Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Phys Chem B ; 128(16): 3885-3897, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38622775

ABSTRACT

Very few drugs have the necessary physicochemical properties to cross the skin's main permeability barrier, the stratum corneum (SC), in sufficient amounts. Propylene glycol (PG) is a chemical penetration enhancer that could be included in topical formulations in order to overcome the barrier properties of the skin and facilitate the transport of drugs across it. Experiments have demonstrated that PG increases the mobility and disorder of SC lipids and may extract cholesterol from the SC, but little is known about the molecular mechanisms of drug permeation enhancement by PG. In this work, we have performed molecular dynamics (MD) simulations to investigate the molecular-level effects of PG on the structure and properties of model SC lipid bilayers. The model bilayers were simulated in the presence of PG concentrations over the range of 0-100% w/w PG, using both an all-atom and a united atom force field. PG was found to localize in the hydrophilic headgroup regions at the bilayer interface, to occupy the lipid-water hydrogen-bonding sites, and to slightly increase lipid tail disorder in a concentration-dependent manner. We showed with MD simulation that PG enhances the permeation of small molecules such as water by interacting with the bilayer interface; the results of our study may be used to guide the design of formulations for transdermal drug delivery with enhanced skin permeation, as well as topical formulations and cosmetic products.


Subject(s)
Lipid Bilayers , Molecular Dynamics Simulation , Propylene Glycol , Skin , Propylene Glycol/chemistry , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Skin/metabolism , Skin/chemistry , Hydrogen Bonding , Skin Absorption/drug effects
2.
Chemistry ; 30(28): e202400308, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38488326

ABSTRACT

Cyclic peptides are increasingly important structures in drugs but their development can be impeded by difficulties associated with their synthesis. Here, we introduce the 3-aminoazetidine (3-AAz) subunit as a new turn-inducing element for the efficient synthesis of small head-to-tail cyclic peptides. Greatly improved cyclizations of tetra-, penta- and hexapeptides (28 examples) under standard reaction conditions are achieved by introduction of this element within the linear peptide precursor. Post-cyclization deprotection of the amino acid side chains with strong acid is realized without degradation of the strained four-membered azetidine. A special feature of this chemistry is that further late-stage modification of the resultant macrocyclic peptides can be achieved via the 3-AAz unit. This is done by: (i) chemoselective deprotection and substitution at the azetidine nitrogen, or by (ii) a click-based approach employing a 2-propynyl carbamate on the azetidine nitrogen. In this way, a range of dye and biotin tagged macrocycles are readily produced. Structural insights gained by XRD analysis of a cyclic tetrapeptide indicate that the azetidine ring encourages access to the less stable, all-trans conformation. Moreover, introduction of a 3-AAz into a representative cyclohexapeptide improves stability towards proteases compared to the homodetic macrocycle.


Subject(s)
Azetidines , Peptides, Cyclic , Azetidines/chemistry , Azetidines/chemical synthesis , Cyclization , Peptides, Cyclic/chemistry , Peptides, Cyclic/chemical synthesis , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/chemical synthesis , Click Chemistry
3.
Phys Chem Chem Phys ; 22(43): 25075-25083, 2020 Nov 21.
Article in English | MEDLINE | ID: mdl-33118559

ABSTRACT

Peptide-based drugs combine advantages of larger biological therapeutics with those of small molecule drugs, but they generally display poor permeability and metabolic stability. Recently, we introduced a new type of peptide bond isostere, in which the backbone carbonyl is replaced with a 3-amino oxetane heterocycle, into short linear peptides with the aim of improving their therapeutic potential. In this study, we have explored the impact of oxetane modification on α-helical peptides to establish whether or not this modification is tolerated in this biologically important structural motif. The oxetane modification was introduced at two positions in a well-characterised helical peptide sequence, and circular dichroism and NMR spectroscopy were used to measure the resulting secondary structure content under different experimental conditions. Our data demonstrated that introduction of an oxetane into the peptide backbone results in a significant loss of helicity, regardless of where in the sequence the modification is placed. The molecular determinants of this destabilisation were then explored using steered molecular dynamics simulations, a computational method analogous to single molecule spectroscopy. Our simulations indicated that oxetane modification introduces a kink in the helical axis, alters the dihedral angles of residues up to three positions away from the modification, and disrupts the (i, i + 4) hydrogen bonding pattern characteristic of α-helices in favour of new, short-range hydrogen bonds. The detailed structural understanding provided in this work can direct future design of chemically modified peptides.


Subject(s)
Ethers, Cyclic/chemistry , Ethers, Cyclic/metabolism , Peptides/chemistry , Models, Molecular , Protein Conformation, alpha-Helical , Protein Stability , Protein Structure, Secondary
4.
J Chem Phys ; 153(3): 034115, 2020 Jul 21.
Article in English | MEDLINE | ID: mdl-32716163

ABSTRACT

Understanding the permeation of molecules through lipid membranes is fundamental for predicting the cellular uptake of solutes and drug delivery mechanisms. In molecular simulations, the usual approach is to compute the free energy (FE) profile of a molecule across a model lipid bilayer, which can then be used to estimate the permeability of the molecule. Umbrella Sampling (US), which involves carrying out a series of biased simulations along a defined reaction coordinate (usually the bilayer normal direction), is a popular method for the computation of such FE profiles. However, US can be challenging to implement because the results are dependent on the strength of the biasing potential and the spacing of windows along the reaction coordinate, which, in practice, are usually optimized by an inefficient trial and error approach. The Steered Molecular Dynamics implementation of the Jarzynski Equality (JE-SMD) has been identified as an alternative to equilibrium sampling methods for measuring the FE change across a reaction coordinate. In the JE-SMD approach, equilibrium FE values are evaluated from the average of rapid non-equilibrium trajectories, thus avoiding the practical issues that come with US. Here, we use three different corrections of the JE-SMD method to calculate the FE change for the translocation of two aromatic substrates, phenylalanine and toluene, across a lipid bilayer and compare the accuracy and computational efficiency of these approaches to the results obtained using US. We show evidence that when computing the free energy profile, the JE-SMD approach suffers from insufficient sampling convergence of the bilayer environment and is dependent on the characteristic of the aromatic substrate itself. We deduce that, despite its drawbacks, US remains the more viable approach of the two for computing the FE profile.


Subject(s)
Lipid Bilayers/chemistry , Molecular Dynamics Simulation , Phospholipids/chemistry , Molecular Conformation , Phenylalanine/chemistry , Thermodynamics , Toluene/chemistry
5.
Nanoscale ; 12(8): 4868-4881, 2020 Feb 27.
Article in English | MEDLINE | ID: mdl-31916561

ABSTRACT

The unique and adjustable properties of nanoparticles present enormous opportunities for their use as targeted drug delivery vectors. For example, nanoparticles functionalized with key surface ligands have been shown to pass through phospholipid bilayers without causing localised disruption. However, the further effects nanoparticles have on multi-component phospholipid bilayers remain unclear. We use coarse-grained computational models to investigate the structural properties of mixed phospholipid bilayers in the presence of ligand-functionalized nanoparticles. Model bilayers are composed of DPPC, DUPC, DFPC and cholesterol, and the nanoparticles are striped with a hydrophobic-ligand band and charged-ligand spherical caps. Our results show that nanoparticles aggregate near unsaturated phospholipid regions, phospholipid bilayer phase-separation is promoted in the presence of nanoparticles, and the heterogeneous components of a phospholipid bilayer play a significant role in the lateral organization of nanoparticles. This study highlights the need for considering the complexity of realistic phospholipid bilayers when optimising ligand functionalized nanoparticles for efficient drug delivery vectors.


Subject(s)
Lipid Bilayers/chemistry , Models, Chemical , Molecular Dynamics Simulation , Nanoparticles/chemistry , Phospholipids/chemistry , Hydrophobic and Hydrophilic Interactions
6.
Chem Sci ; 10(8): 2465-2472, 2019 Feb 28.
Article in English | MEDLINE | ID: mdl-30881675

ABSTRACT

Cyclic peptides are an important source of new drugs but are challenging to produce synthetically. We show that head-to-tail peptide macrocyclisations are greatly improved, as measured by isolated yields, reaction rates and product distribution, by substitution of one of the backbone amide C[double bond, length as m-dash]O bonds with an oxetane ring. The cyclisation precursors are easily made by standard solution- or solid-phase peptide synthesis techniques. Macrocyclisations across a range of challenging ring sizes (tetra-, penta- and hexapeptides) are enabled by incorporation of this turn-inducing element. Oxetane incorporation is shown to be superior to other established amino acid modifications such as N-methylation. The positional dependence of the modification on cyclisation efficiency is mapped using a cyclic peptide of sequence LAGAY. We provide the first direct experimental evidence that oxetane modification induces a turn in linear peptide backbones, through the observation of d NN (i, i + 2) and d αN (i, i + 2) NOEs, which offers an explanation for these improvements. For cyclic peptide, cLAGAY, a combination of NMR derived distance restraints and molecular dynamics simulations are used to show that this modification alters the backbone conformation in proximity to the oxetane, with the flexibility of the ring reduced and a new intramolecular H-bond established. Finally, we incorporated an oxetane into a cyclic pentapeptide inhibitor of Aminopeptidase N, a transmembrane metalloprotease overexpressed on the surface of cancer cells. The inhibitor, cCNGRC, displayed similar IC50 values in the presence or absence of an oxetane at the glycine residue, indicating that bioactivity is fully retained upon amide C[double bond, length as m-dash]O bond replacement.

7.
Phys Chem Chem Phys ; 20(4): 2162-2174, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29116267

ABSTRACT

An understanding of how molecules permeate the complex lipid matrix of the stratum corneum (SC) skin barrier is important for transdermal drug delivery, preventing the adsorption of toxic chemicals and tackling skin diseases. In this paper we present atomistic molecular dynamics simulations of skin-lipid bilayers composed of ceramides, cholesterol (CHOL) and free fatty acids at different lipid compositions and levels of hydration and investigate both perpendicular and lateral permeation pathways through the systems. We show that in fully hydrated bilayers the lipids are heterogeneously distributed, with CHOL-rich domains emerging spontaneously during the simulations. Potential of mean constraint force calculations reveal that the most favourable permeation pathway for water in the direction normal to the bilayer is through a CHOL-rich region, probably due to the disordering effect of CHOL on lipids in the gel-phase. In systems with a low water content (akin to real skin) we find that rather than forming continuous layers, water forms flattened ellipsoid-shaped pools between the lipid headgroups, which are separated by dry regions. This implies that there is no continuous aqueous lateral pathway in the SC and may help to explain why skin is such an effective barrier. We propose that the most probable permeation pathway for a small polar molecule consists of hopping from the headgroup region of one bilayer to the next via a dry region, followed by permeation along the bilayer normal through a CHOL-rich region to the centre of the bilayer where it can diffuse laterally in the lower-density lipidic environment before encountering another CHOL-rich region through which it can exit the bilayer.


Subject(s)
Lipid Bilayers/chemistry , Skin/chemistry , Ceramides/chemistry , Cholesterol/chemistry , Humans , Hydrogen Bonding , Lipid Bilayers/metabolism , Molecular Dynamics Simulation , Permeability , Skin/metabolism , Water/chemistry
8.
Pharm Nanotechnol ; 5(3): 215-219, 2017.
Article in English | MEDLINE | ID: mdl-28847269

ABSTRACT

BACKGROUND: Conventional nanofiber forming peptide amphiphiles comprise a beta sheet forming, short peptide sequence with an alkyl chain attached at one terminus. We report the selfassembly of a peptide amphiphile possessing a mid-chain located alkyl substituent (a T-shaped peptide amphiphile) into nanofiber networks. METHOD: Peptide synthesis was carried out using standard 9-fluorenylmethoxycarbonyl solid phase peptide synthesis protocols, followed by covalent attachment of the alkyl chains to yield target peptide amphiphiles. Self-assembly was then studied using electron microscopy and coarse-grained molecular dynamics simulations. RESULTS: T-shaped peptide amphiphiles self-assembled into nanofibers just like linear peptide amphiphiles, but then unlike linear peptide amphiphiles, T-shaped peptide amphiphiles formed inter-fiber associations and ultimately nanofiber networks. CONCLUSION: Changing the position of the alkyl chain in a peptide amphiphile from the terminal end of the peptide to the middle part of the peptide, to form a T-shaped peptide amphiphile, does not disrupt the molecular interactions required for the self-assembly of the peptide amphiphiles into nanofibers.


Subject(s)
Nanofibers/chemistry , Peptides/chemistry , Surface-Active Agents/chemistry , Drug Delivery Systems/methods , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Nanostructures/chemistry , Particle Size , Protein Conformation , Protein Multimerization
9.
Biomacromolecules ; 17(9): 3033-9, 2016 09 12.
Article in English | MEDLINE | ID: mdl-27476873

ABSTRACT

Antifreeze (glyco) proteins are produced by many cold-acclimatized species to enable them to survive subzero temperatures. These proteins have multiple macroscopic effects on ice crystal growth which makes them appealing for low-temperature applications-from cellular cryopreservation to food storage. Poly(vinyl alcohol) has remarkable ice recrystallization inhibition activity, but its mode of action is uncertain as is the extent at which it can be incorporated into other high-order structures. Here the synthesis and characterization of well-defined block copolymers containing poly(vinyl alcohol) and poly(vinylpyrrolidone) by RAFT/MADIX polymerization is reported, as new antifreeze protein mimetics. The effect of adding a large second hydrophilic block is studied across a range of compositions, and it is found to be a passive component in ice recrystallization inhibition assays, enabling retention of all activity. In the extreme case, a block copolymer with only 10% poly(vinyl alcohol) was found to retain all activity, where statistical copolymers of PVA lose all activity with very minor changes to composition. These findings present a new method to increase the complexity of antifreeze protein mimetic materials, while retaining activity, and also to help understand the underlying mechanisms of action.


Subject(s)
Antifreeze Proteins/chemistry , Biomimetic Materials/chemistry , Cryoprotective Agents/chemistry , Ice , Polyvinyl Alcohol/chemistry , Crystallization , Hydrophobic and Hydrophilic Interactions , Polymerization
10.
Biomacromolecules ; 16(9): 2820-6, 2015 Sep 14.
Article in English | MEDLINE | ID: mdl-26258729

ABSTRACT

Nature has evolved many elegant solutions to enable life to flourish at low temperatures by either allowing (tolerance) or preventing (avoidance) ice formation. These processes are typically controlled by ice nucleating proteins or antifreeze proteins, which act to either promote nucleation, prevent nucleation or inhibit ice growth depending on the specific need, respectively. These proteins can be expensive and their mechanisms of action are not understood, limiting their translation, especially into biomedical cryopreservation applications. Here well-defined poly(vinyl alcohol), synthesized by RAFT/MADIX polymerization, is investigated for its ice nucleation inhibition (INI) activity, in contrast to its established ice growth inhibitory properties and compared to other synthetic polymers. It is shown that ice nucleation inhibition activity of PVA has a strong molecular weight dependence; polymers with a degree of polymerization below 200 being an effective inhibitor at just 1 mg.mL(-1). Other synthetic and natural polymers, both with and without hydroxyl-functional side chains, showed negligible activity, highlighting the unique ice/water interacting properties of PVA. These findings both aid our understanding of ice nucleation but demonstrate the potential of engineering synthetic polymers as new biomimetics to control ice formation/growth processes.


Subject(s)
Biomimetic Materials/chemistry , Ice , Polyvinyl Alcohol/chemistry , Biomimetic Materials/chemical synthesis , Polyvinyl Alcohol/chemical synthesis
11.
PLoS One ; 10(7): e0132706, 2015.
Article in English | MEDLINE | ID: mdl-26181054

ABSTRACT

Keratins are cytoskeletal proteins that hierarchically arrange into filaments, starting with the dimer sub-unit. They are integral to the structural support of cells, in skin, hair and nails. In skin, keratin is thought to play a critical role in conferring the barrier properties and elasticity of skin. In general, the keratin dimer is broadly described by a tri-domain structure: a head, a central rod and a tail. As yet, no atomistic-scale picture of the entire dimer structure exists; this information is pivotal for establishing molecular-level connections between structure and function in intermediate filament proteins. The roles of the head and tail domains in facilitating keratin filament assembly and function remain as open questions. To address these, we report results of molecular dynamics simulations of the entire epithelial human K1/K10 keratin dimer. Our findings comprise: (1) the first three-dimensional structural models of the complete dimer unit, comprising of the head, rod and tail domains; (2) new insights into the chirality of the rod-domain twist gained from analysis of the full domain structure; (3) evidence for tri-subdomain partitioning in the head and tail domains; and, (4) identification of the residue characteristics that mediate non-covalent contact between the chains in the dimer. Our findings are immediately applicable to other epithelial keratins, such as K8/K18 and K5/K14, and to intermediate filament proteins in general.


Subject(s)
Disulfides/chemistry , Intermediate Filaments/chemistry , Keratin-10/chemistry , Keratin-1/chemistry , Protein Multimerization , Amino Acid Sequence , Humans , Hydrogen Bonding , Molecular Dynamics Simulation , Molecular Sequence Data , Protein Folding , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Alignment
12.
Biochim Biophys Acta ; 1848(5): 1248-57, 2015 May.
Article in English | MEDLINE | ID: mdl-25732028

ABSTRACT

Membrane proteins regulate a large number of cellular functions, and have great potential as tools for manipulation of biological systems. Developing these tools requires a robust and quantitative understanding of membrane protein folding and interactions within the bilayer. With this in mind, we have designed a series of proteins to probe the net thermodynamic contribution of well-known sequence motifs to transmembrane helix-helix association in a biological membrane. The proteins were designed from first principles (de novo) using current knowledge about membrane insertion and stabilizing interaction motifs. A simple poly-Leu "scaffold" was decorated with individual helix interaction motifs (G-XXX-G, polar residues, heptad repeat) to create transmembrane helix-helix interactions of increasing strength. The GALLEX assay, an in vivo assay for measurement of transmembrane helix self-association, was combined with computational methods to characterize the relative strength and mode of interaction for each sequence. In addition, the apparent free energy contribution (ΔΔGapp) of each motif to transmembrane helix self-association was measured in a biological membrane, results that are the first of their kind for these de novo designed sequences, and suggest that the free energy barrier to overcoming weak association is quite small (<1.4 kcal mol(-1)) in a natural membrane. By quantifying and rationalizing the contribution of key motifs to transmembrane helix association, our work offers a route to direct the design of novel sequences for use in biotechnology or synthetic biology (e.g. molecular switches) and to predict the effects of sequence modification in known transmembrane domains (for control of cellular processes).


Subject(s)
Computer Simulation , Computer-Aided Design , Escherichia coli Proteins/metabolism , Escherichia coli/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Amino Acid Motifs , Escherichia coli/chemistry , Escherichia coli Proteins/chemistry , Intracellular Membranes/chemistry , Membrane Proteins/chemistry , Protein Folding , Protein Stability , Protein Structure, Secondary , Structure-Activity Relationship , Thermodynamics
13.
Chem Commun (Camb) ; 50(63): 8797-800, 2014 Aug 14.
Article in English | MEDLINE | ID: mdl-24968282

ABSTRACT

A new class of peptidomimetic is reported in which one of the amide C=O bonds of the peptide backbone is replaced by an oxetane ring. They are synthesised by conjugate addition of various α-amino esters to a 3-(nitromethylene)oxetane, reduction of the nitro group and further coupling with N-Z protected amino acids to grow the peptide chain. Structural insights are provided by X-ray diffraction and molecular dynamics simulations.


Subject(s)
Ethers, Cyclic/chemistry , Oligopeptides/chemistry , Amides/chemistry , Amino Acids/chemistry , Models, Molecular , Molecular Conformation , X-Ray Diffraction
14.
Biomacromolecules ; 14(5): 1578-86, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23534826

ABSTRACT

This manuscript reports a detailed study on the ability of poly(vinyl alcohol) to act as a biomimetic surrogate for antifreeze(glyco)proteins, with a focus on the specific property of ice-recrystallization inhibition (IRI). Despite over 40 years of study, the underlying mechanisms that govern the action of biological antifreezes are still poorly understood, which is in part due to their limited availability and challenging synthesis. Poly(vinyl alcohol) (PVA) has been shown to display remarkable ice recrystallization inhibition activity despite its major structural differences to native antifreeze proteins. Here, controlled radical polymerization is used to synthesize well-defined PVA, which has enabled us to obtain the first quantitative structure-activity relationships, to probe the role of molecular weight and comonomers on IRI activity. Crucially, it was found that IRI activity is "switched on" when the polymer chain length increases from 10 and 20 repeat units. Substitution of the polymer side chains with hydrophilic or hydrophobic units was found to diminish activity. Hydrophobic modifications to the backbone were slightly more tolerated than side chain modifications, which implies an unbroken sequence of hydroxyl units is necessary for activity. These results highlight that, although hydrophobic domains are key components of IRI activity, the random inclusion of addition hydrophobic units does not guarantee an increase in activity and that the actual polymer conformation is important.


Subject(s)
Biomimetic Materials/chemistry , Cryoprotective Agents/chemistry , Polyvinyl Alcohol/chemistry , Antifreeze Proteins/chemistry , Crystallization , Glycoproteins/chemistry , Hydrophobic and Hydrophilic Interactions , Ice , Molecular Conformation , Molecular Weight , Polymerization , Quantitative Structure-Activity Relationship
15.
ACS Nano ; 7(2): 1016-26, 2013 Feb 26.
Article in English | MEDLINE | ID: mdl-23289352

ABSTRACT

The delivery of therapeutic peptides and proteins to the central nervous system is the biggest challenge when developing effective neuropharmaceuticals. The central issue is that the blood-brain barrier is impermeable to most molecules. Here we demonstrate the concept of employing an amphiphilic derivative of a peptide to deliver the peptide into the brain. The key to success is that the amphiphilic peptide should by design self-assemble into nanofibers wherein the active peptide epitope is tightly wrapped around the nanofiber core. The nanofiber form appears to protect the amphiphilic peptide from degradation while in the plasma, and the amphiphilic nature of the peptide promotes its transport across the blood-brain barrier. Therapeutic brain levels of the amphiphilic peptide are achieved with this strategy, compared with the absence of detectable peptide in the brain and the consequent lack of a therapeutic response when the underivatized peptide is administered.


Subject(s)
Brain/metabolism , Drug Carriers/chemistry , Enkephalin, Leucine-2-Alanine/analogs & derivatives , Nanofibers/chemistry , Peptides/chemistry , Blood-Brain Barrier/metabolism , Drug Carriers/metabolism , Enkephalin, Leucine-2-Alanine/metabolism , Enkephalin, Leucine-2-Alanine/therapeutic use , Models, Molecular , Nanomedicine , Peptides/metabolism , Protein Conformation
16.
Adv Drug Deliv Rev ; 65(2): 237-50, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22414344

ABSTRACT

Breaching the skin's barrier function by design is an important strategy for delivering drugs and vaccines to the body. However, while there are many proposed approaches for reversibly breaching the skin barrier, our understanding of the molecular processes involved is still rudimentary. Molecular simulation offers an unprecedented molecular-level resolution with an ability to reproduce molecular and bulk level properties. We review the basis of the molecular simulation methodology and give applications of relevance to the skin lipid barrier, focusing on permeation of molecules and chemical approaches for breaching the lipid barrier by design. The bulk kinetic model based on Fick's Law describing absorption of a drug through skin has been reconciled with statistical mechanical quantities such as the local excess chemical potential and local diffusion coefficient within the membrane structure. Applications of molecular simulation reviewed include investigations of the structure and dynamics of simple models of skin lipids, calculation of the permeability of molecules in simple model membranes, and mechanisms of action of the penetration enhancers, DMSO, ethanol and oleic acid. The studies reviewed illustrate the power and potential of molecular simulation to yield important physical insights, inform and rationalize experimental studies, and to predict structural changes, and kinetic and thermodynamic quantities.


Subject(s)
Drug Delivery Systems , Models, Theoretical , Skin Absorption , Computer Simulation , Excipients/chemistry , Humans , Lipid Metabolism , Molecular Dynamics Simulation , Permeability , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism , Skin/metabolism , Thermodynamics , Vaccines/administration & dosage , Vaccines/pharmacokinetics
17.
Biomater Sci ; 1(5): 478-485, 2013 May 02.
Article in English | MEDLINE | ID: mdl-32482011

ABSTRACT

The ability of polyols to act as ice recrystallisation inhibitors (IRI), inspired by antifreeze (glyco)proteins are studied. Poly(vinyl alcohol), PVA, a known IRI active polymer was compared to a panel of mono and polysaccharides, with the aim of elucidating why some polyols are active and others show no activity. When corrected for total hydroxyl concentration all the carbohydrate-based polyols displayed near identical activity with no significant influence of molecular weight. Conversely, PVA was several orders of magnitude more active and its activity displays significant dependence on molecular-weight implying that its mechanism of action is not identical to that of carbohydrates. In a second step, the role of hydrophobicity was studied and it is observed that monosaccharide IRI activity is enhanced by alkylation. Dye-quenching assays demonstrated that PVA is able to present a hydrophobic surface without self-aggregation. Therefore, the ability to present a hydrophobic domain is hypothesised to be essential to obtain high IRI activity, which has many biotechnological applications.

18.
Biochemistry ; 51(12): 2558-68, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22385253

ABSTRACT

Receptor tyrosine kinases bind ligands such as cytokines, hormones, and growth factors and regulate key cellular processes, including cell division. They are also implicated in the development of many types of cancer. One such example is the Neu receptor tyrosine kinase found in rats (homologous to the human ErbB2 protein), which can undergo a valine to glutamic acid (V(664)E) mutation at the center of its α-helical transmembrane domain. This substitution results in receptor activation and oncogenesis. The molecular basis of this dramatic change in behavior upon introduction of the V(664)E mutation has been difficult to pin down, with conflicting results reported in the literature. Here we report the first quantitative, thermodynamic analysis of dimerization and biophysical characterization of the rat Neu transmembrane domain and several mutants in a range of chemical environments. These data have allowed us to identify the effects of the V(664)E mutation in the isolated TM domain with respect to protein-protein and protein-lipid interactions, membrane insertion, and secondary structure. We also report the results from a 100 ns atomistic molecular dynamics simulation of the Neu transmembrane domain in a model membrane bilayer (dipalmitoylphosphatidylcholine). The results from simulation and experiment are in close agreement and suggest that, in the model systems investigated, the V(664)E mutation leads to a weakening of the TM dimer and a change in sequence-dependent interactions. These results are contrary to recent results obtained in mammalian membranes, and the implications of this are discussed.


Subject(s)
Biophysical Phenomena , Cell Membrane/metabolism , Micelles , Molecular Dynamics Simulation , Point Mutation , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Amino Acid Sequence , Animals , Cell Membrane/drug effects , Detergents/chemistry , Escherichia coli/cytology , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Molecular Sequence Data , Oncogenes/genetics , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/chemistry , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Receptor, ErbB-2/genetics , Receptor, ErbB-2/pharmacology , Thermodynamics
19.
J Biol Chem ; 286(40): 34986-97, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21768108

ABSTRACT

Type IX collagen is covalently bound to the surface of type II collagen fibrils within the cartilage extracellular matrix. The N-terminal, globular noncollagenous domain (NC4) of the α1(IX) chain protrudes away from the surface of the fibrils into the surrounding matrix and is available for molecular interactions. To define these interactions, we used the NC4 domain in a yeast two-hybrid screen of a human chondrocyte cDNA library. 73% of the interacting clones encoded fibronectin. The interaction was confirmed using in vitro immunoprecipitation and was further characterized by surface plasmon resonance. Using whole and pepsin-derived preparations of type IX collagen, the interaction was shown to be specific for the NC4 domain with no interaction with the triple helical collagenous domains. The interaction was shown to be of high affinity with nanomolar K(d) values. Analysis of the fibronectin-interacting clones indicates that the constant domain is the likely site of interaction. Type IX collagen and fibronectin were shown to co-localize in cartilage. This novel interaction between the NC4 domain of type IX collagen and fibronectin may represent an in vivo interaction in cartilage that could contribute to the matrix integrity of the tissue.


Subject(s)
Cartilage, Articular/metabolism , Collagen Type IX/metabolism , Fibronectins/metabolism , Animals , Cartilage/metabolism , Cell Line , Chondrocytes/metabolism , DNA, Complementary/metabolism , Humans , Kinetics , Mice , Polymerase Chain Reaction , Protein Interaction Mapping , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Two-Hybrid System Techniques
20.
Biomacromolecules ; 11(12): 3266-74, 2010 Dec 13.
Article in English | MEDLINE | ID: mdl-20964422

ABSTRACT

We use replica-exchange molecular dynamics (REMD) to interrogate molecular structures and properties of four engineered dodecapeptides (in solution, in the absence of a surface) that have been shown to bind to quartz with different propensities. We find that all of the strong-binding peptides feature some polyproline type II secondary structure, have less conformational freedom, and feature fewer intrapeptide hydrogen bonds compared with the weak binder. The regions of contiguous proline content in a given sequence appear to play a role in fostering some of these properties of the strong binders. For preliminary insights into quartz binding, we perform lattice-matching studies between a grid corresponding with the quartz (100) surface and the strong-binding peptide REMD structures. Our findings indicate a commonality among the putative contact residues, even for peptide structures with very different backbone conformations. Furthermore, interpeptide interactions in solution are studied. Our preliminary findings indicate that the strong-binder interpeptide contacts are dominated by weak, nonspecific hydrophobic interactions, while the weak-binding peptide shows more variable behavior due to the distribution of charged residues. In summary, the solution structures of peptides appear to be significant. We propose that these differences in their intra- and interpeptide interactions can influence their propensity to bind onto a solid substrate.


Subject(s)
Peptides/chemistry , Protein Engineering , Quartz , Molecular Dynamics Simulation , Protein Binding , Solutions
SELECTION OF CITATIONS
SEARCH DETAIL
...