Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.170
1.
Sci Rep ; 14(1): 11053, 2024 05 14.
Article En | MEDLINE | ID: mdl-38744900

This study investigated the influence of polyunsaturated fatty acid composition and vitamin E supplementation on oxidative status and immune responses in weanling piglets pre- and post-E. coli challenge. Suckling piglets (n = 24) were randomly selected from two litters for an oral supplementation (1 mL/day) with fish oil or hemp oil and vitamin E supplementation (60 mg natural vitamin E/mL oil) from day 10 to 28 of age. At day 29 and 30 of age, each piglet was orally inoculated with 6.7 × 108 and 3.96 × 108 CFU of F4 and F18 E. coli, respectively. Blood was sampled from all piglets on day 28 before E. coli challenge and on day 35 of age to investigate immunological and oxidative stress markers in plasma. One week after weaning and exposure to E. coli, a general reduction in the α-tocopherol concentration and activity of GPX1 was obtained. Vitamin E supplementation lowered the extent of lipid peroxidation and improved the antioxidative status and immune responses after E. coli challenge. Hemp oil had the greatest effect on antioxidant enzyme activity. Provision of hemp oil and vitamin E to suckling piglets may reduce the incidence of post-weaning diarrhea.


Cannabis , Dietary Supplements , Escherichia coli Infections , Escherichia coli , Fish Oils , Oxidation-Reduction , Vitamin E , Animals , Vitamin E/pharmacology , Swine , Fish Oils/pharmacology , Fish Oils/administration & dosage , Cannabis/chemistry , Oxidation-Reduction/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/veterinary , Escherichia coli Infections/drug therapy , Escherichia coli Infections/prevention & control , Antioxidants/pharmacology , Antioxidants/metabolism , Oxidative Stress/drug effects , Weaning , Lipid Peroxidation/drug effects , Swine Diseases/microbiology , Swine Diseases/immunology , Swine Diseases/prevention & control , Swine Diseases/drug therapy
2.
Arch Microbiol ; 206(6): 249, 2024 May 07.
Article En | MEDLINE | ID: mdl-38713385

Escherichia coli (E. coli) can induce severe clinical bovine mastitis, which is to blame for large losses experienced by dairy farms. Macrophage polarization into various states is in response to pathogen infections. Lycopene, a naturally occurring hydrocarbon carotenoid, relieved inflammation by controlling M1/M2 status of macrophages. Thus, we wanted to explore the effect of lycopene on polarization states of macrophages in E. coli-induced mastitis. Macrophages were cultivated with lycopene for 24, before E. coli inoculation for 6 h. Lycopene (0.5 µmol/L) significantly enhanced cell viabilities and significantly reduced lactic dehydrogenase (LDH) levels in macrophages, whereas 2 and 3 µmol/L lycopene significantly enhanced LDH activities. Lycopene treatment significantly reduced the increase in LDH release, iNOS, CD86, TNF-α, IL-1ß and phosphatase and tensin homolog (PTEN) expressions in E. coli group. 0.5 µmol/L lycopene significantly increased E. coli-induced downregulation of CD206, arginase I (ARG1), indoleamine 2,3-dioxygenase (IDO), chitinase 3-like 3 (YM1), PI3K, AKT, p-AKT, mammalian target of rapamycin (mTOR), p-mTOR, jumonji domain-containing protein-3 (JMJD3) and interferon regulatory factor 4 (IRF4) levels. Moreover, Ginkgolic acid C17:1 (a specific PTEN inhibitor), 740YPDGFR (a specific PI3K activator), SC79 (a specific AKT activator) or CHPG sodium salt (a specific NF-κB activator) significantly decreased CD206, AGR1, IDO and YM1 expressions in lycopene and E. coli-treated macrophages. Therefore, lycopene increased M2 macrophages via inhibiting NOTCH1-PI3K-mTOR-NF-κB-JMJD3-IRF4 pathway in response to E. coli infection in macrophages. These results contribute to revealing the pathogenesis of E. coli-caused bovine mastitis, providing the new angle of the prevention and management of mastitis.


Escherichia coli Infections , Escherichia coli , Interferon Regulatory Factors , Lycopene , Macrophages , NF-kappa B , Phosphatidylinositol 3-Kinases , Receptor, Notch1 , Signal Transduction , TOR Serine-Threonine Kinases , Lycopene/pharmacology , Animals , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , NF-kappa B/metabolism , TOR Serine-Threonine Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Macrophages/drug effects , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Signal Transduction/drug effects , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Interferon Regulatory Factors/metabolism , Interferon Regulatory Factors/genetics , Mice , Cattle , Cell Line , Female , Mastitis, Bovine/microbiology
3.
Front Immunol ; 15: 1368099, 2024.
Article En | MEDLINE | ID: mdl-38665923

Early increase in the level of endothelial progenitor cells (EPCs) in the systemic circulation occurs in patients with septic infection/sepsis. The significance and underlying mechanisms of this response remain unclear. This study investigated the bone marrow EPC response in adult mice with septic infection induced by intravenous injection (i.v.) of Escherichia coli. For in vitro experiments, sorted marrow stem/progenitor cells (SPCs) including lineage(lin)-stem cell factor receptor (c-kit)+stem cell antigen-1 (Sca-1)-, lin-c-kit+, and lin- cells were cultured with or without lipopolysaccharides (LPSs) and recombinant murine vascular endothelial growth factor (VEGF) in the absence and presence of anti-Sca-1 crosslinking antibodies. In a separate set of experiments, marrow lin-c-kit+ cells from green fluorescence protein (GFP)+ mice, i.v. challenged with heat-inactivated E. coli or saline for 24 h, were subcutaneously implanted in Matrigel plugs for 5 weeks. Marrow lin-c-kit+ cells from Sca-1 knockout (KO) mice challenged with heat-inactivated E. coli for 24 h were cultured in the Matrigel medium for 8 weeks. The marrow pool of EPCs bearing the lin-c-kit+Sca-1+VEGF receptor 2 (VEGFR2)+ (LKS VEGFR2+) and LKS CD133+VEGFR2+ surface markers expanded rapidly following septic infection, which was supported by both proliferative activation and phenotypic conversion of marrow stem/progenitor cells. Increase in marrow EPCs and their reprogramming for enhancing angiogenic activity correlated with cell-marked upregulation of Sca-1 expression. Sca-1 was coupled with Ras-related C3 botulinum toxin substrate 2 (Rac2) in signaling the marrow EPC response. Septic infection caused a substantial increase in plasma levels of IFN-γ, VEGF, G-CSF, and SDF-1. The early increase in circulating EPCs was accompanied by their active homing and incorporation into pulmonary microvasculature. These results demonstrate that the marrow EPC response is a critical component of the host defense system. Sca-1 signaling plays a pivotal role in the regulation of EPC response in mice with septic infection.


Endothelial Progenitor Cells , Membrane Proteins , Sepsis , Animals , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/immunology , Sepsis/immunology , Sepsis/metabolism , Mice , Mice, Knockout , Escherichia coli/immunology , Escherichia coli Infections/immunology , Mice, Inbred C57BL , Vascular Endothelial Growth Factor A/metabolism , Antigens, Ly/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/immunology , Cells, Cultured , Male
4.
Theranostics ; 14(6): 2589-2604, 2024.
Article En | MEDLINE | ID: mdl-38646647

Background: The mechanisms underlying the increased mortality of secondary infections during the immunosuppressive phase of sepsis remain elusive. Objectives: We sought to investigate the role of Siglec-F+ neutrophils on splenic T lymphocytes in the immunosuppressed phase of sepsis and on secondary infection in PICS mice, and to elucidate the underlying mechanisms. Methods: We established a mouse model of sepsis-induced immunosuppression followed by secondary infection with LPS or E. coli. The main manifestation of immunosuppression is the functional exhaustion of splenic T lymphocytes. Treg depletion reagent Anti-IL-2, IL-10 blocker Anti-IL-10R, macrophage depletion reagent Liposomes, neutrophil depletion reagent Anti-Ly6G, neutrophil migration inhibitor SB225002, Siglec-F depletion reagent Anti-Siglec-F are all used on PICS mice. The function of neutrophil subsets was investigated by adoptive transplantation and the experiments in vitro. Results: Compared to other organs, we observed a significant reduction in pro-inflammatory cytokines in the spleen, accompanied by a marked increase in IL-10 production, primarily by infiltrating neutrophils. These infiltrating neutrophils in the spleen during the immunosuppressive phase of sepsis undergo phenotypic change in the local microenvironment, exhibiting high expression of neutrophil biomarkers such as Siglec-F, Ly6G, and Siglec-E. Depletion of neutrophils or specifically targeting Siglec-F leads to enhance the function of T lymphocytes and a notable improvement in the survival of mice with secondary infections. Conclusions: We identified Siglec-F+ neutrophils as the primary producers of IL-10, which significantly contributed to T lymphocyte suppression represents a novel finding with potential therapeutic implications.


Neutrophils , Sepsis , Sialic Acid Binding Immunoglobulin-like Lectins , Spleen , Animals , Male , Mice , Cytokines/metabolism , Disease Models, Animal , Escherichia coli Infections/immunology , Immunosuppression Therapy , Interleukin-10/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Sepsis/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
5.
Infect Immun ; 92(5): e0044023, 2024 May 07.
Article En | MEDLINE | ID: mdl-38591882

Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.


Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli Vaccines , Extraintestinal Pathogenic Escherichia coli , Hemolysin Proteins , Animals , Extraintestinal Pathogenic Escherichia coli/genetics , Extraintestinal Pathogenic Escherichia coli/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Mice , Hemolysin Proteins/immunology , Hemolysin Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/immunology , Escherichia coli Vaccines/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Female , Virulence Factors/genetics , Virulence Factors/immunology , Type V Secretion Systems/immunology , Type V Secretion Systems/genetics , Disease Models, Animal , Humans
6.
Cell Rep ; 43(4): 114004, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38522070

During infections, host cells are exposed to pathogen-associated molecular patterns (PAMPs) and virulence factors that stimulate multiple signaling pathways that interact additively, synergistically, or antagonistically. The net effect of such higher-order interactions is a vital determinant of the outcome of host-pathogen interactions. Here, we demonstrate one such complex interplay between bacterial exotoxin- and PAMP-induced innate immune pathways. We show that two caspases activated during enterohemorrhagic Escherichia coli (EHEC) infection by lipopolysaccharide (LPS) and Shiga toxin (Stx) interact in a functionally antagonistic manner; cytosolic LPS-activated caspase-11 cleaves full-length gasdermin D (GSDMD), generating an active pore-forming N-terminal fragment (NT-GSDMD); subsequently, caspase-3 activated by EHEC Stx cleaves the caspase-11-generated NT-GSDMD to render it nonfunctional, thereby inhibiting pyroptosis and interleukin-1ß maturation. Bacteria typically subvert inflammasomes by targeting upstream components such as NLR sensors or full-length GSDMD but not active NT-GSDMD. Thus, our findings uncover a distinct immune evasion strategy where a bacterial toxin disables active NT-GSDMD by co-opting caspase-3.


Caspase 3 , Gasdermins , Intracellular Signaling Peptides and Proteins , Macrophages , Phosphate-Binding Proteins , Pyroptosis , Pyroptosis/drug effects , Phosphate-Binding Proteins/metabolism , Macrophages/metabolism , Macrophages/microbiology , Intracellular Signaling Peptides and Proteins/metabolism , Caspase 3/metabolism , Humans , Animals , Mice , Apoptosis Regulatory Proteins/metabolism , Bacterial Toxins/metabolism , Caspases/metabolism , Lipopolysaccharides/pharmacology , Enterohemorrhagic Escherichia coli/metabolism , Enterohemorrhagic Escherichia coli/pathogenicity , Caspases, Initiator/metabolism , Inflammasomes/metabolism , Mice, Inbred C57BL , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Interleukin-1beta/metabolism
7.
Acta Trop ; 254: 107173, 2024 Jun.
Article En | MEDLINE | ID: mdl-38503364

BACKGROUND: Neonatal and post-weaning diarrhea is a concern disease caused by enterotoxigenic Escherichia coli fimbriae F4 (F4+ETEC) in pig farms. Diarrhea outbreaks are often severe and costly due to the high prevalence and spread of the disease within the same herd. Vaccine is one of strategic solution in protecting pig against F4+ETEC infection in particular pig farm. In present study, we conducted two trials of vaccination with crude F4 fimbriae extract vaccine in pregnant sow and nursery pigs. METHODS: In experiment 1 (20 sows; non-vaccinated control, n=10), we vaccinated pregnant sows (n=10) twice at 4 wk and 2 wk before farrowing and evaluated impact of vaccination on maternal immunity. The sow serum and colostrum were collected before vaccination, 2 and 4 weeks after vaccination, 6 hours after farrowing, respectively, and the piglet's serum from both groups (2 piglet/sow, 10 piglets from each group) were also collected on 3 days old to measure F4 specific IgG, F4 specific IgA using in house ELISA kit. In experiment 2, to optimize doses and dosage of candidate vaccine in piglets, 18 piglets (3 piglets/group) were allocated into five immunized groups and one control group (unimmunized group), we immunized piglets twice at 4 and 6 weeks old with difference doses (i.e., 0, 50, 100, 150, 200 µg), and for a dose 150 µg, we immunized with two dosages at 1 ml and 2 ml. Piglets were challenged with a 3 ml dose of 3 × 109 CFU/ml bacterial culture of enterotoxigenic Escherichia coli (F4+ETEC) in order to evaluate the efficacy of vaccine. After challenging, the clinical sign of the piglets was daily observed and the rectal swab was performed every day for investigation of the fecal shedding of Escherichia coli (F4+ETEC) by using PCR technique. Serum were collected before, 2 and 4 weeks after vaccination and 1 week after challenge to measure F4 specific IgG, F4 specific IgA using in house ELISA kit and cytokines levels (i.e., IL-1 beta, IL-6, IL-8 and TNF alpha) before and 1 week after challenge using commercial ELISA kit. RESULTS: The levels of antibody results showed that in experiment 1, the anti-F4 antibody levels both F4 specific IgG and F4 specific IgA in serum and colostrum of vaccinated sow increased significantly after vaccination. The piglets of immunized sows have antibody level both F4 specific IgG and F4 specific IgA in their serum higher than those piglets of unimmunized sows significantly (p < 0.01). In experiment 2, irrespective of different doses and dosage, there is no difference in term of F4 specific IgG and F4 specific IgA levels among immunized groups. However, all of vaccinated piglets showed F4 specific IgG and F4 specific IgA levels higher and the elimination of Escherichia coli (F4+ETEC) in feces post challenge faster (< 3 days) than unvaccinated group (> 5 days). For cytokines levels, a higher level of IL-1 beta, IL-6, IL-8 and TNF alpha at 1 week after challenge in vaccinated groups was found when compared with the levels in non-vaccinated group. CONCLUSIONS: Our results suggest that crude F4 fimbriae extract autogenous vaccine is a candidate vaccine for protecting piglets against diarrhea disease caused by enterotoxigenic Escherichia coli (F4+ETEC) and vaccination the pregnant sow twice before farrowing is one of strategies to provide maternal derived antibody to the newborn piglets for against enterotoxigenic Escherichia coli (F4+ETEC) during early life.


Antibodies, Bacterial , Enterotoxigenic Escherichia coli , Escherichia coli Infections , Escherichia coli Vaccines , Swine Diseases , Animals , Swine , Female , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Escherichia coli Infections/immunology , Swine Diseases/prevention & control , Swine Diseases/immunology , Swine Diseases/microbiology , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Vaccines/immunology , Escherichia coli Vaccines/administration & dosage , Pregnancy , Antibodies, Bacterial/blood , Colostrum/immunology , Immunoglobulin A/blood , Vaccination/veterinary , Immunoglobulin G/blood , Fimbriae, Bacterial/immunology , Diarrhea/prevention & control , Diarrhea/veterinary , Diarrhea/microbiology , Diarrhea/immunology , Animals, Newborn/immunology , Immunity, Maternally-Acquired
8.
Front Immunol ; 14: 1185232, 2023.
Article En | MEDLINE | ID: mdl-37261344

The present study investigated the expression of cytokines and cellular changes in chickens following vaccination with irradiated avian pathogenic Escherichia coli (APEC) and/or challenge. Four groups of 11-week-old pullets, each consisting of 16 birds were kept separately in isolators before they were sham inoculated (N), challenged only (C), vaccinated (V) or vaccinated and challenged (V+C). Vaccination was performed using irradiated APEC applied via aerosol. For challenge, the homologous strain was administered intratracheally. Birds were sacrificed on 3, 7, 14 and 21 days post challenge (dpc) to examine lesions, organ to body weight ratios and bacterial colonization. Lung and spleen were sampled for investigating gene expression of cytokines mediating inflammation by RT-qPCR and changes in the phenotype of subsets of mononuclear cells by flow cytometry. After re-stimulation of immune cells by co-cultivation with the pathogen, APEC-specific IFN-γ producing cells were determined. Challenged only birds showed more severe pathological and histopathological lesions, a higher probability of bacterial re-isolation and higher organ to body weight ratios compared to vaccinated and challenged birds. In the lung, an upregulation of IL-1ß and IL-6 following vaccination and/or challenge at 3 dpc was observed, whereas in the spleen IL-1ß was elevated. Changes were observed in macrophages and TCR-γδ+ cells within 7 dpc in spleen and lung of challenged birds. Furthermore, an increase of CD4+ cells in spleen and a rise of Bu-1+ cells in lung were present in vaccinated and challenged birds at 3 dpc. APEC re-stimulated lung and spleen mononuclear cells from only challenged pullets showed a significant increase of IFN-γ+CD8α+ and IFN-γ+TCR-γδ+ cells. Vaccinated and challenged chickens responded with a significant increase of IFN-γ+CD8α+ T cells in the lung and IFN-γ+TCR-γδ+ cells in the spleen. Re-stimulation of lung mononuclear cells from vaccinated birds resulted in a significant increase of both IFN-γ+CD8α+ and IFN-γ+TCR-γδ+ cells. In conclusion, vaccination with irradiated APEC caused enhanced pro-inflammatory response as well as the production of APEC-specific IFN-γ-producing γδ and CD8α T cells, which underlines the immunostimulatory effect of the vaccine in the lung. Hence, our study provides insights into the underlying immune mechanisms that account for the defense against APEC.


Escherichia coli Infections , Escherichia coli Vaccines , Animals , Chickens , Female , Escherichia coli Vaccines/administration & dosage , Escherichia coli Vaccines/immunology , Escherichia coli Infections/immunology , Escherichia coli Infections/prevention & control , Escherichia coli Infections/veterinary , Poultry Diseases/immunology , Poultry Diseases/prevention & control , Aerosols
9.
Am J Hum Biol ; 35(8): e23897, 2023 08.
Article En | MEDLINE | ID: mdl-36951242

INTRODUCTION: Multiple studies have reported that milk immune content increases for infants experiencing infectious disease (ID) episodes, suggesting that the immune system of milk (ISOM) offers enhanced protection when needed to combat ID. METHODS: To test the hypothesis that ISOM content and/or activity increases during an infant's ID episode, we characterized milk secretory immunoglobulin A (sIgA; a major ISOM constituent) and in vitro interleukin-6 (IL-6) responses to Salmonella enterica and Escherichia coli, as system-level biomarkers of ISOM activity, in a prospective study among 96 mother-infant dyads in Kilimanjaro, Tanzania. RESULTS: After control for covariates, no milk immune variables (sIgA, Coef: 0.03; 95% CI -0.25, 0.32; in vitro IL-6 response to S. enterica, Coef: 0.23; 95% CI: -0.67, 1.13; IL-6 response to E. coli, Coef: -0.11; 95% CI: -0.98, 0.77) were associated with prevalent ID (diagnosed at the initial participation visit). Among infants experiencing an incident ID (diagnosed subsequent to the initial participation), milk immune content and responses were not substantially higher or lower than the initial visit (sIgA, N: 61; p: 0.788; IL-6 response to S. enterica, N: 56; p: 0.896; IL-6 response to E. coli, N: 36; p: 0.683); this was unchanged by exclusion of infants with ID at the time of initial participation. CONCLUSION: These findings are not consistent with the hypothesis that milk delivers enhanced immune protection when infants experience ID. In environments with a high burden of ID, dynamism may be less valuable to maternal reproductive success than stability in the ISOM.


Escherichia coli Infections , Escherichia coli , Immunoglobulin A, Secretory , Interleukin-6 , Milk, Human , Salmonella Infections , Salmonella enterica , Humans , Female , Milk, Human/chemistry , Interleukin-6/analysis , Interleukin-6/immunology , Salmonella enterica/physiology , Salmonella Infections/immunology , Escherichia coli/physiology , Escherichia coli Infections/immunology , Infant, Newborn , Infant , Tanzania , Prospective Studies , Adult , Cross-Sectional Studies , Immunoenzyme Techniques , Immunoglobulin A, Secretory/analysis , Immunoglobulin A, Secretory/immunology , Longitudinal Studies
10.
Dev Comp Immunol ; 139: 104592, 2023 02.
Article En | MEDLINE | ID: mdl-36414098

Pulmonary collectins have been reported to bind carbohydrates on pathogens and inhibit infection by agglutination, neutralization, and opsonization. In this study, surfactant protein A (SP-A) was identified from goose lung and characterized at expression- and agglutination-functional levels. The deduced amino acid sequence of goose surfactant protein A (gSP-A) has two characteristic structures: a shorter, collagen-like region and a carbohydrate recognition domain. The latter contains two conserved motifs in its Ca2+-binding site: EPN (Glu-Pro-Asn) and WND (Trp-Asn-Asp). Expression analysis using qRT-PCR and fluorescence IHC revealed that gSP-A was highly expressed in the air sac and present in several other tissues, including the lung and trachea. We went on to produce recombinant gSP-A (RgSP-A) using a baculovirus/insect cell system and purified using a Ni2+ affinity column. A biological activity assay showed that all bacterial strains tested in this study were aggregated by RgSP-A, but only Escherichia coli AE17 (E. coli AE17, O2) and E. coli AE158 (O78) were susceptible to RgSP-A-mediated growth inhibition at 2-6 h. Moreover, the swarming motility of the two bacterial strains were weakened with increasing RgSP-A concentration, and their membrane permeability was compromised at 3 h, as determined by flow cytometry and laser confocal microscopy. Therefore, RgSP-A is capable of reducing bacterial viability of E. coli O2 and O78 via an aggregation-dependent mechanism which involves decreasing motility and increasing the bacterial membrane permeability. These data will facilitate detailed studies into the role of gSP-A in innate immune defense as well as for development of antibacterial agents.


Escherichia coli Infections , Escherichia coli , Geese , Immunity, Innate , Pulmonary Surfactant-Associated Protein A , Animals , Escherichia coli/growth & development , Escherichia coli/immunology , Geese/immunology , Geese/microbiology , Pulmonary Surfactant-Associated Protein A/genetics , Pulmonary Surfactant-Associated Protein A/metabolism , Lung/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/veterinary
11.
FASEB J ; 36(11): e22599, 2022 11.
Article En | MEDLINE | ID: mdl-36250902

Emerging evidence suggest that C3aR plays important roles in homeostasis, host defense and disease. Although it is known that C3aR is protective in several models of acute bacterial infections, the role for C3aR in chronic infection is largely unknown. Here we show that C3aR is protective in experimental chronic pyelonephritis. Global C3aR deficient (C3ar-/- ) mice had higher renal bacterial load, more pronounced renal histological lesions, increased renal apoptotic cell accumulation, tissue inflammation and extracellular matrix deposition following renal infection with uropathogenic E. coli (UPEC) strain IH11128, compared to WT control mice. Myeloid C3aR deficient (Lyz2-C3ar-/- ) mice exhibited a similar disease phenotype to global C3ar-/- mice. Pharmacological treatment with a C3aR agonist reduced disease severity in experimental chronic pyelonephritis. Furthermore, macrophages of C3ar-/- mice exhibited impaired ability to phagocytose UPEC. Our data clearly demonstrate a protective role for C3aR against experimental chronic pyelonephritis, macrophage C3aR plays a major role in the protection, and C3aR is necessary for phagocytosis of UPEC by macrophages. Our observation that C3aR agonist curtailed the pathology suggests a therapeutic potential for activation of C3aR in chronic infection.


Escherichia coli Infections , Pyelonephritis , Receptors, Complement , Animals , Mice , Escherichia coli Infections/immunology , Escherichia coli Infections/pathology , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Kidney/microbiology , Kidney/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Pyelonephritis/immunology , Pyelonephritis/microbiology , Pyelonephritis/pathology , Pyelonephritis/prevention & control , Uropathogenic Escherichia coli/pathogenicity , Receptors, Complement/agonists , Receptors, Complement/deficiency , Receptors, Complement/genetics , Receptors, Complement/immunology , Extracellular Matrix/metabolism
12.
Int J Mol Sci ; 23(17)2022 Aug 28.
Article En | MEDLINE | ID: mdl-36077151

Defensins play an important role in fighting bacteria, and are a good candidate for bactericidal agents. However, the function and mechanism of defensins in regulating host responses against bacteria is unclear. In this study, transcriptome analysis was used to study the comprehensive functions of pBD2 in IPEC-J2 cells against E. coli. In total, 230 differentially expressed genes (DEGs) were identified in IPEC-J2 cells between the control and E. coli groups, and were found by KEGG analysis to be involved in many signaling pathways related to immunity. Furthermore, 812 DEGs were observed between E. coli and E. coli +pBD2 groups, involved in the ribosome, oxidative phosphorylation, and certain disease pathways. Among these, 94 overlapping DEGs were in the two DEG groups, and 85 DEGs were reverse expression, which is involved in microRNA in cancer, while PTEN and CDC6 were key genes according to PPI net analysis. The results of qRT-PCR verified those of RNA-seq. The results indicated that pBD2 plays an important role against E. coli by acting on the genes related to immune response, cell cycle, ribosomes, oxidative phosphorylation, etc. The results provide new insights into the potential function and mechanism of pBD2 against E. coli. Meanwhile, this study provides a certain theoretical basis for research and the development of novel peptide drugs.


Defensins/metabolism , Escherichia coli Infections , Escherichia coli , Animals , Cell Line , Escherichia coli Infections/immunology , Gene Expression Profiling , Humans , RNA-Seq , Swine , Transcriptome
13.
Proc Natl Acad Sci U S A ; 119(40): e2206515119, 2022 10 04.
Article En | MEDLINE | ID: mdl-36161923

Antimicrobial peptides (AMPs) are critical to the protection of the urinary tract of humans and other animals from pathogenic microbial invasion. AMPs rapidly destroy pathogens by disrupting microbial membranes and/or augmenting or inhibiting the host immune system through a variety of signaling pathways. We have previously demonstrated that alpha-defensins 1-3 (DEFA1A3) are AMPs expressed in the epithelial cells of the human kidney collecting duct in response to uropathogens. We also demonstrated that DNA copy number variations in the DEFA1A3 locus are associated with UTI and pyelonephritis risk. Because DEFA1A3 is not expressed in mice, we utilized human DEFA1A3 gene transgenic mice (DEFA4/4) to further elucidate the biological relevance of this locus in the murine urinary tract. We demonstrate that the kidney transcriptional and translational expression pattern is similar in humans and the human gene transgenic mouse upon uropathogenic Escherichia coli (UPEC) stimulus in vitro and in vivo. We also demonstrate transgenic human DEFA4/4 gene mice are protected from UTI and pyelonephritis under various UPEC challenges. This study serves as the foundation to start the exploration of manipulating the DEFA1A3 locus and alpha-defensins 1-3 expression as a potential therapeutic target for UTIs and other infectious diseases.


Escherichia coli Infections , Pyelonephritis , Urinary Tract Infections , Uropathogenic Escherichia coli , alpha-Defensins , Animals , DNA Copy Number Variations , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Genetic Loci , Humans , Mice , Mice, Transgenic , Pyelonephritis/genetics , Pyelonephritis/immunology , Pyelonephritis/microbiology , Urinary Tract/microbiology , Urinary Tract Infections/genetics , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , alpha-Defensins/genetics
14.
Front Cell Infect Microbiol ; 12: 996841, 2022.
Article En | MEDLINE | ID: mdl-36093177

Enterotoxigenic Escherichia coli (ETEC) is a major cause of bacterial diarrhea in piglets, leading to economic losses in the pig industry. In past decades, long non-coding RNAs (lncRNAs) have shown to be widely involved in the regulation of host immunity in porcine infection diseases. In this study, we explored the lncRNAs associated with ETEC F41 infection in IPEC-J2 cells by high-throughput sequencing and bioinformatic analysis. A total of 10150 novel porcine lncRNAs were identified. There were 161 differentially expressed (DE) lncRNAs associated with ETEC F41 infection, of which 65 DE lncRNAs were up-regulated and 96 DE lncRNAs were down-regulated. Functional and KEGG enrichment analysis of predicted target genes of DE lncRNAs indicated they are enriched in cell growth and inflammation-related pathways, such as endocytosis, focal adhesion, TGF-ß signaling pathway, and adherens junctions. We revealed a novel candidate lncRNA MPHOSPH9-OT1 that was up-regulated after ETEC infection. The qRT-PCR validation and ELISA assessment showed the knockdown and overexpression of MPHOSPH9-OT1 resulted in significantly down- and up-regulation of cellular mRNA levels and secreted cytokine levels of CXCL8/IL-8, respectively. Meanwhile, MPHOSPH9-OT1 equilibrium is important to maintain the transepithelial electric resistance value and tight junction protein expression of IPEC-J2 cells. This study provides insights into the functionality of novel porcine lncRNAs in host immune responses to ETEC infection.


Cytoskeletal Proteins/genetics , Enterotoxigenic Escherichia coli , Escherichia coli Infections , RNA, Long Noncoding , Swine Diseases , Animals , Enterotoxigenic Escherichia coli/genetics , Escherichia coli Infections/immunology , Interleukin-8/genetics , RNA, Long Noncoding/genetics , RNA-Seq , Swine , Swine Diseases/genetics , Swine Diseases/microbiology
15.
Front Cell Infect Microbiol ; 12: 941939, 2022.
Article En | MEDLINE | ID: mdl-35967844

Lymphostatin is a virulence factor of enteropathogenic E. coli (EPEC) and non-O157 serogroup enterohaemorrhagic E. coli. Previous studies using whole-cell lysates of EPEC showed that lymphostatin inhibits the mitogen-activated proliferation of bulk human peripheral blood mononuclear cells (PBMCs) and the production of cytokines IL-2, IL-4, IL-5, and IFN-γ. Here, we used highly purified lymphostatin and PBMC-derived T cells to show that lymphostatin inhibits anti-CD3/anti-CD28-activated proliferation of human CD4+ and CD8+ T cells and blocks the synthesis of IL-2, IL-4, IL-10 and IFN-γ without affecting cell viability and in a manner dependent on an N-terminal DTD glycosyltransferase motif. Such inhibition was not observed with T cells activated by phorbol 12-myristate 13-acetate and ionomycin, implying that lymphostatin targets T cell receptor signaling. Analysis of the expression of CD69 indicated that lymphostatin suppresses T cell activation at an early stage and no impacts on apoptosis or necrosis were observed. Flow cytometric analysis of the DNA content of lymphostatin-treated CD4+ and CD8+ T cells showed a concentration- and DTD-dependent accumulation of the cells in the G0/G1 phase of the cell cycle, and corresponding reduction of the percentage of cells in S phase. Consistent with this, we found a marked reduction in the abundance of cyclins D3, E and A and loss of phosphorylated Rb over time in activated T cells from 8 donors treated with lymphostatin. Moreover, the cyclin-dependent kinase (cdk) inhibitor p27kip1, which inhibits progression of the cell cycle at G1 by acting on cyclin E-cdk2 or cyclin D-cdk4 complexes, was found to be accumulated in lymphostatin-treated T cells. Analysis of the abundance of phosphorylated kinases involved in signal transduction found that 30 of 39 were reduced in abundance following lymphostatin treatment of T cells from 5 donors, albeit not significantly so. Our data provide novel insights into the mode of action of lymphostatin on human T lymphocytes.


Bacterial Toxins , Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli , T-Lymphocytes , Apoptosis , Bacterial Toxins/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Checkpoints/immunology , Cell Division , Cell Proliferation/physiology , Cytokines/biosynthesis , Cytokines/immunology , Enteropathogenic Escherichia coli/immunology , Enteropathogenic Escherichia coli/pathogenicity , Escherichia coli/immunology , Escherichia coli/pathogenicity , Escherichia coli Infections/immunology , Escherichia coli Proteins/immunology , Humans , Interleukin-2 , Interleukin-4 , Leukocytes, Mononuclear/immunology , Necrosis , T-Lymphocytes/immunology , Virulence Factors/immunology
16.
Proc Natl Acad Sci U S A ; 119(33): e2117904119, 2022 08 16.
Article En | MEDLINE | ID: mdl-35939684

Many urinary tract infections (UTIs) are recurrent because uropathogens persist within the bladder epithelial cells (BECs) for extended periods between bouts of infection. Because persistent uropathogens are intracellular, they are often refractive to antibiotic treatment. The recent discovery of endogenous Lactobacillus spp. in the bladders of healthy humans raised the question of whether these endogenous bacteria directly or indirectly impact intracellular bacterial burden in the bladder. Here, we report that in contrast to healthy women, female patients experiencing recurrent UTIs have a bladder population of Lactobacilli that is markedly reduced. Exposing infected human BECs to L. crispatus in vitro markedly reduced the intracellular uropathogenic Escherichia coli (UPEC) load. The adherence of Lactobacilli to BECs was found to result in increased type I interferon (IFN) production, which in turn enhanced the expression of cathepsin D within lysosomes harboring UPECs. This lysosomal cathepsin D-mediated UPEC killing was diminished in germ-free mice and type I IFN receptor-deficient mice. Secreted metabolites of L. crispatus seemed to be responsible for the increased expression of type I IFN in human BECs. Intravesicular administration of Lactobacilli into UPEC-infected murine bladders markedly reduced their intracellular bacterial load suggesting that components of the endogenous microflora can have therapeutic effects against UTIs.


Antibiosis , Escherichia coli Infections , Interferon Type I , Lactobacillus crispatus , Urinary Bladder , Urinary Tract Infections , Uropathogenic Escherichia coli , Animals , Biological Therapy , Cathepsin D/metabolism , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy , Female , Humans , Immunity, Innate , Interferon Type I/immunology , Lactobacillus crispatus/physiology , Male , Mice , Urinary Bladder/immunology , Urinary Bladder/microbiology , Urinary Tract Infections/immunology , Urinary Tract Infections/microbiology , Urinary Tract Infections/therapy , Uropathogenic Escherichia coli/growth & development
17.
Proc Natl Acad Sci U S A ; 119(31): e2201146119, 2022 08 02.
Article En | MEDLINE | ID: mdl-35878041

Aberrant immune responses, including hyperresponsiveness to Toll-like receptor (TLR) ligands, underlie acute respiratory distress syndrome (ARDS). Type I interferons confer antiviral activities and could also regulate the inflammatory response, whereas little is known about their actions to resolve aberrant inflammation. Here we report that interferon-ß (IFN-ß) exerts partially overlapping, but also cooperative actions with aspirin-triggered 15-epi-lipoxin A4 (15-epi-LXA4) and 17-epi-resolvin D1 to counter TLR9-generated cues to regulate neutrophil apoptosis and phagocytosis in human neutrophils. In mice, TLR9 activation impairs bacterial clearance, prolongs Escherichia coli-evoked lung injury, and suppresses production of IFN-ß and the proresolving lipid mediators 15-epi-LXA4 and resolvin D1 (RvD1) in the lung. Neutralization of endogenous IFN-ß delays pulmonary clearance of E. coli and aggravates mucosal injury. Conversely, treatment of mice with IFN-ß accelerates clearance of bacteria, restores neutrophil phagocytosis, promotes neutrophil apoptosis and efferocytosis, and accelerates resolution of airway inflammation with concomitant increases in 15-epi-LXA4 and RvD1 production in the lungs. Pharmacological blockade of the lipoxin receptor ALX/FPR2 partially prevents IFN-ß-mediated resolution. These findings point to a pivotal role of IFN-ß in orchestrating timely resolution of neutrophil and TLR9 activation-driven airway inflammation and uncover an IFN-ß-initiated resolution program, activation of an ALX/FPR2-centered, proresolving lipids-mediated circuit, for ARDS.


Interferon-beta , Lipoxins , Respiratory Distress Syndrome , Animals , Docosahexaenoic Acids/pharmacology , Docosahexaenoic Acids/therapeutic use , Escherichia coli , Escherichia coli Infections/immunology , Humans , Inflammation/drug therapy , Interferon-beta/immunology , Interferon-beta/pharmacology , Lipoxins/pharmacology , Mice , Receptors, Formyl Peptide/antagonists & inhibitors , Respiratory Distress Syndrome/drug therapy , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Transcriptional Activation/drug effects
18.
Food Funct ; 13(11): 6282-6292, 2022 Jun 06.
Article En | MEDLINE | ID: mdl-35607985

The aims of this study were to evaluate whether a diet supplemented with glyceryl butyrate could attenuate the immune-inflammatory response in piglets challenged with enterotoxigenic Escherichia coli (ETEC), and to explore the mechanisms of its regulation. Eighteen weaning piglets were assigned to three diets: basal diet (CON), antibiotics diet (ATB), and 0.5% glyceryl butyrate diet (GB group). Significantly lower concentrations of IL-1ß, IL-6 and TNF-α in the jejunum and IL-6 in the ileum were observed in the GB group than that in the CON group (P < 0.05). Moreover, a decreasing trend of IL-1ß (P = 0.075) and TNF-α (P = 0.070) was observed in the ileum in the GB group. Correspondingly, the GB group had significantly increased mRNA expression of porcine beta defensins (pBDs) in the jejunum (pBD1, pBD2, pBD114 and pBD129) and ileum (pBD2, pBD3, pBD114 and pBD129) (P < 0.05), and protein abundance of Claudin 1, Occludin, and ZO-1 in the jejunum and ileum (P < 0.05). Further research results showed that the improvement of beta defensins and tight junctions in the GB group was related to the decreased phosphorylation of the NFκB/MAPK pathway. In addition, the results of 16S rDNA sequencing showed that glycerol butyrate supplementation altered the ileal microbiota composition of piglets, increasing the relative abundance of Lactobacillus reuteri, Lactobacillus salivarius, and Lactobacillus agrilis. In summary, glyceryl butyrate attenuated the immune-inflammatory response in piglets challenged with ETEC by inhibiting the NF-κB/MAPK pathways and modulating the gut microbiota, and thus improved piglet intestinal health.


Anti-Inflammatory Agents , Butyrates , Enterotoxigenic Escherichia coli , Escherichia coli Infections , Gastrointestinal Microbiome , Intestines , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Butyrates/pharmacology , Butyrates/therapeutic use , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/drug therapy , Escherichia coli Infections/immunology , Escherichia coli Infections/veterinary , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Inflammation/drug therapy , Inflammation/immunology , Inflammation/veterinary , Interleukin-6 , Intestines/drug effects , Intestines/immunology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Swine , Tumor Necrosis Factor-alpha , beta-Defensins/biosynthesis , beta-Defensins/immunology
19.
J Biol Chem ; 298(5): 101938, 2022 05.
Article En | MEDLINE | ID: mdl-35429501

Mitogen-activated protein kinase phosphatase 1 (Mkp-1) KO mice produce elevated cytokines and exhibit increased mortality and bacterial burden following systemic Escherichia coli infection. To understand how Mkp-1 affects immune defense, we analyzed the RNA-Seq datasets previously generated from control and E. coli-infected Mkp-1+/+ and Mkp-1-/- mice. We found that E. coli infection markedly induced programmed death-ligand 1 (PD-L1) expression and that Mkp-1 deficiency further amplified PD-L1 expression. Administration of a PD-L1-neutralizing monoclonal antibody (mAb) to Mkp-1-/- mice increased the mortality of the animals following E. coli infection, although bacterial burden was decreased. In addition, the PD-L1-neutralizing mAb increased serum interferon (IFN)-γ and tumor necrosis factor alpha, as well as lung- and liver-inducible nitric oxide synthase levels, suggesting an enhanced inflammatory response. Interestingly, neutralization of IFN-α/ß receptor 1 blocked PD-L1 induction in Mkp-1-/- mice following E. coli infection. PD-L1 was potently induced in macrophages by E. coli and lipopolysaccharide in vitro, and Mkp-1 deficiency exacerbated PD-L1 induction with little effect on the half-life of PD-L1 mRNA. In contrast, inhibitors of Janus kinase 1/2 and tyrosine kinase 2, as well as the IFN-α/ß receptor 1-neutralizing mAb, markedly attenuated PD-L1 induction. These results suggest that the beneficial effect of type I IFNs in E. coli-infected Mkp-1-/- mice is, at least in part, mediated by Janus kinase/signal transducer and activator of transcription-driven PD-L1 induction. Our studies also support the notion that enhanced PD-L1 expression contributes to the bactericidal defect of Mkp-1-/- mice.


B7-H1 Antigen , Dual Specificity Phosphatase 1 , Escherichia coli Infections , Gene Expression Regulation , Interferon Type I , Animals , B7-H1 Antigen/genetics , Dual Specificity Phosphatase 1/metabolism , Escherichia coli/genetics , Escherichia coli/immunology , Escherichia coli Infections/immunology , Gene Expression Regulation/immunology , Interferon Type I/genetics , Mice
20.
J Leukoc Biol ; 112(3): 499-512, 2022 09.
Article En | MEDLINE | ID: mdl-35435271

Clearance of airway intruders by immune cells is required to resolve infectious pneumonia. However, the molecular mechanisms underlying this process remain elusive. Here, we demonstrated that alveolar macrophage (AM)-derived neuropilin 2 (NRP2) plays an essential role in controlling severe pneumonia by enhancing microbial clearance. Mice with conditional deletion of the NRP2 gene in AM had persistent bacteria, uncontrolled neutrophil influx, and decreased survival during Escherichia coli-induced pneumonia. In vitro assays demonstrated that NRP2 could bind to CD11b+ Ly6Glo/+ neutrophils and promote their capacities in phagocytosis and killing of bacteria, which is partially contributed to the increased expression of TLR4 and TNF-a. These findings collectively revealed that AM-derived NRP2 protects the lungs from unwanted injury by promoting the clearance of invading pathogens. This study might provide a promising diagnostic biomarker and therapeutic target for severe pneumonia.


Escherichia coli Infections , Macrophages, Alveolar , Neuropilin-2 , Pneumonia, Bacterial , Animals , Escherichia coli Infections/immunology , Lung , Lung Injury , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Neuropilin-2/metabolism , Neutrophils , Phagocytosis , Pneumonia, Bacterial/immunology
...