repeats in the α-synuclein sequence determine its conformation on membranes and influence...

1
while curvature sensing is generally conserved throughout the membrane binding region of aSyn, curvature generation requires specific binding domains. These re- sults will help to form a model for the interplay between aSyn membrane binding activity and membrane remodeling and may have implications for understanding both aSyn’s native role as well as its contribution to PD. 1360-Pos Board B90 Site-Specific Hydration Dynamics Illuminates the Key Structural Features of Membrane-Bound a-Synuclein Samrat Mukhopadhyay, Neha Jain, Karishma Bhasne, M. Hemaswasthi. Indian Institute of Science Education and Research (IISER), Mohali, Mohali, India. a-Synuclein is an intrinsically disordered protein that is preferentially ex- pressed in presynaptic nerve terminals. It undergoes a large-scale conforma- tional rearrangement upon binding to synaptic vesicle membranes. In order to obtain the structural insights into the membrane-bound a-synuclein at the residue-specific resolution, we incorporated single tryptophan at various loca- tions along the sequence. These tryptophans were used as site-specific fluores- cence markers to characterize the structural and dynamical aspects of a-synuclein. The spatial localization of various parts of the protein near the membrane surface was elucidated utilizing a unique and sensitive fluorescence readout, namely, red-edge excitation shift (REES), which originates when a flu- orophore is located in a highly ordered micro-environment. The extent of REES observed at different locations allowed us to directly identify the residues that are localized at the membrane-water interface comprising a thin (~15 A ˚ ) layer of motionally-constrained water molecules (Figure 1). Additionally, we have been able to distinguish subtle but impor- tant structural differences of a-synuclein bound to different lipids membranes. We believe that the structural modulations of a-synuclein on the membrane could potentially be related to its physiological functions as well as to the onset of Parkin- son’s diseases. 1361-Pos Board B91 Determination of Primary Nucleation Mechanisms of a-Synuclein Amyloid Aggregation Francesco A. Aprile, Georg Meisl, Alexander K. Buell, Patrick Flagmeier, Christopher M. Dobson, Michele Vendruscolo, Tuomas P.J. Knowles. University of Cambridge, Cambridge, United Kingdom. Protein conformational diseases represent a class of pathologies in which spe- cific peptides or proteins form aberrant self-assemblies that constitute the hall- mark of several neurodegenerative diseases. Specifically, the formation of intra-neuronal inclusions of the protein a-synuclein (aSyn) is associated with the pathogenesis of Parkinson’s disease (PD). A great interest is in the early stages of aSyn aggregation, for which soluble monomeric proteins are converted into fibrillar nanostructures. It has been shown that at these stages many parallel and competing pathways take contem- poraneously place and it is currently very difficult to address on these mecha- nisms by using standard techniques of molecular investigations. In order to overcome the limitations of standard approaches, we employed ensemble- averaged kinetic studies coupled with microdloplet technology in order to char- acterize the primary nucleation early stages of aSyn amyloid formation and therefore to elucidate the fundamental mechanisms underlying this phenome- non. Testing different aggregation conditions, we have been able to understand that the primary nucleation mechanism underlying aSyn aggregation is not ho- mogeneous, whereas it is catalysed by different factors, including air/water sur- face interactions. The full characterization of all the processes involved in the aggregation mech- anism of aSyn will be fundamental for devising new and innovative therapeutic strategies against PD. Indeed, based on our analysis, we expect that it will be possible to design and screen pharmacological compounds able to selectively inhibit the nucleation steps that trigger either the overall of the process or spe- cifically the formation of the toxic aggregated species. 1362-Pos Board B92 Oligomerisation of Alpha-Synuclein at Physiological Concentrations Marija Iljina. Cambridge University, Cambridge, United Kingdom. Alpha-synuclein is a small intracellular protein naturally abundant in the brain at low-micromolar concentrations. Its fibrillar aggregates are the major constit- uents of intracellular inclusions, known as Lewy bodies, which are the patho- logical hallmarks of Parkinson disease and related neurodegenerative disorders. However, increasing evidence suggest that oligomers, rather than fibrils, are the most toxic and damaging to brain neurons. Single molecule FRET can be used to detect and characterise the low levels of heterogeneous oligomers formed during protein aggregation. In this presentation, I will discuss the recent results of in-vitro studies of alpha-synuclein oligomer formation at physiologically- relevant concentrations using single-molecule FRET spectroscopy and show how these experiments reveal the key microscopic reactions taking place dur- ing the aggregation of alpha-synuclein. 1363-Pos Board B93 Single-Molecule Spectroscopy Reveals Polymer Effects of Disordered Pro- teins in Crowded Environments Andrea Soranno, Iwo Koenig, Madeleine B. Borgia, Hagen Hofmann, Franziska Zosel, Daniel Nettels, Ben Schuler. Biochemistry Department, University of Zu ¨rich, Zu ¨rich, Switzerland. It is currently unclear how the crowded cellular milieu affects the structural dis- tributions of intrinsically disordered proteins (IDPs). Here we employ single- molecule Fo ¨rster resonance energy transfer (FRET) to quantify the effect of molecular crowding on four disordered proteins with very different degrees of charge-induced expansion. For a variety of crowding agents (PEG, PVP, Dextran, PVA), an increasing collapse of the polypeptide chains is observed with increasing concentrations of crowder, as expected from simple consider- ations, such as scaled particle theory. However, we also observe an increasing collapse with increasing size of the crowder, the opposite of what scaled parti- cle theory predicts. Interestingly, the observations can be rationalized quantitatively within the framework of Flory-Huggins theories that take into account the polymeric properties of both the disordered proteins and the crowder. The results provide a step towards understanding the behavior of IDPs and denatured proteins in the presence of polydisperse co-solutes as those characteristic of the cellular environment. 1364-Pos Board B94 Single-Molecule Characterisation of Alpha-Synuclein Oligomers Mathew H. Horrocks 1 , Steven F. Lee 1 , Sonia Gandhi 2 , Marija Iljina 1 , Laura Tosatto 1 , Christopher M. Dobson 1 , David Klenerman 1 . 1 Cambridge University, CAMBRIDGE, United Kingdom, 2 Institute of Neurology, University College London, London, United Kingdom. The pathological hallmark of Parkinson’s disease is the presence of insoluble protein deposits in the brain, which are formed when specific protein molecules misfold and aggregate into highly ordered fibrils. In Parkinson’s disease, the deposits are primarily made up of alpha-synuclein, a protein whose major func- tion is not fully known. Rather than the fibrils themselves being toxic, evidence now points towards the smaller, soluble oligomers formed in the initial stages of the process as being the culprit. We have developed a novel single-molecule fluorescence technique to detect and characterise the oligomers of alpha- synuclein. Using this methodology, we are able to identify the cytotoxic spe- cies, and apply these species to primary neuronal cultures to investigate their damaging effects. 1365-Pos Board B95 Repeats in the a-Synuclein Sequence Determine its Conformation on Membranes and Influence Aggregation Properties Volodymyr Shvadchak 1 , Vinod Subramaniam 2,3 . 1 Nanobiophysics, University of Twente, Enschede, Netherlands, 2 Nanoscale Biophysics, FOM Institute AMOLF, Amsterdam, Netherlands, 3 University of Twente, Enschede, Netherlands. Alpha-Synuclein (aS) is a 140 aa intrinsically disordered and amyloidogenic protein. Its physiological functions are unclear, but are believed to be con- nected to the interaction with synaptic vesicles or membranes of other organelles. The sequence of aS shows partial 11 amino acid periodicity that induces atypical 3/11 helix conformation on membranes. Fluorescence studies of tryptophan mu- tants of aS point to a flexible break at residues 52-54 between two helical domains. Deletion of this flexible 4 aa fragment between two groups of 11 aa repeats does not significantly affect aS membrane binding but strongly decreases the protein aggregation and fibril formation propensity. Moreover, the deletion mutant in- hibits aggregation of wildtype aS, likely by hindering the fibril growth, since the mutant does not appear to co-aggregate into fibrils with wt aS. Introducing additional 11 amino acid repeats into the aS sequence increases af- finity of the modified protein to membranes and slows down the protein aggrega- tion. We believe that the 11 amino acid repeats in the aS sequence play a key role in aS’s ability to switch between a helical conformation on membranes and b-sheets in fibrils. 268a Monday, February 17, 2014

Upload: vinod

Post on 01-Jan-2017

212 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Repeats in the α-Synuclein Sequence Determine its Conformation on Membranes and Influence Aggregation Properties

268a Monday, February 17, 2014

while curvature sensing is generally conserved throughout themembrane bindingregion of aSyn, curvature generation requires specific binding domains. These re-sults will help to form amodel for the interplay between aSynmembrane bindingactivity and membrane remodeling andmay have implications for understandingboth aSyn’s native role as well as its contribution to PD.

1360-Pos Board B90Site-Specific Hydration Dynamics Illuminates the Key Structural Featuresof Membrane-Bound a-SynucleinSamrat Mukhopadhyay, Neha Jain, Karishma Bhasne, M. Hemaswasthi.Indian Institute of Science Education and Research (IISER), Mohali, Mohali,India.a-Synuclein is an intrinsically disordered protein that is preferentially ex-pressed in presynaptic nerve terminals. It undergoes a large-scale conforma-tional rearrangement upon binding to synaptic vesicle membranes. In orderto obtain the structural insights into the membrane-bound a-synuclein at theresidue-specific resolution, we incorporated single tryptophan at various loca-tions along the sequence. These tryptophans were used as site-specific fluores-cence markers to characterize the structural and dynamical aspects ofa-synuclein. The spatial localization of various parts of the protein near themembrane surface was elucidated utilizing a unique and sensitive fluorescencereadout, namely, red-edge excitation shift (REES), which originates when a flu-orophore is located in a highly ordered micro-environment. The extent of REESobserved at different locations allowed us to directly identify the residues thatare localized at the membrane-water interface comprising a thin (~15 A) layerof motionally-constrained water molecules (Figure 1). Additionally, we have

been able to distinguish subtle but impor-tant structural differences of a-synucleinbound to different lipids membranes. Webelieve that the structural modulations ofa-synuclein on the membrane couldpotentially be related to its physiologicalfunctions as well as to the onset of Parkin-son’s diseases.

1361-Pos Board B91Determination of Primary Nucleation Mechanisms of a-SynucleinAmyloid AggregationFrancesco A. Aprile, Georg Meisl, Alexander K. Buell, Patrick Flagmeier,Christopher M. Dobson, Michele Vendruscolo, Tuomas P.J. Knowles.University of Cambridge, Cambridge, United Kingdom.Protein conformational diseases represent a class of pathologies in which spe-cific peptides or proteins form aberrant self-assemblies that constitute the hall-mark of several neurodegenerative diseases. Specifically, the formation ofintra-neuronal inclusions of the protein a-synuclein (aSyn) is associated withthe pathogenesis of Parkinson’s disease (PD).A great interest is in the early stages of aSyn aggregation, for which solublemonomeric proteins are converted into fibrillar nanostructures. It has beenshown that at these stages many parallel and competing pathways take contem-poraneously place and it is currently very difficult to address on these mecha-nisms by using standard techniques of molecular investigations. In order toovercome the limitations of standard approaches, we employed ensemble-averaged kinetic studies coupled with microdloplet technology in order to char-acterize the primary nucleation early stages of aSyn amyloid formation andtherefore to elucidate the fundamental mechanisms underlying this phenome-non. Testing different aggregation conditions, we have been able to understandthat the primary nucleation mechanism underlying aSyn aggregation is not ho-mogeneous, whereas it is catalysed by different factors, including air/water sur-face interactions.The full characterization of all the processes involved in the aggregation mech-anism of aSyn will be fundamental for devising new and innovative therapeuticstrategies against PD. Indeed, based on our analysis, we expect that it will bepossible to design and screen pharmacological compounds able to selectivelyinhibit the nucleation steps that trigger either the overall of the process or spe-cifically the formation of the toxic aggregated species.

1362-Pos Board B92Oligomerisation of Alpha-Synuclein at Physiological ConcentrationsMarija Iljina.Cambridge University, Cambridge, United Kingdom.Alpha-synuclein is a small intracellular protein naturally abundant in the brainat low-micromolar concentrations. Its fibrillar aggregates are the major constit-uents of intracellular inclusions, known as Lewy bodies, which are the patho-logical hallmarks of Parkinson disease and related neurodegenerative disorders.However, increasing evidence suggest that oligomers, rather than fibrils, are the

most toxic and damaging to brain neurons. Single molecule FRET can be usedto detect and characterise the low levels of heterogeneous oligomers formedduring protein aggregation. In this presentation, I will discuss the recent resultsof in-vitro studies of alpha-synuclein oligomer formation at physiologically-relevant concentrations using single-molecule FRET spectroscopy and showhow these experiments reveal the key microscopic reactions taking place dur-ing the aggregation of alpha-synuclein.

1363-Pos Board B93Single-Molecule Spectroscopy Reveals Polymer Effects of Disordered Pro-teins in Crowded EnvironmentsAndrea Soranno, Iwo Koenig, Madeleine B. Borgia, Hagen Hofmann,Franziska Zosel, Daniel Nettels, Ben Schuler.Biochemistry Department, University of Zurich, Zurich, Switzerland.It is currently unclear how the crowded cellular milieu affects the structural dis-tributions of intrinsically disordered proteins (IDPs). Here we employ single-molecule Forster resonance energy transfer (FRET) to quantify the effect ofmolecular crowding on four disordered proteins with very different degreesof charge-induced expansion. For a variety of crowding agents (PEG, PVP,Dextran, PVA), an increasing collapse of the polypeptide chains is observedwith increasing concentrations of crowder, as expected from simple consider-ations, such as scaled particle theory. However, we also observe an increasingcollapse with increasing size of the crowder, the opposite of what scaled parti-cle theory predicts.Interestingly, the observations can be rationalized quantitatively within theframework of Flory-Huggins theories that take into account the polymericproperties of both the disordered proteins and the crowder. The results providea step towards understanding the behavior of IDPs and denatured proteins in thepresence of polydisperse co-solutes as those characteristic of the cellularenvironment.

1364-Pos Board B94Single-Molecule Characterisation of Alpha-Synuclein OligomersMathew H. Horrocks1, Steven F. Lee1, Sonia Gandhi2, Marija Iljina1,Laura Tosatto1, Christopher M. Dobson1, David Klenerman1.1Cambridge University, CAMBRIDGE, United Kingdom, 2Institute ofNeurology, University College London, London, United Kingdom.The pathological hallmark of Parkinson’s disease is the presence of insolubleprotein deposits in the brain, which are formed when specific protein moleculesmisfold and aggregate into highly ordered fibrils. In Parkinson’s disease, thedeposits are primarily made up of alpha-synuclein, a protein whose major func-tion is not fully known. Rather than the fibrils themselves being toxic, evidencenow points towards the smaller, soluble oligomers formed in the initial stagesof the process as being the culprit. We have developed a novel single-moleculefluorescence technique to detect and characterise the oligomers of alpha-synuclein. Using this methodology, we are able to identify the cytotoxic spe-cies, and apply these species to primary neuronal cultures to investigate theirdamaging effects.

1365-Pos Board B95Repeats in the a-Synuclein Sequence Determine its Conformation onMembranes and Influence Aggregation PropertiesVolodymyr Shvadchak1, Vinod Subramaniam2,3.1Nanobiophysics, University of Twente, Enschede, Netherlands, 2NanoscaleBiophysics, FOM Institute AMOLF, Amsterdam, Netherlands, 3University ofTwente, Enschede, Netherlands.Alpha-Synuclein (aS) is a 140 aa intrinsically disordered and amyloidogenicprotein. Its physiological functions are unclear, but are believed to be con-nected to the interaction with synaptic vesicles or membranes of otherorganelles.The sequence of aS shows partial 11 amino acid periodicity that induces atypical3/11 helix conformation on membranes. Fluorescence studies of tryptophan mu-tants of aSpoint to a flexible breakat residues 52-54 between two helical domains.Deletion of this flexible 4 aa fragment between two groups of 11 aa repeats doesnot significantly affect aS membrane binding but strongly decreases the proteinaggregation and fibril formation propensity. Moreover, the deletion mutant in-hibits aggregation of wildtype aS, likely by hindering the fibril growth, sincethe mutant does not appear to co-aggregate into fibrils with wt aS.Introducing additional 11 amino acid repeats into the aS sequence increases af-finity of themodified protein tomembranes and slows down the protein aggrega-

tion. We believe that the 11 amino acidrepeats in the aS sequence play a key rolein aS’s ability to switch between a helicalconformation on membranes and b-sheetsin fibrils.