Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Curr Opin Struct Biol ; 81: 102625, 2023 08.
Article in English | MEDLINE | ID: mdl-37331204

ABSTRACT

A striking feature of nucleic acids and lipid membranes is that they all carry net negative charge and so is true for the majority of intracellular proteins. It is suggested that the role of this negative charge is to assure a basal intermolecular repulsion that keeps the cytosolic content suitably 'fluid' for function. We focus in this review on the experimental, theoretical and genetic findings which serve to underpin this idea and the new questions they raise. Unlike the situation in test tubes, any functional protein-protein interaction in the cytosol is subject to competition from the densely crowded background, i.e. surrounding stickiness. At the nonspecific limit of this stickiness is the 'random' protein-protein association, maintaining profuse populations of transient and constantly interconverting complexes at physiological protein concentrations. The phenomenon is readily quantified in studies of the protein rotational diffusion, showing that the more net negatively charged a protein is the less it is retarded by clustering. It is further evident that this dynamic protein-protein interplay is under evolutionary control and finely tuned across organisms to maintain optimal physicochemical conditions for the cellular processes. The emerging picture is then that specific cellular function relies on close competition between numerous weak and strong interactions, and where all parts of the protein surfaces are involved. The outstanding challenge is now to decipher the very basics of this many-body system: how the detailed patterns of charged, polar and hydrophobic side chains not only control protein-protein interactions at close- and long-range but also the collective properties of the cellular interior as a whole.


Subject(s)
Membrane Proteins , Biophysical Phenomena , Hydrophobic and Hydrophilic Interactions
2.
J Neurochem ; 164(1): 77-93, 2023 01.
Article in English | MEDLINE | ID: mdl-36326589

ABSTRACT

Mutations in the human Superoxide dismutase 1 (hSOD1) gene are well-established cause of the motor neuron disease ALS. Patients and transgenic (Tg) ALS model mice carrying mutant variants develop hSOD1 aggregates in the CNS. We have identified two hSOD1 aggregate strains, which both transmit spreading template-directed aggregation and premature fatal paralysis when inoculated into adult transgenic mice. This prion-like spread of aggregation could be a primary disease mechanism in SOD1-induced ALS. Human SOD1 aggregation has been studied extensively both in cultured cells and under various conditions in vitro. To determine how the structure of aggregates formed in these model systems related to disease-associated aggregates in the CNS, we used a binary epitope-mapping assay to examine aggregates of hSOD1 variants G93A, G85R, A4V, D90A, and G127X formed in vitro, in four different cell lines and in the CNS of Tg mice. We found considerable variability between replicate sets of in vitro-generated aggregates. In contrast, there was a high similarity between replicates of a given hSOD1 mutant in a given cell line, but pronounced variations between different hSOD1 mutants and different cell lines in both structures and amounts of aggregates formed. The aggregates formed in vitro or in cultured cells did not replicate the aggregate strains that arise in the CNS. Our findings suggest that the distinct aggregate morphologies in the CNS could result from a micro-environment with stringent quality control combined with second-order selection by spreading ability. Explorations of pathogenesis and development of therapeutics should be conducted in models that replicate aggregate structures forming in the CNS.


Subject(s)
Amyotrophic Lateral Sclerosis , Mice , Humans , Animals , Superoxide Dismutase-1/genetics , Amyotrophic Lateral Sclerosis/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Mice, Transgenic , Cells, Cultured , Mutation/genetics , Disease Models, Animal
3.
J Phys Chem B ; 125(10): 2521-2532, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33657322

ABSTRACT

Aggregate formation of superoxide dismutase 1 (SOD1) inside motor neurons is known as a major factor in onset of amyotrophic lateral sclerosis. The thermodynamic stability of the SOD1 ß-barrel has been shown to decrease in crowded environments such as inside a cell, but it remains unclear how the thermodynamics of crowding-induced protein destabilization relate to SOD1 aggregation. Here we have examined the effects of a protein crowder, lysozyme, on fibril aggregate formation of the SOD1 ß-barrel. We found that aggregate formation of SOD1 is decelerated even in mildly crowded solutions. Intriguingly, transient diffusive interactions with lysozyme do not significantly affect the static structure of the SOD1 ß-barrel but stabilize an alternative excited "invisible" state. The net effect of crowding is to favor species off the aggregation pathway, thereby explaining the decelerated aggregation in the crowded environment. Our observations suggest that the intracellular environment may have a similar negative (inhibitory) effect on fibril formation of other amyloidogenic proteins in living cells. Deciphering how crowded intracellular environments affect aggregation and fibril formation of such disease-associated proteins will probably become central in understanding the exact role of aggregation in the etiology of these enigmatic diseases.


Subject(s)
Amyotrophic Lateral Sclerosis , Superoxide Dismutase , Diffusion , Humans , Muramidase , Mutation , Superoxide Dismutase-1/genetics
4.
Biochemistry ; 60(10): 735-746, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33635054

ABSTRACT

The structural stability of proteins is found to markedly change upon their transfer to the crowded interior of live cells. For some proteins, the stability increases, while for others, it decreases, depending on both the sequence composition and the type of host cell. The mechanism seems to be linked to the strength and conformational bias of the diffusive in-cell interactions, where protein charge is found to play a decisive role. Because most proteins, nucleotides, and membranes carry a net-negative charge, the intracellular environment behaves like a polyanionic (Z:1) system with electrostatic interactions different from those of standard 1:1 ion solutes. To determine how such polyanion conditions influence protein stability, we use negatively charged polyacetate ions to mimic the net-negatively charged cellular environment. The results show that, per Na+ equivalent, polyacetate destabilizes the model protein SOD1barrel significantly more than monoacetate or NaCl. At an equivalent of 100 mM Na+, the polyacetate destabilization of SOD1barrel is similar to that observed in live cells. By the combined use of equilibrium thermal denaturation, folding kinetics, and high-resolution nuclear magnetic resonance, this destabilization is primarily assigned to preferential interaction between polyacetate and the globally unfolded protein. This interaction is relatively weak and involves mainly the outermost N-terminal region of unfolded SOD1barrel. Our findings point thus to a generic influence of polyanions on protein stability, which adds to the sequence-specific contributions and needs to be considered in the evaluation of in vivo data.


Subject(s)
Ovarian Neoplasms/enzymology , Polyelectrolytes/chemistry , Protein Conformation , Superoxide Dismutase-1/chemistry , Enzyme Stability , Female , Humans , Models, Molecular , Ovarian Neoplasms/drug therapy , Polyelectrolytes/pharmacology , Protein Folding , Thermodynamics
5.
JACS Au ; 1(12): 2385-2393, 2021 Dec 27.
Article in English | MEDLINE | ID: mdl-34977906

ABSTRACT

In solution, the charge of a protein is intricately linked to its stability, but electrospray ionization distorts this connection, potentially limiting the ability of native mass spectrometry to inform about protein structure and dynamics. How the behavior of intact proteins in the gas phase depends on the presence and distribution of ionizable surface residues has been difficult to answer because multiple chargeable sites are present in virtually all proteins. Turning to protein engineering, we show that ionizable side chains are completely dispensable for charging under native conditions, but if present, they are preferential protonation sites. The absence of ionizable side chains results in identical charge state distributions under native-like and denaturing conditions, while coexisting conformers can be distinguished using ion mobility separation. An excess of ionizable side chains, on the other hand, effectively modulates protein ion stability. In fact, moving a single ionizable group can dramatically alter the gas-phase conformation of a protein ion. We conclude that although the sum of the charges is governed solely by Coulombic terms, their locations affect the stability of the protein in the gas phase.

6.
J Phys Chem B ; 124(47): 10698-10707, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33179918

ABSTRACT

In the cytosolic environment, protein crowding and Brownian motions result in numerous transient encounters. Each such encounter event increases the apparent size of the interacting molecules, leading to slower rotational tumbling. The extent of transient protein complexes formed in live cells can conveniently be quantified by an apparent viscosity, based on NMR-detected spin-relaxation measurements, that is, the longitudinal (T1) and transverse (T2) relaxation. From combined analysis of three different proteins and surface mutations thereof, we find that T2 implies significantly higher apparent viscosity than T1. At first sight, the effect on T1 and T2 seems thus nonunifiable, consistent with previous reports on other proteins. We show here that the T1 and T2 deviation is actually not a inconsistency but an expected feature of a system with fast exchange between free monomers and transient complexes. In this case, the deviation is basically reconciled by a model with fast exchange between the free-tumbling reporter protein and a transient complex with a uniform 143 kDa partner. The analysis is then taken one step further by accounting for the fact that the cytosolic content is by no means uniform but comprises a wide range of molecular sizes. Integrating over the complete size distribution of the cytosolic interaction ensemble enables us to predict both T1 and T2 from a single binding model. The result yields a bound population for each protein variant and provides a quantification of the transient interactions. We finally extend the approach to obtain a correction term for the shape of a database-derived mass distribution of the interactome in the mammalian cytosol, in good accord with the existing data of the cellular composition.


Subject(s)
Magnetic Resonance Imaging , Proteins , Animals , Magnetic Resonance Spectroscopy , Viscosity
7.
Proc Natl Acad Sci U S A ; 117(46): 28775-28783, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33148805

ABSTRACT

Although folded proteins are commonly depicted as simplistic combinations of ß-strands and α-helices, the actual properties and functions of these secondary-structure elements in their native contexts are just partly understood. The principal reason is that the behavior of individual ß- and α-elements is obscured by the global folding cooperativity. In this study, we have circumvented this problem by designing frustrated variants of the mixed α/ß-protein S6, which allow the structural behavior of individual ß-strands and α-helices to be targeted selectively by stopped-flow kinetics, X-ray crystallography, and solution-state NMR. Essentially, our approach is based on provoking intramolecular "domain swap." The results show that the α- and ß-elements have quite different characteristics: The swaps of ß-strands proceed via global unfolding, whereas the α-helices are free to swap locally in the native basin. Moreover, the α-helices tend to hybridize and to promote protein association by gliding over to neighboring molecules. This difference in structural behavior follows directly from hydrogen-bonding restrictions and suggests that the protein secondary structure defines not only tertiary geometry, but also maintains control in function and structural evolution. Finally, our alternative approach to protein folding and native-state dynamics presents a generally applicable strategy for in silico design of protein models that are computationally testable in the microsecond-millisecond regime.


Subject(s)
Protein Conformation, alpha-Helical/physiology , Protein Conformation, beta-Strand/physiology , Protein Structure, Secondary/physiology , Crystallography, X-Ray/methods , Hydrogen Bonding , Kinetics , Protein Conformation , Protein Denaturation , Protein Folding , Proteins/chemistry , Thermodynamics
8.
Methods Mol Biol ; 2141: 285-302, 2020.
Article in English | MEDLINE | ID: mdl-32696363

ABSTRACT

In the disordered state, a protein exhibits a high degree of structural freedom, in both space and time. For an ensemble of disordered or unfolded proteins, this means that the ensemble comprises a high diversity of structures, ranging from compact collapsed states to fully extended polypeptide chains. In addition, each chain is highly dynamic and undergoes conformational changes and local dynamics on both fast and slow timescales. The size properties of disordered proteins are thus best described as ensemble averages. A straightforward measure of the size is the hydrodynamic radius, RH, of the ensemble. Since the disordered state is conformationally fluid, the observed RH does not refer to a particular shape or fold. Instead, it should be interpreted as a measure for the average compaction of the structural ensemble. In addition to characterizing the disordered ensemble itself, RH can be used to, with good precision, monitor changes in the ensemble size properties upon functional interactions of the disordered protein, e.g., dimerization, ligand binding, and folding pathways. Here, we present a step-by-step protocol for diffusion measurements using pulsed field gradient nuclear magnetic resonance (PFG NMR) spectroscopy. We describe how to calibrate the magnetic field gradient and offer different schemes for sample preparation. Finally, we describe how to obtain RH directly from the diffusion coefficient as well as from using an internal standard as a reference.


Subject(s)
Hydrodynamics , Intrinsically Disordered Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Calibration , Diffusion , Freeze Drying , Reference Standards
9.
Proc Natl Acad Sci U S A ; 117(19): 10113-10121, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32284426

ABSTRACT

Cellular function is generally depicted at the level of functional pathways and detailed structural mechanisms, based on the identification of specific protein-protein interactions. For an individual protein searching for its partner, however, the perspective is quite different: The functional task is challenged by a dense crowd of nonpartners obstructing the way. Adding to the challenge, there is little information about how to navigate the search, since the encountered surrounding is composed of protein surfaces that are predominantly "nonconserved" or, at least, highly variable across organisms. In this study, we demonstrate from a colloidal standpoint that such a blindfolded intracellular search is indeed favored and has more fundamental impact on the cellular organization than previously anticipated. Basically, the unique polyion composition of cellular systems renders the electrostatic interactions different from those in physiological buffer, leading to a situation where the protein net-charge density balances the attractive dispersion force and surface heterogeneity at close range. Inspection of naturally occurring proteomes and in-cell NMR data show further that the "nonconserved" protein surfaces are by no means passive but chemically biased to varying degree of net-negative repulsion across organisms. Finally, this electrostatic control explains how protein crowding is spontaneously maintained at a constant level through the intracellular osmotic pressure and leads to the prediction that the "extreme" in halophilic adaptation is not the ionic-liquid conditions per se but the evolutionary barrier of crossing its physicochemical boundaries.


Subject(s)
Cell Physiological Phenomena , Extracellular Matrix/metabolism , Proteins/chemistry , Proteins/metabolism , Animals , Biomechanical Phenomena , Humans , Hydrogen-Ion Concentration , Osmolar Concentration , Protein Interaction Maps
10.
J Biol Chem ; 295(21): 7224-7234, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32241918

ABSTRACT

A detailed understanding of the molecular pathways for amyloid-ß (Aß) peptide aggregation from monomers into amyloid fibrils, a hallmark of Alzheimer's disease, is crucial for the development of diagnostic and therapeutic strategies. We investigate the molecular details of peptide fibrillization in vitro by perturbing this process through addition of differently charged metal ions. Here, we used a monovalent probe, the silver ion, that, similarly to divalent metal ions, binds to monomeric Aß peptide and efficiently modulates Aß fibrillization. On the basis of our findings, combined with our previous results on divalent zinc ions, we propose a model that links the microscopic metal-ion binding to Aß monomers to its macroscopic impact on the peptide self-assembly observed in bulk experiments. We found that substoichiometric concentrations of the investigated metal ions bind specifically to the N-terminal region of Aß, forming a dynamic, partially compact complex. The metal-ion bound state appears to be incapable of aggregation, effectively reducing the available monomeric Aß pool for incorporation into fibrils. This is especially reflected in a decreased fibril-end elongation rate. However, because the bound state is significantly less stable than the amyloid state, Aß peptides are only transiently redirected from fibril formation, and eventually almost all Aß monomers are integrated into fibrils. Taken together, these findings unravel the mechanistic consequences of delaying Aß aggregation via weak metal-ion binding, quantitatively linking the contributions of specific interactions of metal ions with monomeric Aß to their effects on bulk aggregation.


Subject(s)
Amyloid beta-Peptides/chemistry , Metals/chemistry , Protein Aggregates , Alzheimer Disease , Humans , Protein Binding , Protein Domains
11.
J Mol Biol ; 432(9): 3050-3063, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32081588

ABSTRACT

Leptin is an important signaling hormone, mostly known for its role in energy expenditure and satiety. Furthermore, leptin plays a major role in other proteinopathies, such as cancer, marked hyperphagia, impaired immune function, and inflammation. In spite of its biological relevance in human health, there are no NMR resonance assignments of the human protein available, obscuring high-resolution characterization of the soluble protein and/or its conformational dynamics, suggested as being important for receptor interaction and biological activity. Here, we report the nearly complete backbone resonance assignments of human leptin. Chemical shift-based secondary structure prediction confirms that in solution leptin forms a four-helix bundle including a pierced lasso topology. The conformational dynamics, determined on several timescales, show that leptin is monomeric, has a rigid four-helix scaffold, and a dynamic domain, including a transiently formed helix. The dynamic domain is anchored to the helical scaffold by a secondary hydrophobic core, pinning down the long loops of leptin to the protein body, inducing motional restriction without a well-defined secondary or tertiary hydrogen bond stabilized structure. This dynamic region is well suited for and may be involved in functional allosteric dynamics upon receptor binding.


Subject(s)
Leptin/chemistry , Leptin/metabolism , Binding Sites , Humans , Hydrogen Bonding , Models, Molecular , Protein Folding , Protein Structure, Secondary
12.
Curr Res Struct Biol ; 2: 68-78, 2020.
Article in English | MEDLINE | ID: mdl-34235470

ABSTRACT

Random encounters between proteins in crowded cells are by no means passive, but found to be under selective control. This control enables proteome solubility, helps to optimise the diffusive search for interaction partners, and allows for adaptation to environmental extremes. Interestingly, the residues that modulate the encounters act mesoscopically through protein surface hydrophobicity and net charge, meaning that their detailed signatures vary across organisms with different intracellular constraints. To examine such variations, we use in-cell NMR relaxation to compare the diffusive behaviour of bacterial and human proteins in both human and Escherichia coli cytosols. We find that proteins that 'stick' in E. coli are generally less restricted in mammalian cells. Furthermore, the rotational diffusion in the mammalian cytosol is less sensitive to surface-charge mutations. This implies that, in terms of protein motions, the mammalian cytosol is more forgiving to surface alterations than E. coli cells. The cellular differences seem not linked to the proteome properties per se, but rather to a 6-fold difference in protein concentrations. Our results outline a scenario in which the tolerant cytosol of mammalian cells, found in long-lived multicellular organisms, provides an enlarged evolutionary playground, where random protein-surface mutations are less deleterious than in short-generational bacteria.

13.
J Am Chem Soc ; 140(48): 16570-16579, 2018 12 05.
Article in English | MEDLINE | ID: mdl-30359015

ABSTRACT

A conspicuous feature of the amyotrophic lateral sclerosis (ALS)-associated protein SOD1 is that its maturation into a functional enzyme relies on local folding of two disordered loops into a catalytic subdomain. To drive the disorder-to-order transition, the protein employs a single Zn2+ ion. The question is then if the entropic penalty of maintaining such disordered loops in the immature apoSOD1 monomer is large enough to explain its unusually low stability, slow folding, and pathological aggregation in ALS. To find out, we determined the effects of systematically altering the SOD1-loop lengths by protein redesign. The results show that the loops destabilize the apoSOD1 monomer by ∼3 kcal/mol, rendering the protein marginally stable and accounting for its aggregation behavior. Yet the effect on the global folding kinetics remains much smaller with a transition-state destabilization of <1 kcal/mol. Notably, this 1/3 transition-state to folded-state stability ratio provides a clear-cut example of the enigmatic disagreement between the Leffler α value from loop-length alterations (typically 1/3) and the "standard" reaction coordinates based on solvent perturbations (typically >2/3). Reconciling the issue, we demonstrate that the disagreement disappears when accounting for the progressive loop shortening that occurs along the folding pathway. The approach assumes a consistent Flory loop entropy scaling factor of c = 1.48 for both equilibrium and kinetic data and has the added benefit of verifying the tertiary interactions of the folding nucleus as determined by phi-value analysis. Thus, SOD1 not only represents a case where evolution of key catalytic function has come with the drawback of a destabilized apo state but also stands out as a well-suited model system for exploring the physicochemical details of protein self-organization.


Subject(s)
Superoxide Dismutase-1/chemistry , Amino Acid Sequence , Biocatalysis , Catalytic Domain , Entropy , Humans , Kinetics , Mutation , Protein Conformation , Protein Folding , Protein Stability , Superoxide Dismutase-1/genetics
14.
Proc Natl Acad Sci U S A ; 114(23): E4556-E4563, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28536196

ABSTRACT

How proteins sense and navigate the cellular interior to find their functional partners remains poorly understood. An intriguing aspect of this search is that it relies on diffusive encounters with the crowded cellular background, made up of protein surfaces that are largely nonconserved. The question is then if/how this protein search is amenable to selection and biological control. To shed light on this issue, we examined the motions of three evolutionary divergent proteins in the Escherichia coli cytoplasm by in-cell NMR. The results show that the diffusive in-cell motions, after all, follow simplistic physical-chemical rules: The proteins reveal a common dependence on (i) net charge density, (ii) surface hydrophobicity, and (iii) the electric dipole moment. The bacterial protein is here biased to move relatively freely in the bacterial interior, whereas the human counterparts more easily stick. Even so, the in-cell motions respond predictably to surface mutation, allowing us to tune and intermix the protein's behavior at will. The findings show how evolution can swiftly optimize the diffuse background of protein encounter complexes by just single-point mutations, and provide a rational framework for adjusting the cytoplasmic motions of individual proteins, e.g., for rescuing poor in-cell NMR signals and for optimizing protein therapeutics.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Amino Acid Substitution , Biophysical Phenomena , Copper Transport Proteins , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Humans , Hydrophobic and Hydrophilic Interactions , Metallochaperones/chemistry , Metallochaperones/genetics , Metallochaperones/metabolism , Models, Molecular , Molecular Chaperones , Mutagenesis, Site-Directed , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs , Protein Transport , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Static Electricity , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
15.
Curr Opin Struct Biol ; 42: 129-135, 2017 02.
Article in English | MEDLINE | ID: mdl-28126529

ABSTRACT

The recent advancement in moving 'biophysical' analysis of proteins in vivo has finally brought us to a position where we can start to make quantitative comparisons with existing in-vitro data. A striking observation is that protein behaviour in live cells seems, after all, not that different from in test tubes, not even at the level of complex mechanisms like protein aggregation. The example examined in this review is the ALS associated protein SOD1 that apparently retains its in-vitro properties in vivo. Does this mean that the protocols for studying proteins in vivo are somehow oversimplified, or that the macromolecular properties and interplay - despite being intrinsically malleable - are evolutionary more 'streamlined' than previously anticipated? Whatever the answer may be the time is now right to put these data to critical biological test.


Subject(s)
Proteins , Animals , Humans , Protein Stability , Proteins/chemistry , Proteins/metabolism , Superoxide Dismutase-1/chemistry , Superoxide Dismutase-1/metabolism
16.
J Am Chem Soc ; 138(48): 15571-15579, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27783493

ABSTRACT

Despite continuing interest in partly unfolded proteins as precursors for aggregation and adverse gain-of-function in human disease, there is yet little known about the local transitions of native structures that possibly lead to such intermediate states. To target this problem, we present here a protein-design strategy that allows real-time detection of rupture and swapping of complete secondary-structure elements in globular proteins-molecular events that have previously been inaccessible experimental analysis. The approach is applied to the dynamic ß-barrel of SOD1, associated with pathologic aggregation in the neurodegenerative disease ALS. Data show that rupture and re-insertion of individual ß-strands do not take place locally but require the SOD1 barrel to unfold globally. The finding questions the very existence of partly unfolded intermediates in the SOD1 aggregation process and presents new clues to the mechanism by which hydrogen bonding maintains global structural integrity.


Subject(s)
Superoxide Dismutase-1/chemistry , Humans , Hydrogen Bonding , Kinetics , Protein Aggregates , Protein Structure, Secondary , Protein Unfolding , Thermodynamics
17.
J Am Chem Soc ; 138(21): 6893-902, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27171340

ABSTRACT

Protein misfolding and formation of cross-ß structured amyloid fibrils are linked to many neurodegenerative disorders. Although recently developed quantitative approaches have started to reveal the molecular nature of self-assembly and fibril formation of proteins and peptides, it is yet unclear how these self-organization events are precisely modulated by microenvironmental factors, which are known to strongly affect the macroscopic aggregation properties. Here, we characterize the explicit effect of ionic strength on the microscopic aggregation rates of amyloid ß peptide (Aß40) self-association, implicated in Alzheimer's disease. We found that physiological ionic strength accelerates Aß40 aggregation kinetics by promoting surface-catalyzed secondary nucleation reactions. This promoted catalytic effect can be assigned to shielding of electrostatic repulsion between monomers on the fibril surface or between the fibril surface itself and monomeric peptides. Furthermore, we observe the formation of two different ß-structured states with similar but distinct spectroscopic features, which can be assigned to an off-pathway immature state (Fß*) and a mature stable state (Fß), where salt favors formation of the Fß fibril morphology. Addition of salt to preformed Fß* accelerates transition to Fß, underlining the dynamic nature of Aß40 fibrils in solution. On the basis of these results we suggest a model where salt decreases the free-energy barrier for Aß40 folding to the Fß state, favoring the buildup of the mature fibril morphology while omitting competing, energetically less favorable structural states.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid/chemistry , Peptide Fragments/chemistry , Alzheimer Disease/metabolism , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Entropy , Kinetics , Microscopy, Electron, Transmission , Nuclear Magnetic Resonance, Biomolecular , Osmolar Concentration , Peptide Fragments/metabolism , Protein Aggregates , Protein Folding , Sodium Chloride/chemistry , Sodium Fluoride/chemistry
18.
Plant Physiol ; 171(2): 932-43, 2016 06.
Article in English | MEDLINE | ID: mdl-27208263

ABSTRACT

Dehydrins are disordered proteins that are expressed in plants as a response to embryogenesis and water-related stress. The molecular function and structural action of the dehydrins are yet elusive, but increasing evidence points to a role in protecting the structure and functional dynamics of cell membranes. An intriguing example is the cold-induced dehydrin Lti30 that binds to membranes by its conserved K segments. Moreover, this binding can be regulated by pH and phosphorylation and shifts the membrane phase transition to lower temperatures, consistent with the protein's postulated function in cold stress. In this study, we reveal how the Lti30-membrane interplay works structurally at atomic level resolution in Arabidopsis (Arabidopsis thaliana). Nuclear magnetic resonance analysis suggests that negatively charged lipid head groups electrostatically capture the protein's disordered K segments, which locally fold up into α-helical segments on the membrane surface. Thus, Lti30 conforms to the general theme of structure-function relationships by folding upon binding, in spite of its disordered, atypically hydrophilic and repetitive sequence signatures. Moreover, the fixed and well-defined structure of the membrane-bound K segments suggests that dehydrins have the molecular prerequisites for higher level binding specificity and regulation, raising new questions about the complexity of their biological function.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/metabolism , Cold Shock Proteins and Peptides/chemistry , Plant Proteins/chemistry , Amino Acid Sequence , Arabidopsis/chemistry , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Cell Membrane/chemistry , Cell Membrane/metabolism , Cold Shock Proteins and Peptides/genetics , Cold Shock Proteins and Peptides/metabolism , Cold Temperature , Hydrogen-Ion Concentration , Models, Molecular , Phosphorylation , Plant Proteins/genetics , Plant Proteins/metabolism , Protein Binding , Protein Conformation , Protein Folding , Sequence Alignment , Static Electricity , Temperature
19.
Proc Natl Acad Sci U S A ; 112(40): 12402-7, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26392565

ABSTRACT

Although protein folding and stability have been well explored under simplified conditions in vitro, it is yet unclear how these basic self-organization events are modulated by the crowded interior of live cells. To find out, we use here in-cell NMR to follow at atomic resolution the thermal unfolding of a ß-barrel protein inside mammalian and bacterial cells. Challenging the view from in vitro crowding effects, we find that the cells destabilize the protein at 37 °C but with a conspicuous twist: While the melting temperature goes down the cold unfolding moves into the physiological regime, coupled to an augmented heat-capacity change. The effect seems induced by transient, sequence-specific, interactions with the cellular components, acting preferentially on the unfolded ensemble. This points to a model where the in vivo influence on protein behavior is case specific, determined by the individual protein's interplay with the functionally optimized "interaction landscape" of the cellular interior.


Subject(s)
Protein Folding , Protein Unfolding , Proteins/chemistry , Thermodynamics , Algorithms , Animals , Catalytic Domain , Cell Line, Tumor , Circular Dichroism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Hydrogen-Ion Concentration , Kinetics , Magnetic Resonance Spectroscopy , Mice , Models, Molecular , Protein Stability , Protein Structure, Tertiary , Proteins/genetics , Proteins/metabolism , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Temperature
20.
Proc Natl Acad Sci U S A ; 112(32): 9878-83, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26221023

ABSTRACT

A longstanding challenge in studies of neurodegenerative disease has been that the pathologic protein aggregates in live tissue are not amenable to structural and kinetic analysis by conventional methods. The situation is put in focus by the current progress in demarcating protein aggregation in vitro, exposing new mechanistic details that are now calling for quantitative in vivo comparison. In this study, we bridge this gap by presenting a direct comparison of the aggregation kinetics of the ALS-associated protein superoxide dismutase 1 (SOD1) in vitro and in transgenic mice. The results based on tissue sampling by quantitative antibody assays show that the SOD1 fibrillation kinetics in vitro mirror with remarkable accuracy the spinal cord aggregate buildup and disease progression in transgenic mice. This similarity between in vitro and in vivo data suggests that, despite the complexity of live tissue, SOD1 aggregation follows robust and simplistic rules, providing new mechanistic insights into the ALS pathology and organism-level manifestation of protein aggregation phenomena in general.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/pathology , Protein Aggregates , Superoxide Dismutase/chemistry , Superoxide Dismutase/metabolism , Animals , Apoproteins/chemistry , Apoproteins/metabolism , Disease Models, Animal , Kinetics , Mice, Transgenic , Mutation/genetics , Protein Unfolding , Spinal Cord/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase-1 , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...