1. Academic Validation
  2. Convergent Evolution of HLA-C Downmodulation in HIV-1 and HIV-2

Convergent Evolution of HLA-C Downmodulation in HIV-1 and HIV-2

  • mBio. 2020 Jul 14;11(4):e00782-20. doi: 10.1128/mBio.00782-20.
Kristina Hopfensperger 1 Jonathan Richard 2 3 Christina M Stürzel 1 Frederic Bibollet-Ruche 4 Richard Apps 5 Marie Leoz 6 Jean-Christophe Plantier 7 Beatrice H Hahn 4 Andrés Finzi 2 3 8 Frank Kirchhoff 1 Daniel Sauter 9
Affiliations

Affiliations

  • 1 Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
  • 2 Centre de Recherche du CHUM, Montreal, Canada.
  • 3 Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Canada.
  • 4 Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
  • 5 NIH Center for Human Immunology, National Institutes of Health, Bethesda, Maryland, USA.
  • 6 Normandie Université, UNIROUEN, UNICAEN, GRAM 2.0, Rouen, France.
  • 7 Normandie Université, UNIROUEN, UNICAEN, GRAM 2.0, Rouen University Hospital, Department of Virology, Laboratory Associated with the National Reference Center on HIV, Rouen, France.
  • 8 Department of Microbiology and Immunology, McGill University, Montreal, Canada.
  • 9 Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany [email protected].
Abstract

HLA-C-mediated antigen presentation induces the killing of human immunodeficiency virus (HIV)-infected CD4+ T cells by cytotoxic T lymphocytes (CTLs). To evade killing, many HIV-1 group M strains decrease HLA-C surface levels using their accessory protein Vpu. However, some HIV-1 group M isolates lack this activity, possibly to prevent the activation of natural killer (NK) cells. Analyzing diverse primate lentiviruses, we found that Vpu-mediated HLA-C downregulation is not limited to pandemic group M but is also found in HIV-1 groups O and P as well as several simian immunodeficiency viruses (SIVs). We show that Vpu targets HLA-C primarily at the protein level, independently of its ability to suppress NF-κB-driven gene expression, and that in some viral lineages, HLA-C downregulation may come at the cost of efficient counteraction of the restriction factor tetherin. Remarkably, HIV-2, which does not carry a vpu gene, uses its accessory protein Vif to decrease HLA-C surface expression. This Vif activity requires intact binding sites for the Cullin5/Elongin ubiquitin ligase complex but is separable from its ability to counteract APOBEC3G. Similar to HIV-1 Vpu, the degree of HIV-2 Vif-mediated HLA-C downregulation varies considerably among different virus isolates. In agreement with opposing selection pressures in vivo, we show that the reduction of HLA-C surface levels by HIV-2 Vif is accompanied by increased NK cell-mediated killing. In summary, our results highlight the complex role of HLA-C in lentiviral infections and demonstrate that HIV-1 and HIV-2 have evolved at least two independent mechanisms to decrease HLA-C levels on infected cells.IMPORTANCE Genome-wide association studies suggest that HLA-C expression is a major determinant of viral load set points and CD4+ T cell counts in HIV-infected individuals. On the one hand, efficient HLA-C expression enables the killing of infected cells by cytotoxic T lymphocytes (CTLs). On the other hand, HLA-C sends inhibitory signals to natural killer (NK) cells and enhances the infectivity of newly produced HIV particles. HIV-1 group M viruses modulate HLA-C expression using the accessory protein Vpu, possibly to balance CTL- and NK cell-mediated immune responses. Here, we show that the second human immunodeficiency virus, HIV-2, can use its accessory protein Vif to evade HLA-C-mediated restriction. Furthermore, our mutational analyses provide insights into the underlying molecular mechanisms. In summary, our results reveal how the two human AIDS viruses modulate HLA-C, a key component of the Antiviral immune response.

Keywords

HIV-1; HIV-2; HLA-C; SIV; Vif; Vpu.

Figures
Products