Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Antiviral Res ; 193: 105084, 2021 09.
Article in English | MEDLINE | ID: mdl-34077807

ABSTRACT

Nipah virus (NiV) and Hendra virus (HeV) are highly pathogenic, bat-borne paramyxoviruses in the genus Henipavirus that cause severe and often fatal acute respiratory and/or neurologic diseases in humans and livestock. There are currently no approved antiviral therapeutics or vaccines for use in humans to treat or prevent NiV or HeV infection. To facilitate development of henipavirus antivirals, a high-throughput screening (HTS) platform was developed based on a well-characterized recombinant version of the nonpathogenic Henipavirus, Cedar virus (rCedV). Using reverse genetics, a rCedV encoding firefly luciferase (rCedV-Luc) was rescued and its utility evaluated for high-throughput antiviral compound screening. The luciferase reporter gene signal kinetics of rCedV-Luc in different human cell lines was characterized and validated as an authentic real-time measure of viral growth. The rCedV-Luc platform was optimized as an HTS assay that demonstrated high sensitivity with robust Z' scores, excellent signal-to-background ratios and coefficients of variation. Eight candidate compounds that inhibited rCedV replication were identified for additional validation and demonstrated that 4 compounds inhibited authentic NiV-Bangladesh replication. Further evaluation of 2 of the 4 validated compounds in a 9-point dose response titration demonstrated potent antiviral activity against NiV-Bangladesh and HeV, with minimal cytotoxicity. This rCedV reporter can serve as a surrogate yet authentic BSL-2 henipavirus platform that will dramatically accelerate drug candidate identification in the development of anti-henipavirus therapies.


Subject(s)
Antiviral Agents/pharmacology , Henipavirus Infections/drug therapy , Henipavirus/drug effects , High-Throughput Screening Assays , Viral Envelope Proteins/metabolism , Cell Line , Genes, Reporter , Henipavirus/physiology , Henipavirus Infections/virology , Humans , Luciferases/genetics , Luciferases/metabolism , Recombination, Genetic , Viral Envelope Proteins/genetics , Virus Internalization/drug effects , Virus Replication/drug effects
2.
Antiviral Res ; 153: 101-113, 2018 05.
Article in English | MEDLINE | ID: mdl-29601894

ABSTRACT

Ebolaviruses, marburgviruses, and henipaviruses are zoonotic pathogens belonging to the Filoviridae and Paramyxoviridae families. They exemplify viruses that continue to spill over into the human population, causing outbreaks characterized by high mortality and significant clinical sequelae in survivors of infection. There are currently no approved small molecule therapeutics for use in humans against these viruses. In this study, we evaluated the antiviral activity of the nucleoside analog 4'-azidocytidine (4'N3-C, R1479) and its 2'-monofluoro- and 2'-difluoro-modified analogs (2'F-4'N3-C and 2'diF-4'N3-C) against representative paramyxoviruses (Nipah virus, Hendra virus, measles virus, and human parainfluenza virus 3) and filoviruses (Ebola virus, Sudan virus, and Ravn virus). We observed enhanced antiviral activity against paramyxoviruses with both 2'diF-4'N3-C and 2'F-4'N3-C compared to R1479. On the other hand, while R1479 and 2'diF-4'N3-C inhibited filoviruses similarly to paramyxoviruses, we observed 10-fold lower filovirus inhibition by 2'F-4'N3-C. To our knowledge, this is the first study to compare the susceptibility of paramyxoviruses and filoviruses to R1479 and its 2'-fluoro-modified analogs. The activity of these compounds against negative-strand RNA viruses endorses the development of 4'-modified nucleoside analogs as broad-spectrum therapeutics against zoonotic viruses of public health importance.


Subject(s)
Antiviral Agents/pharmacology , Cytidine/analogs & derivatives , Ebolavirus/drug effects , Henipavirus/drug effects , Marburgvirus/drug effects , Cytidine/pharmacology , Microbial Sensitivity Tests
3.
Viruses ; 4(2): 280-308, 2012 02.
Article in English | MEDLINE | ID: mdl-22470837

ABSTRACT

The Paramyxoviridae genus Henipavirus is presently represented by the type species Hendra and Nipah viruses which are both recently emerged zoonotic viral pathogens responsible for repeated outbreaks associated with high morbidity and mortality in Australia, Southeast Asia, India and Bangladesh. These enveloped viruses bind and enter host target cells through the coordinated activities of their attachment (G) and class I fusion (F) envelope glycoproteins. The henipavirus G glycoprotein interacts with host cellular B class ephrins, triggering conformational alterations in G that lead to the activation of the F glycoprotein, which facilitates the membrane fusion process. Using the recently published structures of HeV-G and NiV-G and other paramyxovirus glycoproteins, we review the features of the henipavirus envelope glycoproteins that appear essential for mediating the viral fusion process, including receptor binding, G-F interaction, F activation, with an emphasis on G and the mutations that disrupt viral infectivity. Finally, recent candidate therapeutics for henipavirus-mediated disease are summarized in light of their ability to inhibit HeV and NiV entry by targeting their G and F glycoproteins.


Subject(s)
Antiviral Agents/pharmacology , Henipavirus/drug effects , Henipavirus/pathogenicity , Viral Envelope Proteins/metabolism , Viral Fusion Proteins/metabolism , Virus Internalization/drug effects , Drug Therapy/methods , Henipavirus Infections/therapy , Henipavirus Infections/virology , Host-Pathogen Interactions , Humans , Immunotherapy/methods , Models, Molecular , Protein Conformation , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/chemistry , Viral Fusion Proteins/antagonists & inhibitors , Viral Fusion Proteins/chemistry
4.
Curr Opin Virol ; 2(2): 176-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22482714

ABSTRACT

The henipaviruses, Hendra virus and Nipah virus, are classic examples of recently emerged viral zoonoses. In a relatively short time since their discoveries in the mid and late 1990s, respectively, a great deal of new information has been accumulated detailing their biology and certain unique characteristics. Their broad species tropism and abilities to cause severe and often fatal respiratory and/or neurologic disease in both animals and humans has sparked considerable interest in developing effective antiviral strategies to prevent or treat henipavirus infection and disease. Here, recent findings on the few most advanced henipavirus countermeasures are summarized and discussed.


Subject(s)
Antiviral Agents/therapeutic use , Henipavirus Infections/drug therapy , Henipavirus Infections/epidemiology , Henipavirus/physiology , Animals , Disease Outbreaks , Henipavirus/drug effects , Henipavirus/genetics , Henipavirus/isolation & purification , Henipavirus Infections/virology , Humans
5.
PLoS One ; 6(7): e22488, 2011.
Article in English | MEDLINE | ID: mdl-21811620

ABSTRACT

Bats are natural reservoirs for a spectrum of infectious zoonotic diseases including the recently emerged henipaviruses (Hendra and Nipah viruses). Henipaviruses have been observed both naturally and experimentally to cause serious and often fatal disease in many different mammal species, including humans. Interestingly, infection of the flying fox with henipaviruses occurs in the absence of clinical disease. The extreme variation in the disease pattern between humans and bats has led to an investigation into the effects of henipavirus infection on the innate immune response in bat cell lines. We report that henipavirus infection does not result in the induction of interferon expression, and the viruses also inhibit interferon signaling. We also confirm that the interferon production and signaling block in bat cells is not due to differing viral protein expression levels between human and bat hosts. This information, in addition to the known lack of clinical signs in bats following henipavirus infection, suggests that bats control henipavirus infection by an as yet unidentified mechanism, not via the interferon response. This is the first report of henipavirus infection in bat cells specifically investigating aspects of the innate immune system.


Subject(s)
Chiroptera/virology , Henipavirus Infections/immunology , Henipavirus Infections/virology , Henipavirus/immunology , Interferons/biosynthesis , Signal Transduction/immunology , Animals , Cell Line , Genes, Viral/genetics , Henipavirus/drug effects , Henipavirus/genetics , Humans , Interferon Type I/biosynthesis , Interferons/pharmacology , Signal Transduction/drug effects , Viral Proteins/metabolism
6.
J Virol ; 81(19): 10567-74, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17652384

ABSTRACT

Hendra virus (HeV) and Nipah virus (NiV) constitute the Henipavirus genus of paramyxoviruses, both fatal in humans and with the potential for subversion as agents of bioterrorism. Binding of the HeV/NiV attachment protein (G) to its receptor triggers a series of conformational changes in the fusion protein (F), ultimately leading to formation of a postfusion six-helix bundle (6HB) structure and fusion of the viral and cellular membranes. The ectodomain of paramyxovirus F proteins contains two conserved heptad repeat regions, the first (the N-terminal heptad repeat [HRN]) adjacent to the fusion peptide and the second (the C-terminal heptad repeat [HRC]) immediately preceding the transmembrane domain. Peptides derived from the HRN and HRC regions of F are proposed to inhibit fusion by preventing activated F molecules from forming the 6HB structure that is required for fusion. We previously reported that a human parainfluenza virus 3 (HPIV3) F peptide effectively inhibits infection mediated by the HeV glycoproteins in pseudotyped-HeV entry assays more effectively than the comparable HeV-derived peptide, and we now show that this peptide inhibits live-HeV and -NiV infection. HPIV3 F peptides were also effective in inhibiting HeV pseudotype virus entry in a new assay that mimics multicycle replication. This anti-HeV/NiV efficacy can be correlated with the greater potential of the HPIV3 C peptide to interact with the HeV F N peptide coiled-coil trimer, as evaluated by thermal unfolding experiments. Furthermore, replacement of a buried glutamic acid (glutamic acid 459) in the C peptide with valine enhances antiviral potency and stabilizes the 6HB conformation. Our results strongly suggest that conserved interhelical packing interactions in the F protein fusion core are important determinants of C peptide inhibitory activity and offer a strategy for the development of more-potent analogs of F peptide inhibitors.


Subject(s)
Antiviral Agents/pharmacology , Henipavirus/drug effects , Molecular Mimicry , Peptides/pharmacology , Phosphoproteins/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Viral Proteins/pharmacology , Virus Internalization/drug effects , Amino Acid Sequence , Antiviral Agents/chemistry , Cell Line , Conserved Sequence , Hendra Virus/drug effects , Hendra Virus/physiology , Henipavirus/physiology , Humans , Molecular Sequence Data , Mutation , Nipah Virus/drug effects , Nipah Virus/physiology , Paramyxovirinae/drug effects , Peptides/chemistry , Peptides/genetics , Phosphoproteins/chemistry , Phosphoproteins/genetics , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
7.
Virol J ; 2: 57, 2005 Jul 18.
Article in English | MEDLINE | ID: mdl-16026621

ABSTRACT

BACKGROUND: The recent emergence of four new members of the paramyxovirus family has heightened the awareness of and re-energized research on new and emerging diseases. In particular, the high mortality and person to person transmission associated with the most recent Nipah virus outbreaks, as well as the very recent re-emergence of Hendra virus, has confirmed the importance of developing effective therapeutic interventions. We have previously shown that peptides corresponding to the C-terminal heptad repeat (HR-2) of the fusion envelope glycoprotein of Hendra virus and Nipah virus were potent inhibitors of both Hendra virus and Nipah virus-mediated membrane fusion using recombinant expression systems. In the current study, we have developed shorter, second generation HR-2 peptides which include a capped peptide via amidation and acetylation and two poly(ethylene glycol)-linked (PEGylated) peptides, one with the PEG moity at the C-terminus and the other at the N-terminus. Here, we have evaluated these peptides as well as the corresponding scrambled peptide controls in Nipah virus and Hendra virus-mediated membrane fusion and against infection by live virus in vitro. RESULTS: Unlike their predecessors, the second generation HR-2 peptides exhibited high solubility and improved synthesis yields. Importantly, both Nipah virus and Hendra virus-mediated fusion as well as live virus infection were potently inhibited by both capped and PEGylated peptides with IC50 concentrations similar to the original HR-2 peptides, whereas the scrambled modified peptides had no inhibitory effect. These data also indicate that these chemical modifications did not alter the functional properties of the peptides as inhibitors. CONCLUSION: Nipah virus and Hendra virus infection in vitro can be potently blocked by specific HR-2 peptides. The improved synthesis and solubility characteristics of the second generation HR-2 peptides will facilitate peptide synthesis for pre-clinical trial application in an animal model of Henipavirus infection. The applied chemical modifications are also predicted to increase the serum half-life in vivo and should increase the chance of success in the development of an effective antiviral therapy.


Subject(s)
Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Henipavirus/drug effects , Peptides/chemical synthesis , Peptides/pharmacology , Viral Envelope Proteins/antagonists & inhibitors , Virus Internalization/drug effects , Animals , Chlorocebus aethiops , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , HeLa Cells , Henipavirus/physiology , Humans , Inhibitory Concentration 50 , Models, Biological , Vero Cells , Viral Envelope Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...