Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.446
Filter
1.
Microbiol Spectr ; : e0079424, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39365045

ABSTRACT

Bordetella bronchiseptica is a pathogenic bacterium that causes respiratory infections in mammals. Adhesins, toxins, and secretion systems necessary for infection are regulated by the two-component system BvgAS. When the BvgAS system is inactive, there is no transcription of virulence-activated genes, and virulence-repressed genes (vrg) are expressed. The regulation of some vrgs in B. bronchiseptica is dependent upon the virulence-activated gene bvgR. Although having a regulatory role, no DNA-binding domain is described for BvgR. Instead, it contains an EAL domain, usually found in cyclic-di-GMP (c-di-GMP)-specific phosphodiesterases. c-di-GMP is a bacterial second messenger that regulates multiple phenotypes in bacteria, including B. bronchiseptica. The current study aimed to deepen our knowledge about BvgR. We employed RNA-seq analysis to define the BvgR regulon, and then we investigated the phenotypes in which BvgR regulation might be involved such as biofilm formation, cytotoxicity, and virulence. Our result revealed that BvgR inhibits biofilm formation and flagellin expression in virulent phase. Although BvgR has long been considered a repressor protein, our results show that it also upregulates almost 100 genes. This regulation is likely indirect, as BvgR lacks a DNA-binding domain. Notably, among the upregulated genes, we identified 15 associated with the type three secretion system. Consistent with these findings, a B. bronchiseptica strain deficient in bvgR was less cytotoxic than the wild-type strain, elicited a milder immune response, and was less able to persist in the lower respiratory tract of mice.IMPORTANCEBordetella bronchiseptica is a harmful bacterium responsible for respiratory infections in mammals. Its ability to cause disease is tightly regulated by a system called BvgAS. In this study, we focused on understanding the role of a specific gene called bvgR in regulating B. bronchiseptica's virulence factors. Our findings revealed that BvgR, previously thought to primarily repress gene expression, actually plays a complex role in both activating and inhibiting various genes involved in bacterial virulence. This newfound understanding sheds light on the intricate mechanisms underlying B. bronchiseptica's ability to cause infections, providing valuable insights for developing strategies to combat these infections in humans and animals.

2.
mLife ; 3(3): 445-458, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39359676

ABSTRACT

Quorum sensing (QS) inhibition has emerged as a promising target for directed drug design, providing an appealing strategy for developing antimicrobials, particularly against infections caused by drug-resistant pathogens. In this study, we designed and synthesized a total of 33 ß-nitrostyrene derivatives using 1-nitro-2-phenylethane (NPe) as the lead compound, to target the facultative anaerobic bacterial pathogen Serratia marcescens. The QS-inhibitory effects of these compounds were evaluated using S. marcescens NJ01 and the reporter strain Chromobacterium violaceum CV026. Among the 33 new ß-nitrostyrene derivatives, (E)-1-methyl-4-(2-nitrovinyl)benzene (m-NPe, compound 28) was proven to be a potent inhibitor that reduced biofilm formation of S. marcescens NJ01 by 79%. Scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM) results revealed that treatment with m-NPe (50 µg/ml) not only enhanced the susceptibility of the formed biofilms but also disrupted the architecture of biofilms by 84%. m-NPe (50 µg/ml) decreased virulence factors in S. marcescens NJ01, reducing the activity of protease, prodigiosin, and extracellular polysaccharide (EPS) by 36%, 72%, and 52%, respectively. In S. marcescens 4547, the activities of hemolysin and EPS were reduced by 28% and 40%, respectively, outperforming the positive control, vanillic acid (VAN). The study also found that the expression levels of QS- and biofilm-related genes (flhD, fimA, fimC, sodB, bsmB, pigA, pigC, and shlA) were downregulated by 1.21- to 2.32-fold. Molecular dynamics analysis showed that m-NPe could bind stably to SmaR, RhlI, RhlR, LasR, and CviR proteins in a 0.1 M sodium chloride solution. Importantly, a microscale thermophoresis (MST) test revealed that SmaR could be a target protein for the screening of a quorum sensing inhibitor (QSI) against S. marcescens. Overall, this study highlights the efficacy of m-NPe in suppressing the virulence factors of S. marcescens, identifying it as a new potential QSI and antibiofilm agent capable of restoring or improving antimicrobial drug sensitivity.

3.
Front Cell Infect Microbiol ; 14: 1419570, 2024.
Article in English | MEDLINE | ID: mdl-39386171

ABSTRACT

Microbial biofilms play a pivotal role in microbial infections and antibiotic resistance due to their unique properties, driving the urgent need for advanced methodologies to study their behavior comprehensively across varied environmental contexts. While electrochemical biosensors have demonstrated success in understanding the dynamics of biofilms, scientists are now synergistically merging these biosensors with microfluidic technology. This combined approach offers heightened precision, sensitivity, and real-time monitoring capabilities, promising a more comprehensive understanding of biofilm behavior and its implications. Our review delves into recent advancements in electrochemical biosensors on microfluidic chips, specifically tailored for investigating biofilm dynamics, virulence, and properties. Through a critical examination of these advantages, properties and applications of these devices, the review highlights the transformative potential of this technology in advancing our understanding of microbial biofilms in different settings.


Subject(s)
Biofilms , Biosensing Techniques , Electrochemical Techniques , Microfluidics , Biofilms/growth & development , Biosensing Techniques/methods , Biosensing Techniques/instrumentation , Electrochemical Techniques/methods , Electrochemical Techniques/instrumentation , Microfluidics/methods , Microfluidics/instrumentation , Humans , Bacteria , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods
4.
Front Microbiol ; 15: 1399996, 2024.
Article in English | MEDLINE | ID: mdl-39386371

ABSTRACT

Staphylococcus aureus is a bacterial pathogen of considerable significance in public health, capable of inducing a diverse range of infectious diseases. One of the most notorious mechanisms used by S. aureus to survive and colonize the site of infection is its ability to form biofilms. Diflunisal, a non-steroidal anti-inflammatory drug (NSAID), is a known inhibitor of the Agr system in S. aureus, which is key in regulating biofilm formation. This study evaluated the effect of broad-spectrum antibiotics in combination with diflunisal on S. aureus biofilm density. Eight antibiotics were tested independently at different concentrations and in combination with diflunisal to assess their effect on S. aureus biofilm formation. When using the antibiotics alone and with diflunisal, a significant control effect on biofilm formation was observed (p < 0.05), irrespective of diflunisal presence, but did not achieve a complete biofilm growth inhibition. Over time, diflunisal influenced biofilm formation; however, such an effect was correlated with antibiotic concentration and exposure time. With amikacin treatments, biofilm density increased with extended exposure time. In the case of imipenem, doripenem, levofloxacin, and ciprofloxacin, lower doses and absence of diflunisal showed higher control over biofilm growth with longer exposure. However, in all cases, diflunisal did not significantly affect the treatment effect on biofilm formation. In the absence of antibiotics, diflunisal significantly reduced biofilm formation by 53.12% (p < 0.05). This study suggests that diflunisal could be a potential treatment to control S. aureus biofilms, but it does not enhance biofilm inhibition when combined with antibiotics.

5.
Adv Healthc Mater ; : e2402659, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39388414

ABSTRACT

The World Health Organization has warned that without effective action, deaths from drug-resistant bacteria can exceed 10 million annually, making it the leading cause of death. Conventional antibiotics are becoming less effective due to rapid bacterial drug resistance and slowed new antibiotic development, necessitating new strategies. Recently, materials with catalytic/enzymatic properties, known as nanozymes, have been developed, inspired by natural enzymes essential for bacterial eradication. Unlike recent literature reviews that broadly cover nanozyme design and biomedical applications, this review focuses on the latest advancements in nanozymes for combating bacterial drug resistance, emphasizing their design, structural characteristics, applications in combination therapy, and future prospects. This approach aims to promote nanozyme development for combating bacterial drug resistance, especially towards clinical translation.

6.
Pediatr Pulmonol ; 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39392258

ABSTRACT

Nontypeable Haemophilus influenzae (NTHi) and Streptococcus pneumoniae (pneumococcus) are pathobionts that share common environmental niches within the upper respiratory tract. They can form part of the resident upper airway microbiota, but under certain environmental circumstances become pathogenic and induce disease. In children, both organisms have a considerable impact on the healthcare system, commonly causing acute otitis media and pneumonia. They are also associated with chronic biofilm-mediated respiratory infections, such as persistent middle ear effusions and chronic suppurative otitis media, and in the lower airways with protracted bacterial bronchitis and bronchiectasis. Consequently, both organisms are responsible for large numbers of antibiotic prescriptions and substantial healthcare costs. The complex relationship between NTHi and pneumococcal co-interaction during colonization, infection and biofilm formation is poorly understood and a greater understanding is needed to facilitate development of future therapies, and novel interventions and prevention strategies. Co-infections with both bacteria can result in more severe disease, with disease severity likely mediated by their ability to cooperate in some in vivo niches. However, this relationship is not always straightforward, as under certain conditions, these two bacteria compete rather than cooperate. Current opinion supports developing a vaccine targeting NTHi strains, as well as a combined vaccine targeting both NTHi and pneumococci to decrease the respiratory disease burden in young children. This review summarizes our current knowledge of the interactions between NTHi and pneumococci and speculates on the future directions of research to understand how these bacteria co-exist and how to better prevent and treat NTHi and pneumococcal infection.

7.
Bioresour Technol ; : 131567, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39393651

ABSTRACT

Microbial biofilms provide advantages in fermentation processes. However, Corynebacterium glutamicum (C. glutamicum) usually exhibits relatively poor biofilm formation compared to other industrial strains. To develop a biofilm-enhanced fermentation process for C. glutamicum, seven genes potentially related to biofilm formation in C. glutamicum were systematically investigated, which include ppk2B, glgC, virB11, cslA, NCgl2909, NCgl0350 and exeR. Deletion of the NCgl0350, NCgl2909 genes and heterologous expression of the cslA gene were found to increase biofilm amounts by 16.9%, 21.2% and 135%, respectively, compared to the wild-type strain. Meanwhile, the production of L-lysine by engineered strains was assessed in biofilm-based continuous fermentation. The most notable result was observed for the cslA-expressing strain, which produced an average of 26.1% higher L-lysine compared with that of wild-type strain in 6-L bioreactors. In conclusion, this study offers valuable insights into the biofilm formation in C. glutamicum and its application in continuous fermentation processes.

8.
Microb Pathog ; 196: 106993, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39374884

ABSTRACT

Otitis externa is an inflammatory disease of the external ear canal of complex and multifactorial etiology associated with recurrent bacterial infection. This study aimed to assess the antimicrobial and antibiofilm activity of promethazine against bacterial isolates from dogs with otitis externa, as well as the effect of this compound on the dynamics of biofilm formation over 120 h. Planktonic bacterial susceptibility to promethazine was evaluated to determine the minimum inhibitory concentrations (MIC). The minimum biofilm eradication concentration (MBEC) was also determined by broth microdilution. To evaluate the effect on biofilm growth, promethazine was tested at three concentrations MIC, MIC/2 and MIC/8, with daily readings at 48, 72, 96 and 120 h. The MICs of promethazine ranged from 48.83 to 781.25 µg mL-1. Promethazine significantly (P < 0.05) reduced mature biofilm biomass, with MBECs ranging from 48.8 to 6250 µg mL-1 and reduced (P < 0.01) biofilm formation for up to the 120-h, at concentrations corresponding to the MIC obtained against each isolate. Promethazine was effective against microorganisms associated with canine otitis externa. The data suggest that promethazine presents antimicrobial and antibiofilm activity and is a potential alternative to treat and prevent recurrent bacterial otitis in dogs. These results emphasize the importance of drug repurposing in veterinary otology as an alternative to reduce antimicrobial resistance.

9.
Indian J Otolaryngol Head Neck Surg ; 76(5): 3886-3894, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39376426

ABSTRACT

Chronic Suppurative Otitis Media (CSOM) is an inflammation of the mucoperiosteal lining of the middle ear cleft. Recently, biofilms have been discovered to play a pivotal role in the pathogenesis of CSOM. A biofilm is a bacterial aggregation that adheres to the mucosal surfaces and is connected with an extracellular matrix. Biofilms enhance antibiotic resistance, facilitate genetic alterations and amplify competence to combat host immunity. This study aims to identify the spectrum of biofilm-producers in CSOM and investigate their antibiotic sensitivity. Samples (648) were obtained from the deeper part of external auditory meatus of patients with CSOM. Pus samples were collected and processed for culture sensitivity. Biofilms detected. The findings were compiled and statistically analyzed. Out of 500 culture-positive samples, most commonly isolated bacteria was Pseudomonas (62.6%), followed by MRSA (13.8%). Biofilm-producers were 350, with 119 being strong, 167 moderate, and 64 weak. Biofilms were produced by 70% of the isolates, with Pseudomonas producing the most (74.6%), followed by MRSA. Gentamicin was the most effective antibiotic against biofilm-producers. Amoxicillin-Clavulanic Acid, Ceftriaxone, Cefuroxime, and Minocyclin were resistant. Pseudomonas had the highest sensitivity to Levofloxacin (96.6%), followed by Ceftazidime and Ciprofloxacin. Pseudomonas was resistant to Cefuroxime, Amoxicillin-Clavulanic acid and Linezolid. Multi-drug resistance has been widespread among CSOM causal species, particularly in biofilm producers. Thus, screening for biofilm formation, in addition to the standard antibiogram, must be undertaken as part of CSOM protocol. This will address the multi-drug resistance and select an appropriate treatment modality.

10.
Biofouling ; : 1-15, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39391921

ABSTRACT

Extracts of certain fodder grasses may be viewed as powerful agents against infections induced by avian pathogenic Escherichia coli strains. Here we demonstrated ability of Galega orientalis and Rhaponticum carthamoides extracts, alone or in combination with antibiotics, to inhibit growth, viability and biofilm formation in avian pathogenic Escherichia coli strains with different sensitivity to antibiotics and non-pathogenic laboratory strain E. coli BW25113 as well as its mutant derivatives. Modulation of motility and production of extracellular structures in the presence of the extracts correlated with their anti-biofilm effects. Interestingly, an increase in antibacterial action of kanamycin, streptomycin, ciprofloxacin, and cefotaxime on both biofilms and planktonic cultures of the studied strains was observed in the presence of the extracts, including antibiotic resistant APEC strain #45. The extracts alone showed weak prooxidant activity which could contribute to modification of redox-sensitive sites of various regulatory circuits, resulting to synergetic effects in combination with antibiotics.

11.
J Basic Microbiol ; : e2400271, 2024 Oct 11.
Article in English | MEDLINE | ID: mdl-39392011

ABSTRACT

Biofilms are complex communities of microorganisms that can cause significant challenges in various settings, including industrial processes, environmental systems, and human health. The protective nature of biofilms makes them resistant to traditional anti-biofilm strategies, such as chemical agents, mechanical interventions, and surface modifications. To address the limitations of conventional anti-biofilm methods, researchers have explored emerging strategies that encompass the use of natural compounds, nanotechnology-based methods, quorum-sensing inhibition, enzymatic degradation, and antimicrobial photodynamic/sonodynamic therapy. There is an increasing focus on combining multiple anti-biofilm strategies to combat resistance and enhance effectiveness. Researchers are continuously investigating the mechanisms of biofilm formation and developing innovative approaches to overcome the limitations of conventional anti-biofilm methods. These efforts aim to improve the management of biofilms and prevent infections while preserving the environment. This study provides a comprehensive overview of the latest advancements in anti-biofilm strategies. Given the dynamic nature of this field, exploring new approaches is essential to stimulate further research and development initiatives. The effective management of biofilms is crucial for maintaining the health of industrial processes, environmental systems, and human populations.

12.
Sci Rep ; 14(1): 23572, 2024 10 09.
Article in English | MEDLINE | ID: mdl-39384811

ABSTRACT

Klebsiella pneumoniae has been identified as one of the most important opportunistic pathogens responsible for nosocomial infections. Antibiotic resistance and the ability to form biofilms are the two main factors involved in the persistence of infections. Conventional detection methods involve culture isolation and identification followed by biofilm assay that takes 48-72 h. Timely detection of biofilm-forming resistant pathogens is essential to appropriately treat the infection with the right dose and combinations. The present study focuses on evaluating an RT-PCR panel using mrkD, pgaC, and wcaJ genes to screen for biofilm-forming K. pneumoniae from ETA/BAL specimens. The assay accurately identified K. pneumoniae harboring samples with a limit of detection of 1 ng/µl total RNA. Representative culture-negative-PCR-positive samples were subjected to metagenomics which identified K. pneumoniae reads in these samples confirming the specificity of RT-PCR. mrkD and pgaC act as K. pneumoniae specific identification whereas wcaJ acts as a negative marker for biofilm-forming K. pneumoniae. In addition, RT-PCR results correlated well with the phenotypic biofilm-forming assay. This RT-PCR assay is the first of its kind for rapid identification of biofilm-forming K. pneumoniae. The result of this study highlights that the rapid detection of K. pneumoniae biofilms based on the RT-PCR results coupled with clinical conditions would be appropriate to treat emerging infections or to prevent re-infections in clinical settings.


Subject(s)
Biofilms , Biomarkers , Klebsiella Infections , Klebsiella pneumoniae , Biofilms/growth & development , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , Humans , Klebsiella Infections/microbiology , Klebsiella Infections/diagnosis , Bronchoalveolar Lavage Fluid/microbiology , Bacterial Proteins/genetics
13.
Int Wound J ; 21(10): e70059, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39359044

ABSTRACT

Chronic non-healing wounds pose significant challenges due to an elevated inflammatory response caused in part by bacterial contamination (Physiol Rev. 2019;99:665). These wounds lead to billions being spent in the health care system worldwide (N Engl J Med. 2017;376:2367, Int J Pharm. 2014;463:119). We studied the in-vitro and in-vivo antimicrobial effects of a multimodal wound matrix (MWM) against two common wound pathogens, Methicillin-Resistant Staphylococcus aureus (MRSA USA300) and Pseudomonas aeruginosa ATCC 27312 (PA27312) (Int Wound J. 2019;16:634). The in-vitro study conducted was a zone of inhibition test with the two microbes at 104 Log CFU/mL inoculated on Tryptic soy agar with 5% sheep blood (TSAII) plates. Treatments used were MWM, Mupirocin (Positive control for MRSA), Silver Sulfadiazine (Positive Control for PA), Petrolatum and Sterile Saline (both serving as Negative Controls). Treatments were allowed to diffuse into the agar for 3 h and then were incubated for 24 h at 37°C. The in-vivo study utilized a deep dermal porcine wound model (22 × 22 × 3 mm) created on six animals. Three animals were inoculated with MRSA USA300 and the other three with PA27312 with each allowing a 72-h biofilm formation. After 72 h, baseline wounds were assessed for bacterial concentration and all remaining wounds were treated with either MWM alone, Silver Treatment or Untreated Control. Wounds were assessed on days 4, 8 and 12 after treatment application for microbiological analysis. In-vitro, MWM exhibited significant inhibition of MRSA USA300 and PA27312 growth when compared to negative controls (p ≤ 0.05). Likewise, in-vivo, the MWM-treated wounds exhibited a significant (p ≤ 0.05) bacterial reduction compared to all other treatment groups, especially on days 8 and 12 for both pathogens. MWM demonstrated promise in addressing colonized wounds with biofilms. Additional studies on MWM's benefits and comparisons with existing treatments are warranted to optimize wound care strategies (Adv Wound Care. 2021;10:281).


Subject(s)
Disease Models, Animal , Methicillin-Resistant Staphylococcus aureus , Pseudomonas aeruginosa , Wound Infection , Animals , Methicillin-Resistant Staphylococcus aureus/drug effects , Pseudomonas aeruginosa/drug effects , Swine , Wound Infection/drug therapy , Wound Infection/microbiology , Wound Healing/drug effects , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacology , Staphylococcal Infections/drug therapy , Pseudomonas Infections/drug therapy , Microbial Sensitivity Tests , Bandages
14.
Mar Environ Res ; 202: 106777, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39368156

ABSTRACT

As emerging pollutants, microplastics can aggregate microorganisms on their surfaces and form biofilms, enriching antibiotic-resistant bacteria (ARB) and antibiotic resistance genes (ARGs). Consequently, microplastic biofilms have become a focal point of research. Horizontal gene transfer is one of the primary mechanisms by which bacteria acquire antibiotic resistance, with much of the research focusing on suspended bacteria. However, microplastic biofilms, as hotspots for horizontal gene transfer, also merit significant investigation. This study primarily explored and compared the frequency of ARG conjugative transfer between suspended bacteria and microplastic biofilms. The results demonstrated that, compared to suspended bacteria, microplastic biofilms enhanced the frequency of ARG conjugative transfer by 7.2-19.6 times. Among them, biofilms on polyethylene microplastics showed the strongest promotion of conjugation. After the formation of microplastic biofilms, there was a significant increase in bacterial density within the biofilms, which raised the collision frequency of donor and recipient bacteria. Then microplastic biofilms facilitated the gene expression levels of outer membrane proteins, enhanced bacterial gene transfer capabilities, promoted the synthesis of conjugative pili, accelerated the formation of conjugative pairing systems, and elevated the expression levels of genes related to DNA replication and transfer systems, thereby enhancing the conjugative transfer of ARGs within microplastic biofilms. Among different types of microplastic biofilms, polyethylene biofilms exhibited the highest bacterial density, thus showing the highest frequency of ARG conjugation. This study highlights the risks associated with ARG conjugative transfer following the formation of microplastic biofilms and provides insights into the risks of microplastic and antibiotic resistance propagation in estuarine environments.

15.
Biomater Adv ; 166: 214049, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39368439

ABSTRACT

Predicting how tooth and dental material bonds perform in the mouth requires a deep understanding of degrading factors. Yet, this understanding is incomplete, leading to significant uncertainties in designing and evaluating new dental adhesives. The durability of dental bonding interfaces in the oral microenvironment is compromised by bacterial acids, salivary enzymes, and masticatory fatigue. These factors degrade the bond between dental resins and tooth surfaces, making the strength of these bonds difficult to predict. Traditionally studied separately, a combined kinetic analysis of these interactions could enhance our understanding and improvement of dental adhesive durability. To address this issue, we developed and validated an original model to evaluate the bond strength of dental restorations using realistic environments that consider the different mechanical, chemical, and biological degradative challenges working simultaneously: bacteria, salivary esterases, and cyclic loading. We herein describe a comprehensive investigation on dissociating the factors that degrade the bond strength of dental restorations. Our results showed that cariogenic bacteria are the number one factor contributing to the degradation of the bonded interface, followed by cyclic loading and salivary esterases. When tested in combinatorial mode, negative and positive synergies towards the degradation of the interface were observed. Masticatory loads (i.e., cycling loading) enhanced the lactic acid bacterial production and the area occupied by the biofilm at the bonding interface, resulting in more damage at the interface and a reduction of 73 % in bond strength compared to no-degraded samples. Salivary enzymes also produced bond degradation caused by changes in the chemical composition of the resin/adhesive. However, the degradation rates are slowed compared to the bacteria and cyclic loading. These results demonstrate that our synergetic model could guide the design of new dental adhesives for biological applications without laborious trial-and-error experimentation.

16.
Sci Rep ; 14(1): 23078, 2024 10 04.
Article in English | MEDLINE | ID: mdl-39366990

ABSTRACT

Endoscopic lung volume reduction (ELVR) using endobronchial valves (EBV) is a treatment option for a subset of patients with severe chronic obstructive pulmonary disease (COPD), suffering from emphysema and hyperinflation. In this pilot study, we aimed to determine the presence of bacterial biofilm infections on EBV and investigate their involvement in lack of clinical benefits, worsening symptomatology, and increased exacerbations that lead to the decision to remove EBVs. We analyzed ten COPD patients with ELVR who underwent EBV removal. Clinical data were compared to the microbiological findings from conventional EBV culture. In addition, EBV were analyzed by FISHseq, a combination of Fluorescence in situ hybridization (FISH) with PCR and sequencing, for visualization and identification of microorganisms and biofilms. All ten patients presented with clinical symptoms, including pneumonia and recurrent exacerbations. Microbiological cultures from EBV detected several microorganisms in all ten patients. FISHseq showed either mixed or monospecies colonization on the EBV, including oropharyngeal bacterial flora, Staphylococcus aureus, Pseudomonas aeruginosa, Streptococcus spp., and Fusobacterium sp. On 5/10 EBV, FISHseq visualized biofilms, on 1/10 microbial microcolonies, on 3/10 single microorganisms, and on 1/10 no microorganisms. The results of the study demonstrate the presence of biofilms on EBV for the first time and its potential involvement in increased exacerbations and clinical worsening in patients with ELVR. However, further prospective studies are needed to evaluate the clinical relevance of biofilm formation on EBV and appropriate treatment options to avoid infections in patients with ELVR.


Subject(s)
Biofilms , In Situ Hybridization, Fluorescence , Pulmonary Disease, Chronic Obstructive , Humans , Biofilms/growth & development , Pilot Projects , Male , Pulmonary Disease, Chronic Obstructive/microbiology , Female , Aged , Middle Aged , Pneumonectomy/methods
17.
Article in English | MEDLINE | ID: mdl-39406584

ABSTRACT

Complications of cholesteatoma result from characteristic inflammatory and resorptive processes that erode the structures of the middle and inner ear with potential to spread locally. Common intratemporal complications include hearing loss, facial nerve palsy, labyrinthine fistula, and dysgeusia. Extratemporal complications, though less common, may be life-threatening, and include cerebrospinal fluid leak and encephalocele, meningitis, epidural and intraparenchymal abscesses, subdural empyema, and otic hydrocephalus.

18.
Biofouling ; : 1-13, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39377105

ABSTRACT

Candida auris and Staphylococcus aureus are associated with a wide range of infections, as they exhibit multidrug resistance - a growing health concern. In this study, gaseous ozone, and ultraviolet-C (UVC) radiation are applied as infection control measures to inactivate dry biofilms of these organisms on polystyrene surfaces. The dosages utilised herein are 1000 and 3000 ppm.min for ozone and 2864 and 11592 mJ.cm-2 for UVC. Both organisms showed an increased sensitivity to UVC relative to ozone exposure in a bespoke decontamination chamber. While complete inactivation of both organisms (>7.5 CFU log) was realized after 60 mins of UVC application, this could not be achieved with ozonation for the same duration. However, a combined application of ozone and UVC yielded complete inactivation in only 20 mins. For both treatment methods, it was observed that dry biofilms of S. aureus were more difficult to inactivate than dry biofilms of C. auris. Compared to dry biofilms of C. auris, micrographs of wet C. auris biofilms revealed the presence of an abundance of extracellular material after treatments. Interestingly, wet biofilms were more difficult to inactivate than dry biofilms. These insights are crucial to preventing recalcitrant and recurrent infections via contact with contaminated polymeric surfaces.


Oxidative treatment can inactivate dry biofilms formed by C. auris and S. aureus.Both organisms showed an increased sensitivity to UVC compared to ozone.Dry biofilms of S. aureus were more difficult to inactivate than dry biofilms of C. auris.Wet biofilms of C. auris display a spongy appearance compared to its dry biofilms.Wet biofilms of C. auris proved more difficult to inactivate than its dry biofilms.

19.
Arch Microbiol ; 206(11): 432, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39402397

ABSTRACT

Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.


Subject(s)
Bacteria , Bacteriophages , Biofilms , Biofilms/drug effects , Bacteria/drug effects , Bacteria/metabolism , Bacteria/genetics , Bacteriophages/physiology , Extracellular Polymeric Substance Matrix/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology
20.
mSystems ; : e0031724, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39404262

ABSTRACT

Marine biofilms were newly revealed as a giant microbial diversity pool for global oceans. However, the cyanobacterial diversity in marine biofilms within the upper seawater column and its ecological and evolutionary implications remains undetermined. Here, we reconstructed a full picture of modern marine cyanobacteria habitats by re-analyzing 9.3 terabyte metagenomic data sets and 2,648 metagenome-assembled genomes (MAGs). The abundances of cyanobacteria lineages exclusively detected in marine biofilms were up to ninefold higher than those in seawater at similar sample size. Analyses revealed that cyanobacteria in marine biofilms are specialists with strong geographical and environmental constraints on their genome and functional adaption, which is in stark contrast to the generalistic features of seawater-derived cyanobacteria. Molecular dating suggests that the important diversifications in biofilm-forming cyanobacteria appear to coincide with the Great Oxidation Event (GOE), "boring billion" middle Proterozoic, and the Neoproterozoic Oxidation Event (NOE). These new insights suggest that marine biofilms are large and important cyanobacterial factories for the global oceans. IMPORTANCE: Cyanobacteria, highly diverse microbial organisms, play a crucial role in Earth's oxygenation and biogeochemical cycling. However, their connection to these processes remains unclear, partly due to incomplete surveys of oceanic niches. Our study uncovered significant cyanobacterial diversity in marine biofilms, showing distinct niche differentiation compared to seawater counterparts. These patterns reflect three key stages of marine cyanobacterial diversification, coinciding with major geological events in the Earth's history.

SELECTION OF CITATIONS
SEARCH DETAIL