotheses ; 143: 110142, 2020 Oct.
Article in English | MEDLINE | ID: covidwho-671909

ABSTRACT

BACKGROUND: Pulmonary hypertension is a significant complication for some patients with COVID-19 pneumonia, especially those requiring intensive care. Tachyphylaxis to the current therapy, inhaled nitric oxide (iNO), is also common. In vitro, folic acid directly increases nitric oxide (NO) production and extends its duration of action; effects which could be of benefit in reversing pulmonary hypertension and severe hypoxaemia. Our work has shown that, in the systemic circulation, folic acid in high dose rapidly improves nitric oxide mediated vasodilation, by activating endothelial nitric oxide synthase (eNOS). HYPOTHESIS: A similar effect of high dose folic acid on pulmonary endothelial function would be expected from the same mechanism and would lead to improvement in pulmonary perfusion. We therefore hypothesise that folic acid, 5 mg or greater, is a useful therapeutic option for pulmonary hypertension and/or refractory severe hypoxaemia, in patients with severe COVID-19 associated pneumonia in whom NO therapy is considered, with a very low risk of adverse effects.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Folic Acid/therapeutic use , Hypertension, Pulmonary/drug therapy , Nitric Oxide/metabolism , Pandemics , Pneumonia, Viral/complications , Administration, Inhalation , Animals , COVID-19 , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Activation/drug effects , Folic Acid/administration & dosage , Folic Acid/pharmacology , Humans , Hypertension, Pulmonary/complications , Hypoxia/drug therapy , Hypoxia/etiology , Mice , Nitric Oxide/administration & dosage , Nitric Oxide/therapeutic use , Nitric Oxide Synthase Type III/drug effects , SARS-CoV-2 , Tachyphylaxis
SELECTION OF CITATIONS
SEARCH DETAIL