Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
J Med Chem ; 66(4): 2744-2760, 2023 02 23.
Article in English | MEDLINE | ID: covidwho-2242001

ABSTRACT

Enveloped viruses depend on the host endoplasmic reticulum (ER) quality control (QC) machinery for proper glycoprotein folding. The endoplasmic reticulum quality control (ERQC) enzyme α-glucosidase I (α-GluI) is an attractive target for developing broad-spectrum antivirals. We synthesized 28 inhibitors designed to interact with all four subsites of the α-GluI active site. These inhibitors are derivatives of the iminosugars 1-deoxynojirimycin (1-DNJ) and valiolamine. Crystal structures of ER α-GluI bound to 25 1-DNJ and three valiolamine derivatives revealed the basis for inhibitory potency. We established the structure-activity relationship (SAR) and used the Site Identification by Ligand Competitive Saturation (SILCS) method to develop a model for predicting α-GluI inhibition. We screened the compounds against SARS-CoV-2 in vitro to identify those with greater antiviral activity than the benchmark α-glucosidase inhibitor UV-4. These host-targeting compounds are candidates for investigation in animal models of SARS-CoV-2 and for testing against other viruses that rely on ERQC for correct glycoprotein folding.


Subject(s)
1-Deoxynojirimycin , Antiviral Agents , COVID-19 , Glycoside Hydrolase Inhibitors , alpha-Glucosidases , Animals , 1-Deoxynojirimycin/chemistry , 1-Deoxynojirimycin/pharmacology , alpha-Glucosidases/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Endoplasmic Reticulum/enzymology , Glycoproteins , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , SARS-CoV-2/metabolism , Quantitative Structure-Activity Relationship
2.
J Med Chem ; 64(24): 18010-18024, 2021 12 23.
Article in English | MEDLINE | ID: covidwho-1616926

ABSTRACT

Most enveloped viruses rely on the host cell endoplasmic reticulum (ER) quality control (QC) machinery for proper folding of glycoproteins. The key ER α-glucosidases (α-Glu) I and II of the ERQC machinery are attractive targets for developing broad-spectrum antivirals. Iminosugars based on deoxynojirimycin have been extensively studied as ER α-glucosidase inhibitors; however, other glycomimetic compounds are less established. Accordingly, we synthesized a series of N-substituted derivatives of valiolamine, the iminosugar scaffold of type 2 diabetes drug voglibose. To understand the basis for up to 100,000-fold improved inhibitory potency, we determined high-resolution crystal structures of mouse ER α-GluII in complex with valiolamine and 10 derivatives. The structures revealed extensive interactions with all four α-GluII subsites. We further showed that N-substituted valiolamines were active against dengue virus and SARS-CoV-2 in vitro. This study introduces valiolamine-based inhibitors of the ERQC machinery as candidates for developing potential broad-spectrum therapeutics against the existing and emerging viruses.


Subject(s)
Antiviral Agents/pharmacology , Glycoside Hydrolase Inhibitors/pharmacology , Imino Sugars/pharmacology , Inositol/analogs & derivatives , alpha-Glucosidases/metabolism , Animals , Antiviral Agents/chemical synthesis , Antiviral Agents/metabolism , Binding Sites , Chlorocebus aethiops , Crystallography, X-Ray , Dengue Virus/drug effects , Endoplasmic Reticulum/enzymology , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/metabolism , Humans , Imino Sugars/chemical synthesis , Imino Sugars/metabolism , Inositol/chemical synthesis , Inositol/metabolism , Inositol/pharmacology , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Protein Binding , SARS-CoV-2/drug effects , Vero Cells , alpha-Glucosidases/chemistry
3.
Molecules ; 26(10)2021 May 20.
Article in English | MEDLINE | ID: covidwho-1248002

ABSTRACT

Diabetes mellitus (DM) is a chronic disorder and has affected a large number of people worldwide. Insufficient insulin production causes an increase in blood glucose level that results in DM. To lower the blood glucose level, various drugs are employed that block the activity of the α-glucosidase enzyme, which is considered responsible for the breakdown of polysaccharides into monosaccharides leading to an increase in the intestinal blood glucose level. We have synthesized novel 2-(3-(benzoyl/4-bromobenzoyl)-4-hydroxy-1,1-dioxido-2H-benzo[e][1,2]thiazin-2-yl)-N-arylacetamides and have screened them for their in silico and in vitro α-glucosidase inhibition activity. The derivatives 11c, 12a, 12d, 12e, and 12g emerged as potent inhibitors of the α-glucosidase enzyme. These compounds exhibited good docking scores and excellent binding interactions with the selected residues (Asp203, Asp542, Asp327, His600, Arg526) during in silico screening. Similarly, these compounds also showed good in vitro α-glucosidase inhibitions with IC50 values of 30.65, 18.25, 20.76, 35.14, and 24.24 µM, respectively, which were better than the standard drug, acarbose (IC50 = 58.8 µM). Furthermore, a good agreement was observed between in silico and in vitro modes of study.


Subject(s)
Acetamides/chemical synthesis , Acetamides/pharmacology , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/pharmacology , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacology , Thiazines/chemistry , Thiazines/pharmacology , Acetamides/chemistry , Acetamides/therapeutic use , Computer Simulation , Diabetes Mellitus/drug therapy , Drug Evaluation, Preclinical , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/therapeutic use , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/therapeutic use , Inhibitory Concentration 50 , Molecular Docking Simulation , Structure-Activity Relationship , Thiazines/chemical synthesis
4.
Viruses ; 13(5)2021 04 30.
Article in English | MEDLINE | ID: covidwho-1217120

ABSTRACT

Repurposing clinically available drugs to treat the new coronavirus disease 2019 (COVID-19) is an urgent need in the course of the Severe Acute Respiratory Syndrome coronavirus (SARS-CoV-2) pandemic, as very few treatment options are available. The iminosugar Miglustat is a well-characterized drug for the treatment of rare genetic lysosome storage diseases, such as Gaucher and Niemann-Pick type C, and has also been described to be active against a variety of enveloped viruses. The activity of Miglustat is here demonstrated in the micromolar range for SARS-CoV-2 in vitro. The drug acts at the post-entry level and leads to a marked decrease of viral proteins and release of infectious viruses. The mechanism resides in the inhibitory activity toward α-glucosidases that are involved in the early stages of glycoprotein N-linked oligosaccharide processing in the endoplasmic reticulum, leading to a marked decrease of the viral Spike protein. Indeed, the antiviral potential of protein glycosylation inhibitors against SARS-CoV-2 is further highlighted by the low-micromolar activity of the investigational drug Celgosivir. These data point to a relevant role of this approach for the treatment of COVID-19.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Antiviral Agents/pharmacology , Drug Repositioning , Glycoside Hydrolase Inhibitors/pharmacology , Indolizines/pharmacology , SARS-CoV-2/drug effects , 1-Deoxynojirimycin/pharmacology , A549 Cells , Animals , Chlorocebus aethiops , Glycosylation/drug effects , HEK293 Cells , Humans , Spike Glycoprotein, Coronavirus/metabolism , Vero Cells , Virus Release/drug effects , COVID-19 Drug Treatment
5.
Glycobiology ; 31(4): 378-384, 2021 05 03.
Article in English | MEDLINE | ID: covidwho-799105

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic poses an unprecedented challenge for health care and the global economy. Repurposing drugs that have shown promise in inhibiting other viral infections could allow for more rapid dispensation of urgently needed therapeutics. The Spike protein of SARS-CoV-2 is extensively glycosylated with 22 occupied N glycan sites and is required for viral entry. In other glycosylated viral proteins, glycosylation is required for interaction with calnexin and chaperone-mediated folding in the endoplasmic reticulum, and prevention of this interaction leads to unfolded viral proteins and thus inhibits viral replication. As such, we investigated two iminosugars, celgosivir, a prodrug of castanospermine, and UV-4, or N-(9-methoxynonyl)-1-deoxynojirimycin, a deoxynojirimycin derivative. Iminosugars are known inhibitors of the α-glucosidase I and II enzymes and were effective at inhibiting authentic SARS-CoV-2 viral replication in a cell culture system. Celgosivir prevented SARS-CoV-2-induced cell death and reduced viral replication and Spike protein levels in a dose-dependent manner in culture with Vero E6 cells. Castanospermine, the active form of celgosivir, was also able to inhibit SARS-CoV-2, confirming the canonical castanospermine mechanism of action of celgosivir. The monocyclic UV-4 also prevented SARS-CoV-2-induced death and reduced viral replication after 24 h of treatment, although the reduction in viral copies was lost after 48 h. Our findings suggest that iminosugars should be urgently investigated as potential SARS-CoV-2 inhibitors.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , COVID-19 Drug Treatment , Indolizines/pharmacology , SARS-CoV-2/drug effects , Virus Replication/drug effects , 1-Deoxynojirimycin/pharmacology , Animals , COVID-19/virology , Chlorocebus aethiops , Glycoside Hydrolase Inhibitors/pharmacology , Humans , Vero Cells
6.
Biochem Soc Trans ; 48(3): 1287-1295, 2020 Jun 30.
Article in English | MEDLINE | ID: covidwho-592506

ABSTRACT

The ongoing COVID-19 pandemic, caused by SARS-CoV-2, has pushed the health systems of many countries to breaking point and precipitated social distancing measures that have crippled economic activities across the globe. A return to normality is unlikely until effective therapeutics and a vaccine are available. The immediacy of this problem suggests that drug strategies should focus on repurposing approved drugs or late-stage clinical candidates, as these have the shortest path to use in the clinic. Here, we review and discuss the role of host cell N-glycosylation pathways to virus replication and the drugs available to disrupt these pathways. In particular, we make a case for evaluation of the well-tolerated drugs miglitol, celgosivir and especially miglustat for the treatment of COVID-19.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/chemistry , Coronavirus Infections/metabolism , Drug Repositioning/methods , Glycoside Hydrolase Inhibitors/pharmacology , Pneumonia, Viral/metabolism , Antiviral Agents/therapeutic use , COVID-19 , Calnexin/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/virology , Glycoside Hydrolase Inhibitors/therapeutic use , Glycosylation/drug effects , Host-Pathogen Interactions , Humans , Pandemics , Pneumonia, Viral/drug therapy , Pneumonia, Viral/virology , Protein Folding/drug effects , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Virus Replication/drug effects , alpha-Glucosidases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL