+ All Categories
Home > Documents > Aggregation Insolubility Soluble aggregates...

Aggregation Insolubility Soluble aggregates...

Date post: 03-Feb-2021
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
66
Aggregation Insolubility Soluble aggregates Stickiness Main headache for protein purifiers
Transcript
  • Aggregation

    Insolubility

    Soluble aggregates

    Stickiness

    Main headache for protein

    purifiers

  • 03072014 Superose12Analytic MBPAAshortNleD 48hrs concX3ish drop in conc001:10_UV1_280nm 03072014 Superose12Analytic MBPAAshortNleD 48hrs concX3ish drop in conc001:10_UV2_260nm 03072014 Superose12Analytic MBPAAshortNleD 48hrs concX3ish drop in conc001:10_UV3_220nm 03072014 Superose12Analytic MBPAAshortNleD 48hrs concX3ish drop in conc001:10_Fractions 03072014 Superose12Analytic MBPAAshortNleD 48hrs concX3ish drop in conc001:10_Inject

    0

    100

    200

    300

    400

    mAU

    6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 ml

    F3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17

    01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV1_280nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV2_260nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV3_220nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Cond 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Fractions 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Inject

    -20.0

    -10.0

    0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    60.0

    mAU

    60 80 100 120 140 ml

    F3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31

    01072014 MBP AA NleD short NiSeph4ml001:10_UV1_280nm 01072014 MBP AA NleD short NiSeph4ml001:10_UV2_260nm 01072014 MBP AA NleD short NiSeph4ml001:10_Conc 01072014 MBP AA NleD short NiSeph4ml001:10_Fractions 01072014 MBP AA NleD short NiSeph4ml001:10_Inject

    0

    100

    200

    300

    400

    500

    600

    700

    mAU

    40 60 80 100 120 140 ml

    F4 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27

    POOL 14-18

    03072014 Superose12Anal MBPAAshortNleD 48hr no conc001:10_UV1_280nm 03072014 Superose12Anal MBPAAshortNleD 48hr no conc001:10_UV2_260nm 03072014 Superose12Anal MBPAAshortNleD 48hr no conc001:10_UV3_220nm 03072014 Superose12Anal MBPAAshortNleD 48hr no conc001:10_Fractions 03072014 Superose12Anal MBPAAshortNleD 48hr no conc001:10_Inject

    -2.0

    0.0

    2.0

    4.0

    6.0

    8.0

    mAU

    6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 ml

    F3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17

    S P Unb W 4 16 18 28 1 11 16 17 18 19

    Sup & Pellet & Frac 16 (Ni): half amount loaded.

    ~75

    ~25

    ~48

    ~35

    ~63

    Soft aggregation: typical example

    Ni column

    SEC preparative Superose 12

    SEC analyticalSame day

    SEC analyticalAfter 48hr

    Vinograd Nina/ Oded Livnah lab

  • 150 200 250 300 350 400 450 500-50

    0

    50

    100

    150

    200

    250

    300

    OD

    (m

    AU

    )

    ml

    280 nm 260 nm

    100 150 200 250 300 350 400 450 500

    0

    250

    500

    750

    1000 280 nm 260 nm

    OD

    (m

    AU

    )

    ml

    %B

    0

    20

    40

    60

    80

    100

    300 350 400 450 500 550 600

    0

    500

    1000

    1500

    2000

    2500

    mA

    U

    ml

    % B

    0

    20

    40

    60

    80

    100

    100 150 200 250 300 350 400 450

    0

    100

    200

    300

    400

    500

    600

    700

    mA

    U

    ml

    TEV protease production: buffer optimization

    Capture Ni Column Polishing GF step

    Increase lysis buffer volume

    More DNase A in the lysis buffer

    Increase [NaCl] from 0.1 to 0.5

    Ronen Gabizon / Assaf Friedlerlab

  • Multiple pathways by which monomer protein in solution

    can form aggregates

    ‘‘Large’’ aggregates (>10 –mer):

    reversible if non-covalent

    ‘‘Very large’’ aggregates (50nm to

    3000 nm): reversible if non-covalent

    Visible particulates (‘‘snow’’):

    probably irreversible

    S. Webster (2013) PHARMACEUTICAL TECHNOLOGY Volume 37, Issue 11, pp. 42-48

  • • Solubility in a lysate extract & integrity according to PAGE-SDS, are very popular but naïve

    markers of protein health. Since: is the protein correctly folded? Has the proper oligomeric

    conformation? Is the protein active? Is the protein intact?

    • What is the connection between insoluble expression as inclusion body and soluble

    aggregates?

    • Does “solubility during expression” guarantee solubility after purification and storage?

    • Protein solubility and stability (in time), depends of proper conditions: buffer , concentration

    • Correct conditions must be found for each protein

    • Conformation and integrity can change with storage

    Healthy protein: soluble, proper oligomeric conformation and more….

  • At what part of the process does aggregation start?

    Do I have to deal with one, two or all the steps?

    Expression

    Cell lysis/sample

    preparation/buffer

    Purification

    Concentration

    Storage

    Partial solutions can be found by optimizing each of these steps

    Changing a single parameter at a time: time-consuming and often frustrating approach

    How to avoid extensive and laborious screening

    We need a strategy that integrates optimization of all these steps

    A simple and modular toolbox

  • What is good expression?

    Soluble or insoluble (IB) Expression

    It is not enough!!!

    What must be checked is the oligomerization state of the protein:Monomer – Soluble aggregate? / Folded – Unfolded?

    There are soluble aggregates

    Protein yield after first capture step (IMAC, etc)

    It is also not enough!!!

    Oligomerization state over time: Aggregation is time dependent!!!

  • 8

    Fusion proteins

    Optimize purification procedure & purification strategy

    Change expression system: insect, yeast, mammalian, in-vitro (RTS)

    Refolding

    Which domain? How many constructs to obtain

    a soluble domain?

    [IPTG], temperature, induction time, strain, etcChaperones: co-expression, heat shock, chemical chaperones

    What can we do at the expression level?

    Single target, or targets from the same familyPragmatism (HTS): random truncation

    Rational domain designDomain changes

    Limited proteolysis-based domain determination

    MBP, GST, thioredoxin, NusA,SUMO, OthersProteins smaller than 40kDa will be attached to MBP,

    whereas larger proteins will be attached to SUMO

    Change expression conditions

    Consider number of disulfide bridgesLarge of the protein

    How to check correct folding??

  • • Aggregation is considered to be mediated by short “aggregation

    prone” peptide segments

    • Software tools for prediction of APRs: AGGRESCAN, PAGE, TANGO,

    PASTA, SALSA, Zyggregator, and AMYLPRED

    • PROSS (Protein Repair One-Stop Shop): algorithm to optimize

    protein stability. Sarel Fleishman WIS http://pross.weizmann.ac.il/bin/steps

    Aggregation prone regions (APRs) of proteins

    http://pross.weizmann.ac.il/bin/steps

  • Pick host strain

    Pick induction procedure

    Test Purification conditions

    Gel-filtration analysis

    2 days

    2-3 days

    1-2 days

    1-2 days

    Quick expression strategy

    Best host strain

    with higher soluble yield

    and lower IB

    Best affinity binding

    and lower soluble

    aggregates!!!

  • Flowchart: Screening methodology for expressing prone-to-aggregate protein

    Diagnose possible reasons for aggregation

    Clone with 1-2 suitable fusion proteins containing His Tag

    Screen expression conditions in auto-induced media at 25, 30 and 37°C

    IMAC isolation of soluble protein: check by Coomassie staining and Western blot

    Transform to 3-5 selected bacterial strains

    Second round of optimization with selected strains

    Scale-up selected conditions and analyze on IMAC and analytical GF

  • Intrinsic disorder provides several advantages:

    1. Increased interaction surface area

    2. Bind numerous partner ligands via the same region

    3. Interaction may induce a folded conformation

    4. Different ligands may induce different structures

    Wright & Dyson J Mol Biol, 1999Tompa FEBS, 2005

    Uversky & Dunker Science ,2008

    The traditional paradigm:

    “sequence → structure → function”

    An updated paradigm should also include:

    “sequence → disorder → function”

  • Van der Waals and hydrophobic attractions between side-chain and

    backbone atoms

    maximizing hydrogen bonding

    minimizing steric clashes and energetically unfavorable bond

    torsional angles

    maximizing chain entropy

    minimizing (maximizing) electrostatic repulsions (attractions)

    minimizing unfavorable interactions between amino acids and the

    solvent (water) and its co-solutes

    Forces and interactions that drive foldingProtein Aggregation and Its Impact on Product Quality

    Christopher J. Roberts - Curr Opin Biotechnol. 2014 December ; 0: 211–217. doi:10.1016/j.copbio.2014.08.001.

  • Mechanisms of Protein Aggregation Philo J.: Current Pharmac. Biotech. (2009),

    10, 348-351 H.J. Lee et al. / Advanced Drug Delivery

    Reviews (2011)

    1) rapidly reversible non-covalent small oligomers

    2) irreversible non-covalent oligomers

    3) covalent oligomers(disulfide-linked)

    4) Nucleation aggregation

    5) Surface induce alterations

  • The initiation process of“non-native” aggregates

    involves:

    Two or more proteins “misfolding” together to a

    “nuclei” (stable/irreversible)

    Then grow via different mechanisms

    Nucleate aggregation mechanism

    Creation of nuclei as a consequence of other mechnism:

    Adsorption of proteins to hydrophobic interfaces such as those between water and

    air or silicone oil may promote unfolding and subsequent aggregation.

    Adsorption to charged interfaces such water in contact with container materials

    such as glass and stainless steel can also promote non-native aggregation

    Molecular aggregates can ultimately phase separate as macroscopic particles

  • Nucleation-growth process

    Temperature

    Time Protein concentration

    Buffer conditions

    Reversible – Irreversible

    Soluble – Insoluble

    Folded – Unfolded

    Covalent – Non-covalent

    Aggregation: type and characteristics

  • Downstream aggregates are also generated via mechanical stress during filtration, high temperature holds, and excessive air-liquid or solid-liquid interfaces

    H Doss et al. 2019 J. of Chromatography A -https://doi.org/10.1016/j.chroma.2019.03.044

    Common types of mAbs aggregation mechanismsMechanistic and process-related drivers

  • Aggregation: major challenge in therapeutic protein production

    The FDA generally recommends that MAb aggregates be reduced to

  • It is not currently possible to predict a priori when protein aggregation will occur for a given

    protein, BUT, there a number of factors that influence whether and/or how quickly protein

    solutions will aggregate.

    The major factors include :

    • solution conditions

    pH,

    salt concentration and which salt type,

    amount and type of osmolytes

    amphiphilic molecules such as surfactants;

    • temperature

    • pressure

    • air-water interfaces

    • bulk interfaces with water such as stainless steel

    How can one control or mitigate aggregation?

    Protein stabilizer factors

    Aggregation inhibitors protein-protein interaction

    reducers factors

  • Aggregation-prone region on thesurface of the native protein structure

    Each category of additive will improve solubility of some proteins while decreasing the solubility of others

    Osmolytes and kosmotropesenhance protein-protein

    interactions and may increase aggregation

    Chaotropes interact with aggregation-prone surface and improve solubility

    + OsmolytesOr Kosmotropes

    + Chaotropes

    Chaotropes

    Interacts preferentially with protein surfaces

    Such interactions stabilize the exposed surface of the

    protein, resulting in a decrease in the free energy of

    unfolding and an increase in the free energy of aggregation

  • Partially unfolded protein structure exposes hydrophobic residues

    Osmolytes and kosmotropeshide hydrophobic surface

    and therefore may enhance folding

    Chaotropes promote the unfolded (aggregation-

    prone) state

    + OsmolytesOr Kosmotropes + Chaotropes

    Kosmotropics

    Decreased the rate of protein unfolding

    from the native state

    The stabilizing effects can be explained by their

    preferential exclusion from protein surfaces, which

    thermodynamically led to the reduction of protein

    surface exposed to the solvent through unfavorable

    interactions between protein surfaces and additives

    Each category of additive will improve solubility of some

    proteins while decreasing the solubility of others

  • Ideal environment Yamaguchi et al. Biotechnol. J. 2013, 8, 17–31

    • Aggregation rate is decreased, and the native folded protein is stabilized

    (rather than undergoing a reverse transition to intermediate states)

    • Surfactants are most commonly used to protect proteins from shaking/shearing-

    induced aggregation. They inhibit protein aggregation by competing with proteins

    at hydrophobic surfaces.

    • Polysorbate 20 and polysorbate 80 (most common) undergo auto-oxidation

    yielding reactive peroxides, which cause degradation.

    • Sugars protect proteins against dehydration by forming hydrogen bonds with the

    protein (as water substitutes). This increase thermal unfolding temperature and

    inhibits irreversible aggregation of protein molecules

    • Stabilizers do not only stabilize proteins but simultaneously can enhance

    aggregation. So, stabilizers can be used in combination with aggregation inhibitors

    S. Kale and K. Akamanchi - Mol. Pharmaceutics 2016, 13, 4082−4093

  • Additive

    Recommended Initial

    Concentration

    Recommended

    Concentration Range

    Sugars and Osmolytes

    Glycerol 10% 0-40%

    TMAO (trimethylamine N-oxide) 0.5M 0-1 M

    Glucose 0.5M 0-2 M

    Sucrose 0.5M 0-1 M

    Trehalose 0.5M 0-1 M

    Ethylene glycol 10% 0-60%

    D-Sorbitol 0.5M 0.2-1M

    Mannitol 2%

    Xylitol 0.5M 0.2-1M

    Glycine Betaine 1M

    Amino acids and amino acid derivatives

    Glycine 250mM 0.5-2%

    Arginine L-HCl 125mM 0-2 M

    Arginine Ethylester 250mM 0-500 mM

    Proline 250mM 0-1 M

    Potassium Glutamate 250mM 0-500 mM

    Arginine L-HCl + L-glutamic acid (L-Glu) 50mM each

    Non-ionic detergents

    Nonidet P40 (NP40) or Triton X-100 0.01% 0-1%

    Tween 80 or 20 0.1% 0-1%

    DDM: n-Dodecyl β-D-maltoside 0.1% 0.01- 0.5%

    Brij 56: Polyoxyethylene cetyl ether 0.05%

    OG: Octyl glucoside (n-octyl-β-D-glucoside) 0.1% 0.01- 0.5%

    Poloxamer 188 (BASF Pluronic® F68)

    Zwiterionic detergents

    NDSB: Non-detergent Sulfo Betaine 0.5M 0-1M

    CHAPS: 3-[(3-cholamidopropyl)dimethylammonio]-1-

    propanesulfonate

    0.1% 0.01- 0.5%

    Zwittergent 3-14 0.1% 0.001-0.2 %

    LDAO: Lauryldimethylamine N-oxide 0.1% 0.01- 0.5%

    Ionic detergents

    CTAB: cetyltrimethylammonium bromide 0.5%

    Sarkosyl : Sodium lauroyl sarcosinate 0.05% 0.01-0.5%

    SDS: Sodium dodecyl sulfate Up to 0.1%

    Table: Additives Used

    to Stabilize Folding and to

    Prevent AggregationSummary table of different

    publications (De Bernardez Clark 1999, Voziyan 2000, Goloubinoff2001, Bondos 2003, Golovanov

    2004, Hamada 2009, Shukla 2011, Churion 2012, Leibly 2012) and

    from commercial websites (DILYX Biotechnologies OptiSol Protein

    Solubility Screening Kit Application Manual, HAMPTON: Solubility &

    Stability Screen)

    http://www.dilyx.com/protein_solubility_screen_home2http://hamptonresearch.com/default.aspx

  • Mild chaotrope agents and chaotrope salts

    Urea 0.5M 0-2M

    Guanidine HCl 0.5M 0-2M

    N-Methylurea 250mM up to 2.5M

    N-Ethylurea 100mM up to 2M

    N-Methylformamide 3-15%

    NaI 0.2M 0-0.4 M

    CaCl2 10-50 mM 0-0.2M

    MgCl2 10-50 mM 0-0.2M

    Mild and strong kosmotrope salts

    NaCl (weak) 300mM 0-1 M

    KCl (weak) 200mM 0-1 M

    MgSO4 (strong) 100mM 0-0.4 M

    (NH4)2SO4 (strong) 50mM 0-0.2M

    Na2SO4 (strong) 500mM 0-0.2M

    Cs2SO4 (strong) 50mM 0-0.2M

    Potassium citrate 100mM

    Citric Acid 50mM

    Alcohols, Polyols, Polymers, Polyamines, and others

    Ethanol 5-10% Up to 25%

    n-Penthanol 1 to 10mM

    n-Hexanol 0.1 to 10mM

    Cyclohexanol 0.01 to 10mM

    Polyethylene glycol (PEG 3350) 0.3-1.5% 0.1-0.4 g/L

    Polyvinylpyrrolidone 40 (PVP40) 0.05-4%

    Alpha-Cyclodextrin 8-40mM

    Beta-Cyclodextrin 1-5mM

    Putrescine, spermidine, and spermine 0.1M

    Formamide 0.1%

    Reducing Agents

    β-mercaptoethanol (BME) 2 to 5mM 1 to 10mM

    Dithiothreitol (DTT) 1mM 0.1 to 10mM

    tris(2-carboxyethyl)phosphine (TCEP) 1 to 5mM 1 to 50mM

    Table: Additives Used

    to Stabilize Folding and to

    Prevent AggregationSummary table of different

    publications (De Bernardez Clark 1999, Voziyan 2000, Goloubinoff2001, Bondos 2003, Golovanov

    2004, Hamada 2009, Shukla 2011, Churion 2012, Leibly 2012) and

    from commercial websites (DILYX Biotechnologies OptiSol Protein

    Solubility Screening Kit Application Manual, HAMPTON: Solubility &

    Stability Screen)

    http://en.wikipedia.org/wiki/TCEPhttp://www.dilyx.com/protein_solubility_screen_home2http://hamptonresearch.com/default.aspx

  • Flowchart: Screening methodology showing the best additives for purifying prone-to-aggregate proteins

    Parallel bacterial lysis with different families of additives

    Small-scale IMAC purification in the presence of different additives

    Expand the screening of selected families of additives

    Final optimization step: minimize the additive concentration Starting from the lysis

    Starting from the wash & elution

    Non-ionic detergent (0.5%)Zwittergent (0.5%) Osmolyte (0.5M) Urea or GuHCl (1M)Add 0.5M Arg or 50mM Glu/Arg

    only in the elution bufferControl

    Analysis of partially purified protein after overnight incubation at 4°C

    Buffer with high salt (0.5M) + 10% glycerol +/- reducing agents

    a) Supernatant b) pellet c) unbound wash to IMAC and d) eluted protein

    Analytical SEC of the best conditions

    Visual selection of non-turbid

    samples

    Short spin and PAGE-

    SDS analysis

    0

    50

    100

    150

    mAU

    40 60 80 100 120 ml

    Or use a combination of families

  • Best conditions during purification are not necessarily the same for

    other steps like protein concentration or long term stability storage

    Stable conditions after ON incubation: best monomeric / soluble aggregation ratio

    Final Goal of the screening for best additives

    Optimal IMAC conditions:least protein in the unbound fraction and

    higher elution yield

    Selection of optimal lysis conditions: least protein in pellets

  • Example of screening

    methodology

    Ronen Gabizon / Assaf Friedler lab

  • un

    b

    Guanidine

    E1 E2 un

    b

    E2E1

    Urea

    un

    b

    E2E1

    Tween

    un

    b

    Zwitergent 3-14

    E1E2 u

    nb

    E2E1Trehalose

    un

    b

    E2E1

    L-Arg

    Additives

    Example of screening methodology: different additives

    Additives

    HMTV

    HMTV

    Ihab abd-Elrahman / Ben Yehuda Dina lab

  • un

    bGuanidine-HCl

    E1 E2

    HMTV

    HMTV

    E1 E2 E1 E2

    0.2 0.05 0.001

    Tween20 0.05 0.005 0.0001

    E1 E2 E1 E2 E1 E2un

    bExample of screening methodology: additive concentration & time

    Aggregation screening after 24hs 4C

    HMTV

    HMTV

    0.2 0.05 0.001

    Tween20

    0.05 0.005 0.0001

    P S P S P S

    P S P S P S

    Aggregation screening: additive concentration

    Guanidine -HCl

  • MTVHMTV

    • O.N Dialysis with 10mM imidazole and 0.02 M Guanidine and with TEV

    After Negative Ni

    Before TEV

    Example of screening methodology: cut with TEV

  • 1M Guanidine HCl 1M Urea 0.5% Tween 20 0.5% Zwitergent (3-14)

    500mM L-ArgHCl 50mM L-ArgHCl + 50Mm GlutAcid Guy Mayer / Assaf Friedler lab

    Example of screening methodology: different

    additives. 1st optimization

  • Example of screening methodology: different additives. 2nd optimization

    Guy Mayer / Assaf Friedler lab

    Lysis, load, wash and elution in the presence of 1M GuHCl

    Lysis & load in the presence of 1M GuHCl , wash & elution, add 50mM Arg & Glu after elution

  • HLT-VIP1 purification: Buffer screen

    50mM MES pH 6.0 50mM Hepes pH 8.0

    200mM 500mM 200mM 500mM

    Elution buffer

    no additives

    Arg 500mM

    Arg + Glu 50 + 50mM

    Tween20 1%

    Tween80 1%

    Zwittergent 1%

    Trehalose 1M

    TMAO 1M

    LDAO 0.1%

    CHAPS 0.1%

    NDSB-201 0.5M

    Urea 1M

    GndChl 1M

    120102HLTVIP1Superose12prepEndwash001:1_UV1_280nm 120102HLTVIP1Superose12prepEndwash001:1_UV2_260nm 120102HLTVIP1Superose12prepEndwash001:1_UV3_220nm 120102HLTVIP1Superose12prepEndwash001:1_Fractions 120102HLTVIP1Superose12prepEndwash001:1_Inject 120102HLTVIP1Superose12prepEndwash001:1_Logbook

    0

    100

    200

    300

    400

    mAU

    60 80 100 120 140 160 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28

    Lysisbuffer50mM Hepes pH 8.0500mM NaCl10% glycerol1M urea

    NiNTA Batch binding

    Superose 12 prep SEC: same buffer without Urea

    Michal Maes / Assaf Friedler lab

  • Lack of reducing agents

    Ohad Solomon/ Assaf Friedler lab

    Lysis and IMAC purification in buffer without reducing agents and 0.3M NaCl

    Overnight dialysis with low conductivity (50mM NaCl) and AEIX in the presence of 1mM DTT

    Lysis and IMAC purification in buffer with reducing agents and 0.3M NaCl.Add to peak 10mMDTT 50mMArg/Gluand load immediately on SEC in the presence of 1mM DTT & 0.3M NaCl

  • Integrase purification: Buffer screen

    pHis2IN2mut(1-288)

    Grow conditions: Rosetta ON IPTG indiction at 22C

    Batch binding, and wash 20% buffer B

    Buffers A IMAC: HEPES 20 mM pH7.5, 1M NaCl, 10%

    glycerol, 25 mM Imidazole, 2 mM βME (added fresh)

    B: A + 500 mM Imidazole

    Add 2mM EDTA to avoid cloudiness

    Concentrate and add TEV protease 1:10 because of

    the high NaCl under dial buffer A + 50mM Imid

    Negative IMAC

    Gel filtration buffer: HEPES 20 mM pH 7.5, 1 M NaCl,

    10% glycerol, 0.1 mM EDTA, 1 mM DTT

    Concentrate and aliquot. Keep -80C

    150 200 250 300 350 400-10

    0

    10

    20

    30

    40

    50

    60

    mA

    U

    Volume (ml)

    280 nm

    260 nm

    IN(1-288)

    Tzvika Ayhuka and Ronen Gabizon / Assaf Friedler lab

  • Conclusion: Important factors to consider during purification of prone to aggregate proteins

    Protein specific issues: additives, conductivity, buffer, others.

    General Issues as:

    Purification time and Low temperature

    Protein concentration in each step (ratio lysis buffer / cell or

    overloading column, etc)

    Rational selection and arrangements of methods and columns

    Prevention of mechanical or not mechanical stresses to the protein

    such as: freezing / thawing

    exposure to air

    interactions with metal surfaces

  • 150 200 250 300 350 400 450 500-50

    0

    50

    100

    150

    200

    250

    300

    OD

    (m

    AU

    )

    ml

    280 nm 260 nm

    100 150 200 250 300 350 400 450 500

    0

    250

    500

    750

    1000 280 nm 260 nm

    OD

    (m

    AU

    )

    ml

    %B

    0

    20

    40

    60

    80

    100

    300 350 400 450 500 550 600

    0

    500

    1000

    1500

    2000

    2500

    mA

    U

    ml

    % B

    0

    20

    40

    60

    80

    100

    100 150 200 250 300 350 400 450

    0

    100

    200

    300

    400

    500

    600

    700

    mA

    U

    ml

    TEV protease production: buffer optimization

    Capture Ni Column Polishing GF step

    Increase lysis buffer volume

    More DNase A in the lysis buffer

    Increase [NaCl] from 0.1 to 0.5

    Ronen Gabizon / Assaf Friedler lab

  • Strategy: Effect of concentration and time on protein aggregation

    Sil5NiSeph4ml002:1_UV1_280nm Sil5NiSeph4ml002:1_UV2_260nm Sil5NiSeph4ml002:1_Conc Sil5NiSeph4ml002:1_Fractions Sil5NiSeph4ml002:1_Inject Sil5NiSeph4ml002:1_Logbook

    0

    100

    200

    300

    400

    mAU

    0 50 100 150 200 ml

    F3 F4 1 2 3 4 5 6 7 8 9 10 1112 13 14 15 16 17 18 19

    pool 13-15 (12ml) was loaded on sephacryl S200

    next time: longer steps

    Sil6Superdex75prep001:1_UV1_280nm Sil6Superdex75prep001:1_UV2_260nm Sil6Superdex75prep001:1_Fractions Sil6Superdex75prep001:1_Inject Sil6Superdex75prep001:1_Logbook

    -10.0

    0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    mAU

    60 80 100 120 140 160 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39

    97ml = 160kDa

    Next time: Superose 12 or Sephacryl S200

    Sil6SephS200prep001:1_UV1_280nm Sil6SephS200prep001:1_UV2_260nm Sil6SephS200prep001:1_Fractions Sil6SephS200prep001:1_Logbook

    -10.0

    0.0

    10.0

    20.0

    mAU

    150 200 250 300 350 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50

    264ml =50kDa

    Ni column after lysis. Buffers with glycerol and high [NaCl]

    Sephacryl S200 100x2.6cm ~500ml column inmediately after Ni purification

    Superdex 75 100x1.6cm ~200ml column after Ni purification and several hours of

    concentration

    Processing time is critical!!!!Protein concentration between

    IMAC and GF sometimes is problematic

    Michal Maes / Assaf Friedler lab

  • HLTiASPPprolRichSephacrylS200Prep001:1_UV1_280nm HLTiASPPprolRichSephacrylS200Prep001:1_UV2_260nm HLTiASPPprolRichSephacrylS200Prep001:1_Inject HLTiASPPprolRichSephacrylS200Prep001:1_Logbook

    0

    200

    400

    600

    800

    1000

    mAU

    100 150 200 250 300 350 ml

    HLTiASPPprolRichSephacrylS200Prep001:1_UV1_280nm HLTiASPPprolRichSephacrylS200Prep001:1_UV2_260nm HLTiASPPprolRichSephacrylS200Prep001:1_UV3_220nm HLTiASPPprolRichSephacrylS200Prep001:1_Logbook

    -50

    0

    50

    100

    150

    mAU

    100 150 200 250 300 350 ml

    HLTiASPPAnkSH3NiSepharFF16ml001:1_UV1_280nm HLTiASPPAnkSH3NiSepharFF16ml001:1_UV2_260nm HLTiASPPAnkSH3NiSepharFF16ml001:1_Conc HLTiASPPAnkSH3NiSepharFF16ml001:1_Inject HLTiASPPAnkSH3NiSepharFF16ml001:1_Logbook

    0

    500

    1000

    1500

    2000

    2500

    3000

    3500

    mAU

    300 350 400 450 500 550 ml

    Two big changes:

    1) Use X 2 more IMAC resin

    2) Separate aggregates immediately after IMAC

    Strategy: Avoid column overloading & speed process

    IMAC captureSephacryl S200 100x2.6cm ~500ml

    For prone to aggregation proteins sometimes is better to avoid column overloading

    Michal Maes / Assaf Friedler lab

  • DefrozeniASSPAnkSH3Superose12 Anal002:1_UV1_280nm DefrozeniASSPAnkSH3Superose12 Anal002:1_UV2_260nm DefrozeniASSPAnkSH3Superose12 Anal002:1_UV3_220nm DefrozeniASSPAnkSH3Superose12 Anal002:1_Fractions DefrozeniASSPAnkSH3Superose12 Anal002:1_Inject DefrozeniASSPAnkSH3Superose12 Anal002:1_Logbook

    -5.0

    0.0

    5.0

    10.0

    15.0

    20.0

    25.0

    mAU

    6.0 8.0 10.0 12.0 14.0 16.0 18.0 20.0 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 Waste

    GF of Defrozen iASSP Ank SH3

    Strategy: Should TEV protease be cleavage before or after GF separation of soluble aggregates?

    Try to eliminate your soluble aggregates as soon as possible!!!

    Only monomer undergoes TEV protease cleavage(aggregates can not be cleaved)

    HLTiASPPAnkSH3Superose12Prep001:1_UV1_280nm HLTiASPPAnkSH3Superose12Prep001:1_Inject HLTiASPPAnkSH3Superose12PrepDelay002:1_UV1_280nm HLTiASPPAnkSH3Superose12Prep001:1_Logbook

    0

    20

    40

    60

    80

    100

    mAU

    60 80 100 120 140 ml

    Load inmediately after affinity

    Load 12hours after affinity

    After TEV cleavage, monomers are stable

    Load immediately after Ni affinity

    Load 12h after Ni affinity

    +-

    Anat Iosub / Assaf Friedler lab

  • Column selection: develop the best strategy to avoid further aggregation

    Ni column

    TEV protease + dialysis

    Negative Ni column

    GF

    Concentration

    Ni column

    GF immediately

    TEV protease

    Negative Ni column

    and/or Ion exchange

    REVIEW Nature Methods - 5, 135 - 146 (2008)Protein production and purification

    Structural Genomics Consortium

  • chelating2mlAir Imp7Beta001 18 12 12001:1_UV1_280nm chelating2mlAir Imp7Beta001 18 12 12001:1_UV2_260nm chelating2mlAir Imp7Beta001 18 12 12001:1_Conc chelating2mlAir Imp7Beta001 18 12 12001:1_Fractions chelating2mlAir Imp7Beta001 18 12 12001:1_Logbook

    0

    20

    40

    60

    80

    100

    %B

    120.0 130.0 140.0 150.0 160.0 170.0 180.0 190.0 ml

    F4 1 F2 2 3 4 5 6 7 8 9 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 3132

    Fractions 16-22 Peak-833 mAU

    100%B

    20%B

    15%B

    Chelating2mlAir, Imp7598 ImpBeta1-442 from 1L 17C ON 19.12.12

    Imp7BetaSuperose12prepar200ml 18 12 12001:1_UV1_280nm Imp7BetaSuperose12prepar200ml 18 12 12001:1_UV2_260nm Imp7BetaSuperose12prepar200ml 18 12 12001:1_Fractions Imp7BetaSuperose12prepar200ml 18 12 12001:1_Inject Imp7BetaSuperose12prepar200ml 18 12 12001:1_Logbook

    0

    20

    40

    60

    80

    100

    120

    140

    mAU

    60 80 100 120 140 160 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38

    Extra ImpBeta Peak - 106ml

    Complex Peak - 87ml

    Aggregate Peak - 65ml

    Superose12prepar200ml Imp7 598 ImpBeta1-442 + Tween20 0.01% after Ni 20/12/12 Imp7BetaSuperdex75prep320ml 25 04 12001:1_UV1_280nm Imp7BetaSuperdex75prep320ml 25 04 12001:1_UV2_260nm Imp7BetaSuperdex75prep320ml 25 04 12001:1_Fractions Imp7BetaSuperdex75prep320ml 25 04 12001:1_Logbook

    -10.0

    0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    60.0

    mAU

    80 100 120 140 160 180 200 ml

    1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28

    Peak2: 182 ml

    Peak1: 137 mlPeak agregate: 115 ml

    Imp7 (598-C) + Imp Beta (1-442) after Ni and conc - Superdex75prep320ml 25.04.12

    Optimization of Complex Formation

    Co-Purification + Buffer optimization

    Capture: IMAC column

    Superose 12 GF

    Komornik Nadav / Oded Livnah lab

  • Conclusions

    A simple and modular toolbox for designing a minimized

    expression and purification screen

    Protein production in E.coli-based systems following these

    hierarchic rules is more efficient, simpler, and less costly

    Minimized hierarchical screening platform

    Combination of most beneficial approaches:

    rational domain design possible fusion partners

    suitable bacterial hosts optimal induction conditions

  • Conclusions

    Main goal at the expression level is to obtain maximal yield of the correct

    oligomeric conformation along with the minimal presence of insoluble proteins,

    or soluble aggregates, in the shortest possible time.

    Selection of optimal lysis conditions: the least protein in pellets

    Reduce screen to one additive of each family

    Optimal IMAC conditions: least protein in the unbound fraction and a higher

    elution yield

    Best monomeric/soluble aggregation ratio after overnight incubation

    Consider and optimize all the factors that can affect solubility during the

    processing before scale-up: chromatography, concentration and storage

  • It looks difficult!!!But it works….

    Review: Production of prone-to-aggregate proteins

    Mario Lebendiker , Tsafi Danieli

    FEBS Lett. (2014), http://dx.doi.org/10.1016/j.febslet.2013.10.044

    Chapter: A screening methodology for purifying proteins with aggregation

    problems

    Mario Lebendiker, Michal Maess and Assaf Friedler (2015)

    Methods in Molecular Biology (Springer)"Insoluble Proteins“ Vol.1258, pp 261

    http://dx.doi.org/10.1016/j.febslet.2013.10.044

  • Monodispersity, aggregation state

    SEC

    SEC-MALS

    Native PAGE

    Ultrafiltration assays

    Dynamic Light Scattering

    Analytical Ultracentrifugation

    How to check aggregation and stability

    Biological and/or specific activity is extremely important, but, it does not always reflect all the picture

    Stability

    Differential Scanning

    Calorimetry

    Circular Dichroism

    Termal Shift Assay

    Dynamic Light Scattering

    DSF: Differential Scanning Fluorimetry

  • Screens in 24 or 96 wells (or microarrays), covering a range of conditions to be

    tested (pH, salts, additives)

    Protein is mixed with each solution and then screened visually, under a

    microscope or by different analytical methods

    Methods for Screening Solubility Conditions

    PAGE-SDS

    Western blot

    Intrinsic protein fluorescence

    Visual screen: from clear soluble drop to light precipitation, heavy

    aggregation, phase separation (like oil in water), or small crystals

    Light Scattering

    Biological Activity

    Others Circular Dichroism (CD)

    Differential Scanning CalorimetryStability

    Self-Interaction Chromatography (SIC) HSC Technologies Soluble Therapeutics

    Native PAGE

    Spectrophotometer

    Extrinsic dye fluorescence

    Monitor over time or different temperatures or with chemicals (H2O2, urea, etc)

  • Native PAGE

    Like regular SDS-PAGE, but samples are not denatured

    Proteins separate based on size and shape

    Aggregation appears as streaks instead of a discrete band

    Not easy. Not always work. Artifacts

  • Thermal Shift Assay

    • Can be high-throughput

    • Monitor by light scattering or fluorescence

    • Fluorescence: – Upon denaturation, dye binds

    exposed hydrophobic portions of the protein

    – Can use real time PCR machine

    • Can be adapted to search for ligands that bind

    http://www.invitrogen.com/site/us/en/home/Products-and-Services/Applications/PCR/real-time-pcr/real-time-pcr-applications/real-time-pcr-protein-analysis/protein-thermal-shift.html

  • A Novel Protein Aggregation Assay for Biologics Formulation Studies and Production

    ENZO-BioTek: QA/QC - ProteoStat® Fluorescent Reagent for Microplate-based

    Aggregate Quantitation

  • The filter assay detects both soluble and insoluble aggregates

    1. Dilute protein

    1:5 or 1:10 into

    test buffer, final

    volume ~100 µl.

    2. Incubate at

    room tempera-

    ture 1 hour.

    3. Apply protein

    to top of ultrafilter

    unit, spin at 16,000g

    for 15 minutes.

    4. Resuspend re-

    tained aggregates

    in 30 µl H2O.

    5. Detect protein

    in filtrate (Sol) and

    retentate (Agg) by

    SDS-PAGE.

    Bondos & Bicknell (2003) Anal. Biochem. 316 223-231

    Churion & Bondos (2011) Methods Mol. Biol. 896, 415-427

  • OptiSol™ Protein Solubility Screening Kit

    • Filtration assay from Dilyx Biotechnologies

  • Optim 1000 from Avacta

    • Uses an instrument that combines, the powerful analytical capabilities of fluorescence and static light scattering technologies

    • High throughput

    • Measures unfolding temperature and aggregation temperature

  • • When laser light hits a macromolecule, the oscillating electric field of the light induces an oscillating dipole that re-radiates light.

    • The intensity of the radiated light depends on the magnitude of the dipole.

    • The intensity of the scattered light is proportional to the macromolecule concentration.

    • When monomers form a dimer, the intensity of the scattered light increases.

    • By increasing the mass, while keeping the concentration the same, the intensity of the scattered light increases.

    MALS principles(Multi Angle Light Scattering)

    55

    LS- intensity of scattered light

    c – concentration

    K – a constant for a specific solute in a solution

  • Static & Dynamic Light Scattering (LS and DLS) non-invasive measurement for mass and radius

    Light hitting small (

  • Hydrodynamic radius vs Mw Sabine Supman / Max Plank / P4EU course

  • PeakRadius (nm)

    Mw-R (kDa) %Pd %Intensity %Mass %Number

    Peak 1 (True) 8.5 509.2 19.3 21.7 39.7 100.0

    Peak 2 (True) 45.3 25296.0 22.9 55.6 1.4 0.0

    Peak 3 (True) 5546.2

    1938105428.8 28.0 22.7 58.9 0.0

    MBP: highly concentrated sample frozen , unfiltered in 50% glycerol

    DLS of fresh vs frozen sample Sabine Supman / Max Plank / P4EU course

    Radius (nm) %Pd Mw-R (kDa) %Intensity %Mass

    3.9 18.9 78.9 4.0 91.5

    25.0 49.0 6309.9 96.0 8.5

    MBP: highly concentrated sample before freezing

  • Size Exclusion Chromatography (SEC)Size, oligomeric conformation, aggregations, & purity.

    Particles separated by size

    Smaller particles trap in beads and elute later

    Larger particles avoid beads and elute first

    Mini analytical columns for fast results

    Resolution to discriminate monomers from oligomers

    Underestimation of aggregates due to Removal of aggregates by frits and column filtration

    Dilution of sample and thus potential dissociation of reversibly aggregated species

    Non-specific interaction of unfolded, exposed hydrophobic surfaces with the matrix

    Stickyness of properly folded proteins (e.g ß-sheet)

    01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV1_280nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV2_260nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_UV3_220nm 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Cond 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Fractions 01072014 MBP AA NleD short Superose 12 prep200ml001:10_Inject

    -20.0

    -10.0

    0.0

    10.0

    20.0

    30.0

    40.0

    50.0

    60.0

    mAU

    60 80 100 120 140 ml

    F3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31

    HLTp53CTSephacrylS100of500ml004:1_UV1_280nm HLTp53CTSephacrylS100of500ml004:1_UV2_260nm HLTp53CTSephacrylS100of500ml004:1_Fractions HLTp53CTSephacrylS100of500ml004:1_Inject HLTp53CTSephacrylS100of500ml004:1_Logbook

    0

    50

    100

    150

    mAU

    150 200 250 300 350 400 ml

    F3 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

    Fractions around 23 and 32 are higher MW impurities

    POOL 7-14 Difficult HTS!!Reliable QC for low to relatively high MW aggregates

  • DSF: Differential Scanning FluorimetryNanoTemper

    Method for measuring ultra-high resolution protein stability using

    intrinsic tryptophan or tyrosin fluorescence.

    Applications include antibody engineering, membrane protein research,

    formulation and quality control.

    Protein solutions can be analyzed independent of buffer compositions

    (concentration range from more than 200 mg/ml down to 5 µg/ml)

  • Methods to quantify the structural stability of a

    protein: thermal and chemical unfolding

    Thermal unfolding experiments use a constantly

    increasing temperature to monitor protein

    conformational changes over time

    Chemical unfolding experiments employ

    denaturing agents to unfold proteins

    Thermal unfolding occurs over a narrow

    temperature range

    Transition from folded to unfolded - referred to

    as 'melting temperature' or 'Tm' - serves as a

    measure for protein stability

    DSF: Differential Scanning FluorimetryNanoTemper

  • DSF: Differential Scanning FluorimetryNanoTemper

    Fluorescence of the tryptophans (and

    tyrosins) in a protein is strongly dependent on

    their close surroundings

    Changes in protein structure affect both the

    intensity and the emission wavelength of Trp

    fluorescence

    Tm is calculated from the

    changes in tryptophan

    fluorescence intensity, or from

    the ratio of tryptophan

    emission at 330 and 350 nm

  • A) Establishing a protein unfolding standard

    B) Long-term storage of membrane protein

    C) Forced-degradation stress-test on MEK1

    A) Unfolded IgG at different concentrations was mixed with folded IgG and subject to thermal unfolding. The percentage of unfolded IgG in the solution was quantified based on the F350/F330 ratio meassured at 25 °C.

    B) Membrane protein HiTeha were stored at 4 °C and at RT, respectively, and thermal unfolding curves were measured over a time period of 34 days. %unfolded protein was calculated based on the F350/F330 ratio

    C) MEK1 protein was subject to the indicated stresses, and the fraction of unfolded protein was calculated based on the F350/F330 ratio at 25 °C

    Exploring Protein Stability by nanoDSF - Prometheus NT.48

    Melanie Maschberger et al. 2015 - NanoTemper Technologies GmbH, Munich, Germany

  • Type of Stressor Example for Experimental Stress

    Elevated Temperature Incubate 24 hours at 37°C

    Long-Term Storage Store 2 weeks at room temperature

    Ice/Liquid Transition Freeze and thaw 20 times

    Shear Force Force material 20 x through narrow syringe need

    Intense Light Expose samples to direct sunlight or UV light for 1 h

    Chemical Compatibility Add 10 mM of caustic reagent (i.e. heavy metal)

    Surface Exposure Add 5 uL of 10 um diameter glass beads

    Air Oxidation Bubble 10 ml of air through sample

    Chemical Oxidation Add Hydrogen Peroxide

    Experimental stressesOptiSol™ - Protein Solubility Screening Kit Application Manual http://www.dilyx.com/protein_solubility_screen_home2

    http://www.dilyx.com/protein_solubility_screen_home2

  • rAlbumin (15 mg/ml)

    Glycine (20 mg/ml)

    PEG 400 (1 mg/ml)

    Ethylene glycol (10%)

    Polysorbate 80 - Tween-80 (0.82 mg/ml to 8.2 mg/ml) oxidation problems

    Arginine (400mM)

    Histidine (400mM)

    Mannitol (10%)

    Trehalose (500mM)

    TMAO (1.0M)

    Arg/Glu: each amino acid is 50mM

    Excipients applied in the industry to enhance protein stability or avoid aggregation

  • Potential Stabilizers of Influenza Virus-Like ParticlesAggregation

    Kissmann J. et al. Journal of Pharmaceutical Sciences, Vol. X, 1–12 (2010)

    Ascorbic acid 0.15 M

    Aspartic acid 0.075 M

    Lactic acid 0.15 M

    Malic acid 0.15 M

    Arginine 0.3 M

    Diethanolamine 0.3 M

    Guanidine HCl 0.3 M

    Histidine 0.3 M

    Lysine 0.3 M

    Proline 0.3 M

    Glycine 0.3 M

    Brij 35 0.01%

    Brij 35 0.05%

    Brij 35 0.10%

    Tween 20 0.01%

    Tween 20 0.05%

    Tween 20 0.10%

    Tween 80 0.01%

    Tween 80 0.05%

    Tween 80 0.10%

    Pluronic F-68 0.01% Pluronic F-68 0.05% Pluronic F-68 0.10%Albumin (human) 1% Albumin (human) 2.5%Albumin (human) 5% Gelatin (porcine) 2.5% Gelatin (porcine) 5% Lactose 10% Lactose 15%Lactose 20%Trehalose 10%Trehalose 15%Trehalose 20%Sucrose 10%Sucrose 20%

    Dextrose 10%Dextrose 15%Dextrose 20% Mannitol 10%Sorbitol 10%Sorbitol 15%Sorbitol 20%Glycerol 5%Glycerol 10%Glycerol 15%Glycerol 20%α-Cyclodextrin 2.5%2-OH propyl β-CD 5%2-OH propyl β -CD 10%2-OH propyl γ-CD 5%2-OH propyl γ-CD 10%


Recommended