Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
1.
BMC Genomics ; 25(1): 791, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160492

ABSTRACT

Acinetobacter baumannii is a gram-negative bacillus prevalent in nature, capable of thriving under various environmental conditions. As an opportunistic pathogen, it frequently causes nosocomial infections such as urinary tract infections, bacteremia, and pneumonia, contributing to increased morbidity and mortality in clinical settings. Consequently, developing novel vaccines against Acinetobacter baumannii is of utmost importance. In our study, we identified 10 highly conserved antigenic proteins from the NCBI and UniProt databases for epitope mapping. We subsequently screened and selected 8 CTL, HTL, and LBL epitopes, integrating them into three distinct vaccines constructed with adjuvants. Following comprehensive evaluations of immunological and physicochemical parameters, we conducted molecular docking and molecular dynamics simulations to assess the efficacy and stability of these vaccines. Our findings indicate that all three multi-epitope mRNA vaccines designed against Acinetobacter baumannii are promising; however, further animal studies are required to confirm their reliability and effectiveness.


Subject(s)
Acinetobacter baumannii , Bacterial Vaccines , Computational Biology , Acinetobacter baumannii/immunology , Acinetobacter baumannii/genetics , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Computational Biology/methods , Epitopes/immunology , Epitopes/chemistry , Molecular Docking Simulation , Acinetobacter Infections/prevention & control , Acinetobacter Infections/immunology , Epitope Mapping , mRNA Vaccines , Molecular Dynamics Simulation , Humans , RNA, Messenger/genetics , RNA, Messenger/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Bacterial Proteins/immunology , Bacterial Proteins/genetics , Bacterial Proteins/chemistry
2.
PLoS Pathog ; 16(3): e1008374, 2020 03.
Article in English | MEDLINE | ID: mdl-32168364

ABSTRACT

Antimicrobial resistance is increasing in pathogenic bacteria. Yet, the effect of antibiotic exposure on resistant bacteria has been underexplored and may affect pathogenesis. Here we describe the discovery that propagation of the human pathogen Acinetobacter baumannii in an aminoglycoside antibiotic results in alterations to the bacterium that interact with lung innate immunity resulting in enhanced bacterial clearance. Co-inoculation of mice with A. baumannii grown in the presence and absence of the aminoglycoside, kanamycin, induces enhanced clearance of a non-kanamycin-propagated strain. This finding can be replicated when kanamycin-propagated A. baumannii is killed prior to co-inoculation of mice, indicating the enhanced bacterial clearance results from interactions with innate host defenses in the lung. Infection with kanamycin-propagated A. baumannii alters the kinetics of phagocyte recruitment to the lung and reduces pro- and anti-inflammatory cytokine and chemokine production in the lung and blood. This culminates in reduced histopathologic evidence of lung injury during infection despite enhanced bacterial clearance. Further, the antibacterial response induced by killed aminoglycoside-propagated A. baumannii enhances the clearance of multiple clinically relevant Gram-negative pathogens from the lungs of infected mice. Together, these findings exemplify cooperation between antibiotics and the host immune system that affords protection against multiple antibiotic-resistant bacterial pathogens. Further, these findings highlight the potential for the development of a broad-spectrum therapeutic that exploits a similar mechanism to that described here and acts as an innate immunity modulator.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Immunity, Innate/drug effects , Kanamycin/pharmacology , Lung/immunology , Pneumonia, Bacterial/immunology , Acinetobacter Infections/pathology , Acinetobacter baumannii/pathogenicity , Animals , Chemokines/immunology , Female , Lung/pathology , Mice , Mice, Knockout , Phagocytes/pathology , Pneumonia, Bacterial/microbiology
3.
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
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.
Infect Immun ; 88(3)2020 02 20.
Article in English | MEDLINE | ID: mdl-31792075

ABSTRACT

Acinetobacter baumannii is an emerging opportunistic pathogen that primarily infects critically ill patients in nosocomial settings. Because of its rapid acquisition of antibiotic resistance, infections caused by A. baumannii have become extremely difficult to treat, underlying the importance of identifying new antimicrobial targets for this pathogen. Manganese (Mn) is an essential nutrient metal required for a number of bacterial processes, including the response to oxidative stress. Here, we show that exogenous Mn can restore A. baumannii viability in the presence of reactive oxygen species (ROS). This restoration is not dependent on the high-affinity Nramp family Mn transporter, MumT, as a ΔmumT mutant is no more sensitive to hydrogen peroxide (H2O2) killing than wild-type A. baumannii However, mumR, which encodes the transcriptional regulator of mumT, is critical for growth and survival in the presence of H2O2, suggesting that MumR regulates additional genes that contribute to H2O2 resistance. RNA sequencing revealed a role for mumR in regulating the activity of a number of metabolic pathways, including two pathways, phenylacetate and gamma-aminobutyric acid catabolism, which were found to be important for resisting killing by H2O2 Finally, ΔmumR exhibited reduced fitness in a murine model of pneumonia, indicating that MumR-regulated gene products are crucial for protection against the host immune response. In summary, these results suggest that MumR facilitates resistance to the host immune response by activating a transcriptional program that is critical for surviving both Mn starvation and oxidative stress.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Gene Expression Regulation, Bacterial/physiology , Manganese/metabolism , Membrane Transport Proteins/physiology , Oxidative Stress/physiology , Acinetobacter baumannii/genetics , Animals , Immunity, Innate/physiology , Membrane Transport Proteins/genetics , Mice , Reactive Oxygen Species/metabolism
6.
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
7.
PLoS Pathog ; 14(5): e1007056, 2018 05.
Article in English | MEDLINE | ID: mdl-29746596

ABSTRACT

Different pathogens share similar medical settings and rely on similar virulence strategies to cause infections. We have previously applied 3-D computational modeling and bioinformatics to discover novel antigens that target more than one human pathogen. Active and passive immunization with the recombinant N-terminus of Candida albicans Hyr1 (rHyr1p-N) protect mice against lethal candidemia. Here we determine that Hyr1p shares homology with cell surface proteins of the multidrug resistant Gram negative bacterium, Acinetobacter baumannii including hemagglutinin (FhaB) and outer membrane protein A (OmpA). The A. baumannii OmpA binds to C. albicans Hyr1p, leading to a mixed species biofilm. Deletion of HYR1, or blocking of Hyr1p using polyclonal antibodies, significantly reduce A. baumannii binding to C. albicans hyphae. Furthermore, active vaccination with rHyr1p-N or passive immunization with polyclonal antibodies raised against specific peptide motifs of rHyr1p-N markedly improve survival of diabetic or neutropenic mice infected with A. baumannii bacteremia or pneumonia. Antibody raised against one particular peptide of the rHyr1p-N sequence (peptide 5) confers majority of the protection through blocking A. baumannii invasion of host cells and inducing death of the bacterium by a putative iron starvation mechanism. Anti-Hyr1 peptide 5 antibodies also mitigate A. baumannii /C. albicans mixed biofilm formation in vitro. Consistent with our bioinformatic analysis and structural modeling of Hyr1p, anti-Hyr1p peptide 5 antibodies bound to A. baumannii FhaB, OmpA, and an outer membrane siderophore binding protein. Our studies highlight the concept of cross-kingdom vaccine protection against high priority human pathogens such as A. baumannii and C. albicans that share similar ecological niches in immunocompromised patients.


Subject(s)
Fungal Proteins/immunology , Fungal Proteins/pharmacology , Acinetobacter/drug effects , Acinetobacter Infections/immunology , Acinetobacter baumannii/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antibodies, Bacterial/immunology , Bacteria/immunology , Bacterial Infections , Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines/immunology , Biofilms , Candida albicans/metabolism , Candida albicans/pathogenicity , Fungal Proteins/metabolism , Immunization, Passive , Immunotherapy , Mice , Mice, Inbred BALB C , Vaccination
8.
Microb Pathog ; 143: 104114, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32145321

ABSTRACT

Infections caused by multi-drug resistance Acinetobacter baumannii are increasing worldwide. Discovery of the vaccine against this bacterium as a cost-effective and preventive strategy seems necessary. This study has introduced 11 new putative vaccine candidates against A. baumannii using the reverse vaccinology method. We considered 33 genomes of A. baumannii strains and selected the outer membrane and secreted proteins as putative vaccine candidates using Vaxign web tool. Finally, 11 proteins were confirmed as promising vaccine candidates. These targets belonged to proteins involved in cell division (NlpD), fimbria or pili assembly (FimA, PapC, and PapC associated with usher system), iron acquisition (FhuA, BfnH, FatA-like protein, and IutA), DcaP-like protein and two novel hypothetical proteins (HP-1 and HP-2). The analysis of linear and conformational B-cell epitopes showed that the outer membrane proteins including DcaP-like protein and HP-2 had high conserved surface-exposed epitopes that they can consider as excellent putative vaccine targets in the upcoming immunological assays.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii/immunology , Bacterial Vaccines/immunology , Acinetobacter Infections/immunology , Acinetobacter baumannii/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/genetics , Cell Division/immunology , Epitopes/immunology , Fimbriae, Bacterial/immunology , Humans , Vaccinology/methods
9.
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
10.
Infect Immun ; 87(5)2019 03.
Article in English | MEDLINE | ID: mdl-30782860

ABSTRACT

Coagulation and inflammation are interconnected, suggesting that coagulation plays a key role in the inflammatory response to pathogens. A phenome-wide association study (PheWAS) was used to identify clinical phenotypes of patients with a polymorphism in coagulation factor X. Patients with this single nucleotide polymorphism (SNP) were more likely to be hospitalized with hemostatic and infection-related disorders, suggesting that factor X contributes to the immune response to infection. To investigate this, we modeled infections by human pathogens in a mouse model of factor X deficiency. Factor X-deficient mice were protected from systemic Acinetobacter baumannii infection, suggesting that factor X plays a role in the immune response to A. baumannii Factor X deficiency was associated with reduced cytokine and chemokine production and alterations in immune cell population during infection: factor X-deficient mice demonstrated increased abundance of neutrophils, macrophages, and effector T cells. Together, these results suggest that factor X activity is associated with an inefficient immune response and contributes to the pathology of A. baumannii infection.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter Infections/physiopathology , Acinetobacter baumannii/immunology , Factor X/genetics , Factor X/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Phenotype , Polymorphism, Genetic
11.
Infect Immun ; 87(1)2019 01.
Article in English | MEDLINE | ID: mdl-30297527

ABSTRACT

Acinetobacter baumannii is a Gram-negative opportunistic pathogen that causes diverse infections, including pneumonia, bacteremia, and wound infections. Due to multiple intrinsic and acquired antimicrobial-resistance mechanisms, A. baumannii isolates are commonly multidrug resistant, and infections are notoriously difficult to treat. The World Health Organization recently highlighted carbapenem-resistant A. baumannii as a "critical priority" for the development of new antimicrobials because of the risk to human health posed by this organism. Therefore, it is important to discover the mechanisms used by A. baumannii to survive stresses encountered during infection in order to identify new drug targets. In this study, by use of in vivo imaging, we identified hydrogen peroxide (H2O2) as a stressor produced in the lung during A. baumannii infection and defined OxyR as a transcriptional regulator of the H2O2 stress response. Upon exposure to H2O2, A. baumannii differentially transcribes several hundred genes. However, the transcriptional upregulation of genes predicted to detoxify hydrogen peroxide is abolished in an A. baumannii strain in which the transcriptional regulator oxyR is genetically inactivated. Moreover, inactivation of oxyR in both antimicrobial-susceptible and multidrug-resistant A. baumannii strains impairs growth in the presence of H2O2 OxyR is a direct regulator of katE and ahpF1, which encode the major H2O2-degrading enzymes in A. baumannii, as confirmed through measurement of promoter binding by recombinant OxyR in electromobility shift assays. Finally, an oxyR mutant is less fit than wild-type A. baumannii during infection of the murine lung. This work reveals a mechanism used by this important human pathogen to survive H2O2 stress encountered during infection.


Subject(s)
Acinetobacter baumannii/drug effects , Anti-Infective Agents, Local/metabolism , Gene Expression Regulation, Bacterial , Hydrogen Peroxide/metabolism , Oxidants/metabolism , Repressor Proteins/metabolism , Stress, Physiological , Acinetobacter Infections/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/physiology , Animals , Mice
12.
BMC Microbiol ; 19(1): 259, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752683

ABSTRACT

BACKGROUND: Multidrug resistant Acinetobacter baumannii is one of the major infection agents causing nosocomial pneumonia. Therefore, new therapeutic approaches against this bacterium are needed. Surface-exposed proteins from bacterial pathogens are implicated in a variety of virulence-related traits and are considered as promising candidates for vaccine development. RESULTS: We show in this study that a large Blp1 protein from opportunistic pathogen A. baumannii is encoded in all examined clinical strains of globally spread international clonal lineages I (IC I) and II (IC II). The two blp1 gene variants exhibit lineage-specific distribution profile. By characterization of blp1 deletion mutants and their complementation with blp1 alleles we show that blp1 gene is required for A. baumannii biofilm formation and adhesion to epithelial cells in IC I strain but not in the IC II strain. Nevertheless both alleles are functional in restoring the deficient phenotypes of IC I strain. Moreover, the blp1 gene is required for the establishing of A. baumannii virulence phenotype in nematode and murine infection models. Additionally, we demonstrate that C-terminal 711 amino acid fragment of Blp1 elicits an efficient protection to lethal A. baumannii infection in a murine model using active and passive immunization approaches. Antiserum obtained against Blp1-specific antigen provides opsonophagocytic killing of A. baumannii in vitro. CONCLUSIONS: Lineage-specific variants of surface-exposed components of bacterial pathogens complicate the development of new therapeutic approaches. Though we demonstrated different impact of Blp1 variants on adherence of IC I and IC II strains, Blp1-specific antiserum neutralized A. baumannii strains of both clonal lineages. Together with the observed increased survival rate in vaccinated mice these results indicate that A. baumannii Blp1 protein could be considered as a new vaccine candidate.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/pathogenicity , Virulence Factors/genetics , Virulence Factors/immunology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Acinetobacter baumannii/immunology , Animals , Biofilms , Caenorhabditis elegans , Cell Adhesion , Cell Line , Disease Models, Animal , Female , Genetic Variation , Mice
13.
Microb Pathog ; 131: 9-14, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30930220

ABSTRACT

Acinetobacter baumannii is considered as a major cause of nosocomial infection worldwide. Various vaccine formulations have been mostly studied based on secreted or surface-exposed proteins of A. baumannii in murine models. Serum resistance proteins are critical virulence factors in bloodstream infections. AbOmpA and PKF are two major factors involved in serum resistance and could be considered as promising vaccine targets. In this study IgG1, IgG2c, Total-IgG concentrations, survival rates and spleen bacterial loads were studied in C57/BL mice model according to PKF, AbOmpA and AbOmpA + PKF vaccine formulations. The findings showed significant raises of IgG2c and Total-IgG in all three vaccinated groups in comparison with the control group. Whereas, there were low concentrations of IgG1 in all immunization plans. Colony counts of mice spleen showed the bacterial load of PKF plan had the most decrease of bacterial load (DBL = 5 log10 CFU/g). Taken together, this evaluation indicated that PKF vaccination plan induced a polarized Th1 response and rendered an effective protection against bloodstream infection caused by A. baumannii.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/pathogenicity , Antibody Formation/immunology , Bacterial Outer Membrane Proteins/immunology , R Factors/blood , Sepsis/microbiology , Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Animals , Bacterial Load , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cloning, Molecular , Disease Models, Animal , Genes, Bacterial/genetics , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred C57BL , R Factors/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Spleen/immunology , Survival Rate , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Virulence Factors/genetics , Virulence Factors/immunology
14.
Scand J Immunol ; 90(1): e12769, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31006127

ABSTRACT

Acinetobacter baumannii (A baumannii) is an emerging nosocomial pathogenic bacterium which leads to hospital infections. The increase in drug-resistant A baumannii strains makes it difficult to control by using common antibiotics. The development of effective vaccines is an alternative means to avoid A baumannii infections. In the present study, Balb/c mice were inoculated intratracheally with 30 µg of OmpK/Omp22 fusion protein alone or OmpK/Omp22 formulated with MF59 adjuvant. After two times of boosting at day 14 and 21, the antigen-specific antibody levels and the protective immunity against A baumannii challenge were evaluated. The results showed that the OmpK/Omp22 formulated with MF59 immunized mice produced much higher level of antigen-specific antibodies compared to mice immunized with OmpK/Omp22 alone (P < 0.01). Mice immunized with 30 µg of OmpK/Omp22 formulated with MF59 also provided more potent protection post-challenge, which showed lower bacterial loads in the blood and lung tissue, lower level of blood inflammatory cytokines and higher survival rate (83.3%) than mice immunized with OmpK/Omp22 alone (P < 0.001). In conclusion, this study demonstrated that OmpK/Omp22 fusion protein adjuvanted with MF59 induced superior immune response and better protection than OmpK/Omp22 alone through intratracheal inoculation in mice.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/physiology , Adjuvants, Immunologic , Bacterial Outer Membrane Proteins/immunology , Bacterial Vaccines/immunology , Recombinant Fusion Proteins/immunology , Squalene/immunology , Animals , Bacterial Load , Cross Infection , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Mice , Mice, Inbred BALB C , Polysorbates , Trachea/metabolism , Vaccination
15.
J Immunol ; 199(8): 2803-2814, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28855313

ABSTRACT

Acinetobacter baumannii is a bacterial pathogen with increasing impact in healthcare settings, due in part to this organism's resistance to many antimicrobial agents, with pneumonia and bacteremia as the most common manifestations of disease. A significant proportion of clinically relevant A. baumannii strains are resistant to killing by normal human serum (NHS), an observation supported in this study by showing that 12 out of 15 genetically diverse strains of A. baumannii are resistant to NHS killing. To expand our understanding of the genetic basis of A. baumannii serum resistance, a transposon (Tn) sequencing (Tn-seq) approach was used to identify genes contributing to this trait. An ordered Tn library in strain AB5075 with insertions in every nonessential gene was subjected to selection in NHS. We identified 50 genes essential for the survival of A. baumannii in NHS, including already known serum resistance factors, and many novel genes not previously associated with serum resistance. This latter group included the maintenance of lipid asymmetry genetic pathway as a key determinant in protecting A. baumannii from the bactericidal activity of NHS via the alternative complement pathway. Follow-up studies validated the role of eight additional genes identified by Tn-seq in A. baumannii resistance to killing by NHS but not by normal mouse serum, highlighting the human species specificity of A. baumannii serum resistance. The identification of a large number of genes essential for serum resistance in A. baumannii indicates the degree of complexity needed for this phenotype, which might reflect a general pattern that pathogens rely on to cause serious infections.


Subject(s)
Acinetobacter Infections/microbiology , Acinetobacter baumannii/genetics , Blood Bactericidal Activity , Pneumonia/microbiology , Virulence , Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Acinetobacter baumannii/pathogenicity , Animals , Complement Pathway, Alternative/genetics , DNA Transposable Elements/genetics , DNA, Bacterial/analysis , Humans , Lipid Metabolism/genetics , Mice , Pneumonia/immunology , Serum Response Factor/genetics , Species Specificity , Transcriptome , Virulence/genetics
16.
Proc Natl Acad Sci U S A ; 113(34): 9599-604, 2016 08 23.
Article in English | MEDLINE | ID: mdl-27506797

ABSTRACT

Innate cellular immune responses are a critical first-line defense against invading bacterial pathogens. Leukocyte migration from the bloodstream to a site of infection is mediated by chemotactic factors that are often host-derived. More recently, there has been a greater appreciation of the importance of bacterial factors driving neutrophil movement during infection. Here, we describe the development of a zebrafish infection model to study Acinetobacter baumannii pathogenesis. By using isogenic A. baumannii mutants lacking expression of virulence effector proteins, we demonstrated that bacterial drivers of disease severity are conserved between zebrafish and mammals. By using transgenic zebrafish with fluorescent phagocytes, we showed that a mutation of an established A. baumannii global virulence regulator led to marked changes in neutrophil behavior involving rapid neutrophil influx to a localized site of infection, followed by prolonged neutrophil dwelling. This neutrophilic response augmented bacterial clearance and was secondary to an impaired A. baumannii phenylacetic acid catabolism pathway, which led to accumulation of phenylacetate. Purified phenylacetate was confirmed to be a neutrophil chemoattractant. These data identify a previously unknown mechanism of bacterial-guided neutrophil chemotaxis in vivo, providing insight into the role of bacterial metabolism in host innate immune evasion. Furthermore, the work provides a potentially new therapeutic paradigm of targeting a bacterial metabolic pathway to augment host innate immune responses and attenuate disease.


Subject(s)
Acinetobacter Infections/immunology , Acinetobacter baumannii/genetics , Bacterial Proteins/genetics , Chemotaxis/drug effects , Phenylacetates/metabolism , Transcription Factors/genetics , Virulence Factors/genetics , Acinetobacter Infections/microbiology , Acinetobacter Infections/pathology , Acinetobacter baumannii/immunology , Acinetobacter baumannii/metabolism , Acinetobacter baumannii/pathogenicity , Animals , Animals, Genetically Modified , Chemotaxis/immunology , Embryo, Nonmammalian , Female , Gene Expression , Immunity, Innate , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred BALB C , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/microbiology , Phenylacetates/pharmacology , Transcription Factors/deficiency , Virulence , Virulence Factors/deficiency , Zebrafish
17.
Clin Microbiol Rev ; 30(1): 409-447, 2017 01.
Article in English | MEDLINE | ID: mdl-27974412

ABSTRACT

Acinetobacter is a complex genus, and historically, there has been confusion about the existence of multiple species. The species commonly cause nosocomial infections, predominantly aspiration pneumonia and catheter-associated bacteremia, but can also cause soft tissue and urinary tract infections. Community-acquired infections by Acinetobacter spp. are increasingly reported. Transmission of Acinetobacter and subsequent disease is facilitated by the organism's environmental tenacity, resistance to desiccation, and evasion of host immunity. The virulence properties demonstrated by Acinetobacter spp. primarily stem from evasion of rapid clearance by the innate immune system, effectively enabling high bacterial density that triggers lipopolysaccharide (LPS)-Toll-like receptor 4 (TLR4)-mediated sepsis. Capsular polysaccharide is a critical virulence factor that enables immune evasion, while LPS triggers septic shock. However, the primary driver of clinical outcome is antibiotic resistance. Administration of initially effective therapy is key to improving survival, reducing 30-day mortality threefold. Regrettably, due to the high frequency of this organism having an extreme drug resistance (XDR) phenotype, early initiation of effective therapy is a major clinical challenge. Given its high rate of antibiotic resistance and abysmal outcomes (up to 70% mortality rate from infections caused by XDR strains in some case series), new preventative and therapeutic options for Acinetobacter spp. are desperately needed.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter/pathogenicity , Acinetobacter/drug effects , Acinetobacter/metabolism , Acinetobacter Infections/immunology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Clinical Trials as Topic , Community-Acquired Infections/drug therapy , Community-Acquired Infections/immunology , Community-Acquired Infections/microbiology , Cross Infection/drug therapy , Cross Infection/immunology , Cross Infection/microbiology , Drug Resistance, Multiple, Bacterial/drug effects , Humans , Time-to-Treatment , Virulence Factors/immunology
18.
J Am Chem Soc ; 140(28): 8639-8643, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29965749

ABSTRACT

Pseudaminic acid (Pse) has been known for participating in crucial bacterial virulence and thus is an attractive target in the development of glycoconjugate vaccine. Particularly, this therapeutic alternative was suggested to be a potential solution against antibiotic resistant Acinetobacter baumannii that poses a serious global health threat. Also, Pse was found to be involved in the exopolysaccharide (EPS) of mild antibiotic resistant A. baumannii strain 54149 ( Ab-54149) of which specific glycosyl linkage can be depolymerized by phage ΦAB6 tailspike protein (ΦAB6TSP). In this study, we found that the antibodies induced by Ab-54149 EPS was capable of recognizing a range of EPS of other clinical A. baumannii strains, and deemed as a great potential material for vaccination. To efficiently acquire homogeneous EPS-derived oligosaccharide with significant immunogenic activity for the production of glycoconjugate, we used the ΦAB6TSP for the fragmentation of Ab-54149 EPS instead of chemical methods. Moreover, insight into the ligand binding characterization of ΦAB6TSP suggested the branched Pse on the Ab-54149 EPS served as a recognition site of ΦAB6TSP. The serum boosted by ΦAB6TSP-digested product and carrier protein CRM197 conjugate complex displayed specific sensitivity toward Ab-54149 EPS with bacterial killing activity. Strikingly, Pse is an ideal epitope with strong antigenicity, profiting the application of the probe for pathogen detection and glyco-based vaccine.


Subject(s)
Acinetobacter baumannii/immunology , Bacterial Vaccines/immunology , Glycoconjugates/immunology , Polysaccharides, Bacterial/immunology , Sugar Acids/immunology , Vaccines, Conjugate/immunology , Viral Tail Proteins/immunology , Acinetobacter Infections/immunology , Acinetobacter Infections/prevention & control , Glycoside Hydrolases , Humans , Models, Molecular
19.
Microb Pathog ; 114: 147-152, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29155126

ABSTRACT

Lactoperoxidase (Lpo) and Lactoferrin (Lf) were extracted from camel colostrum milk and purified. The antibacterial activity of the two purified proteins was estimated against 14 isolates of multidrug resistance Acinetobacter baumannii. A combination of Lpo and Lf exhibited bactericidal action against A. baumannii in vitro. A mouse model of acute A. baumannii pneumonia was improved. The injection of combined Lpo and Lf after infection leads to significant clearance of A. baumannii rates in lung as well as blood culture P < 0.05 in comparing with control. Furthermore, the results showed a significant P < 0.05 reduction in the Bronchoalveolar lavage albumin concentration, lung injury and lactate dehydrogenase activity in comparing with control. In addition, the combination of Lpo and Lf treatment induced substantial elevation of IL-4 and IL10 concentrations p < 0.0 5 that helped to prevent damage caused by the inflammatory response. We concluded that combination of Lpo and Lf had a major inhibition effect against A. baumannii in comparing with imipenem as well as their immunomodulatory activity against resistant A. baumannii was increased by a synergistic effect of them as a crude combination. This study indicated two combined proteins consider as crucial strategy for practical treatment of pneumonia in the future.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter Infections/immunology , Acinetobacter baumannii/drug effects , Anti-Bacterial Agents/administration & dosage , Colostrum/chemistry , Immunologic Factors/administration & dosage , Lactoferrin/administration & dosage , Lactoperoxidase/administration & dosage , Acinetobacter Infections/genetics , Acinetobacter Infections/microbiology , Acinetobacter baumannii/physiology , Animals , Anti-Bacterial Agents/isolation & purification , Camelus , Colostrum/enzymology , Drug Resistance, Multiple, Bacterial , Drug Synergism , Female , Humans , Immunologic Factors/isolation & purification , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lactoferrin/isolation & purification , Lactoperoxidase/isolation & purification , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests
20.
Microb Pathog ; 118: 18-31, 2018 May.
Article in English | MEDLINE | ID: mdl-29524546

ABSTRACT

Acinetobacter baumannii is an opportunistic multidrug resistant pathogen. TonB-dependent copper receptor is an outer membrane protein and has a role in binding of A. baumannii to host cell via attachment to fibronectin. Moreover, it is highly expressed in biofilm community. In this study the properties of copper receptor were analyzed in silico and its vaccine potential was investigated. TonB-dependent copper and iron receptor domains plus one plug domain at N-terminal were determined by domain analysis. Topology modeling showed 22 ß-strands, 11 loops and 10 periplasmic turns. Interaction of this protein with TonB2 energy transducer was also indicated. Beside the antigenicity, this protein could take part in bacterial virulence. The more preferable 3D structure was selected amongst all 26 predicted structures, refined and used in prediction of ligand binding site and conformational epitope. The results of B and T-cell epitope mapping indicated 8 potential areas in the protein sequence and structure that seems to be able to stimulate both humoral and cellular immune responses. Based on the alignment result, this protein and all selected epitopes are extremely conserved among A. baumannii strains which can be tested as sub unit vaccine by in vivo studies.


Subject(s)
Acinetobacter baumannii/metabolism , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/immunology , Copper/metabolism , Membrane Proteins/chemistry , Membrane Proteins/immunology , Protein Conformation , Acinetobacter Infections/immunology , Acinetobacter Infections/prevention & control , Acinetobacter baumannii/pathogenicity , Amino Acid Sequence , B-Lymphocytes , Bacterial Outer Membrane Proteins/metabolism , Bacterial Vaccines , Binding Sites , Computer Simulation , Epithelial Cells/microbiology , Epitope Mapping , Epitopes/chemistry , Epitopes/immunology , Humans , Immunity, Cellular , Immunity, Humoral , Immunogenicity, Vaccine , Iron/metabolism , Ligands , Models, Molecular , Periplasm/metabolism , Protein Domains , Protein Interaction Domains and Motifs/immunology , Protein Interaction Maps/immunology , Sequence Alignment , T-Lymphocytes , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL