851
|
Very few substitutions in a germ line antibody are required to initiate significant domain exchange. J Virol 2010; 84:10700-7. [PMID: 20702640 DOI: 10.1128/jvi.01111-10] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
2G12 is a broadly neutralizing anti-HIV-1 monoclonal human IgG1 antibody reactive with a high-mannose glycan cluster on the surface of glycoprotein gp120. A key feature of this very highly mutated antibody is domain exchange of the heavy-chain variable region (V(H)) with the V(H) of the adjacent Fab of the same immunoglobulin, which assembles a multivalent binding interface composed of two primary binding sites in close proximity. A non-germ line-encoded proline in the elbow between V(H) and C(H)1 and an extensive network of hydrophobic interactions in the V(H)/V(H)' interface have been proposed to be crucial for domain exchange. To investigate the origins of domain exchange, a germ line version of 2G12 that behaves as a conventional antibody was engineered. Substitution of 5 to 7 residues for those of the wild type produced a significant fraction of domain-exchanged molecules, with no evidence of equilibrium between domain-exchanged and conventional forms. Two substitutions not previously implicated, A(H14) and E(H75), are the most crucial for domain exchange, together with I(H19) at the V(H)/V(H)' interface and P(H113) in the elbow region. Structural modeling gave clues as to why these residues are essential for domain exchange. The demonstration that domain exchange can be initiated by a small number of substitutions in a germ line antibody suggests that the evolution of a domain-exchanged antibody response in vivo may be more readily achieved than considered to date.
Collapse
|
852
|
Pietzsch J, Scheid JF, Mouquet H, Klein F, Seaman MS, Jankovic M, Corti D, Lanzavecchia A, Nussenzweig MC. Human anti-HIV-neutralizing antibodies frequently target a conserved epitope essential for viral fitness. ACTA ACUST UNITED AC 2010; 207:1995-2002. [PMID: 20679402 PMCID: PMC2931156 DOI: 10.1084/jem.20101176] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification and characterization of conserved epitopes on the HIV-1 viral spike that are immunogenic in humans and targeted by neutralizing antibodies is an important step in vaccine design. Antibody cloning experiments revealed that 32% of all HIV-neutralizing antibodies expressed by the memory B cells in patients with high titers of broadly neutralizing antibodies recognize one or more “core” epitopes that were not defined. Here, we show that anti-core antibodies recognize a single conserved epitope on the gp120 subunit. Amino acids D474, M475, R476, which are essential for anti-core antibody binding, form an immunodominant triad at the outer domain/inner domain junction of gp120. The mutation of these residues to alanine impairs viral fusion and fitness. Thus, the core epitope, a frequent target of anti–HIV-neutralizing antibodies, including the broadly neutralizing antibody HJ16, is conserved and indispensible for viral infectivity. We conclude that the core epitope should be considered as a target for vaccine design.
Collapse
Affiliation(s)
- John Pietzsch
- Laboratory of Molecular Immunology and Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
853
|
Anti-gp41 antibodies cloned from HIV-infected patients with broadly neutralizing serologic activity. J Virol 2010; 84:5032-42. [PMID: 20219932 DOI: 10.1128/jvi.00154-10] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Most HIV-infected individuals develop antibodies to the gp120 and gp41 components of the viral spike; however, only a fraction of these individuals mount a broadly neutralizing serum response against HIV. We have cloned anti-HIV antibodies from the memory B-cell compartment of six individuals with variable viral loads and high titers of broadly neutralizing antibodies. Here, we report on the features of the anti-gp41 response in these patients. Competition experiments with previously characterized antibodies targeting defined epitopes on the gp41 ectodomain showed antibodies directed against the "immunodominant region" (cluster I), the carboxy-terminal heptad repeat (cluster II), and the membrane-proximal external region (cluster IV). On the other hand, antibodies directed against the amino-terminal part of the molecule, including the fusion peptide, polar region, and the N-terminal heptad repeat, were not detected. When all patients' data were combined, unique B-cell clones targeting cluster I, II, and IV accounted for 32%, 49%, and 53% of all anti-gp41-reactive B cells, respectively; therefore, no single region was truly immunodominant. Finally, although we found no new neutralizing epitopes or HIV-1-neutralizing activity by any of the gp41 antibodies at concentrations of up to 50 microg/ml, high concentrations of 7 out of 15 anti-cluster I antibodies neutralized tier 2 viruses.
Collapse
|
854
|
Abstract
PURPOSE OF REVIEW Effective vaccine-elicited immunity against HIV-1 infection will likely require broadly neutralizing antibodies to interrupt the fusion-promoting functions of the viral envelope glycoprotein spikes. Efforts in this area have, however, been fraught with challenges. The handful of existing broadly neutralizing monoclonal antibodies has provided information on some of the virus' sites of vulnerability, fueling a decade of structure-informed vaccine design. The fact that very few bnmAbs have been recovered to date illustrates the poor immunogenicity of these epitopes. Recognizing that progress may require more basic information, there has been a notable shift in the field toward identifying new chinks in HIV-1's armor. These efforts are based on the observation that some infected patients develop exceptionally broad serum neutralizing antibodies responses, a better understanding of which would be valuable for vaccine efforts aimed at eliciting similar specificities. RECENT FINDINGS New mapping methodologies are now providing an appreciation of the incidence of specificities similar to the existing known bnmAbs as well as some intriguing insights into novel specificities. SUMMARY The new information emerging from mapping efforts should help to sharpen efforts to isolate new bnmAbs and moreover, may provide crucial information for the rational design of novel vaccine candidates.
Collapse
|
855
|
Abstract
PURPOSE OF REVIEW Several recent advances are permitting a detailed examination of the HIV-specific B-cell response. In this review, we summarize these advances and their implications for understanding the response to HIV during chronic infection or in vaccine. RECENT FINDINGS In HIV-infected patients, aberrant B-cell phenotypes have been associated with diminished humoral responses to other pathogens. HIV-specific B cells are overrepresented in some of these abnormal subsets. Over the past 2 years, flow cytometry-based techniques have been developed to stain HIV-specific B cells. These techniques are permitting a re-examination of frequency, phenotype, and function of HIV-specific B cells. They are also permitting the isolation of HIV-specific B cells in high purity. Immunoglobulin G from sorted HIV-specific B cells is oligoclonal, uses a limited repertoire of immunoglobulin genes, and targets multiple epitopes on Env. SUMMARY It is likely that the defects found in total B cells in HIV-infected patients also play a role in the poorly effective HIV-specific antibody response. A subset of HIV-infected patients produced broadly neutralizing antibodies. Understanding this antibody response, and the B cells that underlie it, may be critical in efforts to elicit neutralizing antibodies against HIV.
Collapse
|
856
|
Identification of human idiotype-specific T cells in lymphoma and myeloma. Curr Top Microbiol Immunol 2010; 344:193-210. [PMID: 20549471 DOI: 10.1007/82_2010_70] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Idiotype protein, among the first identified tumor-specific antigens, has been found to stimulate both humoral and cellular responses in lymphoma and myeloma patients. With the increasing use of B cell depletion treatments such as rituximab in clinic, the cellular response mediated by idiotype-specific T cells has become increasingly important as an adjunct therapy for lymphoma and myeloma. Here, we review the idiotype protein as a tumor antigen and the characteristics of the T cell response elicited idiotype vaccination. We also analyze the T cell epitopes that have been identified in idiotype protein and introduce our new findings of additional T cell epitopes derived from the Ig light chain. Finally, we propose new directions in the generation of idiotype-specific T cells for tumor therapy.
Collapse
|
857
|
Cloning and expression of murine Ig genes from single B cells. J Immunol Methods 2009; 350:183-93. [DOI: 10.1016/j.jim.2009.08.009] [Citation(s) in RCA: 212] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2009] [Accepted: 08/19/2009] [Indexed: 01/03/2023]
|
858
|
Dosenovic P, Chakrabarti B, Soldemo M, Douagi I, Forsell MNE, Li Y, Phogat A, Paulie S, Hoxie J, Wyatt RT, Karlsson Hedestam GB. Selective expansion of HIV-1 envelope glycoprotein-specific B cell subsets recognizing distinct structural elements following immunization. THE JOURNAL OF IMMUNOLOGY 2009; 183:3373-82. [PMID: 19696434 DOI: 10.4049/jimmunol.0900407] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The HIV-1 envelope glycoprotein (Env) functional spike has evolved multiple immune evasion strategies, and only a few broadly neutralizing determinants on the assembled spike are accessible to Abs. Serological studies, based upon Ab binding and neutralization activity in vitro, suggest that vaccination with current Env-based immunogens predominantly elicits Abs that bind nonneutralizing or strain-restricted neutralizing epitopes. However, the fractional specificities of the polyclonal mixture of Abs present in serum, especially those directed to conformational Env epitopes, are often difficult to determine. Furthermore, serological analyses do not provide information regarding how repeated Ag inoculation impacts the expansion and maintenance of Env-specific B cell subpopulations. Therefore, we developed a highly sensitive Env-specific B cell ELISPOT system, which allows the enumeration of Ab-secreting cells (ASC) from diverse anatomical compartments directed against different structural determinants of Env. In this study, we use this system to examine the evolution of B cell responses in mice immunized with engineered Env trimers in adjuvant. We demonstrate that the relative proportion of ASC specific for defined structural elements of Env is altered significantly by homologous booster immunizations. This results in the selective expansion of ASC directed against the variable regions of Env. We suggest that the B cell specificity and compartment analysis described in this study are important complements to serological mapping studies for the examination of B cell responses against subspecificities of a variety of immunogens.
Collapse
Affiliation(s)
- Pia Dosenovic
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
859
|
Abstract
PURPOSE OF REVIEW It has long been known that autologous neutralizing antibodies (AnAbs) exert pressure on the envelope of HIV, resulting in neutralization escape. However, recently, progress has been made in uncovering the precise targets of these potent early antibodies. RECENT FINDINGS AnAbs primarily target variable regions of the HIV-1 envelope, explaining the strain-specificity of these antibodies. Despite high neutralizing potential and cross-reactivity, anti-V3 antibodies do not contribute to autologous neutralization. The V1V2 is commonly immunogenic in early HIV-1 and simian human immunodeficiency virus infections, though the nature of these epitopes remains to be determined. In subtype C viruses, the C3 region is a neutralization target, possibly as a result of its more exposed and amphipathic structure. Autologous neutralization appears to be mediated by very few AnAb specificities that develop sequentially suggesting the possibility of immunological hierarchies for both binding and neutralizing antibodies. The role of AnAbs in preventing superinfection and in restricting virus replication is reexamined in the context of recent data. SUMMARY New studies have greatly contributed toward our understanding of the specificities mediating autologous neutralization and highlighted potential vulnerabilities on transmitted viruses. However, the contribution of AnAbs to the development of neutralization breadth remains to be characterized.
Collapse
Affiliation(s)
- Penny L Moore
- AIDS Virus Research Unit, National Institute for Communicable Diseases, Johannesburg, South Africa.
| | | | | |
Collapse
|
860
|
Neutralizing antibodies generated during natural HIV-1 infection: good news for an HIV-1 vaccine? Nat Med 2009; 15:866-70. [PMID: 19525964 DOI: 10.1038/nm.1949] [Citation(s) in RCA: 339] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Most existing viral vaccines generate antibodies that either block initial infection or help eradicate the virus before it can cause disease. For HIV-1, obstacles to eliciting protective neutralizing antibodies (NAbs) have often seemed insurmountable. The target of HIV-specific NAbs, the viral envelope glycoprotein (Env), is highly variable in amino acid sequence and glycosylation pattern. Conserved elements of HIV-1 Env seem to be poorly immunogenic, and previous attempts to generate broadly reactive NAbs by vaccination have proven ineffective. However, recent studies show that antibodies in the sera of some HIV-1-infected individuals can neutralize diverse HIV-1 isolates. Detailed analyses of these sera provide new insights into the viral epitopes targeted by broadly reactive NAbs. The findings discussed here suggest that the natural NAb response to HIV-1 can inform future vaccine design. A concerted effort of structure-based vaccine design will help guide the development of improved antibody-based vaccines for HIV-1.
Collapse
|
861
|
Circulating CD21low B cells in common variable immunodeficiency resemble tissue homing, innate-like B cells. Proc Natl Acad Sci U S A 2009; 106:13451-6. [PMID: 19666505 DOI: 10.1073/pnas.0901984106] [Citation(s) in RCA: 275] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The homeostasis of circulating B cell subsets in the peripheral blood of healthy adults is well regulated, but in disease it can be severely disturbed. Thus, a subgroup of patients with common variable immunodeficiency (CVID) presents with an extraordinary expansion of an unusual B cell population characterized by the low expression of CD21. CD21(low) B cells are polyclonal, unmutated IgM(+)IgD(+) B cells but carry a highly distinct gene expression profile which differs from conventional naïve B cells. Interestingly, while clearly not representing a memory population, they do share several features with the recently defined memory-like tissue, Fc receptor-like 4 positive B cell population in the tonsils of healthy donors. CD21(low) B cells show signs of previous activation and proliferation in vivo, while exhibiting defective calcium signaling and poor proliferation in response to B cell receptor stimulation. CD21(low) B cells express decreased amounts of homeostatic but increased levels of inflammatory chemokine receptors. This might explain their preferential homing to peripheral tissues like the bronchoalveolar space of CVID or the synovium of rheumatoid arthritis patients. Therefore, as a result of the close resemblance to the gene expression profile, phenotype, function and preferential tissue homing of murine B1 B cells, we suggest that CD21(low) B cells represent a human innate-like B cell population.
Collapse
|
862
|
Bolton DL, Roederer M. Flow cytometry and the future of vaccine development. Expert Rev Vaccines 2009; 8:779-89. [PMID: 19485757 DOI: 10.1586/erv.09.41] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Vaccine research increasingly aims to understand the fundamental mechanisms of protection afforded by licensed and candidate vaccines. Historically, nearly all licensed vaccines have relied on measures of humoral immunity to provide correlates of protection, but cellular immunity is important for protection afforded by some vaccines and will be required for vaccines against TB and malaria. Common means of assessing vaccine-induced immune responses include measuring the frequency and functions of antigen-specific lymphocytes. While diverse assays can provide this information, flow cytometry is unique in its ability to simultaneously report other features of antigen-specific cellular responses. Here, we review the application of flow cytometry to characterizing three areas of immune responses to vaccines or diseases. First, analysis of cellular (T-cell) responses is more mature: polychromatic flow cytometric analysis of T-cell function has already yielded important insight into correlates of protection. Second, antibody and antigen-specific B-cell detection by flow cytometry are being actively developed; to date, these assays are not yet widely used. Finally, flow cytometry can also be used to analyze the contribution of innate immunity to vaccine efficacy and disease pathogenesis.
Collapse
Affiliation(s)
- Diane L Bolton
- Vaccine Research Center, NIAID, NIH, Bethesda, MD 20892, USA.
| | | |
Collapse
|
863
|
Liao HX, Levesque MC, Nagel A, Dixon A, Zhang R, Walter E, Parks R, Whitesides J, Marshall DJ, Hwang KK, Yang Y, Chen X, Gao F, Munshaw S, Kepler TB, Denny T, Moody MA, Haynes BF. High-throughput isolation of immunoglobulin genes from single human B cells and expression as monoclonal antibodies. J Virol Methods 2009; 158:171-9. [PMID: 19428587 PMCID: PMC2805188 DOI: 10.1016/j.jviromet.2009.02.014] [Citation(s) in RCA: 210] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Revised: 02/05/2009] [Accepted: 02/10/2009] [Indexed: 12/28/2022]
Abstract
Defining human B cell repertoires to viral pathogens is critical for design of vaccines that induce broadly protective antibodies to infections such as HIV-1 and influenza. Single B cell sorting and cloning of immunoglobulin (Ig) heavy- and light-chain variable regions (V(H) and V(L)) is a powerful technology for defining anti-viral B cell repertoires. However, the Ig-cloning step is time-consuming and prevents high-throughput analysis of the B cell repertoire. Novel linear Ig heavy- and light-chain gene expression cassettes were designed to express Ig V(H) and V(L) genes isolated from sorted single B cells as IgG1 antibody without a cloning step. The cassettes contain all essential elements for transcriptional and translational regulation, including CMV promoter, Ig leader sequences, constant region of IgG1 heavy- or Ig light-chain, poly(A) tail and substitutable V(H) or V(L) genes. The utility of these Ig gene expression cassettes was established using synthetic V(H) or V(L) genes from an anti-HIV-1 gp41 mAb 2F5 as a model system, and validated further using V(H) and V(L) genes isolated from cloned EBV-transformed antibody-producing cell lines. Finally, this strategy was successfully used for rapid production of recombinant influenza mAbs from sorted single human plasmablasts after influenza vaccination. These Ig gene expression cassettes constitute a highly efficient strategy for rapid expression of Ig genes for high-throughput screening and analysis without cloning.
Collapse
Affiliation(s)
- Hua-Xin Liao
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - Marc C. Levesque
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710
| | - Ashleigh Nagel
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Ashlyn Dixon
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Ruijun Zhang
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Emmanuel Walter
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - John Whitesides
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - Dawn J. Marshall
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Kwan-Ki Hwang
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Yi Yang
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Xi Chen
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
| | - Feng Gao
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - Supriya Munshaw
- Center for Computational Immunology, Duke University Medical Center, Durham, NC, 27710
| | - Thomas B. Kepler
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Center for Computational Immunology, Duke University Medical Center, Durham, NC, 27710
| | - Thomas Denny
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
| | - M. Anthony Moody
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Pediatrics, Duke University Medical Center, Durham, NC, 27710
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, 27710
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710
- Department of Immunology, Duke University Medical Center, Durham, NC, 27710
| |
Collapse
|
864
|
Suzuki H, Fan R, Zhang Z, Brown R, Hall S, Julian BA, Chatham WW, Suzuki Y, Wyatt RJ, Moldoveanu Z, Lee JY, Robinson J, Tomana M, Tomino Y, Mestecky J, Novak J. Aberrantly glycosylated IgA1 in IgA nephropathy patients is recognized by IgG antibodies with restricted heterogeneity. J Clin Invest 2009; 119:1668-77. [PMID: 19478457 DOI: 10.1172/jci38468] [Citation(s) in RCA: 235] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 04/15/2009] [Indexed: 12/13/2022] Open
Abstract
IgA nephropathy (IgAN) is characterized by circulating immune complexes composed of galactose-deficient IgA1 and a glycan-specific IgG antibody. These immune complexes deposit in the glomerular mesangium and induce the mesangioproliferative glomerulonephritis characteristic of IgAN. To define the precise specificities and molecular properties of the IgG antibodies, we generated EBV-immortalized IgG-secreting lymphocytes from patients with IgAN and found that the secreted IgG formed complexes with galactose-deficient IgA1 in a glycan-dependent manner. We cloned and sequenced the heavy- and light-chain antigen-binding domains of IgG specific for galactose-deficient IgA1 and identified an A to S substitution in the complementarity-determining region 3 of the variable region of the gene encoding the IgG heavy chain in IgAN patients. Furthermore, site-directed mutagenesis that reverted the residue to alanine reduced the binding of recombinant IgG to galactose-deficient IgA1. Finally, we developed a dot-blot assay for the glycan-specific IgG antibody that differentiated patients with IgAN from healthy and disease controls with 88% specificity and 95% sensitivity and found that elevated levels of this antibody in the sera of patients with IgAN correlated with proteinuria. Collectively, these findings indicate that glycan-specific antibodies are associated with the development of IgAN and may represent a disease-specific marker and potential therapeutic target.
Collapse
Affiliation(s)
- Hitoshi Suzuki
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
865
|
Scheid JF, Mouquet H, Feldhahn N, Walker BD, Pereyra F, Cutrell E, Seaman MS, Mascola JR, Wyatt RT, Wardemann H, Nussenzweig MC. A method for identification of HIV gp140 binding memory B cells in human blood. J Immunol Methods 2009; 343:65-7. [PMID: 19100741 PMCID: PMC2754789 DOI: 10.1016/j.jim.2008.11.012] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Accepted: 10/31/2008] [Indexed: 11/29/2022]
Abstract
Antibodies to HIV are potentially important reagents for basic and clinical studies. Historically, these reagents have been produced by random cloning of heavy and light chains in phage display libraries [Burton, D.R., Barbas, C.F. III, Persson, M.A.A., Koenig, S., Chanock, R.M., and Lerner, R.A., (1991), A large array of human monoclonal antibodies to type 1 immunodeficiency virus from combinatorial libraries of asymptomatic seropositive individuals. Proc. Natl. Acad. Sci. U. S. A. 88, 10134-10137.] and electrofusion techniques [Buchacher, A., Predl, R., Tauer, C., Purtscher, M., Gruber, G., Heider, R., Steindl, F., Trkola, A., Jungbauer, A., and Katinger, H., (1992), Human monoclonal antibodies against gp41 and gp120 as potential agent for passive immunization. Vaccines 92, 191-195]. Here we describe a method to identify and potentially enrich human memory B cells from HIV infected patients that show serum titers of neutralizing antibodies. When biotinylated gp140 is used to stain peripheral blood mononuclear cells it identifies a distinct population of gp140 binding B cells by flow cytometry.
Collapse
Affiliation(s)
- Johannes F Scheid
- Laboratory of Molecular Immunology, The Rockefeller University, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
866
|
Broad diversity of neutralizing antibodies isolated from memory B cells in HIV-infected individuals. Nature 2009; 458:636-40. [PMID: 19287373 DOI: 10.1038/nature07930] [Citation(s) in RCA: 711] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2009] [Accepted: 02/27/2009] [Indexed: 11/08/2022]
Abstract
Antibodies to conserved epitopes on the human immunodeficiency virus (HIV) surface protein gp140 can protect against infection in non-human primates, and some infected individuals show high titres of broadly neutralizing immunoglobulin (Ig)G antibodies in their serum. However, little is known about the specificity and activity of these antibodies. To characterize the memory antibody responses to HIV, we cloned 502 antibodies from HIV envelope-binding memory B cells from six HIV-infected patients with broadly neutralizing antibodies and low to intermediate viral loads. We show that in these patients, the B-cell memory response to gp140 is composed of up to 50 independent clones expressing high affinity neutralizing antibodies to the gp120 variable loops, the CD4-binding site, the co-receptor-binding site, and to a new neutralizing epitope that is in the same region of gp120 as the CD4-binding site. Thus, the IgG memory B-cell compartment in the selected group of patients with broad serum neutralizing activity to HIV is comprised of multiple clonal responses with neutralizing activity directed against several epitopes on gp120.
Collapse
|
867
|
Discordant memory B cell and circulating anti-Env antibody responses in HIV-1 infection. Proc Natl Acad Sci U S A 2009; 106:3952-7. [PMID: 19225108 DOI: 10.1073/pnas.0813392106] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Long-lived memory B cells (B(Mem)) provide an archive of historic Ab responses. By contrast, circulating Abs typically decline once the immunogen is cleared. Consequently, circulating Abs can underestimate the nature of cognate humoral immunity. On the other hand, the B(Mem) pool should provide a comprehensive picture of Ab specificities that arise over the entire course of infection. To test this hypothesis, we compared circulating Ab and B(Mem) from natural virus suppressors who control HIV-1 without therapy and maintain a relatively intact immune system. We found high frequencies of B(Mem) specific for the conserved neutralizing CD4 induced or CD4 binding site epitopes of gp120, whereas low Ab titers to these determinants were detected in contemporaneous plasma. These data suggest that plasma Ab repertoires can underestimate the breadth of humoral immunity, and analyses of B(Mem) should be included in studies correlating Ab specificity with protective immunity to HIV-1.
Collapse
|
868
|
Smith K, Garman L, Wrammert J, Zheng NY, Capra JD, Ahmed R, Wilson PC. Rapid generation of fully human monoclonal antibodies specific to a vaccinating antigen. Nat Protoc 2009; 4:372-84. [PMID: 19247287 PMCID: PMC2750034 DOI: 10.1038/nprot.2009.3] [Citation(s) in RCA: 571] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We describe herein a protocol for the production of antigen-specific human monoclonal antibodies (hmAbs). Antibody-secreting cells (ASCs) are isolated from whole blood collected 7 d after vaccination and sorted by flow cytometry into single cell plates. The antibody genes of the ASCs are then amplified by RT-PCR and nested PCR, cloned into expression vectors and transfected into a human cell line. The expressed antibodies can then be purified and assayed for binding and neutralization. This method uses established techniques but is novel in their combination and application. This protocol can be completed with as little as 20 ml of human blood and in as little as 28 d when optimal. Although previous methodologies to produce hmAbs, including B-cell immortalization or phage display, can be used to isolate the rare specific antibody even years after immunization, in comparison, these approaches are inefficient, resulting in few relevant antibodies. Although dependent on having an ongoing immune response, the approach described herein can be used to rapidly generate numerous antigen-specific hmAbs in a short time.
Collapse
Affiliation(s)
- Kenneth Smith
- Department of Clinical Immunology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
869
|
B-cell tolerance checkpoints in health and autoimmunity. Curr Opin Immunol 2008; 20:632-8. [DOI: 10.1016/j.coi.2008.09.001] [Citation(s) in RCA: 223] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2008] [Accepted: 09/10/2008] [Indexed: 11/22/2022]
|
870
|
Abstract
A new method allows rapid identification of human monoclonal antibodies from immune or vaccinated individuals.
Collapse
|
871
|
Rohatgi S, Ganju P, Sehgal D. Systematic design and testing of nested (RT-)PCR primers for specific amplification of mouse rearranged/expressed immunoglobulin variable region genes from small number of B cells. J Immunol Methods 2008; 339:205-19. [PMID: 18926828 DOI: 10.1016/j.jim.2008.09.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2008] [Revised: 08/12/2008] [Accepted: 09/18/2008] [Indexed: 11/28/2022]
Abstract
The aim of this study was to develop a highly specific and sensitive (RT-)PCR capable of potentially amplifying the rearranged/expressed VH and VL gene belonging to any mouse immunoglobulin V gene family from a single or a small number of B cells. A database of germline immunoglobulin sequences was used to design 112 primers for a nested (RT-)PCR based strategy to cover all VH, VL, JH, JL, CH and CL gene families/genes from C57BL/6 and BALB/c mice. 93.7% of the primers had 4-fold or less, while 71.4% had no degeneracy. The proportions of germline V genes to which the primers bind with no, up to 1 and up to 2 mismatches are 59.7%, 84.1% and 94.9%, respectively. Most but not all V gene family specific primers designed allow amplification of full-length V genes. The nested primers permit PCR amplification of rearranged V genes belonging to all VH and VL gene families from splenocyte genomic DNA. The V gene family-specific nature of the primers was experimentally confirmed for randomly selected 6 VH and 6 Vkappa families, and all Vlambda genes. The broad V gene family coverage of our primer set was experimentally validated by amplifying the rearranged/expressed VH and VL genes from splenocytes and a panel of 38 hybridomas under conditions where primer mixes and genomic DNA or total RNA was used as starting template. We observed no or low-level cross-family priming. Pooled constant region specific primers allowed efficient RT-PCR amplification of H and L chain isotypes. The expressed VH and VL genes belonging to different V gene families RT-PCR amplified from a mixture of hybridomas in a representative manner. We successfully amplified the expressed VH and Vkappa gene from a single hybridoma cell by RT-PCR and from 10-15 microdissected B cells by genomic PCR. This, first of its kind, comprehensive set of highly sensitive and specific nested primers that provide broad V gene family coverage will open up new avenues and opportunities to study various aspects of mouse B cell biology.
Collapse
Affiliation(s)
- Soma Rohatgi
- National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, India
| | | | | |
Collapse
|
872
|
Bradshaw EM, Kent SC, Tripuraneni V, Orban T, Ploegh HL, Hafler DA, Love JC. Concurrent detection of secreted products from human lymphocytes by microengraving: cytokines and antigen-reactive antibodies. Clin Immunol 2008; 129:10-8. [PMID: 18675591 PMCID: PMC2577144 DOI: 10.1016/j.clim.2008.06.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/07/2008] [Accepted: 06/09/2008] [Indexed: 12/31/2022]
Abstract
Cell surface determinants, cytokines and antibodies secreted by hematopoietic cells are used to classify their lineage and function. Currently available techniques are unable to elucidate multiple secreted proteins while also assigning phenotypic surface-displayed markers to the individual living cells. Here, a soft lithographic method, microengraving, was adapted for the multiplexed interrogation of populations of individual human peripheral blood mononuclear cells for secreted cytokines (IFN-gamma and IL-6), antigen-specific antibodies, and lineage-specific surface-expressed markers. Application of the method to a clinical sample from a recent-onset Type 1 diabetic subject with a positive titer of anti-insulin antibodies showed that approximately 0.58% of circulating CD19(+) B cells secreted proinsulin-reactive antibodies of the IgG isotype and 2-3% of circulating cells secreted IL-6. These data demonstrate the utility of microengraving for interrogating multiple phenotypes of single human cells concurrently and for detecting rare populations of cells by their secreted products.
Collapse
Affiliation(s)
- Elizabeth M. Bradshaw
- Division of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Sally C. Kent
- Division of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Vinay Tripuraneni
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139
| | - Tihamer Orban
- Section of Immunology and Immunogenetics, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115
| | - Hidde L. Ploegh
- Whitehead Institute for Biomedical Research, 9 Cambridge Center, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- The Broad Institute of MIT and Harvard, Seven Cambridge Center, Cambridge, MA 02139
| | - David A. Hafler
- Division of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
- The Broad Institute of MIT and Harvard, Seven Cambridge Center, Cambridge, MA 02139
| | - J. Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA 02139
| |
Collapse
|
873
|
Mietzner B, Tsuiji M, Scheid J, Velinzon K, Tiller T, Abraham K, Gonzalez JB, Pascual V, Stichweh D, Wardemann H, Nussenzweig MC. Autoreactive IgG memory antibodies in patients with systemic lupus erythematosus arise from nonreactive and polyreactive precursors. Proc Natl Acad Sci U S A 2008; 105:9727-32. [PMID: 18621685 PMCID: PMC2474524 DOI: 10.1073/pnas.0803644105] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Indexed: 12/30/2022] Open
Abstract
Persistent autoantibody production in patients with systemic lupus erythematosus (SLE) suggests the existence of autoreactive humoral memory, but the frequency of self-reactive memory B cells in SLE has not been determined. Here, we report on the reactivity of 200 monoclonal antibodies from single IgG+ memory B cells of four SLE patients. The overall frequency of polyreactive and HEp-2 self-reactive antibodies in this compartment was similar to controls. We found 15% of IgG memory B cell antibodies highly reactive and specific for SLE-associated extractable nuclear antigens (ENA) Ro52 and La in one patient with serum autoantibody titers of the same specificity but not in the other three patients or healthy individuals. The germ-line forms of the ENA antibodies were non-self-reactive or polyreactive with low binding to Ro52, supporting the idea that somatic mutations contributed to autoantibody specificity and reactivity. Heterogeneity in the frequency of memory B cells expressing SLE-associated autoantibodies suggests that this variable may be important in the outcome of therapies that ablate this compartment.
Collapse
Affiliation(s)
- Brun Mietzner
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | | | | | | | - Thomas Tiller
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | - Klaus Abraham
- Central Institute for Laboratory Medicine and Pathobiochemistry, Charité Hospital, 13353 Berlin, Germany
| | - Jose B. Gonzalez
- Institute for Clinical Chemistry and Pathobiochemistry, Charité Hospital, 12200 Berlin, Germany; and
| | - Virginia Pascual
- Baylor Institute for Immunology Research, Baylor University, Dallas, TX 75204
| | - Dorothee Stichweh
- Baylor Institute for Immunology Research, Baylor University, Dallas, TX 75204
| | - Hedda Wardemann
- *Max Planck Institute for Infection Biology, 10117 Berlin, Germany
| | | |
Collapse
|