66
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Selection of an HLA-C*03:04-Restricted HIV-1 p24 Gag Sequence Variant Is Associated with Viral Escape from KIR2DL3+ Natural Killer Cells: Data from an Observational Cohort in South Africa

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Background

          Viruses can evade immune surveillance, but the underlying mechanisms are insufficiently understood. Here, we sought to understand the mechanisms by which natural killer (NK) cells recognize HIV-1-infected cells and how this virus can evade NK-cell-mediated immune pressure.

          Methods and Findings

          Two sequence mutations in p24 Gag associated with the presence of specific KIR/HLA combined genotypes were identified in HIV-1 clade C viruses from a large cohort of infected, untreated individuals in South Africa ( n = 392), suggesting viral escape from KIR+ NK cells through sequence variations within HLA class I—presented epitopes. One sequence polymorphism at position 303 of p24 Gag (T Gag303V), selected for in infected individuals with both KIR2DL3 and HLA-C*03: 04, enabled significantly better binding of the inhibitory KIR2DL3 receptor to HLA-C*03:04-expressing cells presenting this variant epitope compared to the wild-type epitope (wild-type mean 18.01 ± 10.45 standard deviation [SD] and variant mean 44.67 ± 14.42 SD, p = 0.002). Furthermore, activation of primary KIR2DL3+ NK cells from healthy donors in response to HLA-C*03:04+ target cells presenting the variant epitope was significantly reduced in comparison to cells presenting the wild-type sequence (wild-type mean 0.78 ± 0.07 standard error of the mean [SEM] and variant mean 0.63 ± 0.07 SEM, p = 0.012). Structural modeling and surface plasmon resonance of KIR/peptide/HLA interactions in the context of the different viral sequence variants studied supported these results. Future studies will be needed to assess processing and antigen presentation of the investigated HIV-1 epitope in natural infection, and the consequences for viral control.

          Conclusions

          These data provide novel insights into how viruses can evade NK cell immunity through the selection of mutations in HLA-presented epitopes that enhance binding to inhibitory NK cell receptors. Better understanding of the mechanisms by which HIV-1 evades NK-cell-mediated immune pressure and the functional validation of a structural modeling approach will facilitate the development of novel targeted immune interventions to harness the antiviral activities of NK cells.

          Abstract

          An analysis from a cohort in South Africa reveals how the HIV virus may escape NK cell immunity by acquiring mutations in HLA-mediated epitopes, which affect binding to NK cell receptors.

          Editors' Summary

          Background

          Throughout life, our immune system—a complex network of cells, tissues, and organs—protects us from attack by viruses, bacteria, parasites, and fungi. The body’s first line of defense against these “pathogens” is the innate immune system, a collection of cells and proteins that is always ready to identify and kill a wide range of foreign invaders. As well as directly killing pathogens, the innate immune system activates the adaptive immune response, which recognizes and kills specific pathogens and is responsible for immunological memory. Most pathogens are dispatched quickly and effectively by the two arms of the immune system, but some infectious agents have found ways to evade the immune response. For example, infection with HIV-1, the virus that causes AIDS, results in prolonged, continuous viral replication even though the human body mounts a vigorous HIV-1-specific immune response. In large part, HIV-1’s evasion of the immune response reflects its ability to kill virus-specific CD4 lymphocytes, which are needed to help other immune system cells kill HIV-1-infected cells. In addition, the proteins on the surface of HIV-1 that are recognized by the human immune system (viral antigens) frequently acquire changes (mutations) that make it harder for the immune system to clear HIV-1 from the human body.

          Why Was This Study Done?

          Viruses evade immune surveillance in many ways, and if we understood the mechanisms underlying immune evasion better, it might be possible to develop targeted immune interventions to deal with viruses such as HIV-1. Here, the researchers investigate how natural killer (NK) cells, a type of lymphocyte that is an important component of the innate antiviral immune response, recognize HIV-infected cells and how HIV-1 evades NK-cell-mediated immune pressure. NK cell activation is determined by the integration of inhibitory and activating signals delivered to the cells by several different receptor families, including the family of killer-cell immunoglobulin-like receptors (KIRs). KIRs mainly bind to human leukocyte antigen (HLA) class I molecules (ligands) on their target cells. HLA class I proteins display fragments (epitopes; peptides recognized by the immune system) of pathogens present in infected cells on the cell surface so that the immune system knows that that cell needs destroying. The binding of distinct KIRs to HLA class I ligands depends on both the sequence of the HLA class I molecule and the sequence of the epitope presented by that HLA class I molecule. Thus, the researchers hypothesized that HIV-1 might evade NK-cell-mediated immune surveillance by acquiring mutations within epitopes presented by HLA class I molecules that enhance the engagement of inhibitory KIRs on NK cells, thereby inhibiting NK cell activity.

          What Did the Researchers Do and Find?

          To investigate this model, the researchers asked whether any polymorphisms (naturally occurring genetic variations) in the HIV-1 gene encoding the p24 Gag protein were selected on a population level in HIV-1-infected individuals expressing specific combinations of KIRs and HLA class I ligands. Using statistical methods to identify KIR/HLA combined genotypes in a large group of untreated HIV-1-infected individuals from South Africa, they showed that a specific sequence polymorphism in p24 Gag was selected for in individuals expressing both HLA-C*03: 04 and KIR2DL3. Functional studies showed that the selection of this variant HIV-1 epitope resulted in better binding of KIR2DL3, an inhibitory KIR, to HLA-C*03:04 than the wild-type epitope. Moreover, the activation of KIR2DL3-positive NK cells from healthy donors in response to HLA-C*03:04-positive target cells presenting the variant epitope was significantly reduced compared to the activation of KIR2DL3-positive NK cells in response to target cells presenting the wild-type epitope.

          What Do These Findings Mean?

          Further studies are needed to assess the consequences of this and other viral sequence variants for viral fitness, the processing and presentation of the mutant epitope during natural infections, and the control of HIV-1 replication in patients. However, these findings provide new insights into how HIV-1 (and possibly other viruses that have a high mutation rate) might evade NK cell immunity through the selection of mutations in HLA-presented epitopes that enhance the binding of inhibitory KIRs to HLA class I/peptide complexes. A better understanding of this molecular mechanism for evasion of immune surveillance should facilitate the development of targeted immune interventions (for example, the use of KIR-blocking antibodies, some of which are already being clinically tested for the treatment of cancer) to maximize the antiviral activities of NK cells.

          Additional Information

          This list of resources contains links that can be accessed when viewing the PDF on a device or via the online version of the article at http://dx.doi.org/10.1371/journal.pmed.1001900.

          Related collections

          Most cited references55

          • Record: found
          • Abstract: found
          • Article: not found

          Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1.

          Allotypes of the natural killer (NK) cell receptor KIR3DL1 vary in both NK cell expression patterns and inhibitory capacity upon binding to their ligands, HLA-B Bw4 molecules, present on target cells. Using a sample size of over 1,500 human immunodeficiency virus (HIV)+ individuals, we show that various distinct allelic combinations of the KIR3DL1 and HLA-B loci significantly and strongly influence both AIDS progression and plasma HIV RNA abundance in a consistent manner. These genetic data correlate very well with previously defined functional differences that distinguish KIR3DL1 allotypes. The various epistatic effects observed here for common, distinct KIR3DL1 and HLA-B Bw4 combinations are unprecedented with regard to any pair of genetic loci in human disease, and indicate that NK cells may have a critical role in the natural history of HIV infection.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The selective downregulation of class I major histocompatibility complex proteins by HIV-1 protects HIV-infected cells from NK cells.

            To avoid detection by CTL, HIV encodes mechanisms for removal of class I MHC proteins from the surface of infected cells. However, class I downregulation potentially exposes the virus-infected cell to attack by NK cells. Human lymphoid cells are protected from NK cell cytotoxicity primarily by HLA-C and HLA-E. We present evidence that HIV-1 selectively downregulates HLA-A and HLA-B but does not significantly affect HLA-C or HLA-E. We then identify the residues in HLA-C and HLA-E that protect them from HIV down-regulation. This selective downregulation allows HIV-infected cells to avoid NK cell-mediated lysis and may represent for HIV a balance between escape from CTL and maintenance of protection from NK cells. These results suggest that subpopulations of CTL and NK cells may be uniquely suited for combating HIV.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Late escape from an immunodominant cytotoxic T-lymphocyte response associated with progression to AIDS.

              The precise role played by HIV-specific cytotoxic T lymphocytes (CTL) in HIV infection remains controversial. Despite strong CTL responses being generated during the asymptomatic phase, the virus persists and AIDS ultimately develops. It has been argued that the virus is so variable, and the virus turnover so great that escape from CTL recognition would occur continually, but so far there is limited evidence for CTL escape. The opposing argument is that evidence for CTL escape is present but hard to find because multiple anti-HIV immune responses are acting simultaneously during the asymptomatic phase of infection. We describe six donors who make a strong CTL response to an immunodominant HLA-B27-restricted epitope. In the two donors who progressed to AIDS, CTL escape to fixation by the same mutation was observed, but only after 9-12 years of epitope stability. CTL escape may play an important role in the pathogenesis of HIV infection.
                Bookmark

                Author and article information

                Contributors
                Role: Academic Editor
                Journal
                PLoS Med
                PLoS Med
                plos
                plosmed
                PLoS Medicine
                Public Library of Science (San Francisco, CA USA )
                1549-1277
                1549-1676
                17 November 2015
                November 2015
                : 12
                : 11
                : e1001900
                Affiliations
                [1 ]Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, United States of America
                [2 ]Heinrich-Pette-Institut, Leibniz Institute for Experimental Virology, Hamburg, Germany
                [3 ]First Department of Internal Medicine, University Medical Center Hamburg—Eppendorf, Hamburg, Germany
                [4 ]HIV Pathogenesis Programme, Doris Duke Medical Research Institute, KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
                [5 ]Computational Biology Center, IBM Thomas J. Watson Research Center, Yorktown Heights, New York, United States of America
                [6 ]Microsoft Research, Los Angeles, California, United States of America
                [7 ]Experimental Immunology, Academic Medical Center, Amsterdam, The Netherlands
                [8 ]Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
                [9 ]Division of Microbiology, New England Primate Research Center, Southborough, Massachusetts, United States of America
                [10 ]Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, Madison, Wisconsin, United States of America
                [11 ]Department of Paediatrics, University of Oxford, Oxford, United Kingdom
                [12 ]Cancer and Inflammation Program, Laboratory of Experimental Immunology, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
                [13 ]First Department of Internal Medicine, Division of Infectious Diseases, University of Cologne, Cologne, Germany
                [14 ]Institute for Medical Microbiology, Immunology and Hygiene, University of Cologne, Cologne, Germany
                [15 ]Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
                [16 ]Max Planck Institute for Infection Biology, Berlin, Germany
                [17 ]KwaZulu-Natal Research Institute for Tuberculosis and HIV, Nelson R. Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
                St. Vincent's Hospital, AUSTRALIA
                Author notes

                PH received travel grants by Abbvie Janssen Cilag and honoraria for speaking at meetings by Janssen Cilag Merck. PH is a member of the Advisory Board “Off label use of otherwise approved drugs in the treatment of HIV-infected patients." MA is a member of the Editorial Board of PLOS Medicine.

                Conceived and designed the experiments: AH CJC WGB NvT AC JC RZ MA. Performed the experiments: AH CJC WGB SK JC. Analyzed the data: JC CJC CK AH CT WGB SK MA AC. Contributed reagents/materials/analysis tools: JS MC PH CK TP DE BW GA. Wrote the first draft of the manuscript: AH MA. Contributed to the writing of the manuscript: AH CJC CT JC MA. Agree with the manuscript’s results and conclusions: AH CT CJC WGB JC NvT JM MJ SK AC CK JS DE GA BW PG MC PH TP RZ TN MA. Enrolled patients: JM MJ CT PG TN. All authors have read, and confirm that they meet, ICMJE criteria for authorship.

                Article
                PMEDICINE-D-14-02557
                10.1371/journal.pmed.1001900
                4648589
                26575988
                fa1f6947-ca06-44b8-8df3-b212d44209d3

                This is an open access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication

                History
                : 12 August 2014
                : 7 October 2015
                Page count
                Figures: 4, Tables: 2, Pages: 27
                Funding
                The work was supported by the Ragon Institute of MGH, MIT and Harvard ( http://www.ragoninstitute.org/index.html), The Heinrich Pette Institute - Leibniz Institute for Experimental Virology ( http://www.hpi-hamburg.de/en/), and the NIH ( http://www.nih.gov/) (R01 AI066031). This project has partly been funded with federal funds from the Frederick National Laboratory for Cancer Research ( http://web.ncifcrf.gov/, contract No. HHSN261200800001E), the Intramural Research Program of the NIH (Frederick National Lab, Center for Cancer Research), and Harvard University Center for AIDS Research (CFAR) ( http://cfar.globalhealth.harvar-d.edu/icb/icb.do). CFAR is an NIH-funded program (P30 AI060354), which is supported by the following NIH Co-Funding and Participating Institutes and Centres: NIAID, NCI, NICHD, NHLBI, NIDA, NIMH, NIA, NCCAM, FIC, and OAR. AH was supported by a German Academic Exchange (DAAD) scholarship ( www.daad.de) and the Koeln Fortune Program ( http://www.medfak.uni-koeln.de/index.php?id=195&L=0). CT was supported by the Wellcome Trust (102468/Z/13/Z). DTE was supported by NIH R01 AI095098. JM was supported by a scholarship from the National Research Foundation. TN holds the South African Research Chair in Systems Biology of HIV/AIDS and is further supported by the Victor Daitz Foundation and an International Early Career Scientist award from the Howard Hughes Medical Institute. The Sinikithemba cohort was funded by the NIH (Grant ROI-AI067073 Contract NOI-AI-15422). PH is funded by the Federal Ministry of Education and Research grant 01KI1017 and supported by DZIF (German Center for Infection Research), www.dzif.de. JMC is an employee of Microsoft Corp; the employer was not involved in the research. The authors have no conflicting financial interests. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Custom metadata
                Clinical data from the South African cohort are stored at the HIV Pathogenesis Programme of the University of KwaZulu-Natal, and are available upon request pending additional approval of the local IRB review committee for patient data release. All Gag-protease sequences obtained in this study are publicly available in the GenBank database under accession numbers HM593106 to HM593510. In accordance with the integrity of data policy of the Heinrich Pette Institute (HPI), all primary data from in vitro experiments have been submitted to the Heinrich Pette Institute (HPI) data repository. Data are available from the HPI data repository upon request for researchers who meet the criteria for access to confidential data. All relevant computational modeling data is available in the Supporting Information files.

                Medicine
                Medicine

                Comments

                Comment on this article