Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.195
Filtrer
1.
Vaccine ; 42(19S1): S101-S124, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39003017

RÉSUMÉ

Invasive non-typhoidal Salmonella (iNTS) disease is an under-recognized high-burden disease causing major health and socioeconomic issues in sub-Saharan Africa (sSA), predominantly among immune-naïve infants and young children, including those with recognized comorbidities such as HIV infection. iNTS disease is primarily caused by Salmonella enterica serovar Typhimurium sequence type (ST) 313 and 'African-restricted clades' of Salmonella Enteritidis ST11 that have emerged across the African continent as a series of epidemics associated with acquisition of new antimicrobial resistance. Due to genotypes with a high prevalence of antimicrobial resistance and scarcity of therapeutic options, these NTS serovars are designated by the World Health Organization as a priority pathogen for research and development of interventions, including vaccines, to address and reduce NTS associated bacteremia and meningitis in sSA. Novel and traditional vaccine technologies are being applied to develop vaccines against iNTS disease, and the results of the first clinical trials in the infant target population should become available in the near future. The "Vaccine Value Profile" (VVP) addresses information related predominantly to invasive disease caused by Salmonella Enteritidis and Salmonella Typhimurium prevalent in sSA. Information is included on stand-alone iNTS disease candidate vaccines and candidate vaccines targeting iNTS disease combined with another invasive serotype, Salmonella Typhi, that is also common across sSA. Out of scope for the first version of this VVP is a wider discussion on either diarrheagenic NTS disease (dNTS) also associated with Salmonella Enteritidis and Salmonella Typhimurium or the development of a multivalent Salmonella vaccines targeting key serovars for use globally. This VVP for vaccines to prevent iNTS disease is intended to provide a high-level, holistic assessment of the information and data that are currently available to inform the potential public health, economic, and societal value of pipeline vaccines and vaccine-like products. Future versions of this VVP will be updated to reflect ongoing activities such as vaccine development strategies and a "Full Vaccine Value Assessment" that will inform the value proposition of an iNTS disease vaccine. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations, and in collaboration with stakeholders from the World Health Organization African Region. All contributors have extensive expertise on various elements of the iNTS disease VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.


Sujet(s)
Salmonelloses , Vaccins antisalmonella , Salmonella enteritidis , Humains , Afrique subsaharienne/épidémiologie , Salmonella enteritidis/immunologie , Salmonella enteritidis/génétique , Salmonella enteritidis/pathogénicité , Salmonelloses/prévention et contrôle , Salmonelloses/épidémiologie , Salmonelloses/microbiologie , Salmonelloses/immunologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Salmonella typhimurium/génétique , Vaccins antisalmonella/immunologie , Vaccins antisalmonella/administration et posologie
2.
Exp Biol Med (Maywood) ; 249: 10081, 2024.
Article de Anglais | MEDLINE | ID: mdl-38974834

RÉSUMÉ

The lack of effective treatment options for an increasing number of cancer cases highlights the need for new anticancer therapeutic strategies. Immunotherapy mediated by Salmonella enterica Typhimurium is a promising anticancer treatment. Candidate strains for anticancer therapy must be attenuated while retaining their antitumor activity. Here, we investigated the attenuation and antitumor efficacy of two S. enterica Typhimurium mutants, ΔtolRA and ΔihfABpmi, in a murine melanoma model. Results showed high attenuation of ΔtolRA in the Galleria mellonella model, and invasion and survival in tumor cells. However, it showed weak antitumor effects in vitro and in vivo. Contrastingly, lower attenuation of the attenuated ΔihfABpmi strain resulted in regression of tumor mass in all mice, approximately 6 days after the first treatment. The therapeutic response induced by ΔihfABpmi was accompanied with macrophage accumulation of antitumor phenotype (M1) and significant increase in the mRNAs of proinflammatory mediators (TNF-α, IL-6, and iNOS) and an apoptosis inducer (Bax). Our findings indicate that the attenuated ΔihfABpmi exerts its antitumor activity by inducing macrophage infiltration or reprogramming the immunosuppressed tumor microenvironment to an activated state, suggesting that attenuated S. enterica Typhimurium strains based on nucleoid-associated protein genes deletion could be immunotherapeutic against cancer.


Sujet(s)
Salmonella typhimurium , Animaux , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Souris , Souris de lignée C57BL , Mélanome/immunologie , Mélanome/génétique , Mélanome/anatomopathologie , Immunothérapie/méthodes , Macrophages/immunologie , Macrophages/métabolisme , Lignée cellulaire tumorale , Mutation , Femelle , Mélanome expérimental/immunologie , Mélanome expérimental/anatomopathologie , Mélanome expérimental/thérapie , Modèles animaux de maladie humaine
3.
Front Immunol ; 15: 1376734, 2024.
Article de Anglais | MEDLINE | ID: mdl-38911854

RÉSUMÉ

Introduction: Non-typhoidal Salmonella (NTS) generally causes self-limiting gastroenteritis. However, older adults (≥65 years) can experience more severe outcomes from NTS infection. We have previously shown that a live attenuated S. Typhimurium vaccine, CVD 1926 (I77 ΔguaBA ΔclpP ΔpipA ΔhtrA), was immunogenic in adult but not aged mice. Here we describe modification of CVD 1926 through deletion of steD, a Salmonella effector responsible for host immune escape, which we hypothesized would increase immunogenicity in aged mice. Methods: Mel Juso and/or mutuDC cells were infected with S. Typhimurium I77, CVD 1926, and their respective steD mutants, and the MHC-II levels were evaluated. Aged (18-month-old) C57BL/6 mice received two doses of PBS, CVD 1926, or CVD 1926 ΔsteD perorally (109 CFU) and the number of FliC-specific CD4+ T cells were determined. Lastly, aged C57BL/6 mice received three doses of PBS, CVD 1926, or CVD 1926 ΔsteD perorally (109 CFU) and then were challenged perorally with wild-type S. Typhimurium SL1344 (108 CFU). These animals were also evaluated for antibody responses. Results: MHC-II induction was higher in cells treated with steD mutants, compared to their respective parental strains. Compared to PBS-vaccinated mice, CVD 1926 ΔsteD elicited significantly more FliC-specific CD4+ T cells in the Peyer's Patches. There were no significant differences in FliC-specific CD4+ T cells in the Peyer's patches or spleen of CVD 1926- versus PBS-immunized mice. CVD 1926 and CVD 1926 ΔsteD induced similar serum and fecal anti-core and O polysaccharide antibody titers after three doses. After two immunizations, the proportion of seroconverters for CVD 1926 ΔsteD was 83% (10/12) compared to 42% (5/12) for CVD 1926. Compared to PBS-immunized mice, mice immunized with CVD 1926 ΔsteD had significantly lower S. Typhimurium counts in the spleen, cecum, and small intestine upon challenge. In contrast, there were no differences in bacterial loads in the tissues of PBS-vaccinated and CVD 1926-immunized animals. Conclusion: These data suggest that the steD deletion enhanced the immunogenicity of our live attenuated S. Typhimurium vaccine. Deletion of immune evasion genes could be a potential strategy to improve the immunogenicity of live attenuated vaccines in older adults.


Sujet(s)
Anticorps antibactériens , Souris de lignée C57BL , Vaccins antisalmonella , Salmonella typhimurium , Vaccins atténués , Animaux , Vaccins antisalmonella/immunologie , Vaccins antisalmonella/administration et posologie , Vaccins antisalmonella/génétique , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Souris , Vaccins atténués/immunologie , Anticorps antibactériens/sang , Anticorps antibactériens/immunologie , Échappement immunitaire , Protéines bactériennes/immunologie , Protéines bactériennes/génétique , Femelle , Délétion de gène , Salmonelloses/immunologie , Salmonelloses/prévention et contrôle , Salmonelloses/microbiologie , Vieillissement/immunologie , Lymphocytes T CD4+/immunologie , Immunogénicité des vaccins
4.
Sci Rep ; 14(1): 14586, 2024 06 25.
Article de Anglais | MEDLINE | ID: mdl-38918457

RÉSUMÉ

Natural killer (NK) cells play a key role in defense against Salmonella infections during the early phase of infection. Our previous work showed that the excretory/secretory products of Ascaris suum repressed NK activity in vitro. Here, we asked if NK cell functionality was influenced in domestic pigs during coinfection with Ascaris and Salmonella enterica serotype Typhimurium. Ascaris coinfection completely abolished the IL-12 and IL-18 driven elevation of IFN-γ production seen in CD16 + CD8α + perforin + NK cells of Salmonella single-infected pigs. Furthermore, Ascaris coinfection prohibited the Salmonella-driven rise in NK perforin levels and CD107a surface expression. In line with impaired effector functions, NK cells from Ascaris-single and coinfected pigs displayed elevated expression of the inhibitory KLRA1 and NKG2A receptors genes, contrasting with the higher expression of the activating NKp46 and NKp30 receptors in NK cells during Salmonella single infection. These differences were accompanied by the highly significant upregulation of T-bet protein expression in NK cells from Ascaris-single and Ascaris/Salmonella coinfected pigs. Together, our data strongly indicate a profound repression of NK functionality by an Ascaris infection which may hinder infected individuals from adequately responding to a concurrent bacterial infection.


Sujet(s)
Ascaridiose , Co-infection , Cellules tueuses naturelles , Maladies des porcs , Animaux , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Ascaridiose/immunologie , Ascaridiose/médecine vétérinaire , Ascaridiose/parasitologie , Co-infection/immunologie , Co-infection/microbiologie , Co-infection/parasitologie , Suidae , Maladies des porcs/parasitologie , Maladies des porcs/immunologie , Maladies des porcs/microbiologie , Salmonelloses animales/immunologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Ascaris suum/immunologie , Interféron gamma/métabolisme , Perforine/métabolisme , Interleukine-12/métabolisme , Protéines à domaine boîte-T/métabolisme , Protéines à domaine boîte-T/génétique , Interleukine-18/métabolisme
5.
Front Immunol ; 15: 1396827, 2024.
Article de Anglais | MEDLINE | ID: mdl-38855102

RÉSUMÉ

Glucocorticoids, which have long served as fundamental therapeutics for diverse inflammatory conditions, are still widely used, despite associated side effects limiting their long-term use. Among their key mediators is glucocorticoid-induced leucine zipper (GILZ), recognized for its anti-inflammatory and immunosuppressive properties. Here, we explore the immunomodulatory effects of GILZ in macrophages through transcriptomic analysis and functional assays. Bulk RNA sequencing of GILZ knockout and GILZ-overexpressing macrophages revealed significant alterations in gene expression profiles, particularly impacting pathways associated with the inflammatory response, phagocytosis, cell death, mitochondrial function, and extracellular structure organization activity. GILZ-overexpression enhances phagocytic and antibacterial activity against Salmonella typhimurium and Escherichia coli, potentially mediated by increased nitric oxide production. In addition, GILZ protects macrophages from pyroptotic cell death, as indicated by a reduced production of reactive oxygen species (ROS) in GILZ transgenic macrophages. In contrast, GILZ KO macrophages produced more ROS, suggesting a regulatory role of GILZ in ROS-dependent pathways. Additionally, GILZ overexpression leads to decreased mitochondrial respiration and heightened matrix metalloproteinase activity, suggesting its involvement in tissue remodeling processes. These findings underscore the multifaceted role of GILZ in modulating macrophage functions and its potential as a therapeutic target for inflammatory disorders, offering insights into the development of novel therapeutic strategies aimed at optimizing the benefits of glucocorticoid therapy while minimizing adverse effects.


Sujet(s)
Macrophages , Mitochondries , Phagocytose , Pyroptose , Facteurs de transcription , Animaux , Mitochondries/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Immunomodulation , Espèces réactives de l'oxygène/métabolisme , Souris knockout , Glucocorticoïdes/pharmacologie , Souris de lignée C57BL , Salmonella typhimurium/immunologie , Escherichia coli/immunologie
6.
Vet Microbiol ; 295: 110151, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38870752

RÉSUMÉ

Porcine circovirus type 2 (PCV2) stands as a predominant etiological agent in porcine circovirus-associated diseases. To manage the spread of the disease, it is necessary to develop a next-generation vaccine expressing PCV2 antigens that target the prevailing genotype such as PCV2d. A bacterial-mediated vaccine delivery by live-attenuated Salmonella has attracted interest for its low-cost production and highly effective vaccine delivery. Thus, in this study, we utilized the advantages of the Salmonella-mediated vaccine delivery by cloning PCV2d cap and rep into a eukaryotic expression plasmid pJHL204 and electroporation into an engineered live-attenuated Salmonella Typhimurium JOL2500 (Δlon, ΔcpxR, ΔsifA, Δasd). The eukaryotic antigen expression by JOL2995 (p204:cap) and JOL2996 (p204:rep) was confirmed in vitro and in vivo which showed efficient antigen delivery. Furthermore, vaccination of mice model with the vaccine candidates elicited humoral and cell-mediated immune responses as depicted by high levels of PCV2-specific antibodies, CD4+ and CD8+ T cells, and neutralizing antibodies, especially by JOL2995 (p204:cap) which correlated with the significant decrease in the viral load in PCV2d-challenged mice. Interestingly, JOL2996 (p204:rep) may not have elicited high levels of neutralizing antibodies and protective efficacy, but it elicited considerably higher cell-mediated immune responses. This study demonstrated Salmonella-mediated vaccine delivery system coupled with the eukaryotic expression vector can efficiently deliver and express the target PCV2d antigens for strong induction of immune response and protective efficacy in mice model, further supporting the potential application of the Salmonella-mediated vaccine delivery system as an effective novel approach in vaccine strategies for PCV2d.


Sujet(s)
Infections à Circoviridae , Circovirus , Vecteurs génétiques , Salmonella typhimurium , Vaccins antiviraux , Animaux , Circovirus/immunologie , Circovirus/génétique , Souris , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Infections à Circoviridae/prévention et contrôle , Infections à Circoviridae/médecine vétérinaire , Infections à Circoviridae/immunologie , Suidae , Antigènes viraux/immunologie , Antigènes viraux/génétique , Souris de lignée BALB C , Anticorps antiviraux/sang , Femelle , Anticorps neutralisants/sang , Protéines de capside/immunologie , Protéines de capside/génétique , Maladies des porcs/prévention et contrôle , Maladies des porcs/immunologie , Maladies des porcs/virologie
7.
Virulence ; 15(1): 2367783, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38937901

RÉSUMÉ

Helicobacter pylori causes globally prevalent infections that are highly related to chronic gastritis and even development of gastric carcinomas. With the increase of antibiotic resistance, scientists have begun to search for better vaccine design strategies to eradicate H. pylori colonization. However, while current strategies prefer to formulate vaccines with a single H. pylori antigen, their potential has not yet been fully realized. Outer membrane vesicles (OMVs) are a potential platform since they could deliver multiple antigens. In this study, we engineered three crucial H. pylori antigen proteins (UreB, CagA, and VacA) onto the surface of OMVs derived from Salmonella enterica serovar Typhimurium (S. Typhimurium) mutant strains using the hemoglobin protease (Hbp) autotransporter system. In various knockout strategies, we found that OMVs isolated from the ΔrfbP ΔfliC ΔfljB ΔompA mutants could cause distinct increases in immunoglobulin G (IgG) and A (IgA) levels and effectively trigger T helper 1- and 17-biased cellular immune responses, which perform a vital role in protecting against H. pylori. Next, OMVs derived from ΔrfbP ΔfliC ΔfljB ΔompA mutants were used as a vector to deliver different combinations of H. pylori antigens. The antibody and cytokine levels and challenge experiments in mice model indicated that co-delivering UreB and CagA could protect against H. pylori and antigen-specific T cell responses. In summary, OMVs derived from the S. Typhimurium ΔrfbP ΔfliC ΔfljB ΔompA mutant strain as the vector while importing H. pylori UreB and CagA as antigenic proteins using the Hbp autotransporter system would greatly benefit controlling H. pylori infection.


Outer membrane vesicles (OMVs), as a novel antigen delivery platform, has been used in vaccine design for various pathogens and even tumors. Salmonella enterica serovar Typhimurium (S. Typhimurium), as a bacterium that is easy to engineer and has both adjuvant efficacy and immune stimulation capacity, has become the preferred bacterial vector for purifying OMVs after Escherichia coli. This study focuses on the design of Helicobacter pylori ;(H. pylori) vaccines, utilizing genetically modified Salmonella OMVs to present several major antigens of H. pylori, including UreB, VacA and CagA. The optimal Salmonella OMV delivery vector and antigen combinations are screened and identified, providing new ideas for the development of H. pylori vaccines and an integrated antigen delivery platform for other difficult to develop vaccines for bacteria, viruses, and even tumors.


Sujet(s)
Antigènes bactériens , Protéines bactériennes , Infections à Helicobacter , Helicobacter pylori , Salmonella typhimurium , Animaux , Infections à Helicobacter/prévention et contrôle , Infections à Helicobacter/immunologie , Infections à Helicobacter/microbiologie , Protéines bactériennes/génétique , Protéines bactériennes/immunologie , Helicobacter pylori/immunologie , Helicobacter pylori/génétique , Souris , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Vaccins antibactériens/immunologie , Vaccins antibactériens/génétique , Femelle , Anticorps antibactériens/immunologie , Anticorps antibactériens/sang , Immunoglobuline G , Génie génétique , Urease/immunologie , Urease/génétique , Modèles animaux de maladie humaine
8.
Vet Microbiol ; 294: 110131, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38805917

RÉSUMÉ

Outer membrane vesicles (OMVs) are membranous structures frequently observed in Gram-negative bacteria that contain bioactive substances. These vesicles are rich in bacterial antigens that can activate the host's immune system, making them a promising candidate vaccine to prevent and manage bacterial infections. The aim of this study was to assess the immunogenicity and protective efficacy of OMVs derived from Salmonella enterica serovar Typhimurium and S. Choleraesuis, while also focusing on enhancing OMV production. Initial experiments showed that OMVs from wild-type strains did not provide complete protection against homologous Salmonella challenge, possible due to the presence of flagella in the purified OMVs samples, which may elicit an unnecessary immune response. To address this, flagellin-deficient mutants of S. Typhimurium and S. Choleraesuis were constructed, designated rSC0196 and rSC0199, respectively. These mutants exhibited reduced cell motility and their OMVs were found to be flagellin-free. Immunization with non-flagellin OMVs derived from rSC0196 induced robust antibody responses and improved survival rates in mice, as compared to the OMVs derived from the wild-type UK-1. In order to enhance OMV production, deletions of ompA or tolR were introduced into rSC0196. The deletion of tolR not only increase the yield of OMVs, but also conferred complete protection against homologous S. Typhimurium challenge in mice. Collectively, these findings indicate that the flagellin-deficient OMVs with a tolR mutation have the potential to serve as a versatile vaccine platform, capable of inducing broad-spectrum protection against significant pathogens.


Sujet(s)
Protéines de la membrane externe bactérienne , Souris de lignée BALB C , Vaccins antisalmonella , Salmonella typhimurium , Animaux , Salmonella typhimurium/immunologie , Salmonella typhimurium/génétique , Souris , Vaccins antisalmonella/immunologie , Protéines de la membrane externe bactérienne/immunologie , Protéines de la membrane externe bactérienne/génétique , Femelle , Flagelline/immunologie , Flagelline/génétique , Salmonelloses animales/prévention et contrôle , Salmonelloses animales/microbiologie , Salmonelloses animales/immunologie , Anticorps antibactériens/sang , Anticorps antibactériens/immunologie , Membrane bactérienne externe/immunologie , Salmonella/immunologie , Salmonella/génétique , Immunogénicité des vaccins , Antigènes bactériens/immunologie
9.
Nat Commun ; 15(1): 4241, 2024 May 18.
Article de Anglais | MEDLINE | ID: mdl-38762500

RÉSUMÉ

Glioblastoma multiforme (GBM) is a highly aggressive brain tumor characterized by invasive behavior and a compromised immune response, presenting treatment challenges. Surgical debulking of GBM fails to address its highly infiltrative nature, leaving neoplastic satellites in an environment characterized by impaired immune surveillance, ultimately paving the way for tumor recurrence. Tracking and eradicating residual GBM cells by boosting antitumor immunity is critical for preventing postoperative relapse, but effective immunotherapeutic strategies remain elusive. Here, we report a cavity-injectable bacterium-hydrogel superstructure that targets GBM satellites around the cavity, triggers GBM pyroptosis, and initiates innate and adaptive immune responses, which prevent postoperative GBM relapse in male mice. The immunostimulatory Salmonella delivery vehicles (SDVs) engineered from attenuated Salmonella typhimurium (VNP20009) seek and attack GBM cells. Salmonella lysis-inducing nanocapsules (SLINs), designed to trigger autolysis, are tethered to the SDVs, eliciting antitumor immune response through the intracellular release of bacterial components. Furthermore, SDVs and SLINs administration via intracavitary injection of the ATP-responsive hydrogel can recruit phagocytes and promote antigen presentation, initiating an adaptive immune response. Therefore, our work offers a local bacteriotherapy for stimulating anti-GBM immunity, with potential applicability for patients facing malignancies at a high risk of recurrence.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Récidive tumorale locale , Salmonella typhimurium , Glioblastome/thérapie , Glioblastome/immunologie , Animaux , Souris , Salmonella typhimurium/immunologie , Mâle , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/immunologie , Tumeurs du cerveau/immunologie , Tumeurs du cerveau/thérapie , Humains , Lignée cellulaire tumorale , Souris de lignée C57BL , Pyroptose , Immunité acquise , Immunité innée , Hydrogels/composition chimique , Immunothérapie/méthodes
10.
Biochemistry (Mosc) ; 89(3): 574-582, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38648774

RÉSUMÉ

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.


Sujet(s)
Adjuvants immunologiques , Flagelline , Vaccins antirabiques , Rage (maladie) , Vaccins inactivés , Vaccins antirabiques/immunologie , Vaccins antirabiques/administration et posologie , Animaux , Flagelline/immunologie , Souris , Rage (maladie)/prévention et contrôle , Rage (maladie)/immunologie , Vaccins inactivés/immunologie , Chiens , Virus de la rage/immunologie , Salmonella typhimurium/immunologie , Femelle , Chats
11.
Vaccine ; 42(15): 3445-3454, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38631956

RÉSUMÉ

Major histocompatibility complex class II (MHC-II) molecules are involved in immune responses against pathogens and vaccine candidates' immunogenicity. Immunopeptidomics for identifying cancer and infection-related antigens and epitopes have benefited from advances in immunopurification methods and mass spectrometry analysis. The mouse anti-MHC-II-DR monoclonal antibody L243 (mAb-L243) has been effective in recognising MHC-II-DR in both human and non-human primates. It has also been shown to cross-react with other animal species, although it has not been tested in livestock. This study used mAb-L243 to identify Staphylococcus aureus and Salmonella enterica serovar Typhimurium peptides binding to cattle and swine macrophage MHC-II-DR molecules using flow cytometry, mass spectrometry and two immunopurification techniques. Antibody cross-reactivity led to identifying expressed MHC-II-DR molecules, together with 10 Staphylococcus aureus peptides in cattle and 13 S. enterica serovar Typhimurium peptides in swine. Such data demonstrates that MHC-II-DR expression and immunocapture approaches using L243 mAb represents a viable strategy for flow cytometry and immunopeptidomics analysis of bovine and swine antigen-presenting cells.


Sujet(s)
Anticorps monoclonaux , Macrophages , Salmonella typhimurium , Staphylococcus aureus , Animaux , Bovins , Suidae/immunologie , Staphylococcus aureus/immunologie , Anticorps monoclonaux/immunologie , Macrophages/immunologie , Salmonella typhimurium/immunologie , Antigènes d'histocompatibilité de classe II/immunologie , Réactions croisées/immunologie , Cytométrie en flux , Spectrométrie de masse , Souris
12.
Poult Sci ; 103(5): 103569, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38447310

RÉSUMÉ

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.


Sujet(s)
Poulets , Chitosane , Protection croisée , Nanoparticules , Maladies de la volaille , Salmonelloses animales , Vaccins antisalmonella , Salmonella enteritidis , Salmonella typhimurium , Animaux , Poulets/immunologie , Salmonella enteritidis/immunologie , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/immunologie , Salmonelloses animales/prévention et contrôle , Salmonelloses animales/immunologie , Chitosane/administration et posologie , Chitosane/pharmacologie , Vaccins antisalmonella/immunologie , Vaccins antisalmonella/administration et posologie , Nanoparticules/administration et posologie , Salmonella typhimurium/immunologie , Administration par voie orale , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/immunologie
13.
Microb Pathog ; 175: 105959, 2023 Feb.
Article de Anglais | MEDLINE | ID: mdl-36581307

RÉSUMÉ

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.


Sujet(s)
Protéines Escherichia coli , Vaccins antisalmonella , Salmonella typhimurium , Animaux , Souris , Anticorps antibactériens/immunologie , Protéines de la membrane externe bactérienne/immunologie , Escherichia coli/génétique , Immunisation , Souris de lignée BALB C , Protéines de fusion recombinantes/génétique , Protéines recombinantes/génétique , Salmonella typhimurium/génétique , Salmonella typhimurium/immunologie , Vaccins atténués , Vaccins synthétiques
14.
Nature ; 609(7926): 348-353, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35978195

RÉSUMÉ

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.


Sujet(s)
Inflammation , Protéines et peptides de signalisation intracellulaire , Macrophages , Nitric oxide synthase type II , Animaux , Arthrite/immunologie , Arthrite/métabolisme , Citrulline/métabolisme , Cyanates/métabolisme , Humains , Inflammation/enzymologie , Inflammation/immunologie , Inflammation/métabolisme , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Macrophages/immunologie , Macrophages/métabolisme , Souris , Nitric oxide synthase type II/métabolisme , Ornithine/métabolisme , Ornithine decarboxylase/métabolisme , Polyamines/métabolisme , Salmonella typhimurium/immunologie
15.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-35163280

RÉSUMÉ

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.


Sujet(s)
Salmonelloses/immunologie , Salmonella typhimurium/immunologie , Analyse spectrale Raman/méthodes , Antibactériens/pharmacologie , Bactéries/effets des médicaments et des substances chimiques , Résistance bactérienne aux médicaments/génétique , Résistance microbienne aux médicaments/effets des médicaments et des substances chimiques , Humains , Salmonelloses/diagnostic , Salmonella typhimurium/effets des médicaments et des substances chimiques , Salmonella typhimurium/pathogénicité
16.
PLoS Pathog ; 18(1): e1009718, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-35073381

RÉSUMÉ

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.


Sujet(s)
Inflammasomes/immunologie , Macrophages/immunologie , Macrophages/microbiologie , Salmonelloses/immunologie , Systèmes de sécrétion de type III/immunologie , Protéines adaptatrices de signalisation CARD/immunologie , Protéines de liaison au calcium/immunologie , Humains , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Protéine inhibitrice de l'apoptose neuronale/immunologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Virulence
17.
J Biol Chem ; 298(1): 101461, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34864057

RÉSUMÉ

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.


Sujet(s)
Inflammasomes , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Salmonelloses , Salmonella typhimurium , Animaux , Caspase-1/génétique , Caspase-1/métabolisme , Interactions hôte-pathogène/immunologie , Inflammasomes/immunologie , Interleukine-1 bêta/génétique , Interleukine-1 bêta/immunologie , Macrophages/immunologie , Souris , Souris de lignée C57BL , Protéine-3 de la famille des NLR contenant un domaine pyrine/génétique , Protéine-3 de la famille des NLR contenant un domaine pyrine/immunologie , Salmonelloses/immunologie , Salmonelloses/microbiologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/isolement et purification
18.
Gut Microbes ; 13(1): 1991776, 2021.
Article de Anglais | MEDLINE | ID: mdl-34719317

RÉSUMÉ

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.


Sujet(s)
Protéines adaptatrices de la transduction du signal/immunologie , Protéines bactériennes/immunologie , Salmonelloses/immunologie , Salmonella typhimurium/immunologie , Protéines adaptatrices de la transduction du signal/génétique , Animaux , Protéines bactériennes/génétique , Microbiome gastro-intestinal , Interactions hôte-pathogène , Humains , Immunité innée , Interleukine-6/génétique , Interleukine-6/immunologie , Souris , Salmonelloses/génétique , Salmonelloses/microbiologie , Salmonella typhimurium/génétique , Facteur de nécrose tumorale alpha/génétique , Facteur de nécrose tumorale alpha/immunologie
19.
Front Immunol ; 12: 729607, 2021.
Article de Anglais | MEDLINE | ID: mdl-34804014

RÉSUMÉ

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.


Sujet(s)
Lymphocytes B/métabolisme , Centre germinatif/métabolisme , Interleukine-4/métabolisme , Muqueuse intestinale/métabolisme , Lymphocytes intra-épithéliaux/métabolisme , Plaques de Peyer/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Animaux , Lymphocytes B/immunologie , Lymphocytes B/microbiologie , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Modèles animaux de maladie humaine , Centre germinatif/immunologie , Centre germinatif/microbiologie , Immunité muqueuse , Immunoglobuline A/immunologie , Immunoglobuline A/métabolisme , Commutation de classe des immunoglobulines , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Lymphocytes intra-épithéliaux/immunologie , Lymphocytes intra-épithéliaux/microbiologie , Activation des lymphocytes , Déplétion lymphocytaire , Souris knockout , Plaques de Peyer/immunologie , Plaques de Peyer/microbiologie , Phénotype , Récepteur lymphocytaire T antigène, gamma-delta/génétique , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Salmonelloses/immunologie , Salmonelloses/métabolisme , Salmonelloses/microbiologie , Salmonella typhimurium/immunologie , Salmonella typhimurium/pathogénicité , Transduction du signal
20.
PLoS Pathog ; 17(10): e1010004, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34695149

RÉSUMÉ

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.


Sujet(s)
Lymphocytes T CD4+/immunologie , Mémoire immunologique/immunologie , Foie/immunologie , Salmonelloses animales/immunologie , Animaux , Femelle , Mâle , Souris , Souris de lignée C57BL , Salmonella typhimurium/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE