Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Mike C. Wolf is active.

Publication


Featured researches published by Mike C. Wolf.


PLOS Pathogens | 2006

Two Key Residues in EphrinB3 Are Critical for Its Use as an Alternative Receptor for Nipah Virus

Oscar A. Negrete; Mike C. Wolf; Hector C. Aguilar; Sven Enterlein; Wei-wei Wang; Elke Mühlberger; Stephen V. Su; Andrea Bertolotti-Ciarlet; Ramon Flick; Benhur Lee

EphrinB2 was recently discovered as a functional receptor for Nipah virus (NiV), a lethal emerging paramyxovirus. Ephrins constitute a class of homologous ligands for the Eph class of receptor tyrosine kinases and exhibit overlapping expression patterns. Thus, we examined whether other ephrins might serve as alternative receptors for NiV. Here, we show that of all known ephrins (ephrinA1–A5 and ephrinB1–B3), only the soluble Fc-fusion proteins of ephrinB3, in addition to ephrinB2, bound to soluble NiV attachment protein G (NiV-G). Soluble NiV-G bound to cell surface ephrinB3 and B2 with subnanomolar affinities (Kd = 0.58 nM and 0.06 nM for ephrinB3 and B2, respectively). Surface plasmon resonance analysis indicated that the relatively lower affinity of NiV-G for ephrinB3 was largely due to a faster off-rate (Koff = 1.94 × 10−3 s−1 versus 1.06 × 10−4 s−1 for ephrinB3 and B2, respectively). EphrinB3 was sufficient to allow for viral entry of both pseudotype and live NiV. Soluble ephrinB2 and B3 were able to compete for NiV-envelope-mediated viral entry on both ephrinB2- and B3-expressing cells, suggesting that NiV-G interacts with both ephrinB2 and B3 via an overlapping site. Mutational analysis indicated that the Leu–Trp residues in the solvent exposed G–H loop of ephrinB2 and B3 were critical determinants of NiV binding and entry. Indeed, replacement of the Tyr–Met residues in the homologous positions in ephrinB1 with Leu–Trp conferred NiV receptor activity to ephrinB1. Thus, ephrinB3 is a bona fide alternate receptor for NiV entry, and two residues in the G–H loop of the ephrin B-class ligands are critical determinants of NiV receptor activity.


Proceedings of the National Academy of Sciences of the United States of America | 2010

A broad-spectrum antiviral targeting entry of enveloped viruses

Mike C. Wolf; Alexander N. Freiberg; Tinghu Zhang; Zeynep Akyol-Ataman; Andrew Grock; Patrick Hong; Jianrong Li; Natalya F. Watson; Angela Q. Fang; Hector C. Aguilar; Matteo Porotto; Anna N. Honko; Robert Damoiseaux; John P. Miller; Sara E. Woodson; Steven Chantasirivisal; Vanessa Fontanes; Oscar A. Negrete; Paul Krogstad; Asim Dasgupta; Anne Moscona; Lisa E. Hensley; Sean P. J. Whelan; Kym F. Faull; Michael E. Jung; Benhur Lee

We describe an antiviral small molecule, LJ001, effective against numerous enveloped viruses including Influenza A, filoviruses, poxviruses, arenaviruses, bunyaviruses, paramyxoviruses, flaviviruses, and HIV-1. In sharp contrast, the compound had no effect on the infection of nonenveloped viruses. In vitro and in vivo assays showed no overt toxicity. LJ001 specifically intercalated into viral membranes, irreversibly inactivated virions while leaving functionally intact envelope proteins, and inhibited viral entry at a step after virus binding but before virus–cell fusion. LJ001 pretreatment also prevented virus-induced mortality from Ebola and Rift Valley fever viruses. Structure–activity relationship analyses of LJ001, a rhodanine derivative, implicated both the polar and nonpolar ends of LJ001 in its antiviral activity. LJ001 specifically inhibited virus–cell but not cell–cell fusion, and further studies with lipid biosynthesis inhibitors indicated that LJ001 exploits the therapeutic window that exists between static viral membranes and biogenic cellular membranes with reparative capacity. In sum, our data reveal a class of broad-spectrum antivirals effective against enveloped viruses that target the viral lipid membrane and compromises its ability to mediate virus–cell fusion.


Journal of Virology | 2006

N-glycans on Nipah virus fusion protein protect against neutralization but reduce membrane fusion and viral entry.

Hector C. Aguilar; Kenneth A. Matreyek; Claire Marie Filone; Sara T. Hashimi; Ernest L. Levroney; Oscar A. Negrete; Andrea Bertolotti-Ciarlet; Daniel Y. Choi; Ian McHardy; Jennifer A. Fulcher; Stephen V. Su; Mike C. Wolf; Luciana Kohatsu; Linda G. Baum; Benhur Lee

ABSTRACT Nipah virus (NiV) is a deadly emerging paramyxovirus. The NiV attachment (NiV-G) and fusion (NiV-F) envelope glycoproteins mediate both syncytium formation and viral entry. Specific N-glycans on paramyxovirus fusion proteins are generally required for proper conformational integrity and biological function. However, removal of individual N-glycans on NiV-F had little negative effect on processing or fusogenicity and has even resulted in slightly increased fusogenicity. Here, we report that in both syncytium formation and viral entry assays, removal of multiple N-glycans on NiV-F resulted in marked increases in fusogenicity (>5-fold) but also resulted in increased sensitivity to neutralization by NiV-F-specific antisera. The mechanism underlying the hyperfusogenicity of these NiV-F N-glycan mutants is likely due to more-robust six-helix bundle formation, as these mutants showed increased fusion kinetics and were more resistant to neutralization by a fusion-inhibitory reagent based on the C-terminal heptad repeat region of NiV-F. Finally, we demonstrate that the fusogenicities of the NiV-F N-glycan mutants were inversely correlated with the relative avidities of NiV-Fs interactions with NiV-G, providing support for the attachment protein “displacement” model of paramyxovirus fusion. Our results indicate that N-glycans on NiV-F protect NiV from antibody neutralization, suggest that this “shielding” role comes together with limiting cell-cell fusion and viral entry efficiencies, and point to the mechanisms underlying the hyperfusogenicity of these N-glycan mutants. These features underscore the varied roles that N-glycans on NiV-F play in the pathobiology of NiV entry but also shed light on the general mechanisms of paramyxovirus fusion with host cells.


PLOS Pathogens | 2010

Ubiquitin-regulated nuclear-cytoplasmic trafficking of the Nipah virus matrix protein is important for viral budding.

Yao E. Wang; Arnold Park; Michael P. Lake; Mickey Pentecost; Betsabe Torres; Tatyana E. Yun; Mike C. Wolf; Alexander N. Freiberg; Benhur Lee

Paramyxoviruses are known to replicate in the cytoplasm and bud from the plasma membrane. Matrix is the major structural protein in paramyxoviruses that mediates viral assembly and budding. Curiously, the matrix proteins of a few paramyxoviruses have been found in the nucleus, although the biological function associated with this nuclear localization remains obscure. We report here that the nuclear-cytoplasmic trafficking of the Nipah virus matrix (NiV-M) protein and associated post-translational modification play a critical role in matrix-mediated virus budding. Nipah virus (NiV) is a highly pathogenic emerging paramyxovirus that causes fatal encephalitis in humans, and is classified as a Biosafety Level 4 (BSL4) pathogen. During live NiV infection, NiV-M was first detected in the nucleus at early stages of infection before subsequent localization to the cytoplasm and the plasma membrane. Mutations in the putative bipartite nuclear localization signal (NLS) and the leucine-rich nuclear export signal (NES) found in NiV-M impaired its nuclear-cytoplasmic trafficking and also abolished NiV-M budding. A highly conserved lysine residue in the NLS served dual functions: its positive charge was important for mediating nuclear import, and it was also a potential site for monoubiquitination which regulates nuclear export of the protein. Concordantly, overexpression of ubiquitin enhanced NiV-M budding whereas depletion of free ubiquitin in the cell (via proteasome inhibitors) resulted in nuclear retention of NiV-M and blocked viral budding. Live Nipah virus budding was exquisitely sensitive to proteasome inhibitors: bortezomib, an FDA-approved proteasome inhibitor for treating multiple myeloma, reduced viral titers with an IC50 of 2.7 nM, which is 100-fold less than the peak plasma concentration that can be achieved in humans. This opens up the possibility of using an “off-the-shelf” therapeutic against acute NiV infection.


PLOS Pathogens | 2013

A Mechanistic Paradigm for Broad-Spectrum Antivirals that Target Virus-Cell Fusion

Frederic Vigant; Jihye Lee; Axel Hollmann; Lukas Bahati Tanner; Zeynep Akyol Ataman; Tatyana Yun; Guanghou Shui; Hector C. Aguilar; Dong Zhang; David Meriwether; Gleyder Roman-Sosa; Lindsey R. Robinson; Terry L. Juelich; Hubert Buczkowski; Sunwen Chou; Miguel A. R. B. Castanho; Mike C. Wolf; Jennifer K. Smith; Ashley C. Banyard; Margaret Kielian; Srinivasa T. Reddy; Markus R. Wenk; Matthias Selke; Nuno C. Santos; Alexander N. Freiberg; Michael E. Jung; Benhur Lee

LJ001 is a lipophilic thiazolidine derivative that inhibits the entry of numerous enveloped viruses at non-cytotoxic concentrations (IC50≤0.5 µM), and was posited to exploit the physiological difference between static viral membranes and biogenic cellular membranes. We now report on the molecular mechanism that results in LJ001s specific inhibition of virus-cell fusion. The antiviral activity of LJ001 was light-dependent, required the presence of molecular oxygen, and was reversed by singlet oxygen (1O2) quenchers, qualifying LJ001 as a type II photosensitizer. Unsaturated phospholipids were the main target modified by LJ001-generated 1O2. Hydroxylated fatty acid species were detected in model and viral membranes treated with LJ001, but not its inactive molecular analog, LJ025. 1O2-mediated allylic hydroxylation of unsaturated phospholipids leads to a trans-isomerization of the double bond and concurrent formation of a hydroxyl group in the middle of the hydrophobic lipid bilayer. LJ001-induced 1O2-mediated lipid oxidation negatively impacts on the biophysical properties of viral membranes (membrane curvature and fluidity) critical for productive virus-cell membrane fusion. LJ001 did not mediate any apparent damage on biogenic cellular membranes, likely due to multiple endogenous cytoprotection mechanisms against phospholipid hydroperoxides. Based on our understanding of LJ001s mechanism of action, we designed a new class of membrane-intercalating photosensitizers to overcome LJ001s limitations for use as an in vivo antiviral agent. Structure activity relationship (SAR) studies led to a novel class of compounds (oxazolidine-2,4-dithiones) with (1) 100-fold improved in vitro potency (IC50<10 nM), (2) red-shifted absorption spectra (for better tissue penetration), (3) increased quantum yield (efficiency of 1O2 generation), and (4) 10–100-fold improved bioavailability. Candidate compounds in our new series moderately but significantly (p≤0.01) delayed the time to death in a murine lethal challenge model of Rift Valley Fever Virus (RVFV). The viral membrane may be a viable target for broad-spectrum antivirals that target virus-cell fusion.


Journal of Virological Methods | 2009

Development of a Neutralization Assay for Nipah Virus Using Pseudotype Particles

Azaibi Tamin; Brian H. Harcourt; Michael K. Lo; James A. Roth; Mike C. Wolf; Benhur Lee; Hana Weingartl; Jean-Christophe Audonnet; William J. Bellini; Paul A. Rota

Nipah virus (NiV) and Hendra virus (HeV) are zoonotic paramyxoviruses capable of causing severe disease in humans and animals. These viruses require biosafety level 4 (BSL-4) containment. Like other paramyxoviruses, the plaque reduction neutralization test (PRNT) can be used to detect antibodies to the surface glycoproteins, fusion (F) and attachment (G), and PRNT titers give an indication of protective immunity. Unfortunately, for NiV and HeV, the PRNT must be performed in BSL-4 containment and takes several days to complete. Thus, we have developed a neutralization assay using VSV pseudotype particles expressing the F and G proteins of NiV (pVSV-NiV-F/G) as target antigens. This rapid assay, which can be performed at BSL-2, was evaluated using serum samples from outbreak investigations and more than 300 serum samples from an experimental NiV vaccination study in swine. The results of the neutralization assays with pVSV-NiV-F/G as antigen showed a good correlation with those of standard PRNT. Therefore, this new method has the potential to be a rapid and cost-effective diagnostic method, especially in locations that lack high containment facilities, and will provide a valuable tool for basic research and vaccine development.


Virology Journal | 2009

A catalytically and genetically optimized β-lactamase-matrix based assay for sensitive, specific, and higher throughput analysis of native henipavirus entry characteristics

Mike C. Wolf; Yao Wang; Alexander N. Freiberg; Hector C. Aguilar; Benhur Lee

Nipah virus (NiV) and Hendra virus (HeV) are the only paramyxoviruses requiring Biosafety Level 4 (BSL-4) containment. Thus, study of henipavirus entry at less than BSL-4 conditions necessitates the use of cell-cell fusion or pseudotyped reporter virus assays. Yet, these surrogate assays may not fully emulate the biological properties unique to the virus being studied. Thus, we developed a henipaviral entry assay based on a β-lactamase-Nipah Matrix (βla-M) fusion protein. We first codon-optimized the bacterial βla and the NiV-M genes to ensure efficient expression in mammalian cells. The βla-M construct was able to bud and form virus-like particles (VLPs) that morphologically resembled paramyxoviruses. βla-M efficiently incorporated both NiV and HeV fusion and attachment glycoproteins. Entry of these VLPs was detected by cytosolic delivery of βla-M, resulting in enzymatic and fluorescent conversion of the pre-loaded CCF2-AM substrate. Soluble henipavirus receptors (ephrinB2) or antibodies against the F and/or G proteins blocked VLP entry. Additionally, a Y105W mutation engineered into the catalytic site of βla increased the sensitivity of our βla-M based infection assays by 2-fold. In toto, these methods will provide a more biologically relevant assay for studying henipavirus entry at less than BSL-4 conditions.


Future Virology | 2007

Pathobiology of henipavirus entry: insights into therapeutic strategies

Mike C. Wolf; Oscar A. Negrete; Benhur Lee

The recently emerged paramyxoviruses, Nipah (NiV) and Hendra (HeV), designated as Biosafety Level 4 pathogens, can cause lethal respiratory and neurological disease in both animals and humans. NiV outbreaks have been associated with efficient transmission amongst livestock (pigs) and mortality rates exceeding 70%, with documented cases of human-to-human transmission. Without vaccines or effective therapeutics, NiV and HeV continue to present an impending threat to global health and economies. The current understanding of henipavirus pathobiology has led to the development of small animal models reflecting certain aspects of the human pathology. In this review, we discuss how these animal models have been critical in testing vaccination strategies and in eliciting neutralizing antibodies against the envelope glycoproteins. Additionally, the discovery of the viral receptor and development of pseudotyped-viral systems have allowed us to explore the multiple opportunities for therapeutic intervention existing...


Archive | 2009

Novel Antiviral Agents for Enveloped Viruses

Benhur Lee; Michael E. Jung; Mike C. Wolf; Tinghu Zhang


Archive | 2011

Broad spectrum antiviral and antiparasitic agents

Benhur Lee; Michael E. Jung; Jihye Lee; Frederic Vigant; Peter J. Bradley; Mike C. Wolf; Bettina E. Hajagos

Collaboration


Dive into the Mike C. Wolf's collaboration.

Top Co-Authors

Avatar

Benhur Lee

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Hector C. Aguilar

Washington State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander N. Freiberg

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar

Oscar A. Negrete

Sandia National Laboratories

View shared research outputs
Top Co-Authors

Avatar

Frederic Vigant

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Jihye Lee

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew Grock

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge