Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
Protein Sci ; 27(7): 1205-1217, 2018 07.
Article in English | MEDLINE | ID: mdl-29417650

ABSTRACT

Protein aggregation is linked with the onset of several neurodegenerative disorders, including Parkinson's disease (PD), which is associated with the aggregation of α-synuclein (αSyn). The structural mechanistic details of protein aggregation, including the nature of the earliest protein-protein interactions, remain elusive. In this study, we have used single molecule force spectroscopy (SMFS) to probe the first dimerization events of the central aggregation-prone region of αSyn (residues 71-82) that may initiate aggregation. This region has been shown to be necessary for the aggregation of full length αSyn and is capable of forming amyloid fibrils in isolation. We demonstrate that the interaction of αSyn71-82 peptides can be studied using SMFS when inserted into a loop of protein L, a mechanically strong and soluble scaffold protein that acts as a display system for SMFS studies. The corresponding fragment of the homolog protein γ-synuclein (γSyn), which has a lower aggregation propensity, has also been studied here. The results from SMFS, together with native mass spectrometry and aggregation assays, demonstrate that the dimerization propensity of γSyn71-82 is lower than that of αSyn71-82 , but that a mixed αSyn71-82 : γSyn71-82 dimer forms with a similar propensity to the αSyn71-82 homodimer, slowing amyloid formation. This work demonstrates the utility of a novel display method for SMFS studies of aggregation-prone peptides, which would otherwise be difficult to study.


Subject(s)
Parkinson Disease/metabolism , Single Molecule Imaging/methods , alpha-Synuclein/chemistry , alpha-Synuclein/metabolism , Crystallography, X-Ray , Humans , Mass Spectrometry , Models, Molecular , Peptides/chemistry , Peptides/metabolism , Protein Aggregates , Protein Binding , Protein Domains , Protein Folding
2.
Chem Sci ; 8(7): 5030-5040, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28970890

ABSTRACT

Although amyloid assembly in vitro is commonly investigated using single protein sequences, fibril formation in vivo can be more heterogeneous, involving co-assembly of proteins of different length, sequence and/or post-translational modifications. Emerging evidence suggests that co-polymerization can alter the rate and/or mechanism of aggregation and can contribute to pathogenicity. Electrospray ionization-ion mobility spectrometry-mass spectrometry (ESI-IMS-MS) is uniquely suited to the study of these heterogeneous ensembles. Here, ESI-IMS-MS combined with analysis of fibrillation rates using thioflavin T (ThT) fluorescence, is used to track the course of aggregation of variants of islet-amyloid polypeptide (IAPP) in isolation and in pairwise mixtures. We identify a sub-population of extended monomers as the key precursors of amyloid assembly, and reveal that the fastest aggregating sequence in peptide mixtures determines the lag time of fibrillation, despite being unable to cross-seed polymerization. The results demonstrate that co-polymerization of IAPP sequences radically alters the rate of amyloid assembly by altering the conformational properties of the mixed oligomers that form.

3.
Curr Opin Chem Biol ; 39: 90-99, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28649012

ABSTRACT

Understanding the mechanisms of amyloid formation and toxicity remain major challenges. Although substantial progress has been made in the development of methods able to identify the species formed during self-assembly and to describe the kinetic mechanisms of aggregation, the structure(s) of non-native species, including potentially toxic oligomers, remain elusive. Moreover, how fibrils contribute to disease remains unclear. Here we review recent advances in the development of small molecules and other reagents that are helping to define the mechanisms of protein aggregation in molecular detail. Such probes form a powerful platform with which to better define the mechanisms of structural conversion into amyloid fibrils and may provide the much-needed stepping stone for future development of successful therapeutic agents.


Subject(s)
Molecular Probes/metabolism , Protein Aggregates , Small Molecule Libraries/metabolism , Animals , Drug Discovery , Humans , Proteins/chemistry , Proteins/metabolism
4.
Methods ; 95: 62-9, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26007606

ABSTRACT

Electrospray ionisation-ion mobility spectrometry-mass spectrometry (ESI-IMS-MS) is a powerful method for the study of conformational changes in protein complexes, including oligomeric species populated during protein self-aggregation into amyloid fibrils. Information on the mass, stability, cross-sectional area and ligand binding capability of each transiently populated intermediate, present in the heterogeneous mixture of assembling species, can be determined individually in a single experiment in real-time. Determining the structural characterisation of oligomeric species and alterations in self-assembly pathways observed in the presence of small molecule inhibitors is of great importance, given the urgent demand for effective therapeutics. Recent studies have demonstrated the capability of ESI-IMS-MS to identify small molecule modulators of amyloid assembly and to determine the mechanism by which they interact (positive, negative, non-specific binding, or colloidal) in a high-throughput format. Here, we demonstrate these advances using self-assembly of Aß40 as an example, and reveal two new inhibitors of Aß40 fibrillation.


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Protein Aggregates/drug effects , Small Molecule Libraries/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Amyloid beta-Peptides/chemistry , High-Throughput Screening Assays , Humans , Hydrogen-Ion Concentration , Ligands , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Conformation , Solutions
5.
Nat Chem Biol ; 12(2): 94-101, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26656088

ABSTRACT

Protein aggregation underlies an array of human diseases, yet only one small-molecule therapeutic targeting this process has been successfully developed to date. Here, we introduce an in vivo system, based on a ß-lactamase tripartite fusion construct, that is capable of identifying aggregation-prone sequences in the periplasm of Escherichia coli and inhibitors that prevent their aberrant self-assembly. We demonstrate the power of the system using a range of proteins, from small unstructured peptides (islet amyloid polypeptide and amyloid ß) to larger, folded immunoglobulin domains. Configured in a 48-well format, the split ß-lactamase sensor readily differentiates between aggregation-prone and soluble sequences. Performing the assay in the presence of 109 compounds enabled a rank ordering of inhibition and revealed a new inhibitor of islet amyloid polypeptide aggregation. This platform can be applied to both amyloidogenic and other aggregation-prone systems, independent of sequence or size, and can identify small molecules or other factors able to ameliorate or inhibit protein aggregation.


Subject(s)
Biological Assay/methods , Protein Aggregation, Pathological , Amyloid beta-Peptides/metabolism , Blotting, Western , Curcumin/pharmacology , Dopamine/chemistry , Dopamine/pharmacology , Humans , Microscopy, Electron, Transmission , Protein Binding/drug effects , Spectrometry, Mass, Electrospray Ionization , beta-Lactamases/chemistry
6.
Analyst ; 140(20): 6990-9, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26193839

ABSTRACT

The precise molecular mechanisms by which different peptides and proteins assemble into highly ordered amyloid deposits remain elusive. The fibrillation of human amylin (also known as islet amyloid polypeptide, hIAPP) and the amyloid-beta peptide (Aß-40) are thought to be pathogenic factors in Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD), respectively. Amyloid diseases may involve co-aggregation of different protein species, in addition to the self-assembly of single precursor sequences. Here we investigate the formation of heterogeneous pre-fibrillar, oligomeric species produced by the co-incubation of hIAPP and Aß-40 using electrospray ionisation-ion mobility spectrometry-mass spectrometry (ESI-IMS-MS)-based methods. Conformational properties and gas-phase stabilities of amyloid oligomers formed from hIAPP or Aß40 alone, and from a 1 : 1 mixture of hIAPP and Aß40 monomers, were determined and compared. We show that co-assembly of the two sequences results in hetero-oligomers with distinct properties and aggregation kinetics properties compared with the homo-oligomers present in solution. The observations may be of key significance to unravelling the mechanisms of amyloid formation in vivo and elucidating how different sequences and/or assembly conditions can result in different fibril structures and/or pathogenic outcomes.


Subject(s)
Amyloid beta-Peptides/chemistry , Islet Amyloid Polypeptide/chemistry , Peptide Fragments/chemistry , Protein Multimerization , Spectrometry, Mass, Electrospray Ionization , Amino Acid Sequence , Humans , Molecular Sequence Data , Protein Stability , Protein Structure, Secondary
7.
Biochemistry ; 54(3): 666-76, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25531836

ABSTRACT

The process of amyloid formation by the normally soluble hormone islet amyloid polypeptide (IAPP) contributes to ß-cell death in type 2 diabetes and in islet transplants. There are no clinically approved inhibitors of islet amyloidosis, and the mode of action of existing inhibitors is not well-understood. Resveratrol, a natural polyphenol, has been reported to inhibit amyloid formation by IAPP and by the Alzheimer's disease Aß peptide. The mechanism of action of this compound is not known, nor is its mode of interaction with IAPP. In this study, we use a series of IAPP variants to examine possible interactions between resveratrol and IAPP. Fluorescence assays, transmission electron microscopy, and mass spectrometry demonstrate that resveratrol is much less effective as an inhibitor of IAPP amyloid formation than the polyphenol (-)-epigallocatechin 3-gallate (EGCG) and, unlike EGCG, does not significantly disaggregate preformed IAPP amyloid fibrils. Resveratrol is also shown to interfere with thioflavin-T assays. His-18 mutants, a truncation mutant, mutants of each of the aromatic residues, and mutants of Arg-11 of IAPP were examined. Mutation of His to Gln or Leu weakens the ability of resveratrol to inhibit amyloid formation by IAPP, as do mutations of Arg-11, Phe-15, or Tyr-37 to Leu, and truncation to form the variant Ac 8-37-IAPP, which removes the first seven residues to eliminate Lys-1 and the N-terminal amino group. In contrast, replacement of Phe-23 with Leu has a smaller effect. The data highlight Phe-15, His-18, and Tyr-37 as being important for IAPP-resveratrol interactions and are consistent with a potential role of the N-terminus and Arg-11 in polypeptide-resveratrol interactions.


Subject(s)
Amino Acids, Aromatic/metabolism , Amyloid/metabolism , Histidine/metabolism , Islet Amyloid Polypeptide/genetics , Islet Amyloid Polypeptide/metabolism , Stilbenes/pharmacology , Amino Acid Sequence , Amyloid/ultrastructure , Arginine/metabolism , Benzothiazoles , Catechin/analogs & derivatives , Catechin/pharmacology , DNA Mutational Analysis , Humans , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/ultrastructure , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Mutant Proteins/ultrastructure , Resveratrol , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship , Thiazoles/metabolism
8.
Nat Chem ; 7(1): 73-81, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25515893

ABSTRACT

The search for therapeutic agents that bind specifically to precursor protein conformations and inhibit amyloid assembly is an important challenge. Identifying such inhibitors is difficult because many protein precursors of aggregation are partially folded or intrinsically disordered, which rules out structure-based design. Furthermore, inhibitors can act by a variety of mechanisms, including specific or nonspecific binding, as well as colloidal inhibition. Here we report a high-throughput method based on ion mobility spectrometry-mass spectrometry (IMS-MS) that is capable of rapidly detecting small molecules that bind to amyloid precursors, identifying the interacting protein species and defining the mode of inhibition. Using this method we have classified a variety of small molecules that are potential inhibitors of human islet amyloid polypeptide (hIAPP) aggregation or amyloid-beta 1-40 aggregation as specific, nonspecific, colloidal or non-interacting. We also demonstrate the ability of IMS-MS to screen for inhibitory small molecules in a 96-well plate format and use this to discover a new inhibitor of hIAPP amyloid assembly.


Subject(s)
Amyloid/metabolism , Small Molecule Libraries/metabolism , Amino Acid Sequence , Amyloid/antagonists & inhibitors , Amyloid beta-Peptides/antagonists & inhibitors , Amyloid beta-Peptides/metabolism , Colloids/chemistry , Colloids/metabolism , Humans , Islet Amyloid Polypeptide/antagonists & inhibitors , Islet Amyloid Polypeptide/metabolism , Molecular Sequence Data , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/metabolism , Protein Binding , Small Molecule Libraries/chemistry , Spectrometry, Mass, Electrospray Ionization
9.
J Am Chem Soc ; 136(2): 660-70, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24372466

ABSTRACT

The molecular mechanisms by which different proteins assemble into highly ordered fibrillar deposits and cause disease remain topics of debate. Human amylin (also known as islet amyloid polypeptide/hIAPP) is found in vivo as amyloid deposits in the pancreatic islets of sufferers of type II diabetes mellitus, and its self-aggregation is thought to be a pathogenic factor in disease and to contribute to the failure of islet transplants. Here, electrospray ionization-ion mobility spectrometry-mass spectrometry (ESI-IMS-MS) has been used to monitor oligomer formation from IAPP. The detection, identification and characterization of oligomers from both human and rat amylin (rIAPP) are described. Oligomers up to and including hexamers have been detected for both peptides. From ESI-IMS-MS derived collision cross sections (CCS), these species are shown to be elongated in conformation. Collision-induced dissociation (CID-MS/MS) revealed differences in the gas-phase stability of the oligomers formed from hIAPP and rIAPP, which may contribute to their differences in amyloid propensity. Using ESI-IMS-MS, the mode of inhibition of amyloid formation from hIAPP using small molecules or co-incubation with rIAPP was also investigated. We show that the polyphenolic compounds epigallocatechin gallate (EGCG) and silibinin bind to specific conformers within a dynamic ensemble of hIAPP monomers, altering the progress of oligomerization and fibril assembly. Hetero-oligomer formation also occurs with rIAPP but leads only to inefficient inhibition. The results indicate that although different small molecules can be effective inhibitors of hIAPP self-assembly, their modes of action are distinct and can be distinguished using ESI-IMS-MS.


Subject(s)
Islet Amyloid Polypeptide/antagonists & inhibitors , Islet Amyloid Polypeptide/analysis , Silymarin/pharmacology , Small Molecule Libraries/pharmacology , Animals , Humans , Islet Amyloid Polypeptide/chemical synthesis , Ligands , Rats , Silybin , Silymarin/chemistry , Small Molecule Libraries/chemistry , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL