Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.012
Filter
1.
Int J Biol Macromol ; 277(Pt 3): 134484, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39102904

ABSTRACT

Given the escalating prevalence of drug-resistant wounds, there is a justified imperative to explore innovative and more efficacious therapies that diverge from conventional, ineffective wound healing approaches. This research has introduced a strategy to address multi-drug resistant (MDR) Pseudomonas aeruginosa infections in a chronic wound model, employing MDR-specific phage Pɸ-Mi-Pa loaded onto mucoadhesive electrospun scaffolds. A cocktail of three isolates of P. aeruginosa-specific lytic phages, Pɸ-Mi-Pa 51, Pɸ-Mi-Pa 120, and Pɸ-Mi-Pa 133 were incorporated into varying ratios of fabricated PCL-PVP polymer. These formulations were assessed for their therapeutic efficacy in achieving bacterial clearance in P. aeruginosa-induced wound infections. The study encompassed biological characterization through in vivo wound healing assessments, histology, and histomorphometry. Additionally, morphological, mechanical, and chemical analyses were conducted on the fabricated PCL-PVP electrospun nanofibrous scaffolds. Three clonal differences of the MDR P. aeruginosa-specific phages (Pɸ-Mi-Pa 51, Pɸ-Mi-Pa 120, and Pɸ-Mi-Pa 133) produced lytic activity and were seen to produce distinct and clear zones of inhibition against MDR P. aeruginosa strains Pa 051, Pa 120 and Pa 133 respectively. The average porosity of the nanofibrous scaffolds PB 1, PB 2, PB 3, and PB 4 were 12.2 ± 0.3 %, 22.1 ± 0.7 %, 31.1 ± 2.4 %, 28.0 ± 0.8 % respectively. In vitro cumulative release of MDR-specific phage Pɸ-Mi-Pa from the mucoadhesive electrospun nanofibrous scaffolds was found to be 70.91 % ± 1.02 % after 12 h of incubation after an initial release of 42.8 % ± 3.01 % after 1 h. Results from the in vivo wound healing study revealed a substantial reduction in wound size, with formulations PB 2 and PB 3 exhibiting the most significant reduction in wound size, demonstrating statistically significant results on day 5 (100 % ± 31.4 %). These findings underscore the potential of bacteriophage-loaded electrospun PCL-PVP nanofibrous scaffolds for treating drug-resistant wounds, generating tissue substitutes, and overcoming certain limitations associated with conventional wound care matrices.


Subject(s)
Disease Models, Animal , Drug Resistance, Multiple, Bacterial , Nanofibers , Pseudomonas Infections , Pseudomonas aeruginosa , Wound Infection , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/virology , Animals , Nanofibers/chemistry , Drug Resistance, Multiple, Bacterial/drug effects , Pseudomonas Infections/drug therapy , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Wound Infection/microbiology , Wound Infection/drug therapy , Wound Infection/therapy , Wound Healing/drug effects , Tissue Scaffolds/chemistry , Rats , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Bacteriophages
2.
PLoS One ; 19(7): e0307079, 2024.
Article in English | MEDLINE | ID: mdl-39012882

ABSTRACT

BACKGROUND: Most of the current bacteriophages (phages) are mostly isolated from environments. However, phages isolated from feces might be more specific to the bacteria that are harmful to the host. Meanwhile, some phages from the environment might affect non-pathogenic bacteria for the host. METHODS: Here, bacteriophages isolated from mouse feces were intratracheally (IT) or intravenously (IV) administered in pneumonia mice caused by Pseudomonas aeruginosa at 2 hours post-intratracheal bacterial administration. As such, the mice with phage treatment, using either IT or IV administration, demonstrated less severe pneumonia as indicated by mortality, serum cytokines, bacteremia, bacterial abundance in bronchoalveolar lavage fluid (BALF), and neutrophil extracellular traps (NETs) in lung tissue (immunofluorescence of neutrophil elastase and myeloperoxidase). RESULTS: Interestingly, the abundance of phages in BALF from the IT and IV injections was similar, supporting a flexible route of phage administration. With the incubation of bacteria with neutrophils, the presence of bacteriophages significantly improved bactericidal activity, but not NETs formation, with the elevated supernatant IL-6 and TNF-α, but not IL-1ß. In conclusion, our findings suggest that bacteriophages against Pseudomonas aeruginosa can be discovered from feces of the host. CONCLUSIONS: The phages attenuate pneumonia partly through an enhanced neutrophil bactericidal activity, but not via inducing NETs formation. The isolation of phages from the infected hosts themselves might be practically useful for future treatment. More studies are warranted.


Subject(s)
Feces , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Pseudomonas aeruginosa/virology , Feces/microbiology , Feces/virology , Mice , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Bronchoalveolar Lavage Fluid/microbiology , Bronchoalveolar Lavage Fluid/virology , Neutrophils/immunology , Bacteriophages/isolation & purification , Bacteriophages/physiology , Extracellular Traps , Pneumonia/microbiology , Pneumonia/therapy , Pneumonia/virology , Cytokines/metabolism , Cytokines/blood , Phage Therapy/methods , Female , Lung/microbiology , Lung/virology , Pneumonia, Bacterial/therapy , Pneumonia, Bacterial/microbiology
3.
Viruses ; 16(7)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-39066163

ABSTRACT

The Gram-negative ESKAPE bacterium Pseudomonas aeruginosa has become a pathogen of serious concern due its extensive multi-drug resistance (MDR) profile, widespread incidences of hospital-acquired infections throughout the United States, and high occurrence in wound infections suffered by warfighters serving abroad. Bacteriophage (phage) therapy has received renewed attention as an alternative therapeutic option against recalcitrant bacterial infections, both as multi-phage cocktails and in combination with antibiotics as synergistic pairings. Environmental screening and phage enrichment has yielded three lytic viruses capable of infecting the MDR P. aeruginosa strain PAO1. Co-administration of each phage with the carbapenem antibiotics ertapenem, imipenem, and meropenem generated enhanced overall killing of bacteria beyond either phage or drug treatments alone. A combination cocktail of all three phages was completely inhibitory to growth, even without antibiotics. The same 3× phage cocktail also disrupted PAO1 biofilms, reducing biomass by over 75% compared to untreated biofilms. Further, the phage cocktail demonstrated broad efficacy as well, capable of infecting 33 out of 100 diverse clinical isolate strains of P. aeruginosa. Together, these results indicate a promising approach for designing layered medical countermeasures to potentiate antibiotic activity and possibly overcome resistance against recalcitrant, MDR bacteria such as P. aeruginosa. Combination therapy, either by synergistic phage-antibiotic pairings, or by phage cocktails, presents a means of controlling mutations that can allow for bacteria to gain a competitive edge.


Subject(s)
Anti-Bacterial Agents , Carbapenems , Drug Resistance, Multiple, Bacterial , Phage Therapy , Pseudomonas Infections , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/drug effects , Carbapenems/pharmacology , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Biofilms/drug effects , Bacteriophages/physiology , Microbial Sensitivity Tests , Humans , Pseudomonas Phages/physiology , Imipenem/pharmacology
4.
Viruses ; 16(7)2024 Jun 29.
Article in English | MEDLINE | ID: mdl-39066214

ABSTRACT

Antimicrobial resistance poses a serious risk to contemporary healthcare since it reduces the number of bacterial illnesses that may be treated with antibiotics, particularly for patients with long-term conditions like cystic fibrosis (CF). People with a genetic predisposition to CF often have recurrent bacterial infections in their lungs due to a buildup of sticky mucus, necessitating long-term antibiotic treatment. Pseudomonas aeruginosa infections are a major cause of CF lung illness, and P. aeruginosa airway isolates are frequently resistant to many antibiotics. Bacteriophages (also known as phages), viruses that infect bacteria, are a viable substitute for antimicrobials to treat P. aeruginosa infections in individuals with CF. Here, we reviewed the utilization of P. aeruginosa bacteriophages both in vivo and in vitro, as well as in the treatment of illnesses and diseases, and the outcomes of the latter.


Subject(s)
Cystic Fibrosis , Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Humans , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Cystic Fibrosis/microbiology , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Animals , Bacteriophages/physiology , Bacteriophages/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
5.
Viruses ; 16(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39066242

ABSTRACT

Pseudomonas aeruginosa is one of the main causes of healthcare-associated infection in Europe that increases patient morbidity and mortality. Multi-resistant pathogens are a major public health issue in burn centers. Mortality increases when the initial antibiotic treatment is inappropriate, especially if the patient is infected with P. aeruginosa strains that are resistant to many antibiotics. Phage therapy is an emerging option to treat severe P. aeruginosa infections. It involves using natural viruses called bacteriophages, which have the ability to infect, replicate, and, theoretically, destroy the P. aeruginosa population in an infected patient. We report here the case of a severely burned patient who experienced relapsing ventilator-associated pneumonia associated with skin graft infection and bacteremia due to extensively drug-resistant P. aeruginosa. The patient was successfully treated with personalized nebulized and intravenous phage therapy in combination with immunostimulation (interferon-γ) and last-resort antimicrobial therapy (imipenem-relebactam).


Subject(s)
Bacteremia , Burns , Drug Resistance, Multiple, Bacterial , Phage Therapy , Pneumonia, Ventilator-Associated , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/drug effects , Pneumonia, Ventilator-Associated/therapy , Pneumonia, Ventilator-Associated/drug therapy , Pneumonia, Ventilator-Associated/microbiology , Phage Therapy/methods , Pseudomonas Infections/therapy , Pseudomonas Infections/drug therapy , Burns/complications , Burns/therapy , Bacteremia/therapy , Bacteremia/drug therapy , Bacteremia/microbiology , Anti-Bacterial Agents/therapeutic use , Male , Recurrence , Bacteriophages/physiology
6.
Virus Res ; 348: 199442, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39074617

ABSTRACT

INTRODUCTION: Pseudomonas aeruginosa is an organism well known for causing significant morbidity and mortality in people living with chronic lung conditions such as cystic fibrosis. We describe the safety, tolerability, and potential efficacy of bronchoscopic and nebulised bacteriophage administration, offering insights into a potential breakthrough for the treatment of chronic infections particularly in children and adolescents. METHOD: A 12-year-old female (F12) and a 17-year-old male (M17), both diagnosed with cystic fibrosis and chronic P. aeruginosa lung infection, underwent bacteriophage treatment (BT). The administration involved bronchoscopic instillation and subsequent nebulisation. This was performed concurrently with intravenous antibiotics and regular physiotherapy delivered in an in-patient setting for 14 days. Microbiological, clinical, and lung function assessments were conducted to assess this treatment modality. RESULTS: No adverse events (fever, localised reaction, wheeze or bronchospasm) occurred during BT. F12 demonstrated a 4% increase, while M17 showed a 5% improvement in FEV1% from their best FEV1% over the past three years following BT. A 12% (F12) and an 8% (M17) improvement from baseline FEV1% was observed. For F12 P. aeruginosa was not isolated from her sputum despite 12 previous hospitalisations for intravenous antibiotics. CONCLUSION: Bronchoscopic and nebulised routes of bacteriophage administration were well-tolerated in these two adolescents. This early report underscores the potential of this treatment modality and encourages clinicians and researchers to actively explore this innovative approach.


Subject(s)
Bronchoscopy , Cystic Fibrosis , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Female , Adolescent , Pseudomonas Infections/therapy , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/drug effects , Male , Child , Cystic Fibrosis/therapy , Nebulizers and Vaporizers , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Phage Therapy/methods , Treatment Outcome , Bacteriophages , Administration, Inhalation
7.
J Burn Care Res ; 45(5): 1336-1340, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-38953562

ABSTRACT

Here, we report about a patient with a full-thickness burn injury of the left lower extremity with approximately 8% of total BSA affected. Initial therapy consisted of necrosectomy and wound coverage with split-thickness graft. The patient developed a wound infection with Pseudomonas aeruginosa, resulting in the failure of the skin graft to achieve complete healing. The case was further complicated by the patient's concurrent presentation of anemia, characterized by a hematocrit level of 19.8% on 11th day after admission. Additionally, the patient refused acceptance of any blood transfusion, adding a significant layer of complexity to the management strategy. In summary, the patient's critical state required an immediate intervention. Due to the contraindication for a further surgical debridement and autograft, we changed the treatment strategy to a conservative approach. First, the wound was debrided employing maggot therapy 17 days after admission. Subsequently, free soft tissue coverage was accomplished using decellularized fish skin dressings on 45th day. This approach yielded satisfactory wound closure. Following an approximately 2-month hospitalization period (52nd day after admission), the patient was discharged with a stable wound condition, nearing complete healing.


Subject(s)
Burns , Debridement , Skin Transplantation , Wound Infection , Humans , Burns/therapy , Burns/surgery , Burns/complications , Animals , Male , Wound Infection/therapy , Wound Infection/microbiology , Wound Healing , Pseudomonas Infections/therapy , Larva , Pseudomonas aeruginosa , Combined Modality Therapy , Fishes , Adult
8.
mSphere ; 9(7): e0070723, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38934592

ABSTRACT

Phage therapy is increasing in relevance as an alternative treatment to combat antibiotic resistant bacteria. Phage cocktails are the state-of-the-art method of administering phages in clinical settings, preferred over monophage treatment because of their ability to eliminate multiple bacterial strains and reduce resistance formation. In our study, we compare monophage applications and phage cocktails to our chosen method of phage sequential treatments. To do so, we isolated four novel bacteriophages capable of infecting Pseudomonas alcaligenes T3, a close relative of P. aeruginosa, and characterized them using sequencing and transmission electron microscopy. While investigating monophage treatments, we observed that different phage concentrations had a strong impact on the timing and amount of resistance formation. When using phage cocktails, we observed that P. alcaligenes were capable of forming resistance in the same timespan it took them to become resistant to single phages. We isolated mutants resistant to each single phage as well as mutants exposed to phage cocktails, resulting in bacteria resistant to all four phages at once. Sequencing these mutants showed that different treatments yielded unique single nucleotide polymorphism mutation patterns. In order to combat resistance formation, we added phages one by one in intervals of 24 h, thus managing to delay resistance development and keeping bacterial growth significantly lower compared to phage cocktails.IMPORTANCEWHO declared antimicrobial resistance a top threat to global health; while antibiotics have stood at the forefront in the fight against bacterial infection, the increasing number of multidrug-resistant bacteria highlights a need to branch out in order to address the threat of antimicrobial resistance. Bacteriophages, viruses solely infecting bacteria, could present a solution due to their abundance, versatility, and adaptability. For this study, we isolated new phages infecting a fast-mutating Pseudomonas alcaligenes strain capable of forming resistance within 30 h. By using a sequential treatment approach of adding one phage after another, we were able to curb bacterial growth significantly more compared to state-of-the-art phage cocktails.


Subject(s)
Phage Therapy , Pseudomonas Phages , Pseudomonas , Phage Therapy/methods , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas/virology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/virology , Mutation , Anti-Bacterial Agents/pharmacology , Bacteriophages/genetics , Bacteriophages/physiology , Bacteriophages/classification , Drug Resistance, Bacterial , Drug Resistance, Multiple, Bacterial
9.
Microbiol Spectr ; 12(8): e0352023, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38912817

ABSTRACT

Pseudomonas aeruginosa infections are getting increasingly serious as antimicrobial resistance spreads. Phage therapy may be a solution to the problem, especially if improved by current advances on phage-host studies. As a mucosal pathogen, we hypothesize that P. aeruginosa and its phages are linked to the bacteriophage adherence to mucus (BAM) model. This means that phage-host interactions could be influenced by mucin presence, impacting the success of phage infections on the P. aeruginosa host and consequently leading to the protection of the metazoan host. By using a group of four different phages, we tested three important phenotypes associated with the BAM model: phage binding to mucin, phage growth in mucin-exposed hosts, and the influence of mucin on CRISPR immunity of the bacterium. Three of the tested phages significantly bound to mucin, while two had improved growth rates in mucin-exposed hosts. Improved phage growth was likely the result of phage exploitation of mucin-induced physiological changes in the host. We could not detect CRISPR activity in our system but identified two putative anti-CRISPR proteins coded by the phage. Overall, the differential responses seen for the phages tested show that the same bacterial species can be targeted by mucosal-associated phages or by phages not affected by mucus presence. In conclusion, the BAM model is relevant for phage-bacterium interactions in P. aeruginosa, opening new possibilities to improve phage therapy against this important pathogen by considering mucosal interaction dynamics.IMPORTANCESome bacteriophages are involved in a symbiotic relationship with animals, in which phages held in mucosal surfaces protect them from invading bacteria. Pseudomonas aeruginosa is one of the many bacterial pathogens threatening humankind during the current antimicrobial resistance crisis. Here, we have tested whether P. aeruginosa and its phages are affected by mucosal conditions. We discovered by using a collection of four phages that, indeed, mucosal interaction dynamics can be seen in this model. Three of the tested phages significantly bound to mucin, while two had improved growth rates in mucin-exposed hosts. These results link P. aeruginosa and its phages to the bacteriophage adherence to the mucus model and open opportunities to explore this to improve phage therapy, be it by exploiting the phenotypes detected or by actively selecting mucosal-adapted phages for treatment.


Subject(s)
Mucins , Mucus , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/physiology , Mucus/microbiology , Mucus/virology , Pseudomonas Phages/physiology , Pseudomonas Phages/genetics , Mucins/metabolism , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Humans , Phage Therapy , Animals , Virus Attachment , Clustered Regularly Interspaced Short Palindromic Repeats
10.
Nat Microbiol ; 9(7): 1828-1841, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38886583

ABSTRACT

Bacteriophages have evolved diverse strategies to overcome host defence mechanisms and to redirect host metabolism to ensure successful propagation. Here we identify a phage protein named Dap1 from Pseudomonas aeruginosa phage PaoP5 that both modulates bacterial host behaviour and contributes to phage fitness. We show that expression of Dap1 in P. aeruginosa reduces bacterial motility and promotes biofilm formation through interference with DipA, a c-di-GMP phosphodiesterase, which causes an increase in c-di-GMP levels that trigger phenotypic changes. Results also show that deletion of dap1 in PaoP5 significantly reduces genome packaging. In this case, Dap1 directly binds to phage HNH endonuclease, prohibiting host Lon-mediated HNH degradation and promoting phage genome packaging. Moreover, PaoP5Δdap1 fails to rescue P. aeruginosa-infected mice, implying the significance of dap1 in phage therapy. Overall, these results highlight remarkable dual functionality in a phage protein, enabling the modulation of host behaviours and ensuring phage fitness.


Subject(s)
Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Pseudomonas aeruginosa , Viral Proteins , Pseudomonas aeruginosa/virology , Pseudomonas aeruginosa/pathogenicity , Pseudomonas aeruginosa/genetics , Animals , Mice , Pseudomonas Phages/genetics , Pseudomonas Phages/physiology , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/immunology , Virulence , Viral Proteins/genetics , Viral Proteins/metabolism , Biofilms/growth & development , Cyclic GMP/metabolism , Cyclic GMP/analogs & derivatives , Female , Bacteriophages/physiology , Bacteriophages/genetics
11.
Int J Biol Macromol ; 271(Pt 2): 132443, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761913

ABSTRACT

Metal-organic frameworks (MOFs)-based therapy opens a new area for antibiotic-drug free infections treatment. In the present study, chitosan membranes (CS) loaded with two concentrations of copper-MOF 10 mg/20 ml (Cu-MOF10/CS) & 20 mg/20 ml (Cu-MOF20/CS) were prepared by a simple lyophilization procedure. FTIR spectra of Cu-MOF10/CS and Cu-MOF20/CS dressings confirmed absence of any undesirable chemical changes after loading Cu-MOF. The SEM images of the synthesized materials (CS, Cu-MOF10/CS & Cu-MOF20/CS) showed interconnected porous structures. Cytocompatibility of the materials was confirmed by fibroblasts cells culturing and the materials were hemocompatible, with blood clotting index <5 %. Cu-MOF20/CS showed comparatively higher effective antibacterial activity against the tested strains; E. coli (149.2 %), P. aeruginosa (165 %) S. aureus (117.8 %) and MRSA (142 %) as compared to Amikacin, CS and Cu-MOF10/CS membranes. Similarly, Cu-MOF20/CS dressing significantly eradicated the biofilms; P. aeruginosa (37 %) and MRSA (52 %) respectively. In full thickness infected wound rat model, on day 23, Cu-MOF10/CS and Cu-MOF20/CS promoted wound healing up to 87.7 % and 82 % respectively. H&E staining of wounded tissues treated with Cu-MOF10/CS & Cu-MOF20/CS demonstrated enhanced neovascularization and re-epithelization along-with reduced inflammation, while trichrome staining exhibited increased collagen deposition. Overall, this study declares Cu-MOFs loaded chitosan dressings a multifunctional platform for the healing of infected wounds.


Subject(s)
Anti-Bacterial Agents , Bandages , Biofilms , Chitosan , Copper , Freeze Drying , Metal-Organic Frameworks , Pseudomonas aeruginosa , Wound Healing , Animals , Chitosan/chemistry , Chitosan/pharmacology , Wound Healing/drug effects , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/pharmacology , Rats , Pseudomonas aeruginosa/drug effects , Porosity , Copper/chemistry , Copper/pharmacology , Biofilms/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Pseudomonas Infections/drug therapy , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Male , Angiogenesis Inducing Agents/pharmacology , Angiogenesis Inducing Agents/chemistry , Wound Infection/drug therapy , Wound Infection/microbiology
12.
Vet Q ; 44(1): 1-9, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38726795

ABSTRACT

Antibiotic-resistant pathogens are a growing global issue, leading to untreatable infectious diseases in both humans and animals. Personalized bacteriophage (phage) therapy, the use of specific anti-bacterial viruses, is currently a leading approach to combat antibiotic-resistant infections. The implementation of phage therapy has primarily been focused on humans, almost neglecting the impact of such infections on the health and welfare of companion animals. Pets also have the potential to spread resistant infections to their owners or the veterinary staff through zoonotic transmission. Here, we showcase personalized phage-antibiotic treatment of a cat with a multidrug-resistant Pseudomonas aeruginosa implant-associated infection post-arthrodesis surgery. The treatment encompassed a tailored combination of an anti-P. aeruginosa phage and ceftazidime, precisely matched to the pathogen. The phage was topically applied to the surgical wound while the antibiotic was administered intramuscularly. After two treatment courses spanning 7 and 3 weeks, the surgical wound, which had previously remained open for five months, fully closed. To the best of our knowledge, this is the first case of personalized phage therapy application in felines, which provides further evidence of the effectiveness of this approach. The successful outcome paves the way for personalized phage-antibiotic treatments against persistent infections therapy in veterinary practice.


Subject(s)
Anti-Bacterial Agents , Cat Diseases , Phage Therapy , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Cats , Phage Therapy/veterinary , Pseudomonas Infections/veterinary , Pseudomonas Infections/drug therapy , Pseudomonas Infections/therapy , Cat Diseases/therapy , Cat Diseases/drug therapy , Cat Diseases/microbiology , Pseudomonas aeruginosa/drug effects , Anti-Bacterial Agents/therapeutic use , Ceftazidime/therapeutic use , Drug Resistance, Multiple, Bacterial , Bacteriophages
13.
Photobiomodul Photomed Laser Surg ; 42(5): 356-365, 2024 May.
Article in English | MEDLINE | ID: mdl-38776546

ABSTRACT

Background: Nosocomial wound infection with Pseudomonas aeruginosa (PA) is a serious complication often responsible for the septic mortality of burn patients. Objective: High-intensity antimicrobial blue light (aBL) treatment may represent an alternative therapy for PA infections and will be investigated in this study. Methods: Antibacterial effects of a light-emitting diode array (450-460 nm; 300 mW/cm2; 15/30 min; 270/540 J/cm2) against PA were determined by suspension assay, biofilm assay, and a human skin wound model and compared with 15-min topically applied 3% citric acid (CA) and wound irrigation solution (Prontosan®; PRT). Results: aBL reduced the bacterial number [2.51-3.56 log10 colony-forming unit (CFU)/mL], whereas PRT or CA treatment achieved a 4.64 or 6.60 log10 CFU/mL reduction in suspension assays. aBL reduced biofilm formation by 60-66%. PRT or CA treatment showed reductions by 25% or 13%. Here, aBL reduced bacterial number in biofilms (1.30-1.64 log10 CFU), but to a lower extend than PRT (2.41 log10 CFU) or CA (2.48 log10 CFU). In the wound skin model, aBL (2.21-2.33 log10 CFU) showed a bacterial reduction of the same magnitude as PRT (2.26 log10 CFU) and CA (2.30 log10 CFU). Conclusions: aBL showed a significant antibacterial efficacy against PA and biofilm formation in a short time. However, a clinical application of aBL in wound therapy requires effective active skin cooling and eye protection, which in turn may limit clinical implementation.


Subject(s)
Biofilms , Pseudomonas Infections , Pseudomonas aeruginosa , Wound Infection , Humans , Pseudomonas aeruginosa/radiation effects , Biofilms/radiation effects , Pseudomonas Infections/therapy , Pseudomonas Infections/radiotherapy , Wound Infection/therapy , Wound Infection/microbiology , Phototherapy , Blue Light
14.
Gene Ther ; 31(7-8): 400-412, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38678160

ABSTRACT

Pseudomonas aeruginosa poses a significant threat to immunocompromised individuals and those with cystic fibrosis. Treatment relies on antibiotics, but persistent infections occur due to intrinsic and acquired resistance of P. aeruginosa towards multiple classes of antibiotics. To date, there are no licensed vaccines for this pathogen, prompting the urgent need for novel treatment approaches to combat P. aeruginosa infection and persistence. Here we validated AAV vectored immunoprophylaxis as a strategy to generate long-term plasma and mucosal expression of highly protective monoclonal antibodies (mAbs) targeting the exopolysaccharide Psl (Cam-003) and the PcrV (V2L2MD) component of the type-III secretion system injectosome either as single mAbs or together as a bispecific mAb (MEDI3902) in a mouse model. When administered intramuscularly, AAV-αPcrV, AAV-αPsl, and AAV-MEDI3902 significantly protected mice challenged intranasally with a lethal dose of P. aeruginosa strains PAO1 and PA14 and reduced bacterial burden and dissemination to other organs. While all AAV-mAbs provided protection, AAV-αPcrV and AAV-MEDI3902 provided 100% and 87.5% protection from a lethal challenge with 4.47 × 107 CFU PAO1 and 87.5% and 75% protection from a lethal challenge with 3 × 107 CFU PA14, respectively. Serum concentrations of MEDI3902 were ~10× lower than that of αPcrV, but mice treated with this vector showed a greater reduction in bacterial dissemination to the liver, lung, spleen, and blood compared to other AAV-mAbs. These results support further investigation into the use of AAV vectored immunoprophylaxis to prevent and treat P. aeruginosa infections and other bacterial pathogens of public health concern for which current treatment strategies are limited.


Subject(s)
Antibodies, Monoclonal , Dependovirus , Genetic Vectors , Pseudomonas Infections , Pseudomonas aeruginosa , Animals , Dependovirus/genetics , Dependovirus/immunology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/genetics , Mice , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Pseudomonas Infections/prevention & control , Pseudomonas Infections/immunology , Pseudomonas Infections/therapy , Antibodies, Monoclonal/immunology , Antibodies, Bispecific , Female , Antigens, Bacterial/immunology , Antigens, Bacterial/genetics , Pneumonia, Bacterial/prevention & control , Pneumonia, Bacterial/therapy , Pneumonia, Bacterial/immunology , Disease Models, Animal , Mice, Inbred C57BL , Humans , Bacterial Toxins , Pore Forming Cytotoxic Proteins
15.
PLoS Biol ; 22(4): e3002566, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38652717

ABSTRACT

Phage therapy is a therapeutic approach to treat multidrug-resistant (MDR) infections that employs lytic bacteriophages (phages) to eliminate bacteria. Despite the abundant evidence for its success as an antimicrobial in Eastern Europe, there is scarce data regarding its effects on the human host. Here, we aimed to understand how lytic phages interact with cells of the airway epithelium, the tissue site that is colonized by bacterial biofilms in numerous chronic respiratory disorders. Using a panel of Pseudomonas aeruginosa phages and human airway epithelial cells (AECs) derived from a person with cystic fibrosis (CF), we determined that interactions between phages and epithelial cells depend on specific phage properties as well as physiochemical features of the microenvironment. Although poor at internalizing phages, the airway epithelium responds to phage exposure by changing its transcriptional profile and secreting antiviral and proinflammatory cytokines that correlate with specific phage families. Overall, our findings indicate that mammalian responses to phages are heterogenous and could potentially alter the way that respiratory local defenses aid in bacterial clearance during phage therapy. Thus, besides phage receptor specificity in a particular bacterial isolate, the criteria to select lytic phages for therapy should be expanded to include mammalian cell responses.


Subject(s)
Cystic Fibrosis , Cytokines , Epithelial Cells , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/virology , Epithelial Cells/virology , Epithelial Cells/metabolism , Epithelial Cells/immunology , Cytokines/metabolism , Cystic Fibrosis/therapy , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Phage Therapy , Bacteriophages/physiology , Bacteriophages/genetics , Respiratory Mucosa/virology , Respiratory Mucosa/metabolism , Respiratory Mucosa/immunology , Pseudomonas Infections/therapy , Pseudomonas Infections/immunology , Pseudomonas Phages/metabolism , Biofilms
16.
Front Cell Infect Microbiol ; 14: 1296777, 2024.
Article in English | MEDLINE | ID: mdl-38469347

ABSTRACT

Phage therapy is a potential approach in the biocontrol of foodborne pathogens. However, the emergence of phage resistance and the narrow host range of most phage isolates continue to limit the antimicrobial efficacy of phages. Here, we investigated the potential of the pqsA gene, encoding the anthranilate-CoA ligase enzyme, as an adjuvant for phage therapy. The knockout of the pqsA gene significantly enhanced the bactericidal effect of phages vB_Pae_QDWS and vB_Pae_S1 against Pseudomonas aeruginosa. Under phage infection pressure, the growth of the PaΔpqsA was significantly inhibited within 8 h compared to the wild-type PAO1. Furthermore, we found that altering phage adsorption is not how PaΔpqsA responds to phage infection. Although pqsA represents a promising target for enhancing phage killing, it may not be applicable to all phages, such as types vB_Pae_W3 and vB_Pae_TR. Our findings provide new material reserves for the future design of novel phage-based therapeutic strategies.


Subject(s)
Bacteriophages , Phage Therapy , Pseudomonas Infections , Pseudomonas Phages , Humans , Pseudomonas aeruginosa/genetics , Pseudomonas Phages/genetics , Pseudomonas Infections/therapy , Mutation
17.
Antimicrob Agents Chemother ; 68(4): e0172823, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38470133

ABSTRACT

Left ventricular assist devices (LVAD) are increasingly used for management of heart failure; infection remains a frequent complication. Phage therapy has been successful in a variety of antibiotic refractory infections and is of interest in treating LVAD infections. We performed a retrospective review of four patients that underwent five separate courses of intravenous (IV) phage therapy with concomitant antibiotic for treatment of endovascular Pseudomonas aeruginosa LVAD infection. We assessed phage susceptibility, bacterial strain sequencing, serum neutralization, biofilm activity, and shelf-life of phage preparations. Five treatments of one to four wild-type virulent phage(s) were administered for 14-51 days after informed consent and regulatory approval. There was no successful outcome. Breakthrough bacteremia occurred in four of five treatments. Two patients died from the underlying infection. We noted a variable decline in phage susceptibility following three of five treatments, four of four tested developed serum neutralization, and prophage presence was confirmed in isolates of two tested patients. Two phage preparations showed an initial titer drop. Phage biofilm activity was confirmed in two. Phage susceptibility alone was not predictive of clinical efficacy in P. aeruginosa endovascular LVAD infection. IV phage was associated with serum neutralization in most cases though lack of clinical effect may be multifactorial including presence of multiple bacterial isolates with varying phage susceptibility, presence of prophages, decline in phage titers, and possible lack of biofilm activity. Breakthrough bacteremia occurred frequently (while the organism remained susceptible to administered phage) and is an important safety consideration.


Subject(s)
Bacteremia , Bacteriophages , Heart-Assist Devices , Phage Therapy , Pseudomonas Infections , Humans , Pseudomonas aeruginosa , Heart-Assist Devices/adverse effects , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Anti-Bacterial Agents/therapeutic use , Prophages , Bacteremia/drug therapy
18.
Exp Eye Res ; 242: 109863, 2024 May.
Article in English | MEDLINE | ID: mdl-38494102

ABSTRACT

PURPOSE: Pseudomonas aeruginosa-induced keratitis is one of the most severe and challenging forms of corneal infection, owing to its associated intense inflammatory reactions leading to corneal necrosis and dense corneal scar with loss of vision. Since mesenchymal stem cells (MSCs) are reported to possess antimicrobial and immunomodulatory properties, they can be tested as an adjuvant treatment along with the antibiotics which are the current standard of care. This study aims to investigate the anti-bacterial and immunomodulatory roles of human bone marrow MSC-derived conditioned medium (MSC-CM) in P. aeruginosa-infected human corneal epithelial cells (HCECs) in vitro. METHODS: The effect of MSC-CM on the growth of clinical isolates of P. aeruginosa was evaluated by colony-forming unit assay. The expression of inflammatory cytokines (IL-6 and TNF-α) and an antimicrobial peptide (Lipocalin 2) in lipopolysaccharide-treated MSCs and HCECs was analyzed through ELISA. Corneal epithelial repair following infection with P. aeruginosa was studied through scratch assay. RESULTS: Compared to control (P. aeruginosa (5*105) incubated in DMEM (1 ml) at 37 °C for 16 h), MSC-CM significantly: i) inhibits the growth of P. aeruginosa (159*109 vs. 104*109 CFU/ml), ii) accelerates corneal epithelial repair following infection with P. aeruginosa (9% vs. 24% closure of the wounded area after 12 h of infection), and iii) downregulates the lipopolysaccharide-induced expression of IL-6, TNF-α and Lipocalin 2 in HCECs. A combination of MSC-CM with an antibiotic, Ciprofloxacin moderately regulated the expression of IL-6, TNF-α, and Lipocalin 2. CONCLUSION: MSC-CM holds promise as an adjunctive therapeutic approach for P. aeruginosa-induced corneal epithelial damage.


Subject(s)
Enzyme-Linked Immunosorbent Assay , Eye Infections, Bacterial , Mesenchymal Stem Cells , Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Eye Infections, Bacterial/microbiology , Eye Infections, Bacterial/metabolism , Eye Infections, Bacterial/pathology , Pseudomonas Infections/microbiology , Pseudomonas Infections/therapy , Pseudomonas Infections/drug therapy , Mesenchymal Stem Cells/metabolism , Epithelium, Corneal/microbiology , Epithelium, Corneal/pathology , Epithelium, Corneal/metabolism , Cells, Cultured , Keratitis/microbiology , Keratitis/metabolism , Keratitis/pathology , Mesenchymal Stem Cell Transplantation/methods , Culture Media, Conditioned/pharmacology , Proof of Concept Study , Interleukin-6/metabolism , Corneal Ulcer/microbiology , Corneal Ulcer/metabolism , Corneal Ulcer/pathology , Corneal Ulcer/drug therapy , Lipocalin-2/metabolism , Tumor Necrosis Factor-alpha/metabolism
19.
mSphere ; 9(2): e0055323, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38299825

ABSTRACT

The bacteriophage is an effective adjunct to existing antibiotic therapy; however, in the course of bacteriophage therapy, host bacteria will develop resistance to bacteriophages, thus affecting the efficacy. Therefore, it is important to describe how bacteria evade bacteriophage attack and the consequences of the biological changes that accompany the development of bacteriophage resistance before the bacteriophage is applied. The specific bacteriophage vB3530 of Pseudomonas aeruginosa (P. aeruginosa) has stable biological characteristics, short incubation period, strong in vitro cleavage ability, and absence of virulence or resistance genes. Ten bacteriophage-resistant strains (TL3780-R) were induced using the secondary infection approach, and the plaque assay showed that vB3530 was less sensitive to TL3780-R. Identification of bacteriophage adsorption receptors showed that the bacterial surface polysaccharide was probably the adsorption receptor of vB3530. In contrast to the TL3780 parental strain, TL3780-R is characterized by the absence of long lipopolysaccharide chains, which may be caused by base insertion of wzy or deletion of galU. It is also intriguing to observe that, in comparison to the parent strain, the bacteriophage-resistant strains TL3780-R mostly exhibited a large cost of fitness (growth rate, biofilm formation, motility, and ability to produce enhanced pyocyanin). In addition, TL3780-R9 showed increased susceptibility to aminoglycosides and chlorhexidine, which may be connected to the loss and down-regulation of mexX expression. Consequently, these findings fully depicted the resistance mechanism of P. aeruginosa to vB3530 and the fitness cost of bacteriophage resistance, laying a foundation for further application of bacteriophage therapy.IMPORTANCEThe bacteriophage is an effective adjunct to existing antibiotic therapy; However, bacteria also develop defensive mechanisms against bacteriophage attack. Thus, there is an urgent need to deeply understand the resistance mechanism of bacteria to bacteriophages and the fitness cost of bacteriophage resistance so as to lay the foundation for subsequent application of the phage. In this study, a specific bacteriophage vB3530 of P. aeruginosa had stable biological characteristics, short incubation period, strong in vitro cleavage ability, and absence of virulence or resistance genes. In addition, we found that P. aeruginosa may lead to phage resistance due to the deletion of galU and the base insertion of wzy, involved in the synthesis of lipopolysaccharides. Simultaneously, we showed the association with the biological state of the bacteria after bacteria acquire bacteriophage resistance, which is extremely relevant to guide the future application of therapeutic bacteriophages.


Subject(s)
Bacteriophages , Pseudomonas Infections , Humans , Bacteriophages/genetics , Pseudomonas aeruginosa , Pseudomonas Infections/therapy , Pseudomonas Infections/microbiology , Anti-Bacterial Agents/pharmacology , Virulence
20.
Microbiol Spectr ; 12(3): e0371923, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38345389

ABSTRACT

The ESKAPEE pathogen Pseudomonas aeruginosa is a common cause of chronic wound and cystic fibrosis lung infections, as well as acute burn and nosocomial infections. Many of these infections are recalcitrant to conventional antibiotic therapies due to both traditional antibiotic resistance mechanisms and antimicrobial tolerance. Recent successes with bacteriophage (phage) therapy to treat chronic human P. aeruginosa infections have led to a renewed interest in isolating and characterizing new P. aeruginosa phages. Here, we isolated and characterized a new lytic phage (termed PIP, pili-infecting phage) capable of infecting P. aeruginosa PA14. PIP is a tailed phage with an icosahedral head and flexible tail containing a genome that is 57,462 bp in length. Phylogenetic analysis reveals that PIP belongs to the subfamily Queuovirinae and genus Nipunavirus but is highly divergent in gene content from known Nipunaviruses. By isolating and characterizing a P. aeruginosa strain that spontaneously evolved resistance to PIP, we show that the receptor for PIP is Type IV pili. In summary, we isolated a new P. aeruginosa phage species with a unique genome, thus increasing the diversity of phages known to infect this important human pathogen.IMPORTANCEThe opportunistic pathogen Pseudomonas aeruginosa causes both acute and chronic human infections. These infections are notoriously difficult to treat due to both antibiotic resistance and antibiotic tolerance. The increasing frequency of antibiotic failure in P. aeruginosa infections has led scientists to explore other treatment options, including bacteriophage (phage) therapy. To this end, there has been a significant effort to identify new Pseudomonas phages. Here, we isolated and characterized a bacteriophage (termed PIP, pili-infecting phage) that infects P. aeruginosa PA14. Examination of the PIP genome revealed that this phage represents a new species in the subclass Queuovirinae. The isolation and characterization of spontaneous PA14 mutants that are resistant to PIP infection revealed Type IV pili as the PIP receptor. Ultimately, this study characterizes a new species of Pseudomonas phage, thus enhancing the known diversity of phages that infect this important pathogen.


Subject(s)
Bacteriophages , Pseudomonas Infections , Pseudomonas Phages , Humans , Bacteriophages/genetics , Pseudomonas aeruginosa/genetics , Phylogeny , Pseudomonas Infections/therapy , Pseudomonas Phages/genetics , Anti-Bacterial Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL