Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Nat Commun ; 9(1): 1822, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29739943

ABSTRACT

Quorum sensing is a promising target for next-generation anti-infectives designed to address evolving bacterial drug resistance. The autoinducer-2 (AI-2) is a key quorum-sensing signal molecule which regulates bacterial group behaviors and is recognized by many Gram-negative and Gram-positive bacteria. Here we report a synthetic mammalian cell-based microbial-control device that detects microbial chemotactic formyl peptides through a formyl peptide sensor (FPS) and responds by releasing AI-2. The microbial-control device was designed by rewiring an artificial receptor-based signaling cascade to a modular biosynthetic AI-2 production platform. Mammalian cells equipped with the microbial-control gene circuit detect formyl peptides secreted from various microbes with high sensitivity and respond with robust AI-2 production, resulting in control of quorum sensing-related behavior of pathogenic Vibrio harveyi and attenuation of biofilm formation by the human pathogen Candida albicans. The ability to manipulate mixed microbial populations through fine-tuning of AI-2 levels may provide opportunities for future anti-infective strategies.


Subject(s)
Biofilms , Biosensing Techniques , Cell Engineering , Homoserine/analogs & derivatives , Lactones/metabolism , Quorum Sensing/genetics , Vibrio/physiology , Animals , Bacterial Proteins/genetics , Candida albicans/metabolism , Cell Line , Drug Resistance, Microbial , Genes, Bacterial , Homoserine/metabolism , Humans , Receptors, Formyl Peptide/metabolism , Reproducibility of Results , Signal Transduction , Synthetic Biology , Vibrio/genetics , Vibrio/pathogenicity
3.
Nat Biomed Eng ; 2(6): 399-415, 2018 06.
Article in English | MEDLINE | ID: mdl-31011195

ABSTRACT

In living organisms, naturally evolved sensors that constantly monitor and process environmental cues trigger corrective actions that enable the organisms to cope with changing conditions. Such natural processes have inspired biologists to construct synthetic living sensors and signalling pathways, by repurposing naturally occurring proteins and by designing molecular building blocks de novo, for customized diagnostics and therapeutics. In particular, designer cells that employ user-defined synthetic gene circuits to survey disease biomarkers and to autonomously re-adjust unbalanced pathological states can coordinate the production of therapeutics, with controlled timing and dosage. Furthermore, tailored genetic networks operating in bacterial or human cells have led to cancer remission in experimental animal models, owing to the network's unprecedented specificity. Other applications of designer cells in infectious, metabolic and autoimmune diseases are also being explored. In this Review, we describe the biomedical applications of synthetic gene circuits in major disease areas, and discuss how the first genetically engineered devices developed on the basis of synthetic-biology principles made the leap from the laboratory to the clinic.


Subject(s)
Communicable Diseases , Gene Regulatory Networks/genetics , Genes, Synthetic/genetics , Synthetic Biology , Theranostic Nanomedicine , Animals , Communicable Disease Control , Communicable Diseases/diagnosis , Communicable Diseases/therapy , Genetic Engineering , Humans , Mice
4.
Nano Lett ; 17(8): 5043-5050, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28703595

ABSTRACT

Current antibiotics gradually lose their efficacy against chronic Pseudomonas aeruginosa infections due to development of increased resistance mediated by biofilm formation, as well as the large arsenal of microbial virulence factors that are coordinated by the cell density-dependent phenomenon of quorum sensing. Here, we address this issue by using synthetic biology principles to rationally engineer quorum-quencher cells with closed-loop control to autonomously dampen virulence and interfere with biofilm integrity. Pathogen-derived signals dynamically activate a synthetic mammalian autoinducer sensor driving downstream expression of next-generation anti-infectives. Engineered cells were able to sensitively score autoinducer levels from P. aeruginosa clinical isolates and mount a 2-fold defense consisting of an autoinducer-inactivating enzyme to silence bacterial quorum sensing and a bipartite antibiofilm effector to dissolve the biofilm matrix. The self-guided cellular device fully cleared autoinducers, potentiated bacterial antibiotic susceptibility, substantially reduced biofilms, and alleviated cytotoxicity to lung epithelial cells. We believe this strategy of dividing otherwise coordinated pathogens and breaking up their shielded stronghold represents a blueprint for cellular anti-infectives in the postantibiotic era.


Subject(s)
Biofilms , Homoserine/analogs & derivatives , Lactones/metabolism , Pseudomonas aeruginosa/metabolism , Quorum Sensing , A549 Cells , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Biofilms/drug effects , Cell Culture Techniques , Cell Survival , DNA/genetics , Drug Resistance, Bacterial , Genetic Vectors , HEK293 Cells , Herpes Simplex Virus Protein Vmw65/genetics , Homoserine/metabolism , Humans , Nuclear Localization Signals , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synthetic Biology , Tobramycin/chemistry , Tobramycin/pharmacology , Trans-Activators/genetics , Virulence , Virulence Factors/biosynthesis
5.
Mol Pharm ; 12(5): 1431-42, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25811325

ABSTRACT

Leptin plays a central role in the control of energy homeostasis and appetite and, thus, has attracted attention for therapeutic approaches in spite of its limited pharmacological activity owing to the very short circulation in the body. To improve drug delivery and prolong plasma half-life, we have fused murine leptin with Pro/Ala/Ser (PAS) polypeptides of up to 600 residues, which adopt random coil conformation with expanded hydrodynamic volume in solution and, consequently, retard kidney filtration in a similar manner as polyethylene glycol (PEG). Relative to unmodified leptin, size exclusion chromatography and dynamic light scattering revealed an approximately 21-fold increase in apparent size and a much larger molecular diameter of around 18 nm for PAS(600)-leptin. High receptor-binding activity for all PASylated leptin versions was confirmed in BIAcore measurements and cell-based dual-luciferase assays. Pharmacokinetic studies in mice revealed a much extended plasma half-life after ip injection, from 26 min for the unmodified leptin to 19.6 h for the PAS(600) fusion. In vivo activity was investigated after single ip injection of equimolar doses of each leptin version. Strongly increased and prolonged hypothalamic STAT3 phosphorylation was detected for PAS(600)-leptin. Also, a reduction in daily food intake by up to 60% as well as loss in body weight of >10% lasting for >5 days was observed, whereas unmodified leptin was merely effective for 1 day. Notably, application of a PASylated superactive mouse leptin antagonist (SMLA) led to the opposite effects. Thus, PASylated leptin not only provides a promising reagent to study its physiological role in vivo but also may offer a superior drug candidate for clinical therapy.


Subject(s)
Leptin/blood , Leptin/pharmacokinetics , Adipokines/metabolism , Animals , Chromatography, Gel , Circular Dichroism , Dynamic Light Scattering , Enzyme-Linked Immunosorbent Assay , Female , HEK293 Cells , Humans , Leptin/chemistry , Male , Mice , Phosphorylation/drug effects , Polyethylene Glycols/chemistry , STAT3 Transcription Factor/metabolism , Surface Plasmon Resonance
6.
Mol Cell ; 55(3): 397-408, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25018017

ABSTRACT

All metabolic activities operate within a narrow pH range that is controlled by the CO2-bicarbonate buffering system. We hypothesized that pH could serve as surrogate signal to monitor and respond to the physiological state. By functionally rewiring the human proton-activated cell-surface receptor TDAG8 to chimeric promoters, we created a synthetic signaling cascade that precisely monitors extracellular pH within the physiological range. The synthetic pH sensor could be adjusted by organic acids as well as gaseous CO2 that shifts the CO2-bicarbonate balance toward hydrogen ions. This enabled the design of gas-programmable logic gates, provided remote control of cellular behavior inside microfluidic devices, and allowed for CO2-triggered production of biopharmaceuticals in standard bioreactors. When implanting cells containing the synthetic pH sensor linked to production of insulin into type 1 diabetic mice developing diabetic ketoacidosis, the prosthetic network automatically scored acidic pH and coordinated an insulin expression response that corrected ketoacidosis.


Subject(s)
Carbon Dioxide/metabolism , Diabetic Ketoacidosis/physiopathology , Microfluidic Analytical Techniques/methods , Receptors, G-Protein-Coupled/genetics , Synthetic Biology/methods , Animals , CHO Cells , Cell Line , Cell Transplantation , Cricetulus , Diabetic Ketoacidosis/therapy , Disease Models, Animal , Female , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Mice , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...