Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 720
Filtrar
1.
Arch Microbiol ; 206(6): 267, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762620

RESUMEN

Acinetobacter species encode for extracellularly secreted Biofilm-associated protein (Bap), a multi-domain protein with variable molecular weights reaching several hundred kilodaltons. Bap is crucial for the development of multi-dimensional structures of mature biofilms. In our investigation, we analyzed 7338 sequences of A. baumannii from the NCBI database and found that Bap or Bap-like protein (BLP) was present in 6422 (87.52%) isolates. Further classification revealed that 12.12% carried Type-1 Bap, 68.44% had Type-2, 6.91% had Type-3, 0.05% had Type-6 or SDF-Type, and 12.51% lacked Bap or BLP. The majority of isolates with Type-1, Type-2, and Type-3 Bap belonged to ST1, ST2, and ST25, respectively. Phylogenetic analysis suggested that Type-1 Bap is the most ancient, while Type-3 and SDF-Type have evolved recently. Studying the interaction of predicted Bap structures with human CEACAM-1 and PIgR showed that Bap with its BIg13 and BIg6 domains interact with the N-terminal domain of CEACAM-1, involving Arg43 and Glu40, involved in CEACAM-1 dimerization. Also, we found that recently evolved Type-3 and SDF-Type Bap showed greater interaction with CEACAM-1 and PIgR. It can be asserted that the evolution of Bap has conferred enhanced virulence characteristics to A. baumannii with increased interaction with CEACAM-1 and PIgR. Using in silico approaches, this study explores the evolutionary, physicochemical, and structural features of A. baumannii Bap and unravels its crucial role in mediating interaction with human CEACAM-1 and PIgR through detailed structure modelling. These findings advance our understanding of A. baumannii Bap and highlight its role in pathogenesis.


Asunto(s)
Acinetobacter baumannii , Proteínas Bacterianas , Biopelículas , Filogenia , Acinetobacter baumannii/genética , Acinetobacter baumannii/química , Acinetobacter baumannii/metabolismo , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Humanos , Infecciones por Acinetobacter/microbiología , Evolución Molecular , Simulación por Computador , Modelos Moleculares
2.
J Bacteriol ; 206(5): e0043523, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38661375

RESUMEN

Acinetobacter baumannii is highly resistant to antimicrobial agents, and XDR strains have become widespread. A. baumannii has developed resistance to colistin, which is considered the last resort against XDR Gram-negative bacteria, mainly caused by lipooligosaccharide (LOS) phosphoethanolamine (pEtN) and/or galactosamine (GalN) modifications induced by mutations that activate the two-component system (TCS) pmrAB. Although PmrAB of A. baumannii has been recognized as a drug resistance factor, its function as TCS, including its regulatory genes and response factors, has not been fully elucidated. In this study, to clarify the function of PmrAB as TCS, we elucidated the regulatory genes (regulon) of PmrAB via transcriptome analysis using pmrAB-activated mutant strains. We discovered that PmrAB responds to low pH, Fe2+, Zn2+, and Al3+. A. baumannii selectively recognizes Fe2+ rather than Fe3+, and a novel region ExxxE, in addition to the ExxE motif sequence, is involved in the environmental response. Furthermore, PmrAB participates in the phosphoethanolamine modification of LOS on the bacterial surface in response to metal ions such as Al3+, contributing to the attenuation of Al3+ toxicity and development of resistance to colistin and polymyxin B in A. baumannii. This study demonstrates that PmrAB in A. baumannii not only regulates genes that play an important role in drug resistance but is also involved in responses to environmental stimuli such as metal ions and pH, and this stimulation induces LOS modification. This study reveals the importance of PmrAB in the environmental adaptation and antibacterial resistance emergence mechanisms of A. baumannii. IMPORTANCE: Antimicrobial resistance (AMR) is a pressing global issue in human health. Acinetobacter baumannii is notably high on the World Health Organization's list of bacteria for which new antimicrobial agents are urgently needed. Colistin is one of the last-resort drugs used against extensively drug-resistant (XDR) Gram-negative bacteria. However, A. baumannii has become increasingly resistant to colistin, primarily by modifying its lipooligosaccharide (LOS) via activating mutations in the two-component system (TCS) PmrAB. This study comprehensively elucidates the detailed mechanism of drug resistance of PmrAB in A. baumannii as well as its biological functions. Understanding the molecular biology of these molecules, which serve as drug resistance factors and are involved in environmental recognition mechanisms in bacteria, is crucial for developing fundamental solutions to the AMR problem.


Asunto(s)
Acinetobacter baumannii , Proteínas Bacterianas , Etanolaminas , Regulación Bacteriana de la Expresión Génica , Lipopolisacáridos , Acinetobacter baumannii/genética , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Lipopolisacáridos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Etanolaminas/farmacología , Etanolaminas/metabolismo , Antibacterianos/farmacología , Metales/metabolismo , Metales/farmacología , Factores de Transcripción
3.
Nat Microbiol ; 9(5): 1244-1255, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38649414

RESUMEN

Carbapenem-resistant Acinetobacter baumannii infections have limited treatment options. Synthesis, transport and placement of lipopolysaccharide or lipooligosaccharide (LOS) in the outer membrane of Gram-negative bacteria are important for bacterial virulence and survival. Here we describe the cerastecins, inhibitors of the A. baumannii transporter MsbA, an LOS flippase. These molecules are potent and bactericidal against A. baumannii, including clinical carbapenem-resistant Acinetobacter baumannii isolates. Using cryo-electron microscopy and biochemical analysis, we show that the cerastecins adopt a serpentine configuration in the central vault of the MsbA dimer, stalling the enzyme and uncoupling ATP hydrolysis from substrate flipping. A derivative with optimized potency and pharmacokinetic properties showed efficacy in murine models of bloodstream or pulmonary A. baumannii infection. While resistance development is inevitable, targeting a clinically unexploited mechanism avoids existing antibiotic resistance mechanisms. Although clinical validation of LOS transport remains undetermined, the cerastecins may open a path to narrow-spectrum treatment modalities for important nosocomial infections.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Antibacterianos , Proteínas Bacterianas , Lipopolisacáridos , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Lipopolisacáridos/metabolismo , Animales , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/tratamiento farmacológico , Ratones , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Transporte Biológico , Pruebas de Sensibilidad Microbiana , Humanos , Microscopía por Crioelectrón , Carbapenémicos/farmacología , Carbapenémicos/metabolismo , Modelos Animales de Enfermedad , Femenino , Transportadoras de Casetes de Unión a ATP
4.
J Appl Microbiol ; 135(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38653725

RESUMEN

AIMS: Acinetobacter baumannii is a nosocomial pathogen known to be multidrug-resistant (MDR), especially to drugs of the carbapenem class. Several factors contribute to resistance, including efflux pumps, ß-lactamases, alteration of target sites, and permeability defects. In addition, outer membrane proteins (OMPs), like porins are involved in the passage of antibiotics, and their alteration could lead to resistance development. This study aimed to explore the possible involvement of porins and OMPs in developing carbapenem resistance due to differential expression. METHODS AND RESULTS: The antibiotic-susceptible and MDR isolates of A. baumannii were first studied for differences in their transcriptional levels of OMP expression and OMP profiles. The antibiotic-susceptible isolates were further treated with imipenem, and it was found that the omp genes were differentially expressed. Six of the nine genes studied were upregulated at 1 h of exposure to imipenem. Their expression gradually decreased with time, further confirmed by their OMP profile and two-dimensional gel electrophoresis. CONCLUSIONS: This study could identify OMPs that were differentially expressed on exposure to imipenem. Hence, this study provides insights into the role of specific OMPs in antibiotic resistance in A. baumannii.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Proteínas de la Membrana Bacteriana Externa , Imipenem , Pruebas de Sensibilidad Microbiana , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Imipenem/farmacología , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Antibacterianos/farmacología , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Infecciones por Acinetobacter/microbiología , Humanos , Porinas/genética , Porinas/metabolismo
5.
Artículo en Inglés | MEDLINE | ID: mdl-38632045

RESUMEN

Narrow-spectrum antibiotics are of great interest given their ability to spare the microbiome and decrease widespread antibiotic resistance compared to broad-spectrum antibiotics. Herein, we screened an in-house library of Actinobacteria strains for selective activity against Acinetobacter baumannii and successfully identified Streptomyces sp. CS-62 as a producer of a natural product with this valuable activity. Analysis of the cultures via high-resolution mass spectrometry and tandem mass spectrometry, followed by comparison with molecules in the Natural Product Atlas and the Global Natural Products Social Molecular Networking platform, suggested a novel natural product. Genome mining analysis initially supported the production of a novel kirromycin derivative. Isolation and structure elucidation via mass spectrometry and Nuclear Magnetic Resonance (NMR) analyses revealed that the active natural product was the known natural product factumycin, exposing omissions and errors in the consulted databases. While public databases are generally very useful for avoiding rediscovery of known molecules, rediscovery remains a problem due to public databases either being incomplete or having errors that result in failed dereplication. Overall, the work describes the ongoing problem of dereplication and the continued need for public database curation.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Streptomyces , Streptomyces/metabolismo , Streptomyces/genética , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/genética , Acinetobacter baumannii/efectos de los fármacos , Antibacterianos/biosíntesis , Antibacterianos/farmacología , Productos Biológicos/metabolismo , Pruebas de Sensibilidad Microbiana
6.
Biophys J ; 123(8): 979-991, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38459695

RESUMEN

COG0523 proteins, also known as nucleotide-dependent metallochaperones, are a poorly understood class of small P-loop G3E GTPases. Multiple family members play critical roles in bacterial pathogen survival during an infection as part of the adaptive response to host-mediated "nutritional immunity." Our understanding of the structure, dynamics, and molecular-level function of COG0523 proteins, apart from the eukaryotic homolog, Zng1, remains in its infancy. Here, we use X-ray absorption spectroscopy to establish that Acinetobacter baumannii (Ab) ZigA coordinates ZnII using all three cysteines derived from the invariant CXCC motif to form an S3(N/O) coordination complex, a feature inconsistent with the ZnII-bound crystal structure of a distantly related COG0523 protein of unknown function from Escherichia coli, EcYjiA. The binding of ZnII and guanine nucleotides is thermodynamically linked in AbZigA, and this linkage is more favorable for the substrate GTP relative to the product GDP. Part of this coupling originates with nucleotide-induced stabilization of the G-domain tertiary structure as revealed by global thermodynamics measurements and hydrogen-deuterium exchange mass spectrometry (HDX-MS). HDX-MS also reveals that the HDX behavior of the G2 (switch 1) loop is highly sensitive to nucleotide status and becomes more exchange labile in the GDP (product)-bound state. Significant long-range perturbation of local stability in both the G-domain and the C-terminal domain define a candidate binding pocket for a client protein that appears sensitive to nucleotide status (GDP versus GTP). We place these new insights into the structure, dynamics, and energetics of intermolecular metal transfer into the context of a model for AbZigA metallochaperone function.


Asunto(s)
Acinetobacter baumannii , Zinc , Humanos , Zinc/metabolismo , Acinetobacter baumannii/metabolismo , Nucleótidos/metabolismo , Bacterias/metabolismo , Guanosina Trifosfato/metabolismo , Unión Proteica , Guanosina Difosfato/metabolismo
7.
Mol Microbiol ; 121(5): 833-849, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38308563

RESUMEN

The nosocomial bacterium Acinetobacter baumannii is protected from antibiotic treatment by acquiring antibiotic resistances and by forming biofilms. Cell attachment, one of the first steps in biofilm formation, is normally induced by environmental metabolites. We hypothesized that vanillic acid (VA), the oxidized form of vanillin and a widely available metabolite, may play a role in A. baumannii cell attachment. We first discovered that A. baumannii actively breaks down VA through the evolutionarily conserved vanABKP genes. These genes are under the control of the repressor VanR, which we show binds directly to VanR binding sites within the vanABKP genes bidirectional promoter. VA in turn counteracts VanR inhibition. We identified a VanR binding site and searched for it throughout the genome, especially in pili encoding promoter genes. We found a VanR binding site in the pilus encoding csu operon promoter and showed that VanR binds specifically to it. As expected, a strain lacking VanR overproduces Csu pili and makes robust biofilms. Our study uncovers the role that VA plays in facilitating the attachment of A. baumannii cells to surfaces, a crucial step in biofilm formation. These findings provide valuable insights into a previously obscure catabolic pathway with significant clinical implications.


Asunto(s)
Acinetobacter baumannii , Adhesión Bacteriana , Proteínas Bacterianas , Biopelículas , Fimbrias Bacterianas , Regulación Bacteriana de la Expresión Génica , Regiones Promotoras Genéticas , Ácido Vanílico , Acinetobacter baumannii/metabolismo , Acinetobacter baumannii/genética , Acinetobacter baumannii/efectos de los fármacos , Ácido Vanílico/metabolismo , Ácido Vanílico/farmacología , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/genética , Fimbrias Bacterianas/metabolismo , Fimbrias Bacterianas/genética , Operón , Sitios de Unión , Benzaldehídos/metabolismo , Benzaldehídos/farmacología
8.
ACS Infect Dis ; 10(3): 870-878, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38311919

RESUMEN

Acinetobacter baumannii is a multidrug-resistant pathogen that has become one of the most challenging pathogens in global healthcare. Several antibiotic-resistant genes, including catB8, have been identified in the A. baumannii genome. CatB8 protein, one of the chloramphenicol acetyltransferases (Cats), is encoded by the catB8 gene. Cats can convert chloramphenicol (chl) to 3-acetyl-chl, leading to bacterial resistance to chl. Here, we present the high-resolution cocrystal structure of CatB8 with chl. The structure that we resolved showed that each monomer of CatB8 binds to four chl molecules, while its homologous protein only binds to one chl molecule. One of the newly discovered chl binding site overlaps with the site of another substrate, acetyl-CoA. Through structure-based biochemical analyses, we identified key residues for chl recruiting and acetylation of chl in CatB8. Our work is of significant importance for understanding the drug resistance of A. baumannii and the effectiveness of antibiotic treatment.


Asunto(s)
Acinetobacter baumannii , Cloranfenicol , Cloranfenicol/farmacología , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Sitios de Unión
9.
Biochemistry ; 63(5): 660-670, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38385972

RESUMEN

Bacterial cells tightly regulate the intracellular concentrations of essential transition metal ions by deploying a panel of metal-regulated transcriptional repressors and activators that bind to operator-promoter regions upstream of regulated genes. Like other zinc uptake regulator (Zur) proteins, Acinetobacter baumannii Zur represses transcription of its regulon when ZnII is replete and binds more weakly to DNA when ZnII is limiting. Previous studies established that Zur proteins are homodimeric and harbor at least two metal sites per protomer or four per dimer. CdII X-ray absorption spectroscopy (XAS) of the Cd2Zn2 AbZur metalloderivative with CdII bound to the allosteric sites reveals a S(N/O)3 first coordination shell. Site-directed mutagenesis suggests that H89 and C100 from the N-terminal DNA binding domain and H107 and E122 from the C-terminal dimerization domain comprise the regulatory metal site. KZn for this allosteric site is 6.0 (±2.2) × 1012 M-1 with a functional "division of labor" among the four metal ligands. N-terminal domain ligands H89 and C100 contribute far more to KZn than H107 and E122, while C100S AbZur uniquely fails to bind to DNA tightly as measured by an in vitro transcription assay. The heterotropic allosteric coupling free energy, ΔGc, is negative, consistent with a higher KZn for the AbZur-DNA complex and defining a bioavailable ZnII set-point of ≈6 × 10-14 M. Small-angle X-ray scattering (SAXS) experiments reveal that only the wild-type Zn homodimer undergoes allosteric switching, while the C100S AbZur fails to switch. These data collectively suggest that switching to a high affinity DNA-binding conformation involves a rotation/translation of one protomer relative to the other in a way that is dependent on the integrity of C100. We place these findings in the context of other Zur proteins and Fur family repressors more broadly.


Asunto(s)
Acinetobacter baumannii , Isoquinolinas , Sulfonamidas , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Proteínas Bacterianas/química , Sitios de Unión , Cadmio , Subunidades de Proteína , Dispersión del Ángulo Pequeño , Zinc/metabolismo , Difracción de Rayos X , Proteínas Represoras/metabolismo , Metales , ADN/metabolismo
10.
Angew Chem Int Ed Engl ; 63(19): e202402405, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38407513

RESUMEN

Antibacterial resistance is a major threat for human health. There is a need for new antibacterials to stay ahead of constantly-evolving resistant bacteria. Nucleic acid therapeutics hold promise as powerful antibiotics, but issues with their delivery hamper their applicability. Here, we exploit the siderophore-mediated iron uptake pathway to efficiently transport antisense oligomers into bacteria. We appended a synthetic siderophore to antisense oligomers targeting the essential acpP gene in Escherichia coli. Siderophore-conjugated PNA and PMO antisense oligomers displayed potent antibacterial properties. Conjugates bearing a minimal siderophore consisting of a mono-catechol group showed equally effective. Targeting the lacZ transcript resulted in dose-dependent decreased ß-galactosidase production, demonstrating selective protein downregulation. Applying this concept to Acinetobacter baumannii also showed concentration-dependent growth inhibition. Whole-genome sequencing of resistant mutants and competition experiments with the endogenous siderophore verified selective uptake through the siderophore-mediated iron uptake pathway. Lastly, no toxicity towards mammalian cells was found. Collectively, we demonstrate for the first time that large nucleic acid therapeutics can be efficiently transported into bacteria using synthetic siderophore mimics.


Asunto(s)
Acinetobacter baumannii , Antibacterianos , Catecoles , Escherichia coli , Sideróforos , Sideróforos/química , Sideróforos/farmacología , Catecoles/química , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Pruebas de Sensibilidad Microbiana , Humanos , Ácidos Nucleicos/química , Ácidos Nucleicos/metabolismo
11.
Int J Mol Sci ; 25(3)2024 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-38339095

RESUMEN

The presence of sub-minimal inhibitory concentration (sub-MIC) antibiotics in our environment is widespread, and their ability to induce antibiotic resistance is inevitable. Acinetobacter baumannii, a pathogen known for its strong ability to acquire antibiotic resistance, has recently shown clinical resistance to the last-line antibiotic tigecycline. To unravel the complex mechanism of A. baumannii drug resistance, we subjected tigecycline-susceptible, -intermediate, and -mildly-resistant strains to successive increases in sub-MIC tigecycline and ultimately obtained tigecycline-resistant strains. The proteome of both key intermediate and final strains during the selection process was analyzed using nanoLC-MS/MS. Among the more than 2600 proteins detected in all strains, we found that RND efflux pump AdeABC was associated with the adaptability of A. baumannii to tigecycline under sub-MIC pressure. qRT-PCR analysis also revealed higher expression of AdeAB in strains that can quickly acquire tigecycline resistance compared with strains that displayed lower adaptability. To validate our findings, we added an efflux pump inhibitor, carbonyl cyanide m-chlorophenyl hydrazine (CCCP), to the medium and observed its ability to inhibit tigecycline resistance in A. baumannii strains with quick adaptability. This study contributes to a better understanding of the mechanisms underlying tigecycline resistance in A. baumannii under sub-MIC pressure.


Asunto(s)
Acinetobacter baumannii , Tigeciclina/farmacología , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Espectrometría de Masas en Tándem , Antibacterianos/farmacología , Antibacterianos/metabolismo , Farmacorresistencia Bacteriana , Pruebas de Sensibilidad Microbiana , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana Múltiple
12.
Nature ; 625(7995): 566-571, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38172634

RESUMEN

Carbapenem-resistant Acinetobacter baumannii (CRAB) has emerged as a major global pathogen with limited treatment options1. No new antibiotic chemical class with activity against A. baumannii has reached patients in over 50 years1. Here we report the identification and optimization of tethered macrocyclic peptide (MCP) antibiotics with potent antibacterial activity against CRAB. The mechanism of action of this molecule class involves blocking the transport of bacterial lipopolysaccharide from the inner membrane to its destination on the outer membrane, through inhibition of the LptB2FGC complex. A clinical candidate derived from the MCP class, zosurabalpin (RG6006), effectively treats highly drug-resistant contemporary isolates of CRAB both in vitro and in mouse models of infection, overcoming existing antibiotic resistance mechanisms. This chemical class represents a promising treatment paradigm for patients with invasive infections due to CRAB, for whom current treatment options are inadequate, and additionally identifies LptB2FGC as a tractable target for antimicrobial drug development.


Asunto(s)
Antibacterianos , Lipopolisacáridos , Proteínas de Transporte de Membrana , Animales , Humanos , Ratones , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/metabolismo , Antibacterianos/clasificación , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Lipopolisacáridos/metabolismo , Pruebas de Sensibilidad Microbiana , Proteínas de Transporte de Membrana/metabolismo , Transporte Biológico/efectos de los fármacos , Modelos Animales de Enfermedad , Infecciones por Acinetobacter/tratamiento farmacológico , Infecciones por Acinetobacter/microbiología , Desarrollo de Medicamentos
13.
J Bacteriol ; 206(1): e0040323, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38084964

RESUMEN

In a recent study by Inga V. Leus, Sean R. Roberts, Anhthu Trinh, Edward W. Yu, and Helen I. Zgurskaya (J Bacteriol, 2023, https://doi.org/10.1128/jb.00217-23), it was found that the clinically relevant resistance-nodulation-cell division (RND)-type AdeABC antibiotic efflux pump from Acinetobacter baumannii exhibits close communication between its antibiotic binding sites. Alterations in one of them can have far-reaching impacts on the drug translocation pathway. These insights could reshape our understanding of RND-type efflux pump mechanisms.


Asunto(s)
Acinetobacter baumannii , Proteínas Bacterianas , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Antibacterianos/farmacología , Antibacterianos/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Transporte Biológico , División Celular , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Farmacorresistencia Bacteriana Múltiple , Pruebas de Sensibilidad Microbiana
14.
J Bacteriol ; 206(1): e0021723, 2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-37850798

RESUMEN

Multidrug efflux is one of the major mechanisms of antibiotic resistance identified in clinical isolates of the human pathogen Acinetobacter baumannii. The multiple antibiotic resistance in this species is often enabled by the overproduction of the tripartite efflux pump AdeABC. In this pump, AdeB is the inner membrane transporter from the resistance-nodulation-division (RND) superfamily of proteins, which is responsible for the recognition and efflux of multiple structurally unrelated compounds. Like other RND transporters, AdeB is a trimeric protein with ligand-binding sites located in the large periplasmic domains. Previous structural studies, however, highlighted the uniqueness of AdeB interactions with ligands. Up to three ligand molecules were bound to one protomer of AdeB, mapping its substrate translocation path. In this study, we introduced single and double substitutions in the identified ligand-binding sites of AdeB. Our results show that the mechanism of substrate translocation by AdeB is different from that of other characterized RND transporters and that the functional interactions between the sites are nonadditive. We identified AdeB mutants with both the loss and the gain of antibiotic susceptibility phenotypes, as well as AdeB mutations making A. baumannii cells overproducing such pump variants even more susceptible to multiple antibiotics than efflux-deficient cells. IMPORTANCE Multidrug efflux pumps of the resistance-nodulation-division superfamily of proteins are important contributors to various aspects of bacterial physiology and antibiotic resistance. Studies of the best-characterized model transporter AcrB from Escherichia coli suggested that these transporters operate by a functional rotation mechanism in which various substrates bind to at least two different binding sites. This study suggests that the mechanism of AdeB is distinct and that the binding sites in this transporter are functionally linked.


Asunto(s)
Acinetobacter baumannii , Proteínas de Escherichia coli , Humanos , Acinetobacter baumannii/genética , Acinetobacter baumannii/metabolismo , Proteínas Bacterianas/metabolismo , Ligandos , Antibacterianos/metabolismo , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Sitios de Unión , Escherichia coli/metabolismo , Farmacorresistencia Bacteriana Múltiple/genética , Pruebas de Sensibilidad Microbiana , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Proteínas de Escherichia coli/metabolismo
15.
Microb Pathog ; 186: 106473, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38048840

RESUMEN

Outer membrane protein A (OmpA) is a critical virulence factor in Acinetobacter baumannii, influencing adhesion, biofilm formation, host immune response, and host cell apoptosis. We investigated the invasion of A549 alveolar epithelial cells by A. baumannii and examined how anti-OmpA antibodies impact these interactions. OmpA was expressed and purified, inducing anti-OmpA antibodies in BALB/c mice. The potential toxicity of OmpA was evaluated in mice by analyzing histology from six organs. A549 cells were exposed to A. baumannii strains 19606 and a clinical isolate. Using cell culture and light microscopy, we scrutinized the effects of anti-OmpA sera on serum resistance, adherence, internalization, and proliferation of A. baumannii in A549 cells. The viability of A549 cells was assessed upon exposure to live A. baumannii and anti-OmpA sera. OmpA-induced antibody demonstrated potent bactericidal effects on both strains of A. baumannii. Both strains formed biofilms, which were reduced by anti-OmpA serum, along with decreased bacterial adherence, internalization, and proliferation in A549 cells. Anti-OmpA serum improved the survival of A549 cells post-infection. Pre-treatment with cytochalasin D hindered bacterial internalization, highlighting the role of actin polymerization in invasion. Microscopic examination revealed varied interactions encompassing adherence, apoptosis, membrane alterations, vacuolization, and damage. A549 cells treated with anti-OmpA serum exhibited improved structures and reduced damage. The findings indicate that A. baumannii can adhere to and proliferate within epithelial cells with OmpA playing a pivotal role in these interactions, and the complex nature of these interactions shapes the intricate course of A. baumannii infection in host cells.


Asunto(s)
Acinetobacter baumannii , Humanos , Animales , Ratones , Acinetobacter baumannii/metabolismo , Células Epiteliales Alveolares , Biopelículas , Bacterias , Proliferación Celular
16.
Mol Microbiol ; 121(2): 196-212, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37918886

RESUMEN

Infections caused by Acinetobacter baumannii, a Gram-negative opportunistic pathogen, are difficult to eradicate due to the bacterium's propensity to quickly gain antibiotic resistances and form biofilms, a protective bacterial multicellular community. The A. baumannii DNA damage response (DDR) mediates the antibiotic resistance acquisition and regulates RecA in an atypical fashion; both RecALow and RecAHigh cell types are formed in response to DNA damage. The findings of this study demonstrate that the levels of RecA can influence formation and dispersal of biofilms. RecA loss results in surface attachment and prominent biofilms, while elevated RecA leads to diminished attachment and dispersal. These findings suggest that the challenge to treat A. baumannii infections may be explained by the induction of the DDR, common during infection, as well as the delicate balance between maintaining biofilms in low RecA cells and promoting mutagenesis and dispersal in high RecA cells. This study underscores the importance of understanding the fundamental biology of bacteria to develop more effective treatments for infections.


Asunto(s)
Acinetobacter baumannii , Acinetobacter baumannii/metabolismo , Daño del ADN , Biopelículas , Antibacterianos/farmacología , Antibacterianos/metabolismo , Farmacorresistencia Bacteriana Múltiple
17.
Biochemistry ; 63(2): 230-240, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38150593

RESUMEN

The first step of histidine biosynthesis in Acinetobacter baumannii, the condensation of ATP and 5-phospho-α-d-ribosyl-1-pyrophosphate to produce N1-(5-phospho-ß-d-ribosyl)-ATP (PRATP) and pyrophosphate, is catalyzed by the hetero-octameric enzyme ATP phosphoribosyltransferase, a promising target for antibiotic design. The catalytic subunit, HisGS, is allosterically activated upon binding of the regulatory subunit, HisZ, to form the hetero-octameric holoenzyme (ATPPRT), leading to a large increase in kcat. Here, we present the crystal structure of ATPPRT, along with kinetic investigations of the rate-limiting steps governing catalysis in the nonactivated (HisGS) and activated (ATPPRT) forms of the enzyme. A pH-rate profile showed that maximum catalysis is achieved above pH 8.0. Surprisingly, at 25 °C, kcat is higher when ADP replaces ATP as substrate for ATPPRT but not for HisGS. The HisGS-catalyzed reaction is limited by the chemical step, as suggested by the enhancement of kcat when Mg2+ was replaced by Mn2+, and by the lack of a pre-steady-state burst of product formation. Conversely, the ATPPRT-catalyzed reaction rate is determined by PRATP diffusion from the active site, as gleaned from a substantial solvent viscosity effect. A burst of product formation could be inferred from pre-steady-state kinetics, but the first turnover was too fast to be directly observed. Lowering the temperature to 5 °C allowed observation of the PRATP formation burst by ATPPRT. At this temperature, the single-turnover rate constant was significantly higher than kcat, providing additional evidence for a step after chemistry limiting catalysis by ATPPRT. This demonstrates allosteric activation by HisZ accelerates the chemical step.


Asunto(s)
ATP Fosforribosil Transferasa , Acinetobacter baumannii , ATP Fosforribosil Transferasa/química , Difosfatos , Acinetobacter baumannii/metabolismo , Dominio Catalítico , Cinética , Adenosina Trifosfato/metabolismo , Catálisis
18.
Appl Microbiol Biotechnol ; 108(1): 3, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38159120

RESUMEN

Acinetobacter baumannii is a Gram-negative opportunistic zoonotic pathogenic bacterium that causes nosocomial infections ranging from minor to life-threatening. The clinical importance of this zoonotic pathogen is rapidly increasing due to the development of multiple resistance mechanisms and the synthesis of numerous virulence factors. Although no flagellum-mediated motility exists, it may move through twitching or surface-associated motility. Twitching motility is a coordinated multicellular movement caused by the extension, attachment, and retraction of type IV pili, which are involved in surface adherence and biofilm formation. Surface-associated motility is a kind of movement that does not need appendages and is most likely driven by the release of extra polymeric molecules. This kind of motility is linked to the production of 1,3-diaminopropane, lipooligosaccharide formation, natural competence, and efflux pump proteins. Since A. baumannii's virulence qualities are directly tied to motility, it is possible that its motility may be used as a specialized preventative or therapeutic measure. The current review detailed the signaling mechanism and involvement of various proteins in controlling A. baumannii motility. As a result, we have thoroughly addressed the role of natural and synthetic compounds that impede A. baumannii motility, as well as the underlying action mechanisms. Understanding the regulatory mechanisms behind A. baumannii's motility features will aid in the development of therapeutic drugs to control its infection. KEY POINTS: • Acinetobacter baumannii exhibits multiple resistance mechanisms. • A. baumannii can move owing to twitching and surface-associated motility. • Natural and synthetic compounds can attenuate A. baumannii motility.


Asunto(s)
Acinetobacter baumannii , Acinetobacter baumannii/metabolismo , Virulencia , Factores de Virulencia/metabolismo , Proteínas Bacterianas/metabolismo , Fimbrias Bacterianas/metabolismo , Biopelículas , Antibacterianos/metabolismo
19.
Int J Mol Sci ; 24(24)2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-38139363

RESUMEN

Antibiotic resistance is a pressing topic, which also affects ß-lactam antibiotic molecules. Until a few years ago, it was considered no more than an interesting species from an academic point of view, Acinetobacter baumanii is today one of the most serious threats to public health, so much so that it has been declared one of the species for which the search for new antibiotics, or new ways to avoid its resistance, is an absolute priority according to WHO. Although there are several molecular mechanisms that are responsible for the extreme resistance of A. baumanii to antibiotics, a class D ß-lactamase is the main cause for the clinical concern of this bacterial species. In this work, we analyzed the A. baumanii OXA-23 protein via molecular dynamics. The results obtained show that this protein is able to assume different conformations, especially in some regions around the active site. Part of the OXA-23 protein has considerable conformational motility, while the rest is less mobile. The importance of these observations for understanding the functioning mechanism of the enzyme as well as for designing new effective molecules for the treatment of A. baumanii is discussed.


Asunto(s)
Acinetobacter baumannii , Acinetobacter baumannii/metabolismo , Antibacterianos/farmacología , Antibacterianos/química , beta-Lactamasas/metabolismo , Simulación de Dinámica Molecular , Pruebas de Sensibilidad Microbiana , Proteínas Bacterianas/metabolismo
20.
Microb Pathog ; 185: 106434, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37913828

RESUMEN

The PmrAB two-component system modulates colistin resistance in Acinetobacter baumannii, but its association with the virulence traits of this bacterium remains uncharacterized. This study explored the role of A. baumannii PmrAB in surface motility, biofilm formation, and outer membrane vesicle (OMV) biogenesis using wild-type (WT) A. baumannii 17978 and ΔpmrA and ΔpmrB mutant strains. The two mutant strains exhibited significantly decreased surface motility compared with that of WT strain by the low expression of abaI, abaR, A1S_0113, A1S_0115, and A1S_0116. Biofilm mass also significantly decreased in the two mutant strains at 12 h of incubation, but restored at 24 h. Under static culture conditions for 12 h, the two mutant strains showed low pgaA expression. However, the other biofilm-associated genes, such as csuC, csuE, ompA, and bap, showed different expression between the two mutant strains. Although the size of OMVs was similar among the three strains, the number of OMVs secreted from the two mutant strains slightly decreased compared with that secreted from the WT strain. Protein concentrations in the OMVs of ΔpmrA mutant significantly decreased compared with those in the OMVs of WT and ΔpmrB strains. Overall, PmrAB modulates virulence traits and OMV biogenesis in A. baumannii.


Asunto(s)
Acinetobacter baumannii , Virulencia/genética , Acinetobacter baumannii/metabolismo , Biopelículas , Transporte Biológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...