Thefiloviruses(EbolaandMarburg)causeseverehemorrhagicfeversyndromeswithhighmortalityrates.As these RNA viruses are classified as NIAID Category A pathogens, the current lack of effective vaccines and antiviral therapeutics for these deadly pathogens is particularly concerning. We and others have established thatefficientfilovirusbuddingiscriticallydependentonthesubversionofhostproteinsTsg101andNedd4and that viral PTAP and PPxY late (L) budding domains are critical for these interactions, respectively. As disruption of virus budding would prevent virus dissemination, we propose to directly evaluate the ability of small molecule inhibitors to disrupt Tsg101PTAP and Nedd4PPxY interactions, thereby preventing virus budding. Our collaborators, Drs. Michael Lee and Mark Olson (USAMRIID, Ft. Detrick, MD), have used the known structures of Tsg101PTAP and Nedd4PPxY interactions to guide the in silico selection/design of competitiveinteractionblockers.Wearecurrentlyevaluatingtheabilityoftopcandidateinhibitorstodisruptthe hostdependent egress of virus particles in viruslike particle (VLP) budding assays. As Tsg101PTAP and Nedd4PPxYinteractionsaretooweakand/ortransienttobedetectedbystandardbiochemicalapproaches, ourlaboratoryhassuccessfullydevelopedapowerfulbimolecularcomplementation(BiMC)approachtodetect theseviralhostinteractionsinlivecells.Wewillusethisinnovativetechniquetodeterminewhethercandidate inhibitors disrupt these virushost interactions. Promising preliminary results with one lead compound (5539 0062)supportthefeasibilityofthisproposal.AsLdomaincontainingmatrixproteinsarerequiredforefficient viruscell separation of many RNA viruses, including retroviruses, arenaviruses, rhabdoviruses, paramyxoviruses,henipaviruses,andfiloviruses,wepredictthattargetingtheinteractiondomainbetween filovirusVP40andhostTsg101andNedd4willserveasbasisforthedevelopmentofnewandpowerfulbroad spectrumantiviraldrugs.Ourgroupofhighlyinteractiveandexpertcollaboratorswillbuilduponourpromising preliminary results obtained following an in silico screen of 4.8 million compounds to functionally validate candidatesmallmoleculesthatinhibitfilovirusTsg101(Aim1)andfilovirusNedd4(Aim2)interactionsduring theR21phaseofthisproposal.IntheR33phaseofthisproposal,wewilldevelopandutilizeFluorescence Lifetime Imaging Microscopy (FLIM) to functionally/mechanistically categorize inhibitors as they disrupt virus host protein interactions in real time and assess their mode of action, thereby laying the foundation for developmentandtestingofmultidrugtherapies(Aim3).Lastly,wewilldeterminetheefficacyofinhibitorsin preclinicallivevirusstudiesusingWTandmutantVP40LdomainVSVrecombinantsalreadygenerated(BSL 2 setting). Moreover, in collaboration with Dr. G. Olinger (USAMRIID), we will test mature lead candidate inhibitorsfortheirabilitytoblockegressoflive,EBOVandMARVinaBSL4setting(Aim4).

Public Health Relevance

Harty,RonaldN. Filoviruses and arenaviruses cause severe hemorrhagic disease in humans and are potential agents of bioterrorism. No vaccines, nor antiviral drugs are currently available for these NIAID Category A pathogens. Here,wewillusecomputermodeling,functionalbuddingassays,andliveimagingtechniquestoidentifyand validatecandidate,hostorientedtherapeuticstoblockvirusbuddingandspread.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Exploratory/Developmental Grants Phase II (R33)
Project #
5R33AI102104-05
Application #
9088302
Study Section
Special Emphasis Panel (NSS)
Program Officer
Davis, Mindy I
Project Start
2012-07-01
Project End
2017-06-30
Budget Start
2016-07-01
Budget End
2017-06-30
Support Year
5
Fiscal Year
2016
Total Cost
Indirect Cost
Name
University of Pennsylvania
Department
Type
DUNS #
042250712
City
Philadelphia
State
PA
Country
United States
Zip Code
19104
Han, Ziying; Sagum, Cari A; Takizawa, Fumio et al. (2017) Ubiquitin Ligase WWP1 Interacts with Ebola Virus VP40 To Regulate Egress. J Virol 91:
Han, Ziying; Sagum, Cari A; Bedford, Mark T et al. (2016) ITCH E3 Ubiquitin Ligase Interacts with Ebola Virus VP40 To Regulate Budding. J Virol 90:9163-71
Han, Ziying; Bart, Stephen M; Ruthel, Gordon et al. (2016) Ebola virus mediated infectivity is restricted in canine and feline cells. Vet Microbiol 182:102-7
Loughran, H Marie; Han, Ziying; Wrobel, Jay E et al. (2016) Quinoxaline-based inhibitors of Ebola and Marburg VP40 egress. Bioorg Med Chem Lett 26:3429-35
Madara, Jonathan J; Han, Ziying; Ruthel, Gordon et al. (2015) The multifunctional Ebola virus VP40 matrix protein is a promising therapeutic target. Future Virol 10:537-546
Okumura, Atsushi; Rasmussen, Angela L; Halfmann, Peter et al. (2015) Suppressor of Cytokine Signaling 3 Is an Inducible Host Factor That Regulates Virus Egress during Ebola Virus Infection. J Virol 89:10399-406
Han, Ziying; Madara, Jonathan J; Herbert, Andrew et al. (2015) Calcium Regulation of Hemorrhagic Fever Virus Budding: Mechanistic Implications for Host-Oriented Therapeutic Intervention. PLoS Pathog 11:e1005220
Han, Ziying; Madara, Jonathan J; Liu, Yuliang et al. (2015) ALIX Rescues Budding of a Double PTAP/PPEY L-Domain Deletion Mutant of Ebola VP40: A Role for ALIX in Ebola Virus Egress. J Infect Dis 212 Suppl 2:S138-45
Lu, Jianhong; Han, Ziying; Liu, Yuliang et al. (2014) A host-oriented inhibitor of Junin Argentine hemorrhagic fever virus egress. J Virol 88:4736-43
Han, Ziying; Lu, Jianhong; Liu, Yuliang et al. (2014) Small-molecule probes targeting the viral PPxY-host Nedd4 interface block egress of a broad range of RNA viruses. J Virol 88:7294-306