Network


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

Hotspot


Dive into the research topics where Genevieve G. Fouda is active.

Publication


Featured researches published by Genevieve G. Fouda.


Journal of Virology | 2008

Initial B-Cell Responses to Transmitted Human Immunodeficiency Virus Type 1: Virion-Binding Immunoglobulin M (IgM) and IgG Antibodies Followed by Plasma Anti-gp41 Antibodies with Ineffective Control of Initial Viremia

Georgia D. Tomaras; Nicole L. Yates; Pinghuang Liu; Li Qin; Genevieve G. Fouda; Leslie L. Chavez; Allan C. deCamp; Robert Parks; Vicki C Ashley; Judith T. Lucas; Myron S. Cohen; Joseph J. Eron; Charles B. Hicks; Hua-Xin Liao; Steven G. Self; Gary Landucci; Donald N. Forthal; Kent J. Weinhold; Brandon F. Keele; Beatrice H. Hahn; Michael L. Greenberg; Lynn Morris; Salim Safurdeen. Abdool Karim; William A. Blattner; David C. Montefiori; George M. Shaw; Alan S. Perelson; Barton F. Haynes

ABSTRACT A window of opportunity for immune responses to extinguish human immunodeficiency virus type 1 (HIV-1) exists from the moment of transmission through establishment of the latent pool of HIV-1-infected cells. A critical time to study the initial immune responses to the transmitted/founder virus is the eclipse phase of HIV-1 infection (time from transmission to the first appearance of plasma virus), but, to date, this period has been logistically difficult to analyze. To probe B-cell responses immediately following HIV-1 transmission, we have determined envelope-specific antibody responses to autologous and consensus Envs in plasma donors from the United States for whom frequent plasma samples were available at time points immediately before, during, and after HIV-1 plasma viral load (VL) ramp-up in acute infection, and we have modeled the antibody effect on the kinetics of plasma viremia. The first detectable B-cell response was in the form of immune complexes 8 days after plasma virus detection, whereas the first free plasma anti-HIV-1 antibody was to gp41 and appeared 13 days after the appearance of plasma virus. In contrast, envelope gp120-specific antibodies were delayed an additional 14 days. Mathematical modeling of the earliest viral dynamics was performed to determine the impact of antibody on HIV replication in vivo as assessed by plasma VL. Including the initial anti-gp41 immunoglobulin G (IgG), IgM, or both responses in the model did not significantly impact the early dynamics of plasma VL. These results demonstrate that the first IgM and IgG antibodies induced by transmitted HIV-1 are capable of binding virions but have little impact on acute-phase viremia at the timing and magnitude that they occur in natural infection.


The Journal of Infectious Diseases | 2003

Changes in the Levels of Chemokines and Cytokines in the Placentas of Women with Plasmodium falciparum Malaria

Amorsolo L. Suguitan; Rose G. F. Leke; Genevieve G. Fouda; Ainong Zhou; Lucy W. Thuita; Simon Metenou; Josephine Fogako; Rosette Megnekou; Diane W. Taylor

Plasmodium falciparum-infected erythrocytes often are sequestered in the placenta and stimulate the accumulation of maternal mononuclear cells. In this study, the role that chemokines and cytokines play in mediating the inflammatory response was investigated. Placental parasites elicited a statistically significant increase in the levels of interferon (IFN)-gamma, tumor necrosis factor (TNF)-alpha, and interleukin (IL)-10, in plasma collected from the intervillous space. Explants of fetal tissue from malaria-positive placentas also secreted significantly enhanced amounts of IFN-gamma. Culture supernatant of maternal intervillous leukocytes obtained from infected placentas contained significantly higher levels of TNF-alpha, IL-10, monocyte chemotactic protein-1, macrophage inflammatory protein (MIP)-1alpha, MIP-1beta, and IFN-gamma inducible protein-10 than did cultures of white blood cells obtained from uninfected placentas. Taken together, these results show that both fetal and maternal cells secrete inflammatory and immunoregulatory cytokines in response to P. falciparum and suggest that beta-chemokines produced by maternal cells contribute to the accumulation of macrophages in the intervillous space.


Journal of Virology | 2011

Cross-Reactive HIV-1-Neutralizing Human Monoclonal Antibodies Identified from a Patient with 2F5-Like Antibodies

Zhongyu Zhu; Haiyan Rebekah Qin; Weizao Chen; Qi Zhao; Xiaoying Shen; Robert J. Schutte; Yanping Wang; Gilad Ofek; Emily Streaker; Ponraj Prabakaran; Genevieve G. Fouda; Hua-Xin Liao; John Owens; Mark K. Louder; Yongping Yang; Kristina-Ana Klaric; M. Anthony Moody; John R. Mascola; Jamie K. Scott; Peter D. Kwong; David C. Montefiori; Barton F. Haynes; Georgia D. Tomaras; Dimiter S. Dimitrov

ABSTRACT The genes encoding broadly HIV-1-neutralizing human monoclonal antibodies (MAbs) are highly divergent from their germ line counterparts. We have hypothesized that such high levels of somatic hypermutation could pose a challenge for elicitation of the broadly neutralizing (bn) Abs and that identification of less somatically mutated bn Abs may help in the design of effective vaccine immunogens. In a quest for such bn Abs, phage- and yeast-displayed antibody libraries, constructed using peripheral blood mononuclear cells (PBMCs) from a patient with bn serum containing Abs targeting the epitope of the bn MAb 2F5, were panned against peptides containing the 2F5 epitope and against the HIV-1 gp140JR-FL. Two MAbs (m66 and m66.6) were identified; the more mutated variant (m66.6) exhibited higher HIV-1-neutralizing activity than m66, although it was weaker than 2F5 in a TZM-bl cell assay. Binding of both MAbs to gp41 alanine substitution mutant peptides required the DKW664–666 core of the 2F5 epitope and two additional upstream residues (L660,663). The MAbs have long (21-residue) heavy-chain third complementarity-determining regions (CDR-H3s), and m66.6 (but not m66) exhibited polyspecific reactivity to self- and non-self-antigens. Both m66 and m66.6 are significantly less divergent from their germ line Ab counterparts than 2F5—they have a total of 11 and 18 amino acid changes, respectively, from the closest VH and Vκ germ line gene products compared to 25 for 2F5. These new MAbs could help explore the complex maturation pathways involved in broad neutralization and its relationship with auto- and polyreactivity and may aid design of vaccine immunogens and development of therapeutics against HIV-1 infection.


American Journal of Pathology | 2004

Plasmodium falciparum-Infected Erythrocytes Adhere Both in the Intervillous Space and on the Villous Surface of Human Placenta by Binding to the Low-Sulfated Chondroitin Sulfate Proteoglycan Receptor

Arivalagan Muthusamy; Rajeshwara N. Achur; Veer P. Bhavanandan; Genevieve G. Fouda; Diane W. Taylor; D. Channe Gowda

In pregnant women infected with Plasmodium falciparum, the parasite-infected red blood cells (IRBCs) sequester in the placenta through chondroitin 4-sulfate (C4S)-mediated adherence. The pattern of IRBC adherence in P. falciparum-infected placenta has been controversial. Moreover, the identity of the chondroitin sulfate proteoglycan (CSPG) receptor, that mediates IRBC adherence, and its location in the placenta have not been established. This study, using immunohistochemical techniques, clearly shows, for the first time, that the low-sulfated CSPGs of the placenta are localized predominantly in the intervillous space. Ex vivo IRBC adherence analyses demonstrate that the IRBCs are adhered to the CSPG receptors in the placenta in a C4S-dependent manner. This IRBC binding pattern was similar to that observed in P. falciparum-infected placentas. These data and the results of dual-fluorescence staining of the endogenous RBCs and syncytiotrophoblasts, and co-localization of CSPG and IRBC adherence unequivocally establish that the low-sulfated CSPGs are the major natural receptors for IRBC adherence in the placenta. Further, it was found that IRBCs adhere mainly in the intervillous space and also at significant levels to the syncytiotrophoblasts. Finally, the ex vivo IRBC adherence method described herein provides a reliable procedure for future studies for the assessment of the efficacy of C4S inhibitors and adhesion inhibitory antibodies.


Clinical and Vaccine Immunology | 2006

Multiplex Assay for Simultaneous Measurement of Antibodies to Multiple Plasmodium falciparum Antigens

Genevieve G. Fouda; Rose Leke; Carole A. Long; Pierre Druilhe; Ainong Zhou; Diane W. Taylor; Armead H. Johnson

ABSTRACT Antibodies to Plasmodium falciparum are classically measured using the enzyme-linked immunosorbent assay (ELISA). Although highly sensitive, this technique is labor-intensive when large numbers of samples must be screened against multiple antigens. The suspension array technology (SAT) might be an alterative to ELISA, as it allows measurement of antibodies against multiple antigens simultaneously with a small volume of sample. This study sought to adapt the new SAT multiplex system for measuring antibodies against nine malarial vaccine candidate antigens, including recombinant proteins from two variants of merozoite surface protein 1, two variants of apical merozoite antigen 1, erythrocyte binding antigen 175, merozoite surface protein 3, and peptides from the circumsporozoite protein, ring erythrocyte surface antigen, and liver-stage antigen 1. Various concentrations of the antigens were coupled to microspheres with different spectral addresses, and plasma samples from Cameroonian adults were screened by SAT in mono- and multiplex formats and by ELISA. Optimal amounts of protein required to perform the SAT assay were 10- to 100-fold less than that needed for ELISA. Excellent agreement was found between the single and multiplex formats (R ≥ 0.96), even when two variants of the same antigen were used. The multiplex assay was rapid, reproducible, required less than 1 μl of plasma, and had a good correlation with ELISA. Thus, SAT provides an important new tool for studying the immune response to malaria rapidly and efficiently in large populations, even when the amount of plasma available is limited, e.g., in studies of neonates or finger-prick blood.


Journal of Virology | 2011

Origin and Evolution of HIV-1 in Breast Milk Determined by Single-Genome Amplification and Sequencing

Jesus F. Salazar-Gonzalez; Maria G. Salazar; Gerald H. Learn; Genevieve G. Fouda; Helen H. Kang; Tatenda Mahlokozera; Andrew B. Wilks; Rachel V. Lovingood; Andrea R. Stacey; Linda Kalilani; Steve Meshnick; Persephone Borrow; David C. Montefiori; Thomas N. Denny; Norman L. Letvin; George M. Shaw; Beatrice H. Hahn; Sallie R. Permar; Aids Vaccine Immunology A

ABSTRACT HIV transmission via breastfeeding accounts for a considerable proportion of infant HIV acquisition. However, the origin and evolution of the virus population in breast milk, the likely reservoir of transmitted virus variants, are not well characterized. In this study, HIV envelope (env) genes were sequenced from virus variants amplified by single-genome amplification from plasmas and milk of 12 chronically HIV-infected, lactating Malawian women. Maximum likelihood trees and statistical tests of compartmentalization revealed interspersion of plasma and milk HIV env sequences in the majority of subjects, indicating limited or no compartmentalization of milk virus variants. However, phylogenetic tree analysis further revealed monotypic virus variants that were significantly more frequent in milk (median proportion of identical viruses, 29.5%; range, 0 to 61%) than in plasma (median proportion of identical viruses, 0%; range, 0 to 26%) (P = 0.002), suggesting local virus replication in the breast milk compartment. Moreover, clonally amplified virus env genes in milk produced functional virus Envs that were all CCR5 tropic. Milk and plasma virus Envs had similar predicted phenotypes and neutralization sensitivities to broadly neutralizing antibodies in both transmitting and nontransmitting mothers. Finally, phylogenetic comparison of longitudinal milk and plasma virus env sequences revealed synchronous virus evolution and new clonal amplification of evolved virus env genes in milk. The limited compartmentalization and the clonal amplification of evolving, functional viruses in milk indicate continual seeding of the mammary gland by blood virus variants, followed by transient local replication of these variants in the breast milk compartment.


Journal of Virology | 2011

HIV-Specific Functional Antibody Responses in Breast Milk Mirror Those in Plasma and Are Primarily Mediated by IgG Antibodies

Genevieve G. Fouda; Nicole L. Yates; Justin Pollara; Xiaoying Shen; Glenn Overman; Tatenda Mahlokozera; Andrew B. Wilks; Helen H. Kang; Jesus F. Salazar-Gonzalez; Maria G. Salazar; Linda Kalilani; Steve Meshnick; Beatrice H. Hahn; George M. Shaw; Rachel V. Lovingood; Thomas N. Denny; Barton F. Haynes; Norman L. Letvin; Guido Ferrari; David C. Montefiori; Georgia D. Tomaras; Sallie R. Permar; Aids Vaccine Immunology

ABSTRACT Despite months of mucosal virus exposure, the majority of breastfed infants born to HIV-infected mothers do not become infected, raising the possibility that immune factors in milk inhibit mucosal transmission of HIV. HIV Envelope (Env)-specific antibodies are present in the milk of HIV-infected mothers, but little is known about their virus-specific functions. In this study, HIV Env-specific antibody binding, autologous and heterologous virus neutralization, and antibody-dependent cell cytotoxicity (ADCC) responses were measured in the milk and plasma of 41 HIV-infected lactating women. Although IgA is the predominant antibody isotype in milk, HIV Env-specific IgG responses were higher in magnitude than HIV Env-specific IgA responses in milk. The concentrations of anti-HIV gp120 IgG in milk and plasma were directly correlated (r = 0.75; P < 0.0001), yet the response in milk was 2 logarithm units lower than in plasma. Similarly, heterologous virus neutralization (r = 0.39; P = 0.010) and ADCC activity (r = 0.64; P < 0.0001) in milk were directly correlated with that in the systemic compartment but were 2 log units lower in magnitude. Autologous neutralization was rarely detected in milk. Milk heterologous virus neutralization titers correlated with HIV gp120 Env-binding IgG responses but not with IgA responses (r = 0.71 and P < 0.0001, and r = 0.17 and P = 0.30). Moreover, IgGs purified from milk and plasma had equal neutralizing potencies against a tier 1 virus (r = 0.65; P < 0.0001), whereas only 1 out of 35 tested non-IgG milk fractions had detectable neutralization. These results suggest that plasma-derived IgG antibodies mediate the majority of the low-level HIV neutralization and ADCC activity in breast milk.


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

Tenascin-C is an innate broad-spectrum, HIV-1–neutralizing protein in breast milk

Genevieve G. Fouda; Frederick H. Jaeger; Joshua D. Amos; Carrie Ho; Erika L. Kunz; Kara Anasti; Lisa Stamper; Brooke E. Liebl; Kimberly H. Barbas; Tomoo Ohashi; M.A. Moseley; Hua-Xin Liao; Harold P. Erickson; S. Munir Alam; Sallie R. Permar

Significance Achieving an AIDS-free generation will require elimination of breast milk transmission of HIV-1, as breastfeeding is a cornerstone of infant survival in developing regions. Antiretroviral prophylaxis considerably reduces postnatal HIV-1 transmission, yet its efficacy is limited by access, adherence, toxicities, and resistance of maternal HIV-1 strains. Alternative, safe strategies of impeding postnatal HIV-1 transmission will be required to eliminate infant HIV-1 infection. In this paper, we identify an innate HIV-neutralizing protein in breast milk, Tenascin-C, which captures and neutralizes HIV-1 virions via binding to the chemokine coreceptor binding site on the HIV-1 Envelope. This protein has the potential to be developed as a prevention strategy for postnatal and other modes of HIV-1 transmission. Achieving an AIDS-free generation will require elimination of postnatal transmission of HIV-1 while maintaining the nutritional and immunologic benefits of breastfeeding for infants in developing regions. Maternal/infant antiretroviral prophylaxis can reduce postnatal HIV-1 transmission, yet toxicities and the development of drug-resistant viral strains may limit the effectiveness of this strategy. Interestingly, in the absence of antiretroviral prophylaxis, greater than 90% of infants exposed to HIV-1 via breastfeeding remain uninfected, despite daily mucosal exposure to the virus for up to 2 y. Moreover, milk of uninfected women inherently neutralizes HIV-1 and prevents virus transmission in animal models, yet the factor(s) responsible for this anti-HIV activity is not well-defined. In this report, we identify a primary HIV-1–neutralizing protein in breast milk, Tenascin-C (TNC). TNC is an extracellular matrix protein important in fetal development and wound healing, yet its antimicrobial properties have not previously been established. Purified TNC captured and neutralized multiclade chronic and transmitted/founder HIV-1 variants, and depletion of TNC abolished the HIV-1–neutralizing activity of milk. TNC bound the HIV-1 Envelope protein at a site that is induced upon engagement of its primary receptor, CD4, and is blocked by V3 loop- (19B and F39F) and chemokine coreceptor binding site-directed (17B) monoclonal antibodies. Our results demonstrate the ability of an innate mucosal host protein found in milk to neutralize HIV-1 via binding to the chemokine coreceptor site, potentially explaining why the majority of HIV-1–exposed breastfed infants are protected against mucosal HIV-1 transmission.


Journal of Virology | 2013

Mucosal Immunization of Lactating Female Rhesus Monkeys with a Transmitted/Founder HIV-1 Envelope Induces Strong Env-Specific IgA Antibody Responses in Breast Milk

Genevieve G. Fouda; Joshua D. Amos; Andrew B. Wilks; Justin Pollara; Caroline A. Ray; Anjali Chand; Erika L. Kunz; Brooke E. Liebl; Kaylan Whitaker; Angela Carville; Shannon D. Smith; Lisa Colvin; David J. Pickup; Herman F. Staats; Glenn Overman; Krissey Eutsey-Lloyd; Robert Parks; Haiyan Chen; Celia C. LaBranche; Susan W. Barnett; Georgia D. Tomaras; Guido Ferrari; David C. Montefiori; Hua-Xin Liao; Norman L. Letvin; Barton F. Haynes; Sallie R. Permar

ABSTRACT We previously demonstrated that vaccination of lactating rhesus monkeys with a DNA prime/vector boost strategy induces strong T-cell responses but limited envelope (Env)-specific humoral responses in breast milk. To improve vaccine-elicited antibody responses in milk, hormone-induced lactating rhesus monkeys were vaccinated with a transmitted/founder (T/F) HIV Env immunogen in a prime-boost strategy modeled after the moderately protective RV144 HIV vaccine. Lactating rhesus monkeys were intramuscularly primed with either recombinant DNA (n = 4) or modified vaccinia virus Ankara (MVA) poxvirus vector (n = 4) expressing the T/F HIV Env C.1086 and then boosted twice intramuscularly with C.1086 gp120 and the adjuvant MF59. The vaccines induced Env-binding IgG and IgA as well as neutralizing and antibody-dependent cellular cytotoxicity (ADCC) responses in plasma and milk of most vaccinated animals. Importantly, plasma neutralization titers against clade C HIV variants MW965 (P = 0.03) and CAP45 (P = 0.04) were significantly higher in MVA-primed than in DNA-primed animals. The superior systemic prime-boost regimen was then compared to a mucosal-boost regimen, in which animals were boosted twice intranasally with C.1086 gp120 and the TLR 7/8 agonist R848 following the same systemic prime. While the systemic and mucosal vaccine regimens elicited comparable levels of Env-binding IgG antibodies, mucosal immunization induced significantly stronger Env-binding IgA responses in milk (P = 0.03). However, the mucosal regimen was not as potent at inducing functional IgG responses. This study shows that systemic MVA prime followed by either intranasal or systemic protein boosts can elicit strong humoral responses in breast milk and may be a useful strategy to interrupt postnatal HIV-1 transmission.


Journal of Clinical Investigation | 2015

Maternal HIV-1 envelope–specific antibody responses and reduced risk of perinatal transmission

Sallie R. Permar; Youyi Fong; Nathan Vandergrift; Genevieve G. Fouda; Peter B. Gilbert; Robert Parks; Frederick H. Jaeger; Justin Pollara; Amanda Martelli; Brooke E. Liebl; Krissey E. Lloyd; Nicole L. Yates; R. Glenn Overman; Xiaoying Shen; Kaylan Whitaker; Haiyan Chen; Jamie Pritchett; Erika Solomon; Emma Friberg; Dawn J. Marshall; John F. Whitesides; Thaddeus C. Gurley; Tarra Von Holle; David Martinez; Fangping Cai; Amit Kumar; Shi Mao Xia; Xiaozhi Lu; Raul Louzao; Samantha Wilkes

Despite the wide availability of antiretroviral drugs, more than 250,000 infants are vertically infected with HIV-1 annually, emphasizing the need for additional interventions to eliminate pediatric HIV-1 infections. Here, we aimed to define humoral immune correlates of risk of mother-to-child transmission (MTCT) of HIV-1, including responses associated with protection in the RV144 vaccine trial. Eighty-three untreated, HIV-1-transmitting mothers and 165 propensity score-matched nontransmitting mothers were selected from the Women and Infants Transmission Study (WITS) of US nonbreastfeeding, HIV-1-infected mothers. In a multivariable logistic regression model, the magnitude of the maternal IgG responses specific for the third variable loop (V3) of the HIV-1 envelope was predictive of a reduced risk of MTCT. Neutralizing Ab responses against easy-to-neutralize (tier 1) HIV-1 strains also predicted a reduced risk of peripartum transmission in secondary analyses. Moreover, recombinant maternal V3-specific IgG mAbs mediated neutralization of autologous HIV-1 isolates. Thus, common V3-specific Ab responses in maternal plasma predicted a reduced risk of MTCT and mediated autologous virus neutralization, suggesting that boosting these maternal Ab responses may further reduce HIV-1 MTCT.

Collaboration


Dive into the Genevieve G. Fouda's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hua-Xin Liao

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge