Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-38562889

RESUMO

Wound infections, exacerbated by the prevalence of antibiotic-resistant bacterial pathogens, necessitate innovative antimicrobial approaches. Polymicrobial infections, often involving Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA), present formidable challenges due to biofilm formation and antibiotic resistance. Hypochlorous acid (HOCl), a potent antimicrobial agent produced naturally by the immune system, holds promise as an alternative therapy. An electrochemical bandage (e-bandage) that generates HOCl in situ was evaluated for treatment of murine wound biofilm infections containing both MRSA and P. aeruginosa with "difficult-to-treat" resistance. Previously, the HOCl-producing e-bandage was shown to reduce wound biofilms containing P. aeruginosa alone. Compared to non-polarized e-bandage (no HOCl production) and Tegaderm only controls, the polarized e-bandages reduced bacterial loads in wounds infected with MRSA plus P. aeruginosa (MRSA: vs Tegaderm only - 1.4 log10 CFU/g, p = 0.0015, vs. non-polarized - 1.1 log10 CFU/g, p = 0.026. P. aeruginosa: vs Tegaderm only - 1.6 log10 CFU/g, p = 0.0015, vs non-polarized - 1.6 log10 CFU/g, p = 0.0032), and MRSA alone (vs Tegaderm only - 1.3 log10 CFU/g, p = 0.0048, vs. non-polarized - 1.1 log10 CFU/g, p = 0.0048), without compromising wound healing or causing tissue toxicity. Addition of systemic antibiotics did not enhance the antimicrobial efficacy of e-bandages, highlighting their potential as standalone therapies. This study provides additional evidence for the HOCl-producing e-bandage as a novel antimicrobial strategy for managing wound infections, including in the context of antibiotic resistance and polymicrobial infections.

2.
bioRxiv ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38586004

RESUMO

Chronic wound infections can be difficult to treat and may lead to impaired healing and worsened patient outcomes. Novel treatment strategies are needed. This study evaluated effects of intermittently produced H2O2 and HOCl, generated via an electrochemical bandage (e-bandage), against methicillin-resistant Staphylococcus aureus biofilms in an agar membrane biofilm model. By changing the working electrode potential, the e-bandage generated either HOCl (1.5 VAg/AgCl) or H2O2 (-0.6 VAg/AgCl). The degree of biocidal activity of intermittent treatment with HOCl and H2O2 correlated with HOCl treatment time; HOCl treatment durations of 0, 1.5, 3, 4.5, and 6 hours (with the rest of the 6 hour total treatment time devoted to H2O2 generation) resulted in mean biofilm reductions of 1.36±0.2, 2.22±0.16, 3.46±0.38, 4.63±0.74 and 7.66±0.5 log CFU/cm2, respectively vs. non-polarized controls, respectively. However, application of H2O2 immediately after HOCl treatment was detrimental to biofilm removal. For example, 3-hours HOCl treatment followed by 3-hours H2O2 resulted in a 1.90±0.84 log CFU/cm2 lower mean biofilm reduction than 3-hours HOCl treatment followed by 3-hours non-polarization. HOCl generated over 3-hours exhibited biocidal activity for at least 7.5-hours after e-bandage operation ceased; 3-hours of HOCl generation followed by 7.5-hours of non-polarization resulted in a biofilm cell reduction of 7.92±0.12 log CFU/cm2 vs. non polarized controls. Finally, intermittent treatment with HOCl (i.e., interspersed with periods of e-bandage non-polarization) for various intervals showed similar effects (approximately 6 log CFU/cm2 reduction vs. non-polarized control) to continuous treatment with HOCl for 3-hours, followed by 3-hours of non-polarization. These findings suggest that timing and sequencing of HOCl and H2O2 treatments are crucial for maximizing biofilm control.

3.
bioRxiv ; 2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38585771

RESUMO

Electrochemical bandages (e-bandages) can be applied to biofilm-infected wounds to generate reactive oxygen species, such as hypochlorous acid (HOCl) or hydrogen peroxide (H 2 O 2 ). The e-bandage-generated HOCl or H 2 O 2 kills biofilms in vitro and in infected wounds on mice. The HOCl-generating e-bandage is more active against biofilms in vitro , although this distinction is less apparent in vivo . The H 2 O 2 -generating e-bandage, more than the HOCl-generating e-bandage, is associated with improved healing of infected wounds. A strategy in which H 2 O 2 and HOCl are generated alternately-for dual action-was explored. The goal was to develop a programmable multimodal wearable potentiostat (PMWP) that could be programmed to generate HOCl or H 2 O 2 , as needed. An ultralow-power microcontroller unit managed operation of the PMWP. The system was operated with a 260-mAh capacity coin battery and weighed 4.6 grams, making it suitable for small animal experiments or human use. The overall cost of a single wearable potentiostat was $6.50 (USD). The device was verified using established electrochemical systems and functioned comparably to a commercial potentiostat. To determine antimicrobial effectiveness, PMWP-controlled e-bandages were tested against clinical isolates of four prevalent chronic wound bacterial pathogens, methicillin-resistant Staphylococcus aureus (MRSA), Pseudomonas aeruginosa, Acinetobacter baumannii , and Enterococcus faecium , and one fungal pathogen of emerging concern, Candida auris . PMWP-controlled e-bandages exhibited broad-spectrum activity against biofilms of all study isolates tested when programmed to deliver HOCl followed by H 2 O 2 . These results show that the PMWP operates effectively and is suitable for animal testing.

4.
Antimicrob Agents Chemother ; 68(2): e0121623, 2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38214514

RESUMO

The growing threat of antibiotic-resistant bacterial pathogens necessitates the development of alternative antimicrobial approaches. This is particularly true for chronic wound infections, which commonly harbor biofilm-dwelling bacteria. A novel electrochemical bandage (e-bandage) delivering low-levels of hypochlorous acid (HOCl) was evaluated against Pseudomonas aeruginosa murine wound biofilms. 5 mm skin wounds were created on the dorsum of mice and infected with 106 colony-forming units (CFU) of P. aeruginosa. Biofilms were formed over 2 days, after which e-bandages were placed on the wound beds and covered with Tegaderm. Mice were administered Tegaderm-only (control), non-polarized e-bandage (no HOCl production), or polarized e-bandage (using an HOCl-producing potentiostat), with or without systemic amikacin. Purulence and wound areas were measured before and after treatment. After 48 hours, wounds were harvested for bacterial quantification. Forty-eight hours of polarized e-bandage treatment resulted in mean biofilm reductions of 1.4 log10 CFUs/g (P = 0.0107) vs non-polarized controls and 2.2 log10 CFU/g (P = 0.004) vs Tegaderm-only controls. Amikacin improved CFU reduction in Tegaderm-only (P = 0.0045) and non-polarized control groups (P = 0.0312) but not in the polarized group (P = 0.3876). Compared to the Tegaderm-only group, there was less purulence in the polarized group (P = 0.009). Wound closure was neither impeded nor improved by either polarized or non-polarized e-bandage treatment. Concurrent amikacin did not impact wound closure or purulence. In conclusion, an HOCl-producing e-bandage reduced P. aeruginosa in wound biofilms with no impairment in wound healing, representing a promising antibiotic-free approach for addressing wound infection.


Assuntos
Infecções por Pseudomonas , Infecção dos Ferimentos , Animais , Camundongos , Pseudomonas aeruginosa , Ácido Hipocloroso , Amicacina , Infecções por Pseudomonas/microbiologia , Infecção dos Ferimentos/microbiologia , Bandagens , Antibacterianos , Biofilmes
5.
bioRxiv ; 2023 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-37790575

RESUMO

A novel electrochemical bandage (e-bandage) delivering low-level hypochlorous acid (HOCl) was evaluated against Pseudomonas aeruginosa murine wound biofilms. 5 mm skin wounds were created on the dorsum of Swiss-Webster mice and infected with 10 6 colony forming units (CFU) of P. aeruginosa . Biofilms were formed over two days, after which e-bandages were placed on the wound beds and covered with Tegaderm™. Mice were administered Tegaderm-only (control), non-polarized e-bandage (no HOCl production), or polarized e-bandage (using an HOCl-producing potentiostat), with or without concurrently administered systemic amikacin. Purulence and wound areas were measured before and after treatment. After 48 hours, animals were sacrificed, and wounds were harvested for bacterial quantification. Forty-eight hours of polarized e-bandage treatment resulted in mean biofilm reductions of 1.4 log 10 CFUs/g (9.0 vs 7.6 log 10 ; p = 0.0107) vs non-polarized controls, and 2.2 log 10 CFU/g (9.8 vs 7.6 log 10 ; p = 0.004) vs Tegaderm only controls. Systemic amikacin improved CFU reduction in Tegaderm-only (p = 0.0045) and non-polarized control groups (p = 0.0312), but not in the polarized group (p = 0.3876). Compared to the Tegaderm only group, there was more purulence reduction in the polarized group (p = 0.009), but not in the non-polarized group (p = 0.064). Wound closure was not impeded or improved by either polarized or non-polarized e-bandage treatment. Concurrent amikacin did not impact wound closure or purulence. In conclusion, an HOCl-producing e-bandage reduced P. aeruginosa in wound biofilms with no impairment in wound healing, representing a promising antibiotic-free approach for addressing wound infections.

6.
J Bacteriol ; 205(10): e0016623, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37756166

RESUMO

The ninth American Society for Microbiology Conference on Biofilms was convened in-person on 13-17 November 2022 in Charlotte, NC. As the first of these conferences since prior to the start of the COVID-19 pandemic, the energy among the participants of the conference was clear, and the meeting was a tremendous success. The mixture of >330 oral and poster presentations resoundingly embodied the vitality of biofilm research across a wide range of topics and multiple scientific disciplines. Special activities, including a pre-conference symposium for early career researchers, further enhanced the attendee experience. As a general theme, the conference was deliberately structured to provide high levels of participation and engagement among early career scientists.


Assuntos
Pandemias , Sociedades Científicas , Humanos , Estados Unidos , Biofilmes
7.
Adv Ther (Weinh) ; 6(5)2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37485434

RESUMO

Biofilms formed by antibiotic-resistant bacteria in wound beds present unique challenges in terms of treating wound infections. In this work, the in vivo activity of a novel electrochemical bandage (e-bandage) composed of carbon fabric and controlled by a wearable potentiostat, designed to continuously deliver low amounts of hydrogen peroxide (H2O2) was evaluated against methicillin-resistant Staphylococcus aureus (MRSA), multidrug-resistant Pseudomonas aeruginosa (MDR-PA) and mixed-species (MRSA and MDR-PA) wound infections. Wounds created on Swiss Webster mice were infected with the above-named bacteria and biofilms allowed to establish on wound beds for 3 days. e-Bandages, which electrochemically reduce dissolved oxygen to H2O2 when polarized at -0.6 VAg/AgCl, were placed atop the infected wound bed and polarized continuously for 48 hours. Polarized e-bandage treatment resulted in significant reductions (p <0.001) of both mono-species and mixed-species wound infections. After e-bandage treatment, electron microscopy showed degradation of bacterial cells, and histopathology showed no obvious alteration to the inflammatory host response. Blood biochemistries showed no abnormalities. Taken all together, results of this work suggest that the described H2O2-producing e-bandage can effectively reduce in vivo MRSA, MDR-PA and mixed-species wound biofilms, and should be further developed as a potential antibiotic-free strategy for treatment of wound infections.

8.
Nature ; 618(7964): 358-364, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37225987

RESUMO

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.


Assuntos
Doença Aguda , Doença Crônica , Genes Bacterianos , Oxigênio , Infecções por Pseudomonas , Pseudomonas aeruginosa , RNA Bacteriano , Animais , Humanos , Oxigênio/metabolismo , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/patogenicidade , Infecções por Pseudomonas/complicações , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , RNA Bacteriano/genética , RNA Bacteriano/metabolismo , Fibrose Cística/microbiologia , Ferimentos e Lesões/microbiologia , Ubiquinona/biossíntese , Anaerobiose , Genes Bacterianos/genética , Sepse/complicações , Sepse/microbiologia
9.
Arthrosc Sports Med Rehabil ; 4(5): e1617-e1621, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36312702

RESUMO

Purpose: To determine the effectiveness of pericapsular nerve group (PENG) block for patients surgically treated for femoroacetabular impingement syndrome (FAIS). Methods: Consecutive patients who underwent surgical treatment of FAIS either with or without preoperative PENG block by a single surgeon were retrospectively identified. Twenty-five patients who received PENG block were matched 1:1 by age, sex, body mass index, and procedure to 25 patients who received no block (NB). A retrospective review of the medical records of consecutive patients undergoing the PENG block was performed. Outcome measures of postanesthesia care unit visual analog scale initial (PACU VAS-initial), maximum (PACU VAS-max), discharge (PACU VAS-discharge), intraoperative fentanyl, pain medications in morphine equivalents (ME), and PACU to discharge times were recorded. Results: Twenty-five patients undergoing a PENG block and 25 patients who did not undergo a block (NB) were identified. No significant differences observed between age, sex, body mass index, surgery time, or procedures performed between the PENG and NB groups, P > .05. Significantly less VAS-initial was observed in the PENG group 3.7 ± 3.2, versus 5.5 ± 2.9 in the NB group, P = .04. Fentanyl usage intraoperatively was 137.3 ± 53.3 µg versus 108.5 ± 39.6 µg in NB versus PENG group respectively, P = .04. Narcotic use was 50.29 ± 11.2 ME versus 34.3 ± 12.1 ME in NB versus PENG group respectively, P = .001. PACU to discharge time was 95.8 ± 31 minutes versus 81.5 ± 19 minutes in NB versus PENG group, respectively, P = .05. No patient in the PENG group demonstrated a motor nerve palsy. Conclusions: For patients undergoing hip arthroscopy for FAIS, the addition of a preoperative PENG block showed a significant decrease in initial PACU pain, PACU narcotic consumption, intraoperative fentanyl usage, and quicker time to discharge without complications when compared to a no block, post-free control group. Level of Evidence: III, retrospective cohort study.

10.
Microbiol Spectr ; 10(5): e0192722, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-35993783

RESUMO

Hyperammonemia (HA) syndrome caused by respiratory infection with ammonia (NH3)-producing Ureaplasma species occurs in 4% of lung transplant recipients (LTRs) and is associated with high mortality. Although Ureaplasma-targeted antibiotic intervention is effective, the threat of antibiotic resistance development and pre-existing resistance make an alternative to antibiotics desirable. Considering that the underlying pathology of Ureaplasma-induced hyperammonemia (UIHA) is dependent upon ureaplasmal urease converting urea to NH3, urease inhibition could represent a targeted treatment approach. Here, the ability of the urease inhibitor, flurofamide, to prevent and treat UIHA was investigated. To confirm that flurofamide is broadly active against Ureaplasma respiratory isolates, the minimum urease inhibitory concentration against 4 isolates of Ureaplasma parvum and 5 isolates of Ureaplasma urealyticum was first determined in vitro. NH3 production by all isolates was inhibited by ≤2 µM flurofamide. To test the ability of flurofamide to prevent and treat UIHA, a mouse model of Ureaplasma respiratory infection was utilized. When animals were administered 6 mg/kg flurofamide via intraperitoneal injection 1 h prior to infection with U. parvum, flurofamide-administered animals exhibited significantly lower blood NH3 levels than did non-prophylaxed animals (10.9 ± 4.0 µmol/L compared to 26.5 ± 17.7 µmol/L; P = 0.0146) 24 h post-treatment. When U. parvum-infected hyperammonemic mice were treated with 6 mg/kg flurofamide, treated animals had significantly greater decreases in blood-NH3 levels 6 h post-treatment than did untreated mice (56.4 ± 17.1% compared to 9.1 ± 33.5% reduction; P = 0.0152). Together, these results indicate that flurofamide is a promising non-antibiotic treatment for UIHA in LTRs. IMPORTANCE Ureaplasma-associated hyperammonemia syndrome occurs in 4% of lung transplant recipients and has historically been almost universally fatal. While Ureaplasma-targeted antibiotics have been shown to be protective, the possibility of underlying resistance and resistance selection render non-antibiotic interventions an interesting approach.


Assuntos
Hiperamonemia , Infecções por Ureaplasma , Camundongos , Animais , Ureaplasma , Hiperamonemia/tratamento farmacológico , Hiperamonemia/prevenção & controle , Hiperamonemia/complicações , Urease/farmacologia , Amônia/farmacologia , Infecções por Ureaplasma/complicações , Infecções por Ureaplasma/tratamento farmacológico , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Ureia/farmacologia
11.
NPJ Biofilms Microbiomes ; 8(1): 49, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35705574

RESUMO

A new technique was used to measure the viscoelasticity of in vivo Pseudomonas aeruginosa biofilms. This was done through ex vivo microrheology measurements of in vivo biofilms excised from mouse wound beds. To our knowledge, this is the first time that the mechanics of in vivo biofilms have been measured. In vivo results are then compared to typical in vitro measurements. Biofilms grown in vivo are more relatively elastic than those grown in a wound-like medium in vitro but exhibited similar compliance. Using various genetically mutated P. aeruginosa strains, it is observed that the contributions of the exopolysaccharides Pel, Psl, and alginate to biofilm viscoelasticity were different for the biofilms grown in vitro and in vivo. In vitro experiments with collagen containing medium suggest this likely arises from the incorporation of host material, most notably collagen, into the matrix of the biofilm when it is grown in vivo. Taken together with earlier studies that examined the in vitro effects of collagen on mechanical properties, we conclude that collagen may, in some cases, be the dominant contributor to biofilm viscoelasticity in vivo.


Assuntos
Biofilmes , Pseudomonas aeruginosa , Animais , Colágeno/metabolismo , Colágeno/farmacologia , Camundongos , Polissacarídeos Bacterianos/metabolismo , Pseudomonas aeruginosa/fisiologia , Substâncias Viscoelásticas , Ferimentos e Lesões/microbiologia
12.
Front Cell Infect Microbiol ; 12: 835754, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35463635

RESUMO

Biofilms are the cause of most chronic bacterial infections. Living within the biofilm matrix, which is made of extracellular substances, including polysaccharides, proteins, eDNA, lipids and other molecules, provides microorganisms protection from antimicrobials and the host immune response. Exopolysaccharides are major structural components of bacterial biofilms and are thought to be vital to numerous aspects of biofilm formation and persistence, including adherence to surfaces, coherence with other biofilm-associated cells, mechanical stability, protection against desiccation, binding of enzymes, and nutrient acquisition and storage, as well as protection against antimicrobials, host immune cells and molecules, and environmental stressors. However, the contribution of specific exopolysaccharide types to the pathogenesis of biofilm infection is not well understood. In this study we examined whether the absence of the two main exopolysaccharides produced by the biofilm former Pseudomonas aeruginosa would affect wound infection in a mouse model. Using P. aeruginosa mutants that do not produce the exopolysaccharides Pel and/or Psl we observed that the severity of wound infections was not grossly affected; both the bacterial load in the wounds and the wound closure rates were unchanged. However, the size and spatial distribution of biofilm aggregates in the wound tissue were significantly different when Pel and Psl were not produced, and the ability of the mutants to survive antibiotic treatment was also impaired. Taken together, our data suggest that while the production of Pel and Psl do not appear to affect P. aeruginosa pathogenesis in mouse wound infections, they may have an important implication for bacterial persistence in vivo.


Assuntos
Infecções por Pseudomonas , Infecção dos Ferimentos , Animais , Proteínas de Bactérias/genética , Biofilmes , Camundongos , Polissacarídeos Bacterianos/metabolismo , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/genética
13.
Microbiol Spectr ; 10(1): e0194221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35171026

RESUMO

Lung transplant recipients (LTRs) are vulnerable to hyperammonemia syndrome (HS) in the early postoperative period, a condition typically unresponsive to nonantibiotic interventions. HS in LTRs is strongly correlated with Ureaplasma infection of the respiratory tract, although it is not well understood what makes LTRs preferentially susceptible to HS compared to other immunocompromised hosts. Ureaplasma species harbor highly active ureases, and postoperative LTRs commonly experience uremia. We hypothesized that uremia could be a potentiating comorbidity, providing increased substrate for ureaplasmal ureases. Using a novel dialyzed flow system, the ammonia-producing capacities of four isolates of Ureaplasma parvum and six isolates of Ureaplasma urealyticum in media formulations relating to normal and uremic host conditions were tested. For all isolates, growth under simulated uremic conditions resulted in increased ammonia production over 24 h, despite similar endpoint bacterial quantities. Further, transcripts of ureC (from the ureaplasmal urease gene cluster) from U. urealyticum IDRL-10763 and ATCC-27816 rose at similar rates under uremic and nonuremic conditions, with similar endpoint populations under the two conditions (despite markedly increased ammonia concentrations under uremic conditions), indicating that the difference in ammonia production by these isolates is due to increased urease activity, not expression. Lastly, uremic mice infected with an Escherichia coli strain harboring a U. urealyticum serovar 8 gene cluster exhibited higher blood ammonia levels compared to nonuremic mice infected with the same strain. Taken together, these data show that U. urealyticum and U. parvum produce more ammonia under uremic conditions compared to nonuremic conditions. This implies that uremia is a plausible contributing factor to Ureaplasma-induced HS in LTRs. IMPORTANCE Ureaplasma-induced hyperammonemia syndrome is a deadly complication affecting around 4% of lung transplant recipients and, to a lesser extent, other solid organ transplant patients. Understanding the underlying mechanisms will inform patient management, potentially decreasing mortality and morbidity. Here, it is shown that uremia is a plausible contributing factor to the pathophysiology of the condition.


Assuntos
Hiperamonemia/complicações , Hiperamonemia/microbiologia , Uremia/complicações , Uremia/microbiologia , Amônia/metabolismo , Animais , Humanos , Hospedeiro Imunocomprometido , Pulmão , Transplante de Pulmão , Camundongos , Transplantados , Ureaplasma , Ureaplasma urealyticum/isolamento & purificação , Sistema Urinário
14.
Soft Matter ; 17(25): 6225-6237, 2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34109345

RESUMO

Pseudomonas aeruginosa is an opportunistic pathogen that causes thousands of deaths every year in part due to its ability to form biofilms composed of bacteria embedded in a matrix of self-secreted extracellular polysaccharides (EPS), e-DNA, and proteins. In chronic wounds, biofilms are exposed to the host extracellular matrix, of which collagen is a major component. How bacterial EPS interacts with host collagen and whether this interaction affects biofilm viscoelasticity is not well understood. Since physical disruption of biofilms is often used in their removal, knowledge of collagen's effects on biofilm viscoelasticity may enable new treatment strategies that are better tuned to biofilms growing in host environments. In this work, biofilms are grown in the presence of different concentrations of collagen that mimic in vivo conditions. In order to explore collagen's interaction with EPS, nine strains of P. aeruginosa with different patterns of EPS production were used to grow biofilms. Particle tracking microrheology was used to characterize the mechanical development of biofilms over two days. Collagen is found to decrease biofilm compliance and increase relative elasticity regardless of the EPS present in the system. However, this effect is minimized when biofilms overproduce EPS. Collagen appears to become a de facto component of the EPS, through binding to bacteria or physical entanglement.


Assuntos
Biofilmes , Pseudomonas aeruginosa , Colágeno , Polissacarídeos Bacterianos , Viscosidade
15.
Int Wound J ; 18(5): 626-638, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33565263

RESUMO

Wound biofilms must be identified to target disruption and bacterial eradication but are challenging to detect with standard clinical assessment. This study tested whether bacterial fluorescence imaging could detect porphyrin-producing bacteria within a biofilm using well-established in vivo models. Mouse wounds were inoculated on Day 0 with planktonic bacteria (n = 39, porphyrin-producing and non-porphyrin-producing species, 107  colony forming units (CFU)/wound) or with polymicrobial biofilms (n = 16, 3 biofilms per mouse, each with 1:1:1 parts Staphylococcus aureus/Escherichia coli/Enterobacter cloacae, 107  CFU/biofilm) that were grown in vitro. Mouse wounds inoculated with biofilm underwent fluorescence imaging up to Day 4 or 5. Wounds were then excised and sent for microbiological analysis. Bacteria-matrix interaction was assessed with scanning electron microscopy (SEM) and histopathology. A total of 48 hours after inoculation with planktonic bacteria or biofilm, red fluorescence was readily detected in wounds; red fluorescence intensified up to Day 4. Red fluorescence from biofilms persisted in excised wound tissue post-wash. SEM and histopathology confirmed bacteria-matrix interaction. This pre-clinical study is the first to demonstrate the fluorescence detection of bacterial biofilm in vivo using a point-of-care wound imaging device. These findings have implications for clinicians targeting biofilm and may facilitate improved visualisation and removal of biofilms.


Assuntos
Infecção dos Ferimentos , Animais , Bactérias , Biofilmes , Camundongos , Imagem Óptica , Sistemas Automatizados de Assistência Junto ao Leito , Infecção dos Ferimentos/diagnóstico
16.
J Microbiol Methods ; 181: 106131, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33385441

RESUMO

Ureaplasma species, including Ureaplasma parvum and Ureaplasma urealyticum, are challenging to culture and maintain. Here, we describe a novel bioreactor for growing high-titer liquid Ureaplasma cultures in a stable manner.


Assuntos
Reatores Biológicos/microbiologia , Infecções por Ureaplasma/microbiologia , Ureaplasma urealyticum/crescimento & desenvolvimento , Ureaplasma/crescimento & desenvolvimento , Humanos
17.
Proc Biol Sci ; 287(1937): 20202272, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33081616

RESUMO

Opportunistic pathogens are associated with a number of chronic human infections, yet the evolution of virulence in these organisms during chronic infection remains poorly understood. Here, we tested the evolution of virulence in the human opportunistic pathogen Pseudomonas aeruginosa in a murine chronic wound model using a two-part serial passage and sepsis experiment, and found that virulence evolved in different directions in each line of evolution. We also assessed P. aeruginosa adaptation to a chronic wound after 42 days of evolution and found that morphological diversity in our evolved populations was limited compared with that previously described in cystic fibrosis (CF) infections. Using whole-genome sequencing, we found that genes previously implicated in P. aeruginosa pathogenesis (lasR, pilR, fleQ, rpoN and pvcA) contained mutations during the course of evolution in wounds, with selection occurring in parallel across all lines of evolution. Our findings highlight that: (i) P. aeruginosa heterogeneity may be less extensive in chronic wounds than in CF lungs; (ii) genes involved in P. aeruginosa pathogenesis acquire mutations during chronic wound infection; (iii) similar genetic adaptations are employed by P. aeruginosa across multiple infection environments; and (iv) current models of virulence may not adequately explain the diverging evolutionary trajectories observed in an opportunistic pathogen during chronic wound infection.


Assuntos
Pseudomonas aeruginosa , Infecção dos Ferimentos/microbiologia , Animais , Camundongos , Infecções por Pseudomonas , Virulência
18.
PLoS Pathog ; 16(6): e1008511, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32555671

RESUMO

The clinical importance of microbiomes to the chronicity of wounds is widely appreciated, yet little is understood about patient-specific processes shaping wound microbiome composition. Here, a two-cohort microbiome-genome wide association study is presented through which patient genomic loci associated with chronic wound microbiome diversity were identified. Further investigation revealed that alternative TLN2 and ZNF521 genotypes explained significant inter-patient variation in relative abundance of two key pathogens, Pseudomonas aeruginosa and Staphylococcus epidermidis. Wound diversity was lowest in Pseudomonas aeruginosa infected wounds, and decreasing wound diversity had a significant negative linear relationship with healing rate. In addition to microbiome characteristics, age, diabetic status, and genetic ancestry all significantly influenced healing. Using structural equation modeling to identify common variance among SNPs, six loci were sufficient to explain 53% of variation in wound microbiome diversity, which was a 10% increase over traditional multiple regression. Focusing on TLN2, genotype at rs8031916 explained expression differences of alternative transcripts that differ in inclusion of important focal adhesion binding domains. Such differences are hypothesized to relate to wound microbiomes and healing through effects on bacterial exploitation of focal adhesions and/or cellular migration. Related, other associated loci were functionally enriched, often with roles in cytoskeletal dynamics. This study, being the first to identify patient genetic determinants for wound microbiomes and healing, implicates genetic variation determining cellular adhesion phenotypes as important drivers of infection type. The identification of predictive biomarkers for chronic wound microbiomes may serve as risk factors and guide treatment by informing patient-specific tendencies of infection.


Assuntos
Microbiota , Polimorfismo de Nucleotídeo Único , Infecções por Pseudomonas , Pseudomonas aeruginosa , Infecções Estafilocócicas , Staphylococcus epidermidis , Cicatrização/genética , Infecção dos Ferimentos , Animais , Doença Crônica , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Estudo de Associação Genômica Ampla , Humanos , Masculino , Camundongos , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/metabolismo , Infecções por Pseudomonas/microbiologia , Infecções por Pseudomonas/patologia , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/patologia , Talina/genética , Talina/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Infecção dos Ferimentos/genética , Infecção dos Ferimentos/metabolismo , Infecção dos Ferimentos/microbiologia , Infecção dos Ferimentos/patologia
19.
Infect Immun ; 88(6)2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32284368

RESUMO

The opportunistic pathogen Pseudomonas aeruginosa is a leading cause of morbidity and mortality worldwide. To survive in both the environment and the host, P. aeruginosa must cope with redox stress. In P. aeruginosa, a primary mechanism for protection from redox stress is the antioxidant glutathione (GSH). GSH is a low-molecular-weight thiol-containing tripeptide (l-γ-glutamyl-l-cysteinyl-glycine) that can function as a reversible reducing agent. GSH plays an important role in P. aeruginosa physiology and is known to modulate several cellular and social processes that are likely important during infection. However, the role of GSH biosynthesis during mammalian infection is not well understood. In this study, we created a P. aeruginosa mutant defective in GSH biosynthesis to examine how loss of GSH biosynthesis affects P. aeruginosa virulence. We found that GSH is critical for normal growth in vitro and provides protection against hydrogen peroxide, bleach, and ciprofloxacin. We also studied the role of P. aeruginosa GSH biosynthesis in four mouse infection models, including the surgical wound, abscess, burn wound, and acute pneumonia models. We discovered that the GSH biosynthesis mutant was slightly less virulent in the acute pneumonia infection model but was equally virulent in the three other models. This work provides new and complementary data regarding the role of GSH in P. aeruginosa during mammalian infection.


Assuntos
Glutationa/biossíntese , Pneumonia Bacteriana/microbiologia , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa/metabolismo , Infecções dos Tecidos Moles/microbiologia , Antibacterianos/farmacologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Desinfetantes/farmacologia , Farmacorresistência Bacteriana , Interações Hospedeiro-Patógeno , Humanos , Viabilidade Microbiana , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/genética , Pseudomonas aeruginosa/crescimento & desenvolvimento
20.
Langmuir ; 36(6): 1585-1595, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-31990563

RESUMO

Biofilms are communities of bacteria embedded in a polymeric matrix which are found in infections and in environments outside the body. Breaking down the matrix renders biofilms more susceptible to physical disruption and to treatments such as antibiotics. Different species of bacteria, and different strains within the same species, produce different types of matrix polymers. This suggests that targeting specific polymers for disruption may be more effective than nonspecific approaches to disrupting biofilm matrixes. In this study, we treated Pseudomonas aeruginosa biofilms with enzymes that are specific to different matrix polymers. We measured the resulting alteration in biofilm mechanics using bulk rheology and changes in structure using electron microscopy. We find that, for biofilms grown in vitro, the effect of enzymatic treatment is greatest when the enzyme is specific to a dominant matrix polymer. Specifically matched enzymatic treatment tends to reduce yield strain and yield stress and increase the rate of biofilm drying, due to increased diffusivity as a result of network compromise. Electron micrographs qualitatively suggest that well-matched enzymatic treatments reduce long-range structure and shorten connecting network fibers. Previous work has shown that generic glycoside hydrolases can cause dispersal of bacteria from in vivo and ex vivo biofilms into a free-swimming state, and thereby make antibiotic treatment more effective. For biofilms grown in wounded mice, we find that well-matched treatments that result in the greatest mechanical compromise in vitro induce the least dispersal ex vivo. Moreover, we find that generic glycoside hydrolases have no measurable effect on the mechanics of biofilms grown in vitro, while previous work has shown them to be highly effective at inducing dispersal in vivo and ex vivo. This highlights the possibility that effective approaches to eradicating biofilms may depend strongly on the growth environment.


Assuntos
Polímeros , Pseudomonas aeruginosa , Animais , Antibacterianos/farmacologia , Biofilmes , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...