Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Fish Shellfish Immunol ; 127: 405-411, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35772679

ABSTRACT

Francisella orientalis infections, known as francisellosis, are one of the most important diseases affecting the production of Nile tilapia, causing high mortality rates in the most susceptible fish stages: fingerlings and juveniles. Antibiotic therapy is the method of choice for treating the disease, as there are no commercially available vaccines. In this study, we developed an inactivated whole-cell vaccine using an isolate of F. orientalis in combination with the aqueous adjuvant Montanide IMS 1312 VG, which was administered to Nile tilapia through immersion. Two immunization trials (1 and 2) were conducted with fish at the fingerling and juvenile stages. For each trial, five different experimental groups were established: a complete vaccine (bacterin in combination with aqueous adjuvant), bacterin, aqueous adjuvant, and positive and negative controls. Thirty days after vaccination, an experimental challenge was performed through intraperitoneal injection of the same F. orientalis isolate. As a result, the vaccinated fingerlings were the only group in which mortality and progression of clinical signs of francisellosis were statistically significantly reduced, although relative percentage of survival (RPS) was low at 50%. In the juvenile group, RPS was higher at 63%, but not statistically significant. Nevertheless, an RPS of only 50% is acceptable for using vaccines in the field. The bacterin and adjuvant treatments alone were not effective, showing an RPS of 37% and 0%, respectively. Post-vaccination mortality was observed in the group exposed only to the adjuvant, which may indicate excessive immune stimulation at this stage. Interestingly, the immune response elicited by the vaccine was unable to eliminate the pathogen from the host; therefore, the surviving animals became carriers. Although the immune response elicited by the vaccine was unable to eliminate the pathogen from the host, this vaccine formulation could be a viable alternative for use in the field and serve as another means of controlling the mortality caused by the pathogen. Our study provides the first report of vaccination, using immersion, against francisellosis at the most susceptible stages of farmed Nile tilapia. Future studies should address the efficiency of immersion vaccines under field conditions.


Subject(s)
Bacterial Vaccines , Cichlids , Fish Diseases/prevention & control , Francisella/immunology , Gram-Negative Bacterial Infections/veterinary , Animals , Bacterial Vaccines/administration & dosage , Francisella/pathogenicity , Gram-Negative Bacterial Infections/prevention & control , Immersion , Mineral Oil , Vaccination/methods , Vaccination/veterinary
2.
Fish Shellfish Immunol ; 113: 86-88, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33826937

ABSTRACT

Although Nile tilapia (Oreochromis niloticus) is a well-established aquaculture species globally, there are a limited number of commercial vaccines available or are used for this species. The majority of diseases affecting farmed tilapia are bacterial, with antibiotics frequently used to treat fish. The current study was performed to optimise the use of mucosal vaccines for tilapia by adapting an existing bacterin vaccine against Francisella noatunensis subsp. orientalis (Fno) as a proof of concept. This vaccine has previously provided excellent protection by injection, however, the preference for tilapia farmers would be to vaccinate fish by immersion or orally, due to the lower cost and ease of application. These vaccination routes, however, are often less efficacious probably due to the lack of adjuvants in immersion and oral vaccines. The aims of this study, therefore, were to optimise the formulation and dose of the Fno vaccine with mucosal adjuvants for oral and immersion delivery. Tilapia fry (av. 6 g) were given three concentrations (high, medium, low; i.e. 1×109, 1×108 and 1×107 CFU mL-1) of antigen combined with the oral adjuvant by oral gavage, to optimise the dose needed to induce an immune response to Fno, and the immune response obtained compared with fish vaccinated by immersion (with and without an immersion adjuvant). Fry were boosted by the same route at 420 degree days (DD), and samples (serum, mucus ) taken at 840 DD for specific antibody responses measured by ELISA and western blotting. Specific IgM titres were significantly elevated in serum and mucus of fish given the high dose adjuvanted vaccine by gavage. In addition, by western blotting with serum, a significant immunogenic reaction was evident between 20 and 37 kDa in the fish given the high dose oral vaccine by gavage. As protection against Fno provided by the injection vaccine was correlated with specific antibody responses these findings suggest the oral vaccine also has potential to provide protection. Further studies are needed to optimise delivery of the vaccine via feed.


Subject(s)
Antibodies, Bacterial/metabolism , Bacterial Vaccines/administration & dosage , Cichlids , Fish Diseases/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/veterinary , Vaccination/veterinary , Administration, Oral , Animals , Antibodies, Bacterial/blood , Gram-Negative Bacterial Infections/immunology
3.
Fish Shellfish Immunol ; 105: 341-349, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32712230

ABSTRACT

Warm-water piscine francisellosis is a granulomatous bacterial disease caused by Francisella orientalis (Fo). The disease has been detected in a wide range of fish species globally, causing mortalities as high as 90% and significant economic losses. Currently there are no commercially available vaccines and few treatment options exist. In the current study, two novel recombinant vaccines were prepared using diatom-expressed IglC or bacterial-expressed GroEL proteins. The vaccine antigens were emulsified with either nanoparticles or a commercially available oil-based adjuvant. Nile tilapia, Oreochromis niloticus, fingerlings were immunized intracoelomically with the recombinant IglC or GroEL vaccines, diatoms alone or phosphate buffer saline. Approximately 840-degree days post-vaccination, fish were challenged via immersion with 106 CFU/mL of wild-type Fo. Twenty-one days post challenge (dpc), the highest relative percent survival was recorded in the IglC-Montanide group (75%), compared to 53%, 50%, 22%, 19% and 16% in the IglC-nanoparticles, GroEL-Montanide, GroEL-nanoparticles, diatoms-Montanide and diatoms-nanoparticles groups, respectively. Protection correlated with significantly higher specific antibody responses in the IglC-Montanide group. Moreover, a significantly lower bacterial load was detected in spleen samples from the IglC-Montanide survivor tilapia compared to the other experimental groups. This is the first report of recombinant vaccines against piscine francisellosis in tilapia. The Fo vaccines described in our study may facilitate development of a safe, cost-effective and highly protective vaccine against francisellosis in farmed tilapia.


Subject(s)
Bacterial Vaccines/immunology , Cichlids/immunology , Fish Diseases/prevention & control , Francisella/immunology , Animals , Bacterial Proteins/immunology , Chaperonin 60/immunology , Fish Diseases/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Gram-Negative Bacterial Infections/veterinary , Vaccines, Synthetic/immunology
4.
J Fish Dis ; 42(8): 1191-1200, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31184398

ABSTRACT

Francisella noatunensis subsp. orientalis is a pathogen of tilapia and other warm-water fish for which no vaccines are commercially available. In this study, a whole cell formalin-inactivated vaccine was developed for the first time using the highly virulent isolate STIR-GUS-F2f7 and the oil-based adjuvant Montanide™ ISA 763A VG. The efficacy of the vaccine was assessed in red Nile tilapia via intraperitoneal (i.p.) injection using homologous experimental infection and correlates of protection such as seral antibody production and bacterial loads in the spleen. For immunization, fish were i.p. injected with 0.1 ml of the vaccine, the adjuvant alone or PBS. At 840 degree days post-vaccination, all fish were i.p. injected with 4.0 × 103 CFU/fish of pathogenic bacteria. The RPS at the end of the trial was 100% in the vaccinated group with significantly higher survival than in the adjuvant and control groups. The RPS in the adjuvant group was 42%, and no significant difference was seen in survival between this and the PBS group. Moreover, significantly higher antibody titres in the serum and significantly lower bacterial loads in the spleen were detected in the vaccinated fish by ELISA and qPCR, respectively. These findings highlight the potential of autogenous vaccines for controlling francisellosis in tilapia.


Subject(s)
Autovaccines/administration & dosage , Cichlids , Francisella/immunology , Gram-Negative Bacterial Infections/prevention & control , Vaccination/veterinary , Animals , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Injections, Intraperitoneal/veterinary , Vaccines, Inactivated/administration & dosage
5.
Article in English | MEDLINE | ID: mdl-31054474

ABSTRACT

The genetic repertoire underlying teleost immunity has been shown to be highly variable. A rare example is Atlantic cod and its relatives Gadiformes that lacks a hallmark of vertebrate immunity: Major Histocompatibility Complex class II. No immunological studies so far have fully unraveled the functionality of this particular immune system. Through global transcriptomic profiling, we investigate the immune response and host-pathogen interaction of Atlantic cod infected with the facultative intracellular bacterium Francisella noatunensis. We find that Atlantic cod displays an overall classic innate immune response with inflammation, acute-phase proteins and cell recruitment through up-regulation of e.g. IL1B, fibrinogen, cathelicidin, hepcidin and several chemotactic cytokines such as the neutrophil attractants CXCL1 and CXCL8. In terms of adaptive immunity, we observe up-regulation of interferon gamma followed by up-regulation of several MHCI transcripts and genes related to antigen transport and loading. Finally, we find up-regulation of immunoglobulins and down-regulation of T-cell and NK-like cell markers. Our analyses also uncover some contradictory transcriptional findings such as up-regulation of anti-inflammatory IL10 as well as down-regulation of the NADPH oxidase complex and myeloperoxidase. This we interpret as the result of host-pathogen interactions where F. noatunensis modulates the immune response. In summary, our results suggest that Atlantic cod mounts a classic innate immune response as well as a neutrophil-driven response. In terms of adaptive immunity, both endogenous and exogenous antigens are being presented on MHCI and antibody production is likely enabled through direct B-cell stimulation with possible neutrophil help. Collectively, we have obtained novel insight in the orchestration of the Atlantic cod immune system and determined likely targets of F. noatunensis host-pathogen interactions.


Subject(s)
Fish Diseases/immunology , Francisella/physiology , Gadus morhua/microbiology , Gram-Negative Bacterial Infections/veterinary , Adaptive Immunity , Animals , Fish Diseases/genetics , Fish Diseases/microbiology , Francisella/immunology , Gadus morhua/genetics , Gadus morhua/immunology , Gene Expression Regulation , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions , Immunity, Innate , Transcriptome
6.
Fish Shellfish Immunol ; 89: 217-227, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30951851

ABSTRACT

Francisellosis, induced by Francisella noatunensis subsp. orientalis (Fno), is an emerging bacterial disease representing a major threat to the global tilapia industry. There are no commercialised vaccines presently available against francisellosis for use in farmed tilapia, and the only available therapeutic practices used in the field are either the prolonged use of antibiotics or increasing water temperature. Recently, an autogenous whole cell-adjuvanted injectable vaccine was developed that gave 100% relative percent survival (RPS) in tilapia challenged with a homologous isolate of Fno. In this study, we evaluated the efficacy of this vaccine against challenge with heterologous Fno isolates. Healthy Nile tilapia, Oreochromis niloticus (∼15 g) were injected intraperitoneally (i.p.) with the vaccine, adjuvant-alone or phosphate buffer saline (PBS) followed by an i.p. challenge with three Fno isolates from geographically distinct locations. The vaccine provided significant protection in all groups of vaccinated tilapia, with a significantly higher RPS of 82.3% obtained against homologous challenge, compared to 69.8% and 65.9% with the heterologous challenges. Protection correlated with significantly higher specific antibody responses, and western blot analysis demonstrated cross-isolate antigenicity with fish sera post-vaccination and post-challenge. Moreover, a significantly lower bacterial burden was detected by qPCR in conjunction with significantly greater expression of IgM, IL-1 ß, TNF-α and MHCII, 72 h post-vaccination (hpv) in spleen samples from vaccinated tilapia compared to fish injected with adjuvant-alone and PBS. The Fno vaccine described in this study may provide a starting point for development a broad-spectrum highly protective vaccine against francisellosis in tilapia.


Subject(s)
Bacterial Vaccines/administration & dosage , Cichlids/immunology , Fish Diseases/prevention & control , Francisella/immunology , Gram-Negative Bacterial Infections/veterinary , Animals , Fish Diseases/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Injections, Intraperitoneal/veterinary , Real-Time Polymerase Chain Reaction/veterinary
7.
Sci Rep ; 9(1): 6045, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988331

ABSTRACT

This work evaluated the effects of dietary supplementation of A-Live (phytogenic) either individually or in combination with Aquaform (potassium diformate, acidifier) on juvenile Nile tilapia (Oreochromis niloticus) growth performance, innate immune parameters, gut microbiome, and resistance against Francisella noatunensis subsp. orientalis challenge. Each experimental group contained 140 fishes (34.3 ± 0.33) in two 150L tanks. The experimental design consisted of five groups: a negative control; treated groups (G1, G2, G3) supplemented with different concentrations of A-Live and Aquaform in the feed; and a positive control (PC) for pathogen infection. Groups G1, G2, G3, and PC were challenged with Francisella spp. after 15 days. After infection, the mortality was significantly lower in groups G1, G2, and G3 (p < 0.01). Furthermore, these groups showed significant increase (p < 0.05) in daily weight gain, feed conversion rate, and specific growth rate. The PC group presented increase (p < 0.05) in the leukocytes and neutrophils number. Innate immunity parameters showed no difference between treatments after infection. Microbiome analysis revealed an increased number of bacteria belonging to the Vibrionaceae family after pathogen infection suggesting a secondary pathogen function of these bacteria. These results validate the beneficial effects of these products in tilapia farming.


Subject(s)
Animal Feed , Cichlids/immunology , Fish Diseases/prevention & control , Formates/administration & dosage , Plant Extracts/administration & dosage , Animals , Aquaculture/methods , Cichlids/microbiology , Dietary Supplements , Disease Resistance/drug effects , Disease Resistance/immunology , Fish Diseases/microbiology , Francisella/drug effects , Francisella/immunology , Francisella/isolation & purification , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Immunity, Innate/drug effects , Microbial Sensitivity Tests
8.
J Immunol ; 202(8): 2254-2265, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30842273

ABSTRACT

The nonreceptor tyrosine kinase c-Abl plays important roles in T cell development and immune responses; however, the mechanism is poorly understood. IFN regulatory factor 3 (IRF3) is a key transcriptional regulator of type I IFN-dependent immune responses against DNA and RNA viruses. The data in this study show that IRF3 is physically associated with c-Abl in vivo and directly binds to c-Abl in vitro. IRF3 is phosphorylated by c-Abl and c-Abl-related kinase, Arg, mainly at Y292. The inhibitor AMN107 inhibits IFN-ß production induced by poly(dA:dT), poly(I:C), and Sendai virus in THP-1 and mouse bone marrow-derived macrophage cells. IRF3-induced transcription of IFN-ß is significantly reduced by the mutation of Y292 to F. Moreover, AMN107 suppresses gene expression of absent in melanoma 2 (AIM2) and subsequently reduces inflammasome activation induced by cytosolic bacteria, dsDNA, and DNA viruses. Consistent with this finding, Francisella tularensis subsp. holarctica live vaccine strain (Ft LVS), which is known as an activator of AIM2 inflammasome, induces death in significantly more C57BL/6 mice treated with the Abl inhibitor AMN107 or c-Abl/Arg small interfering RNA than in untreated mice. This study provides new insight into the function of c-Abl and Arg in regulating immune responses and AIM2 inflammasome activation, especially against Ft LVS infection.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Interferon-beta/immunology , Proto-Oncogene Proteins c-abl/immunology , Animals , Arginine/immunology , DNA-Binding Proteins/immunology , Francisella/immunology , Gene Expression Regulation/drug effects , Humans , Inflammasomes/immunology , Mice , Phosphorylation/drug effects , Pyrimidines/pharmacology , Sendai virus/immunology , THP-1 Cells
9.
Vaccine ; 35(52): 7264-7272, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29153776

ABSTRACT

BACKGROUND: Francisella noatunensis ssp. noatunensis (F.n.n.) is the causative agent of francisellosis in Atlantic cod and constitutes one of the main challenges for future aquaculture on this species. A facultative intracellular bacterium like F.n.n. exert an immunologic challenge against which live attenuated vaccines in general are most effective. Thus, we constructed a deletion in the F.n.n. clpB gene as ΔclpB mutants are among the most promising vaccine candidates in human pathogenic Francisella. PURPOSE: Characterization of F.n.n. ΔclpB using primary Atlantic cod head kidney leukocytes, the zebrafish embryo and adult zebrafish model with focus on potential attenuation, relevant immune responses and immunogenic potential. MAIN RESULTS: Interleukin 1 beta transcription in Atlantic cod leukocytes was significantly elevated from 24 to 96 h post infection with F.n.n. ΔclpB compared to F.n.n. wild-type (wt). Growth attenuation of the deletion mutant in zebrafish embryos was observed by fluorescence microscopy and confirmed by genome quantification by qPCR. In the immunization experiment, adult zebrafish were immunized with 7 × 106 CFU of F.n.n. ΔclpB before challenge four weeks later with 6 × 108 CFU of F.n.n. wt. One day after challenge, immunized zebrafish responded with significantly lower interleukin 8 levels compared to the non-immunized control. Immunized fish were protected against the acute mortality observed in non-immunized zebrafish after challenge and bacterial genomes quantified by qPCR were reduced to a minimum 28 days post challenge, indicating protective immunity stimulated by F.n.n. ΔclpB. CONCLUSION: Deletion mutation of clpB in F.n.n. causes in vitro and in vivo attenuation and elicits a protective immune response in adult zebrafish against a lethal dose of F.n.n. wt. Taken together, the results presented increases the knowledge on protective immune responses against F.n.n.


Subject(s)
Fish Diseases/prevention & control , Francisella/genetics , Gram-Negative Bacterial Infections/veterinary , Zebrafish/microbiology , Animals , Antibody Formation , Aquaculture , Bacterial Proteins/genetics , Disease Models, Animal , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Diseases/mortality , Francisella/immunology , Gadus morhua/immunology , Gadus morhua/microbiology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/prevention & control , Immunogenicity, Vaccine , Interleukin-1beta/genetics , Interleukin-8/biosynthesis , Interleukin-8/immunology , Sequence Deletion , Vaccination , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology , Zebrafish/immunology
10.
Nat Commun ; 8: 15564, 2017 06 05.
Article in English | MEDLINE | ID: mdl-28580947

ABSTRACT

Pyrin domain-only proteins (POPs) are recently evolved, primate-specific proteins demonstrated in vitro as negative regulators of inflammatory responses. However, their in vivo function is not understood. Of the four known POPs, only POP2 is reported to regulate NF-κB-dependent transcription and multiple inflammasomes. Here we use a transgenic mouse-expressing POP2 controlled by its endogenous human promotor to study the immunological functions of POP2. Despite having significantly reduced inflammatory cytokine responses to LPS and bacterial infection, POP2 transgenic mice are more resistant to bacterial infection than wild-type mice. In a pulmonary tularaemia model, POP2 enhances IFN-γ production, modulates neutrophil numbers, improves macrophage functions, increases bacterial control and diminishes lung pathology. Thus, unlike other POPs thought to diminish innate protection, POP2 reduces detrimental inflammation while preserving and enhancing protective immunity. Our findings suggest that POP2 acts as a high-order regulator balancing cellular function and inflammation with broad implications for inflammation-associated diseases and therapeutic intervention.


Subject(s)
Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/prevention & control , Inflammation/immunology , Tularemia/immunology , Tularemia/prevention & control , Animals , Cell Line , Female , HEK293 Cells , HeLa Cells , Humans , Inflammasomes/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Listeria monocytogenes/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis , Neutrophils/immunology , Promoter Regions, Genetic/genetics , Salmonella typhimurium/immunology , Transcription Factor RelA/biosynthesis , Tularemia/microbiology , U937 Cells
11.
J Leukoc Biol ; 102(3): 815-828, 2017 09.
Article in English | MEDLINE | ID: mdl-28550119

ABSTRACT

Francisella novicida is a Gram-negative bacterium that is closely related to the highly virulent facultative intracellular pathogen, Francisella tularensis Data published by us and others demonstrate that F. tularensis virulence correlates directly with its ability to impair constitutive apoptosis and extend human neutrophil lifespan. In contrast, F. novicida is attenuated in humans, and the mechanisms that account for this are incompletely defined. Our published data demonstrate that F. novicida binds natural IgG that is present in normal human serum, which in turn, elicits NADPH oxidase activation that does not occur in response to F. tularensis As it is established that phagocytosis and oxidant production markedly accelerate neutrophil death, we predicted that F. novicida may influence the neutrophil lifespan in an opsonin-dependent manner. To test this hypothesis, we quantified bacterial uptake, phosphatidylserine (PS) externalization, and changes in nuclear morphology, as well as the kinetics of procaspase-3, -8, and -9 processing and activation. To our surprise, we discovered that F. novicida not only failed to accelerate neutrophil death but also diminished and delayed apoptosis in a dose-dependent, but opsonin-independent, manner. In keeping with this, studies of conditioned media (CM) showed that neutrophil longevity could be uncoupled from phagocytosis and that F. novicida stimulated neutrophil secretion of CXCL8. Taken together, the results of this study reveal shared and unique aspects of the mechanisms used by Francisella species to manipulate neutrophil lifespan and as such, advance understanding of cell death regulation during infection.


Subject(s)
Apoptosis/immunology , Francisella/immunology , Neutrophils/immunology , Phagocytosis/immunology , Adult , Caspase 3/immunology , Caspase 8/immunology , Caspase 9/immunology , Enzyme Activation/immunology , Humans , Interleukin-8/immunology , Neutrophils/microbiology
12.
Clin Vaccine Immunol ; 24(5)2017 May.
Article in English | MEDLINE | ID: mdl-28331079

ABSTRACT

Vaccine development against extracellular bacteria has been important for the sustainability of the aquaculture industry. In contrast, infections with intracellular pathogens remain largely an unresolved problem. Francisella noatunensis subsp. orientalis is a Gram-negative, facultative intracellular bacterium that causes the disease francisellosis in fish. Francisellosis is commonly characterized as a chronic granulomatous disease with high morbidity and can result in high mortality depending on the host. In this study, we explored the potential of bacterial membrane vesicles (MVs) as a vaccine agent against F. noatunensis subsp. orientalis Bacterial MVs are spherical structures naturally released from the membrane of bacteria and are often enriched with selected bacterial components such as toxins and signaling molecules. MVs were isolated from broth-cultured F. noatunensis subsp. orientalis in the present work, and proteomic analysis by mass spectrometry revealed that MVs contained a variety of immunogenic factors, including the intracellular growth proteins IglC and IglB, known to be part of a Francisella pathogenicity island (FPI), as well as outer membrane protein OmpA, chaperonin GroEL, and chaperone ClpB. By using flow cytometry and electron microscopy, we observed that F. noatunensis subsp. orientalis mainly infects myelomonocytic cells, both in vivo and in vitro Immunization with MVs isolated from F. noatunensis subsp. orientalis protects zebrafish from subsequent challenge with a lethal dose of F. noatunensis subsp. orientalis To determine if MVs induce a typical acute inflammatory response, mRNA expression levels were assessed by quantitative real-time PCR. Expression of tnfa, il1b, and ifng, as well as mhcii, mpeg1.1, and ighm, was upregulated, thus confirming the immunogenic properties of F. noatunensis subsp. orientalis-derived MVs.


Subject(s)
Bacterial Proteins/immunology , Fish Diseases/prevention & control , Francisella/immunology , Gram-Negative Bacterial Infections/prevention & control , Vaccines/immunology , Aging , Animals , Fish Diseases/immunology , Gram-Negative Bacterial Infections/immunology , Zebrafish
13.
J Immunol ; 198(4): 1531-1542, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087665

ABSTRACT

NKG2D is a potent activating receptor that is expressed on cytotoxic immune cells such as CD8 T and NK cells, where it promotes cytotoxicity after binding stress ligands on infected or transformed cells. On NK cell precursors NKG2D modulates proliferation and maturation. Previously, we observed that NKG2D deficiency affects peripheral B cell numbers. In this study, we show that NKG2D regulates B1a cell development and function. We find that mice deficient for NKG2D have a strong reduction of B1a cell numbers. As a result, NKG2D-deficient mice produce significantly less Ag-specific IgM Abs upon immunization with T cell-independent Ags, and they are more susceptible to Gram-negative sepsis. Klrk1-/- B1a cells are also functionally impaired and they fail to provide protection against Francisella novicida upon adoptive transfer. Using mixed bone marrow chimeric mice, we show that the impact of NKG2D deficiency on B1a cell development is cell intrinsic. No changes in homeostatic turnover and homing of B cells were detectable, limiting the effects of NKG2D to modulation of the hematopoietic development of B1a cells. Using conditional ablation, we demonstrate that the effect of NKG2D on B1a cell development occurs at a developmental stage that precedes the common lymphoid progenitor. Our findings reveal an unexpected new role for NKG2D in the regulation of B1a cell development. The protective effects of this activating receptor therefore reach beyond that of cytotoxic cells, stimulating the immune system to fight bacterial infections by promoting development of innate-like B cells.


Subject(s)
B-Lymphocytes/physiology , Cell Differentiation , Gram-Negative Bacterial Infections/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/prevention & control , Histocompatibility Antigens Class I/immunology , Immunoglobulin M/immunology , Killer Cells, Natural/immunology , Mice , NK Cell Lectin-Like Receptor Subfamily K/deficiency , Spleen/cytology , Spleen/immunology
14.
Microbes Infect ; 19(2): 91-100, 2017 02.
Article in English | MEDLINE | ID: mdl-27965147

ABSTRACT

Respiratory infection of mice with Francisella novicida has recently been used as a model for the highly virulent human pathogen Francisella tularensis. Similar to F. tularensis, even small doses of F. novicida administered by respiratory routes are lethal for inbred laboratory mice. This feature obviously limits study of infection-induced immunity. Parenteral sublethal infections of mice with F. novicida are feasible, but the resulting immune responses are incompletely characterized. Here we use parenteral intradermal (i.d.) and intraperitoneal (i.p.) F. novicida infections of C57BL/6J mice to determine the role of B cells in controlling primary and secondary F. novicida infections. Despite developing comparable levels of F. novicida-primed T cells, B cell knockout mice were much more susceptible to both primary i.d. infection and secondary i.p. challenge than wild type (normal) C57BL/6J mice. Transfer of F. novicida-immune sera to either wild type C57BL/6J mice or to B cell knockout mice did not appreciably impact survival of subsequent lethal F. novicida challenge. However, F. novicida-immune mice that were depleted of T cells after priming but just before challenge survived and cleared secondary i.p. F. novicida challenge. Collectively these results indicate that B cells, if not serum antibodies, play a major role in controlling F. novicida infections in mice.


Subject(s)
B-Lymphocytes/immunology , Francisella/immunology , Gram-Negative Bacterial Infections/mortality , Gram-Negative Bacterial Infections/prevention & control , Respiratory Tract Infections/mortality , Respiratory Tract Infections/prevention & control , Animals , Disease Models, Animal , Injections, Intradermal , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Mice, Knockout , Survival Analysis
15.
Cell ; 167(2): 382-396.e17, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27693356

ABSTRACT

The inflammasome is an intracellular signaling complex, which on recognition of pathogens and physiological aberration, drives activation of caspase-1, pyroptosis, and the release of the pro-inflammatory cytokines IL-1ß and IL-18. Bacterial ligands must secure entry into the cytoplasm to activate inflammasomes; however, the mechanisms by which concealed ligands are liberated in the cytoplasm have remained unclear. Here, we showed that the interferon-inducible protein IRGB10 is essential for activation of the DNA-sensing AIM2 inflammasome by Francisella novicida and contributed to the activation of the LPS-sensing caspase-11 and NLRP3 inflammasome by Gram-negative bacteria. IRGB10 directly targeted cytoplasmic bacteria through a mechanism requiring guanylate-binding proteins. Localization of IRGB10 to the bacterial cell membrane compromised bacterial structural integrity and mediated cytosolic release of ligands for recognition by inflammasome sensors. Overall, our results reveal IRGB10 as part of a conserved signaling hub at the interface between cell-autonomous immunity and innate immune sensing pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Francisella/immunology , GTP Phosphohydrolases/metabolism , Gram-Negative Bacterial Infections/immunology , Host-Pathogen Interactions/immunology , Inflammasomes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , B-Lymphocytes/immunology , Caspases/metabolism , Caspases, Initiator , Cytosol/immunology , Cytosol/microbiology , GTP Phosphohydrolases/genetics , Gram-Negative Bacterial Infections/microbiology , Immunity, Cellular , Immunity, Innate , Inflammasomes/metabolism , Ligands , Mice , Mice, Mutant Strains , Myeloid Cells/immunology , T-Lymphocytes/immunology
16.
J Exp Med ; 213(10): 2081-97, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27551156

ABSTRACT

Lysosomal cathepsins regulate an exquisite range of biological functions, and their deregulation is associated with inflammatory, metabolic, and degenerative diseases in humans. In this study, we identified a key cell-intrinsic role for cathepsin B as a negative feedback regulator of lysosomal biogenesis and autophagy. Mice and macrophages lacking cathepsin B activity had increased resistance to the cytosolic bacterial pathogen Francisella novicida Genetic deletion or pharmacological inhibition of cathepsin B down-regulated mechanistic target of rapamycin activity and prevented cleavage of the lysosomal calcium channel TRPML1. These events drove transcription of lysosomal and autophagy genes via transcription factor EB, which increased lysosomal biogenesis and activation of autophagy initiation kinase ULK1 for clearance of the bacteria. Our results identified a fundamental biological function of cathepsin B in providing a checkpoint for homeostatic maintenance of lysosome populations and basic recycling functions in the cell.


Subject(s)
Cathepsin B/metabolism , Francisella/immunology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Host-Pathogen Interactions , Lysosomes/metabolism , Organelle Biogenesis , Animals , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Bone Marrow Cells/pathology , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Gram-Negative Bacterial Infections/enzymology , Intracellular Space/microbiology , Ion Channel Gating , Macrophages/metabolism , Macrophages/ultrastructure , Mice, Inbred C57BL , Models, Biological , NF-kappa B/metabolism , Signal Transduction , Transient Receptor Potential Channels/metabolism , Up-Regulation
17.
J Mol Biol ; 428(17): 3387-98, 2016 08 28.
Article in English | MEDLINE | ID: mdl-27456933

ABSTRACT

Autophagy is a conserved lysosomal recycling process, which maintains cellular homeostasis during stress and starvation conditions by degrading and recycling proteins, lipids, and carbohydrates, ultimately increasing nutrient availability in eukaryotes. An additional function of autophagy, termed xenophagy, is to detect, capture, and destroy invading microorganisms, such as viruses, bacteria, and protozoa, providing autophagy with a role in innate immunity. Many intracellular pathogens have, however, developed mechanisms to avoid xenophagy and have evolved strategies to take advantage of select autophagic processes to undergo their intracellular life cycle. This review article will discuss the molecular mechanisms used by the intracellular bacterial pathogens Francisella spp. and Brucella spp. to manipulate components of the autophagic pathway, promoting cytosolic growth in the case of Francisella spp. and facilitating cellular egress and cell-to-cell spread in the case of Brucella spp. These examples highlight how successful, highly infectious bacterial pathogens avoid or subvert host autophagy mechanisms normally employed to maintain eukaryotic homeostasis.


Subject(s)
Autophagy , Brucella/immunology , Brucella/pathogenicity , Francisella/immunology , Francisella/pathogenicity , Host-Pathogen Interactions , Immune Evasion , Animals , Humans
18.
Sci Rep ; 6: 28318, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27324690

ABSTRACT

Previous analyses of the Atlantic cod genome showed unique combinations of lacking and expanded number of genes for the immune system. The present study examined lysozyme activity, lysozyme gene distribution and expression in cod. Enzymatic assays employing specific bacterial lysozyme inhibitors provided evidence for presence of g-type, but unexpectedly not for c-type lysozyme activity. Database homology searches failed to identify any c-type lysozyme gene in the cod genome or in expressed sequence tags from cod. In contrast, we identified four g-type lysozyme genes (LygF1a-d) constitutively expressed, although differentially, in all cod organs examined. The active site glutamate residue is replaced by alanine in LygF1a, thus making it enzymatic inactive, while LygF1d was found in two active site variants carrying alanine or glutamate, respectively. In vitro and in vivo infection by the intracellular bacterium Francisella noatunensis gave a significantly reduced LygF1a and b expression but increased expression of the LygF1c and d genes as did also the interferon gamma (IFNγ) cytokine. These results demonstrate a lack of c-type lysozyme that is unprecedented among vertebrates. Our results further indicate that serial gene duplications have produced multiple differentially regulated cod g-type lysozymes with specialised functions potentially compensating for the lack of c-type lysozymes.


Subject(s)
Fish Proteins/genetics , Gadus morhua/genetics , Muramidase/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Chickens/genetics , Fish Diseases/enzymology , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Proteins/chemistry , Fish Proteins/metabolism , Francisella/immunology , Gadus morhua/immunology , Gadus morhua/metabolism , Geese/genetics , Gene Expression , Interferon-gamma/genetics , Interferon-gamma/metabolism , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Models, Molecular , Muramidase/chemistry , Muramidase/metabolism , Organ Specificity/immunology , Phylogeny
19.
PLoS One ; 10(5): e0127278, 2015.
Article in English | MEDLINE | ID: mdl-25993107

ABSTRACT

Inflammasome activation is a two-step process where step one, priming, prepares the inflammasome for its subsequent activation, by step two. Classically step one can be induced by LPS priming followed by step two, high dose ATP. Furthermore, when IL-18 processing is used as the inflammasome readout, priming occurs before new protein synthesis. In this context, how intracellular pathogens such as Francisella activate the inflammasome is incompletely understood, particularly regarding the relative importance of priming versus activation steps. To better understand these events we compared Francisella strains that differ in virulence and ability to induce inflammasome activation for their relative effects on step one vs. step two. When using the rapid priming model, i.e., 30 min priming by live or heat killed Francisella strains (step 1), followed by ATP (step 2), we found no difference in IL-18 release, p20 caspase-1 release and ASC oligomerization between Francisella strains (F. novicida, F. holarctica -LVS and F. tularensis Schu S4). This priming is fast, independent of bacteria viability, internalization and phagosome escape, but requires TLR2-mediated ERK phosphorylation. In contrast to their efficient priming capacity, Francisella strains LVS and Schu S4 were impaired in inflammasome triggering compared to F. novicida. Thus, observed differences in inflammasome activation by F. novicida, LVS and Schu S4 depend not on differences in priming but rather on their propensity to trigger the primed inflammasome.


Subject(s)
Francisella/classification , Francisella/pathogenicity , Inflammasomes/metabolism , Monocytes/microbiology , Adenosine Triphosphate/metabolism , CARD Signaling Adaptor Proteins , Cells, Cultured , Cytoskeletal Proteins/metabolism , Francisella/immunology , Humans , Interleukin-18/metabolism , MAP Kinase Signaling System , Microbial Viability , Monocytes/metabolism , Phosphorylation , Toll-Like Receptor 2/metabolism , Virulence
20.
PLoS One ; 10(4): e0123573, 2015.
Article in English | MEDLINE | ID: mdl-25898318

ABSTRACT

Sepsis is a complex immune disorder that is characterized by systemic hyperinflammation. Alarmins, which are multifunctional endogenous factors, have been implicated in exacerbation of inflammation in many immune disorders including sepsis. Here we show that Galectin-9, a host endogenous ß-galactoside binding lectin, functions as an alarmin capable of mediating inflammatory response during sepsis resulting from pulmonary infection with Francisella novicida, a Gram negative bacterial pathogen. Our results show that this galectin is upregulated and is likely released during tissue damage in the lungs of F. novicida infected septic mice. In vitro, purified recombinant galectin-9 exacerbated F. novicida-induced production of the inflammatory mediators by macrophages and neutrophils. Concomitantly, Galectin-9 deficient (Gal-9-/-) mice exhibited improved lung pathology, reduced cell death and reduced leukocyte infiltration, particularly neutrophils, in their lungs. This positively correlated with overall improved survival of F. novicida infected Gal-9-/- mice as compared to their wild-type counterparts. Collectively, these findings suggest that galectin-9 functions as a novel alarmin by augmenting the inflammatory response in sepsis development during pulmonary F. novicida infection.


Subject(s)
Bronchopneumonia/immunology , Galectins/physiology , Pneumonia, Bacterial/immunology , Tularemia/immunology , Alarmins/physiology , Animals , Bronchopneumonia/metabolism , Bronchopneumonia/microbiology , Female , Francisella/immunology , Inflammation Mediators/metabolism , Lung/immunology , Lung/microbiology , Lung/pathology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Tularemia/metabolism , Tularemia/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...