Network


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

Hotspot


Dive into the research topics where Elena E. Giorgi is active.

Publication


Featured researches published by Elena E. Giorgi.


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.


Journal of Experimental Medicine | 2009

Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection.

Jesus F. Salazar-Gonzalez; Maria G. Salazar; Brandon F. Keele; Gerald H. Learn; Elena E. Giorgi; Hui Li; Julie M. Decker; Shuyi Wang; Joshua Baalwa; Matthias H. Kraus; Nicholas F. Parrish; Katharina S. Shaw; M. Brad Guffey; Katharine J. Bar; Katie L. Davis; Christina Ochsenbauer-Jambor; John C. Kappes; Michael S. Saag; Myron S. Cohen; Joseph Mulenga; Cynthia A. Derdeyn; Susan Allen; Eric Hunter; Martin Markowitz; Peter Hraber; Alan S. Perelson; Tanmoy Bhattacharya; Barton F. Haynes; Bette T. Korber; Beatrice H. Hahn

Identification of full-length transmitted HIV-1 genomes could be instrumental in HIV-1 pathogenesis, microbicide, and vaccine research by enabling the direct analysis of those viruses actually responsible for productive clinical infection. We show in 12 acutely infected subjects (9 clade B and 3 clade C) that complete HIV-1 genomes of transmitted/founder viruses can be inferred by single genome amplification and sequencing of plasma virion RNA. This allowed for the molecular cloning and biological analysis of transmitted/founder viruses and a comprehensive genome-wide assessment of the genetic imprint left on the evolving virus quasispecies by a composite of host selection pressures. Transmitted viruses encoded intact canonical genes (gag-pol-vif-vpr-tat-rev-vpu-env-nef) and replicated efficiently in primary human CD4+ T lymphocytes but much less so in monocyte-derived macrophages. Transmitted viruses were CD4 and CCR5 tropic and demonstrated concealment of coreceptor binding surfaces of the envelope bridging sheet and variable loop 3. 2 mo after infection, transmitted/founder viruses in three subjects were nearly completely replaced by viruses differing at two to five highly selected genomic loci; by 12–20 mo, viruses exhibited concentrated mutations at 17–34 discrete locations. These findings reveal viral properties associated with mucosal HIV-1 transmission and a limited set of rapidly evolving adaptive mutations driven primarily, but not exclusively, by early cytotoxic T cell responses.


Journal of Experimental Medicine | 2009

The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection

Nilu Goonetilleke; Michael K. P. Liu; Jesus F. Salazar-Gonzalez; Guido Ferrari; Elena E. Giorgi; Vitaly V. Ganusov; Brandon F. Keele; Gerald H. Learn; Emma L. Turnbull; Maria G. Salazar; Kent J. Weinhold; Stephen Moore; Norman L. Letvin; Barton F. Haynes; Myron S. Cohen; Peter Hraber; Tanmoy Bhattacharya; Persephone Borrow; Alan S. Perelson; Beatrice H. Hahn; George M. Shaw; Bette T. Korber; Andrew J. McMichael

Identification of the transmitted/founder virus makes possible, for the first time, a genome-wide analysis of host immune responses against the infecting HIV-1 proteome. A complete dissection was made of the primary HIV-1–specific T cell response induced in three acutely infected patients. Cellular assays, together with new algorithms which identify sites of positive selection in the virus genome, showed that primary HIV-1–specific T cells rapidly select escape mutations concurrent with falling virus load in acute infection. Kinetic analysis and mathematical modeling of virus immune escape showed that the contribution of CD8 T cell–mediated killing of productively infected cells was earlier and much greater than previously recognized and that it contributed to the initial decline of plasma virus in acute infection. After virus escape, these first T cell responses often rapidly waned, leaving or being succeeded by T cell responses to epitopes which escaped more slowly or were invariant. These latter responses are likely to be important in maintaining the already established virus set point. In addition to mutations selected by T cells, there were other selected regions that accrued mutations more gradually but were not associated with a T cell response. These included clusters of mutations in envelope that were targeted by NAbs, a few isolated sites that reverted to the consensus sequence, and bystander mutations in linkage with T cell–driven escape.


Journal of Virology | 2009

Quantitating the Multiplicity of Infection with Human Immunodeficiency Virus Type 1 Subtype C Reveals a Non-Poisson Distribution of Transmitted Variants

Melissa-Rose Abrahams; Jeffrey A. Anderson; Elena E. Giorgi; Cathal Seoighe; Koleka Mlisana; Li-Hua Ping; Gayathri Athreya; Florette K. Treurnicht; Brandon F. Keele; Natasha Wood; Jesus F. Salazar-Gonzalez; Tanmoy Bhattacharya; Haitao Chu; Irving Hoffman; Shannon Galvin; Clement Mapanje; Peter N. Kazembe; R. Thebus; Susan A. Fiscus; Winston Hide; Myron S. Cohen; S. S. Abdool Karim; Barton F. Haynes; George M. Shaw; Beatrice H. Hahn; Bette T. Korber; R. Swanstrom; Carolyn Williamson

ABSTRACT Identifying the specific genetic characteristics of successfully transmitted variants may prove central to the development of effective vaccine and microbicide interventions. Although human immunodeficiency virus transmission is associated with a population bottleneck, the extent to which different factors influence the diversity of transmitted viruses is unclear. We estimate here the number of transmitted variants in 69 heterosexual men and women with primary subtype C infections. From 1,505 env sequences obtained using a single genome amplification approach we show that 78% of infections involved single variant transmission and 22% involved multiple variant transmissions (median of 3). We found evidence for mutations selected for cytotoxic-T-lymphocyte or antibody escape and a high prevalence of recombination in individuals infected with multiple variants representing another potential escape pathway in these individuals. In a combined analysis of 171 subtype B and C transmission events, we found that infection with more than one variant does not follow a Poisson distribution, indicating that transmission of individual virions cannot be seen as independent events, each occurring with low probability. While most transmissions resulted from a single infectious unit, multiple variant transmissions represent a significant fraction of transmission events, suggesting that there may be important mechanistic differences between these groups that are not yet understood.


Journal of Experimental Medicine | 2009

Low-dose rectal inoculation of rhesus macaques by SIVsmE660 or SIVmac251 recapitulates human mucosal infection by HIV-1

Brandon F. Keele; Hui Li; Gerald H. Learn; Peter Hraber; Elena E. Giorgi; Truman Grayson; Chuanxi Sun; Yalu Chen; Wendy W. Yeh; Norman L. Letvin; John R. Mascola; Gary J. Nabel; Barton F. Haynes; Tanmoy Bhattacharya; Alan S. Perelson; Bette Korber; Beatrice H. Hahn; George M. Shaw

We recently developed a novel strategy to identify transmitted HIV-1 genomes in acutely infected humans using single-genome amplification and a model of random virus evolution. Here, we used this approach to determine the molecular features of simian immunodeficiency virus (SIV) transmission in 18 experimentally infected Indian rhesus macaques. Animals were inoculated intrarectally (i.r.) or intravenously (i.v.) with stocks of SIVmac251 or SIVsmE660 that exhibited sequence diversity typical of early-chronic HIV-1 infection. 987 full-length SIV env sequences (median of 48 per animal) were determined from plasma virion RNA 1–5 wk after infection. i.r. inoculation was followed by productive infection by one or a few viruses (median 1; range 1–5) that diversified randomly with near starlike phylogeny and a Poisson distribution of mutations. Consensus viral sequences from ramp-up and peak viremia were identical to viruses found in the inocula or differed from them by only one or a few nucleotides, providing direct evidence that early plasma viral sequences coalesce to transmitted/founder viruses. i.v. infection was >2,000-fold more efficient than i.r. infection, and viruses transmitted by either route represented the full genetic spectra of the inocula. These findings identify key similarities in mucosal transmission and early diversification between SIV and HIV-1, and thus validate the SIV–macaque mucosal infection model for HIV-1 vaccine and microbicide research.


PLOS Pathogens | 2010

High Multiplicity Infection by HIV-1 in Men Who Have Sex with Men.

Hui-Hui Li; Katharine J. Bar; Shuyi Wang; Julie M. Decker; Yalu Chen; Chuanxi Sun; Jesus F. Salazar-Gonzalez; Maria G. Salazar; Gerald H. Learn; Charity J. Morgan; Joseph E. Schumacher; Peter Hraber; Elena E. Giorgi; Tanmoy Bhattacharya; Bette T. Korber; Alan S. Perelson; Joseph J. Eron; Myron S. Cohen; Charles B. Hicks; Barton F. Haynes; Martin Markowitz; Brandon F. Keele; Beatrice H. Hahn; George M. Shaw

Elucidating virus-host interactions responsible for HIV-1 transmission is important for advancing HIV-1 prevention strategies. To this end, single genome amplification (SGA) and sequencing of HIV-1 within the context of a model of random virus evolution has made possible for the first time an unambiguous identification of transmitted/founder viruses and a precise estimation of their numbers. Here, we applied this approach to HIV-1 env analyses in a cohort of acutely infected men who have sex with men (MSM) and found that a high proportion (10 of 28; 36%) had been productively infected by more than one virus. In subjects with multivariant transmission, the minimum number of transmitted viruses ranged from 2 to 10 with viral recombination leading to rapid and extensive genetic shuffling among virus lineages. A combined analysis of these results, together with recently published findings based on identical SGA methods in largely heterosexual (HSX) cohorts, revealed a significantly higher frequency of multivariant transmission in MSM than in HSX [19 of 50 subjects (38%) versus 34 of 175 subjects (19%); Fishers exact p = 0.008]. To further evaluate the SGA strategy for identifying transmitted/founder viruses, we analyzed 239 overlapping 5′ and 3′ half genome or env-only sequences from plasma viral RNA (vRNA) and blood mononuclear cell DNA in an MSM subject who had a particularly well-documented virus exposure history 3–6 days before symptom onset and 14–17 days before peak plasma viremia (47,600,000 vRNA molecules/ml). All 239 sequences coalesced to a single transmitted/founder virus genome in a time frame consistent with the clinical history, and a molecular clone of this genome encoded replication competent virus in accord with model predictions. Higher multiplicity of HIV-1 infection in MSM compared with HSX is consistent with the demonstrably higher epidemiological risk of virus acquisition in MSM and could indicate a greater challenge for HIV-1 vaccines than previously recognized.


PLOS ONE | 2010

Transmission of Single HIV-1 Genomes and Dynamics of Early Immune Escape Revealed by Ultra-Deep Sequencing

Will Fischer; Vitaly V. Ganusov; Elena E. Giorgi; Peter Hraber; Brandon F. Keele; Thomas Leitner; Cliff Han; Cheryl D. Gleasner; Lance D. Green; Chien-Chi Lo; Ambarish Nag; Timothy C. Wallstrom; Shuyi Wang; Andrew J. McMichael; Barton F. Haynes; Beatrice H. Hahn; Alan S. Perelson; Persephone Borrow; George M. Shaw; Tanmoy Bhattacharya; Bette T. Korber

We used ultra-deep sequencing to obtain tens of thousands of HIV-1 sequences from regions targeted by CD8+ T lymphocytes from longitudinal samples from three acutely infected subjects, and modeled viral evolution during the critical first weeks of infection. Previous studies suggested that a single virus established productive infection, but these conclusions were tempered because of limited sampling; now, we have greatly increased our confidence in this observation through modeling the observed earliest sample diversity based on vastly more extensive sampling. Conventional sequencing of HIV-1 from acute/early infection has shown different patterns of escape at different epitopes; we investigated the earliest escapes in exquisite detail. Over 3–6 weeks, ultradeep sequencing revealed that the virus explored an extraordinary array of potential escape routes in the process of evading the earliest CD8 T-lymphocyte responses – using 454 sequencing, we identified over 50 variant forms of each targeted epitope during early immune escape, while only 2–7 variants were detected in the same samples via conventional sequencing. In contrast to the diversity seen within epitopes, non-epitope regions, including the Envelope V3 region, which was sequenced as a control in each subject, displayed very low levels of variation. In early infection, in the regions sequenced, the consensus forms did not have a fitness advantage large enough to trigger reversion to consensus amino acids in the absence of immune pressure. In one subject, a genetic bottleneck was observed, with extensive diversity at the second time point narrowing to two dominant escape forms by the third time point, all within two months of infection. Traces of immune escape were observed in the earliest samples, suggesting that immune pressure is present and effective earlier than previously reported; quantifying the loss rate of the founder virus suggests a direct role for CD8 T-lymphocyte responses in viral containment after peak viremia. Dramatic shifts in the frequencies of epitope variants during the first weeks of infection revealed a complex interplay between viral fitness and immune escape.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Phenotypic properties of transmitted founder HIV-1

Nicholas F. Parrish; Feng Gao; Hui Li; Elena E. Giorgi; Hannah J. Barbian; Erica H. Parrish; Lara Zajic; Shilpa S. Iyer; Julie M. Decker; Amit Kumar; Bhavna Hora; Anna Berg; Fangping Cai; Jennifer Hopper; Thomas N. Denny; Hairao Ding; Christina Ochsenbauer; John C. Kappes; Rachel P. Galimidi; Anthony P. West; Pamela J. Bjorkman; Craig B. Wilen; Robert W. Doms; Meagan O'Brien; Nina Bhardwaj; Persephone Borrow; Barton F. Haynes; Mark Muldoon; James Theiler; Bette T. Korber

Defining the virus–host interactions responsible for HIV-1 transmission, including the phenotypic requirements of viruses capable of establishing de novo infections, could be important for AIDS vaccine development. Previous analyses have failed to identify phenotypic properties other than chemokine receptor 5 (CCR5) and CD4+ T-cell tropism that are preferentially associated with viral transmission. However, most of these studies were limited to examining envelope (Env) function in the context of pseudoviruses. Here, we generated infectious molecular clones of transmitted founder (TF; n = 27) and chronic control (CC; n = 14) viruses of subtypes B (n = 18) and C (n = 23) and compared their phenotypic properties in assays specifically designed to probe the earliest stages of HIV-1 infection. We found that TF virions were 1.7-fold more infectious (P = 0.049) and contained 1.9-fold more Env per particle (P = 0.048) compared with CC viruses. TF viruses were also captured by monocyte-derived dendritic cells 1.7-fold more efficiently (P = 0.035) and more readily transferred to CD4+ T cells (P = 0.025). In primary CD4+ T cells, TF and CC viruses replicated with comparable kinetics; however, when propagated in the presence of IFN-α, TF viruses replicated to higher titers than CC viruses. This difference was significant for subtype B (P = 0.000013) but not subtype C (P = 0.53) viruses, possibly reflecting demographic differences of the respective patient cohorts. Together, these data indicate that TF viruses are enriched for higher Env content, enhanced cell-free infectivity, improved dendritic cell interaction, and relative IFN-α resistance. These viral properties, which likely act in concert, should be considered in the development and testing of AIDS vaccines.


Cell | 2013

Protective Efficacy of a Global HIV-1 Mosaic Vaccine against Heterologous SHIV Challenges in Rhesus Monkeys

Dan H. Barouch; Kathryn E. Stephenson; Erica N. Borducchi; Kaitlin M. Smith; Kelly Stanley; Anna McNally; Jinyan Liu; Peter Abbink; Lori F. Maxfield; Michael S. Seaman; Anne-Sophie Dugast; Galit Alter; Melissa Ferguson; Wenjun Li; Patricia L. Earl; Bernard Moss; Elena E. Giorgi; James Szinger; Leigh Anne Eller; Erik Billings; Mangala Rao; Sodsai Tovanabutra; Eric Sanders-Buell; Mo Weijtens; Maria G. Pau; Hanneke Schuitemaker; Merlin L. Robb; Jerome H. Kim; Bette T. Korber; Nelson L. Michael

The global diversity of HIV-1 represents a critical challenge facing HIV-1 vaccine development. HIV-1 mosaic antigens are bioinformatically optimized immunogens designed for improved coverage of HIV-1 diversity. However, the protective efficacy of such global HIV-1 vaccine antigens has not previously been evaluated. Here, we demonstrate the capacity of bivalent HIV-1 mosaic antigens to protect rhesus monkeys against acquisition of infection following heterologous challenges with the difficult-to-neutralize simian-human immunodeficiency virus SHIV-SF162P3. Adenovirus/poxvirus and adenovirus/adenovirus vector-based vaccines expressing HIV-1 mosaic Env, Gag, and Pol afforded a significant reduction in the per-exposure acquisition risk following repetitive, intrarectal SHIV-SF162P3 challenges. Protection against acquisition of infection correlated with vaccine-elicited binding, neutralizing, and functional nonneutralizing antibodies, suggesting that the coordinated activity of multiple antibody functions may contribute to protection against difficult-to-neutralize viruses. These data demonstrate the protective efficacy of HIV-1 mosaic antigens and suggest a potential strategy for the development of a global HIV-1 vaccine. PAPERCLIP:


PLOS Pathogens | 2009

HIV Evolution in Early Infection: Selection Pressures, Patterns of Insertion and Deletion, and the Impact of APOBEC

Natasha Wood; Tanmoy Bhattacharya; Brandon F. Keele; Elena E. Giorgi; Michael Liu; Brian Gaschen; Marcus Daniels; Guido Ferrari; Barton F. Haynes; Andrew J. McMichael; George M. Shaw; Beatrice H. Hahn; Bette T. Korber; Cathal Seoighe

The pattern of viral diversification in newly infected individuals provides information about the host environment and immune responses typically experienced by the newly transmitted virus. For example, sites that tend to evolve rapidly across multiple early-infection patients could be involved in enabling escape from common early immune responses, could represent adaptation for rapid growth in a newly infected host, or could represent reversion from less fit forms of the virus that were selected for immune escape in previous hosts. Here we investigated the diversification of HIV-1 env coding sequences in 81 very early B subtype infections previously shown to have resulted from transmission or expansion of single viruses (n = 78) or two closely related viruses (n = 3). In these cases, the sequence of the infecting virus can be estimated accurately, enabling inference of both the direction of substitutions as well as distinction between insertion and deletion events. By integrating information across multiple acutely infected hosts, we find evidence of adaptive evolution of HIV-1 env and identify a subset of codon sites that diversified more rapidly than can be explained by a model of neutral evolution. Of 24 such rapidly diversifying sites, 14 were either i) clustered and embedded in CTL epitopes that were verified experimentally or predicted based on the individuals HLA or ii) in a nucleotide context indicative of APOBEC-mediated G-to-A substitutions, despite having excluded heavily hypermutated sequences prior to the analysis. In several cases, a rapidly evolving site was embedded both in an APOBEC motif and in a CTL epitope, suggesting that APOBEC may facilitate early immune escape. Ten rapidly diversifying sites could not be explained by CTL escape or APOBEC hypermutation, including the most frequently mutated site, in the fusion peptide of gp41. We also examined the distribution, extent, and sequence context of insertions and deletions, and we provide evidence that the length variation seen in hypervariable loop regions of the envelope glycoprotein is a consequence of selection and not of mutational hotspots. Our results provide a detailed view of the process of diversification of HIV-1 following transmission, highlighting the role of CTL escape and hypermutation in shaping viral evolution during the establishment of new infections.

Collaboration


Dive into the Elena E. Giorgi's collaboration.

Top Co-Authors

Avatar

Bette T. Korber

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Tanmoy Bhattacharya

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Beatrice H. Hahn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

George M. Shaw

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Peter Hraber

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alan S. Perelson

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar

Hui Li

University of Alabama

View shared research outputs
Top Co-Authors

Avatar

Gerald H. Learn

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge