Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Elin S. Gray is active.

Publication


Featured researches published by Elin S. Gray.


Journal of Virology | 2007

Neutralizing Antibody Responses in Acute Human Immunodeficiency Virus Type 1 Subtype C Infection

Elin S. Gray; Penny L. Moore; Isaac A. Choge; Julie M. Decker; F. Bibollet-Ruche; Hui Li; N. Leseka; Florette K. Treurnicht; Koleka Mlisana; George M. Shaw; S. S. Abdool Karim; Carolyn Williamson; Lynn Morris

ABSTRACT The study of the evolution and specificities of neutralizing antibodies during the course of human immunodeficiency virus type 1 (HIV-1) infection may be important in the discovery of possible targets for vaccine design. In this study, we assessed the autologous and heterologous neutralization responses of 14 HIV-1 subtype C-infected individuals, using envelope clones obtained within the first 2 months postinfection. Our data show that potent but relatively strain-specific neutralizing antibodies develop within 3 to 12 months of HIV-1 infection. The magnitude of this response was associated with shorter V1-to-V5 envelope lengths and fewer glycosylation sites, particularly in the V1-V2 region. Anti-MPER antibodies were detected in 4 of 14 individuals within a year of infection, while antibodies to CD4-induced (CD4i) epitopes developed to high titers in 12 participants, in most cases before the development of autologous neutralizing antibodies. However, neither anti-MPER nor anti-CD4i antibody specificity conferred neutralization breadth. These data provide insights into the kinetics, potency, breadth, and epitope specificity of neutralizing antibody responses in acute HIV-1 subtype C infection.


Journal of Virology | 2012

The Development of CD4 Binding Site Antibodies During HIV-1 Infection

Rebecca M. Lynch; Lillian Tran; Mark K. Louder; Stefan D. Schmidt; Myron S. Cohen; Rebecca DerSimonian; Zelda Euler; Elin S. Gray; Salim Safurdeen. Abdool Karim; Jennifer L. Kirchherr; David C. Montefiori; Sengeziwe Sibeko; Kelly A. Soderberg; Georgia D. Tomaras; Zhi-Yong Yang; Gary J. Nabel; Hanneke Schuitemaker; Lynn Morris; Barton F. Haynes; John R. Mascola

ABSTRACT Broadly neutralizing antibodies to the CD4 binding site (CD4bs) of gp120 are generated by some HIV-1-infected individuals, but little is known about the prevalence and evolution of this antibody response during the course of HIV-1 infection. We analyzed the sera of 113 HIV-1 seroconverters from three cohorts for binding to a panel of gp120 core proteins and their corresponding CD4bs knockout mutants. Among sera collected between 99 and 258 weeks post-HIV-1 infection, 88% contained antibodies to the CD4bs and 47% contained antibodies to resurfaced stabilized core (RSC) probes that react preferentially with broadly neutralizing CD4bs antibodies (BNCD4), such as monoclonal antibodies (MAbs) VRC01 and VRC-CH31. Analysis of longitudinal serum samples from a subset of 18 subjects revealed that CD4bs antibodies to gp120 arose within the first 4 to 16 weeks of infection, while the development of RSC-reactive antibodies was more varied, occurring between 10 and 152 weeks post-HIV-1 infection. Despite the presence of these antibodies, serum neutralization mediated by RSC-reactive antibodies was detected in sera from only a few donors infected for more than 3 years. Thus, CD4bs antibodies that bind a VRC01-like epitope are often induced during HIV-1 infection, but the level and potency required to mediate serum neutralization may take years to develop. An improved understanding of the immunological factors associated with the development and maturation of neutralizing CD4bs antibodies during HIV-1 infection may provide insights into the requirements for eliciting this response by vaccination.


Virology | 2009

High Titer HIV-1 V3-Specific Antibodies with Broad Reactivity but Low Neutralizing Potency in Acute Infection and Following Vaccination

Katie L. Davis; Elin S. Gray; Penny L. Moore; Julie M. Decker; Aidy Salomon; David C. Montefiori; Barney S. Graham; Michael C. Keefer; Abraham Pinter; Lynn Morris; Beatrice H. Hahn; George M. Shaw

Identifying the earliest neutralizing antibody specificities that are elicited following infection or vaccination by HIV-1 is an important objective of current HIV/AIDS vaccine research. We have shown previously that transplantation of HIV-1 V3 epitopes into an HIV-2 envelope (Env) scaffold provides a sensitive and specific means to detect and quantify HIV-1 V3 epitope specific neutralizing antibodies (Nabs) in human sera. Here, we employ this HIV-2/HIV-1 V3 scaffolding strategy to study the kinetics of development and breadth of V3-specific Nabs in longitudinal sera from individuals acutely infected with clade C or clade B HIV-1 and in human subjects immunized with clade B HIV-1 immunogens. HIV-2/HIV-1 chimeras containing V3 sequences matched to virus type (HIV-2 or HIV-1), subtype (clade B or C), or strain (autologous or heterologous) were used as test reagents. We found that by 3-8 weeks post infection, 12 of 14 clade C subjects had a median IC(50) V3-specific Nab titer of 1:700 against chimeric viruses containing a heterologous clade C V3. By 5 months post-infection, all 14 subjects were positive for V3-specific Nabs with median titers of 1:8000 against heterologous clade C V3 and 1:1300 against clade B V3. Two acutely infected clade B patients developed heterologous clade B V3-specific Nabs at titers of 1:300 and 1:1800 by 13 weeks of infection and 1:5000 and 1:11000 by 7 months of infection. Titers were not different against chimeras containing autologous clade B V3 sequences. Each of 10 uninfected normal human volunteers who were immunized with clade B HIV-1 Env immunogens, but none of five sham immunized control subjects, developed V3-specific Nabs titers as high as 1:3000 (median 1:1300; range 1:700-1:3000). None of the HIV-1 infected or vaccinated subjects had antibodies that neutralized primary HIV-1 virus strains. These results indicate that high-titer, broadly reactive V3-specific antibodies are among the first to be elicited during acute and early HIV-1 infection and following vaccination but these antibodies lack neutralizing potency against primary HIV-1 viruses, which effectively shield V3 from antibody binding to the functional Env trimer.


Journal of Virology | 2011

Isolation of a Monoclonal Antibody That Targets the Alpha-2 Helix of gp120 and Represents the Initial Autologous Neutralizing-Antibody Response in an HIV-1 Subtype C-Infected Individual

Elin S. Gray; M. Anthony Moody; Constantinos Kurt Wibmer; Xi Chen; Dawn J. Marshall; Joshua D. Amos; Penny L. Moore; Andrew Foulger; Jae-Sung Yu; Bronwen E. Lambson; Salim Safurdeen. Abdool Karim; John Whitesides; Georgia D. Tomaras; Barton F. Haynes; Lynn Morris; Hua-Xin Liao

ABSTRACT The C3-V4 region is a major target of autologous neutralizing antibodies in HIV-1 subtype C infection. We previously identified a Center for AIDS Program of Research in South Africa (CAPRISA) participant, CAP88, who developed a potent neutralizing-antibody response within 3 months of infection that targeted an epitope in the C3 region of the HIV-1 envelope (P. L. Moore et al., PLoS Pathog. 5:e1000598, 2009). Here we showed that these type-specific antibodies could be adsorbed using recombinant gp120 from the transmitted/founder virus from CAP88 but not by gp120 made from other isolates. Furthermore, this activity could be depleted using a chimeric gp120 protein that contained only the C3 region from the CAP88 viral envelope engrafted onto the unrelated CAP63 viral envelope (called 63-88C3). On the basis of this, a differential sorting of memory B cells was performed using gp120s made from 63-88C3 and CAP63 labeled with different fluorochromes as positive and negative probes, respectively. This strategy resulted in the isolation of a highly specific monoclonal antibody (MAb), called CAP88-CH06, that neutralized the CAP88 transmitted/founder virus and viruses from acute infection but was unable to neutralize CAP88 viruses isolated at 6 and 12 months postinfection. The latter viruses contained 2 amino acid changes in the alpha-2 helix of C3 that mediated escape from this MAb. One of these changes involved the introduction of an N-linked glycan at position 339 that occluded the epitope, while the other mutation (either E343K or E350K) was a charge change. Our data validate the use of differential sorting to isolate a MAb targeting a specific epitope in the envelope glycoprotein and provided insights into the mechanisms of autologous neutralization escape.


Archive | 2007

Entry inhibition of HIV-1 subtype C isolates

Lynn Morris; Mia Coetzer; Elin S. Gray; Tonie Cilliers; Kabamba B. Alexandre; Penny L. Moore; James M. Binley

The HIV shows an extraordinary degree of genetic diversity that in rare cases impacts on the efficacy of currently available anti-retroviral therapies [1]. There is less information on how genetic variability might affect the efficacy of a newer class of anti-retrovirals, the entry or fusion inhibitors. This group comprises a diverse collection of compounds that target both viral and host cell components blocking virus attachment and/or fusion and preventing infection and viral integration. Available data suggests that almost all genetic subtypes of HIV-1 engage the CD4 and coreceptor molecules. Thus entry inhibitors that target cellular proteins are likely to be equally efficacious across genetic subtypes. However, the efficacy of those that target the viral envelope glycoprotein is likely to be more impacted, given that as the envelope gene is the most variable of all HIV genes, showing up to 30% difference within HIV-1 and up to 55% between HIV-1 and HIV-2 (www.hiv.lanl.gov). Most entry inhibitors have been designed and tested based on HIV-1 subtype B viruses, and would therefore be expected to be most effective against viruses of this genetic subtype. There are few studies that have specifically explored the phenotypic sensitivity of different HIV subtypes to entry inhibitors. Since HIV-1 subtype C is now the most prevalent subtype globally, causing explosive epidemics in southern Africa, Ethiopia, India and China, we focus our review primarily on viruses of this subtype.


Retrovirology | 2012

HIV-1 subtype C primary isolates exhibit high sensitivity to an anti-gp120 RNA aptamer

Hazel T Mufhandu; Kabamba B. Alexandre; Elin S. Gray; Lynn Morris; Makobetsa Khati

Copyright: 2012 Mufhandu et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Retrovirology | 2012

Affinity maturation pathway of an anti-MPER neutralizing mAb, CAP206-CH12

Nancy Tumba; Elin S. Gray; Bronwen E. Lambson; S. S. Abdool Karim; Hua-Xin Liao; Barton F. Haynes; Munir Alam; Lynn Morris

Background The membrane proximal external region (MPER) of HIV-1 is an important target of broadly cross-reactive mAbs. CAPRISA participant, CAP206, developed antiMPER antibodies early that became cross-neutralizing at 18 months post-infection. This coincided with neutralization of the C4 HIV-2/HIV-1 chimera containing the W670 residue, suggesting changes in an antibody paratope may have resulted in the acquisition of breadth. A neutralizing mAb (CAP206-CH12) was isolated, providing an opportunity to determine which somatic hypermutations contribute to breadth. Methods The putative CAP206-CH12 reverted unmutated ancestor (RUA) was inferred using SoDA (http://www.dulci.org). Batch transient transfections were used to generate recombinant antibodies. Neutralization breadth and potency was tested against autologous, Tier 2, and HIV-2/HIV-1 MPER chimeric viruses using the TZM-bl assay. Binding to MPER peptides was assessed by ELISA and surface plasmon resonance (SPR). Results CAP206-CH12_RUA bound the MPER.03 peptide with a Kd of 120nM, 15-fold weaker than CAP206-CH12 binding, but had no neutralizing activity. Since CAP206CH12 and its RUA differed by 19 residues in the heavy chain and 9 in the light chain, we designed an intermediate precursor (IP) where changes near the CDRs in CAP206-CH12 were reverted back to the germline sequence (11 in the heavy and 5 in the light chain). This CAP206-CH12_IP did not neutralize the C4 chimera suggesting that changes responsible for the affinitymatured CAP206-CH12 neutralizing capacity were among these 16 residues. Chimeric pairs of the light chain IP with the heavy chain of CAP206-CH12 or CAP206-CH12_RUA showed binding to MPER with differences in binding kinetics. Conclusion The reduced binding and neutralizing activity of CAP206CH12_RUA and CAP206-CH12_IP compared to CAP206CH12 suggests a correlation between affinity maturation, neutralization breadth and potency. Ongoing work will assess the affinity maturation of CAP206-CH12 by determining moieties in the antibody paratope associated with effective epitope recognition and the effects of somatic mutations on the evolution of neutralization.


Retrovirology | 2012

Isolation of a clonal lineage of IgA broadly neutralizing antibodies from a chronically infected Tanzanian subject

Ma Moody; Drinker; Thaddeus C. Gurley; Joshua D. Amos; Joshua Eudailey; Lc Armand; Robert Parks; Elin S. Gray; Lynn Morris; Andrés Finzi; Xinzhen Yang; Joseph Sodroski; H Liao; Georgia D. Tomaras; David C. Montefiori; Barton F. Haynes

Background Only ~20% of HIV-1-infected subjects develop broadly neutralizing antibodies (bnAbs) and the origins of such antibodies remains obscure. To date, all isolated bnAbs have been of the IgG isotype. Methods Memory B cells from a chronically infected Tanzanian subject with plasma broad neutralizing activity were labeled with a consensus C envelope (Env) and Env+ cells sorted as single cells. Immunoglobulin heavy and light chain genes were amplified by PCR and analyzed for gene usage and isotype, and then were expressed as recombinant monoclonal antibodies (mAbs). MAbs were assayed for binding to Env proteins and for neutralization of multiple HIV-1 strains. Results We isolated 13 mAbs that bound Env proteins; of these, 11 were members of three clonal lineages, each of which spanned two time points. Six mAbs in one lineage were all IgG1, used V H 1~69, had an average mutation frequency of 12%, mapped to the CD4 contact region, and had modest neutralization activity. In contrast, three mAbs in a second lineage had two members that were IgA2, used V H3~66, had an average mutation frequency of 16%, and neutralized 11/27 (41%) of tier 2 pseudoviruses tested. Conclusion


Retrovirology | 2012

Antibody lineages with evidence of somatic hypermutation persisting for >4 years in a South African subject with broad neutralizing activity

Ma Moody; Ashley M. Trama; Mattia Bonsignori; Chun-Yen Tsao; Drinker; Thaddeus C. Gurley; Joshua D. Amos; Joshua Eudailey; Lc Armand; Robert Parks; Krissey E. Lloyd; S Wang; Katie Seo; J Lee; Kj Jackson; Ramona A. Hoh; T Pham; Krishna M. Roskin; Scott D. Boyd; Andrew Fire; Elin S. Gray; Lynn Morris; H Liao; Georgia D. Tomaras; Thomas B. Kepler; Garnett Kelsoe; Barton F. Haynes

Antibody lineages with evidence of somatic hypermutation persisting for >4 years in a South African subject with broad neutralizing activity M Moody, AM Trama, M Bonsignori, C Tsao, MS Drinker, TC Gurley, JD Amos, JA Eudailey, LC Armand, R Parks, KE Lloyd, S Wang, K Seo, J Lee, KJ Jackson, R Hoh, T Pham, KM Roskin, SD Boyd, AZ Fire, ES Gray, L Morris, H Liao, GD Tomaras, TB Kepler, G Kelsoe, BF Haynes


Retrovirology | 2009

P09-04. Charge changes in the alpha2-helix in the C3 region of the HIV-1 subtype C envelope mediate neutralization escape

Penny L. Moore; Nthabeleng Ranchobe; Bronwen E. Lambson; Elin S. Gray; Koleka Mlisana; S. S. Abdool Karim; Carolyn Williamson; S. Gnanakaran; Lynn Morris

Results Escape in CAP88 was mediated by 2 of 3 amino acid changes in the alpha2-helix by 6 months post-infection An I339N mutation resulted in the formation of a predicted N-linked glycan (PNG) suggesting possible glycan shielding. Sequences also contained either an E343K or an E350K substitution resulting in charge switches from negative to positive residues. Modeling of these non-contiguous changes onto helical wheel diagrams showed both residues were in close physical proximity to I339N. In CAP177, neutralization escape was not associated with changes in the number of PNGs. Three of the 5 changes observed, E337K, Q344K and E351K all lay on the same face of the alpha2-helix. However all 5 substitutions were charge changes from neutral or negatively-charged residues to positively-charged residues. In CAP206, two of the three changes observed in escaped clones (though mutagenesis data was not available) were substitutions also resulting in positive charges, G348R and N351K.

Collaboration


Dive into the Elin S. Gray's collaboration.

Top Co-Authors

Avatar

Lynn Morris

Centre for the AIDS Programme of Research in South Africa

View shared research outputs
Top Co-Authors

Avatar

Penny L. Moore

University of the Witwatersrand

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bronwen E. Lambson

National Health Laboratory Service

View shared research outputs
Top Co-Authors

Avatar

Koleka Mlisana

University of KwaZulu-Natal

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge