Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
PLoS One ; 16(6): e0253489, 2021.
Article in English | MEDLINE | ID: mdl-34138966

ABSTRACT

In the pursuit of suitable and effective solutions to SARS-CoV-2 infection, we investigated the efficacy of several phenolic compounds in controlling key cellular mechanisms involved in its infectivity. The way the SARS-CoV-2 virus infects the cell is a complex process and comprises four main stages: attachment to the cognate receptor, cellular entry, replication and cellular egress. Since, this is a multi-part process, it creates many opportunities to develop effective interventions. Targeting binding of the virus to the host receptor in order to prevent its entry has been of particular interest. Here, we provide experimental evidence that, among 56 tested polyphenols, including plant extracts, brazilin, theaflavin-3,3'-digallate, and curcumin displayed the highest binding with the receptor-binding domain of spike protein, inhibiting viral attachment to the human angiotensin-converting enzyme 2 receptor, and thus cellular entry of pseudo-typed SARS-CoV-2 virions. Both, theaflavin-3,3'-digallate at 25 µg/ml and curcumin above 10 µg/ml concentration, showed binding with the angiotensin-converting enzyme 2 receptor reducing at the same time its activity in both cell-free and cell-based assays. Our study also demonstrates that brazilin and theaflavin-3,3'-digallate, and to a still greater extent, curcumin, decrease the activity of transmembrane serine protease 2 both in cell-free and cell-based assays. Similar pattern was observed with cathepsin L, although only theaflavin-3,3'-digallate showed a modest diminution of cathepsin L expression at protein level. Finally, each of these three compounds moderately increased endosomal/lysosomal pH. In conclusion, this study demonstrates pleiotropic anti-SARS-CoV-2 efficacy of specific polyphenols and their prospects for further scientific and clinical investigations.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/prevention & control , Polyphenols/pharmacology , SARS-CoV-2/drug effects , Spike Glycoprotein, Coronavirus/metabolism , Virus Internalization/drug effects , A549 Cells , Benzopyrans/pharmacology , Biflavonoids/pharmacology , COVID-19/virology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Survival/drug effects , Curcumin/pharmacology , Humans , Protein Binding/drug effects , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Virion/drug effects , Virion/metabolism , Virion/physiology , Virus Attachment/drug effects
2.
Biochem Biophys Res Commun ; 522(4): 862-868, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31806372

ABSTRACT

Ebola virus (EBOV), pathogen of Ebola hemorrhagic fever (EHF), is an enveloped filamental RNA virus. Recently, the EHF crisis occurred in the Democratic Republic of the Congo again highlights the urgency for its clinical treatments. However, no Food and Drug Administration (FDA)-approved therapeutics are currently available. Drug repurposing screening is a time- and cost-effective approach for identifying anti-EBOV therapeutics. Here, by combinatorial screening using pseudovirion and minigenome replicon systems we have identified several FDA-approved drugs with significant anti-EBOV activities. These potential candidates include azithromycin, clomiphene, chloroquine, digitoxin, epigallocatechin-gallate, fluvastatin, tetrandrine and tamoxifen. Mechanistic studies revealed that fluvastatin inhibited EBOV pseudovirion entry by blocking the pathway of mevalonate biosynthesis, while the inhibitory effect of azithromycin on EBOV maybe due to its intrinsic cationic amphiphilic structure altering the homeostasis of later endosomal vesicle similar as tamoxifen. Moreover, based on structure and pathway analyses, the anti-EBOV activity has been extended to other family members of statins, such as simvastatin, and multiple other cardiac glycoside drugs, some of which exhibited even stronger activities. More importantly, in searching for drug interaction, we found various synergy between several anti-EBOV drug combinations, showing substantial and powerful synergistic against EBOV infection. In conclusion, our work illustrates a successful and productive approach to identify new mechanisms and targets for treating EBOV infection by combinatorial screening of FDA-approved drugs.


Subject(s)
Antiviral Agents/analysis , Antiviral Agents/pharmacology , Combinatorial Chemistry Techniques , Drug Approval , Drug Evaluation, Preclinical , Ebolavirus/drug effects , Azithromycin/pharmacology , Cardiac Glycosides/pharmacology , Cell Line , Cholesterol/biosynthesis , Drug Synergism , Ebolavirus/physiology , Fluvastatin/pharmacology , Humans , Mevalonic Acid/metabolism , Models, Biological , Surface-Active Agents/chemistry , Virion/drug effects , Virion/physiology , Virus Internalization/drug effects , Virus Replication/drug effects
3.
Arch Virol ; 163(6): 1649-1655, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29429035

ABSTRACT

Dengue virus (DENV) infection is a major public health problem worldwide; however, specific antiviral drugs against it are not available. Hence, identifying effective antiviral agents for the prevention of DENV infection is important. In this study, we showed that the reportedly highly biologically active green-tea component epigallocatechin gallate (EGCG) inhibited dengue virus infection regardless of infecting serotype, but no or minimal inhibition was observed with other flaviviruses, including Japanese encephalitis virus, yellow fever virus, and Zika virus. EGCG exerted its antiviral effect mainly at the early stage of infection, probably by interacting directly with virions to prevent virus infection. Our results suggest that EGCG specifically targets DENV and might be used as a lead structure to develop an antiviral drug for use against the virus.


Subject(s)
Antiviral Agents/pharmacology , Catechin/analogs & derivatives , Dengue Virus/drug effects , Tea/chemistry , Virion/drug effects , Antiviral Agents/isolation & purification , Catechin/isolation & purification , Catechin/pharmacology , Dengue Virus/physiology , Dose-Response Relationship, Drug , Encephalitis Virus, Japanese/drug effects , Encephalitis Virus, Japanese/physiology , Species Specificity , Virion/physiology , Virus Internalization/drug effects , Yellow fever virus/drug effects , Yellow fever virus/physiology , Zika Virus/drug effects , Zika Virus/physiology
4.
Proc Natl Acad Sci U S A ; 114(46): 12267-12272, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29087346

ABSTRACT

The transmission of viral infections between plant and fungal hosts has been suspected to occur, based on phylogenetic and other findings, but has not been directly observed in nature. Here, we report the discovery of a natural infection of the phytopathogenic fungus Rhizoctonia solani by a plant virus, cucumber mosaic virus (CMV). The CMV-infected R. solani strain was obtained from a potato plant growing in Inner Mongolia Province of China, and CMV infection was stable when this fungal strain was cultured in the laboratory. CMV was horizontally transmitted through hyphal anastomosis but not vertically through basidiospores. By inoculation via protoplast transfection with virions, a reference isolate of CMV replicated in R. solani and another phytopathogenic fungus, suggesting that some fungi can serve as alternative hosts to CMV. Importantly, in fungal inoculation experiments under laboratory conditions, R. solani could acquire CMV from an infected plant, as well as transmit the virus to an uninfected plant. This study presents evidence of the transfer of a virus between plant and fungus, and it further expands our understanding of plant-fungus interactions and the spread of plant viruses.


Subject(s)
Cucumovirus/physiology , Plant Diseases/virology , Rhizoctonia/virology , Solanum tuberosum/virology , Cucumovirus/pathogenicity , Hyphae/virology , Plant Diseases/microbiology , Protoplasts/microbiology , Protoplasts/virology , Solanum tuberosum/microbiology , Virion/pathogenicity , Virion/physiology
5.
PLoS One ; 11(5): e0155003, 2016.
Article in English | MEDLINE | ID: mdl-27171169

ABSTRACT

There is increasing evidence to suggest that the sinus microbiome plays a role in the pathogenesis of chronic rhinosinusitis (CRS). However, the concentration of these microorganisms within the sinuses is still unknown. We show that flow cytometry can be used to enumerate bacteria and virus-like particles (VLPs) in sinus flush samples of CRS patients. This was achieved through trialling 5 sample preparation techniques for flow cytometry. We found high concentrations of bacteria and VLPs in these samples. Untreated samples produced the highest average bacterial and VLP counts with 3.3 ± 0.74 x 10(7) bacteria ml(-1) and 2.4 ± 1.23 x 10(9) VLP ml(-1) of sinus flush (n = 9). These counts were significantly higher than most of the treated samples (p < 0.05). Results showed 10(3) and 10(4) times inter-patient variation for bacteria and VLP concentrations. This wide variation suggests that diagnosis and treatment need to be personalised and that utilising flow cytometry is useful and efficient for this. This study is the first to enumerate bacterial and VLP populations in the maxillary sinus of CRS patients. The relevance of enumeration is that with increasing antimicrobial resistance, antibiotics are becoming less effective at treating bacterial infections of the sinuses, so alternative therapies are needed. Phage therapy has been proposed as one such alternative, but for dosing, the abundance of bacteria is required. Knowledge of whether phages are normally present in the sinuses will assist in gauging the safety of applying phage therapy to sinuses. Our finding, that large numbers of VLP are frequently present in sinuses, indicates that phage therapy may represent a minimally disruptive intervention towards the nasal microbiome. We propose that flow cytometry can be used as a tool to assess microbial biomass dynamics in sinuses and other anatomical locations where infection can cause disease.


Subject(s)
Bacteria/growth & development , Flow Cytometry/methods , Paranasal Sinuses/microbiology , Rhinitis/microbiology , Rhinitis/virology , Sinusitis/microbiology , Sinusitis/virology , Virion/physiology , Body Fluids , Chronic Disease , Fluorescence , Humans , Paranasal Sinuses/virology
6.
Sci Rep ; 6: 19035, 2016 Jan 07.
Article in English | MEDLINE | ID: mdl-26738439

ABSTRACT

APOBEC1 (A1) proteins from lagomorphs and rodents have deaminase-dependent restriction activity against HIV-1, whereas human A1 exerts a negligible effect. To investigate these differences in the restriction of HIV-1 by A1 proteins, a series of chimeric proteins combining rabbit and human A1s was constructed. Homology models of the A1s indicated that their activities derive from functional domains that likely act in tandem through a dimeric interface. The C-terminal region containing the leucine-rich motif and the dimerization domains of rabbit A1 is important for its anti-HIV-1 activity. The A1 chimeras with strong anti-HIV-1 activity were incorporated into virions more efficiently than those without anti-HIV-1 activity, and exhibited potent DNA-mutator activity. Therefore, the C-terminal region of rabbit A1 is involved in both its packaging into the HIV-1 virion and its deamination activity against both viral cDNA and genomic RNA. This study identifies the novel molecular mechanism underlying the target specificity of A1.


Subject(s)
APOBEC-1 Deaminase/pharmacology , Anti-HIV Agents/pharmacology , HIV-1/physiology , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Animals , Drug Evaluation, Preclinical , Escherichia coli/genetics , Genome, Bacterial , Genome, Viral , HEK293 Cells , HIV-1/drug effects , Humans , Mutagens/pharmacology , Mutation , Protein Multimerization , Rabbits , Sequence Homology, Amino Acid , Virion/drug effects , Virion/physiology , Virus Assembly
7.
Hum Vaccin Immunother ; 12(4): 1009-26, 2016 04 02.
Article in English | MEDLINE | ID: mdl-26618392

ABSTRACT

Millions of seasonal and pandemic influenza vaccine doses containing oil-in-water emulsion adjuvant have been administered in order to enhance and broaden immune responses and to facilitate antigen sparing. Despite the enactment of a Global Action Plan for Influenza Vaccines and a multi-fold increase in production capabilities over the past 10 years, worldwide capacity for pandemic influenza vaccine production is still limited. In developing countries, where routine influenza vaccination is not fully established, additional measures are needed to ensure adequate supply of pandemic influenza vaccines without dependence on the shipment of aid from other, potentially impacted first-world countries. Adaptation of influenza vaccine and adjuvant technologies by developing country influenza vaccine manufacturers may enable antigen sparing and corresponding increases in global influenza vaccine coverage capacity. Following on previously described work involving the technology transfer of oil-in-water emulsion adjuvant manufacturing to a Romanian vaccine manufacturing institute, we herein describe the preclinical evaluation of inactivated split virion H5N1 influenza vaccine with emulsion adjuvant, including immunogenicity, protection from virus challenge, antigen sparing capacity, and safety. In parallel with the evaluation of the bioactivity of the tech-transferred adjuvant, we also describe the impact of concurrent antigen manufacturing optimization activities. Depending on the vaccine antigen source and manufacturing process, inclusion of adjuvant was shown to enhance and broaden functional antibody titers in mouse and rabbit models, promote protection from homologous virus challenge in ferrets, and facilitate antigen sparing. Besides scientific findings, the operational lessons learned are delineated in order to facilitate adaptation of adjuvant technologies by other developing country institutes to enhance global pandemic influenza preparedness.


Subject(s)
Adjuvants, Immunologic , Influenza A Virus, H5N1 Subtype/immunology , Influenza Vaccines , Technology Transfer , Drug Evaluation, Preclinical , Emulsions/chemistry , Humans , Influenza A Virus, H5N1 Subtype/physiology , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Oils , Pandemics/prevention & control , Romania , Virion/physiology , Virus Inactivation
8.
PLoS Negl Trop Dis ; 9(12): e0004259, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26625182

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) is an often lethal, acute inflammatory illness that affects a large geographic area. The disease is caused by infection with CCHF virus (CCHFV), a nairovirus from the Bunyaviridae family. Basic research on CCHFV has been severely hampered by biosafety requirements and lack of available strains and molecular tools. We report the development of a CCHF transcription- and entry-competent virus-like particle (tecVLP) system that can be used to study cell entry and viral transcription/replication over a broad dynamic range (~4 orders of magnitude). The tecVLPs are morphologically similar to authentic CCHFV. Incubation of immortalized and primary human cells with tecVLPs results in a strong reporter signal that is sensitive to treatment with neutralizing monoclonal antibodies and by small molecule inhibitors of CCHFV. We used glycoproteins and minigenomes from divergent CCHFV strains to generate tecVLPs, and in doing so, we identified a monoclonal antibody that can prevent cell entry of tecVLPs containing glycoproteins from 3 pathogenic CCHFV strains. In addition, our data suggest that different glycoprotein moieties confer different cellular entry efficiencies, and that glycoproteins from the commonly used strain IbAr10200 have up to 100-fold lower ability to enter primary human cells compared to glycoproteins from pathogenic CCHFV strains.


Subject(s)
Drug Evaluation, Preclinical/methods , Genes, Reporter , Hemorrhagic Fever Virus, Crimean-Congo/physiology , Transcription, Genetic/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Hemorrhagic Fever Virus, Crimean-Congo/genetics , Hemorrhagic Fever Virus, Crimean-Congo/isolation & purification , Molecular Sequence Data , Sequence Analysis, DNA , Virion/genetics , Virion/physiology , Virion/ultrastructure
9.
PLoS One ; 10(2): e0115475, 2015.
Article in English | MEDLINE | ID: mdl-25643242

ABSTRACT

Herpes simplex virus (HSV), a common latent virus in humans, causes certain severe diseases. Extensive use of acyclovir (ACV) results in the development of drug-resistant HSV strains, hence, there is an urgent need to develop new drugs to treat HSV infection. Houttuynia cordata (H. cordata), a natural herbal medicine, has been reported to exhibit anti-HSV effects which is partly NF-κB-dependent. However, the molecular mechanisms by which H. cordata inhibits HSV infection are not elucidated thoroughly. Here, we report that H. cordata water extracts (HCWEs) inhibit the infection of HSV-1, HSV-2, and acyclovir-resistant HSV-1 mainly via blocking viral binding and penetration in the beginning of infection. HCWEs also suppress HSV replication. Furthermore, HCWEs attenuate the first-wave of NF-κB activation, which is essential for viral gene expressions. Further analysis of six compounds in HCWEs revealed that quercetin and isoquercitrin inhibit NF-κB activation and additionally, quercetin also has an inhibitory effect on viral entry. These results indicate that HCWEs can inhibit HSV infection through multiple mechanisms and could be a potential lead for development of new drugs for treating HSV.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 1, Human/drug effects , Herpesvirus 1, Human/physiology , Herpesvirus 2, Human/drug effects , Herpesvirus 2, Human/physiology , Houttuynia/chemistry , Plant Extracts/pharmacology , Acyclovir/pharmacology , Animals , Antiviral Agents/isolation & purification , Cell Line , Drug Resistance, Viral/drug effects , Gene Expression Regulation, Viral/drug effects , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/metabolism , Herpesvirus 2, Human/genetics , Herpesvirus 2, Human/metabolism , Hot Temperature , Humans , NF-kappa B/metabolism , Plant Extracts/isolation & purification , Viral Envelope Proteins/metabolism , Virion/drug effects , Virion/physiology , Virus Internalization/drug effects , Virus Replication/drug effects , Water/chemistry
10.
Cell Microbiol ; 15(2): 190-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23168015

ABSTRACT

Enveloped viruses acquire their membrane from the host by budding at, or wrapping by, cellular membranes. Transmission electron microscopy (TEM) images, however, suggested that the prototype member of the poxviridae, vaccinia virus (VACV), may create its membrane 'de novo' with free open ends exposed in the cytosol. Within the frame of the German-wide priority programme we re-addressed the biogenesis and origin of the VACV membrane using electron tomography (ET), cryo-EM and lipid analysis of purified VACV using mass spectrometry (MS). This review discussed how our data led to a model of unconventional membrane biogenesis involving membrane rupture and the generation of a single open membrane from open membrane intermediates. Lipid analyses of purified virus by MS suggest an ER origin with a relatively low cholesterol content compared with whole cells, confirming published data. Unlike previous reports using thin-layer chromatography, no depletion of phosphatidylethanolamine was detected. We did detect, however, an enrichment for phosphatidic acid, diacylglycerol and phosphatidylinositol in the virion. Our data are discussed in the light of other pathogens that may requirecellular membrane rupture during their intracellular life cycle.


Subject(s)
Cell Membrane Structures/chemistry , Endoplasmic Reticulum/chemistry , Vaccinia virus/chemistry , Virion/chemistry , Cell Membrane Structures/ultrastructure , Cholesterol/analysis , Cryoelectron Microscopy , Diglycerides/analysis , Electron Microscope Tomography , HeLa Cells , Humans , Mass Spectrometry , Phosphatidic Acids/analysis , Phosphatidylethanolamines/analysis , Phosphatidylinositols/analysis , Vaccinia virus/physiology , Vaccinia virus/ultrastructure , Virion/physiology , Virion/ultrastructure
11.
Antivir Chem Chemother ; 19(6): 243-55, 2009.
Article in English | MEDLINE | ID: mdl-19641233

ABSTRACT

BACKGROUND: The development of antiviral drugs has provided crucial new means to mitigate or relieve the debilitating effects of many viral pathogens. Regular use of these drugs has led to generation of resistant strains, making the control of many viral infections very difficult, particularly in HIV-seropositive and AIDS patients. A rich source for the discovery of new HIV infection inhibitors has been, and continues to be, the 'mining' of the large diversity of compounds already available in nature, and specifically those from botanical extracts. METHODS: Using a newly developed direct binding assay with mass spectrometry technology (direct analysis in real-time time-of-flight mass spectrometry), we were able to show that compounds present in extracts of elderberry, cinnamon and green tea bind to and block HIV type-1 (HIV-1) infection in target cells. RESULTS: The compounds that blocked HIV-1 infection were flavonoids and A-type proanthocyanidins. The 50% inhibitory concentration values of these extracts ranged from 0.5 to 201 microg/ml for four different HIV-1 serotypes. Interaction matrices with the elderberry extract and enfuvirtide, a peptide HIV-1 fusion inhibitor, revealed significant super additive effects. This indicates that the compounds in elderberry that prevent HIV-1 infection are likely to bind to viral glycoproteins other than gp41 (the binding site for enfuvirtide). CONCLUSIONS: Optimized elderberry, green tea and cinnamon extracts rich in certain flavonoid compounds were shown to block HIV-1 entry and infection in GHOST cells. As such, these types of botanical extracts could provide a starting point for the development of possible safe and reliable cotherapies for HIV-1-positive individuals, as well as for the identification of new small molecules as leading drug candidates for HIV-1 therapeutics and microbicides.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , HIV-1/physiology , Plant Extracts/pharmacology , Virus Internalization/drug effects , Anti-HIV Agents/chemistry , Anti-HIV Agents/metabolism , Anti-HIV Agents/toxicity , Binding Sites , Camellia sinensis/chemistry , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Line, Tumor , Cinnamomum zeylanicum/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Synergism , Enfuvirtide , HIV Envelope Protein gp41/pharmacology , HIV Infections/prevention & control , HIV-1/metabolism , Humans , Mass Spectrometry , Peptide Fragments/pharmacology , Plant Extracts/chemistry , Plant Extracts/metabolism , Plant Extracts/toxicity , Reproducibility of Results , Sambucus/chemistry , Time Factors , Virion/drug effects , Virion/metabolism , Virion/physiology
12.
Pharmacol Ther ; 124(1): 1-22, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19555718

ABSTRACT

Investigations on the biology of hepatitis C virus (HCV) have been hampered by the lack of small animal models. Efforts have therefore been directed to designing practical and robust cellular models of human origin able to support HCV replication and production in a reproducible, reliable and consistent manner. Many different models based on different forms of virions and hepatoma or other cell types have been described including virus-like particles, pseudotyped particles, subgenomic and full length replicons, virion productive replicons, immortalised hepatocytes, fetal and adult primary human hepatocytes. This review focuses on these different cellular models, their advantages and disadvantages at the biological and experimental levels, and their respective use for evaluating the effect of antiviral molecules on different steps of HCV biology including virus entry, replication, particles generation and excretion, as well as on the modulation by the virus of the host cell response to infection.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Hepacivirus/drug effects , Cells, Cultured , Drug Design , Endocytosis , Hepacivirus/genetics , Hepacivirus/immunology , Hepacivirus/physiology , Hepatocytes/virology , Humans , RNA Interference , RNA, Viral/biosynthesis , Viral Hepatitis Vaccines/immunology , Virion/physiology , Virus Replication
13.
Biol Pharm Bull ; 31(11): 2018-23, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18981566

ABSTRACT

Andrographis paniculata NEES is a medicinal plant that is commonly used in Asia. This work demonstrates that 25 microg/ml of ethanolic extract from A. paniculata (EEAP) and 5 microg/ml of andrographolide, a bioactive compound in EEAP, effectively inhibit the expression of Epstein-Barr virus (EBV) lytic proteins, Rta, Zta and EA-D, during the viral lytic cycle in P3HR1 cells. Transient transfection analysis revealed that the lack of expression of Rta, Zta and EA-D is caused by the inhibition of the transcription of BRLF1 and BZLF1, two EBV immediate-early genes that encode Rta and Zta, respectively. This study finds that the inhibition prevents the virus from producing mature viral particles. Meanwhile, andrographolide is not toxic to P3HR1 cells when the concentration is below 5 microg/ml, indicating that the compound is potentially useful as an anti-EBV drug.


Subject(s)
Antiviral Agents/pharmacology , Diterpenes/pharmacology , Herpesvirus 4, Human/drug effects , Viral Proteins , Virion/drug effects , Andrographis/chemistry , Antiviral Agents/isolation & purification , Cell Line, Tumor , Cell Survival/drug effects , Diterpenes/isolation & purification , Flow Cytometry , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/physiology , Humans , Plant Components, Aerial/chemistry , Transcription, Genetic/drug effects , Viral Proteins/biosynthesis , Viral Proteins/genetics , Virion/genetics , Virion/physiology , Virus Latency/drug effects , Virus Replication/drug effects
14.
J Leukoc Biol ; 79(6): 1166-72, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16574767

ABSTRACT

Acute and chronic alcohol abuse impairs various functions of the immune system and thus, has been implicated as a cofactor in the immunopathogenesis of human immunodeficiency virus (HIV) disease progression. We determined whether naltrexone, an opioid receptor antagonist widely used in the treatment of alcoholism, inhibits alcohol-mediated enhancement of HIV infection of T cells. Alcohol enhanced HIV infection of peripheral blood lymphocytes (PBL) and a human lymphoid cell line (CEMX174). Alcohol increased HIV X4 envelope (Env), not murine leukemia virus Env-pseudotyped infection of CEMX174 cells. Naltrexone antagonized the enhancing effect of alcohol on HIV infection of PBL and CEMX174 cells. The specific mu-opioid receptor antagonist, Cys2, Tyr3, Arg5, Pen7 (CTAP) amide, also blocked the enhancing effect of alcohol on HIV infection. Investigation of the underlying mechanism for the alcohol action showed that alcohol significantly increased endogenous beta-endorphin production and induced mu-opioid receptor mRNA expression in PBL and CEMX174 cells. The role of beta-endorphin in alcohol-mediated enhancement of HIV infection was indicated by the observations that naltrexone and CTAP antagonized ether alcohol- or exogenous beta-endorphin-mediated enhancement of HIV infection. These findings suggest a biological mechanism for the potential therapeutic benefit of naltrexone in treating HIV-infected alcoholics.


Subject(s)
Alcohol Deterrents/pharmacology , Ethanol/pharmacology , HIV-1/physiology , Lymphocytes/drug effects , Naltrexone/pharmacology , Receptors, Opioid, mu/antagonists & inhibitors , T-Lymphocytes/drug effects , beta-Endorphin/physiology , Adult , Alcohol Deterrents/therapeutic use , Alcoholism/complications , Alcoholism/immunology , Cells, Cultured/drug effects , Cells, Cultured/virology , Disease Susceptibility , Drug Evaluation, Preclinical , Female , HIV Infections/etiology , HIV Reverse Transcriptase/analysis , Humans , Hybrid Cells/drug effects , Hybrid Cells/virology , Leukemia Virus, Murine/physiology , Lymphocytes/virology , Male , Middle Aged , Naltrexone/therapeutic use , Peptide Fragments , Peptides/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Opioid, mu/biosynthesis , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/physiology , Somatostatin , T-Lymphocytes/virology , Up-Regulation/drug effects , Virion/physiology , Virus Replication/drug effects , beta-Endorphin/biosynthesis , beta-Endorphin/genetics
15.
J Virol ; 79(2): 1207-14, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15613347

ABSTRACT

Two acidic domains of the Potato leafroll virus (PLRV) coat protein, separated by 55 amino acids and predicted to be adjacent surface features on the virion, were the focus of a mutational analysis. Eleven site-directed mutants were generated from a cloned infectious cDNA of PLRV and delivered to plants by Agrobacterium-mediated mechanical inoculation. Alanine substitutions of any of the three amino acids of the sequence EWH (amino acids 170 to 172) or of D177 disrupted the ability of the coat protein to assemble stable particles and the ability of the viral RNA to move systemically in four host plant species. Alanine substitution of E109, D173, or E176 reduced the accumulation of virus in agrobacterium-infiltrated tissues, the efficiency of systemic infection, and the efficiency of aphid transmission relative to wild-type virus, but the mutations did not affect virion stability. A structural model of the PLRV capsid predicted that the amino acids critical for virion assembly were located within a depression at the center of a coat protein trimer. The other amino acids that affected plant infection and/or aphid transmission were predicted to be located around the perimeter of the depression. PLRV virions play key roles in phloem-limited virus movement in plant hosts as well as in transport and persistence in the aphid vectors. These results identified amino acid residues in a surface-oriented loop of the coat protein that are critical for virus assembly and stability, systemic infection of plants, and movement of virus through aphid vectors.


Subject(s)
Aphids/virology , Capsid Proteins/physiology , Luteovirus/physiology , Solanum tuberosum/virology , Virion/physiology , Virus Assembly , Animals , Capsid Proteins/chemistry
16.
J Immunol ; 170(9): 4840-5, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12707367

ABSTRACT

HIV-1 is an enveloped retrovirus that acquires its outer membrane as the virion exits the cell. Because of the association of apoptosis with the progression of AIDS, HIV-1-infected T cells or macrophages might be expected to express elevated levels of surface phosphatidylserine (PS), a hallmark of programmed cell death. Virions produced by these cells would also be predicted to have PS on the surface of their envelopes. In this study, data are presented that support this hypothesis and suggest that PS is required for macrophage infection. The PS-specific protein annexin V was used to enrich for virus particles and to inhibit HIV-1 replication in primary macrophages, but not T cells. HIV-1 replication was also significantly inhibited with vesicles consisting of PS, but not phosphatidylcholine. PS is specifically required for HIV-1 infection because viruses pseudotyped with vesicular stomatitis virus G and amphotropic murine leukemia virus envelopes were not inhibited by PS vesicles or annexin V. These data indicate that PS is an important cofactor for HIV-1 infection of macrophages.


Subject(s)
Adjuvants, Immunologic/physiology , HIV-1/pathogenicity , Macrophages/virology , Monocytes/virology , Phosphatidylserines/physiology , Adjuvants, Immunologic/antagonists & inhibitors , Adjuvants, Immunologic/biosynthesis , Binding Sites/immunology , Gene Products, env/physiology , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Jurkat Cells , Macrophages/immunology , Macrophages/metabolism , Membrane Fusion/immunology , Monocytes/immunology , Monocytes/metabolism , Phosphatidylserines/antagonists & inhibitors , Phosphatidylserines/biosynthesis , Tumor Cells, Cultured , U937 Cells , Virion/pathogenicity , Virion/physiology , Virus Replication/immunology
17.
Antiviral Res ; 55(3): 447-55, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12206882

ABSTRACT

Casuarinin, a hydrolyzable tannin isolated from the bark of Terminalia arjuna Linn. (Combretaceae), was investigated for its antiviral activity on herpes simplex type 2 (HSV-2) in vitro. Results showed that the IC(50) of casuarinin in XTT and plaque reduction assays were 3.6+/-0.9 and 1.5+/-0.2 microM, respectively. The 50% cytotoxic concentration for cell growth (CC(50)) was 89+/-1 microM. Thus, the selectivity index (SI) (ratio of CC(50) to IC(50)) of casuarinin was 25 and 59 for XTT and plaque reduction assays, respectively. Casuarinin continued to exhibit antiviral activity even added 12 h after infection. During the attachment assay, casuarinin was shown to prevent the attachment of HSV-2 to cells. Furthermore, casuarinin also exhibited an activity in inhibiting the viral penetration. Interestingly, casuarinin was virucidal at a concentration of 25 microM, reducing viral titers up to 100,000-fold. This study concludes that casuarinin possesses anti-herpesvirus activity in inhibiting viral attachment and penetration, and also disturbing the late event(s) of infection.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 2, Human/drug effects , Hydrolyzable Tannins , Tannins/pharmacology , Terminalia/chemistry , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Herpes Simplex/virology , Herpesvirus 2, Human/pathogenicity , Herpesvirus 2, Human/physiology , Plant Bark/chemistry , Plants, Medicinal , Tannins/isolation & purification , Tannins/metabolism , Vero Cells , Viral Plaque Assay , Virion/physiology
18.
Acta Virol ; 44(2): 67-72, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10989696

ABSTRACT

Electron microscopic studies on the stability of immunosorbed (trapped) virions of potato viruses X, S and Y0 (PVX, PVS and PVY0) revealed disintegration and dislodging of PVY0 virions upon incubation with (1) antisera to PVX, PVS, or both diluted in saline, (2) 0.86% NaCl (saline) or 0.1 mol/l CaCl2 but not with 0.1 mol/l CaSO4 or 0.1 mol/l MgSO4. PVX virions, on the other hand, showed partial dislodging upon incubation with an antiserum to PVS diluted in saline, but complete disintegration and dislodging with saline. 0.1 mol/l CaCl2 caused partial dislodging while MgCl2, CaSO4 or MgSO4 (all 0.1 mol/l) had no apparent adverse effect. PVS virions were not affected by saline, CaCl2, MgCl2, CaSO4 or MgSO4 (all 0.1 mol/l) and were only partially dislodged by antisera to PVX or PVY0. Disintegration and/or dislodging of the PVX and PVY0 virions was prevented when (1) they were fixed with glutaraldehyde prior to incubation or (2) the virus extract contained bovine serum albumin (BSA) or (3) heterologous antisera were diluted in 0.1 mol/l phosphate buffer (PB) before use except the PVS antiserum which still caused disintegration and dislodging of PVY0 virions. Prior fixation of virions prevented their disruption and dislodging by saline only in the case of PVY0 but not PVX. On the other hand, BSA reverted the adverse effect of saline but not that of the PVS antiserum on PVY0 virions. The results presented here suggest (1) a disruptive effect of Cl' on PVX and PVY0 virions particularly when it was associated with Na+ and (2) an interaction between the immunosorbed virions of PVX or PVY0 and the antiserum to PVS.


Subject(s)
Carlavirus/physiology , Potexvirus/physiology , Potyvirus/physiology , Serum Albumin, Bovine/pharmacology , Solanum tuberosum/virology , Animals , Carlavirus/drug effects , Carlavirus/immunology , Carlavirus/ultrastructure , Cattle , Hydrogen-Ion Concentration , Ions , Plants, Toxic , Potexvirus/drug effects , Potexvirus/immunology , Potexvirus/ultrastructure , Potyvirus/drug effects , Potyvirus/immunology , Potyvirus/ultrastructure , Nicotiana , Virion/physiology
19.
J Virol ; 74(15): 6790-9, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10888618

ABSTRACT

Enigmatic mechanisms restore the resting state in activated lymphocytes following human immunodeficiency virus type 1 (HIV-1) infection, rarely allowing persistent nonproductive infection. We detail a mechanism whereby cellular factors could establish virological latency. The transcription factors YY1 and LSF cooperate in repression of transcription from the HIV-1 long terminal repeat (LTR). LSF recruits YY1 to the LTR via the zinc fingers of YY1. The first two zinc fingers were observed to be sufficient for this interaction in vitro. A mutant of LSF incapable of binding DNA blocked repression. Like other transcriptional repressors, YY1 can function via recruitment of histone deacetylase (HDAC). We find that HDAC1 copurifies with the LTR-binding YY1-LSF repressor complex, the domain of YY1 that interacts with HDAC1 is required to repress the HIV-1 promoter, expression of HDAC1 augments repression of the LTR by YY1, and the deacetylase inhibitor trichostatin A blocks repression mediated by YY1. This novel link between HDAC recruitment and inhibition of HIV-1 expression by YY1 and LSF, in the natural context of a viral promoter integrated into chromosomal DNA, is the first demonstration of a molecular mechanism of repression of HIV-1. YY1 and LSF may establish transcriptional and virological latency of HIV, a state that has recently been recognized in vivo and has significant implications for the long-term treatment of AIDS.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , HIV Long Terminal Repeat , HIV-1/genetics , Histone Deacetylases/metabolism , Transcription Factors/metabolism , DNA, Viral/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Erythroid-Specific DNA-Binding Factors , HIV-1/metabolism , HeLa Cells , Humans , Promoter Regions, Genetic , RNA-Binding Proteins , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Virion/physiology , YY1 Transcription Factor
20.
J Gen Virol ; 81(Pt 1): 267-72, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10640566

ABSTRACT

cDNA copies of the coat protein (CP) gene of Indian peanut clump virus (IPCV)-H were introduced into cells of Nicotiana benthamiana or Escherichia coli by transformation with vectors based on pROKII or pET respectively. In both plant and bacterial cells, IPCV CP was expressed and assembled to form virus-like particles (VLP). In plant extracts, the smallest preponderant particle length was about 50 nm. Other abundant lengths were about 85 and about 120 nm. The commonest VLP length in bacterial extracts was about 30 nm. Many of the longer VLP appeared to comprise aggregates of shorter particles. The lengths of the supposed 'monomer' VLP corresponded approximately to those expected for encapsidated CP gene transcript RNA. Immunocapture RT-PCR, using primers designed to amplify the CP gene, confirmed that the VLP contained RNA encoding IPCV-H CP. The results show that encapsidation does not require the presence of the 5'-terminal untranslated sequence of the virus RNA and suggest that if there is an 'origin of assembly' motif or sequence, it lies within the CP gene. When transgenic plants expressing IPCV-H CP were inoculated with IPCV-L, a strain that is serologically distinct from IPCV-H, the virus particles that accumulated contained both types of CP.


Subject(s)
Capsid/genetics , Escherichia coli/virology , Nicotiana/virology , Plant Viruses/genetics , Plants, Toxic , RNA Viruses/genetics , Virion/physiology , Arachis/virology , Capsid/metabolism , Escherichia coli/genetics , Gene Expression , Immunoblotting , Microscopy, Electron , Plants, Genetically Modified/virology , Reverse Transcriptase Polymerase Chain Reaction/methods , Nicotiana/genetics , Transformation, Genetic , Virus Assembly
SELECTION OF CITATIONS
SEARCH DETAIL