Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(6): e1011915, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861581

ABSTRACT

Mycobacterium tuberculosis infects two billion people across the globe, and results in 8-9 million new tuberculosis (TB) cases and 1-1.5 million deaths each year. Most patients have no known genetic basis that predisposes them to disease. Here, we investigate the complex genetic basis of pulmonary TB by modelling human genetic diversity with the Diversity Outbred mouse population. When infected with M. tuberculosis, one-third develop early onset, rapidly progressive, necrotizing granulomas and succumb within 60 days. The remaining develop non-necrotizing granulomas and survive longer than 60 days. Genetic mapping using immune and inflammatory mediators; and clinical, microbiological, and granuloma correlates of disease identified five new loci on mouse chromosomes 1, 2, 4, 16; and three known loci on chromosomes 3 and 17. Further, multiple positively correlated traits shared loci on chromosomes 1, 16, and 17 and had similar patterns of allele effects, suggesting these loci contain critical genetic regulators of inflammatory responses to M. tuberculosis. To narrow the list of candidate genes, we used a machine learning strategy that integrated gene expression signatures from lungs of M. tuberculosis-infected Diversity Outbred mice with gene interaction networks to generate scores representing functional relationships. The scores were used to rank candidates for each mapped trait, resulting in 11 candidate genes: Ncf2, Fam20b, S100a8, S100a9, Itgb5, Fstl1, Zbtb20, Ddr1, Ier3, Vegfa, and Zfp318. Although all candidates have roles in infection, inflammation, cell migration, extracellular matrix remodeling, or intracellular signaling, and all contain single nucleotide polymorphisms (SNPs), SNPs in only four genes (S100a8, Itgb5, Fstl1, Zfp318) are predicted to have deleterious effects on protein functions. We performed methodological and candidate validations to (i) assess biological relevance of predicted allele effects by showing that Diversity Outbred mice carrying PWH/PhJ alleles at the H-2 locus on chromosome 17 QTL have shorter survival; (ii) confirm accuracy of predicted allele effects by quantifying S100A8 protein in inbred founder strains; and (iii) infection of C57BL/6 mice deficient for the S100a8 gene. Overall, this body of work demonstrates that systems genetics using Diversity Outbred mice can identify new (and known) QTLs and functionally relevant gene candidates that may be major regulators of complex host-pathogens interactions contributing to granuloma necrosis and acute inflammation in pulmonary TB.

2.
Vaccines (Basel) ; 12(3)2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38543876

ABSTRACT

Mycobacterium bovis Bacillus Calmette-Guérin (BCG) protects against childhood tuberculosis; and unlike most vaccines, BCG broadly impacts immunity to other pathogens and even some cancers. Early in the COVID-19 pandemic, epidemiological studies identified a protective association between BCG vaccination and outcomes of SARS-CoV-2, but the associations in later studies were inconsistent. We sought possible reasons and noticed the study populations often lived in the same country. Since individuals from the same regions can share common ancestors, we hypothesized that genetic background could influence associations between BCG and SARS-CoV-2. To explore this hypothesis in a controlled environment, we performed a pilot study using Diversity Outbred mice. First, we identified amino acid sequences shared by BCG and SARS-CoV-2 spike protein. Next, we tested for IgG reactive to spike protein from BCG-vaccinated mice. Sera from some, but not all, BCG-vaccinated Diversity Outbred mice contained higher levels of IgG cross-reactive to SARS-CoV-2 spike protein than sera from BCG-vaccinated C57BL/6J inbred mice and unvaccinated mice. Although larger experimental studies are needed to obtain mechanistic insight, these findings suggest that genetic background may be an important variable contributing to different associations observed in human randomized clinical trials evaluating BCG vaccination on SARS-CoV-2 and COVID-19.

3.
PLoS Pathog ; 20(3): e1012069, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38452145

ABSTRACT

Mycobacterium tuberculosis (M.tb.) infection leads to over 1.5 million deaths annually, despite widespread vaccination with BCG at birth. Causes for the ongoing tuberculosis endemic are complex and include the failure of BCG to protect many against progressive pulmonary disease. Host genetics is one of the known factors implicated in susceptibility to primary tuberculosis, but less is known about the role that host genetics plays in controlling host responses to vaccination against M.tb. Here, we addressed this gap by utilizing Diversity Outbred (DO) mice as a small animal model to query genetic drivers of vaccine-induced protection against M.tb. DO mice are a highly genetically and phenotypically diverse outbred population that is well suited for fine genetic mapping. Similar to outcomes in people, our previous studies demonstrated that DO mice have a wide range of disease outcomes following BCG vaccination and M.tb. challenge. In the current study, we used a large population of BCG-vaccinated/M.tb.-challenged mice to perform quantitative trait loci mapping of complex infection traits; these included lung and spleen M.tb. burdens, as well as lung cytokines measured at necropsy. We found sixteen chromosomal loci associated with complex infection traits and cytokine production. QTL associated with bacterial burdens included a region encoding major histocompatibility antigens that are known to affect susceptibility to tuberculosis, supporting validity of the approach. Most of the other QTL represent novel associations with immune responses to M.tb. and novel pathways of cytokine regulation. Most importantly, we discovered that protection induced by BCG is a multigenic trait, in which genetic loci harboring functionally-distinct candidate genes influence different aspects of immune responses that are crucial collectively for successful protection. These data provide exciting new avenues to explore and exploit in developing new vaccines against M.tb.


Subject(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculosis Vaccines , Tuberculosis , Humans , Animals , Mice , BCG Vaccine/genetics , Tuberculosis/genetics , Tuberculosis/prevention & control , Tuberculosis/microbiology , Tuberculosis Vaccines/genetics , Vaccination , Genetic Loci , Cytokines/genetics , Antigens, Bacterial
4.
PLoS One ; 18(8): e0289358, 2023.
Article in English | MEDLINE | ID: mdl-37535648

ABSTRACT

Although BCG has been used for almost 100 years to immunize against Mycobacterium tuberculosis, TB remains a global public health threat. Numerous clinical trials are underway studying novel vaccine candidates and strategies to improve or replace BCG, but vaccine development still lacks a well-defined set of immune correlates to predict vaccine-induced protection against tuberculosis. This study aimed to address this gap by examining transcriptional responses to BCG vaccination in C57BL/6 inbred mice, coupled with protection studies using Diversity Outbred mice. We evaluated relative gene expression in blood obtained from vaccinated mice, because blood is easily accessible, and data can be translated to human studies. We first determined that the average peak time after vaccination is 14 days for gene expression of a small subset of immune-related genes in inbred mice. We then performed global transcriptomic analyses using whole blood samples obtained two weeks after mice were vaccinated with BCG. Using comparative bioinformatic analyses and qRT-PCR validation, we developed a working correlate panel of 18 genes that were highly correlated with administration of BCG but not heat-killed BCG. We then tested this gene panel using BCG-vaccinated Diversity Outbred mice and revealed associations between the expression of a subset of genes and disease outcomes after aerosol challenge with M. tuberculosis. These data therefore demonstrate that blood-based transcriptional immune correlates measured within a few weeks after vaccination can be derived to predict protection against M. tuberculosis, even in outbred populations.


Subject(s)
Mycobacterium tuberculosis , Tuberculosis , Animals , Humans , Mice , BCG Vaccine , Collaborative Cross Mice , Mice, Inbred C57BL , Tuberculosis/microbiology , Mycobacterium tuberculosis/genetics , Vaccination
5.
Infect Immun ; 91(7): e0016823, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37338410

ABSTRACT

Tuberculosis is still the leading cause of death globally from any infectious disease, despite the widespread use of the live attenuated vaccine Bacille Calmette Guerin (BCG). While BCG has some efficacy against disseminated TB disease in children, protection wanes into adulthood resulting in over 1.8 million TB deaths per year. This has led to efforts to develop novel vaccine candidates that either replace or boost BCG, as well as to test novel delivery mechanisms to enhance BCG's efficacy. Traditional BCG vaccination is performed as an intradermal (ID) injection but delivering BCG by an alternate route may enhance the depth and breadth of protection. Previously, we demonstrated that phenotypically and genotypically disparate Diversity Outbred (DO) mice have heterogenous responses to M. tuberculosis challenge following intradermal BCG vaccination. Here, we utilize DO mice to examine BCG-induced protection when BCG is delivered systemically via intravenous (IV) administration. We find that DO mice vaccinated with IV BCG had a greater distribution of BCG throughout their organs compared to ID-vaccinated animals. However, compared to ID-vaccinated mice, M. tuberculosis burdens in lungs and spleens were not significantly reduced in animals vaccinated with BCG IV, nor was lung inflammation significantly altered. Nonetheless, DO mice that received BCG IV had increased survival over those vaccinated by the traditional ID route. Thus, our results suggest that delivering BCG by the alternate IV route enhances protection as detected in this diverse small animal model.


Subject(s)
Mycobacterium bovis , Mycobacterium tuberculosis , Tuberculosis , Animals , Mice , BCG Vaccine , Collaborative Cross Mice , Tuberculosis/prevention & control , Vaccination
6.
PLoS One ; 16(3): e0249142, 2021.
Article in English | MEDLINE | ID: mdl-33760886

ABSTRACT

CCR2 is the major chemokine receptor that regulates appropriate trafficking of inflammatory monocytes, but the role of this chemokine receptor and its ligands during primary and secondary infection with intracellular infections remains incompletely understood. Here we used murine infection with the Live Vaccine Strain (LVS) of Francisella tularensis to evaluate the role of CCR2 during primary and secondary parenteral responses to this prototype intracellular bacterium. We find that mice deficient in CCR2 are highly compromised in their ability to survive intradermal infection with LVS, indicating the importance of this receptor during primary parenteral responses. Interestingly, this defect could not be readily attributed to the activities of the known murine CCR2 ligands MCP-1/CCL2, MCP-3/CCL7, or MCP-5/CCL12. Nonetheless, CCR2 knockout mice vaccinated by infection with low doses of LVS generated optimal T cell responses that controlled the intramacrophage replication of Francisella, and LVS-immune CCR2 knockout mice survived maximal lethal Francisella challenge. Thus, fully protective adaptive immune memory responses to this intracellular bacterium can be readily generated in the absence of CCR2.


Subject(s)
Francisella tularensis/physiology , Receptors, CCR2/genetics , Tularemia/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL7/deficiency , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Receptors, CCR2/deficiency , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tularemia/mortality , Tularemia/pathology , Tularemia/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
7.
PLoS One ; 15(8): e0237034, 2020.
Article in English | MEDLINE | ID: mdl-32745117

ABSTRACT

Production of IFN-γ is a key innate immune mechanism that limits replication of intracellular bacteria such as Francisella tularensis (Ft) until adaptive immune responses develop. Previously, we demonstrated that the host cell types responsible for IFN-γ production in response to murine Francisella infection include not only natural killer (NK) and T cells, but also a variety of myeloid cells. However, production of IFN-γ by mouse dendritic cells (DC) is controversial. Here, we directly demonstrated substantial production of IFN-γ by DC, as well as hybrid NK-DC, from LVS-infected wild type C57BL/6 or Rag1 knockout mice. We demonstrated that the numbers of conventional DC producing IFN-γ increased progressively over the course of 8 days of LVS infection. In contrast, the numbers of conventional NK cells producing IFN-γ, which represented about 40% of non-B/T IFN-γ-producing cells, peaked at day 4 after LVS infection and declined thereafter. This pattern was similar to that of hybrid NK-DC. To further confirm IFN-γ production by infected cells, DC and neutrophils were sorted from naïve and LVS-infected mice and analyzed for gene expression. Quantification of LVS by PCR revealed the presence of Ft DNA not only in macrophages, but also in highly purified, IFN-γ producing DC and neutrophils. Finally, production of IFN-γ by infected DC was confirmed by immunohistochemistry and confocal microscopy. Notably, IFN-γ production patterns similar to those in wild type mice were observed in cells derived from LVS-infected TLR2, TLR4, and TLR2xTLR9 knockout (KO) mice, but not from MyD88 KO mice. Taken together, these studies demonstrate the pivotal roles of DC and MyD88 in IFN-γ production and in initiating innate immune responses to this intracellular bacterium.


Subject(s)
Interferon-gamma/metabolism , Myeloid Differentiation Factor 88/genetics , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Francisella tularensis/immunology , Immunity, Innate/immunology , Killer Cells, Natural/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Neutrophils/metabolism , Spleen/metabolism , T-Lymphocytes/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Toll-Like Receptors/immunology , Tularemia/microbiology
8.
mSphere ; 5(2)2020 04 15.
Article in English | MEDLINE | ID: mdl-32295871

ABSTRACT

Many studies of Mycobacterium tuberculosis infection and immunity have used mouse models. However, outcomes of vaccination and challenge with M. tuberculosis in inbred mouse strains do not reflect the full range of outcomes seen in people. Previous studies indicated that the novel Diversity Outbred (DO) mouse population exhibited a spectrum of outcomes after primary aerosol infection with M. tuberculosis Here, we demonstrate the value of this novel mouse population for studies of vaccination against M. tuberculosis aerosol challenge. Using the only currently licensed tuberculosis vaccine, we found that the DO population readily controlled systemic Mycobacterium bovis BCG bacterial burdens and that BCG vaccination significantly improved survival across the DO population upon challenge with M. tuberculosis Many individual DO mice that were vaccinated with BCG and then challenged with M. tuberculosis exhibited low bacterial burdens, low or even no systemic dissemination, little weight loss, and only minor lung pathology. In contrast, some BCG-vaccinated DO mice progressed quickly to fulminant disease upon M. tuberculosis challenge. Across the population, most of these disease parameters were at most modestly correlated with each other and were often discordant. This result suggests the need for a multiparameter metric to better characterize "disease" and "protection," with closer similarity to the complex case definitions used in people. Taken together, these results demonstrate that DO mice provide a novel small-animal model of vaccination against tuberculosis that better reflects the wide spectrum of outcomes seen in people.IMPORTANCE We vaccinated the Diversity Outbred (DO) population of mice with BCG, the only vaccine currently used to protect against tuberculosis, and then challenged them with M. tuberculosis by aerosol. We found that the BCG-vaccinated DO mouse population exhibited a wide range of outcomes, in which outcomes in individual mice ranged from minimal respiratory or systemic disease to fulminant disease and death. The breadth of these outcomes appears similar to the range seen in people, indicating that DO mice may serve as an improved small-animal model to study tuberculosis infection and immunity. Moreover, sophisticated tools are available for the use of these mice to map genes contributing to control of vaccination. Thus, the present studies provided an important new tool in the fight against tuberculosis.


Subject(s)
Collaborative Cross Mice/microbiology , Disease Models, Animal , Tuberculosis Vaccines/immunology , Tuberculosis/genetics , Tuberculosis/immunology , Animals , Collaborative Cross Mice/immunology , Female , Genetic Variation , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis , Tuberculosis/prevention & control , Vaccination
9.
Tuberculosis (Edinb) ; 121: 101914, 2020 03.
Article in English | MEDLINE | ID: mdl-32279870

ABSTRACT

Mycobacteria are important causes of disease in human and animal hosts. Diseases caused by mycobacteria include leprosy, tuberculosis (TB), nontuberculous mycobacteria (NTM) infections and Buruli Ulcer. To better understand and treat mycobacterial disease, clinicians, veterinarians and scientists use a range of discipline-specific approaches to conduct basic and applied research, including conducting epidemiological surveys, patient studies, wildlife sampling, animal models, genetic studies and computational simulations. To foster the exchange of knowledge and collaboration across disciplines, the Many Hosts of Mycobacteria (MHM) conference series brings together clinical, veterinary and basic scientists who are dedicated to advancing mycobacterial disease research. Started in 2007, the MHM series recently held its 8th conference at the Albert Einstein College of Medicine (Bronx, NY). Here, we review the diseases discussed at MHM8 and summarize the presentations on research advances in leprosy, NTM and Buruli Ulcer, human and animal TB, mycobacterial disease comorbidities, mycobacterial genetics and 'omics, and animal models. A mouse models workshop, which was held immediately after MHM8, is also summarized. In addition to being a resource for those who were unable to attend MHM8, we anticipate this review will provide a benchmark to gauge the progress of future research concerning mycobacteria and their many hosts.


Subject(s)
Bacteriology , Biomedical Research , Infectious Disease Medicine , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium/pathogenicity , Tuberculosis/microbiology , Animals , Congresses as Topic , Diffusion of Innovation , Disease Models, Animal , Host-Pathogen Interactions , Humans , Mycobacterium/genetics , Mycobacterium Infections, Nontuberculous/diagnosis , Mycobacterium Infections, Nontuberculous/epidemiology , Tuberculosis/diagnosis , Tuberculosis/epidemiology
10.
Tuberculosis (Edinb) ; 120: 101895, 2020 01.
Article in English | MEDLINE | ID: mdl-32090856

ABSTRACT

New vaccines are needed to combat the public health threat posed by M. tuberculosis (M. tb), but no correlates have been defined to aid vaccine development. Using mouse models, we previously developed an in vitro system that measures the ability of M. tb-immune lymphocytes to control bacterial replication during co-culture with M. tb-infected macrophages. We demonstrated that the degree of in vitro growth control by lymphocytes from mice given vaccines of varying efficacy reflected the relative degree of in vivo protection against lethal challenge. Further, using targeted analyses of gene expression in lymphocytes recovered from co-cultures, we found mediators whose relative expression also correlated with in vitro and in vivo outcomes. Here we advanced those findings by employing genome-wide expression analyses. We first screened splenocytes recovered from co-cultures by microarray, revealing additional genes whose expression correlated with protection. After applying pathway analyses to down-select gene candidates, we used both splenocytes and peripheral blood lymphocytes to validate microarray findings by qRT-PCR. We then subjected data from top candidates to rigorous statistical analyses. Resulting correlate candidates, including CXCL9, IFN-γ, and CCL5, significantly predicted protection with high specificity. These findings therefore refine and extend a panel of relevant immune correlates to advance vaccine development.


Subject(s)
Gene Expression Profiling , Macrophages/drug effects , Mycobacterium tuberculosis/drug effects , T-Lymphocytes/drug effects , Transcriptome , Tuberculosis Vaccines/pharmacology , Animals , Cells, Cultured , Chemokine CCL5/genetics , Chemokine CXCL9/genetics , Coculture Techniques , Genome-Wide Association Study , Interferon-gamma/genetics , Macrophages/immunology , Macrophages/microbiology , Male , Mice, Inbred C57BL , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/growth & development , Mycobacterium tuberculosis/immunology , Oligonucleotide Array Sequence Analysis , T-Lymphocytes/immunology , Vaccination
11.
Pathog Dis ; 76(7)2018 10 01.
Article in English | MEDLINE | ID: mdl-30137434

ABSTRACT

Francisella tularensis is a gram-negative organism found in many regions of the world. F. tularensis can cause a fatal, febrile illness, although these natural tularemia infections are rare in the United States. However, the development of F. tularensis as a potential weapon of bioterrorism during the Cold War spurred the development of a live attenuated vaccine, LVS, from F. tularensis subsp. holarctica in the 1960s. Two colony morphology variants, LVS-G and LVS-R, were generated from parental LVS by plate passage and by acridine orange mutagenesis, respectively. In vaccinated mice, LVS-G and LVS-R exhibit altered immunogenicity and protective capacities. While the exact nature of the mutations in these strains was unknown, previous studies indicated that both had altered lipopolysaccharide structures. To better understand the impact of these mutations on LVS' immunogenicity, we sequenced the genomes of LVS-G and LVS-R as well as our parental laboratory stock of LVS, originally obtained from ATCC, and compared these to the F. tularensis subsp. holarctica LVS genome currently deposited in GenBank. The results indicate that the genomic sequence of ATCC LVS is nearly identical to that of the human LVS vaccine. Furthermore, a limited number of genomic mutations likely account for the phenotypes of LVS-G and LVS-R.


Subject(s)
Francisella tularensis/genetics , Genome, Bacterial , Genomics , Whole Genome Sequencing , Genotype , Humans , Mutation , United States
12.
Microbes Infect ; 18(12): 758-767, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27475899

ABSTRACT

Our laboratory has employed in vitro and in vivo mouse models based on Francisella tularensis Live Vaccine Strain (LVS)-induced protection to elucidate immune correlates for intracellular bacteria. Among the effectors found was GM-CSF, a pleiotropic cytokine that is integral to the development and proliferation of myeloid cells, including alveolar macrophages. GM-CSF has roles in resistance to primary murine infection with several intracellular pathogens, but its role during Francisella infection is unknown. Francisella is an intracellular pathogen that infects lungs after inhalation, primarily invading alveolar macrophages. Here we show that GM-CSF has route-dependent roles during primary infection of mice with LVS. GM-CSF deficient (GM-CSF KO) mice were slightly more susceptible than wild type to intradermal infection, but had increased resistance to intranasal infection. Similarly, these mice had increased resistance to pulmonary infection with virulent F. tularensis (SchuS4). LVS-vaccinated GM-CSF KO mice had normal adaptive immune responses, as measured by T cell activities after LVS intradermal or intranasal vaccination, and survived lethal secondary LVS challenge. GM-CSF KO mice also had robust humoral responses, producing elevated levels of serum antibodies following LVS vaccination compared to wild type mice. Taken together, our data demonstrates that the absence of GM-CSF improves resistance to pulmonary, but not intradermal, infection with Francisella.


Subject(s)
Francisella tularensis/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Mucosal , Nasal Mucosa/immunology , Skin/immunology , Tularemia/immunology , Animals , Antibodies, Bacterial/blood , Disease Models, Animal , Immunity, Cellular , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
13.
Expert Rev Vaccines ; 15(9): 1183-96, 2016 09.
Article in English | MEDLINE | ID: mdl-27010448

ABSTRACT

Renewed interest in Francisella tularensis has resulted in substantial new information about its pathogenesis and immunology, along with development of useful animal models. While understanding of protective immunity against Francisella remains incomplete, data in both animals and humans suggest that inducing T cell-mediated immunity is crucial for successful vaccination with current candidates such as the Live Vaccine Strain (LVS), with specific antibodies and immune B cells playing supporting roles. Consistent with this idea, recent results indicate that measurements of T cell functions and relative gene expression by immune T cells predict vaccine-induced protection in animal models. Because field trials of new vaccines will be difficult to design, using such measurements to derive potential correlates of protection may be important to bridge between animal efficacy studies and people.


Subject(s)
Bacterial Vaccines/immunology , Bacterial Vaccines/isolation & purification , Francisella tularensis/immunology , Tularemia/prevention & control , Animals , Biomarkers/analysis , Disease Models, Animal , Francisella tularensis/pathogenicity , Humans , T-Lymphocytes/immunology , Tularemia/immunology
14.
Clin Vaccine Immunol ; 22(10): 1096-108, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26269537

ABSTRACT

A critical hindrance to the development of a novel vaccine against Mycobacterium tuberculosis is a lack of understanding of protective correlates of immunity and of host factors involved in a successful adaptive immune response. Studies from our group and others have used a mouse-based in vitro model system to assess correlates of protection. Here, using this coculture system and a panel of whole-cell vaccines with varied efficacy, we developed a comprehensive approach to understand correlates of protection. We compared the gene and protein expression profiles of vaccine-generated immune peripheral blood lymphocytes (PBLs) to the profiles found in immune splenocytes. PBLs not only represent a clinically relevant cell population, but comparing the expression in these populations gave insight into compartmentally specific mechanisms of protection. Additionally, we performed a direct comparison of host responses induced when immune cells were cocultured with either the vaccine strain Mycobacterium bovis BCG or virulent M. tuberculosis. These comparisons revealed host-specific and bacterium-specific factors involved in protection against virulent M. tuberculosis. Most significantly, we identified a set of 13 core molecules induced in the most protective vaccines under all of the conditions tested. Further validation of this panel of mediators as a predictor of vaccine efficacy will facilitate vaccine development, and determining how each promotes adaptive immunity will advance our understanding of antimycobacterial immune responses.


Subject(s)
Bacterial Proteins/genetics , Lymphocyte Subsets/immunology , Macrophages/microbiology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Animals , Antigens, Bacterial/immunology , BCG Vaccine/administration & dosage , BCG Vaccine/immunology , Bacterial Proteins/immunology , Cattle , Coculture Techniques , Cytokines/metabolism , Gene Expression Profiling , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/genetics , Spleen/cytology , Spleen/immunology , Tuberculosis Vaccines/administration & dosage
15.
PLoS One ; 9(10): e109898, 2014.
Article in English | MEDLINE | ID: mdl-25296161

ABSTRACT

Our laboratory's investigations into mechanisms of protective immunity against Francisella tularensis Live Vaccine Strain (LVS) have uncovered mediators important in host defense against primary infection, as well as those correlated with successful vaccination. One such potential correlate was IL-12p40, a pleiotropic cytokine that promotes Th1 T cell function as part of IL-12p70. LVS-infected IL-12p40 deficient knockout (KO) mice maintain a chronic infection, but IL-12p35 KO mice clear LVS infection; thus the role that IL-12p40 plays in immunity to LVS is independent of the IL-12p70 heterodimer. IL-12p40 can also partner with IL-23p19 to create the heterodimeric cytokine IL-23. Here, we directly tested the role of IL-23 in LVS resistance, and found IL-23 to be largely dispensable for immunity to LVS following intradermal or intranasal infection. IL-23p19 KO splenocytes were fully competent in controlling intramacrophage LVS replication in an in vitro overlay assay. Further, antibody responses in IL-23p19 KO mice were similar to those of normal wild type mice after LVS infection. IL-23p19 KO mice or normal wild type mice that survived primary LVS infection survived maximal doses of LVS secondary challenge. Thus p40 has a novel role in clearance of LVS infection that is unrelated to either IL-12 or IL-23.


Subject(s)
Bacterial Vaccines , Coinfection/metabolism , Francisella tularensis/physiology , Interleukin-23 Subunit p19/deficiency , Interleukin-23 Subunit p19/genetics , Tularemia/metabolism , Animals , Coinfection/genetics , Coinfection/immunology , Female , Francisella tularensis/immunology , Interleukin-23 Subunit p19/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tularemia/genetics , Tularemia/immunology , Vaccines, Attenuated
16.
Vaccine ; 31(41): 4641-6, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-23906890

ABSTRACT

With tuberculosis continuing to be a major cause of global morbidity and mortality, a new vaccine is urgently needed. Tuberculosis subunit vaccines have been shown to induce robust immune responses in humans and are a potentially safer alternative to BCG for use in HIV-endemic areas. In this study, we investigated the protective efficacy of 16 different novel Mycobacterium tuberculosis antigens using an aerogenic mouse model of pulmonary tuberculosis. These antigens were tested as subunit vaccines formulated in dimethyl dioctadecyl ammonium bromide (DDA) - D(+) with trehalose 6,6 dibenenate (TDB) (DDA/TDB) adjuvant administered alone as monovalent vaccines or in combination. Six of these antigens (Rv1626, Rv1735, Rv1789, Rv2032, Rv2220, and Rv3478) were shown to consistently and significantly reduce bacterial burdens in the lungs of mice relative to nonvaccinated controls. Three of these six (Rv1789, Rv2220, and Rv3478) induced levels of protective immunity that were essentially equivalent to protection induced by the highly immunogenic antigen 85B (>0.5 log10CFU reduction in the lungs relative to naïve mice). Importantly, when these three antigens were combined, protection essentially equivalent to that mediated by BCG was observed. When either Rv1626 or Rv2032 were combined with the highly protective E6-85 fusion protein (antigen 85B fused to ESAT-6), the protection observed was equivalent to BCG-induced protection at one and three months post-aerosol infection and was significantly greater than the protection observed when E6-85 was administered alone at 3 months post-infection. Using multiparameter flow cytometry, monofunctional IFNγ CD4T cells and different multifunctional CD4T cell subsets capable of secreting multiple cytokines (IFNγ, TNFα and/or IL-2) were shown to be induced by the three most protective antigens with splenocyte CD4T cell frequencies significantly greater than observed in naïve controls. The identification of these highly immunogenic TB antigens and antigen combinations should allow for improved immunization strategies against tuberculosis.


Subject(s)
Antigens, Bacterial/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis, Pulmonary/prevention & control , Adjuvants, Immunologic/administration & dosage , Animals , Antigens, Bacterial/administration & dosage , Bacterial Load , CD4-Positive T-Lymphocytes/immunology , Chemistry, Pharmaceutical , Cytokines/biosynthesis , Disease Models, Animal , Female , Flow Cytometry , Lung/immunology , Lung/microbiology , Mice , Mice, Inbred C57BL , Tuberculosis Vaccines/administration & dosage , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
17.
Infect Immun ; 81(2): 585-97, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23230288

ABSTRACT

We employed Francisella tularensis live vaccine strain (LVS) to study mechanisms of protective immunity against intracellular pathogens and, specifically, to understand protective correlates. One potential molecular correlate identified previously was interleukin-6 (IL-6), a cytokine with pleotropic roles in immunity, including influences on T and B cell functions. Given its role as an immune modulator and the correlation with successful anti-LVS vaccination, we examined the role IL-6 plays in the host response to LVS. IL-6-deficient (IL-6 knockout [KO]) mice infected with LVS intradermally or intranasally or anti-IL-6-treated mice, showed greatly reduced 50% lethal doses compared to wild-type (WT) mice. Increased susceptibility was not due to altered splenic immune cell populations during infection or decreased serum antibody production, as IL-6 KO mice had similar compositions of each compared to WT mice. Although LVS-infected IL-6 KO mice produced much less serum amyloid A and haptoglobin (two acute-phase proteins) than WT mice, there were no other obvious pathophysiological differences between LVS-infected WT and IL-6 KO mice. IL-6 KO or WT mice that survived primary LVS infection also survived a high-dose LVS secondary challenge. Using an in vitro overlay assay that measured T cell activation, cytokine production, and abilities of primed splenocytes to control intracellular LVS growth, we found that IL-6 KO total splenocytes or purified T cells were slightly defective in controlling intracellular LVS growth but were equivalent in cytokine production. Taken together, IL-6 is an integral part of a successful immune response to primary LVS infection, but its exact role in precipitating adaptive immunity remains elusive.


Subject(s)
Bacterial Vaccines/immunology , Francisella tularensis/immunology , Interleukin-6/immunology , Tularemia/immunology , Animals , Bacterial Vaccines/metabolism , Bacterial Vaccines/pharmacology , Francisella tularensis/metabolism , Haptoglobins/immunology , Haptoglobins/metabolism , Interleukin-6/metabolism , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Serum Amyloid A Protein/immunology , Serum Amyloid A Protein/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tularemia/metabolism , Tularemia/microbiology , Tularemia/prevention & control , Vaccines, Attenuated/immunology , Vaccines, Attenuated/metabolism , Vaccines, Attenuated/pharmacology
18.
Pharm Res ; 24(10): 1834-43, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17657598

ABSTRACT

PURPOSE: To investigate the use of poly (lactide-co-glycolide) (PLGA) microparticles in respirable sizes as carriers for Antigen 85B (Ag85B), a secreted protein of Mycobacterium tuberculosis, with the ultimate goal of employing them in pulmonary delivery of tuberculosis vaccine. MATERIALS AND METHODS: Recombinant Ag85B was expressed from two Escherichia coli strains and encapsulated by spray-drying in PLGA microspheres with/without adjuvants. These microspheres containing rAg85B were assessed for their ability to deliver antigen to macrophages for subsequent processing and presentation to the specific CD4 T-hybridoma cells DB-1. DB-1 cells recognize the Ag85B(97-112) epitope presented in the context of MHC class II and secrete IL-2 as the cytokine marker. RESULTS: Microspheres suitable for aerosol delivery to the lungs (3.4-4.3 microm median diameter) and targeting alveolar macrophages were manufactured. THP-1 macrophage-like cells exposed with PLGA-rAg85B microspheres induced the DB-1 cells to produce IL-2 at a level that was two orders of magnitude larger than the response elicited by soluble rAg85B. This formulation demonstrated extended epitope presentation. CONCLUSIONS: PLGA microspheres in respirable sizes were effective in delivering rAg85B in an immunologically relevant manner to macrophages. These results are a foundation for further investigation into the potential use of PLGA particles for delivery of vaccines to prevent M. tuberculosis infection.


Subject(s)
Acyltransferases/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , Drug Carriers , Immunity, Cellular , Microspheres , Polyglactin 910/chemistry , Tuberculosis Vaccines/immunology , Acyltransferases/administration & dosage , Acyltransferases/genetics , Administration, Inhalation , Aerosols , Animals , Antigen Presentation , Antigens, Bacterial/administration & dosage , Antigens, Bacterial/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Chemistry, Pharmaceutical , Cloning, Molecular , Delayed-Action Preparations , Drug Compounding , Escherichia coli/genetics , Humans , Hybridomas/immunology , Interleukin-2/metabolism , Kinetics , Lymphocyte Activation , Macrophages/immunology , Particle Size , Recombinant Proteins/immunology , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...