Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 20(6): e1012361, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941361

ABSTRACT

The interactions between a virus and its host vary in space and time and are affected by the presence of molecules that alter the physiology of either the host or the virus. Determining the molecular mechanisms at the basis of these interactions is paramount for predicting the fate of bacterial and phage populations and for designing rational phage-antibiotic therapies. We study the interactions between stationary phase Burkholderia thailandensis and the phage ΦBp-AMP1. Although heterogeneous genetic resistance to phage rapidly emerges in B. thailandensis, the presence of phage enhances the efficacy of three major antibiotic classes, the quinolones, the beta-lactams and the tetracyclines, but antagonizes tetrahydrofolate synthesis inhibitors. We discovered that enhanced antibiotic efficacy is facilitated by reduced antibiotic efflux in the presence of phage. This new phage-antibiotic therapy allows for eradication of stationary phase bacteria, whilst requiring reduced antibiotic concentrations, which is crucial for treating infections in sites where it is difficult to achieve high antibiotic concentrations.


Subject(s)
Anti-Bacterial Agents , Bacteriophages , Burkholderia , Anti-Bacterial Agents/pharmacology , Burkholderia/drug effects , Down-Regulation
2.
ISME Commun ; 3(1): 95, 2023 Sep 08.
Article in English | MEDLINE | ID: mdl-37684358

ABSTRACT

The interactions between bacteria and bacteriophage have important roles in the global ecosystem; in turn changes in environmental parameters affect the interactions between bacteria and phage. However, there is a lack of knowledge on whether clonal bacterial populations harbour different phenotypes that respond to phage in distinct ways and whether the abundance of such phenotypes within bacterial populations is affected by variations in environmental parameters. Here we study the impact of variations in nutrient availability, bacterial growth rate and phage abundance on the interactions between the phage T4 and individual Escherichia coli cells confined in spatial refuges. Surprisingly, we found that fast growing bacteria survive together with all of their clonal kin cells, whereas slow growing bacteria survive in isolation. We also discovered that the number of bacteria that survive in isolation decreases at increasing phage doses possibly due to lysis inhibition in the presence of secondary adsorptions. We further show that these changes in the phenotypic composition of the E. coli population have important consequences on the bacterial and phage population dynamics and should therefore be considered when investigating bacteria-phage interactions in ecological, health or food production settings in structured environments.

3.
PLoS Biol ; 21(4): e3002048, 2023 04.
Article in English | MEDLINE | ID: mdl-37014915

ABSTRACT

One of the deepest branches in the tree of life separates the Archaea from the Bacteria. These prokaryotic groups have distinct cellular systems including fundamentally different phospholipid membrane bilayers. This dichotomy has been termed the lipid divide and possibly bestows different biophysical and biochemical characteristics on each cell type. Classic experiments suggest that bacterial membranes (formed from lipids extracted from Escherichia coli, for example) show permeability to key metabolites comparable to archaeal membranes (formed from lipids extracted from Halobacterium salinarum), yet systematic analyses based on direct measurements of membrane permeability are absent. Here, we develop a new approach for assessing the membrane permeability of approximately 10 µm unilamellar vesicles, consisting of an aqueous medium enclosed by a single lipid bilayer. Comparing the permeability of 18 metabolites demonstrates that diether glycerol-1-phosphate lipids with methyl branches, often the most abundant membrane lipids of sampled archaea, are permeable to a wide range of compounds useful for core metabolic networks, including amino acids, sugars, and nucleobases. Permeability is significantly lower in diester glycerol-3-phosphate lipids without methyl branches, the common building block of bacterial membranes. To identify the membrane characteristics that determine permeability, we use this experimental platform to test a variety of lipid forms bearing a diversity of intermediate characteristics. We found that increased membrane permeability is dependent on both the methyl branches on the lipid tails and the ether bond between the tails and the head group, both of which are present on the archaeal phospholipids. These permeability differences must have had profound effects on the cell physiology and proteome evolution of early prokaryotic forms. To explore this further, we compare the abundance and distribution of transmembrane transporter-encoding protein families present on genomes sampled from across the prokaryotic tree of life. These data demonstrate that archaea tend to have a reduced repertoire of transporter gene families, consistent with increased membrane permeation. These results demonstrate that the lipid divide demarcates a clear difference in permeability function with implications for understanding some of the earliest transitions in cell origins and evolution.


Subject(s)
Archaea , Unilamellar Liposomes , Archaea/genetics , Unilamellar Liposomes/metabolism , Glycerol/metabolism , Cell Membrane/metabolism , Bacteria/metabolism , Membrane Lipids/metabolism , Phospholipids/metabolism , Phosphates/metabolism , Lipid Bilayers/analysis , Lipid Bilayers/metabolism
4.
Commun Biol ; 6(1): 409, 2023 04 14.
Article in English | MEDLINE | ID: mdl-37055536

ABSTRACT

Antimicrobial resistance is an urgent threat to human health, and new antibacterial drugs are desperately needed, as are research tools to aid in their discovery and development. Vancomycin is a glycopeptide antibiotic that is widely used for the treatment of Gram-positive infections, such as life-threatening systemic diseases caused by methicillin-resistant Staphylococcus aureus (MRSA). Here we demonstrate that modification of vancomycin by introduction of an azide substituent provides a versatile intermediate that can undergo copper-catalysed azide-alkyne cycloaddition (CuAAC) reaction with various alkynes to readily prepare vancomycin fluorescent probes. We describe the facile synthesis of three probes that retain similar antibacterial profiles to the parent vancomycin antibiotic. We demonstrate the versatility of these probes for the detection and visualisation of Gram-positive bacteria by a range of methods, including plate reader quantification, flow cytometry analysis, high-resolution microscopy imaging, and single cell microfluidics analysis. In parallel, we demonstrate their utility in measuring outer-membrane permeabilisation of Gram-negative bacteria. The probes are useful tools that may facilitate detection of infections and development of new antibiotics.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Vancomycin , Humans , Vancomycin/pharmacology , Fluorescent Dyes/pharmacology , Azides , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Gram-Positive Bacteria
5.
Elife ; 112022 06 07.
Article in English | MEDLINE | ID: mdl-35670099

ABSTRACT

Phenotypic variations between individual microbial cells play a key role in the resistance of microbial pathogens to pharmacotherapies. Nevertheless, little is known about cell individuality in antibiotic accumulation. Here, we hypothesise that phenotypic diversification can be driven by fundamental cell-to-cell differences in drug transport rates. To test this hypothesis, we employed microfluidics-based single-cell microscopy, libraries of fluorescent antibiotic probes and mathematical modelling. This approach allowed us to rapidly identify phenotypic variants that avoid antibiotic accumulation within populations of Escherichia coli, Pseudomonas aeruginosa, Burkholderia cenocepacia, and Staphylococcus aureus. Crucially, we found that fast growing phenotypic variants avoid macrolide accumulation and survive treatment without genetic mutations. These findings are in contrast with the current consensus that cellular dormancy and slow metabolism underlie bacterial survival to antibiotics. Our results also show that fast growing variants display significantly higher expression of ribosomal promoters before drug treatment compared to slow growing variants. Drug-free active ribosomes facilitate essential cellular processes in these fast-growing variants, including efflux that can reduce macrolide accumulation. We used this new knowledge to eradicate variants that displayed low antibiotic accumulation through the chemical manipulation of their outer membrane inspiring new avenues to overcome current antibiotic treatment failures.


Bacteria can cause an array of diseases ranging from mildly inconvenient to deadly. In fact, every year around the world, five million people succumb to a bacterial infection. Antibiotics can kill bacteria or stop their growth, but many bacterial species are now able to evade these drugs. To be efficient, most antibiotics first need to get inside a bacterium; there, they accumulate until they reach the concentration they need to act. Often, the drugs make their way through channel-like structures ('pores') studded through the external membranes of bacteria and which control the passage of molecules in and out of cells. Resistance usually emerges when genetic changes provide the microorganism with an advantage against antibiotics, or when the microorganism performs the biochemical reactions necessary for life at a slower pace. In contrast, Lapinska, Pagliara et al. decided to examine how genetically similar Escherichia coli bacteria which differed in their growth rate would fare against antibiotics. The drug targeted ribosomes, the machinery that produces proteins in a cell. A combination of techniques was used to follow individual cells, revealing that fast-growing variants better managed to survive. A closer look showed that bacteria which were growing quickly had a surplus of ribosomes, which then produced more pores that could pump the antibiotic out the cell. Next, Lapinska, Pagliara et al. exposed the bacteria to both the antibiotic and a compound that weakens bacterial membrane; this erased the advantage shown by the fast-growing variants. Overall, this work gives a finer understanding of the mechanisms that underlie antibiotic resistance, which could help pave the way to new strategies to combat harmful bacteria.


Subject(s)
Anti-Bacterial Agents , Escherichia coli Proteins , Anti-Bacterial Agents/metabolism , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Macrolides , Microbial Sensitivity Tests , Pseudomonas aeruginosa/metabolism
6.
Commun Biol ; 5(1): 385, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444215

ABSTRACT

The interaction between a cell and its environment shapes fundamental intracellular processes such as cellular metabolism. In most cases growth rate is treated as a proximal metric for understanding the cellular metabolic status. However, changes in growth rate might not reflect metabolic variations in individuals responding to environmental fluctuations. Here we use single-cell microfluidics-microscopy combined with transcriptomics, proteomics and mathematical modelling to quantify the accumulation of glucose within Escherichia coli cells. In contrast to the current consensus, we reveal that environmental conditions which are comparatively unfavourable for growth, where both nutrients and salinity are depleted, increase glucose accumulation rates in individual bacteria and population subsets. We find that these changes in metabolic function are underpinned by variations at the translational and posttranslational level but not at the transcriptional level and are not dictated by changes in cell size. The metabolic response-characteristics identified greatly advance our fundamental understanding of the interactions between bacteria and their environment and have important ramifications when investigating cellular processes where salinity plays an important role.


Subject(s)
Escherichia coli Proteins , Escherichia coli , Bacteria/metabolism , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Glucose/metabolism , Humans , Nutrients/metabolism
7.
Methods Enzymol ; 665: 1-28, 2022.
Article in English | MEDLINE | ID: mdl-35379430

ABSTRACT

Fluorescent probes are extensively applied as useful tools for imaging and determining dynamic processes in bacterial cells. In particular, antibiotic-derived fluorescent probes which can visualize the presence or the localization of antibiotics within bacteria through the monitoring of changes in fluorescence signal, are particularly useful. They form an emerging set of tools for studying the mode of action of their parent antibiotics and examining bacterial resistance and persistence, with the long-term goal of developing fresh approaches to the treatment of drug-resistant bacterial infections. In this chapter, we discuss the applications of antibiotic-based fluorescent probes to visualize bacteria, focusing on the techniques we have utilized to study their localization, penetration and efflux. We describe detailed protocols for analysis of bacteria using microscopy, flow cytometry, and plate reader-based methods based on these probes.


Subject(s)
Anti-Bacterial Agents , Fluorescent Dyes , Anti-Bacterial Agents/pharmacology , Bacteria , Microscopy, Fluorescence
8.
Cell Host Microbe ; 30(1): 31-40.e5, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34932986

ABSTRACT

Phages impose strong selection on bacteria to evolve resistance against viral predation. Bacteria can rapidly evolve phage resistance via receptor mutation or using their CRISPR-Cas adaptive immune systems. Acquisition of CRISPR immunity relies on the insertion of a phage-derived sequence into CRISPR arrays in the bacterial genome. Using Pseudomonas aeruginosa and its phage DMS3vir as a model, we demonstrate that conditions that reduce bacterial growth rates, such as exposure to bacteriostatic antibiotics (which inhibit cell growth without killing), promote the evolution of CRISPR immunity. We demonstrate that this is due to slower phage development under these conditions, which provides more time for cells to acquire phage-derived sequences and mount an immune response. Our data reveal that the speed of phage development is a key determinant of the evolution of CRISPR immunity and suggest that use of bacteriostatic antibiotics can trigger elevated levels of CRISPR immunity in human-associated and natural environments.


Subject(s)
Adaptive Immunity/genetics , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , CRISPR-Cas Systems/immunology , Bacteria/growth & development , Bacteria/immunology , Bacteriophages/genetics , Genome, Bacterial , Humans , Mutation , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/immunology
9.
Nat Commun ; 12(1): 6316, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728631

ABSTRACT

The Ff family of filamentous bacteriophages infect gram-negative bacteria, but do not cause lysis of their host cell. Instead, new virions are extruded via the phage-encoded pIV protein, which has homology with bacterial secretins. Here, we determine the structure of pIV from the f1 filamentous bacteriophage at 2.7 Å resolution by cryo-electron microscopy, the first near-atomic structure of a phage secretin. Fifteen f1 pIV subunits assemble to form a gated channel in the bacterial outer membrane, with associated soluble domains projecting into the periplasm. We model channel opening and propose a mechanism for phage egress. By single-cell microfluidics experiments, we demonstrate the potential for secretins such as pIV to be used as adjuvants to increase the uptake and efficacy of antibiotics in bacteria. Finally, we compare the f1 pIV structure to its homologues to reveal similarities and differences between phage and bacterial secretins.


Subject(s)
Cryoelectron Microscopy/methods , Inovirus/metabolism , Secretin/chemistry , Viral Nonstructural Proteins/chemistry , Amino Acid Sequence , Biological Transport , Protein Structural Elements , Sequence Alignment , Viral Nonstructural Proteins/metabolism
10.
PLoS Biol ; 19(10): e3001406, 2021 10.
Article in English | MEDLINE | ID: mdl-34637438

ABSTRACT

Bacteriophages represent an avenue to overcome the current antibiotic resistance crisis, but evolution of genetic resistance to phages remains a concern. In vitro, bacteria evolve genetic resistance, preventing phage adsorption or degrading phage DNA. In natural environments, evolved resistance is lower possibly because the spatial heterogeneity within biofilms, microcolonies, or wall populations favours phenotypic survival to lytic phages. However, it is also possible that the persistence of genetically sensitive bacteria is due to less efficient phage amplification in natural environments, the existence of refuges where bacteria can hide, and a reduced spread of resistant genotypes. Here, we monitor the interactions between individual planktonic bacteria in isolation in ephemeral refuges and bacteriophage by tracking the survival of individual cells. We find that in these transient spatial refuges, phenotypic resistance due to reduced expression of the phage receptor is a key determinant of bacterial survival. This survival strategy is in contrast with the emergence of genetic resistance in the absence of ephemeral refuges in well-mixed environments. Predictions generated via a mathematical modelling framework to track bacterial response to phages reveal that the presence of spatial refuges leads to fundamentally different population dynamics that should be considered in order to predict and manipulate the evolutionary and ecological dynamics of bacteria-phage interactions in naturally structured environments.


Subject(s)
Bacteriophages/physiology , Environment , Escherichia coli/virology , Computer Simulation , Phenotype , Receptors, Virus/metabolism
11.
ACS Infect Dis ; 7(6): 1848-1858, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34000805

ABSTRACT

Environmental and intracellular stresses can perturb protein homeostasis and trigger the formation and accumulation of protein aggregates. It has been recently suggested that the level of protein aggregates accumulated in bacteria correlates with the frequency of persister and viable but nonculturable cells that transiently survive treatment with multiple antibiotics. However, these findings have often been obtained employing fluorescent reporter strains. This enforced heterologous protein expression facilitates the visualization of protein aggregates but could also trigger the formation and accumulation of protein aggregates. Using microfluidics-based single-cell microscopy and a library of green fluorescent protein reporter strains, we show that heterologous protein expression favors the formation of protein aggregates. We found that persister and viable but nonculturable bacteria surviving treatment with antibiotics are more likely to contain protein aggregates and downregulate the expression of heterologous proteins. Our data also suggest that such aggregates are more basic with respect to the rest of the cell. These findings provide evidence for a strong link between heterologous protein expression, protein aggregation, intracellular pH, and phenotypic survival to antibiotics, suggesting that antibiotic treatments against persister and viable but nonculturable cells could be developed by modulating protein aggregation and pH regulation.


Subject(s)
Escherichia coli , Protein Aggregates , Anti-Bacterial Agents/pharmacology , Bacteria/genetics , Escherichia coli/genetics , Proteomics
12.
Analyst ; 145(7): 2751-2757, 2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32091040

ABSTRACT

A multiplexed biophotonic assay platform has been developed using the localised particle plasmon in gold nanoparticles assembled in an array and functionalised for two assays: total IgG and C-reactive protein (CRP). A protein A/G (PAG) assay, calibrated with a NIST reference material, shows a maximum surface coverage of θmax = 7.13 ± 0.19 mRIU, equivalent to 1.5 ng mm-2 of F(ab)-presenting antibody. The CRP capture antibody has an equivalent surface binding density of θmax = 2.95 ± 0.41 mRIU indicating a 41% capture antibody availability. Free PAG binding to the functionalised anti-CRP surface shows that only 47 ± 3% of CRP capture antibodies are correctly presenting Fab regions for antigen capture. The accuracy and precision of the CRP sensor assay was assessed with 54 blood samples containing spiked CRP in the range 2-160 mg L-1. The mean accuracy was 0.42 mg L-1 with Confidence Interval (CI) at 95% from -14.7 to 13.8 mg L-1 and the precision had a Coefficient of Variation (CV) of 10.6% with 95% CI 0.9%-20.2%. These biophotonic platform performance metrics indicate a CRP assay with 2-160 mg L-1 dynamic range, performed in 8 minutes from 5 µL of whole blood without sample preparation.


Subject(s)
C-Reactive Protein/analysis , Immunoassay/methods , Antibodies/immunology , Antigen-Antibody Reactions , Bacterial Proteins/metabolism , C-Reactive Protein/immunology , C-Reactive Protein/metabolism , Gold/chemistry , Humans , Kinetics , Metal Nanoparticles/chemistry , Reproducibility of Results , Staphylococcal Protein A/metabolism
13.
RSC Chem Biol ; 1(5): 395-404, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-34458770

ABSTRACT

The emerging crisis of antibiotic resistance requires a multi-pronged approach in order to avert the onset of a post-antibiotic age. Studies of antibiotic uptake and localisation in live cells may inform the design of improved drugs and help develop a better understanding of bacterial resistance and persistence. To facilitate this research, we have synthesised fluorescent derivatives of the macrolide antibiotic erythromycin. These analogues exhibit a similar spectrum of antibiotic activity to the parent drug and are capable of labelling both Gram-positive and -negative bacteria for microscopy. The probes localise intracellularly, with uptake in Gram-negative bacteria dependent on the level of efflux pump activity. A plate-based assay established to quantify bacterial labelling and localisation demonstrated that the probes were taken up by both susceptible and resistant bacteria. Significant intra-strain and -species differences were observed in these preliminary studies. In order to examine uptake in real-time, the probe was used in single-cell microfluidic microscopy, revealing previously unseen heterogeneity of uptake in populations of susceptible bacteria. These studies illustrate the potential of fluorescent macrolide probes to characterise and explore drug uptake and efflux in bacteria.

14.
ACS Nano ; 14(2): 1609-1622, 2020 02 25.
Article in English | MEDLINE | ID: mdl-31794180

ABSTRACT

Antimicrobial resistance stimulates the search for antimicrobial forms that may be less subject to acquired resistance. Here we report a conceptual design of protein pseudocapsids exhibiting a broad spectrum of antimicrobial activities. Unlike conventional antibiotics, these agents are effective against phenotypic bacterial variants, while clearing "superbugs" in vivo without toxicity. The design adopts an icosahedral architecture that is polymorphic in size, but not in shape, and that is available in both l and d epimeric forms. Using a combination of nanoscale and single-cell imaging we demonstrate that such pseudocapsids inflict rapid and irreparable damage to bacterial cells. In phospholipid membranes they rapidly convert into nanopores, which remain confined to the binding positions of individual pseudocapsids. This mechanism ensures precisely delivered influxes of high antimicrobial doses, rendering the design a versatile platform for engineering structurally diverse and functionally persistent antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Escherichia coli/drug effects , Protein Engineering , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Cell Survival/drug effects , Microbial Sensitivity Tests , Models, Molecular , Particle Size , Protein Folding , Surface Properties
15.
Philos Trans R Soc Lond B Biol Sci ; 374(1786): 20180442, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31587633

ABSTRACT

Evidence of ageing in the bacterium Escherichia coli was a landmark finding in senescence research, as it suggested that even organisms with morphologically symmetrical fission may have evolved strategies to permit damage accumulation. However, recent work has suggested that ageing is only detectable in this organism in the presence of extrinsic stressors, such as the fluorescent proteins and strong light sources typically used to excite them. Here we combine microfluidics with brightfield microscopy to provide evidence of ageing in E. coli in the absence of these stressors. We report (i) that the doubling time of the lineage of cells that consistently inherits the 'maternal old pole' progressively increases with successive rounds of cell division until it reaches an apparent asymptote, and (ii) that the parental cell divides asymmetrically, with the old pole daughter showing a longer doubling time and slower glucose accumulation than the new pole daughter. Notably, these patterns arise without the progressive accumulation or asymmetric partitioning of observable misfolded-protein aggregates, phenomena previously hypothesized to cause the ageing phenotype. Our findings suggest that ageing is part of the naturally occurring ecologically-relevant phenotype of this bacterium and highlight the importance of alternative mechanisms of damage accumulation in this context. This article is part of a discussion meeting issue 'Single cell ecology'.


Subject(s)
Escherichia coli/physiology , Aging
16.
Sci Adv ; 5(4): eaau3112, 2019 04.
Article in English | MEDLINE | ID: mdl-31001578

ABSTRACT

The aggregates of the Aß peptide associated with Alzheimer's disease are able to both grow in size as well as generate, through secondary nucleation, new small oligomeric species, that are major cytotoxins associated with neuronal death. Despite the importance of these amyloid fibril-dependent processes, their structural and molecular underpinnings have remained challenging to elucidate. Here, we consider two molecular chaperones: the Brichos domain, which suppresses specifically secondary nucleation processes, and clusterin which our results show is capable of inhibiting, specifically, the elongation of Aß fibrils at remarkably low substoichiometric ratios. Microfluidic diffusional sizing measurements demonstrate that this inhibition originates from interactions of clusterin with fibril ends with high affinity. Kinetic experiments in the presence of both molecular chaperones reveal that their inhibitory effects are additive and noncooperative, thereby indicating that the reactive sites associated with the formation of new aggregates and the growth of existing aggregates are distinct.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Protein Aggregates/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/genetics , Clusterin/metabolism , Humans , Kinetics , Microfluidics , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry
17.
J Phys Chem B ; 122(12): 3101-3112, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29488762

ABSTRACT

Protein self-assembly into amyloid fibrils or highly hierarchical superstructures is closely linked to neurodegenerative pathologies as Alzheimer's and Parkinson's diseases. Moreover, protein assemblies also emerged as building blocks for bioinspired nanostructured materials. In both the above mentioned fields, the main challenge is to control the growth and properties of the final protein structure. This relies on a more fundamental understanding of how interactions between proteins can determine structures and functions of biomolecular aggregates. Here, we identify a striking effect of the hydration of the single human insulin molecule and solvent properties in controlling hydrophobicity/hydrophilicity, structures, and morphologies of a superstructure named spherulite, observed in connection to Alzheimer's disease. Depending on the presence of ethanol, such structures can incorporate fluorescent molecules with different physicochemical features and span a range of mechanical properties and morphologies. A theoretical model providing a thorough comprehension of the experimental data is developed, highlighting a direct connection between the intimate physical protein-protein interactions, the growth, and the properties of the self-assembled superstructures. Our findings indicate structural variability as a general property for amyloid-like aggregates and not limited to fibrils. This knowledge is pivotal not only for developing effective strategies against pathological amyloids but also for providing a platform to design highly tunable biomaterials, alternative to elongated protein fibrils.


Subject(s)
Amyloid/chemical synthesis , Ethanol/chemistry , Insulins/chemical synthesis , Amyloid/chemistry , Circular Dichroism , Humans , Hydrophobic and Hydrophilic Interactions , Insulins/chemistry , Microscopy, Atomic Force , Microscopy, Confocal , Microscopy, Electron, Transmission , Neutron Diffraction , Optical Imaging , Scattering, Small Angle , Spectroscopy, Fourier Transform Infrared
18.
Lab Chip ; 18(1): 162-170, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29192926

ABSTRACT

The ability to apply highly controlled electric fields within microfluidic devices is valuable as a basis for preparative and analytical processes. A challenge encountered in the context of such approaches in conductive media, including aqueous buffers, is the generation of electrolysis products at the electrode/liquid interface which can lead to contamination, perturb fluid flows and generally interfere with the measurement process. Here, we address this challenge by designing a single layer microfluidic device architecture where the electric potential is applied outside and downstream of the microfluidic device while the field is propagated back to the chip via the use of a co-flowing highly conductive electrolyte solution that forms a stable interface at the separation region of the device. The co-flowing electrolyte ensures that all the generated electrolysis products, including Joule heat and gaseous products, are flowed away from the chip without coming into contact with the analytes while the single layer fabrication process where all the structures are defined lithographically allows producing the devices in a simple yet highly reproducible manner. We demonstrate that by allowing stable and effective application of electric fields in excess of 100 V cm-1, the described platform provides the basis for rapid separation of heterogeneous mixtures of proteins and protein complexes directly in their native buffers as well as for the simultaneous quantification of their charge states. We illustrate this by probing the interactions in a mixture of an amyloid forming protein, amyloid-ß, and a molecular chaperone, Brichos, known to inhibit the process of amyloid formation. The availability of a platform for applying stable electric fields and its compatibility with single-layer soft-lithography processes opens up the possibility of separating and analysing a wide range of molecules on chip, including those with similar electrophoretic mobilities.


Subject(s)
Electrolysis/instrumentation , Lab-On-A-Chip Devices , Microfluidic Analytical Techniques/instrumentation , Animals , Electrodes , Humans , Models, Chemical , Proteins/analysis , Proteins/chemistry , Proteins/isolation & purification
19.
Phys Chem Chem Phys ; 19(34): 23060-23067, 2017 Aug 30.
Article in English | MEDLINE | ID: mdl-28817152

ABSTRACT

The isoelectric point (pI) of a protein is a key characteristic that influences its overall electrostatic behaviour. The majority of conventional methods for the determination of the isoelectric point of a molecule rely on the use of spatial gradients in pH, although significant practical challenges are associated with such techniques, notably the difficulty in generating a stable and well controlled pH gradient. Here, we introduce a gradient-free approach, exploiting a microfluidic platform which allows us to perform rapid pH change on chip and probe the electrophoretic mobility of species in a controlled field. In particular, in this approach, the pH of the electrolyte solution is modulated in time rather than in space, as in the case for conventional determinations of the isoelectric point. To demonstrate the general approachability of this platform, we have measured the isoelectric points of representative set of seven proteins, bovine serum albumin, ß-lactoglobulin, ribonuclease A, ovalbumin, human transferrin, ubiquitin and myoglobin in microlitre sample volumes. The ability to conduct measurements in free solution thus provides the basis for the rapid determination of isoelectric points of proteins under a wide variety of solution conditions and in small volumes.


Subject(s)
Microfluidics/methods , Proteins/chemistry , Animals , Cattle , Electrophoresis , Humans , Hydrogen-Ion Concentration , Isoelectric Point , Lab-On-A-Chip Devices , Lactoglobulins/chemistry , Myoglobin/chemistry , Serum Albumin, Bovine/chemistry , Transferrin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...