Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 262
Filtrar
1.
Front Immunol ; 12: 701341, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777335

RESUMO

The essential micronutrient Selenium (Se) is co-translationally incorporated as selenocysteine into proteins. Selenoproteins contain one or more selenocysteines and are vital for optimum immunity. Interestingly, many pathogenic bacteria utilize Se for various biological processes suggesting that Se may play a role in bacterial pathogenesis. A previous study had speculated that Francisella tularensis, a facultative intracellular bacterium and the causative agent of tularemia, sequesters Se by upregulating Se-metabolism genes in type II alveolar epithelial cells. Therefore, we investigated the contribution of host vs. pathogen-associated selenoproteins in bacterial disease using F. tularensis as a model organism. We found that F. tularensis was devoid of any Se utilization traits, neither incorporated elemental Se, nor exhibited Se-dependent growth. However, 100% of Se-deficient mice (0.01 ppm Se), which express low levels of selenoproteins, succumbed to F. tularensis-live vaccine strain pulmonary challenge, whereas 50% of mice on Se-supplemented (0.4 ppm Se) and 25% of mice on Se-adequate (0.1 ppm Se) diet succumbed to infection. Median survival time for Se-deficient mice was 8 days post-infection while Se-supplemented and -adequate mice was 11.5 and >14 days post-infection, respectively. Se-deficient macrophages permitted significantly higher intracellular bacterial replication than Se-supplemented macrophages ex vivo, corroborating in vivo observations. Since Francisella replicates in alveolar macrophages during the acute phase of pneumonic infection, we hypothesized that macrophage-specific host selenoproteins may restrict replication and systemic spread of bacteria. F. tularensis infection led to an increased expression of several macrophage selenoproteins, suggesting their key role in limiting bacterial replication. Upon challenge with F. tularensis, mice lacking selenoproteins in macrophages (TrspM) displayed lower survival and increased bacterial burden in the lung and systemic tissues in comparison to WT littermate controls. Furthermore, macrophages from TrspM mice were unable to restrict bacterial replication ex vivo in comparison to macrophages from littermate controls. We herein describe a novel function of host macrophage-specific selenoproteins in restriction of intracellular bacterial replication. These data suggest that host selenoproteins may be considered as novel targets for modulating immune response to control a bacterial infection.


Assuntos
Francisella tularensis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Selenoproteínas/metabolismo , Tularemia/etiologia , Tularemia/metabolismo , Animais , Modelos Animais de Doenças , Suscetibilidade a Doenças , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Camundongos , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/microbiologia , Pneumonia/patologia , Tularemia/mortalidade , Virulência/genética , Fatores de Virulência/genética
2.
Front Immunol ; 12: 716676, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659206

RESUMO

Peptide-based subunit vaccines are coming to the forefront of current vaccine approaches, with safety and cost-effective production among their top advantages. Peptide vaccine formulations consist of multiple synthetic linear epitopes that together trigger desired immune responses that can result in robust immune memory. The advantages of linear compared to conformational epitopes are their simple structure, ease of synthesis, and ability to stimulate immune responses by means that do not require complex 3D conformation. Prediction of linear epitopes through use of computational tools is fast and cost-effective, but typically of low accuracy, necessitating extensive experimentation to verify results. On the other hand, identification of linear epitopes through experimental screening has been an inefficient process that requires thorough characterization of previously identified full-length protein antigens, or laborious techniques involving genetic manipulation of organisms. In this study, we apply a newly developed generalizable screening method that enables efficient identification of B-cell epitopes in the proteomes of pathogenic bacteria. As a test case, we used this method to identify epitopes in the proteome of Francisella tularensis (Ft), a Select Agent with a well-characterized immunoproteome. Our screen identified many peptides that map to known antigens, including verified and predicted outer membrane proteins and extracellular proteins, validating the utility of this approach. We then used the method to identify seroreactive peptides in the less characterized immunoproteome of Select Agent Burkholderia pseudomallei (Bp). This screen revealed known Bp antigens as well as proteins that have not been previously identified as antigens. Although B-cell epitope prediction tools Bepipred 2.0 and iBCE-EL classified many of our seroreactive peptides as epitopes, they did not score them significantly higher than the non-reactive tryptic peptides in our study, nor did they assign higher scores to seroreactive peptides from known Ft or Bp antigens, highlighting the need for experimental data instead of relying on computational epitope predictions alone. The present workflow is easily adaptable to detecting peptide targets relevant to the immune systems of other mammalian species, including humans (depending upon the availability of convalescent sera from patients), and could aid in accelerating the discovery of B-cell epitopes and development of vaccines to counter emerging biological threats.


Assuntos
Mapeamento de Epitopos/métodos , Epitopos de Linfócito B/imunologia , Proteoma , Proteômica , Animais , Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Biologia Computacional/métodos , Francisella tularensis/imunologia , Humanos , Imunização , Camundongos , Peptídeos/imunologia , Proteômica/métodos , Vacinas de Subunidades Antigênicas/imunologia
3.
PLoS Pathog ; 17(8): e1009905, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34449811

RESUMO

B1 cells, a subset of B lymphocytes whose developmental origin, phenotype, and function differ from that of conventional B2 cells, are the main source of "natural" IgM but can also respond to infection by rapidly producing pathogen-specific IgM directed against T-independent antigens. Francisella tularensis (Ft) is a Gram-negative bacterium that causes tularemia. Infection with Ft Live Vaccine Strain activates B1 cells for production of IgM directed against the bacterial LPS in a process incompletely understood. Here we show that immunization with purified Ft LPS elicits production of LPS-specific IgM and IgG3 by B1 cells independently of TLR2 or MyD88. Immunization, but not infection, generated peritoneum-resident memory B1 cells that differentiated into LPS-specific antibody secreting cells (ASC) upon secondary challenge. IL-5 was rapidly induced by immunization with Ft LPS and was required for production of LPS-specific IgM. Antibody-mediated depletion of ILC2 indicated that these cells were the source of IL-5 and were required for IgM production. IL-25, an alarmin that strongly activates ILC2, was rapidly secreted in response to immunization or infection and its administration to mice significantly increased IgM production and B1 cell differentiation to ASC. Conversely, mice lacking IL-17RB, the IL-25 receptor, showed impaired IL-5 induction, IgM production, and B1 ASC differentiation in response to immunization. Administration of IL-5 to Il17rb-/- mice rescued these B1 cells-mediated responses. Il17rb-/- mice were more susceptible to infection with Ft LVS and failed to develop immunity upon secondary challenge suggesting that LPS-specific IgM is one of the protective adaptive immune mechanisms against tularemia. Our results indicated that immunization with Ft LPS triggers production of IL-25 that, through stimulation of IL-5 release by ILC2, promotes B1 cells activation and differentiation into IgM secreting cells. By revealing the existence of an IL-25-ILC2-IL-5 axis our results suggest novel strategies to improve vaccination against T-independent bacterial antigens.


Assuntos
Anticorpos Antibacterianos/imunologia , Subpopulações de Linfócitos B/imunologia , Francisella tularensis/imunologia , Imunoglobulina M/imunologia , Interleucina-5/metabolismo , Interleucinas/metabolismo , Lipopolissacarídeos/farmacologia , Animais , Anticorpos Antibacterianos/metabolismo , Subpopulações de Linfócitos B/metabolismo , Imunidade Inata , Imunoglobulina M/metabolismo , Interleucina-5/genética , Interleucinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/fisiologia , Receptores de Interleucina-17/fisiologia , Receptor 2 Toll-Like/fisiologia , Tularemia/imunologia , Tularemia/microbiologia , Tularemia/patologia
4.
Microbiologyopen ; 10(2): e1170, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33970545

RESUMO

Tularemia is a zoonotic disease of global proportions. Francisella tularensis subspecies tularensis (type A) and holarctica (type B) cause disease in healthy humans, with type A infections resulting in higher mortality. Repeated passage of a type B strain in the mid-20th century generated the Live Vaccine Strain (LVS). LVS remains unlicensed, does not protect against high inhalational doses of type A, and its exact mechanisms of attenuation are poorly understood. Recent data suggest that live attenuated vaccines derived from type B may cross-protect against type A. However, there is a dearth of knowledge regarding virulent type B pathogenesis and its capacity to stimulate the host's innate immune response. We therefore sought to increase our understanding of virulent type B in vitro characteristics using strain OR96-0246 as a model. Adding to our knowledge of innate immune kinetics in macrophages following infection with virulent type B, we observed robust replication of strain OR96-0246 in murine and human macrophages, reduced expression of pro-inflammatory cytokine genes from "wild type" type B-infected macrophages compared to LVS, and delayed macrophage cell death suggesting that virulent type B may suppress macrophage activation. One disruption in LVS is in the gene encoding the chloride transporter ClcA. We investigated the role of ClcA in macrophage infection and observed a replication delay in a clcA mutant. Here, we propose its role in acid tolerance. A greater understanding of LVS attenuation may reveal new mechanisms of pathogenesis and inform strategies toward the development of an improved vaccine against tularemia.


Assuntos
Proteínas de Bactérias/imunologia , Canais de Cloreto/imunologia , Francisella tularensis/imunologia , Imunidade Inata , Tularemia/imunologia , Tularemia/microbiologia , Animais , Proteínas de Bactérias/genética , Canais de Cloreto/genética , Modelos Animais de Doenças , Francisella tularensis/classificação , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Humanos , Cinética , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL
5.
Infect Immun ; 89(7): e0013421, 2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-33875472

RESUMO

Francisella 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 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 the 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 to 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 to vaccine-induced immune responses in a mouse model of respiratory tularemia. We developed a model to vaccinate Aim2- and Nlrp3-deficient (Aim2-/- and Nlrp3-/-) mice using the emrA1 mutant of the 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 those of 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 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.


Assuntos
Vacinas Bacterianas/imunologia , Proteínas de Ligação a DNA/genética , Francisella tularensis/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Tularemia/genética , Tularemia/imunologia , Animais , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Humoral , Imunização , Camundongos , Camundongos Knockout , Mutação , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Atenuadas , Virulência
6.
PLoS One ; 15(8): e0237034, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32745117

RESUMO

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.


Assuntos
Interferon gama/metabolismo , Fator 88 de Diferenciação Mieloide/genética , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Francisella tularensis/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/metabolismo , Baço/metabolismo , Linfócitos T/imunologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/metabolismo , Receptores Toll-Like/imunologia , Tularemia/microbiologia
7.
Sci Rep ; 10(1): 12023, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32694562

RESUMO

Francisella tularensis is a highly infectious intracellular bacterium that causes tularemia by invading and replicating in mammalian myeloid cells. Francisella primarily invades host macrophages, where it escapes phagosomes within a few hours and replicates in the cytoplasm. Less is known about how Francisella traffics within macrophages or exits into the extracellular environment for further infection. Immune T lymphocytes control the replication of Francisella within macrophages in vitro by a variety of mechanisms, but nothing is known about intracellular bacterial trafficking in the face of such immune pressure. Here we used a murine model of infection with a Francisella attenuated live vaccine strain (LVS), which is under study as a human vaccine, to evaluate the hypothesis that immune T cells control intramacrophage bacterial growth by re-directing bacteria into toxic intracellular compartments of infected macrophages. We visualized the interactions of lymphocytes and LVS-infected macrophages using confocal microscopy and characterized LVS intramacrophage trafficking when co-cultured with immune lymphocytes. We focused on the late stages of infection after bacteria escape from phagosomes, through bacterial replication and the death of macrophages. We found that the majority of LVS remained cytosolic in the absence of immune pressure, eventually resulting in macrophage death. In contrast, co-culture of LVS-infected macrophages with LVS-immune lymphocytes halted LVS replication and inhibited the spread of LVS infection between macrophages, but bacteria did not return to vacuoles such as lysosomes or autophagosomes and macrophages did not die. Therefore, immune lymphocytes directly limit intracellular bacterial replication within the cytoplasm of infected macrophages.


Assuntos
Vacinas Bacterianas/imunologia , Citoplasma/microbiologia , Francisella tularensis/imunologia , Macrófagos/microbiologia , Linfócitos T/imunologia , Tularemia/imunologia , Replicação Viral/imunologia , Animais , Citoplasma/imunologia , Modelos Animais de Doenças , Imunização , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagossomos/microbiologia , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Atenuadas
8.
FEBS Lett ; 594(17): 2782-2799, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32484234

RESUMO

Intracellular pathogens affect diverse host cellular defence and metabolic pathways. Here, we used infection with Francisella tularensis to identify SON DNA-binding protein as a central determinant of macrophage activities. RNAi knockdown of SON increases survival of human macrophages following F. tularensis infection or inflammasome stimulation. SON is required for macrophage autophagy, interferon response factor 3 expression, type I interferon response and inflammasome-associated readouts. SON knockdown has gene- and stimulus-specific effects on inflammatory gene expression. SON is required for accurate splicing and expression of GBF1, a key mediator of cis-Golgi structure and function. Chemical GBF1 inhibition has similar effects to SON knockdown, suggesting that SON controls macrophage functions at least in part by controlling Golgi-associated processes.


Assuntos
Autofagia/genética , Proteínas de Ligação a DNA/genética , Francisella tularensis/patogenicidade , Complexo de Golgi/imunologia , Fatores de Troca do Nucleotídeo Guanina/genética , Interações Hospedeiro-Patógeno/genética , Macrófagos/imunologia , Antígenos de Histocompatibilidade Menor/genética , Autofagia/efeitos dos fármacos , Morte Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/imunologia , Francisella tularensis/genética , Francisella tularensis/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/microbiologia , Fatores de Troca do Nucleotídeo Guanina/antagonistas & inibidores , Fatores de Troca do Nucleotídeo Guanina/imunologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Antígenos de Histocompatibilidade Menor/imunologia , Piridinas/farmacologia , Quinolinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Células THP-1 , Acetato de Tetradecanoilforbol/farmacologia
9.
Virulence ; 11(1): 283-294, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32241221

RESUMO

There is a need for development of an effective vaccine against Francisella tularensis, as this potential bioweapon has a high mortality rate and low infectious dose when delivered via the aerosol route. Moreover, this Tier 1 agent has a history of weaponization. We engineered targeted mutations in the Type A strain F. tularensis subspecies tularensis Schu S4 in aro genes encoding critical enzymes in aromatic amino acid biosynthesis. F. tularensis Schu S4ΔaroC, Schu S4ΔaroD, and Schu S4ΔaroCΔaroD mutant strains were attenuated for intracellular growth in vitro and for virulence in vivo and, conferred protection against pulmonary wild-type (WT) F. tularensis Schu S4 challenge in the C57BL/6 mouse model. F. tularensis Schu S4ΔaroD was identified as the most promising vaccine candidate, demonstrating protection against high-dose intranasal challenge; it protected against 1,000 CFU Schu S4, the highest level of protection tested to date. It also provided complete protection against challenge with 92 CFU of a F. tularensis subspecies holarctica strain (Type B). Mice responded to vaccination with Schu S4ΔaroD with systemic IgM and IgG2c, as well as the production of a functional T cell response as measured in the splenocyte-macrophage co-culture assay. This vaccine was further characterized for dissemination, histopathology, and cytokine/chemokine gene induction at defined time points following intranasal vaccination which confirmed its attenuation compared to WT Schu S4. Cytokine, chemokine, and antibody induction patterns compared to wild-type Schu S4 distinguish protective vs. pathogenic responses to F. tularensis and elucidate correlates of protection associated with vaccination against this agent.


Assuntos
Anticorpos Antibacterianos/sangue , Vacinas Bacterianas/imunologia , Citocinas/imunologia , Francisella tularensis/genética , Francisella tularensis/imunologia , Macrófagos/imunologia , Animais , Vacinas Bacterianas/administração & dosagem , Modelos Animais de Doenças , Feminino , Deleção de Genes , Macrófagos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Vacinas Atenuadas/imunologia , Virulência
10.
J Immunol Methods ; 477: 112693, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31689421

RESUMO

Methods used to prepare bone marrow-derived macrophages (BMDMs) may influence the outcomes of immunological assays in which they are used. Supernatant conditioned by growth of L929 cells has often been used to generate mouse macrophages from bone marrow in vitro but is subject to lot-to-lot variability. To reduce experimental variability and to standardize techniques across laboratories, we investigated recombinant M-CSF (rM-CSF) as an alternative supplement for BMDM maturation in the context of macrophage infection, using the intracellular bacterium Live Vaccine Strain (LVS) of Francisella tularensis as a prototype. We compared rM-CSF with L929 supernatant in terms of their effects on mouse and rat macrophage growth, maturation patterns, surface marker expression, and the expression of selected genes. Further, we compared macrophage infectivity and bacterial replication using LVS. Finally, we compared the in vitro function of BMDMs co-cultured with splenocytes from vaccinated animals in terms of their control of intramacrophage bacterial replication, as well as production of cytokines and nitric oxide. We demonstrated that rM-CSF produced BMDMs with similar, or minimal, phenotypic and gene expression outcomes compared to those generated with media containing L929 supernatant. Most importantly, functional outcomes were similar. Taken together, our data support the use of the rM-CSF in cell culture media as an alternative to L929-supplemented media for functional bioassays that use C57BL/6J mouse or Fischer 344 rat BMDMs to study intracellular infections. This comparison therefore facilitates future protocol standardization.


Assuntos
Técnicas de Cultura de Células/métodos , Meios de Cultura/farmacologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Animais , Infecções Bacterianas/imunologia , Vacinas Bacterianas/imunologia , Bioensaio/métodos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Técnicas de Cocultura/métodos , Feminino , Fibroblastos , Francisella tularensis/imunologia , Regulação da Expressão Gênica/imunologia , Imunoensaio/métodos , Linfócitos , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos F344 , Proteínas Recombinantes/metabolismo , Vacinas Atenuadas/imunologia
11.
PLoS One ; 14(12): e0226778, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31877174

RESUMO

Francisella tularensis, a category-A bioterrorism agent causes tularemia. F. tularensis suppresses the immune response of host cells and intracellularly proliferates. However, the detailed mechanisms of immune suppression and intracellular growth are largely unknown. Here we developed a transposon mutant library to identify novel pathogenic factors of F. tularensis. Among 750 transposon mutants of F. tularensis subsp. novicida (F. novicida), 11 were isolated as less cytotoxic strains, and the genes responsible for cytotoxicity were identified. Among them, the function of slt, which encodes soluble lytic transglycosylase (SLT) was investigated in detail. An slt deletion mutant (Δslt) was less toxic to the human monocyte cell line THP-1 vs the wild-type strain. Although the wild-type strain proliferated in THP-1 cells, the number of intracellular Δslt mutant decreased in comparison. The Δslt mutant escaped from phagosomes during the early stages of infection, but the mutant was detected within the autophagosome, followed by degradation in lysosomes. Moreover, the Δslt mutant induced host cells to produce high levels of cytokines such as tumor necrosis factor-α, interleukin (IL)-6, and IL-1ß, compared with the wild-type strain. These results suggest that the SLT of F. novicida is required for immune suppression and escape from autophagy to allow its survival in host cells.


Assuntos
Proteínas de Bactérias/imunologia , Francisella tularensis/imunologia , Glicosiltransferases/imunologia , Tularemia/imunologia , Animais , Linhagem Celular , Francisella tularensis/crescimento & desenvolvimento , Humanos , Evasão da Resposta Imune , Lisossomos/imunologia , Lisossomos/microbiologia , Camundongos , Monócitos/imunologia , Monócitos/microbiologia , Fagossomos/imunologia , Fagossomos/microbiologia , Tularemia/microbiologia
12.
Sci Rep ; 9(1): 11418, 2019 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-31388083

RESUMO

Francisella tularensis (Ft), the causative agent of lethal tularemia, is classified as a category A biological warfare threat agent. While Ft infection is treatable by antibiotics, many failed antibiotic treatments were reported, highlighting the need for effective new treatments. It has been demonstrated that binding of antibody-coated bacteria to the Fc receptor located on phagocytic cells is a key process needed for efficient protection against Ft. Yet, Ft utilizes the same receptor to enter the phagocytic cells in order to escape the immune system. To address the question whether an anti-Ft LPS antibody lacking the ability to bind the Fc receptor may inhibit the entry of Ft into host cells, a soluble scFv (TL1-scFv) was constructed from an anti Ft-LPS antibody (TL1) that was isolated from an immune single-chain (scFv) phage-display library. Bacterial uptake was assessed upon infection of macrophages with Ft live attenuated strain (LVS) in the presence of either TL1 or TL1-scFv. While incubation of LVS in the presence of TL1 greatly enhanced bacterial uptake, LVS uptake was significantly inhibited in the presence of TL1-scFv. These results prompt further experiments probing the therapeutic efficacy of TL1-scFv, alone or in combination with antibiotic treatment.


Assuntos
Anticorpos Antibacterianos/farmacologia , Francisella tularensis/imunologia , Lipopolissacarídeos/imunologia , Fagocitose/efeitos dos fármacos , Anticorpos de Cadeia Única/farmacologia , Tularemia/tratamento farmacológico , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/isolamento & purificação , Anticorpos Antibacterianos/uso terapêutico , Vacinas Bacterianas/administração & dosagem , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Fagocitose/imunologia , Coelhos , Anticorpos de Cadeia Única/sangue , Anticorpos de Cadeia Única/isolamento & purificação , Anticorpos de Cadeia Única/uso terapêutico , Tularemia/sangue , Tularemia/imunologia , Tularemia/microbiologia , Vacinas Atenuadas/administração & dosagem
13.
Artigo em Inglês | MEDLINE | ID: mdl-31334134

RESUMO

Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.


Assuntos
Francisella tularensis/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Moléculas com Motivos Associados a Patógenos/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Tularemia/imunologia , Alarminas/genética , Alarminas/imunologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Francisella tularensis/genética , Francisella tularensis/patogenicidade , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno/genética , Humanos , Macrófagos/imunologia , Macrófagos/microbiologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Fagocitose/genética , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/imunologia , Receptores de Reconhecimento de Padrão/genética , Transdução de Sinais , Tularemia/genética , Tularemia/microbiologia
14.
Nat Commun ; 10(1): 2121, 2019 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-31073183

RESUMO

Pulmonary immune control is crucial for protection against pathogens. Here we identify a pathway that promotes host responses during pulmonary bacterial infection; the expression of CD200 receptor (CD200R), which is known to dampen pulmonary immune responses, promotes effective clearance of the lethal intracellular bacterium Francisella tularensis. We show that depletion of CD200R in mice increases in vitro and in vivo infectious burden. In vivo, CD200R deficiency leads to enhanced bacterial burden in neutrophils, suggesting CD200R normally limits the neutrophil niche for infection. Indeed, depletion of this neutrophil niche in CD200R-/- mice restores F. tularensis infection to levels seen in wild-type mice. Mechanistically, CD200R-deficient neutrophils display significantly reduced reactive oxygen species production (ROS), suggesting that CD200R-mediated ROS production in neutrophils is necessary for limiting F. tularensis colonisation and proliferation. Overall, our data show that CD200R promotes the antimicrobial properties of neutrophils and may represent a novel antibacterial therapeutic target.


Assuntos
Francisella tularensis/patogenicidade , Interações Hospedeiro-Patógeno/imunologia , Glicoproteínas de Membrana/imunologia , Neutrófilos/imunologia , Tularemia/imunologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Macrófagos/imunologia , Macrófagos/microbiologia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Cultura Primária de Células , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Tularemia/microbiologia
15.
Ann Dermatol Venereol ; 146(2): 131-134, 2019 Feb.
Artigo em Francês | MEDLINE | ID: mdl-30638683

RESUMO

BACKGROUND: Tularaemia is a zoonotic disease caused by inoculation with the Gram-negative coccobacillus Francisella tularensis. It was first described in the United States in 1911 and is a rare disease with an annual reported incidence in France between 2002 and 2012 of 0.07 cases per 100,000 habitants. Reporting of the disease in humans has been mandatory in France since 2003. PATIENTS AND METHODS: Herein we report a case of tularaemia following a tick bite in a patient in the north of France. DISCUSSION: Tularaemia is a rare form of zoonosis that should be sought in the event of unexplained adenitis. Clinical presentations vary, and in certain cases only dermatological signs are manifest. Diagnosis is confirmed by bacterial serology. Rapid initiation of suitable antibiotics produces a favourable and benign outcome in most cases. However, the offending organism, which is potentially lethal, is classed as a potential bioterrorism agent.


Assuntos
Doenças Raras/microbiologia , Picadas de Carrapatos/complicações , Tularemia/transmissão , França , Francisella tularensis/imunologia , Virilha , Humanos , Linfadenite/microbiologia , Masculino , Doenças Raras/tratamento farmacológico , Tularemia/tratamento farmacológico
16.
PLoS One ; 13(5): e0198140, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29799870

RESUMO

There are no defined correlates of protection for any intracellular pathogen, including the bacterium Francisella tularensis, which causes tularemia. Evaluating vaccine efficacy against sporadic diseases like tularemia using field trials is problematic, and therefore alternative strategies to test vaccine candidates like the Francisella Live Vaccine Strain (LVS), such as testing in animals and applying correlate measurements, are needed. Recently, we described a promising correlate strategy that predicted the degree of vaccine-induced protection in mice given parenteral challenges, primarily when using an attenuated Francisella strain. Here, we demonstrate that using peripheral blood lymphocytes (PBLs) in this approach predicts LVS-mediated protection against respiratory challenge of Fischer 344 rats with fully virulent F. tularensis, with exceptional sensitivity and specificity. Rats were vaccinated with a panel of LVS-derived vaccines and subsequently given lethal respiratory challenges with Type A F. tularensis. In parallel, PBLs from vaccinated rats were evaluated for their functional ability to control intramacrophage Francisella growth in in vitro co-culture assays. PBLs recovered from co-cultures were also evaluated for relative gene expression using a large panel of genes identified in murine studies. In vitro control of LVS intramacrophage replication reflected the hierarchy of protection. Further, despite variability between individuals, 22 genes were significantly more up-regulated in PBLs from rats vaccinated with LVS compared to those from rats vaccinated with the variant LVS-R or heat-killed LVS, which were poorly protective. These genes included IFN-γ, IL-21, NOS2, LTA, T-bet, IL-12rß2, and CCL5. Most importantly, combining quantifications of intramacrophage growth control with 5-7 gene expression levels using multivariate analyses discriminated protected from non-protected individuals with greater than 95% sensitivity and specificity. The results therefore support translation of this approach to non-human primates and people to evaluate new vaccines against Francisella and other intracellular pathogens.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Francisella tularensis/patogenicidade , Imunização , Sistema Respiratório/microbiologia , Animais , Feminino , Regulação da Expressão Gênica/imunologia , Imunidade Humoral/imunologia , Macrófagos/microbiologia , Análise Multivariada , Ratos , Linfócitos T/imunologia , Virulência
17.
Front Immunol ; 9: 561, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29632532

RESUMO

Francisella tularensis is a remarkably infectious facultative intracellular bacterium of macrophages that causes tularemia. Early evasion of host immune responses contributes to the success of F. tularensis as a pathogen. F. tularensis entry into human monocytes and macrophages is mediated by the major phagocytic receptor, complement receptor 3 (CR3, CD11b/CD18). We recently determined that despite a significant increase in macrophage uptake following C3 opsonization of the virulent Type A F. tularensis spp. tularensis Schu S4, this phagocytic pathway results in limited pro-inflammatory cytokine production. Notably, MAP kinase/ERK activation is suppressed immediately during C3-opsonized Schu S4-CR3 phagocytosis. A mathematical model of CR3-TLR2 crosstalk predicted early involvement of Ras GTPase-activating protein (RasGAP) in immune suppression by CR3. Here, we link CR3-mediated uptake of opsonized Schu S4 by human monocytes and macrophages with inhibition of early signal 1 inflammasome activation, evidenced by limited caspase-1 cleavage and IL-18 release. This inhibition is due to increased RasGAP activity, leading to a reduction in the Ras-ERK signaling cascade upstream of the early inflammasome activation event. Thus, our data uncover a novel signaling pathway mediated by CR3 following engagement of opsonized virulent F. tularensis to limit inflammasome activation in human phagocytic cells, thereby contributing to evasion of the host innate immune system.


Assuntos
Francisella tularensis/imunologia , Inflamassomos/imunologia , Antígeno de Macrófago 1/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Proteínas Ativadoras de ras GTPase/imunologia , Caspase 1/imunologia , Caspase 1/metabolismo , Células Cultivadas , Francisella tularensis/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Inflamassomos/metabolismo , Interleucina-18/imunologia , Interleucina-18/metabolismo , Antígeno de Macrófago 1/metabolismo , Macrófagos/microbiologia , Monócitos/imunologia , Monócitos/microbiologia , Transdução de Sinais/imunologia , Receptor 2 Toll-Like/imunologia , Receptor 2 Toll-Like/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
18.
PLoS One ; 13(4): e0194614, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29684046

RESUMO

Tularemia is a fatal human disease caused by Francisella tularensis, a Gram-negative encapsulated coccobacillus bacterium. Due to its low infectious dose, ease of aerosolized transmission, and lethal effects, the CDC lists F. tularensis as a Category A pathogen, the highest level for a potential biothreat agent. Previous vaccine studies have been conducted with live attenuated, inactivated, and subunit vaccines, which have achieved partial or full protection from F. tularensis live vaccine strain (LVS) challenge, but no vaccine has been approved for human use. We demonstrate the improved efficacy of a multi-antigen subunit vaccine by using Tobacco Mosaic virus (TMV) as an antigen carrier for the F. tularensis SchuS4 proteins DnaK, OmpA, SucB and Tul4 (DOST). The magnitude and quality of immune responses were compared after mice were immunized by subcutaneous or intranasal routes of administration with a TMV-DOST mixture, with or without four different adjuvants. Immune responses varied in magnitude and isotype profile, by antigen, by route of administration, and by protection in an F. tularensis LVS challenge model of disease. Interestingly, our analysis demonstrates an overwhelming IgG2 response to SucB after intranasal dosing, as well as a robust cellular response, which may account for the improved two-dose survival imparted by the tetravalent vaccine, compared to a previous study that tested efficacy of TMV-DOT. Our study provides evidence that potent humoral, cellular and mucosal immunity can be achieved by optimal antigen combination, delivery, adjuvant and appropriate route of administration, to improve vaccine potency and provide protection from pathogen challenge.


Assuntos
Antígenos de Bactérias/imunologia , Vacinas Bacterianas/imunologia , Vírus do Mosaico do Tabaco/genética , Tularemia/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos , Administração Intranasal , Animais , Anticorpos Antibacterianos/análise , Anticorpos Antibacterianos/metabolismo , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Modelos Animais de Doenças , Feminino , Francisella tularensis/imunologia , Imunidade Celular , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Isotipos de Imunoglobulinas/imunologia , Isotipos de Imunoglobulinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Taxa de Sobrevida , Vírus do Mosaico do Tabaco/metabolismo , Tularemia/microbiologia , Tularemia/prevenção & controle , Vacinas Conjugadas/imunologia
19.
mBio ; 8(5)2017 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-29018123

RESUMO

Francisella tularensis is the causative agent of tularemia and a potential bioterrorism agent. In the present study, we isolated, identified, and quantified the proteins present in the membranes of the virulent type A strain, Schu S4, and the attenuated type B strain, LVS (live vaccine strain). Spectral counting of mass spectrometric data showed enrichment for membrane proteins in both strains. Mice vaccinated with whole LVS membranes encapsulated in poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing the adjuvant polyinosinic-polycytidylic acid [poly(I·C)] showed significant protection against a challenge with LVS compared to the results seen with naive mice or mice vaccinated with either membranes or poly(I·C) alone. The PLGA-encapsulated Schu S4 membranes with poly(I·C) alone did not significantly protect mice from a lethal intraperitoneal challenge with Schu S4; however, this vaccination strategy provided protection from LVS challenge. Mice that received the encapsulated Schu S4 membranes followed by a booster of LVS bacteria showed significant protection with respect to a lethal Schu S4 challenge compared to control mice. Western blot analyses of the sera from the Schu S4-vaccinated mice that received an LVS booster showed four immunoreactive bands. One of these bands from the corresponding one-dimensional (1D) SDS-PAGE experiment represented capsule. The remaining bands were excised, digested with trypsin, and analyzed using mass spectrometry. The most abundant proteins present in these immunoreactive samples were an outer membrane OmpA-like protein, FopA; the type IV pilus fiber building block protein; a hypothetical membrane protein; and lipoproteins LpnA and Lpp3. These proteins should serve as potential targets for future recombinant protein vaccination studies.IMPORTANCE The low infectious dose, the high potential mortality/morbidity rates, and the ability to be disseminated as an aerosol make Francisella tularensis a potential agent for bioterrorism. These characteristics led the Centers for Disease Control (CDC) to classify F. tularensis as a Tier 1 pathogen. Currently, there is no vaccine approved for general use in the United States.


Assuntos
Vacinas Bacterianas/imunologia , Francisella tularensis/imunologia , Proteínas de Membrana/imunologia , Tularemia/prevenção & controle , Vacinas de Subunidades Antigênicas/imunologia , Adjuvantes Imunológicos , Animais , Modelos Animais de Doenças , Francisella tularensis/química , Francisella tularensis/patogenicidade , Ácido Láctico , Macrófagos/imunologia , Macrófagos/microbiologia , Espectrometria de Massas , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas , Poli I-C/imunologia , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Proteômica , Tularemia/imunologia , Vacinação , Vacinas Atenuadas/imunologia , Vacinas de Subunidades Antigênicas/genética
20.
Infect Immun ; 85(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28739830

RESUMO

Tularemia is caused by the Gram-negative bacterial pathogen Francisella tularensis Infection of macrophages and their subsequent death are believed to play important roles in the progression of disease. Because complement is a particularly effective opsonin for Francisella, we asked whether complement-dependent uptake of F. tularensis strain SCHU S4 affects the survival of primary human macrophages during infection. Complement component C3 was found to be an essential opsonin in human serum not only for greatly increased uptake of SCHU S4 but also for the induction of macrophage death. Single-cell analysis also revealed that macrophage death did not require a high intracellular bacterial burden. In the presence of C3, macrophage death was observed at 24 h postinfection in a quarter of the macrophages that contained only 1 to 5 bacterial cells. Macrophages infected in the absence of C3 rarely underwent cell death, even when they contained large numbers of bacteria. The need for C3, but not extensive replication of the pathogen, was confirmed by infections with SCHU S4 ΔpurMCD, a mutant capable of phagosome escape but of only limited cytosolic replication. C3-dependent Francisella uptake alone was insufficient to induce macrophage death, as evidenced by the failure of the phagosome escape-deficient mutant SCHU S4 ΔfevR to induce cell death despite opsonization with C3. Together, these findings indicate that recognition of C3-opsonized F. tularensis, but not extensive cytosolic replication, plays an important role in regulating macrophage viability during intracellular infections with type A F. tularensis.


Assuntos
Complemento C3/imunologia , Francisella tularensis/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Morte Celular , Sobrevivência Celular/imunologia , Francisella tularensis/genética , Francisella tularensis/crescimento & desenvolvimento , Francisella tularensis/patogenicidade , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Fagocitose , Fagossomos/imunologia , Fagossomos/microbiologia , Análise de Célula Única
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA