Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Commun ; 15(1): 175, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38168031

ABSTRACT

Bacteriophages are ubiquitous viral predators that have primarily been studied using fast-growing laboratory cultures of their bacterial hosts. However, microbial life in nature is mostly in a slow- or non-growing, dormant state. Here, we show that diverse phages can infect deep-dormant bacteria and suspend their replication until the host resuscitates ("hibernation"). However, a newly isolated Pseudomonas aeruginosa phage, named Paride, can directly replicate and induce the lysis of deep-dormant hosts. While non-growing bacteria are notoriously tolerant to antibiotic drugs, the combination with Paride enables the carbapenem meropenem to eradicate deep-dormant cultures in vitro and to reduce a resilient bacterial infection of a tissue cage implant in mice. Our work might inspire new treatments for persistent bacterial infections and, more broadly, highlights two viral strategies to infect dormant bacteria (hibernation and direct replication) that will guide future studies on phage-host interactions.


Subject(s)
Bacteriophages , Pseudomonas Infections , Animals , Mice , Pseudomonas aeruginosa , Anti-Bacterial Agents/pharmacology , Pseudomonas Infections/microbiology
2.
Sci Transl Med ; 14(662): eabj2381, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36103517

ABSTRACT

Drug-resistant Gram-positive bacterial infections are still a substantial burden on the public health system, with two bacteria (Staphylococcus aureus and Streptococcus pneumoniae) accounting for over 1.5 million drug-resistant infections in the United States alone in 2017. In 2019, 250,000 deaths were attributed to these pathogens globally. We have developed a preclinical glycopeptide antibiotic, MCC5145, that has excellent potency (MIC90 ≤ 0.06 µg/ml) against hundreds of isolates of methicillin-resistant S. aureus (MRSA) and other Gram-positive bacteria, with a greater than 1000-fold margin over mammalian cell cytotoxicity values. The antibiotic has therapeutic in vivo efficacy when dosed subcutaneously in multiple murine models of established bacterial infections, including thigh infection with MRSA and blood septicemia with S. pneumoniae, as well as when dosed orally in an antibiotic-induced Clostridioides difficile infection model. MCC5145 exhibited reduced nephrotoxicity at microbiologically active doses in mice compared to vancomycin. MCC5145 also showed improved activity against biofilms compared to vancomycin, both in vitro and in vivo, and a low propensity to select for drug resistance. Characterization of drug action using a transposon library bioinformatic platform showed a mechanistic distinction from other glycopeptide antibiotics.


Subject(s)
Anti-Infective Agents , Gram-Positive Bacterial Infections , Methicillin-Resistant Staphylococcus aureus , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/pharmacology , Biofilms , Glycopeptides/pharmacology , Glycopeptides/therapeutic use , Lipoglycopeptides/therapeutic use , Mammals , Mice , Microbial Sensitivity Tests , Streptococcus pneumoniae , Vancomycin/pharmacology , Vancomycin/therapeutic use
3.
Swiss Med Wkly ; 151: w20550, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34375986

ABSTRACT

OBJECTIVES: Patients with severe COVID-19 may be at risk of longer term sequelae. Long-term clinical, immunological, pulmonary and radiological outcomes of patients treated with anti-inflammatory drugs are lacking. METHODS: In this single-centre prospective cohort study, we assessed 90-day clinical, immunological, pulmonary and radiological outcomes of hospitalised patients with severe COVID-19 treated with tocilizumab from March 2020 to May 2020. Criteria for tocilizumab administration were oxygen saturation <93%, respiratory rate >30/min, C-reactive protein levels >75 mg/l, extensive area of ground-glass opacities or progression on computed tomography (CT). Descriptive analyses were performed using StataIC 16. RESULTS: Between March 2020 and May 2020, 50 (27%) of 186 hospitalised patients had severe COVID-19 and were treated with tocilizumab. Of these, 52% were hospitalised on the intensive care unit (ICU) and 12% died. Eleven (22%) patients developed at least one microbiologically confirmed super-infection, of which 91% occurred on ICU. Median duration of hospitalisation was 15 days (interquartile range [IQR] 10–24) with 24 days (IQR 14–32) in ICU patients and 10 days (IQR 7–15) in non-ICU patients. At day 90, 41 of 44 survivors (93%) were outpatients. No long-term adverse events or late-onset infections were identified after acute hospital care. High SARS-CoV-2 antibody titres were found in all but one patient, who was pretreated with rituximab. Pulmonary function tests showed no obstructive patterns, but restrictive patterns in two (5.7%) and impaired diffusion capacities for carbon monoxide in 11 (31%) of 35 patients, which predominated in prior ICU patients. Twenty-one of 35 (60%) CT-scans at day 90 showed residual abnormalities, with similar distributions between prior ICU and non-ICU patients. CONCLUSIONS: In this cohort of severe COVID-19 patients, no tocilizumab-related long-term adverse events or late-onset infections were identified. Although chest CT abnormalities were highly prevalent at day 90, the majority of patients showed normal lung function. TRIAL REGISTRATION: ClinicalTrials.gov NCT04351503.


Subject(s)
COVID-19 Drug Treatment , Antibodies, Monoclonal, Humanized , Cohort Studies , Humans , Prospective Studies , SARS-CoV-2
4.
Nucleic Acids Res ; 49(13): e73, 2021 07 21.
Article in English | MEDLINE | ID: mdl-33856484

ABSTRACT

Antibiotic-resistant pathogens often escape antimicrobial treatment by forming protective biofilms in response to quorum-sensing communication via diffusible autoinducers. Biofilm formation by the nosocomial pathogen methicillin-resistant Staphylococcus aureus (MRSA) is triggered by the quorum-sensor autoinducer-2 (AI-2), whose biosynthesis is mediated by methylthioadenosine/S-adenosylhomocysteine nucleosidase (MTAN) and S-ribosylhomocysteine lyase (LuxS). Here, we present a high-throughput screening platform for small-molecular inhibitors of either enzyme. This platform employs a cell-based assay to report non-toxic, bioavailable and cell-penetrating inhibitors of AI-2 production, utilizing engineered human cells programmed to constitutively secrete AI-2 by tapping into the endogenous methylation cycle via ectopic expression of codon-optimized MTAN and LuxS. Screening of a library of over 5000 commercial compounds yielded 66 hits, including the FDA-licensed cytostatic anti-cancer drug 5-fluorouracil (5-FU). Secondary screening and validation studies showed that 5-FU is a potent quorum-quencher, inhibiting AI-2 production and release by MRSA, Staphylococcus epidermidis, Escherichia coli and Vibrio harveyi. 5-FU efficiently reduced adherence and blocked biofilm formation of MRSA in vitro at an order-of-magnitude-lower concentration than that clinically relevant for anti-cancer therapy. Furthermore, 5-FU reestablished antibiotic susceptibility and enabled daptomycin-mediated prevention and clearance of MRSA infection in a mouse model of human implant-associated infection.


Subject(s)
Biofilms/drug effects , Enzyme Inhibitors/pharmacology , Fluorouracil/pharmacology , High-Throughput Screening Assays/methods , Methicillin-Resistant Staphylococcus aureus/drug effects , Quorum Sensing/drug effects , Animals , Bacterial Proteins/antagonists & inhibitors , Carbon-Sulfur Lyases/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Female , Fluorouracil/therapeutic use , HEK293 Cells , Homoserine/analogs & derivatives , Homoserine/biosynthesis , Humans , Lactones , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice, Inbred C57BL , N-Glycosyl Hydrolases/antagonists & inhibitors , Small Molecule Libraries , Staphylococcal Infections/prevention & control
5.
Cell Metab ; 32(3): 457-467.e5, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32738204

ABSTRACT

Serum acetate increases upon systemic infection. Acutely, assimilation of acetate expands the capacity of memory CD8+ T cells to produce IFN-γ. Whether acetate modulates memory CD8+ T cell metabolism and function during pathogen re-encounter remains unexplored. Here we show that at sites of infection, high acetate concentrations are being reached, yet memory CD8+ T cells shut down the acetate assimilating enzymes ACSS1 and ACSS2. Acetate, being thus largely excluded from incorporation into cellular metabolic pathways, now had different effects, namely (1) directly activating glutaminase, thereby augmenting glutaminolysis, cellular respiration, and survival, and (2) suppressing TCR-triggered calcium flux, and consequently cell activation and effector cell function. In vivo, high acetate abundance at sites of infection improved pathogen clearance while reducing immunopathology. This indicates that, during different stages of the immune response, the same metabolite-acetate-induces distinct immunometabolic programs within the same cell type.


Subject(s)
Acetates/metabolism , Anti-Inflammatory Agents/metabolism , CD8-Positive T-Lymphocytes/metabolism , Acetates/blood , Acetates/immunology , Animals , Anti-Inflammatory Agents/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Male , Mice , Mice, Inbred C57BL
6.
Immunity ; 50(1): 152-165.e8, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30611611

ABSTRACT

The ability of the immune system to discriminate self from non-self is essential for eradicating microbial pathogens but is also responsible for allograft rejection. Whether it is possible to selectively suppress alloresponses while maintaining anti-pathogen immunity remains unknown. We found that mice deficient in coronin 1, a regulator of naive T cell homeostasis, fully retained allografts while maintaining T cell-specific responses against microbial pathogens. Mechanistically, coronin 1-deficiency increased cyclic adenosine monophosphate (cAMP) concentrations to suppress allo-specific T cell responses. Costimulation induced on microbe-infected antigen presenting cells was able to overcome cAMP-mediated immunosuppression to maintain anti-pathogen immunity. In vivo pharmacological modulation of this pathway or a prior transfer of coronin 1-deficient T cells actively suppressed allograft rejection. These results define a coronin 1-dependent regulatory axis in T cells important for allograft rejection and suggest that modulation of this pathway may be a promising approach to achieve long-term acceptance of mismatched allografts.


Subject(s)
Graft Rejection/immunology , Heart Transplantation , Infections/immunology , Microfilament Proteins/metabolism , Skin Transplantation , T-Lymphocytes/immunology , Allografts/immunology , Animals , Antigens, Bacterial/immunology , Antigens, Fungal/immunology , Antigens, Viral/immunology , Cells, Cultured , Cyclic AMP/immunology , Graft Survival , Homeostasis/genetics , Humans , Immunity , Immunosuppression Therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Transplantation Tolerance
7.
Int J Pharm ; 550(1-2): 372-379, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30153487

ABSTRACT

Daptomycin (DAP) is a cyclic lipopeptide antibiotic with potential clinical application in orthopedic infections caused by staphylococci. However, it failed to eradicate Staphylococcus aureus in vitro, in intracellular infection studies, as well as in vivo in an experimental model of implant-associated biofilm infections. In this study, the antimicrobial effect of DAP encapsulated in poly(methyl methacrylate)-Eudragit (PMMA-EUD) microparticles (DAP-MPs) on intracellular S. aureus was evaluated in human osteoblast cells using fluorescence in situ hybridization (FISH) analysis. Encapsulated DAP was able to reduce the amount of intracellular S. aureus by 73% compared to blank microparticles (MPs). Then, the advantage of treating with DAP-MPs versus free DAP was evaluated in a murine model of implant-associated biofilm infection. Free DAP showed a >3 log10 decrease in planktonic and adherent bacteria but failed to eradicate adherent methicillin-resistant S. aureus (MRSA), whereas DAP-MPs showed a clearance of planktonic MRSA, significantly reduced adherent MRSA by more than 3 log10 and cured the infection in 60%. This was linked to the prolonged higher DAP concentration within the tissue cage fluid compared to free DAP. To our knowledge, this study provides the first evidence for the high intracellular and in vivo anti-biofilm efficacy of DAP-MPs to target staphylococcal infections.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Daptomycin/administration & dosage , Drug Carriers/administration & dosage , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Acrylic Resins/administration & dosage , Animals , Biofilms/drug effects , Cell Line , Female , Humans , Methicillin-Resistant Staphylococcus aureus/physiology , Mice, Inbred C57BL , Osteoblasts/microbiology , Polymers/administration & dosage
8.
Cell ; 174(2): 259-270.e11, 2018 07 12.
Article in English | MEDLINE | ID: mdl-29937224

ABSTRACT

Many community- and hospital-acquired bacterial infections are caused by antibiotic-resistant pathogens. Methicillin-resistant Staphylococcus aureus (MRSA) predisposes humans to invasive infections that are difficult to eradicate. We designed a closed-loop gene network programming mammalian cells to autonomously detect and eliminate bacterial infections. The genetic circuit contains human Toll-like receptors as the bacterial sensor and a synthetic promoter driving reversible and adjustable expression of lysostaphin, a bacteriolytic enzyme highly lethal to S. aureus. Immunomimetic designer cells harboring this genetic circuit exhibited fast and robust sense-and-destroy kinetics against live staphylococci. When tested in a foreign-body infection model in mice, microencapsulated cell implants prevented planktonic MRSA infection and reduced MRSA biofilm formation by 91%. Notably, this system achieved a 100% cure rate of acute MRSA infections, whereas conventional vancomycin treatment failed. These results suggest that immunomimetic designer cells could offer a therapeutic approach for early detection, prevention, and cure of pathogenic infections in the post-antibiotic era.


Subject(s)
Biomimetics/methods , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/prevention & control , Alkaline Phosphatase/blood , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Disk Diffusion Antimicrobial Tests , Female , HEK293 Cells , Humans , Lipopolysaccharide Receptors/genetics , Lysostaphin/metabolism , Lysostaphin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Mice, Inbred C57BL , Plasmids/genetics , Plasmids/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/veterinary , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Transcription Factor AP-1/metabolism
9.
Nat Commun ; 8(1): 2246, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269769

ABSTRACT

Lipoproteins (Lpp) of Gram-positive bacteria are major players in alerting our immune system. Here, we show that the TLR2 response induced by commensal species Staphylococcus aureus and Staphylococcus epidermidis is almost ten times lower than that induced by noncommensal Staphylococcus carnosus, and this is at least partially due to their different modifications of the Lpp lipid moieties. The N terminus of the lipid moiety is acylated with a long-chain fatty acid (C17) in S. aureus and S. epidermidis, while it is acylated with a short-chain fatty acid (C2) in S. carnosus. The long-chain N-acylated Lpp, recognized by TLR2-TLR1 receptors, silences innate and adaptive immune responses, while the short-chain N-acetylated Lpp, recognized by TLR2-TLR6 receptors, boosts it.


Subject(s)
Adaptation, Physiological/immunology , Bacterial Proteins/metabolism , Immunity, Innate/physiology , Lipids/chemistry , Lipoproteins/metabolism , Staphylococcus aureus/physiology , Staphylococcus epidermidis/physiology , Fatty Acids/metabolism , HEK293 Cells , Humans , Signal Transduction , Th1 Cells/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
10.
Nanomedicine ; 13(1): 11-22, 2017 01.
Article in English | MEDLINE | ID: mdl-27545103

ABSTRACT

The progression in the use of orthopedic implants has led to an increase in the absolute number of implant infections, triggering a search for more effective antibacterial coatings. Nanorattles have recently gained interest in biomedical applications such as drug delivery, as encapsulation of the cargo inside the hollow structure provides a physical protection from the surrounding environment. Here, silver-containing silica nanorattles (Ag@SiO2) were evaluated for their antimicrobial potential and for their impact on cells of the immune system. We show that Ag@SiO2 nanorattles exhibited a clear antibacterial effect against Escherichia coli as well as Staphylococcus aureus found in post-operative infections. Immunotoxicological analyses showed that the particles were taken up through an active phagocytic process by dendritic cells of the immune system and did not affect their viability nor induce unwanted immunological effects. Silver-containing silica nanorattles thus fulfill several prerequisites for an antibacterial coating on surgical implants.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dendritic Cells/drug effects , Metal Nanoparticles/chemistry , Silicon Dioxide/chemistry , Silver/pharmacology , Animals , Cells, Cultured , Dendritic Cells/metabolism , Escherichia coli/drug effects , Mice , Microbial Sensitivity Tests , Phagocytosis , Staphylococcus aureus/drug effects
11.
Antimicrob Agents Chemother ; 60(4): 2292-301, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26833157

ABSTRACT

Many serious bacterial infections are difficult to treat due to biofilm formation, which provides physical protection and induces a sessile phenotype refractory to antibiotic treatment compared to the planktonic state. A key structural component of biofilm is extracellular DNA, which is held in place by secreted bacterial proteins from the DNABII family: integration host factor (IHF) and histone-like (HU) proteins. A native human monoclonal antibody, TRL1068, has been discovered using single B-lymphocyte screening technology. It has low-picomolar affinity against DNABII homologs from important Gram-positive and Gram-negative bacterial pathogens. The disruption of established biofilm was observedin vitroat an antibody concentration of 1.2 µg/ml over 12 h. The effect of TRL1068in vivowas evaluated in a murine tissue cage infection model in which a biofilm is formed by infection with methicillin-resistantStaphylococcus aureus(MRSA; ATCC 43300). Treatment of the established biofilm by combination therapy of TRL1068 (15 mg/kg of body weight, intraperitoneal [i.p.] administration) with daptomycin (50 mg/kg, i.p.) significantly reduced adherent bacterial count compared to that after daptomycin treatment alone, accompanied by significant reduction in planktonic bacterial numbers. The quantification of TRL1068 in sample matrices showed substantial penetration of TRL1068 from serum into the cage interior. TRL1068 is a clinical candidate for combination treatment with standard-of-care antibiotics to overcome the drug-refractory state associated with biofilm formation, with potential utility for a broad spectrum of difficult-to-treat bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Biofilms/drug effects , Foreign Bodies/drug therapy , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Amino Acid Sequence , Animals , Anti-Bacterial Agents/biosynthesis , Anti-Bacterial Agents/isolation & purification , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibody Specificity , B-Lymphocytes/chemistry , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/growth & development , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Daptomycin/pharmacology , Disease Models, Animal , Drug Therapy, Combination , Epitope Mapping , Female , Foreign Bodies/microbiology , Gene Expression , Injections, Intraperitoneal , Integration Host Factors/antagonists & inhibitors , Integration Host Factors/genetics , Integration Host Factors/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , Plankton/drug effects , Plankton/genetics , Plankton/growth & development , Plankton/metabolism , Sequence Alignment , Single-Cell Analysis , Staphylococcal Infections/microbiology
12.
Antimicrob Agents Chemother ; 60(4): 2467-75, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26883700

ABSTRACT

Implant-associated infections (IAIs) are a dreaded complication mainly caused by biofilm-forming staphylococci. Implant surfaces preventing microbial colonization would be desirable. We examined the preventive effect of a silver-coated titanium-aluminum-niobium (TiAlNb) alloy. The surface elicited a strong, inoculum-dependent activity againstStaphylococcus epidermidisandStaphylococcus aureusin an agar inhibition assay. Gamma sterilization and alcohol disinfection did not alter the effect. In a tissue cage mouse model, silver coating of TiAlNb cages prevented perioperative infections in an inoculum-dependent manner and led to a 100% prevention rate after challenge with 2 × 10(6)CFU ofS. epidermidisper cage. InS. aureusinfections, silver coating had only limited effect. Similarly, daptomycin or vancomycin prophylaxis alone did not preventS. aureusinfections. However, silver coating combined with daptomycin or vancomycin prophylaxis thwarted methicillin-resistantS. aureusinfections at a prevention rate of 100% or 33%, respectively. Moreover, silver release from the surface was independent of infection and occurred rapidly after implantation. On day 2, a peak of 82 µg Ag/ml was reached in the cage fluid, corresponding to almost 6× the MIC of the staphylococci. Cytotoxicity toward leukocytes in the cage was low and temporary. Surrounding tissue did not reveal histological signs of silver toxicity.In vitro, no emergence of silver resistance was observed in several clinical strains of staphylococci upon serial subinhibitory silver exposures. In conclusion, our data demonstrate that silver-coated TiAlNb is potent for prevention of IAIs and thus can be considered for clinical application.


Subject(s)
Anti-Bacterial Agents/pharmacology , Coated Materials, Biocompatible/pharmacology , Dental Alloys/pharmacology , Prostheses and Implants/microbiology , Silver/pharmacology , Staphylococcal Infections/prevention & control , Animals , Anti-Bacterial Agents/pharmacokinetics , Cell Survival/drug effects , Daptomycin/pharmacology , Drug Liberation , Female , Kinetics , Leukocytes/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Mice , Mice, Inbred C57BL , Microbial Sensitivity Tests , Silver/pharmacokinetics , Skin , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/growth & development , Vancomycin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...