characterization of oligomerization–aggregation products of neurodegenerative target proteins by...

Upload: gustavo-duarte

Post on 03-Apr-2018

215 views

Category:

Documents


0 download

TRANSCRIPT

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    1/14

    Chapter 27

    Characterization of OligomerizationAggregation Productsof Neurodegenerative Target Proteins by Ion Mobility MassSpectrometry

    Camelia Vlad, Marius Ionut Iurascu, Stefan Slamnoiu,Bastian Hengerer, and Michael Przybylski

    Abstract

    Protein amyloidogenesis is generally considered to be a major cause of two most severe neurodegenerativedisorders, Parkinsons disease (PD) and Alzheimers disease (AD). Formation and accumulation of fibrillaraggregates and plaques derived from a-synuclein (a-Syn) and -amyloid (A) polypeptide in brain havebeen recognized as characteristics of Parkinsons disease and Alzheimers disease. Oligomeric aggregates ofa-Syn and A are considered as neurotoxic intermediate products leading to progressive neurodegenera-tion. However, molecular details of the oligomerization and aggregation pathway(s) and the molecularstructure details are still unclear. We describe here the application of ion-mobility mass spectrometry (IMS-MS) to the identification ofa-Syn and A oligomerizationaggregation products, and to the characteriza-

    tion of different conformational forms. IMS-MS is an analytical technique capable of separating gaseousions based on their size, shape, and topography. IMS-MS studies of soluble a-Syn and A-aggregatesprepared by in vitro incubation over several days were performed on a quadrupole time of flight massspectrometer equipped with a travelling wave ion mobility cell, and revealed the presence of differentconformational states and, remarkably, truncation and proteolytic products of high aggregating reactivity.These results suggest that different polypeptide sequences may contribute to the formation of oligomericaggregates of heterogeneous composition and distinct biochemical properties.

    Key words: Parkinsons disease, a-Synuclein, Alzheimers disease, -Amyloid, Oligomerization, Ionmobility mass spectrometry

    1. Introduction

    Soluble amyloid oligomers have been generally thought to beformed as common intermediates in the amyloid fibrillization path-

    way, and have been implicated as neurotoxic species of amyloid-related neurodegenerative diseases, such as Alzheimers disease(AD) and Parkinsons Disease (PD). In the last few years, increasing

    Vladimir N. Uversky and A. Keith Dunker (eds.), Intrinsically Disordered Protein Analysis:Volume 2, Methods and Experimental Tools, Methods in Molecular Biology, vol. 896,DOI 10.1007/978-1-4614-3704-8_27,# Springer Science+Business Media New York 2012

    399

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    2/14

    evidence has been obtained that soluble oligomers, and not highmolecular weight fibrillar end products, are highly toxic, even whenthey are formed from proteins that are not normally related toneurodegenerative processes (1, 2). Formation and accumulationof fibrillar plaques and misfolding- aggregation products of

    -amyloid peptide (A) and a-synuclein (a-Syn) in brain havebeen recognized as characteristics of Alzheimers disease (AD)and Parkinsons disease (PD) (36). Moreover, in recent years anincreasing number of proteins have been identified as being intrin-sically disordered (Intrinsically Disordered Proteins; IDPs), sug-gesting that they lack stable, folded tertiary structures underphysiological conditions (7). A number of intrinsically disorderedproteins have been shown, or are suspected to be associated

    with human diseases as cancer, cardiovascular disease, amyloidoses,neurodegenerative diseases, and diabetes. IDPs, such as a-synuclein

    involved in Parkinsons disease (8, 9), protein-Tau in Alzheimersdisease (AD) and related tauopathies (10), and prion protein (PrP)in prion disorders (PrD) such as CreutzfeldtJakob Disease (CJD),GerstmannStrausslerScheinker Syndrome and Fatal FamilialInsomnia (FFI) (11, 12) represent potentially crucial targets fordrugs that modulate their proteinprotein interactions.

    a-Synuclein, a protein of 140 amino acids, is mainly expressedin the presynaptic terminals of neurons and has been stronglycorrelated with Parkinsons disease, one of the most severe neuro-degenerative motor disorders (8, 9). a-Syn is a natively unfolded

    protein with unknown function and unspecific conformational het-erogeneity and properties. The main evidence for a causal role ofa-Syn in PD came from the discovery of three rare point mutations ofthe amino acid sequence, A53T (13), A30P (14), and E46K (15) infamilies with autosomal dominant Parkinsons Disease that lead toa-Syn accumulation in Lewy bodies and other pathological inclu-sions at conditions inducing PD (16).

    -Amyloid (A) is a neurotoxic polypeptide containing 3943amino acid residues and is derived from proteolytic cleavage of thetransmembrane A precursor protein (APP). Recently, the forma-

    tion of A-oligomers has become of particular interest, since oligo-meric aggregates have been suggested to be key neurotoxic speciesfor progressive neurodegeneration (3, 17, 18); however, moleculardetails of the pathophysiological degradation of APP, and of the

    A-aggregation pathways and structures are hitherto unclear (19).In recent years, mass spectrometry (MS) has become a major

    analytical tool in structural biology (20). In particular, electrospraymass spectrometry (ESI-MS) has emerged as a powerful techniquefor producing intact gas- phase ions from large biomolecular ions,and ions due to supramolecular complexes (21). However, in con-

    trast to the large number of ESI- mass spectrometric studies ofprotein structures, structure modifications and applications to pro-teomics (2228), the molecular characterization of protein

    400 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    3/14

    misfolding and aggregation species by mass spectrometry hadlittle success; possible explanations are (1) the low intermediateconcentrations and (2) and slow rates of aggregate formationin vitro (6, 7). Conventional soft-ionization mass spectrometrymethods such as ESI-MS and HPLC-MS are not suitable to direct

    in-situ analysis of conformational states and intermediates atdifferent concentrations. Recently, ion mobility mass spectrometry(IMS-MS) is emerging as a new tool to probe complex biomolecu-lar structures from solution phase structures, due to the potential ofIMS-MS for separation of mixtures of protein complexes by con-formational state, and spatial shape and topology (2936). TheIMS-MS instrument employed in this study consists of two parts:(1) an ion mobility drift cell where ions are separated within anelectric field according to their collisional cross sections and (2) aquadrupole time-of-flight mass spectrometer (Synapt-QTOF-MS)

    (33, 34, 3638). Thus, IMS-MS implements a new mode of sepa-ration that allows the differentiation of protein conformationalstates. In the present study, ESI-MS (21) and IMS-MS have beenapplied to the direct analysis of mixtures of a-Syn and A (38)aggregation products in vitro, formed during prolonged incuba-tion times.

    2. Materials

    2.1. Preparation

    and Characterization

    ofa-Synuclein and

    A(140) Oligomers

    Alpha-Syn and A (140) oligomers were prepared by an in vitroincubation procedure for up to 7 days at 37C and at 20C (38, 39)(see Note 2). a-Syn oligomers type A1 were prepared by dissolvingthe protein in 50 mM sodium phosphate buffer (pH 7.0) contain-ing 20 % ethanol, to a final concentration of 7 mM (39). In the caseof oligomers type A2, 10 mM FeCl3 was added, while oligomerstype A1 were prepared without adding FeCl3. After 4 h of shaking,both types of oligomers were lyophilized and resuspended in50 mM sodium phosphate buffer (pH 7.0) containing 10 % ethanol

    with a half of the starting volume. This was followed by incubationfor 7 days at room temperature For A-oligomerization, A(140)was solubilized at a concentration of 1 mg/mL in a buffer containing50 mM Na3PO4, 150 mM NaCl, 0.02 % NaN3 at pH 7.5 (38).a-Syn and A(140) oligomers were separated by gel electrophore-sis and subjected to IMS-MS measurements.

    2.2. Gel Electrophoresis

    ofa-Synuclein and

    A(140) Oligomers

    and Aggregates

    Separation of a-Syn and A (140) oligomers was performed byone-dimensional Tristricine polyacrylamide gel electrophoresis(Tristricine PAGE) on a Mini-Protean II or Mini-Protean 3 elec-

    trophoresis cell (BioRad, M

    unchen, Germany) using 12 % and 15 %polyacrylamide gel electrophoresis, and protein bands visualizedby Coomassie Blue staining. The dimensions of the gel were

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 401

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    4/14

    90 60 1 mm. The proteins were solubilized and denaturedusing a stock solution of the twofold concentrated sample buffercontaining 4 % SDS, 25 % Glycerin, 50 mM TrisHCl, 0.02 %Coomassie, 6 M urea, pH 6.8. Gels were run at 60 V until thetracking dye entered the separating gel and at 120 V until the

    tracking dye reached the bottom of the gel.

    2.3. Buffers and Stock

    Solutions

    The following solutions were prepared in ultrapure water (preparedby purifying deionized water to attain a conductivity of 18 MO):

    1. a-Syn oligomers type A1 buffer: 50 mM sodium phosphatebuffer, pH 7.0. Weigh 1.779 g Na2HPO4 and transfer to a200-ml graduated glass beaker. Add ultrapure water to a

    volume of 200 ml. Mix and adjust pH with HCl (see Note3). Add 40 ml ethanol. Store at 4C.

    2. a-Syn oligomers type A2 buffer: 50 mM sodium phosphatebuffer, pH 7.0. Weigh 1.779 g Na2HPO4 and transfer to aglass beaker. Add ultrapure water to a volume of 200 ml. Mixand adjust pH with HCl (see Note 3). Add 40 ml ethanol and0.3244 mg FeCl3. Store at 4

    C.

    3. A(140) oligomers buffer: 50 mM Na3PO4, 150 mM NaCl,0.02 % NaN3 at pH 7.5. Weigh 1.9 g Na3PO4 and 0.88 gNaCl and transfer to a 100-ml graduated glass beaker contain-ing about 80 ml ultrapure water. Mix and adjust pH with HCl(see Note 3). Make up to 100 ml with ultrapure water. Store

    at 4C.

    For Tristricine PAGE, the following solutions were preparedin ultrapure water.

    4. TrisHCl/SDS buffer: 3 M Tris, 0.3 % SDS, pH 8.45. Weight36.4 g Tris and transfer to a 100 ml graduated glass beakercontaining about 60 ml ultrapure water. Mix and adjust pH

    with HCl to 8.45 (see Note 3). Add 0.3 g SDS and make up to100 ml with ultrapure water.

    5. 2 Tricine sample buffer: 100 mM Tris pH 6.8, 24 % Glyc-

    erol, 8 % SDS, 0.02 % Coomassie brilliant blue G-250. Weigh1.21 g Tris and transfer to a 10-ml graduated glass beaker.

    Adjust pH with HCl to 6.8 (see Note 3). Add 2.4 ml Glycerol,0.8 g SDS (see Note 4), and 2 mg Coomassie brilliant blue G-250. Store at 4C.

    6. 1 Cathode buffer (upper chamber): 100 mM Tris, 100 mMTricine, 0.1 % SDS. Weigh 6.055 g Tris, 8.96 g Tricine, 0.5 gSDS and transfer to a 500-ml graduated glass beaker andmake up with ultrapure water.

    7. 1 Anode buffer (lower chamber): 20 mM Tris, pH 8.96.Weigh 12.11 g Tris and transfer to a 500-ml graduated glassbeaker containing about 400 ml ultrapure water. Mix and

    402 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    5/14

    adjust pH with HCl to 8.96 (see Note 3). Make up withultrapure water to 500 ml.

    8. Ammonium persulfate (APS): 10 % solution in water (seeNote 5).

    9. N0

    ,N0

    ,N0

    ,N0

    -tetramethyl-ethylenediamine (TEMED).10. 15 % separating gel: Mix 7.5 ml Acrylamide, 5 ml TrisHCl/

    SDS and 0.9 ml ultrapure water in a glass beaker. Add 1.58 mlGlycerol and sonicate until total solubilization. Add 75 ml

    APS, 10 ml TEMED and pour the gel into casting platesleaving room at the top for the stacking gel and comb. Addultrapure water to cover top of gel and make flat interface.Polymerize for about 30 min. Check that remaining gel inglass beaker is polymerized before proceeding.

    11. Stacking gel: Mix in a glass beaker 972 ml Acrylamide,

    1.86 ml TrisHCl/SDS, 4.67 ml ultrapure water, 40 mlAPS and 7.5 ml TEMED and pour gel on top of alreadypolymerized separating gel. Insert comb and allow to poly-merize (about 30 min).

    3. Methods

    3.1. Gel ElectrophoreticIsolation and Primary

    Structure

    Characterization

    ofa-Synuclein

    Oligomerization

    Aggregation

    Products

    The in vitro oligomerization products of a-Syn were firstcharacterized by Tristricine PAGE, as described in Subheadings 2.1and 2.2, and the isolated protein bands analyzed by mass spectrom-etry. The electrophoretic separation revealed bands with molecular

    weights corresponding to monomeric (a-Syn)1, dimeric (a-Syn)2,oligomeric (a-Syn)

    nproteins, and three additional bands below the

    a-Syn monomer that were assigned to truncated forms ofa-Syn (seeFig. 1). The band in the region of approximately 37 kDa was excisedfrom the gel, digested with trypsin, and analyzed by HPLC-MS withan ESI-ion trap mass spectrometer (see Note 6). The mass spectro-

    metric data ascertained the presence ofa-Syn peptides by identifica-tion of the corresponding peptide fragments, andidentified ana-Syndimer composed of two intact a-Syn polypeptides. This result wasascertained by N-terminal Edman sequence determination (seeFig. 1c) (see Note 7). Following electrophoretic separation, thebands were transferred onto a PVDF membrane and subjected toEdman sequencing which provided the identification of N-terminaltruncation of the band at 13.9 kDa, a-Syn(5140). Direct massspectrometric analysis of monomer (a-Syn)1 was performed withan Esquire 3000+ ion trap mass spectrometer (Bruker Daltonics,

    Bremen, Germany). The protein band was solubilized in 1 % formicacid as a volatile solvent, to a final concentration of 30 mM, pH 2.5.

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 403

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    6/14

    The spectrum was obtained in the positive ion mode (see Fig. 1b).Inthe ESI mass spectrum, a series of multiply charged ions wereobtained with the most abundant ion at charge state +19.

    3.2. Ion-Mobility Mass

    Spectrometry

    Ion-mobility mass spectrometry was performed with a SYNAPT-QTOF-MS (Waters, Manchester, UK) equipped with an nano-electrospray ionization source (2225). The ions were passedthrough a quadrupole and either set to transmit a substantial massrange or to select a particular m/z before entering the Triwave ionmobility cell. The Triwave system (22) is composed of three

    T-Wave devices (Waters SYNAPT). The first device, theTrap T-Wave, accumulates ions which are stored and gated(500 msec) into the second device, the ion mobility T-Wave cell,

    where they are separated according to their cross section-dependent mobilities (22, 23). The final transfer T-Wave is usedto transport the separated ions into the orthogonal acceleration oa-TOF for MS analysis. The injection volume was 5 ml, and a priordesalting step was performed on a cartridge for 10 min at 20 ml/min with gradient of 1090 % acetonitrile. The mass spectrometricacquisition characteristics were: 3504,000 m/z, cone voltage

    25 V, IMS pressure 0.45 bar, wave height 515 V. IMS-MS dataare inherently three-dimensional, consisting of mass, drift time andintensity (relative abundance) for all ions observed.

    Fig. 1. (a), Primary structure of a-synuclein (a-Syn). (b), TrisTricine PAGE of a-Syn stained with Coomassie Blue:

    molecular weight marker (lane 1); (A1) a-Syn incubated in PBS buffer at room temperature for 7 days (lane 2); (A2) a-Syn

    incubated in PBS buffer with 10 mM FeCl3 at 20C for 7 days (lane 3). (c), Edman sequence determination of (a-Syn)2; ESI-

    MS of a-Syn in 1 % aqueous formic acid at a final concentration of 34.5 mM, pH 2.5 showing the 15+ to 23+ charged

    molecular ions; Edman sequencing of N-terminal truncated polypeptide sequence of 13.9 kDa, a-Syn(5140).

    404 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    7/14

    3.3. Ion Mobility

    Mass Spectrometry

    ofa-Syn- Oligomers

    Figure 2 summarizes the ion-mobility- MS drift time profiles of afreshly prepared a-Syn solution, and a-Syn- oligomers in incuba-tion mixtures type A1, A2 and A1 + A2 after 7 days of incubation inphosphate-buffered saline with 20 % ethanol, 10 mM FeCl3 (driftscope data view). In the different charge state series of ion signals,a-Syn monomer, dimer as well as proteolytic and truncated peptidesequences were identified by molecular weight determinations andpartial tandem-MS sequence determinations (see Figs. 3 and 4a, b).

    The comparison of the extracted drift time mobilograms of theion at m/z 804.1 (18+) between a-Syn oligomers type A1, A2, andA1 + A2 is shown in Fig. 3. In the a-Syn oligomers type A1, twodrift time (conformational) states are present, while in type A2 and

    A1 + A2 even three drift time states are observed indicating threedifferent structural types. The extracted, deconvoluted mass spectraof the drift scope profile due to the a-Syn oligomers, type A1revealed the a-Syn monomer and the intact full-length dimer (seeFig. 4a; m.w. 14,459; 29.919 Da), while the oligomer peak, type

    A1 + A2 showed an additional truncated polypeptide, a-Syn

    (40140) (see Fig. 4b; m.w. 10.437 Da); further proteolyticpeptide fragments of a-Syn with high aggregating reactivity,corresponding to the proteolytic bands observed by gel electropho-resis (see Fig. 1) have been recently identified (40).

    Fig. 2. Ion-mobility drift time of a-Syn after 7 days of incubation in 50 mM sodium phosphate buffer, containing 20 %

    ethanol to a final concentration of 7 mM (A1); Syn after 7 days of incubation in 50 mM sodium phosphate buffer, containing

    20 % ethanol, 10 mM FeCl3 (A2); freshly prepared a-Syn. Signals corresponding to monomers, dimers, and truncated

    peptide sequences are observed.

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 405

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    8/14

    3.4. Ion Mobility Mass

    Spectrometry

    of A-Oligomers

    The soluble fraction of the A(140) fibril preparation obtained byincubation over 5 days at 37C, and a freshly prepared A(140)peptide solution (220 mM) were subjected to comparative analysisby ion mobility- MS (see Fig. 5a, b). In the freshly preparedsolution of A(140), the [M+5H]5+ ion was predominant, whilein soluble fibril preparations ions with increasingly higher chargestates ([M+6H]6+, [M+7H]7+) were most abundant, indicative of

    oligomer formation. In the gel electrophoretic analysis, the forma-tion of A- oligomers can be directly observed (38). The signal-to-noise ratios of the [M+5H]5+ and [M+6H]6+ ions was found to besignificantly lower in the fibril preparation sample compared to thesample of freshly prepared A(140), suggesting a decreasedamount of A(140) monomer due to the formation of oligomers.The extracted ion mobility profiles for the [M+5H]5+ ion of thefreshly prepared A(140) (see Fig. 5c) compared to that of thefibril preparation (see Fig. 5d) indicate the presence of differentconformational states (A and B) with different ion mobilities (cross

    sections), indicative of the oligomerization process. Furthermore,partial oxidation of A at the Met-35 residue has been observed insamples upon formation of oligomers (38).

    sample alphasyn

    Scan

    25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 125 130 135 140 145 150 155 160 165

    %

    0

    100

    25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 125 130 135 140 145 150 155 160 165

    %

    0

    100

    25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 125 130 135 140 145 150 155 160 165

    %

    0

    100

    25 30 35 40 45 50 55 60 65 70 75 80 85 90 95 100 105 110 115 120 125 130 135 140 145 150 155 160 165

    %

    0

    100

    3282_D025_dt_01 TOF MS ES+804.074_804.52 0.10Da

    937

    6.30762.1

    3282_D024_dt_01 TOF MS ES+804.164_804.602 0.10Da

    440

    6.66804.3

    3282_D023_dt_01 TOF MS ES+804.106_804.602 0.10Da

    901

    6.39804.3

    3282_D022_dt_01 TOF MS ES+804.106_804.52 0.10Da

    4.26e3

    6.12;851.5

    A1 +A2(7 days)

    A2

    (7 days)

    a

    b

    c

    d

    A1

    (7 days)

    -Syn(0 days)

    Fig. 3. Comparison of the extracted mobilograms of the ion m/z 804.1 between a-Syn oligomers type A1, A2, A1 + A2. In

    a-Syn oligomers type A1 two structures are observed; in type A2 and A1 + A2 three main forms are present.

    406 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    9/14

    TOF MS ES+14459.4

    mass10000 12000 14000 16000 18000 20000 22000 24000 26000 28000

    %

    0

    1002.30e4

    28919.6

    -Syn (1-140)

    monomer

    -Syn (1-140)

    dimer

    MDVFMKGLSK AKEGVVAAAE KTKQGVAEAA GKTKEGVLYV GSKTKEGVVH GVATVAEKTK EQVTNVGGAV

    VTGVTAVAQK TVEGAGSIAA ATGFVKKDQL GKNEEGAPQE GILEDMPVDP DNEAYEMPSE EGYQDYEPEA

    1

    71 140

    -Syn (1-140)

    Mw exp : 14459.4 Da

    a

    Mw calc : 14460.1 Da

    m = 48.41 ppm

    mass10000 11000 12000 13000 14000 15000 16000 17000 18000 19000

    %

    0

    100

    b

    TOF MS ES+7.93e3

    14461.0010

    10437.0010

    10457.0010

    14477.0010

    -Syn (1-140)

    -Syn (40-140)

    -Syn (1-140)

    Mw exp : 14460.0 Da

    Mw calc : 14460.1 Da

    m = 6.92 ppm

    -Syn (40-140)

    Mw exp : 10437.0 Da

    Mw calc : 10436.4 Da

    m = 57.49 ppm

    Fig. 4. Extracted deconvoluted spectra of a-Syn oligomers: (a), Type A1 oligomers are showing a-Syn monomer plus

    dimer; (b), type A1 + A2 oligomers are containing truncated polypeptide sequence at 10.5 kDa (a-Syn(40140)).

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 407

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    10/14

    m/z400 500 600 700 800 900 1000 1100 1200 1300 1400 1500 1600 1700 1800 1900 2000 2100 2200

    %

    0

    100

    %

    0

    100

    3282_D019_dt_01 61 (5.400) Cm (26:104) TOF MS ES+2.02e3866.4890

    722.2233

    719.2616

    702.7539616.7699

    616.6233

    602.9120

    600.3329

    619.3473

    722.4017

    866.2936

    722.5703

    739.4684

    843.4893

    739.6289

    843.3071

    840.8661

    739.8396

    741.4360

    866.6954

    866.9017

    867.1082

    867.31461083.1299

    900.78231082.6321

    901.0260

    924.3208

    1083.3848

    1083.6399

    1083.8828

    3282_D018_dt_01 62 (5.490) Cm (30:141)

    TOF MS ES+297722.2233

    719.5585

    703.3997

    619.3381

    616.7699

    612.3430

    401.1952610.3362

    461.2322

    619.4943

    722.5703

    866.6737

    722.7389

    866.4781

    732.2323

    739.6188

    866.2610

    739.6589

    741.4160

    866.8800

    867.0756

    878.2579

    878.2799

    878.4877

    900.4834

    900.7712 1083.0813

    5+

    5+

    6+

    6+

    4+

    4+

    7+

    7+

    A

    B

    a

    b

    c

    d

    Scan2 6 2 8 3 0 3 2 3 4 3 6 3 8 4 0 42 4 4 4 6 4 8 5 0 52 5 4 5 6 5 8 6 0 62 6 4 6 6 6 8 7 0 72 74 7 6 7 8 8 0 8 2 8 4 8 6 8 8 9 0 92 9 4 9 6 9 8 100 1 02

    %

    0

    100

    26 28 3 0 3 2 3 4 3 6 38 40 4 2 4 4 4 6 4 8 5 0 5 2 5 4 5 6 58 60 6 2 6 4 6 6 6 8 70 7 2 7 4 7 6 7 8 80 8 2 8 4 8 6 8 8 90 92 9 4 9 6 9 8 100 102

    %

    0

    1005+866.6

    5+866.6

    Drift Time (scans)40 60 80 100

    Fig. 5. Ion mobility MS analysis of (a) freshly prepared A(140); (b) supernatant after 5 days incubation; (c) and (d)extracted ion mobility profiles for m/z 866.6, [M+5H]5+ from freshly prepared A(140) and supernatant, respectively.

    408 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    11/14

    4. Notes

    Ion mobility mass spectrometry has been shown to be an efficienttool for the characterization of in vitro a-Syn and A oligomeriza-tionaggregation products, and provides evidence of (1), the pres-ence of distinct conformational forms of a-Syn and (2), thepresence of at least two different conformational forms involvedin A- aggregation. These results suggest IMS-MS as a powerfultool for the analysis of reactive intermediates in the aggregation ofintrinsically disordered proteins due to its affinity-like gas phaseseparation capability. In the case ofa-Syn, the IMS-MS data iden-tified proteolytic fragments as key molecular species involved inmisfolding and aggregation pathways, suggesting that the IMS-MS method can also be successfully applied to the identification ofhitherto undetected proteolytic truncation products of neurode-generative target proteins. Thus, gel electrophoresis protein Tau,another key protein in Alzheimers disease shows the formation oftruncation/proteolytic fragments in addition to oligomerizationproducts (see Fig. 6). It will be of interest to systematically exploreIMS-MS in the study of aggregation pathways of intrinsically disor-dered proteins.

    1. a-Syn was purified prior to use by analytical RP-HPLC on aUltiMate 3000 system (Dionex, Germering, Germany),

    Fig. 6. SDS-PAGE separation of two isoforms of protein-Tau: Molecular weight marker(lane 1); 2 mg Tau2N/4R protein (lane 2) and 2 mg Tau0N/3R protein (lane 3) showing

    bands corresponding to monomeric (Tau)1, oligomeric (Tau)nv, and truncated forms of Tau,

    isoforms DN(Tau).

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 409

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    12/14

    equipped with a Vydac C4 column (250 4.6 mm I.D.) with5 mm silica (300 A pore size) (Hesperia CA). Linear gradientelution (0 min0 % B; 5 min0 % B; 50 min90 % B) witheluent A: (0.1 % trifluoroacetic acid in water) and eluent B:(0.1 % trifluoroacetic acid in acetonitrilewater 80:20 v/v) was

    employed at a flow rate of 1 ml/min.

    2. A(140) was synthesized by solid phase peptide synthesis on aNovaSyn TGR resin using Fmoc chemistry by a semiautomatedsynthesizer EPS-221 (Abimed, Germany). Fmoc amino acids,NovaSyn TGR resin, PyBop, and other reagents were obtainedfrom Novabiochem (Laufelfingen, Switzerland). The crudeproducts were purified by analytical RP-HPLC (see Note 1).

    3. Concentrated HCl (12N) can be used at first to narrow the gapfrom the starting pH to the necessary pH and further use HCl

    (1N) until the pH 7 is obtained.4. SDS precipitates at 4C. Therefore, the 2 Tricine sample

    buffer needs to be warmed prior to use.

    5. The best is to prepare fresh solution each time.

    6. Protein spots were excised manually from the gel, cutting asclose to the spots as possible, in order to minimize the contami-nation with SDS. After distaining, the dried gel pieces wereswollen in digestion buffer (12 ng/ml TPCK-trypsin in50 mM NH4HCO3) at 4

    C (on ice) for 45 min, and then theywere incubated at 37C overnight (12 h) and at last the peptideswere extracted twice with acetonitrile0.1 % trifluoroacetic acid3:2.

    7. The spots of interest were excised, destained with 100 % meth-anol, and applied into the sequencing cartridge. Sequencedeterminations were performed on an Applied BiosystemsModel 494 Procise Sequencer attached to a Model 140 CMicrogradient System, a 785A Programmable AbsorbanceDetector, and a 610A Data Analysis System.

    Acknowledgments

    We thank Adrian Moise for expert assistance with Edman sequenceand Nick Tomczyk and Emmanuelle Claude, Waters MS- Technol-ogy Centre Manchester, UK, for assistance with the ion mobility-MS. We are grateful to David Clemmer and Nicolas Pearson forhelpful discussion and comments on IMS-MS. This work has been

    supported by the Graduate School Chemical Biology, the ResearchCentre Proteostasis, University of Konstanz, and the Landesstif-tung fur Wissenschaft und Forschung Baden- Wurttemberg.

    410 C. Vlad et al.

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    13/14

    References

    1. Bucciantini M, Giannoni E, Chiti F, Baroni F,Formigli L, Zurdo J, Taddei N, Ramponi G,Dobson CM, Stefani M (2002) Inherent toxic-

    ity of aggregates implies a common mechanismfor protein misfolding diseases. Nature416:507511

    2. Kayed R, Sokolov Y, Edmonds B, McIntireTM, Milton SC, Hall JE, Glabe CG (2004)Permeabilization of lipid bilayers is a commonconformation-dependent activity of solubleamyloid oligomers in protein misfoldingdiseases. J Biol Chem 279:4636346366

    3. Morgan D (2006) Immunotherapy for Alzhei-mers disease. J Alzheimers Dis 9:425432

    4. Shults CW (2006) Lewy bodies. Proc Natl

    Acad Sci U S A 103:166116685. Goedert M (2001) Alpha-synuclein and neuro-degenerative diseases. Nat Rev Neurosci2:492501

    6. Uversky VN (2007) Neuropathology,biochemistry, and biophysics of alpha-synucleinaggregation. J Neurochem 103:1737

    7. Dyson HJ, Wright PE (2005) Intrinsicallyunstructured proteins and their functions. NatRev Mol Cell Biol 6:197208

    8. Maroteaux L, Campanelli JT, Scheller RH(1988) Synuclein: a neuron-specific protein

    localized to the nucleus and presynaptic nerveterminal. J Neurosci 8:28042815

    9. Dawson TM, Dawson VL (2003) Molecularpathways of neurodegeneration in Parkinsonsdisease. Science 302:819822

    10. Tolnay M, Probst A (1999) Review: tau proteinpathology in Alzheimers disease and relateddisorders. Neuropathol Appl Neurobiol25:171187

    11. Johnson RT, Gibbs CJ (1998) Creutzfeldt-Jakob disease and related transmissible spongi-form encephalopathies. N Engl J Med

    339:1994200412. Horiuchi M, Caughey B (1999) Prion protein

    interconversions and the transmissible spongi-form encephalopathies. Structure (London,England) 7:231240

    13. Polymeropoulos MH, Lavedan C, Leroy E, IdeSE, Dehejia A, Dutra A, Pike B, Root H,Rubenstein J, Boyer R et al (1997) Mutationin the alpha-synuclein gene identified infamilies with Parkinsons disease. Science276:20452047

    14. Kruger R, Kuhn W, Muller T, Woitalla D,

    Graeber M, Kosel S, Przuntek H, Epplen JT,Schols L, Riess O (1998) Ala30Pro mutation in

    the gene encoding alpha-synuclein in Parkin-sons disease. Nat Genet 18:106108

    15. Zarranz JJ, Alegre J, Gomez-Esteban JC, Lez-

    cano E, Ros R, Ampuero I, Vidal L, HoenickaJ, Rodriguez O, Atares B et al (2004) The newmutation, E46K, of alpha-synuclein causes Par-kinson and Lewy body dementia. Ann Neurol55:164173

    16. Saha AR, Hill J, Utton MA, Asuni AA, AckerleyS, Grierson AJ, Miller CC, Davies AM, Buch-man VL, Anderton BH et al (2004) Parkin-sons disease alpha-synuclein mutationsexhibit defective axonal transport in culturedneurons. J Cell Sci 117:10171024

    17. Salminen A, Ojala J, Kauppinen A, Kaarniranta

    K, Suuronen T (2009) Inflammation in Alzhei-mers disease: amyloid-beta oligomers triggerinnate immunity defence via pattern recogni-tion receptors. Prog Neurobiol 87:181194

    18. Crews L, Tsigelny I, Hashimoto M, Masliah E(2009) Role of synucleins in Alzheimers dis-ease. Neurotox Res 16:306317

    19. Hull M, Berger M, Heneka M (2006) Disease-modifying therapies in Alzheimers disease:how far have we come? Drugs 66:20752093

    20. Heck AJ, Van Den Heuvel RH (2004) Inves-tigation of intact protein complexes by massspectrometry. Mass Spectrom Rev 23:368389

    21. Fenn JB, Mann M, Meng CK, Wong SF,Whitehouse CM (1989) Electrospray ioniza-tion for mass spectrometry of large biomole-cules. Science 246:6471

    22. Schulenborg T, Schmidt O, van Hall A,Meyer HE, Hamacher M, Marcus K (2006)Proteomics in neurodegenerationdiseasedriven approaches. J Neural Transm113:10551073

    23. Mann M, Hendrickson RC, Pandey A (2001)Analysis of proteins and proteomes by massspectrometry. Annu Rev Biochem 70:437473

    24. Perdivara I, Petrovich R, Allinquant B, Deterd-ing LJ, Tomer KB, Przybylski M (2009) Eluci-dation of O-glycosylation structures of thebeta-amyloid precursor protein by liquidchromatography-mass spectrometry usingelectron transfer dissociation and collisioninduced dissociation. J Proteome Res8:631642

    25. Perdivara I, Deterding L, Moise A, Tomer KB,Przybylski M (2008) Determination of primarystructure and microheterogeneity of a beta-

    amyloid plaque-specific antibody using high-

    27 Characterization of OligomerizationAggregation Products of Neurodegenerative. . . 411

  • 7/28/2019 Characterization of OligomerizationAggregation Products of Neurodegenerative Target Proteins by Ion Mobility Ma

    14/14

    performance LC-tandem mass spectrometry.Anal Bioanal Chem 391:325336

    26. Przybylski M, Glocker MO (1996) Electro-spray mass spectrometry of biomacromolecularcomplexes with noncovalent interactionsnewanalytical perspectives for supramolecular

    chemistry and molecular recognition processes.Angew Chem Int Ed Engl 35:806826

    27. Kebarle P, Verkerk UH (2009) Electrospray:from ions in solution to ions in the gas phase,

    what we know now. Mass Spectrom Rev28:898917

    28. Nemes P, Goyal S, Vertes A (2007) Conforma-tional and noncovalent complexation changesin proteins during electrospray ionization. AnalChem 80:387395

    29. Ruotolo BT, Benesch JL, Sandercock AM,Hyung SJ, Robinson CV (2008) Ion

    mobility-mass spectrometry analysis of largeprotein complexes. Nat Protoc 3:11391152

    30. Bernstein SL, Dupuis NF, Lazo ND, Wytten-bach T, Condron MM, Bitan G, Teplow DB,Shea JE, Ruotolo BT, Robinson CV, BowersMT (2009) Amyloid- protein oligomeriza-tion and the importance of tetramers and dode-camers in the aetiology of Alzheimers Disease.Nat Chem 1:326331

    31. Thalassinos K, Slade SE, Jennings KR, ScrivensJH, Giles K, Wildgoose J, Hoyes J, BatemanRH, Bowers MT (2004) Ion mobility mass

    spectrometry of proteins in a modified com-mercial mass spectrometer. Int J Mass Spec-trom 236:5563

    32. Bleiholder C, Dupuis NF, Wyttenbach T,Bowers MT (2011) Ion mobility-mass spec-trometry reveals a conformational conversionfrom random assembly to -sheet in amyloidfibril formation. Nat Chem 3:172177

    33. Ruotolo BT, Hyung SJ, Robinson PM, Giles K,Bateman RH, Robinson CV (2007) Ionmobility-mass spectrometry reveals long-lived,unfolded intermediates in the dissociation of

    protein complexes. Angew Chem Int Ed Engl46:80018004

    34. Trimpin S, Clemmer DE (2008) Ion mobilityspectrometry/mass spectrometry snapshots forassessing the molecular compositions of com-plex polymeric systems. Anal Chem

    80:9073908335. Kanu AB, Dwivedi P, Tam M, Matz L, Hill HH

    Jr (2008) Ion mobility-mass spectrometry.J Mass Spectrom 43:122

    36. Henderson SC, Valentine SJ, Counterman AE,Clemmer DE (1999) ESI/ion trap/ion mobil-ity/time-of-flight mass spectrometry for rapidand sensitive analysis of biomolecular mixtures.

    Anal Chem 71:291301

    37. Zhou M, Sandercock AM, Fraser CS, RidlovaG, Stephens E, Schenauer MR, Yokoi-Fong T,Barsky D, Leary JA, Hershey JW et al (2008)

    Mass spectrometry reveals modularity and acomplete subunit interaction map of theeukaryotic translation factor eIF3. Proc Natl

    Acad Sci U S A 105:1813918144

    38. Iurascu MI, Cozma C, Tomczyk N, Rontree J,Desor M, Drescher M, Przybylski M (2009)Structural characterization of beta-amyloidoligomer-aggregates by ion mobility massspectrometry and electron spin resonance spec-troscopy. Anal Bioanal Chem 395:25092519

    39. Danzer KM, Haasen D, Karow AR, MoussaudS, Habeck M, Giese A, Kretzschmar H, Hen-

    gerer B, Kostka M (2007) Different species ofalpha-synuclein oligomers induce calciuminflux and seeding. J Neurosci 27:92209232

    40. Vlad C, Lindner K, Karreman C, SchildknechtS, Leist M, Tomczyk N, Rontree J, LangridgeJ, Danzer K, Ciossek T, Petre A, Gross ML,Hengerer B, Przybylski M (2011) Autoproteo-lytic fragments are intermediates in the oligo-merization/aggregation of the Parkinsonsdisease protein alpha-synuclein as revealed byion mobility mass spectrometry. Chem BioChem 12:27402744

    412 C. Vlad et al.