10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Viral Characteristics Associated with the Clinical Nonprogressor Phenotype Are Inherited by Viruses from a Cluster of HIV-1 Elite Controllers

      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

          A small group of HIV-1-infected individuals, called long-term nonprogressors (LTNPs), and in particular a subgroup of LTNPs, elite controllers (LTNP-ECs), display permanent control of viral replication and lack of clinical progression. This control is the result of a complex interaction of host, immune, and viral factors. We identified, by phylogenetic analysis, a cluster of LTNP-ECs infected with very similar low-replication HIV-1 viruses, suggesting the contribution of common viral features to the clinical LTNP-EC phenotype. HIV-1 envelope (Env) glycoprotein mediates signaling and promotes HIV-1 fusion, entry, and infection, being a key factor of viral fitness in vitro, cytopathicity, and infection progression in vivo. Therefore, we isolated full-length env genes from viruses of these patients and from chronically infected control individuals. Functional characterization of the initial events of the viral infection showed that Envs from the LTNP-ECs were ineffective in the binding to CD4 and in the key triggering of actin/tubulin-cytoskeleton modifications compared to Envs from chronic patients. The viral properties of the cluster viruses result in a defective viral fusion, entry, and infection, and these properties were inherited by every virus of the cluster. Therefore, inefficient HIV-1 Env functions and signaling defects may contribute to the low viral replication capacity and transmissibility of the cluster viruses, suggesting a direct role in the LTNP-EC phenotype of these individuals. These results highlight the important role of viral characteristics in the LTNP-EC clinical phenotype. These Env viral properties were common to all the cluster viruses and thus support the heritability of the viral characteristics.

          IMPORTANCE

          HIV-1 long-term nonprogressor elite controller patients, due to their permanent control of viral replication, have been the object of numerous studies to identify the factors responsible for this clinical phenotype. In this work, we analyzed the viral characteristics of the envelopes of viruses from a phylogenetic cluster of LTNP-EC patients. These envelopes showed ineffective binding to CD4 and the subsequent signaling activity to modify actin/tubulin cytoskeletons, which result in low fusion and deficient entry and infection capacities. These Env viral characteristics could explain the nonprogressor clinical phenotype of these patients. In addition, these inefficient env viral properties were present in all viruses of the cluster, supporting the heritability of the viral phenotype.

          Related collections

          Most cited references39

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

          HLA B*5701 is highly associated with restriction of virus replication in a subgroup of HIV-infected long term nonprogressors.

          A unique cohort of HIV-1-infected long term nonprogressors (LTNP) with normal CD4(+) T cell counts and <50 copies/ml of plasma were prospectively recruited for study. HLA typing revealed a dramatic association between the HLA B*5701 class I allele and nonprogressive infection [85% (11 of 13) vs. 9.5% (19 of 200) in progressors; P < 0. 001]. Antigen-specific CD8(+) T cells were enumerated by flow cytometric detection of intracellular IFN-gamma in response to HIV antigens and HLA B*57-gag tetramer staining. No quantitative differences in the total HIV-specific CD8(+) T cell responses were observed between B*57(+) LTNP and five B*57(+) progressors (P = 0.4). Although similar frequencies of peptide specific CD8(+) T cells were also found, the gag-specific CD8(+) T cell response in the LTNP group was highly focused on peptides previously shown to be B*57-restricted. These findings indicate that, within this phenotypically and genotypically distinct cohort, a host immune factor is highly associated with restriction of virus replication and nonprogressive disease. They also strongly suggest a mechanism of virus specific immunity that directly operates through the B*5701 molecule. Further characterization of qualitative differences in the virus-specific responses that distinguish HLA B*57(+) LTNP from progressors may ultimately define mechanisms of effective immune mediated restriction of virus replication.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy.

            J M Coffin (1995)
            Several recent reports indicate that the long, clinically latent phase that characterizes human immunodeficiency virus (HIV) infection of humans is not a period of viral inactivity, but an active process in which cells are being infected and dying at a high rate and in large numbers. These results lead to a simple steady-state model in which infection, cell death, and cell replacement are in balance, and imply that the unique feature of HIV is the extraordinarily large number of replication cycles that occur during infection of a single individual. This turnover drives both the pathogenic process and (even more than mutation rate) the development of genetic variation. This variation includes the inevitable and, in principle, predictable accumulation of mutations such as those conferring resistance to antiviral drugs whose presence before therapy must be considered in the design of therapeutic strategies.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              HIV-1 gp41 fusogenic function triggers autophagy in uninfected cells.

              Cell-expressed HIV-1 envelope glycoproteins (gp120 and gp41, called Env) induce autophagy in uninfected CD4 T cells, leading to their apoptosis, a mechanism most likely contributing to immunodeficiency. The presence of CD4 and CXCR4 on target cells is required for this process, but Env-induced autophagy is independent of CD4 signaling. Here we demonstrate that CXCR4-mediated signaling pathways are not directly involved in autophagy and cell death triggering. Indeed, cells stably expressing mutated forms of CXCR4, unable to transduce different Gi-dependent and -independent signals, still undergo autophagy and cell death after coculture with effector cells expressing Env. After gp120 binding to CD4 and CXCR4, the N terminus fusion peptide (FP) of gp41 is inserted into the target membrane, and gp41 adopts a trimeric extended pre-hairpin intermediate conformation, target of HIV fusion inhibitors such as T20 and C34, before formation of a stable six-helix bundle structure and cell-to-cell fusion. Interestingly, Env-mediated autophagy is triggered in both single cells (hemifusion) and syncytia (complete fusion), and prevented by T20 and C34. The gp41 fusion activity is responsible for Env-mediated autophagy since the Val2Glu mutation in the gp41 FP totally blocks this process. On the contrary, deletion of the C-terminal part of gp41 enhances Env-induced autophagy. These results underline the major role of gp41 in inducing autophagy in the uninfected cells and indicate that the entire process leading to HIV entry into target cells through binding of Env to its receptors, CD4 and CXCR4, is responsible for autophagy and death in the uninfected, bystander cells.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                mBio
                MBio
                mbio
                mbio
                mBio
                mBio
                American Society for Microbiology (1752 N St., N.W., Washington, DC )
                2150-7511
                10 April 2018
                Mar-Apr 2018
                : 9
                : 2
                : e02338-17
                Affiliations
                [a ]Unidad de Virologia Molecular, Laboratorio de Referencia e Investigación en Retrovirus, Centro Nacional de Microbiología (CNM), Instituto de Salud Carlos IIII, Majadahonda, Madrid, Spain
                [b ]Laboratorio de Inmunología Celular y Viral, Unidad de Virología IUETSPC, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna (ULL), Tenerife, Spain
                [c ]Institut de Recerca de la Sida IrsiCaixa, Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
                [d ]Universitat de Vic, Universitat Central de Catalunya, UVIC, Vic, Spain
                [e ]Department of Microbiology and Immunology, Rega Institute, KU Leuven, University of Leuven, Leuven, Belgium
                Medical School, University of Athens
                Author notes
                Address correspondence to Agustín Valenzuela-Fernández, avalenzu@ 123456ull.edu.es , or Cecilio Lopez-Galíndez, clopez@ 123456isciii.es .

                C.C., S.M.-H., D.M.-A., J.B., A.V.-F., and C.L.-G. contributed equally to this article.

                J.B., A.V.-F., and C.L.-G. are co-senior authors.

                Author information
                https://orcid.org/0000-0002-2324-9584
                Article
                mBio02338-17
                10.1128/mBio.02338-17
                5893881
                29636433
                bbf33b67-b20b-49a1-9483-cc69816bda97
                Copyright © 2018 Casado et al.

                This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

                History
                : 15 December 2017
                : 9 March 2018
                Page count
                Figures: 6, Tables: 1, Equations: 0, References: 48, Pages: 18, Words: 10642
                Funding
                Funded by: Ministerio de Economía y Competitividad (MINECO), https://doi.org/10.13039/501100003329;
                Award ID: SAF 2010-17226
                Award ID: SAF 2016-77894-R
                Award ID: FIS PI 13/02269
                Award ID: RIS-RETIC grant RD12/0017/0036
                Award ID: RD12/0016/0028
                Award Recipient : Award Recipient : Award Recipient : Award Recipient :
                Funded by: Ministerio de Economía y Competitividad (MINECO), https://doi.org/10.13039/501100003329;
                Award ID: BIO29 UNLL10-3E-783
                Award ID: SAF2015-64118-R
                Award ID: RIS-RETIC grant RD16/0025/0011
                Award ID: RIS-RETIC RD12/0017/0034
                Award Recipient : Award Recipient : Award Recipient : Award Recipient : Award Recipient : Award Recipient :
                Funded by: Ministerio de Economía y Competitividad (MINECO), https://doi.org/10.13039/501100003329;
                Award ID: FIS PI14/01307
                Award ID: PI17/01518
                Award ID: RD12/0017/0002
                Award ID: RD16/0025/0041
                Award Recipient : Award Recipient : Award Recipient :
                Funded by: Fonds Wetenschappelijk Onderzoek (FWO), https://doi.org/10.13039/501100003130;
                Award ID: No. OT/14/115
                Award ID: G066215N
                Award Recipient :
                Categories
                Research Article
                Custom metadata
                March/April 2018

                Life sciences
                cd4 binding,hiv-1,heritability,ltnp-ec,actin-tubulin modifications,cell signaling,phylogenetic analysis,viral envelope

                Comments

                Comment on this article