Network


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

Hotspot


Dive into the research topics where Michael Citron is active.

Publication


Featured researches published by Michael Citron.


Journal of Virology | 2006

Vaccine-Induced Cellular Immune Responses Reduce Plasma Viral Concentrations after Repeated Low-Dose Challenge with Pathogenic Simian Immunodeficiency Virus SIVmac239

Nancy A. Wilson; Jason S. Reed; Gnankang Napoé; Shari M. Piaskowski; Andy Szymanski; Jessica Furlott; Edna J. Gonzalez; Levi Yant; Nicholas J. Maness; Gemma E. May; Taeko Soma; Matthew R. Reynolds; Eva G. Rakasz; Richard Rudersdorf; Adrian B. McDermott; David H. O'Connor; Thomas C. Friedrich; David B. Allison; Amit Patki; Louis J. Picker; Dennis R. Burton; Jing Lin; Lingyi Huang; Deepa Patel; Gwendolyn Heindecker; Jiang Fan; Michael Citron; Melanie Horton; Fubao Wang; Xiaoping Liang

ABSTRACT The goal of an AIDS vaccine regimen designed to induce cellular immune responses should be to reduce the viral set point and preserve memory CD4 lymphocytes. Here we investigated whether vaccine-induced cellular immunity in the absence of any Env-specific antibodies can control viral replication following multiple low-dose challenges with the highly pathogenic SIVmac239 isolate. Eight Mamu-A*01-positive Indian rhesus macaques were vaccinated with simian immunodeficiency virus (SIV) gag, tat, rev, and nef using a DNA prime-adenovirus boost strategy. Peak viremia (P = 0.007) and the chronic phase set point (P = 0.0192) were significantly decreased in the vaccinated cohort, out to 1 year postinfection. Loss of CD4+ memory populations was also ameliorated in vaccinated animals. Interestingly, only one of the eight vaccinees developed Env-specific neutralizing antibodies after infection. The control observed was significantly improved over that observed in animals vaccinated with SIV gag only. Vaccine-induced cellular immune responses can, therefore, exert a measure of control over replication of the AIDS virus in the complete absence of neutralizing antibody and give us hope that a vaccine designed to induce cellular immune responses might control viral replication.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Design of an HA2-based Escherichia coli expressed influenza immunogen that protects mice from pathogenic challenge

Gayathri Bommakanti; Michael Citron; Robert W. Hepler; Cheryl Callahan; Gwendolyn J. Heidecker; Tariq Ahmad Najar; Xianghan Lu; Joseph G. Joyce; John W. Shiver; Danilo R. Casimiro; Jan ter Meulen; Xiaoping Liang; Raghavan Varadarajan

Influenza HA is the primary target of neutralizing antibodies during infection, and its sequence undergoes genetic drift and shift in response to immune pressure. The receptor binding HA1 subunit of HA shows much higher sequence variability relative to the metastable, fusion-active HA2 subunit, presumably because neutralizing antibodies are primarily targeted against the former in natural infection. We have designed an HA2-based immunogen using a protein minimization approach that incorporates designed mutations to destabilize the low pH conformation of HA2. The resulting construct (HA6) was expressed in Escherichia coli and refolded from inclusion bodies. Biophysical studies and mutational analysis of the protein indicate that it is folded into the desired neutral pH conformation competent to bind the broadly neutralizing HA2 directed monoclonal 12D1, not the low pH conformation observed in previous studies. HA6 was highly immunogenic in mice and the mice were protected against lethal challenge by the homologous A/HK/68 mouse-adapted virus. An HA6-like construct from another H3 strain (A/Phil/2/82) also protected mice against A/HK/68 challenge. Regions included in HA6 are highly conserved within a subtype and are fairly well conserved within a clade. Targeting the highly conserved HA2 subunit with a bacterially produced immunogen is a vaccine strategy that may aid in pandemic preparedness.


Journal of Virology | 2009

Vaccine-Induced Cellular Responses Control Simian Immunodeficiency Virus Replication after Heterologous Challenge

Nancy A. Wilson; Brandon F. Keele; Jason S. Reed; Shari M. Piaskowski; Caitlin E. MacNair; Andrew J. Bett; Xiaoping Liang; Fubao Wang; Elizabeth Thoryk; Gwendolyn J. Heidecker; Michael Citron; Lingyi Huang; Jing Lin; Salvatore Vitelli; Chanook D. Ahn; Masahiko Kaizu; Nicholas J. Maness; Matthew R. Reynolds; Thomas C. Friedrich; John T. Loffredo; Eva G. Rakasz; Stephen Erickson; David B. Allison; Michael Piatak; Jeffrey D. Lifson; John W. Shiver; Danilo R. Casimiro; George M. Shaw; Beatrice H. Hahn; David I. Watkins

ABSTRACT All human immunodeficiency virus (HIV) vaccine efficacy trials to date have ended in failure. Structural features of the Env glycoprotein and its enormous variability have frustrated efforts to induce broadly reactive neutralizing antibodies. To explore the extent to which vaccine-induced cellular immune responses, in the absence of neutralizing antibodies, can control replication of a heterologous, mucosal viral challenge, we vaccinated eight macaques with a DNA/Ad5 regimen expressing all of the proteins of SIVmac239 except Env. Vaccinees mounted high-frequency T-cell responses against 11 to 34 epitopes. We challenged the vaccinees and eight naïve animals with the heterologous biological isolate SIVsmE660, using a regimen intended to mimic typical HIV exposures resulting in infection. Viral loads in the vaccinees were significantly less at both the peak (1.9-log reduction; P < 0.03) and at the set point (2.6-log reduction; P < 0.006) than those in control naïve animals. Five of eight vaccinated macaques controlled acute peak viral replication to less than 80,000 viral RNA (vRNA) copy eq/ml and to less than 100 vRNA copy eq/ml in the chronic phase. Our results demonstrate that broad vaccine-induced cellular immune responses can effectively control replication of a pathogenic, heterologous AIDS virus, suggesting that T-cell-based vaccines may have greater potential than previously appreciated.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Influenza hemagglutinin stem-fragment immunogen elicits broadly neutralizing antibodies and confers heterologous protection.

V. Vamsee Aditya Mallajosyula; Michael Citron; Francesca Ferrara; Xianghan Lu; Cheryl Callahan; Gwendolyn J. Heidecker; Siddhartha P. Sarma; Jessica A. Flynn; Nigel J. Temperton; Xiaoping Liang; Raghavan Varadarajan

Significance Hemagglutinin (HA), the major influenza virus envelope glycoprotein, is the principal target of neutralizing antibodies. Wide diversity and variation of HA entails annual vaccination, as current vaccines typically fail to elicit/boost cross-reactive, broadly neutralizing antibodies (bnAbs). Although several bnAbs bind at the conserved stem of HA making it an attractive universal vaccine candidate, the metastable conformation of this domain imposes challenges in designing a stable, independently folding HA stem immunogen. We rationally designed a stem-fragment immunogen, mimicking the native HA stem that binds conformation-specific bnAbs with high affinity. The immunogen elicited bnAbs and conferred robust protection against lethal, heterologous virus challenge in vivo. Additionally, soluble bacterial expression of such a thermotolerant, disulfide-free immunogen allows for rapid scale-up during pandemic outbreak. Influenza hemagglutinin (HA) is the primary target of the humoral response during infection/vaccination. Current influenza vaccines typically fail to elicit/boost broadly neutralizing antibodies (bnAbs), thereby limiting their efficacy. Although several bnAbs bind to the conserved stem domain of HA, focusing the immune response to this conserved stem in the presence of the immunodominant, variable head domain of HA is challenging. We report the design of a thermotolerant, disulfide-free, and trimeric HA stem-fragment immunogen which mimics the native, prefusion conformation of HA and binds conformation specific bnAbs with high affinity. The immunogen elicited bnAbs that neutralized highly divergent group 1 (H1 and H5 subtypes) and 2 (H3 subtype) influenza virus strains in vitro. Stem immunogens designed from unmatched, highly drifted influenza strains conferred robust protection against a lethal heterologous A/Puerto Rico/8/34 virus challenge in vivo. Soluble, bacterial expression of such designed immunogens allows for rapid scale-up during pandemic outbreaks.


Vaccine | 2009

Comparative immunogenicity evaluations of influenza A virus M2 peptide as recombinant virus like particle or conjugate vaccines in mice and monkeys

Tong-Ming Fu; Karen M. Grimm; Michael Citron; Daniel C. Freed; Jiang Fan; Paul M. Keller; John W. Shiver; Xiaoping Liang; Joseph G. Joyce

Immunization against M2 peptide, also called M2e, from influenza A virus is an innovative vaccine approach for induction of cross-strain protective immunity. Two promising M2 vaccine compositions reported to date are M2 peptide chemically conjugated to carrier proteins or M2 peptide recombinantly expressed on the surface of virus like particles (VLPs) of hepatitis B virus core antigen (HBVc). To conduct a head-to-head comparison of these approaches, we constructed two recombinant HBVc VLPs expressing M2 peptide and prepared two conjugate vaccines with M2 peptide chemically coupled to Neisseria meningitidis outer membrane complex (OMPC) or HBVc VLP, respectively. Here, we showed superior immunogenicity of M2 peptide conjugated to OMPC and M2 peptide expressed on the surface of HBVc antigen based on dose-titration responses in mice. Surprisingly, HBVc expressing M2 peptide was an inferior vaccine in rhesus monkeys, whether as a primary vaccine or as a booster vaccine, when compared with M2-OMPC conjugate vaccine.


Journal of Virology | 2005

Vectored Gag and Env but Not Tat Show Efficacy against Simian-Human Immunodeficiency Virus 89.6P Challenge in Mamu-A*01-Negative Rhesus Monkeys

Xiaoping Liang; Danilo R. Casimiro; William A. Schleif; Fubao Wang; Mary-Ellen Davies; Zhi Qiang Zhang; Tong-Ming Fu; Adam C. Finnefrock; Larry Handt; Michael Citron; Gwendolyn J. Heidecker; Aimin Tang; Minchun Chen; Keith A. Wilson; Lori Gabryelski; Michael McElhaugh; Anthony Carella; Cheryl Moyer; Lingyi Huang; Salvatore Vitelli; Deepa Patel; Jing Lin; Emilio A. Emini; John W. Shiver

ABSTRACT Simian-human immunodeficiency virus (SHIV) challenge studies in rhesus macaques were conducted to evaluate the efficacy of adenovirus-based vaccines in the context of different major histocompatibility complex class I genetic backgrounds and different vaccine compositions. Mamu-A*01 allele-negative rhesus monkeys were immunized with one of the following vaccine constructs: (i) replication-defective recombinant adenovirus type 5 (Ad5) expressing human immunodeficiency virus type 1 (HIV-1) Tat (Ad5/HIVTat); (ii) Ad5 vector expressing simian immunodeficiency virus (SIV) Gag (Ad5/SIVGag); (iii) Ad5 vector expressing the truncated HIV-1jrfl Env, gp140 (Ad5/gp140_jrfl); (iv) Ad5 vector expressing the SHIV-89.6P gp140 (Ad5/gp140_89.6P); or (v) the combination of Ad5/SIVGag and Ad5/gp140_jrfl. Following intravenous challenge with SHIV-89.6P, only those cohorts that received vaccines expressing Gag or Env exhibited an attenuation of the acute viremia and associated CD4-cell lymphopenia. While no prechallenge neutralizing antibody titers were detectable in either Ad5/gp140-vaccinated group, an accelerated neutralizing antibody response was observed in the Ad5/gp140_89.6P-vaccinated group upon viral challenge. The set-point viral loads in the Ad5/SIVGag- and Ad5/gp140_jrfl-vaccinated groups were associated with the overall strength of the induced cellular immune responses. To examine the contribution of Mamu-A*01 allele in vaccine efficacy against SHIV-89.6P challenge, Mamu-A*01-positive monkeys were immunized with Ad5/SIVGag. Vaccine-mediated protection was significantly more pronounced in the Mamu-A*01-positive monkeys than in Mamu-A*01-negative monkeys, suggesting the strong contributions of T-cell epitopes restricted by the Mamu-A*01 molecule. The implications of these results in the development of an HIV-1 vaccine will be discussed.


Journal of Virology | 2012

Design of Escherichia coli-Expressed Stalk Domain Immunogens of H1N1 Hemagglutinin That Protect Mice from Lethal Challenge

Gayathri Bommakanti; Xianghan Lu; Michael Citron; Tariq Ahmad Najar; Gwendolyn J. Heidecker; Jan ter Meulen; Raghavan Varadarajan; Xiaoping Liang

ABSTRACT The hemagglutinin protein (HA) on the surface of influenza virus is essential for viral entry into the host cells. The HA1 subunit of HA is also the primary target for neutralizing antibodies. The HA2 subunit is less exposed on the virion surface and more conserved than HA1. We have previously designed an HA2-based immunogen derived from the sequence of the H3N2 A/HK/68 virus. In the present study, we report the design of an HA2-based immunogen from the H1N1 subtype (PR/8/34). This immunogen (H1HA0HA6) and its circular permutant (H1HA6) were well folded and provided complete protection against homologous viral challenge. Antisera of immunized mice showed cross-reactivity with HA proteins of different strains and subtypes. Although no neutralization was observable in a conventional neutralization assay, sera of immunized guinea pigs competed with a broadly neutralizing antibody, CR6261, for binding to recombinant Viet/04 HA protein, suggesting that CR6261-like antibodies were elicited by the immunogens. Stem domain immunogens from a seasonal H1N1 strain (A/NC/20/99) and a recent pandemic strain (A/Cal/07/09) provided cross-protection against A/PR/8/34 viral challenge. HA2-containing stem domain immunogens therefore have the potential to provide subtype-specific protection.


Journal of Virology | 2005

Characterization of gp120 and its single-chain derivatives, gp120-CD4D12 and gp120-M9: implications for targeting the CD4i epitope in human immunodeficiency virus vaccine design.

Raghavan Varadarajan; Deepak Sharma; Kausik Chakraborty; Mayuri Patel; Michael Citron; Prem Sinha; Ramkishor Yadav; Umar Rashid; Sarah Kennedy; Debra M. Eckert; Romas Geleziunas; David Bramhill; William A. Schleif; Xiaoping Liang; John W. Shiver

ABSTRACT Single-chain derivatives of JRFL gp120 linked to the first two domains of human CD4 (gp120-CD4D12) or to the CD4 miniprotein analog CD4M9 (gp120-M9), have been constructed. Biacore studies revealed that gp120-CD4D12 and gp120-M9 bound to antibody 17b with dissociation constants of 0.8 and 25 nM, respectively, at pH 7.0, while gp120 alone did not bind. The binding of gp120-CD4D12 to 17b is not affected by the addition of excess soluble CD4D12, while the binding of gp120-M9 is enhanced. This finding indicates that the M9 component of the single chain interacts relatively weakly with gp120 and can be displaced by soluble CD4D12. Immunogenicity studies of gp120, gp120-CD4D12, and gp120-M9 were carried out with guinea pigs. All three molecules were highly immunogenic. The resulting antisera were examined for neutralizing activities against various human immunodeficiency virus type 1 isolates. Broadly neutralizing activity was observed only with sera generated against gp120-CD4D12. These antisera were depleted of anti-CD4D12 antibodies by being passed over a column containing immobilized CD4D12. The depleted sera showed a loss of broadly neutralizing activity. Sera that were affinity purified over a column containing immobilized gp120-M9 also lacked such neutralizing activity. This finding suggests that the broadly neutralizing response observed is exclusively due to anti-CD4 antibodies. Competition experiments showed that only antisera generated against gp120-CD4D12 competed with the CD4i antibody 17b and that this activity was not affected by depletion of anti-CD4 antibodies. The data indicate that although antibodies targeting the CD4i epitope were generated by the gp120-CD4D12 immunogen, these antibodies were nonneutralizing.


Virology | 2009

Characterizations of four monoclonal antibodies against M2 protein ectodomain of influenza A virus.

Tong-Ming Fu; Daniel C. Freed; Melanie Horton; Jiang Fan; Michael Citron; Joseph G. Joyce; Victor M. Garsky; Danilo R. Casimiro; Qinjian Zhao; John W. Shiver; Xiaoping Liang

M2 protein of influenza A virus has been implicated as a target for vaccines with broad cross-strain coverage. Studies in small animal models have shown that antibody responses induced by 23-mer M2 peptide vaccines can provide protection against influenza A virus challenge. To study antiviral mechanisms of Merck M2-OMPC conjugate vaccine, we generated and characterized four M2 peptide-specific monoclonal antibodies (mAbs). Here we demonstrated that the protection by our M2 mAbs is independent of NK-mediated effector functions in mice. The protective mAbs preferentially bind to M2 multimers composed of two or more M2 peptides in parallel orientation. Our findings indicate that the protective M2 Ab prefer to bind to epitopes located within the N-terminal 10 amino acids of the M2 peptide, and the epitopes are likely formed by two M2 peptides in parallel orientation. The implications of these results in antiviral mechanisms of immune responses induced by M2 vaccines are discussed.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Vaccination with peptide mimetics of the gp41 prehairpin fusion intermediate yields neutralizing antisera against HIV-1 isolates

Elisabetta Bianchi; Joseph G. Joyce; Michael D. Miller; Adam C. Finnefrock; Xiaoping Liang; Marco Finotto; Paolo Ingallinella; Philip M. McKenna; Michael Citron; Elizabeth Ottinger; Robert W. Hepler; Renee Hrin; Deborah D. Nahas; Chengwei Wu; David C. Montefiori; John W. Shiver; Antonello Pessi; Peter S. Kim

Eliciting a broadly neutralizing polyclonal antibody response against HIV-1 remains a major challenge. One approach to vaccine development is prevention of HIV-1 entry into cells by blocking the fusion of viral and cell membranes. More specifically, our goal is to elicit neutralizing antibodies that target a transient viral entry intermediate (the prehairpin intermediate) formed by the HIV-1 gp41 protein. Because this intermediate is transient, a stable mimetic is required to elicit an immune response. Previously, a series of engineered peptides was used to select a mAb (denoted D5) that binds to the surface of the gp41 prehairpin intermediate, as demonstrated by x-ray crystallographic studies. D5 inhibits the replication of HIV-1 clinical isolates, providing proof-of-principle for this vaccine approach. Here, we describe a series of peptide mimetics of the gp41 prehairpin intermediate designed to permit a systematic analysis of the immune response generated in animals. To improve the chances of detecting weak neutralizing polyclonal responses, two strategies were employed in the initial screening: use of a neutralization-hypersensitive virus and concentration of the IgG fraction from immunized animal sera. This allowed incremental improvements through iterative cycles of design, which led to vaccine candidates capable of generating a polyclonal antibody response, detectable in unfractionated sera, that neutralize tier 1 HIV-1 and simian HIV primary isolates in vitro. Our findings serve as a starting point for the design of more potent immunogens to elicit a broadly neutralizing response against the gp41 prehairpin intermediate.

Collaboration


Dive into the Michael Citron's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Cheryl Callahan

United States Military Academy

View shared research outputs
Top Co-Authors

Avatar

Tong-Ming Fu

United States Military Academy

View shared research outputs
Researchain Logo
Decentralizing Knowledge