scholarly journals Correction: Immunologic Consequences of Francisella tularensis Live Vaccine Strain Infection: Role of the Innate Immune Response in Infection and Immunity

2021 ◽  
pp. ji2100505
Author(s):  
Leah E. Cole ◽  
Karen L. Elkins ◽  
Suzanne M. Michalek ◽  
Nilofer Qureshi ◽  
Linda J. Eaton ◽  
...  
2005 ◽  
Vol 73 (4) ◽  
pp. 2306-2311 ◽  
Author(s):  
Nathalie S. Duckett ◽  
Sofia Olmos ◽  
Douglas M. Durrant ◽  
Dennis W. Metzger

ABSTRACT Francisella tularensis is a gram-negative intracellular bacterium that can induce lethal respiratory infection in humans and rodents. However, little is known about the role of innate or adaptive immunity in protection from respiratory tularemia. In the present study, the role of interleukin-12 (IL-12) in inducing protective immunity in the lungs against intranasal infection of mice with the live vaccine strain (LVS) of F. tularensis was investigated. It was found that gamma interferon (IFN-γ) and IL-12 were strictly required for protection, since mice deficient in IFN-γ, IL-12 p35, or IL-12 p40 all succumbed to LVS doses that were sublethal for wild-type mice. Furthermore, exogenous IL-12 treatment 24 h before intranasal infection with a lethal dose of LVS (10,000 CFU) significantly decreased bacterial loads in the lungs, livers, and spleens of wild-type BALB/c and C57BL/6 mice and allowed the animals to survive infection; such protection was not observed in IFN-γ-deficient mice. The resistance induced by IL-12 to LVS infection was still observed in NK cell-deficient beige mice but not in CD8−/− mice. These results demonstrate that exogenous IL-12 delivered intranasally can prevent respiratory tularemia through a mechanism that is at least partially dependent upon the expression of IFN-γ and CD8 T cells.


2008 ◽  
Vol 76 (9) ◽  
pp. 4311-4321 ◽  
Author(s):  
Roberto De Pascalis ◽  
Betsy C. Taylor ◽  
Karen L. Elkins

ABSTRACT Francisella tularensis, a small gram-negative intracellular bacterium responsible for causing tularemia, is highly pathogenic and classified as a category A agent of bioterrorism. As for other intracellular pathogens, successful protective immune responses to Francisella tularensis require rapid and efficient induction of gamma interferon (IFN-γ) production. Studies using intracellular bacteria such as Listeria monocytogenes as well as Francisella suggest that natural killer (NK) and T cells are important sources of IFN-γ. However, comprehensive characterization of specific sources of IFN-γ produced during Francisella infection in vivo remains incomplete, and depletion of NK cells before infection of mice with the F. tularensis live vaccine strain (LVS) has little impact on the course or outcome of infection. In this study, we determined the cell subpopulations that respond quickly to intradermal F. tularensis LVS infection of mice by producing IFN-γ within hours to a few days. Splenic and liver lymphocytes were obtained from LVS-infected mice and analyzed for IFN-γ mRNA by reverse transcription-PCR, for intracellular cytokine expression by multiparameter flow cytometry, and for ex vivo production of IFN-γ protein by enzyme-linked immunosorbent assay. Cells producing IFN-γ were readily detectable by day 3 after infection, and numbers progressively increased through days 5 to 7. Importantly, the cell types responsible for IFN-γ production were much more varied than expected: these included not only NK cells and T cells, which might be predicted, but also other cells, including dendritic cells (DCs), “NK DCs,” NK T cells, and neutrophils. Most importantly, since RAG-1 knockout mice appeared to exhibit a frequency of IFN-γ-producing cells comparable to that of intact wild-type mice, early IFN-γ production by innate immune cells does not depend on the presence of T or B cells.


2006 ◽  
Vol 176 (11) ◽  
pp. 6888-6899 ◽  
Author(s):  
Leah E. Cole ◽  
Karen L. Elkins ◽  
Suzanne M. Michalek ◽  
Nilofer Qureshi ◽  
Linda J. Eaton ◽  
...  

2006 ◽  
Vol 117 (5) ◽  
pp. 1186-1188 ◽  
Author(s):  
Claudette L. Fuller ◽  
Katherine C. Brittingham ◽  
Matthew J. Hepburn ◽  
James W. Martin ◽  
Patricia L. Petitt ◽  
...  

2013 ◽  
Vol 81 (6) ◽  
pp. 2022-2027 ◽  
Author(s):  
Dennis W. Metzger ◽  
Sharon L. Salmon ◽  
Girish Kirimanjeswara

ABSTRACTWe investigated the role of interleukin-10 (IL-10) in cutaneous and pulmonary infection withFrancisella tularensis. We found that after intradermal challenge of mice with the live vaccine strain (LVS) ofF. tularensis, splenic IL-10 levels increased rapidly and reached a peak 5 days after infection. However, IL-10 expression after infection was detrimental, since IL-10−/−mice showed increased bacterial clearance and were resistant to an infectious dose (>106CFU/mouse) that was uniformly lethal for IL-10+/+mice. Furthermore, IL-10+/+mice treated with neutralizing anti-IL-10R monoclonal antibody were able to survive lethal cutaneous LVS challenge. The presence of IL-10 appeared to restrain the expression of IL-17, since high levels of splenic IL-17 were observed after intradermal LVS infection only in IL-10−/−mice. Furthermore, treatment with neutralizing anti-IL-17R antibody ablated the enhanced survival observed in IL-10−/−mice. However, neutralization of IL-10 activity in IL-17R−/−mice failed to provide protection. Thus, IL-10 suppresses a protective IL-17 response that is necessary for resistance to cutaneous LVS infection. Surprisingly, however, IL-10−/−mice were significantly more susceptible to pulmonary infection with LVS. Finally, although IL-10 is a critical and novel regulator of immunity toF. tularensisLVS infection, its effects were masked during infection with the highly virulent SchuS4 strain. Taken together, these findings suggest that differentially regulating expression of the IL-10 pathway in various tissues could ultimately have prophylactic and therapeutic benefits for protection against tularemia.


2021 ◽  
Author(s):  
Maha Alqahtani ◽  
Zhuo Ma ◽  
Kayla Fantone ◽  
Meenakshi Malik ◽  
Chandra Shekhar Bakshi

Francisella tularensis (F. tularensis) is a facultative intracellular, Gram-negative bacterium that causes a fatal disease known as tularemia. Due to its extremely high virulence, ease of spread by aerosolization, and the potential to be used as a bioterror agent, F. tularensis is classified by the CDC as a Tier 1 Category A Select Agent. Previous studies have demonstrated the roles of inflammasome sensors; absent in melanoma 2 (AIM2) and NLRP3, in the generation of innate immune responses to F. tularensis infection. However, contributions of both the AIM2 and NLRP3 in the development of vaccine-induced adaptive immune responses against F. tularensis are not known. This study determined the contributions of Aim2 and Nlrp3-inflammasome sensors in vaccine-induced immune responses in a mouse model of respiratory tularemia. We developed a model to vaccinate the Aim2 and Nlrp3-deficient mice (Aim2-/- and Nlrp3-/-) using the emrA1 mutant of F. tularensis live vaccine strain (LVS). The results demonstrate that the innate immune responses in Aim2-/- and Nlrp3-/- mice vaccinated with the emrA1 mutant differ from their wild-type counterparts. However, despite these differences in the innate immune responses, both Aim2-/- and Nlrp3-/- mice are fully protected against an intranasal lethal challenge dose of F. tularensis LVS. Moreover, the lack of both Aim2 and Nlrp3 inflammasome sensors does not affect the production of the vaccination-induced antibody and cell-mediated responses. Overall, this study reports a novel finding that both Aim2 and Nlrp3 are dispensable for vaccination-induced immunity against respiratory tularemia caused by F. tularensis.


2015 ◽  
Vol 6 (1) ◽  
Author(s):  
Viktoria V. Мokrozub ◽  
Liudmyla M. Lazarenko ◽  
Liubov M. Sichel ◽  
Lidia P. Babenko ◽  
Petro M. Lytvyn ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document