1
|
Wang S, Chan KW, Wei D, Ma X, Liu S, Hu G, Park S, Pan R, Gu Y, Nazzari AF, Olia AS, Xu K, Lin BC, Louder MK, McKee K, Doria-Rose NA, Montefiori D, Seaman MS, Zhou T, Kwong PD, Arthos J, Kong XP, Lu S. Human CD4-binding site antibody elicited by polyvalent DNA prime-protein boost vaccine neutralizes cross-clade tier-2-HIV strains. Nat Commun 2024; 15:4301. [PMID: 38773089 PMCID: PMC11109196 DOI: 10.1038/s41467-024-48514-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 05/03/2024] [Indexed: 05/23/2024] Open
Abstract
The vaccine elicitation of HIV tier-2-neutralization antibodies has been a challenge. Here, we report the isolation and characterization of a CD4-binding site (CD4bs) specific monoclonal antibody, HmAb64, from a human volunteer immunized with a polyvalent DNA prime-protein boost HIV vaccine. HmAb64 is derived from heavy chain variable germline gene IGHV1-18 and light chain germline gene IGKV1-39. It has a third heavy chain complementarity-determining region (CDR H3) of 15 amino acids. On a cross-clade panel of 208 HIV-1 pseudo-virus strains, HmAb64 neutralized 20 (10%), including tier-2 strains from clades B, BC, C, and G. The cryo-EM structure of the antigen-binding fragment of HmAb64 in complex with a CNE40 SOSIP trimer revealed details of its recognition; HmAb64 uses both heavy and light CDR3s to recognize the CD4-binding loop, a critical component of the CD4bs. This study demonstrates that a gp120-based vaccine can elicit antibodies capable of tier 2-HIV neutralization.
Collapse
Affiliation(s)
- Shixia Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Kun-Wei Chan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Danlan Wei
- Laboratory of Immune Regulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Xiuwen Ma
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Shuying Liu
- SYL Consulting, Thousand Oak, CA, 91320, USA
| | - Guangnan Hu
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Saeyoung Park
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Ying Gu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Alexandra F Nazzari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Kai Xu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Mark K Louder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Krisha McKee
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | | | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02115, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - James Arthos
- Laboratory of Immune Regulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Shan Lu
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01655, USA.
| |
Collapse
|
2
|
Van Ryk D, Vimonpatranon S, Hiatt J, Ganesan S, Chen N, McMurry J, Garba S, Min S, Goes LR, Girard A, Yolitz J, Licavoli I, Wei D, Huang D, Soares MA, Martinelli E, Cicala C, Arthos J. The V2 domain of HIV gp120 mimics an interaction between CD4 and integrin ⍺4β7. PLoS Pathog 2023; 19:e1011860. [PMID: 38064524 PMCID: PMC10732398 DOI: 10.1371/journal.ppat.1011860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/20/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023] Open
Abstract
The CD4 receptor, by stabilizing TCR-MHC II interactions, plays a central role in adaptive immunity. It also serves as the HIV docking receptor. The HIV gp120 envelope protein binds directly to CD4. This interaction is a prerequisite for viral entry. gp120 also binds to ⍺4β7, an integrin that is expressed on a subset of memory CD4+ T cells. HIV tropisms for CD4+ T cells and gut tissues are central features of HIV pathogenesis. We report that CD4 binds directly to ⍺4β7 in a dynamic way, consistent with a cis regulatory interaction. The molecular details of this interaction are related to the way in which gp120 interacts with both receptors. Like MAdCAM-1 and VCAM-1, two recognized ligands of ⍺4β7, the binding interface on CD4 includes 2 sites (1° and accessory), distributed across its two N-terminal IgSF domains (D1 and D2). The 1° site includes a sequence in the G β-strand of CD4 D2, KIDIV, that binds directly to ⍺4β7. This pentapeptide sequence occurs infrequently in eukaryotic proteins. However, a closely related and conserved sequence, KLDIV, appears in the V2 domain of gp120. KLDIV mediates gp120-⍺4β7 binding. The accessory ⍺4β7 binding site on CD4 includes Phe43. The Phe43 aromatic ring protrudes outward from one edge of a loop connecting the C'C" strands of CD4 D1. Phe43 is a principal contact for HIV gp120. It interacts with conserved residues in the recessed CD4 binding pocket. Substitution of Phe43 abrogates CD4 binding to both gp120 and ⍺4β7. As such, the interactions of gp120 with both CD4 and ⍺4β7 reflect elements of their interactions with each other. These findings indicate that gp120 specificities for CD4 and ⍺4β7 are interrelated and suggest that selective pressures which produced a CD4 tropic virus that replicates in gut tissues are linked to a dynamic interaction between these two receptors.
Collapse
Affiliation(s)
- Donald Van Ryk
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Sinmanus Vimonpatranon
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
- Department of Retrovirology, Armed Forces Research Institute of Medical Sciences–United States Component, Bangkok, Thailand
| | - Joe Hiatt
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Sundar Ganesan
- Biological Imaging Section, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Nathalie Chen
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jordan McMurry
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Saadiq Garba
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Susie Min
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Livia R. Goes
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
- Oncovirology Program, Instituto Nacional de Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alexandre Girard
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Jason Yolitz
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Isabella Licavoli
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Danlan Wei
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Dawei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Marcelo A. Soares
- Oncovirology Program, Instituto Nacional de Câncer, Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Genetics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elena Martinelli
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| |
Collapse
|
3
|
Wang S, Chan KW, Wei D, Ma X, Liu S, Hu G, Park S, Pan R, Gu Y, Nazzari AF, Olia AS, Xu K, Lin BC, Louder MK, Doria-Rose NA, Montefiori D, Seaman MS, Zhou T, Kwong PD, Arthos J, Kong XP, Lu S. Human CD4-Binding Site Antibody Elicited by Polyvalent DNA Prime-Protein Boost Vaccine Neutralizes Cross-Clade Tier-2-HIV Strains. RESEARCH SQUARE 2023:rs.3.rs-3360161. [PMID: 37886518 PMCID: PMC10602183 DOI: 10.21203/rs.3.rs-3360161/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The vaccine elicitation of HIV-neutralizing antibodies with tier-2-neutralization breadth has been a challenge. Here, we report the isolation and characteristics of a CD4-binding site specific monoclonal antibody, HmAb64, from a human volunteer immunized with a polyvalent gp120 DNA prime-protein boost vaccine. HmAb64 derived from heavy chain variable germline gene IGHV1-18, light chain germline gene IGKV1-39, and had a 3rd heavy chain complementarity determining region (CDR H3) of 15 amino acids. On a cross-clade panel of 208 HIV-1 pseudo-virus strains, HmAb64 neutralized 21 (10%), including tier-2 neutralization resistant strains from clades B, BC, C, and G. The cryo-EM structure of the antigen-binding fragment of HmAb64 bound to a conformation between prefusion closed and occluded open forms of envelope trimer, using both heavy and light CDR3s to recognize the CD4-binding loop, a critical component of the CD4-binding site. A gp120 subunit-based vaccine can thus elicit an antibody capable of tier 2-HIV neutralization.
Collapse
Affiliation(s)
- Shixia Wang
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Kun-Wei Chan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Danlan Wei
- Laboratory of Immune Regulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Xiuwen Ma
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | | | - Guangnan Hu
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Saeyoung Park
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ying Gu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Alexandra F Nazzari
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Adam S Olia
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Kai Xu
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Bob C Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Mark K Louder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Nicole A Doria-Rose
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | | | - Michael S Seaman
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Peter D Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - James Arthos
- Laboratory of Immune Regulation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shan Lu
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
4
|
Fernandes RA, Perez-Andres M, Blanco E, Jara-Acevedo M, Criado I, Almeida J, Botafogo V, Coutinho I, Paiva A, van Dongen JJM, Orfao A, Faria E. Complete Multilineage CD4 Expression Defect Associated With Warts Due to an Inherited Homozygous CD4 Gene Mutation. Front Immunol 2019; 10:2502. [PMID: 31781092 PMCID: PMC6856949 DOI: 10.3389/fimmu.2019.02502] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Idiopathic T-CD4 lymphocytopenia (ICL) is a rare and heterogeneous syndrome characterized by opportunistic infections due to reduced CD4 T-lymphocytes (<300 cells/μl or <20% T-cells) in the absence of HIV infection and other primary causes of lymphopenia. Molecular testing of ICL has revealed defects in genes not specific to CD4 T-cells, with pleiotropic effects on other cell types. Here we report for the first time an absolute CD4 lymphocytopenia (<0.01 CD4+ T-cells/μl) due to an autosomal recessive CD4 gene mutation that completely abrogates CD4 protein expression on the surface membrane of T-cells, monocytes, and dendritic cells. A 45-year-old female born to consanguineous parents consulted because of exuberant, relapsing, and treatment-refractory warts on her hands and feet since the age of 10 years, in the absence of other recurrent infections or symptoms. Serological studies were negative for severe infections, including HIV 1/2, HTLV-1, and syphilis, but positive for CMV and EBV. Blood analysis showed the absence of CD4+ T-cells (<0.01%) with repeatedly increased counts of B-cells, naïve CD8+ T-lymphocytes, and particularly, CD4/CD8 double-negative (DN) TCRαβ+ TCRγδ- T-cells (30% of T-cells; 400 cells/μl). Flow cytometric staining of CD4 using monoclonal antibodies directed against five different epitopes, located in two different domains of the protein, confirmed no cell surface membrane or intracytoplasmic expression of CD4 on T-cells, monocytes, and dendritic cells but normal soluble CD4 plasma levels. DN T-cells showed a phenotypic and functional profile similar to normal CD4+ T-cells as regards expression of maturation markers, T-helper and T-regulatory chemokine receptors, TCRvβ repertoire, and in vitro cytokine production against polyclonal and antigen-specific stimuli. Sequencing of the CD4 gene revealed a homozygous (splicing) mutation affecting the last bp on intron 7-8, leading to deletion of the juxtamembrane and intracellular domains of the protein and complete abrogation of CD4 expression on the cell membrane. These findings support previous studies in CD4 KO mice suggesting that surrogate DN helper and regulatory T-cells capable of supporting antigen-specific immune responses are produced in the absence of CD4 signaling and point out the need for better understanding the role of CD4 on thymic selection and the immune response.
Collapse
Affiliation(s)
- Rosa Anita Fernandes
- Allergy and Clinical Immunology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Martin Perez-Andres
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Elena Blanco
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Maria Jara-Acevedo
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain.,Sequencing DNA Service, NUCLEUS, University of Salamanca, Salamanca, Spain
| | - Ignacio Criado
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Julia Almeida
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Vitor Botafogo
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Ines Coutinho
- Dermatology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Artur Paiva
- Flow Cytometry Unit-Clinical Pathology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Ciências Biomédicas Laboratoriais, ESTESC-Coimbra Health School, Instituto Politécnico de Coimbra, Coimbra, Portugal.,Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, Coimbra, Portugal
| | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, Netherlands
| | - Alberto Orfao
- Department of Medicine, Cancer Research Centre (IBMCC, USAL-CSIC), Cytometry Service (NUCLEUS), University of Salamanca (USAL), Salamanca, Spain.,Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain.,Biomedical Research Networking Centre on Cancer-CIBER-CIBERONC (CB16/12/00400), Institute of Health Carlos III, Madrid, Spain
| | - Emilia Faria
- Allergy and Clinical Immunology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Bui VC, Nguyen TH. Insights into the interaction of CD4 with anti-CD4 antibodies. Immunobiology 2016; 222:148-154. [PMID: 27773661 DOI: 10.1016/j.imbio.2016.10.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/15/2016] [Indexed: 01/13/2023]
Abstract
Knowledge about the mechanism by which some antibodies can block HIV-1 entry is critical to our understanding of their function and may offer new avenues for controlling the adhesion of HIV-1 to the host cells. While much progress has been made, this mechanism remains unclear. Here, atomic force microscopy, isothermal titration calorimetry (ITC), and circular dichroism spectroscopy were used to measure some biophysical characteristics of the interaction of four-domains (D1-D4) membrane protein CD4 with anti-D3 antibody OKT4 and with HIV-1 entry blocking anti-D1 antibody Leu3a. The results showed that at 37°C they bind with similar binding strength, thermodynamics, and kinetics but with different assembly states. Further analyzing the interactions at different temperatures by ITC showed that binding of CD4 with Leu3a is characteristic for specific hydrophobic binding as well as for protein folding while with OKT4 comes from an extensive additional hydration upon binding and charge-related interactions within the binding site. Comparing these characteristics with those of HIV-1 gp120-CD4 interaction revealed that Leu3a binds to CD4 faster than HIV-1 followed by changing local structure of D1 to which HIV-1 binds leading to a prevention of viral entry.
Collapse
Affiliation(s)
- Van-Chien Bui
- Center for Innovation Competence, Humoral Immune Reactions in Cardiovascular Diseases, University Medicine Greifswald, 17489 Greifswald, Germany; Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Thi-Huong Nguyen
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, 17475 Greifswald, Germany.
| |
Collapse
|
6
|
Cereda PM, Palù G, Rassu M, Toni M, Malwood W, Dettin M, di Bello C. Anti-HIV-1 Activity of CD4 Synthetic Oligopeptides Representative of the Putative gp120 Binding Site. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029100200305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Two CD4 oligopeptides, corresponding to residues (37–53) and (37–55) of the V1 domain of CD4, which recent structural studies propose as the most likely binding site of HIV-1 gp120, have been chemically synthesized by solid-phase techniques, modified by the addition of two side-chain protected cysteines at both termini and purified by HPLC. Their ability to inhibit the infectivity of human immunodeficiency virus type 1 (HIV-1) (HTLV-IIIB, RF and GB8 strains) in different cell lines was monitored by the production of progeny virus, p24 and reverse transcriptase activity in the culture supernatants and by electron microscopy. The results indicated that the peptides inhibited HIV-1 infectivity in a dose-dependent fashion without any detectable cytotoxicity.
Collapse
Affiliation(s)
- P. M. Cereda
- Institute of Microbiology, University of Pavia, Via Brambilla, Italy
| | - G. Palù
- Institute of Microbiology, University of Padova, Via Gabelli, Italy
| | - M. Rassu
- Institute of Microbiology, University of Padova, Via Gabelli, Italy
| | - M. Toni
- Institute of Microbiology, University of Padova, Via Gabelli, Italy
| | - W. Malwood
- National Institute for Medical Research, Mill Hill, London, UK
| | - M. Dettin
- Institute of Industrial Chemistry, University of Padova, Via Marzolo, Italy
| | - C. di Bello
- Institute of Industrial Chemistry, University of Padova, Via Marzolo, Italy
| |
Collapse
|
7
|
Neurath AR, Debnath AK, Strick N, Li YY, Lin K, Jiang S. 3-Hydroxyphthaloyl-β-Lactoglobulin. II. Anti-Human Immunodeficiency Virus Type 1 Activity in in Vitro Environments Relevant to Prevention of Sexual Transmission of the Virus. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029700800208] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
It is anticipated that the rate of sexual transmission of viruses could be substantially decreased by the use of topical chemical barrier methods. Chemical modification of bovine (β-lactoglobulin (β-LG), the major protein of whey, led to the generation of a potent inhibitor (designated 3HP-β-LG) of human immunodeficiency virus type 1 (HIV-1) infection which was also active against herpesviruses. Compounds intended for topical application to prevent sexual transmission of viruses need to maintain their antiviral activity at pH <<7, corresponding to an acidic vaginal environment, and in the presence of seminal fluid. Results presented here show that the binding of 3HP-β-LG to the CD4 receptor for HIV, involved in the anti-HIV-1 activity of this compound, decreases with decreasing pH. The presence of seminal fluid also decreased the binding of 3HP-β-LG to CD4 and diminished the inhibitory effect of the compound on CD4-gp120 binding. 3HP-β-LG was shown to bind Zn++, and the inhibitory effect of seminal fluid could be substantially diminished by chelating Zn++ with ethylenediaminetetraacetate. Saliva had no effect on 3HP-β-LG binding to CD4 or on its interference with gp120-CD4 binding. The decreased 3HP-β-LG-CD4 binding and the concomitant reduction of gp120-CD4 binding inhibition by 3HP-β-LG at low pH and in the presence of seminal fluid could be compensated for by an increase of the 3HP-β-LG concentration and by adding Zn++ chelators to 3HP-β-LG. These results provide a background for the design of 3HP-β-LG formulations for topical use.
Collapse
Affiliation(s)
- AR Neurath
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| | - AK Debnath
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| | - N Strick
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| | - Y-Y Li
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| | - K Lin
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| | - S Jiang
- The Lindsley F Kimball Research Institute of the New York Blood Center, 310 E 67th Street, New York, NY 10021, USA
| |
Collapse
|
8
|
Hyperglycosylated stable core immunogens designed to present the CD4 binding site are preferentially recognized by broadly neutralizing antibodies. J Virol 2014; 88:14002-16. [PMID: 25253346 DOI: 10.1128/jvi.02614-14] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
UNLABELLED The HIV-1 surface envelope glycoprotein (Env) trimer mediates entry into CD4(+) CCR5(+) host cells. Env possesses conserved antigenic determinants, such as the gp120 primary receptor CD4 binding site (CD4bs), a known neutralization target. Env also contains variable regions and protein surfaces occluded within the trimer that elicit nonneutralizing antibodies. Here we engineered additional N-linked glycans onto a cysteine-stabilized gp120 core (0G) deleted of its major variable regions to preferentially expose the conformationally fixed CD4bs. Three, 6, 7, and 10 new NXT/S glycan (G) motifs were engineered into 0G to encode 3G, 6G, 7G, and 10G cores. Following purification, most glycoproteins, except for 10G, were recognized by broadly neutralizing CD4bs-directed antibodies. Gel and glycan mass spectrometry confirmed that additional N-glycans were posttranslationally added to the redesigned cores. Binding kinetics revealed high-affinity recognition by seven broadly neutralizing CD4bs-directed antibodies and low to no binding by non-broadly neutralizing CD4bs-directed antibodies. Rabbits inoculated with the hyperglycosylated cores elicited IgM and IgG responses to each given protein that were similar in their neutralization characteristics to those elicited by parental 0G. Site-specific glycan masking effects were detected in the elicited sera, and the antisera competed with b12 for CD4bs-directed binding specificity. However, the core-elicited sera showed limited neutralization activity. Trimer priming or boosting of the core immunogens elicited tier 1-level neutralization that mapped to both the CD4bs and V3 and appeared to be trimer dependent. Fine mapping at the CD4bs indicated that conformational stabilization of the cores and addition of N-glycans altered the molecular surface of Env sites of vulnerability to neutralizing antibody, suggesting an explanation for why the elicited neutralization was not improved by this rational design strategy. IMPORTANCE Major obstacles to developing an effective HIV-1 vaccine include the variability of the envelope surface glycoproteins and its high-density glycan shield, generated by incorporation of host (human) glycosylation. HIV-1 does harbor highly conserved sites on the exposed envelope protein surface of gp120, one of which is the virus receptor (CD4) binding site. Several broadly neutralizing antibodies elicited from HIV patients do target this gp120 CD4 binding site (CD4bs); however, gp120 immunogens do not elicit broadly neutralizing antibodies. In this study, we targeted the CD4bs by conformational stabilization and additional glycan masking. We used the atomic-level structure to reengineer gp120 cores to preferentially present the cysteine-stabilized CD4bs and to mask (by glycan) nonneutralizing determinants. Importantly, glycan masking did successfully focus antibody responses to the CD4bs; however, the elicited CD4bs-directed antibodies did not neutralize HIV or bind to unmodified gp120, presumably due to the structure-guided modifications of the modified gp120 core.
Collapse
|
9
|
Abstract
The great variability and high glycosylation of gp120 poses a great challenge for the design of a functional immune therapy. The binding region of the CD4 receptor to gp120, however, is well conserved and may constitute a target to limit viral entry and infectivity. Our strategy consists in using a preexisting pool of natural antibodies directed toward the gal(alpha1,3)gal disaccharide and to redirect it to HIV. We here show that using CD4-derived, gp120-binding, synthetic peptides chemically linked to gal(alpha1,3)gal can redirect these natural antibodies and improve the HIV-1 neutralizing activity of the CD4-derived peptides in vitro. Importantly, the binding of the CD4-gal(alpha1,3)gal peptides to HIV-1-infected cells conferred antibody-dependent cellular cytotoxicity after the addition of human sera. Thus, the temporary redirection of naturally occurring antibodies and their biological activities to a new antigen represents a completely new way of targeting a human disease.
Collapse
|
10
|
Kallel C, Masy E, Duthilleul P. [Detection by flow cytometry of T cell subsets secreting IL-2 and UFNy(Gamma): optimalisation for the technic and the establishment of reference values]. ACTA ACUST UNITED AC 2006; 55:222-9. [PMID: 16678358 DOI: 10.1016/j.patbio.2006.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Accepted: 02/24/2006] [Indexed: 11/29/2022]
Abstract
A dysregulation in Th1/Th2 balance has been described for different pathological situations. Knowing the cytokine profile in a given pathology could assist in understanding the disease mechanism and in choosing an immune intervention most effective for the management of this condition. In this work, the production of two Th1 cytokines, IL-2 and IFN- gamma, was analyzed for different T-cell subsets from 20 normal subjects (mean age 33.5 years) and reference values were defined using the flow cytometric analyses. The optimum operating conditions were set as following: mononuclear cells were stimulated with PMA (20 ng/ml) and ionomycin (1 uM) for 6 h in the presence of brefeldin A (10 ug/ml). Cells were fixed with 4% paraformaldehyde and then dually stained, with anti-CD3 or anti-CD4 or anti-CD8 for the membrane and with anticytokine antibody for the intracytoplasma after being permeabilized with 0.5% saponine solution. The frequency determination of cells that produce IL-2 or IFN-gamma revealed large 95% confidence intervals: (CD3-IL-2: 4.60-10.67%, CD8-IL-2: 1.47-23%, CD3-IFN-gamma: 2,97-32,49%, CD4-IFN-gamma: 2.83-21%, CD8-IFN-gamma: 4.60-35.28%). CD4+ lymphocytes produce the majority of IL-2 (85 vs 13% for CD8+). For IFN-gamma, the situation is more balanced, but the CD4+ lymphocytes remain the predominant producer cells (63 vs. 41%).
Collapse
Affiliation(s)
- C Kallel
- Laboratoire de biochimie et d'hématologie, centre hospitalier universitaire Habib-Bourguiba, avenue El-Fardous, 3029 Sfax, Tunisie.
| | | | | |
Collapse
|
11
|
Troadec S, Bès C, Chentouf M, Nguyen B, Briant L, Jacquet C, Chebli K, Pugnière M, Roquet F, Cerutti M, Chardès T. Biological activities on T lymphocytes of a baculovirus-expressed chimeric recombinant IgG1 antibody with specificity for the CDR3-like loop on the D1 domain of the CD4 molecule. Clin Immunol 2006; 119:38-50. [PMID: 16426893 DOI: 10.1016/j.clim.2005.11.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 11/14/2005] [Accepted: 11/22/2005] [Indexed: 12/16/2022]
Abstract
A baculovirus-expressed chimeric recombinant IgG1 (rIgG1) antibody, with Cgamma1 and Ckappa human constant domains, was derived from the murine monoclonal antibody (mAb) 13B8.2, which is specific for the CDR3-like loop of the CD4 molecule and which inhibits HIV-1 replication. Chimeric rIgG1 antibody 13B8.2 blocked, in a dose-dependent manner, antigen presentation through inhibition of subsequent IL-2 secretion by stimulated T cells. The one-way mixed lymphocyte reaction was abrogated by previous addition of baculovirus-produced rIgG1 13B8.2 in the T-cell culture. Anti-proliferative activity of rIgG1 was demonstrated on CD3-activated CD4+ T lymphocytes from healthy donors, such effect being associated with reduced IL-2 secretion of activated T cells. On the other hand, no proliferation inhibition was observed on CD4+ T lymphocytes activated with phorbol ester plus ionomycin, suggesting that rIgG1 13B8.2 preferentially acts on a proximal TCR-induced signaling pathway. Treatment of DBA1/J human CD4-transgenic mice with 100 microg of recombinant antibody for three consecutive days led to in vivo recovery of rIgG1 antibody 13B8.2 both coated on murine T lymphocytes and free in mouse serum, without CD4 depletion or down-modulation. These findings predict that the baculovirus-expressed chimeric rIgG1 anti-CD4 antibody 13B8.2 is a promising candidate for immunotherapy.
Collapse
Affiliation(s)
- Samuel Troadec
- CNRS UMR 5160, Centre de Pharmacologie et Biotechnologie pour la Santé, Faculté de Pharmacie, 15 Avenue Charles Flahault, BP 14491, 34093 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
de Parseval A, Chatterji U, Morris G, Sun P, Olson AJ, Elder JH. Structural mapping of CD134 residues critical for interaction with feline immunodeficiency virus. Nat Struct Mol Biol 2004; 12:60-6. [PMID: 15592478 DOI: 10.1038/nsmb872] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2004] [Accepted: 11/09/2004] [Indexed: 01/02/2023]
Abstract
CD134 is a primary binding receptor for feline immunodeficiency virus (FIV), and with CXCR4 facilitates infection of CD4(+) T cells. Human CD134 fails to support FIV infection. To delineate the regions important for defining virus specificity of CD134, we exchanged domains between human and feline CD134. The binding site for FIV surface glycoprotein (SU) is located in domain 1, in a region distinct from the natural ligand (CD134L)-binding site. Mutagenesis showed that Asp60 and Asp62 are required for interaction with FIV, and modeling studies localized these two residues to the outer edge of domain 1. Substitutions S60D and N62D, in conjunction with H45S, R59G and V64K, imparted both FIV SU binding and receptor function to human CD134. Finally, we demonstrated that soluble CD134 facilitates infection of CD134(-) CXCR4(+) target cells in a manner analogous to CD4 augmentation of HIV infection.
Collapse
Affiliation(s)
- Aymeric de Parseval
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | |
Collapse
|
13
|
Pugach P, Kuhmann SE, Taylor J, Marozsan AJ, Snyder A, Ketas T, Wolinsky SM, Korber BT, Moore JP. The prolonged culture of human immunodeficiency virus type 1 in primary lymphocytes increases its sensitivity to neutralization by soluble CD4. Virology 2004; 321:8-22. [PMID: 15033560 DOI: 10.1016/j.virol.2003.12.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 12/15/2003] [Accepted: 12/15/2003] [Indexed: 10/26/2022]
Abstract
Primary strains of human immunodeficiency virus type 1 (HIV-1) are known to adapt to replication in cell lines in vitro by becoming sensitive to soluble CD4 (sCD4) and neutralizing antibodies (NAb). T-cell lines favor isolation of variants that use CXCR4 as a co-receptor, while primary isolates predominantly use CCR5. We have now studied how a primary R5 isolate, CC1/85, adapts to prolonged replication in primary human peripheral blood mononuclear cells (PBMC). After 19 passages, a variant virus, CCcon.19, had increased sensitivity to both sCD4 and NAb b12 that binds to a CD4-binding site (CD4BS)-associated epitope, but decreased sensitivity to anti-CD4 antibodies. CCcon.19 retains the R5 phenotype, its resistance to other NAbs was unaltered, its sensitivity to various entry inhibitors was unchanged, and its ability to replicate in macrophages was modestly increased. We define CCcon.19 as a primary T-cell adapted (PTCA) variant. Genetic sequence analysis combined with mutagenesis studies on clonal, chimeric viruses derived from CC1/85 and the PTCA variant showed that the most important changes were in the V1/V2 loop structure, one of them involving the loss of an N-linked glycosylation site. Monomeric gp120 proteins expressed from CC1/85 and the PTCA variant did not differ in their affinities for sCD4, suggesting that the structural consequences of the sequence changes were manifested at the level of the native, trimeric Env complex. Overall, the adaptation process probably involves selection for variants with higher CD4 affinity and hence greater fusion efficiency, but this also involves the loss of some resistance to neutralization by agents directed at or near to the CD4BS. The loss of neutralization resistance is of no relevance under in vitro conditions, but NAbs would presumably be a counter-selection pressure against such adaptive changes in vivo, at least when the humoral immune response is intact.
Collapse
Affiliation(s)
- Pavel Pugach
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Bès C, Briant-Longuet L, Cerutti M, Heitz F, Troadec S, Pugnière M, Roquet F, Molina F, Casset F, Bresson D, Péraldi-Roux S, Devauchelle G, Devaux C, Granier C, Chardes T. Mapping the paratope of anti-CD4 recombinant Fab 13B8.2 by combining parallel peptide synthesis and site-directed mutagenesis. J Biol Chem 2003; 278:14265-73. [PMID: 12566463 DOI: 10.1074/jbc.m210694200] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We analyzed antigen-binding residues from the variable domains of anti-CD4 antibody 13B8.2 using the Spot method of parallel peptide synthesis. Sixteen amino acids, defined as Spot critical residues (SCR), were identified on the basis of a 50% decrease in CD4 binding to alanine analogs of reactive peptides. Recombinant Fab 13B8.2 mutants were constructed with alanine residues in place of each of the 16 SCR, expressed in the baculovirus cell system, and purified. CD measurements indicated that the mutated proteins were conformationally intact, with a beta-sheet secondary structure similar to that of wild-type Fab. Compared with the CD4-binding capacity of wild-type Fab 13B8.2, 11 light (Y32-L, W35-L, Y36-L, H91-L, and Y92-L) and heavy chain (H35-H, R38-H, W52-H, R53-H, F100K-H, and W103-H) Fab single mutants showed a decrease in CD4 recognition as demonstrated by enzyme-linked immunosorbent assay, BIAcore, and flow cytometry analyses. The five remaining Fab mutants showed antigen-binding properties similar to those of wild-type Fab. Recombinant Fab mutants that showed decreased CD4 binding also lost their capacity to inhibit human immunodeficiency virus promoter activation and the antigen-presenting ability that wild-type Fab displays. Molecular modeling of the 13B8.2 antibody paratope indicated that most of these critical residues are appropriately positioned inside the putative CD4-binding pocket, whereas the five SCR that were not confirmed by mutagenesis show an unfavorable positioning. Taken together, these results indicate that most of the residues defined by the Spot method as critical matched with important residues defined by mutagenesis in the whole protein context. The identification of critical residues for CD4 binding in the paratope of anti-CD4 recombinant Fab 13B8.2 provides the opportunity for the generation of improved anti-CD4 molecules with more efficient pharmacological properties.
Collapse
Affiliation(s)
- Cédric Bès
- CNRS UMR 5094, Institut de Biotechnologie et Pharmacologie, Faculté de Pharmacie, 15 Avenue Charles Flahault, 34093 Montpellier Cedex 5, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Fouts T, Godfrey K, Bobb K, Montefiori D, Hanson CV, Kalyanaraman VS, DeVico A, Pal R. Crosslinked HIV-1 envelope-CD4 receptor complexes elicit broadly cross-reactive neutralizing antibodies in rhesus macaques. Proc Natl Acad Sci U S A 2002; 99:11842-7. [PMID: 12192089 PMCID: PMC129356 DOI: 10.1073/pnas.182412199] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The identification of HIV envelope structures that generate broadly cross-reactive neutralizing antibodies is a major goal for HIV-vaccine development. In this study, we evaluated one such structure, expressed as either a gp120-CD4 or a gp140-CD4 complex, for its ability to elicit a neutralizing antibody response. In rhesus macaques, covalently crosslinked complexes of soluble human CD4 (shCD4) and HIV-1(IIIB) envelope glycoproteins (gp120 or gp140) generated antibodies that neutralized a wide range of primary HIV-1 isolates regardless of the coreceptor usage or genetic subtype. Ig with cross-reactive neutralizing activity was recovered by affinity chromatography with a chimeric single-chain polypeptide containing sequences for HIV(BaL) gp120 and a mimetic peptide that induces a CD4-triggered envelope structure. These results suggest that covalently crosslinked complexes of the HIV-1 surface envelope glycoprotein and CD4 elicit broadly neutralizing humoral responses that, in part, may be directed against a novel epitope(s) found on the HIV-1 envelope.
Collapse
Affiliation(s)
- Timothy Fouts
- Institute of Human Virology, University of Maryland Biotechnology Institute, 725 West Lombard Street, Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Mandy F, Nicholson J, Autran B, Janossy G. T-cell subset counting and the fight against AIDS: reflections over a 20-year struggle. CYTOMETRY 2002; 50:39-45. [PMID: 12116344 DOI: 10.1002/cyto.10097] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The story of T-lymphocyte subset immunophenotyping technology is reviewed on the occasion of the 20th anniversary of CD4 T-cell enumeration. Over time, immunophenotyping has evolved into precise, reliable, but complicated and expensive technology requiring fresh blood samples. The gating technologies that were universally adapted for clinical flow cytometry for the past decade relied on rapidly deteriorating morphological scatter characteristics of leukocytes. This special issue dedicated to CD4 T-cell enumeration features most of the available new options that will have a significant impact on how this technology will be implemented within the first decade of the 21st century. In a series of original publications, including the new NIH guideline for T-cell subset enumeration, contemporary gating protocols that use immunologically logical parameters are presented as part of the more reliable and affordable immunophenotyping alternative. Some of the improvements addressed here include the costs of the assays and the capacity to monitor interlaboratory and intralaboratory performances. It is clear that an effective attack on the human immunodeficiency virus (HIV) epidemic has to embrace resource-poor regions. Reducing the cost of the assay while improving reliability and durability is a move in the right direction.
Collapse
Affiliation(s)
- Francis Mandy
- National HIV Immunology Laboratory, Health Canada, LCDC 0603B1, Tunney's Pasture, Ottawa, Ontario K1A 0L2, Canada.
| | | | | | | |
Collapse
|
17
|
Guillon C, Schutten M, Boers PHM, Gruters RA, Osterhaus ADME. Antibody-mediated enhancement of human immunodeficiency virus type 1 infectivity is determined by the structure of gp120 and depends on modulation of the gp120-CCR5 interaction. J Virol 2002; 76:2827-34. [PMID: 11861850 PMCID: PMC135957 DOI: 10.1128/jvi.76.6.2827-2834.2002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In this study, we characterized the viral determinants of coreceptor usage in relation to susceptibility to antibody-mediated neutralization or enhancement of infectivity by using chimeras of three highly related human immunodeficiency virus type 1 (HIV-1) isolates of different phenotypes. We found that the V3 region was the main determinant of antibody-mediated enhancement and coreceptor specificity but that the overall structure of gp120 was also important for these properties. Constructs susceptible to antibody-mediated enhancement preferentially use CCR5 as a coreceptor, in contrast to constructs that were neutralized or not affected. Using monoclonal antibodies directed against CD4 or CCR5, we were able to show that antibody-mediated enhancement was CD4 dependent. Altogether, our results suggest that the modulation of the interaction of gp120 with CCR5 is the mechanism underlying antibody-mediated enhancement of HIV-1 infectivity.
Collapse
Affiliation(s)
- Christophe Guillon
- Department of Virology, Erasmus University Rotterdam, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | |
Collapse
|
18
|
Choi YH, Rho WS, Kim ND, Park SJ, Shin DH, Kim JW, Im SH, Won HS, Lee CW, Chae CB, Sung YC. Short peptides with induced beta-turn inhibit the interaction between HIV-1 gp120 and CD4. J Med Chem 2001; 44:1356-63. [PMID: 11311058 DOI: 10.1021/jm000403+] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
To identify novel peptides that inhibit the interaction between human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120 and CD4, we constructed a targeted phage-displayed peptide library in which phenylalanine and proline were fixed at the fourth and sixth positions, respectively, because Phe43 and the adjacent beta-turn of CD4 are critical for interaction with gp120. Two synthetic peptides were selected after three rounds of biopanning against gp120, and one of them, G1 peptide (ARQPSFDLQCGF), exhibited specific inhibition of the interaction between gp120 and CD4 with an IC(50) of about 50 microM. Structural analysis using NMR demonstrated that G1 peptide forms a compact cyclic structure similar to the CD4 region interacting with gp120. Two derivatives of G1 peptide, a linear hexameric peptide (G1-6) and a cyclic nonameric peptide (G1-c), were synthesized based on the structure of the G1 peptide. Interestingly, they showed higher inhibitory activities than did G1 peptide with IC(50)'s of 6 and 1 microM, respectively. Thus, this study might provide a new insight into the development of anti-HIV-1 inhibitors.
Collapse
Affiliation(s)
- Y H Choi
- Department of Molecular Life Science, Pohang University of Science and Technology, Pohang, Kyungbuk, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Briant L, Devaux C. Bioactive CD4 ligands as pre- and/or postbinding inhibitors of HIV-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2001; 48:373-407. [PMID: 10987097 DOI: 10.1016/s1054-3589(00)48012-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- L Briant
- Laboratoire Infections Rétrovirales et Signalisation Cellulaire, CNRS EP 2104, Montpellier, France
| | | |
Collapse
|
20
|
Esser U, Speck RF, Deen KC, Atchison RE, Sweet R, Goldsmith MA. Molecular function of the CD4 D1 domain in coreceptor-mediated entry by HIV type 1. AIDS Res Hum Retroviruses 2000; 16:1845-54. [PMID: 11118070 DOI: 10.1089/08892220050195801] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The surface molecule CD4 plays a key role in initiating cellular entry by the human immunodeficiency virus type 1 (HIV-1), and it is now recognized as acting synergistically with select chemokine receptors (coreceptors) in the infection process. The present study was undertaken to determine whether the extracellular region of CD4 is sufficient to induce fusion of HIV-1 virions with target cells in the absence of its anchoring function. Using pseudotype reporter viruses to quantitate infection, soluble CD4 (sCD4) was tested for its ability to induce fusion by viruses utilizing CCR5 as their coreceptor. We found that sCD4 was competent to replace membrane-bound CD4 to trigger infection mediated by several HIV-1 envelopes. Furthermore, in a comparison of the envelopes of HIV-1 NL4-3 and a chimera containing the gp120 V3 loop of Ba-L, the V3 region was found to be one factor affecting susceptibility to induction by sCD4. In addition, using truncated and mutant derivatives of sCD4, the amino-terminal D1 domain of CD4 was found to be necessary and sufficient for induction of fusion and to require an intact gp120-binding site for this activity. These results delineate determinants on CD4 and gp120 required for fusion induction in collaboration with a coreceptor, and suggest a mechanism whereby CD4 may contribute to viral infection in trans.
Collapse
Affiliation(s)
- U Esser
- Gladstone Institute of Virology and Immunology, San Francisco, California 94141, USA
| | | | | | | | | | | |
Collapse
|
21
|
Biard-Piechaczyk M, Robert-Hebmann V, Richard V, Roland J, Hipskind RA, Devaux C. Caspase-dependent apoptosis of cells expressing the chemokine receptor CXCR4 is induced by cell membrane-associated human immunodeficiency virus type 1 envelope glycoprotein (gp120). Virology 2000; 268:329-44. [PMID: 10704341 DOI: 10.1006/viro.1999.0151] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Human immunodeficiency virus type 1 (HIV-1) envelope glycoproteins interact with CD4 and chemokine receptors on T cells to deliver signals that trigger either activation, anergy, or apoptosis. However, the molecular mechanisms driving these responses remain poorly understood. In this study we demonstrate that apoptosis is induced upon HIV-1 envelope binding to the chemokine receptor CXCR4. Cells expressing a mutant form of CXCR4 with a C-terminal deletion were also sensitive to HIV-1 envelope-mediated apoptosis, indicating that the cytoplasmic tail of CXCR4 is not required to induce the apoptotic pathway. The specificity of this process was analyzed using several inhibitors of gp120-CD4-CXCR4 interaction. Monoclonal antibodies directed against the gp120-binding site on CD4 (ST4) and against CXCR4 (MAB173) prevented the apoptotic signal in a dose-dependent manner. The cell death program was also inhibited by SDF-1alpha, the natural ligand of CXCR4, and by suramin, a G protein inhibitor that binds with a high affinity to the V3 loop of HIV-1 gp120 envelope protein. These results highlight the role played by gp120-binding on CXCR4 to trigger programmed cell death. Next, we investigated the intracellular signal involved in gp120-induced apoptosis. This cell death program was insensitive to pertussis toxin and did not involve activation of the stress- and apoptosis-related MAP kinases p38(MAPK) and SAPK/JNK but was inhibited by a broad spectrum caspase inhibitor (z-VAD.fmk) and a relatively selective inhibitor of caspase 3 (z-DEVD.fmk). Altogether, our results demonstrate that HIV induces a caspase-dependent apoptotic signaling pathway through CXCR4.
Collapse
Affiliation(s)
- M Biard-Piechaczyk
- Laboratoire Infections Rétrovirales et Signalisation Cellulaire, CNRS EP 2104, Institut de Biologie, 4 Boulevard Henri IV, Montpellier Cedex, 34060, France
| | | | | | | | | | | |
Collapse
|
22
|
Podolin PL, Webb EF, Reddy M, Truneh A, Griswold DE. Inhibition of contact sensitivity in human CD4+ transgenic mice by human CD4-specific monoclonal antibodies: CD4+ T-cell depletion is not required. Immunology 2000; 99:287-95. [PMID: 10692049 PMCID: PMC2327156 DOI: 10.1046/j.1365-2567.2000.00946.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Clenoliximab and keliximab are monkey/human chimeric monoclonal antibodies (mAbs) of the immunoglobulin G4 (IgG4) and IgG1 isotypes, respectively, that recognize the same epitope on human CD4. The two mAbs possess identical idiotypes and exhibit equal affinities for CD4. Upon administration of these mAbs to mice that express a human CD4 transgene, but not mouse CD4 (HuCD4/Tg mice), clenoliximab and keliximab exhibited similar kinetics of binding to CD4, and induced the same degree of CD4 modulation from the cell surface, although only keliximab mediated CD4+ T-cell depletion. Epicutaneous sensitization and challenge of HuCD4/Tg mice with the hapten oxazolone resulted in a contact sensitivity response characterized by tissue swelling, and the presence of interferon-gamma (IFN-gamma) and interleukin-4 (IL-4) in the local tissue. Administration of a single 2-mg dose of either clenoliximab or keliximab to HuCD4/Tg mice prior to sensitization significantly reduced post-challenge tissue swelling, and levels of IFN-gamma and IL-4, indicating that CD4+ T-cell depletion is not required for anti-CD4 mAb-mediated inhibition of contact sensitivity. Administration of either mAb prior to challenge failed to inhibit the contact sensitivity response, indicating differential sensitivity of the afferent and efferent phases of the response to inhibition by CD4-specific mAbs. Collectively, these data indicate that CD4 functions as a positive regulatory molecule in the contact sensitivity response.
Collapse
Affiliation(s)
- P L Podolin
- Departments of Immunology and Pulmonary Pharmacology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA
| | | | | | | | | |
Collapse
|
23
|
Shearer MH, Dalgleish AG, Chanh TC, Kennedy RC. Idiotype cascades associated with the CD4-HIV glycoprotein 120 interaction: immunization with anti-idiotypic antibodies induces anti-anti-idiotypic responses with anti-CD4 specificity and in vitro neutralizing activity. AIDS Res Hum Retroviruses 2000; 16:77-86. [PMID: 10628819 DOI: 10.1089/088922200309629] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Anti-idiotypic antibodies (Ab-2), which were generated in baboons against a mouse monoclonal antibody specific for the CD4 molecule expressed on human T cells, were used to produce anti-anti-idiotypic antibodies (Ab-3) in mice. This response induced by Ab-2 immunization of BALB/c mice was classified as anti-anti-idiotype (Ab-3) on the basis of the ability of the mouse Ab-3 to (1) specifically bind the baboon Ab-2 preparation, but not irrelevant baboon IgG preparations, (2) inhibit the binding of the anti-CD4 Ab-1 preparation to the baboon Ab-2, and (3) recognize a second baboon Ab-2, along with a rabbit Ab-2 specific for the monoclonal anti-CD4 Ab-1 preparation. The murine Ab-3 response also recognizes the CD4 molecule expressed on a human CD4+ T cell line, as determined by flow cytometry; recognizes the same epitopes on the CD4 molecule as the Ab-1; inhibits HIV-1 syncytium formation; and neutralizes HIV-1 primary isolates in vitro. These studies suggest that Ab-3 responses can be induced by anti-Id immunization, which serologically mimicks the antigen and Id specificities of the monoclonal anti-CD4 preparation used to generate the anti-Id. Thus, the Ab-3 response exhibits the characteristics of a population that represents the internal image of the Ab-1.
Collapse
Affiliation(s)
- M H Shearer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City 73104, USA
| | | | | | | |
Collapse
|
24
|
Osterhaus AD, Pedersen N, van Amerongen G, Frankenhuis MT, Marthas M, Reay E, Rose TM, Pamungkas J, Bosch ML. Isolation and partial characterization of a lentivirus from talapoin monkeys (Myopithecus talapoin). Virology 1999; 260:116-24. [PMID: 10405363 DOI: 10.1006/viro.1999.9794] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have identified a novel lentivirus prevalent in talapoin monkeys (Myopithecus talapoin), extending previous observations of human immunodeficiency virus-1 cross-reactive antibodies in the serum of these monkeys. We obtained a virus isolate from one of three seropositive monkeys initially available to us. The virus was tentatively named simian immunodeficiency virus from talapoin monkeys (SIVtal). Despite the difficulty of isolating this virus, it was readily passed between monkeys in captivity through unknown routes of transmission. The virus could be propagated for short terms in peripheral blood mononuclear cells of talapoin monkeys but not in human peripheral blood mononuclear cells or human T cell lines. The propagated virus was used to infect a naive talapoin monkey, four rhesus macaques (M. mulatta), and two cynomolgus macaques (M. fascicularis). All animals seroconverted and virus could be reisolated during a short period after experimental infection. A survey of SIVtal-infected captive talapoin monkeys revealed a relative decrease in CD4(+) cell numbers in chronically (>2 years) infected animals. No other signs of immunodeficiency were observed in any of the infected animals. PCR amplification followed by DNA sequencing of two fragments of the polymerase gene revealed that SIVtal is different from the presently known lentiviruses and perhaps most related to the SIV from Sykes monkeys.
Collapse
Affiliation(s)
- A D Osterhaus
- Laboratory of Immunobiology, National Institute for Public Health and Environmental Protection, Bilthoven, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Ferrer M, Harrison SC. Peptide ligands to human immunodeficiency virus type 1 gp120 identified from phage display libraries. J Virol 1999; 73:5795-802. [PMID: 10364331 PMCID: PMC112640 DOI: 10.1128/jvi.73.7.5795-5802.1999] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have used phage-displayed peptide libraries to identify novel ligands to the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120. Screening of libraries of random 12-mers, 7-mers, and cyclic 9-mers produced two families of gp120 binding peptides. Members of a family with the prototype sequence RINNIPWSEAMM (peptide 12p1) inhibit the interaction between gp120 and both four-domain soluble CD4 (4dCD4) and monoclonal antibody (MAb) 17b, a neutralizing antibody that covers the chemokine receptor binding surface on gp120. Peptide 12p1 inhibits the interaction of 4dCD4 with gp120 from three different HIV strains, implying that it binds to a conserved site on gp120. Members of a second family of peptides, with the prototype sequence TSPYEDWQTYLM (peptide 12p2), bind more weakly to gp120. They do not detectably affect its interaction with 4dCD4, but they enhance its binding to MAb 17b. A common sequence motif in the two peptide families and cross-competition for gp120 binding suggest that they have overlapping contacts. Their divergent effects on the affinity of gp120 for MAb 17b may indicate that their binding stabilizes distinct conformational states of gp120. The functional properties of 12p1 suggest that it might be a useful lead for the development of inhibitors of HIV entry.
Collapse
Affiliation(s)
- M Ferrer
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | | |
Collapse
|
26
|
Moulard M, Mabrouk K, Martin I, Van Rietschoten J, Rochat H, Sabatier JM. V3 loop-derived peptide SPC3 inhibits infection of CD4- and galactosylceramide- cells by LAV-2/B. THE JOURNAL OF PEPTIDE RESEARCH : OFFICIAL JOURNAL OF THE AMERICAN PEPTIDE SOCIETY 1999; 53:647-55. [PMID: 10408339 DOI: 10.1034/j.1399-3011.1999.00062.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
SPC3, a synthetic multibranched peptide including the GPGRAF consensus motif of the human immunodeficiency virus type 1 (HIV-1) gp120 V3-loop is a potent inhibitor of HIV infection of human CD4+ lymphocytes, macrophages and CD4-/galactosylceramide+ human colon epithelial cells and is currently tested in phase II clinical trials (FDA protocol 257 A). The antiviral property of SPC3 was further investigated for its ability to inhibit LAV-2/B, an HIV-2 clone with a CD4-independent tropism. SPC3 inhibited the LAV-2/B-mediated infection of B-cell line which does not express the CD4 and the galactosylceramide molecules on their cell surface, suggesting an SPC3-sensitive CD4/galactosylceramide-independent pathway of viral infection in HIV susceptible cells. The molecular mechanism of the peptide inhibition was also investigated. The data suggested that the SPC3-mediated inhibition does not result from a direct competition between SPC3 and gp120 binding to the cell surface of the target cell.
Collapse
Affiliation(s)
- M Moulard
- Centre d'Immunologie de Marseille Luminy, France.
| | | | | | | | | | | |
Collapse
|
27
|
Andersson H, Kappeler F, Hauri HP. Protein targeting to endoplasmic reticulum by dilysine signals involves direct retention in addition to retrieval. J Biol Chem 1999; 274:15080-4. [PMID: 10329713 DOI: 10.1074/jbc.274.21.15080] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dilysine signals confer localization of type I membrane proteins to the endoplasmic reticulum (ER). According to the prevailing model these signals target proteins to the ER by COP I-mediated retrieval from post-ER compartments, whereas the actual retention mechanism in the ER is unknown. We expressed chimeric membrane proteins with a C-terminal -Lys-Lys-Ala-Ala (KKAA) or -Lys-Lys-Phe-Phe (KKFF) dilysine signal in Lec-1 cells. Unlike KKFF constructs, which had access to post-ER compartments, the KKAA chimeras were localized to the ER by confocal microscopy and were neither processed by cis-Golgi-specific enzymes in vivo nor included into ER-derived transport vesicles in an in vitro budding assay, suggesting that KKAA-bearing proteins are permanently retained in the ER. The ER localization was nonsaturable and exclusively mediated by the dilysine signal because mutating the lysines to alanines led to cell surface expression of the chimeras. Although the KKAA signal avidly binds COP I in vitro, the ER retention by this signal does not depend on intact COP I in vivo because it was not affected in an epsilon-COP-deficient cell line. We propose that dilysine ER targeting signals can mediate ER retention in addition to retrieval.
Collapse
Affiliation(s)
- H Andersson
- Department of Pharmacology, Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | | | | |
Collapse
|
28
|
Mondor I, Ugolini S, Sattentau QJ. Human immunodeficiency virus type 1 attachment to HeLa CD4 cells is CD4 independent and gp120 dependent and requires cell surface heparans. J Virol 1998; 72:3623-34. [PMID: 9557643 PMCID: PMC109583 DOI: 10.1128/jvi.72.5.3623-3634.1998] [Citation(s) in RCA: 246] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The binding of human immunodeficiency virus type 1 (HIV-1) (Hx10) virions to two different cell lines was analyzed by using a novel assay based on the detection, by anti-HLA-DR-specific antibodies, of HLA-DR+ virus binding to HLA-DR- cells. Virion attachment to the CD4+-T-cell line A3.01 was highly CD4 dependent in that it was potently inhibited by CD4 monoclonal antibodies (MAbs), and little virus binding to the CD4- sister A2.01 line was observed. By contrast, virion binding to HeLa cells expressing moderate or high levels of CD4 was equivalent to, or lower than, binding to wild-type CD4- HeLa cells. Moreover, several CD4 MAbs did not reduce, but enhanced, HIV-1 attachment to HeLa-CD4 cells. CD4 was required for infection of HeLa cells, however, demonstrating a postattachment role for this receptor. MAbs specific for the V2 and V3 loops and the CD4i epitope of gp120 strongly inhibited virion binding to HeLa-CD4 cells, whereas MAbs specific for the CD4bs and the 2G12 epitopes enhanced attachment. Despite this, all gp120- and gp41-specific MAbs tested neutralized infectivity on HeLa-CD4 cells. HIV-1 attachment to HeLa cells was only partially inhibited by MAbs specific for adhesion molecules present on the virus or target cells but was completely blocked by polyanions such as heparin, dextran sulfate, and pentosan sulfate. Treatment of HeLa-CD4 cells with heparinases completely eliminated HIV attachment and infection, strongly implicating cell surface heparans in the attachment process. CD4 dependence for HIV-1 attachment to target cells is thus highly cell line specific and may be replaced by other ligand-receptor interactions.
Collapse
Affiliation(s)
- I Mondor
- Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | | |
Collapse
|
29
|
Briant L, Robert-Hebmann V, Sivan V, Brunet A, Pouysségur J, Devaux C. Involvement of Extracellular Signal-Regulated Kinase Module in HIV-Mediated CD4 Signals Controlling Activation of Nuclear Factor-κB and AP-1 Transcription Factors. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.4.1875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
Although the molecular mechanisms by which the HIV-1 triggers either T cell activation, anergy, or apoptosis remain poorly understood, it is well established that the interaction of HIV-1 envelope glycoproteins with cell surface CD4 delivers signals to the target cell, resulting in activation of transcription factors such as NF-κB and AP-1. In this study, we report the first evidence indicating that kinases MEK-1 (MAP kinase/Erk kinase) and ERK-1 (extracellular signal-regulated kinase) act as intermediates in the cascade of events that regulate NF-κB and AP-1 activation upon HIV-1 binding to cell surface CD4. We found that CEM cells transfected with dominant negative forms of MEK-1 or ERK-1 do not display NF-κB activation after HIV-1 binding to CD4. In contrast, NF-κB activation was observed in these cells after PMA stimulation. Although the different cell lines studied expressed similar amounts of CD4 and p56lck, HIV-1 replication and HIV-1-induced apoptosis were slightly delayed in cells expressing dominant negative forms of MEK-1 or ERK-1 compared with parental CEM cells and cells expressing a constitutively active mutant form of MEK-1 or wild-type ERK-1. In light of recently published data, we propose that a positive signal initiated following oligomerization of CD4 by the virus is likely to involve a recruitment of active forms of p56lck, Raf-1, MEK-1, and ERK-1, before AP-1 and NF-κB activation.
Collapse
Affiliation(s)
- Laurence Briant
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | - Véronique Robert-Hebmann
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | - Virginie Sivan
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| | | | | | - Christian Devaux
- *Centre de Recherches de Biochimie Macromoléculaire, Laboratoire d’Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France; and
| |
Collapse
|
30
|
Coudronnière N, David C, Hirn M, Devaux C. In vitro selection of HIV-1 resistant to an anti-CD4 monoclonal antibody that inhibits virus transcription. JOURNAL OF ACQUIRED IMMUNE DEFICIENCY SYNDROMES AND HUMAN RETROVIROLOGY : OFFICIAL PUBLICATION OF THE INTERNATIONAL RETROVIROLOGY ASSOCIATION 1998; 17:17-26. [PMID: 9436754 DOI: 10.1097/00042560-199801010-00003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Phase I studies using monoclonal antibodies (mAbs) that bind to the Ig-CDR3-like loop in domain 1 of CD4 (e.g., 13B8-2 mAb) have already been documented for HIV-1-infected patients. In vitro, such mAbs do not inhibit virus to cell fusion but are able to inhibit virus envelope-mediated syncytia formation. Moreover, these mAbs inhibit Tat-induced activation of HIV-1 promoter and HIV-1 transcription in infected CD4+ cells. Here, we report the selection of escaped mutant virus or viruses derived from HIV-1Lai capable of replicating in vitro in the presence of concentrations of 13B8-2 mAb, that usually inhibit HIV-1Lai particle production. The escaped mutant virus or viruses, termed HIV-1Lai13EM, kept the major enzymatic restriction sites found in HIV-1Lai and remained sensitive to anti-CD4 mAb-, soluble CD4-, and recombinant gp120-mediated inhibition of syncytia formation. Possible genetic changes affecting the tat gene or the 5' long terminal repeat (LTR) were investigated. Partial sequence analysis of HIV-1Lai13EM and a control HIV-1Lai grown for 85 days in CEM cells, demonstrated that the first tat exon of these two viruses encoded identical proteins. Although a point mutation G>A was frequently encountered (6 of 13 sequences) in the LTRs of HIV-1Lai13EM at position -188 within the negative regulatory element (NRE), this mutation did not confer the escape mutant phenotype. Our study indicates that the mutant phenotype probably requires genetic changes in a region or regions outside the LTRs.
Collapse
Affiliation(s)
- N Coudronnière
- Laboratoire d'Immunologie des Infections Rétrovirales, Institut de Biologie, Montpellier, France
| | | | | | | |
Collapse
|
31
|
Attanasio R, Pehler K, Scinicariello F. DNA-based immunization induces anti-CD4 antibodies directed primarily to native epitopes. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 1997; 17:207-15. [PMID: 9143878 DOI: 10.1111/j.1574-695x.1997.tb01014.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
DNA-based immunization is one of the most promising strategies to induce protective immunity against a variety of pathogens, presenting clear advantages as compared to the use of recombinant antigens. One of these advantages might be the ability to induce antibodies directed primarily against conformational determinants, as compared to immunization with recombinant proteins. To test this possibility, we have analyzed the antibody responses induced in mice by immunization with either recombinant soluble CD4 (rCD4) or by immunization with plasmid DNA-encoding CD4 (CD4-DNA). Mice immunized with CD4-DNA had lower titers of antibodies able to recognize rCD4 than mice immunized with rCD4. However, immunization with CD4-DNA induced antibodies reactive with the native cell surface CD4 molecule in all mice, whereas only two out of five mice immunized with rCD4 produced antibodies reactive with cell surface CD4, thus demonstrating that the genetic immunization approach may lead to an antibody response more consistent and superior at a qualitative level as compared to immunization with the corresponding recombinant protein. In addition, differences in the kinetics of appearance of antibodies directed against the native CD4 molecule were observed between mice immunized with CD4-DNA or rCD4. In the first case, antibodies reacting with cell surface CD4 were present 28 days after the first immunization, whereas mice immunized with rCD4 produced antibodies directed against the native molecule only following a booster injection. Finally, the two groups of mice produced antibodies with a different isotype distribution. No clear predominance of a specific IgG subclass was detected in the antibody population produced in response to DNA immunization. Conversely, mice immunized with rCD4 produced predominantly antibodies of the IgG1 isotype, indicating generation of a TH2 response. Together, results from this study indicate that the CD4 molecule endogenously produced following DNA immunization is expressed, at least partially, in a native conformation. This feature confers a major advantage to the DNA immunization approach as compared to immunization with the corresponding recombinant protein, which seems to elicit antibodies predominantly directed to epitopes uniquely expressed on the recombinant molecule.
Collapse
Affiliation(s)
- R Attanasio
- Division of Microbiology and Immunology, Yerkes Regional Primate Research Center, Emory University, Atlanta, GA 30329, USA.
| | | | | |
Collapse
|
32
|
Autiero M, Gaubin M, Mani JC, Castejon C, Martin M, el Marhomy S, Guardiola J, Piatier-Tonneau D. Surface plasmon resonance analysis of gp17, a natural CD4 ligand from human seminal plasma inhibiting human immunodeficiency virus type-1 gp120-mediated syncytium formation. EUROPEAN JOURNAL OF BIOCHEMISTRY 1997; 245:208-13. [PMID: 9128743 DOI: 10.1111/j.1432-1033.1997.00208.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We have previously isolated from human seminal plasma a CD4 ligand, the gp17 glycoprotein, which shares sequence identity with three previously identified proteins: secretory actin-binding protein (SABP) from seminal plasma, gross-cystic-disease fluid protein-15 (GCDFP-15) and prolactin-inducible protein (PIP) from breast tumor cells. Functions of these glycoproteins are unknown. To further characterize the physical interaction between gp17 and CD4 we used surface plasmon resonance and demonstrated that gp17-CD4 binding affinity is high. Competition experiments indicated that gp17 interferes with human immunodeficiency virus (HIV) envelope protein/CD4 binding, although it binds to a site distinct from but close to the gp120-binding site. We observed moreover that gp17 inhibits syncytium formation between transfected cells expressing the wild-type HIV-1 envelope glycoprotein and CD4, respectively. Our results suggest that gp17, which may function as an immunomodulatory CD4-binding factor playing a role at insemination, may also play a role in controlling HIV spread in the sexual tract.
Collapse
Affiliation(s)
- M Autiero
- Unité de Génétique Moléculaire et de Biologie du Développement, UPR 420 CNRS, Villejuif, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Milia E, Di Somma MM, Majolini MB, Ulivieri C, Somma F, Piccolella E, Telford JL, Baldari CT. Gene activating and proapoptotic potential are independent properties of different CD4 epitopes. Mol Immunol 1997; 34:287-96. [PMID: 9244341 DOI: 10.1016/s0161-5890(97)00050-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
CD4 engagement triggers an early signaling cascade which initiates late events such as transcription factor activation. The outcome of CD4 engagement is T-cell commitment to alternative, dramatically different fates, such as activation and apoptosis. We have tested a panel of anti-CD4 mAbs specific for different CD4 epitopes, as well as HIV-1 gp120, for the capacity to activate crucial early events such as enhancement of p56(lck) kinase activity and Shc phosphorylation. The same CD4 epitopes were characterized for their capacity both to deliver a gene activating signal and to program T-cells to activation dependent death. No correlation could be found between capacity of specific CD4 epitopes to deliver a gene activating signal and capacity to prime T-cells to apoptosis, suggesting that gene activating and proapoptotic potential are independent functions of CD4 epitopes. Furthermore, while triggering of the calcium pathway appears critical in NF-AT activation, optimal p56(lck) activation and Shc phosphorylation might be required for initiation of the apoptotic pathway.
Collapse
Affiliation(s)
- E Milia
- Department of Evolutionary Biology, University of Siena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Simon JH, Stumbles P, Signoret N, Somoza C, Puklavec M, Sattentau QJ, Barclay AN, James W. Role of CD4 epitopes outside the gp120-binding site during entry of human immunodeficiency virus type 1. J Virol 1997; 71:1476-84. [PMID: 8995673 PMCID: PMC191204 DOI: 10.1128/jvi.71.2.1476-1484.1997] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
CD4 is the primary receptor for human immunodeficiency virus (HIV). The binding site for the surface glycoprotein of HIV type 1 (HIV-1), gp120, has been mapped to the C'-C" region of domain 1 of CD4. Previously, we have shown that a mutant of rat CD4, in which this region was exchanged for that of human CD4, is able to mediate infection of human cells by HIV-1, suggesting that essential interactions between HIV and CD4 are confined to this region. Our observations appeared to conflict with mutagenesis and antibody studies which implicate regions of CD4 outside the gp120-binding site in postbinding events during viral entry. In order to resolve this issue, we have utilized a panel of anti-rat CD4 monoclonal antibodies in conjunction with the rat-human chimeric CD4 to distinguish sequence-specific from steric effects. We find that several antibodies to rat CD4 inhibit HIV infection in cells expressing the chimeric CD4 and that this is probably due to steric hinderance. In addition, we demonstrate that replacement of the rat CDR3-like region with its human homolog does not increase the affinity of the rat-human chimeric CD4 for gp120 or affect the exposure of gp41 following binding to CD4, providing further evidence that this region does not play a crucial role during entry of virus.
Collapse
Affiliation(s)
- J H Simon
- Sir William Dunn School of Pathology, University of Oxford, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Storage stability of the solution formulation of sCD4 determined by DSC in comparison with two functional assays. ACTA ACUST UNITED AC 1996. [DOI: 10.1007/bf01992848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Signoret N, Blanc-Zouaoui D, Kwong PD, Sattentau QJ. Selective effects of electrostatic changes in the CD4 CDR-3-like loop on infection by different human immunodeficiency virus type 1 isolates. AIDS Res Hum Retroviruses 1996; 12:1001-13. [PMID: 8827216 DOI: 10.1089/aid.1996.12.1001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The role of the CDR-3-like loop of the first domain of the CD4 molecule in infection by the human immunodeficiency virus type 1 (HIV-1) is controversial. In an attempt to determine whether the strong negative charge in the CDR-3-like loop influences HIV-1 infection we have substituted by mutagenesis negative for positively charged residues at position 87/88 and 91/92. These mutations were shown to have no obvious effect on CD4 conformation outside of the CDR-3-like loop. Infection of cells expressing the E87K/D88K substitution mutant resulted in a selective reduction in infectivity for certain HIV-1 viruses compared to cells expressing wile-type CD4. Viruses Hx10, HxB2, and MN were 4- to 13-fold less efficient at infecting the E87K/D88K mutant, whereas SF2, RF, and NDK yielded an efficiency of infection similar to, or slightly greater than, that of the wild type. To investigate the step at which infectivity was selectively reduced, we compared early events in the life cycles of Hx10 and SF2 viruses using PCR entry and gp120-binding assays. Both gp120 binding and virus entry were reduced for Hx10 on the mutant CD4-expressing cells as compared to wild-type CD4-expressing cells, whereas no difference was seen in either assay with SF2. Although relatively small in magnitude, the contribution of the CDR-3-like loop to the overall CD4-gp120 interaction may serve to modify the binding and entry of certain virus isolates.
Collapse
Affiliation(s)
- N Signoret
- Centre d'Immunologie de Marseille-Luminy, France
| | | | | | | |
Collapse
|
37
|
Wieder KJ, Chatis P, Boltax J, Wieder I, Nuovo G, Strom TB. Human immunodeficiency virus type 1 entry into murine cell lines and lymphocytes from transgenic mice expressing a glycoprotein 120-binding mutant mouse CD4. AIDS Res Hum Retroviruses 1996; 12:867-76. [PMID: 8798971 DOI: 10.1089/aid.1996.12.867] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Human CD4, the receptor for the gp120 envelope glycoprotein of HIV-1, is the route for viral entry into CD4+ cells; other cellular factors may cooperate with CD4 to facilitate HIV-1 entry into human cells. Human CD4 expressed on murine cells does not readily mediate HIV-1 entry, which may reflect a functional incompatibility of human CD4 with murine cellular components. We postulated that a HIV-1 gp120-binding mutant murine CD4 (L3T4) possessing a minimal number of human amino acid residues could facilitate HIV-1 entry into rodent cells, unlike human CD4. This hypothesis led us to develop a series of murine L3T4 mutants that bear human CD4 gp120-binding region amino acid residues while retaining most L3T4 epitopes. HeLa cell transfectants expressing gp120-binding mutant L3T4 proteins could be infected with HIV-1. Three mouse cell lines expressing these L3T4 mutant proteins could also be infected with HIV-1 as determined by PCR techniques that detect viral DNA and spliced RNAs. Lectin-stimulated polymorphonuclear leukocytes from transgenic mice (SBL mouse) expressing a gp120-binding L3T4 mutant protein were infected with HIV-1 at the same frequency as lectin-stimulated human peripheral blood lymphocytes as determined by in situ PCR analyses. Supernatant p24gag and reverse transcriptase levels in HIV-infected mouse cell cultures, however, were routinely at background levels, unlike HIV-infected human cell cultures. Thus, gp120-binding mutant L3T4 proteins mediate viral entry in all mouse cells that were tested, but high-level viral replication is absent in these cells.
Collapse
Affiliation(s)
- K J Wieder
- Division of Immunology, Beth Israel Hospital and Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | |
Collapse
|
38
|
Weissenhorn W, Chen YH, Reiter C, Federle C, Weiss EH, Riethmüller G, Rieber EP. Structural diversity of monoclonal CD4 antibodies and their capacity to block the HIV gp120/CD4 interaction. Hybridoma (Larchmt) 1996; 15:117-24. [PMID: 8743291 DOI: 10.1089/hyb.1996.15.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A number of monoclonal antibodies have been raised against CD4, the receptor on T cells for the HIV envelope glycoprotein gp120. In the present paper we describe biological activities and sequence analysis of seven CD4 MAb. Five of these MAb preparations compete with HIV/gp120 for CD4 binding. The sequences of the variable regions for these MAb were determined in order to ascertain any correlation with selective V gene usage. A relationship was found between the expressed variable region genes and the CD4 recognition pattern. The VH genes that are used can be subdivided into two major groups expressing either a VH gene belonging to the J558 family or to the VGam family. The usage of the VL genes varies, indicating that the epitope specificity is predominantly determined by the rearranged VH genes. The distinct cross-reactivity pattern of these MAb also correlates with their capacity to block binding of recombinant gp120 to CD4 in vitro. Although five of these MAb were able to block gp120 binding none of the CDR sequences shows a relevant homology to the gp120 sequence. This indicates a steric hinderence mechanism for blocking gp120 binding and not a direct interaction with the receptor binding site on CD4. The data also confirm the failure of these MAb as a potential target for receptor mimicry.
Collapse
|
39
|
Neurath AR, Jiang S, Strick N, Lin K, Li YY, Debnath AK. Bovine beta-lactoglobulin modified by 3-hydroxyphthalic anhydride blocks the CD4 cell receptor for HIV. Nat Med 1996; 2:230-4. [PMID: 8574970 DOI: 10.1038/nm0296-230] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Sexual transmission is the most frequent (86%) route of adult HIV-1 transmission worldwide. In the absence of a prophylactic anti-HIV vaccine, other methods of preventing infection should be implemented. Virucidal spermicides have been considered for this purpose, but their application is contraindicated by adverse effects. Anti-HIV drugs or virus-neutralizing monoclonal antibodies are expensive, suggesting that their wide use in topical chemoprophylaxis is unlikely. This emphasizes the importance of developing other methods for preventing HIV transmission. The target cells for sexual and mucosal HIV transmission include T lymphocytes, monocytes/macrophages and dendritic cells. Therefore, compounds blocking HIV-CD4 binding are expected to inhibit virus transmission. In exploring the possibility that chemical modification of food proteins might lead to compounds with anti-HIV-1 activity, we found that bovine beta-lactoglobulin (beta-LG) modified by 3-hydroxyphthalic anhydride (3HP-beta-LG) (1) blocked at nanomolar concentrations the binding to CD4 of human (HIV) and simian (SIV) immunodeficiency virus surface glycoproteins and monoclonal antibodies specific for the HIV binding site on CD4 and (2) inhibited infection by HIV-1, including primary virus isolates, by HIV-2 and by SIV. The inexpensive and widely available source (whey) for production of 3HP-beta-LG suggests its potential application (nonparenteral) for diminishing the frequency of HIV transmission.
Collapse
Affiliation(s)
- A R Neurath
- Laboratory of Biochemical Virology, Lindsley F. Kimball Research Institute of the New York Blood Center, New York 10021, USA
| | | | | | | | | | | |
Collapse
|
40
|
Indraccolo S, Mion M, Biagiotti R, Romagnani S, Morfini M, Longo G, Zamarchi R, Chieco-Bianchi L, Amadori A. Genetic variability of the human CD4 V2 domain. Immunogenetics 1996; 44:70-2. [PMID: 8613144 DOI: 10.1007/bf02602658] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Affiliation(s)
- S Indraccolo
- Institute of Oncology, University of Padova, Padova, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Benkirane M, Hirn M, Carrière D, Devaux C. Functional epitope analysis of the human CD4 molecule: antibodies that inhibit human immunodeficiency virus type 1 gene expression bind to the immunoglobulin CDR3-like region of CD4. J Virol 1995; 69:6898-903. [PMID: 7474106 PMCID: PMC189606 DOI: 10.1128/jvi.69.11.6898-6903.1995] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We recently demonstrated that monoclonal antibody (MAb) 13B8-2, specific for the immunoglobulin (Ig) complementary determining region 3 (CDR3)-like region of the CD4 molecule, inhibits viral transcription in human immunodeficiency virus (HIV)-infected CEM cells and HIV type 1 (HIV-1) promoter activity. Here, we have studied the capacity of several MAb specific for the D1 domain of CD4, including anti-CDR2-like (Leu-3a and ST4) and anti-CDR3-like (13B8-2 and ST40) MAb, and for the D2 domain of CD4 (BL4) to inhibit both provirus transcription in HIV-1LAI-infected CEM cells and transcription of the chloramphenicol acetyltransferase (CAT) gene under control of the HIV-1 long terminal repeat in transiently transfected CEM cells. We found that HIV-1 promoter activity and provirus transcription are inhibited only by MAb that bind to the CDR3-like region in domain 1 of CD4. Moreover, we demonstrated that the Fab fragment of an anti-CDR3-like region-specific anti-CD4 MAb is a powerful inhibitor of HIV-1 promoter activity. These results have implications for understanding the role of the CDR3-like region in CD4 T-cell signaling, which controls provirus transcription.
Collapse
Affiliation(s)
- M Benkirane
- Laboratoire d'Immunologie des Infections Rétrovirales, Centre National de la Recherche Scientifique UPR 9008-Institut National de la Santé et de la Recherche Médicale U249, Montpellier, France
| | | | | | | |
Collapse
|
42
|
Neurath AR, Debnath AK, Strick N, Li YY, Lin K, Jiang S. Blocking of CD4 cell receptors for the human immunodeficiency virus type 1 (HIV-1) by chemically modified bovine milk proteins: potential for AIDS prophylaxis. J Mol Recognit 1995; 8:304-16. [PMID: 8619951 DOI: 10.1002/jmr.300080504] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The chemical transformation of synthetic combinatorial libraries to increase the diversity of compounds of medicinal interest was reported recently. Chemical modification of natural products represents a complementary approach to accomplish this aim. Modification of lysines by aromatic acid anhydrides, preferentially by 3-hydroxyphthalic and trimellitic anhydrides and trimellitic anhydride chloride, converted commonly available proteins (human and bovine serum albumin and casein) into potent inhibitors of (i) binding between the HIV-1 gp 120 envelope glycoprotein and the CD4 cell receptor, probably owing to their binding to CD4, and (ii) infection by HIV-1. Modified bovine milk proteins are also potent HIV-1 inhibitors and may have potential for anti-HIV-1 prophylaxis.
Collapse
Affiliation(s)
- A R Neurath
- Laboratory of Biochemical Virology, Lindsley F. Kimball Research Institute of the New York Blood Center, New York 10021, USA
| | | | | | | | | | | |
Collapse
|
43
|
Velge-Roussel F, Breton P, Lescure F, Guillon X, Bout D, Hoebeke J. Analysis of human CD4-antibody interaction using the BIAcore system. J Immunol Methods 1995; 183:141-8. [PMID: 7602132 DOI: 10.1016/0022-1759(95)00042-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Interaction between CD4 cell surface protein and HIV-bearing gp120 has been described as the initial step for HIV entry into host cells. Some anti-CD4 antibodies were shown to inhibit this interaction. Biosensor studies using the BIAcore were performed to determine kinetic and thermodynamic parameters of the interaction of one of these antibodies (i.e. IOT4a, clone 13B8-2) with immobilized recombinant soluble CD4 (rsCD4). A non-linear regression method was used to analyze the sensorgrams, showing the existence of a double exponential time curve. A KA of 5.2 x 10(7) M-1 was calculated at 25 degrees C. The complex formation was exothermic (-4.5 kcal.mol-1( and entropically positive (+20 cal.mol-1.K-1). The reaction rate (0.234 x 10(5) M-1.s-1 at 25 degrees C) as well as the enthalpy change of the activated complex (+9.7 kcal.mol-1) are not compatible with a diffusion controlled reaction. The thermodynamic values calculated from equilibrium data corresponded to those calculated from kinetic data confirming the validity of the theoretical approach. As for most antigen-antibody interactions, complex formation was enthalpy driven. The overall positive entropy contribution to the stabilization of the complex is in contrast to that observed for the lysozyme-anti-lysozyme model and is probably due to electrostatic interaction between the epitope and the antibody combining site.
Collapse
Affiliation(s)
- F Velge-Roussel
- CJF INSERM 93-09 Immunologie des Maladies Infectieuses, Equipe Associée INRA d'Immunologie Parasitaire, UFR des Sciences Pharmaceutiques, Tours, France
| | | | | | | | | | | |
Collapse
|
44
|
Pintér C, Siccardi AG, Longhi R, Clivio A. Direct interaction of complement factor H with the C1 domain of HIV type 1 glycoprotein 120. AIDS Res Hum Retroviruses 1995; 11:577-88. [PMID: 7576914 DOI: 10.1089/aid.1995.11.577] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A protein that binds specifically to Env 105-119 (HEDIISLWDQSLKPC) was found in pools of normal human plasma when this peptide was used in affinity chromatography procedures. These samples represented the negative control in experiments aimed at the purification of putative human antibodies to the Env 105-119 region from AIDS sera. In this article we describe the biochemical characterization of this protein, which turned out to be complement factor H (CFH). We propose a functional role for this protein in the complex, early steps of CD4-dependent HIV infection.
Collapse
Affiliation(s)
- C Pintér
- Dipartimento di Biologia e Genetica per le Scienze Mediche, Università di Milano, Milan, Italy
| | | | | | | |
Collapse
|
45
|
Bour S, Geleziunas R, Wainberg MA. The human immunodeficiency virus type 1 (HIV-1) CD4 receptor and its central role in promotion of HIV-1 infection. Microbiol Rev 1995; 59:63-93. [PMID: 7708013 PMCID: PMC239355 DOI: 10.1128/mr.59.1.63-93.1995] [Citation(s) in RCA: 73] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Interactions between the viral envelope glycoprotein gp120 and the cell surface receptor CD4 are responsible for the entry of human immunodeficiency virus type 1 (HIV-1) into host cells in the vast majority of cases. HIV-1 replication is commonly followed by the disappearance or receptor downmodulation of cell surface CD4. This potentially renders cells nonsusceptible to subsequent infection by HIV-1, as well as by other viruses that use CD4 as a portal of entry. Disappearance of CD4 from the cell surface is mediated by several different viral proteins that act at various stages through the course of the viral life cycle, and it occurs in T-cell lines, peripheral blood CD4+ lymphocytes, and monocytes of both primary and cell line origin. At the cell surface, gp120 itself and in the form of antigen-antibody complexes can trigger cellular pathways leading to CD4 internalization. Intracellularly, the mechanisms leading to CD4 downmodulation by HIV-1 are multiple and complex; these include degradation of CD4 by Vpu, formation of intracellular complexes between CD4 and the envelope precursor gp160, and internalization by the Nef protein. Each of the above doubtless contributes to the ultimate depletion of cell surface CD4, although the relative contribution of each mechanism and the manner in which they interact remain to be definitively established.
Collapse
Affiliation(s)
- S Bour
- McGill AIDS Centre, Lady Davis Institute-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
46
|
Benjouad A, Chapuis F, Fenouillet E, Gluckman JC. Multibranched peptide constructs derived from the V3 loop of envelope glycoprotein gp120 inhibit human immunodeficiency virus type 1 infection through interaction with CD4. Virology 1995; 206:457-64. [PMID: 7831801 DOI: 10.1016/s0042-6822(95)80061-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The V3 loop of the gp120 of human immunodeficiency virus type 1 (HIV-1) is assumed to be involved in HIV-1-mediated membrane fusion. V3-derived peptides have been shown either to enhance or to prevent HIV-1 infection. Multibranched peptide constructs (MBPCs) derived from the V3 North American/European consensus sequence were designed to sort out these conflicting findings. At 5 microM, MBPC1 (8-branched GPGRAF) totally, and MBPC2 ([RKSIHIGPGRAFYT]4) partially, inhibited HIV-1LAI infection, whereas the GPGRAF monomer had only a limited effect. A peptide of the entire V3 consensus loop and a control MBPC had no detectable activity. The 5 microM MBPC1 HIV-1-inhibiting concentration was not cytotoxic, nor did it alter T lymphocyte allogeneic, antigen-, or mitogen-induced reactivities, and it was about 5- to 50-fold lower (MBPC2 and MBPC1, respectively) than that resulting in 50% cell death. Analysis of MBPC immunoreactivity showed that MBPC2, but not MBPC1, strongly reacted with human HIV-1 positive sera. Only MBPC2 elicited significant antibody responses in rabbits. The V3-derived MBPCs bound to CD4+ cells, as determined by immunofluorescence analysis. The binding was inhibited either by soluble CD4 or by CD4 monoclonal antibody (mAb) MT151, which recognizes the CDR3 region of the D1 domain of CD4, but not by other CD4 mAbs Leu3a, OKT4A, Q4021, 13B8-2, 5A8, RFT4, nor by the CD26 mAb BA5. Therefore, it appears likely that MBPCs inhibit HIV-1 infection by interacting with the CDR3 region of CD4 or with a region in its vicinity.
Collapse
Affiliation(s)
- A Benjouad
- Laboratoire de Biologie et Génétique des Pathologies Immunitaires, CNRS URA 1463, Paris, France
| | | | | | | |
Collapse
|
47
|
Ramurthy S, Lee MS, Nakanishi H, Shen R, Kahn M. Peptidomimetic antagonists designed to inhibit the binding of CD4 to HIV GP120. Bioorg Med Chem 1994; 2:1007-13. [PMID: 7712121 DOI: 10.1016/s0968-0896(00)82049-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Attempts to enhance the efficacy of our previously reported CD4 CDR2-like (residues 40-45) mimetic 1 by incorporation of the critical guanidine residue Arg-59 of CD4 are described.
Collapse
Affiliation(s)
- S Ramurthy
- Department of Pathobiology, University of Washington, Seattle 98195
| | | | | | | | | |
Collapse
|
48
|
Bour S, Geleziunas R, Wainberg MA. The role of CD4 and its downmodulation in establishment and maintenance of HIV-1 infection. Immunol Rev 1994; 140:147-71. [PMID: 7821926 DOI: 10.1111/j.1600-065x.1994.tb00869.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- S Bour
- Lady Davis Institute-Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
49
|
Ohki K, Kimura T, Jones IM, Morita F, Ikuta K. Multiple effects of CD4 CDR3-related peptide derivatives showing anti-HIV-1 activity on HIV-1 gp120 functions. Vaccine 1994; 12:343-50. [PMID: 8178557 DOI: 10.1016/0264-410x(94)90099-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The interaction of the human immunodeficiency virus type 1 (HIV-1) envelope glycoprotein gp120 with CD4 CDR3-related peptide derivatives showing anti-HIV-1 activity has been studied. Conformational changes in gp120, which could affect its interaction with CD4 and its shedding from virions, were detected by fluorescence spectrum analysis of tryptophan residues after addition of peptide representative of the CD4 CDR3-related region, but not the CD4 CDR2-related region. Interestingly, the addition of scrambled peptide, S1 (with altered amino acid sequence compared with the native CDR3-related peptide but unaltered overall composition), which we recently showed to have stronger anti-HIV-1 activity than the original CDR3-related peptide, had no effects on the conformational change in gp120 or on its interaction with CD4 and its shedding from HIV-1 virions. However, all of the CDR3-related peptides, including S1, showed blocking effects on the binding of antibodies against gp120 V3 loop and C-terminus regions. Thus, we concluded that there were at least two separable activities of the CDR3-related peptides in anti-HIV-1 activity, i.e. induction of conformational changes in gp120, which could affect its binding to CD4 and to gp41 (as observed in native CDR3-related peptides), and inactivation of V3 loop and C-terminus regions in gp120 (as observed in all of the CDR3-related peptides, including S1).
Collapse
Affiliation(s)
- K Ohki
- Section of Serology, Hokkaido University, Sapporo, Japan
| | | | | | | | | |
Collapse
|
50
|
McCarrick MA, Kollman P. Use of molecular dynamics and free energy perturbation calculations in anti-human immunodeficiency virus drug design. Methods Enzymol 1994; 241:370-84. [PMID: 7854189 DOI: 10.1016/0076-6879(94)41074-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- M A McCarrick
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of California, San Francisco 94143
| | | |
Collapse
|