Current antiretroviral drugs keep HIV replication in check, allowing infected individuals to survive for decades. However, antiretroviral therapy does not clear the virus from the body, requiring life-long drug administration to prevent relapse. Chronic antiretroviral drug exposure contributes to clinical metabolic disturbances, organ damage and drives drug resistance, underscoring the need for new pharmaceutical agents to combat HIV disease. To address these critical issues, we have launched a drug discovery campaign targeting the HIV-1 virulence factor, Nef. Unique to primate lentiviruses, Nef is critical for high-titer HIV-1 replication in vivo, immune escape of HIV-infected cells, and AIDS progression. Using a high- throughput screening assay developed during the previous grant period, we have identified several classes of Nef inhibitors with potent antiretroviral activity against Nef-dependent HIV replication in primary human macrophages. In this renewal application, we propose to explore the molecular mechanism of action of these compounds, and to test their efficacy in vivo with the following Specific Aims:
Aim 1. Leverage new Nef-binding compounds to map hot spots for antagonist binding. We have identified seven discrete chemical scaffolds with nanomolar to single-digit micromolar activity against Nef-dependent enhancement of HIV replication in vitro. In this Aim, we propose to combine X-ray crystallography and site-directed mutagenesis to map their binding sites on Nef. We predict that these diverse scaffolds will bind to a common 'hot spot' for Nef inhibitor action, which will enable structure-based analog design for in vivo testing.
Aim 2. Test the hypothesis that Nef antagonists function by preventing Nef dimerization and interactions with host cell effector proteins. Nef interacts with a diverse group of host cell proteins involved in cell-surface receptor trafficking (e.g., AP-1 and AP-2) and signal transduction (e.g., cytoplasmic protein kinases). Using a cell-based fluorescence complementation assay, we will probe inhibitor impact on Nef homodimerization as well as interactions with a diverse panel of effector proteins, and correlate these effects with Nef functions in HIV-infected cells. These studies will identify the chemical scaffolds that affect as many Nef interactions and functions as possible.
Aim 3. Test the hypothesis that Nef antagonists can suppress HIV replication and T-cell loss in a humanized mouse model of AIDS. Humanized mice (NSG mice reconstituted with human immune cells) infected with Nef-deleted HIV-1 exhibit dramatically lower viral loads and substantially less T cell depletion than mice infected with wild-type virus, validating their use to explore Nef inhibitor action at the whole animal level. We will determine the effect of Nef inhibitors on HIV replication, CD4+ T cell loss and immune system function in this model system. These studies represent the first evaluation of small molecule inhibitors of HIV-1 Nef function in vivo, where they are predicted to suppress Nef-dependent enhancement of HIV replication and CD4+ T cell loss.

Public Health Relevance

While current antiretroviral drugs keep HIV replication in check, they do not clear the virus from the body and require life-long drug administration to prevent relapse. Because chronic antiretroviral drug exposure contributes to clinical metabolic disturbances, organ damage and drives drug resistance, we have developed a drug discovery program targeting HIV-1 Nef as an alternative drug target. Nef, an accessory protein encoded by the virus, boosts HIV replication and promotes immune escape of virally infected cells, contributing to HIV persistence. Inhibitors of Nef function may prevent viral spread in vivo and restore recognition of HIV-infected cells by the patient's own immune system, potentially eradicating the virus.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
2R01AI057083-10
Application #
8923752
Study Section
AIDS Discovery and Development of Therapeutics Study Section (ADDT)
Program Officer
Miller, Roger H
Project Start
2003-07-01
Project End
2020-04-30
Budget Start
2015-05-01
Budget End
2016-04-30
Support Year
10
Fiscal Year
2015
Total Cost
Indirect Cost
Name
University of Pittsburgh
Department
Genetics
Type
Schools of Medicine
DUNS #
004514360
City
Pittsburgh
State
PA
Country
United States
Zip Code
15213
Moroco, Jamie A; Alvarado, John Jeff; Staudt, Ryan P et al. (2018) Remodeling of HIV-1 Nef Structure by Src-Family Kinase Binding. J Mol Biol 430:310-321
Shi, Jing; Xiong, Ran; Zhou, Tao et al. (2018) HIV-1 Nef Antagonizes SERINC5 Restriction by Downregulation of SERINC5 via the Endosome/Lysosome System. J Virol 92:
Wu, Mousheng; Alvarado, John J; Augelli-Szafran, Corinne E et al. (2018) A single ?-octyl glucoside molecule induces HIV-1 Nef dimer formation in the absence of partner protein binding. PLoS One 13:e0192512
Shu, Sherry T; Emert-Sedlak, Lori A; Smithgall, Thomas E (2017) Cell-based Fluorescence Complementation Reveals a Role for HIV-1 Nef Protein Dimerization in AP-2 Adaptor Recruitment and CD4 Co-receptor Down-regulation. J Biol Chem 292:2670-2678
Mujib, Shariq; Saiyed, Aamir; Fadel, Saleh et al. (2017) Pharmacologic HIV-1 Nef blockade promotes CD8 T cell-mediated elimination of latently HIV-1-infected cells in vitro. JCI Insight 2:
Emert-Sedlak, Lori A; Loughran, H Marie; Shi, Haibin et al. (2016) Synthesis and evaluation of orally active small molecule HIV-1 Nef antagonists. Bioorg Med Chem Lett 26:1480-1484
Wales, Thomas E; Poe, Jerrod A; Emert-Sedlak, Lori et al. (2016) Hydrogen Exchange Mass Spectrometry of Related Proteins with Divergent Sequences: A Comparative Study of HIV-1 Nef Allelic Variants. J Am Soc Mass Spectrom 27:1048-61
Wales, Thomas E; Hochrein, James M; Morgan, Christopher R et al. (2015) Subtle Dynamic Changes Accompany Hck Activation by HIV-1 Nef and are Reversed by an Antiretroviral Kinase Inhibitor. Biochemistry 54:6382-91
Bayer, Avraham; Delorme-Axford, Elizabeth; Sleigher, Christie et al. (2015) Human trophoblasts confer resistance to viruses implicated in perinatal infection. Am J Obstet Gynecol 212:71.e1-71.e8
Alvarado, John Jeff; Tarafdar, Sreya; Yeh, Joanne I et al. (2014) Interaction with the Src homology (SH3-SH2) region of the Src-family kinase Hck structures the HIV-1 Nef dimer for kinase activation and effector recruitment. J Biol Chem 289:28539-53

Showing the most recent 10 out of 34 publications