Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.185
Filter
1.
Biochemistry (Mosc) ; 89(3): 574-582, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38648774

ABSTRACT

Rabies is a zoonotic disease with high lethality. Most human deaths are associated with the bites received from dogs and cats. Vaccination is the most effective method of preventing rabies disease in both animals and humans. In this study, the ability of an adjuvant based on recombinant Salmonella typhimurium flagellin to increase protective activity of the inactivated rabies vaccine in mice was evaluated. A series of inactivated dry culture vaccine for dogs and cats "Rabikan" (strain Shchelkovo-51) with addition of an adjuvant at various dilutions were used. The control preparation was a similar series of inactivated dry culture vaccine without an adjuvant. Protective activity of the vaccine preparations was evaluated by the NIH potency test, which is the most widely used and internationally recommended method for testing effectiveness of the inactivated rabies vaccines. The value of specific activity of the tested rabies vaccine when co-administered with the adjuvant was significantly higher (48.69 IU/ml) than that of the vaccine without the adjuvant (3.75 IU/ml). Thus, recombinant flagellin could be considered as an effective adjuvant in the composition of future vaccine preparations against rabies virus.


Subject(s)
Adjuvants, Immunologic , Flagellin , Rabies Vaccines , Rabies , Vaccines, Inactivated , Rabies Vaccines/immunology , Rabies Vaccines/administration & dosage , Animals , Flagellin/immunology , Mice , Rabies/prevention & control , Rabies/immunology , Vaccines, Inactivated/immunology , Dogs , Rabies virus/immunology , Salmonella typhimurium/immunology , Female , Cats
2.
Poult Sci ; 103(5): 103569, 2024 May.
Article in English | MEDLINE | ID: mdl-38447310

ABSTRACT

Non-typhoidal Salmonella infection is a significant health and economic burden in poultry industry. Developing an oral vaccine to induce robust mucosal immunity in the intestines of birds, especially cross protection against different Salmonella serotypes is challenging. Therefore, a potent oral vaccine platform that can mitigate different serotypes of Salmonella is warranted for the poultry industry. We reported earlier that the Salmonella enteritidis (SE) immunogenic outer membrane proteins (OMPs) and flagellin (FLA) entrapped in mannose chitosan nanoparticles (OMPs-FLA-mCS NPs) administered prime-boost (d-3 and 3-wk later) by oral inoculation elicits mucosal immunity and reduces challenge SE colonization by over 1 log10 CFU in birds. In this study, we sought to evaluate whether the SE antigens containing OMPs-FLA-mCS NPs vaccine induces cross-protection against Salmonella typhimurium (ST) in broilers. Our data indicated that the OMPs-FLA-mCS NPs vaccine induced higher cross-protective antibody responses compared to commercial Poulvac ST vaccine (contains a modified-live ST bacterium). Particularly, OMPs-FLA-mCS-NP vaccine elicited OMPs and FLA antigens specific increased production of secretory IgA and IgY antibodies in samples collected at both post-vaccination and post-challenge timepoints compared to commercial vaccine group. Notably, the vaccine reduced the challenge ST bacterial load by 0.8 log10 CFU in the cecal content, which was comparable to the outcome of Poulvac ST vaccination. In conclusion, our data suggested that orally administered OMPs-FLA-mCS-NP SE vaccine elicited cross protective mucosal immune responses against ST colonization in broilers. Thus, this candidate vaccine could be a viable option replacing the existing both live and killed Salmonella vaccines for birds.


Subject(s)
Chickens , Chitosan , Cross Protection , Nanoparticles , Poultry Diseases , Salmonella Infections, Animal , Salmonella Vaccines , Salmonella enteritidis , Salmonella typhimurium , Animals , Chickens/immunology , Salmonella enteritidis/immunology , Poultry Diseases/prevention & control , Poultry Diseases/immunology , Salmonella Infections, Animal/prevention & control , Salmonella Infections, Animal/immunology , Chitosan/administration & dosage , Chitosan/pharmacology , Salmonella Vaccines/immunology , Salmonella Vaccines/administration & dosage , Nanoparticles/administration & dosage , Salmonella typhimurium/immunology , Administration, Oral , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
3.
Microb Pathog ; 175: 105959, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36581307

ABSTRACT

The growing emergence of resistant bacteria is the current global concern for the humans and animals. Vaccination could be the desirable method to preventing such infectious diseases. Safe and effective vaccines are urgently needed to manage and prevent Salmonella contamination. Subunit vaccines are safe approaches for the protection against Salmonella spp. The bioinformatics methods were performed to determine the gene structure. Gene cassette (rLPSI) was ordered in pET28a (+), and cloned into E.coli BL21 (DE3), and the recombinant protein was expressed using IPTG (1 mM). Mice were immunized by subcutaneous administration of recombinant protein. Then, the mice were challenged by oral administration of 100LD50 of live S. Typhimurium. The Codon adaptation index of the chimeric gene was multiplied by 0.92. Validation results showed that >90% of residues lie in the desired or extra allowed area of the Ramachandran plot. The recombinant protein (65.9 kDa) was expressed in E.coli. Antibody titers in vaccinated mice were significantly different from those in the control groups. Recombinant protein immunization of the mice provided 90% and 70% protection against 10LD50 and 100LD50 of S. Typhimurium, respectively. In general, the results showed the high efficiency of rLPSI chimeric protein as a protective antigen against S. Typhimurium infection. The rLPSI chimeric protein could be an effective recombinant vaccine candidate against S. Typhimurium infection, but more research is needed.


Subject(s)
Escherichia coli Proteins , Salmonella Vaccines , Salmonella typhimurium , Animals , Mice , Antibodies, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Escherichia coli/genetics , Immunization , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Proteins/genetics , Salmonella typhimurium/genetics , Salmonella typhimurium/immunology , Vaccines, Attenuated , Vaccines, Synthetic
4.
Nature ; 609(7926): 348-353, 2022 09.
Article in English | MEDLINE | ID: mdl-35978195

ABSTRACT

The mammalian immune system uses various pattern recognition receptors to recognize invaders and host damage and transmits this information to downstream immunometabolic signalling outcomes. Laccase domain-containing 1 (LACC1) protein is an enzyme highly expressed in inflammatory macrophages and serves a central regulatory role in multiple inflammatory diseases such as inflammatory bowel diseases, arthritis and clearance of microbial infection1-4. However, the biochemical roles required for LACC1 functions remain largely undefined. Here we elucidated a shared biochemical function of LACC1 in mice and humans, converting L-citrulline to L-ornithine (L-Orn) and isocyanic acid and serving as a bridge between proinflammatory nitric oxide synthase (NOS2) and polyamine immunometabolism. We validated the genetic and mechanistic connections among NOS2, LACC1 and ornithine decarboxylase 1 (ODC1) in mouse models and bone marrow-derived macrophages infected by Salmonella enterica Typhimurium. Strikingly, LACC1 phenotypes required upstream NOS2 and downstream ODC1, and Lacc1-/- chemical complementation with its product L-Orn significantly restored wild-type activities. Our findings illuminate a previously unidentified pathway in inflammatory macrophages, explain why its deficiency may contribute to human inflammatory diseases and suggest that L-Orn could serve as a nutraceutical to ameliorate LACC1-associated immunological dysfunctions such as arthritis or inflammatory bowel disease.


Subject(s)
Inflammation , Intracellular Signaling Peptides and Proteins , Macrophages , Nitric Oxide Synthase Type II , Animals , Arthritis/immunology , Arthritis/metabolism , Citrulline/metabolism , Cyanates/metabolism , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation/metabolism , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Nitric Oxide Synthase Type II/metabolism , Ornithine/metabolism , Ornithine Decarboxylase/metabolism , Polyamines/metabolism , Salmonella typhimurium/immunology
5.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163280

ABSTRACT

The rapid identification of bacterial antibiotic susceptibility is pivotal to the rational administration of antibacterial drugs. In this study, cefotaxime (CTX)-derived resistance in Salmonella typhimurium (abbr. CTXr-S. typhimurium) during 3 months of exposure was rapidly recorded using a portable Raman spectrometer. The molecular changes that occurred in the drug-resistant strains were sensitively monitored in whole cells by label-free surface-enhanced Raman scattering (SERS). Various degrees of resistant strains could be accurately discriminated by applying multivariate statistical analyses to bacterial SERS profiles. Minimum inhibitory concentration (MIC) values showed a positive linear correlation with the relative Raman intensities of I990/I1348, and the R2 reached 0.9962. The SERS results were consistent with the data obtained by MIC assays, mutant prevention concentration (MPC) determinations, and Kirby-Bauer antibiotic susceptibility tests (K-B tests). This preliminary proof-of-concept study indicates the high potential of the SERS method to supplement the time-consuming conventional method and help alleviate the challenges of antibiotic resistance in clinical therapy.


Subject(s)
Salmonella Infections/immunology , Salmonella typhimurium/immunology , Spectrum Analysis, Raman/methods , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Drug Resistance, Bacterial/genetics , Drug Resistance, Microbial/drug effects , Humans , Salmonella Infections/diagnosis , Salmonella typhimurium/drug effects , Salmonella typhimurium/pathogenicity
6.
PLoS Pathog ; 18(1): e1009718, 2022 01.
Article in English | MEDLINE | ID: mdl-35073381

ABSTRACT

Salmonella enterica serovar Typhimurium is a Gram-negative pathogen that uses two distinct type III secretion systems (T3SSs), termed Salmonella pathogenicity island (SPI)-1 and SPI-2, to deliver virulence factors into the host cell. The SPI-1 T3SS enables Salmonella to invade host cells, while the SPI-2 T3SS facilitates Salmonella's intracellular survival. In mice, a family of cytosolic immune sensors, including NAIP1, NAIP2, and NAIP5/6, recognizes the SPI-1 T3SS needle, inner rod, and flagellin proteins, respectively. Ligand recognition triggers assembly of the NAIP/NLRC4 inflammasome, which mediates caspase-1 activation, IL-1 family cytokine secretion, and pyroptosis of infected cells. In contrast to mice, humans encode a single NAIP that broadly recognizes all three ligands. The role of NAIP/NLRC4 or other inflammasomes during Salmonella infection of human macrophages is unclear. We find that although the NAIP/NLRC4 inflammasome is essential for detecting T3SS ligands in human macrophages, it is partially required for responses to infection, as Salmonella also activated the NLRP3 and CASP4/5 inflammasomes. Importantly, we demonstrate that combinatorial NAIP/NLRC4 and NLRP3 inflammasome activation restricts Salmonella replication in human macrophages. In contrast to SPI-1, the SPI-2 T3SS inner rod is not sensed by human or murine NAIPs, which is thought to allow Salmonella to evade host recognition and replicate intracellularly. Intriguingly, we find that human NAIP detects the SPI-2 T3SS needle protein. Critically, in the absence of both flagellin and the SPI-1 T3SS, the NAIP/NLRC4 inflammasome still controlled intracellular Salmonella burden. These findings reveal that recognition of Salmonella SPI-1 and SPI-2 T3SSs and engagement of both the NAIP/NLRC4 and NLRP3 inflammasomes control Salmonella infection in human macrophages.


Subject(s)
Inflammasomes/immunology , Macrophages/immunology , Macrophages/microbiology , Salmonella Infections/immunology , Type III Secretion Systems/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neuronal Apoptosis-Inhibitory Protein/immunology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Virulence
7.
J Biol Chem ; 298(1): 101461, 2022 01.
Article in English | MEDLINE | ID: mdl-34864057

ABSTRACT

Inflammasome signaling results in cell death and release of cytokines from the IL-1 family, which facilitates control over an infection. However, some pathogens such as Salmonella typhimurium (ST) activate various innate immune signaling pathways, including inflammasomes, yet evade these cell death mechanisms, resulting in a chronic infection. Here we investigated inflammasome signaling induced by acute and chronic isolates of ST obtained from different organs. We show that ST isolated from infected mice during the acute phase displays an increased potential to activate inflammasome signaling, which then undergoes a protracted decline during the chronic phase of infection. This decline in inflammasome signaling was associated with reduced expression of virulence factors, including flagella and the Salmonella pathogenicity island I genes. This reduction in cell death of macrophages induced by chronic isolates had the greatest impact on the NLRP3 inflammasome, which correlated with a reduction in caspase-1 activation. Furthermore, rapid cell death induced by Casp-1/11 by ST in macrophages limited the subsequent activation of cell death cascade proteins Casp-8, RipK1, RipK3, and MLKL to prevent the activation of alternative forms of cell death. We observed that the lack of the ability to induce cell death conferred a competitive fitness advantage to ST only during the acute phase of infection. Finally, we show that the chronic isolates displayed a significant attenuation in their ability to infect mice through the oral route. These results reveal that ST adapts during chronic infection by circumventing inflammasome recognition to promote the survival of both the host and the pathogen.


Subject(s)
Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein , Salmonella Infections , Salmonella typhimurium , Animals , Caspase 1/genetics , Caspase 1/metabolism , Host-Pathogen Interactions/immunology , Inflammasomes/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/isolation & purification
8.
Gut Microbes ; 13(1): 1991776, 2021.
Article in English | MEDLINE | ID: mdl-34719317

ABSTRACT

Host engulfment protein ELMO1 generates intestinal inflammation following internalization of enteric bacteria. In Shigella, bacterial effector IpgB1 interacts with ELMO1 and promotes bacterial invasion. IpgB1 belongs to the WxxxE effector family, a motif found in several effectors of enteric pathogens. Here, we have studied the role of WxxxE effectors, with emphasis on Salmonella SifA and whether it interacts with ELMO1 to regulate inflammation. In-silico-analysis of WxxxE effectors was performed using BLAST search and Clustal W program. The interaction of ELMO1 with SifA was assessed by GST pulldown assay and co-immunoprecipitation. ELMO1 knockout mice, and ELMO1-depleted murine macrophage J774 cell lines were challenged with WT and SifA mutant Salmonella. Bacterial effectors containing the WxxxE motif were transfected in WT and ELMO1-depleted J774 cells to assess the inflammatory cytokines. ELMO1 generates differential pro-inflammatory cytokines between pathogenic and nonpathogenic bacteria. WxxxE motif is present in pathogens and in the TIR domain of host proteins. The C-terminal part of ELMO1 interacts with SifA where WxxxE motif is important for interaction. ELMO1-SifA interaction affects bacterial colonization, dissemination, and inflammatory cytokines in vivo. Moreover, ELMO1-SifA interaction increases TNF-α and IL-6 production from the macrophage cell line and is associated with enhanced Rac1 activity. ELMO1 also interacts with WxxxE effectors IpgB1, IpgB2, and Map and induces inflammation after challenge with microbes or microbial ligands. ELMO1 generates a differential response through interaction with the WxxxE motif, which is absent in commensals. ELMO1-WxxxE interaction plays a role in bacterial pathogenesis and induction of inflammatory response.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Bacterial Proteins/immunology , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Bacterial Proteins/genetics , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Immunity, Innate , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Salmonella Infections/genetics , Salmonella Infections/microbiology , Salmonella typhimurium/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
9.
Front Immunol ; 12: 729607, 2021.
Article in English | MEDLINE | ID: mdl-34804014

ABSTRACT

The mucosal immune system is the first line of defense against pathogens. Germinal centers (GCs) in the Peyer's patches (PPs) of the small intestine are constantly generated through stimulation of the microbiota. In this study, we investigated the role of γδ T cells in the GC reactions in PPs. Most γδ T cells in PPs localized in the GCs and expressed a TCR composed of Vγ1 and Vδ6 chains. By using mice with partial and total γδ T cell deficiencies, we found that Vγ1+/Vδ6+ T cells can produce high amounts of IL-4, which drives the proliferation of GC B cells as well as the switch of GC B cells towards IgA. Therefore, we conclude that γδ T cells play a role in sustaining gut homeostasis and symbiosis via supporting the GC reactions in PPs.


Subject(s)
B-Lymphocytes/metabolism , Germinal Center/metabolism , Interleukin-4/metabolism , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/metabolism , Peyer's Patches/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Germinal Center/immunology , Germinal Center/microbiology , Immunity, Mucosal , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Immunoglobulin Class Switching , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/microbiology , Lymphocyte Activation , Lymphocyte Depletion , Mice, Knockout , Peyer's Patches/immunology , Peyer's Patches/microbiology , Phenotype , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella Infections/microbiology , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Signal Transduction
10.
Int J Mol Sci ; 22(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638530

ABSTRACT

Outer Membrane Vesicles (OMV) constitute a promising platform for the development of efficient vaccines. OMV can be decorated with heterologous antigens (proteins or polysaccharides), becoming attractive novel carriers for the development of multicomponent vaccines. Chemical conjugation represents a tool for linking antigens, also from phylogenetically distant pathogens, to OMV. Here we develop two simple and widely applicable conjugation chemistries targeting proteins or lipopolysaccharides on the surface of Generalized Modules for Membrane Antigens (GMMA), OMV spontaneously released from Gram-negative bacteria mutated to increase vesicle yield and reduce potential reactogenicity. A Design of Experiment approach was used to identify optimal conditions for GMMA activation before conjugation, resulting in consistent processes and ensuring conjugation efficiency. Conjugates produced by both chemistries induced strong humoral response against the heterologous antigen and GMMA. Additionally, the use of the two orthogonal chemistries allowed to control the linkage of two different antigens on the same GMMA particle. This work supports the further advancement of this novel platform with great potential for the design of effective vaccines.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Extracellular Vesicles/immunology , Protozoan Proteins/immunology , Protozoan Vaccines/immunology , Animals , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Vaccines/biosynthesis , Female , Lipopolysaccharides/immunology , Mice , Neisseria meningitidis/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/chemistry , Protozoan Vaccines/biosynthesis , Salmonella typhimurium/immunology , Shigella sonnei/immunology
11.
PLoS Pathog ; 17(10): e1010004, 2021 10.
Article in English | MEDLINE | ID: mdl-34695149

ABSTRACT

While Salmonella enterica is seen as an archetypal facultative intracellular bacterial pathogen where protection is mediated by CD4+ T cells, identifying circulating protective cells has proved very difficult, inhibiting steps to identify key antigen specificities. Exploiting a mouse model of vaccination, we show that the spleens of C57BL/6 mice vaccinated with live-attenuated Salmonella serovar Typhimurium (S. Typhimurium) strains carried a pool of IFN-γ+ CD4+ T cells that could adoptively transfer protection, but only transiently. Circulating Salmonella-reactive CD4+ T cells expressed the liver-homing chemokine receptor CXCR6, accumulated over time in the liver and assumed phenotypic characteristics associated with tissue-associated T cells. Liver memory CD4+ T cells showed TCR selection bias and their accumulation in the liver could be inhibited by blocking CXCL16. These data showed that the circulation of CD4+ T cells mediating immunity to Salmonella is limited to a brief window after which Salmonella-specific CD4+ T cells migrate to peripheral tissues. Our observations highlight the importance of triggering tissue-specific immunity against systemic infections.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Liver/immunology , Salmonella Infections, Animal/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Salmonella typhimurium/immunology
12.
Front Immunol ; 12: 719315, 2021.
Article in English | MEDLINE | ID: mdl-34594333

ABSTRACT

Nanoparticle systems are being explored for the display of carbohydrate antigens, characterized by multimeric presentation of glycan epitopes and special chemico-physical properties of nano-sized particles. Among them, outer membrane vesicles (OMVs) are receiving great attention, combining antigen presentation with the immunopotentiator effect of the Toll-like receptor agonists naturally present on these systems. In this context, we are testing Generalized Modules for Membrane Antigens (GMMA), OMVs naturally released from Gram-negative bacteria mutated to increase blebbing, as carrier for polysaccharides. Here, we investigated the impact of saccharide length, density, and attachment site on the immune response elicited by GMMA in animal models, using a variety of structurally diverse polysaccharides from different pathogens (i.e., Neisseria meningitidis serogroup A and C, Haemophilus influenzae type b, and streptococcus Group A Carbohydrate and Salmonella Typhi Vi). Anti-polysaccharide immune response was not affected by the number of saccharides per GMMA particle. However, lower saccharide loading can better preserve the immunogenicity of GMMA as antigen. In contrast, saccharide length needs to be optimized for each specific antigen. Interestingly, GMMA conjugates induced strong functional immune response even when the polysaccharides were linked to sugars on GMMA. We also verified that GMMA conjugates elicit a T-dependent humoral immune response to polysaccharides that is strictly dependent on the nature of the polysaccharide. The results obtained are important to design novel glycoconjugate vaccines using GMMA as carrier and support the development of multicomponent glycoconjugate vaccines where GMMA can play the dual role of carrier and antigen. In addition, this work provides significant insights into the mechanism of action of glycoconjugates.


Subject(s)
Antigens, Bacterial/immunology , Cell Membrane/immunology , Glycoconjugates/immunology , Polysaccharides, Bacterial/immunology , Animals , Antigens, Bacterial/chemistry , Carrier Proteins/chemistry , Carrier Proteins/immunology , Cell Membrane/chemistry , Female , Glycoconjugates/chemistry , Immunity , Mice , Models, Animal , Polysaccharides, Bacterial/chemistry , Salmonella typhimurium/immunology , Vaccines/chemistry , Vaccines/immunology
13.
Int Immunopharmacol ; 101(Pt A): 108185, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607234

ABSTRACT

Typically, the killed form of microorganisms in combination with alum does not produce strong cellular immune responses. A recent investigation has indicated the role of dopamine D2 receptor antagonists like metoclopramide in reducing the polarization of immune responses toward Th2 immunity. This study was performed to evaluate the effects of a combination of alum and metoclopramide on the induction of cellular and humoral immunity in response to a heat-killed preparation ofSalmonella typhimurium(HKST). Wistar rats were immunized with the HKST vaccine alone or in combination with alum, metoclopramide, or the alum-metoclopramide mixture twice with a two-week interval. Fourteen days after the last vaccination, immune responses against S. typhimurium and the protective potential of the vaccines were assessed. The combination of alum and metoclopramide as an adjuvant augmented the potential of the HKST vaccine to enhance lymphocyte proliferation, delayed-type hypersensitivity reaction, and antibody titer. These results were concurrent with the polarization of immune response towards the Th1 response and improving protective immunity against S. typhimurium. Overall, the combination of alum and metoclopramide as an adjuvant synergistically enhanced cellular and humoral immunity after immunization with the HKST vaccine.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Alum Compounds/therapeutic use , Immunity, Cellular/drug effects , Immunity, Humoral/drug effects , Metoclopramide/therapeutic use , Salmonella typhimurium/immunology , Typhoid-Paratyphoid Vaccines/therapeutic use , Adjuvants, Immunologic/administration & dosage , Alum Compounds/adverse effects , Animals , Drug Synergism , Hypersensitivity, Delayed/immunology , Male , Metoclopramide/administration & dosage , Rats , Rats, Wistar , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/prevention & control , Vaccines, Inactivated/therapeutic use
14.
Eur J Immunol ; 51(12): 3228-3238, 2021 12.
Article in English | MEDLINE | ID: mdl-34633664

ABSTRACT

The use of bacteria as an alternative cancer therapy has been reinvestigated in recent years. SL7207: an auxotrophic Salmonella enterica serovar Typhimurium aroA mutant with immune-stimulatory potential has proven a promising strain for this purpose. Here, we show that systemic administration of SL7207 induces melanoma tumor growth arrest in vivo, with greater survival of the SL7207-treated group compared to control PBS-treated mice. Administration of SL7207 is accompanied by a change in the immune phenotype of the tumor-infiltrating cells toward pro-inflammatory, with expression of the TH 1 cytokines IFN-γ, TNF-α, and IL-12 significantly increased. Interestingly, Ly6C+ MHCII+ monocytes were recruited to the tumors following SL7207 treatment and were pro-inflammatory. Accordingly, the abrogation of these infiltrating monocytes using clodronate liposomes prevented SL7207-induced tumor growth inhibition. These data demonstrate a previously unappreciated role for infiltrating inflammatory monocytes underlying bacterial-mediated tumor growth inhibition. This information highlights a possible novel role for monocytes in controlling tumor growth, contributing to our understanding of the immune responses required for successful immunotherapy of cancer.


Subject(s)
Immunotherapy , Melanoma, Experimental , Monocytes/immunology , Salmonella typhimurium/immunology , Th1 Cells/immunology , Animals , Cytokines/immunology , Female , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Salmonella typhimurium/genetics
15.
Microbiol Spectr ; 9(2): e0145721, 2021 10 31.
Article in English | MEDLINE | ID: mdl-34704781

ABSTRACT

The rck open reading frame (ORF) on the pefI-srgC operon encodes an outer membrane protein responsible for invasion of nonphagocytic cell lines and resistance to complement-mediated killing. Until now, the rck ORF was only detected on the virulence plasmids of three serovars of Salmonella subsp. enterica (i.e., Bovismorbificans, Enteritidis, and Typhimurium). The increasing number of Salmonella genome sequences allowed us to use a combination of reference sequences and whole-genome multilocus sequence typing (wgMLST) data analysis to probe the presence of the operon and of rck in a wide array of isolates belonging to all Salmonella species and subspecies. We established the presence of partial or complete operons in 61 subsp. enterica serovars as well as in 4 other subspecies with various syntenies and frequencies. The rck ORF itself was retrieved in 36 subsp. enterica serovars and in two subspecies with either chromosomal or plasmid-borne localization. It displays high conservation of its sequence within the genus, and we demonstrated that most of the allelic variations identified did not alter the virulence properties of the protein. However, we demonstrated the importance of the residue at position 38 (at the level of the first extracellular loop of the protein) in the invasin function of Rck. Altogether, our results highlight that rck is not restricted to the three formerly identified serovars and could therefore have a more important role in virulence than previously expected. Moreover, this work raises questions about the mechanisms involved in rck acquisition and about virulence plasmid distribution and evolution. IMPORTANCE The foodborne pathogen Salmonella is responsible for a wide variety of pathologies depending on the infected host, the infecting serovars, and its set of virulence factors. However, the implication of each of these virulence factors and their role in the specific host-pathogen interplay are not fully understood. The significance of our research is in determining the distribution of one of these factors, the virulence plasmid-encoded invasin and resistance to complement killing protein Rck. In addition to providing elements of reflection concerning the mechanisms of acquisition of specific virulence genes in certain serotypes, this work will help to understand the role of Rck in the pathogenesis of Salmonella.


Subject(s)
Bacterial Outer Membrane Proteins/genetics , Complement System Proteins/immunology , Salmonella Infections/microbiology , Salmonella enterica/genetics , Salmonella typhimurium/genetics , Amino Acid Sequence , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Humans , Operon , Plasmids/genetics , Plasmids/immunology , Salmonella Infections/immunology , Salmonella enterica/immunology , Salmonella enterica/pathogenicity , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Sequence Alignment , Virulence
16.
ACS Synth Biol ; 10(10): 2478-2487, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34525796

ABSTRACT

Neoantigen vaccines are an immunotherapy strategy for treating cancer. The vaccine degrades quickly, so the strategy must include protection and precise targeting for immune cell stimulation. In this study, we engineered attenuated Salmonella typhimurium, which is highly infiltrative to tumors, to act as a carrier for Neoantigen peptide vaccine. Our system used a constitutive promoter vector, so that a single injection of Salmonella expressing Neoantigen could be used without requiring additional induction injections. In vivo experiments on bacteria-treated mice showed that Neoantigen expressed by the engineered carrier infiltrated tumors and resulted in suppressed tumor growth, higher survival rates and longer survival times, a relative increase of CD4 and CD8 T cells, and cytokine release. These results indicate that engineered Salmonella can be used as a carrier for Neoantigen immunotherapy.


Subject(s)
Antigens/therapeutic use , Genetic Engineering , Immunotherapy/methods , Neoplasms, Experimental/therapy , Salmonella typhimurium/immunology , Animals , Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Salmonella typhimurium/genetics , Survival Rate , Tumor Microenvironment
17.
Front Immunol ; 12: 739938, 2021.
Article in English | MEDLINE | ID: mdl-34552598

ABSTRACT

Global increases in the prevalence of antimicrobial resistance highlight the urgent need for novel strategies to combat infectious diseases. Recent studies suggest that host metabolic pathways play a key role in host control of intracellular bacterial pathogens. In this study we explored the potential of targeting host metabolic pathways for innovative host-directed therapy (HDT) against intracellular bacterial infections. Through gene expression profiling in human macrophages, pyruvate metabolism was identified as potential key pathway involved in Salmonella enterica serovar Typhimurium (Stm) infections. Next, the effect of targeting pyruvate dehydrogenase kinases (PDKs) - which are regulators of the metabolic checkpoint pyruvate dehydrogenase complex (PDC) - on macrophage function and bacterial control was studied. Chemical inhibition of PDKs by dichloroacetate (DCA) induced PDC activation and was accompanied with metabolic rewiring in classically activated macrophages (M1) but not in alternatively activated macrophages (M2), suggesting cell-type specific effects of dichloroacetate on host metabolism. Furthermore, DCA treatment had minor impact on cytokine and chemokine secretion on top of infection, but induced significant ROS production by M1 and M2. DCA markedly and rapidly reduced intracellular survival of Stm, but interestingly not Mycobacterium tuberculosis, in human macrophages in a host-directed manner. In conclusion, DCA represents a promising novel HDT compound targeting pyruvate metabolism for the treatment of Stm infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dichloroacetic Acid/pharmacology , Macrophages/drug effects , Protein Kinase Inhibitors/pharmacology , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Salmonella Infections/drug therapy , Salmonella typhimurium/pathogenicity , Cells, Cultured , Energy Metabolism/drug effects , Host-Pathogen Interactions , Humans , Macrophage Activation , Macrophages/enzymology , Macrophages/immunology , Macrophages/microbiology , Phenotype , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Reactive Oxygen Species/metabolism , Salmonella Infections/enzymology , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella typhimurium/immunology
18.
Front Immunol ; 12: 688294, 2021.
Article in English | MEDLINE | ID: mdl-34394082

ABSTRACT

Polarization of macrophages to different functional states is important for mounting responses against pathogen infections. Macrophages are the major target cells of porcine circovirus type 2 (PCV2), which is the primary causative agent of porcine circovirus-associated disease (PCVAD) leading to immense economic losses in the global swine industry. Clinically, PCV2 is often found to increase risk of other pathogenic infections yet the underlying mechanisms remain to be elusive. Here we found that PCV2 infection skewed macrophages toward a M1 status through reprogramming expression of a subset of M1-associated genes and M2-associated genes. Mechanistically, induction of M1-associated genes by PCV2 infection is dependent on activation of nuclear factor kappa B (NF-κB) and c-jun N-terminal kinase (JNK) signaling pathways whereas suppression of M2-associated genes by PCV2 is via inhibiting expression of jumonji domain containing-3 (JMJD3), a histone 3 Lys27 (H3K27) demethylase that regulates M2 activation of macrophages. Finally, we identified that PCV2 capsid protein (Cap) directly inhibits JMJD3 transcription to restrain expression of interferon regulatory factor (IRF4) that controls M2 macrophage polarization. Consequently, sustained infection of PCV2 facilitates bacterial infection in vitro. In summary, these findings showed that PCV2 infection functionally modulated M1 macrophage polarization via targeting canonical signals and epigenetic histone modification, which contributes to bacterial coinfection and virial pathogenesis.


Subject(s)
Actinobacillus Infections/microbiology , Actinobacillus pleuropneumoniae/pathogenicity , Circoviridae Infections/virology , Circovirus/pathogenicity , Coinfection , Macrophages/microbiology , Macrophages/virology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Actinobacillus Infections/immunology , Actinobacillus Infections/metabolism , Actinobacillus pleuropneumoniae/immunology , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Circoviridae Infections/immunology , Circovirus/immunology , Disease Models, Animal , Epigenesis, Genetic , Host-Pathogen Interactions , Interferon Regulatory Factors/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Phenotype , Salmonella Infections/immunology , Salmonella Infections/metabolism , Salmonella typhimurium/immunology , Signal Transduction
19.
Sci Rep ; 11(1): 17214, 2021 08 26.
Article in English | MEDLINE | ID: mdl-34446765

ABSTRACT

Salmonella enterica serovar Gallinarum is a host-restricted bacterial pathogen that causes a serious systemic disease exclusively in birds of all ages. Salmonella enterica serovar Typhimurium is a host-generalist serovar. Dendritic cells (DCs) are key antigen-presenting cells that play an important part in Salmonella host-restriction. We evaluated the differential response of chicken blood monocyte-derived dendritic cells (chMoDCs) exposed to S. Gallinarum or S. Typhimurium. S. Typhimurium was found to be more invasive while S. Gallinarum was more cytotoxic at the early phase of infection and later showed higher resistance against chMoDCs killing. S. Typhimurium promoted relatively higher upregulation of costimulatory and other immune function genes on chMoDCs in comparison to S. Gallinarum during early phase of infection (6 h) as analyzed by real-time PCR. Both Salmonella serovars strongly upregulated the proinflammatory transcripts, however, quantum was relatively narrower with S. Gallinarum. S. Typhimurium-infected chMoDCs promoted relatively higher proliferation of naïve T-cells in comparison to S. Gallinarum as assessed by mixed lymphocyte reaction. Our findings indicated that host restriction of S. Gallinarum to chicken is linked with its profound ability to interfere the DCs function. Present findings provide a valuable roadmap for future work aimed at improved vaccine strategies against this pathogen.


Subject(s)
Dendritic Cells/immunology , Monocytes/immunology , Salmonella typhimurium/immunology , Salmonella/immunology , Animals , B7-1 Antigen/genetics , B7-1 Antigen/immunology , CD40 Antigens/genetics , CD40 Antigens/immunology , Chickens , Cytokines/genetics , Cytokines/immunology , Cytotoxicity, Immunologic/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Gene Expression/immunology , Host-Pathogen Interactions/immunology , Microbial Viability/immunology , Monocytes/cytology , Salmonella/physiology , Salmonella typhimurium/physiology , Species Specificity , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
20.
Science ; 373(6552)2021 07 16.
Article in English | MEDLINE | ID: mdl-34437126

ABSTRACT

Activation of cell-autonomous defense by the immune cytokine interferon-γ (IFN-γ) is critical to the control of life-threatening infections in humans. IFN-γ induces the expression of hundreds of host proteins in all nucleated cells and tissues, yet many of these proteins remain uncharacterized. We screened 19,050 human genes by CRISPR-Cas9 mutagenesis and identified IFN-γ-induced apolipoprotein L3 (APOL3) as a potent bactericidal agent protecting multiple non-immune barrier cell types against infection. Canonical apolipoproteins typically solubilize mammalian lipids for extracellular transport; APOL3 instead targeted cytosol-invasive bacteria to dissolve their anionic membranes into human-bacterial lipoprotein nanodiscs detected by native mass spectrometry and visualized by single-particle cryo-electron microscopy. Thus, humans have harnessed the detergent-like properties of extracellular apolipoproteins to fashion an intracellular lysin, thereby endowing resident nonimmune cells with a mechanism to achieve sterilizing immunity.


Subject(s)
Apolipoproteins L/metabolism , Cell Membrane/metabolism , Cytosol/microbiology , Gram-Negative Bacteria/physiology , Interferon-gamma/immunology , Apolipoproteins L/chemistry , Apolipoproteins L/genetics , Bacterial Outer Membrane/metabolism , Bacteriolysis , CRISPR-Cas Systems , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cell Membrane Permeability , Cells, Cultured , Detergents/metabolism , GTP-Binding Proteins/metabolism , Gene Editing , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/pathogenicity , Gram-Negative Bacteria/ultrastructure , Humans , Immunity, Innate , Lipoproteins/chemistry , Microbial Viability , O Antigens/metabolism , Protein Domains , Salmonella typhimurium/immunology , Salmonella typhimurium/pathogenicity , Salmonella typhimurium/physiology , Salmonella typhimurium/ultrastructure , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...