Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem ; 105: 117732, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38643719

ABSTRACT

Virus entry inhibitors are emerging as an attractive class of therapeutics for the suppression of viral transmission. Naturally occurring pradimicin A (PRM-A) has received particular attention as the first-in-class entry inhibitor that targets N-glycans present on viral surface. Despite the uniqueness of its glycan-targeted antiviral activity, there is still limited knowledge regarding how PRM-A binds to viral N-glycans. Therefore, in this study, we performed binding analysis of PRM-A with synthetic oligosaccharides that reflect the structural motifs characteristic of viral N-glycans. Binding assays and molecular modeling collectively suggest that PRM-A preferentially binds to branched oligomannose motifs of N-glycans via simultaneous recognition of two mannose residues at the non-reducing ends. We also demonstrated, for the first time, that PRM-A can effectively inhibit severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in vitro. Significantly, the anti-SARS-CoV-2 effect of PRM-A is attenuated in the presence of the synthetic branched oligomannose, suggesting that the inhibition of SARS-CoV-2 infection is due to the interaction of PRM-A with the branched oligomannose-containing N-glycans. These data provide essential information needed to understand the antiviral mechanism of PRM-A and suggest that PRM-A could serve as a candidate SARS-CoV-2 entry inhibitor targeting N-glycans.


Subject(s)
Antiviral Agents , Polysaccharides , Pradimicins and Benanomicins , SARS-CoV-2 , Virus Internalization , SARS-CoV-2/drug effects , Polysaccharides/chemistry , Polysaccharides/pharmacology , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/chemical synthesis , Humans , Virus Internalization/drug effects , COVID-19 Drug Treatment , COVID-19/virology , Chlorocebus aethiops , Animals , Vero Cells
2.
Jpn J Infect Dis ; 76(2): 162-166, 2023 Mar 24.
Article in English | MEDLINE | ID: mdl-36575023

ABSTRACT

Biosafety level 4 (BSL-4) laboratories are necessary to study microorganisms that are highly pathogenic to humans and have no prevention or therapeutic measures. Currently, most BSL-4 facilities have suit-type laboratories to conduct experiments on highly pathogenic microorganisms. In 2021, the first Japanese suit-type BSL-4 laboratory was constructed at Nagasaki University. Positive pressure protection suit (PPPS) is a primary barrier that protects and isolates laboratory workers from pathogens and the laboratory environment. Here, we developed a novel PPPS originally designed to be used in the Nagasaki BSL-4 laboratory. We modified several parts of a domestic chemical protective suit, including its front face shield, cuff, and air supply hose, for safe handling of microbiological agents. The improved suit, PS-790BSL4-AL, showed resistance to several chemicals, including quaternary ammonium disinfectant, and did not show any permeation against blood and phages. To validate the suit's integrity, we also established an airtight test that eliminated individual differences for quantitative testing. In conclusion, our developed suit performs sufficiently as a primary barrier and allows for the safe handling of pathogens in our new BSL-4 laboratory.


Subject(s)
Containment of Biohazards , Laboratories , Humans , Japan
3.
Trop Med Health ; 50(1): 30, 2022 Apr 27.
Article in English | MEDLINE | ID: mdl-35477500

ABSTRACT

The coronavirus disease 2019 (COVID 19) pandemic continues to pose a threat to global health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant (B.1.1.529) has spread rapidly worldwide and became dominant in many countries. A natural 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC) has demonstrated antiviral activity in Wuhan, Alpha, Beta, Gamma, and Delta variants of SARS-CoV-2 infections in vitro. In this study, we report antiviral activity of 5-ALA, 5-ALA with SFC led to IC50 of 329 and 765/191, respectively after infection with Omicron variant of SARS-CoV-2 in vitro. Our finding suggests that 5-ALA could be used as antiviral drug candidate to treat Omicron variant infected patients.

4.
Antiviral Res ; 200: 105276, 2022 04.
Article in English | MEDLINE | ID: mdl-35278582

ABSTRACT

Crimean-Congo hemorrhagic fever virus (CCHFV) belongs to the genus Orthonairovirus and is the causative agent of a viral hemorrhagic disease with a case fatality rate of 30%. However, limited studies have been conducted to explore antiviral compounds specific to CCHFV. In this study, we developed a minigenome system of orthonairoviruses, CCHFV and Hazara virus to analyze viral replication and screened an FDA-approved compound library. The transfection of the minigenome components induced marked increase in luciferase expression, indicating the sufficient replication and translation of reporter RNA. Compound library screening identified 14 candidate compounds that significantly decreased luciferase activity. Some of the compounds also inhibited the replication of the infectious Hazara virus. The mechanism of inhibition by tigecycline was further analyzed, and a decrease in the interaction between the viral N protein and RNA by tigecycline was observed. This work provides a basis for validation using animal models and the design of chemical derivatives with stronger activity in future studies on the development of an antiviral against CCHFV.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo , Hemorrhagic Fever, Crimean , Animals , Antiviral Agents/pharmacology , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Hemorrhagic Fever, Crimean/drug therapy , Hemorrhagic Fever, Crimean/prevention & control , Nucleoproteins , RNA , Tigecycline/pharmacology
5.
Antiviral Res ; 199: 105267, 2022 03.
Article in English | MEDLINE | ID: mdl-35227759

ABSTRACT

The central role of Ebola virus (EBOV) VP40 in nascent virion assembly and budding from infected host cells makes it an important therapeutic target. The mechanism of dimerization, following oligomerization of VP40 leading to the production of virus-like particles (VLP) has never been investigated for the development of therapeutic candidates against Ebola disease. Molecular dynamics-based computational screening targeted VP40 dimer with 40,000,000 compounds selected 374 compounds. A novel in vitro screening assay selected two compounds, NUSU#1 and NUSU#2. Conventional VLP assays consistently showed that both compounds inhibited EBOV VP40-mediated VLP production. Intriguingly, NUSU#1 inhibited the VP40-mediated VLP production in other ebolavirus species and the Marburg virus, but did not inhibit Lassa virus Z-mediated VLP production. These results strongly suggested that the selected compounds are potential lead drug candidates against Filovirus disease via disruption of VP40-mediated particle production.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Marburgvirus , Ebolavirus/chemistry , Humans , Viral Matrix Proteins/chemistry , Virus Release
6.
Microorganisms ; 10(1)2022 Jan 04.
Article in English | MEDLINE | ID: mdl-35056548

ABSTRACT

In the initial phase of the novel coronavirus disease (COVID-19) pandemic, a large-scale cluster on the cruise ship Diamond Princess (DP) emerged in Japan. Genetic analysis of the DP strains has provided important information for elucidating the possible transmission process of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on a cruise ship. However, genome-based analyses of SARS-CoV-2 detected in large-scale cruise ship clusters other than the DP cluster have rarely been reported. In the present study, whole-genome sequences of 94 SARS-CoV-2 strains detected in the second large cruise ship cluster, which emerged on the Costa Atlantica (CA) in Japan, were characterized to understand the evolution of the virus in a crowded and confined place. Phylogenetic and haplotype network analysis indicated that the CA strains were derived from a common ancestral strain introduced on the CA cruise ship and spread in a superspreading event-like manner, resulting in several mutations that might have affected viral characteristics, including the P681H substitution in the spike protein. Moreover, there were significant genetic distances between CA strains and other strains isolated in different environments, such as cities under lockdown. These results provide new insights into the unique evolution patterns of SARS-CoV-2 in the CA cruise ship cluster.

7.
Viruses ; 14(1)2022 01 13.
Article in English | MEDLINE | ID: mdl-35062347

ABSTRACT

Ebola virus disease (EVD) is a lethal disease caused by the highly pathogenic Ebola virus (EBOV), and its major symptoms in severe cases include vascular leakage and hemorrhage. These symptoms are caused by abnormal activation and disruption of endothelial cells (ECs) whose mediators include EBOV glycoprotein (GP) without the need for viral replication. However, the detailed molecular mechanisms underlying virus-host interactions remain largely unknown. Here, we show that EBOV-like particles (VLPs) formed by GP, VP40, and NP activate ECs in a GP-dependent manner, as demonstrated by the upregulation of intercellular adhesion molecules-1 (ICAM-1) expression. VLPs-mediated ECs activation showed a different kinetic pattern from that of TNF-α-mediated activation and was associated with apoptotic ECs disruption. In contrast to TNF-α, VLPs induced ICAM-1 overexpression at late time points. Furthermore, screening of host cytoskeletal signaling inhibitors revealed that focal adhesion kinase inhibitors were found to be potent inhibitors of ICAM-1 expression mediated by both TNF-α and VLPs. Our results suggest that EBOV GP stimulates ECs to induce endothelial activation and dysfunction with the involvement of host cytoskeletal signaling factors, which represent potential therapeutic targets for EVD.


Subject(s)
Ebolavirus/physiology , Endothelial Cells/metabolism , Glycoproteins/metabolism , Host-Pathogen Interactions/physiology , Signal Transduction , Apoptosis , Cell Survival , Cytoskeleton , HEK293 Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Integration Host Factors , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/metabolism , Kinetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Virus Replication
8.
Trop Med Health ; 50(1): 6, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34991723

ABSTRACT

BACKGROUND: Genetic variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began to emerge in 2020 and have been spreading globally during the coronavirus disease 2019 (COVID-19) pandemic. Despite the presence of different COVID-19 vaccines, the discovery of effective antiviral therapeutics for the treatment of patients infected with SARS-CoV-2 are still urgently needed. A natural amino acid, 5-aminolevulinic acid (5-ALA), has exhibited both antiviral and anti-inflammatory activities. In a previous study, we demonstrated an in vitro antiviral effect of 5-ALA against SARS-CoV-2 infection without significant cytotoxicity. In the present study, we sought to investigate whether 5-ALA with or without sodium ferrous citrate (SFC) can inhibit in vitro both the original SARS-CoV-2 Wuhan strain and its variants, including the Alpha, Beta, Gamma and Delta strains. METHODS: The antiviral activity of ALA with or without SFC was determined in Vero-E6 cell. The virus inhibition was quantified by real time RT-PCR. RESULTS: Co-administration of 5-ALA and SFC inhibited the Wuhan, Alpha and Delta variants of SARS-CoV-2 with IC50 values of 235, 173 and 397 µM, respectively, and the Beta and Gamma variants with IC50 values of 1311 and 1516 µM. CONCLUSION: Our study suggests that 5-ALA with SFC warrants accelerated clinical evaluation as an antiviral drug candidate for treating patients infected with SARS-CoV-2 variants.

9.
Viruses ; 15(1)2022 12 30.
Article in English | MEDLINE | ID: mdl-36680145

ABSTRACT

Although many arenaviruses cause severe diseases with high fatality rates each year, treatment options are limited to off-label use of ribavirin, and a Food and Drug Administration (FDA)-approved vaccine is not available. To identify novel therapeutic candidates against arenaviral diseases, an RNA polymerase I-driven minigenome (MG) expression system for Lassa virus (LASV) was developed and optimized for high-throughput screening (HTS). Using this system, we screened 2595 FDA-approved compounds for inhibitors of LASV genome replication and identified multiple compounds including pixantrone maleate, a topoisomerase II inhibitor, as hits. Other tested topoisomerase II inhibitors also suppressed LASV MG activity. These topoisomerase II inhibitors also inhibited Junin virus (JUNV) MG activity and effectively limited infection by the JUNV Candid #1 strain, and siRNA knockdown of both topoisomerases (IIα and IIß) restricted JUNV replication. These results suggest that topoisomerases II regulate arenavirus replication and can serve as molecular targets for panarenaviral replication inhibitors.


Subject(s)
Arenavirus , Junin virus , Antiviral Agents/pharmacology , DNA Topoisomerases, Type II/genetics , Junin virus/physiology , Lassa virus , Topoisomerase II Inhibitors/pharmacology , Humans
10.
Sci Rep ; 11(1): 21259, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711897

ABSTRACT

Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is currently a serious public health concern worldwide. Notably, co-infection with other pathogens may worsen the severity of COVID-19 symptoms and increase fatality. Here, we show that co-infection with influenza A virus (IAV) causes more severe body weight loss and more severe and prolonged pneumonia in SARS-CoV-2-infected hamsters. Each virus can efficiently spread in the lungs without interference by the other. However, in immunohistochemical analyses, SARS-CoV-2 and IAV were not detected at the same sites in the respiratory organs of co-infected hamsters, suggesting that either the two viruses may have different cell tropisms in vivo or each virus may inhibit the infection and/or growth of the other within a cell or adjacent areas in the organs. Furthermore, a significant increase in IL-6 was detected in the sera of hamsters co-infected with SARS-CoV-2 and IAV at 7 and 10 days post-infection, suggesting that IL-6 may be involved in the increased severity of pneumonia. Our results strongly suggest that IAV co-infection with SARS-CoV-2 can have serious health risks and increased caution should be applied in such cases.


Subject(s)
COVID-19/complications , Orthomyxoviridae Infections/complications , Pneumonia, Viral/complications , SARS-CoV-2 , Animals , COVID-19/pathology , COVID-19/virology , Coinfection/pathology , Disease Models, Animal , Female , Humans , Interleukin-6/blood , Lung/diagnostic imaging , Lung/pathology , Mesocricetus , Orthomyxoviridae/pathogenicity , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2/pathogenicity , SARS-CoV-2/physiology , Severity of Illness Index , Virus Replication
11.
FEBS Open Bio ; 11(5): 1452-1464, 2021 05.
Article in English | MEDLINE | ID: mdl-33822489

ABSTRACT

Human pathogenic RNA viruses are threats to public health because they are prone to escaping the human immune system through mutations of genomic RNA, thereby causing local outbreaks and global pandemics of emerging or re-emerging viral diseases. While specific therapeutics and vaccines are being developed, a broad-spectrum therapeutic agent for RNA viruses would be beneficial for targeting newly emerging and mutated RNA viruses. In this study, we conducted a screen of repurposed drugs using Sendai virus (an RNA virus of the family Paramyxoviridae), with human-induced pluripotent stem cells (iPSCs) to explore existing drugs that may present anti-RNA viral activity. Selected hit compounds were evaluated for their efficacy against two important human pathogens: Ebola virus (EBOV) using Huh7 cells and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using Vero E6 cells. Selective estrogen receptor modulators (SERMs), including raloxifene, exhibited antiviral activities against EBOV and SARS-CoV-2. Pioglitazone, a PPARγ agonist, also exhibited antiviral activities against SARS-CoV-2, and both raloxifene and pioglitazone presented a synergistic antiviral effect. Finally, we demonstrated that SERMs blocked entry steps of SARS-CoV-2 into host cells. These findings suggest that the identified FDA-approved drugs can modulate host cell susceptibility against RNA viruses.


Subject(s)
Antiviral Agents/pharmacology , Drug Repositioning , RNA Viruses/drug effects , RNA, Viral/antagonists & inhibitors , SARS-CoV-2/drug effects , Animals , Cell Line , Chlorocebus aethiops , Drug Repositioning/methods , Ebolavirus/drug effects , Ebolavirus/physiology , Humans , Induced Pluripotent Stem Cells/virology , Microbial Sensitivity Tests/methods , Pioglitazone/pharmacology , RNA Viruses/physiology , Raloxifene Hydrochloride/pharmacology , SARS-CoV-2/physiology , Selective Estrogen Receptor Modulators/pharmacology , Sendai virus/drug effects , Sendai virus/physiology , Vero Cells , COVID-19 Drug Treatment
12.
Biochem Biophys Res Commun ; 545: 203-207, 2021 03 19.
Article in English | MEDLINE | ID: mdl-33571909

ABSTRACT

The current COVID-19 pandemic requires urgent development of effective therapeutics. 5-amino levulinic acid (5-ALA) is a naturally synthesized amino acid and has been used for multiple purposes including as an anticancer therapy and as a dietary supplement due to its high bioavailability. In this study, we demonstrated that 5-ALA treatment potently inhibited infection of SARS-CoV-2, a causative agent of COVID-19, in cell culture. The antiviral effects could be detected in both human and non-human cells, without significant cytotoxicity. Therefore, 5-ALA is worth to be further investigated as an antiviral drug candidate for COVID-19.


Subject(s)
Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Levulinic Acids/pharmacology , Animals , Antiviral Agents/administration & dosage , COVID-19/prevention & control , COVID-19/virology , Caco-2 Cells , Chlorocebus aethiops , Citric Acid , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Ferrous Compounds/pharmacology , Humans , Levulinic Acids/administration & dosage , Vero Cells , Aminolevulinic Acid
13.
Front Immunol ; 12: 803647, 2021.
Article in English | MEDLINE | ID: mdl-35095889

ABSTRACT

The newly emerged severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causing a spread of coronavirus disease 2019 (COVID-19) globally. In order to end the COVID-19 pandemic, an effective vaccine against SARS-CoV-2 must be produced at low cost and disseminated worldwide. The spike (S) protein of coronaviruses plays a pivotal role in the infection to host cells. Therefore, targeting the S protein is one of the most rational approaches in developing vaccines and therapeutic agents. In this study, we optimized the expression of secreted trimerized S protein of SARS-CoV-2 using a silkworm-baculovirus expression vector system and evaluated its immunogenicity in mice. The results showed that the S protein forming the trimeric structure was the most stable when the chicken cartilage matrix protein was used as the trimeric motif and could be purified in large amounts from the serum of silkworm larvae. The purified S protein efficiently induced antigen-specific antibodies in mouse serum without adjuvant, but its ability to induce neutralizing antibodies was low. After examining several adjuvants, the use of Alum adjuvant was the most effective in inducing strong neutralizing antibody induction. We also examined the adjuvant effect of paramylon from Euglena gracilis when administered with the S protein. Our results highlight the effectiveness and suitable construct design of the S protein produced in silkworms for the subunit vaccine development against SARS-CoV-2.


Subject(s)
Alum Compounds/pharmacology , Aluminum Hydroxide/pharmacology , Bombyx/genetics , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19 Vaccines/immunology , Cell Line , Chickens/genetics , Chickens/immunology , Chlorocebus aethiops , Euglena gracilis/immunology , Euglenozoa Infections/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Pandemics/prevention & control , SARS-CoV-2/immunology , Vaccination/methods , Vero Cells
14.
Antiviral Res ; 160: 175-182, 2018 12.
Article in English | MEDLINE | ID: mdl-30395872

ABSTRACT

Ebola virus disease is a severe disease caused by highly pathogenic Ebolaviruses. Although it shows a high mortality rate in humans, currently there is no licensed therapeutic. During the recent epidemic in West Africa, it was demonstrated that administration of antimalarial medication containing amodiaquine significantly lowered mortality rate of patients infected with the virus. Here, in order to improve its antiviral activity, a series of amodiaquine derivatives were synthesized and tested for Ebola virus infection. We found that multiple compounds were more potent than amodiaquine. The structure-activity relationship analysis revealed that the two independent parts, which are the alkyl chains extending from the aminomethyl group and a halogen bonded to the quinoline ring, were keys for enhancing antiviral potency without increasing toxicity. When these modifications were combined, the antiviral efficacy could be further improved with the selectivity indexes being over 10-times higher than amodiaquine. Mechanistic evaluation demonstrated that the potent derivatives blocked host cell entry of Ebola virus, like the parental amodiaquine. Taken together, our work identified novel potent amodiaquine derivatives, which will aid in further development of effective antiviral therapeutics.


Subject(s)
Amodiaquine/chemical synthesis , Amodiaquine/pharmacology , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Ebolavirus/drug effects , Virus Internalization/drug effects , Amodiaquine/toxicity , Antimalarials/chemical synthesis , Antimalarials/pharmacology , Antimalarials/toxicity , Antiviral Agents/toxicity , Structure-Activity Relationship
15.
J Infect Dis ; 218(suppl_5): S612-S626, 2018 11 22.
Article in English | MEDLINE | ID: mdl-29860496

ABSTRACT

Background: For most classes of drugs, rapid development of therapeutics to treat emerging infections is challenged by the timelines needed to identify compounds with the desired efficacy, safety, and pharmacokinetic profiles. Fully human monoclonal antibodies (mAbs) provide an attractive method to overcome many of these hurdles to rapidly produce therapeutics for emerging diseases. Methods: In this study, we deployed a platform to generate, test, and develop fully human antibodies to Zaire ebolavirus. We obtained specific anti-Ebola virus (EBOV) antibodies by immunizing VelocImmune mice that use human immunoglobulin variable regions in their humoral responses. Results: Of the antibody clones isolated, 3 were selected as best at neutralizing EBOV and triggering FcγRIIIa. Binding studies and negative-stain electron microscopy revealed that the 3 selected antibodies bind to non-overlapping epitopes, including a potentially new protective epitope not targeted by other antibody-based treatments. When combined, a single dose of a cocktail of the 3 antibodies protected nonhuman primates (NHPs) from EBOV disease even after disease symptoms were apparent. Conclusions: This antibody cocktail provides complementary mechanisms of actions, incorporates novel specificities, and demonstrates high-level postexposure protection from lethal EBOV disease in NHPs. It is now undergoing testing in normal healthy volunteers in preparation for potential use in future Ebola epidemics.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/therapeutic use , Hemorrhagic Fever, Ebola/drug therapy , Animals , Antibodies, Monoclonal/isolation & purification , Glycoproteins/immunology , Guinea Pigs , HEK293 Cells , Humans , Macaca mulatta , Male , Mice
16.
Antiviral Res ; 149: 154-163, 2018 01.
Article in English | MEDLINE | ID: mdl-29175127

ABSTRACT

Members of the family Filoviridae cause severe, often fatal disease in humans, for which there are no approved vaccines and only a few experimental drugs tested in animal models. Retro-2, a small molecule that inhibits retrograde trafficking of bacterial and plant toxins inside host cells, has been demonstrated to be effective against a range of bacterial and virus pathogens, both in vitro and in animal models. Here, we demonstrated that Retro-2 and its derivatives, Retro-2.1 and compound 25, blocked infection by Ebola virus and Marburg virus in vitro. We show that the derivatives were more potent inhibitors of infection as compared to the parent compound. Pseudotyped virus assays indicated that the compounds affected virus entry into cells while virus particle localization to Niemann-Pick C1-positive compartments showed that they acted at a late step in virus entry. Our work demonstrates a potential for Retro-type drugs to be developed into anti-filoviral therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Filoviridae/drug effects , Thiophenes/pharmacology , Animals , Antiviral Agents/chemistry , Benzamides/chemistry , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Ebolavirus/drug effects , Ebolavirus/physiology , Filoviridae/physiology , Humans , Marburgvirus/drug effects , Marburgvirus/physiology , Microbial Sensitivity Tests , Molecular Structure , Thiophenes/chemistry , Virus Internalization/drug effects
17.
Chem Biol Interact ; 267: 96-103, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-27712998

ABSTRACT

Medical countermeasures to treat biothreat agent infections require broad-spectrum therapeutics that do not induce agent resistance. A cell-based high-throughput screen (HTS) against ricin toxin combined with hit optimization allowed selection of a family of compounds that meet these requirements. The hit compound Retro-2 and its derivatives have been demonstrated to be safe in vivo in mice even at high doses. Moreover, Retro-2 is an inhibitor of retrograde transport that affects syntaxin-5-dependent toxins and pathogens. As a consequence, it has a broad-spectrum activity that has been demonstrated both in vitro and in vivo against ricin, Shiga toxin-producing O104:H4 entero-hemorrhagic E. coli and Leishmania sp. and in vitro against Ebola, Marburg and poxviruses and Chlamydiales. An effect is anticipated on other toxins or pathogens that use retrograde trafficking and syntaxin-5. Since Retro-2 targets cell components of the host and not directly the pathogen, no selection of resistant pathogens is expected. These lead compounds need now to be developed as drugs for human use.


Subject(s)
Benzamides/pharmacology , Chlamydiales/metabolism , Ebolavirus/metabolism , Leishmania/metabolism , Ricin/metabolism , Shiga Toxins/metabolism , Thiophenes/pharmacology , Animals , Benzamides/chemistry , Body Weight/drug effects , Chlamydiales/drug effects , Ebolavirus/drug effects , Escherichia coli/metabolism , HEK293 Cells , HeLa Cells , Humans , Injections, Intraperitoneal , Leishmania/drug effects , Mice , Mice, Inbred BALB C , Mitomycin/pharmacology , Models, Animal , RAW 264.7 Cells , Ricin/antagonists & inhibitors , Shiga Toxins/antagonists & inhibitors , Thiophenes/chemistry
18.
PLoS Pathog ; 11(11): e1005263, 2015.
Article in English | MEDLINE | ID: mdl-26562011

ABSTRACT

Ebola virus outbreaks, such as the 2014 Makona epidemic in West Africa, are episodic and deadly. Filovirus antivirals are currently not clinically available. Our findings suggest interferon gamma, an FDA-approved drug, may serve as a novel and effective prophylactic or treatment option. Using mouse-adapted Ebola virus, we found that murine interferon gamma administered 24 hours before or after infection robustly protects lethally-challenged mice and reduces morbidity and serum viral titers. Furthermore, we demonstrated that interferon gamma profoundly inhibits Ebola virus infection of macrophages, an early cellular target of infection. As early as six hours following in vitro infection, Ebola virus RNA levels in interferon gamma-treated macrophages were lower than in infected, untreated cells. Addition of the protein synthesis inhibitor, cycloheximide, to interferon gamma-treated macrophages did not further reduce viral RNA levels, suggesting that interferon gamma blocks life cycle events that require protein synthesis such as virus replication. Microarray studies with interferon gamma-treated human macrophages identified more than 160 interferon-stimulated genes. Ectopic expression of a select group of these genes inhibited Ebola virus infection. These studies provide new potential avenues for antiviral targeting as these genes that have not previously appreciated to inhibit negative strand RNA viruses and specifically Ebola virus infection. As treatment of interferon gamma robustly protects mice from lethal Ebola virus infection, we propose that interferon gamma should be further evaluated for its efficacy as a prophylactic and/or therapeutic strategy against filoviruses. Use of this FDA-approved drug could rapidly be deployed during future outbreaks.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/drug therapy , Interferon-gamma/pharmacology , Macrophages/drug effects , Animals , Cells, Cultured , Humans , Macrophages/metabolism , Mice, Inbred BALB C , RNA, Viral/genetics , Virus Replication/drug effects
19.
Science ; 347(6225): 995-8, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25722412

ABSTRACT

Ebola virus causes sporadic outbreaks of lethal hemorrhagic fever in humans, but there is no currently approved therapy. Cells take up Ebola virus by macropinocytosis, followed by trafficking through endosomal vesicles. However, few factors controlling endosomal virus movement are known. Here we find that Ebola virus entry into host cells requires the endosomal calcium channels called two-pore channels (TPCs). Disrupting TPC function by gene knockout, small interfering RNAs, or small-molecule inhibitors halted virus trafficking and prevented infection. Tetrandrine, the most potent small molecule that we tested, inhibited infection of human macrophages, the primary target of Ebola virus in vivo, and also showed therapeutic efficacy in mice. Therefore, TPC proteins play a key role in Ebola virus infection and may be effective targets for antiviral therapy.


Subject(s)
Antiviral Agents/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels/physiology , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/therapy , Molecular Targeted Therapy , Virus Internalization/drug effects , Animals , Antiviral Agents/therapeutic use , BALB 3T3 Cells , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , Calcium Channel Blockers/therapeutic use , Calcium Channels/genetics , Ebolavirus/drug effects , Female , Gene Knockout Techniques , HeLa Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Humans , Macrophages/drug effects , Macrophages/virology , Mice , NADP/analogs & derivatives , NADP/metabolism , RNA Interference , Signal Transduction , Verapamil/pharmacology , Verapamil/therapeutic use
20.
Uirusu ; 65(1): 71-82, 2015.
Article in Japanese | MEDLINE | ID: mdl-26923960

ABSTRACT

Ebola virus is an enveloped virus with filamentous structure and causes a severe hemorrhagic fever in human and nonhuman primates. Host cell entry is the first essential step in the viral life cycle, which has been extensively studied as one of the therapeutic targets. A virus factor of cell entry is a surface glycoprotein (GP), which is an only essential viral protein in the step, as well as the unique particle structure. The virus also interacts with a lot of host factors to successfully enter host cells. Ebola virus at first binds to cell surface proteins and internalizes into cells, followed by trafficking through endosomal vesicles to intracellular acidic compartments. There, host proteases process GPs, which can interact with an intracellular receptor. Then, under an appropriate circumstance, viral and endosomal membranes are fused, which is enhanced by major structural changes of GPs, to complete host cell entry. Recently the basic research of Ebola virus infection mechanism has markedly progressed, largely contributed by identification of host factors and detailed structural analyses of GPs. This article highlights the mechanism of Ebola virus host cell entry, including recent findings.


Subject(s)
Cells/virology , Ebolavirus/physiology , Ebolavirus/pathogenicity , Host-Pathogen Interactions , Membrane Glycoproteins/physiology , Viral Proteins/physiology , Virus Internalization , Animals , Cell Physiological Phenomena , Ebolavirus/growth & development , Humans , Hydrogen-Ion Concentration , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Multivesicular Bodies/physiology , Multivesicular Bodies/virology , Peptide Hydrolases/physiology , Receptors, Virus/physiology , Viral Proteins/chemistry , Viral Proteins/metabolism , Virus Attachment , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...