Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Biofilm ; 7: 100178, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38317668

ABSTRACT

Biofilm formation by the pathobiont Haemophilus influenzae is associated with human nasopharynx colonization, otitis media in children, and chronic respiratory infections in adults suffering from chronic respiratory diseases such as chronic obstructive pulmonary disease (COPD). ß-lactam and quinolone antibiotics are commonly used to treat these infections. However, considering the resistance of biofilm-resident bacteria to antibiotic-mediated killing, the use of antibiotics may be insufficient and require being replaced or complemented with novel strategies. Moreover, unlike the standard minimal inhibitory concentration assay used to assess antibacterial activity against planktonic cells, standardization of methods to evaluate anti-biofilm drug activity is limited. In this work, we detail a panel of protocols for systematic analysis of drug antimicrobial effect on bacterial biofilms, customized to evaluate drug effects against H. influenzae biofilms. Testing of two cinnamaldehyde analogs, (E)-trans-2-nonenal and (E)-3-decen-2-one, demonstrated their effectiveness in both H. influenzae inhibition of biofilm formation and eradication or preformed biofilms. Assay complementarity allowed quantifying the dynamics and extent of the inhibitory effects, also observed for ampicillin resistant clinical strains forming biofilms refractory to this antibiotic. Moreover, cinnamaldehyde analog encapsulation into poly(lactic-co-glycolic acid) (PLGA) polymeric nanoparticles allowed drug vehiculization while maintaining efficacy. Overall, we demonstrate the usefulness of cinnamaldehyde analogs against H. influenzae biofilms, present a test panel that can be easily adapted to a wide range of pathogens and drugs, and highlight the benefits of drug nanoencapsulation towards safe controlled release.

2.
Article in English | MEDLINE | ID: mdl-38403876

ABSTRACT

Microbial biofilms are complex three-dimensional structures where sessile microbes are embedded in a polymeric extracellular matrix. Their resistance toward the host immune system as well as to a diverse range of antimicrobial treatments poses a serious health and development threat, being in the top 10 global public health threats declared by the World Health Organization. In an effort to combat biofilm-related microbial infections, several strategies have been developed to independently eliminate biofilms or to complement conventional antibiotic therapies. However, their limitations leave room for other treatment alternatives, where the application of nanotechnology to biofilm eradication has gained significant relevance in recent years. Their small size, penetration efficiency, and the design flexibility that they present makes them a promising alternative for biofilm infection treatment, although they also present set-backs. This review aims to describe the main possibilities and limitations of nanomedicine against biofilms, while covering the main aspects of biofilm formation and study, and the current therapies for biofilm treatment. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.


Subject(s)
Anti-Infective Agents , Nanomedicine , Biofilms , Anti-Bacterial Agents/therapeutic use , Polymers
3.
Front Cell Infect Microbiol ; 13: 1323619, 2023.
Article in English | MEDLINE | ID: mdl-38156315

ABSTRACT

Introduction: Candida parapsilosis, a pathogenic yeast associated with systemic infections, exhibits metabolic adaptability in response to nutrient availability. Methods: We investigated the impact of RPMI glucose supplemented (RPMId), TSB, BHI and YPD media on C. parapsilosis growth, morphology, susceptibility (caspofungin and amphotericin B), and in vivo virulence (Galleria mellonella) in planktonic and biofilm states. Results: High-glucose media favors growth but hinders metabolic activity and filamentation. Media promoting carbohydrate production reduces biofilm susceptibility. Virulence differences between planktonic cells and biofilm suspensions from the same media shows that biofilm-related factors influence infection outcome depending on nutrient availability. Pseudohyphal growth occurred in biofilms under low oxygen and shear stress, but its presence is not exclusively correlated with virulence. Discussion: This study provides valuable insights into the intricate interplay between nutrient availability and C. parapsilosis pathogenicity. It emphasizes the importance of considering pathogen behavior in diverse conditions when designing research protocols and therapeutic strategies.


Subject(s)
Antifungal Agents , Candida parapsilosis , Antifungal Agents/pharmacology , Antifungal Agents/therapeutic use , Virulence , Culture Media , Biofilms , Microbial Sensitivity Tests , Saccharomyces cerevisiae , Glucose
4.
Cells ; 12(3)2023 01 17.
Article in English | MEDLINE | ID: mdl-36766686

ABSTRACT

Escherichia coli is one of the most common members of the intestinal microbiota. Many of its strains are associated with various inflammatory infections, including urinary or gut infections, especially when displaying antibiotic resistance or in patients with suppressed immune systems. According to recent reports, the biofilm-forming potential of E. coli is a crucial factor for its increased resistance against antibiotics. To overcome the limitations of using antibiotics against resistant E. coli strains, the world is turning once more towards bacteriophage therapy, which is becoming a promising candidate amongst the current personalized approaches to target different bacterial infections. Although matured and persistent biofilms pose a serious challenge to phage therapy, they can still become an effective alternative to antibiotic treatment. Here, we assess the efficiency of clinically isolated phages in phage therapy against representative clinical uropathogenic and invasive biofilm-forming E. coli strains. Our results demonstrate that irrespective of host specificity, bacteriophages producing clear plaques with a high burst size, and exhibiting depolymerizing activity, are good candidates against biofilm-producing E. coli pathogens as verified from our in vitro and in vivo experiments using Galleria mellonella where survival was significantly increased for phage-therapy-treated larvae.


Subject(s)
Bacteriophages , Phage Therapy , Animals , Humans , Escherichia coli , Anti-Bacterial Agents , Biofilms
5.
Front Microbiol ; 13: 959156, 2022.
Article in English | MEDLINE | ID: mdl-36466653

ABSTRACT

Chronic wounds infected by Pseudomonas aeruginosa and Staphylococcus aureus are a relevant health problem worldwide because these pathogens grow embedded in a network of polysaccharides, proteins, lipids, and extracellular DNA, named biofilm, that hinders the transport of antibiotics and increases their antimicrobial tolerance. It is necessary to investigate therapies that improve the penetrability and efficacy of antibiotics. In this context, our main objectives were to study the relationship between P. aeruginosa and S. aureus and how their relationship can affect the antimicrobial treatment and investigate whether functionalized silver nanoparticles can improve the antibiotic therapy. We used an optimized in vitro wound model that mimics an in vivo wound to co-culture P. aeruginosa and S. aureus biofilm. The in vitro wound biofilm was treated with antimicrobial combinatory therapies composed of antibiotics (gentamycin and ciprofloxacin) and biofilm-dispersing free or silver nanoparticles functionalized with enzymes (α-amylase, cellulase, DNase I, or proteinase K) to study their antibiofilm efficacy. The interaction and colocalization of P. aeruginosa and S. aureus in a wound-like biofilm were examined and detailed characterized by confocal and electronic microscopy. We demonstrated that antibiotic monotherapy is inefficient as it differentially affects the two bacterial species in the mixed biofilm, driving P. aeruginosa to overcome S. aureus when using ciprofloxacin and the contrary when using gentamicin. In contrast, dual-antibiotic therapy efficiently reduces both species while maintaining a balanced population. In addition, DNase I nanoparticle treatment had a potent antibiofilm effect, decreasing P. aeruginosa and S. aureus viability to 0.017 and 7.7%, respectively, in combined antibiotics. The results showed that using nanoparticles functionalized with DNase I enhanced the antimicrobial treatment, decreasing the bacterial viability more than using the antibiotics alone. The enzymes α-amylase and cellulase showed some antibiofilm effect but were less effective compared to the DNase I treatment. Proteinase K showed insignificant antibiofilm effect. Finally, we proposed a three-dimensional colocalization model consisting of S. aureus aggregates within the biofilm structure, which could be associated with the low efficacy of antibiofilm treatments on bacteria. Thus, designing a clinical treatment that combines antibiofilm enzymes and antibiotics may be essential to eliminating chronic wound infections.

6.
NPJ Biofilms Microbiomes ; 8(1): 52, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35787627

ABSTRACT

The extracellular matrix protects biofilm cells by reducing diffusion of antimicrobials. Tobramycin is an antibiotic used extensively to treat P. aeruginosa biofilms, but it is sequestered in the biofilm periphery by the extracellular negative charge matrix and loses its efficacy significantly. Dispersal of the biofilm extracellular matrix with enzymes such as DNase I is another promising therapy that enhances antibiotic diffusion into the biofilm. Here, we combine the charge neutralization of tobramycin provided by dextran-based single-chain polymer nanoparticles (SCPNs) together with DNase I to break the biofilm matrix. Our study demonstrates that the SCPNs improve the activity of tobramycin and DNase I by neutralizing the ionic interactions that keep this antibiotic in the biofilm periphery. Moreover, the detailed effects and interactions of nanoformulations with extracellular matrix components were revealed through time-lapse imaging of the P. aeruginosa biofilms by laser scanning confocal microscopy with specific labeling of the different biofilm components.


Subject(s)
Nanoparticles , Tobramycin , Anti-Bacterial Agents/pharmacology , Biofilms , Deoxyribonuclease I , Dextrans , Pseudomonas aeruginosa , Tobramycin/pharmacology
7.
Nanoscale ; 13(44): 18754-18762, 2021 Nov 18.
Article in English | MEDLINE | ID: mdl-34747424

ABSTRACT

Shewanella oneidensis MR-1 is a metal-reducing bacterium that is able to exchange electrons with solid-phase minerals outside the cell. These bacterial cells can produce outer membrane extensions (OMEs) that are tens of nanometers wide and several microns long. The capability of these OMEs to transport electrons is currently under investigation. Tubular chemically fixed OMEs from S. oneidensis have shown good dc conducting properties when measured in an air environment. However, no direct demonstration of the conductivity of the more common bubble-like OMEs has been provided yet, due to the inherent difficulties in measuring it. In the present work, we measured the electrical properties of bubble-like OMEs in a dry air environment by Scanning Dielectric Microscopy (SDM) in force detection mode. We found that at the frequency of the measurements (∼2 kHz), OMEs show an insulating behavior, with an equivalent homogeneous dielectric constant εOME = 3.7 ± 0.7 and no dephasing between the applied ac voltage and the measured ac electric force. The dielectric constant measured for the OMEs is comparable to that obtained for insulating supramolecular protein structures (εprotein = 3-4), pointing towards a rich protein composition of the OMEs, probably coming from the periplasm. Based on the detection sensitivity of the measuring instrument, the upper limit for the ac longitudinal conductivity of bubble-like OMEs in a dry air environment has been set to σOME,ac < 10-5 S m-1, a value several orders of magnitude smaller than the dc conductivity measured in tubular chemically fixed OMEs. The lack of conductivity of bubble-like OMEs can be attributed to the relatively large separation between cytochromes in these larger OMEs and to the suppression of cytochrome mobility due to the dry environmental conditions.


Subject(s)
Shewanella , Electron Transport , Electrons , Periplasm/metabolism
8.
NPJ Biofilms Microbiomes ; 7(1): 62, 2021 08 03.
Article in English | MEDLINE | ID: mdl-34344902

ABSTRACT

Currently, three major circumstances threaten the management of bacterial infections: increasing antimicrobial resistance, expansion of chronic biofilm-associated infections, and lack of an appropriate approach to treat them. To date, the development of accelerated drug susceptibility testing of biofilms and of new antibiofouling systems has not been achieved despite the availability of different methodologies. There is a need for easy-to-use methods of testing the antibiotic susceptibility of bacteria that form biofilms and for screening new possible antibiofilm strategies. Herein, we present a microfluidic platform with an integrated interdigitated sensor (BiofilmChip). This new device allows an irreversible and homogeneous attachment of bacterial cells of clinical origin, even directly from clinical specimens, and the biofilms grown can be monitored by confocal microscopy or electrical impedance spectroscopy. The device proved to be suitable to study polymicrobial communities, as well as to measure the effect of antimicrobials on biofilms without introducing disturbances due to manipulation, thus better mimicking real-life clinical situations. Our results demonstrate that BiofilmChip is a straightforward tool for antimicrobial biofilm susceptibility testing that could be easily implemented in routine clinical laboratories.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteriological Techniques/methods , Biofilms/drug effects , Biofilms/growth & development , Anti-Infective Agents/pharmacology , Bacterial Infections/diagnosis , Bacterial Infections/drug therapy , Bacteriological Techniques/instrumentation , Delivery of Health Care , Humans , Laboratories, Clinical , Microbial Sensitivity Tests , Microfluidics , Mycobacterium tuberculosis , Pseudomonas aeruginosa , Staphylococcus aureus
9.
ACS Appl Mater Interfaces ; 13(13): 14964-14973, 2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33769023

ABSTRACT

The low efficacy of current conventional treatments for bacterial infections increases mortality rates worldwide. To alleviate this global health problem, we propose drug-free enzyme-based nanomotors for the treatment of bacterial urinary-tract infections. We develop nanomotors consisting of mesoporous silica nanoparticles (MSNPs) that were functionalized with either urease (U-MSNPs), lysozyme (L-MSNPs), or urease and lysozyme (M-MSNPs), and use them against nonpathogenic planktonic Escherichia coli. U-MSNPs exhibited the highest bactericidal activity due to biocatalysis of urea into NaHCO3 and NH3, which also propels U-MSNPs. In addition, U-MSNPs in concentrations above 200 µg/mL were capable of successfully reducing 60% of the biofilm biomass of a uropathogenic E. coli strain. This study thus provides a proof-of-concept, demonstrating that enzyme-based nanomotors are capable of fighting infectious diseases. This approach could potentially be extended to other kinds of diseases by selecting appropriate biomolecules.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Muramidase/pharmacology , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Urease/pharmacology , Anti-Bacterial Agents/administration & dosage , Biocatalysis , Biofilms/drug effects , Canavalia/enzymology , Drug Carriers/chemistry , Escherichia coli/physiology , Escherichia coli Infections/drug therapy , Humans , Muramidase/administration & dosage , Urease/administration & dosage , Urinary Tract Infections/drug therapy
10.
Sci Rep ; 10(1): 9390, 2020 06 10.
Article in English | MEDLINE | ID: mdl-32523130

ABSTRACT

Aggregates of Pseudomonas aeruginosa form a protective barrier against antibiotics and the immune system. These barriers, known as biofilms, are associated with several infectious diseases. One of the main components of these biofilms is alginate, a homo- and hetero-polysaccharide that consists of ß-D-mannuronate (M) and α-L-guluronate (G) units. Alginate lyases degrade this sugar and have been proposed as biotherapeutic agents to dissolve P. aeruginosa biofilms. However, there are contradictory reports in the literature regarding the efficacy of alginate lyases against biofilms and their synergistic effect with antibiotics. We found that most positive reports used a commercial crude extract from Flavobacterium multivorum as the alginate lyase source. By using anion exchange chromatography coupled to nano LC MS/MS, we identified two distinct enzymes in this extract, one has both polyM and polyG (polyM/G) degradation activities and it is similar in sequence to a broad-spectrum alginate lyase from Flavobacterium sp. S20 (Alg2A). The other enzyme has only polyG activity and it is similar in sequence to AlyA1 from Zobellia galactanivorans. By characterizing both of these enzymes together with three recombinant alginate lyases (a polyM, a polyG and a polyM/G), we showed that only enzymes with polyM/G activity such as Alg2A and A1-II' (alginate lyase from Sphingomonas sp.) are effective in dissolving biofilms. Furthermore, both activities are required to have a synergistic effect with antibiotics.


Subject(s)
Alginates/metabolism , Bacterial Proteins/therapeutic use , Biological Therapy/methods , Lyases/therapeutic use , Pseudomonas Infections/therapy , Pseudomonas aeruginosa/physiology , Sphingobacterium/metabolism , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/metabolism , Biofilms , Cloning, Molecular , Complex Mixtures , Drug Synergism , Humans , Lyases/metabolism , Substrate Specificity , Tandem Mass Spectrometry
11.
FASEB J ; 34(2): 2912-2928, 2020 02.
Article in English | MEDLINE | ID: mdl-31908067

ABSTRACT

Many notable human pathogens are facultative anaerobes. These pathogens exhibit redundant metabolic pathways and a whole array of regulatory systems to adapt to changing oxygen levels. However, our knowledge of facultative anaerobic pathogens is mostly based on fully aerobic or anaerobic cultures, which does not reflect real infection conditions, while the microaerobic range remains understudied. Here, we examine the behavior of pathogenic and nonpathogenic strains of two facultative anaerobes, Escherichia coli and Pseudomonas aeruginosa, during the aerobic-anaerobic transition. To do so, we introduce a new technique named AnaeroTrans, in which we allow self-consumption of oxygen by steady-state cultures and monitor the system by measuring the gas-phase oxygen concentration. We explore the different behavior of the studied species toward oxygen and examine how this behavior is associated with the targeted infection sites. As a model, we characterize the adaptation profile of the ribonucleotide reductase network, a complex oxygen-dependent enzymatic system responsible for the generation of the deoxyribonucleotides. We also explore the actions of the most important anaerobic regulators and how these regulators influence bacterial fitness. Our results allow us to classify the different elements that compose the aerobic-anaerobic transition into reproducible stages, thus showing the different adaptation mechanisms of the studied species.


Subject(s)
Adaptation, Physiological , Escherichia coli/growth & development , Oxygen/metabolism , Aerobiosis , Escherichia coli Proteins/metabolism , Pseudomonas aeruginosa
12.
Sci Rep ; 9(1): 16284, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31705015

ABSTRACT

The coexistence between species that occurs in some infections remains hard to achieve in vitro since bacterial fitness differences eventually lead to a single organism dominating the mixed culture. Pseudomonas aeruginosa and Staphylococcus aureus are major pathogens found growing together in biofilms in disease-affected lungs or wounds. Herein, we tested and analyzed different culture media, additives and environmental conditions to support P. aeruginosa and S. aureus coexistence in vitro. We have unraveled the potential of DMEM to support the growth of these two organisms in mature cocultured biofilms (three days old) in an environment that dampens the pH rise. Our conditions use equal initial inoculation ratios of both strains and allow the stable formation of separate S. aureus microcolonies that grow embedded in a P. aeruginosa biofilm, as well as S. aureus biofilm overgrowth when bovine serum albumin is added to the system. Remarkably, we also found that S. aureus survival is strictly dependent on a well-characterized phenomenon of oxygen stratification present in the coculture biofilm. An analysis of differential tolerance to gentamicin and ciprofloxacin treatment, depending on whether P. aeruginosa and S. aureus were growing in mono- or coculture biofilms, was used to validate our in vitro coculture conditions.


Subject(s)
Biofilms/growth & development , Coculture Techniques , Environment , Pseudomonas aeruginosa/physiology , Staphylococcus aureus/physiology , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Culture Media , Drug Resistance, Bacterial , Hydrogen-Ion Concentration , Microbial Sensitivity Tests , Oxygen/metabolism , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects
13.
ACS Infect Dis ; 5(9): 1581-1589, 2019 09 13.
Article in English | MEDLINE | ID: mdl-31268675

ABSTRACT

Oleanolic acid (OA) and maslinic acid (MA) are pentacyclic triterpenic compounds that abound in industrial olive oil waste. These compounds have renowned antimicrobial properties and lack cytotoxicity in eukaryotic cells as well as resistance mechanisms in bacteria. Despite these advantages, their antimicrobial activity has only been tested in vitro, and derivatives improving this activity have not been reported. In this work, a set of 14 OA and MA C-28 amide derivatives have been synthesized. Two of these derivatives, MA-HDA and OA-HDA, increase the in vitro antimicrobial activity of the parent compounds while reducing their toxicity in most of the Gram-positive bacteria tested, including a methicillin-resistant Staphylococcus aureus-MRSA. MA-HDA also shows an enhanced in vivo efficacy in a Galleria mellonella invertebrate animal model of infection. A preliminary attempt to elucidate their mechanism of action revealed that these compounds are able to penetrate and damage the bacterial cell membrane. More significantly, their capacity to reduce antibiofilm formation in catheters has also been demonstrated in two sets of conditions: a static and a more challenged continuous-flow S. aureus biofilm.


Subject(s)
Biofilms/drug effects , Gram-Positive Bacteria/physiology , Gram-Positive Bacterial Infections/drug therapy , Lepidoptera/microbiology , Pentacyclic Triterpenes/chemical synthesis , Animals , Bacterial Outer Membrane/drug effects , Cross Infection/drug therapy , Disease Models, Animal , Gram-Positive Bacteria/drug effects , Microbial Sensitivity Tests , Molecular Structure , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Pentacyclic Triterpenes/chemistry , Pentacyclic Triterpenes/pharmacology , Triterpenes/chemistry , Triterpenes/pharmacology
14.
J Med Chem ; 62(2): 512-530, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30507173

ABSTRACT

Six morpholine-(iso)thiosemicarbazone hybrids HL1-HL6 and their Cu(II) complexes with good-to-moderate solubility and stability in water were synthesized and characterized. Cu(II) complexes [Cu(L1-6)Cl] (1-6) formed weak dimeric associates in the solid state, which did not remain intact in solution as evidenced by ESI-MS. The lead proligands and Cu(II) complexes displayed higher antiproliferative activity in cancer cells than triapine. In addition, complexes 2-5 were found to specifically inhibit the growth of Gram-positive bacteria Staphylococcus aureus with MIC50 values at 2-5 µg/mL. Insights into the processes controlling intracellular accumulation and mechanism of action were investigated for 2 and 5, including the role of ribonucleotide reductase (RNR) inhibition, endoplasmic reticulum stress induction, and regulation of other cancer signaling pathways. Their ability to moderately inhibit R2 RNR protein in the presence of dithiothreitol is likely related to Fe chelating properties of the proligands liberated upon reduction.


Subject(s)
Antineoplastic Agents/chemical synthesis , Coordination Complexes/chemistry , Copper/chemistry , Morpholines/chemistry , Thiosemicarbazones/chemistry , Animals , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Crystallography, X-Ray , Humans , Mice , Molecular Conformation , Molecular Docking Simulation , Orphan Nuclear Receptors/antagonists & inhibitors , Orphan Nuclear Receptors/metabolism , Pseudomonas aeruginosa/drug effects , Solubility , Structure-Activity Relationship
15.
Molecules ; 23(11)2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30400165

ABSTRACT

Concerns have been raised about the long-term accumulating effects of triclocarban, a polychlorinated diarylurea widely used as an antibacterial soap additive, in the environment and in human beings. Indeed, the Food and Drug Administration has recently banned it from personal care products. Herein, we report the synthesis, antibacterial activity and cytotoxicity of novel N,N'-diarylureas as triclocarban analogs, designed by reducing one or more chlorine atoms of the former and/or replacing them by the novel pentafluorosulfanyl group, a new bioisostere of the trifluoromethyl group, with growing importance in drug discovery. Interestingly, some of these pentafluorosulfanyl-bearing ureas exhibited high potency, broad spectrum of antimicrobial activity against Gram-positive bacterial pathogens, and high selectivity index, while displaying a lower spontaneous mutation frequency than triclocarban. Some lines of evidence suggest a bactericidal mode of action for this family of compounds.


Subject(s)
Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Carbanilides/chemistry , Carbanilides/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Catheters/microbiology , Humans , Microbial Sensitivity Tests , Molecular Structure , Mutation Rate , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Structure-Activity Relationship
16.
Nanoscale ; 10(40): 19188-19194, 2018 Oct 18.
Article in English | MEDLINE | ID: mdl-30302472

ABSTRACT

The dielectric constant of flagellin proteins in flagellar bacterial filaments ∼10-20 nm in diameter is measured using scanning dielectric microscopy. We obtained for two different bacterial species (Shewanella oneidensis MR-1 and Pseudomonas aeruginosa PAO1) similar relative dielectric constant values εSo = 4.3 ± 0.6 and εPa = 4.5 ± 0.7, respectively, despite their different structure and amino acid sequence. The present results show the applicability of scanning dielectric microscopy to nanoscale filamentous protein complexes and to general 3D macromolecular protein geometries, thus opening new avenues to study the relationship between the dielectric response and protein structure and function.

17.
Front Microbiol ; 9: 986, 2018.
Article in English | MEDLINE | ID: mdl-29867886

ABSTRACT

Pseudomonas aeruginosa is a major pathogenic bacterium in chronic infections and is a model organism for studying biofilms. P. aeruginosa is considered an aerobic bacterium, but in the presence of nitrate, it also grows in anaerobic conditions. Oxygen diffusion through the biofilm generates metabolic and genetic diversity in P. aeruginosa growth, such as in ribonucleotide reductase activity. These essential enzymes are necessary for DNA synthesis and repair. Oxygen availability determines the activity of the three-ribonucleotide reductase (RNR) classes. Class II and III RNRs are active in the absence of oxygen; however, class II RNRs, which are important in P. aeruginosa biofilm growth, require a vitamin B12 cofactor for their enzymatic activity. In this work, we elucidated the conditions in which class II RNRs are active due to vitamin B12 concentration constraints (biosynthesis or environmental availability). We demonstrated that increased vitamin B12 levels during aerobic, stationary and biofilm growth activate class II RNR activity. We also established that the cobN gene is essentially responsible for B12 biosynthesis under planktonic and biofilm growth. Our results unravel the mechanisms of dNTP synthesis by P. aeruginosa during biofilm growth, which appear to depend on the bacterial strain (laboratory-type or clinical isolate).

18.
Sci Rep ; 6: 27232, 2016 06 06.
Article in English | MEDLINE | ID: mdl-27265565

ABSTRACT

The hydrophobic composition of mycobacterial cell walls leads to the formation of clumps when attempting to resuspend mycobacteria in aqueous solutions. Such aggregation may interfere in the mycobacteria-host cells interaction and, consequently, influence their antitumor effect. To improve the immunotherapeutic activity of Mycobacterium brumae, we designed different emulsions and demonstrated their efficacy. The best formulation was initially selected based on homogeneity and stability. Both olive oil (OO)- and mineral oil-in-water emulsions better preserved the mycobacteria viability and provided higher disaggregation rates compared to the others. But, among both emulsions, the OO emulsion increased the mycobacteria capacity to induce cytokines' production in bladder tumor cell cultures. The OO-mycobacteria emulsion properties: less hydrophobic, lower pH, more neutralized zeta potential, and increased affinity to fibronectin than non-emulsified mycobacteria, indicated favorable conditions for reaching the bladder epithelium in vivo. Finally, intravesical OO-M. brumae-treated mice showed a significantly higher systemic immune response, together with a trend toward increased tumor-bearing mouse survival rates compared to the rest of the treated mice. The physicochemical characteristics and the induction of a robust immune response in vitro and in vivo highlight the potential of the OO emulsion as a good delivery vehicle for the mycobacterial treatment of bladder cancer.


Subject(s)
Immunotherapy/methods , Mycobacterium/physiology , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/therapy , Animals , Bacterial Adhesion , Cell Line, Tumor , Cytokines , Emulsions , Fibronectins/metabolism , Humans , Mice , Microbial Viability , Mycobacterium/immunology , Olive Oil/chemistry , Olive Oil/pharmacology , Survival Rate , Treatment Outcome , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...