Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
1.
Iran J Allergy Asthma Immunol ; 20(5): 537-549, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34664813

ABSTRACT

Nosocomial infections caused by Acinetobacter baumannii (A. baumannii) nosocomial infections caused by Acinetobacter baumannii (A. baumannii) are considered as a global serious problem in hospitalized patients because of emerging antibiotic resistance. Immunotherapy approaches are promising to prevent such infections. In our previous study, five antigenic epitopes of outer membrane protein A (OmpA), as the most dangerous virulence molecule in A. baumanii, were predicted in silico. In this study, the investigators evaluated some immunological aspects of the peptides. Five peptides were separately injected into C5BL/6 mice; then the cytokine production (interleukin-4 and interferon-gamma) of splenocytes and opsonophagocytic activity of immunized serum were assessed. To identify the protective function of the peptides, animal models of sepsis and pneumonia infections were actively and passively immunized with selected peptides and pooled sera of immunized mice, respectively. Then, survival rates of them were compared with the non-infected controls. Based on the results, activated spleen cells in P127 peptide-immunized mice exhibited an increase level of IFN-γ compared with the other experimental groups, but not about the IL-4 concentration. The results of opsonophagocytic assay revealed an appropriate killing activity of produced antibodies against A. baumannii in a dose-dependent manner. Further, the survival rates of the mice under passive immunization with the immunized sera or active immunization with P127 peptide were significantly more than those in the control group. Moreover, the survival rate of the P127 peptide immunized group was considerably higher than that among the other peptide-immunized group. In conclusion, findings indicated that peptides derived from outer membrane protein-A can be used as a promising tool for designing the epitope-based vaccines against infections caused by A. baumannii.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Epitopes/immunology , Pneumonia, Bacterial/prevention & control , Sepsis/prevention & control , Acinetobacter Infections/immunology , Acinetobacter Infections/mortality , Animals , Antigens, Bacterial/immunology , Bacterial Vaccines/administration & dosage , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions/immunology , Immunization , Mice , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Prognosis , Sepsis/immunology , Sepsis/mortality , Treatment Outcome
2.
mBio ; 12(5): e0224821, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34517755

ABSTRACT

The human pathogen Acinetobacter baumannii produces and utilizes acinetobactin for iron assimilation. Although two isomeric structures of acinetobactin, one featuring an oxazoline (Oxa) and the other with an isoxazolidinone (Isox) at the core, have been identified, their differential roles as virulence factors for successful infection have yet to be established. This study provides direct evidence that Oxa supplies iron more efficiently than Isox, primarily owing to its specific recognition by the cognate outer membrane receptor, BauA. The other components in the acinetobactin uptake machinery appear not to discriminate these isomers. Interestingly, Oxa was found to form a stable iron complex that is resistant to release of the chelated iron upon competition by Isox, despite their comparable apparent affinities to Fe(III). In addition, both Oxa and Isox were found to be competent iron chelators successfully scavenging iron from host metal sequestering proteins responsible for nutritional immunity. These observations collectively led us to propose a new model for acinetobactin-based iron assimilation at infection sites. Namely, Oxa is the principal siderophore mediating the core Fe(III) supply chain for A. baumannii, whereas Isox plays a minor role in the iron delivery and, alternatively, functions as an auxiliary iron collector that channels the iron pool toward Oxa. The unique siderophore utilization mechanism proposed here represents an intriguing strategy for pathogen adaptation under the various nutritional stresses encountered at infection sites. IMPORTANCE Acinetobacter baumannii has acquired antibiotic resistance at an alarming rate, and it is becoming a serious threat to society, particularly due to the paucity of effective treatment options. Acinetobactin is a siderophore of Acinetobacter baumannii, responsible for active iron supply, and it serves as a key virulence factor to counter host nutritional immunity during infection. While two acinetobactin isomers were identified, their distinctive roles for successful infection of Acinetobacter baumannii remained unsettled. This study clearly identified the isomer containing an oxazoline core as the principal siderophore based on comparative analysis of the specificity of the acinetobactin uptake machinery, the stability of the corresponding iron complexes, and the iron scavenging activity against the host iron sequestering proteins. Our findings are anticipated to stimulate efforts to discover a potent antivirulence agent against Acinetobacter baumannii that exploits the acinetobactin-based iron assimilation mechanism.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter baumannii/metabolism , Imidazoles/chemistry , Imidazoles/metabolism , Oxazoles/chemistry , Oxazoles/metabolism , Acinetobacter Infections/immunology , Acinetobacter Infections/metabolism , Acinetobacter baumannii/chemistry , Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Iron/metabolism , Isomerism , Siderophores/chemistry , Siderophores/metabolism
3.
Elife ; 102021 09 20.
Article in English | MEDLINE | ID: mdl-34544549

ABSTRACT

Vaccination strategies for rapid protection against multidrug-resistant bacterial infection are very important, especially for hospitalized patients who have high risk of exposure to these bacteria. However, few such vaccination strategies exist due to a shortage of knowledge supporting their rapid effect. Here, we demonstrated that a single intranasal immunization of inactivated whole cell of Acinetobacter baumannii elicits rapid protection against broad A. baumannii-infected pneumonia via training of innate immune response in Rag1-/- mice. Immunization-trained alveolar macrophages (AMs) showed enhanced TNF-α production upon restimulation. Adoptive transfer of immunization-trained AMs into naive mice mediated rapid protection against infection. Elevated TLR4 expression on vaccination-trained AMs contributed to rapid protection. Moreover, immunization-induced rapid protection was also seen in Pseudomonas aeruginosa and Klebsiella pneumoniae pneumonia models, but not in Staphylococcus aureus and Streptococcus pneumoniae model. Our data reveal that a single intranasal immunization induces rapid and efficient protection against certain Gram-negative bacterial pneumonia via training AMs response, which highlights the importance and the possibility of harnessing trained immunity of AMs to design rapid-effecting vaccine.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Vaccines/administration & dosage , Klebsiella Infections/prevention & control , Klebsiella pneumoniae/immunology , Macrophages, Alveolar/drug effects , Pneumonia, Bacterial/prevention & control , Pseudomonas Infections/prevention & control , Pseudomonas aeruginosa/immunology , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Administration, Intranasal , Adoptive Transfer , Animals , Cells, Cultured , Disease Models, Animal , Female , Homeodomain Proteins/genetics , Immunity, Innate/drug effects , Klebsiella Infections/immunology , Klebsiella Infections/microbiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/transplantation , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Time Factors , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Vaccination , Vaccines, Inactivated/administration & dosage
4.
Infect Immun ; 89(10): e0016221, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34310884

ABSTRACT

Extremely drug-resistant (XDR) Acinetobacter baumannii is a notorious and frequently encountered pathogen demanding novel therapeutic interventions. An initial monoclonal antibody (MAb), C8, raised against A. baumannii capsule, proved a highly effective treatment against a minority of clinical isolates. To overcome this limitation, we broadened coverage by developing a second antibody for use in a combination regimen. We sought to develop an additional anti-A. baumannii MAb through hybridoma technology by immunizing mice with sublethal inocula of virulent, XDR clinical isolates not bound by MAb C8. We identified a new antibacterial MAb, 65, which bound to strains in a pattern distinct from and complementary to that of MAb C8. MAb 65 enhanced macrophage opsonophagocytosis of targeted strains and markedly improved survival in lethal bacteremic sepsis and aspiration pneumonia murine models of A. baumannii infection. MAb 65 was also synergistic with colistin, substantially enhancing protection compared to monotherapy. Treatment with MAb 65 significantly reduced blood bacterial density, ameliorated cytokine production (interleukin-1ß [IL-1ß], IL-6, IL-10, and tumor necrosis factor), and sepsis biomarkers. We describe a novel MAb targeting A. baumannii that broadens immunotherapeutic strain coverage, is highly potent and effective, and synergistically improves outcomes in combination with antibiotics.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Antibodies, Monoclonal/immunology , Acinetobacter Infections/blood , Acinetobacter Infections/microbiology , Animals , Anti-Bacterial Agents/immunology , Antibodies, Bacterial/immunology , Biomarkers/blood , Colistin/immunology , Cytokines/blood , Cytokines/immunology , Drug Resistance, Multiple, Bacterial/immunology , Mice , Microbial Sensitivity Tests/methods , Sepsis/blood , Sepsis/immunology , Sepsis/microbiology
5.
Sci Rep ; 11(1): 13213, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168196

ABSTRACT

Acinetobacter baumannii (A. baumannii), an opportunistic, gram-negative pathogen, has evoked the interest of the medical community throughout the world because of its ability to cause nosocomial infections, majorly infecting those in intensive care units. It has also drawn the attention of researchers due to its evolving immune evasion strategies and increased drug resistance. The emergence of multi-drug-resistant-strains has urged the need to explore novel therapeutic options as an alternative to antibiotics. Due to the upsurge in antibiotic resistance mechanisms exhibited by A. baumannii, the current therapeutic strategies are rendered less effective. The aim of this study is to explore novel therapeutic alternatives against A. baumannii to control the ailed infection. In this study, a computational framework is employed involving, pan genomics, subtractive proteomics and reverse vaccinology strategies to identify core promiscuous vaccine candidates. Two chimeric vaccine constructs having B-cell derived T-cell epitopes from prioritized vaccine candidates; APN, AdeK and AdeI have been designed and checked for their possible interactions with host BCR, TLRs and HLA Class I and II Superfamily alleles. These vaccine candidates can be experimentally validated and thus contribute to vaccine development against A. baumannii infections.


Subject(s)
Acinetobacter baumannii/immunology , Bacterial Vaccines/immunology , Acinetobacter Infections/immunology , Amino Acid Sequence , Anti-Bacterial Agents/immunology , Computational Biology/methods , Cross Infection/immunology , Epitopes/immunology , Genome, Bacterial/immunology , Genomics/methods , Proteomics/methods , Vaccinology/methods
6.
Front Immunol ; 12: 666742, 2021.
Article in English | MEDLINE | ID: mdl-33936107

ABSTRACT

Vaccines and monoclonal antibodies are promising approaches for preventing and treating infections caused by multidrug resistant Acinetobacter baumannii. However, only partial protection has been achieved with many previously tested protein antigens, which suggests that vaccines incorporating multiple antigens may be necessary in order to obtain high levels of protection. Several aspects that use the wealth of omic data available for A. baumannii have not been fully exploited for antigen identification. In this study, the use of fractionated proteomic and computational data from ~4,200 genomes increased the number of proteins potentially accessible to the humoral response to 8,824 non-redundant proteins in the A. baumannii panproteome. Among them, 59% carried predicted B-cell epitopes and T-cell epitopes recognized by two or more alleles of the HLA class II DP supertype. Potential cross-reactivity with human proteins was detected for 8.9% of antigens at the protein level and 2.7% at the B-cell epitope level. Individual antigens were associated with different infection types by genomic, transcriptomic or functional analyses. High intra-clonal genome density permitted the identification of international clone II as a "vaccitype", in which 20% of identified antigens were specific to this clone. Network-based centrality measurements were used to identify multiple immunologic nodes. Data were formatted, unified and stored in a data warehouse database, which was subsequently used to identify synergistic antigen combinations for different vaccination strategies. This study supports the idea that integration of multi-omic data and fundamental knowledge of the pathobiology of drug-resistant bacteria can facilitate the development of effective multi-antigen vaccines against these challenging infections.


Subject(s)
Acinetobacter baumannii/drug effects , Acinetobacter baumannii/immunology , Bacterial Vaccines/immunology , Drug Resistance, Bacterial/immunology , Epitopes/immunology , Acinetobacter Infections/immunology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/genetics , Antigens, Bacterial/immunology , Epitopes/chemistry , Epitopes/genetics , Genes, Bacterial , Genome, Bacterial , Genomics/methods , Humans
7.
J Infect Dis ; 224(12): 2133-2147, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34036366

ABSTRACT

Monoclonal antibodies (mAbs) are gaining significant momentum as novel therapeutics for infections caused by antibiotic-resistant bacteria. We evaluated the mechanism by which antibacterial mAb therapy protects against Acinetobacter baumannii infections. Anticapsular mAb enhanced macrophage opsonophagocytosis and rescued mice from lethal infections by harnessing complement, macrophages, and neutrophils; however, the degree of bacterial burden did not correlate with survival. Furthermore, mAb therapy reduced proinflammatory (interleukin-1ß [IL-1ß], IL-6, tumor necrosis factor-α [TNF-α]) and anti-inflammatory (IL-10) cytokines, which correlated inversely with survival. Although disrupting IL-10 abrogated the survival advantage conferred by the mAb, IL-10-knockout mice treated with mAb could still survive if TNF-α production was suppressed directly (via anti-TNF-α neutralizing antibody) or indirectly (via macrophage depletion). Thus, even for a mAb that enhances microbial clearance via opsonophagocytosis, clinical efficacy required modulation of pro- and anti-inflammatory cytokines. These findings may inform future mAb development targeting bacteria that trigger the sepsis cascade.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter Infections/immunology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Immunomodulation , Acinetobacter Infections/microbiology , Animals , Anti-Bacterial Agents , Cytokines/blood , Cytokines/immunology , Interleukin-10 , Mice , Opsonization , Tumor Necrosis Factor Inhibitors , Tumor Necrosis Factor-alpha
8.
Int J Nanomedicine ; 16: 1819-1836, 2021.
Article in English | MEDLINE | ID: mdl-33707942

ABSTRACT

BACKGROUND: The development of vaccines is a promising and cost-effective strategy to prevent emerging multidrug-resistant (MDR) Acinetobacter baumannii (A. baumannii) infections. The purpose of this study was to prepare a multiepitope peptide nanovaccine and evaluate its immunogenicity and protective effect in BALB/c mice. METHODS: The B-cell and T-cell epitopes of Omp22 from A. baumannii were predicted using bioinformatics methods and identified by immunological experiments. The optimal epitopes were conjugated in series by 6-aminocaproic acid and chemically synthesized multiepitope polypeptide rOmp22. Then, rOmp22 was encapsulated by chitosan (CS) and poly (lactic-co-glycolic) acid (PLGA) to prepare CS-PLGA-rOmp22 nanoparticles (NPs). The immunogenicity and immunoprotective efficacy of the vaccine were evaluated in BALB/c mice. RESULTS: CS-PLGA-rOmp22 NPs were small (mean size of 272.83 nm) with apparently spherical structures, positively charged (4.39 mV) and nontoxic to A549 cells. A high encapsulation efficiency (54.94%) and a continuous slow release pattern were achieved. Compared with nonencapsulated rOmp22, CS-PLGA-rOmp22 immunized BALB/c mice induced higher levels of rOmp22-specific IgG in serum and IFN-γ in splenocyte supernatant. Additionally, lung injury and bacterial burdens in the lung and blood were suppressed, and potent protection (57.14%-83.3%) against acute lethal intratracheal A. baumannii challenge was observed in BALB/c mice vaccinated with CS-PLGA-rOmp22. CONCLUSION: CS-PLGA-rOmp22 NPs elicited specific IgG antibodies, Th1 cellular immunity and protection against acute lethal intratracheal A. baumannii challenge. Our results indicate that this nanovaccine is a desirable candidate for preventing A. baumannii infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Bacterial Vaccines/immunology , Chitosan/chemistry , Epitopes/immunology , Nanoparticles/chemistry , Peptides/immunology , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , A549 Cells , Acinetobacter Infections/blood , Acinetobacter Infections/microbiology , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Bacterial Load , Body Weight , Epitopes/chemistry , Female , Humans , Immunity, Humoral , Immunization , Immunoglobulin G/immunology , Interferon-gamma/metabolism , Interleukin-4/metabolism , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred BALB C , Nanoparticles/ultrastructure , Peptides/chemistry , Recombinant Proteins/isolation & purification , Spleen/pathology , Survival Analysis
9.
Sci Rep ; 11(1): 2942, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33536477

ABSTRACT

Sepsis is caused by organ dysfunction initiated by an unrestrained host immune response to infection. The emergence of antibiotic-resistant bacteria has rapidly increased in the last decades and has stimulated a firm research platform to combat infections caused by antibiotic-resistant bacteria that cannot be eradicated with conventional antibiotics. Strategies like epigenetic regulators such as lysine demethylase (Kdm) has received attention as a new target. Thus, we sought to investigate the epigenetic mechanisms in sepsis pathophysiology with the aim of discovering new concepts for treatment. A transcriptome analysis of dendritic cells during their inflammatory state identified Kdm as a critical molecule in sepsis regulation. Next, 8-hydroxyquinoline-5-carboxylic acid (IOX1) ability to control endotoxemia induced by Lipopolysaccharide and bacterial sepsis was demonstrated. IOX1 has been shown to regulate endotoxemia and sepsis caused by Escherichia coli and carbapenem-resistant Acinetobacter baumannii and has also contributed to the suppression of multidrug-resistant bacterial growth through the inhibition of DNA Gyrase. These findings show that IOX1 could be a component agent against bacterial sepsis by functioning as a broad-spectrum antibiotic with dual effects.


Subject(s)
Acinetobacter Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/drug therapy , Hydroxyquinolines/pharmacology , Sepsis/drug therapy , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , DNA Gyrase/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli/drug effects , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/metabolism , Humans , Hydroxyquinolines/therapeutic use , Mice , Microbial Sensitivity Tests , Molecular Docking Simulation , Sepsis/immunology , Sepsis/microbiology
10.
Expert Rev Vaccines ; 20(3): 281-295, 2021 03.
Article in English | MEDLINE | ID: mdl-33554671

ABSTRACT

INTRODUCTION: Healthcare-associated infections caused by multidrug-resistant Acinetobacter baumannii are becoming alarming worldwide. However, the pipeline of new antibiotics is very limited. Vaccination is one of the most cost effective and promising strategies to prevent infections and can play an important role in combat multidrug resistance A. baumannii and prevent the development of new drug resistance. AREA COVERED: This review gives an overview of the research and development of A. baumannii vaccines during the past five years (2015-2020), discusses the key progresses and current challenges of the field, and speculates on the future of A. baumannii vaccine development. EXPERT OPINION: Moderate progresses have been made in the research and development of A. baumannii vaccine in the last five years, in particular in the areas of identification of new protein targets, development of multicomponent vaccines, and use of vaccines and antibodies as adjuncts for antibiotics therapies. However, substantial scientific and logistic challenges, such as selection of lead vaccine candidates and formulation, vaccine clinical trials and targeted population, and financial incentives, remain. Thus, innovative strategies will be needed before an A. baumannii vaccine candidate can be brought into late stage of preclinical development in next five years.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii , Bacterial Vaccines , Vaccine Development , Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Animals , Anti-Bacterial Agents/therapeutic use , Humans
11.
Future Microbiol ; 16(3): 143-157, 2021 02.
Article in English | MEDLINE | ID: mdl-33528272

ABSTRACT

Aim: Iron uptake and metabolism pathways are promising targets in vaccine development as an alternative strategy for antibiotics. Methods & methods: HemTR, a putative heme receptor of Acinetobacter baumannii, was expressed and its protectivity against A. baumannii was determined singly or in combination with the siderophore receptor, BauA, in mice. Results: High level of IgG was elicited. There was a delay in mice mortality with reduced bacterial loads in internal organs in the sublethal challenge. Protection was better in the HemTR-BauA group in both lethal and sublethal challenges. Passive transfer of anti-HemTR and anti-BauA partially protected mice against A. baumannii infection. Conclusion: HemTR in combination with other iron receptors could contribute to the development of protective vaccines against A. baumannii.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/immunology , Receptors, Cell Surface/immunology , Sepsis/prevention & control , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Animals , Bacterial Load , Bacterial Outer Membrane Proteins/administration & dosage , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Bacterial Vaccines/immunology , Disease Models, Animal , Female , Heme/immunology , Humans , Mice , Mice, Inbred BALB C , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/genetics , Sepsis/immunology , Sepsis/microbiology
12.
Viruses ; 14(1)2021 12 24.
Article in English | MEDLINE | ID: mdl-35062236

ABSTRACT

Respiratory infections caused by multidrug-resistant Acinetobacter baumannii are difficult to treat and associated with high mortality among critically ill hospitalized patients. Bacteriophages (phages) eliminate pathogens with high host specificity and efficacy. However, the lack of appropriate preclinical experimental models hampers the progress of clinical development of phages as therapeutic agents. Therefore, we tested the efficacy of a purified lytic phage, vB_AbaM_Acibel004, against multidrug-resistant A. baumannii clinical isolate RUH 2037 infection in immunocompetent mice and a human lung tissue model. Sham- and A. baumannii-infected mice received a single-dose of phage or buffer via intratracheal aerosolization. Group-specific differences in bacterial burden, immune and clinical responses were compared. Phage-treated mice not only recovered faster from infection-associated hypothermia but also had lower pulmonary bacterial burden, lower lung permeability, and cytokine release. Histopathological examination revealed less inflammation with unaffected inflammatory cellular recruitment. No phage-specific adverse events were noted. Additionally, the bactericidal effect of the purified phage on A. baumannii was confirmed after single-dose treatment in an ex vivo human lung infection model. Taken together, our data suggest that the investigated phage has significant potential to treat multidrug-resistant A. baumannii infections and further support the development of appropriate methods for preclinical evaluation of antibacterial efficacy of phages.


Subject(s)
Acinetobacter Infections/therapy , Acinetobacter baumannii , Myoviridae/physiology , Phage Therapy , Pneumonia, Bacterial/therapy , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/virology , Animals , Anti-Bacterial Agents/pharmacology , Cytokines/metabolism , Drug Resistance, Multiple, Bacterial , Female , Humans , Lung/immunology , Lung/microbiology , Lung/pathology , Mice , Mice, Inbred C57BL , Phage Therapy/adverse effects , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology
13.
Front Immunol ; 11: 601614, 2020.
Article in English | MEDLINE | ID: mdl-33329595

ABSTRACT

Acinetobacter baumannii is an important opportunistic pathogen that primarily afflicts elderly people. To clarify the pathogenicity of A. baumannii in the elderly, we investigated immune responses to A. baumannii ATCC 19606 infection in klotho knockout (KO) mice, the mouse model of aging. Following intravenous inoculation, the mice seldom displayed severe symptoms. However, the survival rate was 56% at 7 days post-infection. Bacteria were detected in the lungs of klotho KO mice but not klotho wildtype (WT) mice at 7 days post-infection. Neutrophils, eosinophils, interstitial macrophages, and monocyte/dendritic cell subset in the lungs of klotho KO mice were transiently induced after infection with A. baumannii. The number of alveolar macrophages in klotho KO mice was lower than that in klotho WT mice, except for 1 day post-infection. CD11b expression on neutrophils and alveolar macrophages in the lungs of klotho KO mice was seldom upregulated by the infection. These results suggested that immune functions eliminating bacteria in the lungs of klotho KO mice were insufficient. CD11blow conventional DC cells hardly increased in klotho KO mice infected with A. baumannii. Additionally, the production of interleukin (IL)-10 in the sera of klotho KO mice was significantly higher than that in klotho WT mice, whereas that production of interferon-gamma was not detected in the sera of klotho KO mice. These results suggested that acquired immune responses were hardly induced in klotho KO mice. IL-1ß, CXCL1, CXCL2, and CCL2 expression was significantly higher in the lungs of klotho KO mice infected with A. baumannii than in those of klotho WT mice at 1 day post-infection. These results suggested that pulmonary inflammation was elicited in klotho KO mice during early infection. The expression levels of proinflammatory cytokines significantly correlated with TLR9 expression in the lungs of klotho KO mice. The collective results demonstrate an A. baumannii infection state in aged hosts and suggest that pulmonary inflammation and bacterial burden should be noted in aged hosts even in the absence of severe symptoms of A. baumannii infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Cytokines/metabolism , Glucuronidase/deficiency , Lung/immunology , Pneumonia, Bacterial/immunology , Acinetobacter Infections/genetics , Acinetobacter Infections/metabolism , Acinetobacter Infections/microbiology , Acinetobacter baumannii/pathogenicity , Age Factors , Animals , Bacterial Load , Cytokines/genetics , Disease Models, Animal , Glucuronidase/genetics , Host-Parasite Interactions , Klotho Proteins , Lung/metabolism , Lung/microbiology , Male , Mice, Knockout , Pneumonia, Bacterial/genetics , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism
14.
Biomed Res Int ; 2020: 1975875, 2020.
Article in English | MEDLINE | ID: mdl-33134372

ABSTRACT

Acinetobacter baumannii (A. baumannii) is an important opportunistic pathogen widely present in medical environment. Given its complex drug resistance, A. baumannii poses a serious threat to the safety of critically ill patients. Given the limited alternative antibiotics, nonantibiotic-based functional anti-A. baumannii infection proteins must be developed. In this study, we firstly used a series of biological software to predict potential epitopes in the MacB protein sequence and verified them by antibody recognition and lymphocyte proliferation tests. We finally screened out B cell epitope 2, CD8+ T cell epitope 7, and CD4+ T cell epitope 11 and connected them to construct a recombinant antigen epitope (RAE). The determination of IgG in the serum of immunised mice and cytokines in the supernatant of lymphocytes showed that the constructed epitope induced an immune response mediated by Th-1 cells. Finally, the challenge experiment of A. baumannii infection in mice confirmed that the epitope developed based on MacB, especially RAE, provided incomplete immune protection for mice.


Subject(s)
ATP-Binding Cassette Transporters/immunology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/drug effects , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Epitopes, B-Lymphocyte/immunology , Recombinant Fusion Proteins/administration & dosage , ATP-Binding Cassette Transporters/chemistry , ATP-Binding Cassette Transporters/genetics , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/immunology , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , CD4 Antigens/genetics , CD4 Antigens/immunology , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunity, Cellular/drug effects , Immunogenicity, Vaccine , Mice , Mice, Inbred BALB C , Models, Molecular , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Th1 Cells/drug effects , Th1 Cells/immunology , Th1 Cells/microbiology
15.
Int J Mol Sci ; 21(15)2020 Jul 31.
Article in English | MEDLINE | ID: mdl-32752093

ABSTRACT

Acinetobacter baumannii is considered one of the most persistent pathogens responsible for nosocomial infections. Due to the emergence of multidrug resistant strains, as well as high morbidity and mortality caused by this pathogen, A. baumannii was placed on the World Health Organization (WHO) drug-resistant bacteria and antimicrobial resistance research priority list. This review summarizes current studies on mechanisms that protect A. baumannii against multiple stresses caused by the host immune response, outside host environment, and antibiotic treatment. We particularly focus on the ability of A. baumannii to survive long-term desiccation on abiotic surfaces and the population heterogeneity in A. baumannii biofilms. Insight into these protective mechanisms may provide clues for the development of new strategies to fight multidrug resistant strains of A. baumannii.


Subject(s)
Acinetobacter Infections/genetics , Acinetobacter baumannii/genetics , Host-Pathogen Interactions/genetics , Immunity/genetics , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/pathogenicity , Anti-Bacterial Agents/therapeutic use , Biofilms/growth & development , Drug Resistance, Multiple, Bacterial/genetics , Humans , Virulence/genetics
16.
Front Immunol ; 11: 1069, 2020.
Article in English | MEDLINE | ID: mdl-32655550

ABSTRACT

Acinetobacter baumannii (A. baumannii) is becoming a common global concern due to the emergence of multi-drug or pan-drug resistant strains. Confronting the issue of antimicrobial resistance by developing vaccines against the resistant pathogen is becoming a common strategy. In this study, different methods for preparing A. baumannii outer membrane vesicles (AbOMVs) vaccines were developed. sOMV (spontaneously released AbOMV) was extracted from the culture supernatant, while SuOMV (sucrose-extracted AbOMV) and nOMV (native AbOMV) were prepared from the bacterial cells. Three AbOMVs exhibited significant differences in yield, particle size, protein composition, and LPS/DNA content. To compare the protective efficacy of the three AbOMVs, groups of mice were immunized either intramuscularly or intranasally with each AbOMV. Vaccination via both routes conferred significant protection against lethal and sub-lethal A. baumannii challenge. Moreover, intranasal vaccination provided more robust protection, which may be attributed to the induction of significant sIgA response in mucosal sites. Among the three AbOMVs, SuOMV elicited the highest level of protective immunity against A. baumannii infection, whether intramuscular or intranasal immunization, which was characterized by the expression of the most profound specific serum IgG or mucosal sIgA. Taken together, the preparation method had a significant effect on the yield, morphology, and composition of AbOMVs, that further influenced the protective effect against A. baumannii infection.


Subject(s)
Acinetobacter baumannii/immunology , Bacterial Vaccines/isolation & purification , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/pathogenicity , Acinetobacter baumannii/ultrastructure , Administration, Intranasal , Animals , Antibodies, Bacterial/biosynthesis , Antibodies, Bacterial/blood , Antibody Specificity , Bacterial Outer Membrane/immunology , Bacterial Outer Membrane/ultrastructure , Bacterial Outer Membrane Proteins/immunology , Bacterial Outer Membrane Proteins/isolation & purification , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Humans , Immunity, Mucosal , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin G/blood , Immunoglobulin G/classification , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission
17.
Curr Opin Microbiol ; 55: 74-80, 2020 06.
Article in English | MEDLINE | ID: mdl-32388085

ABSTRACT

Acinetobacter baumannii is known for its intrinsic resistance to conventional antibiotic treatment and hypervirulence during infection. This coupled with its extraordinary capacity to survive in myriad harsh environments has led to increasing rates of infection in clinical settings. Numerous studies have characterized the virulence factors and resistance genes in A. baumannii responsible for the detrimental outcomes seen in patients; however, the role of regulatory factors in controlling the expression of these genes remains less well explored. Herein we discuss the latest and most influential findings on the regulatory network of A. baumannii, focusing on the transcription factors, two-component systems, and sRNAs. We place particular focus on those identified as being crucial for sensing and responding to continually changing environments, and influencing survival and virulence when engaging with the human host.


Subject(s)
Acinetobacter baumannii/physiology , Acinetobacter baumannii/pathogenicity , Drug Resistance, Bacterial/genetics , Host-Pathogen Interactions , Virulence/genetics , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Bacterial Proteins , Cell Division/genetics , Cell Wall/metabolism , Gene Expression Regulation, Bacterial , Genes, Bacterial , Humans , Transcription Factors
18.
Infect Immun ; 88(7)2020 06 22.
Article in English | MEDLINE | ID: mdl-32366576

ABSTRACT

Acinetobacter baumannii has emerged as an important etiological agent of hospital-related infections, especially nosocomial pneumonia. The virulence factors of this bacterium and their interactions with the cells and molecules of the immune system just recently began to be extensively studied. Here, we investigated the impact of alveolar macrophages on A. baumannii pneumonia using a mouse model of infection and a flexible tissue culture system. We hypothesized that depletion of macrophages would enhance sepsis and severity of A. baumannii disease. We showed that macrophages are important for modulating the antibacterial function of neutrophils and play an important role in eradicating A. baumannii infection in vivo Our findings suggest that in the absence of macrophages in the lungs, A. baumannii replicates significantly, and host proinflammatory cytokines are considerably reduced. Neutrophils are abundantly recruited to pulmonary tissue, releasing high amounts of reactive oxygen species and causing extensive tissue damage. The ability of A. baumannii to form biofilms and resist oxidative stress in the respiratory tract facilitates systemic dissemination and ultimately death of infected C57BL/6 mice. These results provide novel information regarding A. baumannii pathogenesis and may be important for the development of therapies aimed at reducing morbidity and mortality associated with this emerging bacterial pathogen.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/physiology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Neutrophils/immunology , Sepsis/immunology , Sepsis/microbiology , Acinetobacter Infections/mortality , Acinetobacter Infections/pathology , Animals , Clodronic Acid/pharmacology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Female , Lung/immunology , Lung/metabolism , Lung/microbiology , Lung/pathology , Mice , Models, Biological , Neutrophils/metabolism , Oxidation-Reduction , Prognosis , Reactive Oxygen Species/metabolism , Superoxides/metabolism
19.
Front Immunol ; 11: 708, 2020.
Article in English | MEDLINE | ID: mdl-32391015

ABSTRACT

Acinetobacter baumannii is one of the dominating causes of nosocomial pneumonia, however, very little is known about the host immune response associated with pathogenesis of A. baumannii infection. Here, we used a hypervirulent A. baumannii to establish an acute lethal pneumonia, supported by high bacterial burdens, severe inflammatory cells infiltration and lung damage. The lung transcriptome changes in response to A. baumannii lethal pneumonia were detected by RNA sequencing. The results showed that 6,288 host genes changed expression, with 3,313 upregulated genes and 2,975 downregulated genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that genes related to TNF, cytokine-cytokine receptor interaction, Toll-like receptor, NOD-like receptor, NF-κB, Jak-STAT, HIF-1 signaling pathways, apoptosis, and phagosome were significantly upregulated. Whereas, genes associated with PI3K-AKT signaling pathway, glycolysis/gluconeogenesis, amino acid and fatty acid metabolism were downregulated. Immune cell typing highlighted the inflammatory response of innate immune cells headed by neutrophils. The reliability of RNA sequencing results were verified with selected differentially expressed genes by real-time PCR. This work provides an insight into the pathogenesis of lethal A. baumannii lung infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Immunity, Innate/genetics , Lung/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/mortality , Transcriptome , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Acute Disease , Animals , Disease Models, Animal , Down-Regulation/genetics , Female , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , RNA-Seq , Real-Time Polymerase Chain Reaction , Up-Regulation/genetics
20.
Front Immunol ; 11: 76, 2020.
Article in English | MEDLINE | ID: mdl-32153560

ABSTRACT

Recent years have seen an unprecedented rise in the incidence of multidrug-resistant (MDR) Gram-negative bacteria (GNBs) such as Acinetobacter and Klebsiella species. In view of the shortage of novel drugs in the pipeline, alternative strategies to prevent, and treat infections by GNBs are urgently needed. Previously, we have reported that the Candida albicans hypha-regulated protein Hyr1 shares striking three-dimensional structural homology with cell surface proteins of Acinetobacter baumannii. Moreover, active vaccination with rHyr1p-N or passive immunization with anti-Hyr1p polyclonal antibody protects mice from Acinetobacter infection. In the present study, we use molecular modeling to guide design of monoclonal antibodies (mAbs) generated against Hyr1p and show them to bind to priority surface antigens of Acinetobacter and Klebsiella pneumoniae. The anti-Hyr1 mAbs block damage to primary endothelial cells induced by the bacteria and protect mice from lethal pulmonary infections mediated by A. baumannii or K. pneumoniae. Our current studies emphasize the potential of harnessing Hyr1p mAbs as a cross-kingdom immunotherapeutic strategy against MDR GNBs.


Subject(s)
Acinetobacter Infections/therapy , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/metabolism , Candida albicans/immunology , Fungal Proteins/immunology , Gram-Negative Bacteria/immunology , Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Animals , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Immunoglobulins , Klebsiella pneumoniae/immunology , Mice , Pneumonia/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...