Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
J Biol Chem ; 298(2): 101529, 2022 02.
Article in English | MEDLINE | ID: mdl-34953856

ABSTRACT

Remdesivir (RDV) is a direct-acting antiviral agent that is approved in several countries for the treatment of coronavirus disease 2019 caused by the severe acute respiratory syndrome coronavirus 2. RDV exhibits broad-spectrum antiviral activity against positive-sense RNA viruses, for example, severe acute respiratory syndrome coronavirus and hepatitis C virus, and nonsegmented negative-sense RNA viruses, for example, Nipah virus, whereas segmented negative-sense RNA viruses such as influenza virus or Crimean-Congo hemorrhagic fever virus are not sensitive to the drug. The reasons for this apparent efficacy pattern are unknown. Here, we expressed and purified representative RNA-dependent RNA polymerases and studied three biochemical parameters that have been associated with the inhibitory effects of RDV-triphosphate (TP): (i) selective incorporation of the nucleotide substrate RDV-TP, (ii) the effect of the incorporated RDV-monophosphate (MP) on primer extension, and (iii) the effect of RDV-MP in the template during incorporation of the complementary UTP. We found a strong correlation between antiviral effects and efficient incorporation of RDV-TP. Inhibition in primer extension reactions was heterogeneous and usually inefficient at higher NTP concentrations. In contrast, template-dependent inhibition of UTP incorporation opposite the embedded RDV-MP was seen with all polymerases. Molecular modeling suggests a steric conflict between the 1'-cyano group of the inhibitor and residues of the structurally conserved RNA-dependent RNA polymerase motif F. We conclude that future efforts in the development of nucleotide analogs with a broader spectrum of antiviral activities should focus on improving rates of incorporation while capitalizing on the inhibitory effects of a bulky 1'-modification.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Models, Molecular , RNA Viruses/enzymology , RNA-Dependent RNA Polymerase/antagonists & inhibitors , Adenosine Monophosphate/chemistry , Adenosine Monophosphate/pharmacology , Alanine/chemistry , Alanine/pharmacology , Antiviral Agents/pharmacology , Hepacivirus/drug effects , Hepacivirus/enzymology , Negative-Sense RNA Viruses/drug effects , Negative-Sense RNA Viruses/enzymology , Nipah Virus/drug effects , Nipah Virus/enzymology , Positive-Strand RNA Viruses/drug effects , Positive-Strand RNA Viruses/enzymology , RNA Viruses/drug effects , RNA, Viral/metabolism , RNA-Dependent RNA Polymerase/chemistry , RNA-Dependent RNA Polymerase/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Virus Replication/drug effects
2.
J Infect Dis ; 221(Supplement_4): S480-S492, 2020 May 11.
Article in English | MEDLINE | ID: mdl-32037447

ABSTRACT

Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that causes fatal encephalitis and respiratory disease in humans. There is currently no approved therapeutic for human use against NiV infection. Griffithsin (GRFT) is high-mannose oligosaccharide binding lectin that has shown in vivo broad-spectrum activity against viruses, including severe acute respiratory syndrome coronavirus, human immunodeficiency virus 1, hepatitis C virus, and Japanese encephalitis virus. In this study, we evaluated the in vitro antiviral activities of GRFT and its synthetic trimeric tandemer (3mG) against NiV and other viruses from 4 virus families. The 3mG had comparatively greater potency than GRFT against NiV due to its enhanced ability to block NiV glycoprotein-induced syncytia formation. Our initial in vivo prophylactic evaluation of an oxidation-resistant GRFT (Q-GRFT) showed significant protection against lethal NiV challenge in Syrian golden hamsters. Our results warrant further development of Q-GRFT and 3mG as potential NiV therapeutics.


Subject(s)
Antiviral Agents/pharmacology , Henipavirus Infections/drug therapy , Nipah Virus/drug effects , Plant Lectins/pharmacology , Virus Internalization/drug effects , Animals , Antiviral Agents/therapeutic use , Chlorocebus aethiops , Disease Models, Animal , Drug Evaluation, Preclinical , Female , HEK293 Cells , HeLa Cells , Henipavirus Infections/virology , Humans , Mesocricetus , Nipah Virus/isolation & purification , Plant Lectins/therapeutic use , Vero Cells
3.
J Virol ; 93(3)2019 02 01.
Article in English | MEDLINE | ID: mdl-30429347

ABSTRACT

Ebola virus (EBOV) and Nipah virus (NiV) infection of humans can cause fatal disease and constitutes a public health threat. In contrast, EBOV and NiV infection of fruit bats, the putative (EBOV) or proven (NiV) natural reservoir, is not associated with disease, and it is currently unknown how these animals control the virus. The human interferon (IFN)-stimulated antiviral effector protein tetherin (CD317, BST-2) blocks release of EBOV- and NiV-like particles from cells and is counteracted by the EBOV glycoprotein (GP). In contrast, it is unknown whether fruit bat tetherin restricts virus infection and is susceptible to GP-driven antagonism. Here, we report the sequence of fruit bat tetherin and show that its expression is IFN stimulated and associated with strong antiviral activity. Moreover, we demonstrate that EBOV-GP antagonizes tetherin orthologues of diverse species but fails to efficiently counteract fruit bat tetherin in virus-like particle (VLP) release assays. However, unexpectedly, tetherin was dispensable for robust IFN-mediated inhibition of EBOV spread in fruit bat cells. Thus, the VLP-based model systems mimicking tetherin-mediated inhibition of EBOV release and its counteraction by GP seem not to adequately reflect all aspects of EBOV release from IFN-stimulated fruit bat cells, potentially due to differences in tetherin expression levels that could not be resolved by the present study. In contrast, tetherin expression was essential for IFN-dependent inhibition of NiV infection, demonstrating that IFN-induced fruit bat tetherin exerts antiviral activity and may critically contribute to control of NiV and potentially other highly virulent viruses in infected animals.IMPORTANCE Ebola virus and Nipah virus (EBOV and NiV) can cause fatal disease in humans. In contrast, infected fruit bats do not develop symptoms but can transmit the virus to humans. Why fruit bats but not humans control infection is largely unknown. Tetherin is an antiviral host cell protein and is counteracted by the EBOV glycoprotein in human cells. Here, employing model systems, we show that tetherin of fruit bats displays higher antiviral activity than human tetherin and is largely resistant against counteraction by the Ebola virus glycoprotein. Moreover, we demonstrate that induction of tetherin expression is critical for interferon-mediated inhibition of NiV but, for at present unknown reasons, not EBOV spread in fruit bat cells. Collectively, our findings identify tetherin as an antiviral effector of innate immune responses in fruit bats, which might allow these animals to control infection with NiV and potentially other viruses that cause severe disease in humans.


Subject(s)
Antiviral Agents/pharmacology , Bone Marrow Stromal Antigen 2/pharmacology , Ebolavirus/drug effects , Hemorrhagic Fever, Ebola/virology , Henipavirus Infections/prevention & control , Nipah Virus/drug effects , Virus Replication/drug effects , Animals , Chiroptera , Hemorrhagic Fever, Ebola/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Humans , Immunity, Innate/drug effects , Interferons/pharmacology , Primates , Rodentia , Virus Release
4.
Vox Sang ; 115(3): 146-151, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31930543

ABSTRACT

BACKGROUND: Emerging viruses like severe acute respiratory syndrome coronavirus (SARS-CoV), Crimean-Congo haemorrhagic fever virus (CCHFV) and Nipah virus (NiV) have been identified to pose a potential threat to transfusion safety. In this study, the ability of the THERAFLEX UV-Platelets and THERAFLEX MB-Plasma pathogen inactivation systems to inactivate these viruses in platelet concentrates and plasma, respectively, was investigated. MATERIALS AND METHODS: Blood products were spiked with SARS-CoV, CCHFV or NiV, and then treated with increasing doses of UVC light (THERAFLEX UV-Platelets) or with methylene blue (MB) plus increasing doses of visible light (MB/light; THERAFLEX MB-Plasma). Samples were taken before and after treatment with each illumination dose and tested for residual infectivity. RESULTS: Treatment with half to three-fourths of the full UVC dose (0·2 J/cm2 ) reduced the infectivity of SARS-CoV (≥3·4 log), CCHFV (≥2·2 log) and NiV (≥4·3 log) to the limit of detection (LOD) in platelet concentrates, and treatment with MB and a fourth of the full light dose (120 J/cm2 ) decreased that of SARS-CoV (≥3·1 log), CCHFV (≥3·2 log) and NiV (≥2·7 log) to the LOD in plasma. CONCLUSION: Our study demonstrates that both THERAFLEX UV-Platelets (UVC) and THERAFLEX MB-Plasma (MB/light) effectively reduce the infectivity of SARS-CoV, CCHFV and NiV in platelet concentrates and plasma, respectively.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/radiation effects , Light , Methylene Blue/pharmacology , Nipah Virus/radiation effects , Severe acute respiratory syndrome-related coronavirus/radiation effects , Ultraviolet Rays , Virus Inactivation , Blood Platelets/virology , Blood Transfusion , Hemorrhagic Fever Virus, Crimean-Congo/drug effects , Humans , Nipah Virus/drug effects , Plasma/virology , Severe acute respiratory syndrome-related coronavirus/drug effects
5.
J Virol ; 88(8): 4353-65, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24501399

ABSTRACT

UNLABELLED: Severe acute respiratory syndrome coronavirus (SARS-CoV) and Ebola, Hendra, and Nipah viruses are members of different viral families and are known causative agents of fatal viral diseases. These viruses depend on cathepsin L for entry into their target cells. The viral glycoproteins need to be primed by protease cleavage, rendering them active for fusion with the host cell membrane. In this study, we developed a novel high-throughput screening assay based on peptides, derived from the glycoproteins of the aforementioned viruses, which contain the cathepsin L cleavage site. We screened a library of 5,000 small molecules and discovered a small molecule that can inhibit the cathepsin L cleavage of all viral peptides with minimal inhibition of cleavage of a host protein-derived peptide (pro-neuropeptide Y). The small molecule inhibited the entry of all pseudotyped viruses in vitro and the cleavage of SARS-CoV spike glycoprotein in an in vitro cleavage assay. In addition, the Hendra and Nipah virus fusion glycoproteins were not cleaved in the presence of the small molecule in a cell-based cleavage assay. Furthermore, we demonstrate that the small molecule is a mixed inhibitor of cathepsin L. Our broad-spectrum antiviral small molecule appears to be an ideal candidate for future optimization and development into a potent antiviral against SARS-CoV and Ebola, Hendra, and Nipah viruses. IMPORTANCE: We developed a novel high-throughput screening assay to identify small molecules that can prevent cathepsin L cleavage of viral glycoproteins derived from SARS-CoV and Ebola, Hendra, and Nipah viruses that are required for their entry into the host cell. We identified a novel broad-spectrum small molecule that could block cathepsin L-mediated cleavage and thus inhibit the entry of pseudotypes bearing the glycoprotein derived from SARS-CoV or Ebola, Hendra, or Nipah virus. The small molecule can be further optimized and developed into a potent broad-spectrum antiviral drug.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Ebolavirus/drug effects , Hendra Virus/drug effects , High-Throughput Screening Assays/methods , Nipah Virus/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , Small Molecule Libraries/pharmacology , Cathepsin L/metabolism , Ebolavirus/metabolism , Hendra Virus/metabolism , Humans , Nipah Virus/metabolism , Severe acute respiratory syndrome-related coronavirus/metabolism , Viral Envelope Proteins/metabolism , Virus Diseases/enzymology , Virus Diseases/virology
6.
Vet Pathol ; 52(1): 18-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25352204

ABSTRACT

The reports for Ebola virus Zaire (EBOV), Nipah virus, and Machupo virus (MACV) pathogenesis, in this issue of Veterinary Pathology, are timely considering recent events, both nationally and internationally. EBOV, Nipah virus, and MACV cause highly lethal infections for which no Food and Drug Administration (FDA) licensed vaccines or therapies exist. Not only are there concerns that these agents could be used by those with malicious intent, but shifts in ecological distribution of viral reservoirs due to climate change or globalization could lead to more frequent infections within remote regions than previously seen as well as outbreaks in more populous areas. The current EBOV epidemic shows no sign of abating across 3 West African nations (as of October 2014), including densely populated areas, far outpacing infection rates of previous outbreaks. A limited number of cases have also arisen in the United States and Europe. With few treatment options for these deadly viruses, development of animal models reflective of human disease is paramount to combat these diseases. As an example of this potential, a new treatment compound, ZMapp, that had demonstrated efficacy against EBOV infection in nonhuman primates (NHPs) received an emergency compassionate use exception from the FDA for the treatment of 2 American medical workers infected with EBOV, and they are currently virus free and recovering.


Subject(s)
Arenaviruses, New World/physiology , Disease Models, Animal , Ebolavirus/physiology , Hemorrhagic Fever, American/epidemiology , Hemorrhagic Fever, Ebola/epidemiology , Henipavirus Infections/epidemiology , Nipah Virus/physiology , Animals , Arenaviruses, New World/drug effects , Climate Change , Compassionate Use Trials , Disease Outbreaks , Ebolavirus/drug effects , Epidemics , Europe/epidemiology , Hemorrhagic Fever, American/drug therapy , Hemorrhagic Fever, American/virology , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Henipavirus Infections/drug therapy , Henipavirus Infections/virology , Humans , Internationality , Nipah Virus/drug effects , United States/epidemiology , United States Food and Drug Administration
7.
Emerg Microbes Infect ; 13(1): 2368217, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38865205

ABSTRACT

Nipah virus (NiV), a highly pathogenic Henipavirus in humans, has been responsible for annual outbreaks in recent years. Experiments involving live NiV are highly restricted to biosafety level 4 (BSL-4) laboratories, which impedes NiV research. In this study, we developed transcription and replication-competent NiV-like particles (trVLP-NiV) lacking N, P, and L genes. This trVLP-NiV exhibited the ability to infect and continuously passage in cells ectopically expressing N, P, and L proteins while maintaining stable genetic characteristics. Moreover, the trVLP-NiV displayed a favourable safety profile in hamsters. Using the system, we found the NiV nucleoprotein residues interacting with viral RNA backbone affected viral replication in opposite patterns. This engineered system was sensitive to well-established antiviral drugs, innate host antiviral factors, and neutralizing antibodies. We then established a high-throughput screening platform utilizing the trVLP-NiV, leading to the identification of tunicamycin as a potential anti-NiV compound. Evidence showed that tunicamycin inhibited NiV replication by decreasing the infectivity of progeny virions. In conclusion, this trVLP-NiV system provided a convenient and versatile molecular tool for investigating NiV molecular biology and conducting antiviral drug screening under BSL-2 conditions. Its application will contribute to the development of medical countermeasures against NiV infections.


Subject(s)
Henipavirus Infections , Nipah Virus , Virus Replication , Nipah Virus/physiology , Nipah Virus/genetics , Nipah Virus/drug effects , Animals , Cricetinae , Humans , Henipavirus Infections/virology , Transcription, Genetic , Virion/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Antiviral Agents/pharmacology , Vero Cells , Chlorocebus aethiops , Cell Line , RNA, Viral/genetics
8.
J Virol ; 85(16): 8422-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21653662

ABSTRACT

We have previously described heterotypic peptides from parainfluenza virus that potently inhibit Nipah virus in vitro but are not efficacious in vivo. In contrast, our second-generation inhibitors, featuring a cholesterol moiety, are also efficacious in vivo. The difference between in vitro and in vivo results led us to investigate the basis for this discrepancy. Here, we compare the activities of the compounds in standard laboratory cells and in cells relevant to the natural tropism of Nipah virus, i.e., primary neurons, and show that while our first-generation inhibitors are poorly active in primary neurons, the cholesterol-conjugated compounds are highly potent. These results highlight the advantage of evaluating antiviral potency in cells relevant to natural host target tissue.


Subject(s)
Antiviral Agents/pharmacology , Cholesterol/pharmacology , Neurons/virology , Nipah Virus/drug effects , Peptides/pharmacology , Viral Envelope Proteins/metabolism , Antiviral Agents/chemistry , Cell Line , Cholesterol/metabolism , Epithelial Cells/virology , Humans , Nipah Virus/physiology
9.
PLoS Pathog ; 6(10): e1001168, 2010 Oct 28.
Article in English | MEDLINE | ID: mdl-21060819

ABSTRACT

In the paramyxovirus cell entry process, receptor binding triggers conformational changes in the fusion protein (F) leading to viral and cellular membrane fusion. Peptides derived from C-terminal heptad repeat (HRC) regions in F have been shown to inhibit fusion by preventing formation of the fusogenic six-helix bundle. We recently showed that the addition of a cholesterol group to HRC peptides active against Nipah virus targets these peptides to the membrane where fusion occurs, dramatically increasing their antiviral effect. In this work, we report that unlike the untagged HRC peptides, which bind to the postulated extended intermediate state bridging the viral and cell membranes, the cholesterol tagged HRC-derived peptides interact with F before the fusion peptide inserts into the target cell membrane, thus capturing an earlier stage in the F-activation process. Furthermore, we show that cholesterol tagging renders these peptides active in vivo: the cholesterol-tagged peptides cross the blood brain barrier, and effectively prevent and treat in an established animal model what would otherwise be fatal Nipah virus encephalitis. The in vivo efficacy of cholesterol-tagged peptides, and in particular their ability to penetrate the CNS, suggests that they are promising candidates for the prevention or therapy of infection by Nipah and other lethal paramyxoviruses.


Subject(s)
Cholesterol/therapeutic use , Henipavirus Infections/prevention & control , Nipah Virus/physiology , Paramyxovirinae/physiology , Viral Fusion Proteins/antagonists & inhibitors , Virus Internalization , Amino Acid Motifs/drug effects , Amino Acid Motifs/physiology , Amino Acid Sequence , Animals , Cells, Cultured , Chlorocebus aethiops , Cholesterol/chemistry , Cholesterol/pharmacology , Down-Regulation , Henipavirus Infections/immunology , Henipavirus Infections/therapy , Humans , Models, Biological , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy , Nipah Virus/drug effects , Nipah Virus/immunology , Nipah Virus/pathogenicity , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/therapeutic use , Vero Cells , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/metabolism , Viral Fusion Proteins/physiology
10.
J Virol ; 84(13): 6760-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20357085

ABSTRACT

The fusion of enveloped viruses with the host cell is driven by specialized fusion proteins to initiate infection. The "class I" fusion proteins harbor two regions, typically two heptad repeat (HR) domains, which are central to the complex conformational changes leading to fusion: the first heptad repeat (HRN) is adjacent to the fusion peptide, while the second (HRC) immediately precedes the transmembrane domain. Peptides derived from the HR regions can inhibit fusion, and one HR peptide, T20 (enfuvirtide), is in clinical use for HIV-1. For paramyxoviruses, the activities of two membrane proteins, the receptor-binding protein (hemagglutinin-neuraminidase [HN] or G) and the fusion protein (F), initiate viral entry. The binding of HN or G to its receptor on a target cell triggers the activation of F, which then inserts into the target cell and mediates the membrane fusion that initiates infection. We have shown that for paramyxoviruses, the inhibitory efficacy of HR peptides is inversely proportional to the rate of F activation. For HIV-1, the antiviral potency of an HRC-derived peptide can be dramatically increased by targeting it to the membrane microdomains where fusion occurs, via the addition of a cholesterol group. We report here that for three paramyxoviruses-human parainfluenza virus type 3 (HPIV3), a major cause of lower respiratory tract diseases in infants, and the emerging zoonotic viruses Hendra virus (HeV) and Nipah virus (NiV), which cause lethal central nervous system diseases-the addition of cholesterol to a paramyxovirus HRC-derived peptide increased antiviral potency by 2 log units. Our data suggest that this enhanced activity is indeed the result of the targeting of the peptide to the plasma membrane, where fusion occurs. The cholesterol-tagged peptides on the cell surface create a protective antiviral shield, target the F protein directly at its site of action, and expand the potential utility of inhibitory peptides for paramyxoviruses.


Subject(s)
Antiviral Agents/pharmacology , Hendra Virus/physiology , Nipah Virus/physiology , Parainfluenza Virus 3, Human/physiology , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Chlorocebus aethiops , Hendra Virus/drug effects , Humans , Nipah Virus/drug effects , Parainfluenza Virus 3, Human/drug effects
11.
Biophys Chem ; 270: 106537, 2021 03.
Article in English | MEDLINE | ID: mdl-33450550

ABSTRACT

Nipah virus (NiV) infections are highly contagious and can cause severe febrile encephalitis. An outbreak of NiV infection has reported high mortality rates in Southeast Asian countries including Bangladesh, East Timor, Malaysia, Papua New Guinea, Vietnam, Cambodia, Indonesia, Madagascar, Philippines, Thailand and India. Considering the high risk for an epidemic outbreak, the World Health Organization (WHO) declared NiV as an emerging priority pathogen. However, there are no effective therapeutics or any FDA approved drugs available for the treatment of this infection. Among the known nine proteins of NiV, glycoprotein plays an important role in initiating the entry of viruses and attaching to the host cell receptors. Herein, three antiviral databases consisting of 79,892 chemical entities have been computationally screened against NiV glycoprotein (NiV-G). Particularly, multi-step molecular docking followed by extensive molecular binding interactions analyses, binding free energy estimation, in silico pharmacokinetics, synthetic accessibility and toxicity profile evaluations have been carried out for initial identification of potential NiV-G inhibitors. Further, molecular dynamics (MD) simulation has been performed to understand the dynamic properties of NiV-G protein-bound with proposed five inhibitors (G1-G5) and their interactions behavior, and any conformational changes in NiV-G protein during simulations. Moreover, Molecular Mechanics Poisson-Boltzmann Surface Area (MM-PBSA) based binding free energies (∆G) has been calculated from all MD simulation trajectories to understand the energy contribution of each proposed compound in maintaining and stabilizing the complex binding interactions with NiV-G protein. Proposed compounds showed high negative ∆G values ranging from -166.246 to -226.652 kJ/mol indicating a strong affinity towards the NiV-G protein.


Subject(s)
Antiviral Agents/pharmacology , Glycoproteins/antagonists & inhibitors , Nipah Virus/drug effects , Small Molecule Libraries/pharmacology , Viral Proteins/antagonists & inhibitors , Antiviral Agents/chemistry , Drug Discovery , Glycoproteins/chemistry , Glycoproteins/metabolism , Henipavirus Infections/drug therapy , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Nipah Virus/physiology , Small Molecule Libraries/chemistry , Viral Proteins/chemistry , Viral Proteins/metabolism
12.
J Gen Virol ; 91(Pt 3): 765-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19889926

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) are recently emerged, closely related and highly pathogenic paramyxoviruses that cause severe disease such as encephalitis in animals and humans with fatality rates of up to 75 %. Due to their high case fatality rate following human infection and because of the lack of effective vaccines or therapy, they are classified as Biosafety Level 4 pathogens. A recent study reported that chloroquine, an anti-malarial drug, was effective in preventing NiV and HeV infection in cell culture experiments. In the present study, the antiviral efficacy of chloroquine was analysed, individually and in combination with ribavirin, in the treatment of NiV and HeV infection in in vivo experiments, using a golden hamster model. Although the results confirmed the strong antiviral activity of both drugs in inhibiting viral spread in vitro, they did not prove to be protective in the in vivo model. Ribavirin delayed death from viral disease in NiV-infected hamsters by approximately 5 days, but no significant effect in HeV-infected hamsters was observed. Chloroquine did not protect hamsters when administered either individually or in combination with ribavirin, the latter indicating the lack of a favourable drug-drug interaction.


Subject(s)
Antiviral Agents/therapeutic use , Chloroquine/therapeutic use , Henipavirus Infections/drug therapy , Henipavirus Infections/mortality , Ribavirin/therapeutic use , Animals , Chloroquine/pharmacology , Cricetinae , Disease Models, Animal , Drug Therapy, Combination , Hendra Virus/drug effects , Humans , Mesocricetus , Nipah Virus/drug effects , Ribavirin/pharmacology , Survival Analysis , Treatment Outcome
13.
J Gen Virol ; 91(Pt 11): 2762-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20685931

ABSTRACT

Based on the structural similarity of viral fusion proteins within the family Paramyxoviridae, we tested recently described and newly synthesized acetanilide derivatives for their capacity to inhibit measles virus (MV)-, canine distemper virus (CDV)- and Nipah virus (NiV)-induced membrane fusion. We found that N-(3-cyanophenyl)-2-phenylacetamide (compound 1) has a high capacity to inhibit MV- and CDV-induced (IC(50) µM), but not NiV-induced, membrane fusion. This compound is of outstanding interest because it can be easily synthesized and its cytotoxicity is low [50 % cytotoxic concentration (CC(50)) ≥ 300 µM], leading to a CC(50)/IC(50) ratio of approximately 100. In addition, primary human peripheral blood lymphocytes and primary dog brain cell cultures (DBC) also tolerate high concentrations of compound 1. Infection of human PBMC with recombinant wild-type MV is inhibited by an IC(50) of approximately 20 µM. The cell-to-cell spread of recombinant wild-type CDV in persistently infected DBC can be nearly completely inhibited by compound 1 at 50 µM, indicating that the virus spread between brain cells is dependent on the activity of the viral fusion protein. Our findings demonstrate that this compound is a most applicable inhibitor of morbillivirus-induced membrane fusion in tissue culture experiments including highly sensitive primary cells.


Subject(s)
Antiviral Agents/pharmacology , Benzeneacetamides/pharmacology , Measles virus/drug effects , Measles virus/physiology , Membrane Fusion/drug effects , Virus Internalization/drug effects , Animals , Antiviral Agents/chemistry , Antiviral Agents/toxicity , Benzeneacetamides/chemistry , Benzeneacetamides/toxicity , Cell Survival/drug effects , Cells, Cultured , Chlorocebus aethiops , Distemper Virus, Canine/drug effects , Distemper Virus, Canine/physiology , Dogs , Humans , Inhibitory Concentration 50 , Lymphocytes/drug effects , Neurons/drug effects , Nipah Virus/drug effects , Nipah Virus/physiology
14.
J Virol ; 83(10): 5148-55, 2009 May.
Article in English | MEDLINE | ID: mdl-19264786

ABSTRACT

Nipah (NiV) and Hendra (HeV) viruses are emerging zoonotic paramyxoviruses that cause encephalitis in humans, with fatality rates of up to 75%. We designed a new high-throughput screening (HTS) assay for inhibitors of infection based on envelope glycoprotein pseudotypes. The assay simulates multicycle replication and thus identifies inhibitors that target several stages of the viral life cycle, but it still can be carried out under biosafety level 2 (BSL-2) conditions. These features permit a screen for antivirals for emerging viruses and select agents that otherwise would require BSL-4 HTS facilities. The screening of a small compound library identified several effective molecules, including the well-known compound chloroquine, as highly active inhibitors of pseudotyped virus infection. Chloroquine inhibited infection with live HeV and NiV at a concentration of 1 microM in vitro (50% inhibitory concentration, 2 microM), which is less than the plasma concentrations present in humans receiving chloroquine treatment for malaria. The mechanism for chloroquine's antiviral action likely is the inhibition of cathepsin L, a cellular enzyme that is essential for the processing of the viral fusion glycoprotein and the maturation of newly budding virions. Without this processing step, virions are not infectious. The identification of a compound that inhibits a known cellular target that is important for viral maturation but that had not previously been shown to have antiviral activity for henipaviruses highlights the validity of this new screening assay. Given the established safety profile and broad experience with chloroquine in humans, the results described here provide an option for treating individuals infected by these deadly viruses.


Subject(s)
Antiviral Agents/pharmacology , Chloroquine/pharmacology , Drug Discovery/methods , Hendra Virus/drug effects , Nipah Virus/drug effects , Animals , Chlorocebus aethiops , Hendra Virus/physiology , Henipavirus Infections/drug therapy , Humans , Nipah Virus/physiology , Vero Cells , Viral Envelope Proteins/metabolism , Virus Replication
15.
J Virol ; 83(22): 11979-82, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19759137

ABSTRACT

Hendra virus and Nipah virus, two zoonotic paramyxoviruses in the genus Henipavirus, have recently emerged and continue to cause sporadic disease outbreaks in humans and animals. Mortality rates of up to 75% have been reported in humans, but there are presently no clinically licensed therapeutics for treating henipavirus-induced disease. A recent report indicated that chloroquine, used in malaria therapy for over 70 years, prevented infection with Nipah virus in vitro. Chloroquine was assessed using a ferret model of lethal Nipah virus infection and found to be ineffective against Nipah virus infection in vivo.


Subject(s)
Antiviral Agents/therapeutic use , Chloroquine/therapeutic use , Ferrets/virology , Henipavirus Infections/drug therapy , Nipah Virus/drug effects , Animal Diseases/drug therapy , Animal Diseases/virology , Animals , Antiviral Agents/pharmacology , Chloroquine/pharmacokinetics , Chloroquine/pharmacology , Ferrets/metabolism , Humans , RNA, Viral/metabolism
16.
Sci Rep ; 10(1): 1477, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001794

ABSTRACT

The G-quadruplex (GQ) motifs are considered as potential drug-target sites for several human pathogenic viruses such as Zika, Hepatitis, Ebola, and Human Herpesviruses. The recent outbreaks of Nipah virus (NiV) in India, the highly fatal emerging zoonotic virus is a potential threat to global health security as no anti-viral drug or vaccine in currently available. Therefore, here in the present study, we sought to assess the ability of the putative G-quadruplex forming sequences in the NiV genome to form G-quadruplex structures and act as targets for anti-viral compounds. Bioinformatics analysis underpinned by various biophysical and biochemical techniques (such as NMR, CD, EMSA, DMS footprinting assay) confirmed the presence of two highly conserved G-quadruplex forming sequences (HGQs) in the G and L genes of NiV. These genes encode the cell attachment glycoprotein and RNA-dependent RNA polymerase, respectively and are essential for the virus entry and replication within the host cell. It remains possible that stabilization of these HGQs by the known G-quadruplex binding ligands like TMPyP4 and Braco-19 represents a promising strategy to inhibit the expression of the HGQ harboring genes and thereby stop the viral entry and replication inside the host cell. Accordingly, we report for the first time, that HGQs in Nipah virus genome are targets for G-quadruplex specific ligands; therefore, could serve as potential targets for anti-viral therapy.


Subject(s)
G-Quadruplexes , Genome, Viral , Nipah Virus/genetics , Acridines/pharmacology , Antiviral Agents/pharmacology , Computational Biology , Conserved Sequence , G-Quadruplexes/drug effects , Henipavirus Infections/virology , Humans , Hydrogen Bonding , India , Ligands , Nipah Virus/drug effects , Nipah Virus/physiology , Porphyrins/pharmacology , Virus Internalization , Virus Replication
17.
Curr Mol Pharmacol ; 13(2): 108-125, 2020.
Article in English | MEDLINE | ID: mdl-31657692

ABSTRACT

BACKGROUND: Nipah virus (NiV) and Hendra virus (HeV) of genus Henipavirus are the deadliest zoonotic viruses, which cause severe respiratory ailments and fatal encephalitis in humans and other susceptible animals. The fatality rate for these infections had been alarmingly high with no approved treatment available to date. Viral attachment and fusion with host cell membrane is essential for viral entry and is the most essential event of viral infection. Viral attachment is mediated by interaction of Henipavirus attachment glycoprotein (G) with the host cell receptor: Ephrin B2/B3, while viral fusion and endocytosis are mediated by the combined action of both viral glycoprotein (G) and fusion protein (F). CONCLUSION: This review highlights the mechanism of viral attachment, fusion and also explains the basic mechanism and pathobiology of this infection in humans. The drugs and therapeutics used either experimentally or clinically against NiV and HeV infection have been documented and classified in detail. Some amino acid residues essential for the functionality of G and F proteins were also emphasized. Therapeutic designing to target and block these residues can serve as a promising approach in future drug development against NiV and HeV.


Subject(s)
Antiviral Agents/pharmacology , Drug Design , Hendra Virus/drug effects , Nipah Virus/drug effects , Animals , Hendra Virus/genetics , Henipavirus Infections/physiopathology , Humans , Nipah Virus/genetics , Virus Internalization/drug effects
18.
Antiviral Res ; 175: 104712, 2020 03.
Article in English | MEDLINE | ID: mdl-31935422

ABSTRACT

Nipah virus (NiV) is a highly pathogenic zoonotic paramyxovirus that continues to cause outbreaks in humans characterized by high mortality and significant clinical sequelae in survivors. Currently, no therapeutics are approved for use in humans against NiV infection. Here, we report that 4'-chloromethyl-2'-deoxy-2'-fluorocytidine (ALS-8112) inhibits NiV. ALS-8112 is the parent nucleoside of lumicitabine, which has been evaluated in phase I and II clinical trials to treat pediatric and adult respiratory syncytial virus infection. In this study, we tested ALS-8112 against NiV and other major human respiratory pneumo- and paramyxoviruses in 2 human lung epithelial cell lines, and demonstrated the ability of ALS-8112 to reduce infectious wild-type NiV yield by over 6 orders of magnitude with no apparent cytotoxicity. However, further cytotoxicity testing in primary cells and bone marrow progenitor cells indicated cytotoxicity at higher concentrations of ALS-8112. Our results warrant the evaluation of lumicitabine against NiV infection in relevant animal models.


Subject(s)
Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , Nipah Virus/drug effects , Antiviral Agents/chemistry , Cell Line , Cell Survival/drug effects , Deoxycytidine/chemistry , Deoxycytidine/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/virology , HeLa Cells , Humans , Lung/cytology , Nucleosides/chemistry , Nucleosides/pharmacology , Paramyxoviridae/drug effects
19.
J Virol ; 82(23): 11628-36, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18815311

ABSTRACT

Two members of the paramyxovirus family, Nipah virus (NiV) and Hendra virus (HeV), are recent additions to a growing number of agents of emergent diseases which use bats as a natural host. Identification of ephrin-B2 and ephrin-B3 as cellular receptors for these viruses has enabled the development of immunotherapeutic reagents which prevent virus attachment and subsequent fusion. Here we present the structural analysis of the protein and carbohydrate components of the unbound viral attachment glycoprotein of NiV glycoprotein (NiV-G) at a 2.2-A resolution. Comparison with its ephrin-B2-bound form reveals that conformational changes within the envelope glycoprotein are required to achieve viral attachment. Structural differences are particularly pronounced in the 579-590 loop, a major component of the ephrin binding surface. In addition, the 236-245 loop is rather disordered in the unbound structure. We extend our structural characterization of NiV-G with mass spectrometric analysis of the carbohydrate moieties. We demonstrate that NiV-G is largely devoid of the oligomannose-type glycans that in viruses such as human immunodeficiency virus type 1 and Ebola virus influence viral tropism and the host immune response. Nevertheless, we find putative ligands for the endothelial cell lectin, LSECtin. Finally, by mapping structural conservation and glycosylation site positions from other members of the paramyxovirus family, we suggest the molecular surface involved in oligomerization. These results suggest possible pathways of virus-host interaction and strategies for the optimization of recombinant vaccines.


Subject(s)
Antiviral Agents/pharmacology , Carbohydrates/chemistry , Nipah Virus/chemistry , Vaccines, Synthetic/immunology , Viral Envelope Proteins/chemistry , Viral Vaccines/immunology , Cells, Cultured , Crystallization , Drug Design , Glycosylation , Humans , Nipah Virus/drug effects , Nipah Virus/immunology , Protein Conformation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
20.
Virus Res ; 142(1-2): 92-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19428741

ABSTRACT

We have recently described the development and validation of a high throughput screening assay suitable for henipavirus antiviral identification. While we are confident this assay is robust and effective, we wished to investigate assay performance in a range of alternative cell lines to determine if assay sensitivity and specificity could be improved. We evaluated ten different cell lines for their susceptibility to Hendra and Nipah virus infection and their sensitivity of detection of the effects of the broad spectrum antiviral, ribavirin and nine novel antivirals identified using our initial screening approach. Cell lines were grouped into three categories with respect to viral replication. Virus replicated best in Vero and BSR cells, followed by Hep-2, HeLa, BHK-21 and M17 cells. The lowest levels of RNA replication and viral protein expression were observed in BAEC, MMEC, A549 and ECV304 cells. Eight cell lines appeared to be similarly effective at discriminating the antiviral effects of ribavirin (<2.7-fold difference). The two cells lines most sensitive to the effect of ribavirin (ECV304 and BAEC) also displayed the lowest levels of viral replication while Vero cells were the least sensitive suggesting excess viral replication may limit drug efficacy and cell lines which limit viral replication may result in enhanced antiviral efficacy. However, there was no consistent trend observed with the other nine antivirals tested. While improvements in antiviral sensitivity in other cell lines may indicate an important role in future HTS assays, the slightly lower sensitivity to antiviral detection in Vero cells has inherent advantages in reducing the number of partially effective lead molecules identified during initial screens. Comparison of a panel of 54 novel antiviral compounds identified during routine screening of an in-house compound library in Vero, BHK-21 and BSR cells suggests no clear advantage of screening in either cell type.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , Hendra Virus/physiology , Nipah Virus/physiology , Virus Replication/drug effects , Animals , Cattle , Cell Line , Chlorocebus aethiops , Guinea Pigs , Hendra Virus/drug effects , Humans , Mice , Nipah Virus/drug effects , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL