Network


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

Hotspot


Dive into the research topics where Paul A. Goepfert is active.

Publication


Featured researches published by Paul A. Goepfert.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection

Brandon F. Keele; Elena E. Giorgi; Jesus F. Salazar-Gonzalez; Julie M. Decker; Kimmy T. Pham; Maria G. Salazar; Chuanxi Sun; Truman Grayson; Shuyi Wang; Hui Li; Xiping Wei; Chunlai Jiang; Jennifer L. Kirchherr; Feng Gao; Jeffery A. Anderson; Li Hua Ping; Ronald Swanstrom; Georgia D. Tomaras; William A. Blattner; Paul A. Goepfert; J. Michael Kilby; Michael S. Saag; Eric Delwart; Michael P. Busch; Myron S. Cohen; David C. Montefiori; Barton F. Haynes; Brian Gaschen; Gayathri Athreya; Ha Y. Lee

The precise identification of the HIV-1 envelope glycoprotein (Env) responsible for productive clinical infection could be instrumental in elucidating the molecular basis of HIV-1 transmission and in designing effective vaccines. Here, we developed a mathematical model of random viral evolution and, together with phylogenetic tree construction, used it to analyze 3,449 complete env sequences derived by single genome amplification from 102 subjects with acute HIV-1 (clade B) infection. Viral env genes evolving from individual transmitted or founder viruses generally exhibited a Poisson distribution of mutations and star-like phylogeny, which coalesced to an inferred consensus sequence at or near the estimated time of virus transmission. Overall, 78 of 102 subjects had evidence of productive clinical infection by a single virus, and 24 others had evidence of productive clinical infection by a minimum of two to five viruses. Phenotypic analysis of transmitted or early founder Envs revealed a consistent pattern of CCR5 dependence, masking of coreceptor binding regions, and equivalent or modestly enhanced resistance to the fusion inhibitor T1249 and broadly neutralizing antibodies compared with Envs from chronically infected subjects. Low multiplicity infection and limited viral evolution preceding peak viremia suggest a finite window of potential vulnerability of HIV-1 to vaccine-elicited immune responses, although phenotypic properties of transmitted Envs pose a formidable defense.


Nature Medicine | 2006

Quantum dot semiconductor nanocrystals for immunophenotyping by polychromatic flow cytometry.

Pratip K. Chattopadhyay; David A. Price; Theresa Harper; Michael R. Betts; Joanne Yu; Emma Gostick; Stephen P. Perfetto; Paul A. Goepfert; Richard A. Koup; Stephen C. De Rosa; Marcel P. Bruchez; Mario Roederer

Immune responses arise from a wide variety of cells expressing unique combinations of multiple cell-surface proteins. Detailed characterization is hampered, however, by limitations in available probes and instrumentation. Here, we use the unique spectral properties of semiconductor nanocrystals (quantum dots) to extend the capabilities of polychromatic flow cytometry to resolve 17 fluorescence emissions. We show the need for this power by analyzing, in detail, the phenotype of multiple antigen-specific T-cell populations, revealing variations within complex phenotypic patterns that would otherwise remain obscure. For example, T cells specific for distinct epitopes from one pathogen, and even those specific for the same epitope, can have markedly different phenotypes. The technology we describe, encompassing the detection of eight quantum dots in conjunction with conventional fluorophores, should expand the horizons of flow cytometry, as well as our ability to characterize the intricacies of both adaptive and innate cellular immune responses.


PLOS Pathogens | 2010

Perforin Expression Directly Ex Vivo by HIV-Specific CD8+ T-Cells Is a Correlate of HIV Elite Control

Adam R. Hersperger; Florencia Pereyra; Martha Nason; Korey Demers; Prameet M. Sheth; Lucy Y. Shin; Colin Kovacs; Benigno Rodriguez; Scott F. Sieg; Leia Teixeira-Johnson; Debbie Gudonis; Paul A. Goepfert; Michael M. Lederman; Ian Frank; George Makedonas; Rupert Kaul; Bruce D. Walker; Michael R. Betts

Many immune correlates of CD8+ T-cell-mediated control of HIV replication, including polyfunctionality, proliferative ability, and inhibitory receptor expression, have been discovered. However, no functional correlates using ex vivo cells have been identified with the known ability to cause the direct elimination of HIV-infected cells. We have recently discovered the ability of human CD8+ T-cells to rapidly upregulate perforin—an essential molecule for cell-mediated cytotoxicity—following antigen-specific stimulation. Here, we examined perforin expression capability in a large cross-sectional cohort of chronically HIV-infected individuals with varying levels of viral load: elite controllers (n = 35), viremic controllers (n = 29), chronic progressors (n = 27), and viremic nonprogressors (n = 6). Using polychromatic flow cytometry and standard intracellular cytokine staining assays, we measured perforin upregulation, cytokine production, and degranulation following stimulation with overlapping peptide pools encompassing all proteins of HIV. We observed that HIV-specific CD8+ T-cells from elite controllers consistently display an enhanced ability to express perforin directly ex vivo compared to all other groups. This ability is not restricted to protective HLA-B haplotypes, does not require proliferation or the addition of exogenous factors, is not restored by HAART, and primarily originates from effector CD8+ T-cells with otherwise limited functional capability. Notably, we found an inverse relationship between HIV-specific perforin expression and viral load. Thus, the capability of HIV-specific CD8+ T-cells to rapidly express perforin defines a novel correlate of control in HIV infection.


Journal of Virology | 2007

Escape and Compensation from Early HLA-B57-Mediated Cytotoxic T-Lymphocyte Pressure on Human Immunodeficiency Virus Type 1 Gag Alter Capsid Interactions with Cyclophilin A

Mark A. Brockman; Arne Schneidewind; Matthew P. Lahaie; Aaron G. Schmidt; Toshiyuki Miura; Ivna DeSouza; Faina Ryvkin; Cynthia A. Derdeyn; Susan Allen; Eric Hunter; Joseph Mulenga; Paul A. Goepfert; Bruce D. Walker; Todd M. Allen

ABSTRACT Certain histocompatibility leukocyte antigen (HLA) alleles are associated with improved clinical outcomes for individuals infected with human immunodeficiency virus type 1 (HIV-1), but the mechanisms for their effects remain undefined. An early CD8+ T-cell escape mutation in the dominant HLA-B57-restricted Gag epitope TW10 (TSTLQEQIGW) has been shown to impair HIV-1 replication capacity in vitro. We demonstrate here that this T242N substitution in the capsid protein is associated with upstream mutations at residues H219, I223, and M228 in the cyclophilin A (CypA)-binding loop in B57+ individuals with progressive disease. In an independent cohort of epidemiologically linked transmission pairs, the presence of these substitutions in viruses encoding T242N was associated with significantly higher plasma viremia in donors, further suggesting that these secondary mutations compensated for the replication defect of T242N. Using NL4-3 constructs, we illustrate the ability of these CypA loop changes to partially restore replication of the T242N variant in vitro. Notably, these mutations also enhanced viral resistance to the drug cyclosporine A, indicating a reduced dependence of the compensated virus on CypA that is normally essential for optimal infectivity. Therefore, mutations in TW10 allow HIV-1 to evade a dominant early CD8+ T-cell response, but the benefits of escape are offset by a defect in capsid function. These data suggest that TW10 escape variants undergo a postentry block that is partially overcome by changes in the CypA-binding loop and identify a mechanism for an HIV-1 fitness defect that may contribute to the slower disease progression associated with HLA-B57.


Journal of Virology | 2007

Compensatory Mutation Partially Restores Fitness and Delays Reversion of Escape Mutation within the Immunodominant HLA-B*5703-Restricted Gag Epitope in Chronic Human Immunodeficiency Virus Type 1 Infection

Hayley Crawford; Julia G. Prado; Alasdair Leslie; Stéphane Hué; Isobella Honeyborne; Sharon Reddy; Mary van der Stok; Zenele Mncube; Christian Brander; Christine Rousseau; James I. Mullins; Richard A. Kaslow; Paul A. Goepfert; Susan Allen; Eric Hunter; Joseph Mulenga; Photini Kiepiela; Bruce D. Walker; Philip J. R. Goulder

ABSTRACT HLA-B*5703 is associated with effective immune control in human immunodeficiency virus type 1 (HIV-1) infection. Here we describe an escape mutation within the immunodominant HLA-B*5703-restricted epitope in chronic HIV-1 infection, KAFSPEVIPMF (Gag 162-172), and demonstrate that this mutation reduces viral replicative capacity. Reversion of this mutation following transmission to HLA-B*5703-negative recipients was delayed by the compensatory mutation S165N within the same epitope. These data may help explain the observed association between HLA-B*5703 and long-term control of viremia.


Journal of Experimental Medicine | 2008

Transmission of HIV-1 Gag immune escape mutations is associated with reduced viral load in linked recipients

Paul A. Goepfert; Wendy Lumm; Paul K. Farmer; Philippa C. Matthews; Andrew J. Prendergast; Jonathan M. Carlson; Cynthia A. Derdeyn; Jianming Tang; Richard A. Kaslow; Anju Bansal; Karina Yusim; David Heckerman; Joseph Mulenga; Susan Allen; Philip J. R. Goulder; Eric Hunter

In a study of 114 epidemiologically linked Zambian transmission pairs, we evaluated the impact of human leukocyte antigen class I (HLA-I)–associated amino acid polymorphisms, presumed to reflect cytotoxic T lymphocyte (CTL) escape in Gag and Nef of the virus transmitted from the chronically infected donor, on the plasma viral load (VL) in matched recipients 6 mo after infection. CTL escape mutations in Gag and Nef were seen in the donors, which were subsequently transmitted to recipients, largely unchanged soon after infection. We observed a significant correlation between the number of Gag escape mutations targeted by specific HLA-B allele–restricted CTLs and reduced VLs in the recipients. This negative correlation was most evident in newly infected individuals, whose HLA alleles were unable to effectively target Gag and select for CTL escape mutations in this gene. Nef mutations in the donor had no impact on VL in the recipient. Thus, broad Gag-specific CTL responses capable of driving virus escape in the donor may be of clinical benefit to both the donor and recipient. In addition to their direct implications for HIV-1 vaccine design, these data suggest that CTL-induced viral polymorphisms and their associated in vivo viral fitness costs could have a significant impact on HIV-1 pathogenesis.


Journal of Experimental Medicine | 2009

Evolution of HLA-B*5703 HIV-1 escape mutations in HLA-B*5703–positive individuals and their transmission recipients

Hayley Crawford; Wendy Lumm; Alasdair Leslie; Malinda Schaefer; Debrah I. Boeras; Julia G. Prado; Jianming Tang; Paul Farmer; Thumbi Ndung'u; Shabir Lakhi; Jill Gilmour; Paul A. Goepfert; Bruce D. Walker; Richard A. Kaslow; Joseph Mulenga; Susan Allen; Philip J. R. Goulder; Eric Hunter

HLA-B*57 is the class I allele most consistently associated with control of human immunodeficiency virus (HIV) replication, which may be linked to the specific HIV peptides that this allele presents to cytotoxic T lymphocytes (CTLs), and the resulting efficacy of these cellular immune responses. In two HIV C clade–infected populations in South Africa and Zambia, we sought to elucidate the role of HLA-B*5703 in HIV disease outcome. HLA-B*5703–restricted CTL responses select for escape mutations in three Gag p24 epitopes, in a predictable order. We show that the accumulation of these mutations sequentially reduces viral replicative capacity in vitro. Despite this, in vivo data demonstrate that there is ultimately an increase in viral load concomitant with evasion of all three HLA-B*5703–restricted CTL responses. In HLA-B*5703–mismatched recipients, the previously described early benefit of transmitted HLA-B*5703–associated escape mutations is abrogated by the increase in viral load coincident with reversion. Rapid disease progression is observed in HLA-matched recipients to whom mutated virus is transmitted. These data demonstrate that, although costly escape from CTL responses can progressively attenuate the virus, high viral loads develop in the absence of adequate, continued CTL responses. These data underline the need for a CTL vaccine against multiple conserved epitopes.


PLOS Medicine | 2009

Polyclonal B cell differentiation and loss of gastrointestinal tract germinal centers in the earliest stages of HIV-1 infection

Marc C. Levesque; M. Anthony Moody; Kwan Ki Hwang; Dawn J. Marshall; John F. Whitesides; Joshua D. Amos; Thaddeus C. Gurley; Sallie D. Allgood; Benjamin B. Haynes; Nathan Vandergrift; Steven G. Plonk; Daniel Parker; Myron S. Cohen; Georgia D. Tomaras; Paul A. Goepfert; George M. Shaw; Jörn E. Schmitz; Joseph J. Eron; Nicholas J. Shaheen; Charles B. Hicks; Hua-Xin Liao; Martin Markowitz; Garnett Kelsoe; David M. Margolis; Barton F. Haynes

Studying the effects of early HIV infection on human antibody responses, M. Anthony Moody and colleagues find rapid polyclonal B cell differentiation and structural damage to gut-associated lymphoid tissue.


Journal of Experimental Medicine | 2011

Initial antibodies binding to HIV-1 gp41 in acutely infected subjects are polyreactive and highly mutated

Hua-Xin Liao; Xi Chen; Supriya Munshaw; Ruijun Zhang; Dawn J. Marshall; Nathan Vandergrift; John F. Whitesides; Xiaozhi Lu; Jae-Sung Yu; Kwan-Ki Hwang; Feng Gao; Martin Markowitz; Sonya L. Heath; Katharine J. Bar; Paul A. Goepfert; David C. Montefiori; George C. Shaw; S. Munir Alam; David M. Margolis; Thomas N. Denny; Scott D. Boyd; Eleanor Marshal; Michael Egholm; Birgitte B. Simen; Bozena Hanczaruk; Andrew Fire; Gerald Voss; Garnett Kelsoe; Georgia D. Tomaras; M. Anthony Moody

Many HIV-1 envelope-reactive antibodies shortly after HIV-1 transmission may arise from crow-reactive memory B cells previously stimulated by non-HIV-1 host or microbial antigens


Journal of Theoretical Biology | 2009

Modeling Sequence Evolution in Acute HIV-1 Infection

Ha Youn Lee; Elena E. Giorgi; Brandon F. Keele; Brian Gaschen; Gayathri Athreya; Jesus F. Salazar-Gonzalez; Kimmy T. Pham; Paul A. Goepfert; J. Michael Kilby; Michael S. Saag; Eric Delwart; Michael P. Busch; Beatrice H. Hahn; George M. Shaw; Bette T. Korber; Tanmoy Bhattacharya; Alan S. Perelson

We describe a mathematical model and Monte Carlo (MC) simulation of viral evolution during acute infection. We consider both synchronous and asynchronous processes of viral infection of new target cells. The model enables an assessment of the expected sequence diversity in new HIV-1 infections originating from a single transmitted viral strain, estimation of the most recent common ancestor (MRCA) of the transmitted viral lineage, and estimation of the time to coalesce back to the MRCA. We also calculate the probability of the MRCA being the transmitted virus or an evolved variant. Excluding insertions and deletions, we assume HIV-1 evolves by base substitution without selection pressure during the earliest phase of HIV-1 infection prior to the immune response. Unlike phylogenetic methods that follow a lineage backwards to coalescence, we compare the observed data to a model of the diversification of a viral population forward in time. To illustrate the application of these methods, we provide detailed comparisons of the model and simulations results to 306 envelope sequences obtained from eight newly infected subjects at a single time point. The data from 68 patients were in good agreement with model predictions, and hence compatible with a single-strain infection evolving under no selection pressure. The diversity of the samples from the other two patients was too great to be explained by the model, suggesting multiple HIV-1-strains were transmitted. The model can also be applied to longitudinal patient data to estimate within-host viral evolutionary parameters.

Collaboration


Dive into the Paul A. Goepfert's collaboration.

Researchain Logo
Decentralizing Knowledge