Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Small ; 20(27): e2308814, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38282203

RESUMO

There is a recent resurgence of interest in phage therapy (the therapeutic use of bacterial viruses) as an approach to eliminating difficult-to-treat infections. However, existing approaches for therapeutic phage selection and virulence testing are time-consuming, host-dependent, and facing reproducibility issues. Here, this study presents an innovative approach wherein integrated resonant photonic crystal (PhC) cavities in silicon are used as optical nanotweezers for probing and manipulating single bacteria and single virions with low optical power. This study demonstrates that these nanocavities differentiate between a bacterium and a phage without labeling or specific surface bioreceptors. Furthermore, by tailoring the spatial extent of the resonant optical mode in the low-index medium, phage distinction across phenotypically distinct phage families is demonstrated. The work paves the road to the implementation of optical nanotweezers in phage therapy protocols.


Assuntos
Bacteriófagos , Pinças Ópticas , Vírion , Bacteriófagos/fisiologia
2.
J Infect Dis ; 225(8): 1452-1459, 2022 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33668071

RESUMO

BACKGROUND: The optimal method for delivering phages in the context of ventilator-associated pneumonia (VAP) is unknown. In the current study, we assessed the utility of aerosolized phages (aerophages) for experimental methicillin-resistant Staphylococcus aureus (MRSA) pneumonia. METHODS: Rats were ventilated for 4 hours before induction of pneumonia. Animals received one of the following: (1) aerophages; (2) intravenous (IV) phages; (3) a combination of IV and aerophages; (4) IV linezolid; or (5) a combination of IV linezolid and aerophages. Phages were administered at 2, 12, 24, 48, and 72 hours, and linezolid was administered at 2, 12, 24, 36, 48, 60, and 72 hours. The primary outcome was survival at 96 hours. Secondary outcomes were bacterial and phage counts in tissues and histopathological scoring of the lungs. RESULTS: Aerophages and IV phages each rescued 50% of animals from severe MRSA pneumonia (P < .01 compared with placebo controls). The combination of aerophages and IV phages rescued 91% of animals, which was higher than either monotherapy (P < .05). Standard-of-care antibiotic linezolid rescued 38% of animals. However, linezolid and aerophages did not synergize in this setting (55% survival). CONCLUSIONS: Aerosolized phage therapy showed potential for the treatment of MRSA pneumonia in an experimental animal model and warrants further investigation for application in humans.


Assuntos
Bacteriófagos , Staphylococcus aureus Resistente à Meticilina , Pneumonia Estafilocócica , Pneumonia Associada à Ventilação Mecânica , Animais , Linezolida/uso terapêutico , Pneumonia Estafilocócica/microbiologia , Pneumonia Associada à Ventilação Mecânica/tratamento farmacológico , Ratos
3.
J Biol Chem ; 296: 100639, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33838182

RESUMO

Endolysins are peptidoglycan hydrolases produced at the end of the bacteriophage (phage) replication cycle to lyse the host cell. Endolysins in Gram-positive phages come in a variety of multimodular forms that combine different catalytic and cell wall binding domains. However, the reason why phages adopt endolysins with such complex multidomain architecture is not well understood. In this study, we used the Streptococcus dysgalactiae phage endolysin PlySK1249 as a model to investigate the role of multidomain architecture in phage-induced bacterial lysis and lysis regulation. PlySK1249 consists of an amidase (Ami) domain that lyses bacterial cells, a nonbacteriolytic endopeptidase (CHAP) domain that acts as a dechaining enzyme, and a central LysM cell wall binding domain. We observed that the Ami and CHAP domains synergized for peptidoglycan digestion and bacteriolysis in the native enzyme or when expressed individually and reunified. The CHAP endopeptidase resolved complex polymers of stem-peptides to dimers and helped the Ami domain to digest peptidoglycan to completion. We also found that PlySK1249 was subject to proteolytic cleavage by host cell wall proteases both in vitro and after phage induction. Cleavage disconnected the different domains by hydrolyzing their linker regions, thus hindering their bacteriolytic cooperation and possibly modulating the lytic activity of the enzyme. PlySK1249 cleavage by cell-wall-associated proteases may represent another example of phage adaptation toward the use of existing bacterial regulation mechanism for their own advantage. In addition, understanding more thoroughly the multidomain interplay of PlySK1249 broadens our knowledge on the ideal architecture of therapeutic antibacterial endolysins.


Assuntos
Bacteriólise , Endopeptidases/química , Endopeptidases/metabolismo , N-Acetil-Muramil-L-Alanina Amidase/metabolismo , Peptidoglicano/metabolismo , Fagos de Streptococcus/enzimologia , Streptococcus/crescimento & desenvolvimento , Parede Celular , Domínios Proteicos , Streptococcus/virologia
4.
Rev Med Suisse ; 18(804): 2150-2156, 2022 Nov 16.
Artigo em Francês | MEDLINE | ID: mdl-36382975

RESUMO

The crisis of antibiotic resistance represents a global public health challenge, affecting particularly patients with respiratory infections. The use of (bacterio)phages for the treatment of bacterial infections (phage therapy) seems safe but its effectiveness has not yet been proven by controlled clinical trials. Nevertheless, phage therapy is regaining interest, encouraged by published cases treated successfully with personalized phage combinations as well as significant advances at a preclinical level. Standardized approaches in phage production and treatment administration, as well as future translational studies, are needed to improve our understanding and explore the potential of phage therapy.


La crise de l'antibiorésistance représente un enjeu considérable en santé publique, touchant particulièrement les patients avec des infections respiratoires. L'utilisation des (bactério)phages pour le traitement des infections bactériennes semble sécuritaire mais son efficacité n'a pas encore été formellement démontrée dans des essais cliniques contrôlés. La phagothérapie regagne de l'intérêt comme traitement personnalisé pour les patients qui ne répondent pas aux traitements standards, comme en témoignent les multiples cas publiés ainsi que des découvertes significatives au niveau préclinique. Des approches standardisées concernant la production et l'administration des phages ainsi que des études translationnelles sont nécessaires afin d'améliorer notre compréhension et d'explorer le potentiel de la phagothérapie.


Assuntos
Infecções Bacterianas , Bacteriófagos , Terapia por Fagos , Infecções Respiratórias , Humanos , Infecções Bacterianas/terapia , Infecções Bacterianas/microbiologia , Infecções Respiratórias/terapia , Resistência Microbiana a Medicamentos , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia
5.
Am J Respir Crit Care Med ; 200(9): 1126-1133, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31260638

RESUMO

Rationale: Infections caused by multidrug-resistant bacteria are a major clinical challenge. Phage therapy is a promising alternative antibacterial strategy.Objectives: To evaluate the efficacy of intravenous phage therapy for the treatment of ventilator-associated pneumonia due to methicillin-resistant Staphylococcus aureus in rats.Methods: In a randomized, blinded, controlled experimental study, we compared intravenous teicoplanin (3 mg/kg, n = 12), a cocktail of four phages (2-3 × 109 plaque-forming units/ml of 2003, 2002, 3A, and K; n = 12), and a combination of both (n = 11) given 2, 12, and 24 hours after induction of pneumonia, and then once daily for 4 days. The primary outcome was survival at Day 4. Secondary outcomes were bacterial and phage densities in lungs and spleen, histopathological scoring of infection within the lungs, and inflammatory biomarkers in blood.Measurements and Main Results: Treatment with either phages or teicoplanin increased survival from 0% to 58% and 50%, respectively (P < 0.005). The combination of phages and antibiotics did not further improve outcomes (45% survival). Animal survival correlated with reduced bacterial burdens in the lung (1.2 × 106 cfu/g of tissue for survivors vs. 1.2 × 109 cfu/g for nonsurviving animals; P < 0.0001), as well as improved histopathological outcomes. Phage multiplication within the lung occurred during treatment. IL-1ß increased in all treatment groups over the course of therapy.Conclusions: Phage therapy was as effective as teicoplanin in improving survival and decreasing bacterial load within the lungs of rats infected with methicillin-resistant S. aureus. Combining antibiotics with phage therapy did not further improve outcomes.


Assuntos
Staphylococcus aureus Resistente à Meticilina , Terapia por Fagos , Pneumonia Associada à Ventilação Mecânica/microbiologia , Pneumonia Associada à Ventilação Mecânica/terapia , Infecções Estafilocócicas/terapia , Animais , Antibacterianos/uso terapêutico , Bacteriófagos , Modelos Animais de Doenças , Masculino , Ratos , Ratos Wistar , Infecções Estafilocócicas/microbiologia , Teicoplanina/uso terapêutico
6.
Iran J Med Sci ; 45(4): 298-303, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32801419

RESUMO

BACKGROUND: Escherichia coli (E. coli) is the most common cause of urinary tract infection (UTI) and typically treated with antibiotics. Unrestricted use of antibiotics may lead to the emergence of antibiotic-resistant bacteria. The present study aimed to isolate and characterize phages against E. coli from infected urine samples and to determine the lytic activity of phages against E. coli in vitro. METHODS: The present experimental study was conducted in the Laboratory of Bouali Sina Hospital (Sari, Iran) in May 2018. E. coli was identified from nine urine samples of patients with UTI using conventional microbiological methods. Bacteriophages were isolated from the infected urine specimens, and their lytic activity was determined using the spot test. The titer of the bacteriophages was measured using the double-layer agar technique. The morphology of the bacteriophages was revealed using transmission electron microscopy, and the latent time period and burst size were determined. Data were analyzed using the SPSS software package. RESULTS: E. coli was isolated from nine infected urine samples. The lytic activity of bacteriophages against E. coli was determined using the spot test by observing the formation of inhibition zones. Transmission electron microscopy showed E. coli phages belonging to the Myoviridae family. The latent time period was 20 minutes with a burst size of 1,200 plaque-forming unit (PFU) per infected cell. The results of the double-layer agar assay showed that the titer of bacteriophages was 20×108 PFU/mL. CONCLUSION: The E. coli bacteriophage was isolated from infected urine samples and characterized, and their lytic activity against E. coli was determined in vitro.

7.
Rev Med Suisse ; 15(646): 771-775, 2019 Apr 10.
Artigo em Francês | MEDLINE | ID: mdl-30969489

RESUMO

Phages are viruses able to specifically infect and lyse target bacteria. The use of phages in the treatment of bacterial infections has been limited due of the development of antibiotics since the 1950's. However, phagotherapy is now gaining more interest due to the emergence of multidrug resistant bacteria. Here, we present the benefits and disadvantages of using phages versus antibiotics to treat bacterial infections and describe potential development areas for phagotherapy by reviewing current evidence.


Les phages sont des virus capables d'infecter et de lyser spécifiquement leur bactérie cible. L'utilisation des phages dans le traitement des infections bactériennes, limitée par le développement des antibiotiques à partir des années 1950, regagne de l'intérêt du fait de l'émergence croissante de bactéries multirésistantes. Cet article présente les principes de la phagothérapie en comparant les avantages et inconvénients de cette stratégie par rapport à l'antibiothérapie classique. Il illustre les perspectives d'utilisation des phages à travers une revue des études cliniques existantes.


Assuntos
Infecções Bacterianas , Bacteriófagos , Terapia por Fagos , Antibacterianos , Infecções Bacterianas/terapia , Farmacorresistência Bacteriana Múltipla , Humanos
8.
BMC Bioinformatics ; 19(Suppl 14): 420, 2018 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-30453987

RESUMO

BACKGROUND: Antibiotic resistance and its rapid dissemination around the world threaten the efficacy of currently-used medical treatments and call for novel, innovative approaches to manage multi-drug resistant infections. Phage therapy, i.e., the use of viruses (phages) to specifically infect and kill bacteria during their life cycle, is one of the most promising alternatives to antibiotics. It is based on the correct matching between a target pathogenic bacteria and the therapeutic phage. Nevertheless, correctly matching them is a major challenge. Currently, there is no systematic method to efficiently predict whether phage-bacterium interactions exist and these pairs must be empirically tested in laboratory. Herein, we present our approach for developing a computational model able to predict whether a given phage-bacterium pair can interact based on their genome. RESULTS: Based on public data from GenBank and phagesDB.org, we collected more than a thousand positive phage-bacterium interactions with their complete genomes. In addition, we generated putative negative (i.e., non-interacting) pairs. We extracted, from the collected genomes, a set of informative features based on the distribution of predictive protein-protein interactions and on their primary structure (e.g. amino-acid frequency, molecular weight and chemical composition of each protein). With these features, we generated multiple candidate datasets to train our algorithms. On this base, we built predictive models exhibiting predictive performance of around 90% in terms of F1-score, sensitivity, specificity, and accuracy, obtained on the test set with 10-fold cross-validation. CONCLUSION: These promising results reinforce the hypothesis that machine learning techniques may produce highly-predictive models accelerating the search of interacting phage-bacteria pairs.


Assuntos
Biologia Computacional/métodos , Análise de Dados , Genômica , Aprendizado de Máquina , Algoritmos , Bactérias/virologia , Bacteriófagos/genética , Proteínas/química , Especificidade da Espécie
9.
J Infect Dis ; 215(5): 703-712, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28007922

RESUMO

Background: Increasing antibiotic resistance warrants therapeutic alternatives. Here we investigated the efficacy of bacteriophage-therapy (phage) alone or combined with antibiotics against experimental endocarditis (EE) due to Pseudomonas aeruginosa, an archetype of difficult-to-treat infection. Methods: In vitro fibrin clots and rats with aortic EE were treated with an antipseudomonas phage cocktail alone or combined with ciprofloxacin. Phage pharmacology, therapeutic efficacy, and resistance were determined. Results: In vitro, single-dose phage therapy killed 7 log colony-forming units (CFUs)/g of fibrin clots in 6 hours. Phage-resistant mutants regrew after 24 hours but were prevented by combination with ciprofloxacin (2.5 × minimum inhibitory concentration). In vivo, single-dose phage therapy killed 2.5 log CFUs/g of vegetations in 6 hours (P < .001 vs untreated controls) and was comparable with ciprofloxacin monotherapy. Moreover, phage/ciprofloxacin combinations were highly synergistic, killing >6 log CFUs/g of vegetations in 6 hours and successfully treating 64% (n = 7/11) of rats. Phage-resistant mutants emerged in vitro but not in vivo, most likely because resistant mutations affected bacterial surface determinants important for infectivity (eg, the pilT and galU genes involved in pilus motility and LPS formation). Conclusions: Single-dose phage therapy was active against P. aeruginosa EE and highly synergistic with ciprofloxacin. Phage-resistant mutants had impaired infectivity. Phage-therapy alone or combined with antibiotics merits further clinical consideration.


Assuntos
Antibacterianos/farmacologia , Endocardite/terapia , Terapia por Fagos/métodos , Infecções por Pseudomonas/terapia , Pseudomonas aeruginosa/efeitos dos fármacos , Animais , Ciprofloxacina/farmacologia , Farmacorresistência Bacteriana Múltipla , Endocardite/microbiologia , Feminino , Testes de Sensibilidade Microbiana , Pseudomonas aeruginosa/patogenicidade , Ratos , Ratos Wistar , Virulência
10.
Pharm Res ; 32(7): 2173-9, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25585954

RESUMO

The worldwide antibiotic crisis has led to a renewed interest in phage therapy. Since time immemorial phages control bacterial populations on Earth. Potent lytic phages against bacterial pathogens can be isolated from the environment or selected from a collection in a matter of days. In addition, phages have the capacity to rapidly overcome bacterial resistances, which will inevitably emerge. To maximally exploit these advantage phages have over conventional drugs such as antibiotics, it is important that sustainable phage products are not submitted to the conventional long medicinal product development and licensing pathway. There is a need for an adapted framework, including realistic production and quality and safety requirements, that allows a timely supplying of phage therapy products for 'personalized therapy' or for public health or medical emergencies. This paper enumerates all phage therapy product related quality and safety risks known to the authors, as well as the tests that can be performed to minimize these risks, only to the extent needed to protect the patients and to allow and advance responsible phage therapy and research.


Assuntos
Infecções Bacterianas , Bacteriófagos/crescimento & desenvolvimento , Terapia Biológica , Farmacorresistência Bacteriana Múltipla , Infecções Bacterianas/microbiologia , Infecções Bacterianas/terapia , Bacteriófagos/isolamento & purificação , Terapia Biológica/efeitos adversos , Terapia Biológica/normas , Terapia Biológica/tendências , Humanos
11.
Mol Cell Proteomics ; 11(11): 1123-39, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22843989

RESUMO

Staphylococcus aureus infections involve numerous adhesins and toxins, which expression depends on complex regulatory networks. Adhesins include a family of surface proteins covalently attached to the peptidoglycan via a conserved LPXTG motif. Here we determined the protein and mRNA expression of LPXTG-proteins of S. aureus Newman in time-course experiments, and their relation to fibrinogen adherence in vitro. Experiments were performed with mutants in the global accessory-gene regulator (agr), surface protein A (Spa), and fibrinogen-binding protein A (ClfA), as well as during growth in iron-rich or iron-poor media. Surface proteins were recovered by trypsin-shaving of live bacteria. Released peptides were analyzed by liquid chromatography coupled to tandem mass-spectrometry. To unambiguously identify peptides unique to LPXTG-proteins, the analytical conditions were refined using a reference library of S. aureus LPXTG-proteins heterogeneously expressed in surrogate Lactococcus lactis. Transcriptomes were determined by microarrays. Sixteen of the 18 LPXTG-proteins present in S. aureus Newman were detected by proteomics. Nine LPXTG-proteins showed a bell-shape agr-like expression that was abrogated in agr-negative mutants including Spa, fibronectin-binding protein A (FnBPA), ClfA, iron-binding IsdA, and IsdB, immunomodulator SasH, functionally uncharacterized SasD, biofilm-related SasG and methicillin resistance-related FmtB. However, only Spa and SasH modified their proteomic and mRNA profiles in parallel in the parent and its agr- mutant, whereas all other LPXTG-proteins modified their proteomic profiles independently of their mRNA. Moreover, ClfA became highly transcribed and active in fibrinogen-adherence tests during late growth (24 h), whereas it remained poorly detected by proteomics. On the other hand, iron-regulated IsdA-B-C increased their protein expression by >10-times in iron-poor conditions. Thus, proteomic, transcriptomic, and adherence-phenotype demonstrated differential profiles in S. aureus. Moreover, trypsin peptide signatures suggested differential protein domain exposures in various environments, which might be relevant for anti-adhesin vaccines. A comprehensive understanding of the S. aureus physiology should integrate all three approaches.


Assuntos
Aderência Bacteriana/genética , Perfilação da Expressão Gênica , Proteínas de Membrana/metabolismo , Mutação/genética , Proteômica , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Aderência Bacteriana/efeitos dos fármacos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bases de Dados de Proteínas , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Genótipo , Ferro/farmacologia , Cinética , Lactococcus/efeitos dos fármacos , Lactococcus/metabolismo , Proteínas de Membrana/genética , Viabilidade Microbiana/efeitos dos fármacos , Biblioteca de Peptídeos , Peptídeos/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/crescimento & desenvolvimento , Tripsina/metabolismo
12.
Antimicrob Agents Chemother ; 57(12): 6276-83, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24100496

RESUMO

Beta-hemolytic Streptococcus agalactiae is the leading cause of bacteremia and invasive infections. These diseases are treated with ß-lactams or macrolides, but the emergence of less susceptible and even fully resistant strains is a cause for concern. New bacteriophage lysins could be promising alternatives against such organisms. They hydrolyze the bacterial peptidoglycan at the end of the phage cycle, in order to release the phage progeny. By using a bioinformatic approach to screen several beta-hemolytic streptococci, a gene coding for a lysin was identified on a prophage carried by Streptococcus dysgalactiae subsp. equisimilis SK1249. The gene product, named PlySK1249, harbored an original three-domain structure with a central cell wall-binding domain surrounded by an N-terminal amidase and a C-terminal CHAP domain. Purified PlySK1249 was highly lytic and bactericidal for S. dysgalactiae (2-log10 CFU/ml decrease within 15 min). Moreover, it also efficiently killed S. agalactiae (1.5-log10 CFU/ml decrease within 15 min) but not several streptococcal commensal species. We further investigated the activity of PlySK1249 in a mouse model of S. agalactiae bacteremia. Eighty percent of the animals (n = 10) challenged intraperitoneally with 10(6) CFU of S. agalactiae died within 72 h, whereas repeated injections of PlySK1249 (45 mg/kg 3 times within 24 h) significantly protected the mice (P < 0.01). Thus, PlySK1249, which was isolated from S. dysgalactiae, demonstrated high cross-lytic activity against S. agalactiae both in vitro and in vivo. These encouraging results indicated that PlySK1249 might represent a good candidate to be developed as a new enzybiotic for the treatment of systemic S. agalactiae infections.


Assuntos
Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriemia/tratamento farmacológico , Genoma Bacteriano/genética , Streptococcus agalactiae/efeitos dos fármacos , Animais , Bacteriemia/microbiologia , Escherichia coli/efeitos dos fármacos , Feminino , Camundongos , Staphylococcus aureus/efeitos dos fármacos
13.
Sci Rep ; 13(1): 18204, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875544

RESUMO

S. aureus is a pathogen that frequently causes severe morbidity and phage therapy is being discussed as an alternative to antibiotics for the treatment of S. aureus infections. In this in vitro and animal study, we demonstrated that the activity of anti-staphylococcal phages is severely impaired in 0.5% plasma or synovial fluid. Despite phage replication in these matrices, lysis of the bacteria was slower than phage propagation, and no reduction of the bacterial population was observed. The inhibition of the phages associated with a reduction in phage adsorption, quantified to 99% at 10% plasma. S. aureus is known to bind multiple coagulation factors, resulting in the formation of aggregates and blood clots that might protect the bacterium from the phages. Here, we show that purified fibrinogen at a sub-physiological concentration of 0.4 mg/ml is sufficient to impair phage activity. In contrast, dissolution of the clots by tissue plasminogen activator (tPA) partially restored phage activity. Consistent with these in vitro findings, phage treatment did not reduce bacterial burdens in a neutropenic mouse S. aureus thigh infection model. In summary, phage treatment of S. aureus infections inside the body may be fundamentally challenging, and more investigation is needed prior to proceeding to in-human trials.


Assuntos
Infecções Estafilocócicas , Staphylococcus aureus , Humanos , Animais , Camundongos , Staphylococcus aureus/fisiologia , Ativador de Plasminogênio Tecidual , Líquido Sinovial , Infecções Estafilocócicas/terapia , Infecções Estafilocócicas/microbiologia , Fagos de Staphylococcus/fisiologia , Antibacterianos
14.
Nat Commun ; 14(1): 3629, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37369702

RESUMO

Bacteriophage therapy has been suggested as an alternative or complementary strategy for the treatment of multidrug resistant (MDR) bacterial infections. Here, we report the favourable clinical evolution of a 41-year-old male patient with a Kartagener syndrome complicated by a life-threatening chronic MDR Pseudomonas aeruginosa infection, who is treated successfully with iterative aerosolized phage treatments specifically directed against the patient's isolate. We follow the longitudinal evolution of both phage and bacterial loads during and after phage administration in respiratory samples. Phage titres in consecutive sputum samples indicate in patient phage replication. Phenotypic analysis and whole genome sequencing of sequential bacterial isolates reveals a clonal, but phenotypically diverse population of hypermutator strains. The MDR phenotype in the collected isolates is multifactorial and mainly due to spontaneous chromosomal mutations. All isolates recovered after phage treatment remain phage susceptible. These results demonstrate that clinically significant improvement is achievable by personalised phage therapy even in the absence of complete eradication of P. aeruginosa lung colonization.


Assuntos
Bacteriófagos , Pneumonia , Infecções por Pseudomonas , Masculino , Humanos , Bacteriófagos/genética , Pseudomonas aeruginosa , Pulmão , Farmacorresistência Bacteriana Múltipla , Infecção Persistente , Infecções por Pseudomonas/microbiologia , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico
15.
Microbiol Mol Biol Rev ; 87(4): e0006323, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-37947420

RESUMO

SUMMARYCommunities of microorganisms (microbiota) are present in all habitats on Earth and are relevant for agriculture, health, and climate. Deciphering the mechanisms that determine microbiota dynamics and functioning within the context of their respective environments or hosts (the microbiomes) is crucially important. However, the sheer taxonomic, metabolic, functional, and spatial complexity of most microbiomes poses substantial challenges to advancing our knowledge of these mechanisms. While nucleic acid sequencing technologies can chart microbiota composition with high precision, we mostly lack information about the functional roles and interactions of each strain present in a given microbiome. This limits our ability to predict microbiome function in natural habitats and, in the case of dysfunction or dysbiosis, to redirect microbiomes onto stable paths. Here, we will discuss a systematic approach (dubbed the N+1/N-1 concept) to enable step-by-step dissection of microbiome assembly and functioning, as well as intervention procedures to introduce or eliminate one particular microbial strain at a time. The N+1/N-1 concept is informed by natural invasion events and selects culturable, genetically accessible microbes with well-annotated genomes to chart their proliferation or decline within defined synthetic and/or complex natural microbiota. This approach enables harnessing classical microbiological and diversity approaches, as well as omics tools and mathematical modeling to decipher the mechanisms underlying N+1/N-1 microbiota outcomes. Application of this concept further provides stepping stones and benchmarks for microbiome structure and function analyses and more complex microbiome intervention strategies.


Assuntos
Microbiota , Humanos , Microbiota/genética , Disbiose
16.
FEMS Microbiol Rev ; 46(1)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34289033

RESUMO

Like the sword of Damocles, the threat of a post-antibiotic era is hanging over humanity's head. The scientific and medical community is thus reconsidering bacteriophage therapy (BT) as a partial but realistic solution for treatment of difficult-to-eradicate bacterial infections. Here, we summarize the latest developments in clinical BT applications, with a focus on developments in the following areas: (i) pharmacology of bacteriophages of major clinical importance and their synergy with antibiotics; (ii) production of therapeutic phages; and (iii) clinical trials, case studies and case reports in the field. We address regulatory concerns, which are of paramount importance insofar as they dictate the conduct of clinical trials, which are needed for broader BT application. The increasing amount of new available data confirms the particularities of BT as being innovative and highly personalized. The current circumstances suggest that the immediate future of BT may be advanced within the framework of national BT centers in collaboration with competent authorities, which are urged to adopt incisive initiatives originally launched by some national regulatory authorities.


Assuntos
Infecções Bacterianas , Bacteriófagos , Terapia por Fagos , Antibacterianos/uso terapêutico , Infecções Bacterianas/terapia , Humanos
17.
Viruses ; 14(8)2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-36016414

RESUMO

Background. Recurrent therapeutic failures reported for the treatment of methicillin-resistant Staphylococcus aureus (MRSA) infective endocarditis (IE) with vancomycin may be due to poor bactericidal activity. Alternative antibacterial approaches using bacteriophages may overcome this limitation. Objectives. An experimental rat model of MRSA IE (EE) was used to examine the efficacy of vancomycin combined with a 1:1 bacteriophage (phage) cocktail composed of Herelleviridae vB_SauH_2002 and Routreeviridae 66. Methods. Six hours after inoculation with ca. 5 log10 colony forming units (CFU) of MRSA strain AW7, animals were treated with either: (i) saline, (ii) an equimolar two-phage cocktail (bolus of 1 mL followed by a 0.3 mL/h continuous infusion of 10 log10 plaque forming units (PFU)/mL phage suspension), (iii) vancomycin (at a dose mimicking the kinetics in humans of 0.5 g b.i.d.), or (iv) a combination of both. Bacterial loads in vegetations, and phage loads in vegetations, liver, kidney, spleen, and blood, were measured outcomes. Results. Phage cocktail alone was unable to control the growth of strain AW7 in cardiac vegetations. However, when combined with subtherapeutic doses of vancomycin, a statistically significant decrease of ∆4.05 ± 0.94 log10 CFU/g at 24 h compared to placebo was detected (p < 0.001). The administration of vancomycin was found to significantly impact on the local concentrations of phages in the vegetations and in the organs examined. Conclusions. Lytic bacteriophages as an adjunct treatment to the standard of care antibiotics could potentially improve the management of MRSA IE. Further studies are needed to investigate the impact of antibiotics on phage replication in vivo.


Assuntos
Bacteriófagos , Endocardite Bacteriana , Endocardite , Staphylococcus aureus Resistente à Meticilina , Infecções Estafilocócicas , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Endocardite/tratamento farmacológico , Endocardite Bacteriana/tratamento farmacológico , Endocardite Bacteriana/microbiologia , Testes de Sensibilidade Microbiana , Ratos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Vancomicina/farmacologia , Vancomicina/uso terapêutico
18.
J Am Heart Assoc ; 11(3): e023080, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35043655

RESUMO

Background The potential of phage therapy for the treatment of endovascular Staphylococcus aureus infections remains to be evaluated. Methods and Results The efficacy of a phage cocktail combining Herelleviridae phage vB_SauH_2002 and Podoviriae phage 66 was evaluated against a methicillin-sensitive S. aureus strain in vitro and in vivo in a rodent model of experimental endocarditis. Six hours after bacterial challenge, animals were treated with (1) the phage cocktail. (2) subtherapeutic flucloxacillin dosage, (3) combination of the phage cocktail and flucloxacillin, or (4) saline. Bacterial loads in cardiac vegetations at 30 hours were the primary outcome. Secondary outcomes were phage loads at 30 hours in cardiac vegetations, blood, spleen, liver, and kidneys. We evaluated phage resistance 30 hours post infection in vegetations of rats under combination treatment. In vitro, phages synergized against S. aureus planktonic cells and the cocktail synergized with flucloxacillin to eradicated biofilms. In infected animals, the phage cocktail achieved bacteriostatic effect. The addition of low-dose flucloxacillin elevated bacterial suppression (∆ of -5.25 log10 colony forming unit/g [CFU/g] versus treatment onset, P<0.0001) and synergism was confirmed (∆ of -2.15 log10 CFU/g versus low-dose flucloxacillin alone, P<0.01). Importantly, 9/12 rats given the combination treatment had sterile vegetations at 30 hours. In vivo phage replication was partially suppressed by the antibiotic and selection of resistance to the Podoviridae component of the phage cocktail occurred. Plasma-mediated inhibition of phage killing activity was observed in vitro. Conclusions Combining phages with a low-dose standard of care antibiotic represents a promising strategy for the treatment of S. aureus infective endocarditis.


Assuntos
Bacteriófagos , Endocardite Bacteriana , Endocardite , Infecções Estafilocócicas , Animais , Antibacterianos/farmacologia , Antibacterianos/uso terapêutico , Bacteriófagos/fisiologia , Endocardite/microbiologia , Endocardite Bacteriana/terapia , Floxacilina/farmacologia , Floxacilina/uso terapêutico , Ratos , Infecções Estafilocócicas/terapia , Staphylococcus aureus/fisiologia
19.
Viruses ; 14(11)2022 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-36366553

RESUMO

Erwinia amylovora is a quarantine phytopathogenic bacterium that is the causal agent of fire blight, a destructive disease responsible for killing millions of fruit-bearing plants worldwide, including apple, pear, quince, and raspberry. Efficient and sustainable control strategies for this serious bacterial disease are still lacking, and traditional methods are limited to the use of antibiotics and some basic agricultural practices. This study aimed to contribute to the development of a sustainable control strategy through the identification, characterization, and application of bacteriophages (phages) able to control fire blight on pears. Phages isolated from wastewater collected in the Apulia region (southern Italy) were characterized and evaluated as antibacterial agents to treat experimental fire blight caused by E. amylovora. Transmission electron microscopy (TEM) conducted on purified phages (named EP-IT22 for Erwinia phage IT22) showed particles with icosahedral heads of ca. 90 ± 5 nm in length and long contractile tails of 100 ± 10 nm, typical of the Myoviridae family. Whole genome sequencing (WGS), assembly, and analysis of the phage DNA generated a single contig of 174.346 bp representing a complete circular genome composed of 310 open reading frames (ORFs). EP-IT22 was found to be 98.48% identical to the Straboviridae Erwinia phage Cronus (EPC) (GenBank Acc. n° NC_055743) at the nucleotide level. EP-IT22 was found to be resistant to high temperatures (up to 60 °C) and pH values between 4 and 11, and was able to accomplish a complete lytic cycle within one hour. Furthermore, the viability-qPCR and turbidity assays showed that EP-IT22 (MOI = 1) lysed 94% of E. amylovora cells in 20 h. The antibacterial activity of EP-IT22 in planta was evaluated in E. amylovora-inoculated pear plants that remained asymptomatic 40 days post inoculation, similarly to those treated with streptomycin sulphate. This is the first description of the morphological, biological, and molecular features of EP-IT22, highlighting its promising potential for biocontrol of E. amylovora against fire blight disease.


Assuntos
Bacteriófagos , Erwinia amylovora , Malus , Erwinia amylovora/genética , Bacteriófagos/genética , Doenças das Plantas/prevenção & controle , Doenças das Plantas/microbiologia , Myoviridae/genética
20.
Eur Respir Rev ; 31(166)2022 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-36198417

RESUMO

Lower respiratory tract infections lead to significant morbidity and mortality. They are increasingly caused by multidrug-resistant pathogens, notably in individuals with cystic fibrosis, hospital-acquired pneumonia and lung transplantation. The use of bacteriophages (phages) to treat bacterial infections is gaining growing attention, with numerous published cases of compassionate treatment over the last few years. Although the use of phages appears safe, the lack of standardisation, the significant heterogeneity of published studies and the paucity of robust efficacy data, alongside regulatory hurdles arising from the existing pharmaceutical legislation, are just some of the challenges phage therapy has to overcome. In this review, we discuss the lessons learned from recent clinical experiences of phage therapy for the treatment of pulmonary infections. We review the key aspects, opportunities and challenges of phage therapy regarding formulations and administration routes, interactions with antibiotics and the immune system, and phage resistance. Building upon the current knowledge base, future pre-clinical studies using emerging technologies and carefully designed clinical trials are expected to enhance our understanding and explore the therapeutic potential of phage therapy.


Assuntos
Terapia por Fagos , Pneumonia , Bacteriófagos , Humanos , Legislação de Medicamentos , Terapia por Fagos/efeitos adversos , Pneumonia/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA