Network


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

Hotspot


Dive into the research topics where Maria G. Salazar is active.

Publication


Featured researches published by Maria G. Salazar.


Nature | 2003

Antibody neutralization and escape by HIV-1

Xiping Wei; Julie M. Decker; Shuyi Wang; Huxiong Hui; John C. Kappes; Xiaoyun Wu; Jesus F. Salazar-Gonzalez; Maria G. Salazar; J. Michael Kilby; Michael S. Saag; Natalia L. Komarova; Martin A. Nowak; Beatrice H. Hahn; Peter D. Kwong; George M. Shaw

Neutralizing antibodies (Nab) are a principal component of an effective human immune response to many pathogens, yet their role in HIV-1 infection is unclear. To gain a better understanding of this role, we examined plasma from patients with acute HIV infection. Here we report the detection of autologous Nab as early as 52 days after detection of HIV-specific antibodies. The viral inhibitory activity of Nab resulted in complete replacement of neutralization-sensitive virus by successive populations of resistant virus. Escape virus contained mutations in the env gene that were unexpectedly sparse, did not map generally to known neutralization epitopes, and involved primarily changes in N-linked glycosylation. This pattern of escape, and the exceptional density of HIV-1 envelope glycosylation generally, led us to postulate an evolving ‘glycan shield’ mechanism of neutralization escape whereby selected changes in glycan packing prevent Nab binding but not receptor binding. Direct support for this model was obtained by mutational substitution showing that Nab-selected alterations in glycosylation conferred escape from both autologous antibody and epitope-specific monoclonal antibodies. The evolving glycan shield thus represents a new mechanism contributing to HIV-1 persistence in the face of an evolving antibody repertoire.


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 | 2008

Deciphering Human Immunodeficiency Virus Type 1 Transmission and Early Envelope Diversification by Single-Genome Amplification and Sequencing

Jesus F. Salazar-Gonzalez; Elizabeth Bailes; Kimmy T. Pham; Maria G. Salazar; M. Brad Guffey; Brandon F. Keele; Cynthia A. Derdeyn; Paul Farmer; Eric Hunter; Susan Allen; Olivier Manigart; Joseph Mulenga; Jeffrey A. Anderson; Ronald Swanstrom; Barton F. Haynes; Gayathri Athreya; Bette T. Korber; Paul M. Sharp; George M. Shaw; Beatrice H. Hahn

ABSTRACT Accurate identification of the transmitted virus and sequences evolving from it could be instrumental in elucidating the transmission of human immunodeficiency virus type 1 (HIV-1) and in developing vaccines, drugs, or microbicides to prevent infection. Here we describe an experimental approach to analyze HIV-1 env genes as intact genetic units amplified from plasma virion RNA by single-genome amplification (SGA), followed by direct sequencing of uncloned DNA amplicons. We show that this strategy precludes in vitro artifacts caused by Taq-induced nucleotide substitutions and template switching, provides an accurate representation of the env quasispecies in vivo, and has an overall error rate (including nucleotide misincorporation, insertion, and deletion) of less than 8 × 10−5. Applying this method to the analysis of virus in plasma from 12 Zambian subjects from whom samples were obtained within 3 months of seroconversion, we show that transmitted or early founder viruses can be identified and that molecular pathways and rates of early env diversification can be defined. Specifically, we show that 8 of the 12 subjects were each infected by a single virus, while 4 others acquired more than one virus; that the rate of virus evolution in one subject during an 80-day period spanning seroconversion was 1.7 × 10−5 substitutions per site per day; and that evidence of strong immunologic selection can be seen in Env and overlapping Rev sequences based on nonrandom accumulation of nonsynonymous mutations. We also compared the results of the SGA approach with those of more-conventional bulk PCR amplification methods performed on the same patient samples and found that the latter is associated with excessive rates of Taq-induced recombination, nucleotide misincorporation, template resampling, and cloning bias. These findings indicate that HIV-1 env genes, other viral genes, and even full-length viral genomes responsible for productive clinical infection can be identified by SGA analysis of plasma virus sampled at intervals typical in large-scale vaccine trials and that pathways of viral diversification and immune escape can be determined accurately.


Journal of Virology | 2006

Genetic and Neutralization Properties of Subtype C Human Immunodeficiency Virus Type 1 Molecular env Clones from Acute and Early Heterosexually Acquired Infections in Southern Africa

Ming Li; Jesus F. Salazar-Gonzalez; Cynthia A. Derdeyn; Lynn Morris; Carolyn Williamson; James E. Robinson; Julie M. Decker; Yingying Li; Maria G. Salazar; Victoria R. Polonis; Koleka Mlisana; Salim Safurdeen. Abdool Karim; Kunxue Hong; Kelli M. Greene; Miroslawa Bilska; Jintao Zhou; Susan Allen; Elwyn Chomba; Joseph Mulenga; Cheswa Vwalika; Feng Gao; Ming Zhang; Bette Korber; Eric Hunter; Beatrice H. Hahn; David C. Montefiori

ABSTRACT A standard panel of subtype C human immunodeficiency virus type 1 (HIV-1) Env-pseudotyped viruses was created by cloning, sequencing, and characterizing functional gp160 genes from 18 acute and early heterosexually acquired infections in South Africa and Zambia. In general, the gp120 region of these clones was shorter (most evident in V1 and V4) and less glycosylated compared to newly transmitted subtype B viruses, and it was underglycosylated but no different in length compared to chronic subtype C viruses. The gp120s also exhibited low amino acid sequence variability (12%) in V3 and high variability (39%) immediately downstream of V3, a feature shared with newly transmitted subtype B viruses and chronic viruses of both subtypes. When tested as Env-pseudotyped viruses in a luciferase reporter gene assay, all clones possessed an R5 phenotype and resembled primary isolates in their sensitivity to neutralization by HIV-1-positive plasmas. Results obtained with a multisubtype plasma panel suggested partial subtype preference in the neutralizing antibody response to infection. The clones were typical of subtype C in that all were resistant to 2G12 (associated with loss of N-glycosylation at position 295) and most were resistant to 2F5, but all were sensitive to 4E10 and many were sensitive to immunoglobulin G1b12. Finally, conserved neutralization epitopes in the CD4-induced coreceptor binding domain of gp120 were poorly accessible and were difficult to induce and stabilize with soluble CD4 on Env-pseudotyped viruses. These results illustrate key genetic and antigenic properties of subtype C HIV-1 that might impact the design and testing of candidate vaccines. A subset of these gp160 clones are suitable for use as reference reagents to facilitate standardized assessments of vaccine-elicited neutralizing antibody responses.


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.


Journal of Virology | 2012

Generation of Transmitted/Founder HIV-1 Infectious Molecular Clones and Characterization of Their Replication Capacity in CD4 T Lymphocytes and Monocyte-Derived Macrophages

Christina Ochsenbauer; Tara G. Edmonds; Haitao Ding; Brandon F. Keele; Julie M. Decker; Maria G. Salazar; Jesus F. Salazar-Gonzalez; Robin J. Shattock; Barton F. Haynes; George M. Shaw; Beatrice H. Hahn; John C. Kappes

ABSTRACT Genome sequences of transmitted/founder (T/F) HIV-1 have been inferred by analyzing single genome amplicons of acute infection plasma viral RNA in the context of a mathematical model of random virus evolution; however, few of these T/F sequences have been molecularly cloned and biologically characterized. Here, we describe the derivation and biological analysis of ten infectious molecular clones, each representing a T/F genome responsible for productive HIV-1 clade B clinical infection. Each of the T/F viruses primarily utilized the CCR5 coreceptor for entry and replicated efficiently in primary human CD4+ T lymphocytes. This result supports the conclusion that single genome amplification-derived sequences from acute infection allow for the inference of T/F viral genomes that are consistently replication competent. Studies with monocyte-derived macrophages (MDM) demonstrated various levels of replication among the T/F viruses. Although all T/F viruses replicated in MDM, the overall replication efficiency was significantly lower compared to prototypic “highly macrophage-tropic” virus strains. This phenotype was transferable by expressing the env genes in an isogenic proviral DNA backbone, indicating that T/F virus macrophage tropism mapped to Env. Furthermore, significantly higher concentrations of soluble CD4 were required to inhibit T/F virus infection compared to prototypic macrophage-tropic virus strains. Our findings suggest that the acquisition of clinical HIV-1 subtype B infection occurs by mucosal exposure to virus that is not highly macrophage tropic and that the generation and initial biological characterization of 10 clade B T/F infectious molecular clones provides new opportunities to probe virus-host interactions involved in HIV-1 transmission.


PLOS Pathogens | 2012

Transmitted/Founder and Chronic Subtype C HIV-1 Use CD4 and CCR5 Receptors with Equal Efficiency and Are Not Inhibited by Blocking the Integrin α4β7

Nicholas F. Parrish; Craig B. Wilen; Lauren B. Banks; Shilpa S. Iyer; Jennifer M. Pfaff; Jesus F. Salazar-Gonzalez; Maria G. Salazar; Julie M. Decker; Erica H. Parrish; Anna Berg; Jennifer Hopper; Bhavna Hora; Amit Kumar; Tatenda Mahlokozera; Sally Yuan; Charl Coleman; Marion Vermeulen; Haitao Ding; Christina Ochsenbauer; John C. Tilton; Sallie R. Permar; John C. Kappes; Michael R. Betts; Michael P. Busch; Feng Gao; David C. Montefiori; Barton F. Haynes; George M. Shaw; Beatrice H. Hahn; Robert W. Doms

Sexual transmission of human immunodeficiency virus type 1 (HIV-1) most often results from productive infection by a single transmitted/founder (T/F) virus, indicating a stringent mucosal bottleneck. Understanding the viral traits that overcome this bottleneck could have important implications for HIV-1 vaccine design and other prevention strategies. Most T/F viruses use CCR5 to infect target cells and some encode envelope glycoproteins (Envs) that contain fewer potential N-linked glycosylation sites and shorter V1/V2 variable loops than Envs from chronic viruses. Moreover, it has been reported that the gp120 subunits of certain transmitted Envs bind to the gut-homing integrin α4β7, possibly enhancing virus entry and cell-to-cell spread. Here we sought to determine whether subtype C T/F viruses, which are responsible for the majority of new HIV-1 infections worldwide, share biological properties that increase their transmission fitness, including preferential α4β7 engagement. Using single genome amplification, we generated panels of both T/F (n = 20) and chronic (n = 20) Env constructs as well as full-length T/F (n = 6) and chronic (n = 4) infectious molecular clones (IMCs). We found that T/F and chronic control Envs were indistinguishable in the efficiency with which they used CD4 and CCR5. Both groups of Envs also exhibited the same CD4+ T cell subset tropism and showed similar sensitivity to neutralization by CD4 binding site (CD4bs) antibodies. Finally, saturating concentrations of anti-α4β7 antibodies failed to inhibit infection and replication of T/F as well as chronic control viruses, although the growth of the tissue culture-adapted strain SF162 was modestly impaired. These results indicate that the population bottleneck associated with mucosal HIV-1 acquisition is not due to the selection of T/F viruses that use α4β7, CD4 or CCR5 more efficiently.


Journal of Virology | 2013

Comparison of Viral Env Proteins from Acute and Chronic Infections with Subtype C Human Immunodeficiency Virus Type 1 Identifies Differences in Glycosylation and CCR5 Utilization and Suggests a New Strategy for Immunogen Design

Li Hua Ping; Sarah Joseph; Jeffrey A. Anderson; Melissa Rose Abrahams; Jesus F. Salazar-Gonzalez; Laura P. Kincer; Florette K. Treurnicht; Leslie Arney; Suany Ojeda; Ming Zhang; Jessica Keys; E. Lake Potter; Haitao Chu; Penny L. Moore; Maria G. Salazar; Shilpa S. Iyer; Cassandra B. Jabara; Jennifer L. Kirchherr; Clement Mapanje; Nobubelo Ngandu; Cathal Seoighe; Irving Hoffman; Feng Gao; Yuyang Tang; Celia C. LaBranche; Benhur Lee; Andrew Saville; Marion Vermeulen; Susan A. Fiscus; Lynn Morris

ABSTRACT Understanding human immunodeficiency virus type 1 (HIV-1) transmission is central to developing effective prevention strategies, including a vaccine. We compared phenotypic and genetic variation in HIV-1 env genes from subjects in acute/early infection and subjects with chronic infections in the context of subtype C heterosexual transmission. We found that the transmitted viruses all used CCR5 and required high levels of CD4 to infect target cells, suggesting selection for replication in T cells and not macrophages after transmission. In addition, the transmitted viruses were more likely to use a maraviroc-sensitive conformation of CCR5, perhaps identifying a feature of the target T cell. We confirmed an earlier observation that the transmitted viruses were, on average, modestly underglycosylated relative to the viruses from chronically infected subjects. This difference was most pronounced in comparing the viruses in acutely infected men to those in chronically infected women. These features of the transmitted virus point to selective pressures during the transmission event. We did not observe a consistent difference either in heterologous neutralization sensitivity or in sensitivity to soluble CD4 between the two groups, suggesting similar conformations between viruses from acute and chronic infection. However, the presence or absence of glycosylation sites had differential effects on neutralization sensitivity for different antibodies. We suggest that the occasional absence of glycosylation sites encoded in the conserved regions of env, further reduced in transmitted viruses, could expose specific surface structures on the protein as antibody targets.

Collaboration


Dive into the Maria G. Salazar's collaboration.

Top Co-Authors

Avatar

Jesus F. Salazar-Gonzalez

University of Alabama at Birmingham

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
Top Co-Authors

Avatar

Julie M. Decker

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerald H. Learn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Bette T. Korber

Los Alamos National Laboratory

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge