Yersinia pestis (Yp) - one of the world's most virulent human pathogens - is the gram-negative bacterium that causes pneumonic plague. Virulent, antibiotic-resistant, Yp strains exist and Cold War scientists devised means to effectively aerosolize Yp. Thus, there is grave concern that Yp will be exploited as a bioweapon. To thwart that possibility, it is essential that we develop effective countermeasures. Recent primate studies suggest that the leading vaccine candidate - a recombinant F1-V fusion protein (rF1V) - may not provide sufficient protection. Moreover, resourceful bioweapon engineers could circumvent this vaccine with an F1-negative V-variant strain. While the rF1V vaccine primarily stimulates antibody-mediated humoral immunity, T cell-dependent cellular immunity comprises a second means by which vaccines can prime long-lived protection. However, it is widely accepted that the extreme virulence of Yp results, in large part, from plasmid-encoded factors that dampen inflammation and debilitate phagocytes, thereby compromising cell-mediated defense. Thus, plague vaccine researchers have devoted little attention to cellular immunity. Nevertheless, the Progress Report demonstrates that T cells can protect against pulmonary Yp infection. As such, we propose that next-generation pneumonic plague vaccines should strive to prime both humoral and cellular immunity, as well as incorporate new antigens.
In Aim 1 of this application for grant renewal, we will directly measure the extent to which pulmonary Yp infection suppresses na?ve and effector/memory T cell responses in vivo and identify mechanisms underlying any suppression that exists.
In Aim 2, we will generate T cell clones that confer protection in mice, identify their cognate protein antigens using a series of complementary genetic, molecular and biochemical approaches, and then quantify the utility of these antigens as vaccines. We will also determine which antigens confer synergistic protection when combined with V-specific antibodies. Our findings will allow next-generation F1/V-based vaccines to harness both the neutralizing capacities of antibodies and the antimicrobial capacities of cellular immunity, while simultaneously reducing opportunities for circumvention by bioweapon engineers. Moreover, our studies will also generate powerful new tools for deciphering the fascinating interplay between Yp virulence factors and host defense mechanisms, thereby advancing basic research aimed at exploiting cellular immunity for defense against virulent bacterial pathogens that infect the lung.

Public Health Relevance

To counter the potential use of pneumonic plague as a bioweapon, it is essential that we develop an effective vaccine. Recent animal studies suggest that the current vaccine candidate, which primarily stimulates B cells to make plague-fighting antibodies, may not suffice. This proposal aims to fundamentally improve that vaccine by incorporating the protective capacities of both B and T cells.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Research Project (R01)
Project #
5R01AI061577-06
Application #
7769910
Study Section
Special Emphasis Panel (ZRG1-IMM-K (02))
Program Officer
Mukhopadhyay, Suman
Project Start
2004-09-30
Project End
2014-02-28
Budget Start
2010-03-01
Budget End
2011-02-28
Support Year
6
Fiscal Year
2010
Total Cost
$489,018
Indirect Cost
Name
Trudeau Institute, Inc.
Department
Type
DUNS #
020658969
City
Saranac Lake
State
NY
Country
United States
Zip Code
12983
Szaba, Frank M; Kummer, Lawrence W; Duso, Debra K et al. (2014) TNF? and IFN? but not perforin are critical for CD8 T cell-mediated protection against pulmonary Yersinia pestis infection. PLoS Pathog 10:e1004142
Hickey, Anthony J; Lin, Jr-Shiuan; Kummer, Lawrence W et al. (2013) Intranasal prophylaxis with CpG oligodeoxynucleotide can protect against Yersinia pestis infection. Infect Immun 81:2123-32
Luo, Deyan; Lin, Jr-Shiuan; Parent, Michelle A et al. (2013) Fibrin facilitates both innate and T cell-mediated defense against Yersinia pestis. J Immunol 190:4149-61
Zhang, Yue; Mena, Patricio; Romanov, Galina et al. (2012) A protective epitope in type III effector YopE is a major CD8 T cell antigen during primary infection with Yersinia pseudotuberculosis. Infect Immun 80:206-14
Luo, Deyan; Szaba, Frank M; Kummer, Lawrence W et al. (2011) Protective roles for fibrin, tissue factor, plasminogen activator inhibitor-1, and thrombin activatable fibrinolysis inhibitor, but not factor XI, during defense against the gram-negative bacterium Yersinia enterocolitica. J Immunol 187:1866-76
Lin, Jr-Shiuan; Szaba, Frank M; Kummer, Lawrence W et al. (2011) Yersinia pestis YopE contains a dominant CD8 T cell epitope that confers protection in a mouse model of pneumonic plague. J Immunol 187:897-904
Lin, Jr-Shiuan; Kummer, Lawrence W; Szaba, Frank M et al. (2011) IL-17 contributes to cell-mediated defense against pulmonary Yersinia pestis infection. J Immunol 186:1675-84
Lin, Jr-Shiuan; Park, Steven; Adamovicz, Jeffrey J et al. (2010) TNF? and IFN? contribute to F1/LcrV-targeted immune defense in mouse models of fully virulent pneumonic plague. Vaccine 29:357-62
Robinson, Richard T; Khader, Shabaana A; Martino, Cynthia A et al. (2010) Mycobacterium tuberculosis infection induces il12rb1 splicing to generate a novel IL-12Rbeta1 isoform that enhances DC migration. J Exp Med 207:591-605
Szaba, Frank M; Kummer, Lawrence W; Wilhelm, Lindsey B et al. (2009) D27-pLpxL, an avirulent strain of Yersinia pestis, primes T cells that protect against pneumonic plague. Infect Immun 77:4295-304

Showing the most recent 10 out of 20 publications