Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 169
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(27): e2402422121, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38923984

RESUMEN

Maintenance of DNA integrity is essential to all forms of life. DNA damage generated by reaction with genotoxic chemicals results in deleterious mutations, genome instability, and cell death. Pathogenic bacteria encounter several genotoxic agents during infection. In keeping with this, the loss of DNA repair networks results in virulence attenuation in several bacterial species. Interstrand DNA crosslinks (ICLs) are a type of DNA lesion formed by covalent linkage of opposing DNA strands and are particularly toxic as they interfere with replication and transcription. Bacteria have evolved specialized DNA glycosylases that unhook ICLs, thereby initiating their repair. In this study, we describe AlkX, a DNA glycosylase encoded by the multidrug resistant pathogen Acinetobacter baumannii. AlkX exhibits ICL unhooking activity similar to that of its Escherichia coli homolog YcaQ. Interrogation of the in vivo role of AlkX revealed that its loss sensitizes cells to DNA crosslinking and impairs A. baumannii colonization of the lungs and dissemination to distal tissues during pneumonia. These results suggest that AlkX participates in A. baumannii pathogenesis and protects the bacterium from stress conditions encountered in vivo. Consistent with this, we found that acidic pH, an environment encountered during host colonization, results in A. baumannii DNA damage and that alkX is induced by, and contributes to, defense against acidic conditions. Collectively, these studies reveal functions for a recently described class of proteins encoded in a broad range of pathogenic bacterial species.


Asunto(s)
Acinetobacter baumannii , Daño del ADN , ADN Glicosilasas , Acinetobacter baumannii/patogenicidad , Acinetobacter baumannii/genética , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/metabolismo , ADN Glicosilasas/metabolismo , ADN Glicosilasas/genética , Reparación del ADN , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Animales , Ratones , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Virulencia , Escherichia coli/genética , Escherichia coli/metabolismo
2.
Med Microbiol Immunol ; 212(1): 65-73, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36463365

RESUMEN

Interleukin-17 (IL-17) is involved in host defense against bacterial infection. Little is known about the role of IL-17 in A. baumannii-infected pneumonia. Our objective was to investigate the role of IL-17 in pulmonary A. baumannii infection in a mouse model. We infected C57BL/6 mice intra-tracheally (i.t.) with A. baumannii to establish pneumonia model and found A. baumannii infection elevated IL-17 expression in lungs. IL-17-deficient (Il17-/-) mice were resistant to pulmonary A. baumannii infection, showing improved mice survival, reduced bacteria burdens, and alleviated lung inflammation. Further, treatment of A. baumannii-infected Il17-/- mice with IL-17 exacerbated the severity of pneumonia. These data suggest a pathogenic role of IL-17 in pulmonary A. baumannii infection. Further, the infiltration and phagocytic function of neutrophils in broncho-alveolar lavage fluid were detected by flow cytometry. The results showed that Il17-/- mice had increased neutrophil infiltration and enhanced phagocytosis in neutrophils at the early time of infection. Treatment of mice with IL-17 suppressed phagocytic function of neutrophils. All data suggest that IL-17 promotes susceptibility of mice to pulmonary A. baumannii infection by suppressing neutrophil phagocytosis at early time of infection. Targeting IL-17 might be a potential therapeutic strategy in controlling the outcome of A. baumannii pneumonia.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Neumonía , Animales , Ratones , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Modelos Animales de Enfermedad , Interleucina-17 , Pulmón/patología , Ratones Endogámicos C57BL , Neutrófilos/patología , Neumonía/patología
3.
Exp Biol Med (Maywood) ; 247(3): 282-288, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34713732

RESUMEN

Acinetobacter baumannii is a Gram-negative bacterium responsible for many hospital-acquired infections including ventilator-associated pneumonia and sepsis. We have previously identified A. baumannii thioredoxin A protein (TrxA) as a virulence factor with a multitude of functions including reduction of protein disulfides. TrxA plays an important role in resistance to oxidative stress facilitating host immune evasion in part by alteration of type IV pili and cell surface hydrophobicity. Other virulence factors such as outer membrane vesicles (OMV) shed by bacteria have been shown to mediate bacterial intercellular communication and modulate host immune response. To investigate whether OMVs can be modulated by TrxA, we isolated OMVs from wild type (WT) and TrxA-deficient (ΔtrxA) A. baumannii clinical isolate Ci79 and carried out a functional and proteomic comparison. Despite attenuation of ΔtrxA in a mouse challenge model, pulmonary inoculation of ΔtrxA OMVs resulted in increased lung permeability compared to WT OMVs. Furthermore, ΔtrxA OMVs induced more J774 macrophage-like cell death than WT OMVs. This ΔtrxA OMV-mediated cell death was abrogated when cells were incubated with protease-K-treated OMVs suggesting OMV proteins were responsible for cytotoxicity. We therefore compared WT and mutant OMV proteins using proteomic analysis. We observed that up-regulated and unique ΔtrxA OMV proteins consisted of many membrane bound proteins involved in small molecule transport as well as proteolytic activity. Bacterial OmpA, metalloprotease, and fimbrial protein have been shown to enhance mammalian cell apoptosis through various mechanisms. Differential packaging of these proteins in ΔtrxA OMVs may contribute to the increased cytotoxicity observed in this study.


Asunto(s)
Acinetobacter baumannii/patogenicidad , Proteínas de la Membrana Bacteriana Externa/metabolismo , Membrana Externa Bacteriana/patología , Tiorredoxinas/metabolismo , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/aislamiento & purificación , Animales , Membrana Externa Bacteriana/metabolismo , Vesículas Extracelulares/patología , Interacciones Huésped-Patógeno/fisiología , Humanos , Pulmón/microbiología , Pulmón/patología , Ratones Endogámicos C57BL , Tiorredoxinas/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
4.
PLoS One ; 16(12): e0260627, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855837

RESUMEN

Infectious pneumonia induced by multidrug resistant (MDR) Acinetobacter baumannii strains is among the most common and deadly forms of healthcare acquired infections. Over the years, different strategies have been put in place to increase host susceptibility to MDR A. baumannii, since only a self-limiting pneumonia with no or limited local bacterial replication was frequently obtained in mouse models. Direct instillation into the trachea or intranasal inoculation of the bacterial suspension are the techniques used to induce the infection in most of the preclinical models of pneumonia developed to date. More recently, the oropharyngeal aspiration procedure has been widely described in the literature for a variety of purposes including pathogens administration. Aim of this study was to compare the oropharyngeal aspiration technique to the intranasal inoculation and intratracheal instillation in the ability of inducing a consistent lung infection with two MDR A. baumannii clinical isolates in immunocompromised mice. Moreover, pneumonia obtained by bacteria administration with two out of three techniques, intratracheal and oropharyngeal, was characterised in terms of histopathology of pulmonary lesions, biomarkers of inflammation level and leukocytes cells infiltration extent after mice treatment with either vehicle or the antibiotic tigecycline. The data generated clearly showed that both strains were not able to colonize the lungs when inoculated by intranasal route. By contrast, the bacterial load in lungs of mice intratracheally or oropharyngeally infected significantly increased during 26 hours of monitoring, thus highlighting the ability of these strains to generate the infection when directly instilled into the lower respiratory airways. Furthermore, the intragroup variability of mice was significantly reduced with respect to those intranasally administered. Tigecycline was efficacious in lung bacterial load and cytokines release reduction. Findings were supported by semi-quantitative histopathological evaluation of the pulmonary lesions and by inflammatory biomarkers analysis. To conclude, both intratracheal instillation and oropharyngeal aspiration techniques showed to be suitable methods for inducing a robust and consistent pneumonia infection in mice when difficult MDR A. baumannii clinical isolates were used. Noteworthy, oropharyngeal aspiration not requiring specific technical skills and dedicated equipment, was proven to be a safer, easier and faster technique in comparison to the intratracheal instillation.


Asunto(s)
Infecciones por Acinetobacter/patología , Acinetobacter baumannii/fisiología , Carga Bacteriana/métodos , Neumonía/patología , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/aislamiento & purificación , Administración Intranasal , Animales , Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/microbiología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Huésped Inmunocomprometido , Intubación Intratraqueal , Masculino , Ratones , Orofaringe/microbiología , Orofaringe/patología , Neumonía/tratamiento farmacológico , Neumonía/microbiología , Tigeciclina/farmacología , Tigeciclina/uso terapéutico
5.
Fish Shellfish Immunol ; 117: 82-94, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34311097

RESUMEN

The emergence of carbapenem-resistant Acinetobacter baumannii has increased the risk of nosocomial infections, which pose a huge health threat. There is an urgent need to develop alternative therapies, including broad-spectrum antimicrobial peptides. In this study, we designed, characterized, and studied the antibacterial, antibiofilm effects and possible mode of actions of a novel synthetic peptide Octopromycin, derived from the proline-rich protein 5 of Octopus minor. Octopromycin consists of 38 amino acids, (+5) net positive charge, high hydrophobic residue ratio (36%), and two α-helix secondary structures. The minimum inhibitory concentration and minimum bactericidal concentration against A. baumannii were 50 and 200 µg/mL, respectively. Time-kill kinetics and bacterial viability assays confirmed the concentration-dependent antibacterial activity of Octopromycin. Field emission scanning electron microscopy images clearly showed ultrastructural alterations in Octopromycin-treated A. baumannii cells. Propidium iodide penetrated into Octopromycin-treated A. baumannii cells, demonstrating the loss of cell membrane integrity. Octopromycin treatment increased the production of reactive oxygen species in a concentration-dependent manner, and it inhibited the biofilm formation and showed biofilm eradication activity against A. baumannii. In vitro and in vivo safety evaluation revealed that Octopromycin was nontoxic to HEK293T and Raw 264.7 cells (<400 µg/mL), as well as mice red blood cells (<300 µg/mL), and zebrafish embryos (<4 µg/mL). An in vivo study results revealed that the A. baumannii-infected fish treated with Octopromycin exhibited a significantly higher relative percent survival (37.5%) than the infected mock-treated fish with PBS (16.6%). Furthermore, a decreased bacterial load and fewer alterations in histological analysis confirmed the successful control of A. baumannii by Octopromycin in vivo. Collectively, the results indicate that the antibacterial peptide Octopromycin may achieve rapid control of A. baumannii through multi-target interactions; it presents a desirable therapeutic option for the prevention and control of the infections.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Antibacterianos/uso terapéutico , Péptidos Catiónicos Antimicrobianos/uso terapéutico , Enfermedades de los Peces/tratamiento farmacológico , Octopodiformes , Infecciones por Acinetobacter/patología , Infecciones por Acinetobacter/veterinaria , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/crecimiento & desarrollo , Acinetobacter baumannii/fisiología , Animales , Antibacterianos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , Biopelículas/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Embrión no Mamífero , Eritrocitos/efectos de los fármacos , Enfermedades de los Peces/patología , Células HEK293 , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Ratones , Células RAW 264.7 , Pez Cebra
6.
Respir Res ; 22(1): 159, 2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34022899

RESUMEN

BACKGROUND: Patients in intensive care units (ICUs) often received broad-spectrum antibiotic treatment and Acinetobacter baumannii (A.b.) and Pseudomonas aeruginosa (P.a.) were the most common pathogens causing ventilator-associated pneumonia (VAP). This study aimed to examine the effects and mechanism of mechanical ventilation (MV) on A.b.-induced lung injury and the involvement of alveolar macrophages (AMs). METHODS: C57BL/6 wild-type (WT) and c-Jun N-terminal kinase knockout (JNK1-/-) mice received MV for 3 h at 2 days after nasal instillation of A.b., P.a. (1 × 106 colony-forming unit, CFU), or normal saline. RESULTS: Intranasal instillation of 106 CFU A.b. in C57BL/6 mice induced a significant increase in total cells and protein levels in the bronchoalveolar lavage fluid (BALF) and neutrophil infiltration in the lungs. MV after A.b. instillation increases neutrophil infiltration, interleukin (IL)-6 and vascular cell adhesion molecule (VCAM) mRNA expression in the lungs and total cells, IL-6 levels, and nitrite levels in the BALF. The killing activity of AMs against A.b. was lower than against P.a. The diminished killing activity was parallel with decreased tumor necrosis factor-α production by AMs compared with A.b. Inducible nitric oxide synthase inhibitor, S-methylisothiourea, decreased the total cell number in BALF on mice receiving A.b. instillation and ventilation. Moreover, MV decreased the A.b. and P.a. killing activity of AMs. MV after A.b. instillation induced less total cells in the BALF and nitrite production in the serum of JNK1-/- mice than those of WT mice. CONCLUSION: A.b. is potent in inducing neutrophil infiltration in the lungs and total protein in the BALF. MV enhances A.b.-induced lung injury through an increase in the expression of VCAM and IL-6 levels in the BALF and a decrease in the bacteria-killing activity of AMs. A lower inflammation level in JNK1-/- mice indicates that A.b.-induced VAP causes lung injury through JNK signaling pathway in the lungs.


Asunto(s)
Infecciones por Acinetobacter/enzimología , Acinetobacter baumannii/patogenicidad , Pulmón/enzimología , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Neumonía Asociada al Ventilador/enzimología , Respiración Artificial/efectos adversos , Lesión Pulmonar Inducida por Ventilación Mecánica/enzimología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Interleucina-6/genética , Interleucina-6/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/microbiología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa 8 Activada por Mitógenos/genética , Infiltración Neutrófila , Óxido Nítrico Sintasa de Tipo II/metabolismo , Neumonía Asociada al Ventilador/microbiología , Neumonía Asociada al Ventilador/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo , Lesión Pulmonar Inducida por Ventilación Mecánica/microbiología , Lesión Pulmonar Inducida por Ventilación Mecánica/patología
7.
Sci Rep ; 11(1): 618, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436835

RESUMEN

Acinetobacter baumannii is a highly antibiotic resistant Gram-negative bacterium that causes life-threatening infections in humans with a very high mortality rate. A. baumannii is an extracellular pathogen with poorly understood virulence mechanisms. Here we report that A. baumannii employs the release of outer membrane vesicles (OMVs) containing the outer membrane protein A (OmpAAb) to promote bacterial pathogenesis and dissemination. OMVs containing OmpAAb are taken up by mammalian cells where they activate the host GTPase dynamin-related protein 1 (DRP1). OmpAAb mediated activation of DRP1 enhances its accumulation on mitochondria that causes mitochondrial fragmentation, elevation in reactive oxygen species (ROS) production and cell death. Loss of DRP1 rescues these phenotypes. Our data show that OmpAAb is sufficient to induce mitochondrial fragmentation and cytotoxicity since its expression in E. coli transfers its pathogenic properties to E. coli. A. baumannii infection in mice also induces mitochondrial damage in alveolar macrophages in an OmpAAb dependent manner. We finally show that OmpAAb is also required for systemic dissemination in the mouse lung infection model. In this study we uncover the mechanism of OmpAAb as a virulence factor in A. baumannii infections and further establish the host cell factor required for its pathogenic effects.


Asunto(s)
Infecciones por Acinetobacter/patología , Acinetobacter baumannii/fisiología , Apoptosis , Proteínas de la Membrana Bacteriana Externa/metabolismo , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Infecciones por Acinetobacter/metabolismo , Infecciones por Acinetobacter/microbiología , Proteínas de la Membrana Bacteriana Externa/genética , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo , Virulencia
8.
Biomed Pharmacother ; 135: 111034, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33388597

RESUMEN

Outer membrane protein A (OmpA) of Acinetobacter baumannii (A. baumannii) is associated with autophagy, which plays an important role in its pathogenicity. However, its exact pathophysiological role in the process of lung tissue cell autophagy remains unclear. In this study, animal and cell infection models were established by wild A. baumannii strain and An OmpA knockout mutant (OmpA-/- A. baumannii) strain. The expression levels of markers autophagy, histological change, cell viability and protein expression levels of inflammatory cytokines were examined. OmpA-/-A. baumannii was successfully constructed. The capacities of bacterial adhesion and invasion to host cells increased more obviously in the AB group and the AB + Rapa group than in the OmpA-/- AB group and AB + CQ group. The AB group and AB + Rapa group could produce double membrane vacuoles, endoplasmic reticulum dilation, mitochondrial ridge rupture, and mitochondrial vacuoles. OmpA could lead to increased LC3, AMPK, and PAMPK protein release, and decreased levels of P62, mTOR and pmTOR proteins in vivo and in vitro. OmpA caused lung pathology and the release of inflammatory cytokines. A. baumannii OmpA promotes autophagy in lung cells through the mTOR signalling pathway, which increases the bacterial colonization ability in the double-layer membrane autophagosome formed by the autophagy reaction to escape the clearance of bacteria by the host, promote the release of inflammatory mediators and aggravate the damage to the host.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/metabolismo , Autofagia , Proteínas de la Membrana Bacteriana Externa/metabolismo , Pulmón/microbiología , Neumonía Bacteriana/microbiología , Serina-Treonina Quinasas TOR/metabolismo , Infecciones por Acinetobacter/enzimología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/genética , Animales , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas de la Membrana Bacteriana Externa/genética , Modelos Animales de Enfermedad , Interacciones Huésped-Patógeno , Mediadores de Inflamación/metabolismo , Pulmón/enzimología , Pulmón/patología , Masculino , Mutación , Neumonía Bacteriana/enzimología , Neumonía Bacteriana/patología , Ratas Sprague-Dawley , Transducción de Señal
9.
Viruses ; 14(1)2021 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-35062236

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter/terapia , Acinetobacter baumannii , Myoviridae/fisiología , Terapia de Fagos , Neumonía Bacteriana/terapia , Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/virología , Animales , Antibacterianos/farmacología , Citocinas/metabolismo , Farmacorresistencia Bacteriana Múltiple , Femenino , Humanos , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Terapia de Fagos/efectos adversos , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología
10.
PLoS One ; 15(11): e0242674, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33253325

RESUMEN

Acinetobacter baumannii is a Gram negative nosocomial pathogen that has acquired increasing worldwide notoriety due to its high antibiotic resistance range and mortality rates in hospitalized patients. Therefore, it is necessary to better understand key aspects of A. baumannii pathogenesis such as host-pathogen interactions. In this report, we analyzed both gene expression and cytokine production by human neutrophils infected with A. baumannii. Our assays reveal a proinflammatory response of neutrophils after A. baumannii infection, since intracellular transcription of effector proteins such as COX-2, transcription factors, and proinflammatory cytokines resulted significantly upregulated in neutrophils infected by A. baumannii, compared with unstimulated human neutrophils. Translation and release of CXCL-8, IL-1ß and TNF-α by neutrophils was confirmed by protein quantification in culture supernatants. Results obtained in this report reinforce the importance of human neutrophils in controlling A. baumannii infections but also emphasize the proinflammatory nature of these host-pathogen interactions as a target for future immunomodulatory therapies.


Asunto(s)
Infecciones por Acinetobacter/metabolismo , Acinetobacter baumannii/metabolismo , Citocinas/biosíntesis , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Neutrófilos/metabolismo , Infecciones por Acinetobacter/patología , Humanos , Neutrófilos/microbiología , Neutrófilos/patología
11.
J Comp Pathol ; 178: 56-60, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32800110

RESUMEN

We report the first isolation of Acinetobacter kookii from a Rothschild's giraffe calf (Giraffa camelopardalis rothschildi) that had severe polyarthritis. The isolate was resistant to more than one representative of each of four classes of antibiotics (penicillins, macrolides, lincosamides and tetracyclines). As A. kookii has not been previously associated with disease in humans or animals, it may be an emerging opportunistic pathogen posing a threat to immunocompromised patients. Furthermore, as transmission of Acinetobacter spp. with similar patterns of antimicrobial resistance has been previously reported in human and animal populations, special care should be taken when handling infected animals.


Asunto(s)
Acinetobacter , Artritis/veterinaria , Jirafas , Acinetobacter/efectos de los fármacos , Acinetobacter/aislamiento & purificación , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/patología , Infecciones por Acinetobacter/veterinaria , Animales , Animales de Zoológico/microbiología , Antibacterianos/uso terapéutico , Artritis/microbiología , Enfermedades Transmisibles Emergentes/veterinaria , Resistencia a Medicamentos , Jirafas/microbiología , Masculino
12.
Molecules ; 25(14)2020 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-32708842

RESUMEN

The rapid emergence of antimicrobial resistance in Acinetobacter baumannii coupled with the dried pipeline of novel treatments has driven the search for new therapeutic modalities. Gram-negative bacteria have an extra outer membrane that serves as a permeability barrier for various hydrophobic and/or large compounds. One of the popular approaches to tackle this penetration barrier is use of potentiators or adjuvants in combination with traditional antibiotics. This study reports the in vitro potential of an antimicrobial peptide tridecaptin M in combination with other antibiotics against different strains of A. baumannii. Tridecaptin M sensitized the bacteria to rifampicin, vancomycin, and ceftazidime. Further, we observed that a tridecaptin M and rifampicin combination killed the bacteria completely in 4 h in an ex vivo blood infection model and was superior to rifampicin monotherapy. The study also found that concomitant administration of both compounds is not necessary to achieve the antimicrobial effect. Bacteria pre-treated with tridecaptin M (for 2-4 h) followed by exposure to rifampicin showed similar killing as obtained for combined treatment. Additionally, this combination hampered the survival of persister development in comparison to rifampicin alone. These findings encourage the future investigation of this combination to treat severe infections caused by extremely drug-resistant A. baumannii.


Asunto(s)
Infecciones por Acinetobacter/tratamiento farmacológico , Antibacterianos/farmacología , Péptidos/farmacología , Proteínas Citotóxicas Formadoras de Poros/farmacología , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/patogenicidad , Ceftazidima/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Péptidos/química , Rifampin/farmacología , Vancomicina/farmacología
13.
Infect Immun ; 88(7)2020 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-32366576

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii/fisiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Neutrófilos/inmunología , Sepsis/inmunología , Sepsis/microbiología , Infecciones por Acinetobacter/mortalidad , Infecciones por Acinetobacter/patología , Animales , Ácido Clodrónico/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Femenino , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Ratones , Modelos Biológicos , Neutrófilos/metabolismo , Oxidación-Reducción , Pronóstico , Especies Reactivas de Oxígeno/metabolismo , Superóxidos/metabolismo
14.
Front Immunol ; 11: 708, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32391015

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Acinetobacter baumannii/inmunología , Inmunidad Innata/genética , Pulmón/inmunología , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/mortalidad , Transcriptoma , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Enfermedad Aguda , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Femenino , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/patología , RNA-Seq , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación hacia Arriba/genética
15.
PLoS Pathog ; 16(3): e1008374, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32168364

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Acinetobacter baumannii/inmunología , Inmunidad Innata/efectos de los fármacos , Kanamicina/farmacología , Pulmón/inmunología , Neumonía Bacteriana/inmunología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/patogenicidad , Animales , Quimiocinas/inmunología , Femenino , Pulmón/patología , Ratones , Ratones Noqueados , Fagocitos/patología , Neumonía Bacteriana/microbiología
16.
Sci Rep ; 9(1): 17888, 2019 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-31784652

RESUMEN

Acinetobacter baumannii is a frequent cause of the nosocomial infections. Herein, a novel isothermal amplification technique, multiple cross displacement amplification (MCDA) is employed for detecting all A. baumannii strains and identifying the strains harboring blaOXA-23-like gene. The duplex MCDA assay, which targets the pgaD and blaOXA-23-like genes, could identify the A. baumannii isolates and differentiate these isolates harboring blaOXA-23-like gene. The disposable lateral flow biosensors (LFB) were used for analyzing the MCDA products. A total of sixty-eight isolates, include fifty-three A. baumannii strains and fifteen non-A. baumannii strains, were employed to optimize MCDA methods and determine the sensitivity, specificity and feasibility. The optimal reaction condition is found to be 63 °C within 1 h, with limit of detection at 100 fg templates per tube for pgaD and blaOXA-23-like genes in pure cultures. The specificity of this assay is 100%. Moreover, the practical application of the duplex MCDA-LFB assay was evaluated using clinical samples, and the results obtained from duplex MCDA-LFB method were consistent with conventional culture-based technique. In sum, the duplex MCDA-LFB assay appears to be a reliable, rapid and specific technique to detect all A. baumannii strains and identify these strains harboring blaOXA-23-like gene for appropriate antibiotic therapy.


Asunto(s)
Acinetobacter baumannii/genética , Técnicas Biosensibles/métodos , Farmacorresistencia Bacteriana/genética , beta-Lactamasas/genética , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/enzimología , Acinetobacter baumannii/aislamiento & purificación , Carbapenémicos/farmacología , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Humanos , Límite de Detección , Técnicas de Amplificación de Ácido Nucleico/métodos , beta-Lactamasas/metabolismo
17.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31548324

RESUMEN

Acinetobacter baumannii is an opportunistic bacterial pathogen capable of causing a variety of infections, including pneumonia, sepsis, wound, and burn infections. A. baumannii is an increasing threat to public health due to the prevalence of multidrug-resistant strains, leading the World Health Organization to declare A. baumannii a "Priority 1: Critical" pathogen, for which the development of novel antimicrobials is desperately needed. Zinc (Zn) is an essential nutrient that pathogenic bacteria, including A. baumannii, must acquire from their hosts in order to survive. Consequently, vertebrate hosts have defense mechanisms to sequester Zn from invading bacteria through a process known as nutritional immunity. Here, we describe a Znuptake (Znu) system that enables A. baumannii to overcome this host-imposed Zn limitation. The Znu system consists of an inner membrane ABC transporter and an outer membrane TonB-dependent receptor. Strains of A. baumannii lacking any individual Znu component are unable to grow in Zn-starved conditions, including in the presence of the host nutritional immunity protein calprotectin. The Znu system contributes to Zn-limited growth by aiding directly in the uptake of Zn into A. baumannii cells and is important for pathogenesis in murine models of A. baumannii infection. These results demonstrate that the Znu system allows A. baumannii to subvert host nutritional immunity and acquire Zn during infection.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Proteínas de Transporte de Catión/genética , Zinc/metabolismo , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/patología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL
18.
Autops. Case Rep ; 9(3): e2019106, July-Sept. 2019. ilus
Artículo en Inglés | LILACS | ID: biblio-1016813

RESUMEN

Acinetobacter baumannii has emerged as a pan-resistant superbug causing fatal infections in vulnerable patients. This report is the case of an immunosuppressed transplant patient with a fatal pneumonia due to pan-resistant Acinetobacter baumannii. Alternative therapy for resistant Acinetobacter infection is currently the subject of intense interest and research. This report illustrates the features of this type of emerging infectious disease and reviews some of the novel approaches to treatment.


Asunto(s)
Humanos , Masculino , Persona de Mediana Edad , Infecciones por Acinetobacter/patología , Farmacorresistencia Bacteriana , Neumonía/complicaciones , Autopsia , Infecciones por Acinetobacter/terapia , Resultado Fatal
19.
PLoS One ; 14(7): e0219824, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31318907

RESUMEN

In 2017, the WHO identified Acinetobacter baumannii as the top priority for the development of new antibiotics. Despite the need for new antibiotics, there remains a lack of well validated preclinical tools for A. baumannii. Here, we characterize and validate a mouse model for A. baumannii translational research. Antibiotic sensitivity for meropenem, amikacin, and polymyxin b was determined by the broth microdilution MIC assay. LD100 inoculums, in both blood and lung infection models, were determined in male and female C3HeB/FeJ mice that were challenged with various A. baumannii clinical isolates. Blood (blood infection model) or blood and lung tissue (lung infection model) were collected from infected mice at 2 and 18 hours and the bacterial burden was determined by quantitative culture. Blood chemistry was analyzed using the iStat system. Cytokines (IL-1ß, TNF, IL-6, and IL-10) were measured in the blood and lung homogenate by ELISA assay. Lung sections (H&E stains) were scored by a pathologist. In the blood infection model, the cytokines and physiological data indicate that mice become moribund due to sepsis (low blood pH, falling bicarbonate, and a rising base deficit), whereas mice become moribund due to respiratory failure (low blood pH, rising bicarbonate, and a falling base deficit) in the oral aspiration pneumonia model. We also characterized the timing of changes in various clinical and biomarker endpoints, which can serve as a basis for future interventional studies. Susceptibility was generally similar across gender and infection route. However, we did observe that female mice were approximately 2-fold more sensitive to LAC-4 ColR in the blood infection model. We also observed that female mice were more than 10-fold more resistant to VA-AB41 in the oral aspiration pneumonia model. These results establish parameters to follow in order to assess efficacy of novel preventative and therapeutic approaches for these infections.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Acinetobacter baumannii , Infecciones por Acinetobacter/patología , Acinetobacter baumannii/efectos de los fármacos , Animales , Antibacterianos/farmacología , Biomarcadores , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Masculino , Ratones , Pruebas de Sensibilidad Microbiana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA