Network


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

Hotspot


Dive into the research topics where Lauren A. Hirao is active.

Publication


Featured researches published by Lauren A. Hirao.


Vaccine | 2008

Combined effects of IL-12 and electroporation enhances the potency of DNA vaccination in macaques

Lauren A. Hirao; Ling Wu; Amir S. Khan; David A. Hokey; Jian Yan; Anlan Dai; Michael R. Betts; Ruxandra Draghia-Akli; David B. Weiner

DNA vaccines are a promising technology. Historically, however, the ability of DNA vaccines to induce high response rates and strong immune responses, especially antibody responses, in non-human primates and human clinical trials has proven suboptimal. Here, we performed a pilot study in rhesus macaques to evaluate whether we could improve the immunogenicity of DNA vaccines through the use of adjuvant technology and improved delivery systems. The study consisted of four groups of animals that received: DNA by intramuscular (IM) injection, DNA with plasmid-encoded IL-12 by IM injection, DNA by IM injection with in vivo electroporation (EP), and DNA with IL-12 by IM EP. Each group was immunized three times with optimized HIV gag and env constructs. Vaccine immunogenicity was assessed by IFNgamma ELISpot, CFSE proliferation, polyfunctional flow cytometry, and antibody ELISA. Similar to previous studies, use of IL-12 as an adjuvant increased the gag and env-specific cellular responses. The use of EP to enhance plasmid delivery resulted in dramatically higher cellular as well as humoral responses. Interestingly, the use of EP to administer the DNA and IL-12 adjuvant combination resulted in the induction of higher, more efficient responses such that a 10-fold increase in antigen-specific IFNgamma(+) cells compared to IM DNA immunization was observed after a single immunization. In addition to increases in the magnitude of IFNgamma production in the initial and memory responses, the combined approach resulted in enhancements in the proliferative capacity of antigen-specific CD8(+) T cells and the amount of polyfunctional cells capable of producing IL-2 and TNFalpha in addition to IFNgamma. These data suggest that adjuvant and improved delivery methods may be able to overcome previous immunogenicity limitations in DNA vaccine technology.


Journal of Virology | 2005

Epitope-Mapping Studies Define Two Major Neutralization Sites on the Vaccinia Virus Extracellular Enveloped Virus Glycoprotein B5R

Lydia Aldaz-Carroll; J. Charles Whitbeck; Manuel Ponce de Leon; Huan Lou; Lauren A. Hirao; Stuart N. Isaacs; Bernard Moss; Roselyn J. Eisenberg; Gary H. Cohen

ABSTRACT Vaccinia extracellular enveloped virus (EEV) is critical for cell-to-cell and long-range virus spread both in vitro and in vivo. The B5R gene encodes an EEV-specific type I membrane protein that is essential for efficient EEV formation. The majority of the B5R ectodomain consists of four domains with homology to short consensus repeat domains followed by a stalk. Previous studies have shown that polyclonal antibodies raised against the B5R ectodomain inhibit EEV infection. In this study, our goal was to elucidate the antigenic structure of B5R and relate this to its function. To do this, we produced multimilligram quantities of vaccinia virus B5R as a soluble protein [B5R(275t)] using a baculovirus expression system. We then selected and characterized a panel of 26 monoclonal antibodies (MAbs) that recognize B5R(275t). Five of these MAbs neutralized EEV and inhibited comet formation. Two other MAbs were able only to neutralize EEV, while five others were able only to inhibit comet formation. This suggests that the EEV neutralization and comet inhibition assays measure different viral functions and that at least two different antigenic sites on B5R are important for these activities. We further characterized the MAbs and the antigenic structure of B5R(275t) by peptide mapping and by reciprocal MAb blocking studies using biosensor analysis. The epitopes recognized by neutralizing MAbs were localized to SCR1-SCR2 and/or the stalk of B5R(275t). Furthermore, the peptide and blocking data support the concept that SCR1 and the stalk may be in juxtaposition and may be part of the same functional domain.


Journal of Virology | 2008

Human Immunodeficiency Virus Type 1 Nef Induces Programmed Death 1 Expression through a p38 Mitogen-Activated Protein Kinase-Dependent Mechanism

Karuppiah Muthumani; Andrew Y. Choo; Devon J. Shedlock; Dominick J. Laddy; Senthil G. Sundaram; Lauren A. Hirao; Ling Wu; Khanh P. Thieu; Christopher W. Chung; Karthikbabu Mallil Lankaraman; Pablo Tebas; Guido Silvestri; David B. Weiner

ABSTRACT Chronic viral infection is characterized by the functional impairment of virus-specific T-cell responses. Recent evidence has suggested that the inhibitory receptor programmed death 1 (PD-1) is specifically upregulated on antigen-specific T cells during various chronic viral infections. Indeed, it has been reported that human immunodeficiency virus (HIV)-specific T cells express elevated levels of PD-1 and that this expression correlates with the viral load and inversely with CD4+ T-cell counts. More importantly, antibody blockade of the PD-1/PD-L1 pathway was sufficient to both increase and stimulate virus-specific T-cell proliferation and cytokine production. However, the mechanisms that mediate HIV-induced PD-1 upregulation are not known. Here, we provide evidence that the HIV type 1 (HIV-1) accessory protein Nef can transcriptionally induce the expression of PD-1 during infection in vitro. Nef-induced PD-1 upregulation requires its proline-rich motif and the activation of the downstream kinase p38. Further, inhibition of Nef activity by p38 MAPK inhibitor effectively blocked PD-1 upregulation, suggesting that p38 MAPK activation is an important initiating event in Nef-mediated PD-1 expression in HIV-1-infected cells. These data demonstrate an important signaling event of Nef in HIV-1 pathogenesis.


The Journal of Infectious Diseases | 2011

Multivalent Smallpox DNA Vaccine Delivered by Intradermal Electroporation Drives Protective Immunity in Nonhuman Primates Against Lethal Monkeypox Challenge

Lauren A. Hirao; Ruxandra Draghia-Akli; Jonathan T. Prigge; Maria Yang; Abhishek Satishchandran; Ling Wu; Erika Hammarlund; Amir S. Khan; Tahar Babas; Lowrey Rhodes; Peter Silvera; Mark K. Slifka; Niranjan Y. Sardesai; David B. Weiner

The threat of a smallpox-based bioterrorist event or a human monkeypox outbreak has heightened the importance of new, safe vaccine approaches for these pathogens to complement older poxviral vaccine platforms. As poxviruses are large, complex viruses, they present technological challenges for simple recombinant vaccine development where a multicomponent mixtures of vaccine antigens are likely important in protection. We report that a synthetic, multivalent, highly concentrated, DNA vaccine delivered by a minimally invasive, novel skin electroporation microarray can drive polyvalent immunity in macaques, and offers protection from a highly pathogenic monkeypox challenge. Such a diverse, high-titer antibody response produced against 8 different DNA-encoded antigens delivered simultaneously in microvolumes has not been previously described. These studies represent a significant improvement in the efficiency of the DNA vaccine platform, resulting in immune responses that mimic live viral infections, and would likely have relevance for vaccine design against complex human and animal pathogens.


Current Opinion in Hiv and Aids | 2011

Th17 cells and regulatory T cells in elite control over HIV and SIV.

Dennis Hartigan-O'Connor; Lauren A. Hirao; Joseph M. McCune; Satya Dandekar

Purpose of reviewWe present current findings about two subsets of CD4+ T cells that play an important part in the initial host response to infection with the HIV type 1: those producing IL-17 (Th17 cells) and those with immunosuppressive function (CD25+FoxP3+ regulatory T cells or T-reg). The role of these cells in the control of viral infection and immune activation as well as in the prevention of immune deficiency in HIV-infected elite controllers will be examined. We will also discuss the use of the simian immunodeficiency virus (SIV)-infected macaque model of AIDS to study the interplay between these cells and lentiviral infection in vivo. Recent findingsStudy of Th17 cells in humans and nonhuman primates (NHPs) has shown that depletion of these cells is associated with the dissemination of microbial products from the infected gut, increased systemic immune activation, and disease progression. Most impressively, having a smaller Th17-cell compartment has been found to predict these outcomes. T-reg have been associated with the reduced antiviral T-cell responses but not with the suppression of generalized T cell activation. Both cell subsets influence innate immune responses and, in doing so, may shape the inflammatory milieu of the host at infection. SummaryInteractions between Th17 cells, T-reg, and cells of the innate immune system influence the course of HIV and SIV infection from its earliest stages, even before the appearance of adaptive immunity. Such interactions may be pivotal for elite control over disease progression.


Molecular Therapy | 2010

Comparative Analysis of Immune Responses Induced by Vaccination With SIV Antigens by Recombinant Ad5 Vector or Plasmid DNA in Rhesus Macaques

Lauren A. Hirao; Ling Wu; Abhishek Satishchandran; Amir S. Khan; Ruxandra Draghia-Akli; Adam C. Finnefrock; Andrew J. Bett; Michael R. Betts; Danilo R. Casimiro; Niranjan Y. Sardesai; J. Joseph Kim; John W. Shiver; David B. Weiner

DNA vaccines have undergone important enhancements in their design, formulation, and delivery process. Past literature supports that DNA vaccines are not as immunogenic in nonhuman primates as live vector systems. The most potent recombinant vector system for induction of cellular immune responses in macaques and humans is adenovirus serotype 5 (Ad5), an important benchmark for new vaccine development. Here, we performed a head-to-head evaluation of the Merck Ad5 SIV vaccine and an optimized electroporation (EP) delivered SIV DNA vaccine in macaques. Animals receiving the Ad5 vaccine were immunized three times, whereas the DNA-vaccinated animals were immunized up to four times based on optimized protocols. We observed significant differences in the quantity of IFNgamma responses by enzyme-linked immunosorbent spot (ELISpot), greater proliferative capacity of CD8(+) T cells, and increased polyfunctionality of both CD4(+) and CD8(+) T cells in the DNA-vaccinated group. Importantly, Ad5 immunizations failed to boost following the first immunization, whereas DNA responses were continually boosted with all four immunizations demonstrating a major advantage of these improved DNA vaccines. These optimized DNA vaccines induce very different immune phenotypes than traditional Ad5 vaccines, suggesting that they could play an important role in vaccine research and development.


European Journal of Immunology | 2008

Activation drives PD-1 expression during vaccine-specific proliferation and following lentiviral infection in macaques

David A. Hokey; F. Brad Johnson; Jasmine S. Smith; Joshua L. Weber; Jian Yan; Lauren A. Hirao; Jean D. Boyer; Mark G. Lewis; George Makedonas; Michael R. Betts; David B. Weiner

Recent data supports that increased expression of PD‐1, a negative regulator of immune function, is associated with T cell exhaustion during chronic viral infection. However, PD‐1 expression during acute infection and vaccination has not been studied in great detail in primates. Here, we examine PD‐1 expression on CD3+ T cells following DNA vaccination or lentiviral infection of macaques. Ex vivo peptide stimulation of PBMC from DNA‐vaccinated uninfected macaques revealed a temporal increase in PD‐1 expression in proliferating antigen‐specific CD8+ T cells. Following the initial increase, PD‐1 expression steadily declined as proliferation continued, with a concomitant increase in IFN‐γ secretion. Subsequent examination of PD‐1 expression on T cells from uninfected and lentivirus‐infected non‐vaccinated macaques revealed a significant increase in PD‐1 expression with lentiviral infection, consistent with previous reports. PD‐1 expression was highest on cells with activated memory and effector phenotypes. Despite their decreased telomere length, PD‐1hi T cell populations do not appear to have statistically significant uncapped telomeres, typically indicative of proliferative exhaustion, suggesting a different mechanistic regulation of proliferation by PD‐1. Our data indicate that PD‐1 expression is increased as a result of T cell activation during a primary immune response as well as during persistent immune activation in macaques.


PLOS Pathogens | 2014

Early mucosal sensing of SIV infection by paneth cells induces IL-1β production and initiates gut epithelial disruption.

Lauren A. Hirao; Irina Grishina; Olivier Bourry; William K. Hu; Monsicha Somrit; Sumathi Sankaran-Walters; Chris A. Gaulke; Anne Fenton; Jay A. Li; Robert W. Crawford; Frank Chuang; Ross P. Tarara; Maria L. Marco; Andreas J. Bäumler; Holland Cheng; Satya Dandekar

HIV causes rapid CD4+ T cell depletion in the gut mucosa, resulting in immune deficiency and defects in the intestinal epithelial barrier. Breakdown in gut barrier integrity is linked to chronic inflammation and disease progression. However, the early effects of HIV on the gut epithelium, prior to the CD4+ T cell depletion, are not known. Further, the impact of early viral infection on mucosal responses to pathogenic and commensal microbes has not been investigated. We utilized the SIV model of AIDS to assess the earliest host-virus interactions and mechanisms of inflammation and dysfunction in the gut, prior to CD4+ T cell depletion. An intestinal loop model was used to interrogate the effects of SIV infection on gut mucosal immune sensing and response to pathogens and commensal bacteria in vivo. At 2.5 days post-SIV infection, low viral loads were detected in peripheral blood and gut mucosa without CD4+ T cell loss. However, immunohistological analysis revealed the disruption of the gut epithelium manifested by decreased expression and mislocalization of tight junction proteins. Correlating with epithelial disruption was a significant induction of IL-1β expression by Paneth cells, which were in close proximity to SIV-infected cells in the intestinal crypts. The IL-1β response preceded the induction of the antiviral interferon response. Despite the disruption of the gut epithelium, no aberrant responses to pathogenic or commensal bacteria were observed. In fact, inoculation of commensal Lactobacillus plantarum in intestinal loops led to rapid anti-inflammatory response and epithelial tight junction repair in SIV infected macaques. Thus, intestinal Paneth cells are the earliest responders to viral infection and induce gut inflammation through IL-1β signaling. Reversal of the IL-1β induced gut epithelial damage by Lactobacillus plantarum suggests synergistic host-commensal interactions during early viral infection and identify these mechanisms as potential targets for therapeutic intervention.


Vaccine | 2008

Parameters for DNA vaccination using adaptive constant-current electroporation in mouse and pig models

Ruxandra Draghia-Akli; Amir S. Khan; Patricia A. Brown; Melissa Pope; Ling Wu; Lauren A. Hirao; David B. Weiner

Enhancing the expression of DNA vaccines requires that specific conditions of delivery are optimized. We describe experiments using adaptive constant-current electroporation (EP) in mice and pigs examining parameters such as target muscle, delay between plasmid delivery and onset of EP pulses and DNA vaccine formulation; our studies show that concentrated formulations result in better expression and immunogenicity. Furthermore, various conditions of EP that limit the amount of muscle damage were measured. The results of these studies will help to advance the success of DNA vaccines in animals into success in human clinical trials.


PLOS ONE | 2011

Long-Term Programming of Antigen-Specific Immunity from Gene Expression Signatures in the PBMC of Rhesus Macaques Immunized with an SIV DNA Vaccine

Sarah E. Belisle; Jiangmei Yin; Devon J. Shedlock; Anlan Dai; Jian Yan; Lauren A. Hirao; Michele Kutzler; Mark G. Lewis; Hanne Andersen; Simon M. Lank; Julie A. Karl; David H. O'Connor; Amir R. Khan; Niranjan Y. Sardesai; Jean Chang; Lauri D. Aicher; Robert E. Palermo; David B. Weiner; Michael G. Katze; Jean D. Boyer

While HIV-1-specific cellular immunity is thought to be critical for the suppression of viral replication, the correlates of protection have not yet been determined. Rhesus macaques (RM) are an important animal model for the study and development of vaccines against HIV/AIDS. Our laboratory has helped to develop and study DNA-based vaccines in which recent technological advances, including genetic optimization and in vivo electroporation (EP), have helped to dramatically boost their immunogenicity. In this study, RMs were immunized with a DNA vaccine including individual plasmids encoding SIV gag, env, and pol alone, or in combination with a molecular adjuvant, plasmid DNA expressing the chemokine ligand 5 (RANTES), followed by EP. Along with standard immunological assays, flow-based activation analysis without ex vivo restimulation and high-throughput gene expression analysis was performed. Strong cellular immunity was induced by vaccination which was supported by all assays including PBMC microarray analysis that identified the up-regulation of 563 gene sequences including those involved in interferon signaling. Furthermore, 699 gene sequences were differentially regulated in these groups at peak viremia following SIVmac251 challenge. We observed that the RANTES-adjuvanted animals were significantly better at suppressing viral replication during chronic infection and exhibited a distinct pattern of gene expression which included immune cell-trafficking and cell cycle genes. Furthermore, a greater percentage of vaccine-induced central memory CD8+ T-cells capable of an activated phenotype were detected in these animals as measured by activation analysis. Thus, co-immunization with the RANTES molecular adjuvant followed by EP led to the generation of cellular immunity that was transcriptionally distinct and had a greater protective efficacy than its DNA alone counterpart. Furthermore, activation analysis and high-throughput gene expression data may provide better insight into mechanisms of viral control than may be observed using standard immunological assays.

Collaboration


Dive into the Lauren A. Hirao's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Satya Dandekar

University of California

View shared research outputs
Top Co-Authors

Avatar

Ling Wu

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Amir S. Khan

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar

David A. Hokey

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Anlan Dai

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jean D. Boyer

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Jian Yan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge