Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.758
Filtrar
1.
Mikrobiyol Bul ; 58(2): 135-147, 2024 Apr.
Artículo en Turco | MEDLINE | ID: mdl-38676582

RESUMEN

Pseudomonas aeruginosa is a non-fermentative gram-negative bacillus. Many virulence factors play a role in the pathogenesis of P.aeruginosa. The aim of this study was to early detection of ST111, ST175, ST235, ST253, ST395 which are named high-risk clones with increased epidemic potential due to multidrug resistance in P.aeruginosa isolates by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) method and to evaluate the relationship between high-risk clones and the presence of P.aeruginosa virulence factors and carbapenemase production genes.P.aeruginosa isolates (n= 100) found to be resistant to at least imipenem or meropenem antibiotics isolated from the various clinical samples in the medical microbiology laboratory between 01.01.2021 and 07.06.2022 were included in the study. For the detection of virulence genes uniplex polymerase chain reaction (PCR) for toxA and multiplex PCR for algD, plcN, lasB, plcH were performed in P.aeruginosa isolates. In the detection of carbapenemase genes, two separate multiplex PCRs used for blaKPC , blaNDM , blaVIM , blaOXA-48 and for blaIMP , blaSPM , blaSIM , blaGIM , blaGES . Investigation of the peaks specific to high-risk clones was performed by using VITEK®-MS (bioMérieux, France) system. P.aeruginosa isolates were mostly isolated from intensive care units (45%) and respiratory tract samples (46%). The antibiotic to which the isolates were found to be most susceptible was amikacin, while highest resistance was detected for piperacillin. In PCR results, toxA, lasB, plcH, plcN and algD were detected as 89%, 99%, 98%, 100%, 100%, respectively. When the presence of characteristic peaks belonging to high-risk clones was evaluated with MALDI-TOF MS, ST253 (7%) and ST175 (2%) were detected. The peaks specific to ST235 and ST395 clones were not detected in our study. blaVIM was detected in two isolates and blaGES-5 carbapenemase was detected in two isolates. Virulence factors were detected at high rates in both high-risk clones and other strains and no significant relationship was found between high-risk clones and virulence factors. Early detection of high-risk clones, identification of antimicrobial resistance mechanisms will help to develop strategic treatment options and prevent their worldwide spread.


Asunto(s)
Reacción en Cadena de la Polimerasa , Pseudomonas aeruginosa , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Factores de Virulencia , beta-Lactamasas , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/aislamiento & purificación , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Humanos , beta-Lactamasas/genética , Factores de Virulencia/genética , Proteínas Bacterianas/genética , Infecciones por Pseudomonas/microbiología , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/genética , Imipenem/farmacología , Meropenem/farmacología , Virulencia/genética
2.
Science ; 384(6691): eadl0635, 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38574145

RESUMEN

The retractile type IV pilus (T4P) is important for virulence of the opportunistic human pathogen Pseudomonas aeruginosa. The single-stranded RNA (ssRNA) phage PP7 binds to T4P and is brought to the cell surface through pilus retraction. Using fluorescence microscopy, we discovered that PP7 detaches T4P, which impairs cell motility and restricts the pathogen's virulence. Using cryo-electron microscopy, mutagenesis, optical trapping, and Langevin dynamics simulation, we resolved the structure of PP7, T4P, and the PP7/T4P complex and showed that T4P detachment is driven by the affinity between the phage maturation protein and its bound pilin, plus the pilus retraction force and speed, and pilus bending. Pilus detachment may be widespread among other ssRNA phages and their retractile pilus systems and offers new prospects for antibacterial prophylaxis and therapeutics.


Asunto(s)
Fimbrias Bacterianas , Fagos Pseudomonas , Pseudomonas aeruginosa , Virus ARN , Internalización del Virus , Humanos , Microscopía por Crioelectrón , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/virología , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/virología , Virus ARN/química , Virus ARN/fisiología , Fagos Pseudomonas/química , Fagos Pseudomonas/fisiología , Proteínas Virales/metabolismo
3.
Int J Biol Macromol ; 267(Pt 2): 131677, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38641280

RESUMEN

The research investigates the virulence factors of Pseudomonas aeruginosa (P. aeruginosa), a pathogen known for its ability to cause human infections by releasing various exoenzymes and virulence factors. Particularly relevant in ocular infections, where tissue degeneration can occur, even after bacterial growth has ceased due to the potential role of secreted proteins/enzymes. Clinical isolates of P. aeruginosa, both ocular (146) and non-ocular (54), were examined to determine the frequency and mechanism of virulence factors. Phenotypic characterization revealed the production of alginate, biofilm, phospholipase C, and alkaline protease, while genotypic testing using internal uniplex PCR identified the presence of Exo U, S, T, Y, and LasB genes. Results showed a significant prevalence of Exo U and Y genes in ocular isolates, a finding unique to Indian studies. Additionally, the study noted that ocular isolates often contained all four secretomes, suggesting a potential link between these factors and ocular infections. These findings contribute to understanding the pathogenesis of P. aeruginosa infections, particularly in ocular contexts, and highlights the importance of comprehensive virulence factor analysis in clinical settings.


Asunto(s)
Biopelículas , Pseudomonas aeruginosa , Factores de Virulencia , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Humanos , Biopelículas/crecimiento & desarrollo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Infecciones por Pseudomonas/microbiología , Farmacorresistencia Bacteriana Múltiple/genética , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo , Endopeptidasas
4.
Nucleic Acids Res ; 52(7): 3856-3869, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38477346

RESUMEN

The genetic diversities of subpopulations drive the evolution of pathogens and affect their ability to infect hosts and cause diseases. However, most studies to date have focused on the identification and characterization of adaptive mutations in single colonies, which do not accurately reflect the phenotypes of an entire population. Here, to identify the composition of variant subpopulations within a pathogen population, we developed a streamlined approach that combines high-throughput sequencing of the entire population cells with genotyping of single colonies. Using this method, we reconstructed a detailed quorum-sensing (QS) evolutionary trajectory in Pseudomonas aeruginosa. Our results revealed a new adaptive mutation in the gacS gene, which codes for a histidine kinase sensor of a two-component system (TCS), during QS evolution. This mutation reduced QS activity, allowing the variant to sweep throughout the whole population, while still being vulnerable to invasion by the emerging QS master regulator LasR-null mutants. By tracking the evolutionary trajectory, we found that mutations in gacS facilitated QS-rewiring in the LasR-null mutant. This rapid QS revertant caused by inactive GacS was found to be associated with the promotion of ribosome biogenesis and accompanied by a trade-off of reduced bacterial virulence on host cells. In conclusion, our findings highlight the crucial role of the global regulator GacS in modulating the progression of QS evolution and the virulence of the pathogen population.


Asunto(s)
Proteínas Bacterianas , Evolución Molecular , Mutación , Pseudomonas aeruginosa , Percepción de Quorum , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/genética , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Virulencia/genética , Transactivadores/genética , Transactivadores/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Regulación Bacteriana de la Expresión Génica , Histidina Quinasa/genética , Histidina Quinasa/metabolismo
5.
Nucleic Acids Res ; 52(6): 3433-3449, 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38477394

RESUMEN

The regulation of carbon metabolism and virulence is critical for the rapid adaptation of pathogenic bacteria to host conditions. In Pseudomonas aeruginosa, RccR is a transcriptional regulator of genes involved in primary carbon metabolism and is associated with bacterial resistance and virulence, although the exact mechanism is unclear. Our study demonstrates that PaRccR is a direct repressor of the transcriptional regulator genes mvaU and algU. Biochemical and structural analyses reveal that PaRccR can switch its DNA recognition mode through conformational changes triggered by KDPG binding or release. Mutagenesis and functional analysis underscore the significance of allosteric communication between the SIS domain and the DBD domain. Our findings suggest that, despite its overall structural similarity to other bacterial RpiR-type regulators, RccR displays a more complex regulatory element binding mode induced by ligands and a unique regulatory mechanism.


Asunto(s)
Proteínas Bacterianas , Pseudomonas aeruginosa , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Carbono/metabolismo , Regulación Bacteriana de la Expresión Génica , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Virulencia/genética , Factores de Virulencia/genética
6.
Microb Pathog ; 189: 106609, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38452830

RESUMEN

The emergence of multidrug resistance and increased pathogenicity in microorganisms is conferred by the presence of highly synchronized cell density dependent signalling pathway known as quorum sensing (QS). The QS hierarchy is accountable for the secretion of virulence phenotypes, biofilm formation and drug resistance. Hence, targeting the QS phenomenon could be a promising strategy to counteract the bacterial virulence and drug resistance. In the present study, artocarpesin (ACN), a 6-prenylated flavone was investigated for its capability to quench the synthesis of QS regulated virulence factors. From the results, ACN showed significant inhibition of secreted virulence phenotypes such as pyocyanin (80%), rhamnolipid (79%), protease (69%), elastase (84%), alginate (88%) and biofilm formation (88%) in opportunistic pathogen, Pseudomonas aeruginosa PAO1. Further, microscopic observation of biofilm confirmed a significant reduction in biofilm matrix when P. aeruginosa PAO1 was supplemented with ACN at its sub-MIC concentration. Quantitative gene expression studies showed the promising aspects of ACN in down regulation of several QS regulatory genes associated with production of virulence phenotypes. Upon treatment with sub-MIC of ACN, the bacterial colonization in the gut of Caenorhabditis elegans was potentially reduced and the survival rate was greatly improved. The promising QS inhibition activities were further validated through in silico studies, which put an insight into the mechanism of QS inhibition. Thus, ACN could be considered as possible drug candidate targeting chronic microbial infections.


Asunto(s)
Flavonas , Infecciones por Pseudomonas , Percepción de Quorum , Humanos , Antibacterianos/metabolismo , Proteínas Bacterianas/metabolismo , Biopelículas , Pseudomonas aeruginosa/patogenicidad , Infecciones por Pseudomonas/microbiología , Virulencia/genética , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
7.
Nature ; 618(7964): 358-364, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225987

RESUMEN

The ability to switch between different lifestyles allows bacterial pathogens to thrive in diverse ecological niches1,2. However, a molecular understanding of their lifestyle changes within the human host is lacking. Here, by directly examining bacterial gene expression in human-derived samples, we discover a gene that orchestrates the transition between chronic and acute infection in the opportunistic pathogen Pseudomonas aeruginosa. The expression level of this gene, here named sicX, is the highest of the P. aeruginosa genes expressed in human chronic wound and cystic fibrosis infections, but it is expressed at extremely low levels during standard laboratory growth. We show that sicX encodes a small RNA that is strongly induced by low-oxygen conditions and post-transcriptionally regulates anaerobic ubiquinone biosynthesis. Deletion of sicX causes P. aeruginosa to switch from a chronic to an acute lifestyle in multiple mammalian models of infection. Notably, sicX is also a biomarker for this chronic-to-acute transition, as it is the most downregulated gene when a chronic infection is dispersed to cause acute septicaemia. This work solves a decades-old question regarding the molecular basis underlying the chronic-to-acute switch in P. aeruginosa and suggests oxygen as a primary environmental driver of acute lethality.


Asunto(s)
Enfermedad Aguda , Enfermedad Crónica , Genes Bacterianos , Oxígeno , Infecciones por Pseudomonas , Pseudomonas aeruginosa , ARN Bacteriano , Animales , Humanos , Oxígeno/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Infecciones por Pseudomonas/complicaciones , Infecciones por Pseudomonas/microbiología , Infecciones por Pseudomonas/patología , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Fibrosis Quística/microbiología , Heridas y Lesiones/microbiología , Ubiquinona/biosíntesis , Anaerobiosis , Genes Bacterianos/genética , Sepsis/complicaciones , Sepsis/microbiología
8.
Med. leg. Costa Rica ; 40(1)mar. 2023.
Artículo en Español | LILACS, SaludCR | ID: biblio-1430759

RESUMEN

La Pseudomona aeruginosa es una causa importante de infecciones asociadas a la atención de la salud y en las neumonías adquiridas en la comunidad, rara vez se identifica como el agente patógeno, siendo estas de progresión rápida y de mal pronóstico. Se trata de un menor de un año de edad inmunocompetente el cual fallece en casa una semana después de una lesión en la planta del pie derecho que según familiares le sacaron "pus", tratado con antinflamatorios y analgésicos. Se le realizó necropsia que evidenció cicatriz en planta de pie derecho sin lesiones traumáticas. Pulmones de consistencia indurada, con adherencias y áreas que impresionan necróticas, asociada a efusión pleural. El estudio histológico reportó un proceso infeccioso pulmonar agudo abscedado que se diseminó por continuidad a tejido cardiaco y en estudios microbiológicos de pulmón y bazo se reportó Pseudomona aeruginosa.


Pseudomona aeruginosa is an important cause of health care-associated infections and in community-acquired pneumonias, it is rarely identified as the pathogenic agent, being of rapid progression and poor prognosis. This is a one-year-old immunocompetent minor who died at home one week after a lesion in the sole of the right foot which, according to family members, caused "pus", treated with anti-inflammatory and analgesic drugs. A necropsy was performed, which showed a scar on the sole of the right foot with no traumatic lesions. Lungs of indurated consistency, with adhesions and areas that appear necrotic, associated with pleural effusion. The histological study reported an abscessed acute pulmonary infectious process that spread by continuity to cardiac tissue and microbiological studies of lung and spleen reported Pseudomona aeruginosa.


Asunto(s)
Humanos , Masculino , Lactante , Pericarditis/diagnóstico , Pseudomonas aeruginosa/patogenicidad , Panamá , Neumonía , Absceso , Miocardio
9.
Nature ; 613(7943): 324-331, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36599989

RESUMEN

Pathogens generate ubiquitous selective pressures and host-pathogen interactions alter social behaviours in many animals1-4. However, very little is known about the neuronal mechanisms underlying pathogen-induced changes in social behaviour. Here we show that in adult Caenorhabditis elegans hermaphrodites, exposure to a bacterial pathogen (Pseudomonas aeruginosa) modulates sensory responses to pheromones by inducing the expression of the chemoreceptor STR-44 to promote mating. Under standard conditions, C. elegans hermaphrodites avoid a mixture of ascaroside pheromones to facilitate dispersal5-13. We find that exposure to the pathogenic Pseudomonas bacteria enables pheromone responses in AWA sensory neurons, which mediate attractive chemotaxis, to suppress the avoidance. Pathogen exposure induces str-44 expression in AWA neurons, a process regulated by a transcription factor zip-5 that also displays a pathogen-induced increase in expression in AWA. STR-44 acts as a pheromone receptor and its function in AWA neurons is required for pathogen-induced AWA pheromone response and suppression of pheromone avoidance. Furthermore, we show that C. elegans hermaphrodites, which reproduce mainly through self-fertilization, increase the rate of mating with males after pathogen exposure and that this increase requires str-44 in AWA neurons. Thus, our results uncover a causal mechanism for pathogen-induced social behaviour plasticity, which can promote genetic diversity and facilitate adaptation of the host animals.


Asunto(s)
Caenorhabditis elegans , Feromonas , Pseudomonas aeruginosa , Reproducción , Conducta Sexual Animal , Animales , Femenino , Masculino , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Glucolípidos/metabolismo , Organismos Hermafroditas/fisiología , Feromonas/metabolismo , Pseudomonas aeruginosa/patogenicidad , Pseudomonas aeruginosa/fisiología , Receptores de Feromonas/metabolismo , Reproducción/fisiología , Células Receptoras Sensoriales/metabolismo
10.
Microbiol Spectr ; 11(1): e0330822, 2023 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-36546869

RESUMEN

Pseudomonas aeruginosa is an opportunistic pathogen with multiple strategies to interact with other microbes and host cells, gaining fitness in complicated infection sites. The contact-dependent type VI secretion system (T6SS) is one critical secretion apparatus involved in both interbacterial competition and pathogenesis. To date, only limited numbers of T6SS-effectors have been clearly characterized in P. aeruginosa laboratory strains, and the importance of T6SS diversity in the evolution of clinical P. aeruginosa remains unclear. Recently, we characterized a P. aeruginosa clinical strain LYSZa7 from a COVID-19 patient, which adopted complex genetic adaptations toward chronic infections. Bioinformatic analysis has revealed a putative type VI secretion system (T6SS) dependent lipase effector in LYSZa7, which is a homologue of TseL in Vibrio cholerae and is widely distributed in pathogens. We experimentally validated that this TseL homologue belongs to the Tle2, a subfamily of T6SS-lipase effectors; thereby, we name this effector TseL (TseLPA in this work). Further, we showed the lipase-dependent bacterial toxicity of TseLPA, which primarily targets bacterial periplasm. The toxicity of TseLPA can be neutralized by two immunity proteins, TsiP1 and TsiP2, which are encoded upstream of tseL. In addition, we proved this TseLPA contributes to bacterial pathogenesis by promoting bacterial internalization into host cells. Our study suggests that clinical bacterial strains employ a diversified group of T6SS effectors for interbacterial competition and might contribute to emerging of new epidemic clonal lineages. IMPORTANCE Pseudomonas aeruginosa is one predominant pathogen that causes hospital-acquired infections and is one of the commonest coinfecting bacteria in immunocompromised patients and chronic wounds. This bacterium harbors a diverse accessory genome with a high frequency of gene recombination, rendering its population highly heterogeneous. Numerous Pa lineages coexist in the biofilm, where successful epidemic clonal lineage or strain-specific type commonly acquires genes to increase its fitness over the other organisms. Current studies of Pa genomic diversity commonly focused on antibiotic resistant genes and novel phages, overlooking the contribution of type VI secretion system (T6SS). We characterized a Pa clinical strain LYSZa7 from a COVID-19 patient, which adopted complex genetic adaptations toward chronic infections. We report, in this study, a novel T6SS-lipase effector that is broadly distributed in Pa clinical isolates and other predominant pathogens. The study suggests that hospital transmission may raise the emergence of new epidemic clonal lineages with specified T6SS effectors.


Asunto(s)
COVID-19 , Pseudomonas aeruginosa , Sistemas de Secreción Tipo VI , Humanos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , COVID-19/complicaciones , COVID-19/microbiología , Infección Persistente , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/microbiología , Sistemas de Secreción Tipo VI/genética , Sistemas de Secreción Tipo VI/metabolismo
11.
Proc Natl Acad Sci U S A ; 119(20): e2119434119, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35561220

RESUMEN

The ability of eukaryotic cells to differentiate surface stiffness is fundamental for many processes like stem cell development. Bacteria were previously known to sense the presence of surfaces, but the extent to which they could differentiate stiffnesses remained unclear. Here we establish that the human pathogen Pseudomonas aeruginosa actively measures surface stiffness using type IV pili (TFP). Stiffness sensing is nonlinear, as induction of the virulence factor regulator is peaked with stiffness in a physiologically important range between 0.1 kPa (similar to mucus) and 1,000 kPa (similar to cartilage). Experiments on surfaces with distinct material properties establish that stiffness is the specific biophysical parameter important for this sensing. Traction force measurements reveal that the retraction of TFP is capable of deforming even stiff substrates. We show how slow diffusion of the pilin PilA in the inner membrane yields local concentration changes at the base of TFP during extension and retraction that change with substrate stiffness. We develop a quantitative biomechanical model that explains the transcriptional response to stiffness. A competition between PilA diffusion in the inner membrane and a loss/gain of monomers during TFP extension/retraction produces substrate stiffness-dependent dynamics of the local PilA concentration. We validated this model by manipulating the ATPase activity of the TFP motors to change TFP extension and retraction velocities and PilA concentration dynamics, altering the stiffness response in a predictable manner. Our results highlight stiffness sensing as a shared behavior across biological kingdoms, revealing generalizable principles of environmental sensing across small and large cells.


Asunto(s)
Proteínas Fimbrias , Fimbrias Bacterianas , Pseudomonas aeruginosa , Proteínas Fimbrias/genética , Proteínas Fimbrias/metabolismo , Fimbrias Bacterianas/genética , Fimbrias Bacterianas/fisiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Propiedades de Superficie , Transcripción Genética
12.
ACS Appl Mater Interfaces ; 14(7): 8718-8727, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35138100

RESUMEN

Transparent antimicrobial coatings can maintain the aesthetic appeal of surfaces and the functionality of a touch-screen while adding the benefit of reducing disease transmission. We fabricated an antimicrobial coating of silver oxide particles in a silicate matrix on glass. The matrix was grown by a modified Stöber sol-gel process with vapor-phase water and ammonia. A coating on glass with 2.4 mg of Ag2O per mm2 caused a reduction of 99.3% of SARS-CoV-2 and >99.5% of Pseudomonas aeruginosa, Staphylococcus aureus, and methicillin-resistant Staphylococcus aureus compared to the uncoated glass after 1 h. We envisage that screen protectors with transparent antimicrobial coatings will find particular application to communal touch-screens, such as in supermarkets and other check-out or check-in facilities where a number of individuals utilize the same touch-screen in a short interval.


Asunto(s)
Antiinfecciosos/química , Infecciones Bacterianas/prevención & control , COVID-19/prevención & control , Óxidos/química , Compuestos de Plata/química , Amoníaco/química , Antiinfecciosos/farmacología , Infecciones Bacterianas/microbiología , COVID-19/virología , Vidrio/química , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Óxidos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Silicatos/química , Compuestos de Plata/farmacología , Agua/química
13.
FASEB J ; 36(3): e22057, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35133020

RESUMEN

Non-healing wounds are a major medical challenge, affecting over 6.5 million people in the US alone, with associated healthcare costs of about $16 billion annually. They can result in prolonged hospitalizations, work loss, disability, poor quality of life, and in diabetic patients with foot ulcers, amputation of the affected limb in 25% of patients. Though chronic ulcers may arise from different underlying diseases, the unifying feature is chronic infection, driving persistent inflammation that prolongs the healing process. One of the most frequently cultured or genetically identified pathogens in skin wounds is Pseudomonas aeruginosa. This species avidly forms biofilms in the wound that impede bacterial eradication by the host's immune mechanisms and limit efficacy of systemic antibiotics. Thus, non-antibiotic approaches to limit the growth and biofilm formation of this wound pathogen would be an advance in the treatment of chronic wounds. Prior work has demonstrated that the growth of other microbial species can be modulated by catecholamine agonists and antagonists of the adrenergic receptors (ARs). Here, we demonstrate that not only can the growth of this common wound pathogen be modulated by catecholamines, but also that the beta-AR antagonists can significantly decrease their growth, and importantly, limit their ability to form biofilms. These findings suggest that beta adrenergic antagonists may have a therapeutic role in the treatment of chronic skin wounds.


Asunto(s)
Antagonistas Adrenérgicos/farmacología , Biopelículas , Epinefrina/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Timolol/farmacología , Cicatrización de Heridas , Antagonistas Adrenérgicos/uso terapéutico , Epinefrina/uso terapéutico , Humanos , Infecciones por Pseudomonas/tratamiento farmacológico , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/patogenicidad , Enfermedades Cutáneas Bacterianas/tratamiento farmacológico , Enfermedades Cutáneas Bacterianas/microbiología , Timolol/uso terapéutico
14.
Int J Mol Sci ; 23(3)2022 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-35163191

RESUMEN

Pneumonia is a life-threatening disease often caused by infection with Streptococcus pneumoniae and Pseudomonas aeruginosa. Many of the mediators (e.g., TNF, IL-6R) and junction molecules (e.g., E-cadherin) orchestrating inflammatory cell recruitment and loss of barrier integrity are proteolytically cleaved through a disintegrin and metalloproteinases (ADAMs). We could show by Western blot, surface expression analysis and measurement of proteolytic activity in cell-based assays, that ADAM10 in epithelial cells is upregulated and activated upon infection with Pseudomonas aeruginosa and Exotoxin A (ExoA), but not upon infection with Streptococcus pneumoniae. Targeting ADAM10 by pharmacological inhibition or gene silencing, we demonstrated that this activation was critical for cleavage of E-cadherin and modulated permeability and epithelial integrity. Stimulation with heat-inactivated bacteria revealed that the activation was based on the toxin repertoire rather than the interaction with the bacterial particle itself. Furthermore, calcium imaging experiments showed that the ExoA action was based on the induction of calcium influx. Investigating the extracellular vesicles and their proteolytic activity, we could show that Pseudomonas aeruginosa triggered exosomal release of ADAM10 and proteolytic cleavage in trans. This newly described mechanism could constitute an essential mechanism causing systemic inflammation in patients suffering from Pseudomonas aeruginosa-induced pneumonia stimulating future translational studies.


Asunto(s)
Proteína ADAM10/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Proteínas de la Membrana/metabolismo , Proteolisis , Células A549 , Epitelio/metabolismo , Exosomas/metabolismo , Exosomas/fisiología , Humanos , Inflamación/microbiología , Pseudomonas aeruginosa/metabolismo , Pseudomonas aeruginosa/patogenicidad , Células THP-1
15.
PLoS One ; 17(2): e0263847, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35180238

RESUMEN

BACKGROUND: The interaction between pathogenic bacteria and cholesterol crystals (CCs) has not been investigated. However, CCs are found extensively in atherosclerotic plaques and sclerotic cardiac valves. Interactions between pathogenic bacteria and CCs could provide insights into destabilization of atherosclerotic plaques and bacterial adhesion to cardiac valves. METHODS: Staphylococcus aureus and Pseudomonas aeruginosa were used to assess in vitro bacterial adhesion to CCs and proliferation in the presence of CCs compared to plastic microspheres and glass shards as controls. Ex vivo studies evaluated bacterial adhesion to atherosclerotic rabbit arteries compared to normal arteries and human atherosclerotic carotid plaques compared to normal carotid arteries. Scanning electron microscopy (SEM) was used to visualize bacterial adhesion to CCs and confocal microscopy was used to detect cholesterol binding to bacteria grown in the presence or absence of CCs. RESULTS: In vitro, S. aureus and P. aeruginosa displayed significantly greater adhesion, 36% (p<0.0001) and 89% (p<0.0001), respectively, and growth upon exposure to CCs compared to microspheres or glass shards. Rabbit and human atherosclerotic arteries contained significantly greater bacterial burdens compared to controls (4× (p<0.0004); 3× (p<0.019), respectively. SEM demonstrated that bacteria adhered and appeared to degrade CCs. Consistent with this, confocal microscopy indicated increased cholesterol bound to the bacterial cells. CONCLUSIONS: This study is the first to demonstrate an interaction between bacteria and CCs showing that bacteria dissolve and bind to CCs. This interaction helps to elucidate adhesion of bacteria to sclerotic valves and atherosclerotic plaques that may contribute to endocarditis and plaque destabilization.


Asunto(s)
Aterosclerosis/microbiología , Colesterol/metabolismo , Endocarditis/microbiología , Pseudomonas aeruginosa/patogenicidad , Staphylococcus aureus/patogenicidad , Animales , Aterosclerosis/metabolismo , Colesterol/química , Cristalización , Endocarditis/metabolismo , Humanos , Conejos
16.
Molecules ; 27(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35163966

RESUMEN

Pseudomonas aeruginosa infections have become more challenging to treat and eradicate due to their ability to form biofilms. This study aimed to produce hydrophobic nanoparticles by grafting 11-carbon and three-carbon alkyl chains to a chitosan polymer as a platform to carry and deliver carvacrol for improving its antibacterial and antibiofilm properties. Carvacrol-chitosan nanoparticles showed ζ potential values of 10.5-14.4 mV, a size of 140.3-166.6 nm, and an encapsulation efficiency of 25.1-68.8%. Hydrophobic nanoparticles reduced 46-53% of the biomass and viable cells (7-25%) within P. aeruginosa biofilms. Diffusion of nanoparticles through the bacterial biofilm showed a higher penetration of nanoparticles created with 11-carbon chain chitosan than those formulated with unmodified chitosan. The interaction of nanoparticles with a 50:50 w/w phospholipid mixture at the air-water interface was studied, and values suggested that viscoelasticity and fluidity properties were modified. The modified nanoparticles significantly reduced viable P. aeruginosa in biofilms (0.078-2.0 log CFU·cm-2) and swarming motility (40-60%). Furthermore, the formulated nanoparticles reduced the quorum sensing in Chromobacterium violaceum. This study revealed that modifying the chitosan polarity to synthesize more hydrophobic nanoparticles could be an effective treatment against P. aeruginosa biofilms to decrease its virulence and pathogenicity, mainly by increasing their ability to interact with the membrane phospholipids and penetrate preformed biofilms.


Asunto(s)
Biopelículas/efectos de los fármacos , Cimenos/farmacología , Pseudomonas aeruginosa/efectos de los fármacos , Antibacterianos/farmacología , Quitosano/química , Cimenos/química , Nanopartículas/química , Extractos Vegetales/química , Extractos Vegetales/farmacología , Pseudomonas aeruginosa/patogenicidad , Percepción de Quorum/efectos de los fármacos , Virulencia , Factores de Virulencia
17.
JCI Insight ; 7(4)2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35041620

RESUMEN

Total body irradiation (TBI) targets sensitive bone marrow hematopoietic cells and gut epithelial cells, causing their death and inducing a state of immunodeficiency combined with intestinal dysbiosis and nonproductive immune responses. We found enhanced Pseudomonas aeruginosa (PAO1) colonization of the gut leading to host cell death and strikingly decreased survival of irradiated mice. The PAO1-driven pathogenic mechanism includes theft-ferroptosis realized via (a) curbing of the host antiferroptotic system, GSH/GPx4, and (b) employing bacterial 15-lipoxygenase to generate proferroptotic signal - 15-hydroperoxy-arachidonoyl-PE (15-HpETE-PE) - in the intestines of irradiated and PAO1-infected mice. Global redox phospholipidomics of the ileum revealed that lysophospholipids and oxidized phospholipids, particularly oxidized phosphatidylethanolamine (PEox), represented the major factors that contributed to the pathogenic changes induced by total body irradiation and infection by PAO1. A lipoxygenase inhibitor, baicalein, significantly attenuated animal lethality, PAO1 colonization, intestinal epithelial cell death, and generation of ferroptotic PEox signals. Opportunistic PAO1 mechanisms included stimulation of the antiinflammatory lipoxin A4, production and suppression of the proinflammatory hepoxilin A3, and leukotriene B4. Unearthing complex PAO1 pathogenic/virulence mechanisms, including effects on the host anti/proinflammatory responses, lipid metabolism, and ferroptotic cell death, points toward potentially new therapeutic and radiomitigative targets.


Asunto(s)
Araquidonato 15-Lipooxigenasa/genética , Ferroptosis/genética , Regulación Neoplásica de la Expresión Génica , Leucotrienos/genética , Peróxidos Lipídicos/genética , Pseudomonas aeruginosa/efectos de la radiación , Traumatismos Experimentales por Radiación/genética , Animales , Araquidonato 15-Lipooxigenasa/biosíntesis , Células CACO-2/efectos de la radiación , Femenino , Humanos , Leucotrienos/metabolismo , Peróxidos Lipídicos/metabolismo , Ratones , Ratones Endogámicos C57BL , Pseudomonas aeruginosa/patogenicidad , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología
18.
Microbiol Spectr ; 10(1): e0146321, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35019684

RESUMEN

Anthranilate is a diffusible molecule produced by Pseudomonas aeruginosa and accumulates as P. aeruginosa grows. Anthranilate is an important intermediate for the synthesis of tryptophan and the Pseudomonas quinolone signal (PQS), as well as metabolized by the anthranilate dioxygenase complex (antABC operon products). Here we demonstrate that anthranilate is a key factor that modulates the pathogenicity-related phenotypes of P. aeruginosa and other surrounding bacteria in the environment, such as biofilm formation, antibiotic tolerance, and virulence. We found that the anthranilate levels in P. aeruginosa cultures rapidly increased in the stationary phase and then decreased again, forming an anthranilate peak. Biofilm formation, antibiotic susceptibility, and virulence of P. aeruginosa were significantly altered before and after this anthranilate peak. In addition, these phenotypes were all modified by the mutation of antABC and exogenous addition of anthranilate. Anthranilate also increased the antibiotic susceptibility of other species of bacteria, such as Escherichia coli, Salmonella enterica, Bacillus subtilis, and Staphylococcus aureus. Before the anthranilate peak, the low intracellular anthranilate level was maintained through degradation from the antABC function, in which induction of antABC was also limited to a small extent. The premature degradation of anthranilate, due to its high levels, and antABC expression early in the growth phase, appears to be toxic to the cells. From these results, we propose that by generating an anthranilate peak as a signal, P. aeruginosa may induce some sort of physiological change in surrounding cells. IMPORTANCE Pseudomonas aeruginosa is a notorious pathogen with high antibiotic resistance, strong virulence, and ability to cause biofilm-mediated chronic infection. We found that these characteristics change profoundly before and after the time when anthranilate is produced as an "anthranilate peak". This peak acts as a signal that induces physiological changes in surrounding cells, decreasing their antibiotic tolerance and biofilm formation. This study is important in that it provides a new insight into how microbial signaling substances can induce changes in the pathogenicity-related phenotypes of cells in the environment. In addition, this study shows that anthranilate can be used as an adjuvant to antibiotics.


Asunto(s)
Antibacterianos/farmacología , Biopelículas , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/fisiología , ortoaminobenzoatos/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Biopelículas/efectos de los fármacos , Humanos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidad , Salmonella enterica/efectos de los fármacos , Salmonella enterica/genética , Salmonella enterica/metabolismo , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Virulencia
19.
PLoS Pathog ; 18(1): e1010170, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34986198

RESUMEN

Pseudomonas aeruginosa is an important opportunistic pathogen capable of causing variety of infections in humans. The type III secretion system (T3SS) is a critical virulence determinant of P. aeruginosa in the host infections. Expression of the T3SS is regulated by ExsA, a master regulator that activates the expression of all known T3SS genes. Expression of the exsA gene is controlled at both transcriptional and posttranscriptional levels. Here, we screened a P. aeruginosa transposon (Tn5) insertional mutant library and found rplI, a gene coding for the ribosomal large subunit protein L9, to be a repressor for the T3SS gene expression. Combining real-time quantitative PCR (qPCR), western blotting and lacZ fusion assays, we show that RplI controls the expression of exsA at the posttranscriptional level. Further genetic experiments demonstrated that RplI mediated control of the exsA translation involves 5' untranslated region (5' UTR). A ribosome immunoprecipitation assay and qPCR revealed higher amounts of a 24 nt fragment from exsA mRNA being associated with ribosomes in the ΔrplI mutant. An interaction between RplI and exsA mRNA harboring its 24 nt, but not 12 nt, 5' UTR was confirmed by RNA Gel Mobility Shift and Microscale Thermophoresis assays. Overall, this study identifies the ribosomal large subunit protein L9 as a novel T3SS repressor that inhibits ExsA translation in P. aeruginosa.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica/fisiología , Pseudomonas aeruginosa/patogenicidad , Proteínas Ribosómicas/metabolismo , Transactivadores/metabolismo , Sistemas de Secreción Tipo III/metabolismo , Regiones no Traducidas 5' , Células HeLa , Humanos , Pseudomonas aeruginosa/metabolismo , Transcripción Genética , Virulencia/fisiología , Factores de Virulencia/metabolismo
20.
Biochim Biophys Acta Biomembr ; 1864(1): 183769, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34506798

RESUMEN

Currently, multidrug-resistant bacteria are rapidly increasing worldwide because of the misuse or overuse of antibiotics. In particular, few options exist for treating infections caused by long-persisting oxacillin-resistant strains and recently proliferating carbapenem-resistant strains. Therefore, alternative treatments are urgently needed. The antimicrobial peptide (AMP) Lycosin-II is a peptide consisting of 21 amino acids isolated from the venom of the spider Lycosa singoriensis. Lycosin-II showed strong antibacterial activity and biofilm inhibition effects against gram-positive and gram-negative bacteria including oxacillin-resistant Staphylococcus aureus (S. aureus) and meropenem-resistant Pseudomonas aeruginosa (P. aeruginosa) isolated from patients. In addition, Lycosin-II was not cytotoxic against human foreskin fibroblast Hs27 or hemolytic against sheep red blood cells at the concentration of which exerted antibacterial activity. The mechanism of action of Lycosin-II involves binding to lipoteichoic acid and lipopolysaccharide of gram-positive and gram-negative bacterial membranes, respectively, to destroy the bacterial membrane. Moreover, Lycosin-II showed anti-inflammatory effects by inhibiting the expression of pro-inflammatory cytokines that are increased during bacterial infection in Hs27 cells. These results suggest that Lycosin-II can serve as a therapeutic agent against infections with multidrug-resistant strains.


Asunto(s)
Péptidos Antimicrobianos/química , Biopelículas/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Venenos de Araña/química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Péptidos Antimicrobianos/farmacología , Citocinas/antagonistas & inhibidores , Citocinas/genética , Eritrocitos/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/patogenicidad , Bacterias Grampositivas/efectos de los fármacos , Bacterias Grampositivas/patogenicidad , Humanos , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Fagocitosis/efectos de los fármacos , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/patogenicidad , Ovinos , Venenos de Araña/farmacología , Arañas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...